Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 224(7): 1179-1186, 2021 10 13.
Article in English | MEDLINE | ID: mdl-32492704

ABSTRACT

BACKGROUND: Although effective, some oral pre-exposure prophylaxis (PrEP) users face barriers to adherence using daily pills, which could be reduced by long-acting formulations. Long-acting cabotegravir (CAB LA) is a potential new injectable formulation for human immunodeficiency virus (HIV) PrEP being tested in phase III trials. METHODS: We use a mathematical model of the HIV epidemic in South Africa to simulate CAB LA uptake by population groups with different levels of HIV risk. We compare the trajectory of the HIV epidemic until 2050 with and without CAB LA to estimate the impact of the intervention. RESULTS: Delivering CAB LA to 10% of the adult population could avert more than 15% of new infections from 2023 to 2050. The impact would be lower but more efficient if delivered to populations at higher HIV risk: 127 person-years of CAB LA use would be required to avert one HIV infection within 5 years if used by all adults and 47 person-years if used only by the highest risk women. CONCLUSIONS: If efficacious, a CAB LA intervention could have a substantial impact on the course of the HIV epidemic in South Africa. Uptake by those at the highest risk of infection, particularly young women, could improve the efficiency of any intervention.


Subject(s)
Anti-HIV Agents/administration & dosage , Diketopiperazines/therapeutic use , HIV Infections/drug therapy , HIV Infections/prevention & control , Pre-Exposure Prophylaxis , Pyridones/therapeutic use , Adult , Anti-HIV Agents/therapeutic use , Female , HIV Infections/epidemiology , Humans , Male , Middle Aged , Models, Theoretical , South Africa/epidemiology
2.
Molecules ; 26(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34885762

ABSTRACT

The long-acting parenteral formulation of the HIV integrase inhibitor cabotegravir (GSK744) is currently being developed to prevent HIV infections, benefiting from infrequent dosing and high efficacy. The crystal structure can affect the bioavailability and efficacy of cabotegravir. However, the stability determination of crystal structures of GSK744 have remained a challenge. Here, we introduced an ab initio protocol to determine the stability of the crystal structures of pharmaceutical molecules, which were obtained from crystal structure prediction process starting from the molecular diagram. Using GSK744 as a case study, the ab initio predicted that Gibbs free energy provides reliable further refinement of the predicted crystal structures and presents its capability for becoming a crystal stability determination approach in the future. The proposed work can assist in the comprehensive screening of pharmaceutical design and can provide structural predictions and stability evaluation for pharmaceutical crystals.


Subject(s)
Diketopiperazines/chemistry , HIV Infections/drug therapy , HIV Integrase Inhibitors/chemistry , HIV-1/drug effects , Pyridones/chemistry , Anti-HIV Agents/chemistry , Anti-HIV Agents/therapeutic use , Crystallography, X-Ray , Diketopiperazines/therapeutic use , HIV Infections/genetics , HIV Infections/virology , HIV Integrase Inhibitors/therapeutic use , HIV-1/genetics , HIV-1/ultrastructure , Humans , Pyridones/therapeutic use , Quantum Theory
3.
Bioorg Med Chem Lett ; 30(19): 127440, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32730945

ABSTRACT

The in vitro conversion of (1S,3S)-1-dimethoxylethyl-1,2,3,4-tetrahydro-ß-carboline-3-carboxylic acid, (1S,3S)-DCCA, in rat plasma is monitored by HPLC-FT-ICR-MS. We show that the in vitro conversion of (1S,3S)-DCCA in rat plasma for 1 h leads to forming (6S/12aS)-bisdimethoxyethylheptachpyridone, reflecting intermolecular condensation of (1S,3S)-DCCA, and the in vitro conversion of (6S/12aS)-bisdimethoxyethylheptachpyridone in rat plasma for 1 h leads to forming (6S/12aS)-heptachpyridone, reflecting hydrolysis of (6S/12aS)-bisdimethoxyethylheptachpyridone. At a dose of 1.0 µmol/kg (6S/12aS)-heptachpyridone orally inhibits venous thrombosis and arterial thrombosis in vivo. Bleeding time, clotting time and international normalized ratio show that at this dose (6S/12aS)-heptachpyridone has no bleeding risk, does not lengthen clotting time and does not change the exogenous coagulation pathway. We also show that the reactions promoted by rat plasma are easy to practice by chemical synthesis. Thus our findings build a bridge across the in vivo conversion and the application.


