Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where William S. Luckett is active.

Publication


Featured researches published by William S. Luckett.


Infection and Immunity | 2008

Constitutive Activation of the PrfA regulon enhances the potency of vaccines based on live-attenuated and killed but metabolically active Listeria monocytogenes strains.

Peter Lauer; Bill Hanson; Edward E. Lemmens; Weiqun Liu; William S. Luckett; Meredith Leong; Heather E. Allen; Justin Skoble; Keith S. Bahjat; Nancy E. Freitag; Dirk G. Brockstedt; Thomas W. Dubensky

ABSTRACT Recombinant vaccines derived from the facultative intracellular bacterium Listeria monocytogenes are presently undergoing early-stage clinical evaluation in oncology treatment settings. This effort has been stimulated in part due to preclinical results that illustrate potent activation of innate and adaptive immune effectors by L. monocytogenes vaccines, combined with efficacy in rigorous animal models of malignant and infectious disease. Here, we evaluated the immunologic potency of a panel of isogenic vaccine strains that varied only in prfA. PrfA is an intracellularly activated transcription factor that induces expression of virulence genes and encoded heterologous antigens (Ags) in appropriately engineered vaccine strains. Mutant strains with PrfA locked into a constitutively active state are known as PrfA* mutants. We assessed the impacts of three PrfA* mutants, G145S, G155S, and Y63C, on the immunologic potencies of live-attenuated and photochemically inactivated nucleotide excision repair mutant (killed but metabolically active [KBMA]) vaccines. While PrfA* substantially increased Ag expression in strains grown in broth culture, Ag expression levels were equivalent in infected macrophage and dendritic cell lines, conditions that more closely parallel those in the immunized host. However, only the prfA(G155S) allele conferred significantly enhanced vaccine potency to KBMA vaccines. In the KBMA vaccine background, we show that PrfA*(G155S) enhanced functional cellular immunity following an intravenous or intramuscular prime-boost immunization regimen. These results form the basis of a rationale for including the prfA(G155S) allele in future live-attenuated or KBMA L. monocytogenes vaccines advanced to the clinical setting.


Infection and Immunity | 2009

Impact of Preexisting Vector-Specific Immunity on Vaccine Potency: Characterization of Listeria monocytogenes-Specific Humoral and Cellular Immunity in Humans and Modeling Studies Using Recombinant Vaccines in Mice

Meredith Leong; Johannes Hampl; Weiqun Liu; Shruti Mathur; Keith S. Bahjat; William S. Luckett; Thomas W. Dubensky; Dirk G. Brockstedt

ABSTRACT Recombinant live-attenuated Listeria monocytogenes is currently being developed as a vaccine platform for treatment or prevention of malignant and infectious diseases. The effectiveness of complex biologic vaccines, such as recombinant viral and bacterial vectors, can be limited by either preexisting or vaccine-induced vector-specific immunity. We characterized the level of L. monocytogenes-specific cellular and humoral immunity present in more than 70 healthy adult subjects as a first step to understanding its possible impact on the efficacy of L. monocytogenes-based vaccines being evaluated in early-phase clinical trials. Significant L. monocytogenes-specific humoral immunity was not measured in humans, consistent with a lack of antibodies in mice immunized with wild-type L. monocytogenes. Cellular immune responses specific for listeriolysin O, a secreted bacterial protein required for potency of L. monocytogenes-derived vaccines, were detected in approximately 60% of human donors tested. In mice, while wild-type L. monocytogenes did not induce significant humoral immunity, attenuated L. monocytogenes vaccine strains induced high-titer L. monocytogenes-specific antibodies when given at high doses used for immunization. Passive transfer of L. monocytogenes-specific antiserum to naïve mice had no impact on priming antigen-specific immunity in mice immunized with a recombinant L. monocytogenes vaccine. In mice with preexisting L. monocytogenes-specific immunity, priming of naïve T cells was not prevented, and antigen-specific responses could be boosted by additional vaccinations. For the first time, our findings establish the level of L. monocytogenes-specific cellular immunity in healthy adults, and, together with modeling studies performed with mice, they support the scientific rationale for repeated L. monocytogenes vaccine immunization regimens to elicit a desired therapeutic effect.


Infection and Immunity | 2009

Killed but Metabolically Active Bacillus anthracis Vaccines Induce Broad and Protective Immunity against Anthrax

Justin Skoble; J. W. Beaber; Yi Gao; Julie A. Lovchik; Laurie Sower; Weiqun Liu; William S. Luckett; Johnny W. Peterson; R. Calendar; Daniel A. Portnoy; C. R. Lyons; Thomas W. Dubensky

ABSTRACT Bacillus anthracis is the causative agent of anthrax. We have developed a novel whole-bacterial-cell anthrax vaccine utilizing B. anthracis that is killed but metabolically active (KBMA). Vaccine strains that are asporogenic and nucleotide excision repair deficient were engineered by deleting the spoIIE and uvrAB genes, rendering B. anthracis extremely sensitive to photochemical inactivation with S-59 psoralen and UV light. We also introduced point mutations into the lef and cya genes, which allowed inactive but immunogenic toxins to be produced. Photochemically inactivated vaccine strains maintained a high degree of metabolic activity and secreted protective antigen (PA), lethal factor, and edema factor. KBMA B. anthracis vaccines were avirulent in mice and induced less injection site inflammation than recombinant PA adsorbed to aluminum hydroxide gel. KBMA B. anthracis-vaccinated animals produced antibodies against numerous anthrax antigens, including high levels of anti-PA and toxin-neutralizing antibodies. Vaccination with KBMA B. anthracis fully protected mice against challenge with lethal doses of toxinogenic unencapsulated Sterne 7702 spores and rabbits against challenge with lethal pneumonic doses of fully virulent Ames strain spores. Guinea pigs vaccinated with KBMA B. anthracis were partially protected against lethal Ames spore challenge, which was comparable to vaccination with the licensed vaccine anthrax vaccine adsorbed. These data demonstrate that KBMA anthrax vaccines are well tolerated and elicit potent protective immune responses. The use of KBMA vaccines may be broadly applicable to bacterial pathogens, especially those for which the correlates of protective immunity are unknown.


Transfusion | 2007

Inactivation of parvovirus B19 in human platelet concentrates by treatment with amotosalen and ultraviolet A illumination.

Lynette Sawyer; Deborah Hanson; Grace Castro; William S. Luckett; Thomas W. Dubensky; Adonis Stassinopoulos

BACKGROUND: The human erythrovirus B19 (B19) is a small (18‐ to 26‐nm) nonenveloped virus with a single‐stranded DNA genome of 5.6 kb. B19 is clinically significant and is also generally resistant to pathogen inactivation methods. Photochemical treatment (PCT) with amotosalen and ultraviolet A (UVA) inactivates viruses, bacteria, and protozoa in platelets (PLTs) and plasma prepared for transfusion. In this study, the capacity of PCT to inactivate B19 in human PLT concentrates was evaluated.


Proceedings of the National Academy of Sciences of the United States of America | 2004

Listeria-based cancer vaccines that segregate immunogenicity from toxicity

Dirk G. Brockstedt; Martin A. Giedlin; Meredith Leong; Keith S. Bahjat; Yi Gao; William S. Luckett; Weiqun Liu; David N. Cook; Daniel A. Portnoy; Thomas W. Dubensky


Archive | 2004

Modified free-living microbes, vaccine compositions and methods of use thereof

Thomas W. Dubensky; Dirk G. Brockstedt; Keith S. Bahjat; John E. Hearst; David N. Cook; William S. Luckett


Archive | 2004

Antigen-presenting cell vaccines and methods of use thereof

Thomas W. Dubensky; Dirk G. Brockstedt; Keith S. Bahjat; John E. Hearst; David N. Cook; William S. Luckett


Blood | 2004

Immunization with Live-Attenuated Listeria Encoding CMV Antigen Induces Extensive Expansion of CMV-Specific CD8+ T-Cells Following HSCT: An Alternative to Adoptive Antiviral Immunotherapy.

John D. Roback; Levan J. Lezhava; Elisabeth Hesse; Christopher D. Hillyer; William S. Luckett; Thomas W. Dubensky; Martin Giedlin


Archive | 2007

HOLIN-ENHANCED VACCINES AND REAGENTS, AND METHODS OF USE THEREOF

Peter Lauer; Thomas W. Dubensky; William S. Luckett; William G. Hanson


Blood | 2005

Live-Attenuated and Novel Non-Replicating Listeria Vaccines Encoding CMV Antigen Produce Persistent Functional Antiviral Immunity.

John D. Roback; Levan J. Lezhava; William S. Luckett; Thomas W. Dubensky; Christopher D. Hillyer; Martin Giedlin

Collaboration


Dive into the William S. Luckett's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David N. Cook

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weiqun Liu

University of California

View shared research outputs
Top Co-Authors

Avatar

John E. Hearst

University of California

View shared research outputs
Top Co-Authors

Avatar

Justin Skoble

University of California

View shared research outputs
Top Co-Authors

Avatar

Meredith Leong

University of California

View shared research outputs
Top Co-Authors

Avatar

Peter Lauer

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge