35 results on '"Rebecca L. Sheets"'
Search Results
2. Controlled Human Infection Models To Accelerate Vaccine Development
- Author
-
Robert K. M. Choy, A. Louis Bourgeois, Christian F. Ockenhouse, Richard I. Walker, Rebecca L. Sheets, and Jorge Flores
- Subjects
Microbiology (medical) ,Vaccines ,Infectious Diseases ,Clinical Trials, Phase III as Topic ,General Immunology and Microbiology ,Epidemiology ,Communicable Disease Control ,Vaccine Development ,Public Health, Environmental and Occupational Health ,Humans ,Review ,Models, Biological - Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model’s capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
- Published
- 2022
- Full Text
- View/download PDF
3. Mechanism of ad5 vaccine immunity and toxicity: fiber shaft targeting of dendritic cells.
- Author
-
Cheng Cheng, Jason G D Gall, Wing-pui Kong, Rebecca L Sheets, Phillip L Gomez, C Richter King, and Gary J Nabel
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Recombinant adenoviral (rAd) vectors elicit potent cellular and humoral immune responses and show promise as vaccines for HIV-1, Ebola virus, tuberculosis, malaria, and other infections. These vectors are now widely used and have been generally well tolerated in vaccine and gene therapy clinical trials, with many thousands of people exposed. At the same time, dose-limiting adverse responses have been observed, including transient low-grade fevers and a prior human gene therapy fatality, after systemic high-dose recombinant adenovirus serotype 5 (rAd5) vector administration in a human gene therapy trial. The mechanism responsible for these effects is poorly understood. Here, we define the mechanism by which Ad5 targets immune cells that stimulate adaptive immunity. rAd5 tropism for dendritic cells (DCs) was independent of the coxsackievirus and adenovirus receptor (CAR), its primary receptor or the secondary integrin RGD receptor, and was mediated instead by a heparin-sensitive receptor recognized by a distinct segment of the Ad5 fiber, the shaft. rAd vectors with CAR and RGD mutations did not infect a variety of epithelial and fibroblast cell types but retained their ability to transfect several DC types and stimulated adaptive immune responses in mice. Notably, the pyrogenic response to the administration of rAd5 also localized to the shaft region, suggesting that this interaction elicits both protective immunity and vector-induced fevers. The ability of replication-defective rAd5 viruses to elicit potent immune responses is mediated by a heparin-sensitive receptor that interacts with the Ad5 fiber shaft. Mutant CAR and RGD rAd vectors target several DC and mononuclear subsets and induce both adaptive immunity and toxicity. Understanding of these interactions facilitates the development of vectors that target DCs through alternative receptors that can improve safety while retaining the immunogenicity of rAd vaccines.
- Published
- 2007
- Full Text
- View/download PDF
4. Review of efficacy trials of HIV-1/AIDS vaccines and regulatory lessons learned
- Author
-
TieQun Zhou, Ivana Knezevic, and Rebecca L. Sheets
- Subjects
0301 basic medicine ,medicine.medical_specialty ,education ,Alternative medicine ,Bioengineering ,Context (language use) ,Applied Microbiology and Biotechnology ,World health ,Hiv 1 aids ,03 medical and health sciences ,0302 clinical medicine ,Acquired immunodeficiency syndrome (AIDS) ,Immunology and Microbiology(all) ,Medicine ,030212 general & internal medicine ,Pharmacology ,Medical education ,General Immunology and Microbiology ,business.industry ,Perspective (graphical) ,General Medicine ,AIDS Vaccines ,medicine.disease ,Clinical trial ,030104 developmental biology ,business ,Biotechnology - Abstract
The clinical development of prophylactic HIV-1/AIDS vaccines is confounded by numerous scientific challenges and these in turn result in challenges to regulators reviewing clinical trial applications (CTAs). The search for an HIV-1/AIDS vaccine will only succeed through the conduct of well-designed, well-conducted and well-controlled human efficacy studies. This review summarizes relevant context in which HIV vaccines are being investigated and the six completed efficacy trials of various candidate vaccines and regimens, as well as the lessons learned from them relevant to regulatory evaluation. A companion review focuses on the scientific challenges regulators face and summarizes some current candidates in development. The lessons learned from the completed efficacy trials will enable the development of better designed, potentially more efficient efficacy trials in future. This summary, supported by the World Health Organization (WHO), is unique in that it is meant to aid regulators in understanding the valuable lessons gained from experience in the field to date.
- Published
- 2016
- Full Text
- View/download PDF
5. Now that you want to take your HIV/AIDS vaccine/biological product research concept into the clinic: What are the 'cGMP'?
- Author
-
Rebecca L. Sheets, Vijaya Rangavajhula, Jeffrey K. Pullen, Stuart Z. Shapiro, Vijay L. Mehra, Chris Butler, and Michael Pensiero
- Subjects
AIDS Vaccines ,Vaccine research ,Clinical Trials as Topic ,Medical education ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Public Health, Environmental and Occupational Health ,HIV Infections ,Translational research ,Biological product ,medicine.disease ,Article ,Test (assessment) ,Translational Research, Biomedical ,Product (business) ,Clinical trial ,Infectious Diseases ,Acquired immunodeficiency syndrome (AIDS) ,Government Regulation ,medicine ,Humans ,Molecular Medicine ,business - Abstract
The Division of AIDS Vaccine Research Program funds the discovery and development of HIV/AIDS vaccine candidates. Basic researchers, having discovered a potential vaccine in the laboratory, next want to take that candidate into the clinic to test the concept in humans, to see if it translates. Many of them have heard of "cGMP" and know that they are supposed to make a "GMP product" to take into the clinic, but often they are not very familiar with what "cGMP" means and why these good practices are so important. As members of the Vaccine Translational Research Branch, we frequently get asked "can't we use the material we made in the lab in the clinic?" or "aren't Phase 1 studies exempt from cGMP?" Over the years, we have had many experiences where researchers or their selected contract manufacturing organizations have not applied an appropriate degree of compliance with cGMP suitable for the clinical phase of development. We share some of these experiences and the lessons learned, along with explaining the importance of cGMP, just what cGMP means, and what they can assure, in an effort to de-mystify this subject and facilitate the rapid and safe translational development of HIV vaccines.
- Published
- 2015
- Full Text
- View/download PDF
6. Adventitious agents in viral vaccines: Lessons learned from 4 case studies
- Author
-
Ivana Knezevic, John Petricciani, Rebecca L. Sheets, and Elwyn Griffiths
- Subjects
Circovirus ,education ,Mumps Vaccine ,Adventitious agents ,Bioengineering ,Simian virus 40 ,Biology ,World Health Organization ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Measles ,Viral contamination ,Immunology and Microbiology(all) ,Rotavirus ,medicine ,Animals ,Humans ,Bacteriophages ,Pharmacology ,Biological Products ,Vaccines ,General Immunology and Microbiology ,Regulatory preparedness ,Viral Vaccine ,Rotavirus Vaccines ,RNA-Directed DNA Polymerase ,Viral Vaccines ,General Medicine ,Biological product ,medicine.disease ,biology.organism_classification ,Virology ,Poliomyelitis ,Risk regulation ,Poliovirus Vaccines ,Porcine circovirus ,DNA, Viral ,Immunology ,Public Health ,Drug Contamination ,Measles-Mumps-Rubella Vaccine ,Biotechnology - Abstract
Since the earliest days of biological product manufacture, there have been a number of instances where laboratory studies provided evidence for the presence of adventitious agents in a marketed product. Lessons learned from such events can be used to strengthen regulatory preparedness for the future. We have therefore selected four instances where an adventitious agent, or a signal suggesting the presence of an agent, was found in a viral vaccine, and have developed a case study for each. The four cases are: a) SV40 in polio vaccines; b) bacteriophage in measles and polio vaccines; c) reverse transcriptase in measles and mumps vaccines; and d) porcine circovirus and porcine circovirus DNA sequences in rotavirus vaccines. The lessons learned from each event are discussed. Based in part on those experiences, certain scientific principles have been identified by WHO that should be considered in regulatory risk evaluation if an adventitious agent is found in a marketed vaccine in the future.
- Published
- 2014
- Full Text
- View/download PDF
7. Systematic evaluation of in vitro and in vivo adventitious virus assays for the detection of viral contamination of cell banks and biological products
- Author
-
Nandini Sane, Paula Niksa, Jerald C. Sadoff, Alexander C. Schmidt, Stephen Karakasidis, Valerie B. Randolph, Phillip Minor, James Gombold, Kitti Neumann, Paul Duncan, James A. Richardson, Rebecca L. Sheets, Renita Johnson-Leva, and John Podczasy
- Subjects
Virus quantification ,General Veterinary ,General Immunology and Microbiology ,biology ,viruses ,Viral Vaccine ,Public Health, Environmental and Occupational Health ,Rubella virus ,biology.organism_classification ,medicine.disease_cause ,Virology ,Virus ,Titer ,Infectious Diseases ,Cell culture ,In vivo ,Vesicular stomatitis virus ,medicine ,Molecular Medicine - Abstract
Viral vaccines and the cell substrates used to manufacture them are subjected to tests for adventitious agents, including viruses, contaminate. Some of the compendial methods (in vivo and in vitro in cell culture) were established in the mid-20th century. These methods have not been subjected to current assay validation, as new methods would need to be. This study was undertaken to provide insight into the breadth (selectivity) and sensitivity (limit of detection) of the routine methods, two such validation parameters. Sixteen viral stocks were prepared and characterized. These stocks were tested in serial dilutions by the routine methods to establish which viruses were detected by which methods and above what limit of detection. Sixteen out of sixteen viruses were detected in vitro, though one (bovine viral diarrhea virus) required special conditions to detect and another (rubella virus) was detected with low sensitivity. Many were detected at levels below 1 TCID50 or PFU (titers were established on the production cell line in most cases). In contrast, in vivo, only 6/11 viruses were detected, and 4 of these were detected only at amounts one or more logs above 1 TCID50 or PFU. Only influenza virus and vesicular stomatitis virus were detected at lower amounts in vivo than in vitro. Given the call to reduce, refine, or replace (3Rs) the use of animals in product safety testing and the emergence of new technologies for the detection of viruses, a re-examination of the current adventitious virus testing strategies seems warranted. Suggested pathways forward are offered.
- Published
- 2014
- Full Text
- View/download PDF
8. The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG)
- Author
-
Baevin Carbery, Michael Hendry, Robert T. Chen, Marc Gurwith, Lisa Mac, Richard C. Condit, Stephen J. Seligman, Jean-Louis Excler, Arifa S. Khan, Kenneth I. Berns, Rebecca L. Sheets, Louisa E. Chapman, Anna-Lise Williamson, Najwa Khuri-Bulos, Bettina Klug, and James S. Robertson
- Subjects
Drug-Related Side Effects and Adverse Reactions ,International Cooperation ,viruses ,Genetic Vectors ,Article ,Viral vector ,Immunology and Microbiology(all) ,Viral Vector ,Humans ,Medicine ,Public acceptance ,Vaccines ,Clinical Trials as Topic ,Drug Carriers ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Viral Vaccine ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,Virology ,veterinary(all) ,Clinical trial ,Infectious Diseases ,Immunization ,Molecular Medicine ,Safety ,business - Abstract
Recombinant viral vectors provide an effective means for heterologous antigen expression in vivo and thus represent promising platforms for developing novel vaccines against human pathogens from Ebola to tuberculosis. An increasing number of candidate viral vector vaccines are entering human clinical trials. The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to improve our ability to anticipate potential safety issues and meaningfully assess or interpret safety data, thereby facilitating greater public acceptance when licensed.
- Published
- 2015
- Full Text
- View/download PDF
9. Opinion on adventitious agents testing for vaccines: Why do we worry so much about adventitious agents in vaccines?
- Author
-
Rebecca L. Sheets
- Subjects
General Veterinary ,General Immunology and Microbiology ,Inclusion (disability rights) ,business.industry ,media_common.quotation_subject ,Internet privacy ,Drug Evaluation, Preclinical ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,Context (language use) ,Microbial contamination ,Risk Assessment ,Infectious Diseases ,Immunology ,Animals ,Humans ,Molecular Medicine ,Medicine ,Worry ,Drug Contamination ,Viral contamination ,business ,media_common - Abstract
The manner in which viral vaccines are produced in a biological system makes them vulnerable to microbial contamination. Considerable effort is expended to avoid such contamination and to detect it if it occurred. Is this effort warranted, efficient, scientifically sound, and rational? When asked for my opinion on these matters, I agreed to discuss the basis and historical context for why we do what we do and proffer opinion on what we might do instead or in addition, as we look forward to the inclusion of new strategies and methods in our arsenal. Being an advocate of the 3 R's policy, I invite a re-examination of the traditional in vivo methods in particular. I also advocate for a risk-based approach consistent with "Quality by Design" as a more scientific and rational means of addressing these issues. In the end, vaccinologists need to reassure the public that the vaccines they or their children receive are safe and pure and that all reasonable measures are taken to safeguard them.
- Published
- 2013
- Full Text
- View/download PDF
10. Unique Safety Issues Associated with Virus Vectored Vaccines: Potential for and Theoretical Consequences of Recombination with Wild Type Virus Strains
- Author
-
Richard C. Condit, James S. Robertson, Thomas P. Monath, Vidisha Singh, Marc Gurwith, Rebecca L. Sheets, Robert T. Chen, Denny Kim, Jean-Louis Excler, Lisa M. Mac, Stephen J. Seligman, R. Michael Hendry, Anna-Lise Williamson, and Karin Bok
- Subjects
0301 basic medicine ,viruses ,Genetic Vectors ,Virulence ,Context (language use) ,Biology ,Vaccines, Attenuated ,Virus ,Article ,03 medical and health sciences ,Animals ,Humans ,Vector (molecular biology) ,Recombination, Genetic ,Drug Carriers ,Attenuated vaccine ,General Immunology and Microbiology ,General Veterinary ,Transmission (medicine) ,Viral Vaccine ,Wild type ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,Virology ,030104 developmental biology ,Infectious Diseases ,Viruses ,Molecular Medicine - Abstract
In 2003 and 2013, the World Health Organization convened informal consultations on characterization and quality aspects of vaccines based on live virus vectors. In the resulting reports, one of several issues raised for future study was the potential for recombination of virus-vectored vaccines with wild type pathogenic virus strains. This paper presents an assessment of this issue formulated by the Brighton Collaboration. To provide an appropriate context for understanding the potential for recombination of virus-vectored vaccines, we review briefly the current status of virus-vectored vaccines, mechanisms of recombination between viruses, experience with recombination involving live attenuated vaccines in the field, and concerns raised previously in the literature regarding recombination of virus-vectored vaccines with wild type virus strains. We then present a discussion of the major variables that could influence recombination between a virus-vectored vaccine and circulating wild type virus and the consequences of such recombination, including intrinsic recombination properties of the parent virus used as a vector; sequence relatedness of vector and wild virus; virus host range, pathogenesis and transmission; replication competency of vector in target host; mechanism of vector attenuation; additional factors potentially affecting virulence; and circulation of multiple recombinant vectors in the same target population. Finally, we present some guiding principles for vector design and testing intended to anticipate and mitigate the potential for and consequences of recombination of virus-vectored vaccines with wild type pathogenic virus strains.
- Published
- 2016
11. Adventitious agents, new technology, and risk assessment, 19–20 May 2011, Baltimore, MD
- Author
-
Gopa Raychaudhuri, Rebecca L. Sheets, Johannes Loewer, and John C. Petricciani
- Subjects
Pharmacology ,General Immunology and Microbiology ,Standardization ,Risk analysis (engineering) ,Computer science ,Bioengineering ,General Medicine ,Microbial contamination ,Risk assessment ,Applied Microbiology and Biotechnology ,Biotechnology ,Variety (cybernetics) - Abstract
In May 2011, the International Alliance for Biological Standardization, with the cooperation of WHO, FDA, and NIAID, organized a conference on adventitious agents that might be found in biological products using new technology (http://www.iabs.org/index.php/past-conference-reports/116-baltimore-2011-slides). The implications of such findings on risk assessment also were considered. Topics that were addressed included: a) current routine testing--what are we doing now?; b) recent advances in testing--what tests are being explored/applied?; c) examples of finding agents with "new" techniques; and d) risk assessment, including recent WHO activities. A draft algorithm for risk assessment was discussed in terms of its applicability to a variety of potential new agents and the possibilities for improving it.
- Published
- 2012
- Full Text
- View/download PDF
12. Evaluation of the human host range of bovine and porcine viruses that may contaminate bovine serum and porcine trypsin used in the manufacture of biological products
- Author
-
James C. Richardson, Nandini Sane, Rebecca K. Harston, Rebecca L. Sheets, and Carol J. Marcus-Sekura
- Subjects
Serum ,Porcine parvovirus ,Swine ,Bioengineering ,Virus Physiological Phenomena ,Immunofluorescence ,Applied Microbiology and Biotechnology ,Article ,Host Specificity ,Porcine Trypsin ,medicine ,Animals ,Humans ,Trypsin ,Bovine serum albumin ,Virus classification ,Pharmacology ,Biological Products ,General Immunology and Microbiology ,biology ,medicine.diagnostic_test ,Potential risk ,General Medicine ,biology.organism_classification ,Virology ,Viruses ,biology.protein ,Cattle ,Viral contamination ,Drug Contamination ,Biotechnology - Abstract
Current U.S. requirements for testing cell substrates used in production of human biological products for contamination with bovine and porcine viruses are U.S. Department of Agriculture (USDA) 9CFR tests for bovine serum or porcine trypsin. 9CFR requires testing of bovine serum for seven specific viruses in six families (immunofluorescence) and at least 2 additional families non-specifically (cytopathicity and hemadsorption). 9CFR testing of porcine trypsin is for porcine parvovirus. Recent contaminations suggest these tests may not be sufficient. Assay sensitivity was not the issue for these contaminations that were caused by viruses/virus families not represented in the 9CFR screen. A detailed literature search was undertaken to determine which viruses that infect cattle or swine or bovine or porcine cells in culture also have human host range [ability to infect humans or human cells in culture] and to predict their detection by the currently used 9CFR procedures. There are more viruses of potential risk to biological products manufactured using bovine or porcine raw materials than are likely to be detected by 9CFR testing procedures; even within families, not all members would necessarily be detected. Testing gaps and alternative methodologies should be evaluated to continue to ensure safe, high quality human biologicals.
- Published
- 2011
- Full Text
- View/download PDF
13. Evaluation of cell substrates for the production of biologicals: Revision of WHO recommendations
- Author
-
Glyn Stacey, John Petricciani, Ivana Knezevic, and Rebecca L. Sheets
- Subjects
Pharmacology ,General Immunology and Microbiology ,Standardization ,business.industry ,International standard ,Guidance documents ,Bioengineering ,General Medicine ,Nucleic acid amplification technique ,Applied Microbiology and Biotechnology ,Expert committee ,Engineering management ,Who recommendations ,Immunology ,Medicine ,Production (economics) ,business ,Cell bank ,Biotechnology - Abstract
Evaluating cell substrates for producing vaccines and other biologicals is one of the critical aspects in assuring quality and safety of these products. As part of its mission in setting standards for biological products, WHO provides recommendations for manufacturing and evaluating biologicals. Regular updates of the guidance documents are important to manufacturers and regulators worldwide. WHO Expert Committee on Biological Standardization (ECBS) identified a need for revising the requirements for cell substrates (WHO TRS 878, annex 1). In response, WHO established a Study Group (SG) in 2006 that prepared an updated set of recommendations for using cell substrates for the production of biologicals. A summary of the proposed changes that the SG made in 2007 is available at WHO web site (http://www.who.int/biologicals/publications/meetings/areas/vaccines/cells/en/index.html). Draft revised recommendations were circulated to regulators, manufacturers and other experts for comments in April 2009. The SG held its third meeting on 22-23 April 2009 to review progress in the revision and to propose further improvements. In addition, the experts discussed the need for reference preparations, reference cell banks, and standardization of testing methodologies. The SG proposed clarifications of the rationale for in vivo testing as well as the potential for applying new methods for in vitro testing for detecting microbial agents. In line with this, WHO should conduct review of the current manufacturers' practice in using tests for microbial agents and interpreting these results. Additionally, WHO should take a lead in developing an International Standard for nucleic acid amplification test (NAT) for detecting mycoplasma contamination in cell substrates. WHO Collaborating Centers will lead this initiative, involving other relevant institutions in this area. Finally, advice on the replacement of the WHO Vero reference cell bank 10-87 with respect to the source of cells and re-characterization of the bank was provided. The intended use of the replacement cell bank would be the same as for the current cell bank, which is to serve as a source of well-characterized cells for establishing master cell banks for the production of biologicals. The SG will report outcomes of its discussion to the ECBS at its next meeting in October 2009 for further considerations and advice regarding the proposed course of action.
- Published
- 2010
- Full Text
- View/download PDF
14. An overview of animal cell substrates for biological products
- Author
-
Rebecca L. Sheets and John Petricciani
- Subjects
Mammals ,Pharmacology ,Biological Products ,General Immunology and Microbiology ,Cell substrate ,Cell ,Cell Culture Techniques ,Bioengineering ,Nanotechnology ,General Medicine ,Computational biology ,Oncogenicity ,Biology ,Applied Microbiology and Biotechnology ,Cell Line ,Culture Media ,Phenotype ,medicine.anatomical_structure ,Cellular dna ,medicine ,Animals ,Humans ,Cells, Cultured ,HeLa Cells ,Biotechnology - Abstract
The issue of which cells to use as substrates for the production of biological products, and especially vaccines, has been with us in one form or another ever since the development of cell cultures in the 1950s. The major cell substrate events that occurred over the past 50 years are reviewed briefly. Although numerous conferences were held during that period, incomplete resolution of some cell substrate issues has remained. Specifically, the potential oncogenicity of cellular DNA derived from continuous cell lines, and the tests that are used to rule out the presence of adventitious agents have been recognized as areas that could benefit greatly from studies using state-of-the-art techniques. A collaborative effort involving WHO, NIAID, and IABS resulted from consensus recommendations of a 2004 conference, and the prospects for revised guidance in the near future on the characterization and use of animal cell substrates are bright.
- Published
- 2008
- Full Text
- View/download PDF
15. Biodistribution and Toxicological Safety of Adenovirus Type 5 and Type 35 Vectored Vaccines Against Human Immunodeficiency Virus-1 (HIV-1), Ebola, or Marburg Are Similar Despite Differing Adenovirus Serotype Vector, Manufacturer's Construct, or Gene Inserts
- Author
-
Chris Duffy, Bin He, Edward Koo, Robert T. Bailer, Holly Trotter, Richard A. Koup, T. Manetz, Martha Nason, Rebecca L. Sheets, Judith Stein, Charla Andrews, and Phillip Gomez
- Subjects
Male ,Time Factors ,Genetic Vectors ,Immunology ,Drug Evaluation, Preclinical ,HIV Infections ,Antibodies, Viral ,Toxicology ,medicine.disease_cause ,Injections, Intramuscular ,Polymerase Chain Reaction ,Article ,Adenoviridae ,Viral vector ,Marburg virus disease ,Vaccines, DNA ,medicine ,Animals ,Humans ,Marburg Virus Disease ,Tissue Distribution ,Vector (molecular biology) ,Ebola Vaccines ,Serotyping ,AIDS Vaccines ,Reactogenicity ,Ebola vaccine ,biology ,Viral Vaccine ,Viral Vaccines ,Hemorrhagic Fever, Ebola ,Ebolavirus ,Marburgvirus ,biology.organism_classification ,Virology ,DNA, Viral ,HIV-1 ,Female ,Rabbits - Abstract
The Vaccine Research Center has developed vaccine candidates for different diseases/infectious agents (including HIV-1, Ebola, and Marburg viruses) built on an adenovirus vector platform, based on adenovirus type 5 or 35. To support clinical development of each vaccine candidate, pre-clinical studies were performed in rabbits to determine where in the body they biodistribute and how rapidly they clear, and to screen for potential toxicities (intrinsic and immunotoxicities). The vaccines biodistribute only to spleen, liver (Ad5 only), and/or iliac lymph node (Ad35 only) and otherwise remain in the site of injection muscle and overlying subcutis. Though approximately 10(11) viral particles were inoculated, already by Day 9, all but 10(3) to 10(5) genome copies per mu g of DNA had cleared from the injection site muscle. By three months, the adenovector was cleared with, at most, a few animals retaining a small number of copies in the injection site, spleen (Ad5), or iliac lymph node (Ad35). This pattern of limited biodistribution and extensive clearance is consistent regardless of differences in adenovector type (Ad5 or 35), manufacturer's construct and production methods, or gene-insert. Repeated dose toxicology studies identified treatment-related toxicities confined primarily to the sites of injection, in certain clinical pathology parameters, and in body temperatures (Ad5 vectors) and food consumption immediately post-inoculation. Systemic reactogenicity and reactogenicity at the sites of injection demonstrated reversibility. These data demonstrate the safety and suitability for investigational human use of Ad5 or Ad35 adenovector-based vaccine candidates at doses of up to 2 x 10(11) given intramuscularly to prevent various infectious diseases.
- Published
- 2008
- Full Text
- View/download PDF
16. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Vesicular Stomatitis Virus Human Immunodeficiency Virus-1 gag Vaccine (HVTN 090)
- Author
-
Victoria Chinnell, Gail Broder, Theresa E. Latham, Nadine Rouphael, Mark J. Mulligan, Spyros A. Kalams, Jonathan D. Fuchs, Rebecca L. Sheets, Ian Frank, Shelly Ramirez, Scharla Estep, Sue Li, Theresa Wagner, Reese Isbell, Gina Escamilla, Srilatha Edupuganti, David K. Clarke, Michael A. Egan, Terry J. Higgins, Nidhi Kochar, Ramey Fair, Deb Dunbar, John H. Eldridge, Nicole Frahm, Georgia D. Tomaras, Jenny Tseng, Kyle Rybczyk, Adi Ferrara, Mary Allen, Marc Tremblay, Susan Buchbinder, Michael Pensiero, Jin Bae, Liz Briesemeister, and Marnie Elizaga
- Subjects
safety ,Canarypox ,HIV vaccine ,biology ,viruses ,dose-escalation ,Vesicular stomatitis Indiana virus ,immunogenicity ,biology.organism_classification ,Virology ,Virus ,3. Good health ,Viral vector ,Major Articles ,chemistry.chemical_compound ,Infectious Diseases ,Oncology ,chemistry ,Vesicular stomatitis virus ,Vaccinia ,vesicular stomatitis virus ,Viral load - Abstract
Attenuated, replication-competent viral vector vaccines are highly immunogenic in that they mimic natural infection. They offer the promise of eliciting a more integrated immune response and more abundant and sustained expression of the encoded viral antigens. Although a vaccine to prevent human immunodeficiency virus (HIV) infection remains an urgent global health priority, concerns about the safety of using live-attenuated HIV itself as a vaccine [1] has led the field to focus heavily on the exploration of replication-defective viral vectors to deliver HIV-1 antigens. There are limited data to suggest these vectors are able to induce an optimal, integrated immune response, which may include CD4+ T-cell help for B-cell/antibody and polyfunctional cytotoxic T-cell responses together with long-term immunologic memory [2]. To date, only 1 replication-defective viral vector, based on canarypox, administered sequentially with a gp120 subunit boost vaccine, has demonstrated partial protection from HIV acquisition in an efficacy trial [3]. Currently, only a few replicating viruses, including vaccinia (clinical trial ID NCT01705223), Sendai virus (NCT01705990), measles (NCT01320176), and adenovirus subtype 4 (NCT01989533), have advanced to clinical trials as HIV-1 vaccine vectors. Vesicular stomatitis virus (VSV) is a member of the genus Vesiculovirus in the family Rhabdoviridae. The 2 major serotypes are New Jersey and Indiana. Vesicular stomatitis virus has a single-strand, negative-sense, nonsegmented RNA genome. Similar to Sendai and measles viruses, the virus genomic organization of VSV (Figure (Figure1A)1A) and life cycle make it particularly attractive as a candidate vaccine vector because the genome can accommodate multiple gene inserts, is stable over many generations, and does not undergo recombination. In addition, the VSV genome replicates in the cytoplasm and is incapable of integrating within the genomes of infected host cells. Finally, VSV is a zoonosis, cycling between biting insects and livestock (cattle, horses, and swine). Human infection with VSV is rare in most regions of the world except in certain regions of Central and South America where VSV is endemic. Where VSV infection does occur, it is typically asymptomatic or is associated with an acute influenza-like illness with symptoms such as fever, muscle aches, and malaise [4, 5]. Thus, the general population is largely free of pre-existing, virus-neutralizing immunity—a factor that has limited the clinical utility of HIV-1 vaccines based on adenovirus subtype 5 [6, 7]. Figure 1. (A) Wild-type vesicular stomatitis virus (VSV) genome organization and virion structure. Viral transcription is polar, initiating at the single 3′ promoter and proceeding to the 5′ end of the genome, producing a steep 3′ to 5′ ... Recombinant VSV (rVSV) has been extensively studied as a potential HIV vaccine vector in preclinical, nonhuman primate (NHP) models [8–11]. After 2 sequential immunizations, rhesus macaques that received rVSV expressing HIV-1 env and simian immunodeficiency virus (SIV) gag were all infected but were protected from disease progression for 4 years or more after infection with SHIV 89.6P, maintaining low or undetectable viral load set points and preserved CD4+ T-cell counts [11]. This encouraging level of postchallenge protection from disease suggested that rVSV vectors expressing HIV genes might be an effective HIV vaccine in humans. To maximize safety for first-in-human clinical evaluation, a highly attenuated form of rVSV Indiana was developed. This involved downregulating VSV N protein expression by translocating the N gene further away from the 3′ transcription promoter and truncating the VSV G protein cytoplasmic tail from 29 amino acids found in wild-type virus to a single amino acid [12]. In addition, HIV-1 gag expression was achieved by insertion of the gag gene adjacent to the 3′ transcription promoter (Figure (Figure1B).1B). The resulting vector (rVSVN4CT1gag1) was shown to be safe, well tolerated, and immunogenic in several murine and NHP studies [13, 14]. In the work described here, the HIV Vaccine Trials Network (HVTN) evaluated the safety and immunogenicity of this novel vaccine vector in a phase 1a dose-escalation, first-in-human trial, HVTN 090.
- Published
- 2015
17. Review of efficacy trials of HIV-1/AIDS vaccines and regulatory lessons learned: A review from a regulatory perspective
- Author
-
Rebecca L, Sheets, TieQun, Zhou, and Ivana, Knezevic
- Subjects
AIDS Vaccines ,Acquired Immunodeficiency Syndrome ,Clinical Trials as Topic ,Humans - Abstract
The clinical development of prophylactic HIV-1/AIDS vaccines is confounded by numerous scientific challenges and these in turn result in challenges to regulators reviewing clinical trial applications (CTAs). The search for an HIV-1/AIDS vaccine will only succeed through the conduct of well-designed, well-conducted and well-controlled human efficacy studies. This review summarizes relevant context in which HIV vaccines are being investigated and the six completed efficacy trials of various candidate vaccines and regimens, as well as the lessons learned from them relevant to regulatory evaluation. A companion review focuses on the scientific challenges regulators face and summarizes some current candidates in development. The lessons learned from the completed efficacy trials will enable the development of better designed, potentially more efficient efficacy trials in future. This summary, supported by the World Health Organization (WHO), is unique in that it is meant to aid regulators in understanding the valuable lessons gained from experience in the field to date.
- Published
- 2015
18. Manufacturing and Testing of a Multi-Clade Adenoviral Vector-Based Candidate Vaccine Against Human Immunodeficiency Virus
- Author
-
Bryan T. Butman, Douglas E. Brough, Rebecca L. Sheets, Phil Gomez, Jason G. Gall, Alena Lizonova, Perry Newton, J. Sowers, and Victoria Haque
- Subjects
Human immunodeficiency virus (HIV) ,medicine ,General Earth and Planetary Sciences ,Biology ,Clade ,medicine.disease_cause ,Virology ,General Environmental Science ,Viral vector - Published
- 2006
- Full Text
- View/download PDF
19. Assaying Potency of Novel Vaccines
- Author
-
Rebecca L. Sheets
- Subjects
Pharmacology ,Gerontology ,business.industry ,Drug Discovery ,Immunology ,Molecular Medicine ,Potency ,Medicine ,business - Published
- 2006
- Full Text
- View/download PDF
20. Biodistribution of DNA Plasmid Vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile Virus Is Similar, without Integration, despite Differing Plasmid Backbones or Gene Inserts
- Author
-
Rebecca L. Sheets, Charla A. Andrews, Phillip L. Gomez, Wing Pui Kong, T. Scott Manetz, Martha Nason, Gary J. Nabel, Chris Duffy, and Judith A Stein
- Subjects
Male ,Immunogen ,Genes, Viral ,medicine.medical_treatment ,Mice, Inbred Strains ,Biology ,Toxicology ,medicine.disease_cause ,Severe Acute Respiratory Syndrome ,Injections, Intramuscular ,Article ,DNA vaccination ,Mice ,Plasmid ,medicine ,Vaccines, DNA ,Animals ,Tissue Distribution ,Promoter Regions, Genetic ,Gene ,Acquired Immunodeficiency Syndrome ,Ebola virus ,Viral Vaccine ,Viral Vaccines ,Vector vaccine ,Hemorrhagic Fever, Ebola ,Ebolavirus ,Virology ,Injections, Intravenous ,HIV-1 ,Female ,Adjuvant ,West Nile virus ,West Nile Fever ,Plasmids - Abstract
The Vaccine Research Center has developed a number of vaccine candidates for different diseases/infectious agents (HIV-1, Severe Acute Respiratory Syndrome virus, West Nile virus, and Ebola virus, plus a plasmid cytokine adjuvant-IL-2/Ig) based on a DNA plasmid vaccine platform. To support the clinical development of each of these vaccine candidates, preclinical studies have been performed in mice or rabbits to determine where in the body these plasmid vaccines would biodistribute and how rapidly they would clear. In the course of these studies, it has been observed that regardless of the gene insert (expressing the vaccine immunogen or cytokine adjuvant) and regardless of the promoter used to drive expression of the gene insert in the plasmid backbone, the plasmid vaccines do not biodistribute widely and remain essentially in the site of injection, in the muscle and overlying subcutis. Even though approximately 10(14) molecules are inoculated in the studies in rabbits, by day 8 or 9 ( approximately 1 week postinoculation), already all but on the order of 10(4)-10(6) molecules per microgram of DNA extracted from tissue have been cleared at the injection site. Over the course of 2 months, the plasmid clears from the site of injection with only a small percentage of animals (generally 10-20%) retaining a small number of copies (generally around 100 copies) in the muscle at the injection site. This pattern of biodistribution (confined to the injection site) and clearance (within 2 months) is consistent regardless of differences in the promoter in the plasmid backbone or differences in the gene insert being expressed by the plasmid vaccine. In addition, integration has not been observed with plasmid vaccine candidates inoculated i.m. by Biojector 2000 or by needle and syringe. These data build on the repeated-dose toxicology studies performed (see companion article, Sheets et al., 2006) to demonstrate the safety and suitability for investigational human use of DNA plasmid vaccine candidates for a variety of infectious disease prevention indications.
- Published
- 2006
- Full Text
- View/download PDF
21. Current advances and challenges in HIV-1 vaccines
- Author
-
Rebecca L. Sheets, Rodriguez-Chavez Ir, D'Souza Mp, Bradac Ja, Hill El, Mary Allen, and Michael Pensiero
- Subjects
AIDS Vaccines ,Cellular immunity ,medicine.medical_specialty ,Human immunodeficiency virus (HIV) ,HIV Infections ,Biology ,Clinical disease ,medicine.disease_cause ,Structure and function ,Clinical trial ,Infectious Diseases ,Immune system ,HLA Antigens ,Allergy and Immunology ,Virology ,Medicine public health ,DNA, Viral ,Immunology ,HIV-1 ,medicine ,Humans ,Intensive care medicine ,Mucosal immunity ,Plasmids - Abstract
Recent advances in science, which have aided HIV-1 vaccine development, include an improved understanding of HIV-1 envelope structure and function, expansion of the pipeline with innovative vaccine strategies, promising multi-gene and multi-clade vaccines that elicit cellular immunity, conduct of clinical trials in a global network, and development of validated techniques that enable simultaneous measurement of multiple T cell vaccine-induced immune responses in humans. A common feature of several preventive vaccine strategies now in early clinical trials is their ability in nonhuman primates to attenuate clinical disease rather than completely prevent HIV-1 infection. One vaccine concept has been tested in large-scale clinical trials, two are currently in efficacy trials, and one more is poised to enter efficacy trial in the next few years. Simultaneously, expanded efforts continue to identify new designs that induce mucosal immunity as well as broadly neutralizing antibodies.
- Published
- 2006
- Full Text
- View/download PDF
22. Current advances in HIV vaccines
- Author
-
Mary Allen, Margaret I. Johnston, M. Patricia D'Souza, and Rebecca L. Sheets
- Subjects
AIDS Vaccines ,medicine.medical_specialty ,Human immunodeficiency virus (HIV) ,HIV Infections ,Biology ,medicine.disease_cause ,Vaccine efficacy ,Clinical disease ,Clinical trial ,Infectious Diseases ,Virology ,Medicine public health ,Immunology ,HIV-1 ,medicine ,Humans ,HIV vaccine ,Intensive care medicine ,Hiv envelope - Abstract
Development of a safe and preventive HIV-1 vaccine is a high priority. Recent advances in HIV vaccine development include an improved understanding of HIV envelope structure, development of techniques that enable a detailed analysis of vaccine-induced immune responses in humans, expansion of the pipeline of promising candidate vaccines, and completion of the first vaccine efficacy trials. A common feature of several preventive vaccine strategies in early clinical trials is their ability to attenuate clinical disease rather than completely prevent HIV infection in nonhuman primates. One or more candidate vaccines will likely advance into efficacy trials within the next few years, while efforts to identify new designs that induce broadly neutralizing antibodies continue with incremental success.
- Published
- 2004
- Full Text
- View/download PDF
23. Role of Analytics in Viral Safety
- Author
-
Paul Duncan and Rebecca L. Sheets
- Subjects
Computer science ,Process (engineering) ,business.industry ,media_common.quotation_subject ,Context (language use) ,Process validation ,Biosafety ,Risk analysis (engineering) ,Analytics ,Quality (business) ,Product (category theory) ,business ,Risk assessment ,media_common - Abstract
In summary, this chapter reviews the principles of how the current and routine tests detect adventitious agents, and reviews how novel and emerging methods differ in their detection principles. These facets may permit novel methods to emerge to supplement, refine, or replace the routine methods. We have suggested a framework for risk assessment to assure biosafety in vaccines and suggested quantitative modeling to help crystallize thinking about the place of testing, either routine or novel, in this assurance. We assert that testing for adventitious agents should not be the sole basis on which product biosafety is assured. Appropriate sourcing and quality control of raw and starting materials, adherence to principles of Good Manufacturing Practices, including environmental and personnel monitoring and process validation, and finally, testing as verification are the package needed for maximal assurance of biosafety. Thus, a pathway forward to a new paradigm for adventitious agent testing exists in which detection of a broader array of potential adventitious agents might be included in the testing, with adequate sensitivity to provide the needed assurance of verification that there has been no catastrophic breach, in the context of the overall process, design, and adherence to cGMP. Furthermore, it is our hope that we may be able to implement the 3 Rs policy to reduce, replace, and/or refine the use of animals in product safety testing, at the same time that we provide greater assurance of the biosafety of vaccines.
- Published
- 2014
- Full Text
- View/download PDF
24. Workshop Report: Traditional Approach Preventive HIV Vaccines: What Are the Cell Substrate and Inactivation Issues?1
- Author
-
Rebecca L. Sheets and Karen L. Goldenthal
- Subjects
Infectivity ,Immunology ,Cell ,Biology ,Virology ,Peripheral blood mononuclear cell ,Virus ,law.invention ,chemistry.chemical_compound ,Titer ,Infectious Diseases ,medicine.anatomical_structure ,chemistry ,law ,Cell culture ,Recombinant DNA ,medicine ,DNA - Abstract
A workshop was convened to discuss safety issues for traditional-approach HIV vaccines, especially inactivated vaccines. The topics included issues pertaining to (1) cell substrates used for production and (2) vaccine virus inactivation. The use of cell substrates such as tumor-derived continuous cell lines (TCLs) or virus-transformed. CLs may be the most feasible approach to provide commercial-scale virus yields. However, especially because of concerns about tumorigenicity, TCLs have not been used to produce preventive vaccines for human trials with healthy subjects in the United States. Residual TCL material (e.g., DNA, cellular proteins, viruses) may not be removed during purification of intact HIV virions to the same extent achievable for a recombinant protein. Manufacturing processes, e.g., physicochemical methods of destroying DNA, could decrease tumorigenicity risk. Methods to assess potential for tumorigenicity may need further development. Another potential substrate for viral production that merits further study is human peripheral blood mononuclear cells (PBMCs). Regardless of the cell substrate used, extensive testing for adventitious agents (including non-HIV retroviruses) is needed. Vaccine virus inactivation can be considered in statistical terms, i.e., the probability of a surviving infectious particle. One formula to determine a "safety margin" (SM) is reduction of titer in log10 for all inactivation steps minus initial viral infectivity in log10. Calculations for appropriate SMs should include all sources of variability (e.g., lot-to-lot differences). Ensuring a specified SM, as the lower bound of the 95% confidence interval, for production lots was discussed. Sensitivity and specificity of infectivity assays may present limitations.
- Published
- 1998
- Full Text
- View/download PDF
25. Recombinant feline leukemia virus genes detected in naturally occurring feline lymphosarcomas
- Author
-
Pradip Roy-Burman, Rebecca L. Sheets, Wen-Cheng Jen, and R. Pandey
- Subjects
animal diseases ,viruses ,Molecular Sequence Data ,Immunology ,Biology ,Recombinant virus ,Genes, env ,Polymerase Chain Reaction ,Microbiology ,Feline leukemia virus ,Virus ,law.invention ,Proviruses ,Viral envelope ,law ,Sequence Homology, Nucleic Acid ,hemic and lymphatic diseases ,Virology ,Animals ,Amino Acid Sequence ,Cloning, Molecular ,Gene ,Recombination, Genetic ,Leukemia, Experimental ,Base Sequence ,Sequence Homology, Amino Acid ,Leukemia Virus, Feline ,Lymphoma, Non-Hodgkin ,virus diseases ,Provirus ,biology.organism_classification ,Molecular biology ,Capsid ,Insect Science ,Cats ,Recombinant DNA ,Research Article - Abstract
Using a polymerase chain reaction strategy aimed at detecting recombinant feline leukemia virus (FeLV) genomes with 5' env sequences originating from an endogenous source and 3' env sequences resulting from FeLV subgroup A (FeLV-A), we detected recombinant proviruses in approximately three-fourths of naturally occurring thymic and alimentary feline lymphosarcomas (LSAs) and one-third of the multicentric LSAs from cats determined to be FeLV capsid antigen positive by immunofluorescence assay. In contrast, only 1 of 22 naturally arising FeLV-negative feline LSAs contained recombinant proviruses, and no recombinant env gene was detected in seven samples from normal tissues or tissues from FeLV-positive animals that died from other diseases. Four preferred structural motifs were identified in the recombinants; one is FeLV-B like (recognizing that FeLV-B itself is a product of recombination between FeLV-A and endogenous env genes), and three contain variable amounts of endogenous-like env gene before crossing over to FeLV-A-related sequences: (i) a combination of full-length and deleted env genes with recombination at sites in the middle of the surface glycoprotein (SU), (ii) the entire SU encoded by endogenous-like sequences, and (iii) the entire SU and approximately half of the transmembrane protein encoded by endogenous-like sequences. Additionally, three of the thymic tumors contained recombinant proviruses with mutations in the vicinity of the major neutralizing determinant for the SU protein. These molecular genetic analyses of the LSA DNAs correspond to our previous results in vitro and support the occurrence and association of viral recombinants and mutants in vivo in FeLV-induced leukemogenesis.
- Published
- 1993
- Full Text
- View/download PDF
26. Mode of action of adjuvants: implications for vaccine safety and design
- Author
-
Claire-Anne Siegrist, Hana Golding, Ian D. Gust, William M. Egan, Giuseppe Del Giudice, Pieter Neels, Steven G. Reed, Béatris Mastelic, Paul-Henri Lambert, S. Sohail Ahmed, and Rebecca L. Sheets
- Subjects
Vaccine safety ,Adjuvants, Immunologic/*adverse effects/*pharmacology ,Drug Compounding/methods ,medicine.medical_treatment ,Influenza, Human/epidemiology/prevention & control/therapy ,Human immunodeficiency virus (HIV) ,Bioengineering ,ddc:616.07 ,medicine.disease_cause ,Public Health/methods/trends ,Applied Microbiology and Biotechnology ,Vaccine adjuvant ,medicine ,Communicable Diseases/therapy ,Animals ,Humans ,Mode of action ,Pandemics ,Pharmacology ,Drug compounding ,ddc:618 ,General Immunology and Microbiology ,Maryland ,business.industry ,General Medicine ,United States ,Action (philosophy) ,Influenza A Virus, H1N1 Subtype/immunology ,Drug Design ,Immunology ,Mass Vaccination/adverse effects ,Engineering ethics ,Mass vaccination ,Vaccines/*adverse effects ,Safety ,business ,Adjuvant ,Chemistry, Pharmaceutical/methods/trends ,Biotechnology - Abstract
For decades, the search for new vaccine adjuvants has been largely empirical. A series of new adjuvants and related formulations are now emerging that are acting through identified immunological mechanisms. Understanding adjuvant mechanism of action is crucial for vaccine design, since this allows for directing immune responses towards efficacious disease-specific effector mechanisms and appropriate memory. It is also of great importance to build new paradigms for assessing adjuvant safety at development stages and at regulatory level. This report reflects the conclusions of a group of scientists from academia, regulatory agencies and industry who attended a conference, organized by the International Association for Biologicals (IABS), on the mode of action of adjuvants on 29-30 April 2010 in Bethesda, Maryland, USA, particularly focusing on how understanding adjuvants mode of action can impact on the assessment of vaccine safety and help to develop target-specific vaccines. More information on the conference output can be found on the IABS website, http://www.iabs.org/.
- Published
- 2010
27. Biologically selected recombinants between Feline Leukemia Virus (FeLV) subgroup a and an endogenous FeLV element
- Author
-
Vaclav Klement, Rebecca L. Sheets, Chris K. Grant, Pradip Roy-Burman, and R. Pandey
- Subjects
animal diseases ,viruses ,Molecular Sequence Data ,Enzyme-Linked Immunosorbent Assay ,Biology ,Recombinant virus ,Polymerase Chain Reaction ,Feline leukemia virus ,Virus ,Epitope ,Gene product ,Epitopes ,Plasmid ,Proviruses ,Viral envelope ,hemic and lymphatic diseases ,Virology ,Tumor Cells, Cultured ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Tropism ,Recombination, Genetic ,Base Sequence ,Leukemia Virus, Feline ,Gene Products, env ,virus diseases ,biology.organism_classification ,DNA, Viral ,Mutation ,Plasmids - Abstract
In efforts to elucidate the proximal leukemogens that might be produced during a feline leukemia virus (FeLV) infection of cats, homologous recombinations between molecularly cloned exogenous and endogenous FeLV proviruses of known sequences were examined in cell cultures in vitro. A plasmid containing an infectious member of the most commonly occurring FeLV subgroup (FeLV subgroup A or FeLV-A) was coexpressed with noninfectious constructs containing the envelope (env) gene of an endogenously inherited FeLV-like feline genomic element in transfected feline fibroblasts. The viruses generated were selected for their ability to propagate in human cells which are resistant to infection by the parental ecotropic FeLV-A or the noninfectious endogenous constructs. An analysis of the recombinants thus derived identified a limited number of sites in the env gene which were preferentially utilized in the generation of recombinant FeLVs under the selection conditions used. These sites were clustered in the surface glycoprotein (SU) moiety of the env gene, and it appeared that most, but not all, of the SU gene product of FeLV-A, beginning from the N-terminus, can be replaced by sequences from an endogenous element, still allowing the virus to be biologically viable. In fact, these substitutions in the env gene expanded infectivity of the parental FeLV-A from ecotropic to polytropic cell tropism. Additionally, substitutions in the SU region yielded many recombinants in which a primary neutralizing pentapeptide epitope of FeLV-A was altered because of its variance in the endogenous element. In several of the recombinants, this sequence was also found to be frequently mutated. Consistent with the changes identified in this antibody-binding domain, the recombinant viruses were only weakly inhibited by a monoclonal antibody directed against this epitope, while FeLV-A was highly sensitive to neutralization.
- Published
- 1992
- Full Text
- View/download PDF
28. Inclusion of adolescents in preventive HIV vaccine trials: public health policy and research design at a crossroads
- Author
-
Heather B Jaspan, Coleen K Cunningham, Tim J P Tucker, Peter F Wright, Steve G Self, Rebecca L Sheets, Audrey S Rogers, Linda-Gail Bekker, Craig M Wilson, Ann Duerr, and Judith N Wasserheit
- Subjects
Male ,medicine.medical_specialty ,genetic structures ,Adolescent ,medicine.medical_treatment ,Pterygium surgery ,Physical examination ,medicine ,Humans ,Pharmacology (medical) ,AIDS Vaccines ,medicine.diagnostic_test ,business.industry ,Health Policy ,Immunosuppression ,medicine.disease ,Connective tissue disease ,eye diseases ,Surgery ,Sclera ,Transplantation ,Infectious Diseases ,medicine.anatomical_structure ,Methylprednisolone ,Research Design ,Female ,sense organs ,business ,Complication ,medicine.drug - Abstract
Purpose To report a case of surgically induced necrotizing scleritis (SINS) as a complication of conjunctival autograft after pterygium excision. Methods A 52-year-old man had undergone nasal pterygium excision with conjunctival autograft in the right eye at another facility. He was treated for suspected infective scleritis before presentation. Results The sclera, at the site of prior pterygium excision, showed significant thinning with uveal show. There was active inflammation adjacent to the site of thinning. Systemic studies and the examination were noncontributory. The patient was suspected of having SINS and received pulsed injections of methylprednisolone 1000 mg/d for 3 days. The patient also underwent an emergency scleral patch graft with amniotic membrane graft. Over the next 2 weeks, the scleral graft showed vascularization and was taken well. Conclusions SINS may develop after pterygium surgery with conjunctival autograft. Evidence of connective tissue disease may or may not be found on clinical examination and on laboratory studies. Early diagnosis, prompt immunosuppression, and scleral patch grafting prevents progression and further devastating complications.
- Published
- 2007
29. Mechanism of ad5 vaccine immunity and toxicity: fiber shaft targeting of dendritic cells
- Author
-
Phillip L. Gomez, Rebecca L. Sheets, Wing Pui Kong, Cheng Cheng, C. Richter King, Jason G. D. Gall, and Gary J. Nabel
- Subjects
lcsh:Immunologic diseases. Allergy ,Receptors, Peptide ,Adenoviridae Infections ,Genetic enhancement ,Genetic Vectors ,Immunology ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,Adenoviridae ,Viral vector ,Mice ,Viral Proteins ,Immune system ,Transduction, Genetic ,Immunity ,Virology ,Genetics ,medicine ,Animals ,Humans ,Vector (molecular biology) ,Receptors, Immunologic ,Molecular Biology ,lcsh:QH301-705.5 ,Mice, Inbred BALB C ,Dose-Response Relationship, Drug ,Immunogenicity ,Viral Vaccines ,Dendritic Cells ,Acquired immune system ,Infectious Diseases ,lcsh:Biology (General) ,Mutation ,Receptors, Virus ,Female ,Parasitology ,Rabbits ,lcsh:RC581-607 ,Research Article - Abstract
Recombinant adenoviral (rAd) vectors elicit potent cellular and humoral immune responses and show promise as vaccines for HIV-1, Ebola virus, tuberculosis, malaria, and other infections. These vectors are now widely used and have been generally well tolerated in vaccine and gene therapy clinical trials, with many thousands of people exposed. At the same time, dose-limiting adverse responses have been observed, including transient low-grade fevers and a prior human gene therapy fatality, after systemic high-dose recombinant adenovirus serotype 5 (rAd5) vector administration in a human gene therapy trial. The mechanism responsible for these effects is poorly understood. Here, we define the mechanism by which Ad5 targets immune cells that stimulate adaptive immunity. rAd5 tropism for dendritic cells (DCs) was independent of the coxsackievirus and adenovirus receptor (CAR), its primary receptor or the secondary integrin RGD receptor, and was mediated instead by a heparin-sensitive receptor recognized by a distinct segment of the Ad5 fiber, the shaft. rAd vectors with CAR and RGD mutations did not infect a variety of epithelial and fibroblast cell types but retained their ability to transfect several DC types and stimulated adaptive immune responses in mice. Notably, the pyrogenic response to the administration of rAd5 also localized to the shaft region, suggesting that this interaction elicits both protective immunity and vector-induced fevers. The ability of replication-defective rAd5 viruses to elicit potent immune responses is mediated by a heparin-sensitive receptor that interacts with the Ad5 fiber shaft. Mutant CAR and RGD rAd vectors target several DC and mononuclear subsets and induce both adaptive immunity and toxicity. Understanding of these interactions facilitates the development of vectors that target DCs through alternative receptors that can improve safety while retaining the immunogenicity of rAd vaccines., Author Summary Recombinant adenovirus (rAd) vectors are remarkable for their ability to stimulate potent immune responses and to mediate highly efficient gene transfer. These vectors have been used extensively in human studies with generally acceptable tolerability. As with many bioactive compounds, adenoviruses can also cause potentially serious side effects, as observed in a human gene therapy trial several years ago that led to a fatality. The first manifestation of this toxicity is fever, but the relation of this side effect to the ability of the vector to stimulate immunity was unknown. We show that targeting of rAd vectors induces vaccine responses and toxicity through a previously unrecognized mechanism related to its attachment and entry into cells. We find that both adaptive immunity and fever are mediated by targeting of the rAd vector to dendritic cells and some monocytes, independent of the coxsackievirus and adenovirus receptor and RGD binding domains, mediated instead by the fiber shaft. This finding suggests that a distinct receptor present on dendritic and mononuclear cells mediates both effects. The immunogenicity of rAd vectors is dependent on targeting of virus through a specific fiber region and mediates rAd toxicity, which has implications for vaccine and gene therapy vector design that may help to improve rAd safety and efficacy.
- Published
- 2007
30. Toxicological Safety Evaluation of DNA Plasmid Vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile Virus Is Similar Despite Differing Plasmid Backbones or Gene-Inserts
- Author
-
Phillip L. Gomez, Robert T. Bailer, John Rathmann, Charla Andrews, Rebecca L. Sheets, T. Scott Manetz, and Judith A Stein
- Subjects
Male ,Genes, Viral ,Biology ,Toxicology ,medicine.disease_cause ,Severe Acute Respiratory Syndrome ,Injections, Intramuscular ,Article ,DNA vaccination ,Plasmid ,medicine ,Vaccines, DNA ,Animals ,Tissue Distribution ,Promoter Regions, Genetic ,Acquired Immunodeficiency Syndrome ,Reactogenicity ,Ebola virus ,Viral Vaccine ,Viral Vaccines ,Vector vaccine ,Hemorrhagic Fever, Ebola ,Ebolavirus ,Virology ,Vaccination ,Infectious disease (medical specialty) ,Immunology ,HIV-1 ,Female ,Rabbits ,West Nile virus ,West Nile Fever ,Plasmids - Abstract
The Vaccine Research Center has developed a number of vaccine candidates for different diseases/infectious agents (HIV-1, Severe Acute Respiratory Syndrome virus, West Nile virus, and Ebola virus, plus a plasmid cytokine adjuvant-IL-2/Ig) based on a DNA plasmid vaccine platform. To support the clinical development of each of these vaccine candidates, preclinical studies were performed to screen for potential toxicities (intrinsic and immunotoxicities). All treatment-related toxicities identified in these repeated-dose toxicology studies have been confined primarily to the sites of injection and seem to be the result of both the delivery method (as they are seen in both control and treated animals) and the intended immune response to the vaccine (as they occur with greater frequency and severity in treated animals). Reactogenicity at the site of injection is generally seen to be reversible as the frequency and severity diminished between doses and between the immediate and recovery termination time points. This observation also correlated with the biodistribution data reported in the companion article (Sheets et al., 2006), in which DNA plasmid vaccine was shown to remain at the site of injection, rather than biodistributing widely, and to clear over time. The results of these safety studies have been submitted to the Food and Drug Administration to support the safety of initiating clinical studies with these and related DNA plasmid vaccines. Thus far, standard repeated-dose toxicology studies have not identified any target organs for toxicity (other than the injection site) for our DNA plasmid vaccines at doses up to 8 mg per immunization, regardless of disease indication (i.e., expressed gene-insert) and despite differences (strengths) in the promoters used to drive this expression. As clinical data accumulate with these products, it will be possible to retrospectively compare the safety profiles of the products in the clinic to the results of the repeated-dose toxicology studies, in order to determine the utility of such toxicology studies for signaling potential immunotoxicities or intrinsic toxicities from DNA vaccines. These data build on the biodistribution studies performed (see companion article, Sheets et al., 2006) to demonstrate the safety and suitability for investigational human use of DNA plasmid vaccine candidates for a variety of infectious disease prevention indications.
- Published
- 2006
31. Preventive HIV-1 Vaccines
- Author
-
Donna K. F. Chandler, Karen L. Goldenthal, Therese Cvetkovich, Rebecca L. Sheets, Julienne M. Vaillancourt, and Antonia Geber
- Subjects
Government ,Economic growth ,medicine.medical_specialty ,Acquired immunodeficiency syndrome (AIDS) ,business.industry ,Scale (social sciences) ,Public health ,Human immunodeficiency virus (HIV) ,Medicine ,business ,medicine.disease_cause ,medicine.disease ,World health - Abstract
The World Health Organization (WHO) has estimated that 40 million individuals are living with HIV-1 as of 2003 (UNAIDS/WHO, 2003). In addition, approximately 14,000 people are newly infected with HIV-1 each day, worldwide (UNAIDS/WHO, 2003). Thus, there is tremendous public health interest in developing safe and effective preventive HIV-1 vaccines. Vaccines have proven to be one of the most effective means of preventing serious infectious diseases. The contribution of vaccines toward markedly decreasing the incidence of many of the once-common childhood infectious diseases has been particularly notable (CDC, 1999). Thus, the public health goal is to develop an effective HIV-1 vaccine that will achieve a similar success in controlling the AIDS epidemic. However, the development of an HIV-1 vaccine has a unique and complex set of scientific, clinical, social, ethical, and economic challenges (Amara and Robinson, 2002; Esparza and Osmanov, 2003; Fast et al., 1995; Goldenthal et al.,1998; Klausner et al., 2003; Nathanson and Mathieson, 2000). Collaborative efforts in the development of an effective HIV-1 vaccine are ongoing among government agencies, academic investigators, and pharmaceutical and biotechnology companies. Even more organization on a global scale has been strongly encouraged (Klausner et al., 2003).
- Published
- 2004
- Full Text
- View/download PDF
32. Toxicological Safety Evaluation of DNA Plasmid Vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile Virus Is Similar Despite Differing Plasmid Backbones or Gene-Inserts.
- Author
-
Rebecca L. Sheets, Judith Stein, T. Scott Manetz, Charla Andrews, Robert Bailer, John Rathmann, and Phillip L. Gomez
- Subjects
VACCINES ,DISEASES - Abstract
The Vaccine Research Center has developed a number of vaccine candidates for different diseases/infectious agents (HIV-1, Severe Acute Respiratory Syndrome virus, West Nile virus, and Ebola virus, plus a plasmid cytokine adjuvant—IL-2/Ig) based on a DNA plasmid vaccine platform. To support the clinical development of each of these vaccine candidates, preclinical studies were performed to screen for potential toxicities (intrinsic and immunotoxicities). All treatment-related toxicities identified in these repeated-dose toxicology studies have been confined primarily to the sites of injection and seem to be the result of both the delivery method (as they are seen in both control and treated animals) and the intended immune response to the vaccine (as they occur with greater frequency and severity in treated animals). Reactogenicity at the site of injection is generally seen to be reversible as the frequency and severity diminished between doses and between the immediate and recovery termination time points. This observation also correlated with the biodistribution data reported in the companion article (Sheets et al., 2006), in which DNA plasmid vaccine was shown to remain at the site of injection, rather than biodistributing widely, and to clear over time. The results of these safety studies have been submitted to the Food and Drug Administration to support the safety of initiating clinical studies with these and related DNA plasmid vaccines. Thus far, standard repeated-dose toxicology studies have not identified any target organs for toxicity (other than the injection site) for our DNA plasmid vaccines at doses up to 8 mg per immunization, regardless of disease indication (i.e., expressed gene-insert) and despite differences (strengths) in the promoters used to drive this expression. As clinical data accumulate with these products, it will be possible to retrospectively compare the safety profiles of the products in the clinic to the results of the repeated-dose toxicology studies, in order to determine the utility of such toxicology studies for signaling potential immunotoxicities or intrinsic toxicities from DNA vaccines. These data build on the biodistribution studies performed (see companion article, Sheets et al., 2006) to demonstrate the safety and suitability for investigational human use of DNA plasmid vaccine candidates for a variety of infectious disease prevention indications. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
33. Mobile elements bounded by C4A4 telomeric repeats in oxytricha fallax
- Author
-
Dean Dawson, Deborah Ang, Samuel Cartinhour, Kevin Williams, Alice Lee, Rebecca L. Sheets, and Glenn Herrick
- Subjects
Cell Nucleus ,DNA Replication ,Transposable element ,Genetics ,Macronucleus ,DNA ,Biology ,biology.organism_classification ,Genome ,Chromosomes ,General Biochemistry, Genetics and Molecular Biology ,Transposition (music) ,chemistry.chemical_compound ,chemistry ,DNA Transposable Elements ,Animals ,Oxytricha trifallax ,Ciliophora ,Mobile genetic elements ,Repetitive Sequences, Nucleic Acid ,Sequence (medicine) - Abstract
A novel family of micronuclear elements termed telomere-bearing elements (TBEs) is described. All 1900 family members are eliminated during macronuclear development. We conclude that they are transposons, first because the members are moderately conserved in sequence and probably dispersed in the genome. Second, in two cases, sequence comparison of the termini and flanks of the element with the corresponding empty site indicate that elements cause 3 bp target duplications (AAT) upon insertion; the 3 bp are part of the 5 bp target sequence, AATGA. Lastly, both elements carry 77 or 78 bp inverted terminal repeats. The tip of each inverted terminal repeat is the 17 bp telomere-like sequence 5' C1A4C4A4C4. At least half of the elements have these 17 bp or an extremely similar sequence. One possible pathway for transposition into new micronuclear sites starts in the developing macronucleus with excision to create a free linear form to which telomeres are added, followed by a low frequency of movement to the micronucleus, and insertion into the germ-line micronuclear DNA.
- Published
- 1985
- Full Text
- View/download PDF
34. Nucleotide sequence of yellow fever virus: implications for flavivirus gene expression and evolution
- Author
-
Charles M. Rice, James H. Strauss, Se Jung Shin, Edith M. Lenches, Sean R. Eddy, and Rebecca L. Sheets
- Subjects
Protein Conformation ,viruses ,Biology ,Virus Replication ,Virus ,Viral Proteins ,Kunjin virus ,Protein biosynthesis ,Gene ,Glycoproteins ,Genetics ,Multidisciplinary ,Base Sequence ,Nucleic acid sequence ,RNA ,biology.organism_classification ,Virology ,Biological Evolution ,Flavivirus ,Viral replication ,Gene Expression Regulation ,Genes ,Protein Biosynthesis ,Nucleic Acid Conformation ,RNA, Viral ,Yellow fever virus ,Protein Processing, Post-Translational - Abstract
The sequence of the entire RNA genome of the type flavivirus, yellow fever virus, has been obtained. Inspection of this sequence reveals a single long open reading frame of 10,233 nucleotides, which could encode a polypeptide of 3411 amino acids. The structural proteins are found within the amino-terminal 780 residues of this polyprotein; the remainder of the open reading frame consists of nonstructural viral polypeptides. This genome organization implies that mature viral proteins are produced by posttranslational cleavage of a polyprotein precursor and has implications for flavivirus RNA replication and for the evolutionary relation of this virus family to other RNA viruses.
- Published
- 1985
35. First-in-human phase I clinical trial of a recombinant vesicular stomatitis virus (rVSV)-based preventive HIV-1 vaccine
- Author
-
Marnie Elizaga, Ian Frank, Donald K. Carter, John H. Eldridge, Nidhi Kochar, Rebecca L. Sheets, Mark J. Mulligan, Spyros A. Kalams, Jonathan D. Fuchs, Michael Pensiero, Nicole Frahm, David K. Clarke, and Mary Allen
- Subjects
lcsh:Immunologic diseases. Allergy ,Saliva ,Reactogenicity ,biology ,business.industry ,Phases of clinical research ,Bioinformatics ,Placebo ,biology.organism_classification ,Virology ,chemistry.chemical_compound ,Infectious Diseases ,chemistry ,Vesicular stomatitis virus ,Poster Presentation ,Medicine ,Vaccinia ,lcsh:RC581-607 ,business ,Adverse effect ,Whole blood - Abstract
Methods HVTN 090 enrolled sixty healthy, HIV-1-uninfected adults in a randomized, double-blinded, placebo-controlled dose escalation study. Groups of 12 participants received rVSV Indiana HIV Gag vaccine at 5 dose levels (4.6 x 10 to 3.4 x 10 PFU) (N=10/group) or placebo (N=2/group), delivered intramuscularly at 0 and 2 months. Reactogenicity over 7 days, adverse events (AEs), and viral cultures from whole blood, urine, saliva and swabs of oral lesions were collected. HIV-1-specific CD4+ and CD8+ T-cell responses to Gag peptides were measured 1 and 2 weeks post-boost by intracellular cytokine staining.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.