1
|
Alfaraidi M, Gilks CB, Hoang L. Typing of Vulvar Squamous Cell Carcinoma: Why it is Important? Adv Anat Pathol 2025; 32:20-29. [PMID: 39318249 DOI: 10.1097/pap.0000000000000466] [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: 09/26/2024]
Abstract
The classification of vulvar squamous cell carcinoma (VSCC), as in endometrial cancer, has shifted from the histology-based descriptors toward molecular-based identifiers. Recently, it has been reported that there are 3 genetically distinct and clinically significant subtypes of VSCC: HPV-associated VSCC, HPV-independent/p53 wild-type VSCC, and HPV-independent/p53-mutated VSCC. Each group has different prognostic implications as well as response to treatment, thus reinforcing the need for this 3-tier molecular classification. This molecular subtyping can easily be done on vulvar biopsies using p16 and p53 immunohistochemistry stains to further improve risk prediction and individualized treatment decisions, leading to better patient outcomes.
Collapse
Affiliation(s)
- Mona Alfaraidi
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - C Blake Gilks
- Department of Pathology, Prince Sultan Military Medical Hospital, Riyadh, Saudi Arabia
| | - Lynn Hoang
- Department of Pathology, Prince Sultan Military Medical Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Ordi O, Saco A, Peñuelas N, Blanco-Irazuegui O, Pino MD, Carreras-Dieguez N, Marimon L, Rodrigo-Calvo MT, Morató A, Sisuashvili L, Bustamante M, Cruells A, Darecka K, Vega N, Alós S, Trias I, Fusté P, Parra G, Gut M, Munmany M, Torné A, Jares P, Rakislova N. Whole-Exome Sequencing of Vulvar Squamous Cell Carcinomas Reveals an Impaired Prognosis in Patients With TP53 Mutations and Concurrent CCND1 Gains. Mod Pathol 2024; 37:100574. [PMID: 39089654 DOI: 10.1016/j.modpat.2024.100574] [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: 01/23/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
Very little information is available on the mutational landscape of vulvar squamous cell carcinoma (VSCC), a disease that mainly affects older women. Studies focusing on the mutational patterns of the currently recognized etiopathogenic types of this tumor (human papillomavirus [HPV]-associated [HPV-A], HPV-independent [HPV-I] with TP53 mutation [HPV-I/TP53mut], and HPV-I with wild-type TP53 [HPV-I/TP53wt]) are particularly rare, and there is almost no information on the prognostic implications of these abnormalities.Whole-exome DNA sequencing of 60 VSCC and matched normal tissues from each patient was performed. HPV detection, immunohistochemistry (IHC) for p16, p53, and mismatch repair proteins were also performed. Ten tumors (16.7%) were classified as HPV-A, 37 (61.7%) as HPV-I/TP53mut, and 13 (21.6%) as HPV-I/TP53wt. TP53 was the most frequently mutated gene (66.7%), followed by FAT1 (28.3%), CDKN2A (25.0%), RNF213 (23.3%), NFE2L2 (20%) and PIK3CA (20%). All the 60 tumors (100%) were DNA mismatch repair proficient. Seventeen tumors (28.3%) showed CCND1 gain. Bivariate analysis, adjusted for International Federation of Gynecology and Obstetrics stage, revealed that TP53 mutation, CCND1 gain, and the combination of the 2 alterations were strongly associated with impaired recurrence-free survival (hazard ratio, 4.4; P < .001) and disease-specific survival (hazard ratio, 6.1; P = .002). Similar results were obtained when p53 IHC status was used instead of TP53 status and when considering only HPV-I VSCC. However, in the latter category, p53 IHC maintained its prognostic impact only in combination with CCND1 gains. All tumors carried at least one potentially actionable genomic alteration. In conclusion, VSCCs with CCND1 gain represent a prognostically adverse category among HPV-I/TP53mut tumors. All patients with VSCCs are potential candidates for targeted therapy.
Collapse
Affiliation(s)
- Oriol Ordi
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain; Barcelona Institute of Global Health (ISGlobal)-University of Barcelona, Barcelona, Spain
| | - Adela Saco
- Barcelona Institute of Global Health (ISGlobal)-University of Barcelona, Barcelona, Spain; Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Núria Peñuelas
- Barcelona Institute of Global Health (ISGlobal)-University of Barcelona, Barcelona, Spain
| | - Odei Blanco-Irazuegui
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain
| | - Marta Del Pino
- Barcelona Institute of Global Health (ISGlobal)-University of Barcelona, Barcelona, Spain; Department of Obstetrics and Gynecology, Hospital Clínic - University of Barcelona, Barcelona, Spain
| | - Núria Carreras-Dieguez
- Department of Obstetrics and Gynecology, Hospital Clínic - University of Barcelona, Barcelona, Spain
| | - Lorena Marimon
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain; Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | | | - Alba Morató
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain; Barcelona Institute of Global Health (ISGlobal)-University of Barcelona, Barcelona, Spain
| | - Lia Sisuashvili
- Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Mariona Bustamante
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain
| | - Adrià Cruells
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain
| | - Katarzyna Darecka
- Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Naiara Vega
- Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Silvia Alós
- Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Isabel Trias
- Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Pere Fusté
- Department of Obstetrics and Gynecology, Hospital Clínic - University of Barcelona, Barcelona, Spain
| | - Genis Parra
- Centro Nacional de Análisis Genómico, Barcelona, Spain; Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Marta Gut
- Centro Nacional de Análisis Genómico, Barcelona, Spain; Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Meritxell Munmany
- Department of Obstetrics and Gynecology, Hospital Clínic - University of Barcelona, Barcelona, Spain
| | - Aureli Torné
- Department of Obstetrics and Gynecology, Hospital Clínic - University of Barcelona, Barcelona, Spain
| | - Pedro Jares
- Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain
| | - Natalia Rakislova
- Facultat de Medicina i Ciències de la Salut, Department de Fonaments Clinics, Universitat de Barcelona, Barcelona, Spain; Barcelona Institute of Global Health (ISGlobal)-University of Barcelona, Barcelona, Spain; Department of Pathology, Hospital Clínic of Barcelona-University of Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Catalán-Castorena O, Garibay-Cerdenares OL, Illades-Aguiar B, Rodríguez-Ruiz HA, Zubillaga-Guerrero MI, Leyva-Vázquez MA, Encarnación-Guevara S, Alarcón-Romero LDC. The role of HR-HPV integration in the progression of premalignant lesions into different cancer types. Heliyon 2024; 10:e34999. [PMID: 39170128 PMCID: PMC11336306 DOI: 10.1016/j.heliyon.2024.e34999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024] Open
Abstract
High-risk human papillomavirus (HR-HPV) is associated with the development of different types of cancer, such as cervical, head and neck (including oral, laryngeal, and oropharyngeal), vulvar, vaginal, penile, and anal cancers. The progression of premalignant lesions to cancer depends on factors associated with the host cell and the different epithelia infected by HPV, such as basal cells of the flat epithelium and the cells of the squamocolumnar transformation zone (STZ) found in the uterine cervix and the anal canal, which is rich in heparan sulfate proteoglycans and integrin-like receptors. On the other hand, factors associated with the viral genotype, infection with multiple viruses, viral load, viral persistence, and type of integration determine the viral breakage pattern and the sites at which the virus integrates into the host cell genome (introns, exons, intergenic regions), inducing the loss of function of tumor suppressor genes and increasing oncogene expression. This review describes the role of viral integration and the molecular mechanisms induced by HR-HPV in different types of tissues. The purpose of this review is to identify the common factors associated with the role of integration events in the progression of premalignant lesions in different types of cancer.
Collapse
Affiliation(s)
- Oscar Catalán-Castorena
- Research in Cytopathology and Histochemical Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Olga Lilia Garibay-Cerdenares
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
- CONAHCyT-Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Berenice Illades-Aguiar
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Hugo Alberto Rodríguez-Ruiz
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Ma. Isabel Zubillaga-Guerrero
- Research in Cytopathology and Histochemical Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | - Marco Antonio Leyva-Vázquez
- Molecular Biomedicine Laboratory, Faculty of Chemical-Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| | | | - Luz del Carmen Alarcón-Romero
- Research in Cytopathology and Histochemical Laboratory, Faculty of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, 39089, Mexico
| |
Collapse
|
4
|
Williams KJ. Eradicating Atherosclerotic Events by Targeting Early Subclinical Disease: It Is Time to Retire the Therapeutic Paradigm of Too Much, Too Late. Arterioscler Thromb Vasc Biol 2024; 44:48-64. [PMID: 37970716 DOI: 10.1161/atvbaha.123.320065] [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] [Indexed: 11/17/2023]
Abstract
Recent decades have seen spectacular advances in understanding and managing atherosclerotic cardiovascular disease, but paradoxically, clinical progress has stalled. Residual risk of atherosclerotic cardiovascular disease events is particularly vexing, given recognized lifestyle interventions and powerful modern medications. Why? Atherosclerosis begins early in life, yet clinical trials and mechanistic studies often emphasize terminal, end-stage plaques, meaning on the verge of causing heart attacks and strokes. Thus, current clinical evidence drives us to emphasize aggressive treatments that are delayed until patients already have advanced arterial disease. I call this paradigm "too much, too late." This brief review covers exciting efforts that focus on preventing, or finding and treating, arterial disease before its end-stage. Also included are specific proposals to establish a new evidence base that could justify intensive short-term interventions (induction-phase therapy) to treat subclinical plaques that are early enough perhaps to heal. If we can establish that such plaques are actionable, then broad screening to find them in early midlife individuals would become imperative-and achievable. You have a lump in your coronaries! can motivate patients and clinicians. We must stop thinking of a heart attack as a disease. The real disease is atherosclerosis. In my opinion, an atherosclerotic heart attack is a medical failure. It is a manifestation of longstanding arterial disease that we had allowed to progress to its end-stage, despite knowing that atherosclerosis begins early in life and despite the availability of remarkably safe and highly effective therapies. The field needs a transformational advance to shift the paradigm out of end-stage management and into early interventions that hold the possibility of eradicating the clinical burden of atherosclerotic cardiovascular disease, currently the biggest killer in the world. We urgently need a new evidence base to redirect our main focus from terminal, end-stage atherosclerosis to earlier, and likely reversible, human arterial disease.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Department of Cardiovascular Sciences, Department of Medicine, Lewis Katz School of Medicine at Temple University, PA
| |
Collapse
|
5
|
Corey L, Wallbillich JJ, Wu S, Farrell A, Hodges K, Xiu J, Nabhan C, Guastella A, Kheil M, Gogoi R, Winer I, Bandyopadhyay S, Huang M, Jones N, Wilhite A, Karnezis A, Thaker P, Herzog TJ, Oberley M, Korn WM, Vezina A, Morris R, Ali-Fehmi R. The Genomic Landscape of Vulvar Squamous Cell Carcinoma. Int J Gynecol Pathol 2023; 42:515-522. [PMID: 37131274 PMCID: PMC10417246 DOI: 10.1097/pgp.0000000000000950] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Vulvar squamous cell cancer (VSC) accounts for 90% of vulvar cancers. Next-generation sequencing studies of VSC imply human papillomavirus (HPV) and p53 status play separate roles in carcinogenesis and prognosis. We sought to describe the genomic landscape and analyze the immunologic profiles of VSC with respect to HPV and p53 status. A total of 443 VSC tumors underwent tumor profiling. Next-generation sequencing was performed on genomic DNA isolated from formalin-fixed paraffin-embedded tumor samples. PD-L1, microsatellite instability were tested by fragment analysis, IHC, and next-generation sequencing. Tumor mutational burden-high was defined as >10 mutations per MB. HPV 16/18 positive (HPV+) status was determined using whole exome sequencing on 105 samples. Three cohorts were identified from 105 samples with known HPV: HPV+, HPV-/p53wt, and HPV-/p53mt. Where HPV and p53 status were examined, TP53 mutations were exclusive of HPV+ tumors. In all, 37% of samples were HPV+. Among the 66 HPV- tumors, 52 (78.8%) were HPV-/p53mt and 14 (21.2%) were HPV-/p53wt. The HPV-/p53wt cohort had a higher rate of mutations in the PI3KCA gene (42.9% HPV-/p53wt vs 26.3% HPV+ vs. 5.8% HPV-/p53mt, q =0.028) and alterations in the PI3K/AkT/mTOR pathway (57.1% HPV-/p53wt vs. 34.2% HPV+ vs. 7.7% HPV-/p53mt, q =0.0386) than the other 2 cohorts. Ninety-eight VSC tumors with HPV16/18 information underwent transcriptomic analysis and immune deconvolution method. No differences were observed in immune profiles. The HPV-/p53wt VSC tumors had significantly higher rates of mutations in the PI3KCA gene and alterations in the PI3K/AkT/mTOR pathway, a potential target that merits further investigation in this subgroup.
Collapse
|
6
|
Leung E, Tremblay C, Liao D, Burnett M, Huang L, Sun SZ, Ko JJ. Treatment patterns and outcomes of patients with locally advanced vulvar or vaginal cancer in British Columbia. Gynecol Oncol 2023; 175:107-113. [PMID: 37348429 DOI: 10.1016/j.ygyno.2023.06.013] [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: 04/20/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
OBJECTIVE As vulvar and vaginal cancers are rare malignancies, treatment is extrapolated from the cervical cancer field. Further studies are necessary to evaluate whether surgery, radiotherapy (RT), or combined chemoRT is most beneficial. METHODS A retrospective chart review was conducted on patients diagnosed with vulvar or vaginal cancer in 2000-2017. Descriptive statistics was used to summarize demographic factors. Kaplan-Meier curves, log-rank tests, multivariate analysis with hazard ratios (HR) were conducted to compare survival outcomes, including overall survival (OS), disease-free survival, and cancer-specific survival, between surgery, RT, and chemoRT. RESULTS This study included 688 patients with either vulvar (n = 560, 81%) or vaginal cancer (n = 128, 19%). Median age of diagnosis was 68 (27-98) years. In multivariate survival analysis, vulvar cancer was associated with more likelihood of death (HR: 1.50, p = 0.042) compared to vaginal cancer. For patients who received definitive RT, median OS was 63.8 months with concurrent chemotherapy vs. 46.3 months without for vulvar cancer (p = 0.75); for vaginal, median OS 100.4 with chemotherapy vs. 66.6 months without (p = 0.31). For vulvar cancer patients who received RT (n = 224), adding chemotherapy (n = 100) was not associated with statistically significant OS improvement (HR: 0.989, p = 0.957). Similarly, vaginal cancer patients who received chemoRT (n = 51) did not have significant OS benefit (HR: 0.720, p = 0.331) over patients who received RT (n = 49). CONCLUSIONS In this retrospective study, chemoRT was not associated with significant improvements in survival compared to RT in vulvar or vaginal cancer. Future studies investigating novel therapies to treat these cancers are needed to improve patient outcomes.
Collapse
Affiliation(s)
- Emily Leung
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cassia Tremblay
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Donna Liao
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Madalon Burnett
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Longlong Huang
- Department of Mathematics and Statistics, University of the Fraser Valley, Abbotsford, BC V2S 7M8, Canada
| | - Shaun Z Sun
- Department of Mathematics and Statistics, University of the Fraser Valley, Abbotsford, BC V2S 7M8, Canada; Department of Mathematics, Vancouver Island University, Nanaimo, BC V9R 5S5, Canada
| | - Jenny J Ko
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Medical Oncology, BC Cancer - Abbotsford, Abbotsford, BC V2S 0CS, Canada.
| |
Collapse
|
7
|
Precursors, pathways of carcinogenesis and molecular markers of vulvar squamous cell carcinoma. Literature review. ACTA BIOMEDICA SCIENTIFICA 2023. [DOI: 10.29413/abs.2023-8.1.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023] Open
Abstract
The review analyzes and summarizes the results of the studies on the pathogenesis of vulvar squamous cell carcinoma and its diagnostic features, reviews precursors and molecular subtypes of carcinomas. Despite the relatively low incidence of this tumor, over the past few decades, there has been an upward trend in its incidence, including the incidence among young women. According to the latest World Health Organization classification of lower genital tumors from 2020, vulvar squamous cell carcinoma is divided into human papillomavirus (HPV) associated and HPV-independent. While these carcinomas are often morphologically similar, their mechanisms of carcinogenesis, precursors, and clinical outcomes are different. Just the detection of virus DNA in a tumor is not enough to establish HPV status. Meanwhile immunohistochemical detection of the expression of p16 and p53 proteins allows not only to separate two pathogenetic pathways of carcinogenesis, but also to identify its molecular subtypes. The data on the possible use of p16 and p53 expression as the disease prognosis molecular markers have been obtained. Currently, the tactics of treatment and monitoring patients does not depend on the HPV status of carcinoma; however, the results of recent studies suggest that women with HPV positive vulvar cancer have significantly higher survival rates and a lower risk of recurrence. Understanding the mechanisms of carcinogenesis and improving its diagnosis will advance the assessment of the individual risk of the progression of precancerous lesions, as well as the outcome and the occurrence of tumor recurrence.
Collapse
|
8
|
Della Corte L, Cafasso V, Boccia D, Morra I, De Angelis C, De Placido S, Giampaolino P, Di Carlo C, Bifulco G. How SARS-CoV-2 Infection Impacts the Management of Patients with Vulvar Cancer: Experience in a Third-Level Hospital of Southern Italy. J Pers Med 2023; 13:jpm13020240. [PMID: 36836474 PMCID: PMC9967907 DOI: 10.3390/jpm13020240] [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: 11/24/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Background: Since February 2020, the spread of Coronavirus Disease 2019 (COVID-19) in Italy has induced the government to call for lockdown of any activity apart from primary needs, and changing the lives of each of us. All that has dramatically impacted the management of patients affected by cancer. Patients with vulvar cancer (VC) represent a particularly frail population because they are elderly and affected by multiple comorbidities. The aim of this study is to evaluate the clinical impact of the SARS-CoV-2 infection on VC patients in terms of delay or impossibility of carrying out the scheduled treatment. Methods: The medical records of patients affected by vulvar tumors, referred to "DAI Materno-Infantile" of AOU Federico II of Naples between February 2020 and January 2022 were retrospectively analyzed. The presence of a positive reverse transcription-polymerase chain reaction (RT-PCR) in nasopharyngeal swab defined the positivity to SARS-CoV-2. Results: Twenty-four patients with VC were analyzed and scheduled for treatment. The median age was 70.7 years (range: 59-80). Seven (29.2%) patients were diagnosed with SARS-CoV-2 infection: In three (42.8%) patients, the treatment was delayed with no apparent consequences, in four (57.2%), the treatment was delayed or changed due to cancer progression and, of these four, one died due to respiratory complications of COVID-19, and one died due to oncologic disease progression. Conclusion: COVID-19 caused, in most cases, significant delays in oncologic treatments and high mortality in our series of patients affected by VC.
Collapse
Affiliation(s)
- Luigi Della Corte
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 84014 Naples, Italy
- Correspondence:
| | - Valeria Cafasso
- Department of Public Health, School of Medicine, University of Naples Federico II, 84014 Napoli, Italy
| | - Dominga Boccia
- Department of Public Health, School of Medicine, University of Naples Federico II, 84014 Napoli, Italy
| | - Ilaria Morra
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 84014 Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 84014 Naples, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 84014 Naples, Italy
| | - Pierluigi Giampaolino
- Department of Public Health, School of Medicine, University of Naples Federico II, 84014 Napoli, Italy
| | - Costantino Di Carlo
- Department of Public Health, School of Medicine, University of Naples Federico II, 84014 Napoli, Italy
| | - Giuseppe Bifulco
- Department of Public Health, School of Medicine, University of Naples Federico II, 84014 Napoli, Italy
| |
Collapse
|
9
|
Avila M, Grinsfelder MO, Pham M, Westin SN. Targeting the PI3K Pathway in Gynecologic Malignancies. Curr Oncol Rep 2022; 24:1669-1676. [PMID: 36401704 PMCID: PMC10862662 DOI: 10.1007/s11912-022-01326-9] [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] [Accepted: 07/05/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE OF REVIEW This review explores the PI3K pathway aberrations common in gynecologic malignancies, the relevant therapeutic targets that have been explored to date particularly given their success in endometrial cancers, and predictive biomarkers of response to therapy. RECENT FINDINGS Landmark trials have been noted involving this pathway, particularly in endometrial cancers. One phase II trial of the potent orally bioavailable mTOR inhibitor, everolimus, in combination with letrozole demonstrated an unprecedented clinical benefit rate (CBR) of 40% and high objective response rate (RR) of 32% in hormone agnostic endometrial cancers. This was followed by GOG 3007 that compared everolimus and letrozole to hormonal therapy yielding similar response rates but double progression-free survival rates. The phosphoinositide 3-kinase (PI3K) signaling pathway is implicated in tumorigenesis given its regulation over cell growth, cellular trafficking, and angiogenesis. In gynecologic malignancies, alterations in PI3K signaling are common. Therefore, developing modulators of the PI3K pathway and identifying molecular markers to predict response are of great interest for these cancer types.
Collapse
Affiliation(s)
- Monica Avila
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA
| | - Michaela Onstad Grinsfelder
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA
| | - Melissa Pham
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3279, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Porter VL, Marra MA. The Drivers, Mechanisms, and Consequences of Genome Instability in HPV-Driven Cancers. Cancers (Basel) 2022; 14:4623. [PMID: 36230545 PMCID: PMC9564061 DOI: 10.3390/cancers14194623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022] Open
Abstract
Human papillomavirus (HPV) is the causative driver of cervical cancer and a contributing risk factor of head and neck cancer and several anogenital cancers. HPV's ability to induce genome instability contributes to its oncogenicity. HPV genes can induce genome instability in several ways, including modulating the cell cycle to favour proliferation, interacting with DNA damage repair pathways to bring high-fidelity repair pathways to viral episomes and away from the host genome, inducing DNA-damaging oxidative stress, and altering the length of telomeres. In addition, the presence of a chronic viral infection can lead to immune responses that also cause genome instability of the infected tissue. The HPV genome can become integrated into the host genome during HPV-induced tumorigenesis. Viral integration requires double-stranded breaks on the DNA; therefore, regions around the integration event are prone to structural alterations and themselves are targets of genome instability. In this review, we present the mechanisms by which HPV-dependent and -independent genome instability is initiated and maintained in HPV-driven cancers, both across the genome and at regions of HPV integration.
Collapse
Affiliation(s)
- Vanessa L. Porter
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
11
|
Wei KX, Hoang LN. Squamous and Glandular Lesions of the Vulva and Vagina: What's New and What Remains Unanswered? Surg Pathol Clin 2022; 15:389-405. [PMID: 35715167 DOI: 10.1016/j.path.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A number of changes have been introduced into the 5th Edition of the World Health Organization (WHO) Classification of squamous and glandular neoplasms of the vulva and vagina. This review highlights the major shifts in tumor classification, new entities that have been introduced, recommendations for p16 immunohistochemical testing, biomarker use, molecular findings and practical points for pathologists which will affect clinical care. It also touches upon several issues that still remain answered in these rare but undeniably important women's cancers.
Collapse
Affiliation(s)
- Kelly X Wei
- MD Undergraduate Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lynn N Hoang
- Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
12
|
Williams EA, Montesion M, Lincoln V, Tse JY, Hiemenz MC, Mata DA, Shah BB, Shoroye A, Alexander BM, Werth AJ, Foley-Peres K, Milante RR, Ross JS, Ramkissoon SH, Williams KJ, Adhikari LJ, Zuna RE, LeBoit PE, Lin DI, Elvin JA. HPV51-associated Leiomyosarcoma: A Novel Class of TP53/RB1-Wildtype Tumor With Predilection for the Female Lower Reproductive Tract. Am J Surg Pathol 2022; 46:729-741. [PMID: 35034043 PMCID: PMC9093731 DOI: 10.1097/pas.0000000000001862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Inactivating mutations in tumor suppressor genes TP53 and RB1 are considered central drivers in leiomyosarcomas (LMSs). In high-risk human papillomavirus (HPV)-related tumors, a similar functional outcome is achieved through oncoproteins E6 and E7, which inactivate the p53 and RB1 proteins, respectively. Here, we hypothesized that HPV infection could provide an alternative mechanism for tumorigenesis in a subset of TP53/RB1-wildtype LMS. We evaluated tumor samples from 2585 consecutive unique patients carrying a diagnosis of gynecologic or soft tissue LMS. Tumor DNA and available RNA were analyzed by hybrid-capture-based next-generation sequencing/comprehensive genomic profiling of 406 genes and transcripts (FoundationOneHeme). Of the initial 2585 cases, we excluded 16 based on the presence of molecular alterations that are considered defining for sarcomas other than LMS. In the remaining 2569 cases, we searched for LMS that were TP53/RB1-wildtype (n=486 of 2569; 18.9%). We also searched LMS tumors for HPV sequences that we then classified into genotypes by de novo assembly of nonhuman sequencing reads followed by alignment to the RefSeq database. Among TP53/RB1-wildtype LMS, we identified 18 unique cases harboring HPV sequences. Surprisingly, most (n=11) were HPV51-positive, and these 11 represented all HPV51-positive tumors in our entire LMS database (n=11 of 2569; 0.4%). The absence of genomic alterations in TP53 or RB1 in HPV51-positive LMS represented a marked difference from HPV51-negative LMS (n=2558; 0% vs. 72% [P<0.00001], 0% vs. 53% [P=0.0002]). In addition, compared with HPV51-negative LMS, HPV51-positive LMS were significantly enriched for genomic alterations in ATRX (55% vs. 24%, P=0.027) and TSC1 (18% vs. 0.6%, P=0.0047). All HPV51-positive LMS were in women; median age was 54 years at surgery (range: 23 to 74 y). All known primary sites were from the gynecologic tract or adjacent anogenital area, including 5 cases of vaginal primary site. Histology was heterogeneous, with evaluable cases showing predominant epithelioid (n=5) and spindle (n=5) morphology. In situ hybridization confirmed the presence of high-risk HPV E6/E7 mRNA in tumor cells in three of three evaluable cases harboring HPV51 genomic sequences. Overall, in our pan-LMS analysis, HPV reads were identified in a subset of TP53/RB1-wildtype LMS. For all HPV51-associated LMS, the striking absence of any detectable TP53 or RB1 mutations and predilection for the female lower reproductive tract supports our hypothesis that high-risk HPV can be an alternative tumorigenic mechanism in this distinct class of LMS.
Collapse
Affiliation(s)
- Erik A. Williams
- Departments of Pathology and Dermatology, UCSF Dermatopathology Service, Helen Diller Family Cancer Center, University of California, San Francisco, CA
- Foundation Medicine Inc., Cambridge
| | | | - Vadim Lincoln
- Departments of Pathology and Dermatology, UCSF Dermatopathology Service, Helen Diller Family Cancer Center, University of California, San Francisco, CA
| | | | | | | | | | | | | | - Adrienne J. Werth
- Department of Women’s Health Services, Hartford Hospital, Hartford, CT
| | | | - Riza R. Milante
- Department of Dermatology, Jose R. Reyes Memorial Medical Center, Manila, Philippines
| | - Jeffrey S. Ross
- Foundation Medicine Inc., Cambridge
- Department of Pathology, State University of New York Upstate Medical University, Syracuse, NY
| | - Shakti H. Ramkissoon
- Foundation Medicine Inc., Cambridge
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Kevin Jon Williams
- Departments of Physiology and Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Laura J. Adhikari
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Rosemary E. Zuna
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Philip E. LeBoit
- Departments of Pathology and Dermatology, UCSF Dermatopathology Service, Helen Diller Family Cancer Center, University of California, San Francisco, CA
| | | | | |
Collapse
|
13
|
Salama AM, Momeni-Boroujeni A, Vanderbilt C, Ladanyi M, Soslow R. Molecular landscape of vulvovaginal squamous cell carcinoma: new insights into molecular mechanisms of HPV-associated and HPV-independent squamous cell carcinoma. Mod Pathol 2022; 35:274-282. [PMID: 34650187 PMCID: PMC9450957 DOI: 10.1038/s41379-021-00942-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 02/03/2023]
Abstract
Squamous cell carcinomas of the lower female genital tract may be human papillomavirus-associated or independent. We studied the HPV status, mutational repertoire, histology, and clinical data of 28 samples from 26 patients, 65% with a vulvar primary and 35% with a vaginal primary. These represented invasive vulvovaginal squamous cell carcinomas that underwent clinical tumor-normal targeted massively parallel sequencing analysis. HPV status was determined using the HPV high-risk RNA ISH assay and/or by MSK-IMPACT. Eleven patients had HPV-associated squamous cell carcinoma (four vulvar and seven vaginal) and 15 patients had HPV-independent SqCC (13 vulvar and 2 vaginal). Well-differentiated squamous cell carcinomas were always HPV-independent. HPV-independent moderately and poorly differentiated carcinomas frequently had alterations in the NOTCH signaling pathway (6/7), which were also associated with increased tumor budding (P: 0.002). HPV-associated vulvovaginal squamous cell carcinoma had PIK3CA activating mutations (7/11, 64%) as the most common genomic event, while TERT gene alterations, mainly TERT promoter mutations (14/15 cases, 93%) featured significantly in HPV-independent carcinomas. Other common abnormalities in HPV-independent tumors were TP53 mutations (13/15, 87%), CDKN2A alterations (10/15, 67%), and NOTCH1 and FAT1 mutations (7/15, 47% each). A subset of both HPV-associated and -independent tumors had NOTCH pathway alterations (6/11, 55% and 10/15, 67% respectively), but different genes in this pathway were altered in these tumors. In summary, TERT, TP53, CDKN2A, and NOTCH1 gene alterations strongly point away from an HPV-driven process (odds ratios: 0.01, 0.07, 0, and 0, respectively with p values < 0.02 for all four genes), while PIK3CA activating mutations without the other mutations strongly favors an HPV-driven tumor (odds ratio: 10.12, p value: 0.016). HPV-independent carcinomas are more likely to be moderately-poorly differentiated with intermediate to high tumor cell budding. Cancer cell fraction analysis of HPV-independent squamous carcinomas suggests that TERT and/or NOTCH1 alterations along with TP53 alterations can be the initiating event in these tumors.
Collapse
Affiliation(s)
- Abeer M. Salama
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Chad Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Soslow
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
14
|
Garganese G, Inzani F, Fragomeni SM, Mantovani G, Della Corte L, Piermattei A, Santoro A, Angelico G, Giacò L, Corrado G, Fagotti A, Zannoni GF, Scambia G. The Vulvar Immunohistochemical Panel (VIP) Project: Molecular Profiles of Vulvar Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13246373. [PMID: 34944993 PMCID: PMC8699435 DOI: 10.3390/cancers13246373] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/25/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary This study investigated the immunohistochemical expression of 14 biological markers as potential prognostic/therapeutic factors in vulvar squamous cell carcinoma, comparing 53 node-negative (Group A) and 48 node-positive (Group B) patients. Our results show a significantly higher p16 expression (surrogate of HPV-related tumors) in the vulvar samples of non-metastatic patients. In Group B, PD-L1 positivity and high EGFR expression were found in the vast majority of vulvar and/or nodal specimens. VEGF showed strong/moderate-diffuse expression in almost 14% of all vulvar samples. A mutated p53 and over-expressed PD-L1 showed a significant association with nodal metastasis. Our results support a potential role of immune checkpoint inhibitors and anti-VEGF and anti-EGFR drugs, especially in patients with worse prognosis (metastatic, HPV-independent). A panel including EGFR, VEGF, PDL1, p16, and p53 might be performed routinely in primary tumor and repeated in case of lymph node metastases to identify changes in marker expression. Abstract Introduction: The study’s aim was to investigate the immunohistochemical (IHC) expression of biological markers as potential prognostic/therapeutic factors in vulvar squamous cell carcinoma (VSCC). Methodology: A series of 101 patients surgically treated at our center from 2016 to 2020 were retrospectively enrolled: 53 node-negative (Group A) and 48 node-positive (Group B). A total of 146 samples, 101 from primary tumor (T) and 45 from nodal metastases (N), were investigated. The IHC panel included: p16, p53, MLH1, MSH2, MSH6, PMS2, PD-L1, CD3, HER2/neu, ER, PR, EGFR, VEGF, and CD31. The reactions were evaluated on qualitative and semi-quantitative scales. Generalized Linear Model (GLM) and cluster analysis were performed in R statistical environment. A distance plot compared the IHC panel of T with the correspondent N. Results: In Group A: p16-positive expression (surrogate of HPV-dependent pathway) was significantly higher (20.8% vs. 6.2%, p = 0.04). In Group B: PD-L1 positivity and high EGFR expression were found, respectively, in 77.1% and 97.9% patients (T and/or N). Overall, p16-negative tumors showed a higher PD-L1 expression (60.9% vs. 50.0%). In both groups: tumoral immune infiltration (CD3 expression) was mainly moderate/intense (80% vs. 95%); VEGF showed strong/moderate-diffuse expression in 13.9% of T samples; CD31, related to tumoral microvessel density (MVD), showed no difference between groups; a mutated p53 and over-expressed PD-L1 showed significant association with nodal metastasis, with Odds Ratios (OR) of 4.26 (CI 95% = 1.14–15.87, p = 0.03) and 2.68 (CI 95% = 1.0–7.19, p < 0.05), respectively; since all mismatch repair proteins (MMR) showed a retained expression and ER, PR, and HER2/neu were negative, they were excluded from further analysis. The cluster analysis identified three and four sub-groups of molecular profiles, respectively, in Group A and B, with no difference in prognosis. The molecular signature of each N and corresponding T diverged significantly in 18/41 (43.9%) cases. Conclusions: Our results support a potential role of immune checkpoint inhibitors and anti-VEGF and anti-EGFR drugs especially in patients with worse prognosis (metastatic, HPV-independent). A panel including EGFR, VEGF, PDL1, p16, and p53 might be performed routinely in primary tumor and repeated in case of lymph node metastases to identify changes in marker expression.
Collapse
Affiliation(s)
- Giorgia Garganese
- Dipartimento Scienze della Vita e Sanità Pubblica, Sezione Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.G.); (A.F.); (G.S.)
- Gynecology and Breast Care Center, Mater Olbia Hospital, 07026 Olbia, Italy
| | - Frediano Inzani
- Unità di Gineco-Patologia e Patologia Mammaria, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.P.); (A.S.); (G.A.); (G.F.Z.)
| | - Simona Maria Fragomeni
- Unità di Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (S.M.F.); (L.D.C.); (G.C.)
| | - Giulia Mantovani
- Department of Obstetrics and Gynaecology, Gynaecologic Oncology and Minimally-Invasive Pelvic Surgery, International School of Surgical Anatomy, IRCCS Sacro Cuore-Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
- Correspondence:
| | - Luigi Della Corte
- Unità di Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (S.M.F.); (L.D.C.); (G.C.)
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Alessia Piermattei
- Unità di Gineco-Patologia e Patologia Mammaria, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.P.); (A.S.); (G.A.); (G.F.Z.)
| | - Angela Santoro
- Unità di Gineco-Patologia e Patologia Mammaria, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.P.); (A.S.); (G.A.); (G.F.Z.)
| | - Giuseppe Angelico
- Unità di Gineco-Patologia e Patologia Mammaria, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.P.); (A.S.); (G.A.); (G.F.Z.)
| | - Luciano Giacò
- Bioinformatics Facility Core Research, Gemelli Science and Technology Park, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Giacomo Corrado
- Unità di Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (S.M.F.); (L.D.C.); (G.C.)
| | - Anna Fagotti
- Dipartimento Scienze della Vita e Sanità Pubblica, Sezione Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.G.); (A.F.); (G.S.)
- Unità di Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (S.M.F.); (L.D.C.); (G.C.)
| | - Gian Franco Zannoni
- Unità di Gineco-Patologia e Patologia Mammaria, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.P.); (A.S.); (G.A.); (G.F.Z.)
- Dipartimento Scienze della Vita e Sanità Pubblica, Sezione Anatomia Patologica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giovanni Scambia
- Dipartimento Scienze della Vita e Sanità Pubblica, Sezione Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.G.); (A.F.); (G.S.)
- Unità di Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (S.M.F.); (L.D.C.); (G.C.)
| |
Collapse
|
15
|
Transcriptome Analysis in Vulvar Squamous Cell Cancer. Cancers (Basel) 2021; 13:cancers13246372. [PMID: 34944992 PMCID: PMC8699756 DOI: 10.3390/cancers13246372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary The number of women, especially younger women, diagnosed with vulvar cancer, has been rising mainly due to the infection with human papilloma virus (HPV) over the last years. In contrast to other tumor entities, limited information on the underlying genetic changes is available, and thus treatment advances, especially the development of personalized treatments, are hampered. We aimed to explore the RNA expression profiles in a group of 24 vulvar cancer samples in order to detect potential prognostic markers and therapeutic targets in order to establish to a more profound understanding of vulvar cancer carcinogenesis. Abstract To date, therapeutic strategies in vulvar squamous cell carcinoma (VSCC) are lacking molecular pathological information and targeted therapy hasn’t been approved in the treatment of VSCC, yet. Two etiological pathways are widely accepted: HPV induced vs. HPV independent, associated with chronic skin disease, often harboring TP53 mutations (mut). The aim of this analysis was to analyze the RNA expression patterns for subtype stratification on VSCC samples that can be integrated into the previously performed whole exome sequencing data for the detection of prognostic markers and potential therapeutic targets. We performed multiplex gene expression analysis (NanoString) with 770 genes in 24 prior next generation sequenced samples. An integrative data analysis was performed. Here, 98 genes were differentially expressed in TP53mut vs. HPV+ VSCC, in the TP53mut cohort, where 56 genes were upregulated and 42 were downregulated in comparison to the HPV+ tumors. Aberrant expression was primarily observed in cell cycle regulation, especially in HPV+ disease. Within the TP53mut group, a distinct cluster was identified that was correlated to a significantly worse overall survival (p = 0.017). The RNA expression profiles showed distinct patterns with regard to the known VSCC subtypes and could potentially enable further subclassification in the TP53mut groups
Collapse
|
16
|
Next-Generation Sequencing Whole-Genome Analysis for Targeted Treatment Approach of Metastatic Bartholin Gland Adenocarcinoma: An Emblematic Case Report and Review of the Literature. Diagnostics (Basel) 2021; 11:diagnostics11112085. [PMID: 34829431 PMCID: PMC8624694 DOI: 10.3390/diagnostics11112085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
Bartholin gland adenocarcinoma (BGA) is extremely rare and is characterized by high rates of lymph-node recurrence and distant metastases. No effective palliative treatments are available for metastatic BGA; therefore, advanced BGA remains a challenge for gynecologic oncologists. Considering the rarity of this disease and the lack of a standardized approach, the present study aims to discuss the available literature on current therapies for BGA and to describe an emblematic case treated with a novel tailored approach. A postmenopausal woman with advanced BGA was referred to our department for an adequate evaluation, staging and treatment. Notably, we used PET/CT as a fundamental imaging technique for staging and follow-up. The patient underwent primary surgery followed by standard chemotherapy and pelvic radiotherapy. Three months later, she relapsed, with the appearance of multiple metastatic sites. Considering the evident chemoresistance to standard chemotherapy and the absence of valid therapeutic alternatives for this rare cancer, she was treated with a combination of repeated minimally invasive surgical procedures for all the resectable metastatic lesions and innovative approaches comprising, firstly, chemoimmunotherapy with Nivolumab combined with metronomic vinorelbine, which resulted in a clinical response for approximately 7 months. Upon disease progression, we used a targeted systemic approach based on the whole genomic profile of the primary tumor, which showed PTEN loss, which is predictive of a benefit from an mTOR inhibitor, and a CCND1 amplification, which predicts sensitivity to CDK4/6 inhibitors. Therefore, she received Everolimus, resulting in a significant metabolic response that lasted 12 months. Thereafter, upon further progression of the disease, the patient started Palbociclib treatment, which is currently ongoing, with evidence of a metabolic response. The patient has survived for 54 months from diagnosis, with a good performance status. In conclusion, the present paper confirms the lack of efficacy of conventional therapeutic regimens in the context of advanced, recurrent or metastatic adenocarcinomas of the Bartholin gland. The case report shows how a personalized multidisciplinary approach based on repeated minimally invasive surgery and tailored anticancer treatment based on whole-genome sequencing analysis could be effective and associated with prolonged survival in this rare gynecological cancer.
Collapse
|
17
|
Exploring Differentially Methylated Genes in Vulvar Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13143580. [PMID: 34298793 PMCID: PMC8306700 DOI: 10.3390/cancers13143580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Vulvar squamous cell carcinoma (VSCC) is the most common form of vulvar malignancy, and its incidence has increased in recent years. For better diagnosis and prognostication, and to expand available treatment options, molecular characterization of VSCC is crucial. We sought to identify aberrations in DNA methylation in VSCC, as this has been implicated in the development of several cancers. To this end, we performed genome-wide methylation sequencing on a set of VSCC and normal vulvar tissue using the Infinium MethylationEPIC BeadChip array. We detected 199 genes to be differentially methylated in VSCC compared to normal vulvar tissue. Of these, 194 genes were hyper-methylated, which leads to a loss of function of the genes. As most of these genes are involved in transcription regulator activity, our results suggest that disruption of this process plays an important role in VSCC development. Abstract DNA methylation is the most widely studied mechanism of epigenetic modification, which can influence gene expression without alterations in DNA sequences. Aberrations in DNA methylation are known to play a role in carcinogenesis, and methylation profiling has enabled the identification of biomarkers of potential clinical interest for several cancers. For vulvar squamous cell carcinoma (VSCC), however, methylation profiling remains an under-studied area. We sought to identify differentially methylated genes (DMGs) in VSCC, by performing Infinium MethylationEPIC BeadChip (Illumina) array sequencing, on a set of primary VSCC (n = 18), and normal vulvar tissue from women with no history of vulvar (pre)malignancies (n = 6). Using a false-discovery rate of 0.05, beta-difference (Δβ) of ±0.5, and CpG-island probes as cut-offs, 199 DMGs (195 hyper-methylated, 4 hypo-methylated) were identified for VSCC. Most of the hyper-methylated genes were found to be involved in transcription regulator activity, indicating that disruption of this process plays a vital role in VSCC development. The majority of VSCCs harbored amplifications of chromosomes 3, 8, and 9. We identified a set of DMGs in this exploratory, hypothesis-generating study, which we hope will facilitate epigenetic profiling of VSCCs. Prognostic relevance of these DMGs deserves further exploration in larger cohorts of VSCC and its precursor lesions.
Collapse
|
18
|
Molecular Landscape of Vulvar Squamous Cell Carcinoma. Int J Mol Sci 2021; 22:ijms22137069. [PMID: 34209172 PMCID: PMC8269046 DOI: 10.3390/ijms22137069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023] Open
Abstract
Vulvar squamous cell carcinoma (VSCC) is a rare malignancy with dual pathogenesis, Human papillomavirus (HPV)-associated and HPV-independent, with a poorly explored molecular landscape. We aimed to summarize the findings of the series analyzing molecular hallmarks of this neoplasm. In January 2021, we conducted a comprehensive literature search using Pubmed Medline and Scopus to identify publications focused on genomic profiling of VSCC. Observational studies, including both prospective and retrospective designs, evaluating molecular alterations in VSCC were deemed eligible. A total of 14 studies analyzing 749 VSCC were identified. The study series were heterogeneous in HPV testing and sequencing strategies, included small sets of tumors and cancer genes, and commonly lacked survival analysis. Only one extensive targeted next-generation sequencing-based study comprised a large cohort of 280 VSCC. The mutated genes, their number, and frequencies were highly variable between the series. Overall, TP53 and CDKN2A, followed by PIK3CA, HRAS, and PTEN, were the most frequently studied and mutated genes. Mutations involved in the PI3K/AKT/mTOR pathway, including TP53, HRAS, KRAS, and PIK3CA, have been consistently reported across the studies. However, the role of individual mutations or pathways in the development of VSCC remains unclear. In conclusion, heterogeneity and the small sample size of available molecular series contribute to a limited view of the molecular landscape of VSCC. Large-scale genome- or exome-wide studies with robust HPV testing are necessary to improve the molecular characterization of VSCC.
Collapse
|
19
|
Dasgupta S, Koljenović S, van den Bosch TPP, Swagemakers SMA, van der Hoeven NMA, van Marion R, van der Spek PJ, van Doorn HC, van Kemenade FJ, Ewing-Graham PC. Evaluation of Immunohistochemical Markers, CK17 and SOX2, as Adjuncts to p53 for the Diagnosis of Differentiated Vulvar Intraepithelial Neoplasia (dVIN). Pharmaceuticals (Basel) 2021; 14:ph14040324. [PMID: 33918187 PMCID: PMC8066509 DOI: 10.3390/ph14040324] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/26/2022] Open
Abstract
Histological diagnosis of differentiated vulvar intraepithelial neoplasia (dVIN), the precursor of human papillomavirus (HPV)-independent vulvar squamous cell carcinoma (VSCC), can be challenging, as features of dVIN may mimic those of non-dysplastic dermatoses. To aid the diagnosis, p53-immunohistochemistry (IHC) is commonly used, and mutant expression patterns are used to support a histological diagnosis of dVIN. However, a proportion of dVIN can show wild-type p53-expression, which is characteristic of non-dysplastic dermatoses. Furthermore, recent research has identified a novel precursor of HPV-independent VSCC—the p53-wild-type differentiated exophytic vulvar intraepithelial lesion (de-VIL). Currently, there are no established diagnostic IHC-markers for p53-wild-type dVIN or de-VIL. We evaluated IHC-markers, cytokeratin 17 (CK17), and SRY-box 2 (SOX2), as diagnostic adjuncts for dVIN. For this, IHC-expression of CK17, SOX2, and p53 was studied in dVIN (n = 56), de-VIL (n = 8), and non-dysplastic vulvar tissues (n = 46). For CK17 and SOX2, the percentage of cells showing expression, and the intensity and distribution of expression were recorded. We also performed next generation targeted sequencing (NGTS) on a subset of dVIN (n = 8) and de-VIL (n = 8). With p53-IHC, 74% of dVIN showed mutant patterns and 26% showed wild-type expression. Median percentage of cells expressing CK17 or SOX2 was significantly higher in dVIN (p53-mutant or p53-wild-type) and de-VIL than in non-dysplastic tissues (p < 0.01). Diffuse, moderate-to-strong, full epithelial expression of CK17 or SOX2 was highly specific for dVIN and de-VIL. With NGTS, TP53 mutations were detected in both dVIN and de-VIL. We infer that immunohistochemical markers CK17 and SOX2, when used along with p53, may help support the histological diagnosis of dVIN.
Collapse
Affiliation(s)
- Shatavisha Dasgupta
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
- Correspondence:
| | - Senada Koljenović
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Thierry P. P. van den Bosch
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Sigrid M. A. Swagemakers
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
- Department of Clinical Bioinformatics, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Nick M. A. van der Hoeven
- Department of Gynecology and Obstetrics, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands;
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Ronald van Marion
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Peter J. van der Spek
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
- Department of Clinical Bioinformatics, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Helena C. van Doorn
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Folkert J. van Kemenade
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Patricia C. Ewing-Graham
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| |
Collapse
|
20
|
Dasgupta S, Ewing-Graham PC, Van Den Bosch TPP, Swagemakers SMA, Santegoets LAM, Van Doorn HC, Van Der Spek PJ, Koljenović S, Van Kemenade FJ. Nuclear factor IB is downregulated in vulvar squamous cell carcinoma (VSCC): Unravelling differentially expressed genes in VSCC through gene expression dataset analysis. Oncol Lett 2021; 21:381. [PMID: 33841565 PMCID: PMC8020388 DOI: 10.3892/ol.2021.12642] [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: 11/19/2020] [Accepted: 02/12/2021] [Indexed: 11/25/2022] Open
Abstract
Vulvar squamous cell carcinoma (VSCC) comprises two distinct etiopathological subtypes: i) Human papilloma virus (HPV)-related VSCC, which arises via the precursor high grade squamous intraepithelial lesion (HSIL); and ii) HPV-independent VSCC, which arises via precursor, differentiated vulvar intraepithelial neoplasia (dVIN), driven by TP53 mutations. However, the mechanism of carcinogenesis of VSCC is poorly understood. The current study aimed to gain insight into VSCC carcinogenesis by identifying differentially expressed genes (DEGs) for each VSCC subtype. The expression of certain DEGs was then further assessed by performing immunohistochemistry (IHC) on whole tissue sections of VSCC and its precursors. Statistical analysis of microarrays was performed on two independent gene expression datasets (GSE38228 and a study from Erasmus MC) on VSCC and normal vulva. DEGs were identified that were similarly (up/down) regulated with statistical significance in both datasets. For HPV-related VSCCs, this constituted 88 DEGs, and for HPV-independent VSCCs, this comprised 46 DEGs. IHC was performed on VSCC (n=11), dVIN (n=6), HSIL (n=6) and normal vulvar tissue (n=7) with i) signal transducer and activator of transcription 1 (STAT1; an upregulated DEGs); ii) nuclear factor IB (NFIB; a downregulated DEG); iii) p16 (to determine the HPV status of tissues); and iv) p53 (to confirm the histological diagnoses). Strong and diffuse NFIB expression was observed in the basal and para-basal layers of normal vulvar tissue, whereas NFIB expression was minimal or completely negative in dVIN and in both subtypes of VSCC. In contrast, no discernable difference was observed in STAT1 expression among normal vulvar tissue, dVIN, HSIL or VSCC. By leveraging bioinformatics, the current study identified DEGs that can facilitate research into VSCC carcinogenesis. The results suggested that NFIB is downregulated in VSCC and its relevance as a diagnostic/prognostic biomarker deserves further exploration.
Collapse
Affiliation(s)
- Shatavisha Dasgupta
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Patricia C Ewing-Graham
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Thierry P P Van Den Bosch
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Sigrid M A Swagemakers
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands.,Department of Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Lindy A M Santegoets
- Department of Obstetrics and Gynecology, Reinier de Graaf Gasthuis, 2625 AD Delft, The Netherlands
| | - Helena C Van Doorn
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Peter J Van Der Spek
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands.,Department of Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Senada Koljenović
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| | - Folkert J Van Kemenade
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3000CA Rotterdam, The Netherlands
| |
Collapse
|
21
|
Xing D, Fadare O. Molecular events in the pathogenesis of vulvar squamous cell carcinoma. Semin Diagn Pathol 2021; 38:50-61. [PMID: 33032902 PMCID: PMC7749059 DOI: 10.1053/j.semdp.2020.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Vulvar squamous cell carcinomas (VSCC), which constitute over 90% of vulvar malignancies in adults, are classifiable into 2 subgroups that are mostly clinicopathologically distinct, a classification that is fundamentally based whether or not the tumors are HPV-mediated. In this review, we aim to summarize the recent advances in the understanding of molecular events in the pathogenesis of VSCC, including common and targetable mutations, copy number alterations, epigenetics, noncoding RNAs, and tumor immune microenvironment, which may provide insight into the future management of the disease. These events show substantial differences between the 2 subgroups, although significant areas of overlap exist. Recurrent, driver mutations appear to be substantially more prevalent in HPV(-) VSCC. TP53 mutations are the most common somatic mutations in VSCC overall, and are notably predominant in the HPV(-) VSCC, where 30-88% show a mutation. TP53 mutations are associated with worse patient outcomes, and co-mutations between TP53 and either HRAS, PIK3CA or CDKN2A appear to define subsets with even worse outcomes. A wide variety of other somatic mutations have been identified, including a subset with different mutational frequencies between HPV(+) and HPV(-) VSCC. CDKN2A mutations are common, and have been identified in 21 to 55% of HPV(-) VSCC, and in 2 to 25% of HPV(+) VSCC. Hypermethylation of CDKN2A is the most frequently reported epigenetic alteration in VSCC and the expression of some microRNAs may be associated with patient outcomes. The PTEN/PI3K/AKT/mTOR pathway is commonly altered in HPV(+) VSCC, and is accordingly potentially targetable. HPV-positivity/p16 block expression by immunohistochemistry has been found to be an independent prognostic marker for improved survival in VSCC, and may have some predictive value in VSCC patients treated with definitive radiotherapy. 22-39.3% and 68% of VSCC show EGFR amplification and protein overexpression respectively, although the prognostic and predictive value of an EGFR alteration requires additional study. Recurrent chromosomal gains in VSCCs have been found at 1q, 2q, 3q, 4p, 5p, 7p, 8p, 8q, and 12q, and there may be differential patterns of alterations depending on HPV-status. At least one-third of VSCC patients may potentially benefit from immune checkpoint inhibition therapy, based on a high frequency of PD-L1 expression or amplification, or a high tumor mutational burden. Additional studies are ultimately required to better understand the global landscape of genetic and epigenetic alterations in VSCC, and to identify and test potential targets for clinical application.
Collapse
Affiliation(s)
- Deyin Xing
- Departments of Pathology, Oncology, Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| | - Oluwole Fadare
- Department of Pathology, University of California San Diego Health, La Jolla, CA, United States
| |
Collapse
|