Network


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

Hotspot


Dive into the research topics where Geoffrey W. Stone is active.

Publication


Featured researches published by Geoffrey W. Stone.


Journal of Virology | 2006

Multimeric Soluble CD40 Ligand and GITR Ligand as Adjuvants for Human Immunodeficiency Virus DNA Vaccines

Geoffrey W. Stone; Suzanne Barzee; Victoria Snarsky; Kristin Kee; Celsa A. Spina; Xiao Fang Yu; Richard S. Kornbluth

ABSTRACT For use in humans, human immunodeficiency virus (HIV) DNA vaccines may need to include immunostimulatory adjuvant molecules. CD40 ligand (CD40L), a member of the tumor necrosis factor (TNF) superfamily (TNFSF), is one candidate adjuvant, but it has been difficult to use because it is normally expressed as a trimeric membrane molecule. Soluble trimeric forms of CD40L have been produced, but in vitro data indicate that multimeric, many-trimer forms of soluble CD40L are more active. This multimerization requirement was evaluated in mice using plasmids that encoded either 1-trimer, 2-trimer, or 4-trimer soluble forms of CD40L. Fusion with the body of Acrp30 was used to produce the 2-trimer form, and fusion with the body of surfactant protein D was used to produce the 4-trimer form. Using plasmids for secreted HIV-1 antigens Gag and Env, soluble CD40L was active as an adjuvant in direct proportion to the valence of the trimers (1 < 2 < 4). These CD40L-augmented DNA vaccines elicited strong CD8+ T-cell responses but did not elicit significant CD4+ T-cell or antibody responses. To test the applicability of the multimeric fusion protein approach to other TNFSFs, a 4-trimer construct for the ligand of glucocorticoid-induced TNF family-related receptor (GITR) was also prepared. Multimeric soluble GITR ligand (GITRL) augmented the CD8+ T-cell, CD4+ T-cell, and antibody responses to DNA vaccination. In summary, multimeric CD40L and GITRL are new adjuvants for DNA vaccines. Plasmids for expressing multimeric TNFSF fusion proteins permit the rapid testing of TNFSF molecules in vivo.


Journal of Leukocyte Biology | 2006

Immunostimulatory combinations: designing the next generation of vaccine adjuvants

Richard S. Kornbluth; Geoffrey W. Stone

Agents that activate dendritic cells are essential components for vaccines and can be conceptualized as molecular adjuvants. Other molecular adjuvants affect downstream factors that shape the resulting immune response. This review provides a compendium of recently studied molecular adjuvants, focusing on CD8+ T cell responses, which have important roles in HIV vaccines. Reference is also made to CD8+ T cell antitumor responses, where parallel studies of molecular adjuvants are being pursued. Molecular adjuvants can be considered in the following groups: TNF superfamily molecules such as CD40 ligand; agonists for TLRs; agonists for NAIP, CIITA, HET‐E, TP‐1‐leucine‐rich repeat pathway receptors, such as nucleotide‐binding and oligomerization domain (NOD)1, NOD2, and cryopyrin; chemokines; ILs; CSFs; IFNs; alarmins; and purinergic P2X7 receptor agonists. Complementing these positively acting agents are strategies to reduce the immunosuppressive effects of CD4+CD25+ regulatory T cells and negatively acting factors such as TGF‐β, IL‐10, suppressor of cytokine signaling 1, and programmed cell death‐1 using neutralizing antibodies, antisense, and small interfering RNA. Especially effective are combinations of molecular adjuvants, which can elicit a massive expansion of antigen‐specific CD8+ T cells and show unprecedented efficacy in vaccine and tumor models. Taken together, these new approaches provide significant incremental progress in the development of vaccines to elicit cell‐mediated immunity against HIV and other pathogens.


PLOS ONE | 2009

Nanoparticle-Delivered Multimeric Soluble CD40L DNA Combined with Toll-Like Receptor Agonists as a Treatment for Melanoma

Geoffrey W. Stone; Suzanne Barzee; Victoria Snarsky; Camila Santucci; Brian Tran; Robert Langer; Daniel G. Anderson; Richard S. Kornbluth

Stimulation of CD40 or Toll-Like Receptors (TLR) has potential for tumor immunotherapy. Combinations of CD40 and TLR stimulation can be synergistic, resulting in even stronger dendritic cell (DC) and CD8+ T cell responses. To evaluate such combinations, established B16F10 melanoma tumors were injected every other day X 5 with plasmid DNA encoding a multimeric, soluble form of CD40L (pSP-D-CD40L) either alone or combined with an agonist for TLR1/2 (Pam3CSK4 ), TLR2/6 (FSL-1 and MALP2), TLR3 (polyinosinic-polycytidylic acid, poly(I:C)), TLR4 ( monophosphoryl lipid A, MPL), TLR7 (imiquimod), or TLR9 (Class B CpG phosphorothioate oligodeoxynucleotide, CpG). When used by itself, pSP-D-CD40L slowed tumor growth and prolonged survival, but did not lead to cure. Of the TLR agonists, CpG and poly(I:C) also slowed tumor growth, and the combination of these two TLR agonists was more effective than either agent alone. The triple combination of intratumoral pSP-D-CD40L + CpG + poly(I:C) markedly slowed tumor growth and prolonged survival. This treatment was associated with a reduction in intratumoral CD11c+ dendritic cells and an influx of CD8+ T cells. Since intratumoral injection of plasmid DNA does not lead to efficient transgene expression, pSP-D-CD40L was also tested with cationic polymers that form DNA-containing nanoparticles which lead to enhanced intratumoral gene expression. Intratumoral injections of pSP-D-CD40L-containing nanoparticles formed from polyethylenimine (PEI) or C32 (a novel biodegradable poly(B-amino esters) polymer) in combination with CpG + poly(I:C) had dramatic antitumor effects and frequently cured mice of B16F10 tumors. These data confirm and extend previous reports that CD40 and TLR agonists are synergistic and demonstrate that this combination of immunostimulants can significantly suppress tumor growth in mice. In addition, the enhanced effectiveness of nanoparticle formulations of DNA encoding immunostimulatory molecules such as multimeric, soluble CD40L supports the further study of this technology for tumor immunotherapy.


Clinical and Vaccine Immunology | 2006

Macaque Multimeric Soluble CD40 Ligand and GITR Ligand Constructs Are Immunostimulatory Molecules In Vitro

Geoffrey W. Stone; Suzanne Barzee; Victoria Snarsky; Celsa A. Spina; Jeffrey D. Lifson; Vinod Kumar Bhaskara Pillai; Rama Rao Amara; Francois Villinger; Richard S. Kornbluth

ABSTRACT CD40 ligand (CD40L) and GITR ligand (glucocorticoid-induced tumor necrosis factor receptor-related protein ligand [GITRL]) are tumor necrosis factor superfamily molecules that can be used as vaccine adjuvants. In a previous human immunodeficiency virus (HIV) DNA vaccine study in mice, we found that plasmids expressing multimeric soluble forms of trimeric CD40L (i.e., many trimers) were stronger activators of CD8+ T-cell responses than were single-trimer soluble forms or the natural membrane-bound molecule. This report describes similar multimeric soluble molecules that were constructed for studies in macaques. Both two-trimer and four-trimer forms of macaque CD40L were active in B-cell proliferation assays using macaque and human cells. With human cells, four-trimer macaque GITRL costimulated CD4+ T-cell proliferation and abrogated the immunosuppressive effects of CD4+ CD25+ regulatory T cells on a mixed leukocyte reaction. These molecular adjuvants provide new tools for vaccine development in the simian immunodeficiency virus system and other macaque models.


Antimicrobial Agents and Chemotherapy | 2004

Mechanism of Action of NB2001 and NB2030, Novel Antibacterial Agents Activated by β-Lactamases

Geoffrey W. Stone; Qin Zhang; Rosario Silvestre Castillo; V. Ramana Doppalapudi; Analia R. Bueno; Jean Y. Lee; Qing Li; Maria Vladimir Sergeeva; Gody Khambatta; Nafsika H. Georgopapadakou

ABSTRACT Two potent antibacterial agents designed to undergo enzyme-catalyzed therapeutic activation were evaluated for their mechanisms of action. The compounds, NB2001 and NB2030, contain a cephalosporin with a thienyl (NB2001) or a tetrazole (NB2030) ring at the C-7 position and are linked to the antibacterial triclosan at the C-3 position. The compounds exploit β-lactamases to release triclosan through hydrolysis of the β-lactam ring. Like cephalothin, NB2001 and NB2030 were hydrolyzed by class A β-lactamases (Escherichia coli TEM-1 and, to a lesser degree, Staphylococcus aureus PC1) and class C β-lactamases (Enterobacter cloacae P99 and E. coli AmpC) with comparable catalytic efficiencies (kcat/Km). They also bound to the penicillin-binding proteins of S. aureus and E. coli, but with reduced affinities relative to that of cephalothin. Accordingly, they produced a cell morphology in E. coli consistent with the toxophore rather than the β-lactam being responsible for antibacterial activity. In biochemical assays, they inhibited the triclosan target enoyl reductase (FabI), with 50% inhibitory concentrations being markedly reduced relative to that of free triclosan. The transport of NB2001, NB2030, and triclosan was rapid, with significant accumulation of triclosan in both S. aureus and E. coli. Taken together, the results suggest that NB2001 and NB2030 act primarily as triclosan prodrugs in S. aureus and E. coli.


Journal of Thoracic Oncology | 2009

Regression of Established AB1 Murine Mesothelioma Induced by Peritumoral Injections of CpG Oligodeoxynucleotide Either Alone or in Combination with Poly(I:C) and CD40 Ligand Plasmid DNA

Geoffrey W. Stone; Suzanne Barzee; Victoria Snarsky; Camila Santucci; Brian Tran; Richard S. Kornbluth

Introduction: Stimulation of the CD40 receptor using an agonistic anti-CD40 antibody can slow the growth of AB1 tumors. Stimulation of the GITR receptor may also have antitumor activity by countering the immunosuppressive effects of regulatory CD4+ T cells. Similarly, agonists for Toll-Like Receptors (TLR) such as CpG oligodeoxynucleotides (TLR9 agonist) have activity against AB1 tumors. Combinations of CpG with CD40 ligand and polyinosinic-polycytidylic acid (poly(I:C), TLR3 agonist) may be even stronger than CpG alone. The synergistic effects of these combinations have been tested in other tumor types but not in mesothelioma. Methods: Established AB1 mesothelioma tumors were injected with either plasmid DNA encoding a novel 4-trimer form of murine CD40 ligand (pSP-D-CD40L), GITR ligand (GITRL), or control plasmid DNA. In addition, CpG with or without poly(I:C) was also injected intratumorally. Results: Plasmid injections of pSP-D-CD40L or pSP-D-GITRL, had no significant antitumor effect, possibly reflecting the difficulty of administering DNA injections into this very dense tissue. However, the injection of CpG with or without poly(I:C) strongly suppressed tumor growth and led to long-term tumor-free survival. The response to a triple combination of pSP-D-CD40L + CpG + poly(I:C) was demonstrated by an increase in intratumoral CD8+ T cells and a dramatic increase in F4/80+ macrophages. Conclusions: Intratumoral injections of plasmid DNAs encoding highly active forms of either CD40 ligand or GITR ligand had no significant antitumor effects in this model, although improved DNA delivery techniques could possibly improve this strategy. In contrast, intratumoral CpG injections had significant antitumor effects and there were indications that CpG plus poly(I:C) was even more effective. Taken together, these data confirm previous reports that immune stimulants, especially CpG TLR9 agonists, have potential as a treatment for mesothelioma.


PLOS ONE | 2016

Constitutively Active MAVS Inhibits HIV-1 Replication via Type I Interferon Secretion and Induction of HIV-1 Restriction Factors

Sachin Gupta; James M. Termini; Biju Issac; Elizabeth Guirado; Geoffrey W. Stone

Type I interferon is known to inhibit HIV-1 replication through the induction of interferon stimulated genes (ISG), including a number of HIV-1 restriction factors. To better understand interferon-mediated HIV-1 restriction, we constructed a constitutively active form of the RIG-I adapter protein MAVS. Constitutive MAVS was generated by fusion of full length MAVS to a truncated form of the Epstein Barr virus protein LMP1 (ΔLMP1). Supernatant from ΔLMP1-MAVS-transfected 293T cells contained high levels of type I interferons and inhibited HIV replication in both TZM-bl and primary human CD4+ T cells. Supernatant from ΔLMP1-MAVS-transfected 293T cells also inhibited replication of VSV-G pseudotyped single cycle SIV in TZM-bl cells, suggesting restriction was post-entry and common to both HIV and SIV. Gene array analysis of ΔLMP1-MAVS-transfected 293T cells and trans-activated CD4+ T cells showed significant upregulation of ISG, including previously characterized HIV restriction factors Viperin, Tetherin, MxB, and ISG56. Interferon blockade studies implicated interferon-beta in this response. In addition to direct viral inhibition, ΔLMP1-MAVS markedly enhanced secretion of IFN-β and IL-12p70 by dendritic cells and the activation and maturation of dendritic cells. Based on this immunostimulatory activity, an adenoviral vector (Ad5) expressing ΔLMP1-MAVS was tested as a molecular adjuvant in an HIV vaccine mouse model. Ad5-Gag antigen combined with Ad5-ΔLMP1-MAVS enhanced control of vaccinia-gag replication in a mouse challenge model, with 4/5 animals showing undetectable virus following challenge. Overall, ΔLMP1-MAVS is a promising reagent to inhibit HIV-1 replication in infected tissues and enhance vaccine-mediated immune responses, while avoiding toxicity associated with systemic type I interferon administration.


Archive | 2007

Immunostimulatory Combinations for Vaccine Adjuvants

Richard Syd Kornbluth; Geoffrey W. Stone


Archive | 2014

COMPOSITION COMPRISED OF ANTIGEN LINKED TO A TNF SUPERFAMILY LIGAND

Geoffrey W. Stone; Richard S. Kornbluth


F1000Research | 2014

Multimeric soluble 4-1BBL as a T cell stimulator for adoptive immunotherapy

Richard S. Kornbluth; Victoria Snarsky; Geoffrey W. Stone

Collaboration


Dive into the Geoffrey W. Stone's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suzanne Barzee

University of California

View shared research outputs
Top Co-Authors

Avatar

Brian Tran

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Celsa A. Spina

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel G. Anderson

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge