Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
2.
Vaccine ; 38(2): 101-106, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31706809

ABSTRACT

Respiratory syncytial virus (RSV), is a common cause of serious acute lower respiratory tract illness in infants and young children, causing substantial morbidity and mortality globally. Treatment is mainly supportive and currently there is no licensed preventive vaccine. Clinical trials conducted in the 1960s evaluating a formalin-inactivated RSV vaccine (FI-RSV) in RSV-naïve infants resulted in observations of enhanced respiratory disease (ERD) following subsequent natural RSV infection in vaccinees. In these studies, infants immunized with FI-RSV had higher rates of severe RSV disease compared with controls. This outcome redirected focus on identifying the immunologic mechanisms that precipitated ERD as a prerequisite to further vaccine development. Improved understanding of the immunopathogenesis of ERD derived from animal models has stimulated development of new candidate vaccines and engendered discussions among RSV experts about the safety data needed to advance these products into the clinic, and ultimately, into the target population of RSV-naïve infants. The recognition that multiple products would soon be ready for testing in infants and children prompted the FDA to hold a Vaccines and Related Biological Products Advisory Committee (VRBPAC) meeting to seek perspectives and advice of experts regarding the types and extent of preclinical and clinical data that might be needed to support testing in RSV-naïve infants for specific types of candidate RSV vaccines. Committee members agreed that, if certain conditions are met in preclinical and early clinical studies, it would be reasonable to move forward from studies in adults and older children and into clinical trials evaluating vaccine safety and efficacy in RSV-naïve infants. Herein, we review and summarize perspectives on the discussion regarding recommendations for RSV vaccine development in this population.


Subject(s)
Immunization , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus, Human/immunology , Advisory Committees , Animals , Biological Products/administration & dosage , Child, Preschool , Disease Models, Animal , Humans , Infant , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Severity of Illness Index
3.
Vaccine ; 34(41): 4843-4849, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27566900

ABSTRACT

Respiratory syncytial virus (RSV) is the most common cause of serious acute lower respiratory illness in infants and young children and a significant cause of disease burden in the elderly and immunocompromised. There are no licensed RSV vaccines to address this significant public health need. While advances in vaccine technologies have led to a recent resurgence in RSV vaccine development, the immune correlates of protection against RSV and the immunology of vaccine-associated enhanced respiratory disease (ERD) remain poorly understood. FDA's Center for Biologics Evaluation and Research (CBER) and NIH's National Institute of Allergy and Infectious Diseases (NIAID) organized and co-sponsored an RSV Vaccines Workshop in Bethesda, Maryland on June 1 and 2, 2015. The goal of the conference was to convene scientists, regulators, and industry stakeholders to discuss approaches to RSV vaccine development within the context of three target populations - infants and children, pregnant women, and individuals >60years of age. The agenda included topics related to RSV vaccine development in general, as well as considerations specific to each target population, such as clinical and serological endpoints. The meeting focused on vaccine development for high income countries (HIC), because issues relevant to vaccine development for low and middle income countries (LMIC) have been discussed in other forums. This manuscript summarizes the discussion of clinical, scientific, and regulatory perspectives, research gaps, and lessons learned.


Subject(s)
Biomedical Research , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Aged , Animals , Child, Preschool , Clinical Trials as Topic , Congresses as Topic , Female , Humans , Infant , Maryland , National Institutes of Health (U.S.) , Pregnancy , Respiratory Syncytial Virus, Human , United States , United States Food and Drug Administration
4.
Int J Cancer ; 138(4): 901-11, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26317490

ABSTRACT

We previously determined that human papillomavirus (HPV) virus-like particles (VLPs) and pseudovirions (PsV) did not, respectively, bind to or infect intact epithelium of the cervicovaginal tract. However, they strongly bound heparan sulfate proteoglycans (HSPG) on the basement membrane of disrupted epithelium and infected the keratinocytes that subsequently entered the disrupted site. We here report that HPV capsids (VLP and PsV) have the same restricted tropism for a wide variety of disrupted epithelial and mesothelial tissues, whereas intact tissues remain resistant to binding. However, the HPV capsids directly bind and infect most tumor-derived cell lines in vitro and have analogous tumor-specific properties in vivo, after local or intravenous injection, using orthotopic models for human ovarian and lung cancer, respectively. The pseudovirions also specifically infected implanted primary human ovarian tumors. Heparin and ι-carrageenan blocked binding and infection of all tumor lines tested, implying that tumor cell binding is HSPG-dependent. A survey using a panel of modified heparins indicates that N-sulfation and, to a lesser degree, O-6 sulfation of the surface HSPG on the tumors are important for HPV binding. Therefore, it appears that tumor cells consistently evolve HSPG modification patterns that mimic the pattern normally found on the basement membrane but not on the apical surfaces of normal epithelial or mesothelial cells. Consequently, appropriately modified HPV VLPs and/or PsV could be useful reagents to detect and potentially treat a remarkably broad spectrum of cancers.


Subject(s)
Capsid/metabolism , Human papillomavirus 16/metabolism , Neoplasms/virology , Papillomavirus Infections/virology , Animals , Cell Line, Tumor , Cell Separation , Female , Heparan Sulfate Proteoglycans/metabolism , Humans , Mice
5.
Vaccine ; 33(8): 966-72, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25573034

ABSTRACT

Despite supportive public health policies (e.g., ACIP recommendations), the potential for providing clinical benefit through maternal immunization has yet to be fully realized. For vaccines already licensed and approved for use in adults, specific FDA approval for use during pregnancy to prevent disease in the mother and/or infant may have a significant impact on uptake and usage in pregnant women. In addition, for either a licensed vaccine or a novel vaccine, FDA approval for use during pregnancy would result in labeling that would serve as a resource for practitioners and would facilitate the safe and effective use of the vaccine during pregnancy. In the U.S., while many vaccines are approved for use in adults and most are not contraindicated for use in pregnant women, no vaccine is licensed for use specifically during pregnancy. Among the perceived obstacles hindering the clinical development of vaccines for use in pregnancy, regulatory issues are frequently cited. One aim of this article is to address the perceived regulatory obstacles. General concepts and regulatory considerations for clinical safety and effectiveness evaluations for vaccines indicated for use during pregnancy will be discussed. This discussion is not intended to establish data requirements or to articulate agency policy or guidance regarding specific vaccine products.


Subject(s)
Communicable Disease Control/legislation & jurisprudence , Pregnancy Complications, Infectious/prevention & control , Vaccines , Adult , Clinical Trials as Topic , Female , Humans , Pregnancy , Vaccines/adverse effects , Vaccines/immunology , Vaccines/standards
6.
J Natl Cancer Inst ; 103(9): 737-43, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21483020

ABSTRACT

BACKGROUND: Human papillomavirus (HPV) infection of the genital mucosa is thought to require trauma to the cervicovaginal epithelium. Therefore, we determined whether a cytology specimen collection procedure (Pap smear), which disrupts the epithelium by design, renders the cervix more susceptible to HPV infection in a primate model. METHODS: In a series of female rhesus macaques, a speculum examination was performed with (n = 8) or without (n = 4) a cytology specimen collection procedure as it is commonly practiced in a gynecology clinic. An internal digital examination was performed after specimen collection using Surgilube (n = 4) or 1% iota-carrageenan, a previously indentified HPV inhibitor (n = 4) as the lubricant. The cervix was then inoculated with HPV16 pseudovirions expressing red fluorescent protein. After 3 days, the reproductive tracts were excised and the cervix was cryosectioned. Sections were analyzed by fluorescent confocal microscopy for the number of red fluorescent protein-positive keratinocytes. RESULTS: Substantial infection of the ectocervix, the transformation zone, and the endocervix was detected, but only in conjunction with the cytology specimen collection procedure (cytology using Surgilube vs without cytology using Surgilube, mean = 84 infectious events per section vs mean = 0.05 infectious events per section, difference = 84 infectious events per section, 95% confidence interval = 19 to 384 infectious events per section). When the carrageenan gel was substituted for Surgilube for an internal digital examination, the mean number of infectious events decreased (carrageenan gel vs Surgilube, mean = 3.5 events per section vs mean = 84 infectious events per section difference = 81 events per section, 95% confidence interval = 33 to 213 events per section). CONCLUSIONS: These findings indicate that cytology screening in women might lead to a transient enhancement of susceptibility to HPV infection and that use of a carrageenan-based gel during the examination might mitigate this enhancement.


Subject(s)
Anti-Infective Agents, Local , Carrageenan , Cervix Uteri/virology , Human papillomavirus 16/isolation & purification , Papanicolaou Test , Papillomavirus Infections/etiology , Papillomavirus Infections/prevention & control , Phenylmercury Compounds , Tumor Virus Infections/etiology , Tumor Virus Infections/prevention & control , Vaginal Smears/adverse effects , Vaginal Smears/methods , Animals , Disease Models, Animal , Disease Susceptibility , Female , Gels , Gene Expression Regulation, Viral , Human papillomavirus 16/metabolism , Humans , Keratinocytes/chemistry , Luminescent Proteins/analysis , Macaca mulatta , Microscopy, Confocal , Microscopy, Fluorescence , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Risk Factors , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Virion , Red Fluorescent Protein
7.
Cancer Prev Res (Phila) ; 4(5): 767-73, 2011 May.
Article in English | MEDLINE | ID: mdl-21430072

ABSTRACT

Human papillomavirus (HPV) infection is the most common sexually transmitted infection. Vaccines for HPV infection can reduce the risk of developing cervical cancer. To further improve such vaccines and to explore other methods of preventing or treating viral infection, longitudinal studies in experimental animals are desirable. Here, we describe a newly developed multicolor endoscopic fluorescence imaging system to visualize early HPV infection with fluorescent protein-encoded pseudoviruses (PsV) in the female genital tract of living mice. With this imaging method, the course of HPV PsV infection and the effects of intervention to prevent infection can be monitored in a single mouse over time. Female immunocompetent or athymic mice were pretreated with a vaginal spermicide and then HPV PsV composed of an authentic viral capsid and encapsidating green or red fluorescent protein (GFP or RFP) reporter gene was intravaginally instilled. Expression of GFP or RFP was detected 1 day after PsV challenge, which peaked after 2 or 3 days and decreasing thereafter. No fluorescence was detected in vaccine-treated immunocompetent mice. By using serial infection of the same PsV type (HPV16) encoding either GFP or RFP, different infection patterns of repeated exposure can be monitored. This method offers the ability to monitor experimental virus infections before and after intervention, thereby accelerating the development of appropriate prevention and therapy.


Subject(s)
Endoscopy , Papillomaviridae/physiology , Papillomavirus Infections/diagnosis , Papillomavirus Infections/virology , Animals , Female , Fluorescence , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/therapeutic use
8.
J Virol ; 83(5): 2067-74, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19073722

ABSTRACT

The host factors required for in vivo infection have not been investigated for any papillomavirus. Using a recently developed murine cervicovaginal challenge model, we evaluated the importance of heparan sulfate proteoglycans (HSPGs) in human papillomavirus (HPV) infection of the murine female genital tract. We examined HPV type 16 (HPV16) as well as HPV31 and HPV5, for which some evidence suggests that they may differ from HPV16 in their utilization of HSPGs as their primary attachment factor in vitro. Luciferase-expressing pseudovirus of all three types infected the mouse genital tract, although HPV5, which normally infects nongenital epidermis, was less efficient. Heparinase III treatment of the genital tract significantly inhibited infection of all three types by greater than 90% and clearly inhibited virion attachment to the basement membrane and cell surfaces, establishing that HSPGs are the primary attachment factors for these three viruses in vivo. However, the pseudoviruses differed in their responses to treatment with various forms of heparin, a soluble analog of heparan sulfate. HPV16 and HPV31 infections were effectively inhibited by a highly sulfated form of heparin, but HPV5 was not, although it bound the compound. In contrast, a N-desulfated and N-acylated variant preferentially inhibited HPV5. Inhibition of infection paralleled the relative ability of the variants to inhibit basement membrane and cell surface binding. We speculate that cutaneous HPVs, such as HPV5, and genital mucosal HPVs, such as HPV16 and -31, may have evolved to recognize different forms of HSPGs to enable them to preferentially infect keratinocytes at different anatomical sites.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Human papillomavirus 16/physiology , Papillomavirus Infections/virology , Vagina/virology , Virus Attachment/drug effects , Animals , Capsid/drug effects , Capsid/physiology , Cells, Cultured , Female , Heparin/pharmacology , Heparitin Sulfate/pharmacology , Human papillomavirus 16/drug effects , Mice , Mice, Inbred BALB C , Polysaccharide-Lyases/pharmacology , Receptors, Virus/metabolism , Vagina/metabolism
9.
Proc Natl Acad Sci U S A ; 105(15): 5850-5, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413606

ABSTRACT

Persistent infection with the high-risk subset of genitotropic human papillomavirus (HPV) genotypes is a necessary cause of cervical cancer. Given the global burden of cervical cancer, a low-cost, broadly protective vaccine is needed. RG-1 is a cross-neutralizing and protective monoclonal antibody that recognizes residues 17-36 of HPV16 minor capsid protein L2. Because this epitope is highly conserved in divergent HPV types, we determined whether vaccination with HPV16 L2 17-36 peptide is broadly protective. The peptide was administered to BALB/c mice three times at monthly intervals, either alone or in the context of a synthetic lipopeptide vaccine candidate (P25-P2C-HPV) produced by linkage of the HPV peptide with a broadly recognized T helper epitope (P25) and the Toll-like receptor-2 (TLR2) ligand dipalmitoyl-S-glyceryl cysteine (P2C). In contrast to vaccination with the L2 17-36 peptide or P25-P2C alone, a potent L2-specific antibody response was generated to the P25-P2C-HPV lipopeptide when delivered either s.c. or intranasally. Sera from mice vaccinated with the P25-P2C-HPV lipopeptide neutralized not only HPV16 pseudovirions but also other evolutionarily divergent oncogenic genital (HPV18, HPV45) and cutaneous (HPV5, BPV1) types. The L2-specific antibody response depended on MHC class II, CD40, and MyD88 signaling. Additionally, vaccination with the P25-P2C-HPV lipopeptide protected mice from homologous challenge with HPV16 pseudovirions at cutaneous and genital sites and heterologous challenge with HPV45 pseudovirions. If provided in the appropriate context, therefore, HPV16 L2 17-36 might be used in a totally synthetic cross-protective HPV vaccine.


Subject(s)
Capsid Proteins/immunology , Cross Reactions/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines/chemistry , Animals , Antibodies, Viral , Antibody Formation , Antigens, Viral , Capsid Proteins/therapeutic use , Chromobox Protein Homolog 5 , Epitopes , Humans , Lipoproteins/therapeutic use , Mice , Neutralization Tests , Oncogene Proteins, Viral/therapeutic use , Papillomavirus Vaccines/immunology , Peptide Fragments/therapeutic use
10.
J Virol ; 81(24): 13927-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17928339

ABSTRACT

We generated a monoclonal antibody, RG-1, that binds to highly conserved L2 residues 17 to 36 and neutralizes human papillomavirus 16 (HPV16) and HPV18. Passive immunotherapy with RG-1 was protective in mice. Antiserum to the HPV16 L2 peptide comprising residues 17 to 36 (peptide 17-36) neutralized pseudoviruses HPV5, HPV6, HPV16, HPV 18, HPV31, HPV 45, HPV 52, HPV 58, bovine papillomavirus 1, and HPV11 native virions. Depletion of HPV16 L2 peptide 17-36-reactive antibodies from cross-neutralizing rabbit and human L2-specific sera abolished cross-neutralization and drastically reduced neutralization of the cognate type. This cross-neutralization of diverse HPVs associated with cervical cancer, genital warts, and epidermodysplasia verruciformis suggests the possibility of a broadly protective, peptide-based vaccine.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Capsid Proteins/immunology , Oncogene Proteins, Viral/immunology , Papillomaviridae/classification , Papillomaviridae/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/administration & dosage , Antibodies, Viral/biosynthesis , Cross Reactions , Human papillomavirus 16/immunology , Human papillomavirus 16/pathogenicity , Humans , Immunization, Passive , Mice , Molecular Sequence Data , Neutralization Tests , Papillomavirus Infections/prevention & control
11.
Nat Med ; 13(7): 857-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17603495

ABSTRACT

Genital human papillomavirus (HPV) infection is the most common sexually transmitted infection, and virtually all cases of cervical cancer are attributable to infection by a subset of HPVs (reviewed in ref. 1). Despite the high incidence of HPV infection and the recent development of a prophylactic vaccine that confers protection against some HPV types, many features of HPV infection are poorly understood. It remains worthwhile to consider other interventions against genital HPVs, particularly those that target infections not prevented by the current vaccine. However, productive papillomavirus infection is species- and tissue-restricted, and traditional models use animal papillomaviruses that infect the skin or oral mucosa. Here we report the development of a mouse model of cervicovaginal infection with HPV16 that recapitulates the establishment phase of papillomavirus infection. Transduction of a reporter gene by an HPV16 pseudovirus was characterized by histology and quantified by whole-organ, multispectral imaging. Disruption of the integrity of the stratified or columnar genital epithelium was required for infection, which occurred after deposition of the virus on the basement membrane underlying basal keratinocytes. A widely used vaginal spermicide, nonoxynol-9 (N-9), greatly increased susceptibility to infection. In contrast, carrageenan, a polysaccharide present in some vaginal lubricants, prevented infection even in the presence of N-9, suggesting that carrageenan might serve as an effective topical HPV microbicide.


Subject(s)
Carrageenan/pharmacology , Human papillomavirus 16/physiology , Nonoxynol/pharmacology , Papillomavirus Infections/transmission , Spermatocidal Agents/pharmacology , Animals , Capsid/physiology , Female , Mice , Mucous Membrane/cytology , Mucous Membrane/virology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Vagina/drug effects , Vagina/virology
12.
PLoS Pathog ; 2(7): e69, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16839203

ABSTRACT

Certain sexually transmitted human papillomavirus (HPV) types are causally associated with the development of cervical cancer. Our recent development of high-titer HPV pseudoviruses has made it possible to perform high-throughput in vitro screens to identify HPV infection inhibitors. Comparison of a variety of compounds revealed that carrageenan, a type of sulfated polysaccharide extracted from red algae, is an extremely potent infection inhibitor for a broad range of sexually transmitted HPVs. Although carrageenan can inhibit herpes simplex viruses and some strains of HIV in vitro, genital HPVs are about a thousand-fold more susceptible, with 50% inhibitory doses in the low ng/ml range. Carrageenan acts primarily by preventing the binding of HPV virions to cells. This finding is consistent with the fact that carrageenan resembles heparan sulfate, an HPV cell-attachment factor. However, carrageenan is three orders of magnitude more potent than heparin, a form of cell-free heparan sulfate that has been regarded as a highly effective model HPV inhibitor. Carrageenan can also block HPV infection through a second, postattachment heparan sulfate-independent effect. Carrageenan is in widespread commercial use as a thickener in a variety of cosmetic and food products, ranging from sexual lubricants to infant feeding formulas. Some of these products block HPV infectivity in vitro, even when diluted a million-fold. Clinical trials are needed to determine whether carrageenan-based products are effective as topical microbicides against genital HPVs.


Subject(s)
Carrageenan/pharmacology , Carrageenan/therapeutic use , Papillomaviridae/pathogenicity , Papillomavirus Infections/drug therapy , Papillomavirus Infections/prevention & control , Administration, Topical , Animals , Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/pharmacology , Anti-Infective Agents, Local/therapeutic use , CHO Cells , Capsid/physiology , Carrageenan/chemistry , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Female , HeLa Cells , Heparin/chemistry , Heparin/physiology , Heparitin Sulfate/chemistry , Heparitin Sulfate/physiology , Humans , Hydrogen-Ion Concentration , Mice , Papillomaviridae/drug effects , Papillomaviridae/physiology , Papillomavirus Infections/physiopathology , Polysaccharides/analysis , Polysaccharides/chemistry , Sexually Transmitted Diseases, Viral/drug therapy , Sexually Transmitted Diseases, Viral/prevention & control , Virion/drug effects , Virion/pathogenicity , Virion/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...