RÉSUMÉ
Clinical and preclinical studies have elucidated the favorable effects of Inhibitors of Sodium-Glucose Cotransporter-2 (iSGLT2) in patients and animal models with type 2 diabetes. Notably, these inhibitors have shown significant benefits in reducing hospitalizations and mortality among patients with heart failure. However, despite their incorporation into clinical practice for indications beyond diabetes, the decision-making process regarding their use often lacks a systematic approach. The selection of iSGLT2 remains arbitrary, with only a limited number of studies simultaneously exploring the different classes of them. Currently, no unique guideline establishes their application in both clinical and basic research. This review delves into the prevalent use of iSGLT2 in animal models previously subjected to induced cardiac stress. We have compiled key findings related to cardioprotection across various animal models, encompassing diverse dosages and routes of administration. Beyond their established role in diabetes management, iSGLT2 has demonstrated utility as agents for safeguarding heart health and cardioprotection can be class-dependent among the iSGLT2. These findings may serve as valuable references for other researchers. Preclinical studies play a pivotal role in ensuring the safety of novel compounds or treatments for potential human use. By assessing side effects, toxicity, and optimal dosages, these studies offer a robust foundation for informed decisions, identifying interventions with the highest likelihood of success and minimal risk to patients. The insights gleaned from preclinical studies, which play a crucial role in highlighting areas of knowledge deficiency, can guide the exploration of novel mechanisms and strategies involving iSGLT2.
Sujet(s)
Composés benzhydryliques , Canagliflozine , Cardiotoniques , Glucosides , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Glucosides/usage thérapeutique , Glucosides/pharmacologie , Animaux , Composés benzhydryliques/usage thérapeutique , Composés benzhydryliques/pharmacologie , Humains , Cardiotoniques/usage thérapeutique , Cardiotoniques/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Canagliflozine/usage thérapeutique , Canagliflozine/pharmacologie , Diabète de type 2/traitement médicamenteux , Hypoglycémiants/usage thérapeutique , Hypoglycémiants/pharmacologie , Évaluation préclinique de médicamentRÉSUMÉ
Chikungunya fever is a mosquito-borne disease caused by Chikungunya virus (CHIKV). Treatment of CHIKV infections is currently supportive and does not limit viral replication or symptoms of persistent chronic arthritis. Although there are multiple compounds reported as antivirals active against CHIKV in vitro, there are still no effective and safe antivirals. Thus, active research aims at the identification of new chemical structures with antiviral activity. Here, we report the screen of the Pandemic Response Box library of small molecules against a fully infectious CHIKV reporter virus. Our screening approach successfully identified previously reported CHIKV antiviral compounds within this library and further expanded potentially active hits, supporting the use of reporter-virus-based assays in high-throughput screening format as a reliable tool for antiviral drug discovery. Four molecules were identified as potential drug candidates against CHIKV: MMV1634402 (Brilacidin) and MMV102270 (Diphyllin), which were previously shown to present broad-spectrum antiviral activities, in addition to MMV1578574 (Eravacycline), and the antifungal MMV689401 (Fluopicolide), for which their antiviral potential is uncovered here.
Sujet(s)
Antiviraux , Fièvre chikungunya , Virus du chikungunya , Tests de criblage à haut débit , Bibliothèques de petites molécules , Virus du chikungunya/effets des médicaments et des substances chimiques , Antiviraux/pharmacologie , Antiviraux/composition chimique , Fièvre chikungunya/traitement médicamenteux , Fièvre chikungunya/virologie , Humains , Animaux , Bibliothèques de petites molécules/pharmacologie , Tests de criblage à haut débit/méthodes , Évaluation préclinique de médicament , Réplication virale/effets des médicaments et des substances chimiques , Découverte de médicament , Chlorocebus aethiops , Cellules VeroRÉSUMÉ
American trypanosomiasis or Chagas disease, caused by Trypanosoma cruzi (T. cruzi), affects approximately 6-7 million people worldwide. However, its pharmacological treatment causes several uncomfortable side effects, causing patients' treatment abandonment. Therefore, there is a need for new and better treatments. In this work, the molecular docking of nine hundred twenty-four FDA-approved drugs on three different sites of trypanothione reductase of T. cruzi (TcTR) was carried out to find potential trypanocidal agents. Finally, biological evaluations in vitro and in vivo were conducted with the selected FDA-approved drugs. Digoxin, alendronate, flucytosine, and dihydroergotamine showed better trypanocidal activity than the reference drugs benznidazole and nifurtimox in the in vitro evaluation against the trypomastigotes form. Further, these FDA-approved drugs were able to reduce 20-50% parasitemia in a short time in an in vivo model, although with less efficiency than benznidazole. Therefore, the results suggest a combined therapy of repurposed and canonical drugs against T. cruzi infection.
Sujet(s)
Maladie de Chagas , Simulation de docking moléculaire , NADH, NADPH oxidoreductases , Trypanocides , Trypanosoma cruzi , Trypanocides/pharmacologie , Trypanocides/composition chimique , NADH, NADPH oxidoreductases/antagonistes et inhibiteurs , NADH, NADPH oxidoreductases/composition chimique , NADH, NADPH oxidoreductases/métabolisme , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Trypanosoma cruzi/enzymologie , Maladie de Chagas/traitement médicamenteux , Animaux , Humains , Food and Drug Administration (USA) , Agrément de médicaments , Évaluation préclinique de médicament , États-Unis , SourisRÉSUMÉ
Chagas disease is a tropical neglected disease that affects millions of people worldwide, still demanding a more effective and safer therapy, especially in its chronic phase which lacks a treatment that promotes substantial parasitological cure. The technical note of Romanha and collaborators published in 2010 aimed establish a guideline with the set of minimum criteria and decision gates for the development of new agents against Trypanosoma cruzi with the focus on developing new antichagasic drugs. In this sense, the present review aims to update this technical note, bringing the state of the art and new advances on this topic in recent years.
Sujet(s)
Maladie de Chagas , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Trypanocides , Trypanosoma cruzi , Maladie de Chagas/traitement médicamenteux , Trypanocides/pharmacologie , Trypanocides/usage thérapeutique , Animaux , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Humains , Développement de médicamentRÉSUMÉ
This study presents a new approach for identifying myeloperoxidase (MPO) inhibitors with strong in vivo efficacy. By combining inhibitor-like rules and structure-based virtual screening, the pipeline achieved a 70% success rate in discovering diverse, nanomolar-potency reversible inhibitors and hypochlorous acid (HOCl) scavengers. Mechanistic analysis identified RL6 as a genuine MPO inhibitor and RL7 as a potent HOCl scavenger. Both compounds effectively suppressed HOCl production in cells and neutrophils, with RL6 showing a superior inhibition of neutrophil extracellular trap release (NETosis). In a gout arthritis mouse model, intraperitoneal RL6 administration reduced edema, peroxidase activity, and IL-1ß levels. RL6 also exhibited oral bioavailability, significantly reducing paw edema when administered orally. This study highlights the efficacy of integrating diverse screening methods to enhance virtual screening success, validating the anti-inflammatory potential of potent inhibitors, and advancing the MPO inhibitor research.
Sujet(s)
Goutte articulaire , Myeloperoxidase , Animaux , Myeloperoxidase/antagonistes et inhibiteurs , Myeloperoxidase/métabolisme , Goutte articulaire/traitement médicamenteux , Souris , Humains , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/usage thérapeutique , Mâle , Acide hypochloreux , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Relation structure-activité , Évaluation préclinique de médicamentRÉSUMÉ
Sporotrichosis is recognized as the predominant subcutaneous mycosis in South America, attributed to pathogenic species within the Sporothrix genus. Notably, in Brazil, Sporothrix brasiliensis emerges as the principal species, exhibiting significant sapronotic, zoonotic and enzootic epidemic potential. Consequently, the discovery of novel therapeutic agents for the treatment of sporotrichosis is imperative. The present study is dedicated to the repositioning of pharmaceuticals for sporotrichosis therapy. To achieve this goal, we designed a pipeline with the following steps: (a) compilation and preparation of Sporothrix genome data; (b) identification of orthologous proteins among the species; (c) identification of homologous proteins in publicly available drug-target databases; (d) selection of Sporothrix essential targets using validated genes from Saccharomyces cerevisiae; (e) molecular modeling studies; and (f) experimental validation of selected candidates. Based on this approach, we were able to prioritize eight drugs for in vitro experimental validation. Among the evaluated compounds, everolimus and bifonazole demonstrated minimum inhibitory concentration (MIC) values of 0.5 µg/mL and 4.0 µg/mL, respectively. Subsequently, molecular docking studies suggest that bifonazole and everolimus may target specific proteins within S. brasiliensis- namely, sterol 14-α-demethylase and serine/threonine-protein kinase TOR, respectively. These findings shed light on the potential binding affinities and binding modes of bifonazole and everolimus with their probable targets, providing a preliminary understanding of the antifungal mechanism of action of these compounds. In conclusion, our research advances the understanding of the therapeutic potential of bifonazole and everolimus, supporting their further investigation as antifungal agents for sporotrichosis in prospective hit-to-lead and preclinical investigations.
Sujet(s)
Antifongiques , Repositionnement des médicaments , Génome fongique , Tests de sensibilité microbienne , Sporothrix , Sporotrichose , Sporothrix/effets des médicaments et des substances chimiques , Sporothrix/génétique , Antifongiques/pharmacologie , Sporotrichose/microbiologie , Sporotrichose/traitement médicamenteux , Brésil , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Protéines fongiques/composition chimique , Simulation de docking moléculaire , Génomique , Humains , Évaluation préclinique de médicament , Découverte de médicament , Biologie informatiqueRÉSUMÉ
Proteases represent common targets in combating infectious diseases, including COVID-19. The 3-chymotrypsin-like protease (3CLpro) is a validated molecular target for COVID-19, and it is key for developing potent and selective inhibitors for inhibiting viral replication of SARS-CoV-2. In this review, we discuss structural relationships and diverse subsites of 3CLpro, shedding light on the pivotal role of dimerization and active site architecture in substrate recognition and catalysis. Our analysis of bioinformatics and other published studies motivated us to investigate a novel catalytic mechanism for the SARS-CoV-2 polyprotein cleavage by 3CLpro, centering on the triad mechanism involving His41-Cys145-Asp187 and its indispensable role in viral replication. Our hypothesis is that Asp187 may participate in modulating the pKa of the His41, in which catalytic histidine may act as an acid and/or a base in the catalytic mechanism. Recognizing Asp187 as a crucial component in the catalytic process underscores its significance as a fundamental pharmacophoric element in drug design. Next, we provide an overview of both covalent and non-covalent inhibitors, elucidating advancements in drug development observed in preclinical and clinical trials. By highlighting various chemical classes and their pharmacokinetic profiles, our review aims to guide future research directions toward the development of highly selective inhibitors, underscore the significance of 3CLpro as a validated therapeutic target, and propel the progression of drug candidates through preclinical and clinical phases.
Sujet(s)
Antiviraux , Traitements médicamenteux de la COVID-19 , Protéases 3C des coronavirus , SARS-CoV-2 , Protéases 3C des coronavirus/antagonistes et inhibiteurs , Protéases 3C des coronavirus/métabolisme , Protéases 3C des coronavirus/composition chimique , SARS-CoV-2/enzymologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Humains , Antiviraux/pharmacologie , Antiviraux/composition chimique , Domaine catalytique , Inhibiteurs de protéases/pharmacologie , Inhibiteurs de protéases/composition chimique , COVID-19/virologie , Essais cliniques comme sujet , Réplication virale/effets des médicaments et des substances chimiques , Évaluation préclinique de médicamentRÉSUMÉ
BACKGROUND: Conventional microscopic counting is a widely utilised method for evaluating the trypanocidal effects of drugs on intracellular amastigotes. This is a low-cost approach, but it is time-consuming and reliant on the expertise of the microscopist. So, there is a pressing need for developing technologies to enhance the efficiency of low-cost anti-Trypanosoma cruzi drug screening. OBJECTIVES: In our laboratory, we aimed to expedite the screening of anti-T. cruzi drugs by implementing a fluorescent method that correlates emitted fluorescence from green fluorescent protein (GFP)-expressing T. cruzi (Tc-GFP) with cellular viability. METHODS: Epimastigotes (Y strain) were transfected with the pROCKGFPNeo plasmid, resulting in robust and sustained GFP expression across epimastigotes, trypomastigotes, and intracellular amastigotes. Tc-GFP epimastigotes and intracellular amastigotes were exposed to a serial dilution of benznidazole (Bz). Cell viability was assessed through a combination of microscopic counting, MTT, and fluorimetry. FINDINGS: The fluorescence data indicated an underestimation of the activity of Bz against epimastigotes (IC50 75 µM x 14 µM). Conversely, for intracellular GFP-amastigotes, both fluorimetry and microscopy yielded identical IC50 values. Factors influencing the fluorimetry approach are discussed. MAIN CONCLUSIONS: Our proposed fluorometric assessment is effective and can serve as a viable substitute for the time-consuming microscopic counting of intracellular amastigotes.
Sujet(s)
Protéines à fluorescence verte , Trypanocides , Trypanosoma cruzi , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Trypanosoma cruzi/génétique , Protéines à fluorescence verte/génétique , Trypanocides/pharmacologie , Nitroimidazoles/pharmacologie , Tests de sensibilité parasitaire , Animaux , Concentration inhibitrice 50 , Évaluation préclinique de médicament , Survie cellulaire/effets des médicaments et des substances chimiquesRÉSUMÉ
INTRODUCTION: Chagas disease is spreading faster than expected in different countries, and little progress has been reported in the discovery of new drugs to combat Trypanosoma cruzi infection in humans. Recent clinical trials have ended with small hope. The pathophysiology of this neglected disease and the genetic diversity of parasites are exceptionally complex. The only two drugs available to treat patients are far from being safe, and their efficacy in the chronic phase is still unsatisfactory. AREAS COVERED: This review offers a comprehensive examination and critical review of data reported in the last 10 years, and it is focused on findings of clinical trials and data acquired in vivo in preclinical studies. EXPERT OPINION: The in vivo investigations classically in mice and dog models are also challenging and time-consuming to attest cure for infection. Poorly standardized protocols, availability of diagnosis methods and disease progression markers, the use of different T. cruzi strains with variable benznidazole sensitivities, and animals in different acute and chronic phases of infection contribute to it. More synchronized efforts between research groups in this field are required to put in evidence new promising substances, drug combinations, repurposing strategies, and new pharmaceutical formulations to impact the therapy.
Sujet(s)
Maladie de Chagas , Développement de médicament , Trypanocides , Trypanosoma cruzi , Animaux , Chiens , Humains , Souris , Maladie de Chagas/traitement médicamenteux , Maladie de Chagas/parasitologie , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Maladies négligées/traitement médicamenteux , Maladies négligées/parasitologie , Nitroimidazoles/pharmacologie , Nitroimidazoles/administration et posologie , Trypanocides/pharmacologie , Trypanosoma cruzi/effets des médicaments et des substances chimiquesRÉSUMÉ
Leishmaniasis are neglected infectious diseases caused by kinetoplastid protozoan parasites from the genus Leishmania. These sicknesses are present mainly in tropical regions and almost 1 million new cases are reported each year. The absence of vaccines, as well as the high cost, toxicity or resistance to the current drugs determines the necessity of new treatments against these pathologies. In this review, several compounds with potentialities as new antileishmanial drugs are presented. The discussion is restricted to the preclinical level and molecules are organized according to their chemical nature, source and molecular targets. In this manner, we present antimicrobial peptides, flavonoids, withanolides, 8-aminoquinolines, compounds from Leish-Box, pyrazolopyrimidines, and inhibitors of tubulin polymerization/depolymerization, topoisomerase IB, proteases, pteridine reductase, N-myristoyltransferase, as well as enzymes involved in polyamine metabolism, response against oxidative stress, signaling pathways, and sterol biosynthesis. This work is a contribution to the general knowledge of these compounds as antileishmanial agents.
Sujet(s)
Antiprotozoaires , Leishmania , Leishmaniose , Leishmaniose/traitement médicamenteux , Antiprotozoaires/pharmacologie , Antiprotozoaires/usage thérapeutique , Antiprotozoaires/composition chimique , Leishmania/effets des médicaments et des substances chimiques , Animaux , Humains , Évaluation préclinique de médicament , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Flavonoïdes/usage thérapeutiqueRÉSUMÉ
INTRODUCTION: Human neurodevelopmental and neurodegenerative diseases (NDevDs and NDegDs, respectively) encompass a broad spectrum of disorders affecting the nervous system with an increasing incidence. In this context, the nematode C. elegans, has emerged as a benchmark model for biological research, especially in the field of neuroscience. AREAS COVERED: The authors highlight the numerous advantages of this tiny worm as a model for exploring nervous system pathologies and as a platform for drug discovery. There is a particular focus given to describing the existing models of C. elegans for the study of NDevDs and NDegDs. Specifically, the authors underscore their strong applicability in preclinical drug development. Furthermore, they place particular emphasis on detailing the common techniques employed to explore the nervous system in both healthy and diseased states. EXPERT OPINION: Drug discovery constitutes a long and expensive process. The incorporation of invertebrate models, such as C. elegans, stands as an exemplary strategy for mitigating costs and expediting timelines. The utilization of C. elegans as a platform to replicate nervous system pathologies and conduct high-throughput automated assays in the initial phases of drug discovery is pivotal for rendering therapeutic options more attainable and cost-effective.
Sujet(s)
Caenorhabditis elegans , Modèles animaux de maladie humaine , Développement de médicament , Découverte de médicament , Maladies neurodégénératives , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Animaux , Humains , Découverte de médicament/méthodes , Développement de médicament/méthodes , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/physiopathologie , Tests de criblage à haut débit/méthodes , Évaluation préclinique de médicament/méthodes , Troubles du développement neurologique/traitement médicamenteux , Troubles du développement neurologique/physiopathologie , Maladies du système nerveux/traitement médicamenteux , Maladies du système nerveux/physiopathologieRÉSUMÉ
BACKGROUND: The SARS-CoV-2 pandemic has spurred an unparalleled scientific endeavor to elucidate the virus' structure, infection mechanisms, and pathogenesis. Two-dimensional culture systems have been instrumental in shedding light on numerous aspects of COVID-19. However, these in vitro systems lack the physiological complexity to comprehend the infection process and explore treatment options. Three-dimensional (3D) models have been proposed to fill the gap between 2D cultures and in vivo studies. Specifically, spheroids, composed of lung cell types, have been suggested for studying SARS-CoV-2 infection and serving as a drug screening platform. METHODS: 3D lung spheroids were prepared by coculturing human alveolar or bronchial epithelial cells with human lung stromal cells. The morphology, size, and ultrastructure of spheroids before and after SARS-CoV-2 infection were analyzed using optical and electron microscopy. Immunohistochemistry was used to detect spike protein and, thus, the virus presence in the spheroids. Multiplex analysis elucidated the cytokine release after virus infection. RESULTS: The spheroids were stable and kept their size and morphology after SARS-CoV-2 infection despite the presence of multivesicular bodies, endoplasmic reticulum rearrangement, tubular compartment-enclosed vesicles, and the accumulation of viral particles. The spheroid responded to the infection releasing IL-6 and IL-8 cytokines. CONCLUSION: This study demonstrates that coculture spheroids of epithelial and stromal cells can serve as a cost-effective infection model for the SARS-CoV-2 virus. We suggest using this 3D spheroid as a drug screening platform to explore new treatments related to the cytokines released during virus infection, especially for long COVID treatment.
Sujet(s)
COVID-19 , Évaluation préclinique de médicament , Poumon , SARS-CoV-2 , Sphéroïdes de cellules , Humains , Sphéroïdes de cellules/virologie , COVID-19/virologie , SARS-CoV-2/physiologie , Poumon/virologie , Poumon/anatomopathologie , Traitements médicamenteux de la COVID-19 , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Techniques de coculture , Cytokines/métabolisme , Analyse coût-bénéfice , Cellules épithéliales/virologieRÉSUMÉ
Tuberculosis (TB) remains a primary global health concern, necessitating the discovery and development of new anti-TB drugs, mainly to combat drug-resistant strains. In this context, thiourea derivatives have emerged as promising candidates in TB drug discovery due to their diverse chemical structures and pharmacological properties. This review aimed to explore this potential, identifying and exploring molecular targets for thiourea derivatives in Mycobacterium tuberculosis (Mtb) and the potential application of virtual screening techniques in drug discovery. We have compiled a comprehensive list of possible molecular targets of thiourea derivatives in Mtb. The enzymes are primarily involved in the biosynthesis of various cell wall components, including mycolic acids, peptidoglycans, and arabinans, or targets in the branched-chain amino acid biosynthesis (BCAA) pathway and detoxification mechanisms. We discuss the potential of these targets as critical constituents for the design of novel anti-TB drugs. Besides, we highlight the opportunities that virtual screening methodologies present in identifying potential thiourea derivatives that can interact with these molecular targets. The presented findings contribute to the ongoing efforts in TB drug discovery and lay the foundation for further research in designing and developing more effective treatments against this devastating disease.
Sujet(s)
Antituberculeux , Découverte de médicament , Mycobacterium tuberculosis , Thiourée , Thiourée/composition chimique , Thiourée/pharmacologie , Mycobacterium tuberculosis/effets des médicaments et des substances chimiques , Antituberculeux/composition chimique , Antituberculeux/pharmacologie , Humains , Tuberculose/traitement médicamenteux , Tuberculose/microbiologie , Tuberculose/diagnostic , Évaluation préclinique de médicamentRÉSUMÉ
Leishmaniasis is an infectious disease caused by protozoan species in the genera Leishmania and Endotrypanum. Current antileishmanial drugs are limited due to adverse effects, variable efficacy, the development of resistant parasites, high cost, parenteral administration and lack of availability in endemic areas. Therefore, active searching for new antileishmanial drugs has been done for years, mainly by academia. Drug screening techniques have been a challenge since the intracellular localization of Leishmania amastigotes implies that the host cell may interfere with the quantification of the parasites and the final estimation of the effect. One of the procedures to avoid host cell interference is based on its detergent-mediated lysis and subsequent transformation of viable amastigotes into promastigotes, their proliferation and eventual quantification as an axenic culture of promastigotes. However, the use of detergent involves additional handling of cultures and variability. In the present work, cultures of intracellular amastigotes were incubated for 72 h at 26 °C after exposure to the test compounds and the transformation and proliferation of parasites took place without need of adding any detergent. The assay demonstrated clear differentiation of negative and positive controls (average Z´ = 0.75) and 50% inhibitory concentrations of compounds tested by this method and by the gold standard enumeration of Giemsa-stained cultures were similar (p = 0.5002) and highly correlated (r = 0.9707). This simplified procedure is less labor intensive, the probability of contamination and the experimental error are reduced, and it is appropriate for the automated high throughput screening of compounds.
Sujet(s)
Antiprotozoaires , Leishmania , Leishmaniose , Parasites , Animaux , Évaluation préclinique de médicament , Détergents/pharmacologie , Détergents/usage thérapeutique , Antiprotozoaires/pharmacologieRÉSUMÉ
Since the number of drugs based on natural products (NPs) represents a large source of novel pharmacological entities, NPs have acquired significance in drug discovery. Peru is considered a megadiverse country with many endemic species of plants, terrestrial, and marine animals, and microorganisms. NPs databases have a major impact on drug discovery development. For this reason, several countries such as Mexico, Brazil, India, and China have initiatives to assemble and maintain NPs databases that are representative of their diversity and ethnopharmacological usage. We describe the assembly, curation, and chemoinformatic evaluation of the content and coverage in chemical space, as well as the physicochemical attributes and chemical diversity of the initial version of the Peruvian Natural Products Database (PeruNPDB), which contains 280 natural products. Access to PeruNPDB is available for free ( https://perunpdb.com.pe/ ). The PeruNPDB's collection is intended to be used in a variety of tasks, such as virtual screening campaigns against various disease targets or biological endpoints. This emphasizes the significance of biodiversity protection both directly and indirectly on human health.
Sujet(s)
Produits biologiques , Animaux , Humains , Pérou , Évaluation préclinique de médicament , Produits biologiques/pharmacologie , Produits biologiques/composition chimique , Bases de données factuelles , Découverte de médicamentRÉSUMÉ
Globalization has raised concerns about spreading diseases and emphasized the need for quick and efficient methods for drug screening. Established drug efficacy and toxicity approaches have proven obsolete, with a high failure rate in clinical trials. Organ-on-a-chip has emerged as an essential alternative to outdated techniques, precisely simulating important characteristics of organs and predicting drug pharmacokinetics more ethically and efficiently. Although promising, most organ-on-a-chip devices are still manufactured using principles and materials from the micromachining industry. The abusive use of plastic for traditional drug screening methods and device production should be considered when substituting technologies so that the compensation for the generation of plastic waste can be projected. This critical review outlines recent advances for organ-on-a-chip in the industry and estimates the possibility of scaling up its production. Moreover, it analyzes trends in organ-on-a-chip publications and provides suggestions for a more sustainable future for organ-on-a-chip research and production.
Sujet(s)
Laboratoires sur puces , Humains , Animaux , Évaluation préclinique de médicament , Secteur des soins de santé , Stérilisation/méthodes , Techniques de culture cellulaireRÉSUMÉ
Bruton tyrosine kinase (BTK), a nonreceptor tyrosine kinase, is a major therapeutic target for B-cell-driven malignancies. However, approved covalent BTK inhibitors (cBTKis) are associated with treatment limitations because of off-target side effects, suboptimal oral pharmacology, and development of resistance mutations (eg, C481) that prevent inhibitor binding. Here, we describe the preclinical profile of pirtobrutinib, a potent, highly selective, noncovalent (reversible) BTK inhibitor. Pirtobrutinib binds BTK with an extensive network of interactions to BTK and water molecules in the adenosine triphosphate binding region and shows no direct interaction with C481. Consequently, pirtobrutinib inhibits both BTK and BTK C481 substitution mutants in enzymatic and cell-based assays with similar potencies. In differential scanning fluorimetry studies, BTK bound to pirtobrutinib exhibited a higher melting temperature than cBTKi-bound BTK. Pirtobrutinib, but not cBTKis, prevented Y551 phosphorylation in the activation loop. These data suggest that pirtobrutinib uniquely stabilizes BTK in a closed, inactive conformation. Pirtobrutinib inhibits BTK signaling and cell proliferation in multiple B-cell lymphoma cell lines, and significantly inhibits tumor growth in human lymphoma xenografts in vivo. Enzymatic profiling showed that pirtobrutinib was highly selective for BTK in >98% of the human kinome, and in follow-up cellular studies pirtobrutinib retained >100-fold selectivity over other tested kinases. Collectively, these findings suggest that pirtobrutinib represents a novel BTK inhibitor with improved selectivity and unique pharmacologic, biophysical, and structural attributes with the potential to treat B-cell-driven cancers with improved precision and tolerability. Pirtobrutinib is being tested in phase 3 clinical studies for a variety of B-cell malignancies.
Sujet(s)
Agammaglobulinaemia tyrosine kinase , Lymphomes , Agammaglobulinaemia tyrosine kinase/antagonistes et inhibiteurs , Humains , Animaux , Tests d'activité antitumorale sur modèle de xénogreffe , Lymphomes/traitement médicamenteux , Évaluation préclinique de médicament , Lignée cellulaire tumorale , Souris de lignée NOD , Mâle , Souris SCID , Conformation moléculaire , SourisRÉSUMÉ
This chapter introduces a simple and robust in vitro viability assay to screen bioactive small molecules (e.g., natural, synthetic) against the monomorphic and infective (bloodstream) form of Trypanosoma brucei brucei. The assay relies on a bioluminescent transgenic parasite harboring a genetically encoded copy of a thermostable redshifted firefly luciferase from Photinus pyralis.The major advantages of the assay are simplicity and cost efficiency, along with excellent quality parameters. The bioassay allows estimating parasite numbers and viability (and metabolic state) as a function of bioluminescence (BL) signal. Parasites are grown in the presence of the molecules of interest in a 96-well microplate, and 24 h later, BL is determined with a simple protocol lacking washing steps, using cost-efficient reagents with a reasonable readout time for high-throughput applications.
Sujet(s)
Évaluation préclinique de médicament , Mesures de luminescence , Trypanosoma brucei brucei , Animaux , Évaluation préclinique de médicament/méthodes , Luciférases des lucioles , Mesures de luminescence/méthodes , Trypanosoma brucei brucei/effets des médicaments et des substances chimiquesRÉSUMÉ
To discover new molecules or review the biological activity and toxicity of therapeutic substances, drug development, and research relies on robust biological systems to obtain reliable results. Phenotype-based screenings can transpose the organism's compensatory pathways by adopting multi-target strategies for treating complex diseases, and zebrafish emerged as an important model for biomedical research and drug screenings. Zebrafish's clear correlation between neuro-anatomical and physiological features and behavior is very similar to that verified in mammals, enabling the construction of reliable and relevant experimental models for neurological disorders research. Zebrafish presents highly conserved physiological pathways that are found in higher vertebrates, including mammals, along with a robust behavioral repertoire. Moreover, it is very sensitive to pharmacological/environmental manipulations, and these behavioral phenotypes are detected in both larvae and adults. These advantages align with the 3Rs concept and qualify the zebrafish as a powerful tool for drug screenings and pre-clinical trials. This review highlights important behavioral domains studied in zebrafish larvae and their neurotransmitter systems and summarizes currently used techniques to evaluate and quantify zebrafish larvae behavior in laboratory studies.
Sujet(s)
Agents neuromédiateurs , Danio zébré , Animaux , Comportement animal/physiologie , Évaluation préclinique de médicament/méthodes , Larve/physiologie , Mammifères , Phénotype , Danio zébré/génétiqueRÉSUMÉ
Current human immunodeficiency virus treatments need to be periodically administered lifelong. In this study we assess the effect of repeated doses of an anti-HIV peptide drug candidate in C57BL6 strain. Two schemes of up to 15 administrations and one of 30, daily dosing for 5 days per week, all by the subcutaneous route were evaluated. Different dose concentrations of the peptide were assayed. CIGB-210 treated animals showed no symptoms or abnormal behavior as compared with placebo. All the animals gained weight during the study. Macroscopic evaluation showed no alterations in any of the organs studied. Microscopic analysis of the tissues did not show morphological changes in thymus, stomach, small and large intestines, kidney, brain, or cerebellum. The proliferative response of splenocytes and their capacity to secrete gamma interferon were not compromised by the repeated administration of CIGB-210. There were not statistically significant differences for any of the parameters evaluated during the study among treated and non-treated groups. We can conclude that CIGB-210 is well tolerated in C57BL6 mice in the dose concentration range explored and merits subsequent toxicological studies.