Network


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

Hotspot


Dive into the research topics where Marc Witmer is active.

Publication


Featured researches published by Marc Witmer.


Journal of Medicinal Chemistry | 2008

Discovery of Raltegravir, a Potent, Selective Orally Bioavailable HIV-Integrase Inhibitor for the Treatment of HIV-AIDS Infection

Vincenzo Summa; Alessia Petrocchi; Fabio Bonelli; Benedetta Crescenzi; Monica Donghi; Marco Ferrara; Fabrizio Fiore; Cristina Gardelli; Odalys Gonzalez Paz; Daria J. Hazuda; Philip Jones; Olaf Kinzel; Ralph Laufer; Edith Monteagudo; Ester Muraglia; Emanuela Nizi; Federica Orvieto; Paola Pace; Giovanna Pescatore; Rita Scarpelli; Kara A. Stillmock; Marc Witmer; Michael Rowley

Human immunodeficiency virus type-1 (HIV-1) integrase is one of the three virally encoded enzymes required for replication and therefore a rational target for chemotherapeutic intervention in the treatment of HIV-1 infection. We report here the discovery of Raltegravir, the first HIV-integrase inhibitor approved by FDA for the treatment of HIV infection. It derives from the evolution of 5,6-dihydroxypyrimidine-4-carboxamides and N-methyl-4-hydroxypyrimidinone-carboxamides, which exhibited potent inhibition of the HIV-integrase catalyzed strand transfer process. Structural modifications on these molecules were made in order to maximize potency as HIV-integrase inhibitors against the wild type virus, a selection of mutants, and optimize the selectivity, pharmacokinetic, and metabolic profiles in preclinical species. The good profile of Raltegravir has enabled its progression toward the end of phase III clinical trials for the treatment of HIV-1 infection and culminated with the FDA approval as the first HIV-integrase inhibitor for the treatment of HIV-1 infection.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Diketo acid inhibitor mechanism and HIV-1 integrase: Implications for metal binding in the active site of phosphotransferase enzymes

Jay A. Grobler; Kara A. Stillmock; Binghua Hu; Marc Witmer; Peter J. Felock; Amy S. Espeseth; Abigail Wolfe; Melissa S. Egbertson; Michele Bourgeois; Jeffrey Y. Melamed; John S. Wai; Steve Young; Joseph P. Vacca; Daria J. Hazuda

The process of integrating the reverse-transcribed HIV-1 DNA into the host chromosomal DNA is catalyzed by the virally encoded enzyme integrase (IN). Integration requires two metal-dependent reactions, 3′ end processing and strand transfer. Compounds that contain a diketo acid moiety have been shown to selectively inhibit the strand transfer reaction of IN in vitro and in infected cells and are effective as inhibitors of HIV-1 replication. To characterize the molecular basis of inhibition, we used functional assays and binding assays to evaluate a series of structurally related analogs. These studies focused on investigating the role of the conserved carboxylate and metal binding. We demonstrate that an acidic moiety such as a carboxylate or isosteric heterocycle is not required for binding to the enzyme complex but is essential for inhibition and confers distinct metal-dependent properties on the inhibitor. Binding requires divalent metal and resistance is metal dependent with active site mutants displaying resistance only when the enzymes are evaluated in the context of Mg2+. The mechanism of action of these inhibitors is therefore likely a consequence of the interaction between the acid moiety and metal ion(s) in the IN active site, resulting in a functional sequestration of the critical metal cofactor(s). These studies thus have implications for modeling active site inhibitors of IN, designing and evaluating analogs with improved efficacy, and identifying inhibitors of other metal-dependent phosphotransferases.


Journal of Virology | 2009

Loss of Raltegravir Susceptibility by Human Immunodeficiency Virus Type 1 Is Conferred via Multiple Nonoverlapping Genetic Pathways

Signe Fransen; Soumi Gupta; Robert Danovich; Daria J. Hazuda; Michael I. Miller; Marc Witmer; Christos J. Petropoulos; Wei Huang

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.


Journal of Biological Chemistry | 2003

Inhibition of HIV-1 Ribonuclease H by a Novel Diketo Acid, 4-[5-(Benzoylamino)thien-2-yl]-2,4-dioxobutanoic Acid

Cathryn A. Shaw-Reid; Vandna Munshi; Pia L. Graham; Abigail Wolfe; Marc Witmer; Renee Danzeisen; David B. Olsen; Steven S. Carroll; Mark W. Embrey; John S. Wai; Michael D. Miller; James L. Cole; Daria J. Hazuda

Human immunodeficiency virus-type 1 (HIV-1) reverse transcriptase (RT) coordinates DNA polymerization and ribonuclease H (RNase H) activities using two discrete active sites embedded within a single heterodimeric polyprotein. We have identified a novel thiophene diketo acid, 4-[5-(benzoylamino)thien-2-yl]-2,4-dioxobutanoic acid, that selectively inhibits polymerase-independent RNase H cleavage (IC50 = 3.2 μm) but has no effect on DNA polymerization (IC50 > 50 μm). The activity profile of the diketo acid is shown to be distinct from previously described compounds, including the polymerase inhibitor foscarnet and the putative RNase H inhibitor 4-chlorophenylhydrazone. Both foscarnet and the hydrazone inhibit RNase H cleavage and DNA polymerization activities of RT, yet neither inhibits the RNase H activity of RT containing a mutation in the polymerase active site (D185N) or an isolated HIV-1 RNase H domain chimera containing the α-C helix from Escherichia coli RNase HI, suggesting these compounds affect RNase H indirectly. In contrast, the diketo acid inhibits the RNase H activity of the isolated RNase H domain as well as full-length RT, and inhibition is not affected by the polymerase active site mutation. In isothermal titration calorimetry studies using the isolated RNase H domain, binding of the diketo acid is independent of nucleic acid but strictly requires Mn2+implying a direct interaction between the inhibitor and the RNase H active site. These studies demonstrate that inhibition of HIV-1 RNase H may occur by either direct or indirect mechanisms, and they provide a framework for identifying novel agents such as 4-[5-(benzoylamino)thien- 2-yl]-2,4-dioxobutanoic acid that specifically targets RNase H.


Antimicrobial Agents and Chemotherapy | 2014

In Vitro Characterization of MK-1439, a Novel HIV-1 Nonnucleoside Reverse Transcriptase Inhibitor

Ming-Tain Lai; Meizhen Feng; Jean-Pierre Falgueyret; Paul Tawa; Marc Witmer; Daniel J. DiStefano; Yuan Li; Jason Burch; Nancy Sachs; Meiqing Lu; Elizabeth Cauchon; Louis-Charles Campeau; Jay A. Grobler; Youwei Yan; Yves Ducharme; Bernard Cote; Ernest Asante-Appiah; Daria J. Hazuda; Michael D. Miller

ABSTRACT Nonnucleoside reverse transcriptase inhibitors (NNRTIs) are a mainstay of therapy for treating human immunodeficiency type 1 virus (HIV-1)-infected patients. MK-1439 is a novel NNRTI with a 50% inhibitory concentration (IC50) of 12, 9.7, and 9.7 nM against the wild type (WT) and K103N and Y181C reverse transcriptase (RT) mutants, respectively, in a biochemical assay. Selectivity and cytotoxicity studies confirmed that MK-1439 is a highly specific NNRTI with minimum off-target activities. In the presence of 50% normal human serum (NHS), MK-1439 showed excellent potency in suppressing the replication of WT virus, with a 95% effective concentration (EC95) of 20 nM, as well as K103N, Y181C, and K103N/Y181C mutant viruses with EC95 of 43, 27, and 55 nM, respectively. MK-1439 exhibited similar antiviral activities against 10 different HIV-1 subtype viruses (a total of 93 viruses). In addition, the susceptibility of a broader array of clinical NNRTI-associated mutant viruses (a total of 96 viruses) to MK-1439 and other benchmark NNRTIs was investigated. The results showed that the mutant profile of MK-1439 was superior overall to that of efavirenz (EFV) and comparable to that of etravirine (ETR) and rilpivirine (RPV). Furthermore, E138K, Y181C, and K101E mutant viruses that are associated with ETR and RPV were susceptible to MK-1439 with a fold change (FC) of <3. A two-drug in vitro combination study indicated that MK-1439 acts nonantagonistically in the antiviral activity with each of 18 FDA-licensed drugs for HIV infection. Taken together, these in vitro data suggest that MK-1439 possesses the desired properties for further development as a new antiviral agent.


Bioorganic & Medicinal Chemistry Letters | 2008

Synthesis of 5-(1-H or 1-alkyl-5-oxopyrrolidin-3-yl)-8-hydroxy-[1,6]-naphthyridine-7-carboxamide inhibitors of HIV-1 integrase

Jeffrey Y. Melamed; Melissa S. Egbertson; Sandor L. Varga; Joseph P. Vacca; Greg Moyer; Lori Gabryelski; Peter J. Felock; Kara A. Stillmock; Marc Witmer; William A. Schleif; Daria J. Hazuda; Yvonne M. Leonard; Lixia Jin; Joan D. Ellis; Steven D. Young

HIV-1 integrase catalyzes the insertion of viral DNA into the genome of the host cell. Integrase inhibitor N-(4-fluorobenzyl)-8-hydroxy-1,6-naphthyridine-7-carboxamide selectively inhibits the strand transfer process of integration. 4-Substituted pyrrolidinones possessing various groups on the pyrrolidinone nitrogen were introduced at the 5-position of the naphthyridine scaffold. These analogs exhibit excellent activity against viral replication in a cell-based assay. The preparation of these compounds was enabled by a three-step, two-pot reaction sequence from a common butenolide intermediate.


Methods | 2009

Selection and analysis of HIV-1 integrase strand transfer inhibitor resistant mutant viruses

Marc Witmer; Robert Danovich

This report describes methods for the selection and analysis of antiretroviral resistance to HIV integrase strand transfer inhibitors (InSTIs) in cell culture. The method involves the serial passage of HIV-1 in the presence of increasing concentrations of test inhibitors, followed by the cloning and sequencing of the integrase coding region from the selected viruses. The identified mutations are subsequently re-engineered into a reference wild-type molecular clone, and the resulting replication capacity and level of drug resistance are determined relative to the wild-type virus. Here we describe examples of selection and analysis of InSTI-resistant viruses using four integrase inhibitors from three structurally distinct chemical classes; a diketo acid, two naphthyridines, and a pyrimidinecarboxamide. Each inhibitor selected an independent route to resistance. Interestingly, the shift in the IC50 required to suppress the re-engineered resistant mutant viruses closely matched the concentration of compound used during the selection of drug resistance.


Acta Crystallographica Section D-biological Crystallography | 1999

Purification, solution properties and crystallization of SIV integrase containing a continuous core and C-terminal domain.

Ying Li; Youwei Yan; Joan Zugay-Murphy; Bei Xu; James L. Cole; Marc Witmer; Peter J. Felock; Abigail Wolfe; Daria J. Hazuda; Mohinder K. Sardana; Zhongguo Chen; Lawrence C. Kuo; Vinod V. Sardana

The C-terminal two-thirds segment of integrase derived from the simian immunodeficiency virus has been cloned, expressed in Escherichia coli, and purified to greater than 95% homogeneity. The protein encompasses amino-acid residues 50-293 and contains a F185H substitution to enhance solubility. In dilute solutions at concentrations below 1 mg ml(-1), the enzyme is predominantly dimeric. At the higher concentrations (>10 mg ml(-1)) required to enable crystallization, the enzyme self-associates to form species with molecular weights greater than 200 kDa. Despite the apparent high aggregation in solution, the enzyme crystallizes from a 8%(v/v) polyethylene glycol (molecular weight 6000) solution in a form suitable for X-ray diffraction studies. The resulting single crystals belong to the space group P2(1)2(1)2(1), with unit-cell parameters a = 79.76, b = 99.98, c = 150.2 A, alpha = beta = gamma = 90 degrees and Z = 4. Under X-ray irradiation generated with a rotating-anode generator, the crystals diffract to 2.8 A resolution and allow collection of a native 3 A resolution diffraction data set.


Science | 2000

Inhibitors of strand transfer that prevent integration and inhibit HIV-1 replication in cells

Daria J. Hazuda; Peter J. Felock; Marc Witmer; Abigail Wolfe; Kara A. Stillmock; Jay A. Grobler; Amy S. Espeseth; Lori Gabryelski; William A. Schleif; Carol Blau; Michael D. Miller


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

A naphthyridine carboxamide provides evidence for discordant resistance between mechanistically identical inhibitors of HIV-1 integrase

Daria J. Hazuda; Neville J. Anthony; Robert P. Gomez; Samson M. Jolly; John S. Wai; Linghang Zhuang; Thorsten E. Fisher; Mark W. Embrey; James P. Guare; Melissa S. Egbertson; Joseph P. Vacca; Joel R. Huff; Peter J. Felock; Marc Witmer; Kara A. Stillmock; Robert Danovich; Jay A. Grobler; Michael D. Miller; Amy S. Espeseth; Lixia Jin; I-Wu Chen; Jiunn H. Lin; Kelem Kassahun; Joan D. Ellis; Bradley K. Wong; Wei Xu; Paul G. Pearson; William A. Schleif; Riccardo Cortese; Emilio A. Emini

Collaboration


Dive into the Marc Witmer's collaboration.

Top Co-Authors

Avatar

Peter J. Felock

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Kara A. Stillmock

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

William A. Schleif

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Abigail Wolfe

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

John S. Wai

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Mark W. Embrey

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge