1
|
Kodali N, Bhattaru A, Blanchard I, Sharma Y, Lipner SR. Assessing melanoma prognosis: the interplay between patient profiles, survival, and BRAF, NRAS, KIT, and TWT mutations in a retrospective multi-study analysis. Melanoma Res 2024; 34:419-428. [PMID: 38564430 DOI: 10.1097/cmr.0000000000000968] [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: 04/04/2024]
Abstract
The incidence and prevalence of melanoma are increasing globally, presenting a significant public health concern. The main genetic drivers of melanoma include BRAF, NRAS, KIT and triple wild-type (TWT) mutations. Little is known about the effects of these mutations on outcomes in terms of demographics and patient characteristics. We examined differences in melanoma mortality risk and mutation count across mutation type and patient disease profile. We extrapolated primary melanoma patient data from 14 studies via the cBioportal database. Patients were divided into demographic groups and classified according to BRAF, NRAS, KIT and TWT mutation status. Analyses included two-sample Student t -test and two-way analysis of variance tests analysis with Tukey's post hoc test. Survival outcomes were compared via Kaplan-Meier curve and Cox regression. NRAS-mutated patients exhibited decreased overall survival compared to BRAF-mutated patients. Male patients had higher mutation counts across all gene groups than females, with the fewest TWT mutations in comparison to BRAF, NRAS and KIT mutations. Males also exhibited increased mortality risk for NRAS, KIT and TWT mutations compared to BRAF mutations. An unknown primary melanoma was associated with increased mortality risk across all gene groups. NRAS-mutated acral melanoma patients had an increased mortality risk compared to NRAS-mutated cutaneous melanoma patients. Older patients had a higher mortality risk than younger patients. Patients with heavier versus lower weights had lower mortality risk, which was more pronounced for BRAF-mutated patients. These relationships highlight the importance of demographic and pathologic relationships to aid in risk assessment and personalize treatment plans.
Collapse
Affiliation(s)
- Nilesh Kodali
- Department of Education, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Abhijit Bhattaru
- Department of Education, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Isabella Blanchard
- Department of Education, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Yash Sharma
- Derpartment of Education, UT Southwestern Medical School, Dallas, Texas
| | - Shari R Lipner
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
2
|
Choi ME, Choi EJ, Jung JM, Lee WJ, Jo YS, Won CH. A Narrative Review of the Evolution of Diagnostic Techniques and Treatment Strategies for Acral Lentiginous Melanoma. Int J Mol Sci 2024; 25:10414. [PMID: 39408752 PMCID: PMC11477219 DOI: 10.3390/ijms251910414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Acral melanoma (AM) is a subtype of cutaneous melanoma located on the palms, soles, and nails. The pathogenesis of AM involves mechanical stimulation and characteristic tumor-promoting mutations, such as those in the KIT proto-oncogene. Dermoscopy is useful for diagnosing AM, which is characterized by parallel ridge patterns and irregular diffuse pigmentation. Although histopathological confirmation is the gold standard for diagnosing AM, lesions showing minimal histopathological changes should be considered early-stage AM if they clinically resemble it. Recently, immunohistochemical staining of preferentially expressed antigen in melanoma has been recognized as a useful method to distinguish benign from malignant melanocytic tumors. Research reveals that AM is associated with an immunosuppressive microenvironment characterized by increased numbers of M2 macrophages and regulatory T cells, alongside a decreased number of tumor-infiltrating lymphocytes. Mohs micrographic surgery or digit-sparing wide local excision has been explored to improve quality of life and replace wide local excision or proximal amputation. AM has a worse prognosis than other subtypes, even in the early stages, indicating its inherent aggressiveness.
Collapse
Affiliation(s)
| | | | | | | | | | - Chong Hyun Won
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (M.E.C.); (E.J.C.); (J.M.J.); (W.J.L.); (Y.-S.J.)
| |
Collapse
|
3
|
Colombino M, Casula M, Paliogiannis P, Manca A, Sini MC, Pisano M, Santeufemia DA, Cossu A, Palmieri G. Heterogeneous pathogenesis of melanoma: BRAF mutations and beyond. Crit Rev Oncol Hematol 2024; 201:104435. [PMID: 38977143 DOI: 10.1016/j.critrevonc.2024.104435] [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: 01/31/2024] [Revised: 05/22/2024] [Accepted: 06/29/2024] [Indexed: 07/10/2024] Open
Abstract
Melanoma pathogenesis, conventionally perceived as a linear accumulation of molecular changes, discloses substantial heterogeneity driven by non-linear biological processes, including the direct transformation of melanocyte stem cells. This heterogeneity manifests in diverse biological phenotypes and developmental states, influencing variable responses to treatments. Unveiling the aberrant mechanisms steering melanoma initiation, progression, and metastasis is imperative. Beyond mutations in oncogenic and tumor suppressor genes, the involvement of distinct molecular pathways assumes a pivotal role in melanoma pathogenesis. Ultraviolet (UV) radiations, a principal factor in melanoma etiology, categorizes melanomas based on cumulative sun damage (CSD). The genomic landscape of lesions correlates with UV exposure, impacting mutational load and spectrum of mutations. The World Health Organization's 2018 classification underscores the interplay between sun exposure and genomic characteristics, distinguishing melanomas associated with CSD from those unrelated to CSD. The classification elucidates molecular features such as tumor mutational burden and copy number alterations associated with different melanoma subtypes. The significance of the mutated BRAF gene and its pathway, notably BRAFV600 variants, in melanoma is paramount. BRAF mutations, prevalent across diverse cancer types, present therapeutic avenues, with clinical trials validating the efficacy of targeted therapies and immunotherapy. Additional driver mutations in oncogenes further characterize specific melanoma pathways, impacting tumor behavior. While histopathological examination remains pivotal, challenges persist in molecularly classifying melanocytic tumors. In this review, we went through all molecular characterization that aid in discriminating common and ambiguous lesions. Integration of highly sensitive molecular diagnostic tests into the diagnostic workflow becomes indispensable, particularly in instances where histology alone fails to achieve a conclusive diagnosis. A diagnostic algorithm based on different molecular features inferred by the various studies is here proposed.
Collapse
Affiliation(s)
- Maria Colombino
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy.
| | - Milena Casula
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | | | - Antonella Manca
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Maria Cristina Sini
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Marina Pisano
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | | | - Antonio Cossu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy; Immuno-Oncology & Targeted Cancer Biotherapies, University of Sassari, Sassari, Italy
| |
Collapse
|
4
|
Kodali N, Alomary S, Bhattaru A, Eldaboush A, Schwartz RA, Lipner SR. Gender and melanoma subtype-based prognostic implications of MUC16 and TTN co-occurrent mutations in melanoma: A retrospective multi-study analysis. Cancer Med 2024; 13:e70199. [PMID: 39240165 PMCID: PMC11378355 DOI: 10.1002/cam4.70199] [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: 02/09/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Most primary cutaneous melanomas have pathogenesis driven by ultraviolet exposure and genetic mutations, whereas acral lentiginous melanoma (ALM) and metastatic melanoma are much less, if at all, linked with the former. Thus, we evaluated both ultraviolet related and non-ultraviolet related melanomas. Mutations in the MUC16 and TTN genes commonly occur concurrently in these melanoma patients, but their combined prognostic significance stratified by gender and cancer subtype remains unclear. METHODS The cBioPortal database was queried for melanoma studies and returned 16 independent studies. Data from 2447 melanoma patients were utilized including those with ALM, cutaneous melanoma (CM), and melanoma of unknown primary (MUP). Patients were grouped based on the presence or absence of MUC16 and TTN mutations. Univariate Cox regression and Student's t-tests were used to analyze hazard ratios and total mutation count comparisons, respectively. RESULTS TTN mutations, either alone or concurrently with MUC16 mutations, significantly correlated with worse prognosis overall, in both genders, and in CM patients. ALM patients with both mutations had better prognoses than CM patients, while ALM patients with neither mutation had worse prognosis than CM patients. For MUP patients, only MUC16 mutations correlated with worse prognosis. ALM patients with neither MUC16 nor TTN mutations had significantly more total mutations than MUP patients, followed by CM patients. CONCLUSION TTN mutations are a potential marker of poor prognosis in melanoma, which is amplified in the presence of concurrent MUC16 mutations. ALM patients with neither gene mutations had worse prognosis, suggesting a protective effect of having both MUC16 and TTN mutations. Only MUC16 mutations conferred a worse prognosis for MUP patients. Comprehensive genetic profiling in melanoma patients may facilitate personalized treatment strategies to optimize patient outcomes.
Collapse
Affiliation(s)
- Nilesh Kodali
- Rutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | | | | | - Ahmed Eldaboush
- Department of DermatologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Shari R. Lipner
- Department of DermatologyWeill Cornell MedicineNew YorkNew YorkUSA
| |
Collapse
|
5
|
Smith EA, Belote RL, Cruz NM, Moustafa TE, Becker CA, Jiang A, Alizada S, Chan TY, Seasor TA, Balatico M, Cortes-Sanchez E, Lum DH, Hyngstrom JR, Zeng H, Deacon DC, Grossmann AH, White RM, Zangle TA, Judson-Torres RL. Receptor tyrosine kinase inhibition leads to regression of acral melanoma by targeting the tumor microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.15.599116. [PMID: 38948879 PMCID: PMC11212935 DOI: 10.1101/2024.06.15.599116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Acral melanoma (AM) is an aggressive melanoma variant that arises from palmar, plantar, and nail unit melanocytes. Compared to non-acral cutaneous melanoma (CM), AM is biologically distinct, has an equal incidence across genetic ancestries, typically presents in advanced stage disease, is less responsive to therapy, and has an overall worse prognosis. Independent analysis of published genomic and transcriptomic sequencing identified that receptor tyrosine kinase (RTK) ligands and adapter proteins are frequently amplified, translocated, and/or overexpressed in AM. To target these unique genetic changes, a zebrafish acral melanoma model was exposed to a panel of narrow and broad spectrum multi-RTK inhibitors, revealing that dual FGFR/VEGFR inhibitors decrease acral-analogous melanocyte proliferation and migration. The potent pan-FGFR/VEGFR inhibitor, Lenvatinib, uniformly induces tumor regression in AM patient-derived xenograft (PDX) tumors but only slows tumor growth in CM models. Unlike other multi-RTK inhibitors, Lenvatinib is not directly cytotoxic to dissociated AM PDX tumor cells and instead disrupts tumor architecture and vascular networks. Considering the great difficulty in establishing AM cell culture lines, these findings suggest that AM may be more sensitive to microenvironment perturbations than CM. In conclusion, dual FGFR/VEGFR inhibition may be a viable therapeutic strategy that targets the unique biology of AM.
Collapse
Affiliation(s)
- Eric A Smith
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Rachel L Belote
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Nelly M Cruz
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tarek E Moustafa
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Carly A Becker
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Amanda Jiang
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Shukran Alizada
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Tsz Yin Chan
- Preclinical Research Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Tori A Seasor
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Michael Balatico
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Emilio Cortes-Sanchez
- Immuno Oncology Network Core, The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - David H Lum
- Preclinical Research Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - John R Hyngstrom
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hanlin Zeng
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dekker C Deacon
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Allie H Grossmann
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Richard M White
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Ludwig Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford UK
| | - Thomas A Zangle
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Robert L Judson-Torres
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
6
|
Alhaskawi A, Ezzi SHA, Dong Y, Zhou H, Wang Z, Lai J, Yao C, Kota VG, Abdulla MHAH, Lu H. Recent advancements in the diagnosis and treatment of acral melanoma. J Zhejiang Univ Sci B 2024; 25:106-122. [PMID: 38303495 PMCID: PMC10835211 DOI: 10.1631/jzus.b2300221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/21/2023] [Indexed: 02/03/2024]
Abstract
Acral melanoma (AM) is the most common histologic subtype of melanoma in dark-skinned patients and is associated with a worse prognosis and a high mortality rate, largely due to the inconspicuous nature of early-stage lesions, which can lead to late diagnosis. Because of the overlapping clinical and histopathological features of AM with other forms of cutaneous melanomas, early detection of AM requires a multidisciplinary approach that integrates various diagnostic modalities, including clinical examination, dermoscopy, histopathology, molecular testing, radiological imaging, and blood tests. While surgery is the preferred method of treatment for AM, other therapeutic options may be employed based on the stage and underlying etiology of the disease. Immune checkpoint inhibitors, molecular targeted therapy, radiotherapy, chemotherapy, and oncolytic virotherapy represent promising advanced treatment options for AM. In this review, we provide an overview of the latest advancements in diagnostic and therapeutic methods for AM, highlighting the importance of early detection and the prompt, individualized management of this challenging disease.
Collapse
Affiliation(s)
- Ahmad Alhaskawi
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | - Yanzhao Dong
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Zewei Wang
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingtian Lai
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chengjun Yao
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | | | | | - Hui Lu
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
7
|
Zhang J, Zhang F, Porter KI, Dakup PP, Wang S, Robertson GP, Gaddameedhi S, Zhu J. Telomere dysfunction in Tert knockout mice delays Braf V600E -induced melanoma development. Int J Cancer 2024; 154:548-560. [PMID: 37727982 PMCID: PMC10840707 DOI: 10.1002/ijc.34713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023]
Abstract
Telomerase activation is a crucial step in melanomagenesis, often occurring because of ultraviolet radiation (UVR)-induced mutations at the telomerase gene (TERT) promoter and rendering TERT transcription in response to the activated Raf-MAP kinase pathway by BRAFV600E mutation. Due to the excessively long telomeres in mice, this process does not occur during melanomagenesis in mouse models. To investigate the impact of telomere dysfunction on melanomagenesis, BrafV600E was induced in generations 1 and 4 (G1 and G4) of Tert-/- mice. Our findings revealed that, regardless of UVR exposure, melanoma development was delayed in G4 mice, which had shorter telomeres compared to G1 and wild-type C57BL/6J (G0) mice. Moreover, many G4 tumors displayed an accumulation of excessive DNA damage, as evidenced by increased γH2A.X staining. Tumors from UVR-exposed mice exhibited elevated p53 protein expression. Cultured tumor cells isolated from G4 mice displayed abundant chromosomal fusions and rearrangements, indicative of telomere dysfunction in these cells. Additionally, tumor cells derived from UVB-exposed mice exhibited constitutively elevated expression of mutant p53 proteins, suggesting that p53 was a target of UVB-induced mutagenesis. Taken together, our findings suggest that telomere dysfunction hampers melanomagenesis, and targeting telomere crisis-mediated genomic instability may hold promise for the prevention and treatment of melanoma.
Collapse
Affiliation(s)
- Jinglong Zhang
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99210, USA
| | - Fan Zhang
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99210, USA
| | - Kenneth I. Porter
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99210, USA
| | - Panshak P. Dakup
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99210, USA
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27606, USA
| | - Shuwen Wang
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99210, USA
| | - Gavin P. Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Shobhan Gaddameedhi
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27606, USA
| | - Jiyue Zhu
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99210, USA
| |
Collapse
|
8
|
Zhang J, Tian H, Mao L, Si L. Treatment of acral and mucosal melanoma: Current and emerging targeted therapies. Crit Rev Oncol Hematol 2024; 193:104221. [PMID: 38036156 DOI: 10.1016/j.critrevonc.2023.104221] [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: 04/05/2023] [Revised: 10/14/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023] Open
Abstract
Targeted therapies revolutionized the management of patients with advanced and metastatic cutaneous melanoma. However, despite recent advances in the understanding of the molecular drivers of melanoma and its treatment with targeted therapies, patients with rare and aggressive melanoma subtypes, including acral melanoma (AM) and mucosal melanomas (MM), show limited long-term clinical benefit from current targeted therapies. While patients with AM or MM and BRAF or KIT mutations may benefit from targeted therapies, the frequency of these mutations is relatively low, and there are no genotype-specific treatments for most patients with AM or MM who lack common driver mutations. The poor prognosis of AM and MM can also be attributed to the lack of understanding of their unique molecular landscapes and clinical characteristics, due to being under-represented in preclinical and clinical studies. We review current knowledge of the molecular landscapes of AM and MM, focusing on actionable therapeutic targets and pathways for molecular targeted therapies, to guide the development of more effective targeted therapies for these cancers. Current and emerging strategies for the treatment of these melanoma subtypes using targeted therapies are also summarized.
Collapse
Affiliation(s)
- Jiaran Zhang
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Huichun Tian
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lili Mao
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Lu Si
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
9
|
Carvalho LAD, Aguiar FC, Smalley KSM, Possik PA. Acral melanoma: new insights into the immune and genomic landscape. Neoplasia 2023; 46:100947. [PMID: 37913653 PMCID: PMC10637990 DOI: 10.1016/j.neo.2023.100947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
Acral melanoma is a rare subtype of melanoma that arises on the non-hair bearing skin of the nail bed, palms of the hand and soles of the feet. It is unique among melanomas in not being linked to ultraviolet radiation (UVR) exposure from the sun, and, as such, its incidence is similar across populations who are of Asian, Hispanic, African and European origin. Although research into acral melanoma has lagged behind that of sun-exposed cutaneous melanoma, recent studies have begun to address the unique genetics and immune features of acral melanoma. In this review we will discuss the latest progress in understanding the biology of acral melanoma across different ethnic populations and will outline how these new discoveries can help to guide the therapeutic management of this rare tumor.
Collapse
Affiliation(s)
| | - Flavia C Aguiar
- Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rua Andre Cavalcanti 37, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Keiran S M Smalley
- Department of Tumor Biology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612 USA.
| | - Patricia A Possik
- Division of Basic and Experimental Research, Brazilian National Cancer Institute, Rua Andre Cavalcanti 37, Rio de Janeiro, RJ, 20231-050, Brazil
| |
Collapse
|
10
|
Shi K, Zhang B, Kong BY, Zhang Y, Igartua C, Mohan LS, Quan VL, Panah E, Isales MC, Beaubier N, Taxter TJ, White KP, Zou L, Gerami P. Distinct genomic features in a retrospective cohort of mucosal, acral, and vulvovaginal melanomas. J Am Acad Dermatol 2023; 88:1051-1059. [PMID: 31306728 DOI: 10.1016/j.jaad.2019.07.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/21/2019] [Accepted: 07/03/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Compared with sun-exposed melanomas, less is known regarding the pathogenesis of sun-protected melanomas. Sun-protected melanomas share many epidemiologic factors, but their genetic heterogeneity is not well studied. OBJECTIVE We investigated the genomic profile of acral, mucosal, and vulvovaginal melanomas. We hypothesize that mucosal melanomas, recognized for their uniquely aggressive clinical behavior, have distinct genomic features. METHODS We performed whole transcriptome messenger RNA and DNA (1711 genes) sequencing, messenger RNA expression profiling, tumor mutational burden, ultraviolet signature, and copy number variants analysis on 29 volar/digital acral, 7 mucosal, and 6 vulvovaginal melanomas. RESULTS There was significant genetic heterogeneity, particularly in acral melanomas, with 36% having BRAF alterations, whereas other melanomas had none (P = .0159). Nonzero ultraviolet signatures were more frequent in acral melanomas, suggesting greater ultraviolet involvement. Mucosal melanomas formed a distinct group with increased expression of cell cycle and proliferation genes. Various targetable aberrations were identified, such as AURKA and ERBB2, in mucosal and acral melanomas, respectively. LIMITATIONS The sample size was a small. CONCLUSION There is significant genetic heterogeneity among sun-protected melanomas. Mucosal melanomas have upregulation in cell cycle and proliferation genes, which may explain their aggressive behavior. Ultraviolet radiation plays some role in a subset of acral but not other melanomas.
Collapse
Affiliation(s)
- Katherine Shi
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Bin Zhang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Betty Y Kong
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Yongzhan Zhang
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois
| | | | - Lauren S Mohan
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Victor L Quan
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Elnaz Panah
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Maria Cristina Isales
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | | | | | - Lihua Zou
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Pedram Gerami
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Robert H. Lurie Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
11
|
Abstract
Over the past decade, melanoma has led the field in new cancer treatments, with impressive gains in on-treatment survival but more modest improvements in overall survival. Melanoma presents heterogeneity and transcriptional plasticity that recapitulates distinct melanocyte developmental states and phenotypes, allowing it to adapt to and eventually escape even the most advanced treatments. Despite remarkable advances in our understanding of melanoma biology and genetics, the melanoma cell of origin is still fiercely debated because both melanocyte stem cells and mature melanocytes can be transformed. Animal models and high-throughput single-cell sequencing approaches have opened new opportunities to address this question. Here, we discuss the melanocytic journey from the neural crest, where they emerge as melanoblasts, to the fully mature pigmented melanocytes resident in several tissues. We describe a new understanding of melanocyte biology and the different melanocyte subpopulations and microenvironments they inhabit, and how this provides unique insights into melanoma initiation and progression. We highlight recent findings on melanoma heterogeneity and transcriptional plasticity and their implications for exciting new research areas and treatment opportunities. The lessons from melanocyte biology reveal how cells that are present to protect us from the damaging effects of ultraviolet radiation reach back to their origins to become a potentially deadly cancer.
Collapse
Affiliation(s)
- Patricia P Centeno
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, UK
| | - Valeria Pavet
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, UK
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, UK.
- Oncodrug Ltd, Alderly Park, Macclesfield, UK.
| |
Collapse
|
12
|
Moon S, Kim HJ, Lee Y, Lee YJ, Jung S, Lee JS, Hahn SH, Kim K, Roh JY, Nam S. Oncogenic signaling pathways and hallmarks of cancer in Korean patients with acral melanoma. Comput Biol Med 2023; 154:106602. [PMID: 36716688 DOI: 10.1016/j.compbiomed.2023.106602] [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: 06/20/2022] [Revised: 12/23/2022] [Accepted: 01/22/2023] [Indexed: 01/25/2023]
Abstract
Acral melanoma (AM), a rare subtype of cutaneous melanoma, shows higher incidence in Asians, including Koreans, than in Caucasians. However, the genetic modification associated with AM in Koreans is not well known and has not been comprehensively investigated in terms of oncogenic signaling, and hallmarks of cancer. We performed whole-exome and RNA sequencing for Korean patients with AM and acquired the genetic alterations and gene expression profiles. KIT alterations (previously known to be recurrent alterations in AM) and CDK4/CCND1 copy number amplifications were identified in the patients. Genetic and transcriptomic alterations in patients with AM were functionally converge to the hallmarks of cancer and oncogenic pathways, including 'proliferative signal persistence', 'apoptotic resistance', and 'activation of invasion and metastasis', despite the heterogeneous somatic mutation profiles of Korean patients with AM. This study may provide a molecular understanding for therapeutic strategy for AM.
Collapse
Affiliation(s)
- SeongRyeol Moon
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea
| | - Hee Joo Kim
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea
| | - Yeeun Lee
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea
| | - Yu Joo Lee
- Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea
| | - Sungwon Jung
- Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea
| | - Jin Sook Lee
- Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea; Department of Pediatrics, Seoul National University Hospital Child Cancer and Rare Disease Administration, Seoul National University Children's Hospital, Seoul, 03080, South Korea
| | - Si Houn Hahn
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle Children's Hospital, Seattle, WA, 98105, USA
| | | | - Joo Young Roh
- Department of Dermatology, Ewha Womans University College of Medicine, Seoul Hospital, Seoul, 07804, South Korea.
| | - Seungyoon Nam
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea; Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea; AI Convergence Center for Medical Science, Gachon University College of Medicine, Incheon, 21565, South Korea.
| |
Collapse
|
13
|
Jang H, Ryu HJ, Kim SK. Genetic Characteristics of Primary Cutaneous Malignant Melanoma in Koreans Compared With Western Populations. In Vivo 2023; 37:862-867. [PMID: 36881068 PMCID: PMC10026668 DOI: 10.21873/invivo.13154] [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: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND/AIM Cutaneous melanoma, a melanocyte malignancy, can be divided into many clinical subtypes that differ in presentation, demographics, and genetic profile. In this study, we used next-generation sequencing (NGS) analysis to review genetic alterations in 47 primary cutaneous melanomas in the Korean population and compared them to alterations from melanomas in Western populations. PATIENTS AND METHODS We retrospectively reviewed clinicopathologic and genetic features of 47 patients diagnosed with cutaneous melanomas between 2019-2021 at Severance Hospital, Yonsei University College of Medicine. NGS analysis was performed at diagnosis to evaluate single nucleotide variations (SNVs), copy number variations (CNVs), and genetic fusions. Genetic features in Western cohorts of melanoma were then compared with previous studies performed in the USA: Cohort 1 (n=556), Cohort 2 (n=79), and Cohort 3 (n=38). RESULTS The most common histological classification of melanoma was the acral lentiginous type (23/47, 48.9%). BRAF V600 mutation was most frequent (11/47, 23.4%), but was significantly lower compared to Cohort 1 (240/556, 43.2%) and Cohort 2 (34/79, 43.0%) (p=0.0300). CNV analysis identified amplifications in chromosomes 12q14.1-12q15 (11/47, 23.4%) including CDK4 and MDM2 genes and 11q13.3 (9/47, 19.2%) including CND1, FGF19, FGF3, and FGF4 genes more frequently in the present study population than Cohort 1 (p<0.0001). CONCLUSION These results clearly demonstrated differences in genetic alterations between melanomas in Asian and Western populations. Therefore, BRAF V600 mutation should be considered a significant signaling pathway explaining melanoma pathogenesis occurrence in both Asian and Western populations, whereas loss of chromosome 9p21.3 is unique to melanomas in Western populations.
Collapse
Affiliation(s)
- Hyenguk Jang
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Hyang Joo Ryu
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Sang Kyum Kim
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
14
|
Chen X, Liu T, Wu J, Zhu C, Guan G, Zou C, Guo Q, Ren X, Li C, Cheng P, Cheng W, Wu A. Molecular profiling identifies distinct subtypes across TP53 mutant tumors. JCI Insight 2022; 7:156485. [PMID: 36256461 PMCID: PMC9746906 DOI: 10.1172/jci.insight.156485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 10/14/2022] [Indexed: 01/12/2023] Open
Abstract
Tumor protein 53 mutation (TP53mut) is one of the most important driver events facilitating tumorigenesis, which could induce a series of chain reactions to promote tumor malignant transformation. However, the malignancy progression patterns under TP53 mutation remain less known. Clarifying the molecular landscapes of TP53mut tumors will help us understand the process of tumor development and aid precise treatment. Here, we distilled genetic and epigenetic features altered in TP53mut cancers for cluster-of-clusters analysis. Using integrated classification, we derived 5 different subtypes of TP53mut patients. These subtypes have distinct features in genomic alteration, clinical relevance, microenvironment dysregulation, and potential therapeutics. Among the 5 subtypes, COCA3 was identified as the subtype with worst prognosis, causing an immunosuppressive microenvironment and immunotherapeutic resistance. Further drug efficacy research highlighted olaparib as the most promising therapeutic agents for COCA3 tumors. Importantly, the therapeutic efficacy of olaparib in COCA3 and immunotherapy in non-COCA3 tumors was validated via in vivo experimentation. Our study explored the important molecular events and developed a subtype classification system with distinct targeted therapy strategies for different subtypes of TP53mut tumors. These multiomics classification systems provide a valuable resource that significantly expands the knowledge of TP53mut tumors and may eventually benefit in clinical practice.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianqi Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianqi Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Cunyi Zou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qing Guo
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Ren
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, Shenyang Red Cross Hospital, Shenyang, Liaoning, China
| | - Chen Li
- Department of Orthodontics, Stomatological Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Chiu Y, Weng H, Lin Y, Lin Y, Yeh Y, Perng C, Ma H, Tsai S, Chou T. Genomic profiling with whole‐exome sequencing revealed distinct mutations and novel pathways in Asian melanoma. J Dermatol 2022; 49:1299-1309. [DOI: 10.1111/1346-8138.16579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Yu‐Jen Chiu
- Institute of Clinical Medicine National Yang Ming Chiao Tung University Taipei Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery Taipei Veterans General Hospital Taipei Taiwan
- Department of Surgery, School of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Hui‐Ying Weng
- Biomedical Industry PhD Program National Yang Ming Chiao Tung University Taipei Taiwan
| | - Yen‐Yu Lin
- Department of Pathology and Laboratory Medicine Taipei Veterans General Hospital Taipei Taiwan
| | - Yung‐Feng Lin
- Institute of Molecular and Genomic Medicine National Health Research Institutes Miaoli Taiwan
| | - Yi‐Chen Yeh
- Department of Pathology and Laboratory Medicine Taipei Veterans General Hospital Taipei Taiwan
| | - Chern‐Kang Perng
- Division of Plastic and Reconstructive Surgery, Department of Surgery Taipei Veterans General Hospital Taipei Taiwan
- Department of Surgery, School of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery Taipei Veterans General Hospital Taipei Taiwan
- Department of Surgery, School of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
- Department of Surgery National Defense Medical Center Taipei Taiwan
| | - Shih‐Feng Tsai
- Institute of Molecular and Genomic Medicine National Health Research Institutes Miaoli Taiwan
| | - Teh‐Ying Chou
- Institute of Clinical Medicine National Yang Ming Chiao Tung University Taipei Taiwan
- Department of Pathology and Laboratory Medicine Taipei Veterans General Hospital Taipei Taiwan
| |
Collapse
|
16
|
Moran JMT, Le LP, Nardi V, Golas J, Farahani AA, Signorelli S, Onozato ML, Foreman RK, Duncan LM, Lawrence DP, Lennerz JK, Dias-Santagata D, Hoang MP. Identification of fusions with potential clinical significance in melanoma. Mod Pathol 2022; 35:1837-1847. [PMID: 35871080 DOI: 10.1038/s41379-022-01138-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022]
Abstract
Though uncommon in melanoma, gene fusions may have therapeutic implications. Next generation sequencing-based clinical assays, designed to detect relevant gene fusions, mutations, and copy number changes, were performed on 750 melanomas (375 primary and 375 metastases) at our institution from 2014-2021. These included 599 (80%) cutaneous, 38 (5%) acral, 11 (1.5%) anorectal, 23 (3%) sinonasal, 27 (3.6%) eye (uveal/ conjunctiva), 11 (1.5%) genital (vulva/penile), and 41 (5.5%) melanomas of unknown primary. Sixteen fusions (2%) were detected in samples from 16 patients: 12/599 (2%) cutaneous, 2/38 (5%) acral, 1/9 (11%) vulva, 1/23(4.3%) sinonasal; and 12/16 (75%) fusions were potentially targetable. We identified two novel rearrangements: NAGS::MAST2 and NOTCH1::GNB1; and two fusions that have been reported in other malignancies but not in melanoma: CANT1::ETV4 (prostate cancer) and CCDC6::RET (thyroid cancer). Additional fusions, previously reported in melanoma, included: EML4::ALK, MLPH::ALK, AGAP3::BRAF, AGK::BRAF, CDH3::BRAF, CCT8::BRAF, DIP2B::BRAF, EFNB1::RAF1, LRCH3::RAF1, MAP4::RAF1, RUFY1::RAF1, and ADCY2::TERT. Fusion positive melanomas harbored recurrent alterations in TERT and CDKN2A, among others. Gene fusions were exceedingly rare (0.2%) in BRAF/RAS/NF1-mutant tumors and were detected in 5.6% of triple wild-type melanomas. Interestingly, gene rearrangements were significantly enriched within the subset of triple wild-type melanomas that harbor TERT promoter mutations (18% versus 2%, p < 0.0001). Thirteen (81%) patients were treated with immunotherapy for metastatic disease or in the adjuvant setting. Six of 12 (50%) patients with potentially actionable fusions progressed on immunotherapy, and 3/6 (50%) were treated with targeted agents (ALK and MEK inhibitors), 2 off-label and 1 as part of a clinical trial. One patient with an AGAP3::BRAF fusion positive melanoma experienced a 30-month long response to trametinib. We show that, detecting fusions, especially in triple wild-type melanomas with TERT promoter mutations, may have a clinically significant impact in patients with advanced disease who have failed front-line immunotherapy.
Collapse
Affiliation(s)
- Jakob M T Moran
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Long P Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Josephine Golas
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander A Farahani
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylvia Signorelli
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maristela L Onozato
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruth K Foreman
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lyn M Duncan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donald P Lawrence
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jochen K Lennerz
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Mai P Hoang
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Kim YS, Lee M, Chung YJ. Two subtypes of cutaneous melanoma with distinct mutational signatures and clinico-genomic characteristics. Front Genet 2022; 13:987205. [PMID: 36246650 PMCID: PMC9557124 DOI: 10.3389/fgene.2022.987205] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/23/2022] [Indexed: 11/15/2022] Open
Abstract
Background: To decipher mutational signatures and their associations with biological implications in cutaneous melanomas (CMs), including those with a low ultraviolet (UV) signature. Materials and Methods: We applied non-negative matrix factorization (NMF) and unsupervised clustering to the 96-class mutational context of The Cancer Genome Atlas (TCGA) cohort (N = 466) as well as other publicly available datasets (N = 527). To explore the feasibility of mutational signature-based classification using panel sequencing data, independent panel sequencing data were analyzed. Results: NMF decomposition of the TCGA cohort and other publicly available datasets consistently found two mutational signatures: UV (SBS7a/7b dominant) and non-UV (SBS1/5 dominant) signatures. Based on mutational signatures, TCGA CMs were classified into two clusters: UV-high and UV-low. CMs belonging to the UV-low cluster showed significantly worse overall survival and landmark survival at 1-year than those in the UV-high cluster; low or high UV signature remained the most significant prognostic factor in multivariate analysis. The UV-low cluster showed distinct genomic and functional characteristic patterns: low mutation counts, increased proportion of triple wild-type and KIT mutations, high burden of copy number alteration, expression of genes related to keratinocyte differentiation, and low activation of tumor immunity. We verified that UV-high and UV-low clusters can be distinguished by panel sequencing. Conclusion: Our study revealed two mutational signatures of CMs that divide CMs into two clusters with distinct clinico-genomic characteristics. Our results will be helpful for the clinical application of mutational signature-based classification of CMs.
Collapse
Affiliation(s)
- Yoon-Seob Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Integrated Research Center for Genome Polymorphism (IRCGP), College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, South Korea
| | - Yeun-Jun Chung
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Integrated Research Center for Genome Polymorphism (IRCGP), College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
18
|
Advanced Acral Melanoma Therapies: Current Status and Future Directions. Curr Treat Options Oncol 2022; 23:1405-1427. [PMID: 36125617 PMCID: PMC9526689 DOI: 10.1007/s11864-022-01007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
Melanoma is one of the deadliest malignancies. Its incidence has been significantly increasing in most countries in recent decades. Acral melanoma (AM), a peculiar subgroup of melanoma occurring on the palms, soles, and nails, is the main subtype of melanoma in people of color and is extremely rare in Caucasians. Although great progress has been made in melanoma treatment in recent years, patients with AM have shown limited benefit from current therapies and thus consequently have worse overall survival rates. Achieving durable therapeutic responses in this high-risk melanoma subtype represents one of the greatest challenges in the field. The frequency of BRAF mutations in AM is much lower than that in cutaneous melanoma, which prevents most AM patients from receiving treatment with BRAF inhibitors. However, AM has more frequent mutations such as KIT and CDK4/6, so targeted therapy may still improve the survival of some AM patients in the future. AM may be less susceptible to immune checkpoint inhibitors because of the poor immunogenicity. Therefore, how to enhance the immune response to the tumor cells may be the key to the application of immune checkpoint inhibitors in advanced AM. Anti-angiogenic drugs, albumin paclitaxel, or interferons are thought to enhance the effectiveness of immune checkpoint inhibitors. Combination therapies based on the backbone of PD-1 are more likely to provide greater clinical benefits. Understanding the molecular landscapes and immune microenvironment of AM will help optimize our combinatory strategies.
Collapse
|
19
|
Šerman N, Vranić S, Glibo M, Šerman L, Mokos ZB. Genetic risk factors in melanoma etiopathogenesis and the role of genetic counseling: A concise review. Bosn J Basic Med Sci 2022; 22:673-682. [PMID: 35465855 PMCID: PMC9519167 DOI: 10.17305/bjbms.2021.7378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022] Open
Abstract
Melanoma is a highly aggressive cancer originating from melanocytes. Its etiopathogenesis is strongly related to genetic, epigenetic, and environmental factors. Melanomas encountered in clinical practice are predominantly sporadic, whereas hereditary melanomas account for approximately 10% of the cases. Hereditary melanomas mainly develop due to mutations in the CDKN2A gene, which encodes two tumor suppressor proteins involved in the cell cycle regulation. CDKN2A, along with CDK4, TERT, and POT1 genes, is a high-risk gene for melanoma. Among the genes that carry a moderate risk are MC1R and MITF, whose protein products are involved in melanin synthesis. The environment also contributes to the development of melanoma. Patients at risk of melanoma should be offered genetic counseling to discuss genetic testing options and the importance of skin UV protection, avoidance of sun exposure, and regular preventive dermatological examinations. Although cancer screening cannot prevent the development of the disease, it allows for early diagnosis when the survival rate is the highest.
Collapse
Affiliation(s)
| | - Semir Vranić
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mislav Glibo
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ljiljana Šerman
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence in Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Zrinka Bukvić Mokos
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Dermatology and Venereology, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
20
|
Jung S, Johnson DB. Management of Acral and Mucosal Melanoma: Medical Oncology Perspective. Oncologist 2022; 27:703-710. [PMID: 35640549 PMCID: PMC9355814 DOI: 10.1093/oncolo/oyac091] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Acral and mucosal melanomas (MM) are rare subtypes of melanoma that are biologically and clinically distinct from cutaneous melanoma. Despite the progress in the treatment of cutaneous melanomas with the development of targeted and immune therapies, the therapeutic options for these less common subtypes remain limited. Difficulties in early diagnosis, the aggressive nature of the disease, and the frequently occult sites of origin have also contributed to the poor prognosis associated with acral and MM, with substantially worse long-term prognosis. The rarity of these subtypes has posed significant barriers to better understanding their biological features and investigating novel therapies. Consequently, establishing standardized treatment guidelines has been a challenge. In this review, we provide a brief overview of the current knowledge regarding acral and MM, focusing on their epidemiology, genetic backgrounds, and unique clinical characteristics. Further discussion centers around the management of primary and advanced disease and the role of emerging targeted and immune therapies for these subtypes, specifically focusing on issues relevant to medical oncologists.
Collapse
Affiliation(s)
- Seungyeon Jung
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville TN, USA
| |
Collapse
|
21
|
Bhave P, Ahmed T, Lo SN, Shoushtari A, Zaremba A, Versluis JM, Mangana J, Weichenthal M, Si L, Lesimple T, Robert C, Trojanello C, Wicky A, Heywood R, Tran L, Batty K, Dimitriou F, Stansfeld A, Allayous C, Schwarze JK, Mooradian MJ, Klein O, Mehmi I, Roberts-Thomson R, Maurichi A, Yeoh HL, Khattak A, Zimmer L, Blank CU, Ramelyte E, Kähler KC, Roy S, Ascierto PA, Michielin O, Lorigan PC, Johnson DB, Plummer R, Lebbe C, Neyns B, Sullivan R, Hamid O, Santinami M, McArthur GA, Haydon AM, Long GV, Menzies AM, Carlino MS. Efficacy of anti-PD-1 and ipilimumab alone or in combination in acral melanoma. J Immunother Cancer 2022; 10:e004668. [PMID: 35793872 PMCID: PMC9260790 DOI: 10.1136/jitc-2022-004668] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Acral melanoma is a rare melanoma subtype with poor prognosis. Importantly, these patients were not identified as a specific subgroup in the landmark melanoma trials involving ipilimumab and the anti-programmed cell death protein-1 (PD-1) agents nivolumab and pembrolizumab. There is therefore an absence of prospective clinical trial evidence regarding the efficacy of checkpoint inhibitors (CPIs) in this population. Acral melanoma has lower tumor mutation burden (TMB) than other cutaneous sites, and primary site is associated with differences in TMB. However the impact of this on the effectiveness of immune CPIs is unknown. We examined the efficacy of CPIs in acral melanoma, including by primary site. METHODS Patients with unresectable stage III/IV acral melanoma treated with CPI (anti-PD-1 and/or ipilimumab) were studied. Multivariable logistic and Cox regression analyses were conducted. Primary outcome was objective response rate (ORR); secondary outcomes were progression-free survival (PFS) and overall survival (OS). RESULTS In total, 325 patients were included: 234 (72%) plantar, 69 (21%) subungual and 22 (7%) palmar primary sites. First CPI included: 184 (57%) anti-PD-1, 59 (18%) anti-PD-1/ipilimumab combination and 82 (25%) ipilimumab. ORR was significantly higher with initial anti-PD-1/ipilimumab compared with anti-PD-1 (43% vs 26%, HR 2.14, p=0.0004) and significantly lower with ipilimumab (15% vs 26%, HR 0.49, p=0.0016). Landmark PFS at 1 year was highest for anti-PD-1/ipilimumab at 34% (95% CI 24% to 49%), compared with 26% (95% CI 20% to 33%) with anti-PD-1 and 10% (95% CI 5% to 19%) with ipilimumab. Despite a trend for increased PFS, anti-PD-1/ipilimumab combination did not significantly improve PFS (HR 0.85, p=0.35) or OS over anti-PD-1 (HR 1.30, p=0.16), potentially due to subsequent therapies and high rates of acquired resistance. No outcome differences were found between primary sites. CONCLUSION While the ORR to anti-PD-1/ipilimumab was significantly higher than anti-PD-1 and PFS numerically higher, in this retrospective cohort this benefit did not translate to improved OS. Future trials should specifically include patients with acral melanoma, to help determine the optimal management of this important melanoma subtype.
Collapse
Affiliation(s)
- Prachi Bhave
- Sir Peter MacCallum Cancer Centre Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales, Australia
| | - Tasnia Ahmed
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| | - Alexander Shoushtari
- Medicine, Melanoma and Immunotherapeutics Service, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Anne Zaremba
- Dermatology, University Hospital Essen, Essen, Germany
| | - Judith M Versluis
- Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joanna Mangana
- Dermatology, University Hospital Zürich, Zurich, Switzerland
| | - Michael Weichenthal
- Dermatology, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Lu Si
- Melanoma and Sarcoma, Peking University Cancer Hospital, Beijing, China
| | - Thierry Lesimple
- Research and Medical Oncology, Centre Eugène Marquis, Rennes, France
| | | | - Claudia Trojanello
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione "G.Pascale", Napoli, Italy
| | - Alexandre Wicky
- Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Richard Heywood
- Christie NHS Foundation Trust and Division of Cancer Services, University of Manchester, Manchester, UK
| | - Lena Tran
- Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kathleen Batty
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| | - Florentia Dimitriou
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
- Dermatology, University Hospital Zürich, Zurich, Switzerland
| | - Anna Stansfeld
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Clara Allayous
- Dermatology, Saint-Louis hospital, INSERM U976, AP-HP, Paris, France
| | - Julia K Schwarze
- Medical Oncology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussel, Belgium
| | - Meghan J Mooradian
- Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Oliver Klein
- Medical Oncology, Olivia Newton John Cancer Centre, Austin Health, Melbourne, Victoria, Australia
- Medical Oncology, Warrnambool Hospital, Warrnambool, Victoria, Australia
- Medical Oncology, Peninsula Health, Melbourne, Victoria, Australia
| | - Inderjit Mehmi
- The Angeles Clinic and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, California, USA
| | | | - Andrea Maurichi
- Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Hui-Ling Yeoh
- Medical Oncology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Adnan Khattak
- Medical Oncology, Fiona Stanley Hospital & Edith Cowan Univserity, Perth, Western Australia, Australia
| | - Lisa Zimmer
- Dermatology, University Hospital Essen, Essen, Germany
| | - Christian U Blank
- Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Egle Ramelyte
- Dermatology, University Hospital Zürich, Zurich, Switzerland
| | - Katharina C Kähler
- Dermatology, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | | | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione "G.Pascale", Napoli, Italy
| | | | - Paul C Lorigan
- Christie NHS Foundation Trust and Division of Cancer Services, University of Manchester, Manchester, UK
| | - Douglas B Johnson
- Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ruth Plummer
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Celeste Lebbe
- Université de Paris, AP-HP Department of Dermatology, Hôpital Saint-Louis, Paris, France
| | - Bart Neyns
- Medical Oncology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel, Brussel, Belgium
| | - Ryan Sullivan
- Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, California, USA
| | - Mario Santinami
- Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Grant A McArthur
- Sir Peter MacCallum Cancer Centre Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew M Haydon
- Medical Oncology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Georgina V Long
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales, Australia
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Chen H, Hou K, Yu J, Wang L, Chen X. Nanoparticle-Based Combination Therapy for Melanoma. Front Oncol 2022; 12:928797. [PMID: 35837089 PMCID: PMC9273962 DOI: 10.3389/fonc.2022.928797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 11/18/2022] Open
Abstract
Melanoma is a cutaneous carcinoma, and its incidence is rapidly increasing with every year. The treatment options for melanoma have been comprehensively studied. Conventional treatment methods (e.g., radiotherapy, chemotherapy and photodynamic therapy) with surgical removal inevitably cause serious complications; moreover, resistance is common. Nanoparticles (NPs) combined with conventional methods are new and promising options to treat melanoma, and many combinations have been achieving good success. Due to their physical and biological features, NPs can help target intended melanoma cells more efficiently with less damage. This creates new hope for a better treatment strategy for melanoma with minimum damage and maximum efficacy.
Collapse
Affiliation(s)
- Hongbo Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Hou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Le Wang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Wang M, Banik I, Shain AH, Yeh I, Bastian BC. Integrated genomic analyses of acral and mucosal melanomas nominate novel driver genes. Genome Med 2022; 14:65. [PMID: 35706047 PMCID: PMC9202124 DOI: 10.1186/s13073-022-01068-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/03/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Acral and mucosal melanomas are aggressive subtypes of melanoma, which have a significantly lower burden of somatic mutations than cutaneous melanomas, but more frequent copy number variations, focused gene amplifications, and structural alterations. The landscapes of their genomic alterations remain to be fully characterized. METHODS We compiled sequencing data of 240 human acral and mucosal melanoma samples from 11 previously published studies and applied a uniform pipeline to call tumor cell content, ploidy, somatic and germline mutations, as well as CNVs, LOH, and SVs. We identified genes that are significantly mutated or recurrently affected by CNVs and implicated in oncogenesis. We further examined the difference in the frequency of recurrent pathogenic alterations between the two melanoma subtypes, correlation between pathogenic alterations, and their association with clinical features. RESULTS We nominated PTPRJ, mutated and homozygously deleted in 3.8% (9/240) and 0.8% (2/240) of samples, respectively, as a probable tumor suppressor gene, and FER and SKP2, amplified in 3.8% and 11.7% of samples, respectively, as probable oncogenes. We further identified a long tail of infrequent pathogenic alterations, involving genes such as CIC and LZTR1. Pathogenic germline mutations were observed on MITF, PTEN, ATM, and PRKN. We found BRAF V600E mutations in acral melanomas with fewer structural variations, suggesting that they are distinct and related to cutaneous melanomas. Amplifications of PAK1 and GAB2 were more commonly observed in acral melanomas, whereas SF3B1 R625 codon mutations were unique to mucosal melanomas (12.9%). Amplifications at 11q13-14 were frequently accompanied by fusion to a region on chromosome 6q12, revealing a recurrent novel structural rearrangement whose role remains to be elucidated. CONCLUSIONS Our meta-analysis expands the catalog of driver mutations in acral and mucosal melanomas, sheds new light on their pathogenesis and broadens the catalog of therapeutic targets for these difficult-to-treat cancers.
Collapse
Affiliation(s)
- Meng Wang
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Ishani Banik
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - A Hunter Shain
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Iwei Yeh
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| | - Boris C Bastian
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Shi Q, Liu L, Chen J, Zhang W, Guo W, Wang X, Wang H, Guo S, Yue Q, Ma J, Liu Y, Zhu G, Zhao T, Zhao J, Liu Y, Gao T, Li C. Integrative Genomic Profiling Uncovers Therapeutic Targets of Acral Melanoma in Asian Populations. Clin Cancer Res 2022; 28:2690-2703. [PMID: 35294533 PMCID: PMC9359751 DOI: 10.1158/1078-0432.ccr-21-3344] [Citation(s) in RCA: 4] [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: 09/15/2021] [Revised: 11/18/2021] [Accepted: 03/10/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Acral melanoma is the major subtype of melanoma seen in Asian patients with melanoma and is featured by its insidious onset and poor prognosis. The genomic study that elucidates driving mutational events is fundamental to the development of gene-targeted therapy. However, research on genomic profiles of acral melanoma in Asian patients is still sparse. EXPERIMENTAL DESIGN We carried out whole-exome sequencing (WES) on 60 acral melanoma lesions (with 55 primary samples involved), targeted deep sequencing in a validation cohort of 48 cases, RNA sequencing in 37 acral melanoma samples (all from the 60 undergoing WES), and FISH in 233 acral melanoma specimens (54 of the 60 undergoing WES included). All the specimens were derived from Asian populations. RESULTS BRAF, NRAS, and KIT were discerned as significantly mutated genes (SMG) in acral melanoma. The detected COSMIC signature 3 related to DNA damage repair, along with the high genomic instability score, implied corresponding pathogenesis of acral melanoma. Moreover, the copy number gains of EP300 were associated with the response of acral melanoma to targeted therapy of A485 (a p300 inhibitor) and immune checkpoint blockade treatment. In addition, the temporal order in mutational processes of the samples was reconstructed, and copy-number alterations were identified as early mutational events. CONCLUSIONS Our study provided a detailed view of genomic instability, potential therapeutic targets, and intratumoral heterogeneity of acral melanoma, which might fuel the development of personalized strategies for treating acral melanoma in Asian populations.
Collapse
Affiliation(s)
- Qiong Shi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lin Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weigang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qiao Yue
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jingjing Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guannan Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tao Zhao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianhong Zhao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ying Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
25
|
Klonowska K, Grevelink JM, Giannikou K, Ogorek BA, Herbert ZT, Thorner AR, Darling TN, Moss J, Kwiatkowski DJ. Ultrasensitive profiling of UV-induced mutations identifies thousands of subclinical facial tumors in tuberous sclerosis complex. J Clin Invest 2022; 132:e155858. [PMID: 35358092 PMCID: PMC9106361 DOI: 10.1172/jci155858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
BackgroundTuberous sclerosis complex (TSC) is a neurogenetic syndrome due to loss-of-function mutations in TSC2 or TSC1, characterized by tumors at multiple body sites, including facial angiofibroma (FAF). Here, an ultrasensitive assessment of the extent and range of UV-induced mutations in TSC facial skin was performed.MethodsA multiplex high-sensitivity PCR assay (MHPA) was developed, enabling mutation detection at extremely low (<0.1%) variant allele frequencies (VAFs).ResultsMHPA assays were developed for both TSC2 and TP53, and applied to 81 samples, including 66 skin biopsies. UV-induced second-hit mutation causing inactivation of TSC2 was pervasive in TSC facial skin with an average of 4.8 mutations per 2-mm biopsy at median VAF 0.08%, generating more than 150,000 incipient facial tumors (subclinical "micro-FAFs") in the average TSC subject. The MHPA analysis also led to the identification of a refined UV-related indel signature and a recurrent complex mutation pattern, consisting of both a single-nucleotide or dinucleotide variant and a 1- to 9-nucleotide deletion, in cis.ConclusionTSC facial skin can be viewed as harboring a patchwork of clonal fibroblast proliferations (micro-FAFs) with indolent growth, a small proportion of which develop into clinically observable FAF. Our observations also expand the spectrum of UV-related mutation signatures.FundingThis work was supported by the TSC Alliance; the Engles Family Fund for Research in TSC and LAM; and the NIH, National Heart, Lung, and Blood Institute (U01HL131022-04 and Intramural Research Program).
Collapse
Affiliation(s)
- Katarzyna Klonowska
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joannes M. Grevelink
- Boston Dermatology and Laser Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Krinio Giannikou
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Barbara A. Ogorek
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Aaron R. Thorner
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Thomas N. Darling
- Department of Dermatology, Uniformed Services University, Bethesda, Maryland, USA
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David J. Kwiatkowski
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Li J, Smalley I, Chen Z, Wu JY, Phadke MS, Teer JK, Nguyen T, Karreth FA, Koomen JM, Sarnaik AA, Zager JS, Khushalani NI, Tarhini AA, Sondak VK, Rodriguez PC, Messina JL, Chen YA, Smalley KSM. Single-cell Characterization of the Cellular Landscape of Acral Melanoma Identifies Novel Targets for Immunotherapy. Clin Cancer Res 2022; 28:2131-2146. [PMID: 35247927 PMCID: PMC9106889 DOI: 10.1158/1078-0432.ccr-21-3145] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/10/2021] [Accepted: 03/01/2022] [Indexed: 12/21/2022]
Abstract
PURPOSE Acral melanoma is a rare subtype of melanoma that arises on the non-hair-bearing skin of the palms, soles, and nail beds. In this study, we used single-cell RNA sequencing (scRNA-seq) to map the transcriptional landscape of acral melanoma and identify novel immunotherapeutic targets. EXPERIMENTAL DESIGN We performed scRNA-seq on nine clinical specimens (five primary, four metastases) of acral melanoma. Detailed cell type curation was performed, the immune landscapes were mapped, and key results were validated by analysis of The Cancer Genome Atlas (TCGA) and single-cell datasets. Cell-cell interactions were inferred and compared with those in nonacral cutaneous melanoma. RESULTS Multiple phenotypic subsets of T cells, natural killer (NK) cells, B cells, macrophages, and dendritic cells with varying levels of activation/exhaustion were identified. A comparison between primary and metastatic acral melanoma identified gene signatures associated with changes in immune responses and metabolism. Acral melanoma was characterized by a lower overall immune infiltrate, fewer effector CD8 T cells and NK cells, and a near-complete absence of γδ T cells compared with nonacral cutaneous melanomas. Immune cells associated with acral melanoma exhibited expression of multiple checkpoints including PD-1, LAG-3, CTLA-4, V-domain immunoglobin suppressor of T cell activation (VISTA), TIGIT, and the Adenosine A2A receptor (ADORA2). VISTA was expressed in 58.3% of myeloid cells and TIGIT was expressed in 22.3% of T/NK cells. CONCLUSIONS Acral melanoma has a suppressed immune environment compared with that of cutaneous melanoma from nonacral skin. Expression of multiple, therapeutically tractable immune checkpoints were observed, offering new options for clinical translation.
Collapse
Affiliation(s)
- Jiannong Li
- The Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Inna Smalley
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Zhihua Chen
- The Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Jheng-Yu Wu
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Manali S. Phadke
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Jamie K. Teer
- The Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Thanh Nguyen
- The Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Florian A. Karreth
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - John M. Koomen
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Amod A. Sarnaik
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Jonathan S. Zager
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Nikhil I. Khushalani
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Ahmad A. Tarhini
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Vernon K. Sondak
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Paulo C. Rodriguez
- The Department of Immunology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Jane L. Messina
- The Department of Immunology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Y. Ann Chen
- The Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Keiran S. M. Smalley
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| |
Collapse
|
27
|
Shang Q, Du H, Wu X, Guo Q, Zhang F, Gong Z, Jiao T, Guo J, Kong Y. FMRP ligand circZNF609 destabilizes RAC1 mRNA to reduce metastasis in acral melanoma and cutaneous melanoma. J Exp Clin Cancer Res 2022; 41:170. [PMID: 35534866 PMCID: PMC9087950 DOI: 10.1186/s13046-022-02357-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melanoma is a type of malignant tumor with high aggressiveness and poor prognosis. At present, metastasis of melanoma is still an important cause of death in melanoma patients. However, the potential functions and molecular mechanisms of most circular RNAs (circRNAs) in melanoma metastasis remain unknown. METHODS circRNAs dysregulated in melanoma cell subgroups with different metastatic abilities according to a screening model based on repeated Transwell assays were identified with a circRNA array. The expression and prognostic significance of circZNF609 in skin cutaneous melanoma and acral melanoma cells and tissues were determined by qRT-PCR, nucleoplasmic separation assays and fluorescence in situ hybridization. In vitro wound healing, Transwell and 3D invasion assays were used to analyse melanoma cell metastasis ability. Tail vein injection and intrasplenic injection were used to study in vivo lung metastasis and liver metastasis, respectively. The mechanism of circZNF609 was further evaluated via RNA immunoprecipitation, RNA pull-down, silver staining, and immunofluorescence colocalization assays. RESULTS circZNF609 was stably expressed at low levels in melanoma tissues and cells and was negatively correlated with Breslow depth, clinical stage and prognosis of melanoma patients. circZNF609 inhibited metastasis of acral and cutaneous melanoma in vivo and in vitro. Mechanistically, circZNF609 promoted the binding of FMRP protein and RAC1 mRNA, thereby enhancing the inhibitory effect of FMRP protein on the stability of RAC1 mRNA and ultimately inhibiting melanoma metastasis. CONCLUSIONS Our findings revealed that circZNF609 plays a vital role in the metastasis of acral and cutaneous melanoma through the circRNF609-FMRP-RAC1 axis and indicated that circZNF609 regulates the stability of RAC1 mRNA by combining with FMRP, which might provide insight into melanoma pathogenesis and a new potential target for treatment of melanoma.
Collapse
Affiliation(s)
- Qingfeng Shang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Haizhen Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Xiaowen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Qian Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Fenghao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Ziqi Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Tao Jiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China.
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, China.
| |
Collapse
|
28
|
Broit N, Johansson PA, Rodgers CB, Walpole S, Hayward NK, Pritchard AL. Systematic review and meta-analysis of genomic alterations in acral melanoma. Pigment Cell Melanoma Res 2022; 35:369-386. [PMID: 35229492 PMCID: PMC9540316 DOI: 10.1111/pcmr.13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/15/2022] [Accepted: 02/24/2022] [Indexed: 11/30/2022]
Abstract
Acral melanoma (AM) tumors arise on the palms, soles, fingers, toes, and nailbeds. A comprehensive systematic meta-analysis of AM genomic aberrations has not been conducted to date. A literature review was carried out to identify studies sequencing AM. Whole-genome/exome data from 181 samples were identified. Targeted panel sequencing data from MSK-IMPACT were included as a validation cohort (n = 92), and studies using targeted hot spot sequencing were also collated for BRAF (n = 26 studies), NRAS (n = 21), and KIT (n = 32). Statistical analysis indicated BRAF, NRAS, PTEN, TYRP1, and KIT as significantly mutated genes. Frequent copy-number aberrations were also found for important cancer genes, such as CDKN2A, KIT, MDM2, CCND1, CDK4, and PAK1, among others. Mapping genomic alterations within the context of the hallmarks of cancer identified four components frequently altered, including (i) sustained proliferative signaling and (ii) evading growth suppression, (iii) genome instability and mutation, and (iv) enabling replicative immortality. This analysis provides the largest analysis of genomic aberrations in AM in the literature to date and highlights pathways that may be therapeutically targetable.
Collapse
Affiliation(s)
- Natasa Broit
- Oncogenomics GroupQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Peter A. Johansson
- Oncogenomics GroupQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Chloe B. Rodgers
- Genetics and Immunology GroupUniversity of the Highlands and IslandsInvernessUK
| | - Sebastian T. Walpole
- Oncogenomics GroupQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Nicholas K. Hayward
- Oncogenomics GroupQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Antonia L. Pritchard
- Oncogenomics GroupQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
- Genetics and Immunology GroupUniversity of the Highlands and IslandsInvernessUK
| |
Collapse
|
29
|
Wang J, Peng C, Dai W, Chen X, Meng J, Jiang T. Exploring Tumor Immune Microenvironment and Its Associations With Molecular Characteristics in Melanoma. Front Oncol 2022; 12:821578. [PMID: 35530341 PMCID: PMC9069107 DOI: 10.3389/fonc.2022.821578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe tumor microenvironment (TME), which involves infiltration of multiple immune cells into the tumor tissues, plays an essential role in clinical benefit to therapy. The chemokines and their receptors influence migration and functions of both tumor and immune cells. Also, molecular characteristics are associated with the efficacy of melanoma therapy. However, there lacked exploration of immune characteristics and the association with molecular characteristics.MethodsWe collected the currently available 569 melanoma samples that had both the genomic and transcriptional data from TCGA and SRA databases. We first identified TME subtypes based on the developed immune signatures, and then divided the samples into two immune cohorts based on the immune score. Next, we estimated the compositions of the immune cells of the two cohorts, and performed differential expression genes (DEGs) and functional enrichments. In addition, we investigated the interactions of chemokines and their receptors under immune cells. Finally, we explored the genomic characteristics under different immune subtypes.ResultsTME type D had a better prognosis among the four subtypes. The high-immunity cohort had significantly high 16 immune cells. The 63 upregulated and 384 downregulated genes in the high-immunity cohort were enriched in immune-related biological processes, and keratin, pigmentation and epithelial cells, respectively. The correlations of chemokines and their receptors with immune cell infiltration, such as CCR5-CCL4/CCL5 and CXCR3-CXCL9/CXCL10/CXCL11/CXCL13 axis, showed that the recruitments of 11 immune cells, such as CD4T cells and CD8T cells, were modulated by chemokines and their receptors. The proportions of the four TME subtypes in each molecular subtype were comparable. The two driver genes, CDKN2A and PRB2, had significantly different MAFs between the high-immunity and low-immunity.ConclusionWe dissected the characteristics of immune infiltration, the interactions of chemokines and their receptors under immune cells, and the correlation of molecular and immune characteristics. Our work will enable the reasonable selection of anti-melanoma treatments and accelerate the development of new therapeutic strategies for melanoma.
Collapse
Affiliation(s)
- Jiangyuan Wang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
| | - Wentao Dai
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies) & Shanghai Engineering Research Center of Pharmaceutical Translation, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Gastric Neoplasms, Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiang Chen, ; Jing Meng, ; Taijiao Jiang,
| | - Jing Meng
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
- *Correspondence: Xiang Chen, ; Jing Meng, ; Taijiao Jiang,
| | - Taijiao Jiang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Guangzhou Laboratory, Guangzhou, China
- *Correspondence: Xiang Chen, ; Jing Meng, ; Taijiao Jiang,
| |
Collapse
|
30
|
Weiss JM, Hunter MV, Cruz NM, Baggiolini A, Tagore M, Ma Y, Misale S, Marasco M, Simon-Vermot T, Campbell NR, Newell F, Wilmott JS, Johansson PA, Thompson JF, Long GV, Pearson JV, Mann GJ, Scolyer RA, Waddell N, Montal ED, Huang TH, Jonsson P, Donoghue MTA, Harris CC, Taylor BS, Xu T, Chaligné R, Shliaha PV, Hendrickson R, Jungbluth AA, Lezcano C, Koche R, Studer L, Ariyan CE, Solit DB, Wolchok JD, Merghoub T, Rosen N, Hayward NK, White RM. Anatomic position determines oncogenic specificity in melanoma. Nature 2022; 604:354-361. [PMID: 35355015 PMCID: PMC9355078 DOI: 10.1038/s41586-022-04584-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/25/2022] [Indexed: 12/19/2022]
Abstract
Oncogenic alterations to DNA are not transforming in all cellular contexts1,2. This may be due to pre-existing transcriptional programmes in the cell of origin. Here we define anatomic position as a major determinant of why cells respond to specific oncogenes. Cutaneous melanoma arises throughout the body, whereas the acral subtype arises on the palms of the hands, soles of the feet or under the nails3. We sequenced the DNA of cutaneous and acral melanomas from a large cohort of human patients and found a specific enrichment for BRAF mutations in cutaneous melanoma and enrichment for CRKL amplifications in acral melanoma. We modelled these changes in transgenic zebrafish models and found that CRKL-driven tumours formed predominantly in the fins of the fish. The fins are the evolutionary precursors to tetrapod limbs, indicating that melanocytes in these acral locations may be uniquely susceptible to CRKL. RNA profiling of these fin and limb melanocytes, when compared with body melanocytes, revealed a positional identity gene programme typified by posterior HOX13 genes. This positional gene programme synergized with CRKL to amplify insulin-like growth factor (IGF) signalling and drive tumours at acral sites. Abrogation of this CRKL-driven programme eliminated the anatomic specificity of acral melanoma. These data suggest that the anatomic position of the cell of origin endows it with a unique transcriptional state that makes it susceptible to only certain oncogenic insults.
Collapse
Affiliation(s)
- Joshua M Weiss
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell and Developmental Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Miranda V Hunter
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nelly M Cruz
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arianna Baggiolini
- Developmental Biology, The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohita Tagore
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yilun Ma
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell and Developmental Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sandra Misale
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michelangelo Marasco
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Theresa Simon-Vermot
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nathaniel R Campbell
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics & Systems Biology Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Felicity Newell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Peter A Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - John V Pearson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Graham J Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
- Centre for Cancer Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- New South Wales Health Pathology, Sydney, New South Wales, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Emily D Montal
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ting-Hsiang Huang
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip Jonsson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark T A Donoghue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher C Harris
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tianhao Xu
- Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronan Chaligné
- Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pavel V Shliaha
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald Hendrickson
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cecilia Lezcano
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lorenz Studer
- Developmental Biology, The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte E Ariyan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jedd D Wolchok
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Weill Cornell Medicine, New York, NY, USA
| | | | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Richard M White
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
31
|
Farshidfar F, Rhrissorrakrai K, Levovitz C, Peng C, Knight J, Bacchiocchi A, Su J, Yin M, Sznol M, Ariyan S, Clune J, Olino K, Parida L, Nikolaus J, Zhang M, Zhao S, Wang Y, Huang G, Wan M, Li X, Cao J, Yan Q, Chen X, Newman AM, Halaban R. Integrative molecular and clinical profiling of acral melanoma links focal amplification of 22q11.21 to metastasis. Nat Commun 2022; 13:898. [PMID: 35197475 PMCID: PMC8866401 DOI: 10.1038/s41467-022-28566-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
Acral melanoma, the most common melanoma subtype among non-White individuals, is associated with poor prognosis. However, its key molecular drivers remain obscure. Here, we perform integrative genomic and clinical profiling of acral melanomas from 104 patients treated in North America (n = 37) or China (n = 67). We find that recurrent, late-arising focal amplifications of cytoband 22q11.21 are a leading determinant of inferior survival, strongly associated with metastasis, and linked to downregulation of immunomodulatory genes associated with response to immune checkpoint blockade. Unexpectedly, LZTR1 - a known tumor suppressor in other cancers - is a key candidate oncogene in this cytoband. Silencing of LZTR1 in melanoma cell lines causes apoptotic cell death independent of major hotspot mutations or melanoma subtypes. Conversely, overexpression of LZTR1 in normal human melanocytes initiates processes associated with metastasis, including anchorage-independent growth, formation of spheroids, and an increase in MAPK and SRC activities. Our results provide insights into the etiology of acral melanoma and implicate LZTR1 as a key tumor promoter and therapeutic target.
Collapse
Affiliation(s)
- Farshad Farshidfar
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | | | - Cong Peng
- Xiangya Hospital, Central South University, Changsha, China
| | - James Knight
- Yale Center for Genome Analysis, Yale University, New Haven, CT, 06520, USA
| | | | - Juan Su
- Xiangya Hospital, Central South University, Changsha, China
| | - Mingzhu Yin
- Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Mario Sznol
- Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephan Ariyan
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - James Clune
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Olino
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | | | - Joerg Nikolaus
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Meiling Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Shuang Zhao
- Xiangya Hospital, Central South University, Changsha, China
| | - Yan Wang
- Department of Dermatologic Surgery Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Gang Huang
- Department of Bone and Soft Tissue oncology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Miaojian Wan
- Department of Dermatology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xianan Li
- Department of Bone and Soft Tissue oncology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Jian Cao
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Qin Yan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiang Chen
- Xiangya Hospital, Central South University, Changsha, China.
| | - Aaron M Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| | - Ruth Halaban
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
32
|
Abstract
Activating mutations in RAS genes are the most common genetic driver of human cancers. Yet, drugging this small GTPase has proven extremely challenging and therapeutic strategies targeting these recurrent alterations have long had limited success. To circumvent this difficulty, research has focused on the molecular dissection of the RAS pathway to gain a more-precise mechanistic understanding of its regulation, with the hope to identify new pharmacological approaches. Here, we review the current knowledge on the (dys)regulation of the RAS pathway, using melanoma as a paradigm. We first present a map of the main proteins involved in the RAS pathway, highlighting recent insights into their molecular roles and diverse mechanisms of regulation. We then overview genetic data pertaining to RAS pathway alterations in melanoma, along with insight into other cancers, that inform the biological function of members of the pathway. Finally, we describe the clinical implications of RAS pathway dysregulation in melanoma, discuss past and current approaches aimed at drugging the RAS pathway, and outline future opportunities for therapeutic development. Summary: This Review describes the molecular regulation of the RAS pathway, presents the clinical consequences of its pathological activation in human cancer, and highlights recent advances towards its therapeutic inhibition, using melanoma as an example.
Collapse
Affiliation(s)
- Amira Al Mahi
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, INSERM U1052 CNRS UMR5286, Tumor Escape, Resistance and Immunity Department, 69008 Lyon, France
| | - Julien Ablain
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, INSERM U1052 CNRS UMR5286, Tumor Escape, Resistance and Immunity Department, 69008 Lyon, France
| |
Collapse
|
33
|
Prouteau A, Mottier S, Primot A, Cadieu E, Bachelot L, Botherel N, Cabillic F, Houel A, Cornevin L, Kergal C, Corre S, Abadie J, Hitte C, Gilot D, Lindblad-Toh K, André C, Derrien T, Hedan B. Canine Oral Melanoma Genomic and Transcriptomic Study Defines Two Molecular Subgroups with Different Therapeutical Targets. Cancers (Basel) 2022; 14:cancers14020276. [PMID: 35053440 PMCID: PMC8774001 DOI: 10.3390/cancers14020276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023] Open
Abstract
Simple Summary In humans, mucosal melanoma (MM) is a rare and aggressive cancer. The canine model is frequently and spontaneously affected by MM, thus facilitating the collection of samples and the study of its genetic bases. Thanks to an integrative genomic and transcriptomic analysis of 32 canine MM samples, we identified two molecular subgroups of MM with a different microenvironment and structural variant (SV) content. We demonstrated that SVs are associated with recurrently amplified regions, and identified new candidate oncogenes (TRPM7, GABPB1, and SPPL2A) for MM. Our findings suggest the existence of two MM molecular subgroups that could benefit from dedicated therapies, such as immune checkpoint inhibitors or targeted therapies, for both human and veterinary medicine. Abstract Mucosal melanoma (MM) is a rare, aggressive clinical cancer. Despite recent advances in genetics and treatment, the prognosis of MM remains poor. Canine MM offers a relevant spontaneous and immunocompetent model to decipher the genetic bases and explore treatments for MM. We performed an integrative genomic and transcriptomic analysis of 32 canine MM samples, which identified two molecular subgroups with a different microenvironment and structural variant (SV) content. The overexpression of genes related to the microenvironment and T-cell response was associated with tumors harboring a lower content of SVs, whereas the overexpression of pigmentation-related pathways and oncogenes, such as TERT, was associated with a high SV burden. Using whole-genome sequencing, we showed that focal amplifications characterized complex chromosomal rearrangements targeting oncogenes, such as MDM2 or CDK4, and a recurrently amplified region on canine chromosome 30. We also demonstrated that the genes TRPM7, GABPB1, and SPPL2A, located in this CFA30 region, play a role in cell proliferation, and thus, may be considered as new candidate oncogenes for human MM. Our findings suggest the existence of two MM molecular subgroups that may benefit from dedicated therapies, such as immune checkpoint inhibitors or targeted therapies, for both human and veterinary medicine.
Collapse
Affiliation(s)
- Anais Prouteau
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Stephanie Mottier
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Aline Primot
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Edouard Cadieu
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Laura Bachelot
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Nadine Botherel
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Florian Cabillic
- Laboratoire de Cytogénétique et Biologie Cellulaire, CHU de Rennes, INSERM, INRA, University of Rennes 1, Nutrition Metabolisms and Cancer, 35000 Rennes, France; (F.C.); (L.C.)
| | - Armel Houel
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Laurence Cornevin
- Laboratoire de Cytogénétique et Biologie Cellulaire, CHU de Rennes, INSERM, INRA, University of Rennes 1, Nutrition Metabolisms and Cancer, 35000 Rennes, France; (F.C.); (L.C.)
| | - Camille Kergal
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Sébastien Corre
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Jérôme Abadie
- Laboniris, Department of Biology, Pathology and Food Sciences, Oniris, 44300 Nantes, France;
| | - Christophe Hitte
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - David Gilot
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Catherine André
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Thomas Derrien
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
- Correspondence: (T.D.); (B.H.); Tel.: +33-2-23-23-43-19 (B.H.)
| | - Benoit Hedan
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
- Correspondence: (T.D.); (B.H.); Tel.: +33-2-23-23-43-19 (B.H.)
| |
Collapse
|
34
|
Wan Z, Sun R, Liu YW, Li S, Sun J, Li J, Zhu J, Moharil P, Zhang B, Ren P, Ren G, Zhang M, Ma X, Dai S, Yang D, Lu B, Li S. Targeting metabotropic glutamate receptor 4 for cancer immunotherapy. SCIENCE ADVANCES 2021; 7:eabj4226. [PMID: 34890233 PMCID: PMC8664261 DOI: 10.1126/sciadv.abj4226] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/23/2021] [Indexed: 05/30/2023]
Abstract
In this study, we report a novel role of metabotropic glutamate receptor 4 (GRM4) in suppressing antitumor immunity. We revealed in three murine syngeneic tumor models (B16, MC38, and 3LL) that either genetic knockout (Grm4−/−) or pharmacological inhibition led to significant delay in tumor growth. Mechanistically, perturbation of GRM4 resulted in a strong antitumor immunity by promoting natural killer (NK), CD4+, and CD8+ T cells toward an activated, proliferative, and functional phenotype. Single-cell RNA sequencing and T cell receptor profiling further defined the clonal expansion and immune landscape changes in CD8+ T cells. We further showed that Grm4−/− intrinsically activated interferon-γ production in CD8+ T cells through cyclic adenosine 3′,5′-monophosphate (cAMP)/cAMP response element binding protein–mediated pathway. Our study appears to be of clinical significance as a signature of NKhigh-GRM4low and CD8high-GRM4low correlated with improved survival in patients with melanoma. Targeting GRM4 represents a new approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Zhuoya Wan
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Runzi Sun
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Yang-Wuyue Liu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sihan Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jiang Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Pearl Moharil
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Bei Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Pengfei Ren
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Guolian Ren
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Min Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Shuangshuang Dai
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
35
|
Kolla AM, Vitiello GA, Friedman EB, Sun J, Potdar A, Daou H, Farrow NE, Farley CR, Vetto JT, Han D, Tariq M, Beasley GM, Contreras CM, Lowe M, Zager JS, Osman I, Berman RS, Liebman TN, Stein JA, Lee AY. Acral Lentiginous Melanoma: A United States Multi-Center Substage Survival Analysis. Cancer Control 2021; 28:10732748211053567. [PMID: 34752172 PMCID: PMC8581784 DOI: 10.1177/10732748211053567] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Acral lentiginous melanoma is associated with worse survival than other subtypes of melanoma. Understanding prognostic factors for survival and recurrence can help better inform follow-up care. Objectives To analyze the clinicopathologic features, melanoma-specific survival, and recurrence-free survival by substage in a large, multi-institutional cohort of primary acral lentiginous melanoma patients. Methods Retrospective review of the United States Melanoma Consortium database, a multi-center prospectively collected database of acral lentiginous melanoma patients treated between January 2000 and December 2017. Results Of the 433 primary acral lentiginous melanoma patients identified (median [range] age: 66 [8–97] years; 53% female, 83% white), 66% presented with stage 0–2 disease and the median time of follow-up for the 392 patients included in the survival analysis was 32.5 months (range: 0–259). The 5-year melanoma-specific survivals by stage were 0 = 100%, I = 93.8%, II = 76.2%, III = 63.4%, IIIA = 80.8%, and IV = 0%. Thicker Breslow depth ((HR) = 1.13; 95% CI = 1.05–1.21; P < .001)) and positive nodal status ((HR) = 1.79; 95% CI = 1.00–3.22; P = .050)) were independent prognostic factors for melanoma-specific survival. Breslow depth ((HR = 1.13; 95% CI = 1.07–1.20; P < .001), and positive nodal status (HR = 2.12; 95% CI = 1.38–3.80; P = .001) were also prognostic factors for recurrence-free survival. Conclusion In this cohort of patients, acral lentiginous melanoma was associated with poor outcomes even in early stage disease, consistent with prior reports. Stage IIB and IIC disease were associated with particularly low melanoma-specific and recurrence-free survival. This suggests that studies investigating adjuvant therapies in stage II patients may be especially valuable in acral lentiginous melanoma patients.
Collapse
Affiliation(s)
- Avani M Kolla
- The Ronald O. Perelman Department of Dermatology, 12297NYU Langone Health, New York, NY, USA
| | | | - Erica B Friedman
- Department of Surgery, 12297NYU Langone Health, New York, NY, USA
| | - James Sun
- Department of Cutaneous Oncology, 25301Moffitt Cancer Center, Tampa, FL, USA
| | - Aishwarya Potdar
- Department of Cutaneous Oncology, 25301Moffitt Cancer Center, Tampa, FL, USA.,Department of Surgery, 33697University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hala Daou
- Department of Cutaneous Oncology, 25301Moffitt Cancer Center, Tampa, FL, USA.,Department of Surgery, 33697University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Norma E Farrow
- Department of Surgery, 3065Duke University, Durham, NC, USA
| | - Clara R Farley
- Department of Surgery, 1371Emory University, Atlanta, GA, USA
| | - John T Vetto
- Department of Surgery, 6684Oregon Health & Science University, Portland, OR, USA
| | - Dale Han
- Department of Surgery, 6684Oregon Health & Science University, Portland, OR, USA
| | - Marvi Tariq
- Department of Surgery, 1371Emory University, Atlanta, GA, USA
| | | | - Carlo M Contreras
- Department of Surgery, 2647The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Michael Lowe
- Department of Surgery, 1371Emory University, Atlanta, GA, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, 25301Moffitt Cancer Center, Tampa, FL, USA.,Department of Surgery, 33697University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, 12297NYU Langone Health, New York, NY, USA
| | - Russell S Berman
- Department of Surgery, 12297NYU Langone Health, New York, NY, USA
| | - Tracey N Liebman
- The Ronald O. Perelman Department of Dermatology, 12297NYU Langone Health, New York, NY, USA
| | - Jennifer A Stein
- The Ronald O. Perelman Department of Dermatology, 12297NYU Langone Health, New York, NY, USA
| | - Ann Y Lee
- Department of Surgery, 12297NYU Langone Health, New York, NY, USA
| |
Collapse
|
36
|
Bernardes SS, Ferreira I, Elder DE, Nobre AB, Martínez‐Said H, Adams DJ, Robles‐Espinoza CD, Possik PA. More than just acral melanoma: the controversies of defining the disease. J Pathol Clin Res 2021; 7:531-541. [PMID: 34213090 PMCID: PMC8503895 DOI: 10.1002/cjp2.233] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/13/2021] [Accepted: 06/09/2021] [Indexed: 01/17/2023]
Abstract
Acral melanoma (AM) is a malignant cutaneous melanocytic tumour specifically located on the palms, soles, and nail apparatus, which are areas of glabrous (hairless) skin. Acral lentiginous melanoma, a subtype of AM, represents a histopathological subtype diagnosis of cutaneous melanoma with unique morphological and structural features. Despite clear definitions, the misuse of these terms and the inconsistency in reporting the histopathological features of AM cases have become a major obstacle to the study of the disease. In this review, we discuss the epidemiology, histopathological features, prognosis, and genetic profile of AM, highlighting the differences observed when histopathological subtypes are considered. The increasing global effort to characterise AM cases from ethnically diverse populations would benefit greatly from a more consistent classification of the disease.
Collapse
Affiliation(s)
- Sara S Bernardes
- Program of Immunology and Tumour BiologyBrazilian National Cancer InstituteRio de JaneiroBrazil
- Tissue Microenvironment Laboratory, Department of General PathologyFederal University of Minas GeraisBelo HorizonteBrazil
| | - Ingrid Ferreira
- Experimental Cancer GeneticsWellcome Sanger InstituteHinxtonUK
- Université Libre de BruxellesBrusselsBelgium
| | - David E Elder
- Division of Anatomic PathologyHospital of the University of PennsylvaniaPhiladelphiaPAUSA
| | - Aretha B Nobre
- Division of PathologyBrazilian National Cancer InstituteRio de JaneiroBrazil
- Serviço de Patologia, Maternidade EscolaUniversidade Federal do Rio de JaneiroRio de JaneiroBrazil
| | - Héctor Martínez‐Said
- Servicio de Piel y Partes BlandasInstituto Nacional de CancerologíaCiudad de MéxicoMexico
| | - David J Adams
- Experimental Cancer GeneticsWellcome Sanger InstituteHinxtonUK
| | - Carla Daniela Robles‐Espinoza
- Experimental Cancer GeneticsWellcome Sanger InstituteHinxtonUK
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| | - Patricia A Possik
- Program of Immunology and Tumour BiologyBrazilian National Cancer InstituteRio de JaneiroBrazil
| |
Collapse
|
37
|
Bian SX, Hwang L, Hwang J, Ragab O, In GK, Peng D, Lin E. Acral lentiginous melanoma-Population, treatment, and survival using the NCDB from 2004 to 2015. Pigment Cell Melanoma Res 2021; 34:1049-1061. [PMID: 34273249 PMCID: PMC8599631 DOI: 10.1111/pcmr.12999] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/21/2021] [Accepted: 07/07/2021] [Indexed: 12/27/2022]
Abstract
Acral lentiginous melanoma (ALM) is a rare histological subtype of cutaneous malignant melanoma that typically presents on the palms and soles. To characterize the demographic and treatment characteristics of ALM, we used the National Cancer Database (NCDB) to describe a large multi-institutional cohort of ALM patients, consisting of 4,796 ALM patients from 2004 to 2015. ALM was more likely to be diagnosed at a later stage overall compared with non-ALM cutaneous melanomas, and more likely to be thicker, ulcerated, lymph node positive, and have lymphovascular invasion and positive margins. When stratified by stage, ALM had worse survival compared with non-ALM patients, most notably in stage III patients with 5-year survival of 47.5% versus 56.7%, respectively (p < .001). In ALM patients, older age, male sex, higher comorbidity burden, increased tumor thickness and ulceration, positive lymph nodes, and positive metastasis were independently associated with lower 5-year survival. Multimodality therapy, defined as surgery in addition to systemic therapy and/or radiation therapy, was associated with higher survival in stage III patients but not in other stages. These results call for further investigation into possible treatment intensification in the ALM population in the future.
