1
|
Sara GB, Noa FR, Enrique PR, Miriam CG, Joaquín FR, Ginesa GR. TERT Amplification a Risk Stratification Marker in Papillary Thyroid Carcinoma, Significantly Correlated with Tumor Recurrence and Survival. Endocr Pathol 2025; 36:15. [PMID: 40272676 PMCID: PMC12021720 DOI: 10.1007/s12022-025-09853-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/27/2025]
Abstract
Few studies have analyzed the prevalence of TERT amplification in thyroid cancer, showing discrepancies in various topics. The impact on tumor recurrence and patient survival in papillary thyroid carcinoma (PTC) remains unknown. Thirteen cancer cell lines and 215 tumor samples from 91 patients, who underwent surgery for PTC (41), poorly differentiated thyroid carcinoma (PDC = 15), or anaplastic thyroid carcinoma (ATC = 35), were analyzed. Clonality, spread with tumor dedifferentiation or metastatic PTC cells, and coexistence with TERTp, BRAF, RAS, and PIK3CA mutations were also investigated. TERT amplification was found in 17%, 20%, and 17% of the PTC, PDC, and ATC, respectively. It was more frequent in follicular variant PTC and PTC with distant metastases (86%, P = 0.0448). The cell lines HTh74, SW1736, and T242 had amplification. In PTC, TERT amplification was a subclonal event. The increase in TERT copy number spread in all cases with metastatic PTC cells. In 67% of the PDC and 100% of the ATC, TERT activation segregated with tumor dedifferentiation. TERT amplification correlated with TERTp mutations in PTC (P = 0.0313) and PIK3CA mutations in ATC (P = 0.0272). TERT amplification significantly correlated with vascular invasion (P = 0.03637), distant metastases at diagnosis and/or follow-up (P = 0.04482), metachronous distant metastases (P = 0.03131), death patient status (P = 0.000829), stage at diagnosis (P = 0.01995), and stage III/IV at last follow-up (P = 0.01552). TERT amplification associated independently with tumor-related recurrence and death. Our study shows that PTC can be stratified into clinically prognostic relevant categories based on the presence or not of TERT amplification in the cells.
Collapse
Affiliation(s)
- Gil-Bernabé Sara
- Institute of Biomedicine and Molecular Genetics (IBGM), Valladolid University, Valladolid, Spain
- School of Medicine, Valladolid University, Valladolid, Spain
| | - Feás-Rodríguez Noa
- Institute of Biomedicine and Molecular Genetics (IBGM), Valladolid University, Valladolid, Spain
| | - Pérez-Riesgo Enrique
- Institute of Biomedicine and Molecular Genetics (IBGM), Valladolid University, Valladolid, Spain
- European Miguel de Cervantes University, Valladolid, Spain
| | - Corraliza-Gómez Miriam
- Institute of Biomedicine and Molecular Genetics (IBGM), Valladolid University, Valladolid, Spain
- University of Cádiz, Cádiz, Spain
| | | | - García-Rostán Ginesa
- Institute of Biomedicine and Molecular Genetics (IBGM), Valladolid University, Valladolid, Spain.
- School of Medicine, Valladolid University, Valladolid, Spain.
| |
Collapse
|
2
|
Sadr Z, Ghasemi M, Jafarpour S, Seyfi R, Ghasemi A, Boustanipour E, Khorshid HRK, Ehtesham N. Beginning at the ends: telomere and telomere-based cancer therapeutics. Mol Genet Genomics 2024; 300:1. [PMID: 39638969 DOI: 10.1007/s00438-024-02206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Telomeres, which are situated at the terminal ends of chromosomes, undergo a reduction in length with each cellular division, ultimately reaching a critical threshold that triggers cellular senescence. Cancer cells circumvent this senescence by utilizing telomere maintenance mechanisms (TMMs) that grant them a form of immortality. These mechanisms can be categorized into two primary processes: the reactivation of telomerase reverse transcriptase and the alternative lengthening of telomeres (ALT) pathway, which is dependent on homologous recombination (HR). Various strategies have been developed to inhibit telomerase activation in 85-95% of cancers, including the use of antisense oligonucleotides such as small interfering RNAs and endogenous microRNAs, agents that simulate telomere uncapping, expression modulators, immunotherapeutic vaccines targeting telomerase, reverse transcriptase inhibitors, stabilization of G-quadruplex structures, and gene therapy approaches. Conversely, in the remaining 5-15% of human cancers that rely on ALT, mechanisms involve modifications in the chromatin environment surrounding telomeres, upregulation of TERRA long non-coding RNA, enhanced activation of the ataxia telangiectasia and Rad-3-related protein kinase signaling pathway, increased interactions with nuclear receptors, telomere repositioning driven by HR, and recombination events between non-sister chromatids, all of which present potential targets for therapeutic intervention. Additionally, combinatorial therapy has emerged as a strategy that employs selective agents to simultaneously target both telomerase and ALT, aiming for optimal clinical outcomes. Given the critical role of anti-TMM strategies in cancer treatment, this review provides an overview of the latest insights into the structure and function of telomeres, their involvement in tumorigenesis, and the advancements in TMM-based cancer therapies.
Collapse
Affiliation(s)
- Zahra Sadr
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoumeh Ghasemi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Soheyla Jafarpour
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Seyfi
- Department of Stem Cells Technology and Tissue Regeneration, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Aida Ghasemi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Boustanipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Naeim Ehtesham
- Department of Medical Genetics, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| |
Collapse
|
3
|
Barrutia L, Schuuring E, Rácz E, Diercks GFH, van Kempen LC. Utility and Micro-Costing Framework for TERT Promoter Mutation Analysis in Melanocytic Lesions of Uncertain Malignant Potential: A Retrospective Study in Dutch Local Clinical Practice. Diagnostics (Basel) 2024; 14:1665. [PMID: 39125541 PMCID: PMC11311256 DOI: 10.3390/diagnostics14151665] [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: 04/08/2024] [Revised: 06/24/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
The 2018 WHO edition on the classification of cutaneous melanocytic tumors recognizes eight evolutionary pathways of melanoma and describes tumors of uncertain malignant potential for each. When histology and immunohistochemistry do not support a confident conclusion about its malignant potential, a window of diagnostic uncertainty is created. Mutations in the telomerase reverse transcriptase gene promoter (TERTp) are highly specific for melanoma and can be used as an ancillary technique to acquire a higher level of confidence in the diagnosis. However, little is known about the cost-effectiveness of testing for TERTp mutations. The aims of this study were to determine how often knowledge of the TERTp mutation status contributed to the final diagnosis and to develop a micro-costing framework to calculate cost-effectiveness. A retrospective analysis of all cutaneous melanocytic lesions that were discussed in the Noord-Nederland Melanoma Panel from January 2021 to October 2022 was performed to identify the cases in which the preliminary histopathological diagnosis was uncertain regarding malignancy (ambiguous, likely benign, or likely malignant). For cases in which a TERTp mutation analysis was performed, the final diagnoses were collected, and it was determined whether this impacted the overall conclusion. A micro-costing framework was established to model the financial impact of introducing TERTp mutation analyses and subsequent clinical procedures. The study included 367 cases, of which 175 diagnoses of uncertain malignant potential were initially reported. TERTp mutation analysis was performed for 151/175 (86%). In 38% of these cases, a higher level of confidence regarding malignant potential was obtained. The implementation of TERTp mutation analyses for cutaneous melanocytic proliferations with uncertain malignant potential can narrow the window of diagnostic uncertainty. For the patient group with an initial uncertain diagnosis, the increased cost for molecular testing (86.145 €) was compensated by a reduced overall treatment cost (-122.304 €). A microsimulation model to determine the cost-effectiveness of TERTp mutation analysis projected an overall saving for the healthcare system.
Collapse
Affiliation(s)
- Leire Barrutia
- Department of Pathology and Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Dermatology, Medicine and Toxicology, University of Valladolid, 47002 Valladolid, Spain
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Emõke Rácz
- Department of Dermatology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Gilles F. H. Diercks
- Department of Pathology and Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Léon C. van Kempen
- Department of Pathology and Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Pathology, University of Antwerp, Antwerp University Hospital, 2650 Edegem, Belgium
| |
Collapse
|
4
|
Hsieh MY, Hsu SK, Liu TY, Wu CY, Chiu CC. Melanoma biology and treatment: a review of novel regulated cell death-based approaches. Cancer Cell Int 2024; 24:63. [PMID: 38336727 PMCID: PMC10858604 DOI: 10.1186/s12935-024-03220-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024] Open
Abstract
The incidence of melanoma, the most lethal form of skin cancer, has increased due to ultraviolet exposure. The treatment of advanced melanoma, particularly metastatic cases, remains challenging with poor outcomes. Targeted therapies involving BRAF/MEK inhibitors and immunotherapy based on anti-PD1/anti-CTLA4 antibodies have achieved long-term survival rates of approximately 50% for patients with advanced melanoma. However, therapy resistance and inadequate treatment response continue to hinder further breakthroughs in treatments that increase survival rates. This review provides an introduction to the molecular-level pathogenesis of melanoma and offers an overview of current treatment options and their limitations. Cells can die by either accidental or regulated cell death (RCD). RCD is an orderly cell death controlled by a variety of macromolecules to maintain the stability of the internal environment. Since the uncontrolled proliferation of tumor cells requires evasion of RCD programs, inducing the RCD of melanoma cells may be a treatment strategy. This review summarizes studies on various types of nonapoptotic RCDs, such as autophagy-dependent cell death, necroptosis, ferroptosis, pyroptosis, and the recently discovered cuproptosis, in the context of melanoma. The relationships between these RCDs and melanoma are examined, and the interplay between these RCDs and immunotherapy or targeted therapy in patients with melanoma is discussed. Given the findings demonstrating melanoma cell death in response to different stimuli associated with these RCDs, the induction of RCD shows promise as an integral component of treatment strategies for melanoma.
Collapse
Affiliation(s)
- Ming-Yun Hsieh
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Tzu-Yu Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
5
|
Huber R, Lee J, Borretta L, Tessier-Cloutier B, Lum A, Yip S, Horst BA. TERT promoter mutations in atypical melanocytic lesions: A series of seven cases with adverse melanoma-specific outcome. Hum Pathol 2024; 144:34-39. [PMID: 38224873 DOI: 10.1016/j.humpath.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
The majority of melanocytic proliferations can be readily categorized as benign or malignant based on histologic assessment under the microscope by a trained dermatopathologist. However, a subset of lesions, termed Atypical Melanocytic Proliferations (AMPs), are histologically ambiguous, leading to possible diagnostic error and suboptimal treatment. Mutations in the promoter region of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), are commonly found in melanomas but are rare in melanocytic nevi. In this study, we aimed to determine the prevalence of hot spot TERT promoter (TERT-p) mutations in AMPs with adverse melanoma-specific outcome. Studies were approved by respective institutional review boards. Using a multi-center database, we identified seven cases of melanocytic proliferations with a clinical follow-up period of at least 4 years, which were initially diagnosed as AMPs, and which recurred either as melanoma at site of prior biopsy or as metastatic melanoma. Sequencing of the TERT-p region showed hotspot mutations in three cases (43 %), suggesting that TERT-p mutations are enriched and could aid in the identification of AMPs with adverse outcome. In comparison with existing ancillary techniques for prognostication of AMPs, TERT-p mutation analysis may have advantages in terms of cost effectiveness and turnaround time, and is a promising diagnostic parameter with potential widespread utility.
Collapse
Affiliation(s)
- Reed Huber
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada; Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada
| | - Jonathan Lee
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada; Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada
| | - Lisa Borretta
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | | | - Amy Lum
- Molecular Oncology, BC Cancer Agency, Vancouver, BC, V5Z 1L3, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada; Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada
| | - Basil A Horst
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada; Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
6
|
Caraban BM, Aschie M, Deacu M, Cozaru GC, Pundiche MB, Orasanu CI, Voda RI. A Narrative Review of Current Knowledge on Cutaneous Melanoma. Clin Pract 2024; 14:214-241. [PMID: 38391404 PMCID: PMC10888040 DOI: 10.3390/clinpract14010018] [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: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Cutaneous melanoma is a public health problem. Efforts to reduce its incidence have failed, as it continues to increase. In recent years, many risk factors have been identified. Numerous diagnostic systems exist that greatly assist in early clinical diagnosis. The histopathological aspect illustrates the grim nature of these cancers. Currently, pathogenic pathways and the tumor microclimate are key to the development of therapeutic methods. Revolutionary therapies like targeted therapy and immune checkpoint inhibitors are starting to replace traditional therapeutic methods. Targeted therapy aims at a specific molecule in the pathogenic chain to block it, stopping cell growth and dissemination. The main function of immune checkpoint inhibitors is to boost cellular immunity in order to combat cancer cells. Unfortunately, these therapies have different rates of effectiveness and side effects, and cannot be applied to all patients. These shortcomings are the basis of increased incidence and mortality rates. This study covers all stages of the evolutionary sequence of melanoma. With all these data in front of us, we see the need for new research efforts directed at therapies that will bring greater benefits in terms of patient survival and prognosis, with fewer adverse effects.
Collapse
Affiliation(s)
- Bogdan Marian Caraban
- Clinical Department of Plastic Surgery, Microsurgery-Reconstructive, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Mariana Aschie
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Academy of Medical Sciences of Romania, 030171 Bucharest, Romania
- The Romanian Academy of Scientists, 030167 Bucharest, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| | - Mariana Deacu
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
- Clinical Service of Pathology, Departments of Genetics, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Mihaela Butcaru Pundiche
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Department of General Surgery, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
| | - Cristian Ionut Orasanu
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| | - Raluca Ioana Voda
- Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
- Clinical Service of Pathology, Departments of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), "Ovidius" University of Constanta, 900591 Constanta, Romania
| |
Collapse
|
7
|
El Zarif T, Machaalani M, Nawfal R, Nassar AH, Xie W, Choueiri TK, Pomerantz M. TERT Promoter Mutations Frequency Across Race, Sex, and Cancer Type. Oncologist 2024; 29:8-14. [PMID: 37462445 PMCID: PMC10769781 DOI: 10.1093/oncolo/oyad208] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/23/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Telomerase reverse transcriptase (TERT) gene promoter mutations have been explored, as biomarkers of improved survival for patients with cancer receiving immune checkpoint inhibitors. We sought to investigate their prevalence by race and sex across different cancer types to inform patient selection in clinical trials. RESULTS In this observational study, 31 925 patients with cancer underwent next-generation sequencing of their tumors with 88% (27 970) patients self-reported being Whites, 7.1% (2273) Asians, and 5.3% (1682) Blacks. Examining the distribution of TERT promoter mutations by race, White patients with melanoma harbored more TERT promoter mutations than Asian and Black patients (OR = 25.83; 95%CI, 6.84-217.42; P < .001). In contrast, Asian patients with head and neck cancer (HNC) harbored more TERT promoter mutations compared to White patients (OR = 2.47; 95%CI, 1.39-4.37; P = .004). In addition, the distribution of TERT promoter mutations differed by sex. Males were enriched for TERT gene promoter mutations compared to females with melanoma (OR = 1.82; 95%CI, 1.53-2.16; P < .001), cancer of unknown primary (OR = 1.96; 95%CI, 1.43-2.69; P < .001), hepatobiliary (OR = 3.89; 95%CI, 2.65-5.69; P < .001), and thyroid cancers (OR = 1.42; 95%CI, 1.10-1.84; P = .0087), while females were more enriched for TERT promoter mutations compared to males for HNC (OR = 0.56; 95%CI, 0.39-0.81; P = .0021). CONCLUSIONS The prevalence of TERT gene promoter mutations varies among patients with cancer based on race and sex. These findings inform our understanding of cancer biology and can assist in the design of future clinical trials that leverage drugs targeting TERT promoter dependencies.
Collapse
Affiliation(s)
- Talal El Zarif
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Marc Machaalani
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Rashad Nawfal
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Amin H Nassar
- Department of Hematology/Oncology, Yale New Haven Hospital, New Haven, CT, USA
| | - Wanling Xie
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mark Pomerantz
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Waseh S, Lee JB. Advances in melanoma: epidemiology, diagnosis, and prognosis. Front Med (Lausanne) 2023; 10:1268479. [PMID: 38076247 PMCID: PMC10703395 DOI: 10.3389/fmed.2023.1268479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/13/2023] [Indexed: 06/30/2024] Open
Abstract
Unraveling the multidimensional complexities of melanoma has required concerted efforts by dedicated community of researchers and clinicians battling against this deadly form of skin cancer. Remarkable advances have been made in the realm of epidemiology, classification, diagnosis, and therapy of melanoma. The treatment of advanced melanomas has entered the golden era as targeted personalized therapies have emerged that have significantly altered the mortality rate. A paradigm shift in the approach to melanoma classification, diagnosis, prognosis, and staging is underway, fueled by discoveries of genetic alterations in melanocytic neoplasms. A morphologic clinicopathologic classification of melanoma is expected to be replaced by a more precise molecular based one. As validated, convenient, and cost-effective molecular-based tests emerge, molecular diagnostics will play a greater role in the clinical and histologic diagnosis of melanoma. Artificial intelligence augmented clinical and histologic diagnosis of melanoma is expected to make the process more streamlined and efficient. A more accurate model of prognosis and staging of melanoma is emerging based on molecular understanding melanoma. This contribution summarizes the recent advances in melanoma epidemiology, classification, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Shayan Waseh
- Department of Dermatology, Temple University Hospital, Philadelphia, PA, United States
| | - Jason B. Lee
- Department of Dermatology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
9
|
Liebmann A, Admard J, Armeanu-Ebinger S, Wild H, Abele M, Gschwind A, Seibel-Kelemen O, Seitz C, Bonzheim I, Riess O, Demidov G, Sturm M, Schadeck M, Pogoda M, Bien E, Krawczyk M, Jüttner E, Mentzel T, Cesen M, Pfaff E, Kunc M, Forchhammer S, Forschner A, Leiter-Stöppke U, Eigentler TK, Schneider DT, Schroeder C, Ossowski S, Brecht IB. UV-radiation and MC1R germline mutations are risk factors for the development of conventional and spitzoid melanomas in children and adolescents. EBioMedicine 2023; 96:104797. [PMID: 37716236 PMCID: PMC10511785 DOI: 10.1016/j.ebiom.2023.104797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Genomic characterisation has led to an improved understanding of adult melanoma. However, the aetiology of melanoma in children is still unclear and identifying the correct diagnosis and therapeutic strategies remains challenging. METHODS Exome sequencing of matched tumour-normal pairs from 26 paediatric patients was performed to study the mutational spectrum of melanomas. The cohort was grouped into different categories: spitzoid melanoma (SM), conventional melanoma (CM), and other melanomas (OT). FINDINGS In all patients with CM (n = 10) germline variants associated with melanoma were found in low to moderate melanoma risk genes: in 8 patients MC1R variants, in 2 patients variants in MITF, PTEN and BRCA2. Somatic BRAF mutations were detected in 60% of CMs, homozygous deletions of CDKN2A in 20%, TERTp mutations in 30%. In the SM group (n = 12), 5 patients carried at least one MC1R variant; somatic BRAF mutations were detected in 8.3%, fusions in 25% of the cases. No SM showed a homozygous CDKN2A deletion nor a TERTp mutation. In 81.8% of the CM/SM cases the UV damage signatures SBS7 and/or DBS1 were detected. The patient with melanoma arising in giant congenital nevus (CNM) demonstrated the characteristic NRAS Q61K mutation. INTERPRETATION UV-radiation and MC1R germline variants are risk factors in the development of conventional and spitzoid paediatric melanomas. Paediatric CMs share genomic similarities with adult CMs while the SMs differ genetically from the CM group. Consistent genetic characterization of all paediatric melanomas will potentially lead to better subtype differentiation, treatment, and prevention in the future. FUNDING Found in Acknowledgement.
Collapse
Affiliation(s)
- Alexandra Liebmann
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Sorin Armeanu-Ebinger
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Hannah Wild
- Paediatric Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Michael Abele
- Paediatric Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Axel Gschwind
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Olga Seibel-Kelemen
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Christian Seitz
- Paediatric Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - German Demidov
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Marc Sturm
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Malou Schadeck
- SYNLAB MVZ Human Genetics Freiburg GmbH, Freiburg, Germany
| | - Michaela Pogoda
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany; NGS Competence Center Tübingen, Tübingen, Germany
| | - Ewa Bien
- Department of Paediatrics, Hematology, Oncology, Medical University of Gdansk, Poland
| | - Malgorzata Krawczyk
- Department of Paediatrics, Hematology, Oncology, Medical University of Gdansk, Poland
| | - Eva Jüttner
- Department of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Schleswig-Holstein, Germany
| | - Thomas Mentzel
- Dermatohistopathology Friedrichshafen, Friedrichshafen, Germany
| | - Maja Cesen
- Department of Paediatric Haematology and Oncology, University Hospital Ljubljana, Ljubljana, Slovenia
| | - Elke Pfaff
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Michal Kunc
- Department of Pathomorphology, Medical University of Gdansk, Poland
| | - Stephan Forchhammer
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Tübingen, Germany
| | - Andrea Forschner
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Tübingen, Germany
| | - Ulrike Leiter-Stöppke
- Department of Dermatology, Center for Dermatooncology, University Hospital Tübingen, Tübingen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, Venereology and Allergology, Charite Universitätsmedizin Berlin, Berlin, Germany
| | | | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Ines B Brecht
- Paediatric Hematology and Oncology, University Children's Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
10
|
Hasanau TN, Pisarev EP, Kisil OV, Zvereva ME. The TERT Promoter: A Key Player in the Fight for Cancer Cell Immortality. BIOCHEMISTRY (MOSCOW) 2023; 88:S21-S38. [PMID: 37069112 DOI: 10.1134/s000629792314002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The review describes the role of telomeres and telomerase in tumor progression, as well as various mechanisms of the activation of telomerase reverse transcriptase (TERT) expression in CNS tumors and other cancers. The main mechanism of TERT activation involves acquisition of somatic mutations by the TERT gene promoter (TERTp). The article presents information on the TERTp structure and transcription factors directly interacting with TERTp and regulating its transcription. The prospects of using the mutational status of TERTp as a prognostic marker of CNS malignancies and other tumors with a common profile of TERTp mutations are discussed.
Collapse
Affiliation(s)
- Tsimur N Hasanau
- Natural Compounds Department, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Eduard P Pisarev
- Natural Compounds Department, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Olga V Kisil
- Gause Institute of New Antibiotics, Moscow, 119021, Russia
| | - Maria E Zvereva
- Natural Compounds Department, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
11
|
Towards diagnostic criteria for malignant deep penetrating melanocytic tumors using single nucleotide polymorphism array and next-generation sequencing. Mod Pathol 2022; 35:1110-1120. [PMID: 35184152 DOI: 10.1038/s41379-022-01026-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 11/09/2022]
Abstract
Cutaneous deep penetrating melanocytic neoplasms frequently simulate melanoma and might occasionally progress to metastatic melanoma. Distinguishing deep penetrating nevi (DPN) and deep penetrating melanocytomas (DPM) from malignant deep penetrating tumors (MDPT) is difficult based on histopathology alone, and diagnostic criteria for MDPT are currently lacking. Using a molecular workup, we aimed to provide readily available diagnostic tools for classification of deep penetrating tumors. We used clinical follow-up and Single Nucleotide Polymorphism (SNP) array for tumor classification of 20 deep penetrating neoplasms to identify associations with histopathological, immunohistochemistry, and NGS findings. Ten neoplasms were classified as MDPT, four as DPM, and six as DPN. Two MDPT showed metastases. The following parameters were statistically significantly associated with MDPT: severe nuclear atypia (risk ratio [RR] 2.9, p < 0.05), absence of a nevus component (RR 10.0, p = 0.04), positive PRAME expression (RR 9.0, p = 0.02), complete loss of p16 expression (RR 3.5, p = 0.003), TERT-p and APC mutations (RR 11.0, p = 0.01 and RR 2.7, p = 0.002, respectively), and ≥1 additional pathogenic mutation (RR 9.0, p = 0.02). Ki-67 expression ≥ 5% was not significantly associated with MDPTs, although it was <5% in all DPNs. Three MDPT did not show nuclear β-catenin expression despite having a CTNNB1 (n = 2) or an APC mutation (n = 1). Our findings suggest that complete loss of p16 and positive PRAME expression, a driver mutation in APC, ≥ 1 additional pathogenic mutation, especially in TERT-p, support an MDPT diagnosis in deep penetrating neoplasms. Besides severe nuclear atypia and possibly severe inflammation, we did not identify specific histopathological criteria for malignancy. Non-aberrant nuclear β-catenin expression might not exclude a deep penetrating signature in MDPT.
Collapse
|
12
|
Dobre EG, Constantin C, Neagu M. Skin Cancer Research Goes Digital: Looking for Biomarkers within the Droplets. J Pers Med 2022; 12:jpm12071136. [PMID: 35887633 PMCID: PMC9323323 DOI: 10.3390/jpm12071136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
Skin cancer, which includes the most frequent malignant non-melanoma carcinomas (basal cell carcinoma, BCC, and squamous cell carcinoma, SCC), along with the difficult to treat cutaneous melanoma (CM), pose important worldwide issues for the health care system. Despite the improved anti-cancer armamentarium and the latest scientific achievements, many skin cancer patients fail to respond to therapies, due to the remarkable heterogeneity of cutaneous tumors, calling for even more sophisticated biomarker discovery and patient monitoring approaches. Droplet digital polymerase chain reaction (ddPCR), a robust method for detecting and quantifying low-abundance nucleic acids, has recently emerged as a powerful technology for skin cancer analysis in tissue and liquid biopsies (LBs). The ddPCR method, being capable of analyzing various biological samples, has proved to be efficient in studying variations in gene sequences, including copy number variations (CNVs) and point mutations, DNA methylation, circulatory miRNome, and transcriptome dynamics. Moreover, ddPCR can be designed as a dynamic platform for individualized cancer detection and monitoring therapy efficacy. Here, we present the latest scientific studies applying ddPCR in dermato-oncology, highlighting the potential of this technology for skin cancer biomarker discovery and validation in the context of personalized medicine. The benefits and challenges associated with ddPCR implementation in the clinical setting, mainly when analyzing LBs, are also discussed.
Collapse
Affiliation(s)
- Elena-Georgiana Dobre
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91–95, 050095 Bucharest, Romania;
- Correspondence:
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Monica Neagu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91–95, 050095 Bucharest, Romania;
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
13
|
Dika E, de Biase D, Lambertini M, Alessandrini AM, Acquaviva G, De Leo A, Tallini G, Ricci C, Starace M, Misciali C, Piraccini BM. Mutational landscape in squamous cell carcinoma of the nail unit. Exp Dermatol 2022; 31:854-861. [PMID: 34951714 DOI: 10.1111/exd.14518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/08/2021] [Accepted: 12/21/2021] [Indexed: 02/05/2023]
Abstract
Squamous cell carcinoma (SCC) is the most common malignancy of the nail unit. Pathogenetic mechanisms are yet to be determined, and a deeper molecular characterization of this disease is still necessary. The aim was to obtain a molecular characterization of NU SCC samples using an NGS approach to identify the genetic drivers involved in this tumor. The presence of HPV infection was also assessed. Furthermore, the mutational status was correlated with specific clinical-pathological features for a better insight into the carcinogenesis of this uncommon tumor. We analysed twenty paraffin-embedded nail unit SCC samples from patients diagnosed with primary SCC of the nail unit by next genome sequencing. In the 20 tested samples, the neoplastic cells enrichment ranged from 10% to 50% (mean value: 25.7%). In 14/20 cases (70.0%), at least one mutation was detected; whereas in the other six cases (30.0%), no alterations were observed ('wild-type/WT cases'). Overall, a total of 23 mutations were identified in the 20 specimens. TP53 was the most mutated gene (6/20 cases, 30.0%), while cKit, GNAS, EGFR, DICER1 and CTNNB1 were observed in one sample each (5.0%). No clinical-pathological parameters (age, sex, depth of invasion-DOI, histological subtype, grading and HPV) were significantly associated with the mutational status. The nail unit SCC mutational landscape appeared to be heterogeneous, favouring the hypothesis of a complex pathogenesis and an interaction of multiple elements, including HPV infections. This wealth of information undoubtedly improves our understanding of SCC biology.
Collapse
Affiliation(s)
- Emi Dika
- Dermatology, IRCCS di Policlinico Sant'Orsola, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, Bologna, Italy
| | - Martina Lambertini
- Dermatology, IRCCS di Policlinico Sant'Orsola, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Aurora Maria Alessandrini
- Dermatology, IRCCS di Policlinico Sant'Orsola, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Giorgia Acquaviva
- Department of Medicine, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale), Molecular Diagnostic Unit, University of Bologna, Azienda USL di Bologna, Bologna, Italy
| | - Antonio De Leo
- Department of Medicine, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale), Molecular Diagnostic Unit, University of Bologna, Azienda USL di Bologna, Bologna, Italy
| | - Giovanni Tallini
- Department of Medicine, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale), Molecular Diagnostic Unit, University of Bologna, Azienda USL di Bologna, Bologna, Italy
| | - Costantino Ricci
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Pathology Unit, Maggiore Hospital, Azienda USL di Bologna, Bologna, Italy
| | - Michela Starace
- Dermatology, IRCCS di Policlinico Sant'Orsola, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Cosimo Misciali
- Dermatology, IRCCS di Policlinico Sant'Orsola, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Bianca Maria Piraccini
- Dermatology, IRCCS di Policlinico Sant'Orsola, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
14
|
Fung MA, Vidal CI, Armbrecht EA, Andea AA, Cassarino DS, Comfere NI, Emanuel PO, Ferringer T, Hristov AC, Kim J, Lauer SR, Linos K, Missall TA, Motaparthi K, Novoa RA, Patel R, Shalin SC, Sundram U, Calame A, Bennett DD, Duncan LM, Elston DM, Hosler GA, Hurley YM, Lazar AJ, Lowe L, Messina J, Myles J, Plaza JA, Prieto VG, Reddy V, Schaffer A, Subtil A. Appropriate use criteria for ancillary diagnostic testing in dermatopathology: New recommendations for 11 tests and 220 clinical scenarios from the American Society of Dermatopathology Appropriate Use Criteria Committee. J Cutan Pathol 2022; 49:231-245. [PMID: 34536035 DOI: 10.1111/cup.14135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Appropriate use criteria (AUC) provide patient-centered physician guidance in test selection. An initial set of AUC was reported by the American Society of Dermatopathology (ASDP) in 2018. AUC reflect evidence collected at single timepoints and may be affected by evolving evidence and experience. The objective of this study was to update and expand AUC for selected tests. METHODS RAND/UCLA (RAND Corporation [Santa Monica, CA]/University of California Los Angeles) methodology used includes the following: (a) literature review; (b) review of previously rated tests and previously employed clinical scenarios; (c) selection of previously rated tests for new ratings; (d) development of new clinical scenarios; (e) selection of additional tests; (f) three rating rounds with feedback and group discussion after rounds 1 and 2. RESULTS For 220 clinical scenarios comprising lymphoproliferative (light chain clonality), melanocytic (comparative genomic hybridization, fluorescence in situ hybridization, reverse transcription polymerase chain reaction, telomerase reverse transcriptase promoter), vascular disorders (MYC), and inflammatory dermatoses (periodic acid-Schiff, Gömöri methenamine silver), consensus by panel raters was reached in 172 of 220 (78%) scenarios, with 103 of 148 (70%) rated "usually appropriate" or "rarely appropriate" and 45 of 148 (30%), "appropriateness uncertain." LIMITATIONS The study design only measures appropriateness. Cost, availability, test comparison, and additional clinical considerations are not measured. The possibility that the findings of this study may be influenced by the inherent biases of the dermatopathologists involved in the study cannot be excluded. CONCLUSIONS AUC are reported for selected diagnostic tests in clinical scenarios that occur in dermatopathology practice. Adhering to AUC may reduce inappropriate test utilization and improve healthcare delivery.
Collapse
Affiliation(s)
- Maxwell A Fung
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, California, USA
| | - Claudia I Vidal
- Dermatology Center of Southern Indiana, Bloomington, Indiana, USA
| | - Eric A Armbrecht
- Center for Health Outcomes Research, Saint Louis University, Saint Louis, Missouri, USA
| | - Aleodor A Andea
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David S Cassarino
- Department of Pathology, Kaiser Permanente Southern California, Los Angeles, California, USA
| | - Nneka I Comfere
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Tammie Ferringer
- Department of Dermatology, Geisinger Medical Center, Danville, Pennsylvania, USA
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania, USA
| | - Alexandra C Hristov
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jinah Kim
- Palo Alto Medical Foundation, Palo Alto, California, USA
| | - Scott R Lauer
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Konstantinos Linos
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Tricia A Missall
- Department of Dermatology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Kiran Motaparthi
- Department of Dermatology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Roberto A Novoa
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Rajiv Patel
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sara C Shalin
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Uma Sundram
- Department of Anatomic Pathology, Oakland University William Beaumont School of Medicine and Beaumont Health Systems, Royal Oak, Michigan, USA
| | | | - Daniel D Bennett
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Lyn M Duncan
- Pathology Service and Dermatopathology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dirk M Elston
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gregory A Hosler
- ProPath, Department of Dermatology, Dallas, Texas, USA
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yadira M Hurley
- Department of Dermatology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lori Lowe
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jane Messina
- Department of Dermatology and Cutaneous Surgery, University of South Florida, Moffitt Cancer Center, Tampa, Florida, USA
| | - Jonathan Myles
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jose A Plaza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Dermatology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Victor G Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vijaya Reddy
- Department of Pathology, Rush Medical College, Chicago, Illinois, USA
- Department of Dermatology, Rush Medical College, Chicago, Illinois, USA
| | - András Schaffer
- Division of Dermatology, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Antonio Subtil
- Royal Jubilee Hospital, Victoria, British Columbia, Canada
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
15
|
Mutational Characteristics of Primary Mucosal Melanoma: A Systematic Review. Mol Diagn Ther 2022; 26:189-202. [PMID: 35195858 DOI: 10.1007/s40291-021-00572-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Primary mucosal melanomas (PMMs) are rare and clinically heterogeneous, including head and neck (HNMs), vulvovaginal (VVMs), conjunctival (CjMs), anorectal (ARMs) and penile (PMs) melanomas. While the prognosis of advanced cutaneous melanoma has noticeably improved using treatments with immune checkpoint inhibitors (ICIs) and molecules targeting BRAF and MEK, few advances have been made for PMMs because of their poorer response to ICIs and their different genetic profile. This prompted us to conduct a systematic review of molecular studies of PMMs to clarify their pathogenesis and potential therapeutic targets. METHODS All articles that examined gene mutations in PMMs were identified from the databases and selected based on predefined inclusion criteria. Mutation rate was calculated for all PMMs and each location group by relating the number of mutations identified to the total number of samples analysed. RESULTS Among 1,581 studies identified, 88 were selected. Overall, the frequency of KIT, BRAF and NRAS mutation was 13.5%, 12.9% and 12.1%, respectively. KIT mutation ranged from 6.4% for CjMs to 16.6% for ARMs, BRAF mutation from 8.6% for ARMs to 31.1% for CjMs, and NRAS mutation from 6.2% for ARMs to 18.5% for CjMs. Among 101 other genes analysed, 33 had mutation rates over 10%, including TTN, TSC1, POM121, NF1, MTOR and SF3B1. CONCLUSION In addition to BRAF, NRAS and KIT genes commonly studied, our systematic review identified significantly mutated genes that have already been associated (e.g., TSC1, mTOR, POLE or ATRX) or could be associated with (future) targeted therapies. PROSPERO ID CRD42020185552.
Collapse
|
16
|
Millán-Esteban D, Peña-Chilet M, García-Casado Z, Manrique-Silva E, Requena C, Bañuls J, López-Guerrero JA, Rodríguez-Hernández A, Traves V, Dopazo J, Virós A, Kumar R, Nagore E. Mutational Characterization of Cutaneous Melanoma Supports Divergent Pathways Model for Melanoma Development. Cancers (Basel) 2021; 13:5219. [PMID: 34680367 PMCID: PMC8533762 DOI: 10.3390/cancers13205219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/22/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
According to the divergent pathway model, cutaneous melanoma comprises a nevogenic group with a propensity to melanocyte proliferation and another one associated with cumulative solar damage (CSD). While characterized clinically and epidemiologically, the differences in the molecular profiles between the groups have remained primarily uninvestigated. This study has used a custom gene panel and bioinformatics tools to investigate the potential molecular differences in a thoroughly characterized cohort of 119 melanoma patients belonging to nevogenic and CSD groups. We found that the nevogenic melanomas had a restricted set of mutations, with the prominently mutated gene being BRAF. The CSD melanomas, in contrast, showed mutations in a diverse group of genes that included NF1, ROS1, GNA11, and RAC1. We thus provide evidence that nevogenic and CSD melanomas constitute different biological entities and highlight the need to explore new targeted therapies.
Collapse
Affiliation(s)
- David Millán-Esteban
- School of Medicine, Universidad Católica de València San Vicente Mártir, 46001 Valencia, Spain;
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain; (Z.G.-C.); (J.A.L.-G.)
| | - María Peña-Chilet
- Clinical Bioinformatics Area, Fundación Progreso y Salud, Hospital Virgen del Rocío, 41013 Sevilla, Spain; (M.P.-C.); (J.D.)
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | - Zaida García-Casado
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain; (Z.G.-C.); (J.A.L.-G.)
| | - Esperanza Manrique-Silva
- Department of Dermatology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain; (E.M.-S.); (A.R.-H.)
| | - Celia Requena
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | - José Bañuls
- Department of Dermatology, El Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Hospital General Universitario de Alicante, 03010 Alicante, Spain;
| | - Jose Antonio López-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain; (Z.G.-C.); (J.A.L.-G.)
| | - Aranzazu Rodríguez-Hernández
- Department of Dermatology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain; (E.M.-S.); (A.R.-H.)
| | - Víctor Traves
- Department of Pathological Anatomy, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain;
| | - Joaquín Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud, Hospital Virgen del Rocío, 41013 Sevilla, Spain; (M.P.-C.); (J.D.)
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, 41013 Sevilla, Spain;
- Fundación Progreso y Salud-ELIXIR-es, Hospital Virgen del Rocío, 41013 Sevilla, Spain
| | - Amaya Virós
- Skin Cancer and Aging Lab, Cancer Research UK Manchester Institute, University of Manchester, Manchester SK10 4TG, UK;
| | - Rajiv Kumar
- Division of Functional Genome Analysis, Deutsches Krebsforschüngzentrum, 69120 Heidelberg, Germany;
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska, 142 20 Prague, Czech Republic
- Institute of Medical Biometry and Informatics, University of Heidelberg, 69117 Heidelberg, Germany
| | - Eduardo Nagore
- School of Medicine, Universidad Católica de València San Vicente Mártir, 46001 Valencia, Spain;
- Department of Dermatology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain; (E.M.-S.); (A.R.-H.)
| |
Collapse
|
17
|
Cho WC, Wang WL, Milton DR, Ingram DR, Nagarajan P, Curry JL, Ivan D, Lazar AJ, Hwu WJ, Prieto VG, Torres-Cabala CA, Aung PP. Telomerase Reverse Transcriptase Protein Expression Is More Frequent in Acral Lentiginous Melanoma Than in Other Types of Cutaneous Melanoma. Arch Pathol Lab Med 2021; 145:842-850. [PMID: 33053175 DOI: 10.5858/arpa.2020-0330-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Molecularly distinct from cutaneous melanomas arising from sun-exposed sites, acral lentiginous melanomas (ALMs) typically lack ultraviolet-signature mutations, such as telomerase reverse transcriptase (TERT) promoter mutations. Instead, ALMs show a high degree of copy number alterations, often with multiple amplifications of TERT, which are associated with adverse prognosis. The prognostic value of TERT protein expression in acral melanomas, however, is not established. OBJECTIVE.— To evaluate the frequency and pattern of TERT immunoreactivity and assess the potential utility of TERT expression as a prognostic indicator in ALMs. DESIGN.— TERT expression by immunohistochemistry was analyzed in a series of 57 acral and nonacral melanocytic lesions, including 24 primary and 6 metastatic ALMs. Clinical outcome in patients with ALMs by TERT expression was assessed. RESULTS.— TERT expression was more frequent in ALMs than in nonlentiginous acral melanomas and nonacral cutaneous melanomas, and was absent in acral nevi (P = .01). When present, TERT expression in ALMs was cytoplasmic and more intense than TERT expression in other melanocytic lesions (P = .05) with a higher H-score (P = .01). There was a trend toward decreased overall survival in patients with ALMs with TERT immunoreactivity, but it did not reach statistical significance. Furthermore, no correlation was found between TERT expression and disease-specific survival in patients with ALMs. CONCLUSIONS.— Although TERT protein expression was frequently detected in both primary and metastatic ALMs, TERT immunoreactivity in ALMs did not correlate with survival in our study. Further studies with larger cohorts are needed to elucidate the prognostic value of TERT expression in ALMs.
Collapse
Affiliation(s)
- Woo Cheal Cho
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston
| | - Wei-Lien Wang
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston.,Translational Molecular Pathology (Wang, Ingram, Lazar), The University of Texas MD Anderson Cancer Center, Houston
| | - Denái R Milton
- Biostatistics (Milton), The University of Texas MD Anderson Cancer Center, Houston
| | - Davis R Ingram
- Translational Molecular Pathology (Wang, Ingram, Lazar), The University of Texas MD Anderson Cancer Center, Houston
| | - Priyadharsini Nagarajan
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston
| | - Jonathan L Curry
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston
| | - Doina Ivan
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston
| | - Alexander J Lazar
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston.,Translational Molecular Pathology (Wang, Ingram, Lazar), The University of Texas MD Anderson Cancer Center, Houston.,Melanoma Medical Oncology (Lazar), The University of Texas MD Anderson Cancer Center, Houston. Torres-Cabala and Aung contributed equally to this work
| | - Wen-Jen Hwu
- Genomic Medicine (Hwu), The University of Texas MD Anderson Cancer Center, Houston
| | - Victor G Prieto
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston.,Dermatology (Curry, Ivan, Prieto, Torres-Cabala), The University of Texas MD Anderson Cancer Center, Houston
| | - Carlos A Torres-Cabala
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston.,Dermatology (Curry, Ivan, Prieto, Torres-Cabala), The University of Texas MD Anderson Cancer Center, Houston
| | - Phyu P Aung
- From the Departments of Pathology (Cho, Wang, Nagarajan, Curry, Ivan, Lazar, Prieto, Torres-Cabala, Aung), The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
18
|
Ramani NS, Aung PP, Gu J, Sfamenos S, Sdringola-Maranga C, Nagarajan P, Tetzlaff MT, Curry JL, Ivan D, Diab A, Prieto VG, Hwu WJ, Torres-Cabala CA. TERT amplification but not activation of canonical Wnt/β-catenin pathway is involved in acral lentiginous melanoma progression to metastasis. Mod Pathol 2020; 33:2067-2074. [PMID: 32404956 DOI: 10.1038/s41379-020-0565-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 01/10/2023]
Abstract
Acral lentiginous melanoma (ALM) is a rare tumor that occurs on non-sun exposed skin areas of the hands and feet. Reports suggest that ALM exhibits poor prognosis, although mechanisms driving this remain poorly understood. Alterations in TERT and the Wnt/β-catenin (Wnt) pathway have been suggested to correlate with prognosis of ALM. Thus, immunohistochemical expression of β-catenin and LEF1 along with TERT amplification by FISH was investigated in 34 primary ALMs, 20 metastatic ALMs, 10 primary non-ALMs, and 15 acral nevi. Foot/toe was the most common primary tumor location (85%) for ALM. TERT amplification was detected in 6 of 28 (21.4%) primary ALM, 2 of 8 (25%) primary non-ALM, and 8 of 18 (44.4%) metastatic ALM, the latter showing significantly higher frequency compared with primary melanomas (P = 0.043). Most metastatic ALMs positive for TERT amplification lacked BRAF V600E (87.5%). Cytoplasmic and nonnuclear expression of β-catenin was variably detected in all cases. Metastatic ALM revealed lower expression of β-catenin compared with primary ALM (P = 0.017). No differences in LEF1 expression were detected among the groups; however, acral nevi showed decreased labeling with dermal descent, in contrast to melanoma. No molecular-genetic alteration correlated with prognosis. TERT amplification by FISH is a frequent finding in primary ALM and appears to increase in metastatic tumors, suggesting a role in tumor progression to metastasis. Although TERT amplification has been reported to be infrequent in primary non-ALM, it showed comparable frequency with ALM in our series. Our immunohistochemical findings are not fully supportive of activation of either canonical or noncanonical Wnt cascades in ALM. TERT amplification by FISH and LEF1 immunohistochemistry may help in the differential diagnosis between primary ALM and acral nevus. TERT amplification appears to be a promising target for therapy in patients with metastatic ALM.
Collapse
Affiliation(s)
- Nisha S Ramani
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Phyu P Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun Gu
- Cytogenetics Training Laboratory, School of Health Professions, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven Sfamenos
- Cytogenetics Training Laboratory, School of Health Professions, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chiara Sdringola-Maranga
- Cytogenetics Training Laboratory, School of Health Professions, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyardhisini Nagarajan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael T Tetzlaff
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jonathan L Curry
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Doina Ivan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adi Diab
- Department of Melanoma Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victor G Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen-Jen Hwu
- Department of Melanoma Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos A Torres-Cabala
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
19
|
Pathology-based Biomarkers Useful for Clinical Decisions in Melanoma. Arch Med Res 2020; 51:827-838. [PMID: 32950263 DOI: 10.1016/j.arcmed.2020.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022]
Abstract
The dramatic recent advances in therapy of melanoma require a more personalized and precise diagnostic approach to aid in clinical decisions. Tissue-based biomarkers in pathology have diagnostic, prognostic and predictive relevance. Herein we review the most commonly used pathology-based biomarkers in melanoma. Most of these biomarkers are evaluated through immunohistochemistry (IHC) or fluorescent in situ hybridization (FISH) performed on formalin fixed paraffin embedded tissue (FFPE), and are widely available in clinical pathology laboratories. We describe the utility of MART1/Ki67, p16, PRAME, markers of lymphovascular invasion (D2-40, CD31, D2-40/MITF, CD31/SOX-10), BRAF V600E, NRAS, KIT, BAP1, ALK, NTRK, PD-L1, TERT, PTEN, iNOS, and MMR proteins (MLH1, MSH2, MSH6, PMS2) in the evaluation of melanoma specimens. Correct interpretation and awareness of the significance of these biomarkers is crucial for pathologists, dermatologists, and oncologists who take care of melanoma patients.
Collapse
|