Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
J Am Chem Soc ; 144(7): 2905-2920, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35142215

RESUMO

Drugs targeting SARS-CoV-2 could have saved millions of lives during the COVID-19 pandemic, and it is now crucial to develop inhibitors of coronavirus replication in preparation for future outbreaks. We explored two virtual screening strategies to find inhibitors of the SARS-CoV-2 main protease in ultralarge chemical libraries. First, structure-based docking was used to screen a diverse library of 235 million virtual compounds against the active site. One hundred top-ranked compounds were tested in binding and enzymatic assays. Second, a fragment discovered by crystallographic screening was optimized guided by docking of millions of elaborated molecules and experimental testing of 93 compounds. Three inhibitors were identified in the first library screen, and five of the selected fragment elaborations showed inhibitory effects. Crystal structures of target-inhibitor complexes confirmed docking predictions and guided hit-to-lead optimization, resulting in a noncovalent main protease inhibitor with nanomolar affinity, a promising in vitro pharmacokinetic profile, and broad-spectrum antiviral effect in infected cells.


Assuntos
Antivirais/farmacologia , Proteases 3C de Coronavírus/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , SARS-CoV-2/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antivirais/metabolismo , Antivirais/farmacocinética , Domínio Catalítico , Chlorocebus aethiops , Proteases 3C de Coronavírus/química , Inibidores de Cisteína Proteinase/metabolismo , Inibidores de Cisteína Proteinase/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Humanos , Testes de Sensibilidade Microbiana , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Ligação Proteica , SARS-CoV-2/enzimologia , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacocinética , Células Vero
2.
Biochem Biophys Res Commun ; 555: 134-139, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33813272

RESUMO

There is an urgent need for antivirals targeting the SARS-CoV-2 virus to fight the current COVID-19 pandemic. The SARS-CoV-2 main protease (3CLpro) represents a promising target for antiviral therapy. The lack of selectivity for some of the reported 3CLpro inhibitors, specifically versus cathepsin L, raises potential safety and efficacy concerns. ALG-097111 potently inhibited SARS-CoV-2 3CLpro (IC50 = 7 nM) without affecting the activity of human cathepsin L (IC50 > 10 µM). When ALG-097111 was dosed in hamsters challenged with SARS-CoV-2, a robust and significant 3.5 log10 (RNA copies/mg) reduction of the viral RNA copies and 3.7 log10 (TCID50/mg) reduction in the infectious virus titers in the lungs was observed. These results provide the first in vivo validation for the SARS-CoV-2 3CLpro as a promising therapeutic target for selective small molecule inhibitors.


Assuntos
Amidas/farmacologia , Tratamento Farmacológico da COVID-19 , Proteases 3C de Coronavírus/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Modelos Animais de Doenças , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/enzimologia , Amidas/farmacocinética , Animais , COVID-19/virologia , Catepsina L/antagonistas & inibidores , Linhagem Celular , Cricetinae , Inibidores de Cisteína Proteinase/farmacocinética , Feminino , Humanos , Concentração Inibidora 50 , Masculino , Mesocricetus/virologia , Reprodutibilidade dos Testes , SARS-CoV-2/crescimento & desenvolvimento , Serina Endopeptidases , Especificidade por Substrato , Replicação Viral/efeitos dos fármacos
3.
Bioorg Chem ; 106: 104497, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33261847

RESUMO

The virus SARS CoV-2, which causes the respiratory infection COVID-19, continues its spread across the world and to date has caused more than a million deaths. Although COVID-19 vaccine development appears to be progressing rapidly, scientists continue the search for different therapeutic options to treat this new illness. In this work, we synthesized five new 1-aryl-5-(3-azidopropyl)indol-4-ones and showed them to be potential inhibitors of the SARS CoV-2 main protease (3CLpro). The compounds were obtained in good overall yields and molecular docking indicated favorable binding with 3CLpro. In silico ADME/Tox profile of the new compounds were calculated using the SwissADME and pkCSM-pharmacokinetics web tools, and indicated adequate values of absorption, distribution and excretion, features related to bioavailability. Moreover, low values of toxicity were indicated for these compounds. And drug-likeness levels of the compounds were also predicted according to the Lipinski and Veber rules.


