87 results on '"D. Hazuda"'
Search Results
2. Cellular modulation and HIV reactivation in response to serial treatment of latently HIV infected CD4 T cells with histone deacetylase inhibitors (HDACi)
- Author
-
J. Maxwell, A. Nefedov, C. Dorfmeier, G. Wu, Y. Li, P. Zuck, D. Hazuda, A. Webber, R. Barnard, and B. Howell
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2019
- Full Text
- View/download PDF
3. CD32+ CD4+ T Cells are HIV transcriptionally active rather than a resting reservoir
- Author
-
M. Abdel-Mohsen, C. Tomescu, S. Vadrevu, A. Spivak, L. Kuri-Cervantes, G. Wu, K. Cox, S. Vemula, M. Fair, K. Lynn, M.J. Buzon, J. Martinez-Picado, M. Betts, V. Planelles, K. Mounzer, B. Howell, D. Hazuda, P. Tebas, and L.J. Montaner
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2017
- Full Text
- View/download PDF
4. Higher rectal p24 levels correlate with poor CD4 recovery in treated HIV infection
- Author
-
B.J. Howell, G. Wu, S.L. Goh, P. Zuck, M. Pao, M. Deswal, R. Hoh, J.N. Martin, S.G. Deeks, M. Somsouk, D. Hazuda, and P.W. Hunt
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2017
- Full Text
- View/download PDF
5. The contribution of memory CD4+ T cell subset phenotype to latency reversal efficiency
- Author
-
D.A. Kulpa, A. Talla, S. Ribeiro, R. Barnard, D. Hazuda, N. Chomont, and R. Pierre Sékaly
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2017
- Full Text
- View/download PDF
6. In vitro analysis of different PKC agonists: latency reversal, T-cell activation, cytokine production and isoform selectivity
- Author
-
R. Barnard, D. Tellers, B. Howell, E. Cook, M. Swanson, S. Vemula, J. Li, S. Carroll, and D. Hazuda
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2015
- Full Text
- View/download PDF
7. Memory CD4+ T cell subsets show differential responses to HIV latency-reversing agents
- Author
-
D.A. Kulpa, S. Yuan, A. Talla, A.G. Bebein-Blackwell, J. Brehm, R. Barnard, M. Miller, D. Hazuda, N. Chomont, and R.P. Sékaly
- Subjects
Microbiology ,QR1-502 ,Public aspects of medicine ,RA1-1270 - Published
- 2015
- Full Text
- View/download PDF
8. X-ray structure of simian immunodeficiency virus integrase containing the core and C-terminal domain (residues 50-293)--an initial glance of the viral DNA binding platform
- Author
-
Z, Chen, Y, Yan, S, Munshi, Y, Li, J, Zugay-Murphy, B, Xu, M, Witmer, P, Felock, A, Wolfe, V, Sardana, E A, Emini, D, Hazuda, and L C, Kuo
- Subjects
Models, Molecular ,Binding Sites ,Integrases ,Glutamine ,Molecular Sequence Data ,DNA ,HIV Integrase ,Crystallography, X-Ray ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Solutions ,Avian Sarcoma Viruses ,Catalytic Domain ,Simian Immunodeficiency Virus ,Amino Acid Sequence ,Crystallization ,Protein Structure, Quaternary ,Dimerization - Abstract
The crystal structure of simian immunodeficiency virus (SIV) integrase that contains in a single polypeptide the core and the C-terminal deoxyoligonucleotide binding domain has been determined at 3 A resolution with an R-value of 0.203 in the space group P2(1)2(1)2(1). Four integrase core domains and one C-terminal domain are found to be well defined in the asymmetric unit. The segment extending from residues 114 to 121 assumes the same position as seen in the integrase core domain of avian sarcoma virus as well as human immunodeficiency virus type-1 (HIV-1) crystallized in the absence of sodium cacodylate. The flexible loop in the active site, composed of residues 141-151, remains incompletely defined, but the location of the essential Glu152 residue is unambiguous. The residues from 210-218 that link the core and C-terminal domains can be traced as an extension from the core with a short gap at residues 214-215. The C(alpha) folding of the C-terminal domain is similar to the solution structure of this domain from HIV-1 integrase. However, the dimeric form seen in the NMR structure cannot exist as related by the non-crystallographic symmetry in the SIV integrase crystal. The two flexible loops of the C-terminal domain, residues 228-236 and residues 244-249, are much better fixed in the crystal structure than in the NMR structure with the former in the immediate vicinity of the flexible loop of the core domain. The interface between the two domains encompasses a solvent-exclusion area of 1500 A(2). Residues from both domains purportedly involved in DNA binding are narrowly distributed on the same face of the molecule. They include Asp64, Asp116, Glu152 and Lys159 from the core and Arg231, Leu234, Arg262, Arg263 and Lys264 from the C-terminal domain. A model for DNA binding is proposed to bridge the two domains by tethering the 228-236 loop of the C-terminal domain and the flexible loop of the core.
- Published
- 2000
9. Discovery and analysis of inhibitors of the human immunodeficiency integrase
- Author
-
D, Hazuda, P J, Felock, J C, Hastings, B, Pramanik, and A L, Wolfe
- Subjects
Cell-Free System ,Cations, Divalent ,Virus Assembly ,DNA, Viral ,HIV-1 ,Methods ,Drug Evaluation ,Humans ,HIV Integrase Inhibitors ,Virus Replication ,Antiviral Agents - Abstract
An essential step in the replication of retroviruses is the integration of a DNA copy of the viral genome into the genome of the host cell. Integration encompasses a series of ordered endonucleolytic and DNA strand transfer reactions catalyzed by the viral enzyme, integrase. The requirement for integrase activity in the propagation of HIV-1 in cell culture defines the enzyme as a potential target for chemotherapeutic intervention. We have therefore developed a non-radioisotopic microtiter plate assay which can be used to identify novel inhibitors of integrase from random chemical screens and for the bioassay driven isolation of inhibitors from natural products. This assay uncouples various steps in the reaction pathway and therefore can be exploited to characterize inhibitors. In this monograph we describe a series of modifications to the method which facilitate such mechanistic studies using as an example a series of previously described integrase inhibitors.
- Published
- 1997
10. A system to analyze and identify inhibitors of HIV-1 gene regulation using a defective integrated provirus
- Author
-
V, Byrnes and D, Hazuda
- Subjects
Gene Expression Regulation, Viral ,Transcriptional Activation ,Base Sequence ,Recombinant Fusion Proteins ,Virus Integration ,Molecular Sequence Data ,Defective Viruses ,Genome, Viral ,Oligonucleotides, Antisense ,Transfection ,Jurkat Cells ,Proviruses ,Genes, Reporter ,Genes, tat ,COS Cells ,Gene Products, tat ,HIV-1 ,Animals ,Humans ,tat Gene Products, Human Immunodeficiency Virus ,Cloning, Molecular ,Luciferases - Published
- 1996
11. Evolution of the D-ribose operon on Escherichia coli B/r
- Author
-
M Abou-Sabé, D Hazuda, A Ninfa, and J Pilla
- Subjects
Transposable element ,Operon ,Ribose ,Biology ,medicine.disease_cause ,Microbiology ,Plasmid ,Transduction, Genetic ,Extrachromosomal DNA ,Genes, Regulator ,Escherichia coli ,medicine ,Molecular Biology ,Genetics ,Genetic Complementation Test ,Phosphotransferases ,Chromosome Mapping ,DNA Restriction Enzymes ,Chromosomes, Bacterial ,Molecular biology ,PBR322 ,Complementation ,Phosphotransferases (Alcohol Group Acceptor) ,Enzyme Induction ,DNA Transposable Elements ,bacteria ,L-arabinose operon ,Plasmids ,Research Article - Abstract
The D-ribose operon (rbs) of Escherichia coli K-12 maps at 83 min and is inducible. The rbs operon of E. coli B/r maps at 2 min and is constitutive. Evidence is presented showing that a second inducible copy of the rbs operons is present in E. coli B/r mapping at 83 min. The data indicated that the duplication of the rbs operon represented a transposition of the 83-min region to 2 min. The identification of a second copy of the rbs operon in B/r and the determination of its inducibility were based on the reactivation, through mutagenesis, of inducible rbs expression, mapping by P1 transduction of the mutation site to 83 min, and merodiploid complementation analysis of the D-ribokinase expression in E. coli B/r. We also show that the rbs transposition to 2-min continued to generate transposable elements coding for the 1- to 2-min region of the chromosome and transposing onto extrachromosomal DNA target molecules such as pBR322.
- Published
- 1982
- Full Text
- View/download PDF
12. Impact of HIV-1 Resistance-Associated Mutations on Susceptibility to Doravirine: Analysis of Real-World Clinical Isolates.
- Author
-
Asante-Appiah E, Lai J, Wan H, Yang D, Martin EA, Sklar P, Hazuda D, Petropoulos CJ, Walworth C, and Grobler JA
- Subjects
- Drug Resistance, Viral genetics, HIV Reverse Transcriptase genetics, Humans, Mutation, Pyridones, Reverse Transcriptase Inhibitors pharmacology, Reverse Transcriptase Inhibitors therapeutic use, Triazoles, Anti-HIV Agents pharmacology, Anti-HIV Agents therapeutic use, HIV Infections drug therapy, HIV-1 genetics
- Abstract
Clinical management of human immunodeficiency virus type-1 (HIV-1) infection may be negatively impacted by either acquired or transmitted drug resistance. Here, we aim to extend our understanding of the impact of resistance-associated mutations (RAMs) on the susceptibility of clinical isolates to the nonnucleoside reverse transcriptase inhibitor (NNRTI) doravirine. Clinical isolates from people living with HIV-1 undergoing routine testing for susceptibility to doravirine and other approved NNRTIs (etravirine, rilpivirine, efavirenz, and nevirapine) were collected from August 2018 to August 2019. Susceptibility in the presence/absence of NNRTI and nucleos(t)ide reverse transcriptase inhibitor (NRTI) mutations was determined using cutoffs for relative fold change in inhibition (ratio of the 50% inhibitory concentration [IC
50 ] of patient virus compared with the IC50 of a wild-type reference strain). Biological cutoffs of 3- to 15-fold change were investigated for doravirine, with preestablished cutoffs used for the other NNRTIs. Of 4,070 clinical isolates, 42.9% had ≥1 NNRTI RAM. More isolates were susceptible to doravirine (92.5-96.7%) than to etravirine (91.5%), rilpivirine (89.5%), efavirenz (81.5%), or nevirapine (77.5%). Based on a 3-fold cutoff, doravirine susceptibility was retained in 44.7-65.8% of isolates resistant to another NNRTI and 28.5% of isolates resistant to all other tested NNRTIs. The presence of NRTI RAMs, including thymidine analog mutations, was associated with doravirine hypersusceptibility in some isolates, particularly in the absence of NNRTI RAMs. These results support the favorable resistance profile of doravirine and are of particular importance given the challenge posed by both acquired and transmitted resistance.- Published
- 2021
- Full Text
- View/download PDF
13. Letter to the Editor in Response to Zhou et al.
- Author
-
Troth S, Butterton J, DeAnda CS, Escobar P, Grobler J, Hazuda D, and Painter G
- Published
- 2021
- Full Text
- View/download PDF
14. In Vitro Pharmacokinetic/Pharmacodynamic Modeling of HIV Latency Reversal by Novel HDAC Inhibitors Using an Automated Platform.
- Author
-
Newhard W, Patel M, Cassaday J, Ballard J, Squadroni B, Wu G, Liu J, Yu W, Kozlowski J, Zuck P, Howell B, Hazuda D, Vargo R, and Barnard R
- Subjects
- Automation, Laboratory, Cell Culture Techniques, Cells, Cultured, Gene Expression Regulation, Viral drug effects, HIV genetics, Humans, Jurkat Cells, Virus Replication drug effects, HIV drug effects, HIV Infections drug therapy, HIV Infections virology, Histone Deacetylase Inhibitors pharmacokinetics, Models, Theoretical, Virus Latency drug effects
- Abstract
Antiretroviral therapy is able to effectively control but not eradicate HIV infection, which can persist, leading to the need for lifelong therapy. The existence of latently HIV-infected cells is a major barrier to the eradication of chronic HIV infection. Histone deacetylase inhibitors (HDACis), small molecules licensed for oncology indications, have shown the ability to produce HIV transcripts in vitro and in vivo. The pharmacologic parameters that drive optimal HIV latency reversal in vivo are unknown and could be influenced by such factors as the HDACi binding kinetics, concentration of compound, and duration of exposure. This study evaluates how these parameters affect HIV latency reversal for a series of novel HDACis that differ in their enzymatic on and off rates. Varying cellular exposure, using automated washout methods of HDACi in a Jurkat cell model of HIV latency, led to the investigation of the relationship between pharmacokinetic (PK) properties, target engagement (TE), and pharmacodynamic (PD) responses. Using an automated robotic platform enabled miniaturization of a suspension cell-based washout assay that required multiple manipulations over the 48 h duration of the assay. Quantification of histone acetylation (TE) revealed that HDACis showed early peaks and differences in the durability of response between different investigated HDACis. By expanding the sample times, the shift between TE and PD, as measured by green fluorescent protein, could be fully characterized. The comprehensive data set generated by automating the assays described here was used to establish a PK/PD model for HDACi-induced HIV latency reversal.
- Published
- 2021
- Full Text
- View/download PDF
15. Recommendations for measuring HIV reservoir size in cure-directed clinical trials.
- Author
-
Abdel-Mohsen M, Richman D, Siliciano RF, Nussenzweig MC, Howell BJ, Martinez-Picado J, Chomont N, Bar KJ, Yu XG, Lichterfeld M, Alcami J, Hazuda D, Bushman F, Siliciano JD, Betts MR, Spivak AM, Planelles V, Hahn BH, Smith DM, Ho YC, Buzon MJ, Gaebler C, Paiardini M, Li Q, Estes JD, Hope TJ, Kostman J, Mounzer K, Caskey M, Fox L, Frank I, Riley JL, Tebas P, and Montaner LJ
- Subjects
- CD4-Positive T-Lymphocytes virology, Clinical Trials as Topic, Humans, Mass Screening methods, Viral Load drug effects, Virus Latency drug effects, Anti-Retroviral Agents therapeutic use, Disease Reservoirs virology, HIV Infections diagnosis, HIV Infections drug therapy, HIV-1 drug effects
- Abstract
Therapeutic strategies are being clinically tested either to eradicate latent HIV reservoirs or to achieve virologic control in the absence of antiretroviral therapy. Attaining this goal will require a consensus on how best to measure the numbers of persistently infected cells with the potential to cause viral rebound after antiretroviral-therapy cessation in assessing the results of cure-directed strategies in vivo. Current measurements assess various aspects of the HIV provirus and its functionality and produce divergent results. Here, we provide recommendations from the BEAT-HIV Martin Delaney Collaboratory on which viral measurements should be prioritized in HIV-cure-directed clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
16. Immunogenicity generated by mRNA vaccine encoding VZV gE antigen is comparable to adjuvanted subunit vaccine and better than live attenuated vaccine in nonhuman primates.
- Author
-
Monslow MA, Elbashir S, Sullivan NL, Thiriot DS, Ahl P, Smith J, Miller E, Cook J, Cosmi S, Thoryk E, Citron M, Thambi N, Shaw C, Hazuda D, and Vora KA
- Subjects
- Animals, Antibodies, Viral, Herpesvirus 3, Human, RNA, Messenger, Vaccines, Attenuated, Vaccines, Subunit, Viral Envelope Proteins, Herpes Zoster, Herpes Zoster Vaccine
- Abstract
Shingles is a painful, blistering rash caused by reactivation of latent varicella-zoster virus (VZV) and most frequently occurs in elderly and immunocompromised individuals. Currently, two approved vaccines for the prevention of shingles are on the market, a live attenuated virus vaccine ZOSTAVAX® (Merck & Co., Inc., Kenilworth, NJ, USA) and an AS01
B adjuvanted subunit protein vaccine Shingrix™ (Glaxo Smith Kline, Rockville, MD, USA). Human clinical immunogenicity and vaccine efficacy data is available for these two benchmark vaccines, offering a unique opportunity for comparative analyses with novel vaccine platforms and animal model translatability studies. The studies presented here utilized non-human primates (NHP) to evaluate humoral and cellular immune response by three vaccine modalities: the new platform of lipid nanoparticle (LNP) formulated mRNA encoding VZV gE antigen (VZV gE mRNA/LNP) as compared with well-established platforms of live attenuated VZV (VZV LAV) and adjuvanted VZV gE subunit protein (VZV gE protein/adjuvant). The magnitude of response to vaccination with a single 100-200 μg mRNA dose or two 50 μg mRNA doses of VZV gE mRNA/LNP were comparable to two 50 μg protein doses of VZV gE protein/adjuvant, suggesting the VZV gE mRNA/LNP platform has the potential to elicit a robust immune response, and both modalities generated markedly higher responses than VZV LAV. Additionally, the slopes of decay for VZV-specific antibody titers were roughly similar across all three vaccines, indicating the magnitude of peak immunogenicity was the driving force in determining immune response longevity. Finally, vaccine-induced immunogenicity with VZV LAV and VZV gE protein/adjuvant in NHP closely resembled human clinical trials immune response data for ZOSTAVAX® and Shingrix™, helping to validate NHP as an appropriate preclinical model for evaluating these vaccines., Competing Interests: Declaration of Competing Interest All authors are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Moderna, Cambridge, MA, USA, and Eurofins Lancaster Laboratories Professional Scientific Services, Lancaster, PA, USA., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
17. Modified mRNA/lipid nanoparticle-based vaccines expressing respiratory syncytial virus F protein variants are immunogenic and protective in rodent models of RSV infection.
- Author
-
Espeseth AS, Cejas PJ, Citron MP, Wang D, DiStefano DJ, Callahan C, Donnell GO, Galli JD, Swoyer R, Touch S, Wen Z, Antonello J, Zhang L, Flynn JA, Cox KS, Freed DC, Vora KA, Bahl K, Latham AH, Smith JS, Gindy ME, Ciaramella G, Hazuda D, Shaw CA, and Bett AJ
- Abstract
The RSV Fusion (F) protein is a target for neutralizing antibody responses and is a focus for vaccine discovery; however, the process of RSV entry requires F to adopt a metastable prefusion form and transition to a more stable postfusion form, which displays less potent neutralizing epitopes. mRNA vaccines encode antigens that are translated by host cells following vaccination, which may allow conformational transitions similar to those observed during natural infection to occur. Here we evaluate a panel of chemically modified mRNA vaccines expressing different forms of the RSV F protein, including secreted, membrane associated, prefusion-stabilized, and non-stabilized structures, for conformation, immunogenicity, protection, and safety in rodent models. Vaccination with mRNA encoding native RSV F elicited antibody responses to both prefusion- and postfusion-specific epitopes, suggesting that this antigen may adopt both conformations in vivo. Incorporating prefusion stabilizing mutations further shifts the immune response toward prefusion-specific epitopes, but does not impact neutralizing antibody titer. mRNA vaccine candidates expressing either prefusion stabilized or native forms of RSV F protein elicit robust neutralizing antibody responses in both mice and cotton rats, similar to levels observed with a comparable dose of adjuvanted prefusion stabilized RSV F protein. In contrast to the protein subunit vaccine, mRNA-based vaccines elicited robust CD4+ and CD8+ T-cell responses in mice, highlighting a potential advantage of the technology for vaccines requiring a cellular immune response for efficacy., Competing Interests: Competing interestsA.S.E., P.J.C., M.P.C., D.W., D.J.D., C.C., G.O., J.G., R.S., S.T., Z.W., J.A., L.Z., J.A.F., K.S.C., D.C.F., K.V., A.H.L., J.S.S., M.E.G., D.H., A.J.B. are all employees and/or stockholders of Merck & Co., Inc., Kenilworth, NJ, USA. C.A.S., K.B., and G.C. are employees and/or stockholders of Moderna Inc., Cambridge, MA, USA. G.C., K.B., A.S.E., D.W. and A.J.B. are co-inventors on a patent application related to this work.40, (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
18. Rational Design of Doravirine: From Bench to Patients.
- Author
-
Hwang C, Lai MT, and Hazuda D
- Subjects
- Anti-HIV Agents therapeutic use, Clinical Trials as Topic, Drug Interactions, HIV-1 drug effects, Humans, Pyridones therapeutic use, Reverse Transcriptase Inhibitors therapeutic use, Translational Research, Biomedical, Triazoles therapeutic use, Anti-HIV Agents chemistry, Drug Design, Drug Resistance, Viral, HIV Infections drug therapy, Pyridones chemistry, Reverse Transcriptase Inhibitors chemistry, Triazoles chemistry
- Abstract
Since the approval of nevirapine, the first HIV-1 non-nucleoside reverse transcriptase inhibitor (NNRTI) in 1996, NNRTIs have helped play a critical role in maintaining viral suppression in people living with HIV. The many positive attributes of the class, including potency and long plasma half-life, make them attractive drug discovery targets. Given the availability of multiple once-daily integrase-based treatments for HIV-1 infection, the challenge to develop a new antiretroviral agent that addresses the needs of today's patients is formidable. However, with the increased availability of antiretrovirals for treatment and new pre-exposure prophylaxis guidelines, which should globally expand the use of antiretrovirals in prevention, it will be increasingly important to have access to multiple regimens with options from different classes that are well tolerated and convenient to ensure a sustained impact on the global epidemic. Many attempts to improve upon the NNRTI class have failed to deliver a desirable clinical profile consistent with the current landscape of treatment options. Doravirine is the only NNRTI to successfully advance through phase 3 clinical development and approval in recent years. Learning from the liabilities of approved NNRTIs, as well as past development failures, facilitated a rational approach to the discovery of doravirine by focusing on addressing the known safety/tolerability issues of commonly prescribed NNRTIs, such as central nervous system toxicity with efavirenz and potential cardiotoxicity due to off-target effects on cardiac ion channels with rilpivirine, using structural biology and characterization of resistance in vitro to address resistance liabilities and concentrating on the metabolic profile to limit the potential for drug-drug interactions. These preclinical efforts were critical to the design and selection of doravirine as a novel NNRTI that possessed the desired next-generation profile with the ultimate proof that these attributes translate to patients derived from clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
19. Differentiation into an Effector Memory Phenotype Potentiates HIV-1 Latency Reversal in CD4 + T Cells.
- Author
-
Kulpa DA, Talla A, Brehm JH, Ribeiro SP, Yuan S, Bebin-Blackwell AG, Miller M, Barnard R, Deeks SG, Hazuda D, Chomont N, and Sékaly RP
- Subjects
- DNA, Viral genetics, Gene Expression, Humans, Immunologic Memory physiology, T-Lymphocyte Subsets virology, CD4-Positive T-Lymphocytes virology, Cell Differentiation, HIV Infections virology, HIV-1 physiology, Phenotype, Virus Latency physiology
- Abstract
During antiretroviral therapy (ART), human immunodeficiency virus type 1 (HIV-1) persists as a latent reservoir in CD4
+ T cell subsets in central memory (TCM ), transitional memory (TTM ), and effector memory (TEM ) CD4+ T cells. We have identified differences in mechanisms underlying latency and responses to latency-reversing agents (LRAs) in ex vivo CD4+ memory T cells from virally suppressed HIV-infected individuals and in an in vitro primary cell model of HIV-1 latency. Our ex vivo and in vitro results demonstrate the association of transcriptional pathways of T cell differentiation, acquisition of effector function, and cell cycle entry in response to LRAs. Analyses of memory cell subsets showed that effector memory pathways and cell surface markers of activation and proliferation in the TEM subset are predictive of higher frequencies of cells carrying an inducible reservoir. Transcriptional profiling also demonstrated that the epigenetic machinery (known to control latency and reactivation) in the TEM subset is associated with frequencies of cells with HIV-integrated DNA and inducible HIV multispliced RNA. TCM cells were triggered to differentiate into TEM cells when they were exposed to LRAs, and this increase of TEM subset frequencies upon LRA stimulation was positively associated with higher numbers of p24+ cells. Together, these data highlight differences in underlying biological latency control in different memory CD4+ T cell subsets which harbor latent HIV in vivo and support a role for differentiation into a TEM phenotype in facilitating latency reversal. IMPORTANCE By performing phenotypic analysis of latency reversal in CD4+ T cells from virally suppressed individuals, we identify the TEM subset as the largest contributor to the inducible HIV reservoir. Differential responses of memory CD4+ T cell subsets to latency-reversing agents (LRAs) demonstrate that HIV gene expression is associated with heightened expression of transcriptional pathways associated with differentiation, acquisition of effector function, and cell cycle entry. In vitro modeling of the latent HIV reservoir in memory CD4+ T cell subsets identify LRAs that reverse latency with ranges of efficiency and specificity. We found that therapeutic induction of latency reversal is associated with upregulation of identical sets of TEM -associated genes and cell surface markers shown to be associated with latency reversal in our ex vivo and in vitro models. Together, these data support the idea that the effector memory phenotype supports HIV latency reversal in CD4+ T cells., (Copyright © 2019 Kulpa et al.)- Published
- 2019
- Full Text
- View/download PDF
20. CD32 is expressed on cells with transcriptionally active HIV but does not enrich for HIV DNA in resting T cells.
- Author
-
Abdel-Mohsen M, Kuri-Cervantes L, Grau-Exposito J, Spivak AM, Nell RA, Tomescu C, Vadrevu SK, Giron LB, Serra-Peinado C, Genescà M, Castellví J, Wu G, Del Rio Estrada PM, González-Navarro M, Lynn K, King CT, Vemula S, Cox K, Wan Y, Li Q, Mounzer K, Kostman J, Frank I, Paiardini M, Hazuda D, Reyes-Terán G, Richman D, Howell B, Tebas P, Martinez-Picado J, Planelles V, Buzon MJ, Betts MR, and Montaner LJ
- Subjects
- Anti-Retroviral Agents therapeutic use, CD4-Positive T-Lymphocytes metabolism, HIV Infections drug therapy, Humans, In Vitro Techniques, Lymphocytes metabolism, Receptors, CCR4 metabolism, Receptors, CCR6 metabolism, Receptors, CXCR3 metabolism, HIV Infections metabolism, Receptors, IgG metabolism
- Abstract
The persistence of HIV reservoirs, including latently infected, resting CD4
+ T cells, is the major obstacle to cure HIV infection. CD32a expression was recently reported to mark CD4+ T cells harboring a replication-competent HIV reservoir during antiretroviral therapy (ART) suppression. We aimed to determine whether CD32 expression marks HIV latently or transcriptionally active infected CD4+ T cells. Using peripheral blood and lymphoid tissue of ART-treated HIV+ or SIV+ subjects, we found that most of the circulating memory CD32+ CD4+ T cells expressed markers of activation, including CD69, HLA-DR, CD25, CD38, and Ki67, and bore a TH 2 phenotype as defined by CXCR3, CCR4, and CCR6. CD32 expression did not selectively enrich for HIV- or SIV-infected CD4+ T cells in peripheral blood or lymphoid tissue; isolated CD32+ resting CD4+ T cells accounted for less than 3% of the total HIV DNA in CD4+ T cells. Cell-associated HIV DNA and RNA loads in CD4+ T cells positively correlated with the frequency of CD32+ CD69+ CD4+ T cells but not with CD32 expression on resting CD4+ T cells. Using RNA fluorescence in situ hybridization, CD32 coexpression with HIV RNA or p24 was detected after in vitro HIV infection (peripheral blood mononuclear cell and tissue) and in vivo within lymph node tissue from HIV-infected individuals. Together, these results indicate that CD32 is not a marker of resting CD4+ T cells or of enriched HIV DNA-positive cells after ART; rather, CD32 is predominately expressed on a subset of activated CD4+ T cells enriched for transcriptionally active HIV after long-term ART., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2018
- Full Text
- View/download PDF
21. Long-acting formulations for the treatment of latent tuberculous infection: opportunities and challenges.
- Author
-
Swindells S, Siccardi M, Barrett SE, Olsen DB, Grobler JA, Podany AT, Nuermberger E, Kim P, Barry CE, Owen A, Hazuda D, and Flexner C
- Subjects
- Antitubercular Agents chemistry, Humans, Antitubercular Agents therapeutic use, Delayed-Action Preparations, Latent Tuberculosis drug therapy
- Abstract
Long-acting/extended-release drug formulations have proved very successful in diverse areas of medicine, including contraception, psychiatry and, most recently, human immunodeficiency virus (HIV) disease. Though challenging, application of this technology to anti-tuberculosis treatment could have substantial impact. The duration of treatment required for all forms of tuberculosis (TB) put existing regimens at risk of failure because of early discontinuations and treatment loss to follow-up. Long-acting injections, for example, administered every month, could improve patient adherence and treatment outcomes. We review the state of the science for potential long-acting formulations of existing tuberculosis drugs, and propose a target product profile for new formulations to treat latent tuberculous infection (LTBI). The physicochemical properties of some anti-tuberculosis drugs make them unsuitable for long-acting formulation, but there are promising candidates that have been identified through modeling and simulation, as well as other novel agents and formulations in preclinical testing. An efficacious long-acting treatment for LTBI, particularly for those co-infected with HIV, and if coupled with a biomarker to target those at highest risk for disease progression, would be an important tool to accelerate progress towards TB elimination.
- Published
- 2018
- Full Text
- View/download PDF
22. Identification of proximal biomarkers of PKC agonism and evaluation of their role in HIV reactivation.
- Author
-
Vemula SV, Maxwell JW, Nefedov A, Wan BL, Steve J, Newhard W, Sanchez RI, Tellers D, Barnard RJ, Blair W, Hazuda D, Webber AL, and Howell BJ
- Subjects
- Biomarkers, CD4-Positive T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes virology, Cells, Cultured, Diterpenes chemistry, Diterpenes pharmacology, Drug Agonism, Early Growth Response Protein 1 genetics, Early Growth Response Protein 3 genetics, Gene Expression, HIV Infections virology, Humans, Jurkat Cells, Male, Phorbols pharmacology, Sequence Analysis, RNA, HIV-1 physiology, Protein Kinase C metabolism, Virus Activation drug effects, Virus Latency drug effects
- Abstract
Design: The HIV latent CD4
+ T cell reservoir is broadly recognized as a barrier to HIV cure. Induction of HIV expression using protein kinase C (PKC) agonists is one approach under investigation for reactivation of latently infected CD4+ T cells (Beans et al., 2013; Abreu et al., 2014; Jiang et al., 2014; Jiang and Dandekar, 2015). We proposed that an increased understanding of the molecular mechanisms of action of PKC agonists was necessary to inform on biological signaling and pharmacodynamic biomarkers. RNA sequencing (RNA Seq) was applied to identify genes and pathways modulated by PKC agonists., Methods: Human CD4+ T cells were treated ex vivo with Phorbol 12-myristate 13-acetate, prostatin or ingenol-3-angelate. At 3 h and 24 h post-treatment, cells were harvested and RNA-Seq was performed on RNA isolated from cell lysates. The genes differentially expressed across the PKC agonists were validated by quantitative RT-PCR (qPCR). A subset of genes was evaluated for their role in HIV reactivation using siRNA and CRISPR approaches in the Jurkat latency cell model., Results: Treatment of primary human CD4+ T cells with PKC agonists resulted in alterations in gene expression. qPCR of RNA Seq data confirmed upregulation of 24 genes, including CD69, Egr1, Egr2, Egr3, CSF2, DUSP5, and NR4A1. Gene knockdown of Egr1 and Egr3 resulted in reduced expression and decreased HIV reactivation in response to PKC agonist treatment, indicating a potential role for Egr family members in latency reversal., Conclusion: Overall, our results offer new insights into the mechanism of action of PKC agonists, biomarkers of pathway engagement, and the potential role of EGR family in HIV reactivation., (Copyright © 2016. Published by Elsevier B.V.)- Published
- 2017
- Full Text
- View/download PDF
23. The Combination of Grazoprevir, a Hepatitis C Virus (HCV) NS3/4A Protease Inhibitor, and Elbasvir, an HCV NS5A Inhibitor, Demonstrates a High Genetic Barrier to Resistance in HCV Genotype 1a Replicons.
- Author
-
Lahser FC, Bystol K, Curry S, McMonagle P, Xia E, Ingravallo P, Chase R, Liu R, Black T, Hazuda D, Howe AY, and Asante-Appiah E
- Subjects
- Amides, Benzofurans pharmacology, Carbamates, Cyclopropanes, Drug Therapy, Combination, Genotype, Hepacivirus drug effects, Imidazoles pharmacology, Mutation genetics, Replicon drug effects, Replicon genetics, Ribavirin pharmacology, Sulfonamides, Antiviral Agents pharmacology, Protease Inhibitors pharmacology, Quinoxalines pharmacology
- Abstract
The selection of resistance-associated variants (RAVs) against single agents administered to patients chronically infected with hepatitis C virus (HCV) necessitates that direct-acting antiviral agents (DAAs) targeting multiple viral proteins be developed to overcome failure resulting from emergence of resistance. The combination of grazoprevir (formerly MK-5172), an NS3/4A protease inhibitor, and elbasvir (formerly MK-8742), an NS5A inhibitor, was therefore studied in genotype 1a (GT1a) replicon cells. Both compounds were independently highly potent in GT1a wild-type replicon cells, with 90% effective concentration (EC90) values of 0.9 nM and 0.006 nM for grazoprevir and elbasvir, respectively. No cross-resistance was observed when clinically relevant NS5A and NS3 RAVs were profiled against grazoprevir and elbasvir, respectively. Kinetic analyses of HCV RNA reduction over 14 days showed that grazoprevir and elbasvir inhibited prototypic NS5A Y93H and NS3 R155K RAVs, respectively, with kinetics comparable to those for the wild-type GT1a replicon. In combination, grazoprevir and elbasvir interacted additively in GT1a replicon cells. Colony formation assays with a 10-fold multiple of the EC90 values of the grazoprevir-elbasvir inhibitor combination suppressed emergence of resistant colonies, compared to a 100-fold multiple for the independent agents. The selected resistant colonies with the combination harbored RAVs that required two or more nucleotide changes in the codons. Mutations in the cognate gene caused greater potency losses for elbasvir than for grazoprevir. Replicons bearing RAVs identified from resistant colonies showed reduced fitness for several cell lines and may contribute to the activity of the combination. These studies demonstrate that the combination of grazoprevir and elbasvir exerts a potent effect on HCV RNA replication and presents a high genetic barrier to resistance. The combination of grazoprevir and elbasvir is currently approved for chronic HCV infection., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Primer ID ultra-deep sequencing reveals dynamics of drug resistance-associated variants in breakthrough hepatitis C viruses: relevance to treatment outcome and resistance screening.
- Author
-
Barnard R, Chopra A, James I, Blinco J, Watson MW, Jabara CB, Hazuda D, Lemon SM, Mallal S, and Gaudieri S
- Subjects
- Hepatitis C, Chronic virology, High-Throughput Nucleotide Sequencing, Humans, Mutation, Polymerase Chain Reaction, Proline therapeutic use, Treatment Outcome, Viral Nonstructural Proteins genetics, Antiviral Agents therapeutic use, Drug Resistance, Viral genetics, Hepacivirus genetics, Hepatitis C, Chronic drug therapy, Proline analogs & derivatives
- Abstract
Background: Use of direct-acting antiviral drugs (DAAs) that target HCV may be hampered by the rapid selection of viral strains that harbour drug resistance-associated variants (RAVs). These RAVs are often associated with a fitness cost and tend to occur on low-frequency strains within treatment-naive subjects. To address the clinical relevance of low frequency RAVs in the setting of DAAs, this study utilized a Primer ID ultra-deep sequencing approach to mitigate PCR errors and bias to accurately quantify viral sequences in subjects that failed DAA treatment., Methods: Subjects were enrolled in the follow-up study P05063, and had previous treatment with boceprevir and all had detectable RAVs at virological failure (VF) based on Sanger-based population sequencing. Twelve subjects had three time points available: baseline, VF and follow-up (median 830.5 days). Viral RNA was amplified using unique primer identifiers (Primer IDs) and sequenced using 454 ultra-deep sequencing., Results: The sequencing strategy used in this study improved the detection of clinically relevant low frequency strains bearing RAVs compared to population sequencing and showed that these strains can persist for up to 2 years post-treatment failure. Strains carrying multiple RAVs were common in breakthrough viruses. Putative compensatory mutations were identified., Conclusions: The Primer ID ultra-deep sequencing approach identifies RAVs that can reduce drug sensitivity at levels below the detection threshold for population sequencing. The approach also removes PCR errors and biases, suggesting this sequencing strategy should become the standard approach by which to perform temporal quasispecies studies and resistance screening. ClinicalTrials.gov NCT00689390.
- Published
- 2016
- Full Text
- View/download PDF
25. Postexposure protection of macaques from vaginal SHIV infection by topical integrase inhibitors.
- Author
-
Dobard C, Sharma S, Parikh UM, West R, Taylor A, Martin A, Pau CP, Hanson DL, Lipscomb J, Smith J, Novembre F, Hazuda D, Garcia-Lerma JG, and Heneine W
- Subjects
- Administration, Intravaginal, Administration, Topical, Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Body Fluids, Dose-Response Relationship, Drug, Drug Resistance, Viral drug effects, Female, Gels, Integrase Inhibitors pharmacology, Kinetics, Macaca blood, Progesterone blood, Reverse Transcriptase Inhibitors pharmacology, Reverse Transcriptase Inhibitors therapeutic use, Simian Acquired Immunodeficiency Syndrome blood, Simian Acquired Immunodeficiency Syndrome virology, Simian Immunodeficiency Virus drug effects, Time Factors, Vagina drug effects, Vagina pathology, Viral Load drug effects, Virus Replication drug effects, Integrase Inhibitors therapeutic use, Macaca virology, Post-Exposure Prophylaxis, Simian Acquired Immunodeficiency Syndrome drug therapy, Simian Acquired Immunodeficiency Syndrome prevention & control, Simian Immunodeficiency Virus physiology, Vagina virology
- Abstract
Coitally delivered microbicide gels containing antiretroviral drugs are important for HIV prevention. However, to date, microbicides have contained entry or reverse transcriptase inhibitors that block early steps in virus infection and thus need to be given as a preexposure dose that interferes with sexual practices and may limit compliance. Integrase inhibitors block late steps after virus infection and therefore are more suitable for post-coital dosing. We first determined the kinetics of strand transfer in vitro and confirmed that integration begins about 6 hours after infection. We then used a repeat-challenge macaque model to assess efficacy of vaginal gels containing integrase strand transfer inhibitors when applied before or after simian/human immunodeficiency virus (SHIV) challenge. We showed that gel containing the strand transfer inhibitor L-870812 protected two of three macaques when applied 30 min before SHIV challenge. We next evaluated the efficacy of 1% raltegravir gel and demonstrated its ability to protect macaques when applied 3 hours after SHIV exposure (five of six protected; P < 0.05, Fisher's exact test). Breakthrough infections showed no evidence of drug resistance in plasma or vaginal secretions despite continued gel dosing after infection. We documented rapid vaginal absorption reflecting a short pharmacological lag time and noted that vaginal, but not plasma, virus load was substantially reduced in the breakthrough infection after raltegravir gel treatment. We provide a proof of concept that topically applied integrase inhibitors protect against vaginal SHIV infection when administered shortly before or 3 hours after virus exposure.
- Published
- 2014
- Full Text
- View/download PDF
26. Exploring possibilities.
- Author
-
Hazuda D, Miller M, and Berry J
- Subjects
- Drug Industry, Humans, HIV Infections, Research Personnel
- Published
- 2014
27. Next generation therapeutics.
- Author
-
Carter PJ, Hazuda D, and Wells JA
- Subjects
- Humans, Proteins metabolism, Proteins therapeutic use, Small Molecule Libraries pharmacology, Drug Discovery, Small Molecule Libraries therapeutic use
- Published
- 2013
- Full Text
- View/download PDF
28. Raltegravir once daily or twice daily in previously untreated patients with HIV-1: a randomised, active-controlled, phase 3 non-inferiority trial.
- Author
-
Eron JJ Jr, Rockstroh JK, Reynes J, Andrade-Villanueva J, Ramalho-Madruga JV, Bekker LG, Young B, Katlama C, Gatell-Artigas JM, Arribas JR, Nelson M, Campbell H, Zhao J, Rodgers AJ, Rizk ML, Wenning L, Miller MD, Hazuda D, DiNubile MJ, Leavitt R, Isaacs R, Robertson MN, Sklar P, and Nguyen BY
- Subjects
- Adult, Aged, CD4 Lymphocyte Count, Drug Administration Schedule, Female, HIV Infections mortality, HIV Infections virology, HIV Integrase Inhibitors pharmacokinetics, HIV Integrase Inhibitors therapeutic use, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Pyrrolidinones pharmacokinetics, Pyrrolidinones therapeutic use, Raltegravir Potassium, Treatment Outcome, Viral Load, Young Adult, HIV Infections drug therapy, HIV Integrase Inhibitors administration & dosage, HIV-1 drug effects, Pyrrolidinones administration & dosage
- Abstract
Background: Twice-daily raltegravir with once-daily tenofovir-emtricitabine is an effective initial antiretroviral regimen for patients with HIV-1. On the basis of pharmacokinetic data suggesting efficacy of once-daily raltegravir and because adherence is often improved with once-daily dosing, we aimed to compare these dosing schedules., Methods: In our international, double-blind, randomised, phase 3 non-inferiority study, we enrolled antiretroviral-naive patients with HIV RNA loads of more than 5000 copies per mL and no baseline resistance to tenofovir or emtricitabine at 83 centres worldwide. We randomly allocated patients (1:1) by use of a computer-generated sequence to receive raltegravir once daily (two 400 mg tablets taken together every 24 h), or twice daily (one 400 mg tablet every 12 h), both in combination with once-daily co-formulated tenofovir 300 mg plus emtricitabine 150 mg. The primary outcome was virological response at 48 weeks (viral RNA loads <50 copies per mL) in patients who received at least one dose of study drug, counting non-completers as failure. We assessed non-inferiority in terms of the proportion of patients in both treatment groups who achieved the primary outcome, with a non-inferiority margin of -10%. This study is registered with ClinicalTrials.gov, number NCT00745823., Findings: From Oct 15, 2008, to Nov 2, 2009, we randomly allocated 775 patients, of whom 382 (99%) of 386 patients in the once-daily group and 388 (99%) of 389 in the twice-daily group received at least one dose of study drug. At baseline, 304 (39%) of 770 treated patients had viral loads of more than 100,000 copies per mL and 188 (24%) had CD4 cell counts of fewer than 200 cells per μL. 318 (83%) of 382 patients in the once-daily group had virological response compared with 343 (89%) of 386 in the twice-daily group (difference -5·7%, 95% CI -10·7 to -0·83; p=0·044). Serious adverse events were reported in 26 (7%) of 382 once-daily recipients and 40 (10%) of 388 twice-daily recipients, and adverse events leading to discontinuation occurred in four (1%) patients in each group., Interpretation: Despite high response rates with both regimens, once-daily raltegravir cannot be recommended in place of twice-daily dosing., Funding: Merck., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
29. siRNA screening of a targeted library of DNA repair factors in HIV infection reveals a role for base excision repair in HIV integration.
- Author
-
Espeseth AS, Fishel R, Hazuda D, Huang Q, Xu M, Yoder K, and Zhou H
- Subjects
- 3' Untranslated Regions genetics, Animals, Cell Nucleus genetics, DNA, Complementary genetics, Gene Knockout Techniques, Genetic Vectors genetics, HIV genetics, HIV pathogenicity, HIV Infections genetics, HeLa Cells, Humans, Mice, Reproducibility of Results, Reverse Transcription genetics, Transduction, Genetic, DNA Repair genetics, DNA Repair Enzymes genetics, Gene Library, HIV physiology, HIV Infections virology, RNA, Small Interfering metabolism, Virus Integration genetics
- Abstract
Host DNA repair enzymes have long been assumed to play a role in HIV replication, and many different DNA repair factors have been associated with HIV. In order to identify DNA repair pathways required for HIV infection, we conducted a targeted siRNA screen using 232 siRNA pools for genes associated with DNA repair. Mapping the genes targeted by effective siRNA pools to well-defined DNA repair pathways revealed that many of the siRNAs targeting enzymes associated with the short patch base excision repair (BER) pathway reduced HIV infection. For six siRNA pools targeting BER enzymes, the negative effect of mRNA knockdown was rescued by expression of the corresponding cDNA, validating the importance of the gene in HIV replication. Additionally, mouse embryo fibroblasts (MEFs) lacking expression of specific BER enzymes had decreased transduction by HIV-based retroviral vectors. Examining the role BER enzymes play in HIV infection suggests a role for the BER pathway in HIV integration.
- Published
- 2011
- Full Text
- View/download PDF
30. The base excision repair pathway is required for efficient lentivirus integration.
- Author
-
Yoder KE, Espeseth A, Wang XH, Fang Q, Russo MT, Lloyd RS, Hazuda D, Sobol RW, and Fishel R
- Subjects
- Active Transport, Cell Nucleus, Animals, Cell Line, Cell Nucleus metabolism, Cell Survival, DNA Damage, DNA, Complementary genetics, DNA, Viral genetics, Gene Deletion, HIV Infections genetics, Humans, Lentivirus Infections genetics, Mice, Reverse Transcription genetics, Time Factors, DNA Repair genetics, Lentivirus physiology, Signal Transduction genetics, Virus Integration genetics
- Abstract
An siRNA screen has identified several proteins throughout the base excision repair (BER) pathway of oxidative DNA damage as important for efficient HIV infection. The proteins identified included early repair factors such as the base damage recognition glycosylases OGG1 and MYH and the late repair factor POLß, implicating the entire BER pathway. Murine cells with deletions of the genes Ogg1, Myh, Neil1 and Polß recapitulate the defect of HIV infection in the absence of BER. Defective infection in the absence of BER proteins was also seen with the lentivirus FIV, but not the gammaretrovirus MMLV. BER proteins do not affect HIV infection through its accessory genes nor the central polypurine tract. HIV reverse transcription and nuclear entry appear unaffected by the absence of BER proteins. However, HIV integration to the host chromosome is reduced in the absence of BER proteins. Pre-integration complexes from BER deficient cell lines show reduced integration activity in vitro. Integration activity is restored by addition of recombinant BER protein POLß. Lentiviral infection and integration efficiency appears to depend on the presence of BER proteins.
- Published
- 2011
- Full Text
- View/download PDF
31. Sustained viral response in a hepatitis C virus-infected chimpanzee via a combination of direct-acting antiviral agents.
- Author
-
Olsen DB, Davies ME, Handt L, Koeplinger K, Zhang NR, Ludmerer SW, Graham D, Liverton N, MacCoss M, Hazuda D, and Carroll SS
- Subjects
- Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Cyclopropanes, Dose-Response Relationship, Drug, Drug Administration Schedule, Drug Therapy, Combination, Hepacivirus enzymology, Hepacivirus physiology, Hepatitis C, Chronic virology, Indoles pharmacology, Indoles therapeutic use, Isoindoles, Lactams, Macrocyclic, Leucine analogs & derivatives, Proline analogs & derivatives, Protease Inhibitors administration & dosage, Protease Inhibitors pharmacology, Protease Inhibitors therapeutic use, Sulfonamides, Treatment Outcome, Tubercidin administration & dosage, Tubercidin pharmacology, Tubercidin therapeutic use, Viral Nonstructural Proteins antagonists & inhibitors, Antiviral Agents administration & dosage, Hepacivirus drug effects, Hepatitis C, Chronic drug therapy, Indoles administration & dosage, Pan troglodytes virology, Tubercidin analogs & derivatives, Viral Load drug effects
- Abstract
Efforts to develop novel, interferon-sparing therapies for treatment of chronic hepatitis C (HCV) infection are contingent on the ability of combination therapies consisting of direct antiviral inhibitors to achieve a sustained virologic response. This work demonstrates a proof of concept that coadministration of the nucleoside analogue MK-0608 with the protease inhibitor MK-7009, both of which produced robust viral load declines as monotherapy, to an HCV-infected chimpanzee can achieve a cure of infection.
- Published
- 2011
- Full Text
- View/download PDF
32. MK-7009, a potent and selective inhibitor of hepatitis C virus NS3/4A protease.
- Author
-
Liverton NJ, Carroll SS, Dimuzio J, Fandozzi C, Graham DJ, Hazuda D, Holloway MK, Ludmerer SW, McCauley JA, McIntyre CJ, Olsen DB, Rudd MT, Stahlhut M, and Vacca JP
- Subjects
- Animals, Antiviral Agents pharmacokinetics, Area Under Curve, Cell Line, Cyclopropanes, Dogs, Genotype, Half-Life, Hepacivirus enzymology, Hepacivirus genetics, Humans, Indoles pharmacokinetics, Interferon alpha-2, Interferon-alpha pharmacology, Isoindoles, Lactams, Macrocyclic, Leucine analogs & derivatives, Macaca mulatta, Pan troglodytes, Proline analogs & derivatives, Protease Inhibitors pharmacokinetics, Rats, Recombinant Proteins, Replicon, Substrate Specificity, Sulfonamides, Viral Nonstructural Proteins genetics, Antiviral Agents pharmacology, Hepacivirus drug effects, Indoles pharmacology, Protease Inhibitors pharmacology, Viral Nonstructural Proteins antagonists & inhibitors
- Abstract
The administration of hepatitis C virus (HCV) NS3/4A protease inhibitors to patients with chronic HCV infections has demonstrated that they have dramatic antiviral effects and that compounds acting via this mechanism are likely to form a key component of future anti-HCV therapy. We report here on the preclinical profile of MK-7009, an inhibitor of genotype 1a and 1b proteases at subnanomolar concentrations with modestly shifted potency against genotype 2a and 2b proteases at low nanomolar concentrations. Potent activity was also observed in a cell-based HCV replicon assay in the presence of added human serum (50%). In multiple species evaluated in preclinical studies, the MK-7009 concentrations in the liver were maintained at a significant multiple of the cell-based replicon 50% effective concentration over 12 to 24 h following the administration of moderate oral doses (5 to 10 mg per kg of body weight). MK-7009 also had excellent selectivity against both a range of human proteases and a broad panel of pharmacologically relevant ion channels, receptors, and enzymes. On the basis of this favorable profile, MK-7009 was selected for clinical development and is currently being evaluated in controlled clinical trials with both healthy volunteers and HCV-infected patients.
- Published
- 2010
- Full Text
- View/download PDF
33. Loss of raltegravir susceptibility by human immunodeficiency virus type 1 is conferred via multiple nonoverlapping genetic pathways.
- Author
-
Fransen S, Gupta S, Danovich R, Hazuda D, Miller M, Witmer M, Petropoulos CJ, and Huang W
- Subjects
- Drug Resistance, Viral genetics, Genotype, HIV Infections drug therapy, HIV Integrase genetics, HIV-1 genetics, Humans, Mutagenesis, Site-Directed, Raltegravir Potassium, HIV Integrase Inhibitors pharmacology, HIV-1 drug effects, Pyrrolidinones pharmacology
- Abstract
The human immunodeficiency virus type 1 (HIV-1) integrase mutations N155H and Q148R(H)(K) that reduce susceptibility to the integrase inhibitor raltegravir have been identified in patients failing treatment regimens containing raltegravir. Whether these resistance mutations occur individually or in combination within a single virus genome has not been defined, nor do we fully understand the impact of these primary mutations and other secondary mutations on raltegravir susceptibility and viral replication capacity. To address these important questions, we investigated the raltegravir susceptibility and replication capacity of viruses containing mutations at positions 155 and 148 separately or in combination with secondary mutations selected in subjects failing treatment regimens containing raltegravir. Clonal analysis demonstrated that N155H and Q148R(H)(K) occur independently, not in combination. Viruses containing a Q148R(H)(K) mutation generally displayed larger reductions in raltegravir susceptibility than viruses with an N155H mutation. Analysis of site-directed mutants indicated that E92Q in combination with N155H resulted in a higher level of resistance to raltegravir than N155H alone. Viruses containing a Q148R(H) mutation together with a G140S mutation were more resistant to raltegravir than viruses containing a Q148R(H) mutation alone; however, viruses containing G140S and Q148K were more susceptible to raltegravir than viruses containing a Q148K mutation alone. Both N155H and Q148R(H)(K) mutations reduced the replication capacity, while the addition of secondary mutations either improved or reduced the replication capacity depending on the primary mutation. This study demonstrates distinct genetic pathways to resistance in subjects failing raltegravir regimens and defines the effects of primary and secondary resistance mutations on raltegravir susceptibility and replication capacity.
- Published
- 2009
- Full Text
- View/download PDF
34. Natural polymorphisms of human immunodeficiency virus type 1 integrase and inherent susceptibilities to a panel of integrase inhibitors.
- Author
-
Low A, Prada N, Topper M, Vaida F, Castor D, Mohri H, Hazuda D, Muesing M, and Markowitz M
- Subjects
- Adult, Amino Acid Sequence, Female, HIV Infections drug therapy, HIV Infections virology, HIV Integrase classification, HIV Integrase Inhibitors chemistry, HIV-1 drug effects, HIV-1 genetics, Humans, Male, Molecular Sequence Data, Molecular Structure, Phylogeny, Polymerase Chain Reaction, Polymorphism, Genetic genetics, HIV Integrase genetics, HIV Integrase Inhibitors pharmacology, HIV-1 enzymology
- Abstract
We evaluated the human immunodeficiency virus type 1 (HIV-1) integrase coding region of the pol gene for the presence of natural polymorphisms in patients during early infection (AHI) and with triple-class drug-resistant HIV-1 (MDR). We analyzed selected recombinant viruses containing patient-derived HIV-1 integrase for susceptibility to a panel of strand transfer integrase inhibitors (InSTI). A pretreatment sequence analysis of the integrase coding region was performed for 112 patients identified during acute or early infection and 15 patients with triple-class resistance. A phenotypic analysis was done on 10 recombinant viruses derived from nine patients against a panel of six diverse InSTI. Few of the polymorphisms associated with in vitro InSTI resistance were identified in the samples from newly infected individuals or those patients with MDR HIV-1. We identified polymorphisms V72I, L74I, T97A, V151I, M154I/L, E157Q, V165I, V201I, I203M, T206S, and S230N. V72I was the most common, seen in 63 (56.3%) of the AHI samples. E157Q was the only naturally occurring mutation thought to contribute to resistance to elvitegravir, raltegravir, and L-870,810. None of the patient-derived viruses demonstrated any significant decrease in susceptibility to the drugs tested. In summary, the integrase coding region contains as much natural variation as that seen in protease, but mutations associated with high-level resistance to existing InSTI are rarely, if ever, present in integrase naïve patients, especially those being used clinically. Most of the highly prevalent polymorphisms have little effect on InSTI susceptibility in the absence of specific primary mutations. Baseline testing for integrase susceptibility in InSTI-naïve patients is not currently warranted.
- Published
- 2009
- Full Text
- View/download PDF
35. The challenge of finding a cure for HIV infection.
- Author
-
Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, and Pomerantz RJ
- Subjects
- AIDS Vaccines, Animals, Anti-HIV Agents economics, Antiretroviral Therapy, Highly Active adverse effects, Antiretroviral Therapy, Highly Active economics, CD4-Positive T-Lymphocytes virology, Clinical Trials as Topic, Drug Discovery, HIV physiology, HIV Infections virology, Humans, Viremia drug therapy, Virus Replication drug effects, Anti-HIV Agents therapeutic use, HIV drug effects, HIV Infections drug therapy, Virus Latency drug effects
- Abstract
Although combination therapy for HIV infection represents a triumph for modern medicine, chronic suppressive therapy is required to contain persistent infection in reservoirs such as latently infected CD4+ lymphocytes and cells of the macrophage-monocyte lineage. Despite its success, chronic suppressive therapy is limited by its cost, the requirement of lifelong adherence, and the unknown effects of long-term treatment. This review discusses our current understanding of suppressive antiretroviral therapy, the latent viral reservoir, and the needs for and challenges of attacking this reservoir to achieve a cure.
- Published
- 2009
- Full Text
- View/download PDF
36. Robust antiviral efficacy upon administration of a nucleoside analog to hepatitis C virus-infected chimpanzees.
- Author
-
Carroll SS, Ludmerer S, Handt L, Koeplinger K, Zhang NR, Graham D, Davies ME, MacCoss M, Hazuda D, and Olsen DB
- Subjects
- Animals, Antiviral Agents chemistry, Antiviral Agents pharmacokinetics, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Area Under Curve, Dose-Response Relationship, Drug, Drug Administration Schedule, Hepacivirus genetics, Hepatitis C blood, Hepatitis C virology, Inhibitory Concentration 50, Molecular Structure, Nucleosides chemistry, Nucleosides pharmacokinetics, Nucleosides pharmacology, Nucleosides therapeutic use, Pan troglodytes, RNA, Viral blood, Time Factors, Tubercidin administration & dosage, Tubercidin chemistry, Tubercidin pharmacokinetics, Tubercidin pharmacology, Tubercidin therapeutic use, Viral Load, Antiviral Agents administration & dosage, Hepacivirus drug effects, Hepatitis C drug therapy, Nucleosides administration & dosage, Tubercidin analogs & derivatives
- Abstract
Hepatitis C virus (HCV) infects an estimated 170 million individuals worldwide and is associated with an increased incidence of liver fibrosis, cirrhosis, and hepatocellular carcinoma. Currently approved therapies to treat HCV infection consist of combinations of pegylated alpha interferon and ribavirin which result in a sustained viral response in 40 to 60% of patients. Efforts to develop improved therapies include the development of direct inhibitors of virally encoded enzymes such as the viral RNA-dependent RNA polymerase. A nucleoside analog, 2'-C-methyl-7-deaza-adenosine (MK-0608), has been shown to inhibit viral RNA replication in the subgenomic HCV genotype 1b replicon, with a 50% effective concentration (EC(50)) of 0.3 microM (EC(90) = 1.3 microM). To determine efficacy in vivo, MK-0608 was administered to HCV-infected chimpanzees, resulting in dose- and time-dependent decreases in plasma viral loads. In separate experiments, chimpanzees dosed for 7 days with MK-0608 at 0.2 and 2 mg per kg of body weight per day by intravenous administration experienced average reductions in viral load of 1.0 and >5 log(10) IU/ml, respectively. Two other HCV-infected chimpanzees received daily doses of 1 mg MK-0608 per kg via oral administration. After 37 days of oral dosing, one chimpanzee with a high starting viral load experienced a reduction in viral load of 4.6 log(10), and the viral load in the other chimpanzee fell below the limit of quantification (LOQ) of the HCV TaqMan assay (20 IU/ml). Importantly, viral load remained below the LOQ throughout the duration of dosing and for at least 12 days after dosing ended. The results demonstrate a robust antiviral effect on the administration of MK-0608 to HCV-infected chimpanzees.
- Published
- 2009
- Full Text
- View/download PDF
37. Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid.
- Author
-
Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, and Margolis DM
- Subjects
- CD4-Positive T-Lymphocytes virology, Cell Line, Cells, Cultured, Genes, Reporter, Green Fluorescent Proteins metabolism, HIV Infections virology, Humans, Vorinostat, HIV-1 physiology, Histone Deacetylase Inhibitors, Hydroxamic Acids pharmacology, Virus Activation, Virus Replication
- Abstract
Histone deacetylases (HDACs) act on histones within the nucleosome-bound promoter of human immunodeficiency virus type 1 (HIV-1) to maintain proviral latency. HDAC inhibition leads to promoter expression and the escape of HIV from latency. We evaluated the ability of the potent inhibitor recently licensed for use in oncology, suberoylanilide hydroxamic acid (SAHA; Vorinostat), selective for Class I HDACs, to induce HIV promoter expression in cell lines and virus production from the resting CD4(+) T cells of antiretroviral-treated, aviremic HIV-infected patients. In J89, a Jurkat T cell line infected with a single HIV genome encoding the enhanced green fluorescence protein (EGFP) within the HIV genome, SAHA induced changes at nucleosome 1 of the HIV promoter in chromatin immunoprecipitation (ChIP) assays in concert with EGFP expression. In the resting CD4(+) T cells of antiretroviral-treated, aviremic HIV-infected patients clinically achievable exposures to SAHA induced virus outgrowth ex vivo. These results suggest that potent, selective HDAC inhibitors may allow improved targeting of persistent proviral HIV infection, and define parameters for in vivo studies using SAHA.
- Published
- 2009
- Full Text
- View/download PDF
38. First demonstration of cerebrospinal fluid and plasma A beta lowering with oral administration of a beta-site amyloid precursor protein-cleaving enzyme 1 inhibitor in nonhuman primates.
- Author
-
Sankaranarayanan S, Holahan MA, Colussi D, Crouthamel MC, Devanarayan V, Ellis J, Espeseth A, Gates AT, Graham SL, Gregro AR, Hazuda D, Hochman JH, Holloway K, Jin L, Kahana J, Lai MT, Lineberger J, McGaughey G, Moore KP, Nantermet P, Pietrak B, Price EA, Rajapakse H, Stauffer S, Steinbeiser MA, Seabrook G, Selnick HG, Shi XP, Stanton MG, Swestock J, Tugusheva K, Tyler KX, Vacca JP, Wong J, Wu G, Xu M, Cook JJ, and Simon AJ
- Subjects
- Amyloid beta-Protein Precursor antagonists & inhibitors, Animals, Aspartic Acid Endopeptidases antagonists & inhibitors, Enzyme Inhibitors administration & dosage, Enzyme Inhibitors pharmacology, Humans, Infusions, Intravenous, Macaca mulatta, Mice, Mice, Transgenic, Transfection, ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid beta-Protein Precursor cerebrospinal fluid
- Abstract
beta-Site amyloid precursor protein (APP)-cleaving enzyme (BACE) 1 cleavage of amyloid precursor protein is an essential step in the generation of the potentially neurotoxic and amyloidogenic A beta 42 peptides in Alzheimer's disease. Although previous mouse studies have shown brain A beta lowering after BACE1 inhibition, extension of such studies to nonhuman primates or man was precluded by poor potency, brain penetration, and pharmacokinetics of available inhibitors. In this study, a novel tertiary carbinamine BACE1 inhibitor, tertiary carbinamine (TC)-1, was assessed in a unique cisterna magna ported rhesus monkey model, where the temporal dynamics of A beta in cerebrospinal fluid (CSF) and plasma could be evaluated. TC-1, a potent inhibitor (IC(50) approximately 0.4 nM), has excellent passive membrane permeability, low susceptibility to P-glycoprotein transport, and lowered brain A beta levels in a mouse model. Intravenous infusion of TC-1 led to a significant but transient lowering of CSF and plasma A beta levels in conscious rhesus monkeys because it underwent CYP3A4-mediated metabolism. Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPP beta, A beta 40, A beta 42, and plasma A beta 40 levels. CSF A beta 42 lowering showed an EC(50) of approximately 20 nM with respect to the CSF [TC-1] levels, demonstrating excellent concordance with its potency in a cell-based assay. These results demonstrate the first in vivo proof of concept of CSF A beta lowering after oral administration of a BACE1 inhibitor in a nonhuman primate.
- Published
- 2009
- Full Text
- View/download PDF
39. Emerging pharmacology: inhibitors of human immunodeficiency virus integration.
- Author
-
Hazuda D, Iwamoto M, and Wenning L
- Subjects
- Animals, HIV metabolism, HIV Integrase Inhibitors chemistry, HIV Integrase Inhibitors therapeutic use, Humans, Models, Theoretical, Molecular Structure, Pyrrolidinones chemistry, Pyrrolidinones pharmacokinetics, Pyrrolidinones therapeutic use, Quinolones chemistry, Quinolones pharmacokinetics, Quinolones therapeutic use, Raltegravir Potassium, HIV drug effects, HIV Integrase Inhibitors pharmacokinetics, Pharmacology, Clinical trends, Virus Integration drug effects
- Abstract
The first integrase inhibitor licensed to treat HIV-1 infection was approved in late 2007, more than a decade after the introduction of the first inhibitors of the HIV-1 reverse transcriptase and protease. The unique biochemical and molecular mechanism of action of this novel class of antiretroviral drugs is the fundamental basis for their activity in treating multidrug-resistant HIV-1 infection and is important for understanding both the cellular and in vivo pharmacology and metabolism of these agents. In addition, available pharmacokinetic and drug interaction data for raltegravir and elvitegravir, the two integrase inhibitors that are the most advanced in clinical development to date, are reviewed.
- Published
- 2009
- Full Text
- View/download PDF
40. Discovery and X-ray crystallographic analysis of a spiropiperidine iminohydantoin inhibitor of beta-secretase.
- Author
-
Barrow JC, Stauffer SR, Rittle KE, Ngo PL, Yang Z, Selnick HG, Graham SL, Munshi S, McGaughey GB, Holloway MK, Simon AJ, Price EA, Sankaranarayanan S, Colussi D, Tugusheva K, Lai MT, Espeseth AS, Xu M, Huang Q, Wolfe A, Pietrak B, Zuck P, Levorse DA, Hazuda D, and Vacca JP
- Subjects
- Amyloid Precursor Protein Secretases chemistry, Amyloid Precursor Protein Secretases metabolism, Crystallography, X-Ray, Drug Evaluation, Preclinical, Enzyme Inhibitors chemistry, Hydrogen Bonding, Imidazolidines chemistry, Models, Molecular, Molecular Structure, Piperidines chemistry, Structure-Activity Relationship, Amyloid Precursor Protein Secretases antagonists & inhibitors, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Imidazolidines chemical synthesis, Imidazolidines pharmacology, Piperidines chemical synthesis, Piperidines pharmacology
- Abstract
A high-throughput screen at 100 microM inhibitor concentration for the BACE-1 enzyme revealed a novel spiropiperidine iminohydantoin aspartyl protease inhibitor template. An X-ray cocrystal structure with BACE-1 revealed a novel mode of binding whereby the inhibitor interacts with the catalytic aspartates via bridging water molecules. Using the crystal structure as a guide, potent compounds with good brain penetration were designed.
- Published
- 2008
- Full Text
- View/download PDF
41. Mechanisms of human immunodeficiency virus type 1 concerted integration related to strand transfer inhibition and drug resistance.
- Author
-
Zahm JA, Bera S, Pandey KK, Vora A, Stillmock K, Hazuda D, and Grandgenett DP
- Subjects
- Anti-HIV Agents pharmacology, DNA, Viral drug effects, DNA, Viral metabolism, HIV Integrase chemistry, HIV Integrase drug effects, HIV Integrase genetics, HIV-1 enzymology, HIV-1 genetics, Humans, Inhibitory Concentration 50, Microbial Sensitivity Tests methods, Mutation, Naphthyridines chemistry, Virus Integration genetics, Drug Resistance, Viral drug effects, Drug Resistance, Viral genetics, HIV Integrase Inhibitors pharmacology, HIV-1 drug effects, Keto Acids pharmacology, Naphthyridines pharmacology, Virus Integration drug effects
- Abstract
The "strand transfer inhibitors" of human immunodeficiency virus type-1 (HIV-1) integrase (IN), so named because of their pronounced selectivity for inhibiting strand transfer over 3' OH processing, block virus replication in vivo and ex vivo and prevent concerted integration in vitro. We explored the kinetics of product formation and strand transfer inhibition within reconstituted synaptic complexes capable of concerted integration. Synaptic complexes were formed with viral DNA donors containing either two blunt ends, two 3'-OH-processed ends, or one of each. We determined that one blunt end within a synaptic complex is a sufficient condition for low-nanomolar-range strand transfer inhibition with naphthyridine carboxamide inhibitors L-870,810 and L-870,812. We further explored the catalytic properties and drug resistance profiles of a set of clinically relevant strand transfer inhibitor-resistant HIV-1 IN mutants. The diketo acids and naphthyridine carboxamides, mechanistically similar but structurally distinct strand transfer inhibitors, each select for a distinct set of drug resistance mutations ex vivo. The S153Y and N155S IN resistance mutants were selected with the diketo acid L-841,411, and the N155H mutant was selected with L-810,812. Each mutant exhibited some degree of catalytic impairment relative to the activity of wild type IN, although the N155H mutant displayed near-wild-type IN activities. The resistance profiles indicated that the S153Y mutation potentiates susceptibility to L-870,810 and L-870,812, while the N155S mutation confers resistance to L-870,810 and L-870,812. The N155H mutation confers resistance to L-870,810 and potentiates susceptibility to L-841,411. This study illuminates the interrelated mechanisms of concerted integration, strand transfer inhibition, and resistance to strand transfer inhibitors.
- Published
- 2008
- Full Text
- View/download PDF
42. Human immunodeficiency virus type 1 (HIV-1) integration: a potential target for microbicides to prevent cell-free or cell-associated HIV-1 infection.
- Author
-
Terrazas-Aranda K, Van Herrewege Y, Hazuda D, Lewi P, Costi R, Di Santo R, Cara A, and Vanham G
- Subjects
- Anilides pharmacology, CD4-Positive T-Lymphocytes drug effects, CD4-Positive T-Lymphocytes virology, Cell Line, Cells, Cultured, Coculture Techniques, Dendritic Cells drug effects, Dendritic Cells virology, Female, Furans pharmacology, HIV Infections transmission, HIV Integrase Inhibitors pharmacology, Humans, Male, Naphthyridines pharmacology, Pyrimidines pharmacology, Reverse Transcriptase Inhibitors pharmacology, Thioamides, Anti-HIV Agents pharmacology, HIV Infections prevention & control, HIV-1 drug effects, HIV-1 physiology, Virus Integration drug effects
- Abstract
Conceptually, blocking human immunodeficiency virus type 1 (HIV-1) integration is the last possibility for preventing irreversible cellular infection. Using cocultures of monocyte-derived dendritic cells and CD4(+) T cells, which represent primary targets in sexual transmission, we demonstrated that blocking integration with integrase strand transfer inhibitors (InSTIs), particularly L-870812, could consistently block cell-free and cell-associated HIV-1 infection. In a pretreatment setting in which the compound was present before and during infection and was afterwards gradually diluted during the culture period, the naphthyridine carboxamide L-870812 blocked infection with the cell-free and cell-associated HIV-1 Ba-L strain at concentrations of, respectively, 1,000 and 10,000 nM. The potency of L-870812 was similar to that of the nucleotide reverse transcriptase inhibitor R-9-(2-phosphonylmethoxypropyl) adenine (PMPA) but one or two orders of magnitude lower than those of the nonnucleoside reverse transcriptase inhibitors UC781 and TMC120. In contrast, the diketo acid RDS derivative InSTIs showed clear-cut but weaker antiviral activity than L-870812. Moreover, L-870812 completely blocked subtype C and CRFO2_AG primary isolates, which are prevalent in the African heterosexual epidemic. Furthermore, the addition of micromolar concentrations of L-870812 even 24 h after infection could still block both cell-free and cell-associated Ba-L, opening the prospect of postexposure prophylaxis. Finally, an evaluation of the combined activity of L-870812 with either T20, zidovudine, PMPA, UC781, or TMC120 against replication-deficient HIV-1 Ba-L (env) pseudovirus suggested synergistic activity for all combinations. Importantly, compounds selected for the study by using the coculture model were devoid of acute or delayed cytotoxic effects at HIV-blocking concentrations. Therefore, these findings provide evidence supporting consideration of HIV-1 integration as a target for microbicide development.
- Published
- 2008
- Full Text
- View/download PDF
43. Inhibition of human immunodeficiency virus type 1 concerted integration by strand transfer inhibitors which recognize a transient structural intermediate.
- Author
-
Pandey KK, Bera S, Zahm J, Vora A, Stillmock K, Hazuda D, and Grandgenett DP
- Subjects
- DNA, Viral metabolism, HIV-1 genetics, Humans, Inhibitory Concentration 50, Keto Acids pharmacology, Anti-HIV Agents pharmacology, DNA, Viral drug effects, HIV-1 drug effects, Naphthyridines pharmacology, Virus Integration drug effects
- Abstract
Human immunodeficiency virus type 1 (HIV-1) integrase (IN) inserts the viral DNA genome into host chromosomes. Here, by native agarose gel electrophoresis, using recombinant IN with a blunt-ended viral DNA substrate, we identified the synaptic complex (SC), a transient early intermediate in the integration pathway. The SC consists of two donor ends juxtaposed by IN noncovalently. The DNA ends within the SC were minimally processed (~15%). In a time-dependent manner, the SC associated with target DNA and progressed to the strand transfer complex (STC), the nucleoprotein product of concerted integration. In the STC, the two viral DNA ends are covalently attached to target and remain associated with IN. The diketo acid inhibitors and their analogs effectively inhibit HIV-1 replication by preventing integration in vivo. Strand transfer inhibitors L-870,810, L-870,812, and L-841,411, at low nM concentrations, effectively inhibited the concerted integration of viral DNA donor in vitro. The inhibitors, in a concentration-dependent manner, bound to IN within the SC and thereby blocked the docking onto target DNA, which thus prevented the formation of the STC. Although 3'-OH recessed donor efficiently formed the STC, reactions proceeding with this substrate exhibited marked resistance to the presence of inhibitor, requiring significantly higher concentrations for effective inhibition of all strand transfer products. These results suggest that binding of inhibitor to the SC occurs prior to, during, or immediately after 3'-OH processing. It follows that the IN-viral DNA complex is "trapped" by the strand transfer inhibitors via a transient intermediate within the cytoplasmic preintegration complex.
- Published
- 2007
- Full Text
- View/download PDF
44. Discovery and synthesis of HIV integrase inhibitors: development of potent and orally bioavailable N-methyl pyrimidones.
- Author
-
Gardelli C, Nizi E, Muraglia E, Crescenzi B, Ferrara M, Orvieto F, Pace P, Pescatore G, Poma M, Ferreira Mdel R, Scarpelli R, Homnick CF, Ikemoto N, Alfieri A, Verdirame M, Bonelli F, Paz OG, Taliani M, Monteagudo E, Pesci S, Laufer R, Felock P, Stillmock KA, Hazuda D, Rowley M, and Summa V
- Subjects
- Administration, Oral, Animals, Biological Availability, Blood Proteins metabolism, Cell Line, Tumor, Dogs, HIV Integrase Inhibitors pharmacokinetics, HIV Integrase Inhibitors pharmacology, HIV-1 enzymology, HIV-1 physiology, Humans, Macaca mulatta, Morpholines pharmacokinetics, Morpholines pharmacology, Protein Binding, Pyrimidinones pharmacokinetics, Pyrimidinones pharmacology, Rats, Stereoisomerism, Structure-Activity Relationship, Virus Replication drug effects, HIV Integrase chemistry, HIV Integrase Inhibitors chemical synthesis, HIV-1 drug effects, Morpholines chemical synthesis, Pyrimidinones chemical synthesis
- Abstract
The human immunodeficiency virus type-1 (HIV-1) encodes three enzymes essential for viral replication: a reverse transcriptase, a protease, and an integrase. The latter is responsible for the integration of the viral genome into the human genome and, therefore, represents an attractive target for chemotherapeutic intervention against AIDS. A drug based on this mechanism has not yet been approved. Benzyl-dihydroxypyrimidine-carboxamides were discovered in our laboratories as a novel and metabolically stable class of agents that exhibits potent inhibition of the HIV integrase strand transfer step. Further efforts led to very potent compounds based on the structurally related N-Me pyrimidone scaffold. One of the more interesting compounds in this series is the 2-N-Me-morpholino derivative 27a, which shows a CIC95 of 65 nM in the cell in the presence of serum. The compound has favorable pharmacokinetic properties in three preclinical species and shows no liabilities in several counterscreening assays.
- Published
- 2007
- Full Text
- View/download PDF
45. Dihydroxypyrimidine-4-carboxamides as novel potent and selective HIV integrase inhibitors.
- Author
-
Pace P, Di Francesco ME, Gardelli C, Harper S, Muraglia E, Nizi E, Orvieto F, Petrocchi A, Poma M, Rowley M, Scarpelli R, Laufer R, Gonzalez Paz O, Monteagudo E, Bonelli F, Hazuda D, Stillmock KA, and Summa V
- Subjects
- Animals, Biological Availability, Blood Proteins metabolism, Cell Line, Tumor, Dogs, HIV Integrase Inhibitors pharmacokinetics, HIV Integrase Inhibitors pharmacology, Half-Life, Humans, Macaca mulatta, Protein Binding, Pyridines chemistry, Pyridines pharmacology, Pyrimidines pharmacokinetics, Pyrimidines pharmacology, Rats, Structure-Activity Relationship, Virus Replication, HIV Integrase Inhibitors chemical synthesis, HIV-1 drug effects, Pyridines chemical synthesis, Pyrimidines chemical synthesis
- Abstract
Human immunodeficiency virus type-1 (HIV-1) integrase, one of the three constitutive viral enzymes required for replication, is a rational target for chemotherapeutic intervention in the treatment of AIDS that has also recently been confirmed in the clinical setting. We report here on the design and synthesis of N-benzyl-5,6-dihydroxypyrimidine-4-carboxamides as a class of agents which exhibits potent inhibition of the HIV-integrase-catalyzed strand transfer process. In the current study, structural modifications on these molecules were made in order to examine effects on HIV-integrase inhibitory potencies. One of the most interesting compounds for this series is 2-[1-(dimethylamino)-1-methylethyl]-N-(4-fluorobenzyl)-5,6-dihydroxypyrimidine-4-carboxamide 38, with a CIC95 of 78 nM in the cell-based assay in the presence of serum proteins. The compound has favorable pharmacokinetic properties in preclinical species (rats, dogs, and monkeys) and shows no liabilities in several counterscreening assays, highlighting its potential as a clinically useful antiviral agent.
- Published
- 2007
- Full Text
- View/download PDF
46. LRRTM3 promotes processing of amyloid-precursor protein by BACE1 and is a positional candidate gene for late-onset Alzheimer's disease.
- Author
-
Majercak J, Ray WJ, Espeseth A, Simon A, Shi XP, Wolffe C, Getty K, Marine S, Stec E, Ferrer M, Strulovici B, Bartz S, Gates A, Xu M, Huang Q, Ma L, Shughrue P, Burchard J, Colussi D, Pietrak B, Kahana J, Beher D, Rosahl T, Shearman M, Hazuda D, Sachs AB, Koblan KS, Seabrook GR, and Stone DJ
- Subjects
- Amyloid Precursor Protein Secretases genetics, Amyloid beta-Peptides metabolism, Animals, Aspartic Acid Endopeptidases genetics, Carrier Proteins genetics, Carrier Proteins metabolism, Cells, Cultured, Chromosomes, Human, Pair 10, Enzyme Activation, Humans, Leucine-Rich Repeat Proteins, Membrane Proteins metabolism, Mice, Nerve Tissue Proteins metabolism, Neurons cytology, Neurons metabolism, Nuclear Proteins, Peptide Fragments metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor metabolism, Aspartic Acid Endopeptidases metabolism, Membrane Proteins genetics, Nerve Tissue Proteins genetics, Proteins genetics, Proteins metabolism
- Abstract
Rare familial forms of Alzheimer's disease (AD) are thought to be caused by elevated proteolytic production of the Abeta42 peptide from the beta-amyloid-precursor protein (APP). Although the pathogenesis of the more common late-onset AD (LOAD) is not understood, BACE1, the protease that cleaves APP to generate the N terminus of Abeta42, is more active in patients with LOAD, suggesting that increased amyloid production processing might also contribute to the sporadic disease. Using high-throughput siRNA screening technology, we assessed 15,200 genes for their role in Abeta42 secretion and identified leucine-rich repeat transmembrane 3 (LRRTM3) as a neuronal gene that promotes APP processing by BACE1. siRNAs targeting LRRTM3 inhibit the secretion of Abeta40, Abeta42, and sAPPbeta, the N-terminal APP fragment produced by BACE1 cleavage, from cultured cells and primary neurons by up to 60%, whereas overexpression increases Abeta secretion. LRRTM3 is expressed nearly exclusively in the nervous system, including regions affected during AD, such as the dentate gyrus. Furthermore, LRRTM3 maps to a region of chromosome 10 linked to both LOAD and elevated plasma Abeta42, and is structurally similar to a family of neuronal receptors that includes the NOGO receptor, an inhibitor of neuronal regeneration and APP processing. Thus, LRRTM3 is a functional and positional candidate gene for AD, and, given its receptor-like structure and restricted expression, a potential therapeutic target.
- Published
- 2006
- Full Text
- View/download PDF
47. A genome wide analysis of ubiquitin ligases in APP processing identifies a novel regulator of BACE1 mRNA levels.
- Author
-
Espeseth AS, Huang Q, Gates A, Xu M, Yu Y, Simon AJ, Shi XP, Zhang X, Hodor P, Stone DJ, Burchard J, Cavet G, Bartz S, Linsley P, Ray WJ, and Hazuda D
- Subjects
- Amyloid Precursor Protein Secretases genetics, Aspartic Acid Endopeptidases genetics, Cell Line, Cyclophilins genetics, Cyclophilins metabolism, Gene Expression physiology, Humans, Luciferases metabolism, Microarray Analysis methods, RNA, Messenger metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Transfection methods, Ubiquitin-Protein Ligases genetics, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor metabolism, Aspartic Acid Endopeptidases metabolism, Genome physiology, Ubiquitin-Protein Ligases metabolism
- Abstract
Proteolysis of beta-amyloid precursor protein (APP) into amyloid beta peptide (Abeta) by beta- and gamma-secretases is a critical step in the pathogenesis of Alzheimer's Disease (AD), but the pathways regulating secretases are not fully characterized. Ubiquitinylation, which is dysregulated in AD, may affect APP processing. Here, we describe a screen for APP processing modulators using an siRNA library targeting 532 predicted ubiquitin ligases. Seven siRNA pools diminished Abeta production. Of these, siRNAs targeting PPIL2 (hCyp-60) suppressed beta-site cleavage. Knockdown of PPIL2 mRNA decreased BACE1 mRNA, while overexpression of PPIL2 cDNA enhanced BACE1 mRNA levels. Microarray analysis of PPIL2 or BACE1 knockdown indicated that genes affected by BACE1 knockdown are a subset of those dependent upon PPIL2; suggesting that BACE1 expression is downstream of PPIL2. The association of PPIL2 with BACE expression and its requirement for Abeta production suggests new approaches to discover disease modifying agents for AD.
- Published
- 2006
- Full Text
- View/download PDF
48. Macrocyclic inhibitors of beta-secretase: functional activity in an animal model.
- Author
-
Stachel SJ, Coburn CA, Sankaranarayanan S, Price EA, Wu G, Crouthamel M, Pietrak BL, Huang Q, Lineberger J, Espeseth AS, Jin L, Ellis J, Holloway MK, Munshi S, Allison T, Hazuda D, Simon AJ, Graham SL, and Vacca JP
- Subjects
- Amides chemistry, Amyloid beta-Peptides metabolism, Animals, Blood-Brain Barrier, Brain metabolism, Macrocyclic Compounds chemistry, Macrocyclic Compounds pharmacokinetics, Mice, Molecular Conformation, Phthalic Acids chemistry, Protease Inhibitors chemistry, Protease Inhibitors pharmacokinetics, Stereoisomerism, Structure-Activity Relationship, Tissue Distribution, Amyloid Precursor Protein Secretases metabolism, Macrocyclic Compounds chemical synthesis, Protease Inhibitors chemical synthesis
- Abstract
A macrocyclic inhibitor of beta-secretase was designed by covalently cross-linking the P1 and P3 side chains of an isophthalamide-based inhibitor. Macrocyclization resulted in significantly improved potency and physical properties when compared to the initial lead structures. More importantly, these macrocyclic inhibitors also displayed in vivo amyloid lowering when dosed in a murine model.
- Published
- 2006
- Full Text
- View/download PDF
49. BACE-1 inhibition by a series of psi[CH2NH] reduced amide isosteres.
- Author
-
Coburn CA, Stachel SJ, Jones KG, Steele TG, Rush DM, DiMuzio J, Pietrak BL, Lai MT, Huang Q, Lineberger J, Jin L, Munshi S, Katharine Holloway M, Espeseth A, Simon A, Hazuda D, Graham SL, and Vacca JP
- Subjects
- Amyloid Precursor Protein Secretases, Binding Sites, Immunoassay, Peptides chemistry, Protease Inhibitors pharmacology, Structure-Activity Relationship, Amides chemistry, Endopeptidases metabolism, Protease Inhibitors chemical synthesis
- Abstract
A series of beta-site amyloid precursor protein cleaving enzyme (BACE-1) inhibitors containing a psi(CH2NH) reduced amide bond were synthesized. Incorporation of this reduced amide isostere as a non-cleavable peptide surrogate afforded inhibitors possessing low nanomolar potencies in both an enzymatic and cell-based assay.
- Published
- 2006
- Full Text
- View/download PDF
50. A presenilin-independent aspartyl protease prefers the gamma-42 site cleavage.
- Author
-
Lai MT, Crouthamel MC, DiMuzio J, Pietrak BL, Donoviel DB, Bernstein A, Gardell SJ, Li YM, and Hazuda D
- Subjects
- Amyloid Precursor Protein Secretases, Animals, Blastocyst metabolism, Carbamates pharmacology, Cell Membrane metabolism, Cells, Cultured, DNA genetics, Dipeptides pharmacology, Endopeptidases metabolism, Hydrogen-Ion Concentration, Luminescent Measurements, Membrane Proteins genetics, Membrane Proteins physiology, Mice, Neurons enzymology, Neurons metabolism, Presenilin-1, Presenilin-2, Transfection, Amyloid beta-Peptides metabolism, Aspartic Acid Endopeptidases metabolism
- Abstract
beta-Amyloid peptides (Abeta40 and Abeta42) are the major constituents of amyloid plaques, which are one of the hallmarks of Alzheimer's disease (AD). The Abeta is derived from sequential cleavages of amyloid precursor protein (APP) by beta- and gamma-secretases. gamma-Secretase consists of at least four proteins where presenilins (PS1 and PS2 or PS) are the catalytic subunit involved in the gamma-site cleavage of APP. Secretion of both Abeta40 and Abeta42 is significantly reduced in PS1 knock-out cells and completely abolished in cells deficient for both PS1 and PS2. Consequently, both the PS proteins play essential roles in the production of the secretory of Abeta from cells. Recent studies in primary neurons, however, suggest that PSs are not required for intracellular Abeta42 accumulation; thus the intracellular Abeta42 appears to be generated in a PS-independent manner. Here we present the first biochemical evidence indicating that Abeta, especially Abeta42, can be generated in the absence of PS based on an in vitrogamma-secretase assay employing membranes prepared from PS-deficient Blastocyst-derived (BD) cells. This PS-independent gamma-secretase (PSIG) activity is sensitive to the changes in pH and displays an optimal activity at pH 6.0. Pepstatin A is a potent inhibitor for this proteolytic activity with IC50 of 1.2 nm and 0.4 nm for Abeta40 and Abeta42 generation, respectively. These results indicate that these PS-independent gamma-site cleavages are mediated by an aspartyl protease. More importantly, the PSIG activity displays a distinct preference in mediating the 42-site cleavage over the 40-site cleavage, thereby generating Abeta42 as the predominant product.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.