1
|
A Novel Approach of Antiviral Drugs Targeting Viral Genomes. Microorganisms 2022; 10:microorganisms10081552. [PMID: 36013970 PMCID: PMC9414836 DOI: 10.3390/microorganisms10081552] [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: 06/03/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Outbreaks of viral diseases, which cause morbidity and mortality in animals and humans, are increasing annually worldwide. Vaccines, antiviral drugs, and antibody therapeutics are the most effective tools for combating viral infection. The ongoing coronavirus disease 2019 pandemic, in particular, raises an urgent need for the development of rapid and broad-spectrum therapeutics. Current antiviral drugs and antiviral antibodies, which are mostly specific at protein levels, have encountered difficulties because the rapid evolution of mutant viral strains resulted in drug resistance. Therefore, degrading viral genomes is considered a novel approach for developing antiviral drugs. The current article highlights all potent candidates that exhibit antiviral activity by digesting viral genomes such as RNases, RNA interference, interferon-stimulated genes 20, and CRISPR/Cas systems. Besides that, we introduce a potential single-chain variable fragment (scFv) that presents antiviral activity against various DNA and RNA viruses due to its unique nucleic acid hydrolyzing characteristic, promoting it as a promising candidate for broad-spectrum antiviral therapeutics.
Collapse
|
2
|
Rho SB, Lee SH, Byun HJ, Kim BR, Lee CH. IRF-1 Inhibits Angiogenic Activity of HPV16 E6 Oncoprotein in Cervical Cancer. Int J Mol Sci 2020; 21:ijms21207622. [PMID: 33076322 PMCID: PMC7589982 DOI: 10.3390/ijms21207622] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
HPV16 E6 oncoprotein is a member of the human papillomavirus (HPV) family that contributes to enhanced cellular proliferation and risk of cervical cancer progression via viral infection. In this study, interferon regulatory factor-1 (IRF-1) regulates cell growth inhibition and transcription factors in immune response, and acts as an HPV16 E6-binding cellular molecule. Over-expression of HPV16 E6 elevated cell growth by attenuating IRF-1-induced apoptosis and repressing p21 and p53 expression, but activating cyclin D1 and nuclear factor kappa B (NF-κB) expression. The promoter activities of p21 and p53 were suppressed, whereas NF-κB activities were increased by HPV16 E6. Additionally, the cell viability of HPV16 E6 was diminished by IRF-1 in a dose-dependent manner. We found that HPV16 E6 activated vascular endothelial growth factor (VEGF)-induced endothelial cell migration and proliferation as well as phosphorylation of VEGFR-2 via direct interaction in vitro. HPV16 E6 exhibited potent pro-angiogenic activity and clearly enhanced the levels of hypoxia-inducible factor-1α (HIF-1α). By contrast, the loss of function of HPV16 E6 by siRNA-mediated knockdown inhibited the cellular events. These data provide direct evidence that HPV16 E6 facilitates tumour growth and angiogenesis. HPV16 E6 also activates the PI3K/mTOR signalling cascades, and IRF-1 suppresses HPV16 E6-induced tumourigenesis and angiogenesis. Collectively, these findings suggest a biological mechanism underlying the HPV16 E6-related activity in cervical tumourigenesis.
Collapse
Affiliation(s)
- Seung Bae Rho
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Gyeonggido 411-769, Korea;
| | - Seung-Hoon Lee
- Department of Life Science, Yong In University, Yongin, Gyeonggido 449-714, Korea;
| | - Hyun-Jung Byun
- Phamaceutical Biochemistry, College of Pharmacy and Integrated Research Institute for Drug, Dongguk University, Goyang 100-715, Korea;
| | - Boh-Ram Kim
- Phamaceutical Biochemistry, College of Pharmacy and Integrated Research Institute for Drug, Dongguk University, Goyang 100-715, Korea;
- Correspondence: (B.-R.K.); (C.H.L.); Tel.: +82-31-961-5213 (B.-R.K. & C.H.L.)
| | - Chang Hoon Lee
- Phamaceutical Biochemistry, College of Pharmacy and Integrated Research Institute for Drug, Dongguk University, Goyang 100-715, Korea;
- Correspondence: (B.-R.K.); (C.H.L.); Tel.: +82-31-961-5213 (B.-R.K. & C.H.L.)
| |
Collapse
|
3
|
Duenas-Gonzalez A, Gonzalez-Fierro A. Pharmacodynamics of current and emerging treatments for cervical cancer. Expert Opin Drug Metab Toxicol 2019; 15:671-682. [DOI: 10.1080/17425255.2019.1648431] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Alfonso Duenas-Gonzalez
- Unit of Biomedical Research on Cancer, Instituto de Investigaciones Biomédicas UNAM/Instituto Nacional de Cancerología, Mexico City, Mexico
| | | |
Collapse
|
4
|
Single-Domain Antibodies Represent Novel Alternatives to Monoclonal Antibodies as Targeting Agents against the Human Papillomavirus 16 E6 Protein. Int J Mol Sci 2019; 20:ijms20092088. [PMID: 31035322 PMCID: PMC6539864 DOI: 10.3390/ijms20092088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 01/18/2023] Open
Abstract
Approximately one fifth of all malignancies worldwide are etiologically associated with a persistent viral or bacterial infection. Thus, there is a particular interest in therapeutic molecules which use components of a natural immune response to specifically inhibit oncogenic microbial proteins, as it is anticipated they will elicit fewer off-target effects than conventional treatments. This concept has been explored in the context of human papillomavirus 16 (HPV16)-related cancers, through the development of monoclonal antibodies and fragments thereof against the viral E6 oncoprotein. Challenges related to the biology of E6 as well as the functional properties of the antibodies themselves appear to have precluded their clinical translation. Here, we addressed these issues by exploring the utility of the variable domains of camelid heavy-chain-only antibodies (denoted as VHHs). Through construction and panning of two llama, immune VHH phage display libraries, a pool of potential VHHs was isolated. The interactions of these with recombinant E6 were further characterized using an enzyme-linked immunosorbent assay (ELISA), Western blotting under denaturing and native conditions, and surface plasmon resonance. Three VHHs were identified that bound recombinant E6 with nanomolar affinities. Our results lead the way for subsequent studies into the ability of these novel molecules to inhibit HPV16-infected cells in vitro and in vivo.
Collapse
|
5
|
Maillard PV, van der Veen AG, Poirier EZ, Reis e Sousa C. Slicing and dicing viruses: antiviral RNA interference in mammals. EMBO J 2019; 38:e100941. [PMID: 30872283 PMCID: PMC6463209 DOI: 10.15252/embj.2018100941] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/17/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
To protect against the harmful consequences of viral infections, organisms are equipped with sophisticated antiviral mechanisms, including cell-intrinsic means to restrict viral replication and propagation. Plant and invertebrate cells utilise mostly RNA interference (RNAi), an RNA-based mechanism, for cell-intrinsic immunity to viruses while vertebrates rely on the protein-based interferon (IFN)-driven innate immune system for the same purpose. The RNAi machinery is conserved in vertebrate cells, yet whether antiviral RNAi is still active in mammals and functionally relevant to mammalian antiviral defence is intensely debated. Here, we discuss cellular and viral factors that impact on antiviral RNAi and the contexts in which this system might be at play in mammalian resistance to viral infection.
Collapse
Affiliation(s)
- Pierre V Maillard
- Division of Infection and Immunity, University College London, London, UK
| | | | - Enzo Z Poirier
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | |
Collapse
|
6
|
Pottash AE, Kuffner C, Noonan-Shueh M, Jay SM. Protein-based vehicles for biomimetic RNAi delivery. J Biol Eng 2019; 13:19. [PMID: 30891095 PMCID: PMC6390323 DOI: 10.1186/s13036-018-0130-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/09/2018] [Indexed: 12/30/2022] Open
Abstract
Broad translational success of RNA interference (RNAi) technology depends on the development of effective delivery approaches. To that end, researchers have developed a variety of strategies, including chemical modification of RNA, viral and non-viral transfection approaches, and incorporation with delivery vehicles such as polymer- and lipid-based nanoparticles, engineered and native proteins, extracellular vesicles (EVs), and others. Among these, EVs and protein-based vehicles stand out as biomimetically-inspired approaches, as both proteins (e.g. Apolipoprotein A-1, Argonaute 2, and Arc) and EVs mediate intercellular RNA transfer physiologically. Proteins specifically offer significant therapeutic potential due to their biophysical and biochemical properties as well as their ability to facilitate and tolerate manipulation; these characteristics have made proteins highly successful translational therapeutic molecules in the last two decades. This review covers engineered protein vehicles for RNAi delivery along with what is currently known about naturally-occurring extracellular RNA carriers towards uncovering design rules that will inform future engineering of protein-based vehicles.
Collapse
Affiliation(s)
- Alex Eli Pottash
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Christopher Kuffner
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Madeleine Noonan-Shueh
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Steven M Jay
- 1Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA.,2Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA.,3Program in Molecular and Cellular Biology, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
7
|
Zhu D, Shen H, Tan S, Hu Z, Wang L, Yu L, Tian X, Ding W, Ren C, Gao C, Cheng J, Deng M, Liu R, Hu J, Xi L, Wu P, Zhang Z, Ma D, Wang H. Nanoparticles Based on Poly (β-Amino Ester) and HPV16-Targeting CRISPR/shRNA as Potential Drugs for HPV16-Related Cervical Malignancy. Mol Ther 2018; 26:2443-2455. [PMID: 30241742 DOI: 10.1016/j.ymthe.2018.07.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/24/2018] [Accepted: 07/11/2018] [Indexed: 12/21/2022] Open
Abstract
Persistent high-risk HPV infection is the main cause of cervical cancer. The HPV oncogene E7 plays an important role in HPV carcinogenesis. Currently, HPV vaccines do not offer an effective treatment for women who already present with cervical disease, and recommended periodical cervical screenings are difficult to perform in countries and areas lacking medical resources. Our aim was to develop nanoparticles (NPs) based on poly (β-amino ester) (PBAE) and HPV16 E7-targeting CRISPR/short hairpin RNA (shRNA) to reduce the levels of HPV16 E7 as a preliminary form of a drug to treat HPV infection and its related cervical malignancy. Our NPs showed low toxicity in cells and mouse organs. By reducing the expression of HPV16 E7, our NPs could inhibit the growth of cervical cancer cells and xenograft tumors in nude mice, and they could reverse the malignant cervical epithelium phenotype in HPV16 transgenic mice. The performance of NPs containing shRNA is better than that of NPs containing CRISPR. HPV-targeting NPs consisting of PBAE and CRISPR/shRNA could potentially be developed as drugs to treat HPV infection and HPV-related cervical malignancy.
Collapse
Affiliation(s)
- Da Zhu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui Shen
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Songwei Tan
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zheng Hu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Gynecological Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liming Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lan Yu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xun Tian
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wencheng Ding
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ci Ren
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chun Gao
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jing Cheng
- Department of Obstetrics and Gynecology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ming Deng
- Department of Radiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Rong Liu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junbo Hu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling Xi
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Peng Wu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ding Ma
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Hui Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
8
|
Chabeda A, Yanez RJR, Lamprecht R, Meyers AE, Rybicki EP, Hitzeroth II. Therapeutic vaccines for high-risk HPV-associated diseases. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2018; 5:46-58. [PMID: 29277575 PMCID: PMC5887015 DOI: 10.1016/j.pvr.2017.12.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/13/2017] [Accepted: 12/17/2017] [Indexed: 12/16/2022]
Abstract
Cancer is the second leading cause of death worldwide, and it is estimated that Human papillomavirus (HPV) related cancers account for 5% of all human cancers. Current HPV vaccines are extremely effective at preventing infection and neoplastic disease; however, they are prophylactic and do not clear established infections. Therapeutic vaccines which trigger cell-mediated immune responses for the treatment of established infections and malignancies are therefore required. The E6 and E7 early genes are ideal targets for vaccine therapy due to their role in disruption of the cell cycle and their constitutive expression in premalignant and malignant tissues. Several strategies have been investigated for the development of therapeutic vaccines, including live-vector, nucleic acid, peptide, protein-based and cell-based vaccines as well as combinatorial approaches, with several vaccine candidates progressing to clinical trials. With the current understanding of the HPV life cycle, molecular mechanisms of infection, carcinogenesis, tumour biology, the tumour microenvironment and immune response mechanisms, an approved HPV therapeutic vaccine seems to be a goal not far from being achieved. In this article, the status of therapeutic HPV vaccines in clinical trials are reviewed, and the potential for plant-based vaccine production platforms described.
Collapse
Affiliation(s)
- Aleyo Chabeda
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Romana J R Yanez
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Renate Lamprecht
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Ann E Meyers
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Edward P Rybicki
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Inga I Hitzeroth
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa.
| |
Collapse
|
9
|
Qureshi A, Tantray VG, Kirmani AR, Ahangar AG. A review on current status of antiviral siRNA. Rev Med Virol 2018; 28:e1976. [PMID: 29656441 PMCID: PMC7169094 DOI: 10.1002/rmv.1976] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/18/2018] [Accepted: 02/12/2018] [Indexed: 01/12/2023]
Abstract
Viral diseases like influenza, AIDS, hepatitis, and Ebola cause severe epidemics worldwide. Along with their resistant strains, new pathogenic viruses continue to be discovered so creating an ongoing need for new antiviral treatments. RNA interference is a cellular gene‐silencing phenomenon in which sequence‐specific degradation of target mRNA is achieved by means of complementary short interfering RNA (siRNA) molecules. Short interfering RNA technology affords a potential tractable strategy to combat viral pathogenesis because siRNAs are specific, easy to design, and can be directed against multiple strains of a virus by targeting their conserved gene regions. In this review, we briefly summarize the current status of siRNA therapy for representative examples from different virus families. In addition, other aspects like their design, delivery, medical significance, bioinformatics resources, and limitations are also discussed.
Collapse
Affiliation(s)
- Abid Qureshi
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Vaqar Gani Tantray
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Altaf Rehman Kirmani
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Abdul Ghani Ahangar
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| |
Collapse
|
10
|
Cerqueira C, Thompson CD, Day PM, Pang YYS, Lowy DR, Schiller JT. Efficient Production of Papillomavirus Gene Delivery Vectors in Defined In Vitro Reactions. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:165-179. [PMID: 28497074 PMCID: PMC5423317 DOI: 10.1016/j.omtm.2017.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/13/2017] [Indexed: 11/17/2022]
Abstract
Papillomavirus capsids can package a wide variety of nonviral DNA plasmids and deliver the packaged genetic material to cells, making them attractive candidates for targeted gene delivery vehicles. However, the papillomavirus vectors generated by current methods are unlikely to be suitable for clinical applications. We have developed a chemically defined, cell-free, papillomavirus-based vector production system that allows the incorporation of purified plasmid DNA (pseudogenome) into high-titer papillomavirus L1/L2 capsids. We investigated the incorporation of several DNA forms into a variety of different papillomavirus types, including human and animal types. Our results show that papillomavirus capsids can package and transduce linear or circular DNA under defined conditions. Packaging and transduction efficiencies were surprisingly variable across capsid types, DNA forms, and assembly reaction conditions. The pseudoviruses produced by these methods are sensitive to the same entry inhibitors as cell-derived pseudovirions, including neutralizing antibodies and heparin. The papillomavirus vector production systems developed in this study generated as high as 1011 infectious units/mg of L1. The pseudoviruses were infectious both in vitro and in vivo and should be compatible with good manufacturing practice (GMP) requirements.
Collapse
Affiliation(s)
- Carla Cerqueira
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cynthia D. Thompson
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Patricia M. Day
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuk-Ying S. Pang
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Douglas R. Lowy
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John T. Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Corresponding author: John T. Schiller, Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 37, Room 4112B, Bethesda, MD 20892, USA.
| |
Collapse
|
11
|
Shaikh MH, Clarke DTW, Johnson NW, McMillan NAJ. Can gene editing and silencing technologies play a role in the treatment of head and neck cancer? Oral Oncol 2017; 68:9-19. [PMID: 28438299 DOI: 10.1016/j.oraloncology.2017.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/25/2017] [Accepted: 02/19/2017] [Indexed: 01/04/2023]
Abstract
Conventional treatment strategies have done little to improve the prognosis or disease-free survival in head and neck cancer (HNC) patients. Recent progress in our understanding of molecular aspects of head and neck squamous cell carcinoma (HNSCC) has provided insights into the potential use of molecular targeted therapies in combination with current treatment strategies. Here we review the current understanding of treatment modalities for both HPV-positive and HPV-negative HNSCCs with the potential to use gene editing and silencing technologies therapeutically. The development of sequence-specific RNA interference (RNAi) with its strong gene-specific silencing ability, high target specificity, greater potency and reduced side effects, has shown it to be a promising therapeutic candidate for treating cancers. CRISPR/Cas gene editing is the newest technology with the ability to delete, mutate or replace genes of interest and has great potential for treating HNSCCs. We also discuss the major challenge in using these approaches in HNSCC; that being the choice of target and the ability to deliver the payload. Finally, we highlight the potential combination of RNAi or CRIPSR/Cas with current treatment strategies and outline the possible path to the clinic.
