1
|
Orf Virus-Based Therapeutic Vaccine for Treatment of Papillomavirus-Induced Tumors. J Virol 2020; 94:JVI.00398-20. [PMID: 32404527 DOI: 10.1128/jvi.00398-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Orf virus (ORFV) represents a suitable vector for the generation of efficient, prophylactic antiviral vaccines against different pathogens. The present study investigated for the first time the therapeutic application of ORFV vector-based vaccines against tumors induced by cottontail rabbit papillomavirus (CRPV). ORFV-CRPV recombinants were constructed expressing the early CRPV gene E1, E2, E7, or LE6. In two independent experiments we used in total 23 rabbits which were immunized with a mixture of the four ORFV-CRPV recombinants or empty ORFV vector as a control 5 weeks after the appearance of skin tumors. For the determination of the therapeutic efficacy, the subsequent growth of the tumors was recorded. In the first experiment, we could demonstrate that three immunizations of rabbits with high tumor burden with the combined four ORFV-CRPV recombinants resulted in significant growth retardation of the tumors compared to the control. A second experiment was performed to test the therapeutic effect of 5 doses of the combined vaccine in rabbits with a lower tumor burden than in nonimmunized rabbits. Tumor growth was significantly reduced after immunization, and one vaccinated rabbit even displayed complete tumor regression until the end of the observation period at 26 weeks. Results of delayed-type hypersensitivity (DTH) skin tests suggest the induction of a cellular immune response mediated by the ORFV-CRPV vaccine. The data presented show for the first time a therapeutic potential of the ORFV vector platform and encourage further studies for the development of a therapeutic vaccine against virus-induced tumors.IMPORTANCE Viral vectors are widely used for the development of therapeutic vaccines for the treatment of tumors. In our study we have used Orf virus (ORFV) strain D1701-V for the generation of recombinant vaccines expressing cottontail rabbit papillomavirus (CRPV) early proteins E1, E2, LE6, and E7. The therapeutic efficacy of the ORFV-CRPV vaccines was evaluated in two independent experiments using the outbred CRPV rabbit model. In both experiments the immunization achieved significant suppression of tumor growth. In total, 84.6% of all outbred animals benefited from the ORFV-CRPV vaccination, showing reduction in tumor size and significant tumor growth inhibition, including one animal with complete tumor regression without recurrence.
Collapse
|
2
|
Christensen ND, Budgeon LR, Cladel NM, Hu J. Recent advances in preclinical model systems for papillomaviruses. Virus Res 2016; 231:108-118. [PMID: 27956145 DOI: 10.1016/j.virusres.2016.12.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 12/05/2016] [Indexed: 01/09/2023]
Abstract
Preclinical model systems to study multiple features of the papillomavirus life cycle have greatly aided our understanding of Human Papillomavirus (HPV) biology, disease progression and treatments. The challenge to studying HPV in hosts is that HPV along with most PVs are both species and tissue restricted. Thus, fundamental properties of HPV viral proteins can be assessed in specialized cell culture systems but host responses that involve innate immunity and host restriction factors requires preclinical surrogate models. Fortunately, there are several well-characterized and new animal models of papillomavirus infections that are available to the PV research community. Old models that continue to have value include canine, bovine and rabbit PV models and new rodent models are in place to better assess host-virus interactions. Questions arise as to the strengths and weaknesses of animal PV models for HPV disease and how accurately these preclinical models predict malignant progression, vaccine efficacy and therapeutic control of HPV-associated disease. In this review, we examine current preclinical models and highlight the strengths and weaknesses of the various models as well as provide an update on new opportunities to study the numerous unknowns that persist in the HPV research field.
Collapse
Affiliation(s)
- Neil D Christensen
- Department of Pathology and Microbiology and Immunology, Penn State College of Medicine, 500 University Drive, Hershey PA 17033, USA.
| | - Lynn R Budgeon
- Department of Pathology and Microbiology and Immunology, Penn State College of Medicine, 500 University Drive, Hershey PA 17033, USA
| | - Nancy M Cladel
- Department of Pathology and Microbiology and Immunology, Penn State College of Medicine, 500 University Drive, Hershey PA 17033, USA
| | - Jiafen Hu
- Department of Pathology and Microbiology and Immunology, Penn State College of Medicine, 500 University Drive, Hershey PA 17033, USA
| |
Collapse
|
3
|
Cladel NM, Budgeon LR, Balogh KK, Cooper TK, Hu J, Christensen ND. Mouse papillomavirus MmuPV1 infects oral mucosa and preferentially targets the base of the tongue. Virology 2015; 488:73-80. [PMID: 26609937 DOI: 10.1016/j.virol.2015.10.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/15/2015] [Accepted: 10/29/2015] [Indexed: 01/31/2023]
Abstract
In 2010, a new mouse papillomavirus, MmuPV1, was discovered in a colony of NMRI- Foxn1(nu)/Foxn1(nu) athymic mice in India. This finding was significant because it was the first papillomavirus to be found in a laboratory mouse. In this paper we report successful infections of both dorsal and ventral surfaces of the rostral tongues of outbred athymic nude mice. We also report the observation that the base of the tongue, the area of the tongue often targeted by cancer-associated high-risk papillomavirus infections in humans, is especially susceptible to infection. A suitable animal model for the study of oral papillomavirus infections, co-infections, and cancers has long been sought. The work presented here suggests that such a model is now at hand.
Collapse
Affiliation(s)
- Nancy M Cladel
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, United States; Department of Pathology, Pennsylvania State University College of Medicine, United States.
| | - Lynn R Budgeon
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, United States; Department of Pathology, Pennsylvania State University College of Medicine, United States
| | - Karla K Balogh
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, United States; Department of Pathology, Pennsylvania State University College of Medicine, United States
| | - Timothy K Cooper
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, United States
| | - Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, United States; Department of Pathology, Pennsylvania State University College of Medicine, United States
| | - Neil D Christensen
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, United States; Department of Pathology, Pennsylvania State University College of Medicine, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, United States
| |
Collapse
|
4
|
A novel pre-clinical murine model to study the life cycle and progression of cervical and anal papillomavirus infections. PLoS One 2015; 10:e0120128. [PMID: 25803616 PMCID: PMC4372414 DOI: 10.1371/journal.pone.0120128] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/19/2015] [Indexed: 02/02/2023] Open
Abstract
Background Papillomavirus disease and associated cancers remain a significant health burden in much of the world. The current protective vaccines, Gardasil and Cervarix, are expensive and not readily available to the underprivileged. In addition, the vaccines have not gained wide acceptance in the United States nor do they provide therapeutic value. Papillomaviruses are strictly species specific and thus human viruses cannot be studied in an animal host. An appropriate model for mucosal disease has long been sought. We chose to investigate whether the newly discovered mouse papillomavirus, MmuPV1, could infect mucosal tissues in Foxn1nu/Foxn1nu mice. Methods The vaginal and anal canals of Foxn1nu/Foxn1nu mice were gently abraded using Nonoxynol-9 and “Doctor’s BrushPicks” and MmuPV1 was delivered into the vaginal tract or the anal canal. Results Productive vaginal, cervical and anal infections developed in all mice. Vaginal/cervical infections could be monitored by vaginal lavage. Dysplasias were evident in all animals. Conclusions Anogenital tissues of a common laboratory mouse can be infected with a papillomavirus unique to that animal. This observation will pave the way for fundamental virological and immunological studies that have been challenging to carry out heretofore due to lack of a suitable model system.
Collapse
|
5
|
Secondary infections, expanded tissue tropism, and evidence for malignant potential in immunocompromised mice infected with Mus musculus papillomavirus 1 DNA and virus. J Virol 2013; 87:9391-5. [PMID: 23785210 DOI: 10.1128/jvi.00777-13] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Papillomavirus disease poses a special challenge to people with compromised immune systems. Appropriate models to study infections in these individuals are lacking. We report here the development of a model that will help to address these deficiencies. The MmuPV1 genome was synthesized and used successfully to produce virus from DNA infections in immunocompromised mice. In these early studies, we have demonstrated both primary and secondary infections, expanded tissue tropism, and extensive dysplasia.
Collapse
|
6
|
Jinturkar KA, Rathi MN, Misra A. Gene Delivery Using Physical Methods. CHALLENGES IN DELIVERY OF THERAPEUTIC GENOMICS AND PROTEOMICS 2011:83-126. [DOI: 10.1016/b978-0-12-384964-9.00003-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
7
|
Lactobacillus acidophilus as a live vehicle for oral immunization against chicken anemia virus. Appl Microbiol Biotechnol 2010; 90:77-88. [DOI: 10.1007/s00253-010-3050-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 11/29/2010] [Accepted: 11/30/2010] [Indexed: 10/18/2022]
|
8
|
Moeini H, Omar AR, Rahim RA, Yusoff K. Development of a DNA vaccine against chicken anemia virus by using a bicistronic vector expressing VP1 and VP2 proteins of CAV. Comp Immunol Microbiol Infect Dis 2010; 34:227-36. [PMID: 21146874 DOI: 10.1016/j.cimid.2010.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Accepted: 11/15/2010] [Indexed: 10/18/2022]
Abstract
In the present study, we describe the development of a DNA vaccine against chicken anemia virus. The VP1 and VP2 genes of CAV were amplified and cloned into pBudCE4.1 to construct two DNA vaccines, namely, pBudVP1 and pBudVP2-VP1. In vitro and in vivo studies showed that co-expression of VP1 with VP2 are required to induce significant levels of antibody against CAV. Subsequently, the vaccines were tested in 2-week-old SPF chickens. Chickens immunized with the DNA-plasmid pBudVP2-VP1 showed positive neutralizing antibody titer against CAV. Furthermore, VP1-specific proliferation induction of splenocytes and also high serum levels of Th1 cytokines, IL-2 and IFN-γ were detected in the pBudVP2-VP1-vaccinated chickens. These results suggest that the recombinant DNA plasmid co-expressing VP1 and VP2 can be used as a potential DNA vaccine against CAV.
Collapse
Affiliation(s)
- Hassan Moeini
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | | | | | | |
Collapse
|
9
|
Mucosally delivered peptides prime strong immunity in HLA-A2.1 transgenic rabbits. Vaccine 2010; 28:3706-13. [PMID: 20332046 DOI: 10.1016/j.vaccine.2010.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 03/05/2010] [Accepted: 03/09/2010] [Indexed: 12/22/2022]
Abstract
DNA vaccines delivered subcutaneously by gene-gun have generated strong protective and therapeutic immunity in rabbits. Recent studies have shown that peptides delivered by the mucosal routes also stimulate local and systemic immune responses. Since mucosal delivery is easier to administer and more cost-effective when compared to gene-gun delivery, we were interested to learn whether mucosally delivered peptides would prime protective immunity comparable to that of gene-gun-delivered DNA in rabbits. Our newly developed HLA-A2.1 transgenic rabbit model was used to test the hypothesis. We chose an HLA-A2.1 restricted cottontail rabbit papillomavirus (CRPV) E1 epitope (E1/303-311, MLQEKPFQL) for the peptide immunization studies because it provided complete protection when used as a DNA vaccine. Adjuvant has been widely used to boost immunity for vaccines. In this study, three adjuvants reported to be effective for rabbits (TT helper motif, PADRE and CpG2007) were tested with the peptide vaccine. Peptide alone or fused to TT helper or PADRE to create chimeric peptides was delivered by two mucosal routes (ocular and intranasal) together. Partial protection was found in HLA-A2.1 transgenic rabbits when peptide was delivered mucosally in the presence of adjuvant. When a subsequent booster of a half-dose of the corresponding DNA vaccine was delivered, complete protections were achieved. We conclude that mucosal peptide immunization can be combined with a single DNA vaccination to provide strong protective immunity in rabbits.
Collapse
|
10
|
Hu J, Schell TD, Peng X, Cladel NM, Balogh KK, Christensen ND. Using HLA-A2.1 Transgenic Rabbit Model to Screen and Characterize New HLA-A2.1 Restricted Epitope DNA Vaccines. ACTA ACUST UNITED AC 2010; 1. [PMID: 21572916 DOI: 10.4172/2157-7560.1000101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have established an HLA-A2.1 transgenic rabbit /cottontail rabbit papillomavirus (CRPV) infection model. Using this novel transgenic animal model, we reported earlier that a multivalent epitope DNA vaccine (CRPVE1ep1-5) containing five HLA-A2.1 restricted epitopes from CRPVE1 (42-50, 149-157, 161-169, 245-253 and 303-311) was successful in providing strong and specific protective and therapeutic immunity. Among these five epitopes, two (161-169 and 303-311) have been proven to stimulate strong immunity in both HLA-A2.1 transgenic mouse and rabbit models. In the current study, we further identified the remaining three epitopes (CRPVE1/42-50,149-157, 245-253) in both animal models. CRPVE1/149-157 was able to induce specific CTL responses in HLA-A2.1 transgenic mice by DNA immunization but undetectable by peptide immunization. CRPVE1/42-50 and 245-253 failed to respond in HLA-A2.1 transgenic mice either by peptide or DNA immunization. All the three epitopes when administrated as DNA vaccines, however, were able to stimulate strong protective immunity in HLA-A2.1 transgenic rabbits in a dose dependent manner. Among the five epitopes, two (CRPVE1/ 303-311and CRPVE1/149-157) DNA vaccines also showed specific therapeutic effects in CRPV-infected HLA-A2.1 transgenic rabbits. Taken together, the HLA-A2.1 transgenic rabbit model recognized more epitopes than did the HLA-A2.1 transgenic mouse model. Our data demonstrate that the HLA-A2.1 transgenic rabbit model can complement the HLA-A2.1 transgenic mouse model for the development and testing of new HLA-A2.1 restricted prophylactic and therapeutic T cell based DNA vaccines.
Collapse
Affiliation(s)
- Jiafen Hu
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
11
|
Cladel NM, Hu J, Balogh KK, Christensen ND. Differences in methodology, but not differences in viral strain, account for variable experimental outcomes in laboratories utilizing the cottontail rabbit papillomavirus model. J Virol Methods 2009; 165:36-41. [PMID: 20036285 DOI: 10.1016/j.jviromet.2009.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 12/15/2009] [Accepted: 12/17/2009] [Indexed: 11/18/2022]
Abstract
The cottontail rabbit papillomavirus (CRPV) animal model is used in several laboratories worldwide to investigate immunogenicity, carcinogenicity and life cycle aspects of papillomaviruses. It is the only animal model in which the full life cycle of the virus from initiation of infection to malignant progression can be studied. A major strength of the model is that the viral DNA is infectious. This feature allows for the study of mutant genomes without the need to create infectious mutant virus. Results from laboratory to laboratory have not always been consistent. Different laboratories use different methods for creating infections from DNA and it was postulated that the different challenge methods could play a role in the differential outcomes. Because different laboratories use different strains of CRPV, it was also desirable to test if the difference in CRPV genomes contributed to the differential outcomes. In this study, three of the CRPV strains used most widely (Washington B, Orth CRPV and Hershey CRPV) were cloned into PUC19; the E8 ATG ko mutants for each strain were also generated. We employed the infection technique reported previously in which scarification is done first and is followed with delivery of DNA by pipette 3 days later. The papilloma outgrowth generated by these three wild type constructs and their E8 ATG ko mutants was compared. No significant difference was found among the three strains or their E8 ATG ko mutants. E8 ATG ko mutants induced significantly smaller but persistent papillomas when compared to their respective wild type CRPVs. The gene gun was also used to create infections with both Hershey CRPV DNA and the corresponding E8 ATG ko and was found to lead to less vigorous growth as well as some regressions. Further studies suggested that gene gun delivery might have induced an immune response which then resulted in compromised growth of papillomas. It was concluded that the E8 gene is not required for infection. We suggest that standardized infection methods should be used in laboratories so that inconsistencies in conclusions will be minimized.
Collapse
Affiliation(s)
- Nancy M Cladel
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershy, PA 17033, United States
| | | | | | | |
Collapse
|
12
|
Samara RN, Khleif SN. HPV as a model for the development of prophylactic and therapeutic cancer vaccines. Curr Mol Med 2009; 9:766-73. [PMID: 19689303 PMCID: PMC6948838 DOI: 10.2174/156652409788970625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
HPV has been linked to many human malignancies and, as such, represents a major public health crisis. The understanding of HPV biology, however, has helped tremendously in developing prophylactic vaccines, which should help in decreasing mortality due to HPV infections. Understanding HPV biology has allowed researchers to use the virus as a model for the development of not only prophylactic vaccines, but also therapeutic ones. The advantages of HPV as a model stem from the limited number of proteins encoded by the HPV genome that can be targeted by vaccines, and also from the restricted expression of certain viral proteins during different stages of infection. In this review, we discuss how HPV can be used as a model for the development of both prophylactic and therapeutic vaccines.
Collapse
Affiliation(s)
- Raed N. Samara
- Cancer Vaccine Section, National Cancer Institute, NCI-NNMC, Bldg 10, Rm. 5101, Bethesda, MD 20889, USA
| | - Samir N. Khleif
- Cancer Vaccine Section, National Cancer Institute, NCI-NNMC, Bldg 10, Rm. 5101, Bethesda, MD 20889, USA
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to understand the role of the host immune system in clearing the human papillomavirus (HPV) infection, strategies adopted by HPV to subvert the host immune responses and analyze the challenges to the future immunotherapeutic treatment modalities. RECENT FINDINGS Cervical epithelium provides a protective niche to the virus to subvert the immune responses. The absence of an inflammatory milieu in the cervix makes the resident dendritic and langerhan cells tolerogenic to HPV antigens. CD4 cells predominated in regressing cervical intraepithelial neoplasia lesions, whereas CD8 cells were dominant in invasive carcinoma. A reduced expression of T cell signaling molecule T-cell receptor zeta chain was observed in CD8 lymphocytes. Decreased numbers of NKG2D expressing natural killer and T cells were present in patients with cervical cancer and cervical intraepithelial neoplasia. Increased frequencies of CD4 CD25+ FoxP3+ T regulatory cells were observed in patients with cervical cancer. The Nrp-1+Treg showed greater suppressive activity. A network of Treg and indoleamine 2, 3-dioxygenase expressed in tumor cells facilitates immune escape of tumor cells. SUMMARY The HPV uses different strategies to evade immune recognition. Understanding the immune evasion mechanisms used by HPV will help in designing newer therapeutic strategies for cervical cancer.
Collapse
Affiliation(s)
- Swati Patel
- Chiplunkar Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
| | | |
Collapse
|
14
|
Arora A, Prausnitz MR, Mitragotri S. Micro-scale devices for transdermal drug delivery. Int J Pharm 2008; 364:227-36. [PMID: 18805472 PMCID: PMC2752650 DOI: 10.1016/j.ijpharm.2008.08.032] [Citation(s) in RCA: 255] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 08/13/2008] [Accepted: 08/13/2008] [Indexed: 12/17/2022]
Abstract
Skin makes an excellent site for drug and vaccine delivery due to easy accessibility, immuno-surveillance functions, avoidance of macromolecular degradation in the gastrointestinal tract and possibility of self-administration. However, macromolecular drug delivery across the skin is primarily accomplished using hypodermic needles, which have several disadvantages including accidental needle-sticks, pain and needle phobia. These limitations have led to extensive research and development of alternative methods for drug and vaccine delivery across the skin. This review focuses on the recent trends and developments in this field of micro-scale devices for transdermal macromolecular delivery. These include liquid jet injectors, powder injectors, microneedles and thermal microablation. The historical perspective, mechanisms of action, important design parameters, applications and challenges are discussed for each method.
Collapse
Affiliation(s)
- Anubhav Arora
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
| | | | | |
Collapse
|
15
|
Cladel NM, Hu J, Balogh KK, Christensen ND. CRPV genomes with synonymous codon optimizations in the CRPV E7 gene show phenotypic differences in growth and altered immunity upon E7 vaccination. PLoS One 2008; 3:e2947. [PMID: 18698362 PMCID: PMC2491898 DOI: 10.1371/journal.pone.0002947] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 07/08/2008] [Indexed: 01/24/2023] Open
Abstract
Papillomaviruses use rare codons relative to their hosts. Recent studies have demonstrated that synonymous codon changes in viral genes can lead to increased protein production when the codons are matched to those of cells in which the protein is being expressed. We theorized that the immunogenicity of the virus would be enhanced by matching codons of selected viral genes to those of the host. We report here that synonymous codon changes in the E7 oncogene are tolerated in the context of the cottontail rabbit papillomavirus (CRPV) genome. Papilloma growth rates differ depending upon the changes made indicating that synonymous codons are not necessarily neutral. Immunization with wild type E7 DNA yielded significant protection from subsequent challenge by both wild type and codon-modified genomes. The reduction in growth was most dramatic with the genome containing the greatest number of synonymous codon changes.
Collapse
Affiliation(s)
- Nancy M Cladel
- Jake Gittlen Cancer Research Foundation, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America.
| | | | | | | |
Collapse
|
16
|
Anti-tumor effect of DNA-based vaccination and dSLIM immunomodulatory molecules in mice with Ph+ acute lymphoblastic leukaemia. Vaccine 2008; 26:4669-75. [PMID: 18639600 DOI: 10.1016/j.vaccine.2008.06.094] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2008] [Revised: 06/27/2008] [Accepted: 06/29/2008] [Indexed: 11/22/2022]
Abstract
Since the prognosis of patients with Philadelphia chromosome positive acute lymphoblastic leukaemia (Ph+ ALL) still remains poor, new relapse prevention strategies are needed. We evaluated the pre-immunization of mice with DNA-based vaccines subsequently challenged by the syngeneic Ph+ ALL cell line BM185. Ballistic transfer of minimalistic immunogenically defined gene expression (MIDGE) vectors encoding a BCR-ABLp185 fusion specific peptide or GM-CSF were used for in vivo transfection. DNA-based double stem-loop immunomodulators (dSLIM) were used as immune adjuvant. We present survival and functional data that DNA-based vaccination with BCR-ABLp185 fusion specific sequences, GM-CSF and dSLIM leads to an anti-tumor effect in mice challenged with a lethal Ph+ ALL dose and this effect depends on leukaemia-specific sequences.
Collapse
|
17
|
|
18
|
Badaracco G, Venuti A. Human papillomavirus therapeutic vaccines in head and neck tumors. Expert Rev Anticancer Ther 2007; 7:753-66. [PMID: 17492938 DOI: 10.1586/14737140.7.5.753] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Head and neck cancer represents one of the most challenging diseases as the mortality remains high despite advances in early diagnosis and treatment. Human papillomavirus has been implicated in a third of head and neck squamous cell carcinomas and human papillomavirus type 16 is strongly associated with carcinomas arising from the oropharynx, the tonsil being the preferred infected site. Novel therapeutic approaches including immunotherapy are currently under investigation. Immune vaccines developed against human papillomavirus in the genital area are already available and could simultaneously protect other anatomical localizations; however, prophylactic vaccines are expected to be effective in reducing the incidence of tumors after many years and, therefore, there is an urgent need to improve therapeutic interventions, such as immunotherapy. To date, human papillomavirus therapeutic vaccines are either at the preclinical level or at early phase human trials for genital pathologies. Nevertheless, accumulating evidence from animal and clinical studies suggests that the enhancement of specific and innate immune responses is effective in clearance of the human papillomavirus infection, promoting a cautious optimism regarding the achievement of an efficacious immunotherapy. This article reviews what has been achieved and what remains to be done in the field for the development of future viral vaccines in head and neck tumors.
Collapse
Affiliation(s)
- Gianna Badaracco
- Regina Elena Cancer Institute, Laboratory of Virology, Rome, Italy.
| | | |
Collapse
|
19
|
Brandsma JL, Shlyankevich M, Buonocore L, Roberts A, Becker SM, Rose JK. Therapeutic efficacy of vesicular stomatitis virus-based E6 vaccination in rabbits. Vaccine 2006; 25:751-62. [PMID: 16962690 DOI: 10.1016/j.vaccine.2006.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Revised: 08/04/2006] [Accepted: 08/09/2006] [Indexed: 10/24/2022]
Abstract
Millions of people worldwide are currently infected with human papillomaviruses (HPVs). A therapeutic HPV vaccine would have widespread applicability because HPV-associated lesions are difficult to treat and may progress to carcinoma. We developed three attenuated VSV recombinants expressing the cottontail rabbit papillomavirus (CRPV) early protein E6 for use as vaccines. In cultured cells, two vectors expressed different levels of the E6 protein, and one expressed a ubiquitin-E6 fusion protein. All three were tested for therapeutic efficacy in the cottontail rabbit papillomavirus (CRPV)-rabbit model. Mock vaccination had no effect on papilloma growth. In contrast, inoculation with any of the VSV-E6 vaccines reduced the rate of papilloma growth to as little as 24% the rate in the controls. In five experiments, these effects were achieved after a single immunization. Furthermore, complete papilloma regression occurred in some rabbits observed for 4 months. A VSV-based papillomavirus E6 vaccine could have significant advantages over other therapeutic HPV vaccine candidates described to date.
Collapse
Affiliation(s)
- Janet L Brandsma
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520-8016, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Christensen ND. Cottontail rabbit papillomavirus (CRPV) model system to test antiviral and immunotherapeutic strategies. Antivir Chem Chemother 2006; 16:355-62. [PMID: 16331841 DOI: 10.1177/095632020501600602] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The cottontail rabbit papillomavirus (CRPV)/rabbit model has proven to be the most versatile preclinical model to test antiviral, immunopotentiating and immunotherapeutic strategies for papilloma-virus infections. We have utilized this model for many years and have recently observed significant improvements in the utility of the model. Improvements have included various techniques to infect rabbit skin sites with strains of wild-type and mutant CRPV DNA prepared using standard molecular biological procedures. A better understanding of the virus life cycle in vivo has been gained also from these studies such that we now have several defined strains of CRPV including i) antigenically diverse strains of CRPV, ii) mutant strains of CRPV with reduced growth rates, and iii) mutant strains of CRPV that demonstrate accelerated malignant progression rates. Collectively, these mutant genomes provide laboratories with a powerful pre-clinical model to assess a variety of antiviral therapies. Many of the treatments already tested in the CRPV/rabbit model have shown parallel efficacy against HPV infections in a clinical setting. Some of our recent experiences with the CRPV/rabbit model are outlined in this brief overview.
Collapse
Affiliation(s)
- Neil D Christensen
- The Jake Gittlen Cancer Foundation and Department of Microbiology and Immunology, Pennsylvania State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
21
|
Qian J, Dong Y, Pang YYS, Ibrahim R, Berzofsky JA, Schiller JT, Khleif SN. Combined prophylactic and therapeutic cancer vaccine: Enhancing CTL responses to HPV16 E2 using a chimeric VLP in HLA-A2 mice. Int J Cancer 2006; 118:3022-9. [PMID: 16425257 DOI: 10.1002/ijc.21781] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We identified the strategies to induce a CTL response to human papillomavirus (HPV) 16 E2 in HLA-A2 transgenic mice (AAD). A chimeric HPV16 virus-like particle (VLP) that includes full length HPV16 E7 and E2 (VLP-E7E2) was generated. The combination of E2 and E7 has the advantage that E2 is expressed in early dysplasia and neoplasia lesions, where E7 is expressed in more advance lesions. Since T cell response to E2 is less defined, we first evaluated the strategies to enhancing CD8(+) T cell responses to HPV E7, using different combinations of immune-modulators with VLP-E7E2. Data showed that the CTL response to E7 could be significantly enhanced by coinjection of GM-CSF and anti-CD40 antibodies with chimeric VLP-E7E2 without adjuvant. However, using the same combination, a low level of CD8(+) T cell response to E2 was detected. To enhance the CD8+ T cell response to E2, we analyzed T cell epitopes from E2 sequence. A heterogenous prime-boost with chimeric VLP-E7E2 and E2 peptides was performed. The data showed that the priming with chimeric VLP-E7E2, followed by boosting with E2 peptides, gave a better CTL response than 2 immunizations with E2 peptides. The enhanced immunity is due to the increase of CD11c(+) and CD11c(+) CD40(+) double positive dendritic cells in mice that received immune-modulators, GM-CSF and anti-CD40. Furthermore, the level of anti-L1 antibodies remains similar in mice immunized with chimeric VLP with/without immune-modulators. Thus, the data suggested that the chimeric VLP-E7E2 has a therapeutic potential for the treatment of HPV-associated CINs and cancer without diminishing VLPs potential as a prophylactic vaccine by inducing anti-L1 antibodies against free virus.
Collapse
Affiliation(s)
- Jiahua Qian
- Vaccine Branch, NCI, National Naval Medical Center, Bldg 8, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Vambutas A, DeVoti J, Nouri M, Drijfhout JW, Lipford GB, Bonagura VR, van der Burg SH, Melief CJM. Therapeutic vaccination with papillomavirus E6 and E7 long peptides results in the control of both established virus-induced lesions and latently infected sites in a pre-clinical cottontail rabbit papillomavirus model. Vaccine 2005; 23:5271-80. [PMID: 16054734 DOI: 10.1016/j.vaccine.2005.04.049] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2004] [Revised: 04/27/2005] [Accepted: 04/29/2005] [Indexed: 10/25/2022]
Abstract
This study was performed to test the therapeutic efficacy of overlapping long E6 and E7 peptides, containing both CD4+ T-helper and CD8+ CTL epitopes, on CRPV-induced lesions, which is an appropriate pre-clinical model for HPV diseases, including recurrent respiratory papillomatosis (RRP). Therapeutic peptide vaccination was able to significantly control wart growth (p < 0.01) and abrogate latent CRPV infection (p = 0.0006) compared to controls. Vaccination was associated with a T(H)1 T cell response, as suggested by a strong DTH skin test, antigen-specific proliferation of PBMC and a minimal IgG antibody response. Thus, this study shows promise for treatment of RRP by vaccination with long peptides.
Collapse
Affiliation(s)
- A Vambutas
- North Shore-Long Island Jewish Research Institute, Long Island Jewish Medical Center, New Hyde Park, NY 11040, USA.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Globally, carcinomas of the anogenital tract, in particular cervical cancer, remain some of the most common cancers in women, cervical cancer represents the second most frequent gynecological malignancy and the third leading cause of cancer-related death in women worldwide. The causal relationship between human papilomavirus (HPV) infection and anogenital cancer has prompted substantial interest in the development of both preventive and therapeutic vaccines against high-risk HPV types. In the past decade, several groups have shown encouraging results using experimental vaccination systems in animal models and these results have led to several current prophylactic and therapeutic vaccine clinical trials in humans. Prophylactic vaccination focuses on the induction of high titer neutralizing antibodies that are potentially protective against incident and persistent HPV infection. Two major phase II clinical trials conducted by pharmaceutical companies have demonstrated that their vaccines have 100% efficacy in preventing persistent viral DNA and its associated cellular abnormalities; however, whether they induce long-lasting protective immunity is yet to be determined. At least one US FDA approved prophylactic vaccine targeting the two most common high-risk HPVs is expected to be on the market within the next 2-3 years. Nevertheless, significant reductions in the frequency and onset of cytologic screening and incidences of HPV-related lesions are not expected to become apparent for decades due to the fact that there will be women who are already infected with HPV, the long latency period between infection and development of high-grade lesions, and lesions associated with other high-risk HPV types not being included in the vaccines. Therapeutic vaccines aim to control HPV-associated malignancies by stimulating cellular immune responses that target established HPV infections via viral proteins. Progress in the field of HPV immunotherapy has remained elusive, with clinical trials being limited to small numbers of patients. Potential treatment of precancerous lesions is unique to HPV-associated infection and cancer because of cytologic monitoring and HPV typing. Unlike more common surgical treatments for cervical lesions, active immunotherapy has the potential to address HPV persistence as the cause of lesion development in addition to leaving the patient with long-term immunity that can be reactivated if and when the patient becomes reinfected.
Collapse
Affiliation(s)
- Diane M Da Silva
- Norris Comprehensive Cancer Center and Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California, USA
| | - W Martin Kast
- Norris Comprehensive Cancer Center and Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
24
|
|
25
|
Abstract
A subset of human papillomaviruses (HPVs) promote anogenital malignancy, including cervical cancer, and prevention and treatment strategies that reflect the causal role of HPV are being developed. Vaccines based on HPV virus-like particles induce genotype-specific virus-neutralizing antibody and prevent infection with HPV. Persistent papillomavirus infection is required for the development of papillomavirus-associated cancer and, therefore, therapeutic vaccines are being developed to eliminate established papillomavirus infection. Such vaccines test principles for the growing field of tumour-antigen-specific immunotherapy. This article reviews progress in the field and draws conclusions for the development of future prophylactic and therapeutic viral vaccines.
Collapse
Affiliation(s)
- Ian H Frazer
- Centre for Immunology and Cancer Research, The University of Queensland, Princess Alexandra Hospital, Ipswich Road, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
26
|
Peh WL, Brandsma JL, Christensen ND, Cladel NM, Wu X, Doorbar J. The viral E4 protein is required for the completion of the cottontail rabbit papillomavirus productive cycle in vivo. J Virol 2004; 78:2142-51. [PMID: 14747580 PMCID: PMC369506 DOI: 10.1128/jvi.78.4.2142-2151.2004] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Expression of the papillomavirus E4 protein correlates with the onset of viral DNA amplification. Using a mutant cottontail rabbit papillomavirus (CRPV) genome incapable of expressing the viral E4 protein, we have shown that E4 is required for the productive stage of the CRPV life cycle in New Zealand White and cottontail rabbits. In these lesions, E4 was not required for papilloma development, but the onset of viral DNA amplification and L1 expression were abolished. Viral genome amplification was partially restored when mutant genomes able to express longer forms of E4 were used. These findings suggest that efficient amplification of the CRPV genome is dependent on the expression of a full-length CRPV E4 protein.
Collapse
Affiliation(s)
- Woei L Peh
- Division of Virology, The National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom
| | | | | | | | | | | |
Collapse
|
27
|
Hu J, Cladel NM, Wang Z, Han R, Pickel MD, Christensen ND. GM-CSF enhances protective immunity to cottontail rabbit papillomavirus E8 genetic vaccination in rabbits. Vaccine 2004; 22:1124-30. [PMID: 15003639 DOI: 10.1016/j.vaccine.2003.09.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2003] [Revised: 08/06/2003] [Accepted: 09/29/2003] [Indexed: 11/20/2022]
Abstract
We have reported previously that cottontail rabbit papillomavirus (CRPV) E8 gene immunization induced strong protection against virus challenge. In this study, we primed E8 gene vaccination with mouse granulocyte-macrophage colony-stimulating factor (mGM-CSF), a cytokine that induces differentiation and local recruitment of professional antigen-presenting cells. EIII/JC inbred rabbits were divided into four groups receiving vaccinations with the following constructs: mGM-CSF plus E8, mGM-CSF only, E8 only and vector only. After three immunizations at intervals of 3 weeks, rabbits were challenged with viral DNA at six scarified sites. Papillomas grew on all vaccinated rabbits 4 weeks after inoculation. At week 5, papillomas on four rabbits of mGM-CSF plus E8 and one of E8 only rabbits began to regress. At week 11, all the papillomas on rabbits in the GM-CSF plus E8 vaccination group regressed (regression rate = 100%); regression rates of the mGM-CSF only and E8 only vaccination groups were 50 and 25%, respectively. All papillomas on the vector immunized rabbits remained persistent until the end of the experiment (0%). Antibodies to mGM-CSF were detected in rabbit serum by Western blot. Rabbits vaccinated with E8 plus mGM-CSF or E8 only group had positive Delayed-type hypersensitivity (DTH) skin test to different E8 peptides. These results demonstrated that mGM-CSF could enhance the effects of E8 immunization in rabbits to CRPV infection through cell-mediated immune responses.
Collapse
Affiliation(s)
- Jiafen Hu
- Department of Pathology, The Jake Gittlen Cancer Research Institute, Pennsylvania State University, Hershey, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
28
|
Tian G, Yi JL. Gene cloning of murine α-fetoprotein gene and construction of its eukaryotic expression vector and expression in CHO cells. Shijie Huaren Xiaohua Zazhi 2003; 11:1674-1676. [DOI: 10.11569/wcjd.v11.i11.1674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To clone murine AFP gene, to construct the eukaryotic expression vector of AFP and express it in CHO cells.
METHODS Total RNA was extracted from Hepa1-6 cells, then the murine AFP gene was amplified by RT-PCR and cloned into the eukaryotic expression vector pcDNA3.1. The recombinant vector was transformed into E.coli. DH5α, the positive clones were selected and plasmid DNA was identified by restriction enzyme analysis and sequencing. After transient transfection of CHO-K1 cells with the recombinant vector, Western blotting was used to detect the expression of AFP.
RESULTS The 1.8 kb murine α-fetoprotein gene was successfully cloned from the total RNA of Hepa1-6 cells. Result from restrictiive enzyme analysis and sequencing showed that the murine α-fetoprotein gene was successfully inserted into pcDNA3.1. Result from Western blotting showed that the recombinant vector could express murine α-fetoprotein in CHO-K1 cells.
CONCLUSION The successfully constructed eukaryotic expression vector of murine α-fetoprotein could provide a basis for the research of immunotherapy for hepatocellular carcinoma with pmAFP.
Collapse
Affiliation(s)
- Geng Tian
- Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Ji-Lin Yi
- Department of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
29
|
Davidson EJ, Sehr P, Faulkner RL, Parish JL, Gaston K, Moore RA, Pawlita M, Kitchener HC, Stern PL. Human papillomavirus type 16 E2- and L1-specific serological and T-cell responses in women with vulval intraepithelial neoplasia. J Gen Virol 2003; 84:2089-2097. [PMID: 12867639 DOI: 10.1099/vir.0.19095-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human papillomavirus type 16 (HPV-16)-associated vulval intraepithelial neoplasia (VIN) is frequently a chronic, multifocal high-grade condition with an appreciable risk of progression to vulval cancer. The requirement to treat women with VIN has recently stimulated the use of immunotherapy with E6/E7 oncogene vaccines. Animal models have shown that E2 may also be a useful vaccine target for HPV-associated disease; however, little is known about E2 immunity in humans. This study investigated the prevalence of HPV-16 E2-specific serological and T-cell responses in 18 women with HPV-16-associated VIN and 17 healthy volunteers. E2 responses were determined by full-length E2-GST ELISA with ELISPOT and proliferation assays using E2 C-terminal protein. As positive controls, HPV-16 L1 responses were measured using virus-like particles (VLPs) and L1-GST ELISA with ELISPOT and proliferation using VLPs as antigen. The VIN patients all showed a strong serological response to L1 compared with the healthy volunteers by VLP (15/18 vs 1/17, P<0.001) and L1-GST ELISA (18/18 vs 1/17, P<0.001). In contrast, L1-specific cellular immune responses were detected in a significant proportion of controls but were more prevalent in the VIN patients by proliferation assay (9/17 vs 17/18, P<0.02) and interferon-gamma ELISPOT (9/17 vs 13/18, P=not significant). Similar and low numbers of patients and controls were seropositive for E2-specific Ig (2/18 vs 1/17). In spite of previous studies showing the immunogenicity of E2 in eliciting primary T-cell responses in vitro, there was a low prevalence of E2 responses in the VIN patients and controls (2/18 vs 0/17).
Collapse
Affiliation(s)
- Emma J Davidson
- Academic Unit of Obstetrics & Gynaecology, St Mary's Hospital, Whitworth Park, Manchester M13 0JH, UK
- Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Peter Sehr
- Applied Tumorvirology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Rebecca L Faulkner
- Academic Unit of Obstetrics & Gynaecology, St Mary's Hospital, Whitworth Park, Manchester M13 0JH, UK
- Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Joanna L Parish
- Department of Biochemistry, University of Bristol, Bristol, UK
| | - Kevin Gaston
- Department of Biochemistry, University of Bristol, Bristol, UK
| | - Richard A Moore
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Michael Pawlita
- Applied Tumorvirology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Henry C Kitchener
- Academic Unit of Obstetrics & Gynaecology, St Mary's Hospital, Whitworth Park, Manchester M13 0JH, UK
| | - Peter L Stern
- Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
30
|
Han R, Peng X, Reed CA, Cladel NM, Budgeon LR, Pickel MD, Christensen ND. Gene gun-mediated intracutaneous vaccination with papillomavirus E7 gene delays cancer development of papillomavirus-induced skin papillomas on rabbits. CANCER DETECTION AND PREVENTION 2003; 26:458-67. [PMID: 12507231 DOI: 10.1016/s0361-090x(02)00125-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
High-risk human papillomavirus (HPV) E6 and E7 viral oncogenes are expressed in HPV-associated cancers, and thus represent tumor-specific antigens. We used the cottontail rabbit papillomavirus (CRPV) rabbit model to test whether vaccination with either the E6 or E7 genes alone could prevent or delay carcinoma development. CRPV-induced papillomas on 24 rabbits were allowed to grow for 3 months without any treatment intervention. An immunization protocol using gene gun-mediated intracutaneous administration of DNA plasmids encoding the E6 or the E7 gene or vector only, respectively was initiated at this time point. Carcinoma development was followed up to 24 months after virus infection. Within this period, five rabbits died due to other causes but without carcinoma; one from the vector control group, and two each from the E6- and E7-vaccinated groups. The remaining seven rabbits from the vector control group developed carcinoma within 7-17 months. The remaining six E6-vaccinated rabbits developed cancer within 8-15 months. There was no delay in cancer development for the E6-vaccinated rabbits compared to the vector-injected rabbits. Some delay in cancer development in the remaining E7-vaccinated rabbits was observed; one developed cancer at month 23 and a second was without cancer at month 24. In addition, some E7-vaccinated rabbits with primary skin carcinomas had fewer lung metastases (<2) compared to vector-vaccinated controls (20+). These results suggested that gene gun-mediated intracutaneous immunization with papillomavirus early gene E7 but not E6 delayed carcinoma development of papillomavirus-induced lesions.
Collapse
Affiliation(s)
- Ricai Han
- Department of Pathology, Jake Gittlen Cancer Research Institute, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Stanley MA. Progress in prophylactic and therapeutic vaccines for human papillomavirus infection. Expert Rev Vaccines 2003; 2:381-9. [PMID: 12903803 DOI: 10.1586/14760584.2.3.381] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Virus-like particle (VLP) subunit vaccines composed of the major capsid protein L1 of the genital human papillomaviruses (HPVs) are now in Phase III clinical trials. The vaccines are immunogenic and safe and early results indicate efficacy. VLPs induce strong cell-mediated as well as humoral immune responses and chimeric VLPs including an HPV early protein may have therapeutic potential. Polynucleotide and recombinant viral vaccines encoding nonstructural viral proteins show therapeutic and prophylactic efficacy in animal models and are candidate immunotherapies for established low-grade benign genital infections. Vaccines designed to elicit cytotoxic T-lymphocytes specific for the HPV oncoproteins E6 and E7 show immunogenicity and efficacy in transplantable tumor models in rodents. In Phase I and II trials these vaccines are immunogenic and safe but show limited efficacy.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/immunology
- Antigens, Neoplasm/immunology
- Antigens, Viral/immunology
- Carcinoma, Squamous Cell/prevention & control
- Carcinoma, Squamous Cell/virology
- Clinical Trials, Phase III as Topic
- Condylomata Acuminata/prevention & control
- Condylomata Acuminata/virology
- Disease Models, Animal
- Dog Diseases/immunology
- Dog Diseases/prevention & control
- Dog Diseases/virology
- Dogs
- Female
- Genital Neoplasms, Female/prevention & control
- Genital Neoplasms, Female/virology
- Genital Neoplasms, Male/prevention & control
- Genital Neoplasms, Male/virology
- Humans
- Immunotherapy, Active
- Male
- Middle Aged
- Neoplasms/prevention & control
- Neoplasms/virology
- Oncogene Proteins, Viral/immunology
- Papillomaviridae/genetics
- Papillomaviridae/immunology
- Papillomavirus Infections/prevention & control
- Papillomavirus Infections/therapy
- Papillomavirus Infections/veterinary
- Papillomavirus Vaccines
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Virus Infections/prevention & control
- Tumor Virus Infections/therapy
- Tumor Virus Infections/veterinary
- Uterine Cervical Neoplasms/prevention & control
- Uterine Cervical Neoplasms/virology
- Viral Vaccines/immunology
- Viral Vaccines/therapeutic use
- Uterine Cervical Dysplasia/prevention & control
- Uterine Cervical Dysplasia/virology
Collapse
|
32
|
Abstract
Recent advances in the detection and therapy of carcinoma of the cervix and its squamous intra-epithelial precursor lesions exploit the knowledge that these lesions are a consequence of infection with high risk (HR) human papillomavirus (HPV). HPV infections over-ride cell cycle controls and antibody based immunodetection of proteins that regulate DNA replication may facilitate mass automated cervical smear screening. Detection of HR HPV DNA in smears from selected patient groups will improve detection of high grade precursor lesions and immunodetection of the cell cycle dependent kinase inhibitor p16(INK4a) seems to specifically and sensitively identify HGSIL. Immunisation with HPV early proteins has been shown to have both prophylactic and therapeutic efficacy in animal papillomavirus infections and immunotherapies for low grade intra-epithelial lesions are realistic. Such vaccines are likely to be combined with immunomodulators in order to maximise the response. Immunotherapies for HPV associated high grade pre-cancers and invasive cancers are problematic in view of tumour immune evasion. However, anti-viral chemotherapies may benefit from the neoplastic phenotypic by the induction of: (1) apoptosis as a consequence of small molecule or anti-sense targeting of individual HPV oncoproteins and (2) replicative senescence by down regulation of the early promoter by E2 or small molecule homologues.
Collapse
Affiliation(s)
- Margaret A Stanley
- Department of Pathology, University of Cambridge, Tennis Court Road, UK.
| |
Collapse
|
33
|
Peh WL, Middleton K, Christensen N, Nicholls P, Egawa K, Sotlar K, Brandsma J, Percival A, Lewis J, Liu WJ, Doorbar J. Life cycle heterogeneity in animal models of human papillomavirus-associated disease. J Virol 2002; 76:10401-16. [PMID: 12239317 PMCID: PMC136551 DOI: 10.1128/jvi.76.20.10401-10416.2002] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Animal papillomaviruses are widely used as models to study papillomavirus infection in humans despite differences in genome organization and tissue tropism. Here, we have investigated the extent to which animal models of papillomavirus infection resemble human disease by comparing the life cycles of 10 different papillomavirus types. Three phases in the life cycles of all viruses were apparent using antibodies that distinguish between early events, the onset of viral genome amplification, and the expression of capsid proteins. The initiation of these phases follows a highly ordered pattern that appears important for the production of virus particles. The viruses examined included canine oral papillomavirus, rabbit oral papillomavirus (ROPV), cottontail rabbit papillomavirus (CRPV), bovine papillomavirus type 1, and human papillomavirus types 1, 2, 11, and 16. Each papillomavirus type showed a distinctive gene expression pattern that could be explained in part by differences in tissue tropism, transmission route, and persistence. As the timing of life cycle events affects the accessibility of viral antigens to the immune system, the ideal model system should resemble human mucosal infection if vaccine design is to be effective. Of the model systems examined here, only ROPV had a tissue tropism and a life cycle organization that resembled those of the human mucosal types. ROPV appears most appropriate for studies of the life cycles of mucosal papillomavirus types and for the development of prophylactic vaccines. The persistence of abortive infections caused by CRPV offers advantages for the development of therapeutic vaccines.
Collapse
Affiliation(s)
- Woei Ling Peh
- National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hu J, Han R, Cladel NM, Pickel MD, Christensen ND. Intracutaneous DNA vaccination with the E8 gene of cottontail rabbit papillomavirus induces protective immunity against virus challenge in rabbits. J Virol 2002; 76:6453-9. [PMID: 12050357 PMCID: PMC136259 DOI: 10.1128/jvi.76.13.6453-6459.2002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cottontail rabbit papillomavirus (CRPV)-rabbit model has been used in several studies for testing prophylactic and therapeutic papillomavirus vaccines. Earlier observations had shown that the CRPV nonstructural genes E1, E2, and E6 induced strong to partial protective immunity against CRPV infection. In this study, we found that CRPV E8 immunization eliminated virus-induced papillomas in EIII/JC inbred rabbits (100%) and provided partial protection (55%) against virus challenge in outbred New Zealand White rabbits. CRPV-E8 is a small open reading frame, coding for a 50-amino-acid protein, that is colinear with the CRPV E6 gene and has features similar to those of the bovine papillomavirus and human papillomavirus E5 genes. Papillomas that grew on E8-vaccinated outbred rabbits were significantly smaller than those on vector-vaccinated rabbits (P < 0.01; t test). Delayed-type hypersensitivity skin tests showed that some of the E8-vaccinated rabbits had positive responses to E8-specific peptides.
Collapse
Affiliation(s)
- Jiafen Hu
- Department of Pathology, The Jake Gittlen Cancer Research Institute, College of Medicine, Pennsylvania State University, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
35
|
Ren S, Li M, Smith JM, DeTolla LJ, Furth PA. Low-volume jet injection for intradermal immunization in rabbits. BMC Biotechnol 2002; 2:10. [PMID: 12028591 PMCID: PMC115865 DOI: 10.1186/1472-6750-2-10] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2001] [Accepted: 05/23/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study tested a low-volume (20-30 microl/20-30 microg DNA) jet injection method for intradermal delivery of a DNA vaccine. Jet injection offers the advantages of a needle-less system, low-cost, rapid preparation of the injected DNA solution, and a simple delivery system. More than one construct can be injected simultaneously and the method may be combined with adjuvants. RESULTS Low-volume jet injection targeted delivery of a DNA solution exclusively to the dermis and epidermis of rabbits. A three injection series of plasmid DNA, encoding the Hepatitis B Surface Antigen stimulated a humoral immune response in 2/5 rabbits. One rabbit developed a significant rise in antibody titer after 1 injection and one following 2 injections. There were no significant differences between jet injection and particle bombardment in the maximal antibody titers or number of injections before response. A three injection series of the same plasmid DNA by particle bombardment elicited a significant rise in antibody titer in 3/5 rabbits. One rabbit developed antibody after 1 injection and two after 3 injections. In contrast, 0/5 rabbits receiving DNA by needle and syringe injection responded. In the jet injection and particle bombardment groups, gene expression levels in the skin did not predict response. While immune responses were similar, luciferase gene expression levels in the skin following particle bombardment were 10-100 times higher than jet injection. CONCLUSION Low-volume jet injection is a simple, effective methodology for intradermal DNA immunization.
Collapse
Affiliation(s)
- Shuxun Ren
- Department of Oncology, Lombardi Cancer Center, Georgetown University, 3970 Reservoir Rd, NW, Washington, DC 20007, USA
- Department of Medicine and Institute of Human Virology, University of Maryland Medical School, Baltimore, MD, 21201, USA
| | - Minglin Li
- Department of Oncology, Lombardi Cancer Center, Georgetown University, 3970 Reservoir Rd, NW, Washington, DC 20007, USA
- Department of Medicine and Institute of Human Virology, University of Maryland Medical School, Baltimore, MD, 21201, USA
| | - Joanne M Smith
- Comparative Medicine Program and Department of Pathology, University of Maryland Medical School, Baltimore, MD, 21201, USA
| | - Louis J DeTolla
- Comparative Medicine Program and Department of Pathology, University of Maryland Medical School, Baltimore, MD, 21201, USA
| | - Priscilla A Furth
- Department of Oncology, Lombardi Cancer Center, Georgetown University, 3970 Reservoir Rd, NW, Washington, DC 20007, USA
- Department of Medicine and Institute of Human Virology, University of Maryland Medical School, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland Medical School, Baltimore, MD, 21201, USA
| |
Collapse
|
36
|
Haupt K, Roggendorf M, Mann K. The potential of DNA vaccination against tumor-associated antigens for antitumor therapy. Exp Biol Med (Maywood) 2002; 227:227-37. [PMID: 11910045 DOI: 10.1177/153537020222700403] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Conventional treatment approaches for malignant tumors are highly invasive and sometimes have only a palliative effect. Therefore, there is an increasing demand to develop novel, more efficient treatment options. Increased efforts have been made to apply immunomodulatory strategies in antitumor treatment. In recent years, immunizations with naked plasmid DNA encoding tumor-associated antigens have revealed a number of advantages. By DNA vaccination, antigen-specific cellular as well as humoral immune responses can be generated. The induction of specific immune responses directed against antigens expressed in tumor cells and displayed e.g., by MHC class I complexes can inhibit tumor growth and lead to tumor rejection. The improvement of vaccine efficacy has become a critical goal in the development of DNA vaccination as antitumor therapy. The use of different DNA delivery techniques and coadministration of adjuvants including cytokine genes may influence the pattern of specific immune responses induced. This brief review describes recent developments to optimize DNA vaccination against tumor-associated antigens. The prerequisite for a successful antitumor vaccination is breaking tolerance to tumor-associated antigens, which represent "self-antigens." Currently, immunization with xenogeneic DNA to induce immune responses against self-molecules is under intensive investigation. Tumor cells can develop immune escape mechanisms by generation of antigen loss variants, therefore, it may be necessary that DNA vaccines contain more than one tumor antigen. Polyimmunization with a mixture of tumor-associated antigen genes may have a synergistic effect in tumor treatment. The identification of tumor antigens that may serve as targets for DNA immunization has proceeded rapidly. Preclinical studies in animal models are promising that DNA immunization is a potent strategy for mediating antitumor effects in vivo. Thus, DNA vaccines may offer a novel treatment for tumor patients. DNA vaccines may also be useful in the prevention of tumors with genetic predisposition. By DNA vaccination preventing infections, the development of viral-induced tumors may be avoided.
Collapse
Affiliation(s)
- Katharina Haupt
- Division of Clinical Chemistry, Department of Internal Medicine, University of Essen, 45122 Essen, Germany.
| | | | | |
Collapse
|
37
|
Abstract
Epidemiological and experimental studies have overwhelmingly confirmed human papillomaviruses as important causal agents in anogenital carcinogenesis. A role for human papillomaviruses has also been proposed in a diverse range of other malignancies, and particular interest has focused on non-melanoma skin cancer, the commonest malignancy in fair-skinned populations worldwide. Although the evidence for this is considerably less convincing than for anogenital cancer, important epidemiological and functional data have emerged over the past year that have furthered our understanding of the possible contribution of human papillomaviruses to skin cancer. Epidemiological human papillomavirus DNA detection studies have shown associations with non-melanoma skin cancer, but have also emphasized the ubiquity of epidermodysplasia verruciformis human papillomavirus types in normal skin, hair follicles and benign hyperproliferative disorders, as have seroepidemiological approaches. Functional investigations have demonstrated mechanistically relevant interactions between the virus and ultraviolet radiation, host cytokines and cellular proteins including p53 and the pro-apoptotic protein Bak. Taken together, these data have advanced our understanding of the contribution of human papillomaviruses to malignant transformation in cutaneous keratinocytes, but further research is required before a causal association between human papillomaviruses and skin cancer is reliably confirmed.
Collapse
Affiliation(s)
- Catherine A Harwood
- Imperial Cancer Research Fund Skin Tumour Laboratory, Barts and the London NHS Trust, London, UK.
| | | |
Collapse
|
38
|
Tanaka M, Kaneda Y, Fujii S, Yamano T, Hashimoto K, Huang SKS, Hoon DSB. Induction of a systemic immune response by a polyvalent melanoma-associated antigen DNA vaccine for prevention and treatment of malignant melanoma. Mol Ther 2002; 5:291-9. [PMID: 11863419 DOI: 10.1006/mthe.2002.0537] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Studies have demonstrated that active-specific immunotherapy has potential for controlling melanoma progression. We developed a polyvalent melanoma gene vaccine using a plasmid vector to deliver the immunogenic human melanoma-associated antigens (MAAs) gp100 and TRP-2. The MAA-containing plasmids were delivered individually in vivo using the hemagglutinating virus Japan (HVJ)-anionic liposome delivery system. C57BL/6 mice were immunized weekly by intramuscular (i.m.) injection or intranasal (i.n.) inoculation for 3 weeks. Although both i.m. and i.n. immunization induced Th1 (T helper) and Th2 cell responses to gp100 and TRP2, the i.m. route induced a better Th1 response. MAA-specific IgG2a, IgG1, and delayed-type hypersensitivity (DTH) responses were induced against both MAAs by i.m. immunization. We assessed the vaccine for its prophylactic and therapeutic effect against the murine B16 F10 melanoma. Animals vaccinated and subsequently challenged with a lethal dose of B16 cells were significantly (P<0.01) protected against tumor progression and had significantly (P<0.01) enhanced survival compared with treatment using control plasmid. We also developed a therapeutic model in which mice were given B16 cells and subsequently immunized with the vaccine or treated with control plasmid. In animals treated with the vaccine, tumor growth was significantly (P<0.01) controlled, and survival was prolonged compared with controls. These studies demonstrate that the polyvalent DNA vaccine induces an effective systemic Th response.
Collapse
Affiliation(s)
- Maki Tanaka
- Department of Molecular Oncology, John Wayne Cancer Institute, Santa Monica, California 90404, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Davidson EJ, Brown MD, Burt DJ, Parish JL, Gaston K, Kitchener HC, Stacey SN, Stern PL. Human T cell responses to HPV 16 E2 generated with monocyte-derived dendritic cells. Int J Cancer 2001; 94:807-12. [PMID: 11745482 DOI: 10.1002/ijc.1558] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Persistent infection with human papillomavirus (HPV) type 16 has been implicated in the etiology of cervical cancer. The E2 protein is required early in viral infection and therefore may serve as a useful immune target for a vaccine aimed at prevention or therapy of premalignant lesions. Dendritic cells (DC) prepared from monocytes and pulsed with bacterially produced HPV 16 E2 C-terminus protein were used to stimulate autologous T cells over several rounds of stimulation. T cells were tested for gamma-interferon release by ELISPOT and for cytotoxic activity by (51)chromium release assays. To generate E2-expressing target cells for cytotoxicity assays, we constructed a recombinant vaccinia virus encoding HPV 16 E2, which was used to infect autologous Epstein-Barr virus-transformed lymphoblastoid cell lines (LCL). The results show that DC pulsed with E2 C-terminus protein induce gamma-interferon-releasing T cells as demonstrated by ELISPOT. Furthermore, we demonstrate E2-specific lysis of vaccinia-E2 infected autologous LCL by CD8+ cytotoxic T lymphocytes (CTL). E2-specific CTL did not lyse untreated autologous LCL or LCL infected with wild-type vaccinia and showed low levels of cytotoxicity against natural killer cell-sensitive K562 cells. In addition, T cells stimulated with DC in the absence of E2 failed to demonstrate lysis of vaccinia-E2-labeled targets. Phenotypically, CTL populations were CD3+/CD8+. These results will facilitate the study of naturally occurring T-cell responses to HPV E2 in patients with cervical intraepithelial neoplasia and the development of immunotherapeutic strategies designed to treat this and other HPV-associated diseases.
Collapse
Affiliation(s)
- E J Davidson
- CRC Immunology Group, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Liu WJ, Zhao KN, Gao FG, Leggatt GR, Fernando GJ, Frazer IH. Polynucleotide viral vaccines: codon optimisation and ubiquitin conjugation enhances prophylactic and therapeutic efficacy. Vaccine 2001; 20:862-9. [PMID: 11738751 DOI: 10.1016/s0264-410x(01)00406-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Papillomavirus infection is a major antecedent of anogenital malignancy. We have previously established that the L1 and L2 capsid genes of papillomavirus have suboptimal codon usage for expression in mammalian cells. We now show that the lack of immunogenicity of polynucleotide vaccines based on the L1 gene can be overcome with codon modified L1, which induces strong immune responses, including conformational virus neutralising antibody and delayed type hypersensitivity. Conjugation of a ubiquitin gene to a hybrid gene incorporating L1 and the E7 non-structural papillomavirus protein improved E7 specific CTL responses, and induced protection against an E7 expressing tumour, but induced little neutralising antibody. However, a mixture of ubiquitin conjugated and non-ubiquitin conjugated polynucleotides induced virus neutralising antibody and E7 specific CD8 T cells. An optimal combined prophylactic/therapeutic viral vaccine might therefore comprise ubiquitin conjugated and non-ubiquitinated genes, to induce prophylactic neutralising antibody and therapeutic cell mediated immune responses.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Codon/genetics
- Female
- Genes, Viral
- Humans
- Hypersensitivity, Delayed
- Immunity, Cellular
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neutralization Tests
- Papillomaviridae/genetics
- Papillomaviridae/immunology
- Papillomaviridae/pathogenicity
- Papillomavirus Infections/immunology
- Papillomavirus Infections/prevention & control
- Papillomavirus Infections/therapy
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Virus Infections/immunology
- Tumor Virus Infections/prevention & control
- Tumor Virus Infections/therapy
- Ubiquitin/immunology
- Vaccines, Conjugate/genetics
- Vaccines, Conjugate/pharmacology
- Vaccines, Conjugate/therapeutic use
- Vaccines, DNA/genetics
- Vaccines, DNA/pharmacology
- Vaccines, DNA/therapeutic use
- Viral Vaccines/genetics
- Viral Vaccines/pharmacology
- Viral Vaccines/therapeutic use
Collapse
Affiliation(s)
- W J Liu
- Centre for Immunology and Cancer Research, Princess Alexandra Hospital, University of Queensland, Woolloongabba 4102, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Christensen ND, Han R, Cladel NM, Pickel MD. Combination treatment with intralesional cidofovir and viral-DNA vaccination cures large cottontail rabbit papillomavirus-induced papillomas and reduces recurrences. Antimicrob Agents Chemother 2001; 45:1201-9. [PMID: 11257035 PMCID: PMC90444 DOI: 10.1128/aac.45.4.1201-1209.2001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We used the cottontail rabbit papillomavirus (CRPV) New Zealand White rabbit model to test a combination treatment of large established papillomas with intralesional cidofovir and DNA vaccination to cure sites and reduce recurrences. Intralesional 1% (wt/vol) (0.036 M) cidofovir treatment of rabbit papillomas led to elimination, or "cure," of the papillomas over a 6- to 8-week treatment period (N. D. Christenson, M. D. Pickel, L. R. Budgeon, and J. W. Kreider, Antivir. Res. 48:131-142, 2000). However, recurrences at periods from 1 to 8 weeks after treatment cessation were observed at approximately 50% of cured sites. DNA vaccinations with CRPV E1, E2, E6, and E7 were initiated either after or at the time of intralesional treatments, and the recurrence rates were observed. When DNA vaccinations were started after intralesional cures, recurrence rates were similar to those of vector-vaccinated rabbits. A small proportion of recurrent sites subsequently regressed (4 out of 10, or 40%) in the vaccinated group versus no regression of recurrences in the vector-immunized group (0 out of 19, or 0%), indicating partial effectiveness. In contrast, when DNA vaccinations were conducted during intralesional treatments, a significant reduction of recurrences (from 10 out of 19, or 53%, of sites in vector-immunized rabbits to 3 out of 20, or 15%, of sites in viral-DNA-immunized rabbits) was observed. DNA vaccination without intralesional treatments had a minimal effect on preexisting papillomas. These data indicated that treatment with a combination of antiviral compounds and specific immune stimulation may lead to long-term cures of lesions without the ensuing problem of papilloma recurrence.
Collapse
Affiliation(s)
- N D Christensen
- Pathology, The Jake Gittlen Cancer Research Institute, The Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, USA.
| | | | | | | |
Collapse
|