An orally bioavailable SARS-CoV-2 main protease inhibitor exhibits improved affinity and reduced sensitivity to mutations

Inhibitors of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro) such as nirmatrelvir (NTV) and ensitrelvir (ETV) have proven effective in reducing the severity of COVID-19, but the presence of resistance-conferring mutations in sequenced viral genomes raises concerns about future drug resistance. Second-generation oral drugs that retain function against these mutants are thus urgently needed. We hypothesized that the covalent hepatitis C virus protease inhibitor boceprevir (BPV) could serve as the basis for orally bioavailable drugs that inhibit SARS-CoV-2 Mpro more efficiently than existing drugs. Performing structure-guided modifications of BPV, we developed a picomolar-affinity inhibitor, ML2006a4, with antiviral activity, oral pharmacokinetics, and therapeutic efficacy similar or superior to those of NTV. A crucial feature of ML2006a4 is a derivatization of the ketoamide reactive group that improves cell permeability and oral bioavailability. Last, ML2006a4 was found to be less sensitive to several mutations that cause resistance to NTV or ETV and occur in the natural SARS-CoV-2 population. Thus, anticipatory design can preemptively address potential resistance mechanisms to expand future treatment options against coronavirus variants. ML2006a4 has improved affinity for SARS-CoV-2 Mpro and reduced sensitivity to mutations conferring resistance to nirmatrelvir or ensitrelvir. Editor’s summary The toolkit for preventing and treating patients with COVID-19 has expanded greatly since the pandemic began. Some of the most effective therapeutics for COVID-19 and, potentially, other coronavirus infections are antivirals that inhibit the SARS-CoV-2 main protease (Mpro). Mpro inhibitors include nirmatrelvir, a component of the oral treatment Paxlovid. However, second-generation drugs are needed, because there is a risk that new SARS-CoV-2 variants could become resistant to nirmatrelvir and other antivirals in clinical use. Here, Westberg et al. used the hepatitis C virus protease inhibitor boceprevir as a starting point to make such a next-generation Mpro inhibitor. Their optimized lead candidate, ML2006a4, exhibited robust antiviral activity in vitro and in mice infected with SARS-CoV-2 and could be administered orally. ML2006a4 appeared less sensitive to mutations in the SARS-CoV-2 Mpro, suggesting that the virus would be less likely to become resistant to ML2006a4 in the real world. —Courtney Malo

[1]  J. Teijaro,et al.  Olgotrelvir, a dual inhibitor of SARS-CoV-2 Mpro and cathepsin L, as a standalone antiviral oral intervention candidate for COVID-19. , 2024, i Medicina.

[2]  Yves L Janin On the origins of SARS-CoV-2 main protease inhibitors. , 2024, RSC Medicinal Chemistry.

[3]  Ye-ming Wang,et al.  Oral Simnotrelvir for Adult Patients with Mild-to-Moderate Covid-19. , 2024, New England Journal of Medicine.

[4]  J. Teijaro,et al.  Olgotrelvir, a dual inhibitor of SARS-CoV-2 Mpro and cathepsin L, as a standalone antiviral oral intervention candidate for COVID-19. , 2023, i Medicina.

[5]  A. Ayitou,et al.  Discovery of Nirmatrelvir Resistance Mutations in SARS-CoV-2 3CLpro: A Computational-Experimental Approach , 2023, J. Chem. Inf. Model..

[6]  Gengfu Xiao,et al.  Structure-based development and preclinical evaluation of the SARS-CoV-2 3C-like protease inhibitor simnotrelvir , 2023, Nature communications.

[7]  A. Pruijssers,et al.  Evaluation of in vitro antiviral activity of SARS-CoV-2 Mpro inhibitor pomotrelvir and cross-resistance to nirmatrelvir resistance substitutions , 2023, Antimicrobial Agents and Chemotherapy.

[8]  D. von Laer,et al.  A comprehensive study of SARS-CoV-2 main protease (Mpro) inhibitor-resistant mutants selected in a VSV-based system , 2023, bioRxiv.

[9]  Seo Jung Hong,et al.  Molecular mechanisms of SARS-CoV-2 resistance to nirmatrelvir , 2023, Nature.

[10]  N. Zhong,et al.  Antiviral efficacy of RAY1216 monotherapy and combination therapy with ritonavir in patients with COVID-19: a phase 2, single centre, randomised, double-blind, placebo-controlled trial , 2023, EClinicalMedicine.

[11]  Y. Hirotsu,et al.  Multidrug-resistant mutations to antiviral and antibody therapy in an immunocompromised patient infected with SARS-CoV-2. , 2023, Med.

[12]  N. Zuckerman,et al.  Nirmatrelvir resistance - de novo E166V/L50V mutations in an immunocompromised patient treated with prolonged nirmatrelvir/ritonavir monotherapy leading to clinical and virological treatment failure - a case report. , 2023, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[13]  R. Plemper,et al.  Comparing molnupiravir and nirmatrelvir/ritonavir efficacy and the effects on SARS-CoV-2 transmission in animal models , 2023, Nature communications.

[14]  Hao Tan,et al.  Naturally Occurring Mutations of SARS-CoV-2 Main Protease Confer Drug Resistance to Nirmatrelvir , 2023, ACS central science.

[15]  Y. Kawaoka,et al.  In vitro and in vivo characterization of SARS-CoV-2 resistance to ensitrelvir , 2023, Nature Communications.

[16]  Y. Kawaoka,et al.  In vitro and in vivo characterization of SARS-CoV-2 strains resistant to nirmatrelvir , 2023, Nature Communications.

[17]  M. Siedner,et al.  SARS-CoV-2 virologic rebound with nirmatrelvir-ritonavir therapy , 2023, medRxiv.

[18]  B. Weynand,et al.  Nirmatrelvir-resistant SARS-CoV-2 is efficiently transmitted in female Syrian hamsters and retains partial susceptibility to treatment , 2023, Nature communications.

[19]  W. Chan,et al.  Global prevalence of SARS-CoV-2 3CL protease mutations associated with nirmatrelvir or ensitrelvir resistance , 2023, eBioMedicine.

[20]  J. Vederas,et al.  SARS-CoV-2 Mpro Protease Variants of Concern Display Altered Viral Substrate and Cell Host Target Galectin-8 Processing but Retain Sensitivity toward Antivirals , 2023, ACS central science.

[21]  Yifei Wang,et al.  A new generation Mpro inhibitor with potent activity against SARS-CoV-2 Omicron variants , 2023, Signal Transduction and Targeted Therapy.

[22]  X. Xiong,et al.  Inhibition mechanism and antiviral activity of an α-ketoamide based SARS-CoV-2 main protease inhibitor , 2023, bioRxiv.

[23]  C. Schiffer,et al.  Systematic analyses of the resistance potential of drugs targeting SARS-CoV-2 main protease , 2023, bioRxiv.

[24]  D. Harki,et al.  Rapid resistance profiling of SARS-CoV-2 protease inhibitors , 2023, bioRxiv.

[25]  M. Mina,et al.  The COVID-19 Rebound Study: A Prospective Cohort Study to Evaluate Viral and Symptom Rebound Differences in Participants Treated with Nirmatrelvir Plus Ritonavir Versus Untreated Controls. , 2023, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[26]  T. Uehara,et al.  A phase 2/3 study of S-217622 in participants with SARS-CoV-2 infection (Phase 3 part) , 2023, Medicine.

[27]  G. Oliva,et al.  Structural basis of nirmatrelvir and ensitrelvir activity against naturally occurring polymorphisms of the SARS-CoV-2 main protease , 2023, Journal of Biological Chemistry.

[28]  R. Neher,et al.  Fitness effects of mutations to SARS-CoV-2 proteins , 2023, bioRxiv.

[29]  S. Sarafianos,et al.  Nirmatrelvir Resistance in SARS-CoV-2 Omicron_BA.1 and WA1 Replicons and Escape Strategies , 2023, bioRxiv.

[30]  T. Uehara,et al.  Efficacy and Safety of Ensitrelvir in Patients With Mild-to-Moderate Coronavirus Disease 2019: The Phase 2b Part of a Randomized, Placebo-Controlled, Phase 2/3 Study , 2022, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[31]  J. Bukh,et al.  Nirmatrelvir-resistant SARS-CoV-2 variants with high fitness in an infectious cell culture system , 2022, Science advances.

[32]  Seo Jung Hong,et al.  Multiple pathways for SARS-CoV-2 resistance to nirmatrelvir , 2022, Nature.

[33]  P. Maes,et al.  The Substitutions L50F, E166A, and L167F in SARS-CoV-2 3CLpro Are Selected by a Protease Inhibitor In Vitro and Confer Resistance To Nirmatrelvir , 2022, bioRxiv.

[34]  S. Moro,et al.  SARS-CoV-2 3CLpro mutations selected in a VSV-based system confer resistance to nirmatrelvir, ensitrelvir, and GC376 , 2022, Science Translational Medicine.

[35]  Hao Tan,et al.  Naturally occurring mutations of SARS-CoV-2 main protease confer drug resistance to nirmatrelvir , 2022, bioRxiv.

[36]  C. Jackson,et al.  Predicting antiviral resistance mutations in SARS-CoV-2 main protease with computational and experimental screening , 2022, bioRxiv.

[37]  Rommie E. Amaro,et al.  Transmissible SARS-CoV-2 variants with resistance to clinical protease inhibitors , 2022, bioRxiv.

[38]  M. Head,et al.  Hepatitis C virus NS3/4A inhibitors and other drug-like compounds as covalent binders of SARS-CoV-2 main protease , 2022, Scientific Reports.

[39]  Kai S Yang,et al.  Evolutionary and Structural Insights about Potential SARS-CoV-2 Evasion of Nirmatrelvir , 2022, Journal of medicinal chemistry.

[40]  A. Godzik,et al.  Monitoring for SARS-CoV-2 drug resistance mutations in broad viral populations , 2022, bioRxiv.

[41]  A. Kalgutkar,et al.  Comprehensive Nonclinical Safety Assessment of Nirmatrelvir Supporting Timely Development of the SARS-COV-2 Antiviral Therapeutic, Paxlovid™ , 2022, International journal of toxicology.

[42]  B. Weynand,et al.  A SCID Mouse Model To Evaluate the Efficacy of Antivirals against SARS-CoV-2 Infection , 2022, bioRxiv.

[43]  W. Rabeh,et al.  Key dimer interface residues impact the catalytic activity of 3CLpro, the main protease of SARS-CoV-2 , 2022, Journal of Biological Chemistry.

[44]  J. Chan,et al.  An orally available Mpro inhibitor is effective against wild-type SARS-CoV-2 and variants including Omicron , 2022, Nature Microbiology.

[45]  A. Gribenko,et al.  Genetic Surveillance of SARS-CoV-2 Mpro Reveals High Sequence and Structural Conservation Prior to the Introduction of Protease Inhibitor Paxlovid , 2022, bioRxiv.

[46]  Y. Orba,et al.  Discovery of S-217622, a Noncovalent Oral SARS-CoV-2 3CL Protease Inhibitor Clinical Candidate for Treating COVID-19 , 2022, Journal of medicinal chemistry.

[47]  M. Exline,et al.  Effectiveness of mRNA Vaccination in Preventing COVID-19–Associated Invasive Mechanical Ventilation and Death — United States, March 2021–January 2022 , 2022, MMWR. Morbidity and mortality weekly report.

[48]  M. Baniecki,et al.  Oral Nirmatrelvir for High-Risk, Nonhospitalized Adults with Covid-19 , 2022, The New England journal of medicine.

[49]  G. Nucci,et al.  Innovative Randomized Phase I Study and Dosing Regimen Selection to Accelerate and Inform Pivotal COVID‐19 Trial of Nirmatrelvir , 2022, medRxiv.

[50]  C. Schiffer,et al.  Defining the substrate envelope of SARS-CoV-2 main protease to predict and avoid drug resistance , 2022, Nature Communications.

[51]  C. Schiffer,et al.  Comprehensive fitness landscape of SARS-CoV-2 Mpro reveals insights into viral resistance mechanisms , 2022, bioRxiv.

[52]  B. Weynand,et al.  The oral protease inhibitor (PF-07321332) protects Syrian hamsters against infection with SARS-CoV-2 variants of concern , 2021, Nature Communications.

[53]  Qi Zhang,et al.  Crystal structure of SARS-CoV-2 main protease in complex with protease inhibitor PF-07321332 , 2021, Protein & Cell.

[54]  Malina A. Bakowski,et al.  Preclinical characterization of an intravenous coronavirus 3CL protease inhibitor for the potential treatment of COVID19 , 2021, Nature Communications.

[55]  R. Plemper,et al.  Analysis of SARS-CoV-2 infection dynamic in vivo using reporter-expressing viruses , 2021, Proceedings of the National Academy of Sciences.

[56]  G. Bocharov,et al.  Intracellular Life Cycle Kinetics of SARS-CoV-2 Predicted Using Mathematical Modelling , 2021, Viruses.

[57]  J. Vederas,et al.  Peptidomimetic nitrile warheads as SARS-CoV-2 3CL protease inhibitors. , 2021, RSC medicinal chemistry.

[58]  K. Gajiwala,et al.  An oral SARS-CoV-2 Mpro inhibitor clinical candidate for the treatment of COVID-19 , 2021, Science.

[59]  S. Taliani,et al.  The Alpha Keto Amide Moiety as a Privileged Motif in Medicinal Chemistry: Current Insights and Emerging Opportunities , 2021, Journal of medicinal chemistry.

[60]  Yuquan Wei,et al.  SARS-CoV-2 Mpro inhibitors with antiviral activity in a transgenic mouse model , 2021, Science.

[61]  M. Gilski,et al.  Crystallographic models of SARS-CoV-2 3CLpro: in-depth assessment of structure quality and validation , 2021, IUCrJ.

[62]  Hao Song,et al.  Structural basis for the inhibition of the SARS-CoV-2 main protease by the anti-HCV drug narlaprevir , 2021, Signal Transduction and Targeted Therapy.

[63]  M. Bauer,et al.  Put a ring on it: application of small aliphatic rings in medicinal chemistry. , 2021, RSC medicinal chemistry.

[64]  N. Krogan,et al.  Genetic Screens Identify Host Factors for SARS-CoV-2 and Common Cold Coronaviruses , 2020, Cell.

[65]  M. Bogyo,et al.  Challenges for Targeting SARS-CoV-2 Proteases as a Therapeutic Strategy for COVID-19 , 2020, bioRxiv.

[66]  Vincent A. Voelz,et al.  Open Science Discovery of Potent Non-Covalent SARS-CoV-2 Main Protease Inhibitors , 2020, bioRxiv.

[67]  H. O’Neill,et al.  Malleability of the SARS-CoV-2 3CL Mpro Active-Site Cavity Facilitates Binding of Clinical Antivirals , 2020, Structure.

[68]  R. Hilgenfeld,et al.  SARS-CoV-2 Mpro inhibitors and activity-based probes for patient-sample imaging , 2020, Nature Chemical Biology.

[69]  Lisa E. Gralinski,et al.  A Mouse-Adapted SARS-CoV-2 Induces Acute Lung Injury and Mortality in Standard Laboratory Mice , 2020, Cell.

[70]  Christian Gampe,et al.  Curse or Cure? A Perspective on the Developability of Aldehydes as Active Pharmaceutical Ingredients. , 2020, Journal of medicinal chemistry.

[71]  Huan Sun,et al.  Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease , 2020, Nature Communications.

[72]  Lisa E. Gralinski,et al.  A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures , 2020, Nature.

[73]  Christoph Nitsche,et al.  The SARS-CoV-2 main protease as drug target , 2020, Bioorganic & Medicinal Chemistry Letters.

[74]  John D. Chodera,et al.  SARS-CoV-2 Simulations Go Exascale to Capture Spike Opening and Reveal Cryptic Pockets Across the Proteome , 2020, bioRxiv.

[75]  Ralf Bartenschlager,et al.  SARS-CoV-2 structure and replication characterized by in situ cryo-electron tomography , 2020, Nature Communications.

[76]  M. Diamond,et al.  Growth, detection, quantification, and inactivation of SARS-CoV-2 , 2020, Virology.

[77]  R. D’Aquila,et al.  Broad-spectrum inhibition of coronavirus main and papain-like proteases by HCV drugs , 2020 .

[78]  Luqing Shang,et al.  Comprehensive Insights into the Catalytic Mechanism of Middle East Respiratory Syndrome 3C-Like Protease and Severe Acute Respiratory Syndrome 3C-Like Protease , 2020, ACS catalysis.

[79]  Hualiang Jiang,et al.  Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease , 2020, Science.

[80]  Hualiang Jiang,et al.  Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors , 2020, Nature.

[81]  R. Hilgenfeld,et al.  Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors , 2020, Science.

[82]  R. Hilgenfeld,et al.  α-Ketoamides as Broad-Spectrum Inhibitors of Coronavirus and Enterovirus Replication: Structure-Based Design, Synthesis, and Activity Assessment , 2020, bioRxiv.

[83]  Chunlong Ma,et al.  Boceprevir, GC-376, and calpain inhibitors II, XII inhibit SARS-CoV-2 viral replication by targeting the viral main protease , 2020, Cell Research.

[84]  W. Groutas,et al.  Antiviral Drug Discovery: Norovirus Proteases and Development of Inhibitors , 2019, Viruses.

[85]  J. Wu,et al.  Comparison of antiviral resistance across acute and chronic viral infections , 2018, Antiviral research.

[86]  Michael D Shultz,et al.  Two Decades under the Influence of the Rule of Five and the Changing Properties of Approved Oral Drugs. , 2018, Journal of medicinal chemistry.

[87]  Gwyndaf Evans,et al.  DIALS: implementation and evaluation of a new integration package , 2018, Acta crystallographica. Section D, Structural biology.

[88]  Stacy Ho,et al.  Industry Perspective on Contemporary Protein-Binding Methodologies: Considerations for Regulatory Drug-Drug Interaction and Related Guidelines on Highly Bound Drugs. , 2017, Journal of pharmaceutical sciences.

[89]  D. Greenblatt,et al.  Inhibition of human cytochromes P450 in vitro by ritonavir and cobicistat , 2017, The Journal of pharmacy and pharmacology.

[90]  N. Pedersen,et al.  Efficacy of a 3C-like protease inhibitor in treating various forms of acquired feline infectious peritonitis , 2017, Journal of feline medicine and surgery.

[91]  C. Schiffer,et al.  Improving Viral Protease Inhibitors to Counter Drug Resistance. , 2016, Trends in Microbiology.

[92]  Shery Jacob,et al.  A simple practice guide for dose conversion between animals and human , 2016, Journal of basic and clinical pharmacy.

[93]  N. Pedersen,et al.  Reversal of the Progression of Fatal Coronavirus Infection in Cats by a Broad-Spectrum Coronavirus Protease Inhibitor , 2016, PLoS pathogens.

[94]  Aina E. Cohen,et al.  The Stanford Automated Mounter: pushing the limits of sample exchange at the SSRL macromolecular crystallography beamlines , 2016, Journal of applied crystallography.

[95]  C. Schiffer,et al.  Structural and Thermodynamic Effects of Macrocyclization in HCV NS3/4A Inhibitor MK-5172. , 2016, ACS chemical biology.

[96]  R. Copeland The drug–target residence time model: a 10-year retrospective , 2015, Nature Reviews Drug Discovery.

[97]  Henry B. Wedler,et al.  Predicting hydration propensities of biologically relevant α-ketoamides. , 2015, Bioorganic & medicinal chemistry letters.

[98]  D. Hua,et al.  Broad-Spectrum Inhibitors against 3C-Like Proteases of Feline Coronaviruses and Feline Caliciviruses , 2015, Journal of Virology.

[99]  Yuna Sun,et al.  Peptidyl Aldehyde NK-1.8k Suppresses Enterovirus 71 and Enterovirus 68 Infection by Targeting Protease 3C , 2015, Antimicrobial Agents and Chemotherapy.

[100]  Peng Zhan,et al.  Conformational restriction: an effective tactic in 'follow-on'-based drug discovery. , 2014, Future medicinal chemistry.

[101]  A. Howe,et al.  The Discovery and Development of Boceprevir: A Novel, First-generation Inhibitor of the Hepatitis C Virus NS3/4A Serine Protease , 2013, Journal of clinical and translational hepatology.

[102]  G. Dahl,et al.  Pharmacokinetics and the drug-target residence time concept. , 2013, Drug discovery today.

[103]  Philip R. Evans,et al.  How good are my data and what is the resolution? , 2013, Acta crystallographica. Section D, Biological crystallography.

[104]  M. Perbandt,et al.  3C Protease of Enterovirus 68: Structure-Based Design of Michael Acceptor Inhibitors and Their Broad-Spectrum Antiviral Effects against Picornaviruses , 2013, Journal of Virology.

[105]  K. Battaile,et al.  Broad-Spectrum Antivirals against 3C or 3C-Like Proteases of Picornaviruses, Noroviruses, and Coronaviruses , 2012, Journal of Virology.

[106]  R. Couch,et al.  Immunization with SARS Coronavirus Vaccines Leads to Pulmonary Immunopathology on Challenge with the SARS Virus , 2012, PloS one.

[107]  T. Maurer,et al.  Impact of recovery on fraction unbound using equilibrium dialysis. , 2012, Journal of pharmaceutical sciences.

[108]  H. Chow,et al.  Profiling of Substrate Specificities of 3C-Like Proteases from Group 1, 2a, 2b, and 3 Coronaviruses , 2011, PloS one.

[109]  Stefan Zeuzem,et al.  Boceprevir for previously treated chronic HCV genotype 1 infection. , 2011, The New England journal of medicine.

[110]  M. Golicnik,et al.  Exact and approximate solutions for the decades‐old Michaelis‐Menten equation: Progress‐curve analysis through integrated rate equations , 2011, Biochemistry and molecular biology education : a bimonthly publication of the International Union of Biochemistry and Molecular Biology.

[111]  Yong Zhang,et al.  PKSolver: An add-in program for pharmacokinetic and pharmacodynamic data analysis in Microsoft Excel , 2010, Comput. Methods Programs Biomed..

[112]  G. Chang,et al.  Mutation of Glu-166 Blocks the Substrate-Induced Dimerization of SARS Coronavirus Main Protease , 2010, Biophysical Journal.

[113]  Andrew Hill,et al.  How much ritonavir is needed to boost protease inhibitors? Systematic review of 17 dose-ranging pharmacokinetic trials , 2009, AIDS.

[114]  D. Hazuda,et al.  MK-7009, a Potent and Selective Inhibitor of Hepatitis C Virus NS3/4A Protease , 2009, Antimicrobial Agents and Chemotherapy.

[115]  Kenneth A. Johnson,et al.  Global kinetic explorer: a new computer program for dynamic simulation and fitting of kinetic data. , 2009, Analytical biochemistry.

[116]  Randy J. Read,et al.  Phaser crystallographic software , 2007, Journal of applied crystallography.

[117]  E. Freire,et al.  Long-range cooperative interactions modulate dimerization in SARS 3CLpro. , 2006, Biochemistry.

[118]  Michelle M. Packard,et al.  Severe Acute Respiratory Syndrome Coronavirus Infection of Mice Transgenic for the Human Angiotensin-Converting Enzyme 2 Virus Receptor , 2006, Journal of Virology.

[119]  Yu-An Huang,et al.  Synthesis, crystal structure, structure-activity relationships, and antiviral activity of a potent SARS coronavirus 3CL protease inhibitor. , 2006, Journal of medicinal chemistry.

[120]  M. Hsu,et al.  Mechanism of the Maturation Process of SARS-CoV 3CL Protease , 2005, Journal of Biological Chemistry.

[121]  F. Maldonado,et al.  In Vitro Antiviral Activity and Single-Dose Pharmacokinetics in Humans of a Novel, Orally Bioavailable Inhibitor of Human Rhinovirus 3C Protease , 2005, Antimicrobial Agents and Chemotherapy.

[122]  Robert A Copeland,et al.  Evaluation of enzyme inhibitors in drug discovery. A guide for medicinal chemists and pharmacologists. , 2005, Methods of biochemical analysis.

[123]  D. Sterner,et al.  Expression and purification of SARS coronavirus proteins using SUMO-fusions , 2005, Protein Expression and Purification.

[124]  James P. Cain,et al.  Exploring Ramachandran and chi space: conformationally constrained amino acids and peptides in the design of bioactive polypeptide ligands. , 2004, Current medicinal chemistry.

[125]  B. G. Rao,et al.  Inhibitors of hepatitis C virus NS3.4A protease 2. Warhead SAR and optimization. , 2004, Bioorganic & medicinal chemistry letters.

[126]  T. C. Bruice,et al.  α-Ketoamides and α-Ketocarbonyls: Conformational analysis and development of all-atom OPLS force field , 2000 .

[127]  D. Matthews,et al.  Structure-assisted design of mechanism-based irreversible inhibitors of human rhinovirus 3C protease with potent antiviral activity against multiple rhinovirus serotypes. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[128]  D. Matthews,et al.  Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 4. Incorporation of P1 lactam moieties as L-glutamine replacements. , 1999, Journal of medicinal chemistry.

[129]  G. Murshudov,et al.  Refinement of macromolecular structures by the maximum-likelihood method. , 1997, Acta crystallographica. Section D, Biological crystallography.

[130]  D. Veber,et al.  Inhibition of Thrombin by Peptides Containing Lysyl-α-Keto Carbonyl Derivatives , 1995, Thrombosis and Haemostasis.

[131]  D. Veber,et al.  Amide and α-keto carbonyl inhibitors of thrombin based on arginine and lysine: Synthesis, stability and biological characterization , 1995 .

[132]  Collaborative Computational,et al.  The CCP4 suite: programs for protein crystallography. , 1994, Acta crystallographica. Section D, Biological crystallography.

[133]  H. Wasserman,et al.  (Cyanomethylene)phosphoranes as Novel Carbonyl 1,1-Dipole Synthons: An Efficient Synthesis of .alpha.-Keto Acids, Esters, and Amides , 1994 .

[134]  Y. Cheng,et al.  Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. , 1973, Biochemical pharmacology.

[135]  Yuquan Wei,et al.  SARS-CoV-2 M pro inhibitors with antiviral activity in a transgenic mouse model , 2021 .

[136]  Noel S. Wilson,et al.  Covalent binders in drug discovery. , 2019, Progress in medicinal chemistry.

[137]  M. Manns,et al.  Boceprevir for untreated chronic HCV genotype 1 infection. , 2011, The New England journal of medicine.

[138]  P. Evans,et al.  Scaling and assessment of data quality. , 2006, Acta crystallographica. Section D, Biological crystallography.

[139]  A. W. E. E. K. L. Y. J. O U R N A L D E V O T E D T O T H E A D V A N C E,et al.  S C I E N C E , 2022 .