Assuntos
Antivirais/metabolismo , Azidas/metabolismo , Proteases 3C de Coronavírus/antagonistas & inibidores , Inibidores de Cisteína Proteinase/metabolismo , Indóis/metabolismo , SARS-CoV-2/química , Antivirais/síntese química , Antivirais/farmacocinética , Azidas/síntese química , Azidas/farmacocinética , Domínio Catalítico , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/metabolismo , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/farmacocinética , Indóis/síntese química , Indóis/farmacocinética , Internet , Simulação de Acoplamento Molecular , Ligação Proteica
4.
Bioorg Med Chem Lett ; 30(17): 127377, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32738988

RESUMO

The unprecedented pandemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is threatening global health. The virus emerged in late 2019 and can cause a severe disease associated with significant mortality. Several vaccine development and drug discovery campaigns are underway. The SARS-CoV-2 main protease is considered a promising drug target, as it is dissimilar to human proteases. Sequence and structure of the main protease are closely related to those from other betacoronaviruses, facilitating drug discovery attempts based on previous lead compounds. Covalently binding peptidomimetics and small molecules are investigated. Various compounds show antiviral activity in infected human cells.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antivirais/farmacocinética , Betacoronavirus/enzimologia , Linhagem Celular , Proteases 3C de Coronavírus , Cisteína Endopeptidases/química , Inibidores de Cisteína Proteinase/farmacocinética , Descoberta de Drogas , Humanos , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , SARS-CoV-2 , Especificidade por Substrato , Proteínas não Estruturais Virais/química
5.
Br J Clin Pharmacol ; 85(6): 1072-1083, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30663085

RESUMO

Cathepsin K (CatK) is a cysteine protease abundantly expressed by osteoclasts and localized in the lysosomes and resorption lacunae of these cells. CatK is the principal enzyme responsible for the degradation of bone collagen. Odanacatib is a selective, reversible inhibitor of CatK at subnanomolar potency. The pharmacokinetics of odanacatib have been extensively studied and are similar in young healthy men, postmenopausal women and elderly men, and were qualitatively similar throughout Phase 1 development and in-patient studies. Following 3 weeks of 50 mg once weekly dosing the geometric mean area under the curve from 0 to 168 hours was 41.1 µM h, the concentration at 168 hours was 126 nM and the harmonic mean apparent terminal half-life was 84.8 hr. Odanacatib exposure increased in a less than dose proportional manner due to solubility limited absorption. It is estimated that approximately 70% of the absorbed dose of odanacatib is eliminated via metabolism, 20% is excreted as unchanged drug in the bile or faeces, and 10% is excreted as unchanged drug in the urine. The systemic clearance was low (approximately 13 mL/min). Odanacatib decreases the degradation of bone matrix proteins and reduces the efficiency of bone resorption with target engagement confirmed by a robust decrease in serum C-telopeptides of type 1 collagen (approximately 60%), urinary aminoterminal crosslinked telopeptides of type 1 collagen to creatinine ratio (approximately 50%) and total urine deoxypyridinoline/Cr (approximately 30%), with an increase in serum cross-linked carboxy-terminal telopeptide of type 1 collagen (approximately 55%). The 50-mg weekly dosing regimen evaluated in Phase 3 achieved near maximal reduction in bone resorption throughout the treatment period. The extensive clinical programme for odanacatib, together with more limited clinical experience with other CatK inhibitors (balicatib and ONO-5334), provides important insights into the clinical pharmacology of CatK inhibition and the potential role of CatK in bone turnover and mineral homeostasis. Key findings include the ability of this mechanism to: (i) provide sustained reductions in resorption markers, increases in bone mineral density, and demonstrated fracture risk reduction; (ii) be associated with relative formation-sparing effects such that sustained resorption reduction is achieved without accompanying meaningful reductions in bone formation; and (iii) lead to increases in osteoclast number as well as other osteoclast activity (including build-up of CatK enzyme), which may yield transient increases in resorption following treatment discontinuation and the potential for nonmonotonic responses at subtherapeutic doses.


Assuntos
Compostos de Bifenilo/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Remodelação Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/uso terapêutico , Osteoporose/tratamento farmacológico , Animais , Compostos de Bifenilo/efeitos adversos , Compostos de Bifenilo/farmacocinética , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacocinética , Osso e Ossos/enzimologia , Osso e Ossos/fisiopatologia , Catepsina K/metabolismo , Inibidores de Cisteína Proteinase/efeitos adversos , Inibidores de Cisteína Proteinase/farmacocinética , Feminino , Humanos , Masculino , Osteoporose/enzimologia , Osteoporose/patologia , Transdução de Sinais , Pesquisa Translacional Biomédica , Resultado do Tratamento
6.
J Enzyme Inhib Med Chem ; 34(1): 547-561, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30696325

