1
|
Williams GA, Wu AA, Eugene HC, Tsai YC, Wong M, Nonogaki H, Roden RB, Hung CF, Wu TC, Vang R, Xing D. Clinicopathologic Features and Viral Status of Low-risk HPV6 and HPV11-Associated Squamous Cell Carcinoma of the Uterine Cervix and Vulva. Am J Surg Pathol 2025; 49:458-470. [PMID: 39886739 PMCID: PMC12003062 DOI: 10.1097/pas.0000000000002367] [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] [Indexed: 02/01/2025]
Abstract
Despite being designated as "noncarcinogenic" human papillomavirus (HPV) types, mono-infection with HPV6 or HPV11 has been found in squamous cell carcinomas (SCCs) at specific sites, including the larynx, penis, anus, and rarely, the lower female genital tract. The association between clinicopathologic features, viral status, and the carcinogenic mechanisms related to these low-risk HPVs remains unclear. The current study characterizes a series of low-risk HPV6 and HPV11-associated SCCs of the uterine cervix (6 cases) and vulva (2 cases). The diagnosis of SCC was made through the identification of stromal invasion in 6 cases. In case 2, the diagnosis of cancer was made after metastases to the sigmoid colon and liver. The patient in case 6 was diagnosed with intramucosal papillary SCC given multiple recurrences. While all tumors displayed a similar verruco-papillary architecture, the cytologic features, and immunostaining patterns suggest 2 groups of lesions: one with high-grade cytology and a high Ki-67 proliferation index (>60% of lesional cells), and the other with low-grade cytology and a low Ki-67 (20% to 30% of lesional cells). The detection of HPV6 in 7 of 8 cases underscores its critical role in carcinogenesis at these anatomic sites. Case 8 represented the only patient who was infected with HPV11 and who had a well-controlled human immunodeficiency virus infection. Correlating with viral status, all cases, except case 7, demonstrated a negative or focal p16 staining pattern. In case 7, despite a block pattern of p16 staining often seen in predicting high-risk HPV, we employed several methods to confirm HPV6 as the sole HPV infection. Although this descriptive study does not establish an etiological mechanism for how HPV6/11 leads to malignant transformation, our results exclude the possibility of viral integration through a quantitative polymerase chain reaction-based analysis of the E2/E6 ratio. Our study highlights and expands upon the clinicopathologic features of a distinct group of low-risk HPV6/11-associated SCCs in the cervix and vulva. Although rare, recognizing this group of lesions is important for pathologists and oncologists, as it provides a basis for guiding appropriate prevention strategies and treatment modalities based on the viral type.
Collapse
Affiliation(s)
- Guy A. Williams
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Annie A. Wu
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Henrietta C. Eugene
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ya-Chea Tsai
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Margaret Wong
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Hiro Nonogaki
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Richard B.S. Roden
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Tzyy-Choou Wu
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Russell Vang
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Deyin Xing
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
2
|
De Vleeschauwer SI, van de Ven M, Oudin A, Debusschere K, Connor K, Byrne AT, Ram D, Rhebergen AM, Raeves YD, Dahlhoff M, Dangles-Marie V, Hermans ER. OBSERVE: guidelines for the refinement of rodent cancer models. Nat Protoc 2024; 19:2571-2596. [PMID: 38992214 DOI: 10.1038/s41596-024-00998-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 02/23/2024] [Indexed: 07/13/2024]
Abstract
Existing guidelines on the preparation (Planning Research and Experimental Procedures on Animals: Recommendations for Excellence (PREPARE)) and reporting (Animal Research: Reporting of In Vivo Experiments (ARRIVE)) of animal experiments do not provide a clear and standardized approach for refinement during in vivo cancer studies, resulting in the publication of generic methodological sections that poorly reflect the attempts made at accurately monitoring different pathologies. Compliance with the 3Rs guidelines has mainly focused on reduction and replacement; however, refinement has been harder to implement. The Oncology Best-practices: Signs, Endpoints and Refinements for in Vivo Experiments (OBSERVE) guidelines are the result of a European initiative supported by EurOPDX and INFRAFRONTIER, and aim to facilitate the refinement of studies using in vivo cancer models by offering robust and practical recommendations on approaches to research scientists and animal care staff. We listed cancer-specific clinical signs as a reference point and from there developed sets of guidelines for a wide variety of rodent models, including genetically engineered models and patient derived xenografts. In this Consensus Statement, we systematically and comprehensively address refinement and monitoring approaches during the design and execution of murine cancer studies. We elaborate on the appropriate preparation of tumor-initiating biologicals and the refinement of tumor-implantation methods. We describe the clinical signs to monitor associated with tumor growth, the appropriate follow-up of animals tailored to varying clinical signs and humane endpoints, and an overview of severity assessment in relation to clinical signs, implantation method and tumor characteristics. The guidelines provide oncology researchers clear and robust guidance for the refinement of in vivo cancer models.
Collapse
Affiliation(s)
| | - Marieke van de Ven
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anaïs Oudin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Karlijn Debusschere
- Animal Core Facility VUB, Brussels, Belgium
- Core ARTH Animal Facilities, Medicine and Health Sciences Ghent University, Ghent, Belgium
| | - Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Doreen Ram
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Els R Hermans
- Laboratory Animal Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Chambers L, Haight P, Chalif J, Mehra Y, Spakowicz D, Backes FJ, Cosgrove CM, O’Malley DM, Vargas R, Corr BR, Bae-Jump VL, Arend RC. Bridging the Gap from Bench to Bedside: A Call for In Vivo Preclinical Models to Advance Endometrial Cancer and Cervical Cancer Immuno-oncology Research. Clin Cancer Res 2024; 30:2905-2909. [PMID: 38662438 PMCID: PMC11250463 DOI: 10.1158/1078-0432.ccr-23-2570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/29/2023] [Accepted: 04/23/2024] [Indexed: 07/16/2024]
Abstract
Advanced-stage endometrial and cervical cancers are associated with poor outcomes despite contemporary advances in surgical techniques and therapeutics. Recent clinical trial results have led to a shift in the treatment paradigm for both malignancies, in which immunotherapy is now incorporated as the standard of care up front for most patients with advanced endometrial and cervical cancers as the standard of care. Impressive response rates have been observed, but unfortunately, a subset of patients do not benefit from immunotherapy, and survival remains poor. Continued preclinical research and clinical trial development are crucial for our understanding of resistance mechanisms to immunotherapy and maximization of therapeutic efficacy. In this setting, syngeneic models are preferred over xenograft models as they allow for the evaluation of the tumor-immune interaction in an immunocompetent host, most closely mimicking the tumor-immune interaction in patients with cancer. Unfortunately, significant disparities exist about syngeneic models in gynecologic malignancy, in which queries from multiple large bioscience companies confirm no commercial availability of endometrial or cervical cancer syngeneic cell lines. Published data exist about the recent development of several endometrial and cervical cancer syngeneic cell lines, warranting further investigation. Closing the disparity gap for preclinical models in endometrial and cervical cancers will support physician scientists, basic and translational researchers, and clinical trialists who are dedicated to improving outcomes for our patients with advanced disease and poor prognosis.
Collapse
Affiliation(s)
- Laura Chambers
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, James Hospital and Solove Research Institute
| | - Paulina Haight
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, James Hospital and Solove Research Institute
| | - Julia Chalif
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, James Hospital and Solove Research Institute
| | - Yogita Mehra
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Daniel Spakowicz
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Floor J. Backes
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, James Hospital and Solove Research Institute
| | - Casey M. Cosgrove
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, James Hospital and Solove Research Institute
| | - David M. O’Malley
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, James Hospital and Solove Research Institute
| | - Roberto Vargas
- Division of Gynecologic Oncology, The Cleveland Clinic Foundation, Cleveland, OH
| | - Bradley R. Corr
- Division of Gynecologic Oncology, University of Colorado, Denver, CO
| | - Victoria L. Bae-Jump
- Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C. Arend
- Department of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
4
|
Xiurong D, Xin Z, Neng Y, Li D, Yanzhou W, Kaijian L, Zhiqing L. Genetically engineered mouse model of HPV16 E6-E7 with vaginal-cervical intraepithelial neoplasia and decreased immunity. Heliyon 2024; 10:e29881. [PMID: 38765051 PMCID: PMC11096975 DOI: 10.1016/j.heliyon.2024.e29881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/21/2024] Open
Abstract
Objective To construct models of high-risk human papillomavirus (HPV) infection with precancerous lesions or cervical cancer and explore the immune function. Methods Using CRISPR/Cas9, the expression vector HPV16-E6-E7-Rosa26 was microinjected into fertilized eggs of C57BL/6 N mice using homologous recombination, and the F0 generation was obtained for reproduction. Then, the formation of precancerous lesions was promoted via intramuscular injection of estradiol. Presence of precancerous cervical-vaginal intraepithelial lesions, Ki67 and p16 expression levels, and CD8+ T cell proportions in the spleen were evaluated. Results Two F0 generation mice exhibited correct the homologous recombination. Seven positive mice were identified in the F1 generation. After breeding and mating, 25 homozygous and 11 heterozygous HPV16-E6-E7-engineered mice were obtained from the F2 generation. After estradiol benzoate treatment, the cervical-vaginal epithelium appeared as precancerous lesions with positive Ki67 and p16 expression. The percentage of CD8+ T cells decreased. Conclusion HPV16-E6-E7-Rosa26 induced low immune function in mice, and provides a good model for the basic research of the mechanisms of action of HPV infection-associated precancerous lesions or cervical cancer.
Collapse
Affiliation(s)
- Du Xiurong
- The Army Medical University, Chongqing, 400038, China
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| | - Zhou Xin
- The Army Medical University, Chongqing, 400038, China
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| | - Yang Neng
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| | - Deng Li
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| | - Wang Yanzhou
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| | - Ling Kaijian
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| | - Liang Zhiqing
- The Army Medical University, Chongqing, 400038, China
- Department of Obstetrics and Gynecology, Southwest Hospital of the Third Military Medical University (The first Affiliated Hospital of the Army Medical University), Chongqing, 400038, China
| |
Collapse
|
5
|
Bikorimana J, Abusarah J, Gonçalves M, Farah R, Saad W, Talbot S, Stanga D, Beaudoin S, Plouffe S, Rafei M. An engineered Accum-E7 protein-based vaccine with dual anti-cervical cancer activity. Cancer Sci 2024; 115:1102-1113. [PMID: 38287511 PMCID: PMC11007051 DOI: 10.1111/cas.16096] [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: 05/12/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 01/31/2024] Open
Abstract
Worldwide prevalence of cervical cancer decreased significantly with the use of human papilloma virus (HPV)-targeted prophylactic vaccines. However, these multivalent antiviral vaccines are inert against established tumors, which leave patients with surgical ablative options possibly resulting in long-term reproductive complications and morbidity. In an attempt to bypass this unmet medical need, we designed a new E7 protein-based vaccine formulation using Accum™, a technology platform designed to promote endosome-to-cytosol escape as a means to enhance protein accumulation in target cells. Prophylactic vaccination of immunocompetent mice using the Accum-E7 vaccine (aE7) leads to complete protection from cervical cancer despite multiple challenges conducted with ascending C3.43 cellular doses (0.5-, 1.0-, and 2.0 × 106 cells). Moreover, the humoral response induced by aE7 was higher in magnitude compared with naked E7 protein vaccination and displayed potent inhibitory effects on C3.43 proliferation in vitro. When administered therapeutically to animals with pre-established C3.43 or Tal3 tumors, the vaccine-induced response synergized with multiple immune checkpoint blockers (anti-PD-1, anti-CTLA4, and anti-CD47) to effectively control tumor growth. Mechanistically, the observed therapeutic effect requires cross-presenting dendritic cells as well as CD8 T cells predominantly, with a non-negligible role played by both CD4+ and CD19+ lymphocytes. good laboratory practice (GLP) studies revealed that aE7 is immunogenic and well tolerated by immunocompetent mice with no observed adverse effects despite the use of a fourfold exceeding dose. In a nutshell, aE7 represents an ideal vaccine candidate for further clinical development as it uses a single engineered protein capable of exhibiting both prophylactic and therapeutic activity.
Collapse
Affiliation(s)
- Jean‐Pierre Bikorimana
- Department of Microbiology, Infectious Diseases and ImmunologyUniversité de MontréalMontréalQuebecCanada
| | - Jamilah Abusarah
- Department of Pharmacology and PhysiologyUniversité de MontréalMontréalQuebecCanada
| | - Marina Gonçalves
- Department of Molecular BiologyUniversité de MontréalMontréalQuebecCanada
| | - Roudy Farah
- Department of Microbiology, Infectious Diseases and ImmunologyUniversité de MontréalMontréalQuebecCanada
| | - Wael Saad
- Department of Pharmacology and PhysiologyUniversité de MontréalMontréalQuebecCanada
| | - Sebastien Talbot
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Daniela Stanga
- Defence Therapeutics Inc.VancouverBritish ColumbiaCanada
| | - Simon Beaudoin
- Defence Therapeutics Inc.VancouverBritish ColumbiaCanada
| | | | - Moutih Rafei
- Department of Microbiology, Infectious Diseases and ImmunologyUniversité de MontréalMontréalQuebecCanada
- Department of Pharmacology and PhysiologyUniversité de MontréalMontréalQuebecCanada
- Department of Molecular BiologyUniversité de MontréalMontréalQuebecCanada
| |
Collapse
|
6
|
Skelin J, Tomaić V. Comparative Analysis of Alpha and Beta HPV E6 Oncoproteins: Insights into Functional Distinctions and Divergent Mechanisms of Pathogenesis. Viruses 2023; 15:2253. [PMID: 38005929 PMCID: PMC10674601 DOI: 10.3390/v15112253] [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: 10/27/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Human papillomaviruses (HPVs) represent a diverse group of DNA viruses that infect epithelial cells of mucosal and cutaneous tissues, leading to a wide spectrum of clinical outcomes. Among various HPVs, alpha (α) and beta (β) types have garnered significant attention due to their associations with human health. α-HPVs are primarily linked to infections of the mucosa, with high-risk subtypes, such as HPV16 and HPV18, being the major etiological agents of cervical and oropharyngeal cancers. In contrast, β-HPVs are predominantly associated with cutaneous infections and are commonly found on healthy skin. However, certain β-types, notably HPV5 and HPV8, have been implicated in the development of non-melanoma skin cancers in immunocompromised individuals, highlighting their potential role in pathogenicity. In this review, we comprehensively analyze the similarities and differences between α- and β-HPV E6 oncoproteins, one of the major drivers of viral replication and cellular transformation, and how these impact viral fitness and the capacity to induce malignancy. In particular, we compare the mechanisms these oncoproteins use to modulate common cellular processes-apoptosis, DNA damage repair, cell differentiation, and the immune response-further shedding light on their shared and distinct features, which enable them to replicate at divergent locations of the human body and cause different types of cancer.
Collapse
Affiliation(s)
| | - Vjekoslav Tomaić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia;
| |
Collapse
|
7
|
Peng S, Tu HF, Cheng M, Hu MH, Tsai HL, Tsai YC, Koenig C, Brayton C, Wang H, Chang YN, Arend RC, Levinson K, Roden RBS, Wu TC, Hung CF. Immune responses, therapeutic anti-tumor effects, and tolerability upon therapeutic HPV16/18 E6/E7 DNA vaccination via needle-free biojector. mBio 2023; 14:e0212123. [PMID: 37791765 PMCID: PMC10653862 DOI: 10.1128/mbio.02121-23] [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: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Respectively, HPV16 and HPV18 cause 50% and 20% of cervical cancer cases globally. Viral proteins E6 and E7 are obligate drivers of oncogenic transformation. We recently developed a candidate therapeutic DNA vaccine, pBI-11, that targets HPV16 and HPV18 E6 and E7. Single-site intramuscular delivery of pBI-11 via a needle elicited therapeutic anti-tumor effects in mice and is now being tested in high-risk human papillomavirus+ head and neck cancer patients (NCT05799144). Needle-free biojectors such as the Tropis device show promise due to ease of administration, high patient acceptability, and the possibility of improved delivery. For example, vaccination of patients with the ZyCoV-D DNA vaccine using the Tropis device is effective against COVID19, well tolerated, and licensed. Here we show that split-dose, multi-site administration and intradermal delivery via the Tropis biojector increase the delivery of pBI-11 DNA vaccine, enhance HPV antigen-specific CD8+ T-cell responses, and improve anti-tumor therapeutic effects, suggesting its translational potential to treat HPV16/18 infection and disease.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle Cheng
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ming-Hung Hu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Department of Oncology Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chelsea Koenig
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hao Wang
- Department of Oncology Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Rebecca C. Arend
- Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kimberly Levinson
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B. S. Roden
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - T. C. Wu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Ramos da Silva J, Bitencourt Rodrigues K, Formoso Pelegrin G, Silva Sales N, Muramatsu H, de Oliveira Silva M, Porchia BFMM, Moreno ACR, Aps LRMM, Venceslau-Carvalho AA, Tombácz I, Fotoran WL, Karikó K, Lin PJC, Tam YK, de Oliveira Diniz M, Pardi N, de Souza Ferreira LC. Single immunizations of self-amplifying or non-replicating mRNA-LNP vaccines control HPV-associated tumors in mice. Sci Transl Med 2023; 15:eabn3464. [PMID: 36867683 DOI: 10.1126/scitranslmed.abn3464] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
As mRNA vaccines have proved to be very successful in battling the coronavirus disease 2019 (COVID-19) pandemic, this new modality has attracted widespread interest for the development of potent vaccines against other infectious diseases and cancer. Cervical cancer caused by persistent human papillomavirus (HPV) infection is a major cause of cancer-related deaths in women, and the development of safe and effective therapeutic strategies is urgently needed. In the present study, we compared the performance of three different mRNA vaccine modalities to target tumors associated with HPV-16 infection in mice. We generated lipid nanoparticle (LNP)-encapsulated self-amplifying mRNA as well as unmodified and nucleoside-modified non-replicating mRNA vaccines encoding a chimeric protein derived from the fusion of the HPV-16 E7 oncoprotein and the herpes simplex virus type 1 glycoprotein D (gDE7). We demonstrated that single low-dose immunizations with any of the three gDE7 mRNA vaccines induced activation of E7-specific CD8+ T cells, generated memory T cell responses capable of preventing tumor relapses, and eradicated subcutaneous tumors at different growth stages. In addition, the gDE7 mRNA-LNP vaccines induced potent tumor protection in two different orthotopic mouse tumor models after administration of a single vaccine dose. Last, comparative studies demonstrated that all three gDE7 mRNA-LNP vaccines proved to be superior to gDE7 DNA and gDE7 recombinant protein vaccines. Collectively, we demonstrated the immunogenicity and therapeutic efficacy of three different mRNA vaccines in extensive comparative experiments. Our data support further evaluation of these mRNA vaccines in clinical trials.
Collapse
Affiliation(s)
- Jamile Ramos da Silva
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karine Bitencourt Rodrigues
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Guilherme Formoso Pelegrin
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Natiely Silva Sales
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Hiromi Muramatsu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mariângela de Oliveira Silva
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Bruna F M M Porchia
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.,Laboratory of Tumor Immunology, Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo, SP 05508-000, Brazil.,ImunoTera Soluções Terapêuticas Ltda., São Paulo, SP 05508-000, Brazil
| | - Ana Carolina Ramos Moreno
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Luana Raposo M M Aps
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.,ImunoTera Soluções Terapêuticas Ltda., São Paulo, SP 05508-000, Brazil
| | - Aléxia Adrianne Venceslau-Carvalho
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - István Tombácz
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wesley Luzetti Fotoran
- Department of Parasitology, Institute for Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | | | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T1Z3, Canada
| | - Mariana de Oliveira Diniz
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luís Carlos de Souza Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.,Scientific Platform Pasteur USP, University of São Paulo, São Paulo, SP, 05508-020, Brazil
| |
Collapse
|
9
|
Peng S, Xing D, Ferrall L, Tsai YC, Hung CF, Wu TC. Identification of human MHC-I HPV18 E6/E7-specific CD8 + T cell epitopes and generation of an HPV18 E6/E7-expressing adenosquamous carcinoma in HLA-A2 transgenic mice. J Biomed Sci 2022; 29:80. [PMID: 36224625 PMCID: PMC9554842 DOI: 10.1186/s12929-022-00864-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 09/28/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Human Papillomavirus type 18 (HPV18) is a high-risk HPV that is commonly associated with cervical cancer. HPV18 oncogenes E6 and E7 are associated with the malignant transformation of cells, thus the identification of human leukocyte antigen (HLA)-restricted E6/E7 peptide-specific CD8 + T cell epitopes and the creation of a HPV18 E6/E7 expressing cervicovaginal tumor in HLA-A2 transgenic mice will be significant for vaccine development. METHODS In the below study, we characterized various human HLA class I-restricted HPV18 E6 and E7-specific CD8 + T cells mediated immune responses in HLA class I transgenic mice using DNA vaccines encoding HPV18E6 and HPV18E7. We then confirmed HLA-restricted E6/E7 specific CD8 + T cell epitopes using splenocytes from vaccinated mice stimulated with HPV18E6/E7 peptides. Furthermore, we used oncogenic DNA plasmids encoding HPV18E7E6(delD70), luciferase, cMyc, and AKT to create a spontaneous cervicovaginal carcinoma model in HLA-A2 transgenic mice. RESULTS Therapeutic HPV18 E7 DNA vaccination did not elicit any significant CD8 + T cell response in HLA-A1, HLA-24, HLA-B7, HLA-B44 transgenic or wild type C57BL/6 mice, but it did generate a strong HLA-A2 and HLA-A11 restricted HPV18E7-specific CD8 + T cell immune response. We found that a single deletion of aspartic acid (D) at location 70 in HPV18E6 DNA abolishes the presentation of HPV18 E6 peptide (aa67-75) by murine MHC class I. We found that the DNA vaccine with this mutant HPV18 E6 generated E6-specific CD8 + T cells in HLA-A2. HLA-A11, HLA-A24 and HLA-b40 transgenic mice. Of note, HLA-A2 restricted, HPV18 E7 peptide (aa7-15)- and HPV18 E6 peptide (aa97-105)-specific epitopes are endogenously processed by HPV18 positive Hela-AAD (HLA-A*0201/Dd) cells. Finally, we found that injection of DNA plasmids encoding HPV18E7E6(delD70), AKT, cMyc, and SB100 can result in the development of adenosquamous carcinoma in the cervicovaginal tract of HLA-A2 transgenic mice. CONCLUSIONS We characterized various human HLA class I-restricted HPV18 E6/E7 peptide specific CD8 + T cell epitopes in human HLA class I transgenic mice. We demonstrated that HPV18 positive Hela cells expressing chimeric HLA-A2 (AAD) do present both HLA-A2-restricted HPV18 E7 (aa7-15)- and HPV18 E6 (aa97-105)-specific CD8 + T cell epitopes. A mutant HPV18E6 that had a single deletion at location 70 obliterates the E6 presentation by murine MHC class I and remains oncogenic. The identification of these human MHC restricted HPV antigen specific epitopes as well as the HPV18E6/E7 expressing adenosquamous cell carcinoma model may have significant future translational potential.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, USA
| | - Deyin Xing
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, USA
| | - Louise Ferrall
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, USA
| | - Ya-Chea Tsai
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, MD, USA.
- The Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans St., Baltimore, MD, 21231, USA.
| | - T-C Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University, Baltimore, MD, USA.
- The Johns Hopkins Medical Institutions, CRB II Room 309, 1550 Orleans St., Baltimore, MD, 21231, USA.
| |
Collapse
|
10
|
Yilmaz V, Louca P, Potamiti L, Panayiotidis M, Strati K. A novel lineage-tracing mouse model for studying early MmuPV1 infections. eLife 2022; 11:72638. [PMID: 35533001 PMCID: PMC9084889 DOI: 10.7554/elife.72638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Human papillomaviruses are DNA viruses that ubiquitously infect humans and have been associated with hyperproliferative lesions. The recently discovered mouse specific papillomavirus (MmuPV1) provides the opportunity to study papillomavirus infections in vivo in the context of a common laboratory mouse model (Mus musculus). To date, a major challenge in the field has been the lack of tools to identify, observe, and characterize individually the papillomavirus hosting cells and also trace the progeny of these cells over time. Here, we present the successful generation of an in vivo lineage-tracing model of MmuPV1-harboring cells and their progeny by means of genetic reporter activation. Following the validation of the system both in vitro and in vivo, we used it to provide a proof-of-concept of its utility. Using flow-cytometry analysis, we observed increased proliferation dynamics and decreased MHC-I cell surface expression in MmuPV1-treated tissues which could have implications in tissue regenerative capacity and ability to clear the virus. This model is a novel tool to study the biology of the MmuPV1 host-pathogen interactions.
Collapse
Affiliation(s)
- Vural Yilmaz
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Panayiota Louca
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Louiza Potamiti
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Mihalis Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Katerina Strati
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
11
|
Peng S, Xing D, Ferrall L, Tsai YC, Roden RBS, Hung CF, Wu TC. Development of a Spontaneous HPV16 E6/E7-Expressing Head and Neck Squamous Cell Carcinoma in HLA-A2 Transgenic Mice. mBio 2022; 13:e0325221. [PMID: 35089069 PMCID: PMC8725581 DOI: 10.1128/mbio.03252-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/23/2021] [Indexed: 01/10/2023] Open
Abstract
Human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) is a growing global health problem. HPV16 has been attributed to a majority of HPV-associated HNSCCs. In order to test candidate immunotherapies, we developed a spontaneous HPV16-driven HNSCC model in HLA-A2 (AAD) transgenic mice. We sought to eliminate the confounding effects of dominant HPV antigen presentation through murine major histocompatibility complex class I (MHC-I) via epitope mutagenesis (without compromising tumorigenicity). We generated HPV16 E6(R55K)(delK75) and E7(N53S) expression constructs with mutations in known dominant H-2Db epitopes and characterized their presentation through murine and human MHC-I molecules using in vitro and in vivo activation of HPV16 E6/E7 antigen-specific CD8+ T cells. In addition, we tested the ability of E6(R55K)(delK75) and E7(N53S) for oncogenicity. The mutated E7(N53S) abolished the presentation of murine H-2Db-restricted HPV16 E7 peptide (i.e., amino acids [aa] 49 to 57) cytotoxic T lymphocyte (CTL) epitope and resulted in HLA-A2-restricted presentation of the HPV16 E7 (aa 11 to 20)-specific CTL epitope. The mutated E6(R55K)(delK75) abolished the activation of murine MHC-I-restricted E6-specific CD8+ T cell-mediated immune responses in C57BL/6 mice. In addition, the vaccination led to the activation of human HLA-A2-restricted E6-specific CD8+ T cell-mediated immune responses in HLA-A2 (AAD) transgenic mice. Injection of DNA plasmids encoding LucE7(N53S)E6(R55K)(delK75), AKT, c-Myc, and SB100 followed by electroporation results in development of squamous cell carcinoma in the oral/pharyngeal cavity of all of the HLA-A2 (AAD) transgenic mice (5/5), with 2/5 tumor-bearing mice developing metastatic carcinoma in the neck lymph nodes. IMPORTANCE Our data indicate that mutated HPV16 E6(R55K)(delK75) and mutated HPV16 E7(N53S) DNA abolishes the presentation of HPV16 E6 and E7 through murine MHC-I and results in their presentation through human HLA-A2 molecules. Additionally, the mutated HPV16 E6 and E7 remain oncogenic. Our approach is potentially applicable to different human MHC-I transgenic mice for the identification of human MHC-I restricted HPV16 E6/E7-specific CTL epitopes as well as the generation of spontaneous HPV E6/E7-expressing oral/pharyngeal carcinoma.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Deyin Xing
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Louise Ferrall
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Ya-Chea Tsai
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B. S. Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - T.-C. Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Avdoshina DV, Kondrashova AS, Belikova MG, Bayurova EO. Murine Models of Chronic Viral Infections and Associated Cancers. Mol Biol 2022; 56:649-667. [PMID: 36217336 PMCID: PMC9534466 DOI: 10.1134/s0026893322050028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/07/2022]
Abstract
Viruses are now recognized as bona fide etiologic factors of human cancer. Carcinogenic viruses include Epstein– Barr virus (EBV), high-risk human papillomaviruses (HPVs), hepatitis B virus (HBV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), human immunodeficiency virus type 1 (HIV-1, indirectly), and several candidate human cancer viruses. It is estimated that 15% of all human tumors worldwide are caused by viruses. Tumor viruses establish long-term persistent infections in humans, and cancer is an accidental side effect of viral replication strategies. Viruses are usually not complete carcinogens, supporting the concept that cancer results from the accumulation of multiple cooperating events, in which human cancer viruses display different, often opposing roles. The laboratory mouse Mus musculus is one of the best in vivo experimental systems for modeling human pathology, including viral infections and cancer. However, mice are unsusceptible to infection with the known carcinogenic viruses. Many murine models were developed to overcome this limitation and to address various aspects of virus-associated carcinogenesis, from tumors resulting from xenografts of human tissues and cells, including cancerous and virus infected, to genetically engineered mice susceptible to viral infections and associated cancer. The review considers the main existing models, analyzes their advantages and drawbacks, describes their applications, outlines the prospects of their further development.
Collapse
Affiliation(s)
- D. V. Avdoshina
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia
| | - A. S. Kondrashova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia
| | - M. G. Belikova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia ,Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia ,Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - E. O. Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Products, Russian Academy of Sciences (Polio Institute), 108819 Moscow, Russia ,Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| |
Collapse
|
13
|
Yoshida H, Shiraishi K, Kato T. Molecular Pathology of Human Papilloma Virus-Negative Cervical Cancers. Cancers (Basel) 2021; 13:cancers13246351. [PMID: 34944973 PMCID: PMC8699825 DOI: 10.3390/cancers13246351] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cervical cancer is the fourth most common cancer in women worldwide and is predominantly caused by infection with human papillomavirus (HPV). However, a small subset of cervical cancers tests negative for HPV, including true HPV-independent cancers and false-negative cases. True HPV-negative cancers appear to be more prevalent in certain pathological adenocarcinoma subtypes, such as gastric- and clear-cell-type adenocarcinomas. Moreover, HPV-negative cervical cancers have proven to be a biologically distinct tumor subset that follows a different pathogenetic pathway to HPV-associated cervical cancers. HPV-negative cervical cancers are often diagnosed at an advanced stage with a poor prognosis and are expected to persist in the post-HPV vaccination era; therefore, it is important to understand HPV-negative cancers. In this review, we provide a concise overview of the molecular pathology of HPV-negative cervical cancers, with a focus on their definitions, the potential causes of false-negative HPV tests, and the histology, genetic profiles, and pathogenesis of HPV-negative cancers.
Collapse
Affiliation(s)
- Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Correspondence: ; Tel.: +81-3-3457-5201
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo 104-0045, Japan;
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| |
Collapse
|
14
|
Tseng SH, Liu L, Peng S, Kim J, Ferrall L, Hung CF, Wu TC. Control of Spontaneous HPV16 E6/E7 Expressing Oral Cancer in HLA-A2 (AAD) Transgenic Mice with Therapeutic HPV DNA Vaccine. J Biomed Sci 2021; 28:63. [PMID: 34517865 PMCID: PMC8436567 DOI: 10.1186/s12929-021-00759-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/06/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Human Papillomavirus type 16 (HPV16) has been associated with a subset of head and neck cancers. Two HPV encoded oncogenic proteins, E6 and E7, are important for the malignant progression of HPV-associated cancers. A spontaneous HPV16 E6/E7-expressing oral tumor model in human HLA-A2 (AAD) transgenic mice will be important for the development of therapeutic HPV vaccines for the control of HPV-associated head and neck cancers. METHODS In the current studies, we characterized the HLA-A2 restricted HPV16 E7-specific CD8 + T cell mediated immune responses in the HLA-A2 (AAD) transgenic mice using a therapeutic naked DNA vaccine encoding calreticulin (CRT) linked to a mutated E7(N53S). We also employed oncogenic DNA plasmids that encoded HPV16E6/E7/Luc, NRasG12V, and sleeping beauty transposase for the transfection into the submucosal of oral cavity of the transgenic mice with electroporation to create a spontaneous oral tumor. Furthermore, we characterized the therapeutic antitumor effects of CRT/E7(N53S) DNA vaccine using the spontaneous HPV16 E6/E7-expressing oral tumor model in HLA-A2 (AAD) transgenic mice. RESULTS We found that CRT/E7(N53S) DNA vaccine primarily generated human HPV16 E7 peptide (aa11-20) specific CD8 + T cells, as compared to the wild-type CRT/E7 vaccine, which primarily generated murine H-2Db restricted E7 peptide (aa49-57) specific CD8 + T cell responses. We also observed transfection of the oncogenic DNA plasmids with electroporation generated spontaneous oral tumor in all of the injected mice. Additionally, treatment with CRT/E7(N53S) DNA vaccine intramuscularly followed by electroporation resulted in significant antitumor effects against the spontaneous HPV16 E6/E7-expressing oral tumors in HLA-A2 (AAD) transgenic mice. CONCLUSIONS Taken together, the data indicated that the combination of HPV16 E6/E7-expressing DNA, NRasG12V DNA and DNA encoding sleeping beauty transposase is able to generate spontaneous oral tumor in HLA-A2 (AAD) transgenic mice, which can be successfully controlled by treatment with CRT/E7(N53S) DNA vaccine. The translational potential of our studies are discussed.
Collapse
Affiliation(s)
- Ssu-Hsueh Tseng
- Department of Pathology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA
| | - Li Liu
- Department of Pathology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA
| | - Shiwen Peng
- Department of Pathology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA
| | - Jinhwi Kim
- Department of Obstetrics and Gynecology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-Ro, Uijeongbu, Gyeonggi-do, 11765, Republic of Korea
| | - Louise Ferrall
- Department of Pathology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Department of Oncology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Department of Obstetrics and Gynecology, CRB II, Johns Hopkins University, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Departments of Pathology, Oncology, and Obstetrics and Gynecology, The Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans St, Baltimore, MD, 21231, USA.
| | - T -C Wu
- Department of Pathology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Department of Oncology, Johns Hopkins University, CRB II, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Department of Obstetrics and Gynecology, CRB II, Johns Hopkins University, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Department of Molecular Microbiology and Immunology, CRB II, Johns Hopkins University, 1550 Orleans St, Baltimore, MD, 21287, USA. .,Departments of Pathology, Oncology, Obstetrics and Gynecology, and Molecular Microbiology and Immunology, The Johns Hopkins Medical Institutions, CRB II Room 309, 1550 Orleans St, Baltimore, MD, 21231, USA.
| |
Collapse
|