Subject(s)
Carbazoles/therapeutic use , Diketopiperazines/therapeutic use , Fibrinolytic Agents/therapeutic use , Venous Thrombosis/drug therapy , Animals , Blood/metabolism , Carbazoles/chemical synthesis , Carbazoles/metabolism , Diketopiperazines/chemical synthesis , Diketopiperazines/metabolism , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/metabolism , Hydrolysis , Male , Rats, Sprague-Dawley , Vena Cava, Inferior/drug effects
4.
Bioorg Med Chem ; 27(9): 1836-1844, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30910474

ABSTRACT

MBRI-001, a deuterium-substituted plinabulin derivative, has been reported to have better pharmacokinetic and similar antitumor effects in comparison with plinabulin. In this approach, we further carried out its polymorphs, co-crystal structure of MBRI-001-tubulin and tubulin inhibition study. Among the different polymorphs, Form F (MBRI-001/H2O) was prepared and evaluated, which had better physical stability and suitable process for scale-up production. Co-crystal structure of MBRI-001-tubulin (PDB:5XI5) was prepared and analyzed. The result of tubulin polymerization assay demonstrated that MBRI-001 could inhibit tubulin polymerization which was similar as plinabulin. Subsequently, the anti-proliferative activities of plinabulin and MBRI-001 were evaluated against two different human lung cancer cell lines. In vivo study, MBRI-001 revealed similar antitumor inhibition in comparison with plinabulin in A549 xenograft tumor model. Therefore, we suggested that MBRI-001 could be developed as a promising anti-cancer agent in near future.


Subject(s)
Diketopiperazines/chemistry , Tubulin Modulators/chemistry , Tubulin/chemistry , Animals , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Deuterium/chemistry , Diketopiperazines/metabolism , Diketopiperazines/pharmacology , Diketopiperazines/therapeutic use , Humans , Mice , Mice, Nude , Molecular Conformation , Molecular Dynamics Simulation , Neoplasms/drug therapy , Protein Structure, Tertiary , Tubulin/metabolism , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use
6.
Bioorg Med Chem ; 26(16): 4687-4692, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30119994

ABSTRACT

MBRI-001 was demonstrated preliminary better pharmacokinetics and antitumor effects than that of plinabulin in vivo. In this approach, we further carried out systematic pharmacokinetic and pharmacodynamic study of MBRI-001 in vitro and in vivo. MBRI-001 was tested stable in rat plasma and more stable in liver microsomes than plinabulin in vitro. In vivo, MBRI-001 could be distributed rapidly and widely in various tissues, especially the concentration of MBRI-001 in lung was remarkably higher than other tissues. Excretion study indicated that MBRI-001 might been decomposed and excreted as metabolites. Additionally, the combination treatment of MBRI-001 and gefitinib revealed better antitumor inhibition rate than monotherapy in vivo. Therefore, we suggest that MBRI-001 could be developed as a promising anti-cancer agent in near future.


Subject(s)
Antineoplastic Agents/chemistry , Diketopiperazines/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Blood Proteins/chemistry , Blood Proteins/metabolism , Cell Line, Tumor , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Deuterium/chemistry , Diketopiperazines/metabolism , Diketopiperazines/pharmacokinetics , Diketopiperazines/therapeutic use , Female , Half-Life , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Protein Binding , Rats , Rats, Wistar , Tissue Distribution
7.
Xenobiotica ; 48(5): 467-477, 2018 May.
Article in English | MEDLINE | ID: mdl-28485193

ABSTRACT

1. Breast cancer resistance protein (BCRP) plays an important role in drug absorption, distribution and excretion. It is challenging to evaluate BCRP functions in preclinical models because commonly used BCRP inhibitors are nonspecific or unstable in animal plasma. 2. In this work, in vitro absorption, distribution, metabolism and elimination (ADME) assays and pharmacokinetic (PK) experiments in Bcrp knockout (KO) (Abcg2-/-) and wild-type (WT) FVB mice and Wistar rats were conducted to characterize the preclinical properties of a novel selective BCRP inhibitor (ML753286, a Ko143 analog). 3. ML753286 is a potent inhibitor for BCRP, but not for P-glycoprotein (P-gp), organic anion-transporting polypeptide (OATP) or major cytochrome P450s (CYPs). It has high permeability, but is not an efflux transporter substrate. ML753286 has low to medium clearance in rodent and human liver S9 fractions, and is stable in plasma cross species. Bcrp inhibition affects oral absorption and clearance of sulfasalazine in rodents. A single dose of ML753286 at 50-300 mg/kg orally, and at 20 mg/kg intravenously or 25 mg/kg orally inhibits Bcrp functions in mice and rats, respectively. 4. These findings confirm that ML753286 is a useful selective inhibitor to evaluate BCRP/Bcrp activity in vitro and in rodent model systems.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Absorption, Physiological , Breast Neoplasms/drug therapy , Diketopiperazines/pharmacokinetics , Diketopiperazines/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Caco-2 Cells , Cell Membrane Permeability/drug effects , Diketopiperazines/blood , Diketopiperazines/chemistry , Dogs , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Macaca fascicularis , Male , Mice, Knockout , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Neoplasm Proteins/metabolism , Rats , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use , Time Factors
9.
Toxicol Appl Pharmacol ; 305: 194-202, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27312871

ABSTRACT

PURPOSE: Vascular smooth muscle cell (VSMC) proliferation plays a critical role in the pathogenesis of atherosclerosis and restenosis. This study investigated piperazinedione derived compound TW-01-mediated inhibitory effects on VSMC proliferation and intimal hyperplasia. METHODS: Cell proliferation was determined using [(3)H]-thymidine incorporation and MTT assay; cell cycle distribution was measured using flow cytometry; proteins and mRNA expression were determined using western blotting and RT-PCR analyses; DNA binding activity of nuclear factor-κB (NF-κB), as measured using enzyme-linked immunosorbent assays (ELISA); in vivo effects of TW-01 were determined using balloon angioplasty in the rat. RESULTS: TW-01 significantly inhibited cell proliferation. At the concentrations used, no cytotoxic effects were observed. Three predominant signaling pathways were inhibited by TW-01: (a) extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein kinase (MAPK) activation and its downstream effectors of c-fos, c-jun, and c-myc; (b) DNA binding activity of nuclear factor-κB (NF-κB); and, (c) Akt/protein kinase B (PKB) and cell cycle progression. Furthermore, TW-01 also inhibited Ras activation, a shared upstream event of each of these signaling cascades. In vascular injury studies, oral administration of TW-01 significantly suppressed intimal hyperplasia induced by balloon angioplasty. CONCLUSION: The present study suggests that TW-01 might be a potential candidate for atherosclerosis treatment.


Subject(s)
Angioplasty, Balloon/adverse effects , Cell Proliferation/drug effects , Coronary Restenosis/drug therapy , Diketopiperazines/therapeutic use , Hyperplasia/drug therapy , Pyridines/therapeutic use , Animals , Carotid Artery, Common/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Diketopiperazines/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Pyridines/pharmacology , Rats, Wistar , Tunica Intima/drug effects , Tunica Intima/pathology , ras Proteins/antagonists & inhibitors
10.
Med Res Rev ; 35(5): 937-67, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25850821

ABSTRACT

Although fungi produce highly structurally diverse metabolites, many of which have served as excellent sources of pharmaceuticals, no fungi-derived agent has been approved as a cancer drug so far. This is despite a tremendous amount of research being aimed at the identification of fungal metabolites with promising anticancer activities. This review discusses the results of clinical testing of fungal metabolites and their synthetic derivatives, with the goal to evaluate how far we are from an approved cancer drug of fungal origin. Also, because in vivo studies in animal models are predictive of the efficacy and toxicity of a given compound in a clinical situation, literature describing animal cancer testing of compounds of fungal origin is reviewed as well. Agents showing the potential to advance to clinical trials are also identified. Finally, the technological challenges involved in the exploitation of fungal biodiversity and procurement of sufficient quantities of clinical candidates are discussed, and potential solutions that could be pursued by researchers are highlighted.


Subject(s)
Antineoplastic Agents/chemistry , Fungi/chemistry , Neoplasms/drug therapy , Androstadienes/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Aphidicolin/therapeutic use , Biological Products/chemistry , Clinical Trials as Topic , Cyclohexanes/therapeutic use , Diketopiperazines/therapeutic use , Disease Models, Animal , Drug Design , Drug Resistance, Neoplasm , Fatty Acids, Unsaturated/therapeutic use , Female , Humans , Macrolides/therapeutic use , Male , Mice , Polycyclic Sesquiterpenes , Sesquiterpenes/therapeutic use , Trichothecenes/therapeutic use , Wortmannin
11.
Biol Pharm Bull ; 37(3): 378-86, 2014.
Article in English | MEDLINE | ID: mdl-24583858

ABSTRACT

In a recent study, we isolated the diketopiperazine disulfide glionitrin A from the co-culture broth of a mine drainage-derived fungus (Aspergillus fumigatus KMC901) and bacterium (Sphingomonas KMK001). Here, we investigated the antitumor activity of glionitrin A and its underlying molecular mechanisms in human prostate cancer DU145 cells. Glionitrin A showed significant cytotoxicity, promoting cell cycle arrest and apoptosis. Glionitrin A-treated cells exhibited elevated levels of phospho-histone 2AX (Ser139), a marker of DNA damage, and accumulated in both S phase and G2/M phase due to the activation of checkpoints associated with the ataxia-telangiectasia-mutated and ataxia-telangiectasia-mutated-Rad3-related Chk1/2 pathway downstream of p53-binding protein 1 phosphorylation at Ser1778. In addition, glionitrin A induced apoptosis through both caspase-dependent and -independent pathways. Glionitrin A activated caspase-8, -9 and -3 and also released endonuclease G from the mitochondria to the nucleus in a dose-dependent manner. Our in vivo study performed in nude mice bearing xenografts of DU145 cells showed that glionitrin A dramatically reduced the tumor volume by an average of 38.2% (5 mg/kg, per os (p.o.)) and 71.3% (10 mg/kg, p.o.) at 27 d after the beginning of treatment. Taken together, these findings provide a detailed description of the mechanism underlying the biological activity of the new natural product glionitrin A, which has the potential to be developed as an anti-prostate cancer agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/metabolism , Biological Products/therapeutic use , Checkpoint Kinase 2/metabolism , Diketopiperazines/therapeutic use , Prostatic Neoplasms/drug therapy , Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Aspergillus fumigatus , Biological Products/pharmacology , Caspase 1/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , Diketopiperazines/pharmacology , Endodeoxyribonucleases/metabolism , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Prostatic Neoplasms/metabolism , Sphingomonas , Tumor Suppressor p53-Binding Protein 1
12.
Int J Mol Sci ; 15(9): 16911-35, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25250910

ABSTRACT

Synthesis and anticonvulsant potential of certain new 6-aryl-9-substituted-6,9-diazaspiro[4.5]decane-8,10-diones (6a-l) and 1-aryl-4-substituted-1,4-diazaspiro[5.5] undecane-3,5-diones (6m-x) are reported. The intermediates 1-[(aryl)(cyanomethyl)amino] cycloalkanecarboxamides (3a-f) were prepared via adopting Strecker synthesis on the proper cycloalkanone followed by partial hydrolysis of the obtained nitrile functionality and subsequent N-cyanomethylation. Compounds 3a-f were subjected to complete nitrile hydrolysis to give the respective carboxylic acid derivatives 4a-f which were cyclized under mild conditions to give the spiro compounds 5a-f. Ultimately, compounds 5a-f were alkylated or aralkylated to give the target compounds 6a-i and 6m-u. On the other hand, compounds 6j-l and 6v-x were synthesized from the intermediates 5a-f through alkylation, dehydration and finally tetrazole ring formation. Anticonvulsant screening of the target compounds 6a-x revealed that compound 6g showed an ED50 of 0.0043 mmol/kg in the scPTZ screen, being about 14 and 214 fold more potent than the reference drugs, Phenobarbital (ED50 = 0.06 mmol/kg) and Ethosuximide (ED50 = 0.92 mmol/kg), respectively. Compound 6e exhibited an ED50 of 0.019 mmol/kg, being about 1.8 fold more potent than that of the reference drug, Diphenylhydantoin (ED50 = 0.034 mmol/kg) in the MES screen. Interestingly, all the test compounds 6a-x did not show any minimal motor impairment at the maximum administered dose in the neurotoxicity screen.


Subject(s)
Anticonvulsants/chemical synthesis , Diketopiperazines/chemical synthesis , Spiro Compounds/chemical synthesis , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Convulsants/toxicity , Diketopiperazines/chemistry , Diketopiperazines/pharmacology , Diketopiperazines/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electroshock , Male , Mice , Molecular Structure , Pentylenetetrazole/toxicity , Random Allocation , Rotarod Performance Test , Seizures/chemically induced , Seizures/etiology , Seizures/prevention & control , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Structure-Activity Relationship
13.
Lung Cancer ; 195: 107932, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39173229

ABSTRACT

BACKGROUND: Plinabulin is a GEF-H1 releasing agent with an immune-enhancing function. We report results from a multicenter Phase I/II study (NCT03575793) assessing plinabulin in combination with nivolumab and ipilimumab for the treatment of recurrent SCLC. METHODS: In Phase I, patients were enrolled using a 3 + 3 design to determine dose-limiting toxicities (DLTs) and recommended Phase 2 dose (RP2D). Patients received nivolumab (1 mg/kg), ipilimumab (3 mg/kg), and plinabulin (in escalating doses) on day 1 of each 21-day cycle for 4 cycles followed by maintenance with plinabulin and nivolumab. In phase II, patients with recurrent PD(L)1 inhibitor resistant SCLC were enrolled. The primary objective was median progression-free survival (PFS). RESULTS: Between 9/2018 and 2/2023, 39 patients were enrolled, and 36 patients received study treatment and were evaluable for safety (16 in Phase I; 20 in Phase II). In the phase I dose-escalation, there were 2 DLTs; grade 3 altered mental status lasting <24 h and grade 3 infusion reaction. The Plinabulin RP2D was determined to be 30 mg/m2. Common TRAEs were vomiting (44 %), nausea (42 %), and infusion reaction (36 %); 6 % of patients had a ≥grade 3 TRAE. Five patients (14 %) had ≥grade 3 irAEs; there were no cases of immune-related pneumonitis. In the efficacy analysis in 27 patients, the median PFS was 1.6 months (95 % CI 1.2 to 2.7) and the trial did not meet the pre-specified target median PFS of 3.5 months. Four patients treated at 30 mg/m2 had PR (confirmed 1, unconfirmed 3); 5 patients had SD with a CBR of 33 %. Two of 8 patients treated in phase I at the lower 20 mg/m2 dose had confirmed PR, with 1 patient on the drug regimen for >90 cycles. The median OS and follow-up time were 5.5 months and 2.5 months respectively. CONCLUSIONS: Plinabulin in combination with nivolumab and ipilimumab was tolerable at the dose of 30 mg/m2. While the clinical responses in PD-1 resistant SCLC were limited, some patients had a long duration of response. The number of ≥grade 3 irAE with the combination were lower than expected.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Ipilimumab , Lung Neoplasms , Nivolumab , Small Cell Lung Carcinoma , Humans , Ipilimumab/administration & dosage , Ipilimumab/therapeutic use , Ipilimumab/adverse effects , Male , Nivolumab/administration & dosage , Nivolumab/therapeutic use , Nivolumab/adverse effects , Female , Middle Aged , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/mortality , Aged , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Adult , Neoplasm Recurrence, Local/drug therapy , Aged, 80 and over , Diketopiperazines/administration & dosage , Diketopiperazines/therapeutic use
14.
J Sex Med ; 10(10): 2506-17, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23937679

ABSTRACT

AIM: To assess the efficacy and safety of the selective oxytocin receptor antagonist epelsiban in the treatment of premature ejaculation (PE). METHODS: Double-blind, randomized, parallel-group, placebo-controlled, stopwatch-monitored, phase 2, multicenter study (GSK557296; NCT01021553) conducted in men (N=77) 18-55 years of age, with PE defined as per International Society for Sexual Medicine consensus definition. Patients provided informed consent prior to a 4-week un-medicated run-in to determine baseline intravaginal ejaculatory latency times (IELT) recorded in an electronic diary. Patients needed to make a minimum of four intercourse attempts and have a mean IELT<65 seconds to be considered for randomization. Men with moderate-to-severe erectile dysfunction were excluded from the study. Eligible patients were randomized to placebo, epelsiban 50 mg, or 150 mg, taken 1 hour before sexual activity. Active treatment IELT times were recorded in an electronic diary, along with subjective measures of intercourse satisfaction, over an 8-week treatment period. The Modified Index of Premature Ejaculation and International Index of Erectile Function were completed at study visits. MAIN OUTCOME MEASURES: Stopwatch timed IELT recordings and a modified version of the patient-reported outcome questionnaire the IPE were used in this study to determine the effect of epelsiban when taken orally prior to intercourse in subjects diagnosed with PE. RESULTS: The baseline (mean) IELT for patients pretreatment was (0.52, 0.63, and 0.59 minutes) for placebo, epelsiban 50 mg and 150 mg, respectively. On-treatment, average geometric least squares means of the median IELT values (mean) were slightly higher in the 50 mg and 150 mg groups (0.72 and 0.69 minutes), respectively, vs. the placebo group (0.62 minutes). Headache was the most common adverse event, and rates were similar across all groups. CONCLUSIONS: Epelsiban 50 mg and 150 mg were well tolerated, but did not result in a clinically or statistically significant change in IELT in men with PE, compared with placebo.


Subject(s)
Diketopiperazines/therapeutic use , Ejaculation/drug effects , Hormone Antagonists/therapeutic use , Morpholines/therapeutic use , Premature Ejaculation/drug therapy , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Diketopiperazines/adverse effects , Diketopiperazines/pharmacokinetics , Double-Blind Method , Genotype , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Least-Squares Analysis , Male , Middle Aged , Morpholines/adverse effects , Morpholines/pharmacokinetics , Netherlands , Patient Satisfaction , Pharmacogenetics , Premature Ejaculation/diagnosis , Premature Ejaculation/metabolism , Premature Ejaculation/physiopathology , Premature Ejaculation/psychology , Reaction Time , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Sexual Behavior/drug effects , Surveys and Questionnaires , Time Factors , Treatment Outcome , United States , Young Adult
15.
Org Biomol Chem ; 10(47): 9501-6, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23111956

ABSTRACT

Effusin A (1), a spirobicyclic N,O-acetal derivative with an unprecedented 3',3a',5',6'-tetrahydrospiro[piperazine-2,2'-pyrano[2,3,4-de]chromene] ring system, and a spiro-polyketide-diketopiperazine hybrid dihydrocryptoechinulin D (2) were isolated from a mangrove rhizosphere soil derived fungus, Aspergillus effuses H1-1. Their structures were determined by detailed spectroscopic analysis. Effusin A (1) and dihydrocryptoechinulin D (2) occurred as racemates, the enantiomers of which were separated and characterized by online HPLC-ECD analysis and their absolute configurations were determined by the solution TDDFT ECD calculation approach. The cytotoxic effects of 1 and 2 were preliminarily evaluated and 2 showed potent activity on P388 cells with an IC(50) value of 1.83 µM. The target of racemic 2 was also investigated and the (12R,28S,31S)-2 enantiomer showed selectivity against topoisomerase I.


Subject(s)
Aspergillus/chemistry , Diketopiperazines/chemistry , Rhizosphere , Soil , Cell Line, Tumor , Cell Proliferation/drug effects , Diketopiperazines/pharmacokinetics , Diketopiperazines/therapeutic use , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Soil/chemistry , Wetlands
16.
Curr Opin HIV AIDS ; 17(4): 186-191, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35762372

ABSTRACT

PURPOSE OF REVIEW: Tenofovir-based oral PrEP has been effective in reducing population-level HIV incidence in multiple settings, although disparities remain. Injectable cabotegravir-based PrEP is an alternative that may be attractive to individuals with adherence challenges or who do not desire to take a daily medication. We review promises and challenges of cabotegravir-based PrEP. RECENT FINDINGS: Cabotegravir has demonstrated higher effectiveness than oral PrEP in two randomized trials, with a hazard ratio of 0.31 for HIV incidence among MSM and transgender women across multiple settings [95% confidence interval (CI) 0.18-0.62] and 0.11 for cisgender women in sub-Saharan Africa (95% CI 0.040.32). Cabotegravir was also highly effective among populations with disproportionate HIV incidence. Although cabotegravir breakthrough was rare, diagnosis was delayed with use of antigen/antibody-based HIV tests, and resistance occurred with breakthrough infections. Implementation will need to overcome several challenges, including HIV RNA laboratory monitoring not being widely available, requirement for additional staff time and clinic space, and need to provide oral medication during interruptions in dosing. SUMMARY: Cabotegravir-based PrEP is a highly effective additional PrEP option that will expand HIV prevention options. For successful roll-out, strategies for streamlined and accessible delivery of cabotegravir in real-world settings will need to be developed.


Subject(s)
Diketopiperazines , HIV Infections , HIV Integrase Inhibitors , Pre-Exposure Prophylaxis , Pyridones , Sexual and Gender Minorities , Diketopiperazines/therapeutic use , Female , HIV Infections/drug therapy , HIV Integrase Inhibitors/therapeutic use , Homosexuality, Male , Humans , Male , Pyridones/therapeutic use
17.
JAMA Netw Open ; 5(1): e2145446, 2022 01 04.
Article in English | MEDLINE | ID: mdl-35084480

ABSTRACT

Importance: Prevention of chemotherapy-induced neutropenia (CIN) and its clinical consequences is an unmet need for which plinabulin, a selective immunomodulating microtubule-binding agent, is being tested. Objective: To demonstrate noninferiority between plinabulin and pegfilgrastim for days of severe neutropenia in cycle 1 in patients with solid tumors treated with docetaxel. Design, Setting, and Participants: The Plinabulin vs Pegfilgrastim for the Prevention of Docetaxel-Induced Neutropenia in Patients With Solid Tumors (PROTECTIVE-1) double-blind phase 3 randomized clinical trial was performed in multiple centers in China, Russia, Ukraine, and the US. Participants included patients with breast, prostate, or non-small cell lung cancer treated with single-agent docetaxel chemotherapy. Data were collected from June 1, 2018, to January 31, 2019. The database was locked on February 18, 2021. Data analysis was based on intention to treat and safety and performed from October 5, 2018, to February 23, 2021. Interventions: Plinabulin, 40 mg, plus placebo or pegfilgrastim, 6 mg, plus placebo. Main Outcomes and Measures: The primary end point was day of severe neutropenia in cycle 1. Additional end points included clinical consequences of CIN (febrile neutropenia, hospitalizations, infections, antibiotic use, and modifications of chemotherapy dose), patient-reported outcomes for bone pain score, markers for immune suppression (neutrophil-to-lymphocyte ratio [NLR] of >5), immature neutrophils (band, promyelocyte, and myelocyte counts >0), and safety. Results: Among the 105 patients included in the analysis (65 [6.19%] women; median age, 59 [range, 31-81] years), the primary end point was met within a noninferiority margin of 0.65 days, with a mean difference of 0.52 days (98.52% CI, 0.40-0.65 days). Grade 4 neutropenia frequency in cycle 1 was not significantly different. Plinabulin had earlier onset of action with less grade 4 neutropenia in week 1 of cycle 1. Plinabulin had fewer adverse clinical consequences with rates of febrile neutropenia (0 of 52 vs 1 of 53 [1.9%]), infections (4 of 52 [7.7%] vs 8 of 53 [15.1%]), chemotherapy dose delay of more than 7 days (2 of 52 [3.8%] vs 3 of 53 [5.7%]), and permanent chemotherapy discontinuation (7 of 52 [13.5%] vs 14 of 53 [26.4%]). Patients receiving plinabulin had significantly less bone pain (difference, -0.67 [95% CI, -1.17 to -0.16]; P = .01) and a better immunosuppressive profile (NLR >5 at day 8, 2 of 52 [3.8%] vs 24 of 51 [46.0%]; P < .001). Plinabulin was well tolerated, with comparable safety to pegfilgrastim. Conclusions and Relevance: Plinabulin has comparable efficacy to pegfilgrastim for the prevention of CIN, with better safety and a better immunosuppressive profile. Plinabulin's same-day dosing compared with pegfilgrastim's next-day dosing offers distinct advantages, including reducing use of health care services. Trial Registration: ClinicalTrials.gov Identifier: NCT03102606.


Subject(s)
Antineoplastic Agents/adverse effects , Diketopiperazines/therapeutic use , Docetaxel/adverse effects , Filgrastim/therapeutic use , Neoplasms/drug therapy , Neutropenia/prevention & control , Polyethylene Glycols/therapeutic use , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Double-Blind Method , Female , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Neutropenia/chemically induced , Prostatic Neoplasms/drug therapy , Treatment Outcome
18.
Med Res Rev ; 31(4): 576-604, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20027670

ABSTRACT

A short, efficient and highly stereoselective synthesis has been developed for a series of 6-indanyl-3-alkyl-7-aryl/heterocyclic-(3R, 6R, 7R)-2, 5-diketopiperazine amides that are potent and selective oxytocin (OT) antagonists. Property-based design using an estimate of human oral absorption enabled focus to be directed to those templates with the greatest chance of delivering high bioavailability in humans. This led to the 2', 4'-difluorophenyl dimethylamide 40, a highly potent (pK(i) =9.2) and selective OT antagonist (>1,000-fold selectivity vs. the human vasopressin receptors V1a, V2, and V1b) with good oral bioavailability (>50%) in the rat and dog. Increased solubility and an improved Cyp450 profile was achieved with a range of 2'-substituted 7-(1',3'-oxazol-4'-yl)-(3R,6R,7R)-2,5-diketopiperazine amides and branching at the α-carbon of the 3-butyl group led to a superior rat pharmacokinetic profile that resulted in the discovery of the 2'-methyl-1',3'-oxazol-4'-yl morpholine amide derivative 74 GSK221149A (Retosiban), which had the best oral exposure and bioavailability in the rat. Retosiban has sub-nanomolar affinity (K(i) =0.65 nM) for the oxytocin receptor with >1400-fold selectivity over the closely related vasopressin receptors. It has good solubility, low protein binding and has a good Cyp450 profile with no significant inhibition IC(50) >100 µM. Retosiban is >15-fold more potent at the human oxytocin receptor than atosiban (a marketed i.v, peptide OT antagonist) and it has been shown to be an effective tocolytic by i.v. and by oral administration in rats, and was selected for progression as a potential clinical candidate for preterm labor.


Subject(s)
Diketopiperazines/pharmacokinetics , Diketopiperazines/therapeutic use , Drug Design , Obstetric Labor, Premature/drug therapy , Oxytocin/antagonists & inhibitors , Administration, Oral , Biological Availability , Diketopiperazines/administration & dosage , Diketopiperazines/chemistry , Female , Humans , Oxytocin/metabolism , Pregnancy
19.
J Appl Microbiol ; 110(1): 304-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21122037

ABSTRACT

AIMS: Research is to identify the bioactive secondary metabolites produced by Aspergillus sp. KMD 901 isolated from marine sediment and to assess their apoptosis-inducing effects. METHODS AND RESULTS: Aspergillus sp. KMD 901 was isolated from marine sediment obtained from the East Sea of Korea. An ethyl acetate extract of KMD 901 exhibited potent cytotoxic activity towards five cancer cell lines (HCT116, AGS, A549, MCF-7 and HepG2). Sequencing of the internal transcribed spacer (ITS) region in this strain allowed us to identify KMD 901 as a strain of Aspergillus versicolor. The cytotoxic compounds from Aspergillus sp. KMD 901 were purified by reversed-phase high-performance liquid chromatography and identified as diketopiperazine disulfides through spectroscopic analyses including extensive 2D NMR and mass spectrometry. The diketopiperazine disulfides were found to induce apoptosis in HCT116 cells based on cell morphology, DNA fragmentation observed by agarose gel electrophoresis, Annexin-V/PI staining using a flow cytometer and cleavage of poly (ADP-ribose) polymerase (PARP), caspase-3, caspase-8, caspase-9 and Bcl-2 family proteins (Bcl-2, Bcl-xL and Bax) using Western blotting analysis. Further study using an in vivo xenograft model showed inhibitory effects of acetylapoaranotin (2) on tumour proliferation. CONCLUSION: A new diketopiperazine disulfide, deoxyapoaranotin (3), along with previously described acetylaranotin (1) and acetylapoaranotin (2) was separated from Aspergillus sp. KMD 901 and found to have direct cytotoxic and apoptosis-inducing effects towards HCT116 colon cancer cell lines. SIGNIFICANCE AND IMPACT OF THE STUDY: These results suggest that the diketopiperazine disulfides produced from Aspergillus sp., KMD 901, could be candidates for the development of apoptosis-inducing antitumour agents. Also, this study indicates that marine natural products as potential source of pharmaceuticals.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis , Aspergillus/metabolism , Diketopiperazines/toxicity , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Aspergillus/isolation & purification , Caspases/metabolism , Cell Line, Tumor , Diketopiperazines/chemistry , Diketopiperazines/metabolism , Diketopiperazines/therapeutic use , Disulfides/chemistry , Disulfides/metabolism , Disulfides/therapeutic use , Disulfides/toxicity , Geologic Sediments/microbiology , HCT116 Cells , Humans , Mice , Mice, Nude , Oceans and Seas , Oxepins/chemistry , Oxepins/metabolism , Oxepins/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
20.
Biomolecules ; 11(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34680148

ABSTRACT

Cyclic dipeptides, also know as diketopiperazines (DKP), the simplest cyclic forms of peptides widespread in nature, are unsurpassed in their structural and bio-functional diversity. DKPs, especially those containing proline, due to their unique features such as, inter alia, extra-rigid conformation, high resistance to enzyme degradation, increased cell permeability, and expandable ability to bind a diverse of targets with better affinity, have emerged in the last years as biologically pre-validated platforms for the drug discovery. Recent advances have revealed their enormous potential in the development of next-generation theranostics, smart delivery systems, and biomaterials. Here, we present an updated review on the biological and structural profile of these appealing biomolecules, with a particular emphasis on those with anticancer properties, since cancers are the main cause of death all over the world. Additionally, we provide a consideration on supramolecular structuring and synthons, based on the proline-based DKP privileged scaffold, for inspiration in the design of compound libraries in search of ideal ligands, innovative self-assembled nanomaterials, and bio-functional architectures.


Subject(s)
Diketopiperazines/chemistry , Dipeptides/chemistry , Neoplasms/drug therapy , Proline/chemistry , Diketopiperazines/therapeutic use , Dipeptides/genetics , Dipeptides/therapeutic use , Drug Discovery , Humans , Neoplasms/genetics , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Peptides, Cyclic/therapeutic use , Proline/genetics , Proline/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL