Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Clin Microbiol Infect ; 25(2): 210-216, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29684633

ABSTRACT

OBJECTIVES: To evaluate the efficacy of a carrageenan-based lubricant gel in reducing the risk of genital human papillomavirus (HPV) infections in women. METHODS: We conducted a planned interim analysis of a randomized, double-blind, placebo-controlled, phase 2B trial. Women aged 18 years and older were randomly assigned (1:1) to a carrageenan-based gel or a placebo gel to be self-applied every other day for the first month and before and after each intercourse during follow-up. Assessments were performed at 0.5, 1, 3, 6, 9 and 12 months. The primary outcome was incidence of a new infection by an HPV type that was not present at baseline. Intention-to-treat analyses were performed. RESULTS: Between January 2013 and June 2017, a total of 280 participants were randomly assigned to the carrageenan (n = 141) or the placebo (n = 139) arm. All participants were included in safety analyses, but three (1%) were excluded from efficacy analyses (HPV results unavailable for two participants in the carrageenan and one participant in the placebo arm). The median follow-up time was 9.2 months (interquartile range, 1.9-13.2 months). A total of 59 (42%) of 139 participants in the carrageenan arm and 78 (57%) of 138 participants in the placebo arm became infected by at least one new HPV type (hazard ratio = 0.64, 95% confidence interval = 0.45-0.89, p 0.009). A total of 62 (44%) of 141 participants in the carrageenan arm versus 43 (31%) of 139 participants in the placebo arm reported an adverse event (p 0.02), none of which was deemed related to the gels. CONCLUSIONS: Our trial's interim analysis suggests that using a carrageenan-based lubricant gel can reduce the risk of genital HPV infections in women.


Subject(s)
Carrageenan , Gels , Papillomaviridae , Papillomavirus Infections/prevention & control , Uterine Cervical Diseases/prevention & control , Uterine Cervical Diseases/virology , Administration, Intravaginal , Adult , Double-Blind Method , Female , Humans
2.
Mucosal Immunol ; 6(2): 393-404, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22968420

ABSTRACT

Human papillomaviruses (HPV)-related cervical cancer is the second leading cause of cancer death in women worldwide. Despite active development, HPV E6/E7 oncogene-specific therapeutic vaccines have had limited clinical efficacy to date. Here, we report that intravaginal (IVAG) instillation of CpG-ODN (TLR9 agonist) or poly-(I:C) (TLR3 agonist) after subcutaneous E7 vaccination increased ~fivefold the number of vaccine-specific interferon-γ-secreting CD8 T cells in the genital mucosa (GM) of mice, without affecting the E7-specific systemic response. The IVAG treatment locally increased both E7-specific and total CD8 T cells, but not CD4 T cells. This previously unreported selective recruitment of CD8 T cells from the periphery by IVAG CpG-ODN or poly-(I:C) was mediated by TLR9 and TLR3/melanoma differentiation-associated gene 5 signaling pathways, respectively. For CpG, this recruitment was associated with a higher proportion of GM-localized CD8 T cells expressing both CCR5 and CXCR3 chemokine receptors and E-selectin ligands. Most interestingly, IVAG CpG-ODN following vaccination led to complete regression of large genital HPV tumors in 75% of mice, instead of 20% with vaccination alone. These findings suggest that mucosal application of immunostimulatory molecules might substantially increase the effectiveness of parenterally administered vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genital Neoplasms, Female/immunology , Genital Neoplasms, Female/metabolism , Papillomaviridae/immunology , Papillomavirus Vaccines/immunology , Toll-Like Receptors/agonists , Animals , CD8-Positive T-Lymphocytes/metabolism , Cervix Uteri/immunology , Cervix Uteri/metabolism , Cervix Uteri/virology , DEAD-box RNA Helicases/metabolism , Female , Genital Neoplasms, Female/mortality , Genital Neoplasms, Female/virology , Humans , Immunization , Interferon-Induced Helicase, IFIH1 , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Poly I-C/administration & dosage , Poly I-C/pharmacology , Receptors, CCR5/metabolism , Receptors, CXCR3/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Sialoglycoproteins/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism , Toll-Like Receptors/metabolism
3.
Mucosal Immunol ; 3(5): 475-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20555315

ABSTRACT

Mucosal immunization may be important for protection against pathogens whose transmission and pathogenesis target the mucosal tissue. The capsid proteins of human papillomavirus (HPV) confer tropism for the basal epithelium and can encapsidate DNA during self-assembly to form pseudovirions (PsVs). Therefore, we produced mucosal vaccine vectors by HPV PsV encapsidation of DNA plasmids expressing an experimental antigen derived from the M and M2 proteins of respiratory syncytial virus (RSV). Intravaginal (IVag) delivery elicited local and systemic M-M2-specific CD8+ T-cell and antibody responses in mice that were comparable to an approximately 10,000-fold higher dose of naked DNA. A single HPV PsV IVag immunization primed for M-M2-specific-IgA in nasal and vaginal secretions. Based on light emission and immunofluorescent microscopy, immunization with HPV PsV-encapsidated luciferase- and red fluorescent protein (RFP)-expressing plasmids resulted in transient antigen expression (<5 days), which was restricted to the vaginal epithelium. HPV PsV encapsidation of plasmid DNA is a novel strategy for mucosal immunization that could provide new vaccine options for selected mucosal pathogens.


Subject(s)
Mucous Membrane/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Viruses/physiology , Virion/metabolism , Administration, Intravaginal , Administration, Mucosal , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Epithelium/virology , Female , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Lymphocyte Activation , Mice , Mice, Inbred Strains , Mucous Membrane/immunology , Mucous Membrane/pathology , Mucous Membrane/virology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/transmission , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/pathogenicity , Vaccines, DNA , Vagina/pathology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Virion/genetics , Virion/pathogenicity
4.
Int J STD AIDS ; 14(4): 258-65, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12716496

ABSTRACT

A population at low risk for developing cervical cancer in Southern Brazil was studied to identify the main determinants of serological response to human papillomavirus (HPV). Enzyme-linked immunosorbent assay tests were performed in 976 women to detect serum IgG antibodies against HPV 16 L1 virus-like particles (VLPs) and HPVs 16, 18, 6 and 11 L1 VLPs as a mixture of antigens. Women with four or more sexual partners were more likely to be seropositive than women with one partner (HPV 16 serology odds ratio [OR]=3.06, 95% confidence interval [CI]: 2.0-4.8; HPV 6/11/16/18 serology OR=4.64, 95% CI: 3.0-7.2). HPV DNA and both serological responses were associated. Those positives to HPV 16 serology were twice as likely to have a cytological diagnosis of squamous intraepithelial lesions (SILs) than seronegatives (OR=2.07; 95% CI: 1.0-4.5, and OR=1.73; 95% CI: 0.8-3.8). Seropositivity to HPV 16 and HPV 6/11/16/18 antigens seem to be better markers of past sexual activity than current HPV infection, and humoral response to HPV 16 or HPV 6/11/16/18 may not be a strong indicator of cervical lesions in populations at low risk for cervical lesions.


Subject(s)
Antibodies, Viral/immunology , Papillomaviridae/immunology , Uterine Cervical Neoplasms/virology , Adolescent , Adult , Aged , Enzyme-Linked Immunosorbent Assay , Female , Humans , Middle Aged , Papillomaviridae/isolation & purification , Risk Factors , Uterine Cervical Neoplasms/epidemiology
5.
Expert Opin Biol Ther ; 1(4): 571-81, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11727495

ABSTRACT

Non-infectious human papillomavirus-like particles (VLP), composed of the L1 major capsid protein, are under active development as vaccines to prevent cervical cancer. They would presumably function primarily by generating virion-neutralising antibodies against the genital human papillomavirus (HPV) types that are the principal cause of most cervical cancers. Early phase clinical studies indicate that the VLP vaccines are well tolerated and able to consistently induce high titres of virus type-specific neutralising antibodies. Two types of second-generation VLP-based subunit vaccines with therapeutic implications, both related and unrelated to papillomavirus infection, are in preclinical development. One type seeks to induce cell-mediated immune responses, especially cytotoxic lymphocytes (CTL), against non-structural papillomavirus proteins, proteins of other viruses, or tumour associated antigens. The target antigen is incorporated into the VLPs as a fusion protein of L1 or the L2 minor capsid protein. In mouse models, this approach has generated potent CTL responses after low dose vaccination in the absence adjuvant. The second type of therapeutic VLP-based vaccine seeks to generate autoantibodies to self-antigens. The display of self polypeptides in the context of the highly ordered array of repetitive elements on the papillomavirus VLP surface abrogates the ability of the humoral immune system to functionally distinguish between foreign and self. High titre and high avidity auto-reactive IgG antibodies have been induced to both soluble (TNF-alpha) and cell surface (CCR5) central self-antigens. Vaccines based on this approach could potentially be effective alternatives to monoclonal antibody (mAb)-based therapies for a variety of disease targets.


Subject(s)
Capsid/immunology , Oncogene Proteins, Viral/immunology , Papillomaviridae/genetics , Papillomavirus Vaccines , Uterine Cervical Neoplasms/drug therapy , Viral Vaccines/therapeutic use , Adult , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , B-Lymphocytes/immunology , Capsid/genetics , Capsid Proteins , Clinical Trials as Topic , Dendritic Cells/immunology , Female , Humans , Oncogene Proteins, Viral/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control
6.
J Virol ; 75(21): 10493-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581419

ABSTRACT

Coexpression of bovine papillomavirus L1 with L2 mutants lacking either eight N-terminal or nine C-terminal amino acids that encode positively charged domains resulted in wild-type levels of viral genome encapsidation. Despite wild-type binding to the cell surface, the resulting virions were noninfectious. An L2 mutant encoding a scrambled version of the nine C-terminal residues restored infectivity, in contrast to an L2 mutant encoding a scrambled version of the N-terminal residues.


Subject(s)
Bovine papillomavirus 1/physiology , Capsid Proteins , Capsid/physiology , Nuclear Proteins , Animals , Capsid/chemistry , Cell Line , Cricetinae , Neoplasm Proteins/analysis , Transcription Factors/analysis , Tumor Suppressor Proteins , Virion/physiology , Virus Assembly
7.
J Clin Invest ; 108(3): 415-23, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489935

ABSTRACT

High avidity and long-lasting autoantibodies to a self-polypeptide (TNF-alpha) were generated after parenteral vaccination of mice with low doses of virus-like particle-based (VLP-based) vaccines that were constructed by linking mouse TNF-alpha peptides to the surface of papillomavirus VLPs. High-titer autoantibodies were induced with or without coadministration of potent conventional adjuvants, but were enhanced by coadministration of CFA. Compared with immunization with the fusion protein alone, attachment to VLPs increased autoantibody titers 1,000-fold. A comparison of Ab responses against the self (TNF-alpha) and foreign components of the fusion protein showed that VLP conjugation abrogated the ability of the humoral immune system to distinguish between self and foreign. Similar levels of IgM were detected to self and foreign epitopes regardless of the assembly state of the antigen, suggesting that conjugation of self-peptides to VLPs promotes survival or expansion of mature autoreactive B cells. In a mouse model, vaccination with conjugated particles inhibited development of type II collagen-induced arthritis. Together, these results suggest a potentially flexible method to efficiently generate autoantibodies against specific self-proteins that mediate arthritis and other diseases.


Subject(s)
Autoantibodies/biosynthesis , Autoantigens/administration & dosage , Papillomaviridae/immunology , Amino Acid Sequence , Animals , Arthritis/etiology , Arthritis/immunology , Arthritis/pathology , Arthritis/prevention & control , Autoantigens/genetics , Immunoglobulin M/biosynthesis , Male , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccination , Vaccines, Conjugate/administration & dosage
8.
Am J Vet Res ; 62(8): 1212-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11497440

ABSTRACT

OBJECTIVE: To determine expression of a transforming gene (E5) of bovine papillomavirus in sarcoids, other tumors, and normal skin samples collected from horses with and without sarcoids. SAMPLE POPULATION: 23 sarcoids and 6 samples of normal skin obtained from 16 horses with sarcoids, 2 samples of normal skin and 2 papillomas obtained from horses without sarcoids, and 1 papilloma obtained from a cow. PROCEDURE: Protein was extracted from tissue samples collected from horses and incubated with agarose beads covalently coupled to Staphylococcus aureus protein A and an anti-E5 polyclonal antibody. Following incubation, proteins were eluted from the beads and electrophoresed on a 14% polyacrylamide gel and transferred to a polyvinylidene difluoride membrane. The E5 protein was detected by use of western blot analysis, using a chemiluminescence detection system. RESULTS: All 23 sarcoids had positive results for expression of E5 protein. Quantity of viral protein appeared to vary among sarcoids. All other tissues examined had negative results for E5 protein. Highest expression for E5 protein was observed in biologically aggressive fibroblastic variants of sarcoids, compared with expression in quiescent tumors. CONCLUSIONS AND CLINICAL RELEVANCE: This study documented that activation and expression of the E5 gene is evident in sarcoids obtained from horses. These data support the conclusion that infection with bovine papillomavirus is important in the initiation or progression of sarcoids in horses. Treatment strategies designed to increase immune recognition of virally infected cells are warranted.


Subject(s)
Bovine papillomavirus 1/genetics , Horse Diseases/virology , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/veterinary , Sarcoidosis/veterinary , Skin Diseases, Infectious/veterinary , Tumor Virus Infections/veterinary , Animals , Blotting, Western/veterinary , Bovine papillomavirus 1/metabolism , DNA, Viral/chemistry , DNA, Viral/genetics , Electrophoresis, Polyacrylamide Gel/veterinary , Gene Expression Regulation, Viral , Horse Diseases/pathology , Horses , Oncogene Proteins, Viral/biosynthesis , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Polymerase Chain Reaction/veterinary , Precipitin Tests/veterinary , Sarcoidosis/pathology , Sarcoidosis/virology , Skin Diseases, Infectious/pathology , Skin Diseases, Infectious/virology , Transcription, Genetic/genetics , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
9.
Int Immunol ; 13(5): 633-41, 2001 May.
Article in English | MEDLINE | ID: mdl-11312251

ABSTRACT

Human papillomavirus virus-like particles (HPV VLP) and chimeric VLP are immunogens that are able to elicit potent anti-viral/tumor B and T cell responses. To investigate the immunogenicity of VLP, we determined which cells of the immune system are able to bind HPV-16 VLP. VLP were found to bind very well to human and mouse immune cells that expressed markers of antigen-presenting cells (APC) such as MHC class II, CD80 and CD86, including dendritic cells, macrophages and B cells. mAb blocking studies identified Fc gamma RIII (CD16) as one of the molecules to which the VLP can bind both on immune cells and foreskin epithelium. However, transfection of a CD16(-) cell line with CD16 did not confer binding of VLP. Splenocytes from Fc gamma RIII knockout mice showed a 33% decrease in VLP binding overall and specifically to subsets of APC. These combined data support a role for CD16 as an accessory molecule in an HPV VLP-receptor complex, possibly contributing to the immunogenicity of HPV VLP.


Subject(s)
Papillomaviridae/immunology , Animals , Antibodies, Blocking , Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Base Sequence , Cell Line , Chimera/immunology , DNA Primers/genetics , Humans , In Vitro Techniques , Male , Mice , Mice, Knockout , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Receptors, IgG/genetics , Receptors, IgG/metabolism , Transfection , Tumor Virus Infections/immunology
10.
J Immunol ; 166(9): 5346-55, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313370

ABSTRACT

The role of viral structural proteins in the initiation of adaptive immune responses is poorly understood. To address this issue, we focused on the effect of noninfectious papillomavirus-like particles (VLPs) on dendritic cell (DC) activation. We found that murine bone marrow-derived dendritic cells (BMDCs) effectively bound and rapidly internalized bovine papillomavirus VLPS: Exposure to fully assembled VLPs of bovine papillomavirus, human papillomavirus (HPV)16 or HPV18, but not to predominately disordered HPV16 capsomers, induced acute phenotypic maturation of BMDCS: Structurally similar polyomavirus VLPs bound to the DC surface and were internalized, but failed to induce maturation. DCs that had incorporated HPV16 VLPs produced proinflammatory cytokines IL-6 and TNF-alpha; however, the release of these cytokines was delayed relative to LPS activation. Production of IL-12p70 by VLP-exposed DCs required the addition of syngeneic T cells or rIFN-gamma. Finally, BMDCs pulsed with HPV16 VLPs induced Th1-dominated primary T cell responses in vitro. Our data provide evidence that DCs respond to intact papillomavirus capsids and that they play a central role in VLP-induced immunity. These results offer a mechanistic explanation for the striking ability of papillomavirus VLP-based vaccines to induce potent T and B cell responses even in the absence of adjuvant.


Subject(s)
Bovine papillomavirus 1/immunology , Capsid Proteins , Dendritic Cells/immunology , Dendritic Cells/virology , Papillomaviridae/immunology , Virion/immunology , Animals , BK Virus/immunology , Capsid/immunology , Capsid/metabolism , Cattle , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Interphase/immunology , JC Virus/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Protein Binding/immunology , Th1 Cells/immunology , Th1 Cells/virology , Virus Assembly/genetics , Virus Assembly/immunology
11.
J Virol ; 75(9): 4332-42, 2001 May.
Article in English | MEDLINE | ID: mdl-11287582

ABSTRACT

BPHE-1 cells, which harbor 50 to 200 viral episomes, encapsidate viral genome and generate infectious bovine papillomavirus type 1 (BPV1) upon coexpression of capsid proteins L1 and L2 of BPV1, but not coexpression of BPV1 L1 and human papillomavirus type 16 (HPV16) L2. BPV1 L2 bound in vitro via its C-terminal 85 residues to purified L1 capsomers, but not with intact L1 virus-like particles in vitro. However, when the efficiency of BPV1 L1 coimmunoprecipitation with a series of BPV1 L2 deletion mutants was examined in vivo, the results suggested that residues 129 to 246 and 384 to 460 contain independent L1 interaction domains. An L2 mutant lacking the C-terminal L1 interaction domain was impaired for encapsidation of the viral genome. Coexpression of BPV1 L1 and a chimeric L2 protein composed of HPV16 L2 residues 1 to 98 fused to BPV1 L2 residues 99 to 469 generated infectious virions. However, inefficient encapsidation was seen when L1 was coexpressed with either BPV1 L2 with residues 91 to 246 deleted or with BPV1 L2 with residues 1 to 225 replaced with HPV16 L2. Impaired genome encapsidation did not correlate closely with impairment of the L2 proteins either to localize to promyelocytic leukemia oncogenic domains (PODs) or to induce localization of L1 or E2 to PODs. We conclude that the L1-binding domain located near the C terminus of L2 may bind L1 prior to completion of capsid assembly, and that both L1-binding domains of L2 are required for efficient encapsidation of the viral genome.


Subject(s)
Bovine papillomavirus 1/metabolism , Capsid Proteins , Capsid/metabolism , Genome, Viral , Virus Assembly/physiology , Animals , Binding Sites , Bovine papillomavirus 1/genetics , Capsid/genetics , Cattle , Cell Line , Cricetinae , Humans , Mutagenesis , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomaviridae/metabolism , Precipitin Tests
12.
N Engl J Med ; 344(15): 1125-31, 2001 Apr 12.
Article in English | MEDLINE | ID: mdl-11297703

ABSTRACT

BACKGROUND: Oncogenic human papillomaviruses (HPVs), especially HPV type 16 (HPV-16), cause anogenital epithelial cancers and are suspected of causing epithelial cancers of the head and neck. METHODS: To examine the relation between head and neck cancers and HPVs, we performed a nested case-control study within a joint Nordic cohort in which serum samples were collected from almost 900,000 subjects. Samples collected at enrollment from 292 persons in whom squamous-cell carcinoma of the head and neck developed, on average, 9.4 years after enrollment and from 1568 matched controls were analyzed for antibodies against HPV-16, HPV-18, HPV-33, and HPV-73 and for cotinine levels as a marker of smoking habits. Polymerase-chain-reaction (PCR) analyses for HPV DNA were performed in tumor tissue from 160 of the study patients with cancer. RESULTS: After adjustment for cotinine levels, the odds ratio for squamous-cell carcinoma of the head and neck in subjects who were seropositive for HPV-16 was 2.2 (95 percent confidence interval, 1.4 to 3.4). No increased risk was observed for other HPV types. Fifty percent of oropharyngeal and 14 percent of tongue cancers contained HPV-16 DNA, according to PCR analysis. CONCLUSIONS: HPV-16 infection may be a risk factor for squamous-cell carcinoma of the head and neck.


Subject(s)
Antibodies, Viral/blood , Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/virology , Papillomaviridae , Papillomavirus Infections/complications , Tumor Virus Infections/complications , Adult , Carcinoma, Squamous Cell/blood , Case-Control Studies , Cohort Studies , Cotinine/blood , Female , Head and Neck Neoplasms/blood , Humans , Male , Middle Aged , Odds Ratio , Papillomaviridae/classification , Papillomaviridae/immunology , Risk Factors
13.
Cancer Epidemiol Biomarkers Prev ; 10(2): 101-6, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11219765

ABSTRACT

Genital human papillomavirus (HPV) infection is generally considered to be sexually transmitted. However, nonsexual spread of the virus has also been suggested. The goal of this study was to assess: (a) the role of sexual intercourse in the transmission of HPV; (b) the determinants for seroconversion; and (c) the correlation between HPV DNA, abnormal cervical cytology, and serological response to HPV16. One hundred virgins and 105 monogamous women were randomly selected from a population-based cohort study in Copenhagen, Denmark, in which the women were examined twice with 2-year interval (interview, cervical swabs, Pap smear, blood samples). The presence of HPV DNA was determined by GP5+/6+ primers based HPV-PCR-EIA. HPV 16 virus-like particles (VLP) antibodies were detected by ELISA. All of the virgins were both HPV DNA negative and seronegative to VLP16, except for one woman who was weakly HPV 6 DNA positive. Only those virgins who initiated sexual activity became HPV DNA positive and/or VLP16 positive. The most important determinant of HPV DNA acquisition was the number of partners between the two examinations. The only significant risk factor for HPV 16 VLP seroconversion among women acquiring HPV DNA was HPV type. Our results show that sexual intercourse is important in the transmission of HPV, and that HPV 16 VLP seroconversion and the development of cervical lesions only occur after HPV transmission. Remarkably, no cervical lesions were found in HPV 16 DNA positive women who had seroconverted. Although based on small numbers, this may suggest that the development of antibodies had a protective effect.


Subject(s)
Coitus , Disease Transmission, Infectious/statistics & numerical data , Papillomaviridae/isolation & purification , Papillomavirus Infections/transmission , Sexually Transmitted Diseases, Viral/transmission , Tumor Virus Infections/transmission , Adult , Cohort Studies , DNA, Viral/analysis , Denmark/epidemiology , Female , Humans , Incidence , Longitudinal Studies , Papillomavirus Infections/diagnosis , Papillomavirus Infections/epidemiology , Polymerase Chain Reaction/methods , Prospective Studies , Risk Assessment , Risk Factors , Sexually Transmitted Diseases, Viral/diagnosis , Sexually Transmitted Diseases, Viral/epidemiology , Tumor Virus Infections/diagnosis , Tumor Virus Infections/epidemiology
14.
J Natl Cancer Inst ; 93(4): 284-92, 2001 Feb 21.
Article in English | MEDLINE | ID: mdl-11181775

ABSTRACT

BACKGROUND: Studies in animal models have shown that systemic immunization with a papillomavirus virus-like particle (VLP) vaccine composed of L1, a major structural viral protein, can confer protection against subsequent experimental challenge with the homologous virus. Here we report results of a double-blind, placebo-controlled, dose-escalation trial to evaluate the safety and immunogenicity of a human papillomavirus (HPV) type 16 (HPV16) L1 VLP vaccine in healthy adults. METHODS: Volunteers were given intramuscular injections with placebo or with 10- or 50-microg doses of HPV16 L1 VLP vaccine given without adjuvant or with alum or MF59 as adjuvants at 0, 1, and 4 months. All vaccine recipients were monitored for clinical signs and symptoms for 7 days after each inoculation. Immune responses were measured by an HPV16 L1 VLP-based enzyme-linked immunosorbent assay (ELISA) and by an HPV16 pseudovirion neutralization assay. The antibody titers were given as the reciprocals of the highest dilution showing positive reactivity in each assay. All statistical tests were two-sided. RESULTS: The prevaccination geometric mean ELISA titer for six seropositive individuals was 202 (range, 40--640). All vaccine formulations were well tolerated, and all subjects receiving vaccine seroconverted. Serum antibody responses at 1 month after the third injection were dose dependent in recipients of vaccine without adjuvant or with MF59 but were similar at both doses when alum was the adjuvant. With the higher dose, the geometric means of serum ELISA antibody titers (95% confidence intervals) to purified VLP 1 month after the third injection were as follows: 10,240 (1499 to 69 938) without adjuvant, 10,240 (1114 to 94 145) with MF59, and 2190 (838 to 5723) with alum. Responses of subjects within each group were similar. Neutralizing and ELISA antibody titers were highly correlated (Spearman correlation =.85), confirming that ELISA titers are valid proxies for neutralizing antibodies. CONCLUSIONS: The HPV16 L1 VLP vaccine is well tolerated and is highly immunogenic even without adjuvant, with the majority of the recipients achieving serum antibody titers that were approximately 40-fold higher than what is observed in natural infection.


Subject(s)
Antibodies, Viral/blood , Papillomaviridae/immunology , Papillomavirus Vaccines , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Alum Compounds/administration & dosage , Baculoviridae , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization Schedule , Immunoglobulins/blood , Injections, Intramuscular , Male , Polysorbates/administration & dosage , Recombinant Proteins , Reference Values , Squalene/administration & dosage , Viral Vaccines/administration & dosage
15.
J Natl Cancer Inst Monogr ; (28): 50-4, 2001.
Article in English | MEDLINE | ID: mdl-11158207

ABSTRACT

Papillomavirus-like particle (VLP)-based subunit vaccines have undergone rapid development over the past 8 years. Three types are being investigated. The most basic type is composed of only the L1 major capsid protein and is designed to prevent genital human papillomavirus (HPV) infection by inducing virus-neutralizing antibodies. On the basis of positive results in animal models, clinical trials of this type of vaccine for HPV16, and other types, are currently under way. Preliminary results have been encouraging in that systemic immunization with the L1 VLPs induced high serum titers of neutralizing antibodies without substantial adverse effects. The second type of vaccine incorporates other papillomavirus polypeptides into the VLPs as L1 or L2 fusion proteins. These chimeric VLPs are designed to increase the therapeutic potential of an HPV vaccine by inducing cell-mediated responses to nonstructural viral proteins, such as E7. Studies in mice indicate that these vaccines generate potent antitumor cytotoxic lymphocyte (CTL) responses while retaining the ability to induce high-titer neutralizing antibodies. It is likely that prophylactic and therapeutic clinical trials of chimeric VLPs will be initiated in the near future. The third type of VLP-based vaccine is designed to induce autoantibodies against central self-antigens by incorporating self-peptides into the outer surface of VLPs, a process that could have therapeutic potential in various disease settings unrelated to HPV infection. In a recent proof of concept study, a peptide from an external loop of mouse CCR5 protein was inserted into a neutralizing epitope of L1. In mice, the particles generated by this chimeric L1 were able to induce high titers of CCR5 antibodies that specifically recognized the surface of CCR5-transfected cells and blocked in vitro infection of an M-tropic human immunodeficiency virus strain.


Subject(s)
Capsid Proteins , Papillomaviridae/immunology , Papillomavirus Vaccines , Vaccines, Subunit , Viral Vaccines , Animals , Antibodies, Viral/biosynthesis , Autoantibodies/biosynthesis , Clinical Trials as Topic , Cytotoxicity, Immunologic , Female , HIV Infections/prevention & control , Humans , Immunization , Male , Mice , Oncogene Proteins, Viral/immunology , Papillomaviridae/ultrastructure , Papillomavirus Infections/prevention & control , Rabbits , Receptors, CCR5/immunology , Receptors, HIV/immunology , Recombinant Fusion Proteins/immunology , Sexually Transmitted Diseases, Viral/prevention & control , Transfection , Tumor Virus Infections/prevention & control , Vaccines, Subunit/immunology , Viral Vaccines/immunology , Virion/ultrastructure
16.
Virology ; 279(1): 354-60, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11145916

ABSTRACT

Human papillomaviruses, mainly type 16 (HPV16), are responsible for cervical intraepithelial neoplasia, which can lead, in association with other factors, to cervical cancer. Both Salmonella recombinant vaccine strains assembling HPV16 virus-like particles (VLPs) and HPV16 VLPs purified from insect cells are able to induce HPV16 neutralizing antibodies in genital secretions of mice after nasal immunization. Anti-HPV16-specific antibodies in cervical secretions of women may prevent genital infection with HPV16, although this cannot be critically evaluated in the absence of an experimental model for genital papillomavirus infection. Induction of HPV16-specific cell-mediated immunity in the genital mucosa could improve the efficacy of a vaccine and a mucosal route of immunization might be necessary to do so. It has been shown that systemic immunization of mice with purified HPV16 VLPs confers protection against an HPV16-expressing tumor cell challenge through the induction of cytotoxic T-lymphocytes. Using the same C3 tumor model, we show that intranasal immunization of mice with purified HPV16 VLPs in a prophylactic setting also induces anti-tumor immunity. More interestingly, mucosal vaccination of mice with a Salmonella recombinant strain stably expressing HPV16 L1 VLPs also induces anti-tumor immunity in prophylactic as well as in therapeutic settings. Our data suggest that attenuated Salmonella strains expressing chimeric VLPs containing nonstructural viral proteins might be a promising candidate vaccine against cervical cancer by inducing both neutralizing antibodies and cell-mediated immunity.


Subject(s)
Cancer Vaccines/immunology , Immunity, Mucosal , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Salmonella typhimurium/genetics , Tumor Virus Infections/immunology , Viral Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Viral/analysis , Cancer Vaccines/administration & dosage , Cells, Cultured , Female , Insecta , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus Infections/prevention & control , Tumor Cells, Cultured , Tumor Virus Infections/prevention & control , Vaccination , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Virion/immunology , Virion/isolation & purification , Virion/metabolism
17.
Virology ; 279(1): 361-9, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11145917

ABSTRACT

Genital HPV genotypes are generally distinct serotypes, but whether variants within a genotype can represent serologic subtypes is unclear. In this study we used serum from human volunteers vaccinated with HPV16 L1 VLPs from variant 114K, to examine cross-neutralization of variants from each of the five major phylogenetic branches of HPV16. Recombinant Semliki Forest virus-derived pseudovirions for each variant were generated and combined with serum from vaccines, and the mixture was monitored for infectivity in a standard C127 cell focal transformation assay. Sera from all 10 VLP-immunized individuals had neutralizing activity against each of the variant pseudovirions. For each of the sera, variant titers differed by only fourfold or less from the median titer. Therefore, from a vaccine perspective, HPV16 variants belong to a single serotype. Vaccination with HPV16 114K L1 VLPs generates antibodies that should confer a similar degree of protection against all known phylogenetic branches of HPV16.


Subject(s)
Antibodies, Viral/immunology , Papillomaviridae/immunology , Viral Vaccines/immunology , Virion/immunology , Antibodies, Viral/blood , Cross Reactions , Humans , Immunization , Neutralization Tests , Papillomaviridae/classification , Papillomaviridae/genetics , Papillomavirus Infections/prevention & control , Phylogeny , Serotyping , Tumor Virus Infections/prevention & control , Viral Vaccines/administration & dosage , Virion/metabolism , Virus Assembly
18.
Virology ; 290(2): 350-60, 2001 Nov 25.
Article in English | MEDLINE | ID: mdl-11883199

ABSTRACT

Chimeric human papillomavirus virus-like particles (HPV cVLPs) carrying HPV16 E7 protein are potent vaccines for inducing cell-mediated immunity (CMI) against HPV-induced tumors in animal models. We tested the hypothesis that virion-neutralizing antibodies generated during an initial vaccination might prevent effective boosting of CMI to the cVLPs. Mice with circulating HPV16-neutralizing antibodies, generated by direct immunization with wild-type VLPs or by passive transfer of hyperimmune anti-HPV16 VLP mouse sera, were subsequently vaccinated with HPV16 E7-containing cVLPs. Mice with preexisting neutralizing antibodies were not protected from HPV16 E7-positive TC-1 tumor challenge, compared to the protection seen in mice lacking these antibodies. Antibody-coated VLPs bound very inefficiently to receptor-positive cell lines, suggesting that one of the mechanisms of antibody interference is blocking of VLP binding to its receptor and thereby uptake of VLPs by antigen-presenting cells. Our results suggest that repetitive vaccination with a cVLP for induction of cellular immune responses to an incorporated antigen may be of limited effectiveness due to the presence of neutralizing antibodies against the capsid proteins induced after the first application. This limitation could potentially be overcome by boosting with cVLPs containing the same target antigen incorporated into other papillomavirus-type VLPs.


Subject(s)
Antibodies, Viral/immunology , Papillomaviridae/immunology , Papillomavirus Vaccines , Viral Vaccines/immunology , Animals , Female , HeLa Cells , Humans , Immunization, Passive , Mice , Mice, Inbred C57BL , Neutralization Tests , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins , Recombination, Genetic , Virion/immunology
19.
J Clin Virol ; 19(1-2): 67-74, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11091149

ABSTRACT

BACKGROUND: the knowledge that sexually transmitted infection with one of a limited number of human papillomaviruses (HPVs) is a central cause of almost all cervical cancers affords the opportunity to prevent this common cancer through anti-viral vaccination. OBJECTIVE: the spectacular success of vaccines in preventing several other viral diseases offers hope that immunoprophylaxis against the relevant HPVs could lead to a major reduction in cervical cancer incidence. RESULTS AND CONCLUSION: the results of preclinical studies and early phase clinical trials of virus-like particle (VLP) based subunit vaccines have been very encouraging. However, unique aspects of papillomavirus biology and genital tract infections, and the lack of sexual a transmission model for papillomavirus, make it far from certain that effective prophylactic vaccination against genital HPV infection will be easily achieved. Future clinical efficacy trials will likely test the hypothesis that parenteral injection of VLPs can induce antibody mediated and type specific protection against genital tract HPV infection and subsequent development of premalignant neoplastic disease.


Subject(s)
Papillomaviridae/immunology , Papillomavirus Vaccines , Uterine Cervical Neoplasms/prevention & control , Viral Vaccines/administration & dosage , Animals , Drug Design , Drug Evaluation, Preclinical , Female , Uterine Cervical Neoplasms/immunology , Viral Vaccines/immunology , Virion/immunology , Virion/isolation & purification
20.
Virology ; 270(2): 254-7, 2000 May 10.
Article in English | MEDLINE | ID: mdl-10792983

ABSTRACT

Vaccination with virus-like particles (VLP), comprising both L1 and L2 of human papillomavirus (HPV) genital types 6, 16, and 18, induces predominantly type-specific neutralizing antibodies. L2 polypeptide vaccines protect animals against experimental challenge with homologous papillomavirus and cross-reactive epitopes are present in HPV L2. To assess L2-specific cross-neutralization of HPV genotypes, sheep were immunized with purified, bacterially expressed HPV6, 16, or 18 L2. In addition to neutralizing the homologous HPV type in vitro, antisera to each HPV L2 also cross-neutralized both heterologous HPV types. This suggests that unlike VLP-based prophylactic HPV vaccines, an L2 polypeptide vaccine may provide broad-spectrum protection.


Subject(s)
Capsid/immunology , Immunodominant Epitopes/immunology , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Tumor Virus Infections/immunology , Viral Vaccines/immunology , Cross Reactions , Epitopes/immunology , Humans , Papillomavirus Infections/prevention & control , Tumor Virus Infections/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...