1
|
Fernandez-Flores A. "Pathway Leap": A New Molecular Phenomenon to Consider in the Pathogenesis of Melanocytic Tumors. Am J Dermatopathol 2025; 47:178-183. [PMID: 39787342 DOI: 10.1097/dad.0000000000002916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
INTRODUCTION The current WHO classification of melanocytic tumors distinguishes 9 pathogenic routes. This classification is based on the conceptual interpretation that melanocytic tumors evolve from benign counterparts, accumulating mutations, eventually developing into melanomas with metastatic and potentially lethal capacity. In this article, we present a molecular study of 2 melanocytic tumors that suggest a "leap" from pathogenic routes IV to I. MATERIALS AND METHODS Two recent melanocytic tumors were selected, each exhibiting 2 contiguous melanocytic populations of distinct morphology, without separation between them. One population corresponded to a common melanocytic nevus (with morphology consistent with route I), while the other population displayed epithelioid morphology, consistent with route IV. Immunohistochemical studies were performed in both cases, as well as molecular studies using PCR to search for mutations in the NRAS and BRAF genes. For the molecular study, both populations were manually separated by microdissection. RESULTS In both cases, the melanocytic population consistent with route I showed a BRAF mutation. In both cases, the epithelioid population did not present a BRAF mutation. No NRAS mutations were observed in any of the populations. CONCLUSIONS These findings suggest the existence of a molecular phenomenon of "leap" between pathways, which we have termed "pathway leap." This could explain the enigmatic group of tumors that the WHO classifies under the heading of "combined nevi." This group could be more frequent than suspected, because microdissection is not a technique commonly used in the daily diagnosis of melanocytic tumors.
Collapse
Affiliation(s)
- Angel Fernandez-Flores
- Department of Cellular Pathology, Hospital El Bierzo, Ponferrada, Spain
- Department of Cellular Pathology, Hospital de la Reina, Ponferrada, Spain ; and
- Department of Research. Institute for Biomedical Research of A Coruña (INIBIC). University of A Coruña (UDC). A Coruña, Spain
| |
Collapse
|
2
|
Kerkour T, Zhou C, Hollestein L, Mooyaart A. Genetic Concordance in Primary Cutaneous Melanoma and Matched Metastasis: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:16281. [PMID: 38003476 PMCID: PMC10671327 DOI: 10.3390/ijms242216281] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Studying primary melanoma and its corresponding metastasis has twofold benefits. Firstly, to better understand tumor biology, and secondly, to determine which sample should be examined in assessing drug targets. This study systematically analyzed all the literature on primary melanoma and its matched metastasis. Following PRISMA guidelines, we searched multiple medical databases for relevant publications from January 2000 to December 2022, assessed the quality of the primary-level studies using the QUIPS tool, and summarized the concordance rate of the most reported genes using the random-effects model. Finally, we evaluated the inter-study heterogeneity using the subgroup analysis. Thirty-one studies investigated the concordance of BRAF and NRAS in 1220 and 629 patients, respectively. The pooled concordance rate was 89.4% [95% CI: 84.5; 93.5] for BRAF and 97.8% [95% CI: 95.8; 99.4] for NRAS. When high-quality studies were considered, only BRAF mutation status consistency increased. Five studies reported the concordance status of c-KIT (93%, 44 patients) and TERT promoter (64%, 53 patients). Lastly, three studies analyzed the concordance of cancer genes involved in the signaling pathways, apoptosis, and proliferation, such as CDKN2A (25%, four patients), TP53 (44%, nine patients), and PIK3CA (20%, five patients). Our study found that the concordance of known drug targets (mainly BRAF) during melanoma progression is higher than in previous meta-analyses, likely due to advances in molecular techniques. Furthermore, significant heterogeneity exists in the genes involved in the melanoma genetic makeup; although our results are based on small patient samples, more research is necessary for validation.
Collapse
Affiliation(s)
- Thamila Kerkour
- Department of Dermatology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (T.K.); (C.Z.); (L.H.)
| | - Catherine Zhou
- Department of Dermatology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (T.K.); (C.Z.); (L.H.)
| | - Loes Hollestein
- Department of Dermatology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (T.K.); (C.Z.); (L.H.)
| | - Antien Mooyaart
- Department of Pathology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
3
|
Maher NG, Scolyer RA, Colebatch AJ. Biology and genetics of acquired and congenital melanocytic naevi. Pathology 2023; 55:169-177. [PMID: 36635156 DOI: 10.1016/j.pathol.2022.12.344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Acquired and congenital melanocytic naevi are common benign neoplasms. Understanding their biology and genetics will help clinicians and pathologists correctly diagnose melanocytic tumours, and generate insights into naevus aetiology and melanomagenesis. Genomic data from published studies analysing acquired and congenital melanocytic naevi, including oncogenic driver mutations, common melanoma associated mutations, copy number aberrations, somatic mutation signature patterns, methylation profile, and single nucleotide polymorphisms, were reviewed. Correlation of genomic changes to dermoscopic features, particular anatomic sites and total body naevus counts, was also performed. This review also highlights current scientific theories and evidence concerning naevi growth arrest. Acquired and congenital melanocytic naevi show simple genomes, typically characterised by mutually exclusive single oncogenic driver mutations in either BRAF or NRAS genes. Genomic differences exist between acquired and congenital naevi, common and dysplastic naevi, and by dermoscopic features. Acquired naevi show a higher rate of BRAF hotspot mutations and a lower rate of NRAS hotspot mutations compared to congenital naevi. Dysplastic naevi show upregulation of follicular keratinocyte-related genes compared to common naevi. Anatomical locations and DNA signatures of naevi implicates ultraviolet radiation and non-ultraviolet radiation pathways in naevogenesis. DNA driver point mutations in acquired and congenital melanocytic naevi have been well characterised. Future research is required to better understand transcriptional and epigenetic changes in naevi, as well as those regulating naevus growth arrest and cell environment signalling.
Collapse
Affiliation(s)
- Nigel G Maher
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Andrew J Colebatch
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
4
|
Field carcinogenesis and biological significance of the potential of the bystander effect: carcinogenesis, therapeutic response, and tissue regeneration. Surg Today 2022; 53:545-553. [PMID: 35576018 DOI: 10.1007/s00595-022-02524-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
Abstract
The "bystander effect" is a transmission phenomenon mediating communication from target to non-target cells, as well as cell-to-cell interactions between neighboring and distantly located cells. In this narrative review, we describe the fundamental and clinical significance of the bystander effect with respect to cell-to-cell interactions in carcinogenesis, therapeutic response, and tissue regeneration. In carcinogenesis, the bystander effect mediates communications between tumor microenvironments and non-malignant epithelial cells and has been suggested to impact heterogeneous tumorigenic cells in tumors and cancerized fields. In therapeutic response, the bystander effect mediates communications between drug-sensitive and drug-resistant cells and may transmit both drug efficacy and resistance. Therefore, control of therapeutic response transmission via the bystander effect might offer a promising future cancer treatment. Finally, in tissue regeneration, circulating cells and stromal cells may differentiate into various cells for the purpose of tissue regeneration under direction of the bystander effect arising from surrounding cells in a defective space. We hope that the findings we present will promote the development of innovative cancer therapies and tissue regeneration methodologies from the viewpoint of cell-to-cell interactions through the bystander effect.
Collapse
|
5
|
Mitogen-activating protein kinase pathway alterations in Langerhans cell histiocytosis. Curr Opin Oncol 2021; 33:101-109. [PMID: 33315630 DOI: 10.1097/cco.0000000000000707] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Langerhans cell histiocytosis (LCH) is a rare disorder characterized by the infiltration of involved tissues by specialized dendritic cells. The demonstration of the constant activation of the mitogen-activated protein kinase (MAPK) pathway in LCH lesions has been a breakthrough in the understanding of the pathogenesis of this rare disease. We will summarize the current knowledge on MAPK alterations in LCH and the new therapeutic options indicated by these findings. RECENT FINDINGS Since the description of the B-Raf proto-oncogene, serine/threonine kinase (BRAF)V600E mutation in LCH lesions, several other molecular alterations affecting the MAPK pathway have been identified in most cases. Based on these driver alterations, LCH cells were shown to be derived from hematopoietic precursors, which yielded the current concept of LCH as a myeloid inflammatory neoplasia. MAPK pathway inhibitors have emerged as an innovative therapy in severe forms of LCH, resulting in virtually no acquired resistance. However, although they are highly effective, their effect is only temporary, as the disease relapses upon discontinuation of the treatment. SUMMARY LCH is an inflammatory myeloid neoplastic disorder, driven by mutations activating the MAPK pathway. MAPK-targeted treatments represent an important stepforward in the management of patients with severe progressive LCH.
Collapse
|
6
|
PERK mediates resistance to BRAF inhibition in melanoma with impaired PTEN. NPJ Precis Oncol 2021; 5:68. [PMID: 34282258 PMCID: PMC8289936 DOI: 10.1038/s41698-021-00207-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/22/2021] [Indexed: 12/02/2022] Open
Abstract
Targeting mutant BRAF in patients with melanomas harboring this oncogene has been highly successful as a first-line treatment, but other mutations may affect its efficacy and alter the route of acquired resistance resulting in recurrence and poor prognosis. As an evolving strategy, melanoma treatment needs to be expanded to include targets based on newly discovered emerging molecules and pathways. We here show that PERK plays a critical role in BRAF inhibitor-acquired resistance in melanoma with impaired PTEN. Inhibition of PERK by either shRNA or a pharmacological inhibitor blocked the growth of BRAF inhibitor-resistant melanoma with impaired PTEN in vitro and in vivo, suggesting an effective approach against melanomas with mutant BRAF and PTEN deficiency. Our current findings, along with our previous discovery that the AXL/AKT axis mediates resistance to BRAF inhibition in melanoma with wild-type PTEN, provide new insights toward a strategy for combating BRAF inhibition-acquired resistance in BRAF mutant melanoma with different PTEN statuses.
Collapse
|
7
|
Muzza M, Pogliaghi G, Persani L, Fugazzola L, Colombo C. Combined Mutational and Clonality Analyses Support the Existence of Intra-Tumor Heterogeneity in Papillary Thyroid Cancer. J Clin Med 2021; 10:jcm10122645. [PMID: 34208446 PMCID: PMC8234519 DOI: 10.3390/jcm10122645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Despite its potential clinical impact, intra-tumor genetic heterogeneity (ITH) has been scantly investigated in papillary thyroid cancer (PTC). We studied ITH in PTC by combining, for the first time, data derived from the evaluation of the normalized allelic frequencies (NAF) of the mutation/s, using a customized MassARRAY panel, and those obtained by the HUMARA clonality assay. Among tumors with a single mutation, 80% of cases with NAF 50 ± 5% were clonal, consistent with the presence of a single mutated clone, while 20% of cases showed a polyclonal pattern, suggesting the presence of the same mutation in two or more clones. Differently, all cases with NAF < 45% were polyclonal. Among tumors with double mutation, cases with both mutations showing NAF 50 ± 5% were monoclonal, consistent with the presence of a single clone harboring both mutations. On the other hand, all cases with double mutation at NAF < 45% were polyclonal, indicating the presence of two clones with different mutations. Finally, no significant differences in the clinico-pathological characteristics were found between monoclonal and polyclonal tumors. In conclusion, the present study adds insights into the concept of ITH in PTC, which warrants attention because the occurrence of this phenomenon is likely to affect the response to targeted drugs.
Collapse
Affiliation(s)
- Marina Muzza
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (M.M.); (G.P.)
| | - Gabriele Pogliaghi
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (M.M.); (G.P.)
| | - Luca Persani
- Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (L.P.); (C.C.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Laura Fugazzola
- Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (L.P.); (C.C.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
- Correspondence:
| | - Carla Colombo
- Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, 20095 Milan, Italy; (L.P.); (C.C.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| |
Collapse
|
8
|
Sevilla A, Morales MC, Ezkurra PA, Rasero J, Velasco V, Cancho-Galan G, Sánchez-Diez A, Mujika K, Penas C, Smith I, Asumendi A, Cortés JM, Boyano MD, Alonso S. BRAF V600E mutational load as a prognosis biomarker in malignant melanoma. PLoS One 2020; 15:e0230136. [PMID: 32168325 PMCID: PMC7069620 DOI: 10.1371/journal.pone.0230136] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/21/2020] [Indexed: 11/19/2022] Open
Abstract
Analyzing the mutational load of driver mutations in melanoma could provide valuable information regarding its progression. We aimed at analyzing the heterogeneity of mutational load of BRAF V600E in biopsies of melanoma patients of different stages, and investigating its potential as a prognosis factor. Mutational load of BRAF V600E was analyzed by digital PCR in 78 biopsies of melanoma patients of different stages and 10 nevi. The BRAF V600E load was compared among biopsies of different stages. Results showed a great variability in the load of V600E (0%-81%). Interestingly, we observed a significant difference in the load of V600E between the early and late melanoma stages, in the sense of an inverse correlation between BRAF V600E mutational load and melanoma progression. In addition, a machine learning approach showed that the mutational load of BRAF V600E could be a good predictor of metastasis in stage II patients. Our results suggest that BRAF V600E is a promising biomarker of prognosis in stage II patients.
Collapse
Affiliation(s)
- Arrate Sevilla
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, UPV/EHU, Leioa, Spain
| | - M. Celia Morales
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
| | - Pilar A. Ezkurra
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
| | - Javier Rasero
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
| | - Verónica Velasco
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Pathology, Cruces University Hospital, Barakaldo, Spain
| | - Goikoane Cancho-Galan
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Pathology, Basurto University Hospital, Bilbao, Spain
| | - Ana Sánchez-Diez
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Dermatology, Basurto University Hospital, Bilbao, Spain
| | - Karmele Mujika
- Department of Oncology, Onkologikoa Hospital, Donostia, Spain
- Biodonostia Health Research Institute, Donostia-San Sebastián, Spain
| | - Cristina Penas
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
| | - Isabel Smith
- Department of Zoology and Animal Cell Biology, Faculty of Science and Technology, UPV/EHU, Leioa, Spain
| | - Aintzane Asumendi
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
| | - Jesús M. Cortés
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque: The Basque Foundation for Science, Bilbao, Spain
| | - Maria Dolores Boyano
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
- Biocruces-Bizkaia Health Research Institute, Barakaldo, Spain
| | - Santos Alonso
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, UPV/EHU, Leioa, Spain
| |
Collapse
|
9
|
Fernandes M, Barcelos D, Comodo AN, Guimarães DP, Lopes Carapeto FC, Cardili L, de Sousa Morães L, Cerutti Ap J, Landman Ap G. Acral Lentiginous Melanomas Harbour Intratumor Heterogeneity in BRAF Exon 15, With Mutations Distinct From V600E/V600K. Am J Dermatopathol 2019; 41:733-740. [PMID: 31021835 DOI: 10.1097/dad.0000000000001418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The choice of appropriate therapeutic strategies may be influenced by intratumor heterogeneity and makes cancer treatment considerably more challenging. We aimed to evaluate the heterogeneity of BRAF exon 15 mutations in different areas of acral lentiginous melanoma (ALM). The entire exon 15 was sequenced in 4 different areas of paraffin-embedded samples from 26 patients with ALM. A total of 26 of 49 cases of ≥1 mm in depth of ALM identified by clinical, anatomical, and pathological data fulfilled the inclusion and exclusion criteria for this study. Tumors had a mean Breslow depth of 7.2 mm and an average mitotic index of 3 mitosis/mm. Mutations distinct from the common V600E and V600K were detected in 31%, and intratumor heterogeneity was observed in 31% of samples. Interestingly, 63.5% of all mutations had been previously associated with cancer. Most (62.5%) of the missense BRAF exon 15 mutations found in the ALM samples examined here were deemed "detrimental" for protein function according to at least 2 functional prediction programs, and 3 mutations (37.5%) were predicted to be "neutral," with no effect on protein function. BRAF exon 15 mutations were detected frequently in ALM and displayed heterogeneity, a finding to be further investigated.
Collapse
Affiliation(s)
- Mariana Fernandes
- Department of Pathology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Denise Barcelos
- Department of Pathology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | - Leonardo Cardili
- Department of Pathology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lais de Sousa Morães
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
| | - Janete Cerutti Ap
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
| | - Gilles Landman Ap
- Department of Pathology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Orchard GE, Wojcik K, Rickaby W, Martin B, Semkova K, Shams F, Stefanato CM. Immunohistochemical detection of V600E BRAF mutation is a useful primary screening tool for malignant melanoma. Br J Biomed Sci 2019; 76:77-82. [DOI: 10.1080/09674845.2019.1592885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- GE Orchard
- Viapath Analytics, Dermatopathology Department, St John’s Institute of Dermatology, St. Thomas’ Hospital
| | - K Wojcik
- Viapath Analytics, Dermatopathology Department, St John’s Institute of Dermatology, St. Thomas’ Hospital
| | - W Rickaby
- Dermatopathology Department, St. John’s Institute of Dermatology, St. Thomas’ Hospital
| | - B Martin
- Dermatopathology Department, St. John’s Institute of Dermatology, St. Thomas’ Hospital
| | - K Semkova
- Dermatopathology Department, St. John’s Institute of Dermatology, St. Thomas’ Hospital
| | - F Shams
- Imperial College Healthcare NHS Trust, St. Mary’s Hospital
| | - CM Stefanato
- Dermatopathology Department, St. John’s Institute of Dermatology, St. Thomas’ Hospital
| |
Collapse
|
11
|
Stark MS, Tan JM, Tom L, Jagirdar K, Lambie D, Schaider H, Soyer HP, Sturm RA. Whole-Exome Sequencing of Acquired Nevi Identifies Mechanisms for Development and Maintenance of Benign Neoplasms. J Invest Dermatol 2018; 138:1636-1644. [PMID: 29476775 DOI: 10.1016/j.jid.2018.02.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 01/08/2023]
Abstract
The melanoma transformation rate of an individual nevus is very low despite the detection of oncogenic BRAF or NRAS mutations in 100% of nevi. Acquired melanocytic nevi do, however, mimic melanoma, and approximately 30% of all melanomas arise within pre-existing nevi. Using whole-exome sequencing of 30 matched nevi, adjacent normal skin, and saliva we sought to identify the underlying genetic mechanisms for nevus development. All nevi were clinically, dermoscopically, and histopathologically documented. In addition to identifying somatic mutations, we found mutational signatures relating to UVR mirroring those found in cutaneous melanoma. In nevi we frequently observed the presence of the UVR mutation signature compared with adjacent normal skin (97% vs. 10%, respectively). Copy number aberration analysis showed that for nevi with copy number loss of tumor suppressor genes, this loss was balanced by loss of potent oncogenes. Moreover, reticular and nonspecific patterned nevi showed an increased (P < 0.0001) number of copy number aberrations compared with globular nevi. The mutation signature data generated in this study confirms that UVR strongly contributes to nevogenesis. Copy number changes reflect at a genomic level the dermoscopic differences of acquired melanocytic nevi. Finally, we propose that the balanced loss of tumor suppressor genes and oncogenes is a protective mechanism of acquired melanocytic nevi.
Collapse
Affiliation(s)
- Mitchell S Stark
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia.
| | - Jean-Marie Tan
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Lisa Tom
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kasturee Jagirdar
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | | | - Helmut Schaider
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia; Department of Dermatology, Princess Alexandra Hospital, Brisbane, Australia
| | - H Peter Soyer
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia; Department of Dermatology, Princess Alexandra Hospital, Brisbane, Australia
| | - Richard A Sturm
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Abstract
Tumorigenesis begins long before the growth of a clinically detectable lesion and, indeed, even before any of the usual morphological correlates of pre-malignancy are recognizable. Field cancerization, which is the replacement of the normal cell population by a cancer-primed cell population that may show no morphological change, is now recognized to underlie the development of many types of cancer, including the common carcinomas of the lung, colon, skin, prostate and bladder. Field cancerization is the consequence of the evolution of somatic cells in the body that results in cells that carry some but not all phenotypes required for malignancy. Here, we review the evidence of field cancerization across organs and examine the biological mechanisms that drive the evolutionary process that results in field creation. We discuss the clinical implications, principally, how measurements of the cancerized field could improve cancer risk prediction in patients with pre-malignant disease.
Collapse
Affiliation(s)
- Kit Curtius
- Centre for Tumour Biology, Barts Cancer Institute, EC1M 6BQ London, UK
| | - Nicholas A Wright
- Centre for Tumour Biology, Barts Cancer Institute, EC1M 6BQ London, UK
| | - Trevor A Graham
- Centre for Tumour Biology, Barts Cancer Institute, EC1M 6BQ London, UK
| |
Collapse
|
13
|
Testa U, Castelli G, Pelosi E. Melanoma: Genetic Abnormalities, Tumor Progression, Clonal Evolution and Tumor Initiating Cells. Med Sci (Basel) 2017; 5:E28. [PMID: 29156643 PMCID: PMC5753657 DOI: 10.3390/medsci5040028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/31/2017] [Accepted: 11/08/2017] [Indexed: 12/11/2022] Open
Abstract
Melanoma is an aggressive neoplasia issued from the malignant transformation of melanocytes, the pigment-generating cells of the skin. It is responsible for about 75% of deaths due to skin cancers. Melanoma is a phenotypically and molecularly heterogeneous disease: cutaneous, uveal, acral, and mucosal melanomas have different clinical courses, are associated with different mutational profiles, and possess distinct risk factors. The discovery of the molecular abnormalities underlying melanomas has led to the promising improvement of therapy, and further progress is expected in the near future. The study of melanoma precursor lesions has led to the suggestion that the pathway of tumor evolution implies the progression from benign naevi, to dysplastic naevi, to melanoma in situ and then to invasive and metastatic melanoma. The gene alterations characterizing melanomas tend to accumulate in these precursor lesions in a sequential order. Studies carried out in recent years have, in part, elucidated the great tumorigenic potential of melanoma tumor cells. These findings have led to speculation that the cancer stem cell model cannot be applied to melanoma because, in this malignancy, tumor cells possess an intrinsic plasticity, conferring the capacity to initiate and maintain the neoplastic process to phenotypically different tumor cells.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
14
|
Oliveira ÉAD, Lima DSD, Cardozo LE, Souza GFD, de Souza N, Alves-Fernandes DK, Faião-Flores F, Quincoces JAP, Barros SBDM, Nakaya HI, Monteiro G, Maria-Engler SS. Toxicogenomic and bioinformatics platforms to identify key molecular mechanisms of a curcumin-analogue DM-1 toxicity in melanoma cells. Pharmacol Res 2017; 125:178-187. [PMID: 28882690 DOI: 10.1016/j.phrs.2017.08.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/31/2017] [Accepted: 08/30/2017] [Indexed: 12/17/2022]
Abstract
Melanoma is a highly invasive and metastatic cancer with high mortality rates and chemoresistance. Around 50% of melanomas are driven by activating mutations in BRAF that has led to the development of potent anti-BRAF inhibitors. However resistance to anti-BRAF therapy usually develops within a few months and consequently there is a need to identify alternative therapies that will bypass BRAF inhibitor resistance. The curcumin analogue DM-1 (sodium 4-[5-(4-hydroxy-3-methoxy-phenyl)-3-oxo-penta-1,4-dienyl]-2-methoxy-phenolate) has substantial anti-tumor activity in melanoma, but its mechanism of action remains unclear. Here we use a synthetic lethal genetic screen in Saccharomyces cerevisiae to identify 211 genes implicated in sensitivity to DM-1 toxicity. From these 211 genes, 74 had close human orthologues implicated in oxidative phosphorylation, insulin signaling and iron and RNA metabolism. Further analysis identified 7 target genes (ADK, ATP6V0B, PEMT, TOP1, ZFP36, ZFP36L1, ZFP36L2) with differential expression during melanoma progression implicated in regulation of tumor progression, cell differentiation, and epithelial-mesenchymal transition. Of these TOP1 and ADK were regulated by DM-1 in treatment-naïve and vemurafenib-resistant melanoma cells respectively. These data reveal that the anticancer effect of curcumin analogues is likely to be mediated via multiple targets and identify several genes that represent candidates for combinatorial targeting in melanoma.
Collapse
Affiliation(s)
- Érica Aparecida de Oliveira
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Diogenes Saulo de Lima
- Computational Systems Biology Laboratory, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Lucas Esteves Cardozo
- Computational Systems Biology Laboratory, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | | | - Nayane de Souza
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Debora Kristina Alves-Fernandes
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Fernanda Faião-Flores
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | | | - Silvia Berlanga de Moraes Barros
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Helder I Nakaya
- Computational Systems Biology Laboratory, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Gisele Monteiro
- Biochemical Pharmaceutical Technology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil
| | - Silvya Stuchi Maria-Engler
- Skin Biology Group, Clinical Chemistry and Toxicology Department, School of Pharmaceutical Sciences, University of Sao Paulo, FCF/USP, Sao Paulo, Brazil.
| |
Collapse
|
15
|
Amaral T, Sinnberg T, Meier F, Krepler C, Levesque M, Niessner H, Garbe C. The mitogen-activated protein kinase pathway in melanoma part I - Activation and primary resistance mechanisms to BRAF inhibition. Eur J Cancer 2017; 73:85-92. [PMID: 28169047 DOI: 10.1016/j.ejca.2016.12.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 02/07/2023]
Abstract
Mitogen-activated protein kinase (MAPK) pathway has an important role in normal cells and can be activated under physiological conditions. MAPK pathway activation is a fundamental step in several intracellular processes requiring a sequential phosphorylation of the different pathway components. In normal cells, when MAPK pathway activation occurs, it leads to cell growth and differentiation. In order to prevent persistent MAPK pathway activation, physiological upstream negative feedback also takes place. In cells harbouring BRAFV600 mutations, the process leading to MAPK pathway activation is different, and the negative physiological feedback does not exist thus leading to permanent MAPK pathway activation, which ultimately can lead to uncontrolled proliferation. Targeted therapy with rapidly accelerated fibrosarcoma - B (BRAF) and/or mitogen-activated extracellular signal-regulated kinase kinase (MEK) inhibitors is indicated in patients with metastatic melanoma harboring BRAFV600 mutations. However, several different resistance mechanisms to this therapy were identified. In this review, we focus on primary or intrinsic resistance mechanisms to BRAF and MEK inhibition. In this setting, although a BRAF mutation is identified, there is no response to treatment with either BRAF or MEK inhibitor.
Collapse
Affiliation(s)
- Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Portuguese Air Force Health Direction, Paço do Lumiar, 1649-020 Lisbon, Portugal.
| | - Tobias Sinnberg
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74D, 01307 Dresden, Germany
| | - Clemens Krepler
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Mitchell Levesque
- University of Zürich Hospital, Department of Dermatology, Wagistrasse 14, CH-8952 Schlieren, Zürich, Switzerland
| | - Heike Niessner
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany
| |
Collapse
|
16
|
Abstract
Melanomas on sun-exposed skin are heterogeneous tumours, which can be subtyped on the basis of their cumulative levels of exposure to ultraviolet (UV) radiation. A melanocytic neoplasm can also be staged by how far it has progressed, ranging from a benign neoplasm, such as a naevus, to a malignant neoplasm, such as a metastatic melanoma. Each subtype of melanoma can evolve through distinct evolutionary trajectories, passing through (or sometimes skipping over) various stages of transformation. This Review delineates several of the more common progression trajectories that occur in the patient setting and proposes models for tumour evolution that integrate genetic, histopathological, clinical and biological insights from the melanoma literature.
Collapse
Affiliation(s)
- A Hunter Shain
- University of California, San Francisco, Departments of Dermatology and Pathology and Helen Diller Family Comprehensive Cancer Center, Box 3111, San Francisco, CA 94143, USA
| | - Boris C Bastian
- University of California, San Francisco, Departments of Dermatology and Pathology and Helen Diller Family Comprehensive Cancer Center, Box 3111, San Francisco, CA 94143, USA
| |
Collapse
|
17
|
Cellular Blue Nevus Diagnosed following Excision of Melanoma: A Challenge in Diagnosis. Case Rep Pathol 2016; 2016:8107671. [PMID: 27313934 PMCID: PMC4899595 DOI: 10.1155/2016/8107671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/07/2016] [Accepted: 03/27/2016] [Indexed: 12/17/2022] Open
Abstract
A case of a 41-year-old woman with a history of nodular melanoma (NM), associated with an indurated dome-shaped blue-black nodule with a diameter of 1.2 cm in the gluteal region, is presented. Clinical diagnosis of the lesion, present from birth, was blue nevus. Recently, the nodule has been showing a mild enlargement and thus complete resection was performed. Histological analysis revealed a pigmented lesion with an expansive pattern of extension into the dermis and the subcutaneous adipose tissue. The lesion displayed an alveolar pattern as well as a pigmented dendritic cell pattern. The histology was consistent with cellular blue nevus (CBN); however, the history of NM which was excised one year earlier, as well as the clinical information about the slow growing lesion, included a differential diagnosis of CBN, borderline melanocytic tumor, and malignant blue nevus. Additional immunohistochemical (HMB-45, p16, and Ki-67) and molecular (BRAF V600E mutation) analyses were performed on both lesions: the CBN-like and the previously excised NM. Along with lesion history and histological analyses, p16 staining and BRAF were useful diagnostic tools for confirming the benign nature of CBN in this case.
Collapse
|
18
|
Woltsche N, Schwab C, Deinlein T, Hofmann-Wellenhof R, Zalaudek I. Dermoscopy in the era of dermato-oncology: from bed to bench side and retour. Expert Rev Anticancer Ther 2016; 16:531-41. [DOI: 10.1586/14737140.2016.1168700] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
19
|
Bennett DC. Genetics of melanoma progression: the rise and fall of cell senescence. Pigment Cell Melanoma Res 2015; 29:122-40. [PMID: 26386262 DOI: 10.1111/pcmr.12422] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022]
Abstract
There are many links between cell senescence and the genetics of melanoma, meaning both familial susceptibility and somatic-genetic changes in sporadic melanoma. For example, CDKN2A, the best-known melanoma susceptibility gene, encodes two effectors of cell senescence, while other familial melanoma genes are related to telomeres and their maintenance. This article aimed to analyze our current knowledge of the genetic or epigenetic driver changes necessary to generate a cutaneous metastatic melanoma, the commonest order in which these occur, and the relation of these changes to the biology and pathology of melanoma progression. Emphasis is laid on the role of cell senescence and the escape from senescence leading to cellular immortality, the ability to divide indefinitely.
Collapse
Affiliation(s)
- Dorothy C Bennett
- Molecular Cell Sciences Research Centre, St George's, University of London, Cranmer Terrace, London, UK
| |
Collapse
|
20
|
Néel A, Artifoni M, Donadieu J, Lorillon G, Hamidou M, Tazi A. Histiocytose langerhansienne de l’adulte. Rev Med Interne 2015; 36:658-67. [DOI: 10.1016/j.revmed.2015.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 01/30/2015] [Accepted: 04/30/2015] [Indexed: 12/21/2022]
|
21
|
Palmieri G, Ombra M, Colombino M, Casula M, Sini M, Manca A, Paliogiannis P, Ascierto PA, Cossu A. Multiple Molecular Pathways in Melanomagenesis: Characterization of Therapeutic Targets. Front Oncol 2015; 5:183. [PMID: 26322273 PMCID: PMC4530319 DOI: 10.3389/fonc.2015.00183] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/26/2015] [Indexed: 12/12/2022] Open
Abstract
Molecular mechanisms involved in pathogenesis of malignant melanoma have been widely studied and novel therapeutic treatments developed in recent past years. Molecular targets for therapy have mostly been recognized in the RAS–RAF–MEK–ERK and PI3K–AKT signaling pathways; small-molecule inhibitors were drawn to specifically target key kinases. Unfortunately, these targeted drugs may display intrinsic or acquired resistance and various evidences suggest that inhibition of a single effector of the signal transduction cascades involved in melanoma pathogenesis may be ineffective in blocking the tumor growth. In this sense, a wider comprehension of the multiple molecular alterations accounting for either response or resistance to treatments with targeted inhibitors may be helpful in assessing, which is the most effective combination of such therapies. In the present review, we summarize the known molecular mechanisms underlying either intrinsic and acquired drug resistance either alternative roads to melanoma pathogenesis, which may become targets for innovative anticancer approaches.
Collapse
Affiliation(s)
- Giuseppe Palmieri
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - MariaNeve Ombra
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche , Avellino , Italy
| | - Maria Colombino
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - Milena Casula
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - MariaCristina Sini
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - Antonella Manca
- Unità di Genetica dei Tumori, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche , Sassari , Italy
| | - Panagiotis Paliogiannis
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche, Università di Sassari , Sassari , Italy
| | | | - Antonio Cossu
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche, Università di Sassari , Sassari , Italy
| |
Collapse
|
22
|
Aksenenko MB, Kirichenko AK, Ruksha TG. Russian study of morphological prognostic factors characterization in BRAF-mutant cutaneous melanoma. Pathol Res Pract 2015; 211:521-7. [PMID: 25888143 DOI: 10.1016/j.prp.2015.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/21/2015] [Accepted: 03/25/2015] [Indexed: 01/16/2023]
Abstract
Melanoma is one of the aggressive cancer types causing the majority of deaths in skin cancer patients. Mutational screening of the tumor revealed a number of driver mutations in oncogenes which enabled melanoma classification into a few molecular subtypes. BRAF is a key component of mitogen-activated kinase pathway; its activating mutation leads to accelerated melanoma cells proliferation, invasion and survival. Somatic mutations in BRAF were reported in various malignancies, including thyroid cancer, colorectal cancer and melanoma. Specific features of BRAF-positive tumors could have clinical implications as mutational alterations may have an impact on the biological behavior of the tumor and prognosis of the disease. In the present study, the frequency of BRAF V600E mutation was evaluated in Russian patients with melanocytic lesions, of which 41.25% were primary melanoma and 60% were melanocytic nevi. Melanoma patients with trunk localization were of younger age in the BRAF-positive group as compared with BRAF-negative patients. Immunohistochemical evaluations of Ki-67 expression, as well as matrix metalloproteinase-2, -9, were found to be equal in BRAF-positive and BRAF-negative tumors. MMP-2/MMP-9 immunoreactivity was observed in stromal and/or melanocytic cells both in melanoma and nevi patients. Besides tumor cells, MMP-9 expression was observed in lymphocytes in 27.2% of BRAF-positive and in 19.1% of BRAF-negative patients. Histopathological prognostic markers (Breslow thickness, mitotic index, ulceration, tumor infiltrating lymphocytes pattern) did not show any differences depending on BRAF V600E mutational status. The frequency of BRAF-positive melanomas in Russian cohort is similar to other Caucasian population rates. BRAF V600E mutation harboring tumors are more often observed in younger patients without specific features of morphological prognostic factors.
Collapse
Affiliation(s)
- M B Aksenenko
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia.
| | - A K Kirichenko
- Department of Pathological Anatomy, Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - T G Ruksha
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia.
| |
Collapse
|
23
|
Obert J, Tazi A. [Pulmonary manifestations of Langerhans cell histiocytosis]. Rev Mal Respir 2015; 32:850-66. [PMID: 26003197 DOI: 10.1016/j.rmr.2015.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/13/2015] [Indexed: 01/20/2023]
Abstract
Pulmonary Langerhans cell histiocytosis is a rare diffuse cystic interstitial pneumonia of unknown etiology that occurs selectively in young smokers of both genders. The multicenter studies conducted by the reference center have better defined the short and medium terms natural history of the disease and the clinical management of patients. A substantial proportion of patients experience a dramatic decline in their lung function soon after diagnosis. Importantly, smoking cessation is associated with a decreased risk of subsequent deterioration. Cladribine, a purine analogue, chemotherapy may dramatically improve lung function in patients with progressive pulmonary Langerhans cell histiocytosis, but this treatment should be used only in the setting of clinical research. Specific pulmonary hypertension therapies (anti-endothelin receptors, inhibitors of phosphodiesterases) may be used with caution in specialized centres for patients with severe pulmonary hypertension, and seem to be well tolerated. The recent identification of the V600E mutation of the BRAF oncogene in approximately half of the Langerhans cell histiocytosis lesions, including pulmonary granulomas, represents an important step forward in the understanding of the pathogenesis of Langerhans cell histiocytosis. Potentially it opens the way to targeted therapies.
Collapse
Affiliation(s)
- J Obert
- Service de pneumologie, Centre national de référence de l'histiocytose langerhansienne, hôpital Saint-Louis, Assistance publique-Hôpitaux de Paris, 1, avenue Claude-Vellefaux, 75475 Paris cedex 10, France
| | - A Tazi
- Service de pneumologie, Centre national de référence de l'histiocytose langerhansienne, hôpital Saint-Louis, Assistance publique-Hôpitaux de Paris, 1, avenue Claude-Vellefaux, 75475 Paris cedex 10, France; U1153 CRESS, équipe de recherche en biostatistiques et épidémiologie clinique, université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France.
| |
Collapse
|
24
|
Day F, Muranyi A, Singh S, Shanmugam K, Williams D, Byrne D, Pham K, Palmieri M, Tie J, Grogan T, Gibbs P, Sieber O, Waring P, Desai J. A mutant BRAF V600E-specific immunohistochemical assay: correlation with molecular mutation status and clinical outcome in colorectal cancer. Target Oncol 2014; 10:99-109. [PMID: 24859797 PMCID: PMC4363480 DOI: 10.1007/s11523-014-0319-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/09/2014] [Indexed: 12/12/2022]
Abstract
The B-type Raf kinase (BRAF) V600E mutation is a well-established biomarker for poor prognosis in metastatic colorectal cancer (mCRC) and is a highly attractive drug target. A barrier to the development of new therapies targeting BRAF V600E in mCRC is the low prevalence of mutations (approximately 10 %) and the current need for access to sequencing-based technologies which are not routinely available outside of large cancer centres. Availability of a standardised immunohistochemistry (IHC) test, more suited to routine pathology practice, would provide much broader access to patient identification. We sought to evaluate the accuracy and clinical utility of a recently developed BRAF V600E IHC method as a prognostic biomarker in a large cohort of community-based CRC patients. Archival tumour samples from 505 patients with stage I–IV CRC were immunohistochemically tested with two antibodies, pBR1 for total BRAF and VE1 for BRAF V600E. Cases were assessed by two blinded pathologists, and results were compared to BRAF V600E mutation status determined using DNA sequencing. Discordant cases were retested with a BRAF V600E SNaPshot assay. BRAF mutation status was correlated with overall survival (OS) in stage IV CRC. By DNA sequencing and IHC, 505 and 477 patients were respectively evaluable. Out of 477 patients, 56 (11. 7 %) had BRAF V600E mutations detected by sequencing and 63 (13.2 %) by IHC. Using DNA sequencing results as the reference, sensitivity and specificity for IHC were 98.2 % (55/56) and 98.1 % (413/421), respectively. IHC had a positive predictive value (PPV) of 87.3 % (55/63) and a negative predictive value (NPV) of 99.8 % (413/414). Compared to DNA sequencing plus retesting of available discordant cases by SNaPshot assay, IHC using the VE1 antibody had a 100 % sensitivity (59/59), specificity (416/416), NPV (416/416) and PPV (59/59). Stage IV CRC patients with BRAF V600E protein detected by IHC exhibited a significantly shorter overall survival (hazard ratio = 2.20, 95 % CI 1.26–3.83, p = 0.005), consistent with other published series. Immunohistochemistry using the BRAF V600E VE1 antibody is an accurate diagnostic assay in CRC. The test provides a simple, clinically applicable method of testing for the BRAF V600E mutation in routine practice.
Collapse
Affiliation(s)
- Fiona Day
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
| | | | | | | | - David Williams
- Department of Pathology, Austin Health, Heidelberg, VIC Australia
| | - David Byrne
- Pathology Department, Peter MacCallum Cancer Centre, East Melbourne, VIC Australia
| | - Kym Pham
- Faculty of Medicine, Dentistry and Health Sciences, Department of Pathology, University of Melbourne, Parkville, VIC Australia
| | - Michelle Palmieri
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
| | - Jeanne Tie
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville, VIC Australia
- Department of Medical Oncology, Western Hospital, Footscray, VIC Australia
| | | | - Peter Gibbs
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville, VIC Australia
- Department of Medical Oncology, Western Hospital, Footscray, VIC Australia
| | - Oliver Sieber
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
| | - Paul Waring
- Faculty of Medicine, Dentistry and Health Sciences, Department of Pathology, University of Melbourne, Parkville, VIC Australia
| | - Jayesh Desai
- Ludwig Colon Cancer Initiative Laboratory, Ludwig Institute for Cancer Research, Parkville, VIC Australia
- Faculty of Medicine, Dentistry and Health Sciences, Department of Surgery, University of Melbourne, Parkville, VIC Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville, VIC Australia
- Department of Medical Oncology, Western Hospital, Footscray, VIC Australia
| |
Collapse
|
25
|
Grichnik JM, Ross AL, Schneider SL, Sanchez MI, Eller MS, Hatzistergos KE. How, and from which cell sources, do nevi really develop? Exp Dermatol 2014; 23:310-3. [DOI: 10.1111/exd.12363] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2014] [Indexed: 12/16/2022]
Affiliation(s)
- James M. Grichnik
- Department of Dermatology and Cutaneous Surgery; University of Miami; Miller School of Medicine; Miami FL USA
- Sylvester Comprehensive Cancer Center; University of Miami; Miller School of Medicine; Miami FL USA
- Interdisciplinary Stem Cell Institute; University of Miami Miller School of Medicine; Miami FL USA
| | - Andrew L. Ross
- Department of Dermatology and Cutaneous Surgery; University of Miami; Miller School of Medicine; Miami FL USA
| | - Samantha L. Schneider
- Department of Dermatology and Cutaneous Surgery; University of Miami; Miller School of Medicine; Miami FL USA
| | - Margaret I. Sanchez
- Department of Dermatology and Cutaneous Surgery; University of Miami; Miller School of Medicine; Miami FL USA
- Sylvester Comprehensive Cancer Center; University of Miami; Miller School of Medicine; Miami FL USA
| | - Mark S. Eller
- Sylvester Comprehensive Cancer Center; University of Miami; Miller School of Medicine; Miami FL USA
| | | |
Collapse
|
26
|
Bastian BC. The molecular pathology of melanoma: an integrated taxonomy of melanocytic neoplasia. ANNUAL REVIEW OF PATHOLOGY 2014; 9:239-71. [PMID: 24460190 PMCID: PMC4831647 DOI: 10.1146/annurev-pathol-012513-104658] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Melanomas comprise multiple biologically distinct categories, which differ in cell of origin, age of onset, clinical and histologic presentation, pattern of metastasis, ethnic distribution, causative role of UV radiation, predisposing germ-line alterations, mutational processes, and patterns of somatic mutations. Neoplasms are initiated by gain-of-function mutations in one of several primary oncogenes, which typically lead to benign melanocytic nevi with characteristic histologic features. The progression of nevi is restrained by multiple tumor-suppressive mechanisms. Secondary genetic alterations override these barriers and promote intermediate or overtly malignant tumors along distinct progression trajectories. The current knowledge about the pathogenesis and clinical, histologic, and genetic features of primary melanocytic neoplasms is reviewed and integrated into a taxonomic framework.
Collapse
Affiliation(s)
- Boris C Bastian
- Departments of Dermatology and Pathology, Cardiovascular Research Institute, University of California, San Francisco, California 94158-9001;
| |
Collapse
|
27
|
Capovilla M. [Cellular and molecular mechanisms of carcinogenic side effects and resistance to BRAF inhibitors in metastatic melanoma with BRAFV600 mutation: state of the knowledge]. Ann Pathol 2013; 33:375-85. [PMID: 24331719 DOI: 10.1016/j.annpat.2013.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 01/07/2023]
Abstract
Cutaneous melanoma is a malignant tumor with a high metastatic potential. If an early treatment is associated with a favorable outcome, the prognosis of metastatic melanoma remains poor. Advances in molecular characterization of cancers, notably the discovery of BRAF gene mutations in metastatic melanoma, allowed to the recent development of targeted therapies against mutated BRAF protein. Despite high tumor response rates observed in clinical trials, these new drugs are associated with frequent secondary tumor resistance occurrence and paradoxical carcinogenic side effects. The cellular and molecular mechanisms of these carcinogenic side effects and secondary resistance are not yet fully elucidated and are actually intensely studied. This review of the literature focus on the mechanisms of these carcinogenic side effects and on the tumor resistance associated with anti-BRAF targeted therapies.
Collapse
MESH Headings
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Carcinoma, Squamous Cell/chemically induced
- Cell Transformation, Neoplastic/drug effects
- Drug Resistance, Neoplasm/genetics
- Enzyme Activation/drug effects
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Genes, ras
- Humans
- Indoles/adverse effects
- Indoles/pharmacology
- Indoles/therapeutic use
- Intercellular Signaling Peptides and Proteins/metabolism
- Keratoacanthoma/chemically induced
- Leukemia/chemically induced
- MAP Kinase Signaling System/drug effects
- Melanoma/chemically induced
- Melanoma/drug therapy
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/secondary
- Models, Biological
- Molecular Targeted Therapy
- Mutation, Missense
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neoplasms, Second Primary/chemically induced
- Neoplastic Stem Cells/enzymology
- Nevus, Pigmented/enzymology
- Nevus, Pigmented/pathology
- Point Mutation
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Proto-Oncogene Proteins B-raf/antagonists & inhibitors
- Proto-Oncogene Proteins B-raf/genetics
- Proto-Oncogene Proteins B-raf/physiology
- Proto-Oncogene Proteins c-raf/biosynthesis
- Proto-Oncogene Proteins c-raf/physiology
- Skin Neoplasms/chemically induced
- Sulfonamides/adverse effects
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Tumor Microenvironment
- Vemurafenib
Collapse
Affiliation(s)
- Mathieu Capovilla
- Service de pathologie, centre François-Baclesse, 3, avenue Général-Harris, BP 5026, 14076 Caen cedex 05, France.
| |
Collapse
|
28
|
Colombino M, Lissia A, Capone M, De Giorgi V, Massi D, Stanganelli I, Fonsatti E, Maio M, Botti G, Caracò C, Mozzillo N, Ascierto PA, Cossu A, Palmieri G. Heterogeneous distribution of BRAF/NRAS mutations among Italian patients with advanced melanoma. J Transl Med 2013; 11:202. [PMID: 23987572 PMCID: PMC3765741 DOI: 10.1186/1479-5876-11-202] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 08/27/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Prevalence and distribution of pathogenetic mutations in BRAF and NRAS genes were evaluated in multiple melanoma lesions from patients with different geographical origin within the same Italian population. METHODS Genomic DNA from a total of 749 tumor samples (451 primary tumors and 298 metastases) in 513 consecutively-collected patients with advanced melanoma (AJCC stages III and IV) was screened for mutations in exon 15 of BRAF gene and, at lower extension (354/513; 69%), in the entire coding DNA of NRAS gene by automated direct sequencing. Among tissues, 236 paired samples of primary melanomas and synchronous or asynchronous metastases were included into the screening. RESULTS Overall, mutations were detected in 49% primary melanomas and 51% metastases, for BRAF gene, and 15% primary tumors and 16% secondaries, for NRAS gene. A heterogeneous distribution of mutations in both genes was observed among the 451 primary melanomas according to patients' geographical origin: 61% vs. 42% (p = 0.0372) BRAF-mutated patients and 2% vs. 21% (p < 0.0001) NRAS-mutated cases were observed in Sardinian and non-Sardinian populations, respectively. Consistency in BRAF/NRAS mutations among paired samples was high for lymph node (91%) and visceral metastases (92.5%), but significantly lower for brain (79%; p = 0.0227) and skin (71%; p = 0.0009) metastases. CONCLUSIONS Our findings about the two main alterations occurring in the different tumor tissues from patients with advanced melanoma may be helpful in improving the management of such a disease.
Collapse
Affiliation(s)
- Maria Colombino
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti 07100, Sassari, Italy
| | - Amelia Lissia
- Institute of Pathology, University Hospital (AOU), Sassari, Italy
| | | | | | - Daniela Massi
- Institute of Pathology, University of Florence, Florence, Italy
| | - Ignazio Stanganelli
- Skin Cancer Unit, Istituto Scientifico Romagnolo Tumori (IRST), Meldola, Italy
| | - Ester Fonsatti
- Department of Oncology, University Hospital (AOU), Siena, Italy
| | - Michele Maio
- Department of Oncology, University Hospital (AOU), Siena, Italy
| | - Gerardo Botti
- National Tumour Institute “Fondazione Pascale”, Naples, Italy
| | - Corrado Caracò
- National Tumour Institute “Fondazione Pascale”, Naples, Italy
| | - Nicola Mozzillo
- National Tumour Institute “Fondazione Pascale”, Naples, Italy
| | | | - Antonio Cossu
- Institute of Pathology, University Hospital (AOU), Sassari, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Traversa La Crucca 3, Baldinca Li Punti 07100, Sassari, Italy
| |
Collapse
|
29
|
Zebary A, Omholt K, Vassilaki I, Höiom V, Lindén D, Viberg L, Kanter-Lewensohn L, Johansson CH, Hansson J. KIT, NRAS, BRAF and PTEN mutations in a sample of Swedish patients with acral lentiginous melanoma. J Dermatol Sci 2013; 72:284-9. [PMID: 23993026 DOI: 10.1016/j.jdermsci.2013.07.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 06/27/2013] [Accepted: 07/20/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Acral lentiginous melanoma (ALM) accounts for <10% of all melanomas in Caucasians. Although the involvement of KIT, NRAS and BRAF mutations is well known in ALM, the impact of these mutations on clinicopathological features has not been established. OBJECTIVE To define the KIT, NRAS, BRAF and PTEN mutation frequencies in Swedish patients with ALM and to evaluate the impact of mutation status on patient and tumor characteristics. METHODS Tumor cells were microdissected from 88 primary ALMs and 16 paired metastases and analyzed for KIT, NRAS and BRAF mutations. A subset of 25 ALMs was also evaluated for PTEN mutations. RESULTS BRAF mutations were identified in 17% of the primary ALMs. Both NRAS and KIT mutations were found at a similar frequency of 15%. Only one of the ALMs that were screened for PTEN harbored a mutation (4%). The KIT, NRAS and BRAF mutation status in paired primary and metastatic ALMs was identical. Patients with BRAF mutated tumors were significantly younger (57 years) than those with BRAF wild-type tumors (73 years, p=0.028). BRAF mutations were significantly more common in females (p=0.011) and more often found in tumors located on the feet (p=0.039). Anatomical site was an independent prognostic factor for overall survival; patients with ALMs on the hands or under fingernails had a better prognosis than those with tumors on the feet or under toenails (p=0.025). CONCLUSION Our results confirm the presence of KIT, NRAS and BRAF mutations in ALM and provide evidence that mutations in these genes occur at similar frequencies. Our results also show that PTEN is mutated in a small subset of ALM tumors.
Collapse
Affiliation(s)
- Abdlsattar Zebary
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yeh I, von Deimling A, Bastian BC. Clonal BRAF mutations in melanocytic nevi and initiating role of BRAF in melanocytic neoplasia. J Natl Cancer Inst 2013; 105:917-9. [PMID: 23690527 DOI: 10.1093/jnci/djt119] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BRAF(V600E) mutations are frequent in melanomas originating from intermittently sun-exposed skin and also in common acquired melanocytic nevi, suggesting that BRAF mutation is an early event in melanocytic neoplasia. All neoplastic melanocytes within such a nevus would be expected to carry the BRAF mutation, and thus we evaluated the frequency of cells with BRAF(V600E) mutations within acquired nevi by droplet digital polymerase chain reaction. In BRAF-mutant nevi the number of BRAF mutant alleles equaled the number of wild-type (WT) alleles in the neoplastic cell population, consistent with a fully clonal heterozygous BRAF mutation. The allelic ratio of BRAF(V600E) to BRAF(WT) in the eight VE1-positive nevi, adjusted for degree of stromal contamination, ranged from 0.84 to 1.12 with an average ratio of 1.01. This was confirmed by immunohistochemistry with an antibody specific for BRAF(V600E), which uniformly labeled the neoplastic cells without any evidence of heterogeneity. We found BRAF(V600E) mutations in the melanocytic nevi to be fully clonal, strongly suggesting that BRAF-activating mutations typically are early initiating events in melanocytic neoplasia.
Collapse
Affiliation(s)
- Iwei Yeh
- Department of Dermatology, University of California, San Francisco, USA.
| | | | | |
Collapse
|
31
|
Wilmott JS, Menzies AM, Haydu LE, Capper D, Preusser M, Zhang YE, Thompson JF, Kefford RF, von Deimling A, Scolyer RA, Long GV. BRAF(V600E) protein expression and outcome from BRAF inhibitor treatment in BRAF(V600E) metastatic melanoma. Br J Cancer 2013; 108:924-31. [PMID: 23403819 PMCID: PMC3590666 DOI: 10.1038/bjc.2013.29] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND To examine the association between level and patterns of baseline intra-tumoural BRAF(V600E) protein expression and clinical outcome of BRAF(V600E) melanoma patients treated with selective BRAF inhibitors. METHODS Fifty-eight BRAF(V600E) metastatic melanoma patients treated with dabrafenib or vemurafenib on clinical trials had pre-treatment tumour BRAF(V600E) protein expression immunohistochemically (IHC) assessed using the BRAF V600E mutant-specific antibody VE1. Sections were examined for staining intensity (score 1-3) and percentage of immunoreactive tumour cells, and from this an immunoreactive score (IRS) was derived (intensity × per cent positive/10). The presence of intra-tumoural heterogeneity for BRAF(V600E) protein expression was also assessed. BRAF(V600E) expression was correlated with RECIST response, time to best response (TTBR), progression-free survival (PFS) and overall survival (OS). RESULTS Expression was generally high (median IRS 28 (range 5-30)) and homogeneous (78%). Expression of mutated protein BRAF(V600E) as measured by intensity, per cent immunoreactive cells, or IRS did not correlate with RECIST response, TTBR, PFS or OS, including on multivariate analysis. Heterogeneity of staining was seen in 22% of cases and did not correlate with outcome. CONCLUSION In the current study population, IHC-measured pre-treatment BRAF(V600E) protein expression does not predict response or outcome to BRAF inhibitor therapy in BRAF(V600E) metastatic melanoma patients.
Collapse
Affiliation(s)
- J S Wilmott
- Melanoma Institute Australia, Sydney, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hiatt JB, Pritchard CC, Salipante SJ, O'Roak BJ, Shendure J. Single molecule molecular inversion probes for targeted, high-accuracy detection of low-frequency variation. Genome Res 2013; 23:843-54. [PMID: 23382536 PMCID: PMC3638140 DOI: 10.1101/gr.147686.112] [Citation(s) in RCA: 256] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The detection and quantification of genetic heterogeneity in populations of cells is fundamentally important to diverse fields, ranging from microbial evolution to human cancer genetics. However, despite the cost and throughput advances associated with massively parallel sequencing, it remains challenging to reliably detect mutations that are present at a low relative abundance in a given DNA sample. Here we describe smMIP, an assay that combines single molecule tagging with multiplex targeted capture to enable practical and highly sensitive detection of low-frequency or subclonal variation. To demonstrate the potential of the method, we simultaneously resequenced 33 clinically informative cancer genes in eight cell line and 45 clinical cancer samples. Single molecule tagging facilitated extremely accurate consensus calling, with an estimated per-base error rate of 8.4 × 10(-6) in cell lines and 2.6 × 10(-5) in clinical specimens. False-positive mutations in the single molecule consensus base-calls exhibited patterns predominantly consistent with DNA damage, including 8-oxo-guanine and spontaneous deamination of cytosine. Based on mixing experiments with cell line samples, sensitivity for mutations above 1% frequency was 83% with no false positives. At clinically informative sites, we identified seven low-frequency point mutations (0.2%-4.7%), including BRAF p.V600E (melanoma, 0.2% alternate allele frequency), KRAS p.G12V (lung, 0.6%), JAK2 p.V617F (melanoma, colon, two lung, 0.3%-1.4%), and NRAS p.Q61R (colon, 4.7%). We anticipate that smMIP will be broadly adoptable as a practical and effective method for accurately detecting low-frequency mutations in both research and clinical settings.
Collapse
Affiliation(s)
- Joseph B Hiatt
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | |
Collapse
|
33
|
Mehnert JM, Kluger HM. Driver mutations in melanoma: lessons learned from bench-to-bedside studies. Curr Oncol Rep 2013; 14:449-57. [PMID: 22723080 DOI: 10.1007/s11912-012-0249-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The identification of somatic driver mutations in human samples has allowed for the development of a molecular classification for melanoma. Recent breakthroughs in the treatment of metastatic melanoma have arisen as a result of these significant new insights into the molecular biology of the disease, particularly the development of inhibitors of activating BRAF(V600E) mutations. In this article the roles of several mutations known to be involved in the malignant transformation of melanocytes are reviewed including BRAF, PTEN, NRAS, ckit, and p16 as well as some of the emerging mutations in cutaneous and uveal melanoma. The bench to bedside collaborations that resulted in these discoveries are summarized, and potential therapeutic strategies to target driver mutations in specific patient subsets are discussed.
Collapse
Affiliation(s)
- Janice M Mehnert
- The Cancer Institute of New Jersey, 195 Little Albany Street Rm 5543, New Brunswick, NJ 08903, USA.
| | | |
Collapse
|
34
|
Immunohistochemistry Is Highly Sensitive and Specific for the Detection of V600E BRAF Mutation in Melanoma. Am J Surg Pathol 2013; 37:61-5. [DOI: 10.1097/pas.0b013e31826485c0] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Badalian-Very G, Vergilio JA, Fleming M, Rollins BJ. Pathogenesis of Langerhans cell histiocytosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:1-20. [PMID: 22906202 DOI: 10.1146/annurev-pathol-020712-163959] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Langerhans cell histiocytosis (LCH) combines in one nosological category a group of diseases that have widely disparate clinical manifestations but are all characterized by accumulation of proliferating cells with surface markers and ultrastructural features similar to cutaneous Langerhans cells (LCs). Despite this unified nosology, important questions about LCH remain unanswered. First, despite having phenotypic features of LCs, LCH cell gene-expression patterns differ from those in LCs. Although this observation suggests that LCH may arise from an earlier precursor, it is not necessarily inconsistent with the hypothesis that LCs are the cell of origin for LCH. Second, LCH's prominent inflammatory component and occasional benign clinical course suggest that LCH may not be a neoplasm. However, the demonstration that LCH cells are clonal, along with the recent discovery of activating BRAF mutations in LCH cells, strongly suggests that LCH is a neoplastic disease. These new observations point the way to rationally targeted therapies.
Collapse
Affiliation(s)
- Gayane Badalian-Very
- Department of Medical Oncology, Dana-Farber Cancer Institute, and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | |
Collapse
|
36
|
Colombino M, Capone M, Lissia A, Cossu A, Rubino C, De Giorgi V, Massi D, Fonsatti E, Staibano S, Nappi O, Pagani E, Casula M, Manca A, Sini M, Franco R, Botti G, Caracò C, Mozzillo N, Ascierto PA, Palmieri G. BRAF/NRAS Mutation Frequencies Among Primary Tumors and Metastases in Patients With Melanoma. J Clin Oncol 2012; 30:2522-9. [DOI: 10.1200/jco.2011.41.2452] [Citation(s) in RCA: 359] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Purpose The prevalence of BRAF, NRAS, and p16CDKN2A mutations during melanoma progression remains inconclusive. We investigated the prevalence and distribution of mutations in these genes in different melanoma tissues. Patients and Methods In all, 291 tumor tissues from 132 patients with melanoma were screened. Paired samples of primary melanomas (n = 102) and synchronous or asynchronous metastases from the same patients (n = 165) were included. Tissue samples underwent mutation analysis (automated DNA sequencing). Secondary lesions included lymph nodes (n = 84), and skin (n = 36), visceral (n = 25), and brain (n = 44) sites. Results BRAF/NRAS mutations were identified in 58% of primary melanomas (43% BRAF; 15% NRAS); 62% in lymph nodes, 61% subcutaneous, 56% visceral, and 70% in brain sites. Mutations were observed in 63% of metastases (48% BRAF; 15% NRAS), a nonsignificant increase in mutation frequency after progression from primary melanoma. Of the paired samples, lymph nodes (93% consistency) and visceral metastases (96% consistency) presented a highly similar distribution of BRAF/NRAS mutations versus primary melanomas, with a significantly less consistent pattern in brain (80%) and skin metastases (75%). This suggests that independent subclones are generated in some patients. p16CDKN2A mutations were identified in 7% and 14% of primary melanomas and metastases, with a low consistency (31%) between secondary and primary tumor samples. Conclusion In the era of targeted therapies, assessment of the spectrum and distribution of alterations in molecular targets among patients with melanoma is needed. Our findings about the prevalence of BRAF/NRAS/p16CDKN2A mutations in paired tumor lesions from patients with melanoma may be useful in the management of this disease.
Collapse
Affiliation(s)
- Maria Colombino
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Mariaelena Capone
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Amelia Lissia
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Antonio Cossu
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Corrado Rubino
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Vincenzo De Giorgi
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Daniela Massi
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Ester Fonsatti
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Stefania Staibano
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Oscar Nappi
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Elena Pagani
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Milena Casula
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Antonella Manca
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - MariaCristina Sini
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Renato Franco
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Gerardo Botti
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Corrado Caracò
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Nicola Mozzillo
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Paolo A. Ascierto
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Giuseppe Palmieri
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| |
Collapse
|
37
|
Tran SL, Haferkamp S, Scurr LL, Gowrishankar K, Becker TM, Desilva C, Thompson JF, Scolyer RA, Kefford RF, Rizos H. Absence of distinguishing senescence traits in human melanocytic nevi. J Invest Dermatol 2012; 132:2226-34. [PMID: 22513787 DOI: 10.1038/jid.2012.126] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cellular senescence permanently restricts the replicative capacity of cells in response to various stress signals, including aberrant activation of oncogenes. The presence of predictive senescence markers in human premalignant lesions suggests that senescence may function as a genuine tumor suppressor. These markers are not exclusive to the senescence program, however, and it is possible that their expression in vivo does not discriminate irreversible from reversible forms of proliferative arrest. In this study, we aimed to clarify whether human nevus cells can be distinguished from primary and transformed melanocytes by examining the expression of eight senescence markers, including those previously purported to define nevi as senescent tumors. Specifically, we analyzed effectors of senescence, including p16(INK4a), p53, and DNA damage (γ-H2AX), as well as predictive markers of senescence including Ki67, PML, senescence-associated β-galactosidase, heterochromatic foci (H3K9Me, 4'-6-diamidino-2-phenylindole), and nuclear size. We found that these commonly accepted senescence markers do not in fact distinguish nevi from precursor/normal and transformed/malignant melanocytes. We conclude that on the basis of current evidence it cannot be reasonably inferred that nevi are permanently growth arrested via senescence.
Collapse
Affiliation(s)
- Sieu L Tran
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sakaizawa K, Goto Y, Kiniwa Y, Uchiyama A, Harada K, Shimada S, Saida T, Ferrone S, Takata M, Uhara H, Okuyama R. Mutation analysis of BRAF and KIT in circulating melanoma cells at the single cell level. Br J Cancer 2012; 106:939-46. [PMID: 22281663 PMCID: PMC3305957 DOI: 10.1038/bjc.2012.12] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: The availability of molecular-targeted therapies for the treatment of melanoma has emphasised the need to identify mutations in target genes such as BRAF and KIT. Circulating tumour cells (CTC) are present in the peripheral blood of a significant proportion of cancer patients. Methods: High molecular weight melanoma-associated antigen (HMW-MAA) was used to isolate melanoma cells from peripheral blood as it is selectively expressed at high levels on melanomas. The HMW-MAA-positive cells were isolated using immunomagnetic beads. After removing CD45+ cells, CTC were identified by staining with MART-1- and gp100-specific antibodies (HMW-MAA+, CD45−, MART-1/gp100+). Single, isolated CTC were then subjected to BRAF and KIT mutational analysis. Results: CTC (HMW-MAA+, CD45−, MART-1/gp100+) were isolated from the blood of 11 patients and BRAF and KIT were sequenced in nine and four patients, respectively. The BRAF sequences identified in the CTC were inconsistent with those identified in autologous melanoma tumours in three patients and the KIT sequences were inconsistent in three patients. In addition, polyclonal BRAF mutations were identified in one patient and concomitant mutations in BRAF and KIT were identified in another patient. Conclusion: Melanoma cells show clonal heterogeneity. Therefore, CTC genotyping may be crucial for successful molecular-targeted therapy.
Collapse
Affiliation(s)
- K Sakaizawa
- Department of Dermatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Intra- and inter-tumor heterogeneity of BRAF(V600E))mutations in primary and metastatic melanoma. PLoS One 2012; 7:e29336. [PMID: 22235286 PMCID: PMC3250426 DOI: 10.1371/journal.pone.0029336] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/25/2011] [Indexed: 12/22/2022] Open
Abstract
The rationale for using small molecule inhibitors of oncogenic proteins as cancer therapies depends, at least in part, on the assumption that metastatic tumors are primarily clonal with respect to mutant oncogene. With the emergence of BRAF(V600E) as a therapeutic target, we investigated intra- and inter-tumor heterogeneity in melanoma using detection of the BRAF(V600E) mutation as a marker of clonality. BRAF mutant-specific PCR (MS-PCR) and conventional sequencing were performed on 112 tumors from 73 patients, including patients with matched primary and metastatic specimens (n = 18). Nineteen patients had tissues available from multiple metastatic sites. Mutations were detected in 36/112 (32%) melanomas using conventional sequencing, and 85/112 (76%) using MS-PCR. The better sensitivity of the MS-PCR to detect the mutant BRAF(V600E) allele was not due to the presence of contaminating normal tissue, suggesting that the tumor was comprised of subclones of differing BRAF genotypes. To determine if tumor subclones were present in individual primary melanomas, we performed laser microdissection and mutation detection via sequencing and BRAF(V600E)-specific SNaPshot analysis in 9 cases. Six of these cases demonstrated differing proportions of BRAF(V600E)and BRAF(wild-type) cells in distinct microdissected regions within individual tumors. Additional analyses of multiple metastatic samples from individual patients using the highly sensitive MS-PCR without microdissection revealed that 5/19 (26%) patients had metastases that were discordant for the BRAF(V600E) mutation. In conclusion, we used highly sensitive BRAF mutation detection methods and observed substantial evidence for heterogeneity of the BRAF(V600E) mutation within individual melanoma tumor specimens, and among multiple specimens from individual patients. Given the varied clinical responses of patients to BRAF inhibitor therapy, these data suggest that additional studies to determine possible associations between clinical outcomes and intra- and inter-tumor heterogeneity could prove fruitful.
Collapse
|
40
|
Abstract
The widely accepted paradigm for tumorigenesis begins with rate-limiting mutations in a key growth control gene resulting in immediate lesion growth. Tumor progression occurs as cells within the tumor acquire additional carcinogenic mutations. However, there is clear evidence that the road to cancer can begin long before the growth of a clinically detectable lesion - indeed, long before any of the usual morphological correlates of preneoplasia are recognizable. Field cancerization, the replacement of the normal cell population by a histologically nondysplastic but protumorigenic mutant cell clone, underlies the development of many cancer types, and in this article we review field cancerization in the GI tract. We present the evidence that field cancerization can underpin tumorigenesis in all gastrointestinal compartments, discuss the homeostatic mechanisms that could permit clone spread and highlight how an understanding of the mechanisms driving field cancerization is a means to study human stem cell biology. Finally, we discuss how appropriate recognition of the role of field cancerization in tumorigenesis could impact patient care.
Collapse
Affiliation(s)
- Trevor A Graham
- Histopathology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK.
| | | | | |
Collapse
|
41
|
|
42
|
Badalian-Very G, Vergilio JA, Degar BA, Rodriguez-Galindo C, Rollins BJ. Recent advances in the understanding of Langerhans cell histiocytosis. Br J Haematol 2011; 156:163-72. [PMID: 22017623 DOI: 10.1111/j.1365-2141.2011.08915.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Langerhans cell histiocytosis (LCH) is a proliferative disease of cells that share phenotypic characteristics with the primary antigen presenting cells of the epidermis. Its clinical manifestations are highly variable, extending from very benign forms to a disseminated, aggressive disease that causes significant mortality. Although many of the fundamental pathogenetic features of LCH have been enigmatic, recent advances have led to a much clearer understanding of the disease. In particular, careful molecular analyses of mouse models and human LCH samples suggest that LCH's cell of origin may not be the epidermal LC itself but a myeloid-derived precursor. Advanced genomic technologies have revealed the presence of activating, somatic BRAF mutations in the majority of patient specimens. Together, these observations have produced a new picture of LCH as a myeloid neoplasm. These advances are likely to have profound implications for the use of targeted therapeutics in LCH.
Collapse
Affiliation(s)
- Gayane Badalian-Very
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
43
|
Swoboda A, Rasin-Streden D, Schanab O, Okamoto I, Pehamberger H, Petzelbauer P, Mikula M. Identification of genetic disparity between primary and metastatic melanoma in human patients. Genes Chromosomes Cancer 2011; 50:680-8. [PMID: 21584902 DOI: 10.1002/gcc.20890] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 04/14/2011] [Accepted: 04/15/2011] [Indexed: 06/01/2025] Open
Abstract
It is commonly accepted that cancer cell progression is accompanied by accumulation of genetic changes. Here we searched for copy number variations in melanoma and asked whether homozygous losses always cumulate during tumor cell progression. Therefore we investigated either melanoma cell lines or tissue derived from the primary lesion and from the lymph node metastasis of the same individual patient. In vitro studies of melanoma cell lines revealed high migratory and anchorage independent growth of metastasis-derived cells. Surprisingly, whole genome DNA analysis of a primum-derived cell line revealed a total of 10 homozygous losses, whereas the matched metastasis-derived cell line only shared five of those losses. We further tested these cells in a mouse model for intradermal melanoma growth and detected fast growth of the metastasis-derived cell line and no growth of primum-derived cells. Additionally, we screened matched pairs of patient-derived melanoma primum and metastasis samples and we could also identify a case with homozygous deletions exclusively present in the primary lesion. Therefore, we suggest that tumor cell progression at the metastatic niche can occur parallel and independently from the primary tumor. We propose that for mutation-targeted therapy genotyping should be performed not only from primary, but also from metastatic melanoma.
Collapse
MESH Headings
- Adult
- Animals
- Base Sequence
- Cell Line, Tumor
- Cell Proliferation
- Cell Shape
- Chromosome Deletion
- Chromosomes, Human, Pair 1/genetics
- Chromosomes, Human, Pair 10/genetics
- Chromosomes, Human, Pair 13/genetics
- Chromosomes, Human, Pair 4/genetics
- DNA Copy Number Variations
- Female
- Genome-Wide Association Study
- Humans
- Lymphatic Metastasis
- Male
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/pathology
- Mice
- Mice, SCID
- Neoplasm Invasiveness
- Neoplasm Transplantation
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- X Chromosome/genetics
Collapse
Affiliation(s)
- Alexander Swoboda
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Währingergürtel 18-20,Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
44
|
Gaisa NT, Graham TA, McDonald SA, Poulsom R, Heidenreich A, Jakse G, Knuechel R, Wright NA. Clonal architecture of human prostatic epithelium in benign and malignant conditions. J Pathol 2011; 225:172-80. [PMID: 21898875 DOI: 10.1002/path.2959] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 06/24/2011] [Accepted: 06/27/2011] [Indexed: 12/25/2022]
Abstract
The location of stem cells in the epithelium of the prostatic acinus remains uncertain, as does the cellular origin of prostatic neoplasia. Here, we apply lineage tracing to visualize the clonal progeny of stem cells in benign and malignant human prostates and understand the clonal architecture of this epithelium. Cells deficient for the mitochondrially-encoded enzyme cytochrome c oxidase (CCO) were identified in 27 frozen prostatectomy specimens using dual colour enzyme histochemistry and individual CCO-normal and -deficient cell areas were laser-capture microdissected. PCR-sequencing of the entire mitochondrial genome (mtDNA) of cells from CCO-deficient areas found to share mtDNA mutations not present in adjacent CCO-normal cells, thus proving a clonal origin. Immunohistochemistry was performed to visualize the three cell lineages normally present in the prostatic epithelium. Entire CCO-deficient acini, and part-deficient acini were found. Deficient patches spanned either basal or luminal cells, but sometimes also both epithelial cell types in normal, hyperplastic or atrophic epithelium, and prostatic intraepithelial neoplasia (PIN). Patches comprising both PIN and invasive cancer were observed. Each cell area within a CCO-deficient patch contained an identical mtDNA mutation, defining the patch as a clonal unit. CCO-deficient patches in benign epithelium contained basal, luminal and endocrine cells, demonstrating multilineage differentiation and therefore the presence of a stem cell. Our results demonstrate that the normal, atrophic, hypertrophic and atypical (PIN) epithelium of human prostate contains stem cell-derived clonal units that actively replenish the epithelium during ageing. These deficient areas usually included the basal compartment indicating the basal layer as the location of the stem cell. Importantly, single clonal units comprised both PIN and invasive cancer, supporting PIN as the pre-invasive lesion for prostate cancer.
Collapse
Affiliation(s)
- Nadine T Gaisa
- Institute of Pathology, RWTH, Aachen University, Aachen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Champion KJ, Bunag C, Estep AL, Jones JR, Bolt CH, Rogers RC, Rauen KA, Everman DB. Germline mutation in BRAF codon 600 is compatible with human development: de novo p.V600G mutation identified in a patient with CFC syndrome. Clin Genet 2011; 79:468-74. [PMID: 20735442 DOI: 10.1111/j.1399-0004.2010.01495.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BRAF, the protein product of BRAF, is a serine/threonine protein kinase and one of the direct downstream effectors of Ras. Somatic mutations in BRAF occur in numerous human cancers, whereas germline BRAF mutations cause cardio-facio-cutaneous (CFC) syndrome. One recurrent somatic mutation, p.V600E, is frequently found in several tumor types, such as melanoma, papillary thyroid carcinoma, colon cancer, and ovarian cancer. However, a germline mutation affecting codon 600 has never been described. Here, we present a patient with CFC syndrome and a de novo germline mutation involving codon 600 of BRAF, thus providing the first evidence that a pathogenic germline mutation involving this critical codon is not only compatible with development but can also cause the CFC phenotype. In vitro functional analysis shows that this mutation, which replaces a valine with a glycine at codon 600 (p.V600G), leads to increased ERK and ELK phosphorylation compared to wild-type BRAF but is less strongly activating than the cancer-associated p.V600E mutation.
Collapse
Affiliation(s)
- K J Champion
- Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Despite recent advances, the biology underlying nevogenesis remains unclear. Activating mutations in NRAS, HRAS, BRAF, and GNAQ have been identified in benign nevi. Their presence roughly correlates with congenital, Spitz, acquired, and blue nevi, respectively. These mutations are likely to play a critical role in driving nevogenesis. While each mutation is able to activate the MAP kinase pathway, they also interact with a host of different proteins in other pathways. The different melanocytic developmental pathways activated by each mutation cause the cells to migrate, proliferate, and differentiate to different extents within the skin. This causes each mutation to give rise to a characteristic growth pattern. The exact location and differentiation state of the cell of origin for benign moles remains to be discovered. Further research is necessary to fully understand nevus development given that most of the same developmental pathways are also present in melanoma.
Collapse
|
47
|
Puzanov I, Burnett P, Flaherty KT. Biological challenges of BRAF inhibitor therapy. Mol Oncol 2011; 5:116-23. [PMID: 21393075 DOI: 10.1016/j.molonc.2011.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 01/27/2011] [Indexed: 12/19/2022] Open
Abstract
Activating mutations in BRAF, a constituent of the map kinase pathway, were first discovered as being most prevalent in melanoma in 2002. Only recently have potent and selective, orally available inhibitors of BRAF emerged for clinical testing and demonstrated clear evidence of tumor regression in the majority of patients whose tumors harbor a BRAF mutation. While these early observations suggest that the BRAF targeted therapy will become part of the standard treatment paradigm for patients with advanced melanoma, it is also clear that a majority of these responses are incomplete and temporary. Therefore, the focus of the melanoma field has shifted to understanding the limits of the first generation of selective BRAF inhibitors with regard to safety and efficacy, the context of somatic genetic changes that accompany BRAF, and the combination regimens that target distinct elements of melanoma pathophysiology.
Collapse
Affiliation(s)
- Igor Puzanov
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
48
|
Zalaudek I, Guelly C, Pellacani G, Hofmann-Wellenhof R, Trajanoski S, Kittler H, Scope A, Marghoob AA, Longo C, Leinweber B, Ferrara G, Saida T, Grichnik JM, Argenziano G, Becker JC. The Dermoscopical and Histopathological Patterns of Nevi Correlate with the Frequency of BRAF Mutations. J Invest Dermatol 2011; 131:542-5. [DOI: 10.1038/jid.2010.332] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
49
|
Zipser MC, Eichhoff OM, Widmer DS, Schlegel NC, Schoenewolf NL, Stuart D, Liu W, Gardner H, Smith PD, Nuciforo P, Dummer R, Hoek KS. A proliferative melanoma cell phenotype is responsive to RAF/MEK inhibition independent of BRAF mutation status. Pigment Cell Melanoma Res 2011; 24:326-33. [DOI: 10.1111/j.1755-148x.2010.00823.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Polyclonality of BRAF mutations in primary melanoma and the selection of mutant alleles during progression. Br J Cancer 2011; 104:464-8. [PMID: 21224857 PMCID: PMC3049568 DOI: 10.1038/sj.bjc.6606072] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Oncogenic BRAF mutation had been considered to be a founder event in the formation of melanocytic tumours; however, we recently argued against this notion by showing marked polyclonality of BRAF mutations in acquired melanocytic nevi (Lin et al, J Natl Cancer Inst., 2009; 101:1423-7). Here, we tested whether similar heterogeneity of BRAF mutations exists in primary melanomas. METHODS We isolated and sequenced single melanoma cells from five primary melanoma tissues using antibodies against human high-molecular-weight melanoma-associated antigen. We also examined 10 primary melanomas by the sensitive Mutector assay detecting the BRAF(V600E) mutation, as well as by cloning and sequencing of separated alleles. Furthermore, we estimated the frequency of BRAF mutant alleles in paired samples of primary tumour and recurrence or metastasis in three patients. RESULTS Single-cell mutation analyses revealed that four of five primary melanomas contained both BRAF-wild-type and BRAF-mutant tumour cells. Tumour heterogeneity in terms of BRAF mutations was also shown in 8 of 10 primary melanomas. Selection of BRAF mutant alleles during progression was demonstrated in all the three patients. CONCLUSION Acquisition of a BRAF mutation is not a founder event, but may be one of the multiple clonal events in melanoma development, which is selected for during the progression.
Collapse
|