1
|
Hojný J, Hrudka J, Prouzová Z, Kendall Bártů M, Krkavcová E, Dvořák J, Michálková R, Čapka D, Zavillová N, Matěj R, Waldauf P. Altered TP53, CDKN2A, ATM, EPHA7, POT1, CHEK1, GRIN2A, and EGFR Predict Shorter Survival in Penile Squamous Cell Carcinoma. Mod Pathol 2025; 38:100689. [PMID: 39694330 DOI: 10.1016/j.modpat.2024.100689] [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: 04/24/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024]
Abstract
Penile squamous cell carcinoma (pSCC) represents an uncommon malignancy characterized by stagnant mortality, psychosexual distress, and a highly variable prognosis. Currently, the World Health Organization distinguishes between human papillomavirus (HPV)-related and HPV-independent pSCC. Recently, there has been an evolving line of research documenting the enrichment of HPV-independent pSCC with a high tumor mutational burden (TMB) and programmed death ligand-1 expression, as well as clusters of genes associated with HPV status. In this study, we conducted comprehensive next-generation sequencing DNA profiling of 146 pSCC samples using a panel consisting of 355 genes associated with tumors. This profiling was correlated with immunohistochemical markers and prognostic clinical data. A survival analysis of recurrent genomic events (found in ≥10 cases) was performed. TP53, CDKN2A, ATM, EPHA7, POT1, CHEK1, GRIN2A, and EGFR alterations were associated with significantly shortened overall survival in univariate and multivariate analysis. HPV positivity, diagnosed through both p16 immunohistochemistry and HPV DNA analysis, displayed no impact on survival but was associated with high-grade, lymphatic invasion, programmed death ligand-1 negativity/weak expression, and low TMB. FAT1, TP53, CDKN2A, CASP8, and HRAS were more often mutated in HPV-independent pSCC. In contrast, HPV-associated pSCCs were enriched by EPHA7, ATM, GRIN2A, and CHEK1 mutations. PIK3CA, FAT1, FBXW7, and KMT2D mutations were associated with high TMB. NOTCH1, TP53, CDKN2A, POT1, KMT2D, ATM, CHEK1, EPHA3, and EGFR alterations were related to adverse clinicopathologic signs, such as advanced stage, high tumor budding, and lymphovascular invasion. We detected 160 alterations with potential treatment implications, with 21.2% of samples showing alterations in the homologous recombination repair pathway. To the best of our knowledge, this study describes the largest cohort of pSCC with complex molecular pathologic, clinical, and prognostic analysis correlating with prognosis.
Collapse
Affiliation(s)
- Jan Hojný
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | - Jan Hrudka
- Department of Pathology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, Czech Republic.
| | - Zuzana Prouzová
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic; Department of Pathology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Michaela Kendall Bártů
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | - Eva Krkavcová
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | - Jiří Dvořák
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | - Romana Michálková
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | - David Čapka
- Department of Urology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Nicolette Zavillová
- Department of Urology, Third Faculty of Medicine of Charles University, Thomayer University Hospital, Prague, Czech Republic
| | - Radoslav Matěj
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic; Department of Pathology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, Czech Republic; Department of Pathology and Molecular Medicine, Third Faculty of Medicine, Charles University, Thomayer University Hospital, Prague, Czech Republic
| | - Petr Waldauf
- Department of Anaesthesia and Intensive Care Medicine, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| |
Collapse
|
2
|
Tsang DA, Tam SYC, Oh CC. Molecular Alterations in Cutaneous Squamous Cell Carcinoma in Immunocompetent and Immunosuppressed Hosts-A Systematic Review. Cancers (Basel) 2023; 15:1832. [PMID: 36980718 PMCID: PMC10046480 DOI: 10.3390/cancers15061832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/26/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
The characterization of cutaneous squamous cell carcinoma (cSCC) at the molecular level is lacking in the current literature due to the high mutational burden of this disease. Immunosuppressed patients afflicted with cSCC experience considerable morbidity and mortality. In this article, we review the molecular profile of cSCC among the immunosuppressed and immunocompetent populations at the genetic, epigenetic, transcriptomic, and proteometabolomic levels, as well as describing key differences in the tumor immune microenvironment between these two populations. We feature novel biomarkers from the recent literature which may serve as potential targets for therapy.
Collapse
Affiliation(s)
- Denise Ann Tsang
- Department of Dermatology, Singapore General Hospital, Singapore 169608, Singapore;
| | - Steve Y. C. Tam
- Education Resource Centre, Singapore General Hospital, Singapore 169608, Singapore
| | - Choon Chiat Oh
- Department of Dermatology, Singapore General Hospital, Singapore 169608, Singapore;
- Duke-NUS Medical School, Singapore 169608, Singapore
| |
Collapse
|
3
|
Vigoda M, Mathieson C, Evans N, Hale C, Jennings J, Lucero O, Jeng S, Bottomly D, Clayburgh D, Andersen P, Li R, Petrisor D, Tyner JW, McWeeney S, Kulesz-Martin M. Functional proteomics of patient derived head and neck squamous cell carcinoma cells reveal novel applications of trametinib. Cancer Biol Ther 2022; 23:310-318. [PMID: 35343367 PMCID: PMC8966983 DOI: 10.1080/15384047.2022.2055420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In this study, we report a differential response of mitogen-activated protein kinase–kinase (MEK) inhibitor trametinib in 20 head and neck squamous cell carcinoma (HNSCC) patients’ tumor-derived cell cultures. Relatively sensitive and resistant cases to trametinib were identified using high throughput metabolic assays and validated in extended dose response studies in vitro. High throughput metabolic assays exploring combination therapies with trametinib were subjected to synergy models and maximal synergistic dose analyses. These yielded several candidates, including axtinib, GDC-0032, GSK-690693, and SGX-523. The combination regimen of trametinib and AXL/MET/VEGFR inhibitor glesatinib showed initial efficacy both in vitro and in vivo (92% reduction in tumor volume). Sensitivity was validated in vivo in a patient-derived xenograft (PDX) model in which trametinib as a single agent effected reduction in tumor volume up to 72%. Reverse Phase Protein Arrays (RPPA) demonstrated differentially expressed proteins and phosphoproteins upon trametinib treatment. Furthermore, resistant cell lines showed a compensatory mechanism via increases in MAPK and non-MAPK pathway proteins that may represent targets for future combination regimens. Intrinsic-targeted options have potential to address paucity of medical treatment options for HNSCC cancer patients, enhance response to extrinsic targeted agents, and/or reduce morbidity as neoadjuvant to surgical treatments.
Collapse
Affiliation(s)
- Myles Vigoda
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Michigan State University College of Osteopathic Medicine, East Lansing, MI, USA
| | - Chase Mathieson
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Nathaniel Evans
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics and Clinical Epidemiolog, Oregon Health & Science University, Portland, OR, USA
| | - Carolyn Hale
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Jennifer Jennings
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Olivia Lucero
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Sophia Jeng
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel Clayburgh
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Peter Andersen
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Ryan Li
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Daniel Petrisor
- Department of Otolaryngology Head and Neck Surgery, Oregon Health & Science University, Operative Care Division, Portland VA Health Care System, Portland, OR, USA
| | - Jeffrey W Tyner
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR, USA
| | - Shannon McWeeney
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics and Clinical Epidemiolog, Oregon Health & Science University, Portland, OR, USA
| | - Molly Kulesz-Martin
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
4
|
Chen X, Yu D, Zhou H, Zhang X, Hu Y, Zhang R, Gao X, Lin M, Guo T, Zhang K. The role of EphA7 in different tumors. Clin Transl Oncol 2022; 24:1274-1289. [PMID: 35112312 DOI: 10.1007/s12094-022-02783-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/18/2022] [Indexed: 12/06/2022]
Abstract
Ephrin receptor A7 (EphA7) is a member of the Eph receptor family. It is widely involved in signal transduction between cells, regulates cell proliferation and differentiation, and participates in developing neural tubes and brain. In addition, EphA7 also has a dual role of tumor promoter and tumor suppressor. It can participate in cell proliferation, migration and apoptosis through various mechanisms, and affect tumor differentiation, staging and prognosis. EphA7 may be a potential diagnostic marker and tumor treatment target. This article reviews the effects of EphA7 on a variety of tumor biological processes and pathological characteristics, as well as specific effects and regulatory mechanisms.
Collapse
Affiliation(s)
- Xiangyi Chen
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Dechen Yu
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Haiyu Zhou
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China. .,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China. .,Xigu District People's Hospital, Lanzhou, 730030, China.
| | - Xiaobo Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Yicun Hu
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Ruihao Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Xidan Gao
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Maoqiang Lin
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Taowen Guo
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| | - Kun Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730030, China.,Gansu Key Laboratory of Bone and Joint Diseases, Lanzhou, 730030, China.,Xigu District People's Hospital, Lanzhou, 730030, China
| |
Collapse
|
5
|
Comprehensive Mutational and Phenotypic Characterization of New Metastatic Cutaneous Squamous Cell Carcinoma Cell Lines Reveal Novel Drug Susceptibilities. Int J Mol Sci 2020; 21:ijms21249536. [PMID: 33333825 PMCID: PMC7765308 DOI: 10.3390/ijms21249536] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a common skin cancer. Most patients who develop metastases (2–5%) present with advanced disease that requires a combination of radical surgery and adjuvant radiation therapy. There are few effective therapies for refractory disease. In this study, we describe novel patient-derived cell lines from cSCC metastases of the head and neck (designated UW-CSCC1 and UW-CSCC2). The cell lines genotypically and phenotypically resembled the original patient tumor and were tumorogenic in mice. Differences in cancer-related gene expression between the tumor and cell lines after various culturing conditions could be largely reversed by xenografting and reculturing. The novel drug susceptibilities of UW-CSCC1 and an irradiated subclone UW-CSCC1-R to drugs targeting cell cycle, PI3K/AKT/mTOR, and DNA damage pathways were observed using high-throughput anti-cancer and kinase-inhibitor compound libraries, which correlate with either copy number variations, targetable mutations and/or the upregulation of gene expression. A secondary screen of top hits in all three cell lines including PIK3CA-targeting drugs supports the utility of targeting the PI3K/AKT/mTOR pathway in this disease. UW-CSCC cell lines are thus useful preclinical models for determining targetable pathways and candidate therapeutics.
Collapse
|