1
|
Aiyenuro A, Griffin H, Schichl K, Omar T, Ordi J, Kelly H, Walker C, Pino MD, Desai K, de Sanjosé S, Schiffman M, Doorbar J. Role of Reserve Cells in Metaplasia and the Development of Human Papillomavirus-Associated High-Grade Squamous Intraepithelial Lesions at the Cervical Transformation Zone. J Transl Med 2025; 105:104166. [PMID: 40204232 DOI: 10.1016/j.labinv.2025.104166] [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: 05/29/2024] [Revised: 03/04/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Squamous cervical cancers generally arise as a result of persistent infection with high-risk human papillomaviruses (hrHPVs) and occur near the squamocolumnar junction (SCJ) and within the transformation zone (TZ). The susceptibility of the TZ to HPV-related carcinogenesis appears linked to epithelial cell plasticity, with squamous metaplasia originating from a specialized stem cell population at this site. Two alternative cell populations have been implicated: keratin (K)7+ve cuboidal cells located at the SCJ vs a more broadly distributed K17+ve cervical reserve cell population. To distinguish between the hypotheses, we utilized multiplex immunofluorescence and large-scale digital imaging to map cell populations at the TZ of 165 women with and without hrHPV infections. Our results did not reveal a distinct population of K7+ cuboidal cells at the SCJ but found instead that the cuboidal and columnar cells of the TZ express K7 and K8 throughout and lack the p63 transcription factor required for epithelial stratification. Squamous metaplasia and reserve cells, which are defined by their subcolumnar location and pattern of biomarker expression (K5/K17/P63), were conspicuous at cervical crypt entrances within the TZ extending proximally toward the endocervix. In HPV-infected tissue, crypt-entrance regions with thin high-grade squamous intraepithelial lesion pathology showed prominent expression of hrHPV E6/E7 mRNA, as detected by fluorescence in situ hybridization, and p16/MCM expression, with infection also apparent in neighboring reserve cells. In some instances, normal/uninfected reserve cells (E6/E7 mRNA-ve) and squamous metaplasia were not only seen close to these regions of hrHPV infection but also extended well beyond the infected area both laterally and by depth. Our results confirm that the reserve cells underneath the columnar epithelia at TZ have the potential to undergo malignant squamous transformation via reserve cell proliferation, in agreement with previous histopathological studies. These translational findings highlight the importance of understanding the molecular biology of the epithelial sites where HPV cancers develop and suggest that in high-risk individuals, treatment strategies should target a wider area than previously thought.
Collapse
Affiliation(s)
- Ademola Aiyenuro
- Department of Pathology, University of Cambridge, United Kingdom
| | - Heather Griffin
- Department of Pathology, University of Cambridge, United Kingdom
| | | | - Tanvier Omar
- Department of Anatomical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Charlotte Maxeke Academic Hospital, Johannesburg, South Africa
| | - Jaume Ordi
- Institute Clinic of Gynecology, Obstetrics, and Neonatology-University of Barcelona, Barcelona, Spain; IS Global, Barcelona, Spain
| | - Helen Kelly
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Caroline Walker
- Department of Pathology, University of Cambridge, United Kingdom
| | - Marta Del Pino
- Institute Clinic of Gynecology, Obstetrics, and Neonatology-University of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Kanan Desai
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Silvia de Sanjosé
- IS Global, Barcelona, Spain; Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland.
| | - Mark Schiffman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland.
| | - John Doorbar
- Department of Pathology, University of Cambridge, United Kingdom.
| |
Collapse
|
2
|
Huerta-Padilla V, Marrero-Rodríguez D, Taniguchi-Ponciano K, López AE, Candanedo-González F, Salcedo E, Valdivia-Flores A, Rodriguez-Esquivel M, Virgilio LG, López-Romero R, Nambo-Lucio MDJ, Meza-Toledo SE, Bandala C, Meraz MA, Salcedo M. Thymopoietin- α, - β, and - γ Isoforms Increased Expression in Cervical Cancer Cells. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:1668482. [PMID: 40242184 PMCID: PMC12003041 DOI: 10.1155/cjid/1668482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/20/2024] [Indexed: 04/18/2025]
Abstract
Cervical cancer (CC) is a public health concern related to the human papillomavirus (HPV) persistent infection. Minichromosome maintenance 2 (MCM2) has been postulated as a surrogate marker for HPV infection. Thymopoietin (TMPO) is a nuclear protein regulated by E2F such as MCM2 or p16. TMPO can give rise to six different isoforms. Herein, both the mRNA and protein levels of TMPO isoforms were analyzed in cervical cells. TMPO expression was selected and analyzed through in silico in several databases from the healthy cervix and cervical lesions. TMPO RNA expression was evaluated in cervical samples and cell lines by RT-PCR and protein expression by Western-blot and immunohistochemistry assays. TMPO and MCM2 immunostaining were evaluated in cervical smears. The clinical-pathological correlation analysis was performed using Kruskal-Wallis or Χ 2 tests. TMPO is overexpressed in 74% of CC cells and all CC cell lines. Moreover, negative immunostaining was observed in normal cervical tissue, compared to strong expression for cervical lesions. Interestingly, TMPO-α, -β, -δ, -ε, and -γ are expressed in all cervical cells and tissues, but a differential expression for α, -β, and -γ isoforms among the cervical cells was observed as overexpressed when HPV is present. Also, the immunostaining of both MCM2 and TMPO was quite similar, but TMPO expression was more sensitive and specific than MCM2 protein. The present study has revealed that TMPO protein expression could be a potential molecular marker for cervical transformed cells, highlighting the TMPO-α, -β, and -γ isoforms as a promising molecular marker of HPV infection.
Collapse
Affiliation(s)
- Víctor Huerta-Padilla
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
- Departamento de Bioquimica, Laboratorio de Quimioterapia Experimental, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Daniel Marrero-Rodríguez
- Endocrine Diseases Research Unit, Specialties Hospital, National Medical Center SXXI, Mexican Institute of Social Security, Mexico City, Mexico
| | - Keiko Taniguchi-Ponciano
- Endocrine Diseases Research Unit, Specialties Hospital, National Medical Center SXXI, Mexican Institute of Social Security, Mexico City, Mexico
| | - Ariana E. López
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | - Fernando Candanedo-González
- Anatomo-Pathology Service, Oncology Hospital, National Medical Center SXXI, Mexican Institute of Social Security, Mexico City, Mexico
| | - Emmanuel Salcedo
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | | | - Miriam Rodriguez-Esquivel
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | - Laura Gómez Virgilio
- Centro de Investigación y de Estudios Avanzados, Molecular Biomedicine Department, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Ricardo López-Romero
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | | | - Sergio E. Meza-Toledo
- Departamento de Bioquimica, Laboratorio de Quimioterapia Experimental, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Cindy Bandala
- Departamento de Medicina Traslacional aplicada a Neurociencias, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Marco A. Meraz
- Centro de Investigación y de Estudios Avanzados, Molecular Biomedicine Department, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mauricio Salcedo
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| |
Collapse
|
3
|
Ovestad IT, Dalen I, Soreng K, Akbari S, Lapin M, Janssen EAM, Austdal M, Munk AC, Gudlaugsson E. Gene expression levels associated with impaired immune response and increased proliferation could serve as biomarkers for women following cervical cancer screening programmes. Front Immunol 2024; 15:1507193. [PMID: 39712018 PMCID: PMC11659148 DOI: 10.3389/fimmu.2024.1507193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Human papilloma virus (HPV) infections vary in their oncogenic potential, and whether an infection progresses to cervical intraepithelial neoplasia (CIN) also depends on the immune response. Therefore, the aim of the present study was to explore biomarkers related to the immune system and cell proliferation, in combination with HPV classified as having high (HOP) or low oncogenic potential (LOP), that can possibly guide a more accurate identification of women following cervical cancer screening programmes in need for immediate follow-up with a biopsy. A next-generation sequencing transcriptomic immune profile analysis applied to 28 persistent CIN3 lesions and 14 normal biopsies identified four genes, the immune markers ARG1 and HLA-DQB2 and the tumour markers CDKN2A and KRT7, as possible markers for differentiating between CIN3 and normal tissue. To validate these findings, analysis of the relative gene expression of these markers by use of reverse transcriptase real-time quantitative polymerase chain reaction was performed in an independent cohort of 264 (82 normal, 64 CIN1, and 118 CIN2/CIN3) biopsies, and the data were combined with information on the HOP- or LOP-HPV identified in the biopsies. Statistical analysis was performed with receiver operating characteristic curves, reporting area under the curve (AUC) with 95% confidence intervals (CIs), and logistic regression. Statistically significantly higher median expression levels of CDKN2A (p < 0.001) and KRT7 (p = 0.045) and significantly lower expression of ARG1 (p = 0.012) were found in biopsies with HOP-HPV infections, with no difference detected for HLA-DQB2 (p = 0.82). Models using expression levels of CDKN2A (AUC, 0.91; 95% CI, 0.86-0.95), KRT7 (0.86, 0.81-0.91), or ARG1 (0.78, 0.70-0.85) together with HOP/LOP-HPV class were significantly better than HPV class alone (0.72, 0.66-0.79) in discriminating CIN2/3 versus CIN1 (p < 0.001, p < 0.001, and p = 0.014, respectively).
Collapse
Affiliation(s)
- Irene T. Ovestad
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Ingvild Dalen
- Section of Biostatistics, Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Kristiane Soreng
- National HPV Reference laboratory Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
| | - Saleha Akbari
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Morten Lapin
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Emiel AM Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, Stavanger, Norway
| | - Marie Austdal
- Section of Biostatistics, Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Ane Cecilie Munk
- Department of Gynaecology, Sorlandet Hospital, Kristiansand, Norway
| | - Einar Gudlaugsson
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
4
|
Fehér E, Kaszab E, Mótyán JA, Máté D, Bali K, Hoitsy M, Sós E, Jakab F, Bányai K. Structural similarity of human papillomavirus E4 and polyomaviral VP4 exhibited by genomic analysis of the common kestrel (Falco tinnunculus) polyomavirus. Vet Res Commun 2024; 48:309-315. [PMID: 37688754 PMCID: PMC10810995 DOI: 10.1007/s11259-023-10210-1] [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: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 09/11/2023]
Abstract
Polyomaviruses are widely distributed viruses of birds that may induce developmental deformities and internal organ disorders primarily in nestlings. In this study, polyomavirus sequence was detected in kidney and liver samples of a common kestrel (Falco tinnunculus) that succumbed at a rescue station in Hungary. The amplified 5025 nucleotide (nt) long genome contained the early (large and small T antigen, LTA and STA) and late (viral proteins, VP1, VP2, VP3) open reading frames (ORFs) typical for polyomaviruses. One of the additional putative ORFs (named VP4) showed identical localization with the VP4 and ORF-X of gammapolyomaviruses, but putative splicing sites could not be found in its sequence. Interestingly, the predicted 123 amino acid (aa) long protein sequence showed the highest similarity with human papillomavirus E4 early proteins in respect of the aa distribution and motif arrangement implying similar functions. The LTA of the kestrel polyomavirus shared <59.2% nt and aa pairwise identity with the LTA sequence of other polyomaviruses and formed a separated branch in the phylogenetic tree among gammapolyomaviruses. Accordingly, the kestrel polyomavirus may be the first member of a novel species within the Gammapolyomavirus genus, tentatively named Gammapolyomavirus faltin.
Collapse
Affiliation(s)
- Enikő Fehér
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary.
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary.
- National Laboratory of Virology, Szentágothai Research Centre, University of Pécs, Pécs, Hungary.
| | - Eszter Kaszab
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
- Institute of Metagenomics, University of Debrecen, Debrecen, Hungary
| | - János András Mótyán
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dóra Máté
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
| | - Krisztina Bali
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
| | - Márton Hoitsy
- Conservation and Veterinary Services, Budapest Zoo and Botanical Garden, Budapest, Hungary
- Department of Exotic Animal and Wildlife Medicine, University of Veterinary Medicine, Budapest, Hungary
| | - Endre Sós
- Conservation and Veterinary Services, Budapest Zoo and Botanical Garden, Budapest, Hungary
- Department of Exotic Animal and Wildlife Medicine, University of Veterinary Medicine, Budapest, Hungary
| | - Ferenc Jakab
- National Laboratory of Virology, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Krisztián Bányai
- HUN-REN Veterinary Medical Research Institute, Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, Budapest, Hungary
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
5
|
Kirk A, Graham SV. The human papillomavirus late life cycle and links to keratinocyte differentiation. J Med Virol 2024; 96:e29461. [PMID: 38345171 DOI: 10.1002/jmv.29461] [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: 08/30/2023] [Revised: 12/21/2023] [Accepted: 01/25/2024] [Indexed: 02/15/2024]
Abstract
Regulation of human papillomavirus (HPV) gene expression is tightly linked to differentiation of the keratinocytes the virus infects. HPV late gene expression is confined to the cells in the upper layers of the epithelium where the virus capsid proteins are synthesized. As these proteins are highly immunogenic, and the upper epithelium is an immune-privileged site, this spatial restriction aids immune evasion. Many decades of work have contributed to the current understanding of how this restriction occurs at a molecular level. This review will examine what is known about late gene expression in HPV-infected lesions and will dissect the intricacies of late gene regulation. Future directions for novel antiviral approaches will be highlighted.
Collapse
Affiliation(s)
- Anna Kirk
- Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Sheila V Graham
- Centre for Virus Research, University of Glasgow, Glasgow, UK
| |
Collapse
|
6
|
Rosendo-Chalma P, Antonio-Véjar V, Ortiz Tejedor JG, Ortiz Segarra J, Vega Crespo B, Bigoni-Ordóñez GD. The Hallmarks of Cervical Cancer: Molecular Mechanisms Induced by Human Papillomavirus. BIOLOGY 2024; 13:77. [PMID: 38392296 PMCID: PMC10886769 DOI: 10.3390/biology13020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024]
Abstract
Human papillomaviruses (HPVs) and, specifically, high-risk HPVs (HR-HPVs) are identified as necessary factors in the development of cancer of the lower genital tract, with CaCU standing out as the most prevalent tumor. This review summarizes ten mechanisms activated by HR-HPVs during cervical carcinogenesis, which are broadly associated with at least seven of the fourteen distinctive physiological capacities of cancer in the newly established model by Hanahan in 2022. These mechanisms involve infection by human papillomavirus, cellular tropism, genetic predisposition to uterine cervical cancer (CaCU), viral load, viral physical state, regulation of epigenetic mechanisms, loss of function of the E2 protein, deregulated expression of E6/E7 oncogenes, regulation of host cell protein function, and acquisition of the mesenchymal phenotype.
Collapse
Affiliation(s)
- Pedro Rosendo-Chalma
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer of Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (IIB-UNAM), Mexico City 14080, Mexico
- Unidad Académica de Posgrado, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
| | - Verónica Antonio-Véjar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Guerrero, Mexico
| | - Jonnathan Gerardo Ortiz Tejedor
- Unidad Académica de Posgrado, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
- Carrera de Bioquímica y Farmacia, Universidad Católica de Cuenca, Cuenca 010101, Ecuador
| | - Jose Ortiz Segarra
- Carrera de Medicina, Facultad de Ciencias Médicas, Universidad de Cuenca, Cuenca 010107, Ecuador
| | - Bernardo Vega Crespo
- Carrera de Medicina, Facultad de Ciencias Médicas, Universidad de Cuenca, Cuenca 010107, Ecuador
| | | |
Collapse
|
7
|
Reuschenbach M, Doorbar J, Del Pino M, Joura EA, Walker C, Drury R, Rauscher A, Saah AJ. Prophylactic HPV vaccines in patients with HPV-associated diseases and cancer. Vaccine 2023; 41:6194-6205. [PMID: 37704498 DOI: 10.1016/j.vaccine.2023.08.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/15/2023]
Abstract
Individuals with human papillomavirus (HPV)-related disease remain at risk for subsequent HPV infection and related disease after treatment of specific lesions. Prophylactic HPV vaccines have shown benefits in preventing subsequent HPV-related disease when administered before or soon after treatment. Based on our understanding of the HPV life cycle and vaccine mechanism of action, prophylactic HPV vaccination is not expected to clear active persistent HPV infection or unresected HPV-associated dysplastic tissue remaining after surgery. However, vaccination may reasonably be expected to prevent new HPV infections caused by a different HPV type as well as re-infection with the same HPV type, whether from a new exposure to an infected partner or through autoinoculation from an adjacent or distant productively infected site. In this review, we describe the evidence for using prophylactic HPV vaccines in patients with HPV-associated disease before, during, or after treatment and discuss potential mechanisms by which individuals with HPV-associated disease may or may not benefit from prophylactic vaccines. We also consider how precise terminology relating to the use of prophylactic vaccines in this population is critical to avoid the incorrect implication that prophylactic vaccines have direct therapeutic potential, which would be counter to the vaccine's mechanism of action, as well as considered off-label. In other words, the observed effects occur through the known mechanism of action of prophylactic HPV vaccines, namely by preventing virus of the same or a different HPV type from infecting the patient after the procedure.
Collapse
Affiliation(s)
- Miriam Reuschenbach
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, USA; MSD Sharp & Dohme GmbH, Levelingstraße 4a, 81673 Munich, Germany.
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Marta Del Pino
- Hospital Clínic de Barcelona, Universitat de Barcelona, Gran Via de les Corts Catalanes, 585, 08007 Barcelona, Spain
| | - Elmar A Joura
- Medical University of Vienna, Department of Gynecology and Obstetrics, Comprehensive Cancer Center, BT86/E 01, Spitalgasse 23, 1090 Vienna, Austria
| | - Caroline Walker
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | | | | | - Alfred J Saah
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, USA
| |
Collapse
|
8
|
Yin W, Egawa N, Zheng K, Griffin H, Tian P, Aiyenuro A, Bornstein J, Doorbar J. HPV E6 inhibits E6AP to regulate epithelial homeostasis by modulating keratinocyte differentiation commitment and YAP1 activation. PLoS Pathog 2023; 19:e1011464. [PMID: 37379354 DOI: 10.1371/journal.ppat.1011464] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
Human papillomaviruses (HPV) cause persistent infections by modulating epithelial homeostasis in cells of the infected basal layer. Using FUCCI and cell-cell competition assays, we have identifed regulatory roles for E6AP and NHERF1, which are the primary HPV11 E6 cellular targets, as well as being targets of the high-risk E6 proteins, in processes governing epithelial homeostasis (i.e. cell density, cell cycle entry, commitment to differentiation and basal layer delamination). Depletion of E6AP, or expression of HPV11 or 16E6 increased keratinocyte cell density and cell cycle activity, and delayed the onset of differentiation; phenotypes which were conspicuously present in HPV11 and 16 infected patient tissue. In line with proposed E6 functions, in HPV11 condyloma tissue, E6AP and NHERF1 were significantly reduced when compared to uninfected epithelium. In experimental systems, loss of HPV11 E6/E6AP binding abolished 11E6's homeostasis regulatory functions, while loss of E6/NHERF1 binding reduced the cell density threshold at which differentiation was triggered. By contrast, a NHERF1-binding mutant of 16E6 was not compromised in its homeostasis functions, while E6AP appeared essential. RNA sequencing revealed similar transcriptional profiles in both 11 and 16E6-expressing cells and E6AP-/- cells, with YAP target genes induced, and keratinocyte differentiation genes being downregulated. HPV11 E6-mediated Yap activation was observed in 2D and 3D (organotypic raft) cell culture systems and HPV-infected lesions, with both NHERF1, which is a regulator of the Hippo and Wnt pathways, and E6AP, playing an important role. As the conserved binding partner of Alpha group HPV E6 proteins, the precise role of E6AP in modulating keratinocyte phenotype and associated signalling pathways has not previously been defined. Our study suggests a model in which the preserved functions of the low and high-risk Alpha E6 proteins modulate epithelial homeostasis via E6AP activity, and lead to alteration of multiple downstream pathways, including those involving NHERF1 and YAP.
Collapse
Affiliation(s)
- Wen Yin
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ke Zheng
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Heather Griffin
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Pu Tian
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ademola Aiyenuro
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Jacob Bornstein
- Gynecologist & Obstetrician, Colposcopy, Azrieli Faculty of Medicine of Bar-Ilan University, and Galilee Medical Center-Nahariya
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
9
|
Ahmed SMQ, Laha S, Das R, Ifthikar MA, Das SP. MCM10 expression is linked to cervical cancer aggressiveness. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1009903. [PMID: 39086679 PMCID: PMC11285692 DOI: 10.3389/fmmed.2023.1009903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 02/03/2023] [Indexed: 08/02/2024]
Abstract
Cervical cancer screening is a challenge mainly in developing countries. In developed countries, both incidence and mortality rates have been decreasing due to well organized screening programs. One of the potential biomarkers being exploited are the minichromosome maintenance proteins (MCMs), which show both specificity and sensitivity. MCM2-7 are involved in DNA replication initiation and elongation, and the MCM subunits are highly expressed in malignant tissues. Unlike other MCMs, MCM10, which is not part of the core helicase complex, is a critical determinant of origin activation and its levels are limiting in cancer cells. In this study, we performed bioinformatic analysis on the expression profile of all DNA replication associated MCM proteins in cervical cancer. MCM10 showed a relatively higher expression profile compared to the other MCMs. The mRNA expression levels of the MCMs were significantly increased in tumour tissues compared to normal, and MCM10 showed a fold change of 3.4. In order to understand if MCM10 is associated with the aggressiveness of cervical cancer, we looked into the mRNA expression pattern of MCM10 in three cervical cancer cell lines and one normal cervical cell line. MCM10 expression was significantly higher in the case of the more aggressive cancer cell line HeLa compared to controls. MCM10, therefore, can serve as a prominent biomarker for cancer progression and thus aid in early detection to control the spread of cancer cells. Our results show that MCM10 expression levels in cervical cancer cell lines are associated with cancer aggressiveness, demonstrating its clinical significance.
Collapse
Affiliation(s)
| | - Suparna Laha
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Ranajit Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Mariam Anjum Ifthikar
- Department of Oncology, Yenepoya Medical College Hospital, Yenepoya (Deemed to be University), Mangalore, India
| | - Shankar Prasad Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
10
|
Przybylski M, Pruski D, Millert-Kalińska S, Krzyżaniak M, de Mezer M, Frydrychowicz M, Jach R, Żurawski J. Expression of E4 Protein and HPV Major Capsid Protein (L1) as A Novel Combination in Squamous Intraepithelial Lesions. Biomedicines 2023; 11:biomedicines11010225. [PMID: 36672733 PMCID: PMC9855969 DOI: 10.3390/biomedicines11010225] [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: 11/30/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
We aim to describe the relationship between the immunohistochemical expression patterns of HPV E4 markers and the presence of HPV major capsid protein (L1) in cervical tissues obtained by biopsy of patients with abnormal liquid-based cytology (LBC) results, HR HPV infections, or clinically suspicious cervix. A novel HPV-encoded marker, SILgrade-E4 (XR-E4-1), and an HPV (clone K1H8) antibody were used to demonstrate the expression in terminally differentiated epithelial cells with a productive HPV infection in the material. A semiquantitative analysis was performed based on light microscope images. The level of E4 protein decreased with the disease severity. Patients with LSIL-CIN 1 and HSIL-CIN 2 diagnoses had significantly lower levels of HPV major capsid protein (L1) than those without confirmed cervical lesions. Our analysis confirms a higher incidence of L1 in patients with molecularly diagnosed HPV infections and excluded lesions of LSIL-CIN 1 and HSIL-CIN 2. Further studies on the novel biomarkers might help assess the chances of the remission of lesions such as LSIL-CIN 1 and HSIL-CIN 2. Higher levels of E4 protein and L1 may confirm a greater probability of the remission of lesions and incidental infections. In the cytological verification or HPV-dependent screening model, testing for E4 protein and L1 expression may indicate a group with a lower risk of progression of histopathologically diagnosed lesions.
Collapse
Affiliation(s)
- Marcin Przybylski
- Gynecology Specialised Practise, 60-682 Poznań, Poland
- Department of Obstetrics and Gynecology, District Public Hospital in Poznan, 60-479 Poznań, Poland
| | - Dominik Pruski
- Department of Obstetrics and Gynecology, District Public Hospital in Poznan, 60-479 Poznań, Poland
- Gynecology Specialised Practise, 60-408 Poznań, Poland
- Correspondence:
| | - Sonja Millert-Kalińska
- Department of Obstetrics and Gynecology, District Public Hospital in Poznan, 60-479 Poznań, Poland
- Doctoral School, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| | - Monika Krzyżaniak
- Department of Pathology, Hospital of Lord’s Transfiguration, Poznan University of Medical Sciences, 61-848 Poznań, Poland
| | - Mateusz de Mezer
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznań, Poland
| | | | - Robert Jach
- Department of Gynecological Endocrinology, Jagiellonian University Medical College, 31-008 Cracow, Poland
| | - Jakub Żurawski
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznań, Poland
| |
Collapse
|
11
|
Shiraz A, Egawa N, Pelt DM, Crawford R, Nicholas AK, Romashova V, Sasieni P, Griffin H, Doorbar J. Cervical cell lift: A novel triage method for the spatial mapping and grading of precancerous cervical lesions. EBioMedicine 2022; 82:104157. [PMID: 35863292 PMCID: PMC9301573 DOI: 10.1016/j.ebiom.2022.104157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 06/14/2022] [Accepted: 06/28/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Primary HPV screening, due to its low specificity, requires an additional liquid-based cytology (LBC) triage test. However, even with LBC triage there has been a near doubling in the number of patients referred for colposcopy in recent years, the majority having low-grade disease. METHODS To counter this, a triage test that generates a spatial map of the cervical surface at a molecular level has been developed which removes the subjectivity associated with LBC by facilitating identification of lesions in their entirety. 50 patients attending colposcopy were recruited to participate in a pilot study to evaluate the test. For each patient, cells were lifted from the cervix onto a membrane (cervical cell lift, CCL) and immunostained with a biomarker of precancerous cells, generating molecular maps of the cervical surface. These maps were analysed to detect high-grade lesions, and the results compared to the final histological diagnosis. FINDINGS We demonstrated that spatial molecular mapping of the cervix has a sensitivity of 90% (95% CI 69-98) (positive predictive value 81% (95% CI 60-92)) for the detection of high-grade disease, and that AI-based analysis could aid disease detection through automated flagging of biomarker-positive cells. INTERPRETATION Spatial molecular mapping of the CCL improved the rate of detection of high-grade disease in comparison to LBC, suggesting that this method has the potential to decisively identify patients with clinically relevant disease that requires excisional treatment. FUNDING CRUK Early Detection Project award, Jordan-Singer BSCCP award, Addenbrooke's Charitable Trust, UK-MRC, Janssen Pharmaceuticals/Advanced Sterilisation Products, and NWO.
Collapse
Affiliation(s)
- Aslam Shiraz
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK; Department of Gynae-Oncology, Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Daniël M Pelt
- Leiden Institute of Advanced Computer Science (LIACS), Leiden University, Niels Bohrweg 1, 2333 CA Leiden, the Netherlands
| | - Robin Crawford
- Department of Gynae-Oncology, Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Adeline K Nicholas
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Veronika Romashova
- Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Peter Sasieni
- Cancer Prevention Group, School of Cancer & Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, King's College London, London, UK
| | - Heather Griffin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| |
Collapse
|
12
|
Saito M, Rajesh A, Innes C, van der Griend R, Fitzgerald P, Simcock B, Sykes P, Hibma M. Blimp-1 is a prognostic indicator for progression of cervical intraepithelial neoplasia grade 2. J Cancer Res Clin Oncol 2022; 148:1991-2002. [PMID: 35386001 PMCID: PMC9294030 DOI: 10.1007/s00432-022-03993-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Progression of cervical intraepithelial neoplasia (CIN) to higher grade disease is associated with persistent human papillomavirus (HPV) infection and an absence of immune-mediated regression. However, the immune microenvironment that distinguishes progression from persistent or regressing lesions has not been well defined. METHODS A total of 69 patients under the age of 25 with high-risk HPV-positive cytology and biopsy-confirmed p16-positive CIN2 were included in the study. Biopsies were stained using 20 antibodies to a range of immune markers. Based on a 2-year follow-up, samples were analysed in "progressor" (CIN3 +) or "persister/regressor" (CIN1, 2 or normal) groups. RESULTS Progression was most strongly associated with Blimp-1 positive cell staining in the lesion (P = 0.0019) and with low numbers of infiltrating CD4 cells in the dermal region beneath the lesion (P = 0.0022). The presence of CD4, CD8 and T bet-positive cells in the dermal region most strongly correlated with CD11c cells in the persister/regressor but not the progressor group. CONCLUSION High numbers of Blimp-1 + cells in CIN2 lesions may predict progression to more severe disease. Measurement of Blimp-1 may have diagnostic utility for the determination of the need to treat women with cervical pre-cancer. HIGHLIGHTS CIN2 progression is associated with high numbers of Blimp-1 positive cells in the lesion. Detection of Blimp-1 in the lesion may have utility as a prognostic test to inform the need to treat CIN2.
Collapse
Affiliation(s)
- Mayumi Saito
- Department of Pathology, Dunedin School of Medicine, University of Otago, P O Box 56, Dunedin, New Zealand
| | - Aarthi Rajesh
- Department of Pathology, Dunedin School of Medicine, University of Otago, P O Box 56, Dunedin, New Zealand
| | - Carrie Innes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | | | | | - Bryony Simcock
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Merilyn Hibma
- Department of Pathology, Dunedin School of Medicine, University of Otago, P O Box 56, Dunedin, New Zealand.
| |
Collapse
|
13
|
Damgaard RK, Jenkins D, de Koning MN, Quint WG, Stoler MH, Doorbar J, Kahlert J, Gravitt PE, Steiniche T, Petersen LK, Hammer A. Performance of HPV E4 and p16 INK4a biomarkers in predicting regression of cervical intraepithelial neoplasia grade 2 (CIN2): protocol for a historical cohort study. BMJ Open 2022; 12:e059593. [PMID: 35793925 PMCID: PMC9260811 DOI: 10.1136/bmjopen-2021-059593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Cervical intraepithelial neoplasia grade 2 (CIN2) represents a spectrum of lesions with variable progression and regression. Pathological diagnosis of CIN2 is subjective and poorly reproducible. Accurate diagnosis and identification of different patterns of CIN2 related to outcome are essential to reduce the risks of overtreatment or undertreatment. It is important to explore novel methods for risk stratification of CIN2 to enable targeted treatment of women at high risk of progression or persistent disease and follow-up of women at low risk. The combination of the novel biomarker human papillomavirus (HPV) E4 with p16INK4a targets steps in the transition from a productive oncogenic HPV infection (CIN1) to a transformed lesion (CIN3) within CIN2. Previous cross-sectional studies suggest that HPV E4 combined with p16INK4a may be valuable for risk assessment of CIN2. However, data on HPV E4/p16INK4a as a predictor for CIN2 regression is lacking. METHODS AND ANALYSIS We will conduct a historical cohort study including 500 women aged 23-40 years with a first CIN2 diagnosis in Aarhus, Denmark during 2000-2010. Women will be eligible if they have undergone active surveillance and have no previous record of hysterectomy, cone biopsy, and CIN2 or worse. Women will be randomly selected through the Danish Pathology Databank. Tissue samples from women included will be sectioned for p16INK4a and HPV E4 immunohistochemical staining in addition to conventional hematoxylin and eosin (H&E) staining. A positive result will be defined as HPV E4 positive. Through the Danish Pathology Databank, we will collect results on all subsequent cervical biopsies. Regression will be used as the primary outcome. ETHICS AND DISSEMINATION The study has been approved by the Ethical Committee in Central Denmark Region (1-10-72-60-20) and registered at the Faculty of Health, Aarhus University. Results will be published in a peer-reviewed journal and presented at scientific meetings. TRIAL REGISTRATION NUMBER NCT05049252.
Collapse
Affiliation(s)
- Rikke Kamp Damgaard
- Dep. Clinical Medicine, Aarhus University, Aarhus, Denmark
- Dep. Gynecology and Obstetrics, Center for Research and Education, Gødstrup Hospital, Herning, Denmark
| | - David Jenkins
- Viroclinics-DDL, DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | | | - Wim Gv Quint
- Viroclinics-DDL, DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - Mark H Stoler
- Dep. Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - John Doorbar
- Dep. Pathology, Division of Virology, University of Cambridge, Cambridge, UK
| | - Johnny Kahlert
- Dep. Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Patti E Gravitt
- National Cancer Institute, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Torben Steiniche
- Dep. Clinical Medicine, Aarhus University, Aarhus, Denmark
- Dep. Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Lone Kjeld Petersen
- Dep. Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
- OPEN, University of Southern Denmark, Odense, Denmark
| | - Anne Hammer
- Dep. Clinical Medicine, Aarhus University, Aarhus, Denmark
- Dep. Gynecology and Obstetrics, Center for Research and Education, Gødstrup Hospital, Herning, Denmark
| |
Collapse
|
14
|
Known Benefits and Unknown Risks of Active Surveillance of Cervical Intraepithelial Neoplasia Grade 2. Obstet Gynecol 2022; 139:680-686. [PMID: 35271554 DOI: 10.1097/aog.0000000000004705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/30/2021] [Indexed: 01/18/2023]
Abstract
Cervical intraepithelial neoplasia grade 2 (CIN 2) is an equivocal diagnosis with high interobserver variation. Owing to high regression rates of 50%, many countries recommend active surveillance of CIN 2, especially in women younger than age 25-30 years, where regression rates are even higher (ie, 60%). Additionally, excisional treatment is associated with increased risk of reproductive harm, particularly preterm birth. Active surveillance typically consists of semi-annual follow-up visits for up to 2 years, including colposcopy and either cytology, testing for human papillomavirus, or both. Excisional treatment is recommended for progression or persistent disease after 2 years. Because active surveillance in younger women is relatively new, knowledge on subsequent risk of cervical cancer is limited. Considering human papillomavirus latency, women undergoing active surveillance might be at higher risk of cervical cancer than women undergoing excisional treatment. Furthermore, there are limited data describing preferences of women for the management of CIN 2, and it is also unclear how active surveillance may affect planning for future pregnancy. In this context, biomarkers for risk stratification of CIN 2 into either high or low probability of progression would allow for targeted treatment. Currently, immunohistochemical staining for p16 is used to clarify the histologic diagnosis, but whether it or other biomarkers can be used for risk-stratification in clinical management of women with CIN 2 remains unknown. In conclusion, active surveillance of CIN 2 needs further investigation, including understanding the long-term cervical cancer risk and evaluation of markers that may enable risk stratification of CIN 2.
Collapse
|
15
|
Doorbar J, Zheng K, Aiyenuro A, Yin W, Walker CM, Chen Y, Egawa N, Griffin HM. Principles of epithelial homeostasis control during persistent human papillomavirus infection and its deregulation at the cervical transformation zone. Curr Opin Virol 2021; 51:96-105. [PMID: 34628359 DOI: 10.1016/j.coviro.2021.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/22/2022]
Abstract
Human papillomaviruses establish a reservoir of infection in the epithelial basal layer. To do this they limit their gene expression to avoid immune detection and modulate epithelial homeostasis pathways to inhibit the timing of basal cell delamination and differentiation to favour persistence. For low-risk Alpha papillomaviruses, which cause benign self-limiting disease in immunocompetent individuals, it appears that cell competition at the lesion edge restricts expansion. These lesions may be considered as self-regulating homeostatic structures, with epithelial cells of the hair follicles and sweat glands, which are proposed targets of the Beta and Mu papillomaviruses, showing similar restrictions to their expansion across the epithelium as a whole. In the absence of immune control, which facilitates deregulated viral gene expression, such lesions can expand, leading to problematic papillomatosis in afflicted individuals. By contrast, he high-risk Alpha HPV types can undergo deregulated viral gene expression in immunocompetent hosts at a number of body sites, including the cervical transformation zone (TZ) where they can drive the formation of neoplasia. Homeostasis at the TZ is poorly understood, but involves two adjacent epithelial cell population, one of which has the potential to stratify and to produce a multilayed squamous epithelium. This process of metaplasia involves a specialised cell type known as the reserve cell, which has for several decades been considered as the cell of origin of cervical cancer. It is becoming clear that during evolution, HPV gene products have acquired functions directly linked to their requirements to modify the normal processes of epithelial homestasis at their various sites of infection. These protein functions are beginning to provide new insight into homeostasis regulation at different body sites, and are likely to be central to our understanding of HPV epithelial tropisms.
Collapse
Affiliation(s)
- John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom.
| | - Ke Zheng
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Ademola Aiyenuro
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Wen Yin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Caroline M Walker
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Yuwen Chen
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| | - Heather M Griffin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB8 9UP, United Kingdom
| |
Collapse
|
16
|
Güzel C, van Sten-Van't Hoff J, de Kok IMCM, Govorukhina NI, Boychenko A, Luider TM, Bischoff R. Molecular markers for cervical cancer screening. Expert Rev Proteomics 2021; 18:675-691. [PMID: 34551656 DOI: 10.1080/14789450.2021.1980387] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Cervical cancer remains a significant healthcare problem, notably in low- to middle-income countries. While a negative test for hrHPV has a predictive value of more than 99.5%, its positive predictive value is less than 10% for CIN2+ stages. This makes the use of a so-called triage test indispensable for population-based screening to avoid referring women, that are ultimately at low risk of developing cervical cancer, to a gynecologist. This review will give an overview of tests that are based on epigenetic marker panels and protein markers. AREAS COVERED There is a medical need for molecular markers with a better predictive value to discriminate hrHPV-positive women that are at risk of developing cervical cancer from those that are not. Areas covered are epigenetic and protein markers as well as health economic considerations in view of the fact that most cases of cervical cancer arise in low-to-middle-income countries. EXPERT OPINION While there are biomarker assays based on changes at the nucleic acid (DNA methylation patterns, miRNAs) and at the protein level, they are not widely used in population screening. Combining nucleic acid-based and protein-based tests could improve the overall specificity for discriminating CIN2+ lesions that carry a low risk of progressing to cervical cancer within the screening interval from those that carry an elevated risk. The challenge is to reduce unnecessary referrals without an undesired increase in false-negative diagnoses resulting in cases of cervical cancer that could have been prevented. A further challenge is to develop tests for low-and middle-income countries, which is critical to reduce the worldwide burden of cervical cancer.
Collapse
Affiliation(s)
- Coşkun Güzel
- Erasmus MC, Department of Neurology, University of Groningen, Rotterdam, The Netherlands
| | | | | | - Natalia I Govorukhina
- Department of Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
| | | | - Theo M Luider
- Erasmus MC, Department of Neurology, University of Groningen, Rotterdam, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
van der Zee RP, Meijer CJLM, Cuming T, Kreuter A, van de Sandt MM, Quint WGV, de Vries HJC, Prins JM, Steenbergen RDM. Characterisation of anal intraepithelial neoplasia and anal cancer in HIV-positive men by immunohistochemical markers p16, Ki-67, HPV-E4 and DNA methylation markers. Int J Cancer 2021; 149:1833-1844. [PMID: 34310698 PMCID: PMC9292283 DOI: 10.1002/ijc.33748] [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: 04/12/2021] [Revised: 06/07/2021] [Accepted: 07/05/2021] [Indexed: 11/05/2022]
Abstract
Human papillomavirus (HPV)-induced anal intraepithelial neoplasia (AIN, graded 1-3) is highly prevalent in HIV-positive (HIV+) men who have sex with men (MSM), but only a minority of lesions progresses to cancer. Our study aimed to characterise comprehensively anal tissue samples from a cross-sectional series (n = 104) of HIV+ MSM and longitudinal series (n = 40) of AIN2/3 progressing to cancer using different biomarkers. The cross-sectional series consisted of 8 normal, 26 AIN1, 45 AIN2, 15 AIN3 and 10 anal squamous cell carcinoma. Tissue sections were immunohistochemically (IHC) stained for p16 (viral transformation marker), Ki-67 (cellular proliferation marker) and HPV-E4 (viral production marker). We evaluated the expression of IHC markers and compared it with DNA methylation, a marker for malignant transformation. E4 positivity decreased, whereas p16 and Ki-67 scores and methylation marker positivity increased (P values < .001) with increasing severity of anal lesions. Within AIN2, a heterogeneous biomarker pattern was observed concerning E4, p16 and methylation status, reflecting the biological heterogeneity of these lesions. In the longitudinal series, all AIN2/3 and carcinomas showed high p16 and Ki-67 expression, strong methylation positivity and occasional E4 positivity. We earlier showed that high methylation levels are associated with progression to cancer. The observed E4 expression in some AIN2/3 during the course of progression to cancer and absence of E4 in a considerable number of AIN1 lesions make the potential clinical significance of E4 expression difficult to interpret. Our data show that IHC biomarkers can help to characterise AIN; however, their prognostic value for cancer risk stratification, next to objective methylation analysis, appears to be limited.
Collapse
Affiliation(s)
- Ramon P van der Zee
- Department of Pathology, Cancer Center Amsterdam (CCA), Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Internal Medicine, Division of Infectious Diseases, Amsterdam Institute for Infection and Immunity (AII), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Dermatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Chris J L M Meijer
- Department of Pathology, Cancer Center Amsterdam (CCA), Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Tamzin Cuming
- Anal Neoplasia Service, Homerton University Hospital, London, UK
| | - Alexander Kreuter
- Department of Dermatology, Venerology and Allergology, Helios St. Elisabeth Hospital Oberhausen, University Witten/Herdecke, Oberhausen, Germany
| | | | - Wim G V Quint
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - Henry J C de Vries
- Department of Dermatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Infectious Diseases, STI Outpatient Clinic, Public Health Service of Amsterdam (GGD Amsterdam), Amsterdam, The Netherlands
| | - Jan M Prins
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam Institute for Infection and Immunity (AII), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Renske D M Steenbergen
- Department of Pathology, Cancer Center Amsterdam (CCA), Amsterdam University Medical Centers (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Vink FJ, Dick S, Heideman DAM, De Strooper LMA, Steenbergen RDM, Lissenberg-Witte BI, Floore A, Bonde JH, Oštrbenk Valenčak A, Poljak M, Petry KU, Hillemanns P, van Trommel NE, Berkhof J, Bleeker MCG, Meijer CJLM. Classification of high-grade cervical intraepithelial neoplasia by p16 ink4a , Ki-67, HPV E4 and FAM19A4/miR124-2 methylation status demonstrates considerable heterogeneity with potential consequences for management. Int J Cancer 2021; 149:707-716. [PMID: 33729551 PMCID: PMC8252755 DOI: 10.1002/ijc.33566] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023]
Abstract
High‐grade cervical intraepithelial neoplasia (CIN2 and CIN3) represents a heterogeneous disease with varying cancer progression risks. Biomarkers indicative for a productive human papillomavirus (HPV) infection (HPV E4) and a transforming HPV infection (p16ink4a, Ki‐67 and host‐cell DNA methylation) could provide guidance for clinical management in women with high‐grade CIN. This study evaluates the cumulative score of immunohistochemical expression of p16ink4a (Scores 0‐3) and Ki‐67 (Scores 0‐3), referred to as the “immunoscore” (IS), in 262 CIN2 and 235 CIN3 lesions derived from five European cohorts in relation to immunohistochemical HPV E4 expression and FAM19A4/miR124‐2 methylation in the corresponding cervical scrape. The immunoscore classification resulted in 30 lesions within IS group 0‐2 (6.0%), 151 lesions within IS group 3‐4 (30.4%) and 316 lesions within IS group 5‐6 (63.6%). E4 expression decreased significantly from CIN2 to CIN3 (P < .001) and with increasing immunoscore group (Ptrend < .001). Methylation positivity increased significantly from CIN2 to CIN3 (P < .001) and with increasing immunoscore group (Ptrend < .001). E4 expression was present in 9.8% of CIN3 (23/235) and in 12.0% of IS group 5‐6 (38/316). Notably, in a minority (43/497, 8.7%) of high‐grade lesions, characteristics of both transforming HPV infection (DNA hypermethylation) and productive HPV infection (E4 expression) were found simultaneously. Next, we stratified all high‐grade CIN lesions, based on the presumed cancer progression risk of the biomarkers used, into biomarker profiles. These biomarker profiles, including immunoscore and methylation status, could help the clinician in the decision for immediate treatment or a “wait and see” policy to reduce overtreatment of high‐grade CIN lesions.
What's new?
Treating all high‐grade cervical intraepithelial neoplasia (CIN2/3) with excisional therapy leads to overtreatment, as these lesions have varying cancer progression risks. Here, the authors evaluated expression patterns of p16ink4a, Ki‐67 and the HPV E4 protein, and methylation of FAM19A4/miR124‐2 in high‐grade CIN. The biomarker expression patterns revealed the high degree of heterogeneity among CIN2/3 lesions. Biomarker profiles based on the presumed cancer progression risks were established and could guide clinicians in choosing whether to treat immediately or wait and see.
Collapse
Affiliation(s)
- Frederique J Vink
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Stèfanie Dick
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniëlle A M Heideman
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Lise M A De Strooper
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Renske D M Steenbergen
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Birgit I Lissenberg-Witte
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Arno Floore
- Self-screen B.V., Amsterdam, The Netherlands
| | - Jesper H Bonde
- Molecular Pathology Laboratory, Department of Pathology, Hvidovre Hospital, Hvidovre, Denmark
| | - Anja Oštrbenk Valenčak
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mario Poljak
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Karl U Petry
- Department of Gynecologic Oncology, Klinikum Wolfsburg, Wolfsburg, Germany
| | - Peter Hillemanns
- Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Nienke E van Trommel
- Antoni van Leeuwenhoek, Netherlands Cancer Institute, Department of Gynecologic Oncology, Centre of Gynecologic Oncology Amsterdam, Amsterdam, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maaike C G Bleeker
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Chris J L M Meijer
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Coordinated Expression of HPV-6 Genes with Predominant E4 and E5 Expression in Laryngeal Papilloma. Microorganisms 2021; 9:microorganisms9030520. [PMID: 33802595 PMCID: PMC7998961 DOI: 10.3390/microorganisms9030520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/17/2022] Open
Abstract
Laryngeal papilloma (LP) associated with human papillomavirus (HPV)-6 or -11 infection shows aggressive growth. However, the detailed molecular mechanism of virus-driven tumorigenesis has not been uncovered fully. HPV-6 viral gene expression and dynamic alterations were investigated with in situ localization of viral DNA and RNA in 13 patients with HPV-6-infected laryngeal papilloma. The average viral load was 4.80 × 105 ± 1.86 × 105 copies/ng DNA. E4, E5a, and E5b mRNAs accounted for 96% of the expression of 9 mRNAs. The alteration of viral DNA load during recurrence paralleled the mRNA expression levels, and the expression of all mRNAs showed a similar curve. E4, E5a, and E5b were expressed in the middle to upper part of the epithelium and were co-expressed in the same cells. E4 immunohistochemistry demonstrated an extensively positive reaction in the upper cell layer in accordance with E4 mRNA expression. These results suggest that individual viral genes are coordinately expressed for viral replication, virus release, and immunosurveillance avoidance. The newly developed E4-specific monoclonal antibody can be applied to further functional studies and clinical applications such as targeted molecular therapies.
Collapse
|
20
|
Egawa N, Shiraz A, Crawford R, Saunders-Wood T, Yarwood J, Rogers M, Sharma A, Eichenbaum G, Doorbar J. Dynamics of papillomavirus in vivo disease formation & susceptibility to high-level disinfection-Implications for transmission in clinical settings. EBioMedicine 2021; 63:103177. [PMID: 33421945 PMCID: PMC7806788 DOI: 10.1016/j.ebiom.2020.103177] [Citation(s) in RCA: 17] [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: 09/29/2020] [Revised: 11/20/2020] [Accepted: 12/04/2020] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND High-level disinfection protects tens-of-millions of patients from the transmission of viruses on reusable medical devices. The efficacy of high-level disinfectants for preventing human papillomavirus (HPV) transmission has been called into question by recent publications, which if true, would have significant public health implications. METHODS Evaluation of the clinical relevance of these published findings required the development of novel methods to quantify and compare: (i) Infectious titres of lab-produced, clinically-sourced, and animal-derived papillomaviruses, (ii) The papillomavirus dose responses in the newly developed in vitro and in vivo models, and the kinetics of in vivo disease formation, and (iii) The efficacy of high-level disinfectants in inactivating papillomaviruses in these systems. FINDINGS Clinical virus titres obtained from cervical lesions were comparable to those obtained from tissue (raft-culture) and in vivo models. A mouse tail infection model showed a clear dose-response for disease formation, that papillomaviruses remain stable and infective on fomite surfaces for at least 8 weeks without squames and up to a year with squames, and that there is a 10-fold drop in virus titre with transfer from a fomite surface to a new infection site. Disinfectants such as ortho-phthalaldehyde and hydrogen peroxide, but not ethanol, were highly effective at inactivating multiple HPV types in vitro and in vivo. INTERPRETATION Together with comparable results presented in a companion manuscript from an independent laboratory, this work demonstrates that high-level disinfectants inactivate HPV and highlights the need for standardized and well-controlled methods to assess HPV transmission and disinfection. FUNDING Advanced Sterilization Products, UK-MRC (MR/S024409/1 and MC-PC-13050) and Addenbrookes Charitable Trust.
Collapse
Affiliation(s)
- Nagayasu Egawa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Aslam Shiraz
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Robin Crawford
- Department of Gynae-Oncology, Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Taylor Saunders-Wood
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Jeremy Yarwood
- Advanced Sterilization Products, Inc., 33 Technology Drive, Irvine, CA 926184, USA
| | - Marc Rogers
- Advanced Sterilization Products, Inc., 33 Technology Drive, Irvine, CA 926184, USA
| | - Ankur Sharma
- Advanced Sterilization Products, Inc., 33 Technology Drive, Irvine, CA 926184, USA
| | - Gary Eichenbaum
- Johnson & Johnson, Office of the Chief Medical Officer, 410 George Street, New Brunswick, NJ, 08901, USA
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| |
Collapse
|
21
|
Use of Cap Analysis Gene Expression to detect human papillomavirus promoter activity patterns at different disease stages. Sci Rep 2020; 10:17991. [PMID: 33093512 PMCID: PMC7582169 DOI: 10.1038/s41598-020-75133-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 10/12/2020] [Indexed: 11/16/2022] Open
Abstract
Transcription of human papillomavirus (HPV) genes proceeds unidirectionally from multiple promoters. Direct profiling of transcription start sites (TSSs) by Cap Analysis Gene Expression (CAGE) is a powerful strategy for examining individual HPV promoter activity. The objective of this study was to evaluate alterations of viral promoter activity during infection using CAGE technology. We used CAGE-based sequencing of 46 primary cervical samples, and quantitatively evaluated TSS patterns in the HPV transcriptome at a single-nucleotide resolution. TSS patterns were classified into two types: early promoter-dominant type (Type A) and late promoter-dominant type (Type B). The Type B pattern was more frequently found in CIN1 and CIN2 lesions than in CIN3 and cancer samples. We detected transcriptomes from multiple HPV types in five samples. Interestingly, in each sample, the TSS patterns of both HPV types were the same. The viral gene expression pattern was determined by the differentiation status of the epithelial cells, regardless of HPV type. We performed unbiased analyses of TSSs across the HPV genome in clinical samples. Visualising TSS pattern dynamics, including TSS shifts, provides new insights into how HPV infection status relates to disease state.
Collapse
|
22
|
Molina MA, Carosi Diatricch L, Castany Quintana M, Melchers WJ, Andralojc KM. Cervical cancer risk profiling: molecular biomarkers predicting the outcome of hrHPV infection. Expert Rev Mol Diagn 2020; 20:1099-1120. [PMID: 33044104 DOI: 10.1080/14737159.2020.1835472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Cervical cancer affects half a million women worldwide annually. Given the association between high-risk human papillomavirus (hrHPV) infection and carcinogenesis, hrHPV DNA testing became an essential diagnostic tool. However, hrHPV alone does not cause the disease, and, most importantly, many cervical lesions regress to normal in a year because of the host immune system. Hence, the low specificity of hrHPV DNA tests and their inability to predict the outcome of infections have triggered a further search for biomarkers. AREAS COVERED We evaluated the latest viral and cellular biomarkers validated for clinical use as primary screening or triage for cervical cancer and assessed their promise for prevention as well as potential use in the future. The literature search focused on effective biomarkers for different stages of the disease, aiming to determine their significance in predicting the outcome of hrHPV infections. EXPERT OPINION Biomarkers such as p16/Ki-67, hrHPV genotyping, hrHPV transcriptional status, and methylation patterns have demonstrated promising results. Their eventual implementation in the screening programs may support the prompt diagnosis of hrHPV infection and its progression to cancer. These biomarkers will help in making clinical management decisions on time, thus, saving the lives of hrHPV-infected women, particularly in developing countries.
Collapse
Affiliation(s)
- Mariano A Molina
- Department of Microbiology, Faculty of Science, Radboud University , Nijmegen, The Netherlands.,Department of Medical Microbiology, Radboud university medical center , Nijmegen, The Netherlands
| | | | - Marina Castany Quintana
- Department of Medical Microbiology, Radboud university medical center , Nijmegen, The Netherlands
| | - Willem Jg Melchers
- Department of Medical Microbiology, Radboud university medical center , Nijmegen, The Netherlands
| | - Karolina M Andralojc
- Department of Medical Microbiology, Radboud university medical center , Nijmegen, The Netherlands.,Department of Biochemistry, Radboud Institute for Molecular Life Sciences , Nijmegen, The Netherlands
| |
Collapse
|
23
|
Kremer WW, Vink FJ, van Zummeren M, Dreyer G, Rozendaal L, Doorbar J, Bleeker MCG, Meijer CJLM. Characterization of cervical biopsies of women with HIV and HPV co-infection using p16 ink4a, ki-67 and HPV E4 immunohistochemistry and DNA methylation. Mod Pathol 2020; 33:1968-1978. [PMID: 32249820 DOI: 10.1038/s41379-020-0528-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/23/2022]
Abstract
This study aims to characterize cervical intraepithelial neoplasia (CIN) in women living with HIV using biomarkers. Immunohistochemical (IHC) staining for human papillomavirus (HPV) E4 protein indicates CIN with productive HPV infection, whereas Ki-67 and p16ink4a indicate CIN with transforming characteristics, which may be further characterized using DNA hypermethylation, indicative for advanced transforming CIN. Cervical biopsies (n = 175) from 102 HPV positive women living with HIV were independently reviewed by three expert pathologists. The consensus CIN grade was used as reference standard. IHC staining patterns were scored for Ki-67 (0-3), p16ink4a (0-3), and E4 (0-2) and correlated to methylation levels of four cellular genes in corresponding cervical scrapes. Reference standards and immunoscores were obtained from 165 biopsies:15 no dysplasia, 91 CIN1, 31 CIN2, and 28 CIN3. Ki-67 and p16ink4a scores increased with increasing CIN grade, while E4 positivity was highest in CIN1 and CIN2 lesions. E4 positive CIN1 lesions had higher Ki-67 and p16ink4a scores and higher methylation levels compared with E4 negative CIN1 lesions. E4 positive biopsies with low cumulative Ki-67/p16 ink4a immunoscores (0-3) had significantly higher methylation levels compared with E4 negative biopsies. No significant differences in Ki-67 and p16ink4a scores and methylation levels were observed between E4 negative and positive CIN2 or CIN3 lesions. The presence of high methylation levels in scrapes of CIN lesions with IHC characteristics of both productive (E4 positive) and transforming infections (increased Ki-67/p16ink4a expression) in women living with HIV might indicate a rapid aggressive course of HPV infections towards cancer in these women.
Collapse
Affiliation(s)
- Wieke W Kremer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Frederique J Vink
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Marjolein van Zummeren
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Greta Dreyer
- Department of Obstetrics and Gynecology, University of Pretoria, Pretoria, South Africa
| | - Lawrence Rozendaal
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Maaike C G Bleeker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Chris J L M Meijer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Shiraz A, Crawford R, Egawa N, Griffin H, Doorbar J. The early detection of cervical cancer. The current and changing landscape of cervical disease detection. Cytopathology 2020; 31:258-270. [PMID: 32301535 DOI: 10.1111/cyt.12835] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 01/04/2023]
Abstract
Cervical cancer prevention has undergone dramatic changes over the past decade. With the introduction of human papillomavirus (HPV) vaccination, some countries have seen a dramatic decline in HPV-mediated cervical disease. However, widespread implementation has been limited by economic considerations and the varying healthcare priorities of different countries, as well as by vaccine availability and, in some instances, vaccine hesitancy amongst the population/government. In this environment, it is clear that cervical screening will retain a critical role in the prevention of cervical cancer and will in due course need to adapt to the changing incidence of HPV-associated neoplasia. Cervical screening has, for many years, been performed using Papanicolaou staining of cytology samples. As our understanding of the role of HPV in cervical cancer progression has advanced, and with the availability of sensitive detection systems, cervical screening now incorporates HPV testing. Although such tests improve disease detection, they are not specific, and cannot discriminate high-grade from low-grade disease. This has necessitated the development of effective triage approaches to stratify HPV-positive women according to their risk of cancer progression. Although cytology triage remains the mainstay of screening, novel strategies under evaluation include DNA methylation, biomarker detection and the incorporation of artificial intelligence systems to detect cervical abnormalities. These tests, which can be partially anchored in a molecular understanding of HPV pathogenesis, will enhance the sensitivity of disease detection and improve patient outcomes. This review will provide insight on these innovative methodologies while explaining their scientific basis drawing from our understanding of HPV tumour biology.
Collapse
Affiliation(s)
- Aslam Shiraz
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Gynae-Oncology, Addenbrookes Hospital, Cambridge, UK
| | - Robin Crawford
- Department of Gynae-Oncology, Addenbrookes Hospital, Cambridge, UK
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Heather Griffin
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Castle PE, Adcock R, Cuzick J, Wentzensen N, Torrez-Martinez NE, Torres SM, Stoler MH, Ronnett BM, Joste NE, Darragh TM, Gravitt PE, Schiffman M, Hunt WC, Kinney WK, Wheeler CM. Relationships of p16 Immunohistochemistry and Other Biomarkers With Diagnoses of Cervical Abnormalities: Implications for LAST Terminology. Arch Pathol Lab Med 2020; 144:725-734. [PMID: 31718233 PMCID: PMC8575174 DOI: 10.5858/arpa.2019-0241-oa] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
CONTEXT.— Lower Anogenital Squamous Terminology (LAST) standardization recommended p16INK4a immunohistochemistry (p16 IHC) for biopsies diagnosed morphologically as cervical intraepithelial neoplasia (CIN) grade 2 (CIN2) to classify them as low-grade or high-grade squamous intraepithelial lesions (HSILs). OBJECTIVE.— To describe the relationships of p16 IHC and other biomarkers associated with cervical cancer risk with biopsy diagnoses. DESIGN.— A statewide, stratified sample of cervical biopsies diagnosed by community pathologists (CPs), including 1512 CIN2, underwent a consensus, expert pathologist panel (EP) review (without p16 IHC results), p16 IHC interpretation by a third pathology group, and human papillomavirus (HPV) genotyping, results of which were grouped hierarchically according to cancer risk. Antecedent cytologic interpretations were also available. RESULTS.— Biopsies were more likely to test p16 IHC positive with increasing severity of CP diagnoses, overall (Ptrend ≤ .001) and within each HPV risk group (Ptrend ≤ .001 except for low-risk HPV [Ptrend < .010]). All abnormal grades of CP-diagnosed biopsies were more likely to test p16 IHC positive with a higher HPV risk group (Ptrend < .001), and testing p16 IHC positive was associated with higher HPV risk group than testing p16 IHC negative for each grade of CP-diagnosed biopsies (P < .001). p16 IHC-positive, CP-diagnosed CIN2 biopsies were less likely than CP-diagnosed CIN3 biopsies to test HPV16 positive, have an antecedent HSIL+ cytology, or to be diagnosed as CIN3+ by the EP (P < .001 for all). p16 IHC-positive, CP-diagnosed CIN1 biopsies had lower HPV risk groups than p16 IHC-negative, CP-diagnosed CIN2 biopsies (P < .001). CONCLUSIONS.— p16 IHC-positive, CP-diagnosed CIN2 appears to be lower cancer risk than CP-diagnosed CIN3. LAST classification of "HSIL" diagnosis, which includes p16 IHC-positive CIN2, should annotate the morphologic diagnosis (CIN2 or CIN3) to inform all management decisions, which is especially important for young (<30 years) women diagnosed with CIN2 for whom surveillance rather than treatment is recommended.
Collapse
Affiliation(s)
- Philip E Castle
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Rachael Adcock
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Jack Cuzick
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Nicolas Wentzensen
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Norah E Torrez-Martinez
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Salina M Torres
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Mark H Stoler
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Brigitte M Ronnett
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Nancy E Joste
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Teresa M Darragh
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Patti E Gravitt
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Mark Schiffman
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - William C Hunt
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Walter K Kinney
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| | - Cosette M Wheeler
- From Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York (Dr Castle); Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom (Ms Adcock and Dr Cuzick); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland (Drs Wentzensen and Schiffman); the Department of Pathology, University of New Mexico Cancer Center, Albuquerque (Ms Torrez-Martinez, Dr Torres, Dr Joste, Dr Gravitt, Mr Hunt, and Dr Wheeler); the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Stoler); the Department of Pathology, Johns Hopkins University, Baltimore, Maryland (Dr Ronnett); the Department of Pathology, University of California, San Francisco (Dr Darragh); and Sacramento, California (Dr Kinney)
| |
Collapse
|
26
|
Leeman A, Jenkins D, Marra E, van Zummeren M, Pirog E, van de Sandt M, van Eeden A, Schim van der Loeff M, Doorbar J, de Vries H, van Kemenade F, Meijer C, Quint W. Grading immunohistochemical markers p16 INK4a and HPV E4 identifies productive and transforming lesions caused by low- and high-risk HPV within high-grade anal squamous intraepithelial lesions. Br J Dermatol 2020; 182:1026-1033. [PMID: 31302935 PMCID: PMC7187351 DOI: 10.1111/bjd.18342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Because current guidelines recognise high-grade anal squamous intraepithelial lesions (HSILs) and low-grade SILs (LSILs), and recommend treatment of all HSILs although not all progress to cancer, this study aims to distinguish transforming and productive HSILs by grading immunohistochemical (IHC) biomarkers p16INK 4a (p16) and E4 in low-risk human papillomavirus (lrHPV) and high-risk (hr)HPV-associated SILs as a potential basis for more selective treatment. METHODS Immunostaining for p16 and HPV E4 was performed and graded in 183 biopsies from 108 HIV-positive men who have sex with men. The causative HPV genotype of the worst lesion was identified using the HPV SPF10-PCR-DEIA-LiPA25 version 1 system, with laser capture microdissection for multiple infections. The worst lesions were scored for p16 (0-4) to identify activity of the hrHPV E7 gene, and panHPV E4 (0-2) to mark HPV production and life cycle completion. RESULTS There were 37 normal biopsies, 60 LSILs and 86 HSILs, with 85% of LSILs caused by lrHPV and 93% of HSILs by hrHPV. No normal biopsy showed E4, but 43% of LSILs and 37% of HSILs were E4 positive. No differences in E4 positivity rates were found between lrHPV and hrHPV lesions. Most of the lesions caused by lrHPV (90%) showed very extensive patchy p16 staining; p16 grade in HSILs was variable, with frequency of productive HPV infection dropping with increasing p16 grade. CONCLUSIONS Combined p16/E4 IHC identifies productive and nonproductive HSILs associated with hrHPV within the group of HSILs defined by the Lower Anogenital Squamous Terminology recommendations. This opens the possibility of investigating selective treatment of advanced transforming HSILs caused by hrHPV, and a 'wait and see' policy for productive HSILs. What's already known about this topic? For preventing anal cancer in high-risk populations, all patients with high-grade squamous intraepithelial lesions (HSILs) are treated, even though this group of lesions is heterogeneous, the histology is variable and regression is frequent. What does this study add? By adding human papillomavirus (HPV) E4 immunohistochemistry to p16 INK4a (p16), and grading expression of both markers, different biomarker expression patterns that reflect the heterogeneity of HSILs can be identified. Moreover, p16/E4 staining can separate high-risk HPV-associated HSILs into productive and more advanced transforming lesions, providing a potential basis for selective treatment.
Collapse
Affiliation(s)
- A. Leeman
- DDL Diagnostic LaboratoryVisseringlaan 252288 ERRijswijkthe Netherlands
| | - D. Jenkins
- DDL Diagnostic LaboratoryVisseringlaan 252288 ERRijswijkthe Netherlands
| | - E. Marra
- Public Health Service of AmsterdamDepartment of Infectious DiseasesAmsterdamthe Netherlands
| | - M. van Zummeren
- Cancer Center AmsterdamDepartment of PathologyVU University Medical CenterAmsterdamthe Netherlands
| | - E.C. Pirog
- Department of PathologyWeill Cornell Medical CollegeNew YorkNYU.S.A
| | - M.M. van de Sandt
- DDL Diagnostic LaboratoryVisseringlaan 252288 ERRijswijkthe Netherlands
| | - A. van Eeden
- Department of Internal MedicineDC KliniekenAmsterdamthe Netherlands
| | | | - J. Doorbar
- Department of PathologyUniversity of CambridgeCambridgeU.K
| | - H.J.C. de Vries
- Public Health Service of AmsterdamDepartment of Infectious DiseasesAmsterdamthe Netherlands
- Department of DermatologyAmsterdam UMCUniversity of Amsterdam, and Amsterdam Infection and Immunity Institute (AI&II)Amsterdamthe Netherlands
| | - F.J. van Kemenade
- Erasmus MC University Medical CenterDepartment of PathologyRotterdamthe Netherlands
| | - C.J.L.M. Meijer
- Cancer Center AmsterdamDepartment of PathologyVU University Medical CenterAmsterdamthe Netherlands
| | - W.G.V. Quint
- DDL Diagnostic LaboratoryVisseringlaan 252288 ERRijswijkthe Netherlands
| |
Collapse
|
27
|
Hendawi N, Niklander S, Allsobrook O, Khurram SA, Bolt R, Doorbar J, Speight PM, Hunter KD. Human papillomavirus (HPV) can establish productive infection in dysplastic oral mucosa, but HPV status is poorly predicted by histological features and p16 expression. Histopathology 2020; 76:592-602. [PMID: 31617604 DOI: 10.1111/his.14019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/04/2019] [Accepted: 10/14/2019] [Indexed: 01/14/2023]
Abstract
AIMS Previous studies have reported the presence of high-risk human papillomavirus (HR-HPV) in a subset of dysplastic oral epithelial lesions. Many cases show a histological spectrum of atypia similar to that seen in non-human papillomavirus (HPV) severe epithelial dysplasia, but some studies have suggested that HPV status can be inferred on the basis of histological features. We aimed to assess the utility of such histological features and p16 as surrogate markers of HPV infection in a retrospective cohort of 33 cases of severe epithelial dysplasia, with matched clinicopathological data and histological features. METHODS AND RESULTS Tissue sections were assessed for the expression of p16, minichromosome maintenance 2, HPV E4 and HPV L1 by the use of immunohistochemistry. HPV16/18 E6 and E7 expression was assessed by the use of RNA in-situ hybridisation (RNAScope). In the cohort, 18.2% of cases (6/33) were HR-HPV-positive, with no age/gender differences between the HPV-positive and HPV-negative groups. HPV E4 and HPV L1 were expressed in surface keratinocytes in four of six (66%) HPV-positive cases, indicative of productive HPV infection. Lack of p16 expression was predictive of HPV-negative status, but sensitivity and specificity varied according to the cut-off. Histologically, the presence of karyorrhectic nuclei and abnormal mitotic figures was higher in HPV-positive lesions (P < 0.05), but the predictive specificity and sensitivity were suboptimal (sensitivity, 0.75; specificity, 0.52). CONCLUSIONS This study demonstrates, for the first time, that a minority of severely dysplastic oral lesions harbour productive, biologically relevant HPV infection. Consideration should be given to the specific assessment of HPV status in severe epithelial dysplasia cases, as both p16 status and the presence of karyorrhectic cells are poor predictive markers of HPV status.
Collapse
Affiliation(s)
- Naeima Hendawi
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
- Department of Oral Pathology, University of Benghazi, Benghazi, Libya
| | - Sven Niklander
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
- Facultad de Odontologia, Universidad Andres Bello, Viña del Mar, Chile
| | - Olive Allsobrook
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
| | - Syed Ali Khurram
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
| | - Robert Bolt
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Paul M Speight
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
| | - Keith D Hunter
- Unit of Oral and Maxillofacial Medicine and Pathology, University of Sheffield, Sheffield, UK
- Oral Pathology and Biology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
28
|
Leeman A, Jenkins D, Del Pino M, Ordi J, Torné A, Doorbar J, Meijer CJLM, van Kemenade FJ, Quint WGV. Expression of p16 and HPV E4 on biopsy samples and methylation of FAM19A4 and miR124-2 on cervical cytology samples in the classification of cervical squamous intraepithelial lesions. Cancer Med 2020; 9:2454-2461. [PMID: 32022461 PMCID: PMC7131853 DOI: 10.1002/cam4.2855] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/29/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022] Open
Abstract
The decision to treat a cervical squamous intraepithelial lesion (SIL) by loop electrosurgical excision procedure (LEEP) relies heavily on a colposcopy-directed biopsy showing high-grade (H)SIL. Diagnosis is often supported by p16, an immunohistochemical (IHC) biomarker of high-risk (hr)HPV E7 gene activity. Additional potential markers include methylation of tumor suppressor genes FAM19A4/miR124-2 in cervical cytology for advanced transforming HSIL and the IHC marker HPV E4 for productive, potentially regressing lesions. In 318 women referred for colposcopy, we investigated the relationship between staining patterns of p16 and E4 IHC in the worst biopsy, and the relation of these to FAM19A4/miR124-2 methylation status in cytology. E4-positive staining decreased with increasing SIL/CIN grade from 41% in LSIL to 3% in HSIL/CIN3. E4 positivity increased with grade of p16 when p16 expression was limited to the lower two third of the epithelium (r = 0.378), but fell with expression over. Loss of E4 expression in the worst lesion was associated with the methylation of FAM19A4/miR124-2. We also examined whether these biomarkers can predict the histological outcome of the LEEP biopsy in a subgroup of 119 who underwent LEEP treatment. About 85% of women with ≥lower two third p16 staining/E4-negative HSIL biopsies and 65% with limited p16 staining/E4-positive HSIL biopsies had ≥HSIL in the LEEP specimen (P = .025). p16 expression in a biopsy is related both to viral production and transformation, while decreased E4 expression relates to methylation, indicating advanced HSIL. p16 expression in ≥2/3 of the epithelium and absent E4 indicate likely HSIL on a subsequent LEEP specimen.
Collapse
Affiliation(s)
| | - David Jenkins
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - Marta Del Pino
- Institute of Gynecology, Obstetrics and Neonatology, Hospital Clínic-Institut d´Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Faculty of Medicine-University of Barcelona, Barcelona, Spain
| | - Jaume Ordi
- Department of Pathology, ISGlobal, Hospital Clinic of Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Aureli Torné
- Institute of Gynecology, Obstetrics and Neonatology, Hospital Clínic-Institut d´Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Faculty of Medicine-University of Barcelona, Barcelona, Spain
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Chris J L M Meijer
- Amsterdam Medical Center, Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Wim G V Quint
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| |
Collapse
|
29
|
Griffin H, Mudhar HS, Rundle P, Shiraz A, Mahmood R, Egawa N, Quint W, Rennie IG, Doorbar J. Human papillomavirus type 16 causes a defined subset of conjunctival in situ squamous cell carcinomas. Mod Pathol 2020; 33:74-90. [PMID: 31485010 PMCID: PMC6930848 DOI: 10.1038/s41379-019-0350-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 11/08/2022]
Abstract
Squamous cell carcinoma of the conjunctiva is associated with a number of risk factors, including HIV infection, iatrogenic immunosuppression and atopy. In addition, several studies have suggested an involvement of HPV, based on the presence of viral DNA, but did not establish whether there was active infection or evidence of causal disease association. In this manuscript, 31 cases of conjunctival in situ squamous cell carcinoma were classified as HPV DNA-positive or -negative, before being analysed by immunohistochemistry to establish the distribution of viral and cellular biomarkers of HPV gene expression. Our panel included p16INK4a, TP53 and MCM, but also the virally encoded E4 gene product, which is abundantly expressed during productive infection. Subsequent in situ detection of HPV mRNA using an RNAscope approach confirmed that early HPV gene expression was occurring in the majority of cases of HPV DNA-positive conjunctival in situ squamous cell carcinoma, with all of these cases occurring in the atopic group. Viral gene expression correlated with TP53 loss, p16INK4a elevation, and extensive MCM expression, in line with our general understanding of E6 and E7's role during transforming infection at other epithelial sites. A characteristic E4 expression pattern was detected in only one case. HPV mRNA was not detected in lower grades of dysplasia, and was not observed in cases that were HPV DNA-negative. Our study demonstrates an active involvement of HPV in the development of a subset of conjunctival in situ squamous cell carcinoma. No high-risk HPV types were detected other than HPV16. It appears that the conjunctiva is a vulnerable epithelial site for HPV-associated transformation. These cancers are defined by their pattern of viral gene expression, and by the distribution of surrogate markers of HPV infection.
Collapse
Affiliation(s)
- Heather Griffin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Hardeep Singh Mudhar
- National Specialist Ophthalmic Pathology Service, Department of Histopathology, E-Floor, Royal Hallamshire Hospital, Glossop Rd, Sheffield, S10 2JF, UK
| | - Paul Rundle
- Sheffield Ocular Oncology Service, Department of Ophthalmology, Royal Hallamshire Hospital, Glossop Rd, Sheffield, S10 2JF, UK
| | - Aslam Shiraz
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Radma Mahmood
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Wim Quint
- Delft Diagnostic Laboratories, Visseringlaan 25, 2288 ER, Rijswijk, The Netherlands
| | - Ian G Rennie
- Sheffield Ocular Oncology Service, Department of Ophthalmology, Royal Hallamshire Hospital, Glossop Rd, Sheffield, S10 2JF, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| |
Collapse
|
30
|
Ramsauer AS, Wachoski-Dark GL, Fraefel C, Tobler K, Brandt S, Knight CG, Favrot C, Grest P. Paving the way for more precise diagnosis of EcPV2-associated equine penile lesions. BMC Vet Res 2019; 15:356. [PMID: 31640696 PMCID: PMC6805557 DOI: 10.1186/s12917-019-2097-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/16/2019] [Indexed: 01/15/2023] Open
Abstract
Background There is growing evidence that equine papillomavirus type 2 (EcPV2) infection is causally associated with the development of equine genital squamous cell carcinomas (SCCs). Early stages of disease present clinically as plaques or wart-like lesions which can gradually progress to tumoural lesions. Histologically these lesions are inconsistently described as benign hyperplasia, papilloma, penile intraepithelial neoplasia (PIN), carcinoma in situ (CIS) or SCC. Guidelines for histological classification of early SCC precursor lesions are not precisely defined, leading to potential misdiagnosis. The aim of this study was to identify histologic criteria and diagnostic markers allowing for a more accurate diagnosis of EcPV2-associated equine penile lesions. Results A total of 61 archived equine penile lesions were histologically re-assessed and classified as benign hyperplasia, papilloma, CIS or SCC. From these, 19 representative lesions and adjacent normal skin were comparatively analysed for the presence of EcPV2 DNA and transcripts using PCR and RNA in situ hybridisation (RISH). All lesional samples were positive by EcPV2 PCR and RISH, while adjacent normal skin was negative. RISH analysis yielded signal distribution patterns that allowed distinction of early (hyperplasia, papilloma) from late stage lesions (CIS, SCC). Subsequently, the 19 lesions were further assessed for expression of p53, Ki67, MCM7 and MMP1 by immunohistochemistry (IHC). All four proteins were expressed in both normal and lesional tissue. However, p53 expression was up-regulated in basal keratinocyte layers of papillomas, CIS and SCCs, as well as in upper keratinocyte layers of CIS and SCCs. MCM7 expression was only up-regulated in upper proliferating keratinocyte layers of papillomas, CIS and SCCs. Conclusion This study proposes combining a refined histological protocol for analysis of equine penile lesions with PCR- and/or RISH based EcPV2-screening and p53/MCM7 IHC to more accurately determine the type of lesion. This may help to guide the choice of optimum treatment strategy, especially at early stages of disease.
Collapse
Affiliation(s)
- Anna Sophie Ramsauer
- Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland. .,Dermatology Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Garrett Louis Wachoski-Dark
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Cornel Fraefel
- Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Kurt Tobler
- Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Sabine Brandt
- Research Group Oncology, Equine Clinic, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Cameron Greig Knight
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Claude Favrot
- Dermatology Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Paula Grest
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Kaur G, Balasubramaniam SD, Lee YJ, Balakrishnan V, Oon CE. Minichromosome Maintenance Complex (MCM) Genes Profiling and MCM2 Protein Expression in Cervical Cancer Development. Asian Pac J Cancer Prev 2019; 20:3043-3049. [PMID: 31653153 PMCID: PMC6982666 DOI: 10.31557/apjcp.2019.20.10.3043] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Minichromosome maintenance complex (MCM) proteins are essential for the process of DNA replication and cell division. This study aimed to evaluate MCM genes expression profiles and MCM2 protein in HPV-associated cervical carcinogenesis. METHODOLOGY MCM2, 4, 5 and 7 genes expression profiles were evaluated in three cervical tissue samples each of normal cervix, human papillomavirus (HPV)-infected low grade squamous intraepithelial lesion (LSIL), high grade squamous intraepithelial lesion (HSIL) and squamous cell carcinoma (SCC), using Human Transcriptome Array 2.0 and validated by nCounter® PanCancer Pathway NanoString Array. Immunohistochemical expression of MCM2 protein was semi-quantitatively assessed by histoscore in tissue microarrays containing 9 cases of normal cervix, 10 LSIL, 10 HSIL and 42 cases of SCC. RESULTS MCM2, 4, 5 and 7 genes expressions were upregulated with increasing fold change during the progression from LSIL to HSIL and the highest in SCC. MCM2 gene had the highest fold change in SCC compared to normal cervix. Immunohistochemically, MCM2 protein was localised in the nuclei of basal cells of normal cervical epithelium and dysplastic-neoplastic cells of CIN and SCC. There was a significant difference in MCM2 protein expression between the histological groups (P = 0.039), and histoscore was the highest in HSIL compared to normal cervix (P = 0.010). CONCLUSION The upregulation of MCM genes expressions in cervical carcinogenesis reaffirms MCM as a proliferative marker in DNA replication pathway, whereby proliferation of dysplastic and cancer cells become increasingly dysregulated and uncontrolled. A strong expression of MCM2 protein in HSIL may aid as a concatenated screening tool in detecting pre-cancerous cervical lesions.
Collapse
Affiliation(s)
- Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | | | - Yung Jen Lee
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| |
Collapse
|
32
|
Ramsauer AS, Kubacki J, Favrot C, Ackermann M, Fraefel C, Tobler K. RNA-seq analysis in equine papillomavirus type 2-positive carcinomas identifies affected pathways and potential cancer markers as well as viral gene expression and splicing events. J Gen Virol 2019; 100:985-998. [PMID: 31084699 DOI: 10.1099/jgv.0.001267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Equine papillomavirus type 2 (EcPV2) was discovered only recently, but it is found consistently in the context of genital squamous cell carcinomas (SCCs). Since neither cell cultures nor animal models exist, the characterization of this potential disease agent relies on the analysis of patient materials. To analyse the host and viral transcriptome in EcPV2-affected horses, genital tissue samples were collected from horses with EcPV2-positive lesions as well as from healthy EcPV2-negative horses. It was determined by RNA-seq analysis that there were 1957 differentially expressed (DE) host genes between the SCC and control samples. These genes were most abundantly related to DNA replication, cell cycle, extracellular matrix (ECM)-receptor interaction and focal adhesion. By comparison to other cancer studies, MMP1 and IL8 appeared to be potential marker genes for the development of SCCs. Analysis of the viral reads revealed the transcriptional activity of EcPV2 in all SCC samples. While few reads mapped to the structural viral genes, the majority of reads mapped to the non-structural early (E) genes, in particular to E6, E7 and E2/E4. Within these reads a distinct pattern of splicing events, which are essential for the expression of different genes in PV infections, was observed. Additionally, in one sample the integration of EcPV2 DNA into the host genome was detected by DNA-seq and confirmed by PCR. In conclusion, while host MMP1 and IL8 expression and the presence of EcPV2 may be useful markers in genital SCCs, further research on EcPV2-related pathomechanisms may focus on cell cycle-related genes, the viral genes E6, E7 and E2/E4, and integration events.
Collapse
Affiliation(s)
- Anna Sophie Ramsauer
- 2 Dermatology Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,1 Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Jakub Kubacki
- 1 Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Claude Favrot
- 2 Dermatology Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Mathias Ackermann
- 1 Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Cornel Fraefel
- 1 Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Kurt Tobler
- 1 Institute of Virology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Refining our understanding of cervical neoplasia and its cellular origins. PAPILLOMAVIRUS RESEARCH 2019; 7:176-179. [PMID: 30974183 PMCID: PMC6477515 DOI: 10.1016/j.pvr.2019.04.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 11/26/2022]
Abstract
Human papillomaviruses (HPV) cause cancer at a number of vulnerable epithelial sites, including the cervix, the anus and the oropharynx, with cervical cancer being the most significant in terms of numbers. The cervix has a complex epithelial organisation, and comprises the stratified epithelium of the ectocervix, the columnar epithelium of the endocervix, and the cervical transformation zone (TZ). Most cervical cancers arise at the TZ, which is a site where a stratified squamous epithelium can develop via metaplasia from a simple columnar epithelium. It is thought that this process is mediated by the cervical reserve cell, a specialised type of stem cell that is located at the TZ, which has been proposed as the target cell for HPV infection. Reserve cells may be derived from the basal cells of the ectocervix, or may originate from the cuboidal cells found at the squamo columnar junction. It appears that HPV infection of these diverse cell types, including the columnar cells of the endocervix, facilitates deregulated viral gene expression and the development of neoplasia, with different epithelial sites having different cancer risk. It is envisaged that these concepts may explain the vulnerability of the oropharynx, and other TZ regions where HPV-associated cancers arise.
Collapse
|
34
|
Presence or Absence of Significant HPVE4 Expression in High-grade Anal Intraepithelial Neoplasia With p16/Ki-67 Positivity Indicates Distinct Patterns of Neoplasia: A Study Combining Immunohistochemistry and Laser Capture Microdissection PCR. Am J Surg Pathol 2019; 42:463-471. [PMID: 29438174 DOI: 10.1097/pas.0000000000000984] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Progression of anal intraepithelial neoplasia (AIN) involves transition from productive to transforming human papillomavirus (HPV) infection. Grading aims to distinguish productive low-grade AIN from high-grade anal intraepithelial neoplasia (HGAIN) with risk of cancer. We describe immunohistochemical patterns in AIN adding a novel marker for initiation of the productive phase of the HPV life cycle (panHPVE4) to those for cell cycle activity (Ki-67) and transforming activity of HPVE7 gene (p16). We studied 67 anal biopsies for suspected anal neoplasia (17 normal, 15 AIN1, 20 AIN2, 15 AIN3) from 54 men who have sex with men at New York Presbyterian Hospital, USA. Two pathologists generated consensus AIN and immunogrades. Whole tissue and laser capture microdissection samples from multiple HPV-infected biopsies were tested for HPV with SPF10-PCR-DEIA-LiPA25, version 1. (Para)basal Ki-67 expression distinguished normal from AIN (≥lower-third Ki-67) with sensitivity 0.92 and specificity 1.0. Ki-67 did not distinguish grades of AIN. Null/patchy p16 versus diffuse ≥lower-third patterns discriminated HGAIN (sensitivity, 1.0; specificity, 0.84). There was marked heterogeneity in E4 expression within HGAIN. Most AIN2 (14/20) was E4 versus 0/15 AIN3 (sensitivity, 0.70; specificity 1.0). HPV was detected in 63 (94%) biopsies, with 49 (77.8%) high-risk HPV. HPV16 was the most frequent (13%). Multiple HPV genotypes were found in 15 (24%) biopsies and laser capture microdissection -polymerase chain reaction confirmed specific HPV types in E4 +/- AIN. Although Ki-67 discriminated AIN and p16 HGAIN, E4/p16 staining shows that most AIN2 is different from transformed AIN3 in showing both entry into productive HPV infection and transforming activity.
Collapse
|
35
|
Hammer A, de Koning MN, Blaakaer J, Steiniche T, Doorbar J, Griffin H, Mejlgaard E, Svanholm H, Quint WG, Gravitt PE. Whole tissue cervical mapping of HPV infection: Molecular evidence for focal latent HPV infection in humans. PAPILLOMAVIRUS RESEARCH 2019; 7:82-87. [PMID: 30772498 PMCID: PMC6389775 DOI: 10.1016/j.pvr.2019.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/01/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022]
Abstract
In this study, we aimed to provide molecular evidence of HPV latency in humans and discuss potential challenges of conducting studies on latency. We analyzed the entire cervix of two women who underwent hysterectomy unrelated to cervical abnormality. The cervices were sectioned into 242 and 186 sets respectively, and each set was tested separately for HPV using the SPF10-PCR-DEIA-LiPA25 system. To identify whether there was any evidence of transforming or productive infection, we used the biomarkers E4 and P16INK4a to stain slides immediately adjacent to HPV-positive sections. HPV was detected in both cervices. In patient 1, 1/242 sets was positive for HPV31. In patient 2, 13/186 sets were positive for HPV18 and 1/186 was positive for HPV53. The infection was very focal in both patients, and there was no sign of a transforming or productive infection, as evaluated by the markers E4 and P16INK4a. Had we only analyzed one set from each block, the probability of detecting the infection would have been 32.3% and 2%, respectively.Our findings support the idea that HPV may be able to establish latency in the human cervix; however, the risk associated with a latent HPV infection remains unclear.
Collapse
Affiliation(s)
- Anne Hammer
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark.
| | | | - Jan Blaakaer
- Department of Obstetrics and Gynecology, Odense University Hospital, Denmark
| | - Torben Steiniche
- Department of Clinical Medicine, Aarhus University, Denmark; Department of Pathology, Aarhus University Hospital, Denmark
| | - John Doorbar
- Department of Pathology, University of Cambridge, United Kingdom
| | - Heather Griffin
- Department of Pathology, University of Cambridge, United Kingdom
| | - Else Mejlgaard
- Department of Pathology, Aarhus University Hospital, Denmark
| | | | - Wim Gv Quint
- DDL Diagnostic Laboratory, Rijswijk, the Netherlands
| | - Patti E Gravitt
- Department of Global Health, Milken Institute School of Public Health, the George Washington University, USA
| |
Collapse
|
36
|
Fudulu A, Albulescu A, Anton G. Human papillomaviruses' proteins with clinical utility. J Immunoassay Immunochem 2018; 40:81-90. [PMID: 30518287 DOI: 10.1080/15321819.2018.1553790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cervical cancer, the fourth leading cause of cancer-associated deaths among women worldwide, is associated with human papilloma virus (HPV) infection. Despite the prophylactic HPV vaccination and the implementation of cervical and HPV-based screening programs, a significant increase in cervical cancer incidence is estimated by the year 2020. Thus, further development of diagnostic tools that allow detection and risk assesment in genital HPV infection is necessary. A special interest is focused on the HPV viral proteins whose expression might be of use either as primary screening tool or in conjunction with other markers (cellular proteins, HPV DNA, PAP test).
Collapse
Affiliation(s)
- Alina Fudulu
- a Department of Molecular Virology , Stefan S Nicolau Institute of Virology , Bucharest , Romania
| | - Adrian Albulescu
- a Department of Molecular Virology , Stefan S Nicolau Institute of Virology , Bucharest , Romania.,b National Institute of Chemical-Pharmaceutical R&D , Bucharest , Romania
| | - Gabriela Anton
- a Department of Molecular Virology , Stefan S Nicolau Institute of Virology , Bucharest , Romania
| |
Collapse
|
37
|
Zummeren MV, Kremer WW, Leeman A, Bleeker MCG, Jenkins D, Sandt MVD, Doorbar J, Heideman DAM, Steenbergen RDM, Snijders PJF, Kenter GG, Quint WGV, Berkhof J, Meijer CJLM. HPV E4 expression and DNA hypermethylation of CADM1, MAL, and miR124-2 genes in cervical cancer and precursor lesions. Mod Pathol 2018; 31:1842-1850. [PMID: 30135508 DOI: 10.1038/s41379-018-0101-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023]
Abstract
In this study, we evaluate the expression of human papillomavirus E4 protein (marker for the onset of a productive infection) and hypermethylation of host-cell CADM1, MAL, and miR124-2 genes (marker for an advanced, transforming infection) in cervical intraepithelial neoplasia (CIN) and cancer. A total of 115 cervical lesions were categorized by 3 pathologists into no dysplasia, CIN1, CIN2, CIN3, or cancer by classical histomorphological grading criteria, and by an immunoscore (cumulative value: 0-6) grading system based on Ki-67 (score: 0-3) and p16ink4a (score: 0-3) expression. Lesions were immunostained for E4 protein and analyzed for hypermethylation of CADM1, MAL, or miR124-2 genes. Expression of E4 and hypermethylation levels were related to CIN grade based on both classical and immunoscore grading. Hypermethylation increased with severity of the lesion as defined by both classical histomorphological grading and immunoscore criteria, and was always present in carcinomas (22/22). Extensive E4 expression decreased with increasing CIN grade and immunoscore, being most frequent in classically graded CIN1 or in lesions with cumulative immunoscore 1-3 and absent in carcinomas. High-grade lesions (CIN2/3 or immunoscore: 4-6) showed less E4 expression, which was inversely related to an increasing hypermethylation. Extensive E4 expression, as observed in a small proportion of high-grade lesions (6/49 and 8/43, respectively), was mostly associated with a negative methylation marker status (5/6 and 7/8, respectively). Our results illustrate the gradual transition of productive CIN (reflected by extensive E4 expression), to advanced transforming CIN (reflected by extensive hypermethylation) and cancer. Expression patterns of E4 and hypermethylation status of host-cell genes, may be used to identify cervical lesions at risk for cervical cancer, providing a better guidance for clinicians on treatment decisions.
Collapse
Affiliation(s)
- Marjolein van Zummeren
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Wieke W Kremer
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Maaike C G Bleeker
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - David Jenkins
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | | | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Daniëlle A M Heideman
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Renske D M Steenbergen
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter J F Snijders
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Gemma G Kenter
- Department of Gynecology, Center for Gynecologic Oncology Amsterdam, Amsterdam, The Netherlands
| | - Wim G V Quint
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Chris J L M Meijer
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Thomas M, Banks L. Upsetting the Balance: When Viruses Manipulate Cell Polarity Control. J Mol Biol 2018; 430:3481-3503. [PMID: 29680664 PMCID: PMC7094317 DOI: 10.1016/j.jmb.2018.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/12/2018] [Accepted: 04/14/2018] [Indexed: 12/20/2022]
Abstract
The central importance of cell polarity control is emphasized by the frequency with which it is targeted by many diverse viruses. It is clear that in targeting key polarity control proteins, viruses affect not only host cell polarity, but also influence many cellular processes, including transcription, replication, and innate and acquired immunity. Examination of the interactions of different virus proteins with the cell and its polarity controls during the virus life cycles, and in virally-induced cell transformation shows ever more clearly how intimately all cellular processes are linked to the control of cell polarity.
Collapse
|
39
|
Stevenson A, Kavanagh K, Pan J, Stevenson L, Griffin H, Doorbar J, Scott E, Deeny M, Cuschieri K, Graham SV. Risk stratification of cervical disease using detection of human papillomavirus (HPV) E4 protein and cellular MCM protein in clinical liquid based cytology samples. J Clin Virol 2018; 108:19-25. [PMID: 30218891 PMCID: PMC6224362 DOI: 10.1016/j.jcv.2018.08.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022]
Abstract
Cytospinning is a viable method for preparing LBC cells for antibody staining. We assessed the performance of a dual biomarker (one viral: HPVE4, one cellular: MCM2) in risk stratification of cervical disease. MCM2 was significantly associated with CIN2+ (p = 0.03). HPVE4 was associated with CIN1/normal (p = 0.06). The dual biomarker approach may be useful to risk stratify cervical disease especially in resource-poor settings.
Background While human papillomavirus (HPV) DNA testing offers high sensitivity for the detection of significant cervical disease, its specificity is suboptimal given the high prevalence of transient HPV infections (CIN1 or less). Biomarkers to identify those suffering from low grade disease from those with high grade disease could save healthcare costs and reduce patient anxiety. Objective The objective of the present work was to develop and test an immunohistochemistry (IHC)-based dual viral and cellular biomarker strategy which was applicable to liquid based cytology (LBC) samples. Study design We developed a novel IHC assay for detection of HPV E4 and cellular minichromosome maintenance (MCM) proteins in routinely taken cervical LBC samples using cytospin-prepared slides. The assay was applied to a prospective cohort of Scottish women referred to a colposcopy clinic due to preceding cytological abnormalities. The performance of the biomarkers for detection of clinically insignificant (CIN1 or less) versus significant disease was determined. Results A total of 81 women were recruited representing 64 cases of <=CIN1 and 28 of CIN2 + . Biomarker performance relative to histopathology outcomes showed high levels of MCM detection was significantly associated with CIN2+ (p = 0.03) while E4 was detected more frequently in <=CIN1 (p = 0.06). Conclusions Combined detection of a host proliferation marker and a marker of viral gene expression could allow triage of cases of clinically insignificant disease prior to colposcopy. However, there was overlap between distributions of MCM levels in CIN2+ and <=CIN1 suggesting that additional biomarkers would be required for improved specificity. Combined with cytospin-prepared slides this approach could provide a means of risk stratification of disease in low resource settings.
Collapse
Affiliation(s)
- Andrew Stevenson
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH, Scotland, UK.
| | - Kim Kavanagh
- Mathematics and Statistics, Livingstone Tower, University of Strathclyde, Glasgow G1 1XQ, Scotland, UK.
| | - Jiafeng Pan
- Mathematics and Statistics, Livingstone Tower, University of Strathclyde, Glasgow G1 1XQ, Scotland, UK.
| | - Lynne Stevenson
- Veterinary Diagnostic Services, School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH, Scotland, UK.
| | - Heather Griffin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| | - Evelyn Scott
- Stobhill Hospital, 133 Balornock Rd, Glasgow G21 3UW, Scotland, UK
| | - Miriam Deeny
- Stobhill Hospital, 133 Balornock Rd, Glasgow G21 3UW, Scotland, UK.
| | - Kate Cuschieri
- Scottish HPV Reference Laboratory, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, EH16 4SA, Scotland, UK.
| | - Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH, Scotland, UK.
| |
Collapse
|
40
|
Pottier C, Kriegsmann M, Alberts D, Smargiasso N, Baiwir D, Mazzucchelli G, Herfs M, Fresnais M, Casadonte R, Delvenne P, Pauw E, Longuespée R. Microproteomic Profiling of High‐Grade Squamous Intraepithelial Lesion of the Cervix: Insight into Biological Mechanisms of Dysplasia and New Potential Diagnostic Markers. Proteomics Clin Appl 2018; 13:e1800052. [DOI: 10.1002/prca.201800052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/06/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Charles Pottier
- Mass Spectrometry LaboratoryGIGA‐ResearchDepartment of ChemistryUniversity of Liège Liège Belgium
- Department of Medical OncologyUniversity of Liège Liège Belgium
| | - Mark Kriegsmann
- Institute of pathologyUniversity of Heidelberg Heidelberg Germany
| | - Deborah Alberts
- Mass Spectrometry LaboratoryGIGA‐ResearchDepartment of ChemistryUniversity of Liège Liège Belgium
| | - Nicolas Smargiasso
- Mass Spectrometry LaboratoryGIGA‐ResearchDepartment of ChemistryUniversity of Liège Liège Belgium
| | | | - Gabriel Mazzucchelli
- Mass Spectrometry LaboratoryGIGA‐ResearchDepartment of ChemistryUniversity of Liège Liège Belgium
| | - Michael Herfs
- Laboratory of Experimental PathologyGIGA‐CancerDepartment of PathologyUniversity of Liège Liège Belgium
| | - Margaux Fresnais
- Department of Clinical Pharmacology and PharmacoepidemiologyUniversity of Heidelberg Heidelberg Germany
- German Cancer Consortium (DKTK)‐German Cancer Research Center (DKFZ) Heidelberg Germany
| | | | - Philippe Delvenne
- Laboratory of Experimental PathologyGIGA‐CancerDepartment of PathologyUniversity of Liège Liège Belgium
| | - Edwin Pauw
- Mass Spectrometry LaboratoryGIGA‐ResearchDepartment of ChemistryUniversity of Liège Liège Belgium
| | - Rémi Longuespée
- Mass Spectrometry LaboratoryGIGA‐ResearchDepartment of ChemistryUniversity of Liège Liège Belgium
- Institute of pathologyUniversity of Heidelberg Heidelberg Germany
- Proteopath GmbH Trier Germany
| |
Collapse
|
41
|
Purdie KJ, Proby CM, Rizvi H, Griffin H, Doorbar J, Sommerlad M, Feltkamp MC, der Meijden EV, Inman GJ, South AP, Leigh IM, Harwood CA. The Role of Human Papillomaviruses and Polyomaviruses in BRAF-Inhibitor Induced Cutaneous Squamous Cell Carcinoma and Benign Squamoproliferative Lesions. Front Microbiol 2018; 9:1806. [PMID: 30154763 PMCID: PMC6102365 DOI: 10.3389/fmicb.2018.01806] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Human papillomavirus (HPV) has long been proposed as a cofactor in the pathogenesis of cutaneous squamous cell carcinoma (cSCC). More recently, the striking clinico-pathological features of cSCCs that complicate treatment of metastatic melanoma with inhibitors targeting BRAF mutations (BRAFi) has prompted speculation concerning a pathogenic role for oncogenic viruses. Here, we investigate HPV and human polyomaviruses (HPyV) and correlate with clinical, histologic, and genetic features in BRAFi-associated cSCC. Materials and Methods: Patients receiving BRAFi treatment were recruited at Barts Health NHS Trust. HPV DNA was detected in microdissected frozen samples using reverse line probe technology and degenerate and nested PCR. HPV immunohistochemistry was performed in a subset of samples. Quantitative PCR was performed to determine the presence and viral load of HPyVs with affinity for the skin (HPyV6, HPyV7, HPyV9, MCPyV, and TSPyV). These data were correlated with previous genetic mutational analysis of H, K and NRAS, NOTCH1/2, TP53, CDKN2A, CARD11, CREBBP, TGFBR1/2. Chromosomal aberrations were profiled using single nucleotide polymorphism (SNP) arrays. Results: Forty-five skin lesions from seven patients treated with single agent vemurafenib in 2012–2013 were analyzed: 12 cSCC, 19 viral warts (VW), 2 actinic keratosis (AK), 5 verrucous keratosis/other squamoproliferative (VK/SP) lesions, one melanocytic lesion and 6 normal skin samples. Significant histologic features of viral infection were seen in 10/12 (83%) cSCC. HPV DNA was detected in 18/19 (95%) VW/SP, 9/12 (75%) cSCC, 4/5 (80%) SP, and 3/6 (50%) normal skin samples and in 1/12 cases assessed by immunohistochemistry. HPyV was co-detected in 22/30 (73%) of samples, usually at low viral load, with MCPyV and HPyV7 the most common. SNP arrays confirmed low levels of chromosomal abnormality and there was no significant correlation between HPV or HPyV detection and individual gene mutations or overall mutational burden. Conclusion: Despite supportive clinicopathologic evidence, the role for HPV and HPyV infection in the pathogenesis of BRAFi-induced squamoproliferative lesions remains uncertain. Synergistic oncogenic mechanisms are plausible although speculative. Nonetheless, with the prospect of a significant increase in the adjuvant use of these drugs, further research is justified and may provide insight into the pathogenesis of other BRAFi-associated malignancies.
Collapse
Affiliation(s)
- Karin J Purdie
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Charlotte M Proby
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Hasan Rizvi
- Department of Pathology, Barts Health NHS Trust, London, United Kingdom
| | - Heather Griffin
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - John Doorbar
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Mary Sommerlad
- Department of Dermatology, Barts Health NHS Trust, London, United Kingdom
| | - Mariet C Feltkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Els Van der Meijden
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Gareth J Inman
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Irene M Leigh
- Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Dermatology, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
42
|
van Zummeren M, Leeman A, Kremer WW, Bleeker MCG, Jenkins D, van de Sandt M, Heideman DAM, Steenbergen R, Snijders PJF, Quint WGV, Berkhof J, Meijer CJLM. Three-tiered score for Ki-67 and p16 ink4a improves accuracy and reproducibility of grading CIN lesions. J Clin Pathol 2018; 71:981-988. [PMID: 30012698 PMCID: PMC6225805 DOI: 10.1136/jclinpath-2018-205271] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/13/2018] [Accepted: 06/16/2018] [Indexed: 02/04/2023]
Abstract
Aims To investigate the accuracy and reproducibility of a scoring system for cervical intraepithelial neoplasia (CIN1–3) based on immunohistochemical (IHC) biomarkers Ki-67 and p16ink4a. Methods 115 cervical tissue specimens were reviewed by three expert gynaecopathologists and graded according to three strategies: (1) CIN grade based on H&E staining only; (2) immunoscore based on the cumulative score of Ki-67 and p16ink4a only (0–6); and (3) CIN grade based on H&E supported by non-objectified IHC 2 weeks after scoring 1 and 2. The majority consensus diagnosis of the CIN grade based on H&E supported by IHC was used as the Reference Standard. The proportion of test positives (accuracy) and the absolute agreements across pathologists (reproducibility) of the three grading strategies within each Reference Standard category were calculated. Results We found that immunoscoring with positivity definition 6 yielded the highest proportion of test positives for Reference Standard CIN3 (95.5%), in combination with the lowest proportion of test positives in samples with CIN1 (1.8%). The proportion of test positives for CIN3 was significantly lower for sole H&E staining (81.8%) or combined H&E and IHC grading (84.8%) with positivity definition ≥CIN3. Immunoscore 6 also yielded high absolute agreements for CIN3 and CIN1, but the absolute agreement was low for CIN2. Conclusions The higher accuracy and reproducibility of the immunoscore opens the possibility of a more standardised and reproducible definition of CIN grade than conventional pathology practice, allowing a more accurate comparison of CIN-based management strategies and evaluation of new biomarkers to improve the understanding of progression of precancer from human papillomavirus infection to cancer.
Collapse
Affiliation(s)
- Marjolein van Zummeren
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Wieke W Kremer
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Maaike C G Bleeker
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - David Jenkins
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | | | - Daniëlle A M Heideman
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Renske Steenbergen
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter J F Snijders
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Wim G V Quint
- DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Chris J L M Meijer
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Wechsler EI, Tugizov S, Herrera R, Da Costa M, Palefsky JM. E5 can be expressed in anal cancer and leads to epidermal growth factor receptor-induced invasion in a human papillomavirus 16-transformed anal epithelial cell line. J Gen Virol 2018; 99:631-644. [PMID: 29624161 DOI: 10.1099/jgv.0.001061] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We created the first human papillomavirus (HPV)-16-immortalized anal epithelial cell line, known as AKC2 cells to establish an in vitro model of HPV-16-induced anal carcinogenesis. Consistent with detection of E6, E7 and E5 expression in anal cancer biopsies, AKC2 cells expressed high levels of all three HPV oncogenes. Also, similar to findings in anal cancer biopsies, epidermal growth factor receptor (EGFR) was overexpressed in AKC2 cells. AKC2 cells exhibited a poorly differentiated and invasive phenotype in three-dimensional raft culture and inhibition of EGFR function abrogated AKC2 invasion. Reducing E5 expression using E5-targeted siRNAs in AKC2 cells led to knockdown of E5 expression, but also HPV-16 E2, E6 and E7 expression. AKC2 cells treated with E5-targeted siRNA had reduced levels of total and phosphorylated EGFR, and reduced invasion. Rescue of E6/E7 expression with simultaneous E5 knockdown confirmed that E5 plays a key role in EGFR overexpression and EGFR-induced invasion.
Collapse
Affiliation(s)
- Erin Isaacson Wechsler
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Sharof Tugizov
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rossana Herrera
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Maria Da Costa
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Joel M Palefsky
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
44
|
Ramos IR, Meade AD, Ibrahim O, Byrne HJ, McMenamin M, McKenna M, Malkin A, Lyng FM. Raman spectroscopy for cytopathology of exfoliated cervical cells. Faraday Discuss 2018; 187:187-98. [PMID: 27032537 DOI: 10.1039/c5fd00197h] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cervical cancer is the fourth most common cancer affecting women worldwide but mortality can be decreased by early detection of pre-malignant lesions. The Pap smear test is the most commonly used method in cervical cancer screening programmes. Although specificity is high for this test, it is widely acknowledged that sensitivity can be poor mainly due to the subjective nature of the test. There is a need for new objective tests for the early detection of pre-malignant cervical lesions. Over the past two decades, Raman spectroscopy has emerged as a promising new technology for cancer screening and diagnosis. The aim of this study was to evaluate the potential of Raman spectroscopy for cervical cancer screening using both Cervical Intraepithelial Neoplasia (CIN) and Squamous Intraepithelial Lesion (SIL) classification terminology. ThinPrep® Pap samples were recruited from a cervical screening population. Raman spectra were recorded from single cell nuclei and subjected to multivariate statistical analysis. Normal and abnormal ThinPrep® samples were discriminated based on the biochemical fingerprint of the cells using Principal Component Analysis (PCA). Principal Component Analysis - Linear Discriminant Analysis (PCA-LDA) was employed to build classification models based on either CIN or SIL terminology. This study has shown that Raman spectroscopy can be successfully applied to the study of routine cervical cytology samples from a cervical screening programme and that the use of CIN terminology resulted in improved sensitivity for high grade cases.
Collapse
Affiliation(s)
- I R Ramos
- DIT Centre for Radiation and Environmental Science, FOCAS Research Institute, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland. and School of Physics, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland
| | - A D Meade
- School of Physics, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland
| | - O Ibrahim
- DIT Centre for Radiation and Environmental Science, FOCAS Research Institute, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland. and School of Physics, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland
| | - H J Byrne
- FOCAS Research Institute, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland
| | - M McMenamin
- Cytopathology Department, Altnagelvin Hospital, Western Health and Social Care Trust, Derry, Northern Ireland, UK
| | - M McKenna
- Cytopathology Department, Altnagelvin Hospital, Western Health and Social Care Trust, Derry, Northern Ireland, UK
| | - A Malkin
- School of Biological Sciences, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland
| | - F M Lyng
- DIT Centre for Radiation and Environmental Science, FOCAS Research Institute, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland. and School of Physics, Dublin Institute of Technology, Kevin St, Dublin 8, Ireland
| |
Collapse
|
45
|
Biryukov J, Myers JC, McLaughlin-Drubin ME, Griffin HM, Milici J, Doorbar J, Meyers C. Mutations in HPV18 E1^E4 Impact Virus Capsid Assembly, Infectivity Competence, and Maturation. Viruses 2017; 9:v9120385. [PMID: 29257050 PMCID: PMC5744159 DOI: 10.3390/v9120385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 01/24/2023] Open
Abstract
The most highly expressed protein during the productive phase of the human papillomavirus (HPV) life cycle is E1^E4. Its full role during infection remains to be established. HPV E1^E4 is expressed during both the early and late stages of the virus life cycle and contributes to viral genome amplification. In an attempt to further outline the functions of E1^E4, and determine whether it plays a role in viral capsid assembly and viral infectivity, we examined wild-type E1^E4 as well as four E1^E4 truncation mutants. Our study revealed that HPV18 genomes containing the shortest truncated form of E1^E4, the 17/18 mutant, produced viral titers that were similar to wild-type virus and significantly higher compared to virions containing the three longer E1^E4 mutants. Additionally, the infectivity of virus containing the shortest E1^E4 mutation was equivalent to wild-type and significantly higher than the other three mutants. In contrast, infectivity was completely abrogated for virus containing the longer E1^E4 mutants, regardless of virion maturity. Taken together, our results indicate for the first time that HPV18 E1^E4 impacts capsid assembly and viral infectivity as well as virus maturation.
Collapse
Affiliation(s)
- Jennifer Biryukov
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (J.B.); (J.C.M.); (M.E.M.-D.); (J.M.)
| | - Jocelyn C. Myers
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (J.B.); (J.C.M.); (M.E.M.-D.); (J.M.)
| | - Margaret E. McLaughlin-Drubin
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (J.B.); (J.C.M.); (M.E.M.-D.); (J.M.)
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Heather M. Griffin
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (H.M.G.); (J.D.)
| | - Janice Milici
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (J.B.); (J.C.M.); (M.E.M.-D.); (J.M.)
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (H.M.G.); (J.D.)
| | - Craig Meyers
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (J.B.); (J.C.M.); (M.E.M.-D.); (J.M.)
- Correspondence: ; Tel.: +1-717-531-6240
| |
Collapse
|
46
|
Kranjec C, Holleywood C, Libert D, Griffin H, Mahmood R, Isaacson E, Doorbar J. Modulation of basal cell fate during productive and transforming HPV-16 infection is mediated by progressive E6-driven depletion of Notch. J Pathol 2017; 242:448-462. [PMID: 28497579 PMCID: PMC5601300 DOI: 10.1002/path.4917] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/13/2017] [Accepted: 04/24/2017] [Indexed: 11/10/2022]
Abstract
In stratified epithelia such as the epidermis, homeostasis is maintained by the proliferation of cells in the lower epithelial layers and the concomitant loss of differentiated cells from the epithelial surface. These differentiating keratinocytes progressively stratify and form a self‐regenerating multi‐layered barrier that protects the underlying dermis. In such tissue, the continual loss and replacement of differentiated cells also limits the accumulation of oncogenic mutations within the tissue. Inactivating mutations in key driver genes, such as TP53 and NOTCH1, reduce the proportion of differentiating cells allowing for the long‐term persistence of expanding mutant clones in the tissue. Here we show that through the expression of E6, HPV‐16 prevents the early fate commitment of human keratinocytes towards differentiation and confers a strong growth advantage to human keratinocytes. When E6 is expressed either alone or with E7, it promotes keratinocyte proliferation at high cell densities, through the combined inactivation of p53 and Notch1. In organotypic raft culture, the activity of E6 is restricted to the basal layer of the epithelium and is enhanced during the progression from productive to abortive or transforming HPV‐16 infection. Consistent with this, the expression of p53 and cleaved Notch1 becomes progressively more disrupted, and is associated with increased basal cell density and reduced commitment to differentiation. The expression of cleaved Notch1 is similarly disrupted also in HPV‐16‐positive cervical lesions, depending on neoplastic grade. When taken together, these data depict an important role of high‐risk E6 in promoting the persistence of infected keratinocytes in the basal and parabasal layers through the inactivation of gene products that are commonly mutated in non‐HPV‐associated neoplastic squamous epithelia. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Christian Kranjec
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK.,The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - Christina Holleywood
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - Diane Libert
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Heather Griffin
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK.,The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - Radma Mahmood
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - Erin Isaacson
- The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK.,The Francis Crick Institute Mill Hill Laboratory, The Ridgeway, Mill Hill, London, UK
| |
Collapse
|
47
|
Schmidt D. [Modern biomarkers for precancerous lesions of the uterine cervix : Histological-cytological correlation and use]. DER PATHOLOGE 2017; 37:534-541. [PMID: 27638536 DOI: 10.1007/s00292-016-0231-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The correlation between the cytological findings from the PAP smear and the histological outcome in cases where the cytological findings must be histologically verified, is an integral component of the German screening program for cervical cancer. These data are collected nationwide as part of a benchmarking process by the individual Associations of Statutory Health Insurance Physicians (KV) in the federal states and reported to the National Association of Statutory Health Insurance Physicians (KBV) in Berlin. In most cases there is a good correlation between cytology and histology but in some cases either a different grade of severity of cervical intraepithelial neoplasia (CIN) is found or the histological findings are negative. The reasons for a lack of correlation can be insufficient sampling in the cytology or the biopsy or a misinterpretation of the individual findings. Although the findings from H&E sections are considered to be the gold standard in the histological evaluation, it has long been known that the interobserver agreement in these preparations is only moderate. A significant improvement becomes apparent, firstly by the classification of cervical cancer precursors into low-grade and high-grade groups and secondly by the targeted application of biomarkers, in particular p16 and Ki-67, according to the recommendations of the lower anogenital squamous terminology standardization (LAST) project. The biomarkers p16 and Ki-67 should be used in the differential diagnostics between reactive and reparative alterations and for further differentiation of a CIN grade 2 but not to confirm a CIN grade 3. It is still unclear whether p16 is suitable as a prognostic marker for low-grade lesions.
Collapse
Affiliation(s)
- D Schmidt
- Institut für Pathologie, Gereonstr. 14a, 41747, Viersen, Deutschland.
| |
Collapse
|
48
|
Egawa N, Wang Q, Griffin HM, Murakami I, Jackson D, Mahmood R, Doorbar J. HPV16 and 18 genome amplification show different E4-dependence, with 16E4 enhancing E1 nuclear accumulation and replicative efficiency via its cell cycle arrest and kinase activation functions. PLoS Pathog 2017; 13:e1006282. [PMID: 28306742 PMCID: PMC5371391 DOI: 10.1371/journal.ppat.1006282] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/29/2017] [Accepted: 03/08/2017] [Indexed: 12/03/2022] Open
Abstract
To clarify E1^E4’s role during high-risk HPV infection, the E4 proteins of HPV16 and 18 were compared side by side using an isogenic keratinocyte differentiation model. While no effect on cell proliferation or viral genome copy number was observed during the early phase of either virus life cycle, time-course experiments showed that viral genome amplification and L1 expression were differently affected upon differentiation, with HPV16 showing a much clearer E4 dependency. Although E4 loss never completely abolished genome amplification, its more obvious contribution in HPV16 focused our efforts on 16E4. As previously suggested, in the context of the virus life cycle, 16E4s G2-arrest capability was found to contribute to both genome amplification success and L1 accumulation. Loss of 16E4 also lead to a reduced maintenance of ERK, JNK and p38MAPK activity throughout the genome amplifying cell layers, with 16E4 (but not 18E4) co-localizing precisely with activated cytoplasmic JNK in both wild type raft tissue, and HPV16-induced patient biopsy tissue. When 16E1 was co-expressed with E4, as occurs during genome amplification in vivo, the E1 replication helicase accumulated preferentially in the nucleus, and in transient replication assays, E4 stimulated viral genome amplification. Interestingly, a 16E1 mutant deficient in its regulatory phosphorylation sites no longer accumulated in the nucleus following E4 co-expression. E4-mediated stabilisation of 16E2 was also apparent, with E2 levels declining in organotypic raft culture when 16E4 was absent. These results suggest that 16E4-mediated enhancement of genome amplification involves its cell cycle inhibition and cellular kinase activation functions, with E4 modifying the activity and function of viral replication proteins including E1. These activities of 16E4, and the different kinase patterns seen here with HPV18, 31 and 45, may reflect natural differences in the biology and tropisms of these viruses, as well as differences in E4 function. In HPV induced lesions, the most abundant protein expressed in the productive stage of viral life cycle is E1^E4 (E4), with its expression being coincident with viral genome amplification. To clarify the role of E4 in the high-risk HPV life cycle, we carried out a comparative analysis of E4 function in HPV16 and 18 using an isogenic keratinocyte cell-line background. Our results show that E1^E4 contributes to virus genome replication efficiency and life cycle completion rather than being essential. These effects were seen more dramatically with HPV16. The difference between HPV16 and HPV18 in our system suggests important tropism differences between these viruses. HPV16 E4’s contribution to the virus life cycle is mediated by several activities, including its G2 arrest function, as well as its role in activating members of the MAPK pathway, including ERK, p38, and most notably pJNK. These 16 E4 functions facilitated the nuclear localization of the E1 virus helicase and enhanced E1/E2 dependent viral genome amplification as well as stabilising E2. We suspect that the massive accumulation of E4 in the upper epithelial layers may however underlie a more critical role for E4 post-genome amplification.
Collapse
Affiliation(s)
- Nagayasu Egawa
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
| | - Qian Wang
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
| | - Heather M. Griffin
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
| | - Isao Murakami
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
| | - Deborah Jackson
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
| | - Radma Mahmood
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Wakeham K, Kavanagh K, Cuschieri K, Millan D, Pollock KG, Bell S, Burton K, Reed NS, Graham SV. HPV status and favourable outcome in vulvar squamous cancer. Int J Cancer 2017; 140:1134-1146. [PMID: 27864932 DOI: 10.1002/ijc.30523] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/24/2016] [Accepted: 10/07/2016] [Indexed: 11/11/2022]
Abstract
It is universally accepted that high-risk human papillomavirus (HR-HPV) is the cause of cervical dysplasia and cancer. More recently, it has been shown that HPV is also a marker of clinical outcome in oropharyngeal cancer. However, contemporary information is lacking on both the prevalence of HPV infection in vulvar cancer (VSCC), its precursor lesion, vulvar intraepithelial neoplasia (VIN) and the influence of HPV-status on the prognosis of this malignancy. We have conducted a detailed population-based study to examine rates of progression of VIN to VSCC, type-specific HPV prevalence in vulvar disease and the influence of HPV status on clinical outcome in VSCC. We observed that the age at which women are diagnosed with VSCC is falling and there is a significant time gap between first diagnosis of VIN and progression to invasive disease. HR-HPV infection was detected in 87% (97/112) cases of VIN and 52% cases (32/62) of VSCC. The presence of HR-HPV in squamous intraepithelial lesion was associated with lower rates of progression to invasive cancer (hazard ratio, 0.22, p = 0.001). In the adjusted analysis, HR-HPV was associated with improved progression-free survival of VSCC compared to those with HPV negative tumours (hazard ratio, 0.32, p = 0.02).
Collapse
Affiliation(s)
- Katie Wakeham
- Sussex Cancer Centre, Brighton and Sussex University Hospital
- Institute of Cancer Sciences, University of Glasgow
| | - Kim Kavanagh
- Department of Mathematics and Statistics, University of Strathclyde
| | - Kate Cuschieri
- Scottish HPV Reference Laboratory, Royal Infirmary of Edinburgh
| | - David Millan
- Department of Pathology, The Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde
| | | | - Sarah Bell
- Department of Pathology, The Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde
| | - Kevin Burton
- Department of Gynaecological Oncology, Glasgow Royal Infirmary
| | | | - Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow
| |
Collapse
|
50
|
Yajid AI, Zakariah MA, Mat Zin AA, Othman NH. Potential Role of E4 Protein in Human Papillomavirus Screening: a Review. Asian Pac J Cancer Prev 2017; 18:315-319. [PMID: 28345325 PMCID: PMC5454721 DOI: 10.22034/apjcp.2017.18.2.315] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In 2006, cervical cancer was reported as the second most common cancer in women of Malaysia. This type of cancer has been shown to correlate with persistent high risk human papillomavirus (HPV) infection. Although HPV is well known to induce cervical cancer, knowledge of pathways that link the latent stage of the viral replication cycle to precancerous and cancerous stages remains incomplete. However, it is interesting to note that the virus can be isolated from tissues ranging from normal to low-grade squamous intraepithelial lesions as well as high-grade intraepithelial lesions (HSILs), thus prompting scientists to develop HPV detection methods for screening. Detection of HPV using viral proteins such as L1 and E1 is proposed to be very useful in assisting the management of high risk infection and cervical cancer. These tests however can lead to false positive results, largely due to the exisstence of asymptomatic or transient HPV infections within any given individual. Somes observation indicate that use of HPV proteins such as E6 and E7 might lead to false positive results. However, one particular HPV protein, E4 shows potential as an accurate marker of the tissue state following HPV infection. E4 expression has been shown to correlate with the levels of HPV DNA incorporation by the host. Thus, it is possible that E4 could serve as a useful marker to define stages of viral carcinogenesis.
Collapse
Affiliation(s)
- Aidy Irman Yajid
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.
| | | | | | | |
Collapse
|