Collapse
Affiliation(s)
- Mushfiq H Shaikh
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia; School of Medical Science, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia; Understanding Chronic Conditions Program, Menzies Health Institute Queensland, Australia.
| | - Daniel T W Clarke
- School of Medical Science, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia; Understanding Chronic Conditions Program, Menzies Health Institute Queensland, Australia.
| | - Newell W Johnson
- School of Dentistry and Oral Health, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia; Understanding Chronic Conditions Program, Menzies Health Institute Queensland, Australia.
| | - Nigel A J McMillan
- School of Medical Science, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia; Understanding Chronic Conditions Program, Menzies Health Institute Queensland, Australia.
| |
Collapse
|
12
|
Li L, Xu C, Long J, Shen D, Zhou W, Zhou Q, Yang J, Jiang M. E6 and E7 gene silencing results in decreased methylation of tumor suppressor genes and induces phenotype transformation of human cervical carcinoma cell lines. Oncotarget 2016; 6:23930-43. [PMID: 26329329 PMCID: PMC4695162 DOI: 10.18632/oncotarget.4525] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 06/02/2015] [Indexed: 01/01/2023] Open
Abstract
In SiHa and CaSki cells, E6 and E7-targeting shRNA specifically and effectively knocked down human papillomavirus (HPV) 16 E6 and E7 at the transcriptional level, reduced the E6 and E7 mRNA levels by more than 80% compared with control cells that expressed a scrambled-sequence shRNA. E6 and E7 repression resulted in down-regulation of DNA methyltransferase mRNA and protein expression, decreased DNA methylation and increased mRNA expression levels of tumor suppressor genes, induced a certain apoptosis and inhibited proliferation in E6 and E7 shRNA-infected SiHa and CaSki cells compared with the uninfected cells. Repression of E6 and E7 oncogenes resulted in restoration of DNA methyltransferase suppressor pathways and induced apoptosis in HPV16-positive cervical carcinoma cell lines. Our findings suggest that the potential carcinogenic mechanism of HPV16 through influencing DNA methylation pathway to activate the development of cervical cancer exist, and maybe as a candidate therapeutic strategy for cervical and other HPV-associated cancers.
Collapse
Affiliation(s)
- Liming Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Cui Xu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jia Long
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Danbei Shen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wuqing Zhou
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Qiyan Zhou
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jia Yang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Mingjun Jiang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
13
|
Serrano-Olvera A, Cetina L, Coronel J, Dueñas-González A. Emerging drugs for the treatment of cervical cancer. Expert Opin Emerg Drugs 2015; 20:165-82. [PMID: 25578210 DOI: 10.1517/14728214.2015.1002768] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Worldwide, most cervical cancer (CC) patients require the use of drug therapy either adjuvant, concurrent with radiation or palliative. AREAS COVERED This review briefly discusses the current achievements in treating CC with an emphasis in emerging agents. EXPERT OPINION Concurrent cisplatin with radiation and lately, gemcitabine-cisplatin chemoradiation has resulted in small but significant improvements in the treatment of locally advanced and high-risk early-stage patients. So far, only antiangiogenic therapy with bevacizumab added to cisplatin chemoradiation has demonstrated safety and encouraging results in a Phase II study. In advanced disease, cisplatin doublets yield median survival rates not exceeding 14 months. The first Phase III study of bevacizumab, added to standard chemotherapy cisplatin- or non-cisplatin-containing doublet, has shown significant increase in both overall survival and progression-free survival. Further studies are needed before bevacizumab plus chemotherapy can be considered the standard of care for advanced disease. The characterization of the mutational landscape of CC and developments of novel targeted therapies may result in more effective and individualized treatments for CC. The potential efficacy of knocking down the key alterations in CC, E6 and E7 human papilloma virus oncoproteins must not be overlooked.
Collapse
|
14
|
Samimi M, Gardair C, Nicol JTJ, Arnold F, Touzé A, Coursaget P. Merkel cell polyomavirus in merkel cell carcinoma: clinical and therapeutic perspectives. Semin Oncol 2014; 42:347-58. [PMID: 25843739 DOI: 10.1053/j.seminoncol.2014.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Merkel cell carcinoma (MCC) is a rare and often aggressive cutaneous cancer with a poor prognosis. The incidence of this cancer increases with age, immunodeficiency and sun exposure. Merkel cell polyomavirus (MCPyV), a new human polyomavirus identified in 2008, is detected in the majority of the MCCs and there is a growing body of evidence that healthy human skin harbors resident or transient MCPyV. A causal link between MCPyV and MCC has been evidenced and this is the first polyomavirus to be clearly implicated as a causal agent underlying a human cancer, and MCPyV was recently classified as a 2A carcinogen. MCC is thus a rare tumor caused by a very common viral skin infection. The aim of this review is to provide a basic overview of the epidemiological, clinical, and pathological characteristics of MCC, to present the current knowledge on MCPyV polyomavirus and its causal association with MCC development, and to describe the therapeutic implications of this causal link.
Collapse
Affiliation(s)
- Mahtab Samimi
- Université François Rabelais, Tours, France; CHRU de Tours-Hôpital Trousseau, Service de Dermatologie, Tours, France; Unité Mixte de Recherche INRA-Univerity of Tours N°1282, Tours, France
| | - Charlotte Gardair
- CHRU de Tours-Hôpital Trousseau, Service d׳Anatomie et Cytologie Pathologiques, Tours, France
| | - Jérome T J Nicol
- Université François Rabelais, Tours, France; Unité Mixte de Recherche INRA-Univerity of Tours N°1282, Tours, France
| | - Francoise Arnold
- Université François Rabelais, Tours, France; Unité Mixte de Recherche INRA-Univerity of Tours N°1282, Tours, France
| | - Antoine Touzé
- Université François Rabelais, Tours, France; Unité Mixte de Recherche INRA-Univerity of Tours N°1282, Tours, France
| | | |
Collapse
|
15
|
Duenas-Gonzalez A, Serrano-Olvera A, Cetina L, Coronel J. New molecular targets against cervical cancer. Int J Womens Health 2014; 6:1023-31. [PMID: 25525394 PMCID: PMC4266260 DOI: 10.2147/ijwh.s49471] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cervical cancer is the third most commonly diagnosed cancer worldwide and the fourth leading cause of cancer death in women. Major advances but still insufficient achievements in the treatment of locally advanced and high-risk early stage patients have occurred in the last decade with the incorporation of concurrent cisplatin with radiation and, lately, gemcitabine added to cisplatin chemoradiation. Despite a number of clinical studies incorporating molecular-targeted therapy as radiosensitizers being in progress, so far, only antiangiogenic therapy with bevacizumab added to cisplatin chemoradiation has demonstrated safety and shown encouraging results in a Phase II study. In advanced disease, cisplatin doublets do not have a great impact on the natural history of the disease with median survival rates not exceeding 13 months. The first Phase III study of bevacizumab, added to cisplatin or a non-cisplatin-containing doublet, showed significant increase in both overall survival and progression-free survival. Further studies are needed before bevacizumab plus chemotherapy can be considered the standard of care for advanced disease. Characterization of the mutational landscape of cervical cancer has already been initiated, indicating that, for now, few of these targetable alterations match with available agents. Progress in both the mutational landscape knowledge and developments of novel targeted therapies may result in more effective and individualized treatments for cervical cancer. The potential efficacy of knocking down the key alterations in cervical cancer – E6 and E7 human papillomavirus oncoproteins – must not be overlooked.
Collapse
Affiliation(s)
- Alfonso Duenas-Gonzalez
- Unit of Biomedical Research in Cancer, Instituto de Investigaciones Biomedicas UNAM/Instituto Nacional de Cancerologia, Mexico City, Mexico ; ISSEMyM Cancer Center, Toluca, Mexico
| | | | - Lucely Cetina
- Division of Clinical Research, Instituto Nacional de Cancerologia, Mexico City, Mexico
| | - Jaime Coronel
- Division of Clinical Research, Instituto Nacional de Cancerologia, Mexico City, Mexico
| |
Collapse
|
16
|
Leija-Montoya AG, Benítez-Hess ML, Toscano-Garibay JD, Alvarez-Salas LM. Characterization of an RNA aptamer against HPV-16 L1 virus-like particles. Nucleic Acid Ther 2014; 24:344-55. [PMID: 25111024 PMCID: PMC4162430 DOI: 10.1089/nat.2013.0469] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 07/15/2014] [Indexed: 01/05/2023] Open
Abstract
The human papillomavirus (HPV) capsid is mainly composed of the L1 protein that can self-assemble into virus-like particles (VLPs) that are structurally and immunologically similar to the infectious virions. We report here the characterization of RNA aptamers that recognize baculovirus-produced HPV-16 L1 VLPs. Interaction and slot-blot binding assays showed that all isolated aptamers efficiently bound HPV-16 VLPs, although the Sc5-c3 aptamer showed the highest specificity and affinity (Kd=0.05 pM). Sc5-c3 secondary structure consisted of a hairpin with a symmetric bubble and an unstructured 3'end. Biochemical and genetic analyses showed that the Sc5-c3 main loop is directly involved on VLPs binding. In particular, binding specificity appeared mediated by five non-consecutive nucleotide positions. Experiments using bacterial-produced HPV-16 L1 resulted in low Sc5-c3 binding, suggesting that recognition of HPV-16 L1 VLPs relies on quaternary structure features not present in bacteria-produced L1 protein. Sc5-c3 produced specific and stable binding to HPV-16 L1 VLPs even in biofluid protein mixes and thus it may provide a potential diagnostic tool for active HPV infection.
Collapse
Affiliation(s)
- Ana Gabriela Leija-Montoya
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México D.F., México
| | - María Luisa Benítez-Hess
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México D.F., México
| | | | - Luis Marat Alvarez-Salas
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México D.F., México
| |
Collapse
|
17
|
Durzyńska J. IGF axis and other factors in HPV-related and HPV-unrelated carcinogenesis (review). Oncol Rep 2014; 32:2295-306. [PMID: 25333772 PMCID: PMC4240475 DOI: 10.3892/or.2014.3505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022] Open
Abstract
The insulin-like growth factor (IGF) axis promotes the growth of cells, tissues and organs. IGF-1 is mainly produced in the liver but is also secreted from local tissues. In the circulation, IGF-1 is bound to insulin-like binding proteins (IGFBPs), and when released it activates the insulin-like growth factor receptor (IGF-1R). The signal is further transmitted by intracellular signaling pathways leading to gene expression that regulates, among others, cell proliferation and survival. This review presents the IGF axis in the context of cell transformation and cancer development. Aspects involving IGF-1 deficiency and protection from cancer are also briefly described. Furthermore, human papillomaviruses (HPVs) interplaying with IGF axis components in cervical cancer development are described. These small dsDNA viruses are divided into low-risk and high-risk HPVs with regard to the potency of their oncogenic actions; they mainly infect epithelial or mucosal cells. Special attention is drawn to expression of two major HPV oncogenes (E6 and E7) initiating and maintaining cervical carcinogenesis, which is a multistep and multifactorial process; therefore, involvement of additional factors such as mitochondrial DNA changes, sex hormones, retinoic and folic acids are also discussed. Finally, IGF axis components and HPV oncogenes as targets in anticancer treatment are presented which include IGF-1R downregulation, RNA interference and anti-HPV therapeutic vaccines. The review concludes that despite an enormous advancement in research on IGF and HPV-related cancers, more molecular studies and clinical trials are needed before commercialized therapies are widely available for oncology patients.
Collapse
Affiliation(s)
- Julia Durzyńska
- Department of Molecular Virology, Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, 60-614 Poznań, Poland
| |
Collapse
|
18
|
Human Papillomavirus Type 16 Pseudovirions with Few Point Mutations in L1 Major Capsid Protein FG Loop Could Escape Actual or Future Vaccination for Potential Use in Gene Therapy. Mol Biotechnol 2014; 56:479-86. [DOI: 10.1007/s12033-014-9745-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
19
|
Qureshi A, Thakur N, Kumar M. VIRsiRNApred: a web server for predicting inhibition efficacy of siRNAs targeting human viruses. J Transl Med 2013; 11:305. [PMID: 24330765 PMCID: PMC3878835 DOI: 10.1186/1479-5876-11-305] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/22/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Selection of effective viral siRNA is an indispensable step in the development of siRNA based antiviral therapeutics. Despite immense potential, a viral siRNA efficacy prediction algorithm is still not available. Moreover, performances of the existing general mammalian siRNA efficacy predictors are not satisfactory for viral siRNAs. Therefore, we have developed "VIRsiRNApred" a support vector machine (SVM) based method for predicting the efficacy of viral siRNA. METHODS In the present study, we have employed a new dataset of 1725 viral siRNAs with experimentally verified quantitative efficacies tested under heterogeneous experimental conditions and targeting as many as 37 important human viruses including HIV, Influenza, HCV, HBV, SARS etc. These siRNAs were divided into training (T1380) and validation (V345) datasets. Important siRNA sequence features including mono to penta nucleotide frequencies, binary pattern, thermodynamic properties and secondary structure were employed for model development. RESULTS During 10-fold cross validation on T1380 using hybrid approach, we achieved a maximum Pearson Correlation Coefficient (PCC) of 0.55 between predicted and actual efficacy of viral siRNAs. On V345 independent dataset, our best model achieved a maximum correlation of 0.50 while existing general siRNA prediction methods showed PCC from 0.05 to 0.18. However, using leave one out cross validation PCC was improved to 0.58 and 0.55 on training and validation datasets respectively. SVM performed better than other machine learning techniques used like ANN, KNN and REP Tree. CONCLUSION VIRsiRNApred is the first algorithm for predicting inhibition efficacy of viral siRNAs which is developed using experimentally verified viral siRNAs. We hope this algorithm would be useful in predicting highly potent viral siRNA to aid siRNA based antiviral therapeutics development. The web server is freely available at http://crdd.osdd.net/servers/virsirnapred/.
Collapse
Affiliation(s)
| | | | - Manoj Kumar
- Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research, Sector 39-A, Chandigarh 160036, India.
| |
Collapse
|
20
|
Weyland M, Griveau A, Bejaud J, Benoit JP, Coursaget P, Garcion E. Lipid nanocapsule functionalization by lipopeptides derived from human papillomavirus type-16 capsid for nucleic acid delivery into cancer cells. Int J Pharm 2013; 454:756-64. [PMID: 23769994 DOI: 10.1016/j.ijpharm.2013.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/06/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
Abstract
Plasmid DNA (pDNA) and small interfering RNAs (siRNAs) are very useful tools for the treatment of cancer. However, pDNA and siRNAs efficacy is restricted by their negative charge and susceptibility to degradation by endonucleases that prevent them penetrating tissue and cellular barriers such as the plasma and endolysosomal membranes. Viral vectors have some advantages but their use is largely limited by their immunogenicity. On the other hand, synthetic nanoparticles have advantage of being relatively non-immunogenic but their ability to deliver nucleic acids remains less efficient than their viral counterparts. The present study is focussed on the development and evaluation of biomimetic lipid nanocapsules (LNCs) functionalized with a L1 papillomavirus type-16 capsid-derived lipopeptide on their surface, for transfection of U87MG glioma cells and Caco-2 colorectal adenocarcinoma cells with pDNA or siRNAs. Since the L1-peptide has been described as a nuclear localization signal able to complex with nucleic acids and bind to heparan sulfate on the cell surface, the structure and function of L1-peptide bound to LNCs (L1-LNCs) were investigated. Although L1-LNCs were shown to complex with both pDNA and siRNAs, the pDNA-L1-LNC complexes showed only weak transfection efficiency. In contrast, siRNA-L1-LNC complexes appeared as effective repressors of targeted messengers.
Collapse
Affiliation(s)
- M Weyland
- Inserm U1066, Micro et nanomédecines biomimétiques, F-49933 Angers, France
| | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Dueñas-Gonzalez A, Cetina L, Coronel J, Cervantes-Madrid D. Emerging drugs for cervical cancer. Expert Opin Emerg Drugs 2012; 17:203-18. [DOI: 10.1517/14728214.2012.683409] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
23
|
Ajay AK, Meena AS, Bhat MK. Human papillomavirus 18 E6 inhibits phosphorylation of p53 expressed in HeLa cells. Cell Biosci 2012; 2:2. [PMID: 22244155 PMCID: PMC3285035 DOI: 10.1186/2045-3701-2-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 01/13/2012] [Indexed: 11/22/2022] Open
Abstract
Background In HPV infected cells p53 function is abrogated by E6 and even ectopically expressed p53 is unable to perform tumor suppressor functions. In addition to facilitating its degradation, E6 may also inhibit p53 transactivity, though the mechanisms are still poorly understood. It has been reported that inhibition of p300, an acetyltransferase responsible for p53 acetylation is inactivated by E6. Activation of overexpressed p53 to cause cell growth inhibition is facilitated by its phosphorylation. Previously, we reported that non-genotoxically overexpressed p53 in HeLa cells needs to be phosphorylated to perform its cell growth inhibitory functions. Since over expressed p53 by itself was not activated, we hypothesized an inhibitory role for E6. Results Majority of reports proposes E6 mediated degradation of p53 as a possible reason for its inactivation. However, results presented here for the first time demonstrate that overexpressed p53 is not directly associated with E6 and therefore free, yet it is not functionally active in HPV positive cells. Also, the stability of overexpressed p53 does not seem to be an issue because inhibition of proteasomal degradation did not increase the half-life of overexpressed p53, which is more than endogenous p53. However, inhibition of proteasomal degradation prevents the degradation of endogenous p53. These findings suggest that overexpressed p53 and endogenous p53 are differentially subjected to proteasomal degradation and the reasons for this discrepancy remain unclear. Our studies demonstrate that p53 over expression has no effect on anchorage independent cell-growth and E6 nullifies its cell growth inhibitory effect. E6 overexpression abrogates OA induced p53 occupancy on the p21 promoter and cell death as well. E6 did not decrease p53 protein but phospho-p53 level was significantly reduced. Conclusion We report for the first time that E6 de-activates p53 by inhibiting its phosphorylation. This prevents p53 binding to p21 promoter and thereby restraining its cell-growth inhibitory functions. Our study provides new evidence indicating that viral protein E6 inhibits p53 transactivity by mechanism independent of degradation pathway.
Collapse
Affiliation(s)
- Amrendra K Ajay
- National Centre for Cell Science, NCCS Complex, Pune University Campus, Ganeshkhind, Pune - 411007, India.
| | | | | |
Collapse
|
24
|
Zhou J, Peng C, Li B, Wang F, Zhou C, Hong D, Ye F, Cheng X, Lü W, Xie X. Transcriptional gene silencing of HPV16 E6/E7 induces growth inhibition via apoptosis in vitro and in vivo. Gynecol Oncol 2011; 124:296-302. [PMID: 22056554 DOI: 10.1016/j.ygyno.2011.10.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 10/20/2011] [Accepted: 10/25/2011] [Indexed: 10/15/2022]
Abstract
OBJECTIVE Transcriptional silencing of HPV oncogenes using short interfering RNA (siRNA) blocks E6/E7 expression. Our objective was to estimate the effective value of E6/E7 specific siRNA-induced transcriptional gene silencing as a potential therapeutic strategy for cervical cancer. METHODS In vitro studies were performed by employing two categories of siRNA targeting promoter of E6/E7 gene and E7 transcript, respectively, and inhibitory effect of both siRNAs was further observed in vitro and on xenograft in BALB/c mice that were inoculated with siRNA transfected SiHa cells and parental SiHa cells followed by siRNA intratumoral injection in vivo. Tumor volume and growth curves were assessed. Furthermore, cellular proliferation and apoptosis of inoculated tumors were determined by immunohistochemistry staining and TUNEL assay. RESULTS The two most active siRNA sequences specifically knockdown E6/E7 expressions at mRNA level in HPV16 positive Siha cells, increased p53 and decreased p16 expressions at protein level, inhibited cell proliferation, and induced cell apoptosis in vitro. Furthermore, both siRNAs effectively inhibited tumor formation and growth no matter in mice with siRNA transfected cells in vitro or with siRNA intratumoral injection in vivo. TUNEL staining and FCM assay consistently showed that tumor retardation was through induction of cellular apoptosis. CONCLUSION RNAi targeting the promoter of HPV16 E6/E7 acts effectively in vitro and in vivo, especially through intratumoral delivery, and may be a candidate therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Jiansong Zhou
- Women's Reproductive Health Laboratory of Zhejiang Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The simian virus 40 and murine polyomaviruses were shown to be DNA tumor viruses in their natural hosts and/or heterologous experimental hosts in the mid-20th Century. The first two human polyomaviruses, the BK polyomavirus and JC polyomavirus, were discovered in 1971 and were shown to induce severe disease in immunocompromised patients, but their involvement in human cancers is still a matter for debate. The discovery of a polyomavirus associated with Merkel cell carcinoma (Merkel cell polyomavirus) in 2008 resulted in a renewed interest in the Polyomaviridae family, leading to the discovery of new human polyomaviruses. This review addresses the involvement of the nine human polyomaviruses and simian virus 40 in human diseases, with a particular focus on their prevalence and the humoral response directed against structural antigens in the general population and in subjects presenting specific diseases.
Collapse
Affiliation(s)
- Jérôme TJ Nicol
- Université François Rabelais, INSERM U618 Equipe Vecteurs, Virus, Vaccins. Faculté des Sciences Pharmaceutiques Philippe Maupas, 31 avenue Monge, 37200 TOURS, France
| | - Antoine Touzé
- Université François Rabelais, INSERM U618 Equipe Vecteurs, Virus, Vaccins. Faculté des Sciences Pharmaceutiques Philippe Maupas, 31 avenue Monge, 37200 TOURS, France
| |
Collapse
|
26
|
Buonaguro FM, Tornesello ML, Buonaguro L. New adjuvants in evolving vaccine strategies. Expert Opin Biol Ther 2011; 11:827-32. [PMID: 21609186 DOI: 10.1517/14712598.2011.587802] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adjuvants are becoming the key players of vaccine formulations to enhance the immunogenicity of subunit (peptides, proteins, virus-like particles (VLPs)) and DNA vaccines, as well as to reach the current new goals of preventing and/or treating chronic infectious diseases and cancers. Induction of humoral response, in particular neutralizing antibodies able to inhibit the binding of pathogens to their cellular receptors, remains a major goal of vaccines targeted to prevent acute lytic infections; induction/modulation of cellular immunity is, however, critical to fight latently/chronically infected cells as well as cancer cells. The new adjuvants, included in vaccine preparations, are currently able to modify the presentation of epitopes to the immune system with a specific T(H)1 versus T(H)2 polarization efficacy. A paradigm of the relevance of these new adjuvants is the immunological result obtained with the inclusion of monophosphoryl lipid A in the formulation of L1-based human papillomavirus (HPV)-naked VLPs. In the May issue of this journal, Garcon and colleagues describe the highly enhanced humoral and memory B cellular immunity of the AS04-adjuvanted HPV vaccine, which results in a long-lasting and broad spectrum immunity.
Collapse
Affiliation(s)
- Franco M Buonaguro
- Molecular Biology and Viral Oncology, Dpt of Experimental Oncology, Istituto Nazionale Tumori Fond Pascale, Napoli, Italy.
| | | | | |
Collapse
|
27
|
Effects of HMGB1 expression suppressed by siRNA on cell cycle and proliferation of human cervical cancer cell line HeLa. ACTA ACUST UNITED AC 2010. [DOI: 10.1007/s11805-010-0507-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Combelas N, Saussereau E, Fleury MJJ, Ribeiro T, Gaitan J, Duarte-Forero DF, Coursaget P, Touzé A. Papillomavirus pseudovirions packaged with the L2 gene induce cross-neutralizing antibodies. J Transl Med 2010; 8:28. [PMID: 20334659 PMCID: PMC2852459 DOI: 10.1186/1479-5876-8-28] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 03/24/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Current vaccines against HPVs are constituted of L1 protein self-assembled into virus-like particles (VLPs) and they have been shown to protect against natural HPV16 and HPV18 infections and associated lesions. In addition, limited cross-protection has been observed against closely related types. Immunization with L2 protein in animal models has been shown to provide cross-protection against distant papillomavirus types, suggesting that the L2 protein contains cross-neutralizing epitopes. However, vaccination with L2 protein or L2 peptides does not induce high titers of anti-L2 antibodies. In order to develop a vaccine with the potential to protect against other high-risk HPV types, we have produced HPV58 pseudovirions encoding the HPV31 L2 protein and compared their capacity to induce cross-neutralizing antibodies with that of HPV L1 and HPV L1/L2 VLPs. METHODS The titers of neutralizing antibodies against HPV16, HPV18, HPV31 and HPV58 induced in Balb/c mice were compared after immunization with L2-containing vaccines. RESULTS Low titers of cross-neutralizing antibodies were detected in mice when immunized with L1/L2 VLPs, and the highest levels of cross-neutralizing antibodies were observed in mice immunized with HPV 58 L1/L2 pseudovirions encoding the HPV 31 L2 protein. CONCLUSIONS The results obtained indicate that high levels of cross-neutralizing antibodies are only observed after immunization with pseudovirions encoding the L2 protein. HPV pseudovirions thus represent a possible new strategy for the generation of a broad-spectrum vaccine to protect against high-risk HPVs and associated neoplasia.
Collapse
Affiliation(s)
- Nicolas Combelas
- Inserm U618 Protéases et vectorisation pulmonaires, Tours, University François Rabelais, Tours, France and IFR 136 Agents Transmissibles et Infectiologie, Tours, France.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Buonaguro FM, Tornesello ML, Buonaguro L. Virus-like particle vaccines and adjuvants: the HPV paradigm. Expert Rev Vaccines 2009; 8:1379-98. [PMID: 19803760 DOI: 10.1586/erv.09.81] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Complex antigen structures currently represent the most-studied approach for prophylactic as well as therapeutic vaccines. Different types of complex vaccines, including virus-like particles and virosomes, have been developed depending on the nature of the viral pathogen they are trying to replicate (enveloped vs naked) or the modality to express antigenic epitopes (i.e., the binding of envelope protein on liposomic structures). The complex structure of these vaccines provides them with some adjuvanted properties, not uniformly present for all virus-like particle types. The further inclusion of specific adjuvants in vaccine preparations can modify the presentation modality of such particles to the immune system with a specific Th1 versus Th2 polarization efficacy. A paradigm of the relevance of these new adjuvants are the immunological results obtained with the inclusion of monophosphoryl lipid A adjuvant in the formulation of L1-based human papillomavirus-naked virus-like particles to reduce a Th1 cellular immunity impairment, peculiar for alum-derived adjuvants, along with the induction of highly enhanced humoral and memory B-cellular immunity.
Collapse
Affiliation(s)
- Franco Maria Buonaguro
- Molecular Biology and Viral Oncology, Department of Experimental Oncology, Istituto Nazionale Tumori Fond Pascale, Via Mariano Semmola 142, 80131 Napoli, Italy.
| | | | | |
Collapse
|
30
|
Wang SL, Yao HH, Qin ZH. Strategies for short hairpin RNA delivery in cancer gene therapy. Expert Opin Biol Ther 2009; 9:1357-68. [DOI: 10.1517/14712590903236843] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|