1
|
MITF Downregulation Induces Death in Human Mast Cell Leukemia Cells and Impairs IgE-Dependent Degranulation. Int J Mol Sci 2023; 24:ijms24043515. [PMID: 36834926 PMCID: PMC9961600 DOI: 10.3390/ijms24043515] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Activating mutations in KIT (CD117) have been associated with several diseases, including gastrointestinal stromal tumors and mastocytosis. Rapidly progressing pathologies or drug resistance highlight the need for alternative treatment strategies. Previously, we reported that the adaptor molecule SH3 binding protein 2 (SH3BP2 or 3BP2) regulates KIT expression at the transcriptional level and microphthalmia-associated transcription factor (MITF) expression at the post-transcriptional level in human mast cells and gastrointestinal stromal tumor (GIST) cell lines. Lately, we have found that the SH3BP2 pathway regulates MITF through miR-1246 and miR-5100 in GIST. In this study, miR-1246 and miR-5100 were validated by qPCR in the SH3BP2-silenced human mast cell leukemia cell line (HMC-1). MiRNA overexpression reduces MITF and MITF-dependent target expression in HMC-1. The same pattern was observed after MITF silencing. In addition, MITF inhibitor ML329 treatment reduces MITF expression and affects the viability and cell cycle progression in HMC-1. We also examine whether MITF downregulation affected IgE-dependent mast cell degranulation. MiRNA overexpression, MITF silencing, and ML329 treatment reduced IgE-dependent degranulation in LAD2- and CD34+-derived mast cells. These findings suggest MITF may be a potential therapeutic target for allergic reactions and deregulated KIT mast-cell-mediated disorders.
Collapse
|
2
|
Molecular Features of Preinvasive and Invasive Vulvar Neoplasms. J Low Genit Tract Dis 2023; 27:40-46. [PMID: 36083687 DOI: 10.1097/lgt.0000000000000701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Neoplasms arising from the vulva are uncommon and comprise various subtypes. Given the recent advancements in the molecular aspects of oncologic pathology and how they have impacted cancer treatment, an understanding of recent innovations in the molecular features of vulvar lesions is important. MATERIALS AND METHODS Systematic literature search was performed on PubMed, Google Scholar, and Scopus databases for molecular and genetic characteristics of vulvar neoplasms. Peer-reviewed literature published in English is included. RESULTS Squamous cell carcinoma (SCC) and its precursors are the predominant neoplasm at this site. Human papillomavirus (HPV) plays a crucial role in the pathogenesis of some of these lesions. Human papillomavirus-associated SCC follows the carcinogenic pathway driven by viral proteins E6 and E7 while HPV-independent SCC shows a high incidence of mutation of TP53 and CDKN2A genes. Mutations in the genes involving the PI3K-Akt pathway play an important role in the pathogenesis of both types of SCC. Among other vulvar malignancies, melanoma, and vulvar Paget disease (VPD) pose a significant clinical challenge and have unique molecular characteristics. Compared with dermal cutaneous melanoma, vulvar melanoma shows a higher rate of mutation of cKIT and NRAS genes and a lower rate of mutations in BRAF . Less than 20% of VPD shows amplification of ERBB2 and seldom shows mutation in genes involving the PI3K-Akt pathway. CONCLUSIONS Several potentially targetable molecular pathways have emerged as they have been shown to be involved in the tumorigenesis of SCC, melanoma, and VPD.
Collapse
|
3
|
Mahumud RA, Shahjalal M. The Emerging Burden of Genetic Instability and Mutation in Melanoma: Role of Molecular Mechanisms. Cancers (Basel) 2022; 14:cancers14246202. [PMID: 36551688 PMCID: PMC9776466 DOI: 10.3390/cancers14246202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Melanoma is a severe skin cancer affecting thousands of people and a growing public health concern worldwide. The potential hallmarks of melanoma are genetic instability and mutation (GIAM), which are driving mechanisms for phenotypic variation and adaptation in melanoma. In metastatic melanoma, DNA repair-associated genes are frequently expressed at higher levels than in primary cancers, suggesting melanoma cells rely on genetic stability to spread distantly. The tumour microenvironment is affected by genomic instability and melanoma mutation (GIMM), which plays significant roles in developing GIMM and their contributions to the overall disease burden. The GIAM is the crucial vulnerability of cancer cells, determining their sensitivity to harmful treatments, including radiation and many chemotherapeutics. The high incidence of melanoma is typically associated with genetic modifications, and several clinical and genetic interventions have been critical in easing the burden.
Collapse
Affiliation(s)
- Rashidul Alam Mahumud
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Correspondence:
| | - Md. Shahjalal
- Department of Public Health, North South University, Dhaka 1229, Bangladesh
| |
Collapse
|
4
|
Lorenz A, Kozłowski M, Lenkiewicz S, Kwiatkowski S, Cymbaluk-Płoska A. Cutaneous Melanoma versus Vulvovaginal Melanoma-Risk Factors, Pathogenesis and Comparison of Immunotherapy Efficacy. Cancers (Basel) 2022; 14:5123. [PMID: 36291906 PMCID: PMC9600893 DOI: 10.3390/cancers14205123] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 12/01/2022] Open
Abstract
Cutaneous melanoma is a relatively common neoplasm, with fairly well understood pathogenesis, risk factors, prognosis and therapeutic protocols. The incidence of this disease is increasing every year. The situation is different for rare malignancies such as vulvar melanomas and for the even rarer vaginal melanomas. The risk factors for vulvovaginal tumors are not fully understood. The basis of treatment in both cases is surgical resection; however, other types of treatments such as immunotherapy are available. This paper focuses on comparing the pathogenesis and risk factors associated with these neoplasms as well as the efficacy of two groups of drugs-anti-PD-L1 and anti-CTLA4 inhibitors-against both cutaneous melanoma and melanoma of the lower genital tract (vulva and vagina). In the case of cutaneous melanoma, the situation looks more optimistic than for vulvovaginal melanoma, which has a much worse prognosis and, as it turns out, shows a poorer response to immune therapy.
Collapse
Affiliation(s)
- Anna Lorenz
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Mateusz Kozłowski
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Sebastian Lenkiewicz
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
5
|
Abstract
Melanoma is a relentless type of skin cancer which involves myriad signaling pathways which regulate many cellular processes. This makes melanoma difficult to treat, especially when identified late. At present, therapeutics include chemotherapy, surgical resection, biochemotherapy, immunotherapy, photodynamic and targeted approaches. These interventions are usually administered as either a single-drug or in combination, based on tumor location, stage, and patients' overall health condition. However, treatment efficacy generally decreases as patients develop treatment resistance. Genetic profiling of melanocytes and the discovery of novel molecular factors involved in the pathogenesis of melanoma have helped to identify new therapeutic targets. In this literature review, we examine several newly approved therapies, and briefly describe several therapies being assessed for melanoma. The goal is to provide a comprehensive overview of recent developments and to consider future directions in the field of melanoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Pavan Kumar Dhanyamraju, Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA17033, USA. Tel: +1-6096474712, E-mail:
| | - Trupti N. Patel
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
6
|
Khaddour K, Maahs L, Avila-Rodriguez AM, Maamar Y, Samaan S, Ansstas G. Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches. Cancers (Basel) 2021; 13:5847. [PMID: 34831002 PMCID: PMC8616477 DOI: 10.3390/cancers13225847] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Melanomas exhibit the highest rate of somatic mutations among all different types of cancers (with the exception of BCC and SCC). The accumulation of a multimode of mutations in the driver oncogenes are responsible for the proliferative, invasive, and aggressive nature of melanomas. High-resolution and high-throughput technology has led to the identification of distinct mutational signatures and their downstream alterations in several key pathways that contribute to melanomagenesis. This has enabled the development of individualized treatments by targeting specific molecular alterations that are vital for cancer cell survival, which has resulted in improved outcomes in several cancers, including melanomas. To date, BRAF and MEK inhibitors remain the only approved targeted therapy with a high level of evidence in BRAFV600E/K mutant melanomas. The lack of approved precision drugs in melanomas, relative to other cancers, despite harboring one of the highest rates of somatic mutations, advocates for further research to unveil effective therapeutics. In this review, we will discuss potential druggable mutations and the ongoing research of novel individualized treatment approaches targeting non-BRAF mutations in melanomas.
Collapse
Affiliation(s)
- Karam Khaddour
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Lucas Maahs
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Ana Maria Avila-Rodriguez
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Yazan Maamar
- Division of Hematology and Oncology, Department of Medicine, University of Tishreen Lattakia, Lattakia 2217, Syria;
| | - Sami Samaan
- Department of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - George Ansstas
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
7
|
Wohlmuth C, Wohlmuth-Wieser I. Vulvar Melanoma: Molecular Characteristics, Diagnosis, Surgical Management, and Medical Treatment. Am J Clin Dermatol 2021; 22:639-651. [PMID: 34125416 PMCID: PMC8421300 DOI: 10.1007/s40257-021-00614-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2021] [Indexed: 12/13/2022]
Abstract
Ten percent of all women have pigmented vulvar lesions. Fortunately, most of these are benign but 1% of all melanomas in women affect the vulva. While the mortality rate of cutaneous melanoma has dropped by 7% annually during the last 5 years, the prognosis of vulvar melanoma remains dismal: the 5-year overall survival rate is 47% compared with 92% for cutaneous melanoma. The current evidence suggests that this likely results from a combination of delayed diagnosis and different tumor biology, treatment strategies, and treatment response. Although many landmark trials on checkpoint inhibitors included mucosal and vulvar melanomas, the results were often not reported separately. Post-hoc analyses indicate overall response rates between 19 and 37% for checkpoint inhibitors. A recently published retrospective study on vulvar melanomas suggests an objective response in 33.3% with a similar safety profile to cutaneous melanoma. Tyrosine kinase inhibitors may be considered in recurrent disease if a c-KIT mutation is present.
Collapse
Affiliation(s)
- Christoph Wohlmuth
- Department of Obstetrics and Gynecology, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria.
| | - Iris Wohlmuth-Wieser
- Department of Dermatology and Allergology, Paracelsus Medical University, Salzburg, Austria
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Protein tyrosine phosphatase receptor type E (PTPRE) regulates the activation of wild-type KIT and KIT mutants differently. Biochem Biophys Rep 2021; 26:100974. [PMID: 33732906 PMCID: PMC7937656 DOI: 10.1016/j.bbrep.2021.100974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 11/20/2022] Open
Abstract
Activation of receptor tyrosine kinases needs tight control by tyrosine phosphatases to keep their normal function. In this study, we investigated the regulation of activation of the type III receptor tyrosine kinase KIT by protein tyrosine phosphatase receptor type E (PTPRE). We found that PTPRE can associate with wild-type KIT and inhibit KIT activation in a dose-dependent manner, although the activation of wild-type KIT is dramatically inhibited even when PTPRE is expressed at low level. The D816V mutation of KIT is the most frequently found oncogenic mutation in mastocytosis, and we found that PTPRE can associate and inhibit the activation of KIT/D816V in a dose dependent manner, but the inhibition is much weaker compared with wild-type KIT. Similar to mastocytosis, KIT mutations are the main oncogenic mutations in gastrointestinal stromal tumors (GISTs) although GISTs carry different types of KIT mutations. We further studied the regulation of the activation of GISTs-type KIT mutants and other mastocytosis-type KIT mutants by PTPRE. Indeed, PTPRE can almost block the activation of GISTs-type KIT mutants, while the activation of mastocytosis-type KIT mutants is more resistant to the inhibition of PTPRE. Taken together, our results suggest that PTPRE can associate with KIT, and inhibit the activation of both wild-type KIT and GISTs-type KIT mutants, while the activation of mastocytosis-type KIT mutants is more resistant to PTPRE. PTPRE associates with wild-type KIT and KIT mutants. PTPRE inhibits the activation of both wild-type KIT and GISTs-type KIT mutants dramatically. The activation of mastocytosis-type KIT mutants are more resistant to the inhibition of PTPRE.
Collapse
|
9
|
Radu A, Bejenaru C, Ţolea I, Maranduca MA, Brănişteanu DC, Bejenaru LE, Petrariu FD, Stoleriu G, Brănişteanu DE. Immunohistochemical study of CD117 in various cutaneous melanocytic lesions. Exp Ther Med 2020; 21:78. [PMID: 33363589 PMCID: PMC7725021 DOI: 10.3892/etm.2020.9510] [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: 09/09/2020] [Accepted: 10/09/2020] [Indexed: 12/27/2022] Open
Abstract
The aim of the present study was to carried out a comparative immunohistochemical evaluation of CD117 (c-Kit), a biomarker that evaluates both tumor progression and prognosis, in different melanocytic lesions, to emphasize the significance of this biomarker in malignant melanoma (MM). The study was performed on 55 cases, represented by a control group, which included 5 cases of simple nevi and 5 cases of dysplastic nevi, as well as a study group consisting of 35 cases of primary MM and 10 metastases (one intestinal, 3 cutaneous - one satellite and two distant as well as 6 in the lymph nodes). The study group included 15 cases of superficial spreading melanoma (SSM), 10 cases of nodular melanoma (NM), 3 lentigo maligna melanoma (LMM), 3 cases of acral lentiginous melanoma (ALM) and 4 cases of amelanotic MM. CD117 was found to be massively involved in the process of tumorigenesis of cutaneous malignancies, being immunohistochemically undetectable in benign neural lesions, but densely expressed in dysplastic lesions and in situ melanoma areas. In invasive cutaneous MMs, CD117 expression tended to decrease with neoplasia progression proceding into the tumorigenic, vertical growth phase, being lower in the profound dermal component of tumors and in nodular MMs. To eliminate the epidermal barriers and gain a proliferative advantage to allow the transition to the vertical growth phase, it seems that MM should lose expression of c-Kit. Cutaneous metastases were found to express CD117 at a level comparable to their primary tumors, suggesting that other mechanisms interfere directly with the metastatic process and not loss of c-Kit expression by itself. CD117 overexpression in cutaneous melanocytic lesions correlates significantly with increased immunostaining intensity, suggesting that the immunohistochemical evaluation of CD117 may be a good method for screening patients, who could benefit from personalized therapy with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Antonia Radu
- Department of Pharmaceutical Botany, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania
| | - Cornelia Bejenaru
- Department of Pharmaceutical Botany, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania
| | - Ion Ţolea
- Department of Dermatology, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania
| | - Minela Aida Maranduca
- Department of Physiology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iasi 700115, Romania
| | | | - Ludovic Everard Bejenaru
- Department of Pharmacognosy and Phytotherapy, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania
| | - Florin Dumitru Petrariu
- Department of Preventive Medicine and Interdisciplinarity, 'Grigore T. Popa' University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Gabriela Stoleriu
- Clinical Department, Faculty of Medicine and Pharmacy, 'Dunarea de Jos' University, Galati 800008, Romania
| | - Daciana Elena Brănişteanu
- Department of Dermatology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iasi 700115, Romania
| |
Collapse
|
10
|
Ammar UM, Abdel-Maksoud MS, Ali EM, Mersal KI, Ho Yoo K, Oh CH. Structural optimization of imidazothiazole derivatives affords a new promising series as B-Raf V600E inhibitors; synthesis, in vitro assay and in silico screening. Bioorg Chem 2020; 100:103967. [DOI: 10.1016/j.bioorg.2020.103967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
|
11
|
Comodo-Navarro AN, Fernandes M, Barcelos D, Carapeto FCL, Guimarães DP, de Sousa Moraes L, Cerutti J, Iwamura ESM, Landman G. Intratumor Heterogeneity of KIT Gene Mutations in Acral Lentiginous Melanoma. Am J Dermatopathol 2020; 42:265-271. [PMID: 31393283 DOI: 10.1097/dad.0000000000001475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Melanoma is an aggressive skin malignancy, and the acral lentiginous melanoma (ALM) subtype affects non-sun-exposed sites such as the volar surface of the hands and feet and the subungual region and is most common in Asians, Hispanics, and Afro-descendants. The presence of different clones within the same tumor seems to influence the aggressiveness of tumors. Patients with mutations in the KIT gene have shown a good response to tyrosine kinase inhibitor therapy. We tested the hypothesis of intratumor heterogeneity through analysis of KIT gene mutations in ALM and determined the correlation between KIT mutations and demographic, clinical, and histopathological variables. Twenty-five ALM samples were examined. We selected up to four different regions per tumor for sequencing by the Sanger method for analysis of KIT gene exon 11 and exon 13 mutations. Advanced lesions were predominant, and the main histopathological characteristics of lesions were Breslow index >4.0 mm (17/25, 68%), Clark level IV/V (21/25, 84%), ulceration (16/25, 64%), and >3 mitoses/mm (8/25, 32%). KIT gene mutations were detected in 11/25 cases (44%), and all these 11 cases displayed intratumor heterogeneity, that is, at least 2 tumor regions had different mutational profiles. The predicted effect of most mutations detected was detrimental to protein function. No significant correlations between histopathological variables and either KIT mutations or intratumor heterogeneity were observed. The hypothesis of intratumor heterogeneity of KIT gene mutations in acral lentiginous melanoma was supported.
Collapse
Affiliation(s)
| | - Mariana Fernandes
- Departamento de Patologia, Universidade Federal de São Paulo, São Paulo, Brazil; and
| | - Denise Barcelos
- Departamento de Patologia, Universidade Federal de São Paulo, São Paulo, Brazil; and
| | | | | | - Lais de Sousa Moraes
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Janete Cerutti
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Gilles Landman
- Departamento de Patologia, Universidade Federal de São Paulo, São Paulo, Brazil; and
| |
Collapse
|
12
|
Yazdani M, Jaafari MR, Verdi J, Alani B, Noureddini M, Badiee A. Ex vivo-generated dendritic cell-based vaccines in melanoma: the role of nanoparticulate delivery systems. Immunotherapy 2020; 12:333-349. [DOI: 10.2217/imt-2019-0173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Melanoma is a poor immunogenic cancer and many treatment strategies have been used to enhance specific or nonspecific immunity against it. Dendritic cell (DC)-based cancer vaccine is the most effective therapies that have been used so far. Meanwhile, the efficacy of DC-based immunotherapy relies on critical factors relating to DCs such as the state of maturation and proper delivery of antigens. In this regard, the use of nanoparticulate delivery systems for effective delivery of antigen to ex vivo-generated DC-based vaccines that also poses adjuvanticity would be an ideal approach. In this review article, we attempt to summarize the role of different types of nanoparticulate antigen delivery systems used in the development of ex vivo-generated DC-based vaccines against melanoma and describe their adjuvanticity in mediation of DC maturation, cytoplasmic presentation of antigens to MHC class I molecules, which led to potent antigen-specific immune responses. As were represented, cationic liposomes were the most used approach, which suggest its potential applicability as delivery systems for further experiments in combination with either adjuvants or monoclonal antibodies.
Collapse
Affiliation(s)
- Mona Yazdani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Mahdi Noureddini
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 91778-99191, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 91778-99191, Iran
| |
Collapse
|
13
|
González-Ruiz L, González-Moles MÁ, González-Ruiz I, Ruiz-Ávila I, Ayén Á, Ramos-García P. An update on the implications of cyclin D1 in melanomas. Pigment Cell Melanoma Res 2020; 33:788-805. [PMID: 32147907 DOI: 10.1111/pcmr.12874] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/03/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Cyclin D1 is a protein encoded by the CCND1 gene, located on 11q13 chromosome, which is a key component of the physiological regulation of the cell cycle. CCND1/cyclin D1 is upregulated in several types of human tumors including melanoma and is currently classified as an oncogene that promotes uncontrolled cell proliferation. Despite the demonstrated importance of CCND1/cyclin D1 as a central oncogene in several types of human tumors, its knowledge in melanoma is still limited. This review examines data published on upregulation of the CCND1 gene and cyclin D1 protein in the melanoma setting, focusing on the pathways and molecular mechanisms involved in the activation of the gene and on the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Lucia González-Ruiz
- Dermatology Service, Ciudad Real General University Hospital, Ciudad Real, Spain
| | | | | | - Isabel Ruiz-Ávila
- Biohealth Research Institute, Granada, Spain.,Pathology Service, San Cecilio Hospital Complex, Granada, Spain
| | - Ángela Ayén
- Dermatology Service, San Cecilio Hospital Complex, Granada, Spain
| | | |
Collapse
|
14
|
Weisberg E, Meng C, Case AE, Sattler M, Tiv HL, Gokhale PC, Buhrlage SJ, Liu X, Yang J, Wang J, Gray N, Stone RM, Adamia S, Dubreuil P, Letard S, Griffin JD. Comparison of effects of midostaurin, crenolanib, quizartinib, gilteritinib, sorafenib and BLU-285 on oncogenic mutants of KIT, CBL and FLT3 in haematological malignancies. Br J Haematol 2019; 187:488-501. [PMID: 31309543 DOI: 10.1111/bjh.16092] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/21/2019] [Indexed: 12/27/2022]
Abstract
Mutations in two type-3 receptor tyrosine kinases (RTKs), KIT and FLT3, are common in both acute myeloid leukaemia (AML) and systemic mastocytosis (SM) and lead to hyperactivation of key signalling pathways. A large number of tyrosine kinase inhibitors (TKIs) have been developed that target either FLT3 or KIT and significant clinical benefit has been demonstrated in multiple clinical trials. Given the structural similarity of FLT3 and KIT, it is not surprising that some of these TKIs inhibit both of these receptors. This is typified by midostaurin, which has been approved by the US Food and Drug Administration for mutant FLT3-positive AML and for KIT D816V-positive SM. Here, we compare the in vitro activities of the clinically available FLT3 and KIT inhibitors with those of midostaurin against a panel of cells expressing a variety of oncogenic FLT3 or KIT receptors, including wild-type (wt) FLT3, FLT3-internal tandem duplication (ITD), FLT3 D835Y, the resistance mutant FLT3-ITD+ F691L, KIT D816V, and KIT N822K. We also examined the effects of these inhibitors in vitro and in vivo on cells expressing mutations in c-CBL found in AML that result in hypersensitization of RTKs, such as FLT3 and KIT. The results show a wide spectrum of activity of these various mutations to these clinically available TKIs.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Chengcheng Meng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Abigail E Case
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hong L Tiv
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Prafulla C Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Xiaoxi Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jing Yang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Nathanael Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Sophia Adamia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patrice Dubreuil
- CRCM, [Signalling, Haematopoiesis and Mechanism of Oncogenesis, Equipe Labellisée Ligue Contre le Cancer], Inserm, U1068; Institut Paoli-Calmettes; Aix-Marseille University, Marseille, France
| | - Sebastien Letard
- CRCM, [Signalling, Haematopoiesis and Mechanism of Oncogenesis, Equipe Labellisée Ligue Contre le Cancer], Inserm, U1068; Institut Paoli-Calmettes; Aix-Marseille University, Marseille, France
| | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Apsel Winger B, Cortopassi WA, Garrido Ruiz D, Ding L, Jang K, Leyte-Vidal A, Zhang N, Esteve-Puig R, Jacobson MP, Shah NP. ATP-Competitive Inhibitors Midostaurin and Avapritinib Have Distinct Resistance Profiles in Exon 17-Mutant KIT. Cancer Res 2019; 79:4283-4292. [PMID: 31270078 DOI: 10.1158/0008-5472.can-18-3139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 05/05/2019] [Accepted: 06/26/2019] [Indexed: 01/08/2023]
Abstract
KIT is a type-3 receptor tyrosine kinase that is frequently mutated at exon 11 or 17 in a variety of cancers. First-generation KIT tyrosine kinase inhibitors (TKI) are ineffective against KIT exon 17 mutations, which favor an active conformation that prevents these TKIs from binding. The ATP-competitive inhibitors, midostaurin and avapritinib, which target the active kinase conformation, were developed to inhibit exon 17-mutant KIT. Because secondary kinase domain mutations are a common mechanism of TKI resistance and guide ensuing TKI design, we sought to define problematic KIT kinase domain mutations for these emerging therapeutics. Midostaurin and avapritinib displayed different vulnerabilities to secondary kinase domain substitutions, with the T670I gatekeeper mutation being selectively problematic for avapritinib. Although gatekeeper mutations often directly disrupt inhibitor binding, we provide evidence that T670I confers avapritinib resistance indirectly by inducing distant conformational changes in the phosphate-binding loop. These findings suggest combining midostaurin and avapritinib may forestall acquired resistance mediated by secondary kinase domain mutations. SIGNIFICANCE: This study identifies potential problematic kinase domain mutations for next-generation KIT inhibitors midostaurin and avapritinib.
Collapse
Affiliation(s)
- Beth Apsel Winger
- Division of Hematology/Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Wilian A Cortopassi
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Diego Garrido Ruiz
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Lucky Ding
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California
| | - Kibeom Jang
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California
| | - Ariel Leyte-Vidal
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California
| | - Na Zhang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California.,Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Rosaura Esteve-Puig
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Neil P Shah
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California.
| |
Collapse
|
16
|
Cowey CL, Liu FX, Boyd M, Aguilar KM, Krepler C. Real-world treatment patterns and clinical outcomes among patients with advanced melanoma: A retrospective, community oncology-based cohort study (A STROBE-compliant article). Medicine (Baltimore) 2019; 98:e16328. [PMID: 31305421 PMCID: PMC6641721 DOI: 10.1097/md.0000000000016328] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Recently, the effectiveness of novel immune checkpoint inhibitors and BRAF-directed therapies has been demonstrated in advanced melanoma trial populations. Limited research, however, has evaluated the impact of these therapies in a real-world setting. The aim of this study was to evaluate treatment patterns and clinical outcomes among advanced melanoma patients treated with modern therapies within community oncology clinics. Adult patients with advanced melanoma who initiated treatment within the US Oncology Network between 1/1/14 and 12/31/16 were included. Data were sourced from electronic healthcare records. Patients were followed through 12/31/17. Descriptive analyses were performed to assess patient and treatment characteristics and Kaplan-Meier methods were used for time-to-event outcomes. In total, 484 patients met eligibility criteria (32.0% with brain metastasis, 12.6% with Eastern Cooperative Oncology Group performance status ≥2). In the first-line (1L) setting during the study period, 37.0% received anti-PD1 monotherapies, 26.4% ipilimumab monotherapy, 19.8% BRAF/MEK combination therapy, 6.4% BRAF or MEK monotherapy, 4.1% ipilimumab/nivolumab combination therapy and 6.2% other regimens. Differences in baseline demographic and clinical characteristics were observed across treatment groups. For the overall study population, the median (95% confidence interval) estimates for overall survival, time to next treatment and progression-free survival were 20.7 (16.0, 26.8), 5.8 (5.3, 6.5), and 4.9 (4.2, 5.7) months, respectively. The results of this study provide real-world insight into advanced melanoma treatment trends and clinical outcomes, including high utilization of immunotherapies and BRAF/MEK combination therapy. Future research can explore underlying differences in patient subpopulations and the sequence of therapies across lines of therapy.
Collapse
|
17
|
Mo X, Preston S, Zaidi MR. Macroenvironment-gene-microenvironment interactions in ultraviolet radiation-induced melanomagenesis. Adv Cancer Res 2019; 144:1-54. [PMID: 31349897 DOI: 10.1016/bs.acr.2019.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cutaneous malignant melanoma is one of the few major cancers that continue to exhibit a positive rate of increase in the developed world. A wealth of epidemiological data has undisputedly implicated ultraviolet radiation (UVR) from sunlight and artificial sources as the major risk factor for melanomagenesis. However, the molecular mechanisms of this cause-and-effect relationship remain murky and understudied. Recent efforts on multiple fronts have brought unprecedented expansion of our knowledge base on this subject and it is now clear that melanoma is caused by a complex interaction between genetic predisposition and environmental exposure, primarily to UVR. Here we provide an overview of the effects of the macroenvironment (UVR) on the skin microenvironment and melanocyte-specific intrinsic (mostly genetic) landscape, which conspire to produce one of the deadliest malignancies.
Collapse
Affiliation(s)
- Xuan Mo
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Sarah Preston
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - M Raza Zaidi
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
18
|
Clinical Pharmacokinetic and Pharmacodynamic Considerations in the (Modern) Treatment of Melanoma. Clin Pharmacokinet 2019; 58:1029-1043. [DOI: 10.1007/s40262-019-00753-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
19
|
Etchevers HC, Dupin E, Le Douarin NM. The diverse neural crest: from embryology to human pathology. Development 2019; 146:146/5/dev169821. [PMID: 30858200 DOI: 10.1242/dev.169821] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 02/07/2019] [Indexed: 01/13/2023]
Abstract
We review here some of the historical highlights in exploratory studies of the vertebrate embryonic structure known as the neural crest. The study of the molecular properties of the cells that it produces, their migratory capacities and plasticity, and the still-growing list of tissues that depend on their presence for form and function, continue to enrich our understanding of congenital malformations, paediatric cancers and evolutionary biology. Developmental biology has been key to our understanding of the neural crest, starting with the early days of experimental embryology and through to today, when increasingly powerful technologies contribute to further insight into this fascinating vertebrate cell population.
Collapse
Affiliation(s)
- Heather C Etchevers
- Aix-Marseille Université, INSERM, MMG, U1251, 27 boulevard Jean Moulin 13005 Marseille, France
| | - Elisabeth Dupin
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Nicole M Le Douarin
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| |
Collapse
|
20
|
Ahmad F, Avabhrath N, Natarajan S, Parikh J, Patole K, Das BR. Molecular evaluation of BRAF V600 mutation and its association with clinicopathological characteristics: First findings from Indian malignant melanoma patients. Cancer Genet 2019; 231-232:46-53. [PMID: 30803557 DOI: 10.1016/j.cancergen.2019.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/04/2018] [Accepted: 01/07/2019] [Indexed: 12/24/2022]
Abstract
Mutations in the BRAF gene have been described to occur in two-third of melanomas. The objective of the study was to establish the frequency of BRAF V600E/K/R mutation in a series of melanomas from Indian origin and to correlate mutation status with clinicopathological features. Seventy melanoma cases were evaluated for BRAF V600 mutation by pyrosequencing. Overall, BRAF mutations were detected in 30% of the patients. All mutations observed were missense type (GTG > GAG) resulting in p.V600E, while none showed V600K/R mutation. The frequency of BRAF V600E mutations were more in patients with onset age of 50 years. BRAF mutations were significantly associated with tumor site wherein more mutations were seen in tumors from head and neck and extremities region. Acral and mucosal tumor subtype showed a mutation frequency of 31% and 20%, respectively. Epithelial cell morphology tends to harbor frequent BRAF V600E mutation (36%) than other morphological subtypes. Tumors with ulceration and necrosis showed increased BRAF mutation rate (32.5% and 33%) respectively. In conclusion, this is the first study to report a mutation frequency of 30% in this cohort. Our results demonstrated that the BRAF V600E mutation is a frequent event in Indian melanomas, and represents an important molecular target for novel therapeutic approaches.
Collapse
Affiliation(s)
- Firoz Ahmad
- Research and Development, Division, SRL Ltd, Plot no.1, Prime Square building, S.V.Road, Goregaon (W), Mumbai, India
| | - Nagashree Avabhrath
- Research and Development, Division, SRL Ltd, Plot no.1, Prime Square building, S.V.Road, Goregaon (W), Mumbai, India
| | - Sripriya Natarajan
- Research and Development, Division, SRL Ltd, Plot no.1, Prime Square building, S.V.Road, Goregaon (W), Mumbai, India
| | - Jeenal Parikh
- Histopathology Division, Division, SRL Ltd, Plot no.1, Prime Square building, S.V.Road, Goregaon (W), Mumbai, India
| | - Kamlakar Patole
- Histopathology Division, Division, SRL Ltd, Plot no.1, Prime Square building, S.V.Road, Goregaon (W), Mumbai, India
| | - Bibhu Ranjan Das
- Research and Development, Division, SRL Ltd, Plot no.1, Prime Square building, S.V.Road, Goregaon (W), Mumbai, India.
| |
Collapse
|
21
|
Pharmacokinetic and cytokine profiles of melanoma patients with dabrafenib and trametinib-induced pyrexia. Cancer Chemother Pharmacol 2019; 83:693-704. [DOI: 10.1007/s00280-019-03780-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/13/2019] [Indexed: 02/07/2023]
|
22
|
Lambert A, Salleron J, Lion M, Rouyer M, Lozano N, Leroux A, Merlin JL, Harlé A. Comparison of Three Real-Time PCR Assays for the Detection of PIK3CA Somatic Mutations in Formalin-Fixed Paraffin Embedded Tissues of Patients with Breast Carcinomas. Pathol Oncol Res 2018; 25:1117-1123. [PMID: 30426328 DOI: 10.1007/s12253-018-0538-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/09/2018] [Indexed: 01/27/2023]
Abstract
Breast cancer is the leading cause of cancer-related death in women worldwide. Mutations of the PIK3CA gene are found in approximately 25% of breast carcinomas and are reported as activators of the PI3K/AKT/mTOR pathway. This study aims to compare three assays for the somatic mutation detection of PIK3CA gene in FFPE tissues of patients with breast cancer. We compared Cobas® PIK3CA Mutation Test (Roche Diagnostics, Meylan, France), PCR amplification-refractory mutation system Scorpions® (ARMS) and High-Resolution Melting PCR assay (HRM) for the detection of PIK3CA mutations. Discrepant samples were assessed using Next Generation Sequencing (NGS). 46 FFPE breast carcinomas samples of patients treated for breast cancer have been assessed for PIK3CA mutations using the three PCR assays. Among the 46 samples, 17 (37.8%), 13 (28.36%) and 19 (41.3%) had a PIK3CA mutation, with Cobas®, ARMS and HRM assays respectively. Three different mutations of PIK3CA have been detected for one sample. Calculated kappa were 0.95[0.86;1] between Cobas® and HRM, 0.75[0.55;0.95] between Cobas® and ARMS and 0.72[0.51;0.92] between HRM and ARMS. Five samples were found with discrepant results. Our study shows that the Cobas® assay is suitable for PIK3CA mutation assessment in patients with breast cancer. HRM assay is also suitable for PIK3CA mutation assessment but requires a mutation characterization with a specific assay.
Collapse
Affiliation(s)
- A Lambert
- Département d'Oncologie Médicale, Institut de Cancérologie de Lorraine, 54519, Vandoeuvre les Nancy, France
| | - J Salleron
- Département de Biostatistiques, Institut de Cancérologie de Lorraine, 54519, Vandoeuvre les Nancy, France
| | - M Lion
- CNRS UMR 7039 CRAN, Institut de Cancérologie de Lorraine, Service de Biopathologie, Université de Lorraine, 6 avenue de Bourgogne CS 30519, 54519, 54500, Vandoeuvre-lès-Nancy Cedex, France
| | - M Rouyer
- CNRS UMR 7039 CRAN, Institut de Cancérologie de Lorraine, Service de Biopathologie, Université de Lorraine, 6 avenue de Bourgogne CS 30519, 54519, 54500, Vandoeuvre-lès-Nancy Cedex, France
| | - N Lozano
- CNRS UMR 7039 CRAN, Institut de Cancérologie de Lorraine, Service de Biopathologie, Université de Lorraine, 6 avenue de Bourgogne CS 30519, 54519, 54500, Vandoeuvre-lès-Nancy Cedex, France
| | - A Leroux
- CNRS UMR 7039 CRAN, Institut de Cancérologie de Lorraine, Service de Biopathologie, Université de Lorraine, 6 avenue de Bourgogne CS 30519, 54519, 54500, Vandoeuvre-lès-Nancy Cedex, France
| | - J L Merlin
- CNRS UMR 7039 CRAN, Institut de Cancérologie de Lorraine, Service de Biopathologie, Université de Lorraine, 6 avenue de Bourgogne CS 30519, 54519, 54500, Vandoeuvre-lès-Nancy Cedex, France
| | - Alexandre Harlé
- CNRS UMR 7039 CRAN, Institut de Cancérologie de Lorraine, Service de Biopathologie, Université de Lorraine, 6 avenue de Bourgogne CS 30519, 54519, 54500, Vandoeuvre-lès-Nancy Cedex, France.
| |
Collapse
|
23
|
Capo A, Goteri G, Mozzicafreddo G, Serresi S, Giacchetti A. Melanoma and mastocytosis: is really only a coincidence? Clin Exp Dermatol 2018; 44:76-77. [PMID: 30178486 DOI: 10.1111/ced.13717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2018] [Indexed: 11/30/2022]
Affiliation(s)
- A Capo
- Dermatology Unit, Istituto Nazionale di Riposo e Cura per Anziani, INRCA-IRCCS Hospital, via della Montagnola 81, Ancona, 60127, Italy
| | - G Goteri
- Unit of Pathologic Anatomy and Histopathology, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche Region, United Ancona Hospitals, Torrette, Ancona, Italy
| | - G Mozzicafreddo
- Dermatology Unit, Istituto Nazionale di Riposo e Cura per Anziani, INRCA-IRCCS Hospital, via della Montagnola 81, Ancona, 60127, Italy
| | - S Serresi
- Dermatology Unit, Istituto Nazionale di Riposo e Cura per Anziani, INRCA-IRCCS Hospital, via della Montagnola 81, Ancona, 60127, Italy
| | - A Giacchetti
- Dermatology Unit, Istituto Nazionale di Riposo e Cura per Anziani, INRCA-IRCCS Hospital, via della Montagnola 81, Ancona, 60127, Italy
| |
Collapse
|
24
|
Zhang Y, Zhan X, Peng S, Cai Y, Zhang YS, Liu Y, Wang Z, Yu Y, Wang Y, Shi Q, Zeng X, Yuan K, Zhou N, Joshi R, Zhang M, Zhang Z, Min W. Targeted-gene silencing of BRAF to interrupt BRAF/MEK/ERK pathway synergized photothermal therapeutics for melanoma using a novel FA-GNR-siBRAF nanosystem. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:1679-1693. [PMID: 29684526 DOI: 10.1016/j.nano.2018.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/20/2018] [Accepted: 04/09/2018] [Indexed: 01/16/2023]
Abstract
Melanoma is significantly associated with mutant BRAF gene, a suitable target for siRNA-based anti-melanoma therapy. However, a tumor-specific delivery system is a major hurdle for clinical applications. Here, we developed a novel nano-carrier, FA-GNR-siBRAF for safe topical application, which consists of folic acid (FA) as the tumor-targeting moiety, golden nanorods (GNR) providing photothermal capability to kill tumor cells under laser irradiation, and siRNA specifically silencing BRAF (siBRAF). The in vitro and in vivo results revealed that FA-GNR-siBRAF displayed high transfection rates, and subsequently induced remarkable gene knockdown of BRAF, resulting in suppression of melanoma growth due to the interruption of the MEK/ERK pathway. Combinatorial photothermal effects and BRAF knockdown by FA-GNR-siBRAF effectively killed tumor cells through apoptosis, with enhanced efficiency than individual treatments. Therefore, the FA-GNR-siBRAF simultaneously induced BRAF gene silencing and photothermal effects which achieved synergistic efficacy in the treatment of melanoma, paving a new path for developing clinical treatment methods for melanoma.
Collapse
Affiliation(s)
- Yujuan Zhang
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China; Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA.
| | - Xuelin Zhan
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Shanshan Peng
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China; Department of Surgery, Pathology and Oncology, University of Western Ontario, London, Canada
| | - Ying Cai
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Yanling Liu
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Zhigang Wang
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yanrong Yu
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yifan Wang
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Qiaofa Shi
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
| | - Xiaoping Zeng
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
| | - Keng Yuan
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Nanjin Zhou
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Rakesh Joshi
- Department of Surgery, Pathology and Oncology, University of Western Ontario, London, Canada
| | - Meng Zhang
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China; Department of Surgery, Pathology and Oncology, University of Western Ontario, London, Canada
| | - Zhuxu Zhang
- Department of Surgery, Pathology and Oncology, University of Western Ontario, London, Canada
| | - Weiping Min
- Institute of immunotherapy and College of Basic Medicine of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China; Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China; Department of Surgery, Pathology and Oncology, University of Western Ontario, London, Canada.
| |
Collapse
|
25
|
Abstract
Melanoma represents the most aggressive and the deadliest form of skin cancer. Current therapeutic approaches include surgical resection, chemotherapy, photodynamic therapy, immunotherapy, biochemotherapy, and targeted therapy. The therapeutic strategy can include single agents or combined therapies, depending on the patient’s health, stage, and location of the tumor. The efficiency of these treatments can be decreased due to the development of diverse resistance mechanisms. New therapeutic targets have emerged from studies of the genetic profile of melanocytes and from the identification of molecular factors involved in the pathogenesis of the malignant transformation. In this review, we aim to survey therapies approved and under evaluation for melanoma treatment and relevant research on the molecular mechanisms underlying melanomagenesis.
Collapse
Affiliation(s)
- Beatriz Domingues
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Manuel Lopes
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Hospital S João, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Helena Pópulo
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Hirota S. Differential diagnosis of gastrointestinal stromal tumor by histopathology and immunohistochemistry. Transl Gastroenterol Hepatol 2018; 3:27. [PMID: 29971258 DOI: 10.21037/tgh.2018.04.01] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 03/26/2018] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors in the gastrointestinal (GI) tract. GISTs account for approximately 80% of the clinically relevant GI mesenchymal tumors. Although most GISTs show spindle cell morphology, 10-15% of GISTs show pure epithelioid configuration. Therefore, not only spindle cell tumors but also epithelioid cell ones developing in the GI tract are subject to the differential diagnoses of GISTs. GISTs are basically positive for KIT, a receptor tyrosine kinase (RTK) encoded by protooncogene c-kit, by immunohistochemistry, but approximately 5% of GISTs are only weakly or barely positive for KIT. Since almost all spindle cell type GISTs are strongly and diffusely positive for KIT regardless of different genetic subtypes, diagnosis of the spindle cell type GISTs is not difficult. On the other hand, epithelioid cell type GISTs show different staining patterns of KIT in different genetic backgrounds. Approximately half of the epithelioid cell type GISTs with platelet-derived growth factor receptor alpha (PDGFRA) gene mutation might show weak or undetectable staining of KIT. On the other hand, almost all GISTs are negative for desmin, which is a positive marker for mature smooth muscle cells, and S100 protein, which is a Schwann cell marker. Smooth muscle tumors such as leiomyomas and leiomyosarcomas, which usually show the spindle cell morphology, consist of approximately 10% of the clinically relevant GI mesenchymal tumors and are almost positive for desmin and negative for KIT and S100 protein. Schwannomas which nearly always show the spindle cell pattern, comprise up to 5% of the GI mesenchymal tumors, and almost all of them are positive for S100 protein and negative for KIT and desmin. Thus, most GI mesenchymal tumors are differentially diagnosed by immunohistochemistry (IHC) of KIT, desmin and S100 protein. However, mesenchymal tumors such as desmoids, solitary fibrous tumors (SFTs), inflammatory myofibroblastic tumors (IMTs), perivascular epithelioid cell tumors (PEComas), inflammatory fibroid polyps (IFPs), rarely develop in the GI tract, and have to be correctly diagnosed through detection of specific immunohistochemical markers and/or unique genetic aberrations.
Collapse
Affiliation(s)
- Seiichi Hirota
- Department of Surgical Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
27
|
Toscano de Mendonça UB, Cernea CR, Matos LL, Monteiro de Araujo Lima RR. Analysis of KIT gene mutations in patients with melanoma of the head and neck mucosa: a retrospective clinical report. Oncotarget 2018; 9:22886-22894. [PMID: 29796159 PMCID: PMC5955421 DOI: 10.18632/oncotarget.25094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/22/2018] [Indexed: 11/25/2022] Open
Abstract
Unlike their cutaneous counterparts, head and neck mucosal malignant melanomas (HNMM) are more aggressive, and their prognostic markers have not been fully elucidated. This study, comprising 28 patients with HNMM, aimed to establish the relationship between different mutations and outcome, define the incidence of KIT mutations in HNMM, and identify the correlation among therapeutic options, histopathological findings, demographic data, and clinical response. Clinical analysis included patient characteristics, staging, primary and palliative treatments, and disease-free survival and overall survival (OS). Progression-free survival and OS were analyzed. Paraffin blocks were selected following histologic analyses, enabling DNA extraction. PCR amplification of exons 9, 11, 13, and 17, with different DNA concentrations, was performed. Patients were predominantly females (57%) and aged 27–85 years. All patients underwent surgery; 17 received adjuvant radiotherapy, and recurrences occurred in 82% patients. Oncologic mutations in KIT were found in 7 of 7 tumors, 3 in exon 9, 3 in exon 11, and 1 in exon 13. Predictive factors for recurrence were mitotic rate, vascular invasion, and perineural spread. There were no significant differences in DFS and OS according to KIT mutation. Our study results suggest that some patients might benefit from appropriate targeted therapy with kinase inhibitors.
Collapse
Affiliation(s)
| | - Claudio Roberto Cernea
- Department of Head and Neck Surgery, School of Medicine, University of São Paulo, Sao Paulo-SP, Brazil
| | - Leandro Luongo Matos
- Department of Head and Neck Surgery, School of Medicine, University of São Paulo, Sao Paulo-SP, Brazil
| | | |
Collapse
|
28
|
Tiedje V, Ting S, Herold T, Synoracki S, Latteyer S, Moeller LC, Zwanziger D, Stuschke M, Fuehrer D, Schmid KW. NGS based identification of mutational hotspots for targeted therapy in anaplastic thyroid carcinoma. Oncotarget 2018; 8:42613-42620. [PMID: 28489587 PMCID: PMC5522092 DOI: 10.18632/oncotarget.17300] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/11/2017] [Indexed: 12/13/2022] Open
Abstract
CONTEXT Anaplastic thyroid carcinoma (ATC) represents one of the most aggressive carcinomas with no consistent survival benefit when treated with conventional radiochemotherapy. Approaches targeting "oncogene addiction" of ATC are increasingly explored and first promising results have been reported in single case studies. OBJECTIVE To determine the prevalence of mutations in known thyroid oncogenes and signalling pathways amendable to targeted therapy in a large cohort of ATC. RESULTS In 118 ATC (57 male/ 61 female) a total of 165 mutations were found. Genes involved in the MAPK/ERK and PI3K pathway (BRAF 11.0%, HRAS 4.2%, KRAS 7.6%, NRAS 7.6%, PI3KCA 11.8%) were altered in 33%. Targetable receptor tyrosine kinases were mutated in 11%. The most frequently altered genes were TERT in 86/118 (73%) and p53 in 65/118 (55%) cases. No mutations were found analysing ALK, KIT, MET and mTOR. MATERIALS AND METHODS Next generation sequencing (NGS) was performed in FFPE samples from 118 ATC using MiSeq (Illumina) and CLC Cancer Research Workbench (CLCbio; Qiagen) for mutation analysis in: ALK, BRAF, CDKN2A, EGFR, ERBB2, HRAS, KIT, KRAS, MET, mTOR, NRAS, PDGFRA, PI3KCA, p53, RB1, RET and TSC2. Sanger sequencing was used to detect TERT promotor mutations. CONCLUSIONS To our knowledge this is the largest study analysing mutations for targeted therapy of ATC. We found that 33% of ATC harbour mutations in pathways amendable to targeted therapy. Molecular screening in ATC is suggested for targeted therapies since current conventional treatment for ATC proved mainly futile.
Collapse
Affiliation(s)
- Vera Tiedje
- Department of Endocrinology and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Saskia Ting
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Thomas Herold
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Synoracki
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Soeren Latteyer
- Department of Endocrinology and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Lars C Moeller
- Department of Endocrinology and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Denise Zwanziger
- Department of Endocrinology and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Martin Stuschke
- Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Dagmar Fuehrer
- Department of Endocrinology and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| | - Kurt Werner Schmid
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
| |
Collapse
|
29
|
Abstract
Sinonasal mucosal melanoma (SNMM) is a rare oncological entity that comprises most head and neck mucosal melanomas. SNMM has distinctive genetic background, different from cutaneous melanoma. Survival outcomes among SNMM patients are poor; while there is no clear consensus on the optimal management of SNMM, the primary treatment modality is generally considered to be wide surgical excision, and radiation therapy (RT) is often used in the postoperative adjuvant setting to improve locoregional control. Systemic therapies have demonstrated little or no survival benefit, and most SNMM patients die of distant metastatic disease. Owing to the rarity of the disease, the literature describing treatment approaches for SNMM is lacking and largely limited to isolated case reports and retrospective series. Here, we describe contemporary diagnostic and therapeutic approaches to SNMM based on the most recent molecular and outcome data.
Collapse
|
30
|
Soheili F, Jalili Z, Rahbar M, Khatooni Z, Mashayekhi A, Jafari H. Novel mutation of GATA4 gene in Kurdish population of Iran with nonsyndromic congenital heart septals defects. CONGENIT HEART DIS 2018; 13:295-304. [PMID: 29377543 DOI: 10.1111/chd.12571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/07/2017] [Accepted: 12/10/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND The mutations in GATA4 gene induce inherited atrial and ventricular septation defects, which is the most frequent forms of congenital heart defects (CHDs) constituting about half of all cases. METHOD We have performed High resolution melting (HRM) mutation scanning of GATA4 coding exons of nonsyndrome 100 patients as a case group including 39 atrial septal defects (ASD), 57 ventricular septal defects (VSD) and four patients with both above defects and 50 healthy individuals as a control group. Our samples are categorized according to their HRM graph. The genome sequencing has been done for 15 control samples and 25 samples of patients whose HRM analysis were similar to healthy subjects for each exon. The PolyPhen-2 and MUpro have been used to determine the causative possibility and structural stability prediction of GATA4 sequence variation. RESULTS The HRM curve analysis exhibit that 21 patients and 3 normal samples have deviated curves for GATA4 coding exons. Sequencing analysis has revealed 12 nonsynonymous mutations while all of them resulted in stability structure of protein 10 of them are pathogenic and 2 of them are benign. Also we found two nucleotide deletions which one of them was novel and one new indel mutation resulting in frame shift mutation, and 4 synonymous variations or polymorphism in 6 of patients and 3 of normal individuals. Six or about 50% of these nonsynonymous mutations have not been previously reported. CONCLUSION Our results show that there is a spectrum of GATA4 mutations resulting in septal defects.
Collapse
Affiliation(s)
- Fariborz Soheili
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, IR, Iran.,Department of Marine Biology, Faculty of Marine Sciences, Chabahar Maritime University, Chabahar, IR, Iran
| | - Zahra Jalili
- Department of Cardiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, IR, Iran
| | - Mahtab Rahbar
- Department of Pathology, Faculty of Medicine, Iran Medical University of Medical Science, Tehran, IR, Iran
| | - Zahed Khatooni
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, IR, Iran
| | - Amir Mashayekhi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University Tehran, IR, Iran
| | - Hossein Jafari
- Department of Statistic and Basic Science, Chabahar Maritime University, Chabahar, IR, Iran
| |
Collapse
|
31
|
Vacca D, Cancila V, Gulino A, Lo Bosco G, Belmonte B, Di Napoli A, Florena AM, Tripodo C, Arancio W. Real-time detection of BRAF V600E mutation from archival hairy cell leukemia FFPE tissue by nanopore sequencing. Mol Biol Rep 2017; 45:1-7. [DOI: 10.1007/s11033-017-4133-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 12/08/2017] [Indexed: 12/01/2022]
|
32
|
Cardoso L, Stevenson M, Thakker RV. Molecular genetics of syndromic and non-syndromic forms of parathyroid carcinoma. Hum Mutat 2017; 38:1621-1648. [PMID: 28881068 PMCID: PMC5698716 DOI: 10.1002/humu.23337] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 08/21/2017] [Accepted: 09/04/2017] [Indexed: 12/23/2022]
Abstract
Parathyroid carcinoma (PC) may occur as part of a complex hereditary syndrome or an isolated (i.e., non-syndromic) non-hereditary (i.e., sporadic) endocrinopathy. Studies of hereditary and syndromic forms of PC, which include the hyperparathyroidism-jaw tumor syndrome (HPT-JT), multiple endocrine neoplasia types 1 and 2 (MEN1 and MEN2), and familial isolated primary hyperparathyroidism (FIHP), have revealed some genetic mechanisms underlying PC. Thus, cell division cycle 73 (CDC73) germline mutations cause HPT-JT, and CDC73 mutations occur in 70% of sporadic PC, but in only ∼2% of parathyroid adenomas. Moreover, CDC73 germline mutations occur in 20%-40% of patients with sporadic PC and may reveal unrecognized HPT-JT. This indicates that CDC73 mutations are major driver mutations in the etiology of PCs. However, there is no genotype-phenotype correlation and some CDC73 mutations (e.g., c.679_680insAG) have been reported in patients with sporadic PC, HPT-JT, or FIHP. Other genes involved in sporadic PC include germline MEN1 and rearranged during transfection (RET) mutations and somatic alterations of the retinoblastoma 1 (RB1) and tumor protein P53 (TP53) genes, as well as epigenetic modifications including DNA methylation and histone modifications, and microRNA misregulation. This review summarizes the genetics and epigenetics of the familial syndromic and non-syndromic (sporadic) forms of PC.
Collapse
Affiliation(s)
- Luís Cardoso
- Department of EndocrinologyDiabetes and MetabolismCentro Hospitalar e Universitário de CoimbraPraceta Prof Mota PintoCoimbraPortugal
- Radcliffe Department of MedicineAcademic Endocrine UnitOxford Centre for DiabetesEndocrinology and MetabolismUniversity of OxfordOxfordUnited Kingdom
| | - Mark Stevenson
- Radcliffe Department of MedicineAcademic Endocrine UnitOxford Centre for DiabetesEndocrinology and MetabolismUniversity of OxfordOxfordUnited Kingdom
| | - Rajesh V. Thakker
- Radcliffe Department of MedicineAcademic Endocrine UnitOxford Centre for DiabetesEndocrinology and MetabolismUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
33
|
Helbig D, Quaas A, Mauch C, Merkelbach-Bruse S, Büttner R, Emberger M, Wobser M, Rüsseler V, Pütz K, Binot E, Rehker J, Budczies J, Ihle MA. Copy number variations in atypical fibroxanthomas and pleomorphic dermal sarcomas. Oncotarget 2017; 8:109457-109467. [PMID: 29312620 PMCID: PMC5752533 DOI: 10.18632/oncotarget.22691] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/13/2017] [Indexed: 11/25/2022] Open
Abstract
Atypical fibroxanthomas (AFX) and pleomorphic dermal sarcomas (PDS) are frequent cutaneous sarcomas typically arising on sun-exposed skin in elderly patients. In contrast to AFX, which generally do not recur after complete excision, PDS locally recur in up to 50% and metastasize in up to 20%. We recently detected characteristic UV-induced TP53 mutations as potential driver mutation in almost all PDS investigated as well as activating PIK3CA and RAS gene mutations in around one third of our tumors representing targets for personalized treatments in patients with unresectable or metastasized PDS. In the present study, we identified amplifications and deletions in a small part of the PDS (6 of 27 cases) but not in AFX suggesting that copy number variations (CNV) might not be an initial event in tumor development but rather important during tumor progression. In addition to BRAF, KNSTRN, IDH1 and PDGFRA amplification, CNV analyses revealed deletions in the CDKN2A, KIT and PDGFRA genes. In cases where an appropriate FISH assay was established, the CNV results could be verified by FISH analysis. Amplification of BRAF, KIT or PDGFRA and/or losses of CDKN2A might represent bad prognostic markers, although larger studies are needed to clarify their association with prognosis or progression in PDS.
Collapse
Affiliation(s)
- Doris Helbig
- Department of Dermatology, University Hospital Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Cornelia Mauch
- Department of Dermatology, University Hospital Cologne, Cologne, Germany
| | | | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Marion Wobser
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Vanessa Rüsseler
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Katharina Pütz
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Elke Binot
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jan Rehker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jan Budczies
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | | |
Collapse
|
34
|
Adler NR, Wolfe R, Kelly JW, Haydon A, McArthur GA, McLean CA, Mar VJ. Tumour mutation status and sites of metastasis in patients with cutaneous melanoma. Br J Cancer 2017; 117:1026-1035. [PMID: 28787433 PMCID: PMC5625668 DOI: 10.1038/bjc.2017.254] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/13/2017] [Accepted: 07/07/2017] [Indexed: 11/23/2022] Open
Abstract
Background: Cutaneous melanoma can metastasise haematogenously and/or lymphogenously to form satellite/in-transit, lymph node or distant metastasis. This study aimed to determine if BRAF and NRAS mutant and wild-type tumours differ in their site of first tumour metastasis and anatomical metastatic pathway. Methods: Prospective cohort of patients with a histologically confirmed primary cutaneous melanoma at three tertiary referral centres in Melbourne, Australia from 2010 to 2015. Multinomial regression determined clinical, histological and mutational factors associated with the site of first metastasis and metastatic pathway. Results: Of 1048 patients, 306 (29%) developed metastasis over a median 4.7 year follow-up period. 73 (24%), 192 (63%) and 41 (13%) developed distant, regional lymph node and satellite/in-transit metastasis as the first site of metastasis, respectively. BRAF mutation was associated with lymph node metastasis (adjusted RRR 2.46 95% CI 1.07–5.69, P=0.04) and sentinel lymph node positivity (adjusted odds ratio [aOR] OR 1.55, 95% CI 1.14–2.10, P=0.005). BRAF mutation and NRAS mutation were associated with increased odds of developing liver metastasis (aOR 3.09, 95% CI 1.49–6.42, P=0.003; aOR 3.17, 95% CI 1.32–7.58, P=0.01) and central nervous system (CNS) metastasis (aOR 4.65, 95% CI 2.23–9.69, P<0.001; aOR 4.03, 95% CI 1.72–9.44, P=0.001). NRAS mutation was associated with lung metastasis (aOR 2.44, 95% CI 1.21–4.93, P=0.01). Conclusions: BRAF mutation was found to be associated with lymph node metastasis as first metastasis and sentinel lymph node positivity. BRAF and NRAS mutations were associated with CNS and liver metastasis and NRAS mutation with lung metastasis. If these findings are validated in additional prospective studies, a role for heightened visceral organ surveillance may be warranted in patients with tumours harbouring these somatic mutations.
Collapse
Affiliation(s)
- Nikki R Adler
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria 3004, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - John W Kelly
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria 3004, Australia
| | - Andrew Haydon
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria 3004, Australia.,Department of Medical Oncology, Alfred Hospital, Melbourne, Victoria 3004, Australia
| | - Grant A McArthur
- Divisions of Research and Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia
| | - Catriona A McLean
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria 3004, Australia.,Department of Anatomical Pathology, Alfred Hospital, Melbourne, Victoria 3004, Australia
| | - Victoria J Mar
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Victoria 3004, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.,Skin and Cancer Foundation, Carlton, Victoria 3053, Australia
| |
Collapse
|
35
|
Comparison of Five Different Assays for the Detection of BRAF Mutations in Formalin-Fixed Paraffin Embedded Tissues of Patients with Metastatic Melanoma. Mol Diagn Ther 2017; 21:209-216. [DOI: 10.1007/s40291-017-0258-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
36
|
Noeparast A, Teugels E, Giron P, Verschelden G, De Brakeleer S, Decoster L, De Grève J. Non-V600 BRAF mutations recurrently found in lung cancer predict sensitivity to the combination of Trametinib and Dabrafenib. Oncotarget 2016; 8:60094-60108. [PMID: 28947956 PMCID: PMC5601124 DOI: 10.18632/oncotarget.11635] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 06/09/2016] [Indexed: 12/12/2022] Open
Abstract
Approximately half of BRAF-mutated Non-small cell lung cancers (NSCLCs) harbor a non-V600 BRAF mutation, accounting for ∼40,000 annual deaths worldwide. Recent studies have revealed the benefits of combined targeted therapy with a RAF-inhibitor (Dabrafenib) and a MEK-inhibitor (Trametinib) in treating V600 BRAF mutant cancers, including NSCLC. In contrast, sensitivity of non-V600 BRAF mutations to these inhibitors is not documented. Non-V600 mutations can either increase or impair BRAF kinase activity. However, impaired BRAF kinases can still activate the ERK pathway in a CRAF-dependent manner. Herein, beyond describing a cohort of BRAF mutant NSCLC patients and functionally analyzing 13 tumor-derived BRAF mutations, we demonstrate that both types of non-V600 BRAF mutations can be sensitive to clinically relevant doses of Dabrafenib and Trametinib in HEK293T cells, in lung epithelial cellular model (BEAS-2B) and in human cancer cell lines harboring non-V600 BRAF mutations. ERK activity induced by both types of these mutations is further reduced by combinatorial drug treatment. Moreover, the combination leads to more prolonged ERK inhibition and has anti-proliferative and pro-apoptotic effects in cells harboring both types of non-V600 BRAF mutations. This study provides a basis for the clinical exploration of non-V600 BRAF mutant lung cancers upon treatment with Trametinib and Dabrafenib.
Collapse
Affiliation(s)
- Amir Noeparast
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Erik Teugels
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Philippe Giron
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gil Verschelden
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sylvia De Brakeleer
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lore Decoster
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jacques De Grève
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
37
|
Abbaspour Babaei M, Kamalidehghan B, Saleem M, Huri HZ, Ahmadipour F. Receptor tyrosine kinase (c-Kit) inhibitors: a potential therapeutic target in cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2443-59. [PMID: 27536065 PMCID: PMC4975146 DOI: 10.2147/dddt.s89114] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
c-Kit, a receptor tyrosine kinase, is involved in intracellular signaling, and the mutated form of c-Kit plays a crucial role in occurrence of some cancers. The function of c-Kit has led to the concept that inhibiting c-Kit kinase activity can be a target for cancer therapy. The promising results of inhibition of c-Kit for treatment of cancers have been observed in some cancers such as gastrointestinal stromal tumor, acute myeloid leukemia, melanoma, and other tumors, and these results have encouraged attempts toward improvement of using c-Kit as a capable target for cancer therapy. This paper presents the findings of previous studies regarding c-Kit as a receptor tyrosine kinase and an oncogene, as well as its gene targets and signaling pathways in normal and cancer cells. The c-Kit gene location, protein structure, and the role of c-Kit in normal cell have been discussed. Comprehending the molecular mechanism underlying c-Kit-mediated tumorogenesis is consequently essential and may lead to the identification of future novel drug targets. The potential mechanisms by which c-Kit induces cellular transformation have been described. This study aims to elucidate the function of c-Kit for future cancer therapy. In addition, it has c-Kit inhibitor drug properties and their functions have been listed in tables and demonstrated in schematic pictures. This review also has collected previous studies that targeted c-Kit as a novel strategy for cancer therapy. This paper further emphasizes the advantages of this approach, as well as the limitations that must be addressed in the future. Finally, although c-Kit is an attractive target for cancer therapy, based on the outcomes of treatment of patients with c-Kit inhibitors, it is unlikely that Kit inhibitors alone can lead to cure. It seems that c-Kit mutations alone are not sufficient for tumorogenesis, but do play a crucial role in cancer occurrence.
Collapse
Affiliation(s)
| | - Behnam Kamalidehghan
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-e Pajoohesh; Medical Genetics Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Saleem
- Department of Urology; Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota; Section of Molecular Therapeutics & Cancer Health Disparity, The Hormel Institute, Austin, MN, USA
| | - Hasniza Zaman Huri
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Clinical Investigation Centre, University Malaya Medical Centre, Lembah Pantai, Kuala Lumpur, Malaysia
| | - Fatemeh Ahmadipour
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Aya F, Fernandez-Martinez A, Gaba L, Victoria I, Tosca M, Pineda E, Gascon P, Prat A, Arance A. Sequential treatment with immunotherapy and BRAF inhibitors in BRAF-mutant advanced melanoma. Clin Transl Oncol 2016; 19:119-124. [PMID: 27147251 DOI: 10.1007/s12094-016-1514-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Immunotherapy (IT) agents and BRAF inhibitors (BRAFi) are effective treatments for patients with advanced BRAF-mutant melanoma although the optimal sequence remains to be elucidated. The aim of this study was to compare the outcomes of two different cohorts of patients treated with BRAFi first, then IT or the reverse sequence. PATIENTS AND METHODS This is a retrospective study on two groups of patients: a cohort was treated first with BRAFi followed by immunotherapy (BRAFi-IT) and the other cohort with the reverse sequence (IT-BRAFi). Baseline characteristics and clinical outcomes were compared between the two cohorts. RESULTS A total of 25 patients were included in the study. Sixteen patients were given BRAFi-IT sequence and nine received IT-BRAFi sequence. No differences were observed in the characteristics of patients prior to each treatment between cohorts. Objective response rate (ORR) achieved by BRAFi were not different among groups. ORR achieved by IT was higher when administered after BRAFi (43.8 vs 0 %). Survival rates at 1-2 years were similar in both cohorts and median overall survival was not different for BRAFi-IT and IT-BRAFi (log rank test p = 0.97). CONCLUSIONS No differences were observed in OS between the two cohorts. These results support the indistinct use of IT or BRAFi as initial treatment in patients with metastatic BRAF-mutant melanoma, although higher rate of response to IT was observed when administered after BRAFi. Prospective randomized clinical trials are needed on this issue.
Collapse
Affiliation(s)
- F Aya
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain. .,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - A Fernandez-Martinez
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - L Gaba
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - I Victoria
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - M Tosca
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Pineda
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - P Gascon
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - A Prat
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - A Arance
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
39
|
Detection of BRAF Mutations Using a Fully Automated Platform and Comparison with High Resolution Melting, Real-Time Allele Specific Amplification, Immunohistochemistry and Next Generation Sequencing Assays, for Patients with Metastatic Melanoma. PLoS One 2016; 11:e0153576. [PMID: 27111917 PMCID: PMC4844167 DOI: 10.1371/journal.pone.0153576] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/31/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Metastatic melanoma is a severe disease with one of the highest mortality rate in skin diseases. Overall survival has significantly improved with immunotherapy and targeted therapies. Kinase inhibitors targeting BRAF V600 showed promising results. BRAF genotyping is mandatory for the prescription of anti-BRAF therapies. METHODS Fifty-nine formalin-fixed paraffin-embedded melanoma samples were assessed using High-Resolution-Melting (HRM) PCR, Real-time allele-specific amplification (RT-ASA) PCR, Next generation sequencing (NGS), immunohistochemistry (IHC) and the fully-automated molecular diagnostics platform IdyllaTM. Sensitivity, specificity, positive predictive value and negative predictive value were calculated using NGS as the reference standard to compare the different assays. RESULTS BRAF mutations were found in 28(47.5%), 29(49.2%), 31(52.5%), 29(49.2%) and 27(45.8%) samples with HRM, RT-ASA, NGS, IdyllaTM and IHC respectively. Twenty-six (81.2%) samples were found bearing a c.1799T>A (p.Val600Glu) mutation, three (9.4%) with a c.1798_1799delinsAA (p.Val600Lys) mutation and one with c.1789_1790delinsTC (p.Leu597Ser) mutation. Two samples were found bearing complex mutations. CONCLUSIONS HRM appears the less sensitive assay for the detection of BRAF V600 mutations. The RT-ASA, IdyllaTM and IHC assays are suitable for routine molecular diagnostics aiming at the prescription of anti-BRAF therapies. IdyllaTM assay is fully-automated and requires less than 2 minutes for samples preparation and is the fastest of the tested assays.
Collapse
|
40
|
Clinicopathological characteristics and mutation profiling in primary cutaneous melanoma. Am J Dermatopathol 2016; 37:389-97. [PMID: 25357015 DOI: 10.1097/dad.0000000000000241] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The incidence of mutations in malignant melanoma varies remarkably according to the subtype of melanoma, and this in itself is affected by racial and geographical factors. Studies screening melanoma case series for different types of mutations are relatively rare. METHOD The authors analyzed the frequency of various somatic point mutations of 10 genes in 106 primary cutaneous melanoma cases. The mutations (BRAF, NRAS, KIT, CDKN2A, KRAS, HRAS, PIK3CA, STK11, GNAQ, CTNNB1) were evaluated with real-time PCR-based PCR-Array through allele-specific amplification, and the results were correlated with various clinicopathological characteristics. RESULTS Mutations were found in 64.2% of the melanomas overall. BRAF (42.5%), NRAS (15.1%), and CDKN2A (13.2%) were the 3 most common mutations. BRAF and NRAS mutations were more frequent in nodular and superficial spreading melanomas (P < 0.001). Associations with BRAF mutation were as follows: male gender [odds ratio (OR) = 2.4], younger age (OR = 2.7), superficial spreading (OR = 15.6) and nodular melanoma (OR = 9.5), trunk localization (OR = 6.3), and intermittent sun exposure (OR = 4.6). A considerable percentage of V600K (44.4%) mutations were found among the BRAF mutations, whereas KIT mutations (3.8%) were less frequent. Multiple mutations were detected in 13.2% of the melanomas. The most common co-occurrences were in the BRAF, NRAS, and CDKN2A genes. CONCLUSIONS The authors analyzed 10 somatic mutations in the main subtypes of primary cutaneous melanomas from the western region of Turkey. Mutations were found in 64.2% of the melanomas overall. The most common mutations were in the BRAF and NRAS genes. In addition to other less common mutations, a notable number of multiple mutations were encountered. The multiplicity and concurrence of mutations in this study may provide further study areas for personalized targeted therapy.
Collapse
|
41
|
Silva IP, Salhi A, Giles KM, Vogelsang M, Han SW, Ismaili N, Lui KP, Robinson EM, Wilson MA, Shapiro RL, Pavlick A, Zhong J, Kirchhoff T, Osman I. Identification of a Novel Pathogenic Germline KDR Variant in Melanoma. Clin Cancer Res 2015; 22:2377-85. [PMID: 26631613 DOI: 10.1158/1078-0432.ccr-15-1811] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/17/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE The application of pan-cancer next-generation sequencing panels in the clinical setting has facilitated the identification of low frequency somatic mutations and the testing of new therapies in solid tumors using the "basket trial" scheme. However, little consideration has been given to the relevance of nonsynonymous germline variants, which are likely to be uncovered in tumors and germline and which may be relevant to prognostication and prediction of treatment response. EXPERIMENTAL DESIGN We analyzed matched tumor and normal DNA from 34 melanoma patients using an Ion Torrent cancer-associated gene panel. We elected to study the germline variant Q472H in the kinase insert domain receptor (KDR), which was identified in 35% of melanoma patients in both a pilot and an independent 1,223 patient cohort. Using patient-derived melanoma cell lines and human samples, we assessed proliferation, invasion, VEGF levels, and angiogenesis by analyzing tumor microvessel density (MVD) using anti-CD34 antibody. RESULTS Serum VEGF levels and tumor MVD were significantly higher in Q472H versus KDR wild-type (WD) patients. Primary cultures derived from melanomas harboring the KDR variant were more proliferative and invasive than KDR wild type. Finally, using a VEGFR2 antibody, we showed that KDR Q472H cells were sensitive to targeted inhibition of VEGFR2, an effect that was not observed in KDR WT cells. CONCLUSIONS Our data support the integration of germline analysis into personalized treatment decision-making and suggest that patients with germline KDR variant might benefit from antiangiogenesis treatment. Clin Cancer Res; 22(10); 2377-85. ©2015 AACR.
Collapse
Affiliation(s)
- Ines P Silva
- The Ronald O. Perelman Department of Dermatology, New York, New York. The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Amel Salhi
- The Ronald O. Perelman Department of Dermatology, New York, New York. The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Keith M Giles
- The Ronald O. Perelman Department of Dermatology, New York, New York. The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Matjaz Vogelsang
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. Departments of Population Health and Environmental Medicine, New York, New York
| | - Sung W Han
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. NYU Center for Health Informatics and Bioinformatics, New York, New York
| | | | - Kevin P Lui
- The Ronald O. Perelman Department of Dermatology, New York, New York. The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Eric M Robinson
- The Ronald O. Perelman Department of Dermatology, New York, New York. The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York
| | - Melissa A Wilson
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. Department of Medicine, New York, New York
| | - Richard L Shapiro
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. Department of Surgery, New York, New York
| | - Anna Pavlick
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. Department of Medicine, New York, New York
| | - Judy Zhong
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. NYU Center for Health Informatics and Bioinformatics, New York, New York
| | - Tomas Kirchhoff
- The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York. Departments of Population Health and Environmental Medicine, New York, New York
| | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, New York, New York. The Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York, New York.
| |
Collapse
|
42
|
Flisikowski K, Flisikowska T, Sikorska A, Perkowska A, Kind A, Schnieke A, Switonski M. Germline gene polymorphisms predisposing domestic mammals to carcinogenesis. Vet Comp Oncol 2015; 15:289-298. [PMID: 26575426 DOI: 10.1111/vco.12186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 09/15/2015] [Accepted: 09/20/2015] [Indexed: 12/31/2022]
Abstract
Cancer is a complex disease caused in part by predisposing germline gene polymorphisms. Knowledge of carcinogenesis in companion mammals (dog and cat) and some livestock species (pig and horse) is quite advanced. The prevalence of certain cancers varies by breed in these species, suggesting the presence of predisposing genetic variants in susceptible breeds. This review summarizes the present understanding of germline gene polymorphisms, including BRCA1, BRCA2, MC1R, KIT, NRAS and RAD51, associated with predisposition to melanoma, mammary cancer, osteosarcoma and histiocytic sarcoma in dogs, cats, pigs and horses. The predisposing variants in these species are discussed in the context of human germline gene polymorphisms associated with the same types of cancer.
Collapse
Affiliation(s)
- K Flisikowski
- Chair of Livestock Biotechnology, Technical University of Munich, Freising, Germany
| | - T Flisikowska
- Chair of Livestock Biotechnology, Technical University of Munich, Freising, Germany
| | - A Sikorska
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Poznan, Poland
| | - A Perkowska
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Poznan, Poland
| | - A Kind
- Chair of Livestock Biotechnology, Technical University of Munich, Freising, Germany
| | - A Schnieke
- Chair of Livestock Biotechnology, Technical University of Munich, Freising, Germany
| | - M Switonski
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Poznan, Poland
| |
Collapse
|
43
|
Lyu J, Wu Y, Li C, Wang R, Song H, Ren G, Guo W. Mutation scanning of BRAF
,NRAS
,KIT,
and GNAQ
/GNA11
in oral mucosal melanoma: a study of 57 cases. J Oral Pathol Med 2015; 45:295-301. [PMID: 26399561 DOI: 10.1111/jop.12358] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Jiong Lyu
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Yunteng Wu
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Chaojun Li
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Runxiang Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Hao Song
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Guoxin Ren
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Wei Guo
- Department of Oral and Maxillofacial-Head and Neck Oncology; Ninth people's Hospital, Shanghai Jiaotong University School of Medicine; Shanghai China
| |
Collapse
|
44
|
|
45
|
Abstract
Melanoma is increasing in incidence and represents an aggressive type of cancer. Efforts have focused on identifying genetic factors in melanoma carcinogenesis to guide prevention, screening, early detection, and targeted therapy. This article reviews the hereditary risk factors associated with melanoma and the known molecular pathways and genetic mutations associated with this disease. This article also explores the controversies associated with genetic testing and the latest advances in identifying genetic targets in melanoma, which offer promise for future application in the multidisciplinary management of melanoma.
Collapse
Affiliation(s)
- Omar M Rashid
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, SRB 4.24012, Tampa, FL 33612, USA; Bienes Comprehensive Cancer Center, Holy Cross Hospital, 4725 N Federal Highway, Fort Lauderdale, FL 33308, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, SRB 4.24012, Tampa, FL 33612, USA.
| |
Collapse
|
46
|
Riahi A, Kharrat M, Lariani I, Chaabouni-Bouhamed H. High-resolution melting (HRM) assay for the detection of recurrent BRCA1/BRCA2 germline mutations in Tunisian breast/ovarian cancer families. Fam Cancer 2015; 13:603-9. [PMID: 25069718 DOI: 10.1007/s10689-014-9740-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Germline deleterious mutations in the BRCA1/BRCA2 genes are associated with an increased risk for the development of breast and ovarian cancer. Given the large size of these genes the detection of such mutations represents a considerable technical challenge. Therefore, the development of cost-effective and rapid methods to identify these mutations became a necessity. High resolution melting analysis (HRM) is a rapid and efficient technique extensively employed as high-throughput mutation scanning method. The purpose of our study was to assess the specificity and sensitivity of HRM for BRCA1 and BRCA2 genes scanning. As a first step we estimate the ability of HRM for detection mutations in a set of 21 heterozygous samples harboring 8 different known BRCA1/BRCA2 variations, all samples had been preliminarily investigated by direct sequencing, and then we performed a blinded analysis by HRM in a set of 68 further sporadic samples of unknown genotype. All tested heterozygous BRCA1/BRCA2 variants were easily identified. However the HRM assay revealed further alteration that we initially had not searched (one unclassified variant). Furthermore, sequencing confirmed all the HRM detected mutations in the set of unknown samples, including homozygous changes, indicating that in this cohort, with the optimized assays, the mutations detections sensitivity and specificity were 100 %. HRM is a simple, rapid and efficient scanning method for known and unknown BRCA1/BRCA2 germline mutations. Consequently the method will allow for the economical screening of recurrent mutations in Tunisian population.
Collapse
Affiliation(s)
- Aouatef Riahi
- Laboratoire Génétique Humaine, Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
| | | | | | | |
Collapse
|
47
|
Iurlo A, Gianelli U, Beghini A, Spinelli O, Orofino N, Lazzaroni F, Cambiaghi S, Intermesoli T, Rambaldi A, Cortelezzi A. Identification of kit(M541L) somatic mutation in chronic eosinophilic leukemia, not otherwise specified and its implication in low-dose imatinib response. Oncotarget 2015; 5:4665-70. [PMID: 25015329 PMCID: PMC4148089 DOI: 10.18632/oncotarget.1941] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Activating mutations of KIT receptor tyrosine kinase have been reported in different neoplasms. The M541L KIT substitution (KITM541L) has been described to be associated with pediatric mastocytosis, to enhance growth rate of the affected cells and to confer higher sensitivity to imatinib therapy. We investigated the presence of KITM541L in five males with chronic eosinophilic leukemia, not otherwise specified (CEL, NOS), all negative for Platelet-derived growth factor-alpha (PDGFR) or PDGFRbeta abnormalities, which responded to imatinib therapy. To assess whether the mutation was constitutive or somatic in nature, we evaluated its presence analyzing either the neoplastic or normal cell population (epidermal cells or CD3-positive T lymphocytes). KITM541L substitution was found in 4 out of 5 patients and in all it was somatic in nature. All patients were treated with low dose imatinib (100 mg daily orally), achieving complete and persistent clinical and hematological remission (median follow-up 74 months). One patient relapsed after 50 months. Our study strongly suggests to search for the KITM541L in patients with CEL, NOS, negative for PDGFRalpha and PDGFRbeta abnormalities, to identify a subgroup of cases who may benefit from low dose imatinib therapy.
Collapse
Affiliation(s)
- Alessandra Iurlo
- Hematology and Transplantation Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy; Oncohematology Unit of the Elderly, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu YP, Wu HY, Yang X, Xu HQ, Chen D, Huang Q, Fu WL. Diagnostic accuracy of high resolution melting analysis for detection of KRAS mutations: a systematic review and meta-analysis. Sci Rep 2014; 4:7521. [PMID: 25515911 PMCID: PMC4268648 DOI: 10.1038/srep07521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 11/27/2014] [Indexed: 01/01/2023] Open
Abstract
Increasing evidence points to a negative correlation between KRAS mutations and patients' responses to anti-EGFR monoclonal antibody treatment. Therefore, patients must undergo KRAS mutation detection to be eligible for treatment. High resolution melting analysis (HRM) is gaining increasing attention in KRAS mutation detection. However, its accuracy has not been systematically evaluated. We conducted a meta-analysis of published articles, involving 13 articles with 1,520 samples, to assess its diagnostic accuracy compared with DNA sequencing. The quality of included articles was assessed using the revised Quality Assessment for Studies of Diagnostic Accuracy (QUADAS-2) tools. Random effects models were applied to analyze the performance of pooled characteristics. The overall sensitivity and specificity of HRM were 0.99 (95% confidence interval [CI]: 0.98-1.00) and 0.96 (95%CI: 0.94-0.97), respectively. The area under the summary receiver operating characteristic curve was 0.996. High sensitivity and specificity, less labor, rapid turn-around and the closed-tube format of HRM make it an attractive choice for rapid detection of KRAS mutations in clinical practice. The burden of DNA sequencing can be reduced dramatically by the implementation of HRM, but positive results still need to be sequenced for diagnostic confirmation.
Collapse
Affiliation(s)
- Yue-Ping Liu
- 1] Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China [2] Department of Laboratory Medicine, 477TH Hospital of PLA, Xiangyang City, 400013, Hubei Province, PR China
| | - Hai-Yan Wu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| | - Xiang Yang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| | - Han-Qing Xu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| | - Dong Chen
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| | - Qing Huang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| | - Wei-Ling Fu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| |
Collapse
|
49
|
Mucosal Melanoma of the Head and Neck: A Systematic Review of the Literature. Int J Radiat Oncol Biol Phys 2014; 90:1108-18. [DOI: 10.1016/j.ijrobp.2014.03.042] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/14/2014] [Accepted: 03/25/2014] [Indexed: 02/03/2023]
|
50
|
Wu S, Kuo H, Li WQ, Canales AL, Han J, Qureshi AA. Association between BRAFV600E and NRASQ61R mutations and clinicopathologic characteristics, risk factors and clinical outcome of primary invasive cutaneous melanoma. Cancer Causes Control 2014; 25:1379-86. [PMID: 25048604 PMCID: PMC4220546 DOI: 10.1007/s10552-014-0443-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 07/10/2014] [Indexed: 12/19/2022]
Abstract
PURPOSE Previous studies suggest that solar UV exposure in early life is predictive of cutaneous melanoma risk in adulthood, whereas the relation of BRAF mutation with sun exposure and disease prognosis has been less certain. We investigated the associations between BRAF(V600E) and NRAS(Q61R) mutations and known risk factors, clinicopathologic characteristics and clinical outcomes of melanoma in a case series of primary invasive cutaneous melanoma from the Nurses' Health Study (NHS). METHODS Somatic BRAF(V600E) and NRAS(Q61R) mutations of 127 primary invasive melanomas from the NHS cohort were determined by pyrosequencing using formalin-fixed, paraffin-embedded block tissues. Logistic regression analyses were performed to detect the associations of mutations with melanoma risk factors, and Kaplan-Meier method was used to examine associations between mutations and survival. RESULTS The odds ratios for harboring BRAF(V600E) mutations were 5.54 (95% CI 1.19-25.8, p(trend) = 0.02) for women residing in states with UV index ≥ 7 versus those residing in states with UV index ≤5 at 30 years of age. Patients with BRAF(V600E) mutations tended to have shorter melanoma-specific survival when compared to patients with wild type at both loci (median survival time 110 vs. 159 months) (p = 0.03). No association was found between NRASQ61R mutation and melanoma risk factors or melanoma-specific survival. CONCLUSIONS BRAF(V600E) mutations in primary cutaneous melanomas were associated with residence in locations with medium and high UV indices in mid-life. BRAF(V600E) mutation may be associated with an unfavorable prognosis among melanoma patients.
Collapse
Affiliation(s)
- Shaowei Wu
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Helen Kuo
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wen-Qing Li
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Alvaro Laga Canales
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana, USA
- Department of Dermatology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abrar A. Qureshi
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence, RI, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|