RESUMO

We report computer-aided design of new lactone-chalcone and isatin-chalcone (HLCIC) inhibitors of the falcipain-2 (PfFP-2). 3D models of 15 FP-2:HLCIC1-15 complexes with known observed activity (IC50exp) were prepared to establish a quantitative structure-activity (QSAR) model and linear correlation between relative Gibbs free energy of enzyme:inhibitor complex formation (ΔΔGcom) and IC50exp: pIC50exp = -0.0236 × ΔΔGcom+5.082(#); R2 = 0.93. A 3D pharmacophore model (PH4) derived from the QSAR directed our effort to design novel HLCIC analogues. During the design, an initial virtual library of 2621440 HLCIC was focused down to 18288 drug-like compounds and finally, PH4 screened to identify 81 promising compounds. Thirty-three others were added from an intuitive substitution approach intended to fill better the enzyme S2 pocket. One hundred and fourteen theoretical IC50 (IC50pre) values were predicted by means of (#) and their pharmacokinetics (ADME) profiles. More than 30 putative HLCICs display IC50pre 100 times superior to that of the published most active training set inhibitor HLCIC1.


Assuntos
Chalconas/química , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacologia , Desenho de Fármacos , Isatina/química , Lactonas/química , Plasmodium falciparum/enzimologia , Domínio Catalítico , Chalconas/farmacologia , Desenho Assistido por Computador , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacocinética , Concentração Inibidora 50 , Isatina/farmacologia , Lactonas/farmacologia , Modelos Moleculares , Sondas Moleculares , Relação Quantitativa Estrutura-Atividade , Termodinâmica
7.
J Transl Med ; 16(1): 125, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743078

RESUMO

BACKGROUND: Cathepsin K is an attractive therapeutic target for diseases in which bone resorption is excessive such as osteoporosis and osteoarthritis (OA). The current paper characterized the pharmacological profile of the potent and selective cathepsin K inhibitor, MIV-711, in vitro and in cynomolgus monkeys, and assessed translation to human based on a single dose clinical study in man. METHODS: The potency and selectivity of MIV-711 were assessed in vitro using recombinant enzyme assays and differentiated human osteoclasts. MIV-711 was administered to healthy cynomolgus monkeys (3-30 µmol/kg, p.o.). Plasma levels of MIV-711 and the bone resorption biomarker CTX-I were measured after single dose experiments, and urine levels of CTX-I, NTX-I and CTX-II biomarkers were measured after repeat dose experiments. The safety, pharmacokinetics and pharmacodynamics (serum CTX-I) of MIV-711 were assessed in human healthy subjects after single ascending doses from 20 to 600 mg. RESULTS: MIV-711 was a potent inhibitor of human cathepsin K (Ki: 0.98 nmol/L) with > 1300-fold selectivity towards other human cathepsins. MIV-711 inhibited human osteoclast-mediated bone resorption with an IC50 value of 43 nmol/L. Single oral doses of MIV-711 to monkeys reduced plasma levels of CTX-I in a dose-dependent fashion by up to 57% at trough. The effect on CTX-I was linearly correlated to the plasma exposure of MIV-711, while the efficacy duration outlasted plasma exposure. Repeat oral dosing with MIV-711 also reduced urinary levels of the bone resorption biomarkers CTX-I (by 93%) and NTX-I (by 71%) and the cartilage degradation biomarker CTX-II (by 71%). MIV-711 was safe and well-tolerated when given as single ascending doses to healthy subjects. MIV-711 reduced serum CTX-I levels in a dose-dependent manner by up to 79% at trough. The relationship between MIV-711 exposure and effects on these biomarkers in humans was virtually identical when compared to the corresponding monkey data. CONCLUSIONS: MIV-711 is a potent and selective cathepsin K inhibitor with dose-dependent effects on biomarkers of bone and cartilage degradation in monkey and human. Taken together, MIV-711 shows promise for the treatment of bone and cartilage related disorders in humans, such as OA. Trial Registration EudraCT number 2011-003024-12, registered on June 22nd 2011.


Assuntos
Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Compostos Orgânicos/farmacologia , Administração Oral , Adulto , Animais , Biomarcadores/urina , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Inibidores de Cisteína Proteinase/administração & dosagem , Inibidores de Cisteína Proteinase/sangue , Inibidores de Cisteína Proteinase/farmacocinética , Feminino , Humanos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Compostos Orgânicos/administração & dosagem , Compostos Orgânicos/sangue , Compostos Orgânicos/farmacocinética , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Adulto Jovem
8.
J Transl Med ; 16(1): 56, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29523155

RESUMO

BACKGROUND: MIV-711 is a highly potent and selective cathepsin K inhibitor. The current article summarizes the therapeutic effects of MIV-711 on joint pathology in rabbits subjected to anterior cruciate ligament transection (ACLT), and the prophylactic effects on joint pathology in dogs subjected to partial medial meniscectomy, two surgical models of osteoarthritis (OA). METHODS: Starting 1 week after surgery, rabbits were dosed daily via oral gavage with either MIV-711 or vehicle (n = 7/group) for 7 weeks. The four treatment groups were: (1) sham + vehicle; (2) ACLT + vehicle; (3) ACLT + MIV-711, 30 µmol/kg and (4) ACLT + MIV-711, 100 µmol/kg. Subchondral bone and articular cartilage structures were assessed by µCT, histomorphometry, and scoring. Dogs subjected to partial medial meniscectomy received either MIV-711 (30 µmol/kg) or vehicle (n = 15/group) via oral gavage once daily, starting 1 day before meniscectomy, for 28 days. Cartilage degradation was assessed at the macroscopic and microscopic levels. The exposures of MIV-711 were assessed in both studies and biomarkers reflecting bone resorption (HP-1 in rabbits, CTX-I in dogs) and cartilage degradation (CTX-II) were measured. RESULTS: In ACLT rabbits, MIV-711 decreased HP-1 levels by up to 72% (p < 0.001) and CTX-II levels by up to 74% (p < 0.001) compared to ACLT vehicle controls. ACLT surgery significantly reduced the total thickness of the subchondral bone plate and reduced trabecular bone volume in the femur and tibia. These effects were reversed by MIV-711. ACLT resulted in cartilage thickening, which was attenuated by MIV-711. MIV-711 did not affect osteophyte formation or Mankin scores. In dogs, MIV-711 reduced CTX-I and CTX-II levels by 86% (p < 0.001) and 80% (p < 0.001), respectively. Synovial CTX-II levels were reduced by 55-57% (p < 0.001) compared to baseline. MIV-711-treated animals had 25-37% lower macroscopic scores in the femur condyles and 13-33% lower macroscopic scores in the tibial plateaus. CONCLUSIONS: MIV-711 prevents subchondral bone loss and partially attenuates cartilage pathology in two animal models of OA. These beneficial effects of MIV-711 on joint pathology are observed in conjunction with decreases in bone and cartilage biomarkers that have been shown to be clinically attainable in human. The data support the further development of MIV-711 for the treatment of OA.


Assuntos
Lesões do Ligamento Cruzado Anterior/tratamento farmacológico , Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/uso terapêutico , Articulações/patologia , Osteoartrite/tratamento farmacológico , Animais , Ligamento Cruzado Anterior , Biomarcadores/sangue , Biomarcadores/urina , Reabsorção Óssea/patologia , Cartilagem Articular/diagnóstico por imagem , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Inibidores de Cisteína Proteinase/sangue , Inibidores de Cisteína Proteinase/farmacocinética , Inibidores de Cisteína Proteinase/farmacologia , Modelos Animais de Doenças , Cães , Feminino , Articulações/diagnóstico por imagem , Articulações/efeitos dos fármacos , Masculino , Compostos Orgânicos , Osteoartrite/sangue , Osteoartrite/diagnóstico por imagem , Osteoartrite/patologia , Análise de Componente Principal , Coelhos
9.
Xenobiotica ; 48(12): 1258-1267, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29160126

RESUMO

1. LASSBio-1736 ((E)-1-4(trifluoromethyl) benzylidene)-5-(2-4-dichlorozoyl) carbonylhydrazine) is proposed to be an oral cysteine protease leishmanicidal inhibitor. 2. This work aimed to investigate plasma pharmacokinetics, protein binding and tissue distribution of LASSBio-1736 in male Wistar rats. 3. LASSBio-1736 was administered to male Wistar rats at doses of 3.2 mg/kg intravenously and 12.6 mg/kg oral and intraperitoneal. The individual plasma-concentration profiles were determined by HPLC-UV and evaluated by non-compartmental and population pharmacokinetic analysis (Monolix 2016R1, Lixoft). Tissue distribution was evaluated after iv injection of 3.2 mg/kg drug by non-compartmental approach. 4. After intravenous administration, Vdss (1.79 L/kg), t ½ (23.1 h) and CLtot (56.1 mL/h/kg) were determined, and they were statistically similar (α =0.05) to oral and intraperitoneal pharmacokinetic parameters. The plasma profiles obtained after intravenous, oral and intraperitoneal administration of the compound were best fitted to a three-compartment and one-compartment open model with first-order absorption. 5. The intraperitoneal and oral bioavailability were around 40 and 15%, respectively. 6. Liver, spleen and skin tissues showed penetration of 340, 130 and 40%, respectively, with t ½ like plasma values. 7. LASSBio-1736 protein binding was 95 ± 2%. 8. The t ½, CLtot and tissue distribution of the compound agreed with the desired drug characteristics for leishmanicidal activity.


Assuntos
Antiprotozoários/farmacologia , Antiprotozoários/farmacocinética , Inibidores de Cisteína Proteinase/farmacologia , Inibidores de Cisteína Proteinase/farmacocinética , Animais , Leishmaniose/sangue , Leishmaniose/tratamento farmacológico , Masculino , Ratos , Ratos Wistar
10.
Am J Physiol Heart Circ Physiol ; 302(3): H582-93, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22081706

RESUMO

Our laboratory has shown that λ-carrageenan-induced peripheral inflammatory pain (CIP) can alter tight junction (TJ) protein expression and/or assembly leading to changes in blood-brain barrier xenobiotic permeability. However, the role of reactive oxygen species (ROS) and subsequent oxidative stress during CIP is unknown. ROS (i.e., superoxide) are known to cause cellular damage in response to pain/inflammation. Therefore, we examined oxidative stress-associated effects at the blood-brain barrier (BBB) in CIP rats. During CIP, increased staining of nitrosylated proteins was detected in hind paw tissue and enhanced presence of protein adducts containing 3-nitrotyrosine occurred at two molecular weights (i.e., 85 and 44 kDa) in brain microvessels. Tempol, a pharmacological ROS scavenger, attenuated formation of 3-nitrotyrosine-containing proteins in both the hind paw and in brain microvessels when administered 10 min before footpad injection of λ-carrageenan. Similarly, CIP increased 4-hydroxynoneal staining in brain microvessels and this effect was reduced by tempol. Brain permeability to [(14)C]sucrose and [(3)H]codeine was increased, and oligomeric assemblies of occludin, a critical TJ protein, were altered after 3 h CIP. Tempol attenuated both [(14)C]sucrose and [(3)H]codeine brain uptake as well as protected occludin oligomers from disruption in CIP animals, suggesting that ROS production/oxidative stress is involved in modulating BBB functional integrity during pain/inflammation. Interestingly, tempol administration reduced codeine analgesia in CIP animals, indicating that oxidative stress during pain/inflammation may affect opioid delivery to the brain and subsequent efficacy. Taken together, our data show for the first time that ROS pharmacological scavenging is a viable approach for maintaining BBB integrity and controlling central nervous system drug delivery during acute inflammatory pain.


Assuntos
Barreira Hematoencefálica , Permeabilidade Capilar/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Proteínas de Membrana/metabolismo , Neuralgia , Xenobióticos/farmacocinética , Doença Aguda , Aldeídos/farmacocinética , Analgésicos Opioides/farmacocinética , Animais , Antioxidantes/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar/imunologia , Radioisótopos de Carbono , Codeína/farmacocinética , Inibidores de Cisteína Proteinase/farmacocinética , Hiperalgesia/tratamento farmacológico , Hiperalgesia/imunologia , Hiperalgesia/metabolismo , Masculino , Proteínas de Membrana/imunologia , Neuralgia/tratamento farmacológico , Neuralgia/imunologia , Neuralgia/metabolismo , Neurite (Inflamação)/tratamento farmacológico , Neurite (Inflamação)/imunologia , Neurite (Inflamação)/metabolismo , Ocludina , Estresse Oxidativo/imunologia , Ratos , Ratos Sprague-Dawley , Marcadores de Spin , Sacarose/farmacocinética , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/imunologia , Junções Íntimas/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
11.
Bioengineered ; 13(2): 3350-3361, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35048792

RESUMO

The COVID-19 new variants spread rapidly all over the world, and until now scientists strive to find virus-specific antivirals for its treatment. The main protease of SARS-CoV-2 (Mpro) exhibits high structural and sequence homology to main protease of SARS-CoV (93.23% sequence identity), and their sequence alignment indicated 12 mutated/variant residues. The sequence alignment of SARS-CoV-2 main protease led to identification of only one mutated/variant residue with no significant role in its enzymatic process. Therefore, Mpro was considered as a high-profile drug target in anti-SARS-CoV-2 drug discovery. Apigenin analogues to COVID-19 main protease binding were evaluated. The detailed interactions between the analogues of Apigenin and SARS-CoV-2 Mpro inhibitors were determined as hydrogen bonds, electronic bonds and hydrophobic interactions. The binding energies obtained from the molecular docking of Mpro with Boceprevir, Apigenin, Apigenin 7-glucoside-4'-p-coumarate, Apigenin 7-glucoside-4'-trans-caffeate and Apigenin 7-O-beta-d-glucoside (Cosmosiin) were found to be -6.6, -7.2, -8.8, -8.7 and -8.0 kcal/mol, respectively. Pharmacokinetic parameters and toxicological characteristics obtained by computational techniques and Virtual ADME studies of the Apigenin analogues confirmed that the Apigenin 7-glucoside-4'-p-coumarate is the best candidate for SARS-CoV-2 Mpro inhibition.


Assuntos
Antivirais/farmacologia , Apigenina/farmacologia , Tratamento Farmacológico da COVID-19 , Proteases 3C de Coronavírus/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/enzimologia , Sequência de Aminoácidos , Antivirais/química , Antivirais/farmacocinética , Apigenina/química , Apigenina/farmacocinética , Bioengenharia , COVID-19/virologia , Simulação por Computador , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/genética , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Glucosídeos/química , Glucosídeos/farmacocinética , Glucosídeos/farmacologia , Humanos , Simulação de Acoplamento Molecular , Fitoterapia , Domínios Proteicos , SARS-CoV-2/genética
12.
Chem Biol Interact ; 335: 109348, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33278462

RESUMO

The disease, COVID-19, is caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2) for which there is currently no treatment. The SARS-CoV-2 main protease (Mpro) is an important enzyme for viral replication. Small molecules that inhibit this protease could lead to an effective COVID-19 treatment. The 2-pyridone scaffold was previously identified as a possible key pharmacophore to inhibit SARS-CoV-2 Mpro. A search for natural, antimicrobial products with the 2-pyridone moiety was undertaken herein, and their calculated potency as inhibitors of SARS-CoV-2 Mpro was investigated. Thirty-three natural products containing the 2-pyridone scaffold were identified from the literature. An in silico methodology using AutoDock was employed to predict the binding energies and inhibition constants (Ki values) for each 2-pyridone-containing compound with SARS-CoV-2 Mpro. This consisted of molecular optimization of the 2-pyridone compound, docking of the compound with a crystal structure of SARS-CoV-2 Mpro, and evaluation of the predicted interactions and ligand-enzyme conformations. All compounds investigated bound to the active site of SARS-CoV-2 Mpro, close to the catalytic dyad (His-41 and Cys-145). Thirteen molecules had predicted Ki values <1 µM. Glu-166 formed a key hydrogen bond in the majority of the predicted complexes, while Met-165 had some involvement in the complex binding as a close contact to the ligand. Prominent 2-pyridone compounds were further evaluated for their ADMET properties. This work has identified 2-pyridone natural products with calculated potent inhibitory activity against SARS-CoV-2 Mpro and with desirable drug-like properties, which may lead to the rapid discovery of a treatment for COVID-19.


Assuntos
Antivirais/metabolismo , Produtos Biológicos/metabolismo , Proteases 3C de Coronavírus/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Piridonas/metabolismo , SARS-CoV-2/enzimologia , Antivirais/química , Antivirais/farmacocinética , Produtos Biológicos/química , Produtos Biológicos/farmacocinética , Células CACO-2 , Domínio Catalítico , Proteases 3C de Coronavírus/química , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacocinética , Humanos , Ligação de Hidrogênio , Simulação de Acoplamento Molecular , Estrutura Molecular , Ligação Proteica , Piridonas/química , Piridonas/farmacocinética
13.
Bioorg Med Chem Lett ; 20(17): 5184-90, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20656488

RESUMO

Succinic acid amides have been found to be effective P2-P3 scaffold replacements for peptidic ICE inhibitors. Heteroarylalkyl fragments occupying the P4 position provided access to compounds with nM affinities. Utilization of an acylal prodrug moiety was required to overcome biopharmaceutical issues which led to the identification of 17f, a potential clinical candidate.


Assuntos
Amidas/química , Inibidores de Caspase , Inibidores de Cisteína Proteinase/farmacologia , Ácido Succínico/química , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacocinética , Meia-Vida , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Estereoisomerismo , Relação Estrutura-Atividade
14.
Bioorg Med Chem Lett ; 20(5): 1524-7, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20149657

RESUMO

Morphing structural features of HTS-derived chemotypes led to the discovery of novel 2-cyano-pyrimidine inhibitors of cathepsin K with good pharmacokinetic profiles, for example, compound 20 showed high catK potency (IC(50)=4nM), >580-fold selectivity over catL and catB, and oral bioavailability in the rat of 52%.


Assuntos
Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/química , Pirimidinas/química , Administração Oral , Animais , Sítios de Ligação , Catepsina K/metabolismo , Linhagem Celular , Cristalografia por Raios X , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/farmacocinética , Desenho de Fármacos , Ensaios de Triagem em Larga Escala , Humanos , Pirimidinas/síntese química , Pirimidinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
15.
Bioorg Med Chem Lett ; 20(3): 887-92, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20061146

RESUMO

MK-0674 is a potent and selective cathepsin K inhibitor from the same structural class as odanacatib with a comparable inhibitory potency profile against Cat K. It is orally bioavailable and exhibits long half-life in pre-clinical species. In vivo studies using deuterated MK-0674 show stereoselective epimerization of the alcohol stereocenter via an oxidation/reduction cycle. From in vitro incubations, two metabolites could be identified: the hydroxyleucine and the glucuronide conjugate which were confirmed using authentic synthetic standards.


Assuntos
Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/farmacocinética , Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/administração & dosagem , Inibidores de Cisteína Proteinase/farmacocinética , Descoberta de Drogas/métodos , Administração Oral , Animais , Disponibilidade Biológica , Compostos de Bifenilo/química , Catepsina K/metabolismo , Inibidores de Cisteína Proteinase/química , Cães , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Macaca mulatta , Coelhos , Ratos
16.
Biochem Pharmacol ; 174: 113808, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31930961

RESUMO

Separase, a sister chromatid cohesion-resolving enzyme, is an oncogene and overexpressed in many human cancers. Sepin-1 (2,2-dimethyl-5-nitro-2H-benzimidazole-1,3-dioxide) is a potent separase inhibitor that impedes cancer cell growth, cell migration, and wound healing, suggesting that Sepin-1 possesses a great potential to target separase-overexpressing tumors. As a part of the IND-enabling studies to bring Sepin-1 to clinic, herein we report the results from a 28-day repeat-dose pharmacokinetic study of Sepin-1 in rats. Sepin-1 was intravenously administered to Sprague-Dawley rats once daily for 28 days at three different (5, 10, and 20 mg/kg) doses. Blood samples were collected after administration of doses on days 1 and 28. Sepin-1 is unstable and isomerizes in basic solutions, but it is stable in acidic buffer such as citrate-buffered saline (pH 4.0). UHPLC-MS analysis indicated Sepin-1 was rapidly metabolized in vivo. One of the major metabolites was an amine adduct of 2,2-dimethyl-5-nitro-2H-benzimidazole (named Sepin-1.55). The concentration of Sepin-1.55 in blood samples was Sepin-1 dose-dependent and used for pharmacokinetic analysis of Sepin-1. Tmax was approximately 5-15 min. The data suggest that no Sepin-1 accumulation occurred from daily repeat dosing and similar exposures on the first and final day of dosing. Data also suggest a gender difference, namely that female rats have more exposure and slower clearance than male rats. The data support that Sepin-1 is a potential drug candidate that can be further developed to treat Separase-overexpressing human tumors.


Assuntos
Benzimidazóis , Inibidores de Cisteína Proteinase , Separase/antagonistas & inibidores , Animais , Benzimidazóis/química , Benzimidazóis/farmacocinética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacocinética , Estabilidade de Medicamentos , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Separase/sangue
17.
Bioanalysis ; 11(16): 1469-1481, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31512492

RESUMO

Aim: An innovative Atg4B inhibitor, S130, exhibited a negative influence on colorectal cancer cells in vitro and in vivo. To assist reliable toxicodynamic and pharmacokinetic evaluation, an LC-MS/MS assay of S130 in rat plasma must be necessary. Results: An LC-MS/MS assay for determination of S130 in rat plasma has been first developed and fully verified whose values met the admissible limits as per the US FDA guidelines. Chromatographic separation was achieved by using an isocratic elution after 3 min. MS was conducted under the ESI+ mode fitted with selected reaction monitoring. The calibration curve proved acceptable linearity over 0.50-800 ng/ml. Conclusion: The developed LC-MS/MS assay of S130 in rat plasma is easily applicable in pharmacokinetics study and the further toxicological evaluation.


Assuntos
Cromatografia Líquida/métodos , Neoplasias Colorretais/enzimologia , Inibidores de Cisteína Proteinase/sangue , Espectrometria de Massas em Tandem/métodos , Animais , Cromatografia Líquida/normas , Inibidores de Cisteína Proteinase/farmacocinética , Feminino , Humanos , Camundongos , Ratos , Padrões de Referência , Espectrometria de Massas em Tandem/normas
18.
Antimicrob Agents Chemother ; 52(7): 2346-54, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18443109

RESUMO

Cysteine protease inhibitors kill malaria parasites and are being pursued for development as antimalarial agents. Because they have multiple targets within bloodstream-stage parasites, workers have assumed that resistance to these inhibitors would not be acquired easily. In the present study, we used in vitro selection to generate a parasite resistant to growth inhibition by leupeptin, a broad-profile cysteine and serine protease inhibitor. Resistance was not associated with upregulation of cysteine protease activity, reduced leupeptin sensitivity of this activity, or expression level changes for putative cysteine or serine proteases in the parasite genome. Instead, it was associated with marked changes in the plasmodial surface anion channel (PSAC), an ion channel on infected erythrocytes that functions in nutrient and bulky organic solute uptake. Osmotic fragility measurements, electrophysiological recordings, and leupeptin uptake studies revealed selective reductions in organic solute permeability via PSAC, altered single-channel gating, and reduced inhibitor affinity. These changes yielded significantly reduced leupeptin uptake and could fully account for the acquired resistance. PSAC represents a novel route for the uptake of bulky hydrophilic compounds acting against intraerythrocytic parasite targets. Drug development based on such compounds should proceed cautiously in light of possible resistance development though the selection of PSAC mutants.


Assuntos
Resistência a Medicamentos/fisiologia , Eritrócitos/parasitologia , Canais Iônicos/metabolismo , Leupeptinas/farmacocinética , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Antimaláricos/farmacocinética , Transporte Biológico Ativo , Permeabilidade da Membrana Celular , Inibidores de Cisteína Proteinase/farmacocinética , Genes de Protozoários , Humanos , Técnicas In Vitro , Canais Iônicos/genética , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/genética , Proteínas de Protozoários/genética
19.
Chem Biol ; 14(5): 499-511, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17524981

RESUMO

The papain-family cathepsins are cysteine proteases that are emerging as promising therapeutic targets for a number of human disease conditions ranging from osteoporosis to cancer. Relatively few selective inhibitors for this family exist, and the in vivo selectivity of most existing compounds is unclear. We present here the synthesis of focused libraries of epoxysuccinyl-based inhibitors and their screening in crude tissue extracts. We identified a number of potent inhibitors that display selectivity for endogenous cathepsin targets both in vitro and in vivo. Importantly, the selectivity patterns observed in crude extracts were generally retained in vivo, as assessed by active-site labeling of tissues from treated animals. Overall, this study identifies several important compound classes and highlights the use of activity-based probes to assess pharmacodynamic properties of small-molecule inhibitors in vivo.


Assuntos
Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/farmacologia , Compostos de Epóxi/síntese química , Compostos de Epóxi/farmacologia , Papaína/antagonistas & inibidores , Animais , Cromatografia Líquida de Alta Pressão , Inibidores de Cisteína Proteinase/farmacocinética , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Compostos de Epóxi/farmacocinética , Indicadores e Reagentes , Injeções Intraperitoneais , Intubação Gastrointestinal , Masculino , Espectrometria de Massas , Camundongos , Biblioteca de Peptídeos , Proteômica , Ratos
20.
Bioorg Med Chem ; 16(2): 838-53, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17996450

RESUMO

Hologram quantitative structure-activity relationships (HQSAR) were applied to a data set of 41 cruzain inhibitors. The best HQSAR model (Q(2)=0.77; R(2)=0.90) employing Surflex-Sim, as training and test sets generator, was obtained using atoms, bonds, and connections as fragment distinctions and 4-7 as fragment size. This model was then used to predict the potencies of 12 test set compounds, giving satisfactory predictive R(2) value of 0.88. The contribution maps obtained from the best HQSAR model are in agreement with the biological activities of the study compounds. The Trypanosoma cruzi cruzain shares high similarity with the mammalian homolog cathepsin L. The selectivity toward cruzain was checked by a database of 123 compounds, which corresponds to the 41 cruzain inhibitors used in the HQSAR model development plus 82 cathepsin L inhibitors. We screened these compounds by ROCS (Rapid Overlay of Chemical Structures), a Gaussian-shape volume overlap filter that can rapidly identify shapes that match the query molecule. Remarkably, ROCS was able to rank the first 37 hits as being only cruzain inhibitors. In addition, the area under the curve (AUC) obtained with ROCS was 0.96, indicating that the method was very efficient to distinguishing between cruzain and cathepsin L inhibitors.


Assuntos
Catepsinas/química , Catepsinas/farmacocinética , Cisteína Endopeptidases/química , Cisteína Endopeptidases/farmacocinética , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacocinética , Proteínas de Protozoários/antagonistas & inibidores , Animais , Catepsina L , Catepsinas/farmacologia , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Proteínas de Protozoários/metabolismo , Relação Quantitativa Estrutura-Atividade , Trypanosoma cruzi/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA