Network


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

Hotspot


Dive into the research topics where Joseph Hesselgesser is active.

Publication


Featured researches published by Joseph Hesselgesser.


Nature | 2014

Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys

James B. Whitney; Alison L. Hill; Srisowmya Sanisetty; Pablo Penaloza-MacMaster; Jinyan Liu; Mayuri Shetty; Lily Parenteau; Crystal Cabral; Jennifer Shields; Stephen Blackmore; Jeffrey Y. Smith; Amanda L. Brinkman; Lauren Peter; Sheeba Mathew; Kaitlin M. Smith; Erica N. Borducchi; Daniel I. S. Rosenbloom; Mark G. Lewis; Jillian Hattersley; Bei Li; Joseph Hesselgesser; Romas Geleziunas; Merlin L. Robb; Jerome H. Kim; Nelson L. Michael; Dan H. Barouch

The viral reservoir represents a critical challenge for human immunodeficiency virus type 1 (HIV-1) eradication strategies. However, it remains unclear when and where the viral reservoir is seeded during acute infection and the extent to which it is susceptible to early antiretroviral therapy (ART). Here we show that the viral reservoir is seeded rapidly after mucosal simian immunodeficiency virus (SIV) infection of rhesus monkeys and before systemic viraemia. We initiated suppressive ART in groups of monkeys on days 3, 7, 10 and 14 after intrarectal SIVMAC251 infection. Treatment with ART on day 3 blocked the emergence of viral RNA and proviral DNA in peripheral blood and also substantially reduced levels of proviral DNA in lymph nodes and gastrointestinal mucosa as compared with treatment at later time points. In addition, treatment on day 3 abrogated the induction of SIV-specific humoral and cellular immune responses. Nevertheless, after discontinuation of ART following 24 weeks of fully suppressive therapy, virus rebounded in all animals, although the monkeys that were treated on day 3 exhibited a delayed viral rebound as compared with those treated on days 7, 10 and 14. The time to viral rebound correlated with total viraemia during acute infection and with proviral DNA at the time of ART discontinuation. These data demonstrate that the viral reservoir is seeded rapidly after intrarectal SIV infection of rhesus monkeys, during the ‘eclipse’ phase, and before detectable viraemia. This strikingly early seeding of the refractory viral reservoir raises important new challenges for HIV-1 eradication strategies.


PLOS Pathogens | 2014

Histone Deacetylase Inhibitor Romidepsin Induces HIV Expression in CD4 T Cells from Patients on Suppressive Antiretroviral Therapy at Concentrations Achieved by Clinical Dosing

Datsen George Wei; Vicki Chiang; Elizabeth Fyne; Mini Balakrishnan; Tiffany Barnes; Michael Graupe; Joseph Hesselgesser; Alivelu Irrinki; Jeffrey P. Murry; George Stepan; Kirsten M. Stray; Angela Tsai; Helen Yu; Jonathan Spindler; Mary Kearney; Celsa A. Spina; Deborah McMahon; Jacob Lalezari; Derek D. Sloan; John W. Mellors; Romas Geleziunas; Tomas Cihlar

Persistent latent reservoir of replication-competent proviruses in memory CD4 T cells is a major obstacle to curing HIV infection. Pharmacological activation of HIV expression in latently infected cells is being explored as one of the strategies to deplete the latent HIV reservoir. In this study, we characterized the ability of romidepsin (RMD), a histone deacetylase inhibitor approved for the treatment of T-cell lymphomas, to activate the expression of latent HIV. In an in vitro T-cell model of HIV latency, RMD was the most potent inducer of HIV (EC50 = 4.5 nM) compared with vorinostat (VOR; EC50 = 3,950 nM) and other histone deacetylase (HDAC) inhibitors in clinical development including panobinostat (PNB; EC50 = 10 nM). The HIV induction potencies of RMD, VOR, and PNB paralleled their inhibitory activities against multiple human HDAC isoenzymes. In both resting and memory CD4 T cells isolated from HIV-infected patients on suppressive combination antiretroviral therapy (cART), a 4-hour exposure to 40 nM RMD induced a mean 6-fold increase in intracellular HIV RNA levels, whereas a 24-hour treatment with 1 µM VOR resulted in 2- to 3-fold increases. RMD-induced intracellular HIV RNA expression persisted for 48 hours and correlated with sustained inhibition of cell-associated HDAC activity. By comparison, the induction of HIV RNA by VOR and PNB was transient and diminished after 24 hours. RMD also increased levels of extracellular HIV RNA and virions from both memory and resting CD4 T-cell cultures. The activation of HIV expression was observed at RMD concentrations below the drug plasma levels achieved by doses used in patients treated for T-cell lymphomas. In conclusion, RMD induces HIV expression ex vivo at concentrations that can be achieved clinically, indicating that the drug may reactivate latent HIV in patients on suppressive cART.


Nature | 2016

Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys

Erica N. Borducchi; Crystal Cabral; Kathryn E. Stephenson; Jinyan Liu; Peter Abbink; David Ng’ang’a; Joseph P. Nkolola; Amanda L. Brinkman; Lauren Peter; Benjamin C. Lee; Jessica Jimenez; David Jetton; Jade Mondesir; Shanell Mojta; Abishek Chandrashekar; Katherine Molloy; Galit Alter; Jeffrey M. Gerold; Alison L. Hill; Mark G. Lewis; Maria G. Pau; Hanneke Schuitemaker; Joseph Hesselgesser; Romas Geleziunas; Jerome H. Kim; Merlin L. Robb; Nelson L. Michael; Dan H. Barouch

The development of immunologic interventions that can target the viral reservoir in HIV-1-infected individuals is a major goal of HIV-1 research. However, little evidence exists that the viral reservoir can be sufficiently targeted to improve virologic control following discontinuation of antiretroviral therapy. Here we show that therapeutic vaccination with Ad26/MVA (recombinant adenovirus serotype 26 (Ad26) prime, modified vaccinia Ankara (MVA) boost) and stimulation of TLR7 (Toll-like receptor 7) improves virologic control and delays viral rebound following discontinuation of antiretroviral therapy in SIV-infected rhesus monkeys that began antiretroviral therapy during acute infection. Therapeutic vaccination with Ad26/MVA resulted in a marked increase in the magnitude and breadth of SIV-specific cellular immune responses in virologically suppressed, SIV-infected monkeys. TLR7 agonist administration led to innate immune stimulation and cellular immune activation. The combination of Ad26/MVA vaccination and TLR7 stimulation resulted in decreased levels of viral DNA in lymph nodes and peripheral blood, and improved virologic control and delayed viral rebound following discontinuation of antiretroviral therapy. The breadth of cellular immune responses correlated inversely with set point viral loads and correlated directly with time to viral rebound. These data demonstrate the potential of therapeutic vaccination combined with innate immune stimulation as a strategy aimed at a functional cure for HIV-1 infection.


Antimicrobial Agents and Chemotherapy | 2009

Preclinical Evaluation of GS-9160, a Novel Inhibitor of Human Immunodeficiency Virus Type 1 Integrase

Gregg S. Jones; Fang Yu; Ameneh Zeynalzadegan; Joseph Hesselgesser; Xiaowu Chen; James K. Chen; Haolun Jin; Choung U. Kim; Matthew Blake Wright; Romas Geleziunas; Manuel Tsiang

ABSTRACT GS-9160 is a novel and potent inhibitor of human immunodeficiency virus type 1 (HIV-1) integrase (IN) that specifically targets the process of strand transfer. It is an authentic inhibitor of HIV-1 integration, since treatment of infected cells results in an elevation of two-long terminal repeat circles and a decrease of integration junctions. GS-9160 has potent and selective antiviral activity in primary human T lymphocytes producing a 50% effective concentration (EC50) of ∼2 nM, with a selectivity index (50% cytotoxic concentration/EC50) of ∼2,000. The antiviral potency of GS-9160 decreased by 6- to 10-fold in the presence of human serum. The antiviral activity of GS-9160 is synergistic in combination with representatives from three different classes of antiviral drugs, namely HIV-1 protease inhibitors, nonnucleoside reverse transcriptase inhibitors, and nucleotide reverse transcriptase inhibitors. Viral resistance selections performed with GS-9160 yielded a novel pattern of mutations within the catalytic core domain of IN; E92V emerged initially, followed by L74M. While E92V as a single mutant conferred 12-fold resistance against GS-9160, L74M had no effect as a single mutant. Together, these mutations conferred 67-fold resistance to GS-9160, indicating that L74M may potentiate the resistance caused by E92V. The pharmacokinetic profile of GS-9160 in healthy human volunteers revealed that once-daily dosing was not likely to achieve antiviral efficacy; hence, the clinical development of this compound was discontinued.


Journal of Medicinal Chemistry | 2013

Identification and Optimization of Pteridinone Toll-like Receptor 7 (TLR7) Agonists for the Oral Treatment of Viral Hepatitis

Paul A. Roethle; Ryan Mcfadden; Hong Yang; Paul Hrvatin; Hon C. Hui; Michael Graupe; Brian M. Gallagher; Jessica Jade Chao; Joseph Hesselgesser; Paul Duatschek; Jim Zheng; Daniel B. Tumas; Jason K. Perry; Randall L. Halcomb

Pteridinone-based Toll-like receptor 7 (TLR7) agonists were identified as potent and selective alternatives to the previously reported adenine-based agonists, leading to the discovery of GS-9620. Analogues were optimized for the immunomodulatory activity and selectivity versus other TLRs, based on differential induction of key cytokines including interferon α (IFN-α) and tumor necrosis factor α (TNF-α). In addition, physicochemical properties were adjusted to achieve desirable in vivo pharmacokinetic and pharmacodynamic properties. GS-9620 is currently in clinical evaluation for the treatment of chronic hepatitis B (HBV) infection.


Journal of Pharmacology and Experimental Therapeutics | 2013

Pharmacokinetic and Pharmacodynamic Properties of GS-9620, a Novel Toll-Like Receptor 7 Agonist, Demonstrate Interferon-Stimulated Gene Induction without Detectable Serum Interferon at Low Oral Doses

Abigail Fosdick; Jim Zheng; Stefan Pflanz; Christian R. Frey; Joseph Hesselgesser; Randall L. Halcomb; Grushenka H.I. Wolfgang; Daniel B. Tumas

GS-9620 [8-(3-(pyrrolidin-1-ylmethyl)benzyl)-4-amino-2-butoxy-7,8-dihydropteridin-6(5H)-one] is a potent, orally bioavailable small-molecule agonist of Toll-like receptor 7 (TLR7) developed for finite treatment of chronic hepatitis B viral (HBV) infection, with the goal of inducing a liver-targeted antiviral effect without inducing the adverse effects associated with current systemic interferon-α (IFN-α) therapies. We characterized the pharmacodynamic response of GS-9620 in CD-1 mice and cynomolgus monkeys following intravenous or oral administration and showed that GS-9620 induces the production of select chemokines and cytokines, including IFN-α and interferon-stimulated genes (ISGs). It is noteworthy that we also demonstrated that, in animals and healthy human volunteers, oral administration of GS-9620 can induce a type I interferon-dependent antiviral innate immune response, as measured by whole-blood mRNA of the ISGs 2′5′-oligoadenylate synthetase 1 (OAS1) and myxovirus resistance 1 (MX1), without the induction of detectable systemic IFN-α, i.e., a presystemic response. Additionally, presystemic induction of hepatic OAS1 and MX1 mRNA was observed in CD-1 mice in the absence of detectable systemic IFN-α. We propose that the mechanism of this presystemic response is likely its high intestinal absorption, which facilitates localized activation of TLR7, probably in plasmacytoid dendritic cells at the level of gut-associated lymphoid tissue and/or the liver. This localized response is further supported by data that indicate only minimal contributions of systemic immune stimulation to the overall pharmacodynamic response to orally administered GS-9620. These data demonstrate that GS-9620 can induce an antiviral innate immune response without inducing a systemic IFN-α response and thus suggest the therapeutic potential of this approach in the treatment of chronic HBV infection.


Antimicrobial Agents and Chemotherapy | 2016

Elevated Plasma Viral Loads in Romidepsin-Treated Simian Immunodeficiency Virus-Infected Rhesus Macaques on Suppressive Combination Antiretroviral Therapy.

Gregory Q. Del Prete; Kelli Oswald; Abigail Lara; Rebecca Shoemaker; Jeremy Smedley; Rhonda Macallister; Vicky Coalter; Adam Wiles; Rodney Wiles; Yuan Li; Randy Fast; Rebecca Kiser; Jim Zheng; W. Gregory Alvord; Charles M. Trubey; Michael Piatak; Claire Deleage; Brandon F. Keele; Jacob D. Estes; Joseph Hesselgesser; Romas Geleziunas; Jeffrey D. Lifson

ABSTRACT Replication-competent human immunodeficiency virus (HIV) persists in infected people despite suppressive combination antiretroviral therapy (cART), and it represents a major obstacle to HIV functional cure or eradication. We have developed a model of cART-mediated viral suppression in simian human immunodeficiency virus (SIV) mac239-infected Indian rhesus macaques and evaluated the impact of the histone deacetylase inhibitor (HDACi) romidepsin (RMD) on viremia in vivo. Eight macaques virologically suppressed to clinically relevant levels (<30 viral RNA copies/ml of plasma), using a three-class five-drug cART regimen, received multiple intravenous infusions of either RMD (n = 5) or saline (n = 3) starting 31 to 54 weeks after cART initiation. In vivo RMD treatment resulted in significant transient increases in acetylated histone levels in CD4+ T cells. RMD-treated animals demonstrated plasma viral load measurements for each 2-week treatment cycle that were significantly higher than those in saline control-treated animals during periods of treatment, suggestive of RMD-induced viral reactivation. However, plasma virus rebound was indistinguishable between RMD-treated and control-treated animals for a subset of animals released from cART. These findings suggest that HDACi drugs, such as RMD, can reactivate residual virus in the presence of suppressive antiviral therapy and may be a valuable component of a comprehensive HIV functional cure/eradication strategy.


Antimicrobial Agents and Chemotherapy | 2017

TLR7 Agonist GS-9620 Is a Potent Inhibitor of Acute HIV-1 Infection in Human Peripheral Blood Mononuclear Cells

Rujuta A. Bam; Derek Hansen; Alivelu Irrinki; Andrew S. Mulato; Gregg S. Jones; Joseph Hesselgesser; Christian R. Frey; Tomas Cihlar; Stephen R. Yant

ABSTRACT GS-9620 is a potent and selective oral Toll-like receptor 7 (TLR7) agonist that directly activates plasmacytoid dendritic cells (pDCs). GS-9620 suppressed hepatitis B virus (HBV) in animal models of chronic infection and transiently activated HIV expression ex vivo in latently infected peripheral blood mononuclear cells (PBMCs) from virally suppressed patients. Currently, GS-9620 is under clinical evaluation for treating chronic HBV infection and for reducing latent reservoirs in virally suppressed HIV-infected patients. Here, we investigated the in vitro anti-HIV-1 activity of GS-9620. GS-9620 potently inhibited viral replication in PBMCs, particularly when it was added 24 to 48 h prior to HIV infection (50% effective concentration = 27 nM). Depletion of pDCs but not other immune cell subsets from PBMC cultures suppressed GS-9620 antiviral activity. Although GS-9620 was inactive against HIV in purified CD4+ T cells and macrophages, HIV replication was potently inhibited by conditioned medium derived from GS-9620-treated pDC cultures when added to CD4+ T cells prior to infection. This suggests that GS-9620-mediated stimulation of PBMCs induced the production of a soluble factor(s) inhibiting HIV replication in trans. GS-9620-treated PBMCs primarily showed increased production of interferon alpha (IFN-α), and cotreatment with IFN-α-blocking antibodies reversed the HIV-1-inhibitory effect of GS-9620. Additional studies demonstrated that GS-9620 inhibited a postentry event in HIV replication at a step coincident with or prior to reverse transcription. The simultaneous activation of HIV-1 expression and inhibition of HIV-1 replication are important considerations for the clinical evaluation of GS-9620 since these antiviral effects may help restrict potential local HIV spread upon in vivo latency reversal.


Nature Medicine | 2018

Early antiretroviral therapy limits SIV reservoir establishment to delay or prevent post-treatment viral rebound

Afam A. Okoye; Scott G. Hansen; Mukta Vaidya; Yoshinori Fukazawa; Haesun Park; Derick M. Duell; Richard Lum; Colette M. Hughes; Abigail B. Ventura; Emily Ainslie; Julia C. Ford; David W. Morrow; Roxanne M. Gilbride; Alfred W. Legasse; Joseph Hesselgesser; Romas Geleziunas; Yuan Li; Kelli Oswald; Rebecca Shoemaker; Randy Fast; William J. Bosche; Bhavesh R. Borate; Paul T. Edlefsen; Michael K. Axthelm; Louis J. Picker; Jeffrey D. Lifson

Prophylactic vaccination of rhesus macaques with rhesus cytomegalovirus (RhCMV) vectors expressing simian immunodeficiency virus (SIV) antigens (RhCMV/SIV) elicits immune responses that stringently control highly pathogenic SIV infection, with subsequent apparent clearance of the infection, in ~50% of vaccinees. In contrast, here, we show that therapeutic RhCMV/SIV vaccination of rhesus macaques previously infected with SIV and given continuous combination antiretroviral therapy (cART) beginning 4–9 d post-SIV infection does not mediate measurable SIV reservoir clearance during over 600 d of follow-up on cART relative to RhCMV/control vaccination. However, none of the six animals started on cART on day four or five, across both RhCMV/SIV- and RhCMV/control-vaccinated groups, those rhesus macaques with SIV reservoirs most closely resembling those of prophylactically RhCMV/SIV-vaccinated and protected animals early in their course, showed post-cART viral rebound with up to nine months of follow-up. Moreover, at necropsy, these rhesus macaques showed little to no evidence of replication-competent SIV. These results suggest that the early SIV reservoir is limited in durability and that effective blockade of viral replication and spread in this critical time window by either pharmacologic or immunologic suppression may result in reduction, and potentially loss, of rebound-competent virus over a period of ~two years.Early and prolonged administration of antiretroviral therapy to SIV-infected and post-exposure-vaccinated rhesus macaques was associated with absence or delay of detectable virus after therapy interruption.


Nature Medicine | 2015

B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers

Yoshinori Fukazawa; Richard Lum; Afam A. Okoye; Haesun Park; Kenta Matsuda; Jin Young Bae; Shoko I. Hagen; Rebecca Shoemaker; Claire Deleage; Carissa Lucero; David R. Morcock; Tonya Swanson; Alfred W. Legasse; Michael K. Axthelm; Joseph Hesselgesser; Romas Geleziunas; Vanessa M. Hirsch; Paul T. Edlefsen; Michael Piatak; Jacob D. Estes; Jeffrey D. Lifson; Louis J. Picker

Collaboration


Dive into the Joseph Hesselgesser's collaboration.

Top Co-Authors

Avatar

Dan H. Barouch

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Derek D. Sloan

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Erica N. Borducchi

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jinyan Liu

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lauren Peter

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge