Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 225
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cancer Treat Rev ; 103: 102334, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34974243

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) has been investigated as a promising therapeutic target in select cancers with a mutated version of the enzyme (mtIDH1). With only one phase III trial published to date and two indications approved for routine clinical use by the FDA, we reviewed the entire clinical trial portfolio to broadly understand mtIDH1 inhibitor activity in patients. We queried PubMed.gov and ClinicalTrials.gov to identify published and ongoing clinical trials related to IDH1 and cancer. Progression-free survival (PFS), overall survival (OS), 2-hydroxyglutarate levels, and adverse events were summarized. To date, ten clinical trials investigating mtIDH1 inhibitors among patients with diverse malignancies (cholangiocarcinoma, acute myeloid leukemia, chondrosarcoma, glioma) have been published. Almost every trial (80%) has investigated ivosidenib. In multiple phase I trials, ivosidenib treatment resulted in promising radiographic and biochemical responses with improved survival outcomes (relative to historic data) among patients with both solid and hematologic mtIDH1 malignancies. Among patients enrolled in a phase III trial with advanced cholangiocarcinoma, ivosidenib resulted in a PFS rate of 32% at 6 months, as compared to 0% with placebo. There was a 5.2 month increase in OS with ivosidenib relative to placebo, after considering crossover. The treatment-specific grade ≥3 adverse event rate of ivosidenib was 2%-26% among all patients, and was just 3.6% among 284 patients who had a solid tumor across four trials. Although <1% of malignancies harbor IDH1 mutations, small molecule mtIDH1 inhibitors, namely ivosidenib, appear to be biologically active and well tolerated in patients with solid and hematologic mtIDH1 malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Glycine/analogs & derivatives , Isocitrate Dehydrogenase/antagonists & inhibitors , Neoplasms/drug therapy , Pyridines/therapeutic use , Aniline Compounds/adverse effects , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Benzimidazoles/adverse effects , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Clinical Trials as Topic , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Glycine/adverse effects , Glycine/pharmacology , Glycine/therapeutic use , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasms/mortality , Pyridines/adverse effects , Pyridines/pharmacology
2.
Drug Des Devel Ther ; 15: 4649-4664, 2021.
Article in English | MEDLINE | ID: mdl-34803375

ABSTRACT

BACKGROUND: The interaction of small molecules with direct targets constitutes the molecular initiation events of drug efficacy and toxicity. Aconitine, an active compound of the Aconitum species, has various pharmacological effects but is strongly toxic to the heart. The direct targets of aconitine-induced cardiotoxicity remain unclear. METHODS: We predicted the toxic targets of aconitine based on network pharmacology and followed a novel proteomic approach based on the "drug affinity responsive target stability" technology combined with LC-MS/MS to identify the direct targets of aconitine. The identified targets were analysed from the perspective of multilevel and multidimensional bioinformatics through a network integration method. The binding sites were investigated via molecular docking to explore the toxicity mechanism and predict the direct targets of aconitine. Finally, atomic force microscopy (AFM) imaging was performed to verify the affinity of aconitine to the direct targets. RESULTS: PTGS2, predicted by network pharmacology as a toxic target, encodes cyclooxygenase 2 (COX-2), which is closely related to myocardial injury. Furthermore, cytosolic phospholipase A2 (cPLA2) is the upstream signal protein of PTGS2, and it is a key enzyme in the metabolism of arachidonic acid during an inflammatory response. We determined cPLA2 as a direct target, and AFM imaging verified that aconitine could bind to cPLA2 well; thus, aconitine may cause the expression of PTGS2/COX-2 and release inflammatory factors, thereby promoting myocardial injury and dysfunction. CONCLUSION: We developed a complete set of methods to predict and verify the direct targets of aconitine, and cPLA2 was identified as one. Overall, the novel strategy provides new insights into the discovery of direct targets and the molecular mechanism of toxic components that are found in traditional Chinese medicine.


Subject(s)
Aconitine/adverse effects , Drugs, Chinese Herbal/adverse effects , Enzyme Inhibitors/adverse effects , Phospholipases A2, Cytosolic/antagonists & inhibitors , Aconitine/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Enzyme Inhibitors/chemistry , Medicine, Chinese Traditional , Molecular Conformation , Molecular Docking Simulation , Network Pharmacology , Phospholipases A2, Cytosolic/analysis , Phospholipases A2, Cytosolic/metabolism , Rats , Structure-Activity Relationship
3.
Eur J Pharmacol ; 912: 174583, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34678238

ABSTRACT

Anemia is a common feature and complication of chronic kidney disease (CKD). Erythropoiesis-stimulating agents (ESAs) and recombinant human erythropoietin have been used widely in renal anemia treatment. Recently, hypoxia-inducible factor-prolyl hydroxylase domain inhibitors (HIF-PHIs) that may improve the treatment of renal anemia patients were launched. Previous studies indicated that HIF-PHIs may decrease hepcidin levels and modulate iron metabolism, thereby increasing total iron-binding capacity and reducing the need for iron supplementation. Furthermore, HIF-PHIs can reduce inflammation and oxidative stress in CKD. Recombinant erythropoietin has become a routine treatment for patients with CKD and end-stage renal disease with relatively few adverse effects. However, higher doses of recombinant erythropoietin have been demonstrated to be an independent predictor of mortality in patients under hemodialysis. Phase III clinical trials of HIF-PHIs in patients with anemia and dialysis-dependent CKD have shown their efficacy and safety in both non-dialysis and dialysis CKD patients. However, HIFα binds to specific hypoxia-response elements in the vascular endothelial growth factor or retinoic acid-related orphan receptor gamma t (RORγt) promoter, which may be involved in the progression of cancer, psoriasis, and rheumatoid arthritis. In this paper, we have summarized the mechanism, clinical application, and clinical trials of HIF-PHIs in the treatment of renal anemia and aimed to provide an overview of the new drugs in clinical practice, as well as reconsider the advantages and disadvantages of HIF-PHIs and ESAs. Presently, there are not enough clinical studies examining the effects of long-term administration of HIF-PHIs. Therefore, further studies will be needed.


Subject(s)
Anemia/drug therapy , Anemia/metabolism , Enzyme Inhibitors/pharmacology , Hematinics/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Anemia/etiology , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiovascular Diseases/chemically induced , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Erythropoietin/adverse effects , Erythropoietin/pharmacology , Erythropoietin/therapeutic use , Hematinics/adverse effects , Hematinics/therapeutic use , Humans , Renal Insufficiency, Chronic/complications
4.
J Pharmacol Exp Ther ; 377(2): 242-253, 2021 05.
Article in English | MEDLINE | ID: mdl-33622769

ABSTRACT

Enhanced signaling of the endocannabinoid (eCB) system through inhibition of the catabolic enzymes monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) has received increasing interest for development of candidate analgesics. This study compared effects of MAGL and FAAH inhibitors with effects of ∆9-tetrahydrocannabinol (THC) using a battery of pain-stimulated, pain-depressed, and pain-independent behaviors in male and female mice. Intraperitoneal injection of dilute lactic acid (IP acid) served as an acute visceral noxious stimulus to stimulate two behaviors (stretching, facial grimace) and depress two behaviors (rearing, nesting). Nesting and locomotion were also assessed in the absence of IP acid as pain-independent behaviors. THC and a spectrum of six eCB catabolic enzyme inhibitors ranging from MAGL- to FAAH-selective were assessed for effectiveness to alleviate pain-related behaviors at doses that did not alter pain-independent behaviors. The MAGL-selective inhibitor MJN110 produced the most effective antinociceptive profile, with 1.0 mg/kg alleviating IP acid effects on stretching, grimace, and nesting without altering pain-independent behaviors. MJN110 effects on IP acid-depressed nesting had a slow onset and long duration (40 minutes to 6 hours), were blocked by rimonabant, and tended to be greater in females. As inhibitors increased in FAAH selectivity, antinociceptive effectiveness decreased. PF3845, the most FAAH-selective inhibitor, produced no antinociception up to doses that disrupted locomotion. THC decreased IP acid-stimulated stretching and grimace at doses that did not alter pain-independent behaviors; however, it did not alleviate IP acid-induced depression of rearing or nesting. These results support further consideration of MAGL-selective inhibitors as candidate analgesics for acute inflammatory pain. SIGNIFICANCE STATEMENT: This study characterized a spectrum of endocannabinoid catabolic enzyme inhibitors ranging in selectivity from monoacylglycerol lipase-selective to fatty acid amide hydrolase-selective in a battery of pain-stimulated, pain-depressed, and pain-independent behaviors previously pharmacologically characterized in a companion paper. This battery provides a method for prioritizing candidate analgesics by effectiveness to alleviate pain-related behaviors at doses that do not alter pain-independent behaviors, with inclusion of pain-depressed behaviors increasing translational validity and decreasing susceptibility to motor-depressant false positives.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Behavior, Animal/drug effects , Dronabinol/pharmacology , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Analgesics/adverse effects , Animals , Dronabinol/adverse effects , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Enzyme Inhibitors/adverse effects , Female , Male , Mice , Mice, Inbred ICR , Nociception/drug effects
6.
Leukemia ; 34(11): 2903-2913, 2020 11.
Article in English | MEDLINE | ID: mdl-32733012

ABSTRACT

The mutant IDH1 (mIDH1) inhibitor BAY1436032 demonstrated robust activity in preclinical AML models, supporting clinical evaluation. In the current dose-escalation study, BAY1436032 was orally administered to 27 mIDH1 AML subjects across 4 doses ranging from 300 to 1500 mg twice-daily. BAY1436032 exhibited a relatively short half-life and apparent non-linear pharmacokinetics after continuous dosing. Most subjects experienced only partial target inhibition as indicated by plasma R-2HG levels. BAY1436032 was safe and a maximum tolerated dose was not identified. The median treatment duration for all subjects was 3.0 months (0.49-8.5). The overall response rate was 15% (4/27; 1 CRp, 1 PR, 2 MLFS), with responding subjects experiencing a median treatment duration of 6.0 months (3.9-8.5) and robust R-2HG decreases. Thirty percent (8/27) achieved SD, with a median treatment duration of 5.5 months (3.1-7.0). Degree of R-2HG inhibition and clinical benefit did not correlate with dose. Although BAY1436032 was safe and modestly effective as monotherapy, the low overall response rate and incomplete target inhibition achieved at even the highest dose tested do not support further clinical development of this investigational agent in AML.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Enzyme Inhibitors/therapeutic use , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Molecular Targeted Therapy , Mutation , Adult , Aged , Aniline Compounds/administration & dosage , Aniline Compounds/adverse effects , Aniline Compounds/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Bone Marrow/pathology , DNA Mutational Analysis , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Prognosis , Survival Analysis , Treatment Outcome
7.
J Comput Aided Mol Des ; 34(11): 1117-1132, 2020 11.
Article in English | MEDLINE | ID: mdl-32833084

ABSTRACT

There is a pressing need to improve the efficiency of drug development, and nowhere is that need more clear than in the case of neglected diseases like malaria. The peculiarities of pyrimidine metabolism in Plasmodium species make inhibition of dihydroorotate dehydrogenase (DHODH) an attractive target for antimalarial drug design. By applying a pair of complementary quantitative structure-activity relationships derived for inhibition of a truncated, soluble form of the enzyme from Plasmodium falciparum (s-PfDHODH) to data from a large-scale phenotypic screen against cultured parasites, we were able to identify a class of antimalarial leads that inhibit the enzyme and abolish parasite growth in blood culture. Novel analogs extending that class were designed and synthesized with a goal of improving potency as well as the general pharmacokinetic and toxicological profiles. Their synthesis also represented an opportunity to prospectively validate our in silico property predictions. The seven analogs synthesized exhibited physicochemical properties in good agreement with prediction, and five of them were more active against P. falciparum growing in blood culture than any of the compounds in the published lead series. The particular analogs prepared did not inhibit s-PfDHODH in vitro, but advanced biological assays indicated that other examples from the class did inhibit intact PfDHODH bound to the mitochondrial membrane. The new analogs, however, killed the parasites by acting through some other, unidentified mechanism 24-48 h before PfDHODH inhibition would be expected to do so.


Subject(s)
Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Malaria, Falciparum/drug therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Plasmodium falciparum/drug effects , Quinolones/chemistry , Antimalarials/adverse effects , Antimalarials/pharmacokinetics , Dihydroorotate Dehydrogenase , Drug Design , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Molecular Structure , Quantitative Structure-Activity Relationship , Quinolones/adverse effects , Quinolones/pharmacokinetics
8.
Cochrane Database Syst Rev ; 2: CD003481, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32045960

ABSTRACT

BACKGROUND: Indomethacin is used as standard therapy to close a patent ductus arteriosus (PDA) but is associated with reduced blood flow to several organs. Ibuprofen, another cyclo-oxygenase inhibitor, may be as effective as indomethacin with fewer adverse effects. OBJECTIVES: To determine the effectiveness and safety of ibuprofen compared with indomethacin, other cyclo-oxygenase inhibitor(s), placebo, or no intervention for closing a patent ductus arteriosus in preterm, low-birth-weight, or preterm and low-birth-weight infants. SEARCH METHODS: We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL 2017, Issue 10), MEDLINE via PubMed (1966 to 30 November 2017), Embase (1980 to 30 November 2017), and CINAHL (1982 to 30 November 2017). We searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA: Randomised or quasi-randomised controlled trials of ibuprofen for the treatment of a PDA in preterm, low birth weight, or both preterm and low-birth-weight newborn infants. DATA COLLECTION AND ANALYSIS: Data collection and analysis conformed to the methods of the Cochrane Neonatal Review Group. We used the GRADE approach to assess the quality of evidence. MAIN RESULTS: We included 39 studies enrolling 2843 infants. Ibuprofen (IV) versus placebo: IV Ibuprofen (3 doses) reduced the failure to close a PDA compared with placebo (typical relative risk (RR); 0.62 (95% CI 0.44 to 0.86); typical risk difference (RD); -0.18 (95% CI -0.30 to -0.06); NNTB 6 (95% CI 3 to 17); I2 = 65% for RR and I2 = 0% for RD; 2 studies, 206 infants; moderate-quality the evidence). One study reported decreased failure to close a PDA after single or three doses of oral ibuprofen compared with placebo (64 infants; RR 0.26, 95% CI 0.11 to 0.62; RD -0.44, 95% CI -0.65 to -0.23; NNTB 2, 95% CI 2 to 4; I2 test not applicable). Ibuprofen (IV or oral) compared with indomethacin (IV or oral): Twenty-four studies (1590 infants) comparing ibuprofen (IV or oral) with indomethacin (IV or oral) found no significant differences in failure rates for PDA closure (typical RR 1.07, 95% CI 0.92 to 1.24; typical RD 0.02, 95% CI -0.02 to 0.06; I2 = 0% for both RR and RD; moderate-quality evidence). A reduction in NEC (necrotising enterocolitis) was noted in the ibuprofen (IV or oral) group (18 studies, 1292 infants; typical RR 0.68, 95% CI 0.49 to 0.94; typical RD -0.04, 95% CI -0.07 to -0.01; NNTB 25, 95% CI 14 to 100; I2 = 0% for both RR and RD; moderate-quality evidence). There was a statistically significant reduction in the proportion of infants with oliguria in the ibuprofen group (6 studies, 576 infants; typical RR 0.28, 95% CI 0.14 to 0.54; typical RD -0.09, 95% CI -0.14 to -0.05; NNTB 11, 95% CI 7 to 20; I2 = 24% for RR and I2 = 69% for RD; moderate-quality evidence). The serum/plasma creatinine levels 72 hours after initiation of treatment were statistically significantly lower in the ibuprofen group (11 studies, 918 infants; MD -8.12 µmol/L, 95% CI -10.81 to -5.43). For this comparison, there was high between-study heterogeneity (I2 = 83%) and low-quality evidence. Ibuprofen (oral) compared with indomethacin (IV or oral): Eight studies (272 infants) reported on failure rates for PDA closure in a subgroup of the above studies comparing oral ibuprofen with indomethacin (IV or oral). There was no significant difference between the groups (typical RR 0.96, 95% CI 0.73 to 1.27; typical RD -0.01, 95% CI -0.12 to 0.09; I2 = 0% for both RR and RD). The risk of NEC was reduced with oral ibuprofen compared with indomethacin (IV or oral) (7 studies, 249 infants; typical RR 0.41, 95% CI 0.23 to 0.73; typical RD -0.13, 95% CI -0.22 to -0.05; NNTB 8, 95% CI 5 to 20; I2 = 0% for both RR and RD). There was low-quality evidence for these two outcomes. There was a decreased risk of failure to close a PDA with oral ibuprofen compared with IV ibuprofen (5 studies, 406 infants; typical RR 0.38, 95% CI 0.26 to 0.56; typical RD -0.22, 95% CI -0.31 to -0.14; NNTB 5, 95% CI 3 to 7; moderate-quality evidence). There was a decreased risk of failure to close a PDA with high-dose versus standard-dose of IV ibuprofen (3 studies 190 infants; typical RR 0.37, 95% CI 0.22 to 0.61; typical RD - 0.26, 95% CI -0.38 to -0.15; NNTB 4, 95% CI 3 to 7); I2 = 4% for RR and 0% for RD); moderate-quality evidence). Early versus expectant administration of IV ibuprofen, echocardiographically-guided IV ibuprofen treatment versus standard IV ibuprofen treatment, continuous infusion of ibuprofen versus intermittent boluses of ibuprofen, and rectal ibuprofen versus oral ibuprofen were studied in too few trials to allow for precise estimates of any clinical outcomes. AUTHORS' CONCLUSIONS: Ibuprofen is as effective as indomethacin in closing a PDA. Ibuprofen reduces the risk of NEC and transient renal insufficiency. Therefore, of these two drugs, ibuprofen appears to be the drug of choice. The effectiveness of ibuprofen versus paracetamol is assessed in a separate review. Oro-gastric administration of ibuprofen appears as effective as IV administration. To make further recommendations, studies are needed to assess the effectiveness of high-dose versus standard-dose ibuprofen, early versus expectant administration of ibuprofen, echocardiographically-guided versus standard IV ibuprofen, and continuous infusion versus intermittent boluses of ibuprofen. Studies are lacking evaluating the effect of ibuprofen on longer-term outcomes in infants with PDA.


Subject(s)
Ductus Arteriosus, Patent/drug therapy , Ibuprofen/therapeutic use , Infant, Low Birth Weight , Cyclooxygenase Inhibitors/therapeutic use , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Humans , Ibuprofen/adverse effects , Indomethacin/adverse effects , Indomethacin/therapeutic use , Infant, Newborn , Infant, Premature , Randomized Controlled Trials as Topic
9.
Regul Toxicol Pharmacol ; 111: 104553, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31843592

ABSTRACT

New effective compounds to treat tuberculosis are urgently needed. IQG-607 is an orally active anti-tuberculosis drug candidate, with promising preliminary safety profile and anti-mycobacterial activity in both in vitro and in vivo models of tuberculosis infection. Here, we evaluated the mutagenic and genotoxic effects of IQG-607, and its interactions with CYP450 isoforms. Moreover, we describe for the first time a combination study of IQG-607 in Mycobacterium tuberculosis-infected mice. Importantly, IQG-607 had additive effects when combined with the first-line anti-tuberculosis drugs rifampin and pyrazinamide in mice. IQG-607 presented weak to moderate inhibitory potential against CYP450 isoforms 3A4, 1A2, 2C9, 2C19, 2D6, and 2E1. The Salmonella mutagenicity test revealed that IQG-607 induced base pair substitution mutations in the strains TA100 and TA1535. However, in the presence of human metabolic S9 fraction, no mutagenic effect was detected in any strain. Additionally, IQG-607 did not increase micronucleus frequencies in mice, at any dose tested, 25, 100, or 250 mg/kg. The favorable activity in combination with first-line drugs and mild to moderate toxic events described in this study suggest that IQG-607 represents a candidate for clinical development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ferrous Compounds/adverse effects , Ferrous Compounds/pharmacology , Isoniazid/analogs & derivatives , Mycobacterium tuberculosis/drug effects , Salmonella typhimurium/drug effects , Tuberculosis/drug therapy , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Chromosome Aberrations , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Ferrous Compounds/administration & dosage , Isoniazid/administration & dosage , Isoniazid/adverse effects , Isoniazid/pharmacology , Male , Mice , Microbial Sensitivity Tests , Mutagenicity Tests , Mycobacterium tuberculosis/genetics , Salmonella typhimurium/genetics , Tuberculosis/microbiology
10.
Int J Dermatol ; 59(2): 178-183, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31681985

ABSTRACT

BACKGROUND: Skin depigmentation is increasingly oriented toward plant extracts because of harmfulness of depigmenting active ingredients used in cosmetics and dermatology. Reconstructed human pigmented epidermis (RHPE) is the closest in vitro model to human skin and offers the possibility to test the global depigmenting effect of a plant extract. These co-cultures of keratinocytes and melanocytes are the most advanced and newest models for testing depigmentation, and until now very few studies have been done with these cultures. We investigated the cytotoxicity and the inhibitory effect on tyrosinase and melanogenesis of four extracts from Combretum micranthum (G. Don) leaves, Anacardium occidentale (L.) fruits, Moringa oleifera (Lam.) seeds, and Adansonia digitata (L.) seeds. METHODS: The vegetal extracts were obtained by ultrasound-assisted extraction and the vegetal oils by maceration. Anti-tyrosinase properties of two aqueous extracts were evaluated. Then, the cytotoxicity and depigmenting effects of these plant extracts were tested in vitro with RHPE model delivered by SkinEthic® . RESULTS: Antityrosinase activities were found to be 84.58% and 31.02% for C. micranthum and A. occidentale, respectively. All extracts, except A. occidentale, showed to be nontoxic. C. micranthum, M. oleifera, A. digitata, and mixture of M. oleifera and A. digitata extracts have shown, for the first time, an in vitro depigmenting activity equivalent or even more important than kojic acid. CONCLUSIONS: These natural extracts coming from Senegal botanical biodiversity could be used in cosmetic and dermatology as alternative agents to achieve skin depigmentation. Further study should be focused on the mechanism of action of these plant extracts.


Subject(s)
Enzyme Inhibitors/pharmacology , Phytotherapy , Plant Extracts/pharmacology , Skin Lightening Preparations/pharmacology , Skin Pigmentation/drug effects , Adansonia , Anacardium , Cell Survival/drug effects , Combretum , Enzyme Inhibitors/adverse effects , Fruit , Humans , Keratinocytes , Melanins/biosynthesis , Melanocytes/drug effects , Monophenol Monooxygenase/antagonists & inhibitors , Moringa oleifera , Phytotherapy/adverse effects , Plant Extracts/adverse effects , Plant Leaves , Protein Biosynthesis/drug effects , Seeds , Skin Lightening Preparations/adverse effects , Tissue Culture Techniques
11.
Expert Opin Investig Drugs ; 29(2): 135-141, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31519114

ABSTRACT

Introduction: De novo lipogenesis (DNL) plays a major role in fatty acid metabolism and contributes significantly to triglyceride accumulation within the hepatocytes in patients with nonalcoholic steatohepatitis (NASH). Acetyl-CoA carboxylase (ACC) converts acetyl-CoA to malonyl CoA and is a rate-controlling step in DNL. Furthermore, malonyl-CoA is an important regulator of hepatic mitochondrial fat oxidation through its ability to inhibit carnitine palmitoyltransferase I. Therefore, inhibiting ACC pharmacologically represents an attractive approach to treating NASH.Areas covered: This article summarizes preclinical and clinical data on the efficacy and safety of the liver-targeted ACC inhibitor GS-0976 (Firsocostat) for the treatment of NASH. In a phase 2 trial that included 126 patients with NASH and fibrosis, GS-0976 20 mg daily for 12 weeks showed significant relative reduction in liver fat by 29%; however, treatment was associated with an increase in plasma triglycerides with 16 patients having levels >500 mg/dL.Expert opinion: Preclinical and preliminary clinical data support the development of GS-0976 as treatment for NASH. ACC-induced hypertriglyceridemia can be mitigated by fish oil or fibrates, but the long-term cardiovascular effects require further investigations.


Subject(s)
Enzyme Inhibitors/therapeutic use , Isobutyrates/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Oxazoles/therapeutic use , Pyrimidines/therapeutic use , Acetyl-CoA Carboxylase/antagonists & inhibitors , Animals , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Fatty Acids/metabolism , Humans , Isobutyrates/adverse effects , Isobutyrates/pharmacology , Lipogenesis/drug effects , Non-alcoholic Fatty Liver Disease/physiopathology , Oxazoles/adverse effects , Oxazoles/pharmacology , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Triglycerides/blood
12.
Pharmacol Res ; 150: 104510, 2019 12.
Article in English | MEDLINE | ID: mdl-31678209

ABSTRACT

Herbal medicines are widely used as alternative or complementary therapies worldwide to treat and prevent chronic diseases. However, herbal medicines coadministration with therapeutic drugs may cause dramatic clinical herb-drug/herb interactions (HDIs/HHIs) that may result in low drug efficacy or serious toxic reactions. Phase II metabolism enzyme UDP-glucuronosyltransferases (UGTs) play a significant detoxification role in vivo. Most drugs and non-drug xenobiotics undergo phase II metabolic transformations to be more polar compounds that are more easily excreted. Herbal medicines are a mixed and chemically varied group that includes flavonoids, stilbenes, coumarins, quinones, and terpenes, which are potential substrates and inhibitors of UGTs. Although increasing studies about glucuronidation metabolism and the inhibition toward UGTs of many herbal medicines have been reported, it is still difficult to determine which compounds from herbal medicines are substrates or inhibitors of UGTs. This article gives an overview of UGTs studies, which mainly focuses on glucuronidation of herbal constituents as substrates catalyzed by UGTs, potential herbal inhibitors for UGTs. We summarize the negative effects of UGT1A polymorphism and single nucleotide polymorphisms (SNPs), relevant clinical situations of HDIs/HHIs induced by inhibition of UGTs, and propose establishing classification criteria for inhibitors. Finally, we also discuss future research and strategic directions to advance the understanding of the potential HDIs/HHIs and suggest some additional studies revealing more information on UGT-mediated HDIs/HHIs.


Subject(s)
Enzyme Inhibitors/adverse effects , Glucuronosyltransferase/antagonists & inhibitors , Herb-Drug Interactions , Animals , Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Plants, Medicinal , Polymorphism, Genetic , Substrate Specificity
13.
Expert Opin Ther Pat ; 29(3): 199-213, 2019 03.
Article in English | MEDLINE | ID: mdl-30760060

ABSTRACT

INTRODUCTION: Aldose reductase (ALR2) is both the key enzyme of the polyol pathway, whose activation under hyperglycemic conditions leads to the development of chronic diabetic complications, and the crucial promoter of inflammatory and cytotoxic conditions, even under a normoglycemic status. Accordingly, it represents an excellent drug target and a huge effort is being done to disclose novel compounds able to inhibit it. AREAS COVERED: This literature survey summarizes patents and patent applications published over the last 5 years and filed for natural, semi-synthetic and synthetic ALR2 inhibitors. Compounds described have been discussed and analyzed from both chemical and functional angles. EXPERT OPINION: Several ALR2 inhibitors with a promising pre-clinical ability to address diabetic complications and inflammatory diseases are being developed during the observed timeframe. Natural compounds and plant extracts are the prevalent ones, thus confirming the use of phytopharmaceuticals as an increasingly pursued therapeutic trend also in the ALR2 inhibitors field. Intriguing hints may be taken from synthetic derivatives, the most significant ones being represented by the differential inhibitors ARDIs. Differently from classical ARIs, these compounds should fire up the therapeutic efficacy of the class while minimizing its side effects, thus overcoming the existing limits of this kind of inhibitors.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , Aldehyde Reductase/metabolism , Animals , Diabetes Complications/drug therapy , Diabetes Complications/enzymology , Enzyme Inhibitors/adverse effects , Humans , Inflammation/drug therapy , Inflammation/enzymology , Patents as Topic
14.
Arch Toxicol ; 93(2): 227-251, 2019 02.
Article in English | MEDLINE | ID: mdl-30499019

ABSTRACT

Folate is vital for cell development and growth. It is involved in one-carbon transfer reactions essential for the synthesis of purines and pyrimidines. It also acts in conjunction with cobalamin (vitamin B12) as a fundamental cofactor in the remethylation cycle that converts homocysteine to methionine. A deficiency in folate or vitamin B12 can lead to elevated homocysteine level, which has been identified as an independent risk factor in several health-related conditions. Adequate folate levels are essential in women of childbearing age and in pregnant women, and folate deficiency is associated with several congenital malformations. Low folate levels can be caused by dietary deficiencies, a genetic predisposition or treatment with medicines that affect folate concentration. Women who are pregnant or of child-bearing age commonly use medicines, so it is important to identify the basic biochemical mechanisms by which medicines interfere with the folate-homocysteine-methionine pathway. This review focuses on prescription medicines associated with folate disruption. It also summarizes their undesirable/toxic effects. Recommendations regarding folate supplementation during medical therapy are also reviewed.


Subject(s)
Enzyme Inhibitors/adverse effects , Folic Acid Antagonists/adverse effects , Folic Acid Deficiency/etiology , Folic Acid/metabolism , Homocysteine/metabolism , Methionine/metabolism , Enzyme Inhibitors/therapeutic use , Female , Folic Acid/pharmacology , Folic Acid Deficiency/complications , Humans , Pregnancy , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/antagonists & inhibitors , Vitamin B 12 Deficiency/etiology
15.
Sci Rep ; 8(1): 12142, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108271

ABSTRACT

Fatty acid amide hydrolase (FAAH) is an important enzyme for lipid metabolism and an interesting pharmacological target, given its role in anandamide breakdown. The FAAH-/- genotype is the most widely used mouse model to investigate the effects of a complete pharmacological inhibition of this enzyme. In this paper, we explore, by means of label-free SWATH proteomics, the changes in protein expression occurring in the liver of FAAH-/- knockout (KO) mice. We identified several altered biological processes and pathways, like fatty acid synthesis and glycolysis, which explain the observed phenotype of this mouse. We also observed the alteration of other proteins, like carboxylesterases and S-methyltransferases, apparently not immediately related to FAAH, but known to have important biological roles. Our study, reporting more than 3000 quantified proteins, offers an in-depth analysis of the liver proteome of this model.


Subject(s)
Amidohydrolases/genetics , Liver/enzymology , Proteomics/methods , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Computational Biology , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/adverse effects , Fatty Acids/biosynthesis , Glycolysis/genetics , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Models, Animal , Proteome/metabolism , Tandem Mass Spectrometry/methods
16.
Clin Microbiol Infect ; 24 Suppl 2: S41-S52, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29426804

ABSTRACT

BACKGROUND: The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biologic therapies. AIMS: To review, from an infectious diseases perspective, the safety profile of therapies targeting cell surface receptors and associated signaling pathways among cancer patients and to suggest preventive recommendations. SOURCES: Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT: Vascular endothelial growth factor (VEGF)-targeted agents (bevacizumab and aflibercept) are associated with a meaningful increase in the risk of infection, likely due to drug-induced neutropaenia, although no clear benefit is expected from the universal use of anti-infective prophylaxis. VEGF tyrosine kinase inhibitors (i.e. sorafenib or sunitinib) do not seem to significantly affect host's susceptibility to infection, and universal anti-infective prophylaxis is not recommended either. Anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (cetuximab or panitumumab) induce neutropaenia and secondary skin and soft tissue infection in cases of severe papulopustular rash. Systemic antibiotics (doxycycline or minocycline) should be administered to prevent the latter complication, whereas no recommendation can be established on the benefit from antiviral, antifungal or anti-Pneumocystis prophylaxis. A lower risk of infection is reported for anti-ErbB2/HER2 monoclonal antibodies (trastuzumab and pertuzumab) and ErbB receptor tyrosine kinase inhibitors (including dual-EGFR/ErbB2 inhibitors such as lapatinib or neratinib) compared to conventional chemotherapy, presumably as a result of the decreased occurrence of drug-induced neutropaenia. IMPLICATIONS: With the exception of VEGF-targeted agents, the overall risk of infection associated with the reviewed therapies seems to be low.


Subject(s)
Biological Therapy/adverse effects , Communicable Diseases/therapy , Molecular Targeted Therapy/adverse effects , Signal Transduction/drug effects , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Biological Therapy/methods , Clinical Trials as Topic , Communicable Disease Control , Consensus , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Humans , Immunocompromised Host , Molecular Targeted Therapy/methods , Receptors, Cell Surface/antagonists & inhibitors , Vascular Endothelial Growth Factor A/drug effects
17.
J Med Food ; 21(3): 207-214, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29315017

ABSTRACT

Nonceliac gluten sensitivity (NCGS) is a gluten-related gastrointestinal disorder distinct from celiac disease (CD) and gluten allergy that is not easy to diagnose due to the lack of biomarkers. It is characterized by intestinal symptoms and extraintestinal manifestations with the consumption of gluten-containing foods. In contrast to CD, NCGS patients do not present a genetic predisposition or intestinal villi atrophy. Recent studies question the proinflammatory triggering activity of α-gliadin fraction contained in wheat, since it has been demonstrated that the amylase-trypsin inhibitors (ATIs) exert a strong activating effect on the innate immune response. We aimed to analyze the role of ATIs in the activation of innate immunity and in the development of the symptoms characteristic of NCGS. A systematic literature search was made using databases such as MEDLINE, SciELO, Science Direct, and Scopus, with focus on key words such as "amylase-trypsin inhibitors," "wheat," "gluten," and "celiac." Many studies are available on the structure, inhibition mechanism, and immune system effects of ATIs, mainly focused on IgE-mediated reactions. Recently, with the increase of NCGS interest, has increased the literature on the capacity of ATIs contained in wheat to activate the innate immune system. Literature published to date questions the relationship between activation of the innate immune system and gluten in NCGS. ATIs may have acted as interfering contaminant of gluten and appear as potential activator of innate immunity in NCGS patients. In view of their potential impact, more interventional studies are needed to demonstrate the proinflammatory effect of ATIs.


Subject(s)
Edible Grain/adverse effects , Enzyme Inhibitors/adverse effects , Food Intolerance/etiology , Glutens/adverse effects , Plant Proteins/adverse effects , Trypsin Inhibitors/adverse effects , alpha-Amylases/antagonists & inhibitors , Animals , Edible Grain/chemistry , Enzyme Inhibitors/analysis , Enzyme Inhibitors/metabolism , Food Intolerance/immunology , Food Intolerance/metabolism , Food Intolerance/physiopathology , Glutens/metabolism , Hordeum/adverse effects , Hordeum/chemistry , Humans , Immunity, Innate , Immunity, Mucosal , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Plant Proteins/analysis , Plant Proteins/metabolism , Secale/adverse effects , Secale/chemistry , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Triticum/adverse effects , Triticum/chemistry , Trypsin Inhibitors/analysis , Trypsin Inhibitors/metabolism
18.
Ann Anat ; 212: 27-36, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28455132

ABSTRACT

Nω-Nitro-l-arginine methyl ester (l-NAME)-induced hypertension and cardiovascular remodeling are associated with oxidative stress and inflammation. Garcinia mangostana Linn., has been reported to have antioxidant and anti-inflammatory properties. This study investigated whether G. mangostana pericarp extract (GME) could prevent l-NAME-induced hemodynamic alterations, cardiovascular remodeling, oxidative stress and inflammation in rats. Male Sprague-Dawley rats were given 40mg/kg/day of l-NAME in drinking water to induce hypertension, and were simultaneously treated with GME at a dose of 200mg/kg/day. Rats that received l-NAME for five weeks had high blood pressure, left ventricular hypertrophy and thickening of aortic wall. Vascular superoxide production, plasma malondialdehyde (MDA), and plasma tumor necrosis factor alpha (TNF-α) were significantly increased in l-NAME-hypertensive rats (p<0.05). This was consistent with up-regulation of the p47phox NADPH oxidase subunit and iNOS protein expression in aortic tissues (p<0.05). Low levels of plasma nitric oxide metabolites were observed in l-NAME hypertension. GME prevented the development of hypertension and cardiovascular remodeling induced by l-NAME with reduced oxidative stress and inflammation. These data suggest that GME had a protective effect against l-NAME-induced hypertension and cardiovascular remodeling via suppressing p47phox NADPH oxidase subunit and iNOS protein expression resulting in enhancing NO bioavailability.


Subject(s)
Cardiovascular System/drug effects , Garcinia mangostana/chemistry , Hypertension/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Free Radical Scavengers/metabolism , Fruit/chemistry , Hypertension/chemically induced , Hypertension/complications , Hypertension/metabolism , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/prevention & control , Inflammation/etiology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/adverse effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Plant Extracts/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Ventricular Remodeling/drug effects
19.
Cancer Lett ; 400: 325-335, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28323035

ABSTRACT

Natural products are considered as promising tools for the prevention and treatment of cancer. The enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase unit of polycomb repressor complexes such as PRC2 complex that has oncogenic roles through interference with growth and metastatic potential. Several agents targeting EZH2 has been discovered but they often induce side effects in clinical trials. Recently, EZH2 has emerged as a potential target of natural products with documented anti-cancer effects and this discloses a new scenario for the development of EZH2 inhibitory strategies with agents with low cytotoxic detrimental effects. In fact, several natural products such as curcumin, triptolide, ursolic acid, sulforaphane, davidiin, tanshindiols, gambogic acid, berberine and Alcea rosea have been shown to serve as EZH2 modulators. Mechanisms like inhibition of histone H3K4, H3K27 and H3K36 trimethylation, down-regulation of matrix metalloproteinase expression, competitive binding to the S-adenosylmethionine binding site of EZH2 and modulation of tumor-suppressive microRNAs have been demonstrated to mediate the EZH2-inhibitory activity of the mentioned natural products. This review summarizes the pathways that are regulated by various natural products resulting in the suppression of EZH2, and provides a plausible molecular mechanism for the putative anti-cancer effects of these compounds.


Subject(s)
Antineoplastic Agents/therapeutic use , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Half-Life , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/pathology , Signal Transduction/drug effects
20.
Diabetes Obes Metab ; 19(8): 1165-1170, 2017 08.
Article in English | MEDLINE | ID: mdl-28261955

ABSTRACT

AIMS: Hypothalamic injury-associated obesity (HIAO) results from damage to the hypothalamus that often occurs with surgical removal/radiation therapy of tumours in the hypothalamic region, such as craniopharyngioma. There is currently no rigorously studied pharmaceutical treatment for the intractable weight gain and cardiometabolic consequences that occur in patients with HIAO. We aimed to assess efficacy, safety and tolerability of beloranib treatment for 4 to 8 weeks in patients with HIAO. MATERIALS AND METHODS: This Phase 2a, double-blind, placebo-controlled study included 14 patients with HIAO, randomized to receive beloranib 1.8 mg or placebo subcutaneously twice weekly for 4 weeks with an optional 4-week open-label extension in which all patients received beloranib. The primary endpoint was change in weight from baseline to Week 4. RESULTS: Participants were 64% female, with a mean (SD) age of 32 (9) years, BMI of 43 (7) kg/m2 and weight of 126 (22) kg. Compared with placebo (N = 4), beloranib 1.8 mg (N = 8) resulted in a mean (95% CI) difference in weight of -3.2 (-5.4, -0.9) kg after 4 weeks. Weight loss continued through the 8 weeks in patients randomized to beloranib (mean -6.2 [-8.2, -4.1] kg). Beloranib treatment was associated with improvements in high-sensitivity CRP. Adverse events were mild to moderate. No patients who received beloranib discontinued treatment. CONCLUSION: Beloranib treatment resulted in progressive weight loss in patients with HIAO that was comparable to that observed with beloranib in patients with exogenous obesity. These findings indicate a novel mechanism for treating obesity in patients with HIAO.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Appetite Depressants/therapeutic use , Cardiovascular Diseases/prevention & control , Cinnamates/therapeutic use , Cyclohexanes/therapeutic use , Epoxy Compounds/therapeutic use , Glycoproteins/antagonists & inhibitors , Hypothalamus/injuries , Metabolic Syndrome/prevention & control , Obesity, Morbid/drug therapy , Sesquiterpenes/therapeutic use , Adult , Aminopeptidases/metabolism , Appetite Depressants/administration & dosage , Appetite Depressants/adverse effects , Biomarkers/blood , Body Mass Index , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cinnamates/administration & dosage , Cinnamates/adverse effects , Cohort Studies , Cyclohexanes/administration & dosage , Cyclohexanes/adverse effects , Double-Blind Method , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Epoxy Compounds/administration & dosage , Epoxy Compounds/adverse effects , Female , Follow-Up Studies , Glycoproteins/metabolism , Humans , Injections, Subcutaneous , Male , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Methionyl Aminopeptidases , Obesity, Morbid/blood , Obesity, Morbid/etiology , Obesity, Morbid/physiopathology , Proof of Concept Study , Risk , Sesquiterpenes/administration & dosage , Sesquiterpenes/adverse effects , Weight Loss/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL