Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Drug Metab Dispos ; 49(9): 718-728, 2021 09.
Article in English | MEDLINE | ID: mdl-34135089

ABSTRACT

For drug development, species differences in drug-metabolism reactions present obstacles for predicting pharmacokinetics in humans. We characterized the species differences in hydrolases among humans and mice, rats, dogs, and cynomolgus monkeys. In this study, to expand the series of such studies, we attempted to characterize marmoset hydrolases. We measured hydrolase activities for 24 compounds using marmoset liver and intestinal microsomes, as well as recombinant marmoset carboxylesterase (CES) 1, CES2, and arylacetamide deacetylase (AADAC). The contributions of CES1, CES2, and AADAC to hydrolysis in marmoset liver microsomes were estimated by correcting the activities by using the ratios of hydrolase protein levels in the liver microsomes and those in recombinant systems. For six out of eight human CES1 substrates, the activities in marmoset liver microsomes were lower than those in human liver microsomes. For two human CES2 substrates and three out of seven human AADAC substrates, the activities in marmoset liver microsomes were higher than those in human liver microsomes. Notably, among the three rifamycins, only rifabutin was hydrolyzed by marmoset tissue microsomes and recombinant AADAC. The activities for all substrates in marmoset intestinal microsomes tended to be lower than those in liver microsomes, which suggests that the first-pass effects of the CES and AADAC substrates are due to hepatic hydrolysis. In most cases, the sums of the values of the contributions of CES1, CES2, and AADAC were below 100%, which indicated the involvement of other hydrolases in marmosets. In conclusion, we clarified the substrate preferences of hydrolases in marmosets. SIGNIFICANCE STATEMENT: This study confirmed that there are large differences in hydrolase activities between humans and marmosets by characterizing marmoset hydrolase activities for compounds that are substrates of human CES1, CES2, or arylacetamide deacetylase. The data obtained in this study may be useful for considering whether marmosets are appropriate for examining the pharmacokinetics and efficacies of new chemical entities in preclinical studies.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Hydrolases , Intestine, Small/enzymology , Liver/enzymology , Microsomes/enzymology , Rifamycins/pharmacokinetics , Animals , Callithrix , Carboxylesterase/metabolism , Drug Development/methods , Enzyme Activation/physiology , Enzyme Assays/methods , Humans , Hydrolases/classification , Hydrolases/metabolism , Recombinant Proteins/metabolism , Species Specificity , Substrate Specificity
2.
BJOG ; 125(7): 804-811, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29130574

ABSTRACT

BACKGROUND: Rifamycin antibiotics are commonly used for treatment of tuberculosis, but may reduce the effectiveness of hormonal contraception (HC). OBJECTIVES: To determine whether interactions between rifamycins and HC result in decreased effectiveness or increased toxicity of either therapy. SEARCH STRATEGY: We searched MEDLINE, Embase, Cochrane and clinicaltrials.gov through May 2017. SELECTION CRITERIA: We included trials, cohort, and case-control studies addressing pregnancy rates, pharmacodynamics or pharmacokinetic (PK) outcomes when HC and rifamycins were administered together versus apart. Of 7291 original records identified, 11 met inclusion criteria after independent review by two authors. DATA COLLECTION AND ANALYSIS: Two authors independently abstracted study details and assessed study quality using the United States Preventive Services Task Force grading system. Findings are reported descriptively. MAIN RESULTS: Studies only addressed combined oral contraceptives (COCs) and none reported pregnancy rates. Quality ranged from good to poor. Rifampin increased the frequency of ovulation in two of four studies, and reduced estrogen and/or progestin exposure in five studies. Rifabutin led to smaller PK changes than rifampin in two studies. In one study each, rifaximin and rifalazil did not alter hormone PK. CONCLUSIONS: No studies evaluated pregnancy risk or non-oral HCs. PK and ovulation outcomes support a clinically concerning drug interaction between COCs and rifampin, and to a lesser extent rifabutin. Data are limited for other rifamycins. TWEETABLE ABSTRACT: Rifampin and rifabutin reduce systemic exposure of oral contraceptives, but no studies have evaluated pregnancy risk.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Contraceptives, Oral, Combined/pharmacokinetics , Rifabutin/pharmacokinetics , Rifamycins/pharmacokinetics , Adult , Drug Interactions , Drug Therapy, Combination , Female , Humans , Middle Aged , Ovulation/drug effects , Pregnancy , Treatment Outcome
3.
Antimicrob Agents Chemother ; 60(8): 4860-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27270284

ABSTRACT

Rifapentine (RPT) is a rifamycin antimycobacterial and, as part of a combination therapy, is indicated for the treatment of pulmonary tuberculosis (TB) caused by Mycobacterium tuberculosis Although the results from a number of studies indicate that rifapentine has the potential to shorten treatment duration and enhance completion rates compared to other rifamycin agents utilized in antituberculosis drug regimens (i.e., regimens 1 to 4), its optimal dose and exposure in humans are unknown. To help inform such an optimization, a physiologically based pharmacokinetic (PBPK) model was developed to predict time course, tissue-specific concentrations of RPT and its active metabolite, 25-desacetyl rifapentine (dRPT), in humans after specified administration schedules for RPT. Starting with the development and verification of a PBPK model for rats, the model was extrapolated and then tested using human pharmacokinetic data. Testing and verification of the models included comparisons of predictions to experimental data in several rat tissues and time course RPT and dRPT plasma concentrations in humans from several single- and repeated-dosing studies. Finally, the model was used to predict RPT concentrations in the lung during the intensive and continuation phases of a current recommended TB treatment regimen. Based on these results, it is anticipated that the PBPK model developed in this study will be useful in evaluating dosing regimens for RPT and for characterizing tissue-level doses that could be predictors of problems related to efficacy or safety.


Subject(s)
Antitubercular Agents/pharmacokinetics , Rifampin/analogs & derivatives , Animals , Antibiotics, Antitubercular/pharmacokinetics , Antibiotics, Antitubercular/pharmacology , Antitubercular Agents/pharmacology , Drug Administration Schedule , Drug Therapy, Combination/methods , Humans , Lung/drug effects , Lung/microbiology , Mycobacterium tuberculosis/drug effects , Rats , Rifampin/pharmacokinetics , Rifampin/pharmacology , Rifamycins/pharmacokinetics , Rifamycins/pharmacology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology
4.
Antimicrob Agents Chemother ; 58(7): 4014-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798277

ABSTRACT

A randomized, double-blind study comparing single-dose chlamydia therapies of oral rifalazil (25 mg) and azithromycin (1 g) was conducted in 82 women with uncomplicated genital Chlamydia trachomatis infection. The microbiologic cure rate of C. trachomatis with rifalazil (n = 33) was 84.8% at the visit on day 22 to 26 (test-of-cure visit), versus 92.1% with azithromycin (n = 38), and the number of treatment failures in each group was 5 and 3, respectively. The difference in cure rate was -7.3%, with a lower limit of the 95% confidence interval (95% CI) of -22.5, and thus, noninferiority was not established at the prespecified margin (lower limit of CI of -15%). The overall treatment-emergent adverse event (TEAE) and treatment-related TEAE rates were lower in the rifalazil group (68% and 55%) than in the azithromycin group (71% and 62%), respectively. Subjects classified as treatment failures at day 22 to 26 had a lower mean plasma concentration of rifalazil at the visit on day 8 to 12 than those classified as treatment cures, but this difference was not significant; however, the levels were similar for both groups at the visit on day 22 to 26. A single 25-mg dose of rifalazil was well tolerated and eradicated C. trachomatis in most of these women with uncomplicated genital C. trachomatis infection. (The study was registered at clinicaltrials.gov under registration no. NCT01631201).


Subject(s)
Anti-Bacterial Agents/therapeutic use , Azithromycin/therapeutic use , Chlamydia Infections/drug therapy , Chlamydia trachomatis , Genital Diseases, Female/drug therapy , Rifamycins/therapeutic use , Adult , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Azithromycin/adverse effects , Azithromycin/pharmacokinetics , Chlamydia Infections/microbiology , Double-Blind Method , Endpoint Determination , Female , Genital Diseases, Female/microbiology , Humans , Rifamycins/adverse effects , Rifamycins/pharmacokinetics , Treatment Outcome , Young Adult
5.
Pharmacol Res ; 85: 39-44, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24836868

ABSTRACT

Rifaximin is an antibiotic, locally acting in the gastrointestinal tract, which may exist in different crystal as well as amorphous forms. The branded rifaximin formulation contains the polymorph rifaximin-α, whose systemic bioavailability is very limited. This study was performed to compare the pharmacokinetics of this formulation with that of a generic product, whose composition in terms of solid state forms of the active pharmaceutical ingredient was found to be different. Two tablets (2×200mg) of branded and generic formulations were given to 24 healthy volunteers of either sex, according to a single-blind, randomized, two-treatment, single-dose, two-period, cross-over design. Plasma and urinary samples were collected at preset times (for 24h or 48h, respectively) after dosing, and assayed for rifaximin concentrations by high-performance liquid chromatography-mass spectrometry. Rifaximin plasma and urine concentration-time profiles showed relevant differences when generic and branded rifaximin were compared. Most pharmacokinetic parameters were significantly higher after administration of generic rifaximin than after rifaximin-α. In particular, the differences for Cmax, AUC and cumulative urinary excretion between the generic formulation and the branded product ranged from 165% to 345%. The few adverse events recorded were not serious and not related to study medications. The results of the present investigation demonstrate different systemic bioavailability of generic and branded formulations of rifaximin. As a consequence, the therapeutic results obtained with rifaximin-α should not be translated sic et simpliciter to the generic formulations of rifaximin, which do not claim containing only rifaximin-α and will display significantly higher systemic absorption in both health and disease.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Drugs, Generic/pharmacokinetics , Rifamycins/pharmacokinetics , Adolescent , Adult , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Biological Availability , Cross-Over Studies , Female , Healthy Volunteers , Humans , Intestinal Absorption , Male , Middle Aged , Rifamycins/blood , Rifamycins/urine , Rifaximin , Single-Blind Method , Young Adult
6.
Expert Rev Clin Pharmacol ; 17(7): 537-547, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38339997

ABSTRACT

INTRODUCTION: Tuberculosis (TB) is a leading infectious disease cause of mortality worldwide, especially for people living with human immunodeficiency virus (PLWH). Treating TB in PLWH can be challenging due to numerous drug interactions. AREAS COVERED: This review discusses drug interactions between antitubercular and antiretroviral drugs. Due to its clinical importance, initiation of antiretroviral therapy in patients requiring TB treatment is discussed. Special focus is placed on the rifamycin class, as it accounts for the majority of interactions. Clinically relevant guidance is provided on how to manage these interactions. An additional section on utilizing therapeutic drug monitoring (TDM) to optimize drug exposure and minimize toxicities is included. EXPERT OPINION: Antitubercular and antiretroviral coadministration can be successfully managed. TDM can be used to optimize drug exposure and minimize toxicity risk. As new TB and HIV drugs are discovered, additional research will be needed to assess for clinically relevant drug interactions.


Subject(s)
Anti-HIV Agents , Antitubercular Agents , Drug Interactions , Drug Monitoring , HIV Infections , Tuberculosis , Humans , Tuberculosis/drug therapy , HIV Infections/drug therapy , HIV Infections/complications , Antitubercular Agents/administration & dosage , Antitubercular Agents/adverse effects , Antitubercular Agents/pharmacokinetics , Antitubercular Agents/pharmacology , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Drug Monitoring/methods , Rifamycins/administration & dosage , Rifamycins/pharmacokinetics , Rifamycins/adverse effects
7.
Antimicrob Agents Chemother ; 57(4): 1648-53, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23335744

ABSTRACT

During active tuberculosis a spectrum of physiologically different Mycobacterium tuberculosis bacilli reside in human tissues. Subpopulations of the pathogen survive antibiotic treatment for a prolonged time in a dormant state of phenotypic drug resistance, a phenomenon independent of genetic mutations. Here, we used an established culture model of nutrient deprivation to shift down M. tuberculosis from growth to nonreplicating survival, which is characterized by a drastic loss of drug susceptibility. Liquid chromatography coupled with mass spectrometry techniques were employed to quantify drug penetration in replicating and nutrient-starved nonreplicating bacilli. We found that intracellular concentrations of fluoroquinolones, rifamycins, and linezolid were lower in nonreplicating M. tuberculosis. Studies with pump inhibitors suggest that the observed differences were independent of efflux processes. We conclude that decreased drug permeability contributes to phenotypic drug resistance of dormant M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Acetamides/pharmacokinetics , Acetamides/pharmacology , Antitubercular Agents/pharmacokinetics , Fluoroquinolones/pharmacokinetics , Fluoroquinolones/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Linezolid , Microbial Sensitivity Tests , Oxazolidinones/pharmacokinetics , Oxazolidinones/pharmacology , Rifamycins/pharmacokinetics , Rifamycins/pharmacology
8.
Curr Med Chem ; 29(16): 2846-2862, 2022.
Article in English | MEDLINE | ID: mdl-34365945

ABSTRACT

Rifamycins are considered a milestone for tuberculosis (TB) treatment because of their proficient sterilizing ability. Currently, available TB treatments are complicated and need a long duration, which ultimately leads to failure of patient compliance. Some new rifamycin derivatives, i.e., rifametane, TNP-2092 (rifamycin-quinolizinonehybrid), and TNP-2198 (rifamycin-nitromidazole hybrid) are under clinical trials, which are attempting to overcome the problems associated with TB treatment. The undertaken review is intended to compare the pharmacokinetics, pharmacodynamics and safety profiles of these rifamycins, including rifalazil, another derivative terminated in phase II trials, and already approved rifamycins. The emerging resistance of microbes is an imperative consideration associated with antibiotics. Resistance development potential of microbial strains against rifamycins and an overview of chemistry, as well as structure-activity relationship (SAR) of rifamycins, are briefly described. Moreover, issues associated with rifamycins are discussed as well. We expect that newly emerging rifamycins shall appear as potential tools for TB treatment in the near future.


Subject(s)
Rifamycins , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Humans , Microbial Sensitivity Tests , Rifamycins/pharmacokinetics , Rifamycins/therapeutic use
9.
Antimicrob Agents Chemother ; 55(5): 2122-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21402860

ABSTRACT

The new oral 200-mg rifamycin SV MMX modified-release tablets, designed to deliver rifamycin SV directly into the colonic lumen, offer considerable advantages over the existing immediate-release antidiarrheic formulations. In two pharmacokinetics studies of healthy volunteers, the absorption, urinary excretion, and fecal elimination of rifamycin SV after single- and multiple-dose regimens of the new formulation were investigated. Concentrations in plasma of >2 ng/ml were infrequently and randomly quantifiable after single and multiple oral doses. The systemic exposure to rifamycin SV after single and multiple oral doses of MMX tablets under fasting and fed conditions or following a four-times-a-day (q.i.d.) or a twice-a-day (b.i.d.) regimen could be considered negligible. With both oral regimens, the drug was confirmed to be very poorly absorbable systemically. The amount of systemically absorbed antibiotic excreted by the renal route is far lower than 0.01% of the administered dose after both the single- and multiple-dose regimens. The absolute bioavailability, calculated as the mean percent ratio between total urinary excretion amounts (ΣXu) after a single intravenous injection and after a single oral dose under fasting conditions, was 0.0410±0.0617. The total elimination of the unchanged rifamycin SV with feces was 87% of the administered oral dose. No significant effect of rifamycin SV on vital signs, electrocardiograms, or laboratory parameters was observed.


Subject(s)
Rifamycins/administration & dosage , Rifamycins/pharmacokinetics , Tablets/administration & dosage , Tablets/pharmacokinetics , Adult , Aged , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/blood , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/urine , Female , Humans , Male , Middle Aged , Rifamycins/blood , Rifamycins/urine
10.
Xenobiotica ; 40(1): 24-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19919292

ABSTRACT

Transporters are carrier proteins that may influence pharmacokinetic, pharmacodynamic, and toxicological characteristics of drugs. The development of validated in vitro transporter models is imperative to support regulatory submissions of drug candidates. This study is focused on utilizing human embryonic kidney (HEK) 293 cell cultures genetically transfected with the human organic anion transporting polypeptides (OATP) 1B1 transporter to identify substrates and inhibitors in drug development. The kinetics of OATP1B1-mediated uptake of [(3)H]-oestradiol 17beta-glucuronide and inhibition of uptake by rifamycin SV were used to determine K(m), V(max), and IC(50) values over a range of passage numbers to investigate accuracy and precision. The mean K(m) and V(max) values were found to be 6.3 +/- 1.2 microM and 460 +/- 96 pmol min(-1) mg(-1), respectively. The mean IC(50) value for rifamycin SV was 0.23 +/- 0.07 microM on uptake of 1 microM [(3)H]-oestradiol 17beta-glucuronide. These data were similar to previously reported values (accuracy greater than 82%), reproducible (precision less than 29%) and exhibited low standard deviations (SDs) obviating the need to study test compounds on more than one occasion. [(3)H]-oestrone 3-sulfate and [(3)H]-pravastatin exhibited concentration-dependent OATP1B1 uptake, and statistically significant differences were observed at each concentration between uptake rates of HEK293-OATP1B1 and HEK293-MOCK cells (uptake ratios greater than or equal to 3). Propranolol showed no positive uptake ratio. Bezafibrate and gemfibrozil exhibited concentration-dependent inhibition of OATP1B1-mediated uptake of [(3)H]-oestradiol 17beta-glucuronide with mean IC(50) values of 16 and 27 microM, respectively. Based on the validation results, acceptance criteria to identify a test compound as a substrate and/or inhibitor using these specific cell lines were determined. These validated OATP1B1 assays were robust, reproducible, and suitable for routine in vitro evaluation of candidate drugs.


Subject(s)
Drug Interactions , Organic Anion Transporters/metabolism , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/pharmacology , Bezafibrate/pharmacokinetics , Bezafibrate/pharmacology , Biological Transport , Cell Line , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , Estradiol/pharmacology , Gemfibrozil/pharmacokinetics , Gemfibrozil/pharmacology , Humans , Hypolipidemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Kidney/drug effects , Kidney/metabolism , Kinetics , Liver-Specific Organic Anion Transporter 1 , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Rifamycins/pharmacokinetics , Rifamycins/pharmacology
11.
Ann Pharmacother ; 43(1): 77-84, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19092143

ABSTRACT

OBJECTIVE: To review the effectiveness and safety of rifaximin in the treatment of hepatic encephalopathy (HE). DATA SOURCES: MEDLINE (1990-October 2008) was searched using the terms rifaximin, rifamycins, hepatic encephalopathy, liver cirrhosis, and acute liver failure. Other sources included the bibliographies of pertinent articles as well as programs and abstracts from infectious diseases and gastrointestinal diseases meetings. STUDY SELECTION AND DATA EXTRACTION: All English-language articles identified from the search were evaluated. All primary literature that addressed the efficacy and safety of rifaximin in the treatment of HE was included in this review. DATA SYNTHESIS: HE is a complex neuropsychiatric syndrome seen in patients with liver failure. It is characterized by disturbances in consciousness and behavior, personality changes, fluctuating neurologic signs, asterixis, and electroencephalographic changes. Although the etiology of HE is unknown, the accumulation of several gut-derived toxins such as mercaptans, ammonia, and benzodiazepine has been implicated. Current treatment options for HE include agents that reduce the concentration of these toxins, such as nonabsorbable disaccharides and antibiotics. Several studies have evaluated the use of rifaximin in the treatment of HE. They include a dose-finding study, 9 open-label studies, and 4 double-blind studies comparing rifaximin with either nonabsorbable disaccharides or antibiotics. Commonly used outcomes in most of these studies were changes in portal systemic encephalopathy index and the improvement in HE grade. Despite various limitations of the studies, rifaximin showed superior efficacy compared with lactulose for the treatment of HE, similar efficacy to paromomycin, and similar or greater efficacy than neomycin. Rifaximin was found to be associated with fewer hospitalizations, fewer days of hospitalization, and lower hospitalization charges than were seen with lactulose. Rifaximin also had a better tolerance profile than the comparative agents. CONCLUSIONS: Rifaximin appears to be an effective and safe treatment option for HE. Better-designed studies are needed to characterize its efficacy in the treatment of HE.


Subject(s)
Gastrointestinal Agents/therapeutic use , Hepatic Encephalopathy/drug therapy , Rifamycins/therapeutic use , Animals , Clinical Trials as Topic/methods , Gastrointestinal Agents/pharmacokinetics , Hepatic Encephalopathy/metabolism , Humans , Rifamycins/pharmacokinetics , Rifaximin , Severity of Illness Index , Treatment Outcome
12.
Biomed Chromatogr ; 23(11): 1145-50, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19444797

ABSTRACT

A highly sensitive and selective on-line two-dimensional reversed-phase liquid chromatography/electrospray ionization-tandem mass spectrometry (2D-LC-ESI/MS/MS) method was developed and validated to determine rifaximin in rat serum by direct injection. The 2D-LC-ESI/MS/MS system consisted of a restricted access media column for trapping proteins as the first dimension and a Waters C(18 )column as second dimension using 0.1% aqueous acetic acid:acetonitrile as mobile phase in a gradient elution mode. Rifampacin was used as an internal standard. The linear dynamic range was 0.5-10 ng/mL (r(2) > 0.998). Acceptable precision and accuracy were obtained over the calibration range. The assay was successfully used in analysis of rat serum to support pharmacokinetic studies.


Subject(s)
Anti-Infective Agents/blood , Rifamycins/blood , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Animals , Anti-Infective Agents/pharmacokinetics , Calibration , Rats , Rats, Wistar , Reproducibility of Results , Rifamycins/pharmacokinetics , Rifaximin , Sensitivity and Specificity
13.
Biomed Chromatogr ; 23(6): 563-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19101925

ABSTRACT

A simple and rapid reversed-phase HPLC method for determination of rifaximin in rat serum and urine was developed. Separation of rifaximin from biological matrix was achieved by direct injection of rat serum and urine onto a restricted-access medium, Supelco LC-Hisep, a shielded hydrophobic stationary phase, using acetonitrile:water:acetic acid (18:82:0.1 v/v/v) as a mobile phase. The linear range was 0.10-20 microg/mL (r(2 )> 0.999, n = 6), intraday and interday variation was <6.10%. The limits of detection and quantification were 0.03 (signal-to-noise ratio >3) and 0.10 microg/mL (signal-to-noise ratio >10), respectively. The method was successfully applied to pharmacokinetic studies of rifaximin after an oral administration to rats.


Subject(s)
Anti-Infective Agents/blood , Anti-Infective Agents/urine , Chromatography, High Pressure Liquid/methods , Rifamycins/blood , Rifamycins/urine , Animals , Anti-Infective Agents/pharmacokinetics , Chromatography, High Pressure Liquid/economics , Chromatography, High Pressure Liquid/instrumentation , Hydrophobic and Hydrophilic Interactions , Rats , Rats, Wistar , Rifamycins/pharmacokinetics , Rifaximin , Sensitivity and Specificity , Time Factors
14.
Clin Drug Investig ; 39(7): 691-697, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31172447

ABSTRACT

Rifamycin SV MMX® (Aemcolo™; Relafalk™) is a novel oral formulation of the antibacterial rifamycin SV that uses MultiMatrix (MMX®) technology to enable colonic delivery. Specifically, the active ingredient (rifamycin SV) is released throughout the colon, where it acts locally in the intestinal lumen; systemic absorption is minimal. Rifamycin SV MMX® exhibits antibacterial activity against a broad spectrum of clinically relevant enteropathogens and is available in the EU and the USA for the treatment of adults with traveller's diarrhoea. In two multinational, phase III studies, rifamycin SV MMX® (400 mg twice daily for 3 days) effectively shortened the duration of non-dysenteric traveller's diarrhoea in adults, being significantly more effective than placebo and noninferior to ciprofloxacin in reducing median time to last unformed stool. As expected (given its poor systemic absorption), rifamycin SV MMX® was well tolerated in this patient population, with the overall incidence of treatment-emergent adverse events generally similar to those of placebo and ciprofloxacin. Current evidence indicates that twice-daily rifamycin SV MMX® is an effective and well tolerated treatment option for shortening the duration of non-dysenteric traveller's diarrhoea in adults.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Diarrhea/drug therapy , Rifamycins/therapeutic use , Adult , Anti-Bacterial Agents/pharmacokinetics , Humans , Placebos , Rifamycins/pharmacokinetics
15.
MAbs ; 11(6): 1162-1174, 2019.
Article in English | MEDLINE | ID: mdl-31219754

ABSTRACT

DSTA4637S, a novel THIOMAB™ antibody-antibiotic conjugate (TAC) against Staphylococcus aureus (S. aureus), is currently being investigated as a potential therapy for complicated S. aureus bloodstream infections. DSTA4637S is composed of a monoclonal THIOMABTM IgG1 recognizing S. aureus linked to a rifamycin-class antibiotic (dmDNA31) via a protease-cleavable linker. The pharmacokinetics (PK) of DSTA4637A (a liquid formulation of DSTA4637S) and its unconjugated antibody MSTA3852A were characterized in rats and monkeys. Systemic concentrations of three analytes, total antibody (TAb), antibody-conjugated dmDNA31 (ac-dmDNA31), and unconjugated dmDNA31, were measured to describe complex TAC PK in nonclinical studies. In rats and monkeys, following intravenous administration of a single dose of DSTA4637A, systemic concentration-time profiles of both TAb and ac-dmDNA31 were bi-exponential, characterized by a short distribution phase and a long elimination phase as expected for a monoclonal antibody-based therapeutic. Systemic exposures of both TAb and ac-dmDNA31 were dose proportional over the dose range tested, and ac-dmDNA31 cleared 2-3 times faster than TAb. Unconjugated dmDNA31 plasma concentrations were low (<4 ng/mL) in every study regardless of dose. In this report, an integrated semi-mechanistic PK model for two analytes (TAb and ac-dmDNA31) was successfully developed and was able to well describe the complicated DSTA4637A PK in mice, rats and monkeys. DSTA4637S human PK was predicted reasonably well using this model with allometric scaling of PK parameters from monkey data. This work provides insights into PK behaviors of DSTA4637A in preclinical species and informs clinical translatability of these observed results and further clinical development. Abbreviations: ADC: Antibody-drug conjugate; AUCinf: time curve extrapolated to infinity; ac-dmDNA31: antibody-conjugated dmDNA31; Cmax: maximum concentration observed; DAR: drug-to-antibody ratio; CL: clearance; CLD: distribution clearance; CL1: systemic clearance of all DAR species; kDC: deconjugation rate constant; PK: Pharmacokinetics; IV: Intravenous; IgG: Immunoglobulin G; mAb: monoclonal antibody; S. aureus: Staphylococcus aureus; TAC: THIOMABTM antibody-antibiotic conjugate; TDC: THIOMABTM antibody-drug conjugate; TAb: total antibody; t1/2, λz: terminal half-life; vc linker: valine-citrulline linker; Vss: volume of distribution at steady state; Vc: volume of distribution for the central compartment; Vp: the volume of distribution for the peripheral compartment.


Subject(s)
Antibodies, Bacterial , Antibodies, Monoclonal , Immunoconjugates , Immunoglobulin G , Rifamycins , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Macaca fascicularis , Male , Rats , Rats, Sprague-Dawley , Rifamycins/immunology , Rifamycins/pharmacokinetics , Rifamycins/pharmacology
16.
J Antimicrob Chemother ; 62(1): 156-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18400804

ABSTRACT

OBJECTIVES: ABI-0043 is a novel benzoxazinorifamycin derivative, which derives its potent bactericidal activity by the specific inhibition of bacterial RNA polymerase. We evaluated the in vitro pharmacodynamics and bactericidal activity of ABI-0043 against clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-susceptible S. aureus (MSSA). METHODS: Using time-kill studies at a wide range of concentrations of ABI-0043, we evaluated the killing activity against four clinical isolates of S. aureus over 24 h. An integrated pharmacokinetic/pharmacodynamic area measure was applied to all cfu data and was fitted to a Hill-type mathematical model to evaluate pharmacodynamics. RESULTS: Bacterial killing for ABI-0043 occurred rapidly and in a concentration-dependent manner. Bactericidal activity was achieved within 4 h at > or =16 x MIC against all isolates. Bacterial reductions were greatest at > or =64 x MIC against MRSA and MSSA isolates, as a >4 log(10) cfu/mL reduction was observed as early as 2 h, and sustained throughout 24 h. The pharmacodynamics of ABI-0043 was well described by a Hill-type model, with a steep sigmoidicity constant and a low EC(50) against all isolates. CONCLUSIONS: ABI-0043 displayed rapid and sustained bactericidal activity against S. aureus clinical isolates. ABI-0043 represents a promising antistaphylococcal agent to combat serious S. aureus infections. Further, pharmacokinetic, pharmacodynamic and in vivo studies are warranted to determine its ultimate place in antibacterial therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Rifamycins/pharmacology , Rifamycins/pharmacokinetics , Staphylococcus aureus/drug effects , Colony Count, Microbial , Dose-Response Relationship, Drug , Humans , Methicillin Resistance , Microbial Viability , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification , Time Factors
17.
Drug Metab Dispos ; 36(10): 1998-2004, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18606743

ABSTRACT

A novel reaction system was developed for the production of metabolites of poorly water-soluble parent compounds using mammalian liver microsomes. The system includes the selection and use of an appropriate hydrophobic polymeric resin as a reservoir for the hydrophobic parent compounds and its metabolites. The utility of the extractive biotransformation approach was shown for the production of a low-yielding, synthetically challenging 32-hydroxylated metabolite of the antibiotic rifalazil using mouse liver microsomes. To address the low solubility and reactivity of rifalazil in the predominantly aqueous microsomal catalytic system, a variety of strategies were tested for the enhanced delivery of hydrophobic substrates, including the addition of mild detergents, polyvinylpyrrolidone, glycerol, bovine serum albumin, and hydrophobic polymeric resins. The latter strategy was identified as the most suitable for the production of 32-hydroxy-rifalazil, resulting in up to 13-fold enhancement of the volumetric productivity compared with the standard aqueous system operating at the solubility limit of rifalazil. The production process was optimized for a wide range of reaction parameters; the most important for improving volumetric productivity included the type and amount of the polymeric resin, cofactor recycling system, concentrations of the biocatalyst and rifalazil, reaction temperature, and agitation rate. The optimized extractive biotransformation system was used to synthesize 32-hydroxy-rifalazil on a multimilligram scale.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Rifamycins/chemical synthesis , Animals , Anti-Bacterial Agents/pharmacokinetics , Biotransformation , Chromatography, High Pressure Liquid , Female , Male , Mass Spectrometry , Mice , Microsomes, Liver/metabolism , Rifamycins/pharmacokinetics , Solubility
20.
Expert Opin Drug Metab Toxicol ; 14(7): 753-760, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29897844

ABSTRACT

INTRODUCTION: Rifaximin is a poorly absorbable antibiotic with a broad-spectrum activity against both Gram-negative and -positive bacteria. It is active in the small intestine due to its high bile solubility, whereas in the aqueous environment of the colon, it shows limited efficacy against highly susceptible bacteria. These unique pharmacokinetic properties limit its systemic effects and can correct gut microflora imbalances. Thus, rifaximin has become a major therapeutic agent in several gastrointestinal diseases in which an imbalance in gut microflora may play a role, including diarrhea predominant irritable bowel syndrome (IBS-D). Area covered: This is an up-to-date review focusing on the efficacy of rifaximin in the treatment of IBS-D in both adult and pediatric populations. We will review the pharmacokinetic properties of rifaximin, including its absorption in health and disease, mechanisms of action, and interactions with other drugs. Expert opinion: Given its safety profile and its negligible absorption, rifaximin is a suitable treatment for IBS-D, in both adults and children. The possibility of modulating gut microbiota composition without side effects has made this drug an appealing therapeutic agent in highly prevalent gastrointestinal diseases. However, to date, monitoring for the development of resistant bacterial strains during long-term rifaximin use is still lacking.


Subject(s)
Diarrhea/drug therapy , Irritable Bowel Syndrome/drug therapy , Rifamycins/therapeutic use , Adult , Animals , Anti-Infective Agents/adverse effects , Anti-Infective Agents/pharmacokinetics , Anti-Infective Agents/therapeutic use , Child , Diarrhea/etiology , Diarrhea/microbiology , Drug Interactions , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/therapeutic use , Gastrointestinal Microbiome/drug effects , Humans , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/physiopathology , Rifamycins/adverse effects , Rifamycins/pharmacokinetics , Rifaximin
SELECTION OF CITATIONS
SEARCH DETAIL