1
|
Liao HC, Liu SJ. Advances in nucleic acid-based cancer vaccines. J Biomed Sci 2025; 32:10. [PMID: 39833784 PMCID: PMC11748563 DOI: 10.1186/s12929-024-01102-w] [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: 07/07/2024] [Accepted: 11/05/2024] [Indexed: 01/22/2025] Open
Abstract
Nucleic acid vaccines have emerged as crucial advancements in vaccine technology, particularly highlighted by the global response to the COVID-19 pandemic. The widespread administration of mRNA vaccines against COVID-19 to billions globally marks a significant milestone. Furthermore, the approval of an mRNA vaccine for Respiratory Syncytial Virus (RSV) this year underscores the versatility of this technology. In oncology, the combination of mRNA vaccine encoding neoantigens and immune checkpoint inhibitors (ICIs) has shown remarkable efficacy in eliciting protective responses against diseases like melanoma and pancreatic cancer. Although the use of a COVID-19 DNA vaccine has been limited to India, the inherent stability at room temperature and cost-effectiveness of DNA vaccines present a viable option that could benefit developing countries. These advantages may help DNA vaccines address some of the challenges associated with mRNA vaccines. Currently, several trials are exploring the use of DNA-encoded neoantigens in combination with ICIs across various cancer types. These studies highlight the promising role of nucleic acid-based vaccines as the next generation of immunotherapeutic agents in cancer treatment. This review will delve into the recent advancements and current developmental status of both mRNA and DNA-based cancer vaccines.
Collapse
Affiliation(s)
- Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 307378, Taiwan.
| |
Collapse
|
2
|
Mohammadi M, Saha A, Giles-Davis W, Xiang Z, Novikov M, Hasanpourghadi M, C. J. Ertl H. Preclinical Immunogenicity and Efficacy Studies for Therapeutic Vaccines for Human Papillomavirus-Type-16-Associated Cancer. Vaccines (Basel) 2024; 12:616. [PMID: 38932345 PMCID: PMC11209626 DOI: 10.3390/vaccines12060616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The objective of this study was to conduct preclinical immunogenicity and efficacy studies with several therapeutic vaccines for human papillomavirus (HPV)-16-associated cancers expressing the early antigens E5, E6, and E7 with or without E2. The viral oncoproteins were either expressed by themselves as fusion proteins or the fusion proteins were inserted genetically into herpes simplex virus (HSV)-1 glycoprotein D (gD) which, upon binding to the herpes virus entry mediator (HVEM), inhibits an early T cell checkpoint mediated by the B and T cell mediator (BTLA). This, in turn, lowers the threshold for T cell activation and augments and broadens CD8+ T cell responses to the antigens. The fusion antigens were expressed by chimpanzee adenovirus (AdC) vectors. Expression of the HPV antigens within gD was essential for vaccine immunogenicity and efficacy against challenge with TC-1 cells, which express E7 and E6 of HPV-16 but neither E5 nor E2. Unexpectedly, inclusion of E2 increased both CD8+ T cell responses to the other oncoproteins of HPV-16 and the effectiveness of the vaccines to cause the regression of sizable TC-1 tumors.
Collapse
|
3
|
Zhang Y, Liu S, Chen M, Ou Q, Tian S, Tang J, He Z, Chen Z, Wang C. Preimmunization with Listeria-vectored cervical cancer vaccine candidate strains can establish specific T-cell immune memory and prevent tumorigenesis. BMC Cancer 2024; 24:288. [PMID: 38439023 PMCID: PMC10910769 DOI: 10.1186/s12885-024-12046-7] [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: 09/30/2023] [Accepted: 02/23/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Although HPV prophylactic vaccines can provide effective immune protection against high-risk HPV infection, studies have shown that the protective effect provided by them would decrease with the increased age of vaccination, and they are not recommended for those who are not in the appropriate age range for vaccination. Therefore, in those people who are not suitable for HPV prophylactic vaccines, it is worth considering establishing memory T-cell immunity to provide long-term immune surveillance and generate a rapid response against lesional cells to prevent tumorigenesis. METHODS In this study, healthy mice were preimmunized with LM∆E6E7 and LI∆E6E7, the two Listeria-vectored cervical cancer vaccine candidate strains constructed previously by our laboratory, and then inoculated with tumor cells 40 d later. RESULTS The results showed that preimmunization with LM∆E6E7 and LI∆E6E7 could establish protective memory T-cell immunity against tumor antigens in mice, which effectively eliminate tumor cells. 60% of mice preimmunized with vaccines did not develop tumors, and for the remaining mice, tumor growth was significantly inhibited. We found that preimmunization with vaccines may exert antitumor effects by promoting the enrichment of T cells at tumor site to exert specific immune responses, as well as inhibiting intratumoral angiogenesis and cell proliferation. CONCLUSION Altogether, this study suggests that preimmunization with LM∆E6E7 and LI∆E6E7 can establish memory T-cell immunity against tumor antigens in vivo, which provides a viable plan for preventing tumorigenesis and inhibiting tumor progression.
Collapse
Affiliation(s)
- Yunwen Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Shen Zhen Biomed Alliance Biotech Group Co., Ltd., Shenzhen, China
| | - Sijing Liu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mengdie Chen
- Shen Zhen Biomed Alliance Biotech Group Co., Ltd., Shenzhen, China
| | - Qian Ou
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Sicheng Tian
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jing Tang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zhiqun He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zhaobin Chen
- Shen Zhen Biomed Alliance Biotech Group Co., Ltd., Shenzhen, China.
| | - Chuan Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Bhattacharjee R, Kumar L, Dhasmana A, Mitra T, Dey A, Malik S, Kim B, Gundamaraju R. Governing HPV-related carcinoma using vaccines: Bottlenecks and breakthroughs. Front Oncol 2022; 12:977933. [PMID: 36176419 PMCID: PMC9513379 DOI: 10.3389/fonc.2022.977933] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Human papillomavirus (HPV) contributes to sexually transmitted infection, which is primarily associated with pre-cancerous and cancerous lesions in both men and women and is among the neglected cancerous infections in the world. At global level, two-, four-, and nine-valent pure L1 protein encompassed vaccines in targeting high-risk HPV strains using recombinant DNA technology are available. Therapeutic vaccines are produced by early and late oncoproteins that impart superior cell immunity to preventive vaccines that are under investigation. In the current review, we have not only discussed the clinical significance and importance of both preventive and therapeutic vaccines but also highlighted their dosage and mode of administration. This review is novel in its way and will pave the way for researchers to address the challenges posed by HPV-based vaccines at the present time.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Dehradun, India
| | - Tamoghni Mitra
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, India
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Bonglee Kim, ; Rohit Gundamaraju,
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Lab, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
- *Correspondence: Bonglee Kim, ; Rohit Gundamaraju,
| |
Collapse
|
5
|
Zanotto C, Paolini F, Radaelli A, De Giuli Morghen C. Construction of a recombinant avipoxvirus expressing the env gene of Zika virus as a novel putative preventive vaccine. Virol J 2021; 18:50. [PMID: 33663531 PMCID: PMC7931497 DOI: 10.1186/s12985-021-01519-x] [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: 07/13/2020] [Accepted: 02/19/2021] [Indexed: 12/03/2022] Open
Abstract
Background Zika virus (ZIKV) has been declared a public health emergency that requires development of an effective vaccine, as it might represent an international threat. Methods Here, two novel DNA-based (pVAXzenv) and fowlpox-based (FPzenv) recombinant putative vaccine candidates were constructed that contained the cPrME genes of ZIKV. The env gene inserted into the fowlpox vector was verified for correct transgene expression by Western blotting and by immunofluorescence in different cell lines. The production of virus-like particles as a result of env gene expression was also demonstrated by electron microscopy. BALB/c mice were immunosuppressed with dexamethasone and immunized following a prime–boost strategy in a heterologous protocol where pVAXzenv was followed by FPzenv, to evaluate the immunogenicity of the Env protein. The mice underwent a challenge with an epidemic ZIKV after the last boost. Results These data show that the ZIKV Env protein was correctly expressed in both normal human lung fibroblasts (MRC-5 cells) and green monkey kidney (Vero) cells infected with FPzenv, and that the transgene expression lasted for more than 2 weeks. After mucosal administration of FPzenv, the immunized mice showed specific and significantly higher humoral responses compared to the control mice. However, virus neutralizing antibodies were not detected using plaque reduction assays. Conclusions Although BALB/c mice appear to be an adequate model for ZIKV infection, as it mimics the natural mild infection in human beings, inadequate immune suppression seemed to occur by dexamethasone and different immune suppression strategies should be applied before challenge to reveal any protection of the mice.
Collapse
Affiliation(s)
- Carlo Zanotto
- Laboratory of Molecular Virology and Recombinant Vaccine Development, Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy
| | - Francesca Paolini
- HPV-UNIT, Laboratory of Virology, Regina Elena National Cancer Institute, Via delle Messi d'Oro, 156, 00158, Rome, Italy
| | - Antonia Radaelli
- Laboratory of Molecular Virology and Recombinant Vaccine Development, Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy.
| | | |
Collapse
|
6
|
Belnoue E, Leystra AA, Carboni S, Cooper HS, Macedo RT, Harvey KN, Colby KB, Campbell KS, Vanderveer LA, Clapper ML, Derouazi M. Novel Protein-Based Vaccine against Self-Antigen Reduces the Formation of Sporadic Colon Adenomas in Mice. Cancers (Basel) 2021; 13:cancers13040845. [PMID: 33671373 PMCID: PMC7923075 DOI: 10.3390/cancers13040845] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Colorectal cancer remains a leading cause of cancer-related mortality worldwide. However, high-risk populations with a genetic predisposition for colorectal cancer could benefit greatly from novel and efficacious immunopreventive strategies that afford long-lasting protection. The achaete-scute family bHLH transcription factor 2 (Ascl2) has been identified as a promising target for immunoprevention of colorectal cancer, based on its induction during the formation and progression of colorectal tumors and its minimal expression observed in healthy tissue. The goal of the present study was to determine the efficacy of a protein-based vaccine targeting Ascl2 in combination with an anti-PD-1 treatment in a spontaneous colorectal cancer mouse model. This novel vaccine strategy promotes potent tumor-specific immunity, and prevents the formation of colon adenomas in mice. The results demonstrate that Ascl2 is a promising target for immunoprevention for individuals at elevated risk of developing colorectal cancer. Abstract Novel immunopreventive strategies are emerging that show great promise for conferring long-term protection to individuals at high risk of developing colorectal cancer. The KISIMA vaccine platform utilizes a chimeric protein comprising: (1) a selected tumor antigen; (2) a cell-penetrating peptide to improve antigen delivery and epitope presentation, and (3) a TLR2/4 agonist to serve as a self-adjuvant. This study examines the ability of a KISIMA vaccine against achaete-scute family bHLH transcription factor 2 (Ascl2), an early colon cancer antigen, to reduce colon tumor formation by stimulating an anti-tumor immune response. Vaccine administrations were well-tolerated and led to circulating antibodies and antigen-specific T cells in a mouse model of colorectal cancer. To assess preventive efficacy, the vaccine was administered to mice either alone or in combination with the immune checkpoint inhibitor anti-PD-1. When delivered to animals prior to colon tumor formation, the combination strategy significantly reduced the development of colon microadenomas and adenomas, as compared to vehicle-treated controls. This response was accompanied by an increase in the intraepithelial density of CD3+ T lymphocytes. Together, these data indicate that the KISIMA-Ascl2 vaccine shows great potential to be a safe and potent immunopreventive intervention for individuals at high risk of developing colorectal cancer.
Collapse
Affiliation(s)
- Elodie Belnoue
- AMAL Therapeutics, Fondation pour Recherches Médicales, 64 avenue de la Roseraie, 1205 Geneva, Switzerland; (E.B.); (S.C.)
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Alyssa A. Leystra
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (A.A.L.); (H.S.C.); (R.T.M.); (K.N.H.); (L.A.V.)
| | - Susanna Carboni
- AMAL Therapeutics, Fondation pour Recherches Médicales, 64 avenue de la Roseraie, 1205 Geneva, Switzerland; (E.B.); (S.C.)
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Harry S. Cooper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (A.A.L.); (H.S.C.); (R.T.M.); (K.N.H.); (L.A.V.)
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA
| | - Rodrigo T. Macedo
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (A.A.L.); (H.S.C.); (R.T.M.); (K.N.H.); (L.A.V.)
| | - Kristen N. Harvey
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (A.A.L.); (H.S.C.); (R.T.M.); (K.N.H.); (L.A.V.)
| | - Kimberly B. Colby
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (K.B.C.); (K.S.C.)
| | - Kerry S. Campbell
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (K.B.C.); (K.S.C.)
| | - Lisa A. Vanderveer
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (A.A.L.); (H.S.C.); (R.T.M.); (K.N.H.); (L.A.V.)
| | - Margie L. Clapper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA; (A.A.L.); (H.S.C.); (R.T.M.); (K.N.H.); (L.A.V.)
- Correspondence: (M.L.C.); (M.D.)
| | - Madiha Derouazi
- AMAL Therapeutics, Fondation pour Recherches Médicales, 64 avenue de la Roseraie, 1205 Geneva, Switzerland; (E.B.); (S.C.)
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
- Correspondence: (M.L.C.); (M.D.)
| |
Collapse
|
7
|
Sabet F, Mosavat A, Ahmadi Ghezeldasht S, Basharkhah S, Shamsian SAA, Abbasnia S, Shamsian K, Rezaee SA. Prevalence, genotypes and phylogenetic analysis of human papillomaviruses (HPV) in northeast Iran. Int J Infect Dis 2020; 103:480-488. [PMID: 33310023 DOI: 10.1016/j.ijid.2020.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Human papillomaviruses (HPV) are the main etiology of invasive cervical cancer. Together HPV and viral hepatitis account for the cause of 25% of cancers in developing countries. To evaluate the association between population movements and the spread of HPV, this study looked at prevalence, genotypes, and phylogenetic assessment of HPV in Great Khorasan, a pilgrimage-tourism province in northeast Iran. METHODS From March 2013 to July 2018, 567 samples were collected from three groups in Khorasan: Razavi and North Khorasan provinces (highly mobile population); South Khorasan province (conservative and desert); and diverse group (tourists). RESULTS HPV prevalence was 48.4% in Razavi and North Khorasan (first group); 19.9% in South Khorasan (second group); and 33.6% in the diverse group. The four most common HPV genotypes were HPV-6, 11, 51 and 16, in the first group; HPV-6, 11, 16 and 58 in the second group; and HPV-6, 11, 16 and 53/89 in the diverse group. The most frequent genotypes that are known as high risk for cervical cancer were HPV-51 in the first group, HPV-16 in the second group and the diverse group. Among low-risk genotypes, HPV-6, and HPV-11 were more frequent in all groups. DNA sequencing and phylogenetic analysis of 20 HPV-positive samples showed that the distributions of the HPV genotypes were HPV-6 (50%), 11 (10%), 67 (5%), 16 (15%), 31 (10%), 54 (5%), and 89 (5%). CONCLUSIONS The findings show that areas associated with population movement should be frequently monitored for infectious diseases, while conservative and less populated areas have less risk for virus spread and endemicity. Health authorities should focus more on the establishment of HPV diagnostic facilities, screening, vaccination, and enhancement of public knowledge in these regions.
Collapse
Affiliation(s)
- Faezeh Sabet
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Razavi Khorasan, Mashhad, Iran.
| | - Sanaz Ahmadi Ghezeldasht
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Razavi Khorasan, Mashhad, Iran.
| | - Samira Basharkhah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Ali Akbar Shamsian
- Department of Mycology and Parasitology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Razavi Khorasan, Mashhad, Iran.
| | - Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khosrow Shamsian
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Razavi Khorasan, Mashhad, Iran.
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Zhao Y, Han Z, Zhang X, Zhang X, Sun J, Ma D, Liu S. Construction and immune protection evaluation of recombinant virus expressing Newcastle disease virus F protein by the largest intergenic region of fowlpox virus NX10. Virus Genes 2020; 56:734-748. [PMID: 33009986 DOI: 10.1007/s11262-020-01799-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/26/2020] [Indexed: 01/27/2023]
Abstract
Fowlpox virus (FPV) is used as a vaccine vector to prevent diseases in poultry and mammals. The insertion site is considered as one of the main factors influencing foreign gene expression. Therefore, the identification of insertion sites that can stably and efficiently express foreign genes is crucial for the construction of recombinant vaccines. In this study, we found that the insertion of foreign genes into ORF054 and the ORF161/ORF162 intergenic region of the FPV genome did not affect replication, and that the foreign genes inserted into the intergenic region were more efficiently expressed than when they were inserted into a gene. Based on these results, the recombinant virus rFPVNX10-NDV F-E was constructed and immune protection against virulent FPV and Newcastle disease virus (NDV) was evaluated. Tests for anti-FPV antibodies in the vaccinated chickens were positive within 14 days post-vaccination. After challenge with FPV102, no clinical signs of FP were observed in vaccinated chickens, as compared to that in the control group (unvaccinated), which showed 100% morbidity. Low levels of NDV-specific neutralizing antibodies were detected in vaccinated chickens before challenge. After challenge with NDV ck/CH/LHLJ/01/06, all control chickens died within 4 days post-challenge, whereas 5/15 vaccinated chickens died between 4 and 12 days post-challenge. Vaccination provided an immune protection rate of 66.7%, whereas the control group showed 100% mortality. These results indicate that the ORF161/ORF162 intergenic region of FPVNX10 can be used as a recombination site for foreign gene expression in vivo and in vitro.
Collapse
Affiliation(s)
- Yan Zhao
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, 150030, China.,Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Zongxi Han
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Xiaocai Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Xuemei Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Junfeng Sun
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Deying Ma
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, 150030, China.
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China.
| |
Collapse
|
9
|
Shah UJ, Nasiruddin M, Dar SA, Khan MKA, Akhter MR, Singh N, Rabaan AA, Haque S. Emerging biomarkers and clinical significance of HPV genotyping in prevention and management of cervical cancer. Microb Pathog 2020; 143:104131. [PMID: 32169490 DOI: 10.1016/j.micpath.2020.104131] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/10/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022]
Abstract
Cervical cancer is a growing and serious problem world-wide in women, but more acute in developing countries especially in Indian subcontinent. The main causative agent for the disease is Human Papilloma Virus (HPV). The history of the cervical cancer goes back to eighteenth century as the HPV infection is reported since 1800s. Presently, the genetic structure of HPV is well defined. Several screening tests including cytology and visual based screening and high risk HPV testing are available. Also available are various clinical and commercial diagnostic tests. However due to the lack of awareness and population-based screening programs, the morbidity and mortality rate is alarmingly high. There are new emerging biomarkers including E6/E7 mRNA, p16ink4a, markers of aberrant S-phase induction, chromosomal abnormalities and miRNAs along with advanced genotyping methods. These markers have clinical significance and are helpful in disease prevention and management. Further, recent advancement in the field of metagenomics has increased the prospects of identifying newer microbes, viruses hitherto reported thus far in the context of HPV infection. Analysis of HPV cases using modern tools including genotyping using more powerful biomarkers is envisaged to enhance the prospects of early diagnosis, better prognosis, more reliable treatment and eventual management of the disease.
Collapse
Affiliation(s)
- Ushma Jaykamal Shah
- MedGenome Labs Ltd., Kailash Cancer Hospital and Research Center, Muni Seva Ashram, P.O. Goraj, Tal. Waghodia, Dist. Vadodara, 391760, Gujarat, India
| | - Mohammad Nasiruddin
- Anand Diagnostic Laboratory (A Neuberg Associate), Neuberg Anand Reference Laboratory, Anand Tower, 54, Bowring Hospital Road, Shivajinagar, Bangalore - 560001, India.
| | - Sajad Ahmad Dar
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| | - Md Khurshid Alam Khan
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, Tamil Nadu, India
| | - Mohammad Riyaz Akhter
- MedGenome Labs Ltd., 3rd Floor, Narayana Nethralaya Building, Narayana Health City, # 258/A, Bommasandra, Hosur Road, Bangalore, 560099, Karnataka, India
| | - Nidhi Singh
- Department of Obstetrics and Gynecology, Prasad Institute of Medical Sciences, Lucknow, 226401, Uttar Pradesh, India
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Saudi Aramco, Dhahran, 31311, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia.
| |
Collapse
|
10
|
Wang HF, Wang SS, Tang YJ, Chen Y, Zheng M, Tang YL, Liang XH. The Double-Edged Sword-How Human Papillomaviruses Interact With Immunity in Head and Neck Cancer. Front Immunol 2019; 10:653. [PMID: 31001266 PMCID: PMC6454067 DOI: 10.3389/fimmu.2019.00653] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/11/2019] [Indexed: 02/05/2023] Open
Abstract
Patients with human papilloma virus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) have remarkably better prognosis, which differs from HPV-negative oropharyngeal squamous cell carcinoma (OPSCC) with respect to clinical, genomic, molecular, and immunological aspects, especially having the characteristics of high levels of immune cell infiltration and high degrees of immunosuppression. This review will summarize immune evasion mechanisms in HPV-positive HNSCC, analyze the host various immune responses to HPV and abundant numbers of infiltrating immune cell, and discuss the differences between HPV-positive HNSCC with cervical cancer. A deeper understanding of the immune landscape will help new concepts to emerge in immune-checkpoint oncology, which might be a valuable add-on to established concepts.
Collapse
Affiliation(s)
- Hao-Fan Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Sha-Sha Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, Department of Oral Pathology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, Department of Oral Pathology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| |
Collapse
|
11
|
Cordeiro MN, De Lima RDCP, Paolini F, Melo ARDS, Campos APF, Venuti A, De Freitas AC. Current research into novel therapeutic vaccines against cervical cancer. Expert Rev Anticancer Ther 2018; 18:365-376. [DOI: 10.1080/14737140.2018.1445527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Marcelo Nazário Cordeiro
- Laboratório de Estudos Moleculares e Terapia Experimental – LEMTE, Brazil Universidade Federal de Pernambuco – UFPE, Brazil
| | - Rita de Cássia Pereira De Lima
- Laboratório de Estudos Moleculares e Terapia Experimental – LEMTE, Brazil Universidade Federal de Pernambuco – UFPE, Brazil
| | - Francesca Paolini
- HPV-Unit UOSD Immunology and Tumor Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alanne Rayssa da Silva Melo
- Laboratório de Estudos Moleculares e Terapia Experimental – LEMTE, Brazil Universidade Federal de Pernambuco – UFPE, Brazil
| | - Ana Paula Ferreira Campos
- Laboratório de Estudos Moleculares e Terapia Experimental – LEMTE, Brazil Universidade Federal de Pernambuco – UFPE, Brazil
| | - Aldo Venuti
- HPV-Unit UOSD Immunology and Tumor Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Antonio Carlos De Freitas
- Laboratório de Estudos Moleculares e Terapia Experimental – LEMTE, Brazil Universidade Federal de Pernambuco – UFPE, Brazil
| |
Collapse
|
12
|
Liu C, Xie Y, Sun B, Geng F, Zhang F, Guo Q, Wu H, Yu B, Wu J, Yu X, Kong W, Zhang H. MUC1- and Survivin-based DNA Vaccine Combining Immunoadjuvants CpG and interleukin-2 in a Bicistronic Expression Plasmid Generates Specific Immune Responses and Antitumour Effects in a Murine Colorectal Carcinoma Model. Scand J Immunol 2018; 87:63-72. [PMID: 29193199 DOI: 10.1111/sji.12633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/16/2017] [Indexed: 12/30/2022]
Abstract
DNA vaccination is a promising cancer treatment due to its safety, but poor immunogenicity limits its application. However, immunoadjuvants, heterogeneous prime-boost strategies and combination with conventional treatments can be used to improve the antitumour immune effects. A CpG motif and interleukin-2 (IL-2) cytokine are often used as adjuvants. In this study, a DNA vaccine containing a CpG motif was constructed to evaluate its adjuvant effect. The results show that the cytotoxicity of the DNA vaccine was increased fivefold, and survival lifetime was prolonged twofold by the CpG motif adjuvant. To simplify the industrial production process, a bicistronic plasmid was constructed to carry the fusion genes of survivin/MUC1 (MS) and IL-2 and with a CpG motif in its backbone. The results showed that the antitumour effect of the bicistronic vaccine was the same as that of the two vaccine co-injected regime. Furthermore, the vaccine could suppress metastatic tumour foci by 69.1% in colorectal carcinoma-bearing mice. Moreover, the vaccine induced survivin- and MUC1-specific immune responses in splenocytes and induced the immune promoting factor CCL-19 and GM-CSF upregulated, while metastatic-associated factor MMP-9 and immunosuppressing factor PD-L1 downregulated in tumour tissue. When combining the vaccine with the chemotherapy drug oxaliplatin, the survival was prolonged by about 2.5-fold. In conclusion, the DNA vaccine containing a CpG motif in bicistronic form showed good effects on colorectal cancer by inhibiting both tumour growth and metastasis, and combination with oxaliplatin could improve its antitumour effects.
Collapse
Affiliation(s)
- C Liu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
| | - Y Xie
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
| | - B Sun
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - F Geng
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
| | - F Zhang
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
| | - Q Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
| | - H Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - B Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - J Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - X Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - W Kong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - H Zhang
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
13
|
Bissa M, Forlani G, Zanotto C, Tosi G, De Giuli Morghen C, Accolla RS, Radaelli A. Fowlpoxvirus recombinants coding for the CIITA gene increase the expression of endogenous MHC-II and Fowlpox Gag/Pro and Env SIV transgenes. PLoS One 2018; 13:e0190869. [PMID: 29385169 PMCID: PMC5791965 DOI: 10.1371/journal.pone.0190869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/21/2017] [Indexed: 01/12/2023] Open
Abstract
A complete eradication of an HIV infection has never been achieved by vaccination and the search for new immunogens that can induce long-lasting protective responses is ongoing. Avipoxvirus recombinants are host-restricted for replication to avian species and they do not have the undesired side effects induced by vaccinia recombinants. In particular, Fowlpox (FP) recombinants can express transgenes over long periods and can induce protective immunity in mammals, mainly due to CD4-dependent CD8+ T cells. In this context, the class II transactivator (CIITA) has a pivotal role in triggering the adaptive immune response through induction of the expression of class-II major histocompatibility complex molecule (MHC-II), that can present antigens to CD4+ T helper cells. Here, we report on construction of novel FPgp and FPenv recombinants that express the highly immunogenic SIV Gag-pro and Env structural antigens. Several FP-based recombinants, with single or dual genes, were also developed that express CIITA, driven from H6 or SP promoters. These recombinants were used to infect CEF and Vero cells in vitro and determine transgene expression, which was evaluated by real-time PCR and Western blotting. Subcellular localisation of the different proteins was evaluated by confocal microscopy, whereas HLA-DR or MHC-II expression was measured by flow cytometry. Fowlpox recombinants were also used to infect syngeneic T/SA tumour cells, then injected into Balb/c mice to elicit MHC-II immune response and define the presentation of the SIV transgene products in the presence or absence of FPCIITA. Antibodies to Env were measured by ELISA. Our data show that the H6 promoter was more efficient than SP to drive CIITA expression and that CIITA can enhance the levels of the gag/pro and env gene products only when infection is performed by FP single recombinants. Also, CIITA expression is higher when carried by FP single recombinants than when combined with FPgp or FPenv constructs and can induce HLA-DR cell surface expression. However, in-vivo experiments did not show any significant increase in the humoral response. As CIITA already proved to elicit immunogenicity by improving antigen presentation, further in-vivo experiments should be performed to increase the immune responses. The use of prime/boost immunisation protocols and the oral administration route of the recombinants may enhance the immunogenicity of Env peptides presented by MHC-II and provide CD4+ T-cell stimulation.
Collapse
Affiliation(s)
- Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, Milan, Italy
| | - Greta Forlani
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli 32, Milan, Italy
| | - Giovanna Tosi
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Carlo De Giuli Morghen
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli 32, Milan, Italy
- Catholic University “Our Lady of Good Counsel”, Rr. Dritan Hoxha, Tirana, Albania
| | - Roberto S. Accolla
- Department of Experimental Medicine, University of Insubria, Via O. Rossi 9, Varese, Italy
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, Milan, Italy
- CNR Institute of Neurosciences, Cellular and Molecular Pharmacology Section, University of Milan, via Vanvitelli 32, Milan, Italy
- * E-mail:
| |
Collapse
|
14
|
Sauermann U, Radaelli A, Stolte-Leeb N, Raue K, Bissa M, Zanotto C, Krawczak M, Tenbusch M, Überla K, Keele BF, De Giuli Morghen C, Sopper S, Stahl-Hennig C. Vector Order Determines Protection against Pathogenic Simian Immunodeficiency Virus Infection in a Triple-Component Vaccine by Balancing CD4 + and CD8 + T-Cell Responses. J Virol 2017; 91:e01120-17. [PMID: 28904195 PMCID: PMC5686736 DOI: 10.1128/jvi.01120-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/06/2017] [Indexed: 12/15/2022] Open
Abstract
An effective AIDS vaccine should elicit strong humoral and cellular immune responses while maintaining low levels of CD4+ T-cell activation to avoid the generation of target cells for viral infection. The present study investigated two prime-boost regimens, both starting vaccination with single-cycle immunodeficiency virus, followed by two mucosal boosts with either recombinant adenovirus (rAd) or fowlpox virus (rFWPV) expressing SIVmac239 or SIVmac251 gag/pol and env genes, respectively. Finally, vectors were switched and systemically administered to the reciprocal group of animals. Only mucosal rFWPV immunizations followed by systemic rAd boost significantly protected animals against a repeated low-dose intrarectal challenge with pathogenic SIVmac251, resulting in a vaccine efficacy (i.e., risk reduction per exposure) of 68%. Delayed viral acquisition was associated with higher levels of activated CD8+ T cells and Gag-specific gamma interferon (IFN-γ)-secreting CD8+ cells, low virus-specific CD4+ T-cell responses, and low Env antibody titers. In contrast, the systemic rFWPV boost induced strong virus-specific CD4+ T-cell activity. rAd and rFWPV also induced differential patterns of the innate immune responses, thereby possibly shaping the specific immunity. Plasma CXCL10 levels after final immunization correlated directly with virus-specific CD4+ T-cell responses and inversely with the number of exposures to infection. Also, the percentage of activated CD69+ CD8+ T cells correlated with the number of exposures to infection. Differential stimulation of the immune response likely provided the basis for the diverging levels of protection afforded by the vaccine regimen.IMPORTANCE A failed phase II AIDS vaccine trial led to the hypothesis that CD4+ T-cell activation can abrogate any potentially protective effects delivered by vaccination or promote acquisition of the virus because CD4+ T helper cells, required for an effective immune response, also represent the target cells for viral infection. We compared two vaccination protocols that elicited similar levels of Gag-specific immune responses in rhesus macaques. Only the animal group that had a low level of virus-specific CD4+ T cells in combination with high levels of activated CD8+ T cells was significantly protected from infection. Notably, protection was achieved despite the lack of appreciable Env antibody titers. Moreover, we show that both the vector and the route of immunization affected the level of CD4+ T-cell responses. Thus, mucosal immunization with FWPV-based vaccines should be considered a potent prime in prime-boost vaccination protocols.
Collapse
Affiliation(s)
- Ulrike Sauermann
- Unit of Infection Models, Deutsches Primatenzentrum GmbH, Goettingen, Germany
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Nicole Stolte-Leeb
- Unit of Infection Models, Deutsches Primatenzentrum GmbH, Goettingen, Germany
| | - Katharina Raue
- Unit of Infection Models, Deutsches Primatenzentrum GmbH, Goettingen, Germany
| | - Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Christian-Albrechts University, Kiel, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Klaus Überla
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Carlo De Giuli Morghen
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Catholic University Our Lady of Good Counsel, Tirana, Albania
| | - Sieghart Sopper
- Clinic for Hematology and Oncology, Medical University Innsbruck, Tyrolean Cancer Research Center, Innsbruck, Austria
| | | |
Collapse
|
15
|
Choi YJ, Park JS. Clinical significance of human papillomavirus genotyping. J Gynecol Oncol 2016; 27:e21. [PMID: 26768784 PMCID: PMC4717226 DOI: 10.3802/jgo.2016.27.e21] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/07/2015] [Accepted: 10/30/2015] [Indexed: 12/21/2022] Open
Abstract
Cervical cancer is the fourth most common cancer in women worldwide, and the human papillomavirus (HPV) is the main causative agent for its development. HPV is a heterogeneous virus, and a persistent infection with a high-risk HPV contributes to the development of cancer. In recent decades, great advances have been made in understanding the molecular biology of HPV, and HPV’s significance in cervical cancer prevention and management has received increased attention. In this review, we discuss the role of HPV genotyping in cervical cancer by addressing: clinically important issues in HPV virology; the current application of HPV genotyping in clinical medicine; and potential future uses for HPV genotyping.
Collapse
Affiliation(s)
- Youn Jin Choi
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Sup Park
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
16
|
Illiano E, Bissa M, Paolini F, Zanotto C, De Giuli Morghen C, Franconi R, Radaelli A, Venuti A. Prime-boost therapeutic vaccination in mice with DNA/DNA or DNA/Fowlpox virus recombinants expressing the Human Papilloma Virus type 16 E6 and E7 mutated proteins fused to the coat protein of Potato virus X. Virus Res 2016; 225:82-90. [PMID: 27664839 DOI: 10.1016/j.virusres.2016.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/16/2016] [Accepted: 09/19/2016] [Indexed: 11/26/2022]
Abstract
The therapeutic antitumor potency of a prime-boost vaccination strategy was explored, based on the mutated, nontransforming forms of the E6 (E6F47R) and E7 (E7GGG) oncogenes of Human Papilloma Virus type 16 (HPV16), fused to the Potato virus X (PVX) coat protein (CP) sequence. Previous data showed that CP fusion improves the immunogenicity of tumor-associated antigens and may thus increase their efficacy. After verifying the correct expression of E6F47RCP and E7GGGCP inserted into DNA and Fowlpox virus recombinants by Western blotting and immunofluorescence, their combined use was evaluated for therapy in a pre-clinical mouse model of HPV16-related tumorigenicity. Immunization protocols were applied using homologous (DNA/DNA) or heterologous (DNA/Fowlpox) prime-boost vaccine regimens. The humoral immune responses were determined by ELISA, and the therapeutic efficacy evaluated by the delay in tumor appearance and reduced tumor volume after inoculation of syngeneic TC-1* tumor cells. Homologous DNA/DNA genetic vaccines were able to better delay tumor appearance and inhibit tumor growth when DNAE6F47RCP and DNAE7GGGCP were administered in combination. However, the heterologous DNA/Fowlpox vaccination strategy was able to delay tumor appearance in a higher number of animals when E6F47RCP and in particular E7GGGCP were administered alone.
Collapse
Affiliation(s)
- Elena Illiano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Francesca Paolini
- Laboratory of Virology HPV-UNIT, Regina Elena National Cancer Institute, Rome, Italy.
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.
| | - Carlo De Giuli Morghen
- Cellular and Molecular Pharmacology Section, CNR Institute of Neurosciences, University of Milan, Milan, Italy; Catholic University "Our Lady of Good Counsel", Tirana, Albania.
| | - Rosella Franconi
- Laboratory of Biomedical Technologies, Division of Health Protection Technologies, Department for Sustainability, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Casaccia Research Centre, Rome, Italy.
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Cellular and Molecular Pharmacology Section, CNR Institute of Neurosciences, University of Milan, Milan, Italy.
| | - Aldo Venuti
- Laboratory of Virology HPV-UNIT, Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
17
|
Aggarwal C. DNA-based immunotherapy for HPV-associated head and neck cancer. Immunotherapy 2016; 8:1187-92. [DOI: 10.2217/imt-2016-0055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) accounts for 3% of all cancers. Most patients present with locally advanced disease, where multimodality therapies are used with curative intent. Despite favorable early local treatment results, about one third of the patients will eventually develop metastatic disease. Immunotherapy offers a novel therapeutic strategy beyond cytotoxic chemotherapy, with initial approvals in melanoma and non-small-cell lung cancer. HPV-associated SCCHN is a distinct subset, with unique epidemiology and treatment outcomes. Both subsets of SCCHN (HPV-related or not) are particularly favorable for immunotherapy, as immune evasion and dysregulation have been shown to play a key role in the initiation and progression of disease. This review focuses on the latest developments in immunotherapy in SCCHN, with a particular focus on DNA-based approaches including vaccine and adoptive cellular therapies.
Collapse
Affiliation(s)
- Charu Aggarwal
- Assistant Professor, University of Pennsylvania, Department of Medicine, Hematology–Oncology Division, 624 South Pavilion, Perelman Center for Advanced Medicine, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Leng CY, Low HC, Chua LL, Chong ML, Sulaiman H, Azwa I, Roberts JM, Kamarulzaman A, Rajasuriar R, Woo YL. Human papillomavirus 16 (HPV16) and HPV52 E6-specific immunity in HIV-infected adults on combination antiretroviral therapy. HIV Med 2016; 18:321-331. [PMID: 27649852 DOI: 10.1111/hiv.12432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Human papillomavirus (HPV)-associated cancers disproportionately affect those infected with HIV despite effective combination antiretroviral therapy (cART). The primary aim of this study was to quantify HPV16 and HPV52 E6-specific interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) T-cell responses, a correlate of protective immunity, in the first year following cART initiation and subsequently in those patients with suboptimal (sIR) and optimal (oIR) immune reconstitution. METHODS Ninety-four HIV-infected patients were recruited to the study; a longitudinal cohort of patients recruited just prior to commencing cART and followed up for 48 weeks (n = 27), and a cross-sectional cohort (n = 67) consisting of patients with sIR (CD4 T-cell count < 350 cells/μL) and oIR (CD4 T-cell count > 500 cells/μL) after a minimum of 2 years on cART. Controls (n = 29) consisted of HIV-negative individuals. IFN-γ ELISPOT responses against HPV16 and HPV52 E6 were correlated to clinical characteristics, anal and oral HPV carriage, T-cell maturational subsets, markers of activation, senescence and T-regulatory cells. RESULTS HPV16 and HPV52 E6-specific T-cell responses were detected in only one of 27 patients (3.7%) during the initial phase of immune recovery. After at least 2 years of cART, those who achieved oIR had significantly higher E6-specific responses (9 of 34; 26.5%) compared with those with sIR (2 of 32; 6.3%) (P = 0.029). Apart from higher CD4 T-cell counts and lower CD4 T-cell activation, no other immunological correlates were associated with the detection of HPV16 and HPV52 E6-specific responses. CONCLUSIONS HPV16 and HPV52 E6-specific IFN-γ T-cell responses, a correlate of protective immunity, were detected more frequently among HIV-infected patients who achieved optimal immune recovery on cART (26.5%) compared with those with suboptimal recovery (6.3%).
Collapse
Affiliation(s)
- C Y Leng
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - H C Low
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - L L Chua
- University of Malaya Cancer Research Institute (UMCRI), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - M L Chong
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - H Sulaiman
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - I Azwa
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - J M Roberts
- Douglass Hanly Moir Pathology, Macquarie Park, New South Wales, Australia
| | - A Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - R Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Y L Woo
- Centre of Excellence for Research in AIDS (CERiA), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,University of Malaya Cancer Research Institute (UMCRI), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Illiano E, Demurtas OC, Massa S, Di Bonito P, Consalvi V, Chiaraluce R, Zanotto C, De Giuli Morghen C, Radaelli A, Venuti A, Franconi R. Production of functional, stable, unmutated recombinant human papillomavirus E6 oncoprotein: implications for HPV-tumor diagnosis and therapy. J Transl Med 2016; 14:224. [PMID: 27465494 PMCID: PMC4963926 DOI: 10.1186/s12967-016-0978-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/13/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High-risk human papillomaviruses (HR-HPVs) types 16 and 18 are the main etiological agents of cervical cancer, with more than 550,000 new cases each year worldwide. HPVs are also associated with other ano-genital and head-and-neck tumors. The HR-HPV E6 and E7 oncoproteins are responsible for onset and maintenance of the cell transformation state, and they represent appropriate targets for development of diagnostic and therapeutic tools. METHODS The unmutated E6 gene from HPV16 and HPV18 and from low-risk HPV11 was cloned in a prokaryotic expression vector for expression of the Histidine-tagged E6 protein (His6-E6), according to a novel procedure. The structural properties were determined using circular dichroism and fluorescence spectroscopy. His6-E6 oncoprotein immunogenicity was assessed in a mouse model, and its functionality was determined using in vitro GST pull-down and protein degradation assays. RESULTS The His6-tagged E6 proteins from HPV16, HPV18, and HPV11 E6 genes, without any further modification in the amino-acid sequence, were produced in bacteria as soluble and stable molecules. Structural analyses of HPV16 His6-E6 suggests that it maintains correct folding and conformational properties. C57BL/6 mice immunized with HPV16 His6-E6 developed significant humoral immune responses. The E6 proteins from HPV16, HPV18, and HPV11 were purified according to a new procedure, and investigated for protein-protein interactions. HR-HPV His6-E6 bound p53, the PDZ1 motif from MAGI-1 proteins, the human discs large tumor suppressor, and the human ubiquitin ligase E6-associated protein, thus suggesting that it is biologically active. The purified HR-HPV E6 proteins also targeted the MAGI-3 and p53 proteins for degradation. CONCLUSIONS This new procedure generates a stable, unmutated HPV16 E6 protein, which maintains the E6 properties in in vitro binding assays. This will be useful for basic studies, and for development of diagnostic kits and immunotherapies in preclinical mouse models of HPV-related tumorigenesis.
Collapse
Affiliation(s)
- Elena Illiano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.,Laboratory of Biomedical Technologies (SSPT-TECS-TEB), Department for Sustainability, Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and the Environment (ENEA), 'Casaccia' Research Centre, Via Anguillarese 301, 00123, Rome, Italy
| | - Olivia Costantina Demurtas
- Laboratory of Biotechnology (SSPT-BIOAG-BIOTEC), Department for Sustainability, Division Biotechnology and Agroindustry, Italian National Agency for New Technologies, Energy and the Environment (ENEA), 'Casaccia' Research Centre, Via Anguillarese 301, 00123 Rome, Italy
| | - Silvia Massa
- Laboratory of Biotechnology (SSPT-BIOAG-BIOTEC), Department for Sustainability, Division Biotechnology and Agroindustry, Italian National Agency for New Technologies, Energy and the Environment (ENEA), 'Casaccia' Research Centre, Via Anguillarese 301, 00123 Rome, Italy
| | - Paola Di Bonito
- Department of Infectious Diseases, Istituto Superiore Sanità, Viale Regina Elena 299, 00185, Rome, Italy
| | - Valerio Consalvi
- 'A. Rossi Fanelli' Department of Biochemical Sciences, University of Rome 'La Sapienza', P.le Aldo Moro 5, 00185, Rome, Italy
| | - Roberta Chiaraluce
- 'A. Rossi Fanelli' Department of Biochemical Sciences, University of Rome 'La Sapienza', P.le Aldo Moro 5, 00185, Rome, Italy
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy
| | - Carlo De Giuli Morghen
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy.,Catholic University 'Our Lady of Good Counsel', Tirana, Albania
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.,Cellular and Molecular Pharmacology Section, CNR Institute of Neurosciences, University of Milan, 20129, Milan, Italy
| | - Aldo Venuti
- HPV-UNIT, Ridait Department, Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| | - Rosella Franconi
- Laboratory of Biomedical Technologies (SSPT-TECS-TEB), Department for Sustainability, Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and the Environment (ENEA), 'Casaccia' Research Centre, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
20
|
Prime-boost vaccine strategy against viral infections: Mechanisms and benefits. Vaccine 2016; 34:413-423. [DOI: 10.1016/j.vaccine.2015.11.062] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/01/2023]
|
21
|
Panatto D, Amicizia D, Bragazzi NL, Rizzitelli E, Tramalloni D, Valle I, Gasparini R. Human Papillomavirus Vaccine. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015:231-322. [DOI: 10.1016/bs.apcsb.2015.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|