Collapse
Affiliation(s)
- Shelly X Bian
- Department of Radiation Oncology, University of Southern California, Los Angeles, CA, USA
| | - Lindsay Hwang
- Department of Radiation Oncology, University of Southern California, Los Angeles, CA, USA
| | - Jennifer Hwang
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Omar Ragab
- Department of Radiation Oncology, University of Southern California, Los Angeles, CA, USA
| | - Gino K In
- Department of Medicine, Division of Oncology, University of Southern California, Los Angeles, CA, USA
- Department of Dermatology, University of Southern California, Los Angeles, CA, USA
| | - David Peng
- Department of Dermatology, University of Southern California, Los Angeles, CA, USA
| | - Eugene Lin
- Department of Medicine, Division of Nephrology, University of Southern California, Los Angeles, CA, USA
- Leonard D Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
38
|
Jung T, Haist M, Kuske M, Grabbe S, Bros M. Immunomodulatory Properties of BRAF and MEK Inhibitors Used for Melanoma Therapy-Paradoxical ERK Activation and Beyond. Int J Mol Sci 2021; 22:ijms22189890. [PMID: 34576054 PMCID: PMC8469254 DOI: 10.3390/ijms22189890] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/21/2022] Open
Abstract
The advent of mitogen-activated protein kinase (MAPK) inhibitors that directly inhibit tumor growth and of immune checkpoint inhibitors (ICI) that boost effector T cell responses have strongly improved the treatment of metastatic melanoma. In about half of all melanoma patients, tumor growth is driven by gain-of-function mutations of BRAF (v-rat fibrosarcoma (Raf) murine sarcoma viral oncogene homolog B), which results in constitutive ERK activation. Patients with a BRAF mutation are regularly treated with a combination of BRAF and MEK (MAPK/ERK kinase) inhibitors. Next to the antiproliferative effects of BRAF/MEKi, accumulating preclinical evidence suggests that BRAF/MEKi exert immunomodulatory functions such as paradoxical ERK activation as well as additional effects in non-tumor cells. In this review, we present the current knowledge on the immunomodulatory functions of BRAF/MEKi as well as the non-intended effects of ICI and discuss the potential synergistic effects of ICI and MAPK inhibitors in melanoma treatment.
Collapse
|
39
|
Belote RL, Le D, Maynard A, Lang UE, Sinclair A, Lohman BK, Planells-Palop V, Baskin L, Tward AD, Darmanis S, Judson-Torres RL. Human melanocyte development and melanoma dedifferentiation at single-cell resolution. Nat Cell Biol 2021; 23:1035-1047. [PMID: 34475532 DOI: 10.1038/s41556-021-00740-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/18/2021] [Indexed: 12/13/2022]
Abstract
In humans, epidermal melanocytes are responsible for skin pigmentation, defence against ultraviolet radiation and the deadliest common skin cancer, melanoma. Although there is substantial overlap in melanocyte development pathways between different model organisms, species-dependent differences are frequent and the conservation of these processes in human skin remains unresolved. Here, we used a single-cell enrichment and RNA-sequencing pipeline to study human epidermal melanocytes directly from the skin, capturing transcriptomes across different anatomical sites, developmental age, sexes and multiple skin tones. We uncovered subpopulations of melanocytes that exhibit anatomical site-specific enrichment that occurs during gestation and persists through adulthood. The transcriptional signature of the volar-enriched subpopulation is retained in acral melanomas. Furthermore, we identified human melanocyte differentiation transcriptional programs that are distinct from gene signatures generated from model systems. Finally, we used these programs to define patterns of dedifferentiation that are predictive of melanoma prognosis and response to immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Rachel L Belote
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Daniel Le
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Microchemistry, Proteomics, Lipidomics and Next Generation Sequencing, Genentech Inc, South San Francisco, CA, USA
| | - Ashley Maynard
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ursula E Lang
- Department of Dermatology, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Adriane Sinclair
- Department of Urology and Division of Pediatric Urology, University of California, San Francisco, CA, USA
| | - Brian K Lohman
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Vicente Planells-Palop
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Laurence Baskin
- Department of Urology and Division of Pediatric Urology, University of California, San Francisco, CA, USA
| | - Aaron D Tward
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Spyros Darmanis
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Department of Microchemistry, Proteomics, Lipidomics and Next Generation Sequencing, Genentech Inc, South San Francisco, CA, USA.
| | - Robert L Judson-Torres
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA.
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
40
|
Comprehensive molecular profiling of UV-induced metastatic melanoma in Nme1/Nme2-deficient mice reveals novel markers of survival in human patients. Oncogene 2021; 40:6329-6342. [PMID: 34433909 PMCID: PMC8595820 DOI: 10.1038/s41388-021-01998-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
Hepatocyte growth factor-overexpressing mice that harbor a deletion of the Ink4a/p16 locus (HP mice) form melanomas with low metastatic potential in response to UV irradiation. Here we report that these tumors become highly metastatic following hemizygous deletion of the Nme1 and Nme2 metastasis suppressor genes (HPN mice). Whole genome sequencing of melanomas from HPN mice revealed a striking increase in lung metastatic activity that is associated with missense mutations in eight signature genes (Arhgap35, Atp8b4, Brca1, Ift172, Kif21b, Nckap5, Pcdha2 and Zfp869). RNA-seq analysis of transcriptomes from HP and HPN primary melanomas identified a 32-gene signature (HPN lung metastasis signature) for which decreased expression is strongly associated with lung metastatic potential. Analysis of transcriptome data from The Cancer Genome Atlas revealed expression profiles of these genes that predict improved survival of patients with cutaneous or uveal melanoma. Silencing of three representative HPN lung metastasis signature genes (ARRDC3, NYNRIN, RND3) in human melanoma cells resulted in increased invasive activity, consistent with roles for these genes as mediators of the metastasis suppressor function of NME1 and NME2. In conclusion, our studies have identified a family of genes that mediate suppression of melanoma lung metastasis, and which may serve as prognostic markers and/or therapeutic targets for clinical management of metastatic melanoma.
Collapse
|
41
|
Kruslin B, Gatalica Z, Hes O, Skenderi F, Miettinen M, Contreras E, Xiu J, Ellis M, Florento E, Vranic S, Swensen J. TERT Gene Fusions Characterize a Subset of Metastatic Leydig Cell Tumors. Clin Genitourin Cancer 2021; 19:333-338. [PMID: 33741265 PMCID: PMC9907364 DOI: 10.1016/j.clgc.2021.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Metastatic Leydig cell tumors (LCT) are rare, difficult-to-treat malignancies without known underlying molecular-genetic events. An index case of metastatic LCT showed an LDLR-TERT gene fusion upon routine genetic profiling for detection of therapeutic targets, which was then followed by an investigation into a cohort of additional LCTs. PATIENTS AND METHODS Twenty-nine LCT (27 male and 2 female patients) were profiled using next-generation sequencing and immunohistochemistry. RESULTS TERT gene fusions were detected only in testicular metastatic LCTs, in 3 of 7 successfully analyzed cases (RMST:TERT, LDLR:TERT, and B4GALT5:TERT). TOP1 and CCND3 amplifications were identified in the case with a B4GALT5:TERT fusion. A TP53 mutation was detected in 1 metastatic tumor without a TERT fusion. Five primary (4 testicular and 1 ovarian) LCTs showed multiple gene amplifications, without a consistent pattern. A single metastatic ovarian LCT showed BAP1 mutation and copy number amplifications affecting the NPM1, PCM1, and SS18 genes. At the protein level, 4 of 7 metastatic and 6 of 10 primary testicular LCTs overexpressed Topo1. Androgen receptor was overexpressed in 10 of 13 primary testicular tumors and 2 of 5 metastatic testicular LCTs (without detectable ARv7 messenger RNA or ARv7 protein). Only 1 metastatic testicular LCT exhibited a high tumor mutational burden; all tested cases were microsatellite instability stable and did not express programmed cell death ligand 1. CONCLUSIONS Our study for the first time identified TERT gene fusions as a main genetic alteration and a potential therapeutic target in metastatic LCTs. Topo1 and androgen receptor may guide decisions on chemotherapy and/or hormone therapy for selected individual patients.
Collapse
Affiliation(s)
- Bozo Kruslin
- Clinical Department of Pathology and Cytology “Ljudevit Jurak”, University Hospital Centre “Sestre milosrdnice”, Zagreb, Croatia,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Zoran Gatalica
- Caris Life Sciences, Phoenix, Arizona,Department of Pathology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma
| | - Ondrej Hes
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Pilsen, Czech Republic
| | - Faruk Skenderi
- Department of Pathology, Clinical Center, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Markku Miettinen
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | | | | | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar.
| | | |
Collapse
|
42
|
Eddy K, Chen S. Glutamatergic Signaling a Therapeutic Vulnerability in Melanoma. Cancers (Basel) 2021; 13:3874. [PMID: 34359771 PMCID: PMC8345431 DOI: 10.3390/cancers13153874] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/15/2021] [Accepted: 07/29/2021] [Indexed: 01/03/2023] Open
Abstract
Like other cancers, melanomas are associated with the hyperactivation of two major cell signaling cascades, the MAPK and PI3K/AKT pathways. Both pathways are activated by numerous genes implicated in the development and progression of melanomas such as mutated BRAF, RAS, and NF1. Our lab was the first to identify yet another driver of melanoma, Metabotropic Glutamate Receptor 1 (protein: mGluR1, mouse gene: Grm1, human gene: GRM1), upstream of the MAPK and PI3K/AKT pathways. Binding of glutamate, the natural ligand of mGluR1, activates MAPK and PI3K/AKT pathways and sets in motion the deregulated cellular responses in cell growth, cell survival, and cell metastasis. In this review, we will assess the proposed modes of action that mediate the oncogenic properties of mGluR1 in melanoma and possible application of anti-glutamatergic signaling modulator(s) as therapeutic strategy for the treatment of melanomas.
Collapse
Affiliation(s)
- Kevinn Eddy
- Graduate Program in Cellular and Molecular Pharmacology, School of Graduate Studies, Rutgers University, Piscataway, NJ 08854, USA;
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Graduate Program in Cellular and Molecular Pharmacology, School of Graduate Studies, Rutgers University, Piscataway, NJ 08854, USA;
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Environmental & Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
43
|
Bagaev A, Kotlov N, Nomie K, Svekolkin V, Gafurov A, Isaeva O, Osokin N, Kozlov I, Frenkel F, Gancharova O, Almog N, Tsiper M, Ataullakhanov R, Fowler N. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021; 39:845-865.e7. [PMID: 34019806 DOI: 10.1016/j.ccell.2021.04.014] [Citation(s) in RCA: 549] [Impact Index Per Article: 183.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/14/2020] [Accepted: 04/23/2021] [Indexed: 12/18/2022]
Abstract
The clinical use of molecular targeted therapy is rapidly evolving but has primarily focused on genomic alterations. Transcriptomic analysis offers an opportunity to dissect the complexity of tumors, including the tumor microenvironment (TME), a crucial mediator of cancer progression and therapeutic outcome. TME classification by transcriptomic analysis of >10,000 cancer patients identifies four distinct TME subtypes conserved across 20 different cancers. The TME subtypes correlate with patient response to immunotherapy in multiple cancers, with patients possessing immune-favorable TME subtypes benefiting the most from immunotherapy. Thus, the TME subtypes act as a generalized immunotherapy biomarker across many cancer types due to the inclusion of malignant and microenvironment components. A visual tool integrating transcriptomic and genomic data provides a global tumor portrait, describing the tumor framework, mutational load, immune composition, anti-tumor immunity, and immunosuppressive escape mechanisms. Integrative analyses plus visualization may aid in biomarker discovery and the personalization of therapeutic regimens.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nathan Fowler
- BostonGene, Waltham, MA 02453, USA; Department of Lymphoma and Myeloma, Unit 0429, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
44
|
Yeo PM, Lim ZV, Tan WDV, Zhao X, Chia HY, Tan SH, Teo MCC, Tan MWP. Melanoma in Singapore: A 20-year review of disease and treatment outcomes. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2021; 50:456-466. [PMID: 34195752 DOI: 10.47102/annals-acadmedsg.2020535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Melanomas in Asians have different clinicopathological characteristics and prognosis from melanomas in Caucasians. This study reviewed the epidemiology and treatment outcomes of cutaneous melanoma diagnosed at a tertiary referral dermatology centre in Singapore, which has a multiracial population. The study also determined whether Asians had comparable relapse-free and overall survival periods to Caucasians in Singapore. METHOD This is a retrospective review of cutaneous melanoma cases in our centre between 1996 and 2015. RESULTS Sixty-two cases of melanoma were diagnosed in 61 patients: 72.6% occurred in Chinese, 19.4% in Caucasians and 3.2% in Indians, with an over-representation of Caucasians. Superficial spreading melanoma, acral lentiginous melanoma and nodular melanoma comprised 37.1%, 35.5% and 22.6% of the cases, respectively. The median time interval to diagnosis was longer in Asians than Caucasians; median Breslow's thickness in Asians were significantly thicker than in Caucasians (2.6mm versus 0.9mm, P=0.018) and Asians tend to present at a later stage. The mortality rates for Asians and Caucasians were 52% and 0%, respectively. CONCLUSION More physician and patient education on skin cancer awareness is needed in our Asian-predominant population for better outcomes.
Collapse
|
45
|
Oba J, Woodman SE. The genetic and epigenetic basis of distinct melanoma types. J Dermatol 2021; 48:925-939. [PMID: 34008215 DOI: 10.1111/1346-8138.15957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022]
Abstract
Melanoma represents the deadliest skin cancer. Recent therapeutic developments, including targeted and immune therapies have revolutionized clinical management and improved patient outcome. This progress was achieved by rigorous molecular and functional studies followed by robust clinical trials. The identification of key genomic alterations and gene expression profiles have propelled the understanding of distinct characteristics within melanoma subtypes. The aim of this review is to summarize and highlight the main genetic and epigenetic findings of melanomas and highlight their pathological and therapeutic importance.
Collapse
Affiliation(s)
- Junna Oba
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Scott E Woodman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
46
|
Bobos M. Histopathologic classification and prognostic factors of melanoma: a 2021 update. Ital J Dermatol Venerol 2021; 156:300-321. [PMID: 33982546 DOI: 10.23736/s2784-8671.21.06958-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite the rapid recent advances in molecular analysis of tumors, which allow large-scale and high-resolution genomics, the "gold standard" for melanoma diagnosis continues to be histopathology, in conjunction with clinical characteristics and sometimes with important support of immunohistochemistry. Observations, where postulated that cutaneous melanomas may arise through two distinct pathways, discoveries such as that BRAFV600E mutations were mostly common in melanomas on sun-exposed skin with little solar elastosis and seminal works for melanoma progression and evolution set the groundwork for the new WHO Classification of Melanoma: a classification of melanoma that not only encompasses histologic but also clinical, epidemiologic, and genetic characteristics. The melanomas were divided into those etiologically related to sun exposure and those that are not, based on their mutational signatures, anatomic site, and epidemiology. On the basis of degree of associated solar elastosis melanomas on the sun exposed skin were further divided by the histopathologic degree of cumulative solar damage (CSD) of the surrounding skin, into low and high CSD. On the low-CSD group of melanomas are included superficial spreading melanomas, while the high-CSD melanomas encompasses lentigo maligna and desmoplastic melanomas. The "non-CSD" classification includes acral melanomas, some melanomas in congenital nevi, melanomas in blue nevi, Spitz melanomas, mucosal melanomas, and uveal melanomas. Nodular and nevoid melanoma may occur in any pathway. A group of intermediate tumors termed melanocytoma is proposed for tumors that in addition to mutations that activate the MAPK pathway, harbor multiple driver mutations, and they are either low-grade or high-grade, to indicate that they may carry a higher risk of malignant transformation. In this review a summary of the most recent WHO classification of melanoma is provided. A short analysis of essential histopathologic prognostic parameters is also provided. The new classification of melanoma discriminates distinct types of melanoma based on their clinicopathologic, and genomic characteristics. Undoubtedly, melanoma research will continue to evolve as new clinical, pathological, molecular data accumulates. The challenge of the forthcoming years is to better characterize the intermediate category of melanocytic lesions.
Collapse
Affiliation(s)
- Mattheos Bobos
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Alexandrian Campus, Sindos, Thessaloniki, Greece -
| |
Collapse
|
47
|
Patton EE, Mueller KL, Adams DJ, Anandasabapathy N, Aplin AE, Bertolotto C, Bosenberg M, Ceol CJ, Burd CE, Chi P, Herlyn M, Holmen SL, Karreth FA, Kaufman CK, Khan S, Kobold S, Leucci E, Levy C, Lombard DB, Lund AW, Marie KL, Marine JC, Marais R, McMahon M, Robles-Espinoza CD, Ronai ZA, Samuels Y, Soengas MS, Villanueva J, Weeraratna AT, White RM, Yeh I, Zhu J, Zon LI, Hurlbert MS, Merlino G. Melanoma models for the next generation of therapies. Cancer Cell 2021; 39:610-631. [PMID: 33545064 PMCID: PMC8378471 DOI: 10.1016/j.ccell.2021.01.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.
Collapse
Affiliation(s)
- E Elizabeth Patton
- MRC Human Genetics Unit and Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | - Kristen L Mueller
- Melanoma Research Alliance, 730 15th Street NW, Washington, DC 20005, USA.
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Niroshana Anandasabapathy
- Department of Dermatology, Meyer Cancer Center, Program in Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY 10026, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Corine Bertolotto
- Université Côte d'Azur, Nice, France; INSERM, Biology and Pathologies of Melanocytes, Team 1, Equipe Labellisée Ligue 2020, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Marcus Bosenberg
- Departments of Dermatology, Pathology, and Immunobiology, Yale University, New Haven, CT, USA
| | - Craig J Ceol
- Program in Molecular Medicine and Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology, and Genetics, The Ohio State University, Biomedical Research Tower, Room 918, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Sheri L Holmen
- Department of Surgery, University of Utah Health Sciences Center, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Florian A Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Charles K Kaufman
- Washington University School of Medicine, Department of Medicine, Division of Oncology, Department of Developmental Biology, McDonnell Science Building, 4518 McKinley Avenue, St. Louis, MO 63110, USA
| | - Shaheen Khan
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany; Member of the German Center for Lung Research (DZL), German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, LKI, KU Leuven, 3000 Leuven, Belgium; Trace, Department of Oncology, LKI, KU Leuven, 3000 Leuven, Belgium
| | - Carmit Levy
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - David B Lombard
- Department of Pathology, Institute of Gerontology, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology and Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Kerrie L Marie
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Richard Marais
- CRUK Manchester Institute, The University of Manchester, Alderley Park, Macclesfield SK10 4TG, UK
| | - Martin McMahon
- Department of Dermatology & Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Carla Daniela Robles-Espinoza
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Santiago de Querétaro 76230, Mexico; Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Ze'ev A Ronai
- Cancer Center, Sanford Burnham Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Maria S Soengas
- Spanish National Cancer Research Centre, 28029 Madrid, Spain
| | - Jessie Villanueva
- The Wistar Institute, Molecular and Cellular Oncogenesis Program, Philadelphia, PA, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, and Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Richard M White
- Department of Cancer Biology & Genetics and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iwei Yeh
- Departments of Dermatology and Pathology, University of California, San Francisco, CA, USA
| | - Jiyue Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Marc S Hurlbert
- Melanoma Research Alliance, 730 15th Street NW, Washington, DC 20005, USA
| | - Glenn Merlino
- Center for Cancer Research, NCI, NIH, 37 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
48
|
The Genomic Landscape of Thyroid Cancer Tumourigenesis and Implications for Immunotherapy. Cells 2021; 10:cells10051082. [PMID: 34062862 PMCID: PMC8147376 DOI: 10.3390/cells10051082] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer is the most prevalent endocrine malignancy that comprises mostly indolent differentiated cancers (DTCs) and less frequently aggressive poorly differentiated (PDTC) or anaplastic cancers (ATCs) with high mortality. Utilisation of next-generation sequencing (NGS) and advanced sequencing data analysis can aid in understanding the multi-step progression model in the development of thyroid cancers and their metastatic potential at a molecular level, promoting a targeted approach to further research and development of targeted treatment options including immunotherapy, especially for the aggressive variants. Tumour initiation and progression in thyroid cancer occurs through constitutional activation of the mitogen-activated protein kinase (MAPK) pathway through mutations in BRAF, RAS, mutations in the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) pathway and/or receptor tyrosine kinase fusions/translocations, and other genetic aberrations acquired in a stepwise manner. This review provides a summary of the recent genetic aberrations implicated in the development and progression of thyroid cancer and implications for immunotherapy.
Collapse
|
49
|
Martí JM, Garcia-Diaz A, Delgado-Bellido D, O'Valle F, González-Flores A, Carlevaris O, Rodríguez-Vargas JM, Amé JC, Dantzer F, King GL, Dziedzic K, Berra E, de Álava E, Amaral AT, Hammond EM, Oliver FJ. Selective modulation by PARP-1 of HIF-1α-recruitment to chromatin during hypoxia is required for tumor adaptation to hypoxic conditions. Redox Biol 2021; 41:101885. [PMID: 33581682 PMCID: PMC7878192 DOI: 10.1016/j.redox.2021.101885] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The adaptation to hypoxia is mainly controlled by the HIF transcription factors. Increased expression/activity of HIF-1α correlates with poor prognosis in cancer patients. PARP-1 inhibitors are used in the clinic to treat BRCAness breast/ovarian cancer and have been shown to regulate the hypoxic response; therefore, their use could be expanded. METHODS In this work by integrating molecular/cell biology approaches, genome-wide ChIP-seq, and patient samples, we elucidate the extent to which PARP-1 exerts control over HIF-1-regulated genes. RESULTS In human melanoma, PARP-1 and HIF-1α expression are strongly associated. In response to a hypoxic challenge poly(ADP-ribose) (PAR) is synthesized, HIF-1α is post-transcriptionally modified (PTM) and stabilized by PARylation at specific K/R residues located at its C-terminus. Using an unbiased ChIP-seq approach we demonstrate that PARP-1 dictates hypoxia-dependent HIF-recruitment to chromatin in a range of HIF-regulated genes while analysis of HIF-binding motifs (RCGTG) reveals a restriction on the recognition of hypoxia responsive elements in the absence of PARP-1. Consequently, the cells are poorly adapted to hypoxia, showing a reduced fitness during hypoxic induction. CONCLUSIONS These data characterize the fine-tuning regulation by PARP-1/PARylation of HIF activation and suggest that PARP inhibitors might have therapeutic potential against cancer types displaying HIF-1α over-activation.
Collapse
Affiliation(s)
- Juan Manuel Martí
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, and CIBERONC, 18100, Granada, Spain
| | - Angel Garcia-Diaz
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, and CIBERONC, 18100, Granada, Spain
| | - Daniel Delgado-Bellido
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, and CIBERONC, 18100, Granada, Spain
| | - Francisco O'Valle
- Pathology Department, School of Medicine, IBIMER, CIBM, University of Granada, Spain and Biosanitary Research Institute (IBS. GRANADA), University of Granada, Granada, Spain
| | - Ariannys González-Flores
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, and CIBERONC, 18100, Granada, Spain
| | - Onintza Carlevaris
- CIC BioGUNE, Parque Tecnológico de Bizkaia- Ed. 801A, 48160, Derio, Spain, CIBERONC
| | - José Manuel Rodríguez-Vargas
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire D'Excellence Medalis, UMR7242, Centre National de La Recherche Scientifique/Université de Strasbourg, Institut de Recherche de L'Ecole de Biotechnologie de Strasbourg, Boulevard S. Brant, BP10413, 67412, Illkirch, France
| | - Jean Christophe Amé
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire D'Excellence Medalis, UMR7242, Centre National de La Recherche Scientifique/Université de Strasbourg, Institut de Recherche de L'Ecole de Biotechnologie de Strasbourg, Boulevard S. Brant, BP10413, 67412, Illkirch, France
| | - Françoise Dantzer
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire D'Excellence Medalis, UMR7242, Centre National de La Recherche Scientifique/Université de Strasbourg, Institut de Recherche de L'Ecole de Biotechnologie de Strasbourg, Boulevard S. Brant, BP10413, 67412, Illkirch, France
| | - George L King
- Section of Vascular Cell Biology and Complications, Dianne Nunnally Hoppes Laboratory for Diabetes Complications, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Klaudia Dziedzic
- CIC BioGUNE, Parque Tecnológico de Bizkaia- Ed. 801A, 48160, Derio, Spain, CIBERONC
| | - Edurne Berra
- CIC BioGUNE, Parque Tecnológico de Bizkaia- Ed. 801A, 48160, Derio, Spain, CIBERONC
| | - E de Álava
- Institute of Biomedicine of Sevilla (IBiS), Virgen Del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - A T Amaral
- Institute of Biomedicine of Sevilla (IBiS), Virgen Del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Ester M Hammond
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - F Javier Oliver
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, and CIBERONC, 18100, Granada, Spain.
| |
Collapse
|
50
|
Elefanti L, Zamuner C, Del Fiore P, Stagni C, Pellegrini S, Dall’Olmo L, Fabozzi A, Senetta R, Ribero S, Salmaso R, Mocellin S, Bassetto F, Cavallin F, Tosi AL, Galuppini F, Dei Tos AP, Menin C, Cappellesso R. The Molecular Landscape of Primary Acral Melanoma: A Multicenter Study of the Italian Melanoma Intergroup (IMI). Int J Mol Sci 2021; 22:3826. [PMID: 33917086 PMCID: PMC8067752 DOI: 10.3390/ijms22083826] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022] Open
Abstract
Acral melanoma (AM) is a rare and aggressive subtype of melanoma affecting the palms, soles, and nail apparatus with similar incidence among different ethnicities. AM is unrelated to ultraviolet radiation and has a low mutation burden but frequent chromosomal rearrangements and gene amplifications. Next generation sequencing of 33 genes and somatic copy number variation (CNV) analysis with genome-wide single nucleotide polymorphism arrays were performed in order to molecularly characterize 48 primary AMs of Italian patients in association with clinicopathological and prognostic features. BRAF was the most commonly mutated gene, followed by NRAS and TP53, whereas TERT promoter, KIT, and ARID1A were less frequently mutated. Gains and losses were recurrently found in the 1q, 6p, 7, 8q, 20 and 22 chromosomes involving PREX2, RAC1, KMT2C, BRAF, CCND1, TERT, and AKT3 genes, and in the 6q, 9, 10, 11q and 16q chromosomes including CDKN2A, PTEN, and ADAMTS18 genes, respectively. This study confirmed the variety of gene mutations and the high load of CNV in primary AM. Some genomic alterations were associated with histologic prognostic features. BRAF mutations, found with a higher rate than previously reported, correlated with a low Breslow thickness, low mitotic count, low CNV of the AMs, and with early-stage of disease.
Collapse
Affiliation(s)
- Lisa Elefanti
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (L.E.); (S.P.)
| | - Carolina Zamuner
- Anatomy and Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy;
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (P.D.F.); (L.D.); (S.M.)
| | - Camilla Stagni
- Department of Molecular Medicine, University of Padua, 35128 Padua, Italy;
| | - Stefania Pellegrini
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (L.E.); (S.P.)
| | - Luigi Dall’Olmo
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (P.D.F.); (L.D.); (S.M.)
| | - Alessio Fabozzi
- Oncology Unit 3, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy;
| | - Rebecca Senetta
- Pathology Unit, Department of Oncology, University of Turin, 10124 Turin, Italy;
| | - Simone Ribero
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10124 Turin, Italy;
| | - Roberto Salmaso
- Pathological Anatomy Unit, Padua University Hospital, 35128 Padua, Italy; (R.S.); (A.P.D.T.); (R.C.)
| | - Simone Mocellin
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (P.D.F.); (L.D.); (S.M.)
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, 35128 Padua, Italy
| | - Franco Bassetto
- Plastic Surgery Unit, Padua University Hospital, 35128 Padua, Italy;
- Department of Neurosciences (DNS), University of Padua, 35128 Padua, Italy
| | | | - Anna Lisa Tosi
- Pathological Anatomy Unit, AULSS5, Santa Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Francesca Galuppini
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy;
| | - Angelo Paolo Dei Tos
- Pathological Anatomy Unit, Padua University Hospital, 35128 Padua, Italy; (R.S.); (A.P.D.T.); (R.C.)
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy;
| | - Chiara Menin
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (L.E.); (S.P.)
| | - Rocco Cappellesso
- Pathological Anatomy Unit, Padua University Hospital, 35128 Padua, Italy; (R.S.); (A.P.D.T.); (R.C.)
| |
Collapse
|