Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 87(11): 6127-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536682

ABSTRACT

While the oncogenic human papillomavirus (HPV) types with the greatest medical impact are clustered within the α9 and α7 species, a significant fraction of cervical cancers are caused by α5, α6, and α11 viruses. Benign genital warts are caused principally by the α10 viruses HPV6 and HPV11. In an effort to achieve broad protection against both cervical cancer- and genital wart-associated types, we produced at high levels in bacteria a multimeric protein (α11-88x8) fusing eight polypeptides corresponding to a protective domain comprising L2 residues ∼11 to 88 derived from HPV6 (α10), HPV16 (α9), HPV18 (α7), HPV31 (α9), HPV39 (α7), HPV51 (α5), HPV56 (α6), and HPV73 (α11) and a truncated derivative with the last three units deleted (α11-88x5). Mice were immunized three times with α11-88x8 or α11-88x5 adjuvanted with alum or the licensed HPV vaccines and challenged intravaginally with HPV6, HPV16, HPV26, HPV31, HPV33, HPV35, HPV45, HPV51, HPV56, HPV58, or HPV59 pseudovirions. The α11-88x5 and α11-88x8 vaccines induced similarly robust protection against each HPV type tested and indistinguishable HPV16-neutralizing antibody titers. Passive transfer of α11-88x8 antisera was protective. Further, rabbit antisera to α11-88x8 and α11-88x5 similarly neutralized native HPV18 virions. These findings suggest that immunologic competition between units is not a significant issue and that it is not necessary to include a unit of L2 derived from each species to achieve broader protection against diverse medically significant HPV types than is achieved with the licensed HPV vaccines.


Subject(s)
Alphapapillomavirus/classification , Alphapapillomavirus/genetics , Capsid Proteins/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Phylogeny , Alphapapillomavirus/immunology , Animals , Antibodies, Viral/immunology , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Female , Genotype , Humans , Mice , Mice, Inbred BALB C , Papillomavirus Infections/immunology , Papillomavirus Vaccines/genetics , Papillomavirus Vaccines/immunology , Rabbits
2.
PLoS One ; 8(1): e55538, 2013.
Article in English | MEDLINE | ID: mdl-23383218

ABSTRACT

We sought to define the protective epitopes within the amino terminus of human papillomavirus (HPV) type 16 minor capsid protein L2. Passive transfer of mice with rabbit antisera to HPV16 L2 peptides 17-36, 32-51 and 65-81 provided significant protection against vaginal HPV16 challenge, whereas antisera to 47-66, 108-120 or 373-392 did not. Vaccination with L1 virus-like particles induces a high titer, but generally type-restricted neutralizing antibody response. Conversely, vaccination with L2 11-88, especially multimers thereof, induces antibodies that neutralize a broad range of papillomavirus types, albeit at lower titers than for L1 VLP. With the intent of enhancing the immunogenicity and the breadth of protection by focusing the immune response to the key protective epitopes, we designed L2 fusion proteins consisting of residues ∼11-88 of eight divergent mucosal HPV types 6, 16, 18, 31, 39, 51, 56, 73 (11-88×8) or residues ∼13-47 of fifteen HPV types (13-47×15). The 11-88×8 was significantly more immunogenic than 13-47×15 in Balb/c mice regardless of the adjuvant used, suggesting the value of including the 65-81 protective epitope in the vaccine. Since the L2 47-66 peptide antiserum failed to elicit significant protection, we generated an 11-88×8 construct deleted for this region in each subunit (11-88×8Δ). Mice were vaccinated with 11-88×8 and 11-88×8Δ to determine if deletion of this non-protective epitope enhanced the neutralizing antibody response. However, 11-88×8Δ was significantly less immunogenic than 11-88×8, and even the addition of a known T helper epitope, PADRE, to the construct (11-88×8ΔPADRE) failed to recover the immunogenicity of 11-88×8 in C57BL/6 mice, suggesting that while L2 47-66 is not a critical protective or T helper epitope, it nevertheless contributes to the immunogenicity of the L2 11-88×8 multimer vaccine.


Subject(s)
Capsid Proteins/immunology , Human papillomavirus 16/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid Proteins/chemistry , Epitopes/chemistry , Epitopes/immunology , Female , Humans , Mice , Neutralization Tests , Oncogene Proteins, Viral/chemistry , Papillomavirus Infections/prevention & control , Peptides/chemistry , Peptides/immunology , Rabbits , Vagina/immunology , Vagina/virology
3.
J Natl Cancer Inst ; 101(11): 782-92, 2009 Jun 03.
Article in English | MEDLINE | ID: mdl-19470949

ABSTRACT

BACKGROUND: Vaccination with minor capsid protein L2 induces antibodies that cross-neutralize diverse papillomavirus types. However, neutralizing antibody titers against the papillomavirus type from which the L2 vaccine was derived are generally higher than the titers against heterologous types, which could limit effectiveness against heterologous types. We hypothesized that vaccination with concatenated multitype L2 fusion proteins derived from known cross-protective epitopes of several divergent human papillomavirus (HPV) types might enhance immunity across clinically relevant HPV genotypes. METHODS: Antibody responses of mice (n = 120) and rabbits (n = 23) to vaccination with HPV-16 amino-terminal L2 polypeptides or multitype L2 fusion proteins, namely, 11-200 x 3 (HPV types 6, 16, 18), 11-88 x 5 (HPV types 1, 5, 6, 16, 18), or 17-36 x 22 (five cutaneous, two mucosal low-risk, and 15 oncogenic types), that were formulated alone or in GPI-0100, alum, or 1018 ISS adjuvants were compared with vaccination with L1 virus-like particles (VLPs), including Gardasil, a licensed quadrivalent HPV L1 vaccine, and a negative control. Mice were challenged with HPV-16 pseudovirions 4 months after vaccination. Statistical tests were two-sided. RESULTS: The HPV-16 L2 polypeptides generated robust HPV-16-neutralizing antibody responses, albeit lower than those to HPV-16 L1 VLPs, and lower responses against other HPVs. In contrast, vaccination with the multitype L2 fusion proteins 11-200 x 3 and 11-88 x 5 induced high serum neutralizing antibody titers against all heterologous HPVs tested. 11-200 x 3 formulated in GPI-0100 adjuvant or alum with 1018 ISS protected mice against HPV-16 challenge (reduction in HPV-16 infection vs phosphate-buffered saline control, P < .001) 4 months after vaccination as well as HPV-16 L1 VLPs, but 11-200 x 3 alone or formulated with either alum or 1018 ISS was less effective (reduction in HPV-16 infection, P < .001). CONCLUSION: Concatenated multitype L2 proteins in adjuvant have potential as pan-oncogenic HPV vaccines.


Subject(s)
Antibodies, Viral/immunology , B-Lymphocytes/immunology , Capsid Proteins/immunology , Human papillomavirus 16/immunology , Neutralization Tests , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Analysis of Variance , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Capsid Proteins/therapeutic use , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18 , Humans , Immune Sera , Mice , Mice, Inbred BALB C , Neutralization Tests/methods , Oncogene Proteins, Viral/therapeutic use , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Papillomavirus Vaccines/therapeutic use , Rabbits , Time Factors , Virion
SELECTION OF CITATIONS
SEARCH DETAIL