34 results on '"Krasimir A. Spasov"'
Search Results
2. Structural Studies and Structure Activity Relationships for Novel Computationally Designed Non-nucleoside Inhibitors and Their Interactions With HIV-1 Reverse Transcriptase
- Author
-
Kathleen M. Frey, Nicole Bertoletti, Albert H. Chan, Joseph A. Ippolito, Mariela Bollini, Krasimir A. Spasov, William L. Jorgensen, and Karen S. Anderson
- Subjects
non-nucleoside reverse transcriptase inhibitors ,HIV RT ,structural studies ,computational chemistry ,drug design ,Biology (General) ,QH301-705.5 - Abstract
Reverse transcriptase (RT) from the human immunodeficiency virus continues to be an attractive drug target for antiretroviral therapy. June 2022 will commemorate the 30th anniversary of the first Human Immunodeficiency Virus (HIV) RT crystal structure complex that was solved with non-nucleoside reverse transcriptase inhibitor nevirapine. The release of this structure opened opportunities for designing many families of non-nucleoside reverse transcriptase inhibitors (NNRTIs). In paying tribute to the first RT-nevirapine structure, we have developed several compound classes targeting the non-nucleoside inhibitor binding pocket of HIV RT. Extensive analysis of crystal structures of RT in complex with the compounds informed iterations of structure-based drug design. Structures of seven additional complexes were determined and analyzed to summarize key interactions with residues in the non-nucleoside inhibitor binding pocket (NNIBP) of RT. Additional insights comparing structures with antiviral data and results from molecular dynamics simulations elucidate key interactions and dynamics between the nucleotide and non-nucleoside binding sites.
- Published
- 2022
- Full Text
- View/download PDF
3. Long‐acting and extended‐release implant and nanoformulations with a synergistic antiretroviral two‐drug combination controls HIV‐1 infection in a humanized mouse model
- Author
-
Jagadish Beloor, Shalley N. Kudalkar, Gina Buzzelli, Fan Yang, Hanna K. Mandl, Jyothi K. Rajashekar, Krasimir A. Spasov, William L. Jorgensen, W. Mark Saltzman, Karen S. Anderson, and Priti Kumar
- Subjects
Compound I ,drug synergy ,EFdA ,HIV ,humanized mice ,implants ,Chemical engineering ,TP155-156 ,Biotechnology ,TP248.13-248.65 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract The HIV pandemic has affected over 38 million people worldwide with close to 26 million currently accessing antiretroviral therapy (ART). A major challenge in the long‐term treatment of HIV‐1 infection is nonadherence to ART. Long‐acting antiretroviral (LA‐ARV) formulations, that reduce dosing frequency to less than once a day, are an urgent need that could tackle the adherence issue. Here, we have developed two LA‐ART interventions, one an injectable nanoformulation, and the other, a removable implant, for the delivery of a synergistic two‐drug ARV combination comprising a pre‐clinical nonnucleoside reverse transcriptase inhibitor (NNRTI), Compound I, and the nucleoside reverse transcriptase inhibitor (NRTI), 4′‐ethynyl‐2‐fluoro‐2′‐deoxyadenosine. The nanoformulation is poly(lactide‐co‐glycolide)‐based and the implant is a copolymer of ω‐pentadecalactone and p‐dioxanone, poly(PDL‐co‐DO), a novel class of biocompatible, biodegradable materials. Both the interventions, packaged independently with each ARV, released sustained levels of the drugs, maintaining plasma therapeutic indices for over a month, and suppressed viremia in HIV‐1‐infected humanized mice for up to 42 days with maintenance of CD4+ T cells. These data suggest promise in the use of these new drugs as LA‐ART formulations in subdermal implant and injectable mode.
- Published
- 2022
- Full Text
- View/download PDF
4. Potent Noncovalent Inhibitors of the Main Protease of SARS-CoV-2 from Molecular Sculpting of the Drug Perampanel Guided by Free Energy Perturbation Calculations
- Author
-
Zhuobin Liang, Shalley N. Kudalkar, Krasimir A. Spasov, Julian Tirado-Rives, Scott J. Miller, Shuo Zhang, Karen S. Anderson, Maya Deshmukh, Brett D. Lindenbach, William L. Jorgensen, Joseph A. Ippolito, Farren J. Isaacs, Elizabeth A Stone, Mohammad Mehdi Ghahremanpour, Yuka Takeo, and Chun-Hui Zhang
- Subjects
Drug ,Protease ,Coronavirus disease 2019 (COVID-19) ,010405 organic chemistry ,Chemistry ,Drug discovery ,General Chemical Engineering ,medicine.medical_treatment ,media_common.quotation_subject ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,General Chemistry ,010402 general chemistry ,01 natural sciences ,Combinatorial chemistry ,0104 chemical sciences ,Free energy perturbation ,Perampanel ,chemistry.chemical_compound ,medicine ,Ic50 values ,QD1-999 ,Research Article ,media_common - Abstract
Starting from our previous finding of 14 known drugs as inhibitors of the main protease (Mpro) of SARS-CoV-2, the virus responsible for COVID-19, we have redesigned the weak hit perampanel to yield multiple noncovalent, nonpeptidic inhibitors with ca. 20 nM IC50 values in a kinetic assay. Free-energy perturbation (FEP) calculations for Mpro-ligand complexes provided valuable guidance on beneficial modifications that rapidly delivered the potent analogues. The design efforts were confirmed and augmented by determination of high-resolution X-ray crystal structures for five analogues bound to Mpro. Results of cell-based antiviral assays further demonstrated the potential of the compounds for treatment of COVID-19. In addition to the possible therapeutic significance, the work clearly demonstrates the power of computational chemistry for drug discovery, especially FEP-guided lead optimization., Noncovalent inhibitors with low-nanomolar potency are reported for the main protease of SARS-CoV-2. X-ray crystal structures of complexes are provided, and antiviral activity is also demonstrated using infected cells.
- Published
- 2021
- Full Text
- View/download PDF
5. Covalent Inhibition of Wild-Type HIV-1 Reverse Transcriptase Using a Fluorosulfate Warhead
- Author
-
Shengyan Jin, José A. Cisneros, Karen S. Anderson, Krasimir A. Spasov, Kara J. Cutrona, M. Valhondo, Haichan Niu, Nicole Bertoletti, Zachary J Carter, William L. Jorgensen, and Joseph A. Ippolito
- Subjects
chemistry.chemical_classification ,Catechol ,010405 organic chemistry ,Stereochemistry ,Organic Chemistry ,Wild type ,Crystal structure ,01 natural sciences ,Biochemistry ,In vitro ,Reverse transcriptase ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,Enzyme ,chemistry ,Covalent bond ,Drug Discovery ,Mass spectrum - Abstract
[Image: see text] Covalent inhibitors of wild-type HIV-1 reverse transcriptase (CRTIs) are reported. Three compounds derived from catechol diether non-nucleoside inhibitors (NNRTIs) with addition of a fluorosulfate warhead are demonstrated to covalently modify Tyr181 of HIV-RT. X-ray crystal structures for complexes of the CRTIs with the enzyme are provided, which fully demonstrate the covalent attachment, and confirmation is provided by appropriate mass shifts in ESI-TOF mass spectra. The three CRTIs and six noncovalent analogues are found to be potent inhibitors with both IC(50) values for in vitro inhibition of WT RT and EC(50) values for cytopathic protection of HIV-1-infected human T-cells in the 5–320 nM range.
- Published
- 2021
- Full Text
- View/download PDF
6. Structural investigation of <scp>2‐naphthyl</scp> phenyl ether inhibitors bound to <scp>WT</scp> and <scp>Y181C</scp> reverse transcriptase highlights key features of the <scp>NNRTI</scp> binding site
- Author
-
Vincent N. Duong, Karen S. Anderson, Krasimir A. Spasov, Won-Gil Lee, Albert H. Chan, Joseph A. Ippolito, and William L. Jorgensen
- Subjects
Mutation ,Ether ,Drug resistance ,Resistance mutation ,medicine.disease_cause ,Biochemistry ,Reverse transcriptase ,chemistry.chemical_compound ,chemistry ,Viral life cycle ,medicine ,Potency ,Binding site ,Molecular Biology - Abstract
Human immunodeficiency virus (HIV)-1 remains as a global health issue that is primarily treated with highly active antiretroviral therapy, a combination of drugs that target the viral life cycle. One class of these drugs are non-nucleoside reverse transcriptase inhibitors (NNRTIs) that target the viral reverse transcriptase (RT). First generation NNRTIs were troubled with poor pharmacological properties and drug resistance, incentivizing the development of improved compounds. One class of developed compounds are the 2-naphthyl phenyl ethers, showing promising efficacy against the Y181C RT mutation. Further biochemical and structural work demonstrated differences in potency against the Y181C mutation and binding mode of the compounds. This work aims to understand the relationship between the binding mode and ability to overcome drug resistance using macromolecular x-ray crystallography. Comparison of 2-naphthyl phenyl ethers bound to Y181C RT reveal that compounds that interact with the invariant W229 are more capable of retaining efficacy against the resistance mutation. Additional modifications to these compounds at the 4-position, computationally designed to compensate for the Y181C mutation, do not demonstrate improved potency. Ultimately, we highlight important considerations for the development of future HIV-1 drugs that are able to combat drug resistance.
- Published
- 2020
- Full Text
- View/download PDF
7. Design, synthesis, and biological testing of biphenylmethyloxazole inhibitors targeting HIV-1 reverse transcriptase
- Author
-
Zachary J. Carter, Klarissa Hollander, Krasimir A. Spasov, Karen S. Anderson, and William L. Jorgensen
- Subjects
Organic Chemistry ,Clinical Biochemistry ,Drug Discovery ,Pharmaceutical Science ,Molecular Medicine ,Molecular Biology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
8. Molecular and cellular studies evaluating a potent 2-cyanoindolizine catechol diether NNRTI targeting wildtype and Y181C mutant HIV-1 reverse transcriptase
- Author
-
Krasimir A. Spasov, William L. Jorgensen, Kathleen M. Frey, Won-Gil Lee, Shalley N. Kudalkar, Karen S. Anderson, Tsuneo Sasaki, and Zachary T. K. Gannam
- Subjects
Anti-HIV Agents ,Clinical Biochemistry ,Mutant ,Catechols ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,Article ,chemistry.chemical_compound ,Drug Discovery ,Potency ,Binding site ,Molecular Biology ,Catechol ,Molecular Structure ,010405 organic chemistry ,Organic Chemistry ,Wild type ,HIV Reverse Transcriptase ,Reverse transcriptase ,In vitro ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry ,Drug Design ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Indolizine - Abstract
The development of efficacious NNRTIs for HIV/AIDS therapy is commonly met with the emergence of drug resistant strains, including the Y181C variant. Using a computationally-guided approach, we synthesized the catechol diether series of NNRTIs, which display sub-nanomolar potency in cellular assays. Among the most potent were a series of 2-cyanoindolizine substituted catechol diethers, including Compound 1. We present here a thorough evaluation of this compound, including biochemical, cellular, and structural studies. The compound demonstrates low nanomolar potency against both WT and Y181C HIV-1 RT in in vitro and cellular assays. Our crystal structures of both the wildtype and mutant forms of RT in complex with Compound 1 allow the interrogation of this compound’s features that allow it to maintain strong efficacy against the drug resistant mutant. Among these are compensatory shifts in the NNRTI binding pocket, persistence of multiple hydrogen bonds, and van der Waals contacts throughout the binding site. Further, the fluorine at the C6 position of the indolizine moiety makes multiple favorable interactions with both RT forms. The present study highlights the indolizine-substituted catechol diether class of NNRTIs as promising therapeutic candidates possessing optimal pharmacological properties and significant potency against multiple RT variants.
- Published
- 2019
- Full Text
- View/download PDF
9. Optimization of Triarylpyridinone Inhibitors of the Main Protease of SARS-CoV-2 to Low-Nanomolar Antiviral Potency
- Author
-
Maria-Elena Liosi, Krasimir A. Spasov, Zhuobin Liang, Chun-Hui Zhang, Karen S. Anderson, Scott J. Miller, Joseph A. Ippolito, Brett D. Lindenbach, Klarissa Hollander, Julian Tirado-Rives, Maya Deshmukh, Raquel A. Reilly, Farren J. Isaacs, William L. Jorgensen, Shuo Zhang, and Elizabeth A Stone
- Subjects
chemistry.chemical_classification ,Letter ,Protease ,SARS-CoV-2 ,COVID-19 drugs ,medicine.medical_treatment ,Organic Chemistry ,protease inhibitors ,Uracil ,lead optimization ,Biochemistry ,Combinatorial chemistry ,Free energy perturbation ,chemistry.chemical_compound ,Enzyme ,chemistry ,Drug Discovery ,medicine ,Vero cell ,Potency ,Cytotoxicity ,EC50 - Abstract
Non-covalent inhibitors of the main protease (Mpro) of SARS-CoV-2 having a pyridinone core were previously reported with IC50 values as low as 0.018 μM for inhibition of enzymatic activity and EC50 values as low as 0.8 μM for inhibition of viral replication in Vero E6 cells. The series has now been further advanced by consideration of placement of substituted five-membered-ring heterocycles in the S4 pocket of Mpro and N-methylation of a uracil ring. Free energy perturbation calculations provided guidance on the choice of the heterocycles, and protein crystallography confirmed the desired S4 placement. Here we report inhibitors with EC50 values as low as 0.080 μM, while remdesivir yields values of 0.5–2 μM in side-by-side testing with infectious SARS-CoV-2. A key factor in the improvement is enhanced cell permeability, as reflected in PAMPA measurements. Compounds 19 and 21 are particularly promising as potential therapies for COVID-19, featuring IC50 values of 0.044–0.061 μM, EC50 values of ca. 0.1 μM, good aqueous solubility, and no cytotoxicity.
- Published
- 2021
10. Design, Conformation, and Crystallography of 2-Naphthyl Phenyl Ethers as Potent Anti-HIV Agents
- Author
-
Krasimir A. Spasov, Won-Gil Lee, William L. Jorgensen, Karen S. Anderson, and Albert H. Chan
- Subjects
0301 basic medicine ,Hydrolase inhibitor ,Catechol ,Chemistry ,Anti hiv ,Stereochemistry ,Organic Chemistry ,Substituent ,virus diseases ,Phenyl Ethers ,Crystal structure ,010402 general chemistry ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Drug Discovery ,Transferase ,Conformational isomerism - Abstract
Catechol diethers that incorporate a 7-cyano-2-naphthyl substituent are reported as non-nucleoside inhibitors of HIV-1 reverse transcriptase (NNRTIs). Many of the compounds have 1–10 nM potencies toward wild-type HIV-1. An interesting conformational effect allows two unique conformers for the naphthyl group in complexes with HIV-RT. X-ray crystal structures for 4a and 4f illustrate the alternatives.
- Published
- 2016
- Full Text
- View/download PDF
11. Reply to Pandey et al.: Understanding the efficacy of a potential antiretroviral drug candidate in humanized mouse model of HIV infection
- Author
-
José A. Cisneros, Krasimir A. Spasov, Jagadish Beloor, Won-Gil Lee, Karen S. Anderson, W. Mark Saltzman, William L. Jorgensen, Shalley N. Kudalkar, Priti Kumar, and Elias Quijano
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Multidisciplinary ,Anti-HIV Agents ,business.industry ,Human immunodeficiency virus (HIV) ,HIV Infections ,Antiretroviral drug ,medicine.disease_cause ,humanities ,Plasma viral load ,Disease Models, Animal ,Mice ,03 medical and health sciences ,030104 developmental biology ,Internal medicine ,Humanized mouse ,HIV-1 ,medicine ,Animals ,Letters ,business ,Viral load - Abstract
We appreciate Pandey et al.’s (1) interest in further understanding our published work (2). Our responses are given below the excerpts from the Letter.Our first and foremost concern is the data presented in their figure 6C. We fail to understand why there was a fall in the plasma viral loads after day 19 in the control group (i.e., the group not given any intervention). Furthermore, not only was the fall in the plasma viral load significant (to below the limit of detection
- Published
- 2018
- Full Text
- View/download PDF
12. Discovery and crystallography of bicyclic arylaminoazines as potent inhibitors of HIV-1 reverse transcriptase
- Author
-
Ricardo Gallardo-Macias, Albert H. Chan, Kathleen M. Frey, William L. Jorgensen, Karen S. Anderson, Won-Gil Lee, and Krasimir A. Spasov
- Subjects
Models, Molecular ,Pyrimidine ,Anti-HIV Agents ,Stereochemistry ,Clinical Biochemistry ,Substituent ,Human immunodeficiency virus (HIV) ,Pharmaceutical Science ,medicine.disease_cause ,Biochemistry ,Article ,Cell Line ,Bridged Bicyclo Compounds ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Aqueous solubility ,medicine ,Humans ,Molecular Biology ,Cell Proliferation ,Dose-Response Relationship, Drug ,Molecular Structure ,Bicyclic molecule ,Triazines ,Organic Chemistry ,HIV ,HIV Reverse Transcriptase ,Reverse transcriptase ,Solubility ,chemistry ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Azabicyclo Compounds - Abstract
Non-nucleoside inhibitors of HIV-1 reverse transcriptase (HIV-RT) are reported that incorporate a 7-indolizinylamino or 2-naphthylamino substituent on a pyrimidine or 1,3,5-triazine core. The most potent compounds show below 10 nanomolar activity towards wild-type HIV-1 and variants bearing Tyr181Cys and Lys103Asn/Tyr181Cys resistance mutations. The compounds also feature good aqueous solubility. Crystal structures for two complexes enhance the analysis of the structure–activity data.
- Published
- 2015
- Full Text
- View/download PDF
13. Potent Inhibitors Active against HIV Reverse Transcriptase with K101P, a Mutation Conferring Rilpivirine Resistance
- Author
-
Robert F. Siliciano, Sarah B. Laskey, Mariela Bollini, Krasimir A. Spasov, Kathleen M. Frey, Karen S. Anderson, Andrea C. Mislak, William L. Jorgensen, William T. Gray, and Won-Gil Lee
- Subjects
Efavirenz ,Resistance ,Population ,Non-nucleoside reverse transcriptase inhibitors ,Biology ,medicine.disease_cause ,Biochemistry ,purl.org/becyt/ford/1 [https] ,chemistry.chemical_compound ,Drug Discovery ,purl.org/becyt/ford/1.4 [https] ,medicine ,Transferase ,education ,chemistry.chemical_classification ,Mutation ,education.field_of_study ,Otras Ciencias Químicas ,Organic Chemistry ,Ciencias Químicas ,HIV ,virus diseases ,Virology ,Reverse transcriptase ,Rreverse transcriptase ,Discovery and development of non-nucleoside reverse-transcriptase inhibitors ,Enzyme ,chemistry ,Rilpivirine ,CIENCIAS NATURALES Y EXACTAS ,Mutations - Abstract
Catechol diether compounds have nanomolar antiviral and enzymatic activity against HIV with reverse transcriptase (RT) variants containing K101P, a mutation that confers high-level resistance to FDA-approved non-nucleoside inhibitors efavirenz and rilpivirine. Kinetic data suggests that RT (K101P) variants are as catalytically fit as wild-type and thus can potentially increase in the viral population as more antiviral regimens include efavirenz or rilpivirine. Comparison of wild-type structures and a new crystal structure of RT (K101P) in complex with a leading compound confirms that the K101P mutation is not a liability for the catechol diethers while suggesting that key interactions are lost with efavirenz and rilpivirine. Fil: Gray, William T.. University of Yale; Estados Unidos Fil: Frey, Kathleen M.. University of Yale; Estados Unidos Fil: Laskey, Sarah B.. University Johns Hopkins; Estados Unidos Fil: Mislak, Andrea C.. University of Yale; Estados Unidos Fil: Spasov, Krasimir A.. University of Yale; Estados Unidos Fil: Lee, Won Gil. University of Yale; Estados Unidos Fil: Bollini, Mariela. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. University of Yale; Estados Unidos Fil: Siliciano, Robert F.. University Johns Hopkins; Estados Unidos. Howard Hughes Medial Institute; Estados Unidos Fil: Jorgensen, William L.. University of Yale; Estados Unidos Fil: Anderson, Karen S.. University of Yale; Estados Unidos
- Published
- 2015
- Full Text
- View/download PDF
14. Structure-Based Evaluation of Non-nucleoside Inhibitors with Improved Potency and Solubility That Target HIV Reverse Transcriptase Variants
- Author
-
Karen S. Anderson, Kathleen M. Frey, Krasimir A. Spasov, William L. Jorgensen, David E. Puleo, and Mariella Bollini
- Subjects
Models, Molecular ,Anti-HIV Agents ,Stereochemistry ,Allosteric regulation ,Mutant ,HIV Infections ,Crystallography, X-Ray ,medicine.disease_cause ,01 natural sciences ,Article ,Cell Line ,HIV reverse transcritase variants ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Humans ,Point Mutation ,030304 developmental biology ,0303 health sciences ,Mutation ,Catechol ,biology ,010405 organic chemistry ,Point mutation ,Otras Ciencias Naturales y Exactas ,virus diseases ,Active site ,HIV Reverse Transcriptase ,Reverse transcriptase ,0104 chemical sciences ,3. Good health ,Solubility ,chemistry ,HIV-1 ,catechol diether ,biology.protein ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Nucleoside ,CIENCIAS NATURALES Y EXACTAS - Abstract
The development of novel non-nucleoside inhibitors (NNRTIs) with activity against variants of HIV reverse transcriptase (RT) is crucial for overcoming treatment failure. The NNRTIs bind in an allosteric pocket in RT ∼10 Å away from the active site. Earlier analogues of the catechol diether compound series have picomolar activity against HIV strains with wild-type RT but lose potency against variants with single Y181C and double K103N/Y181C mutations. As guided by structure-based and computational studies, removal of the 5-Cl substitution of compound 1 on the catechol aryl ring system led to a new analogue compound 2 that maintains greater potency against Y181C and K103N/Y181C variants and better solubility (510 μg/mL). Crystal structures were determined for wild-type, Y181C, and K103N/Y181C RT in complex with both compounds 1 and 2 to understand the structural basis for these findings. Comparison of the structures reveals that the Y181C mutation destabilizes the binding mode of compound 1 and disrupts the interactions with residues in the pocket. Compound 2 maintains the same conformation in wild-type and mutant structures, in addition to several interactions with the NNRTI binding pocket. Comparison of the six crystal structures will assist in the understanding of compound binding modes and future optimization of the catechol diether series. Fil: Frey, Kathleen M.. University of Yale; Estados Unidos Fil: Puleo, David. University of Yale; Estados Unidos Fil: Spasov, Krasimir A.. University of Yale; Estados Unidos Fil: Bollini, Mariela. University of Yale; Estados Unidos. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Centro de Investigaciones en Bionanociencias "Elizabeth Jares Erijman"; Argentina Fil: Jorgensen, William L.. University of Yale; Estados Unidos Fil: Anderson, Karen S.. University of Yale; Estados Unidos
- Published
- 2015
- Full Text
- View/download PDF
15. From in silico hit to long-acting late-stage preclinical candidate to combat HIV-1 infection
- Author
-
Karen S. Anderson, W. Mark Saltzman, Won-Gil Lee, Krasimir A. Spasov, Jagadish Beloor, Priti Kumar, José A. Cisneros, William L. Jorgensen, Shalley N. Kudalkar, and Elias Quijano
- Subjects
0301 basic medicine ,Drug ,Virtual screening ,Multidisciplinary ,business.industry ,media_common.quotation_subject ,030106 microbiology ,virus diseases ,Pharmacology ,Efficacy ,03 medical and health sciences ,030104 developmental biology ,Pharmacokinetics ,In vivo ,Potency ,Medicine ,business ,Viral load ,Combination drug ,media_common - Abstract
The HIV-1 pandemic affecting over 37 million people worldwide continues, with nearly one-half of the infected population on highly active antiretroviral therapy (HAART). Major therapeutic challenges remain because of the emergence of drug-resistant HIV-1 strains, limitations because of safety and toxicity with current HIV-1 drugs, and patient compliance for lifelong, daily treatment regimens. Nonnucleoside reverse transcriptase inhibitors (NNRTIs) that target the viral polymerase have been a key component of the current HIV-1 combination drug regimens; however, these issues hamper them. Thus, the development of novel more effective NNRTIs as anti-HIV-1 agents with fewer long-term liabilities, efficacy on new drug-resistant HIV-1 strains, and less frequent dosing is crucial. Using a computational and structure-based design strategy to guide lead optimization, a 5 µM virtual screening hit was transformed to a series of very potent nanomolar to picomolar catechol diethers. One representative, compound I, was shown to have nanomolar activity in HIV-1-infected T cells, potency on clinically relevant HIV-1 drug-resistant strains, lack of cytotoxicity and off-target effects, and excellent in vivo pharmacokinetic behavior. In this report, we show the feasibility of compound I as a late-stage preclinical candidate by establishing synergistic antiviral activity with existing HIV-1 drugs and clinical candidates and efficacy in HIV-1-infected humanized [human peripheral blood lymphocyte (Hu-PBL)] mice by completely suppressing viral loads and preventing human CD4+ T-cell loss. Moreover, a long-acting nanoformulation of compound I [compound I nanoparticle (compound I-NP)] in poly(lactide-coglycolide) (PLGA) was developed that shows sustained maintenance of plasma drug concentrations and drug efficacy for almost 3 weeks after a single dose.
- Published
- 2017
- Full Text
- View/download PDF
16. Covalent inhibitors for eradication of drug-resistant HIV-1 reverse transcriptase: From design to protein crystallography
- Author
-
Karen S. Anderson, Krasimir A. Spasov, Amanda B. Buckingham, Albert H. Chan, José A. Cisneros, Zaritza O. Petrova, Shalley N. Kudalkar, William L. Jorgensen, and Won-Gil Lee
- Subjects
0301 basic medicine ,Drug ,Models, Molecular ,Spectrometry, Mass, Electrospray Ionization ,Nevirapine ,Efavirenz ,Anti-HIV Agents ,Protein Conformation ,media_common.quotation_subject ,Drug resistance ,010402 general chemistry ,Crystallography, X-Ray ,01 natural sciences ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Resistance, Viral ,medicine ,Humans ,Point Mutation ,Polymerase ,media_common ,Multidisciplinary ,biology ,virus diseases ,Biological Sciences ,Virology ,Reverse transcriptase ,HIV Reverse Transcriptase ,0104 chemical sciences ,Discovery and development of non-nucleoside reverse-transcriptase inhibitors ,Kinetics ,030104 developmental biology ,chemistry ,Amino Acid Substitution ,Rilpivirine ,Drug Design ,biology.protein ,HIV-1 ,Reverse Transcriptase Inhibitors ,medicine.drug - Abstract
Development of resistance remains a major challenge for drugs to treat HIV-1 infections, including those targeting the essential viral polymerase, HIV-1 reverse transcriptase (RT). Resistance associated with the Tyr181Cys mutation in HIV-1 RT has been a key roadblock in the discovery of nonnucleoside RT inhibitors (NNRTIs). It is the principal point mutation that arises from treatment of HIV-infected patients with nevirapine, the first-in-class drug still widely used, especially in developing countries. We report covalent inhibitors of Tyr181Cys RT (CRTIs) that can completely knock out activity of the resistant mutant and of the particularly challenging Lys103Asn/Tyr181Cys variant. Conclusive evidence for the covalent modification of Cys181 is provided from enzyme inhibition kinetics, mass spectrometry, protein crystallography, and antiviral activity in infected human T-cell assays. The CRTIs are also shown to be selective for Cys181 and have lower cytotoxicity than the approved NNRTI drugs efavirenz and rilpivirine.
- Published
- 2017
17. Illuminating HIV gp120-ligand recognition through computationally-driven optimization of antibody-recruiting molecules
- Author
-
Mark Krystal, Nannan Zhou, Robert A. Domaoal, Krasimir A. Spasov, Don T. Li, William L. Jorgensen, Ran N. Tao, Christopher G. Parker, Markus K. Dahlgren, Takuji Shoda, David Spiegel, Sangil Lee, Navneet Jawanda, Richard E. Sutton, Eugene F. Douglass, and Karen S. Anderson
- Subjects
biology ,Cellular Assay ,Human immunodeficiency virus (HIV) ,Nanotechnology ,General Chemistry ,Computational biology ,medicine.disease_cause ,Ligand (biochemistry) ,Article ,medicine ,biology.protein ,Molecule ,Hiv gp120 ,Computational analysis ,Antibody - Abstract
Here we report on the structure-based optimization of antibody-recruiting molecules targeting HIV gp120 (ARM-H). These studies have leveraged a combination of medicinal chemistry, biochemical and cellular assay analysis, and computation. Our findings have afforded an optimized analog of ARM-H, which is ~1000 fold more potent in gp120-binding and MT-2 antiviral assays than our previously reported derivative. Furthermore, computational analysis, taken together with experimental data, provides evidence that azaindole- and indole-based attachment inhibitors bind gp120 at an accessory hydrophobic pocket beneath the CD4-binding site and can also adopt multiple unique binding modes in interacting with gp120. These results are likely to prove highly enabling in the development of novel HIV attachment inhibitors, and more broadly, they suggest novel applications for ARMs as probes of conformationally flexible systems.
- Published
- 2014
- Full Text
- View/download PDF
18. Picomolar Inhibitors of HIV Reverse Transcriptase Featuring Bicyclic Replacement of a Cyanovinylphenyl Group
- Author
-
Kathleen M. Frey, Karen S. Anderson, Won-Gil Lee, William L. Jorgensen, Krasimir A. Spasov, Ricardo Gallardo-Macias, and Mariela Bollini
- Subjects
Models, Molecular ,Anti-HIV Agents ,Stereochemistry ,Microbial Sensitivity Tests ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Biochemistry ,Article ,Catalysis ,Virus ,Bridged Bicyclo Compounds ,Structure-Activity Relationship ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Structure–activity relationship ,Reactivity (chemistry) ,chemistry.chemical_classification ,Catechol ,Dose-Response Relationship, Drug ,Molecular Structure ,Bicyclic molecule ,General Chemistry ,HIV Reverse Transcriptase ,Reverse transcriptase ,Enzyme ,chemistry ,Rilpivirine ,HIV-1 ,Reverse Transcriptase Inhibitors ,Monte Carlo Method - Abstract
Members of the catechol diether class are highly potent non-nucleoside inhibitors of HIV-1 reverse transcriptase (NNRTIs). The most active compounds yield EC50 values below 0.5 nM in assays using human T-cells infected by wild-type HIV-1. However, these compounds such as rilpivirine, the most recently FDA-approved NNRTI, bear a cyanovinylphenyl (CVP) group. This is an uncommon substructure in drugs that gives reactivity concerns. In the present work, computer simulations were used to design bicyclic replacements for the CVP group. The predicted viability of a 2-cyanoindolizinyl alternative was confirmed experimentally and provided compounds with 0.4 nM activity against the wild-type virus. The compounds also performed well with EC50 values of 10 nM against the challenging HIV-1 variant that contains the Lys103Asn/Tyr181Cys double mutation in the RT enzyme. Indolyl and benzofuranyl analogues were also investigated; the most potent compounds in these cases have EC50 values toward wild-type HIV-1 near 10 nM and high-nanomolar activities toward the double-variant. The structural expectations from the modeling were much enhanced by obtaining an X-ray crystal structure at 2.88 Å resolution for the complex of the parent 2-cyanoindolizine 10b and HIV-1 RT. The aqueous solubilities of the most potent indolizine analogues were also measured to be ~40 μg/mL, which is similar to that for the approved drug efavirenz and ~1000-fold greater than for rilpivirine.
- Published
- 2013
- Full Text
- View/download PDF
19. Optimization of diarylazines as anti-HIV agents with dramatically enhanced solubility
- Author
-
Karen S. Anderson, José A. Cisneros, William L. Jorgensen, Mariela Bollini, and Krasimir A. Spasov
- Subjects
Models, Molecular ,Anti-HIV Agents ,Clinical Biochemistry ,Substituent ,Pharmaceutical Science ,Etravirine ,Microbial Sensitivity Tests ,Pharmacology ,Biochemistry ,Article ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Binding site ,Solubility ,Molecular Biology ,Cell Line, Transformed ,Cell Proliferation ,Dose-Response Relationship, Drug ,Molecular Structure ,Anti hiv ,Organic Chemistry ,Reverse transcriptase ,Hydrazines ,chemistry ,Rilpivirine ,HIV-1 ,Molecular Medicine ,medicine.drug - Abstract
Non-nucleoside inhibitors of HIV-1 reverse transcriptase are reported that have ca. 100-fold greater solubility than the structurally related drugs etravirine and rilpivirine, while retaining high anti-viral activity. The solubility enhancements come from strategic placement of a morpholinylalkoxy substituent in the entrance channel of the NNRTI binding site. Compound 4d shows low-nanomolar activity similar to etravirine towards wild-type HIV-1 and key viral variants.
- Published
- 2013
- Full Text
- View/download PDF
20. Discovery of dimeric inhibitors by extension into the entrance channel of HIV-1 reverse transcriptase
- Author
-
Karen S. Anderson, Anil R. Ekkati, Mariela Bollini, Krasimir A. Spasov, Robert A. Domaoal, and William L. Jorgensen
- Subjects
Models, Molecular ,Anti-HIV Agents ,Stereochemistry ,Amino Acid Motifs ,Clinical Biochemistry ,Human immunodeficiency virus (HIV) ,Pharmaceutical Science ,Crystallography, X-Ray ,medicine.disease_cause ,Biochemistry ,Article ,immune system diseases ,Drug Discovery ,medicine ,Humans ,Computer Simulation ,Molecular Biology ,Molecular Structure ,Chemistry ,Drug discovery ,Organic Chemistry ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,HIV Reverse Transcriptase ,Reverse transcriptase ,Entrance channel ,HIV-1 ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Triazenes ,Dimerization - Abstract
Design of non-nucleoside inhibitors of HIV-1 reverse transcriptase is being pursued with computational guidance. Extension of azine-containing inhibitors into the entrance channel between Lys103 and Glu138 has led to the discovery of potent and structurally novel derivatives including dimeric inhibitors in an NNRTI-linker-NNRTI motif.
- Published
- 2012
- Full Text
- View/download PDF
21. Computationally-Guided Optimization of a Docking Hit to Yield Catechol Diethers as Potent Anti-HIV Agents
- Author
-
Vinay V. Thakur, Krasimir A. Spasov, Ricardo Gallardo-Macias, Karen S. Anderson, Mariela Bollini, Robert A. Domaoal, and William L. Jorgensen
- Subjects
Models, Molecular ,Quantitative structure–activity relationship ,Anti-HIV Agents ,Stereochemistry ,T-Lymphocytes ,Catechols ,Quantitative Structure-Activity Relationship ,Stereoisomerism ,Article ,Free energy perturbation ,chemistry.chemical_compound ,Drug Discovery ,Humans ,Computer Simulation ,Catechol ,Halogen bond ,Molecular Structure ,Uracil ,HIV Reverse Transcriptase ,chemistry ,Docking (molecular) ,HIV-1 ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Linker ,Ethers ,Protein Binding - Abstract
A 5-μM docking hit has been optimized to an extraordinarily potent (55 pM) non-nucleoside inhibitor of HIV reverse transcriptase. Use of free energy perturbation (FEP) calculations to predict relative free energies of binding aided the optimizations by identifying optimal substitution patterns for phenyl rings and a linker. The most potent resultant catechol diethers feature terminal uracil and cyanovinylphenyl groups. A halogen bond with Pro95 likely contributes to the extreme potency of compound 42. In addition, several examples are provided illustrating failures of attempted grafting of a substructure from a very active compound onto a seemingly related scaffold to improve its activity.
- Published
- 2011
- Full Text
- View/download PDF
22. Structural fold, conservation and Fe(II) binding of the intracellular domain of prokaryote FeoB
- Author
-
Tai Huang Huang, Krasimir A. Spasov, Yuh-Ju Sun, Vinzenz M. Unger, Yi Chung Chen, Gang Dong, Kuo Wei Hung, Jai Hui Chen, Edward T. Eng, Chwan Deng Hsiao, and Yi-Wei Chang
- Subjects
Models, Molecular ,Protein Folding ,Protein Conformation ,Molecular Sequence Data ,Crystallography, X-Ray ,Article ,Protein structure ,Bacterial Proteins ,Species Specificity ,GTP-Binding Proteins ,Structural Biology ,Amino Acid Sequence ,Ferrous Compounds ,Binding site ,Cation Transport Proteins ,Binding Sites ,Sequence Homology, Amino Acid ,biology ,Chemistry ,biology.organism_classification ,Recombinant Proteins ,Protein Structure, Tertiary ,Transport protein ,Pyrococcus furiosus ,Kinetics ,Klebsiella pneumoniae ,Crystallography ,Structural Homology, Protein ,G-domain ,Biophysics ,Protein folding ,Ferrous iron transport ,Binding domain - Abstract
FeoB is a G-protein coupled membrane protein essential for Fe(II) uptake in prokaryotes. Here, we report the crystal structures of the intracellular domain of FeoB (NFeoB) from Klebsiella pneumoniae (KpNFeoB) and Pyrococcus furiosus (PfNFeoB) with and without bound ligands. In the structures, a canonical G-protein domain (G domain) is followed by a helical bundle domain (S-domain), which despite its lack of sequence similarity between species is structurally conserved. In the nucleotide-free state, the G-domain's two switch regions point away from the binding site. This gives rise to an open binding pocket whose shallowness is likely to be responsible for the low nucleotide-binding affinity. Nucleotide binding induced significant conformational changes in the G5 motif which in the case of GMPPNP binding was accompanied by destabilization of the switch I region. In addition to the structural data, we demonstrate that Fe(II)-induced foot printing cleaves the protein close to a putative Fe(II)-binding site at the tip of switch I, and we identify functionally important regions within the S-domain. Moreover, we show that NFeoB exists as a monomer in solution, and that its two constituent domains can undergo large conformational changes. The data show that the S-domain plays important roles in FeoB function.
- Published
- 2010
- Full Text
- View/download PDF
23. Structural Basis of Membrane Invagination by F-BAR Domains
- Author
-
Olivier Destaing, Rushika M. Perera, Krasimir A. Spasov, Adam Frost, Aurélien Roux, Edward H. Egelman, Pietro De Camilli, and Vinzenz M. Unger
- Subjects
Dynamins ,Models, Molecular ,Green Fluorescent Proteins ,Biology ,Fatty Acid-Binding Proteins ,Transfection ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Minor Histocompatibility Antigens ,Cell membrane ,Membrane bending ,03 medical and health sciences ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,BAR domain ,030304 developmental biology ,Membrane invagination ,0303 health sciences ,Membrane tubulation ,Biochemistry, Genetics and Molecular Biology(all) ,Bilayer ,Cell Membrane ,Cryoelectron Microscopy ,030302 biochemistry & molecular biology ,Membrane Proteins ,Protein Structure, Tertiary ,Cell biology ,medicine.anatomical_structure ,Membrane ,Membrane protein ,COS Cells ,Liposomes ,Biophysics ,Carrier Proteins ,Microtubule-Associated Proteins - Abstract
SummaryBAR superfamily domains shape membranes through poorly understood mechanisms. We solved structures of F-BAR modules bound to flat and curved bilayers using electron (cryo)microscopy. We show that membrane tubules form when F-BARs polymerize into helical coats that are held together by lateral and tip-to-tip interactions. On gel-state membranes or after mutation of residues along the lateral interaction surface, F-BARs adsorb onto bilayers via surfaces other than their concave face. We conclude that membrane binding is separable from membrane bending, and that imposition of the module's concave surface forces fluid-phase bilayers to bend locally. Furthermore, exposure of the domain's lateral interaction surface through a change in orientation serves as the crucial trigger for assembly of the helical coat and propagation of bilayer bending. The geometric constraints and sequential assembly of the helical lattice explain how F-BAR and classical BAR domains segregate into distinct microdomains, and provide insight into the spatial regulation of membrane invagination.
- Published
- 2008
- Full Text
- View/download PDF
24. Characterization of a Novel Prokaryotic GDP Dissociation Inhibitor Domain from the G Protein Coupled Membrane Protein FeoB
- Author
-
Krasimir A. Spasov, Amir Reza Jalilian, Edward T. Eng, and Vinzenz M. Unger
- Subjects
G protein ,Amino Acid Motifs ,Molecular Sequence Data ,Biology ,Guanosine Diphosphate ,Models, Biological ,Article ,chemistry.chemical_compound ,GTP-binding protein regulators ,GTP-Binding Proteins ,Structural Biology ,rho-Specific Guanine Nucleotide Dissociation Inhibitors ,Amino Acid Sequence ,Cation Transport Proteins ,Molecular Biology ,Guanine Nucleotide Dissociation Inhibitors ,Sequence Homology, Amino Acid ,Escherichia coli Proteins ,GDP binding ,Protein Structure, Tertiary ,Transport protein ,Prokaryotic Cells ,chemistry ,Membrane protein ,Biochemistry ,Guanosine diphosphate ,Ferrous iron transport - Abstract
The FeoB family of membrane embedded G proteins are involved with high affinity Fe(II) uptake in prokaryotes. Here, we report that FeoB harbors a novel GDP dissociation inhibitor-like domain that specifically stabilizes GDP-binding through an interaction with the switch I region of the G protein. We show that the stabilization of GDP binding is conserved between species despite a high degree of sequence variability in their guanine nucleotide dissociation inhibitor (GDI)-like domains, and demonstrate that the presence of the membrane embedded domain increases GDP-binding affinity roughly 150-fold over the level accomplished by action of the GDI-like domain alone. To our knowledge, this is the first example for a prokaryotic GDI, targeting a bacterial G protein-coupled membrane process. Our findings suggest that Fe(II) uptake in bacteria involves a G protein regulatory pathway reminiscent of signaling mechanisms found in higher-order organisms.
- Published
- 2008
- Full Text
- View/download PDF
25. Picomolar Inhibitors of HIV-1 Reverse Transcriptase: Design and Crystallography of Naphthyl Phenyl Ethers
- Author
-
Won-Gil Lee, Krasimir A. Spasov, Ricardo Gallardo-Macias, Kathleen M. Frey, William L. Jorgensen, Karen S. Anderson, and Mariela Bollini
- Subjects
Catechol ,Chemistry ,Stereochemistry ,ANTI-HIV AGENTS ,PROTEIN CRYSTALLOGRAPHY ,Organic Chemistry ,Substituent ,Human immunodeficiency virus (HIV) ,Ciencias Químicas ,virus diseases ,Phenyl Ethers ,Crystal structure ,medicine.disease_cause ,Biochemistry ,Reverse transcriptase ,Virus ,chemistry.chemical_compound ,Química Orgánica ,Drug Discovery ,Hydrolase ,medicine ,NNRTIS ,CIENCIAS NATURALES Y EXACTAS - Abstract
Catechol diethers that incorporate a 6-cyano-1-naphthyl substituent have been explored as non-nucleoside inhibitors of HIV-1 reverse transcriptase (NNRTIs). Promising compounds are reported that show midpicomolar activity against the wild-type virus and sub-20 nM activity against viral variants bearing Tyr181Cys and Lys103Asn mutations in HIV-RT. An X-ray crystal structure at 2.49 Å resolution is also reported for the key compound 6e with HIV-RT. Fil: Won-Gil Lee. University of Yale; Estados Unidos Fil: Frey, Kathleen M.. University of Yale; Estados Unidos Fil: Ricardo Gallardo-Macias. University of Yale; Estados Unidos Fil: Spasov, Krasimir A.. University of Yale; Estados Unidos Fil: Bollini, Mariela. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Houssay; Argentina. University of Yale; Estados Unidos Fil: Anderson, Karen S.. University of Yale; Estados Unidos Fil: Jorgensen, William L.. University of Yale; Estados Unidos
- Published
- 2014
26. Structure-based evaluation of C5 derivatives in the catechol diether series targeting HIV-1 reverse transcriptase
- Author
-
Krasimir A. Spasov, William L. Jorgensen, Mariela Bollini, William T. Gray, Ricardo Gallardo-Macias, Karen S. Anderson, and Kathleen M. Frey
- Subjects
Stereochemistry ,Catechols ,Substituent ,Halogen bonds ,Crystal structure ,Molecular Dynamics Simulation ,Biochemistry ,Article ,purl.org/becyt/ford/1 [https] ,Structure-Activity Relationship ,Molecular dynamics ,chemistry.chemical_compound ,Drug Discovery ,purl.org/becyt/ford/1.4 [https] ,Structure–activity relationship ,Uracil ,Pharmacology ,Binding Sites ,Hydrogen bond ,Organic Chemistry ,Ciencias Químicas ,Hydrogen Bonding ,HIV Reverse Transcriptase ,Reverse transcriptase ,Protein Structure, Tertiary ,Química Orgánica ,chemistry ,Drug Design ,HIV-1 ,Alkoxy group ,Structure based drug design ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,HIV-1 reverse transcriptase ,Nonnucleoside reverse transcriptase inhibitors ,CIENCIAS NATURALES Y EXACTAS ,Structure activity relationships - Abstract
Using a computationally driven approach, a class of inhibitors with picomolar potency known as the catechol diethers were developed targeting the non-nucleoside-binding pocket of HIV-1 reverse transcriptase. Computational studies suggested that halogen-bonding interactions between the C5 substituent of the inhibitor and backbone carbonyl of conserved residue Pro95 might be important. While the recently reported crystal structures of the reverse transcriptase complexes confirmed the interactions with the non-nucleoside-binding pocket, they revealed the lack of a halogen-bonding interaction with Pro95. To understand the effects of substituents at the C5 position, we determined additional crystal structures with 5-Br and 5-H derivatives. Using comparative structural analysis, we identified several conformations of the ethoxy uracil dependent on the strength of a van der Waals interaction with the Cγ of Pro95 and the C5 substitution. The 5-Cl and 5-F derivatives position the ethoxy uracil to make more hydrogen bonds, whereas the larger 5-Br and smaller 5-H position the ethoxy uracil to make fewer hydrogen bonds. EC50 values correlate with the trends observed in the crystal structures. The influence of C5 substitutions on the ethoxy uracil conformation may have strategic value, as future derivatives can possibly be modulated to gain additional hydrogen-bonding interactions with resistant variants of reverse transcriptase. Fil: Frey, Kathleen M.. University of Yale; Estados Unidos Fil: Gray, William T.. University of Yale; Estados Unidos Fil: Spasov, Krasimir A.. University of Yale; Estados Unidos Fil: Bollini, Mariela. University of Yale; Estados Unidos. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina Fil: Gallardo Macias, Ricardo. University of Yale; Estados Unidos Fil: Jorgensen, William L.. University of Yale; Estados Unidos Fil: Anderson, Karen S.. University of Yale; Estados Unidos
- Published
- 2014
- Full Text
- View/download PDF
27. First three-dimensional structure of Toxoplasma gondii thymidylate synthase-dihydrofolate reductase: insights for catalysis, interdomain interactions, and substrate channeling
- Author
-
Melissa A. Vargo, Hitesh Sharma, Mark J. Landau, Krasimir A. Spasov, and Karen S. Anderson
- Subjects
Models, Molecular ,Substrate channeling ,Protozoan Proteins ,Reductase ,Biochemistry ,Thymidylate synthase ,Catalysis ,Protein Structure, Secondary ,Article ,Substrate Specificity ,Folic Acid ,Imaging, Three-Dimensional ,Multienzyme Complexes ,Catalytic Domain ,Dihydrofolate reductase ,parasitic diseases ,Transferase ,heterocyclic compounds ,chemistry.chemical_classification ,Binding Sites ,biology ,Mutagenesis ,Toxoplasma gondii ,Thymidylate Synthase ,biology.organism_classification ,enzymes and coenzymes (carbohydrates) ,Kinetics ,Tetrahydrofolate Dehydrogenase ,Enzyme ,chemistry ,biology.protein ,Toxoplasma - Abstract
Most species, such as humans, have monofunctional forms of thymidylate synthase (TS) and dihydrofolate reductase (DHFR) that are key folate metabolism enzymes making critical folate components required for DNA synthesis. In contrast, several parasitic protozoa, including Toxoplasma gondii , contain a unique bifunctional thymidylate synthase-dihydrofolate reductase (TS-DHFR) having the catalytic activities contained on a single polypeptide chain. The prevalence of T. gondii infections across the world, especially for those immunocompromised, underscores the need to understand TS-DHFR enzyme function and to find new avenues to exploit for the design of novel antiparasitic drugs. As a first step, we have solved the first three-dimensional structures of T. gondii TS-DHFR at 3.7 Å and of a loop truncated TS-DHFR, removing several flexible surface loops in the DHFR domain, improving resolution to 2.2 Å. Distinct structural features of the TS-DHFR homodimer include a junctional region containing a kinked crossover helix between the DHFR domains of the two adjacent monomers, a long linker connecting the TS and DHFR domains, and a DHFR domain that is positively charged. The roles of these unique structural features were probed by site-directed mutagenesis coupled with presteady state and steady state kinetics. Mutational analysis of the crossover helix region combined with kinetic characterization established the importance of this region not only in DHFR catalysis but also in modulating the distal TS activity, suggesting a role for TS-DHFR interdomain interactions. Additional kinetic studies revealed that substrate channeling occurs in which dihydrofolate is directly transferred from the TS to DHFR active site without entering bulk solution. The crystal structure suggests that the positively charged DHFR domain governs this electrostatically mediated movement of dihydrofolate, preventing release from the enzyme. Taken together, these structural and kinetic studies reveal unique, functional regions on the T. gondii TS-DHFR enzyme that may be targeted for inhibition, thus paving the way for designing species specific inhibitors.
- Published
- 2013
28. Extension into the entrance channel of HIV-1 reverse transcriptase—Crystallography and enhanced solubility
- Author
-
Mariela Bollini, Krasimir A. Spasov, José A. Cisneros, William L. Jorgensen, Karen S. Anderson, Kalyan Das, Joseph D. Bauman, Eddy Arnold, and Kathleen M. Frey
- Subjects
Models, Molecular ,NNRTI ,Stereochemistry ,Anti-HIV Agents ,Clinical Biochemistry ,Human immunodeficiency virus (HIV) ,Substituent ,Pharmaceutical Science ,Crystal structure ,Anti-HIV agent ,medicine.disease_cause ,Crystallography, X-Ray ,Biochemistry ,Article ,purl.org/becyt/ford/1 [https] ,chemistry.chemical_compound ,Structure-Activity Relationship ,Drug Discovery ,medicine ,purl.org/becyt/ford/1.4 [https] ,Solubility ,Triazene ,Enzyme Inhibitors ,Molecular Biology ,Drug solubility ,Dose-Response Relationship, Drug ,Molecular Structure ,Organic Chemistry ,Ciencias Químicas ,virus diseases ,Reverse transcriptase ,HIV Reverse Transcriptase ,Entrance channel ,Crystallography ,Pyrimidines ,Química Orgánica ,chemistry ,Crystal structures ,HIV-1 ,Molecular Medicine ,CIENCIAS NATURALES Y EXACTAS - Abstract
Non-nucleoside inhibitors of HIV-1 reverse transcriptase (HIV-RT) are reported that feature extension into the entrance channel near Glu138. Complexes of the parent anilinylpyrimidine 1 and the morpholinoethoxy analog 2j with HIV-RT have received crystallographic characterization confirming the designs. Measurement of aqueous solubilities of 2j, 2k, the parent triazene 2a, and other NNRTIs demonstrate profound benefits for addition of the morpholinyl substituent. Fil: Bollini, Mariela. University of Yale; Estados Unidos. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina Fil: Frey, Kathleen M.. University of Yale. School of Medicine; Estados Unidos Fil: Cisneros, José A.. University of Yale; Estados Unidos Fil: Spasov, Krasimir A.. University of Yale. School of Medicine; Estados Unidos Fil: Das, Kalyan. Rutgers University; Estados Unidos Fil: Bauman, Joseph D.. Rutgers University; Estados Unidos Fil: Arnold, Eddy. Rutgers University; Estados Unidos Fil: Anderson, Karen S.. University of Yale. School of Medicine; Estados Unidos Fil: Jorgensen, William L.. University of Yale; Estados Unidos
- Published
- 2013
29. Optimization of benzyloxazoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase to enhance Y181C potency
- Author
-
Mariela Bollini, Julian Tirado-Rives, Karen S. Anderson, William L. Jorgensen, Krasimir A. Spasov, and Ricardo Gallardo-Macias
- Subjects
Models, Molecular ,Anti-HIV agents ,Anti-HIV Agents ,Stereochemistry ,Clinical Biochemistry ,Substituent ,Human immunodeficiency virus (HIV) ,Pharmaceutical Science ,medicine.disease_cause ,Biochemistry ,Article ,Virus ,Free-energy calculations ,Free energy perturbation ,Structure-Activity Relationship ,chemistry.chemical_compound ,Computer-aided drug design ,Drug Discovery ,medicine ,Humans ,Potency ,Molecular Biology ,Chemistry ,Otras Ciencias Químicas ,Organic Chemistry ,Ciencias Químicas ,HIV Reverse Transcriptase ,Reverse transcriptase ,Drug Design ,HIV-1 ,NNRTIs ,Reverse Transcriptase Inhibitors ,Molecular Medicine ,Nucleoside ,Isopropyl ,CIENCIAS NATURALES Y EXACTAS - Abstract
Design of non-nucleoside inhibitors of HIV-1 reverse transcriptase with improved activity towards Tyr181Cys containing variants was pursued with the assistance of free energy perturbation (FEP) calculations. Optimization of the 4-R substituent in 1 led to ethyl and isopropyl analogs 1e and 1f with 1–7 nM potency towards both the wild-type virus and a Tyr181C variant. Fil: Bollini, Mariela. University of Yale; Estados Unidos. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina Fil: Gallardo Macias, Ricardo. University of Yale; Estados Unidos Fil: Spasov, Krasimir A.. University of Yale. School of Medicine; Estados Unidos Fil: Tirado Rives, Julian. University of Yale; Estados Unidos Fil: Anderson, Karen S.. University of Yale. School of Medicine; Estados Unidos Fil: Jorgensen, William L.. University of Yale; Estados Unidos
- Published
- 2012
30. Efficient discovery of potent anti-HIV agents targeting the Tyr181Cys variant of HIV reverse transcriptase
- Author
-
Krasimir A. Spasov, Vinay V. Thakur, Robert A. Domaoal, William L. Jorgensen, Karen S. Anderson, and Mariela Bollini
- Subjects
Models, Molecular ,Pyrimidine ,Molecular model ,Stereochemistry ,Anti-HIV Agents ,Protein Conformation ,Substituent ,Biochemistry ,Catalysis ,Article ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Colloid and Surface Chemistry ,Protein structure ,Drug Discovery ,Nitriles ,Potency ,Structure–activity relationship ,Molecular Targeted Therapy ,Chemistry ,Drug discovery ,Triazines ,General Chemistry ,Reverse transcriptase ,HIV Reverse Transcriptase ,Pyrimidines ,Amino Acid Substitution ,HIV-1 ,Thermodynamics - Abstract
Non-nucleoside reverse transcriptase inhibitors (NNRTIs) that interfere with the replication of human immunodeficiency virus (HIV) are being pursued with guidance from molecular modeling including free-energy perturbation (FEP) calculations for protein-inhibitor binding affinities. The previously reported pyrimidinylphenylamine 1 and its chloro analogue 2 are potent anti-HIV agents; they inhibit replication of wild-type HIV-1 in infected human T-cells with EC(50) values of 2 and 10 nM, respectively. However, they show no activity against viral strains containing the Tyr181Cys (Y181C) mutation in HIV-RT. Modeling indicates that the problem is likely associated with extensive interaction between the dimethylallyloxy substituent and Tyr181. As an alternative, a phenoxy group is computed to be oriented in a manner diminishing the contact with Tyr181. However, this replacement leads to a roughly 1000-fold loss of activity for 3 (2.5 μM). The present report details the efficient, computationally driven evolution of 3 to novel NNRTIs with sub-10 nM potency toward both wild-type HIV-1 and Y181C-containing variants. The critical contributors were FEP substituent scans for the phenoxy and pyrimidine rings and recognition of potential benefits of addition of a cyanovinyl group to the phenoxy ring.
- Published
- 2011
31. Quasi-Biomimetic Ring Contraction Catalyzed by a Cysteine-Based Nucleophile: Total Synthesis of Sch-642305, Some Analogs and their Putative anti-HIV Activities
- Author
-
Alpay Dermenci, Karen S. Anderson, Scott J. Miller, Robert A. Domaoal, Krasimir A. Spasov, and Philipp S. Selig
- Subjects
Chemistry ,Stereochemistry ,Anti hiv ,Total synthesis ,Nanotechnology ,General Chemistry ,Sch 642305 ,Article ,Catalysis ,chemistry.chemical_compound ,Nucleophile ,Rauhut–Currier reaction ,Biomimetic synthesis ,Cysteine - Abstract
Cysteine plays a number of important functional and structural roles in Nature, often in the realm of catalysis. Herein, we present an example of a cysteine-catalyzed Rauhut-Currier reaction for a potentially biomimetic synthesis of Sch-642305 and related analogs. In this key step of the synthesis we discuss interesting new discoveries and the importance of substrate-catalyst recognition, as well as cysteine's structural features. Also, we investigate the activity of Sch-642305 and four analogs in HIV-infected T-cells.
- Published
- 2011
32. Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse transcriptase; cyano group alternatives
- Author
-
Karen S. Anderson, Jacob G. Zeevaart, Cheryl S. Leung, William L. Jorgensen, Robert A. Domaoal, Mariela Bollini, Vinay V. Thakur, and Krasimir A. Spasov
- Subjects
Azoles ,Models, Molecular ,Stereochemistry ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Human immunodeficiency virus (HIV) ,Pharmaceutical Science ,medicine.disease_cause ,Biochemistry ,Reverse transcriptase ,Article ,Free energy perturbation ,Drug Discovery ,medicine ,Anti-hiv drugs ,HIV-1 ,Molecular Medicine ,Reverse Transcriptase Inhibitors ,Free energies ,Molecular Biology ,Nucleoside - Abstract
Design of non-nucleoside inhibitors of HIV-1 reverse transcriptase is being pursued with the assistance of free energy perturbation (FEP) calculations to predict relative free energies of binding. Extension of azole-containing inhibitors into an ‘eastern’ channel between Phe227 and Pro236 has led to the discovery of potent and structurally novel derivatives.
- Published
- 2010
33. Mechanistic Insights into Membrane Remodeling Through BAR‐Domain Proteins
- Author
-
Carsten Mim, Vinzenz M. Unger, Edward H. Egelman, Pietro De Camilli, Rushika M. Perera, Adam Frost, and Krasimir A. Spasov
- Subjects
Chemistry ,Membrane remodeling ,Genetics ,Biophysics ,BAR domain ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2009
- Full Text
- View/download PDF
34. Computationally-GuidedOptimization ofa Docking Hit to Yield Catechol Diethers as Potent Anti-HIV Agents.
- Author
-
Mariela Bollini, RobertA. Domaoal, Vinay V. Thakur, Ricardo Gallardo-Macias, Krasimir A. Spasov, Karen S. Anderson, and William L. Jorgensen
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.