Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
AAPS J ; 22(1): 3, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31712917

ABSTRACT

Multiple approaches such as mathematical deconvolution and mechanistic oral absorption models have been used to predict in vivo drug dissolution in the gastrointestinal (GI) tract. However, these approaches are often validated by plasma pharmacokinetic profiles, but not by in vivo drug dissolution due to the limited data available regarding the local GI environment. It is also challenging to predict and validate in vivo dissolution in different regions of the GI tract (stomach, duodenum, jejunum, and ileum). In this study, the dynamic fluid compartment absorption and transport (DFCAT) model was used to predict the in vivo dissolution profiles of ibuprofen, which was administered as an 800-mg immediate-release tablet to healthy subjects, in different regions of the GI tract. The prediction was validated with concentration time-courses of ibuprofen (BCS class 2a) in different regions of the GI tract that we have obtained over the past few years. The computational model predicted that the dissolution of ibuprofen was minimal in the stomach (2%), slightly more in the duodenum (6.3%), and primarily dissolved in the jejunum (63%) and the ileum (25%). The detailed model prediction of drug dissolution in different regions of GI can provide a quantitative reference of in vivo dissolution that may provide valuable insight in developing in vitro tests for drug product optimization and quality.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Drug Liberation , Ibuprofen/pharmacokinetics , Intestinal Absorption , Models, Theoretical , Administration, Oral , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Gastrointestinal Tract , Humans , Ibuprofen/administration & dosage , Proof of Concept Study
2.
mSphere ; 4(2)2019 03 13.
Article in English | MEDLINE | ID: mdl-30867328

ABSTRACT

Although the microbiota in the proximal gastrointestinal (GI) tract have been implicated in health and disease, much about these microbes remains understudied compared to those in the distal GI tract. This study characterized the microbiota across multiple proximal GI sites over time in healthy individuals. As part of a study of the pharmacokinetics of oral mesalamine administration, healthy, fasted volunteers (n = 8; 10 observation periods total) were orally intubated with a four-lumen catheter with multiple aspiration ports. Samples were taken from stomach, duodenal, and multiple jejunal sites, sampling hourly (≤7 h) to measure mesalamine (administered at t = 0), pH, and 16S rRNA gene-based composition. We observed a predominance of Firmicutes across proximal GI sites, with significant variation compared to stool. The microbiota was more similar within individuals over time than between subjects, with the fecal microbiota being unique from that of the small intestine. The stomach and duodenal microbiota displayed highest intraindividual variability compared to jejunal sites, which were more stable across time. We observed significant correlations in the duodenal microbial composition with changes in pH; linear mixed models identified positive correlations with multiple Streptococcus operational taxonomic units (OTUs) and negative correlations with multiple Prevotella and Pasteurellaceae OTUs. Few OTUs correlated with mesalamine concentration. The stomach and duodenal microbiota exhibited greater compositional dynamics than the jejunum. Short-term fluctuations in the duodenal microbiota were correlated with pH. Given the unique characteristics and dynamics of the proximal GI tract microbiota, it is important to consider these local environments in health and disease states.IMPORTANCE The gut microbiota are linked to a variety of gastrointestinal diseases, including inflammatory bowel disease. Despite this importance, microbiota dynamics in the upper gastrointestinal tract are understudied. Our article seeks to understand what factors impact microbiota dynamics in the healthy human upper gut. We found that the upper gastrointestinal tract contains consistently prevalent bacterial OTUs that dominate the overall community. Microbiota variability is highest in the stomach and duodenum and correlates with pH.


Subject(s)
Bacteria/classification , Fasting , Gastrointestinal Microbiome , Intestine, Small/microbiology , Stomach/microbiology , Administration, Oral , Adolescent , Adult , Bacteria/isolation & purification , Feces/microbiology , Female , Firmicutes/classification , Firmicutes/isolation & purification , Healthy Volunteers , Humans , Hydrogen-Ion Concentration , Intubation, Gastrointestinal , Linear Models , Male , Middle Aged , Pasteurellaceae/classification , Pasteurellaceae/isolation & purification , RNA, Ribosomal, 16S/genetics , Spatio-Temporal Analysis , Young Adult
3.
J Neurogastroenterol Motil ; 25(1): 100-112, 2019 Jan 31.
Article in English | MEDLINE | ID: mdl-30646481

ABSTRACT

BACKGROUND/AIMS: High-resolution methods have advanced esophageal and anorectal manometry interpretation but are incompletely established for intestinal manometry. We characterized normal fasting duodeno-jejunal manometry parameters not measurable by standard techniques using clustered closely-spaced recordings. METHODS: Ten fasting recordings were performed in 8 healthy controls using catheters with 3-4 gastrointestinal manometry clusters with 1-2 cm channel spacing. Migrating motor complex phase III characteristics were quantified. Spatial-temporal contour plots measured propagation direction and velocity of individual contractions. Coupling was defined by pressure peak continuity within clusters. RESULTS: Twenty-three phase III complexes (11 antral, 12 intestinal origin) with 157 (95% CI, 104-211) minute periodicities, 6.99 (6.25-7.74) minute durations, 10.92 (10.68-11.16) cycle/minute frequencies, 73.6 (67.7-79.5) mmHg maximal amplitudes, and 4.20 (3.18-5.22) cm/minute propagation velocities were recorded. Coupling of individual contractions was 39.1% (32.1-46.1); 63.0% (54.4-71.6) of contractions were antegrade and 32.8% (24.1-41.5) were retrograde. Individual phase III contractions propagated > 35 fold faster (2.48 cm/sec; 95% CI, 2.25-2.71) than complexes themselves. Phase III complexes beyond the proximal jejunum were longer in duration (P = 0.025) and had poorer contractile coupling (P = 0.025) than proximal complexes. Coupling was greater with 1 cm channel spacing vs 2 cm (P < 0.001). CONCLUSIONS: Intestinal manometry using clustered closely-spaced pressure ports characterizes novel antegrade and retrograde propagation and coupling properties which degrade in more distal jejunal segments. Coupling is greater with more closely-spaced recordings. Applying similar methods to dysmotility syndromes will define the relevance of these methods.

4.
Mol Pharm ; 15(12): 5468-5478, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30417648

ABSTRACT

Exploring the intraluminal behavior of an oral drug product in the human gastrointestinal (GI) tract remains challenging. Many in vivo techniques are available to investigate the impact of GI physiology on oral drug behavior in fasting state conditions. However, little is known about the intraluminal behavior of a drug in postprandial conditions. In a previous report, we described the mean solution and total concentrations of ibuprofen after oral administration of an immediate-release (IR) tablet in fed state conditions. In parallel, blood samples were taken to assess systemic concentrations. The purpose of this work was to statistically evaluate the impact of GI physiology (e.g., pH, contractile events) within and between individuals (intra and intersubject variability) for a total of 17 healthy subjects. In addition, a pharmacokinetic (PK) analysis was performed by noncompartmental analysis, and PK parameters were correlated with underlying physiological factors (pH, time to phase III contractions postdose) and study parameters (e.g., ingested amount of calories, coadministered water). Moreover, individual plasma profiles were deconvoluted to assess the fraction absorbed as a function of time, demonstrating the link between intraluminal and systemic behavior of the drug. The results demonstrated that the in vivo dissolution of ibuprofen depends on the present gastric pH and motility events at the time of administration. Both intraluminal factors were responsible for explaining 63% of plasma Cmax variability among all individuals. For the first time, an in-depth analysis was performed on a large data set derived from an aspiration/motility study, quantifying the impact of physiology on systemic behavior of an orally administered drug product in fed state conditions. The data obtained from this study will help us to develop an in vitro biorelevant dissolution approach and optimize in silico tools in order to predict the in vivo performance of orally administered drug products, especially in fed state conditions.


Subject(s)
Drug Liberation , Gastric Absorption/physiology , Ibuprofen/pharmacokinetics , Postprandial Period/physiology , Stomach/physiology , Administration, Oral , Adult , Area Under Curve , Biological Availability , Biological Variation, Individual , Biological Variation, Population/physiology , Computer Simulation , Datasets as Topic , Female , Food-Drug Interactions/physiology , Gastric Emptying/physiology , Healthy Volunteers , Humans , Hydrogen-Ion Concentration , Ibuprofen/administration & dosage , Male , Middle Aged , Models, Biological , Solubility , Tablets , Young Adult
5.
Mol Pharm ; 15(12): 5454-5467, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30372084

ABSTRACT

The goal of this project was to explore and to statistically evaluate the responsible gastrointestinal (GI) factors that are significant factors in explaining the systemic exposure of ibuprofen, between and within human subjects. In a previous study, we determined the solution and total concentrations of ibuprofen as a function of time in aspirated GI fluids, after oral administration of an 800 mg IR tablet (reference standard) of ibuprofen to 20 healthy volunteers in fasted state conditions. In addition, we determined luminal pH and motility pressure recordings that were simultaneously monitored along the GI tract. Blood samples were taken to determine ibuprofen plasma levels. In this work, an in-depth statistical and pharmacokinetic analysis was performed to explain which underlying GI variables are determining the systemic concentrations of ibuprofen between (inter-) and within (intra-) subjects. In addition, the obtained plasma profiles were deconvoluted to link the fraction absorbed with the fraction dissolved. Multiple linear regressions were performed to explain and quantitatively express the impact of underlying GI physiology on systemic exposure of the drug (in terms of plasma Cmax/AUC and plasma Tmax). The exploratory analysis of the correlation between plasma Cmax/AUC and the time to the first phase III contractions postdose (TMMC-III) explains ∼40% of the variability in plasma Cmax for all fasted state subjects. We have experimentally shown that the in vivo intestinal dissolution of ibuprofen is dependent upon physiological variables like, in this case, pH and postdose phase III contractions. For the first time, this work presents a thorough statistical analysis explaining how the GI behavior of an ionized drug can explain the systemic exposure of the drug based on the individual profiles of participating subjects. This creates a scientifically based and rational framework that emphasizes the importance of including pH and motility in a predictive in vivo dissolution methodology to forecast the in vivo performance of a drug product. Moreover, as no extensive first-pass metabolism is considered for ibuprofen, this study demonstrates how intraluminal drug behavior is reflecting the systemic exposure of a drug.


Subject(s)
Drug Liberation , Fasting/physiology , Gastrointestinal Absorption/physiology , Gastrointestinal Tract/physiology , Ibuprofen/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Biological Availability , Biological Variation, Individual , Biological Variation, Population/physiology , Datasets as Topic , Female , Healthy Volunteers , Humans , Hydrogen-Ion Concentration , Ibuprofen/administration & dosage , Male , Middle Aged , Models, Biological , Solubility , Tablets , Young Adult
6.
Cell Host Microbe ; 24(3): 364-378.e6, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30212649

ABSTRACT

In addition to high-fat diet (HFD) and inactivity, inflammation and microbiota composition contribute to obesity. Inhibitory immune receptors, such as NLRP12, dampen inflammation and are important for resolving inflammation, but their role in obesity is unknown. We show that obesity in humans correlates with reduced expression of adipose tissue NLRP12. Similarly, Nlrp12-/- mice show increased weight gain, adipose deposition, blood glucose, NF-κB/MAPK activation, and M1-macrophage polarization. Additionally, NLRP12 is required to mitigate HFD-induced inflammasome activation. Co-housing with wild-type animals, antibiotic treatment, or germ-free condition was sufficient to restrain inflammation, obesity, and insulin tolerance in Nlrp12-/- mice, implicating the microbiota. HFD-fed Nlrp12-/- mice display dysbiosis marked by increased obesity-associated Erysipelotrichaceae, but reduced Lachnospiraceae family and the associated enzymes required for short-chain fatty acid (SCFA) synthesis. Lachnospiraceae or SCFA administration attenuates obesity, inflammation, and dysbiosis. These findings reveal that Nlrp12 reduces HFD-induced obesity by maintaining beneficial microbiota.


Subject(s)
Gastrointestinal Microbiome , Intracellular Signaling Peptides and Proteins/immunology , Obesity/immunology , Obesity/microbiology , Adipose Tissue/immunology , Adult , Aged , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Female , Homeostasis , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/genetics , Obesity/metabolism
7.
Eur J Pharm Biopharm ; 129: 162-174, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29857136

ABSTRACT

The goal of this study was to create a mass transport model (MTM) model for gastric emptying and upper gastrointestinal (GI) appearance that can capture the in vivo concentration-time profiles of the nonabsorbable drug phenol red in solution in the stomach and upper small intestine by direct luminal measurement while simultaneously recording the contractile activity (motility) via manometry. We advanced from a one-compartmental design of the stomach to a much more appropriate, multi-compartmental 'mixing tank' gastric model that reflects drug distribution along the different regions of the stomach as a consequence of randomly dosing relative to the different contractile phases of the migrating motor complex (MMC). To capture the intraluminal phenol red concentrations in the different segments of the GI tract both in fasted and fed state conditions, it was essential to include a bypass flow compartment ('magenstrasse') to facilitate the transport of the phenol red solution directly to the duodenum (fasted state) or antrum (fed state). The fasted and fed state models were validated with external reference data from an independent aspiration study using another nonabsorbable marker (paromomycin). These results will be essential for the development and optimization of computational programs for GI simulation and absorption prediction, providing a realistic gastric physiologically-based pharmacokinetic (PBPK) model based on direct measurement of gastric concentrations of the drug in the stomach.


Subject(s)
Gastric Emptying/drug effects , Intestinal Absorption , Intestine, Small/drug effects , Models, Biological , Stomach/physiology , Administration, Oral , Adult , Fasting , Female , Healthy Volunteers , Humans , Intestine, Small/physiology , Male , Middle Aged , Paromomycin/pharmacology , Phenolsulfonphthalein/pharmacology , Postprandial Period , Solubility , Stomach/drug effects , Young Adult
8.
AAPS J ; 19(6): 1682-1690, 2017 11.
Article in English | MEDLINE | ID: mdl-28980204

ABSTRACT

Gastrointestinal (GI) fluid volume and its dynamic change are integral to study drug disintegration, dissolution, transit, and absorption. However, key questions regarding the local volume and its absorption, secretion, and transit remain unanswered. The dynamic fluid compartment absorption and transit (DFCAT) model is proposed to estimate in vivo GI volume and GI fluid transport based on magnetic resonance imaging (MRI) quantified fluid volume. The model was validated using GI local concentration of phenol red in human GI tract, which was directly measured by human GI intubation study after oral dosing of non-absorbable phenol red. The measured local GI concentration of phenol red ranged from 0.05 to 168 µg/mL (stomach), to 563 µg/mL (duodenum), to 202 µg/mL (proximal jejunum), and to 478 µg/mL (distal jejunum). The DFCAT model characterized observed MRI fluid volume and its dynamic changes from 275 to 46.5 mL in stomach (from 0 to 30 min) with mucus layer volume of 40 mL. The volumes of the 30 small intestine compartments were characterized by a max of 14.98 mL to a min of 0.26 mL (0-120 min) and a mucus layer volume of 5 mL per compartment. Regional fluid volumes over 0 to 120 min ranged from 5.6 to 20.38 mL in the proximal small intestine, 36.4 to 44.08 mL in distal small intestine, and from 42 to 64.46 mL in total small intestine. The DFCAT model can be applied to predict drug dissolution and absorption in the human GI tract with future improvements.


Subject(s)
Drug Liberation , Intestinal Absorption , Administration, Oral , Gastric Emptying , Gastrointestinal Transit , Humans , Magnetic Resonance Imaging , Phenolsulfonphthalein/metabolism
10.
Mol Pharm ; 14(12): 4295-4304, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28937221

ABSTRACT

In vivo drug dissolution in the gastrointestinal (GI) tract is largely unmeasured. The purpose of this clinical study was to evaluate the in vivo drug dissolution and systemic absorption of the BCS class IIa drug ibuprofen under fed and fasted conditions by direct sampling of stomach and small intestinal luminal content. Expanding current knowledge of drug dissolution in vivo will help to establish physiologically relevant in vitro models predictive of drug dissolution. A multilumen GI catheter was orally inserted into the GI tract of healthy human subjects. Subjects received a single oral dose of ibuprofen (800 mg tablet) with 250 mL of water under fasting and fed conditions. The GI catheter facilitated collection of GI fluid from the stomach, duodenum, and jejunum. Ibuprofen concentration in GI fluid supernatant and plasma was determined by LC-MS/MS. A total of 23 subjects completed the study, with 11 subjects returning for an additional study visit (a total of 34 completed study visits). The subjects were primarily white (61%) and male (65%) with an average age of 30 years. The subjects had a median [min, max] weight of 79 [52, 123] kg and body mass index of 25.7 [19.4, 37.7] kg/m2. Ibuprofen plasma levels were higher under fasted conditions and remained detectable for 28 h under both conditions. The AUC0-24 and Cmax were lower in fed subjects vs fasted subjects, and Tmax was delayed in fed subjects vs fasted subjects. Ibuprofen was detected immediately after ingestion in the stomach under fasting and fed conditions until 7 h after dosing. Higher levels of ibuprofen were detected in the small intestine soon after dosing in fasted subjects compared to fed. In contrast to plasma drug concentration, overall gastric concentrations remained higher under fed conditions due to increased gastric pH vs fasting condition. The gastric pH increased to near neutrality after feedingbefore decreasing to acidic levels after 7 h. Induction of the fed state reduced systemic levels but increased gastric levels of ibuprofen, which suggest that slow gastric emptying and transit dominate the effect for plasma drug concentration. The finding of high levels of ibuprofen in stomach and small intestine 7 h post dosing was unexpected. Future work is needed to better understand the role of various GI parameters, such as motility and gastric emptying, on systemic ibuprofen levels in order to improve in vitro predictive models.


Subject(s)
Absorption, Physiological/physiology , Drug Liberation/physiology , Gastrointestinal Tract/physiology , Ibuprofen/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Biopharmaceutics , Fasting/physiology , Female , Gastric Emptying/physiology , Healthy Volunteers , Humans , Intestinal Absorption/physiology , Male , Middle Aged , Postprandial Period , Solubility , Tablets , Young Adult
11.
Mol Pharm ; 14(12): 4281-4294, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28737409

ABSTRACT

In this study, we determined the pH and buffer capacity of human gastrointestinal (GI) fluids (aspirated from the stomach, duodenum, proximal jejunum, and mid/distal jejunum) as a function of time, from 37 healthy subjects after oral administration of an 800 mg immediate-release tablet of ibuprofen (reference listed drug; RLD) under typical prescribed bioequivalence (BE) study protocol conditions in both fasted and fed states (simulated by ingestion of a liquid meal). Simultaneously, motility was continuously monitored using water-perfused manometry. The time to appearance of phase III contractions (i.e., housekeeper wave) was monitored following administration of the ibuprofen tablet. Our results clearly demonstrated the dynamic change in pH as a function of time and, most significantly, the extremely low buffer capacity along the GI tract. The buffer capacity on average was 2.26 µmol/mL/ΔpH in fasted state (range: 0.26 and 6.32 µmol/mL/ΔpH) and 2.66 µmol/mL/ΔpH in fed state (range: 0.78 and 5.98 µmol/mL/ΔpH) throughout the entire upper GI tract (stomach, duodenum, and proximal and mid/distal jejunum). The implication of this very low buffer capacity of the human GI tract is profound for the oral delivery of both acidic and basic active pharmaceutical ingredients (APIs). An in vivo predictive dissolution method would require not only a bicarbonate buffer but also, more significantly, a low buffer capacity of dissolution media to reflect in vivo dissolution conditions.


Subject(s)
Body Fluids/chemistry , Gastrointestinal Motility/physiology , Gastrointestinal Tract/physiology , Ibuprofen/pharmacokinetics , Intestinal Absorption/physiology , Absorption, Physiological , Administration, Oral , Adult , Body Fluids/physiology , Buffers , Drug Liberation , Healthy Volunteers , Humans , Hydrogen-Ion Concentration , Intestinal Mucosa/physiology , Manometry , Middle Aged , Solubility , Tablets , Therapeutic Equivalency , Time Factors , Young Adult
13.
Nat Immunol ; 18(5): 541-551, 2017 05.
Article in English | MEDLINE | ID: mdl-28288099

ABSTRACT

Inflammatory bowel diseases involve the dynamic interaction of host genetics, the microbiome and inflammatory responses. Here we found lower expression of NLRP12 (which encodes a negative regulator of innate immunity) in human ulcerative colitis, by comparing monozygotic twins and other patient cohorts. In parallel, Nlrp12 deficiency in mice caused increased basal colonic inflammation, which led to a less-diverse microbiome and loss of protective gut commensal strains (of the family Lachnospiraceae) and a greater abundance of colitogenic strains (of the family Erysipelotrichaceae). Dysbiosis and susceptibility to colitis associated with Nlrp12 deficency were reversed equally by treatment with antibodies targeting inflammatory cytokines and by the administration of beneficial commensal Lachnospiraceae isolates. Fecal transplants from mice reared in specific-pathogen-free conditions into germ-free Nlrp12-deficient mice showed that NLRP12 and the microbiome each contributed to immunological signaling that culminated in colon inflammation. These findings reveal a feed-forward loop in which NLRP12 promotes specific commensals that can reverse gut inflammation, while cytokine blockade during NLRP12 deficiency can reverse dysbiosis.


Subject(s)
Clostridiales/physiology , Colitis, Ulcerative/immunology , Colon/physiology , Firmicutes/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Microbiota , RNA, Ribosomal, 16S/analysis , Animals , Biodiversity , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colon/microbiology , Dextran Sulfate , Feces/microbiology , Gene-Environment Interaction , Humans , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/genetics , Microbiota/immunology , Symbiosis , Twins, Monozygotic
14.
Nat Microbiol ; 2: 16267, 2017 Jan 23.
Article in English | MEDLINE | ID: mdl-28112760

ABSTRACT

Mesalamine serves as the gold standard in treating ulcerative colitis. However, its precise mechanism(s) of action remains unclear. Here, we show that mesalamine treatment rapidly decreases polyphosphate levels in diverse bacteria, including members of the human gut microbiome. This decrease sensitizes bacteria towards oxidative stress, reduces colonization and attenuates persister cell and biofilm formation, suggesting that mesalamine aids in diminishing the capacity of bacteria to persist within chronically inflamed environments.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gastrointestinal Microbiome/drug effects , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/physiology , Mesalamine/pharmacology , Polyphosphates/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biofilms/drug effects , Cecum/microbiology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Escherichia coli/drug effects , Feces/microbiology , Gram-Negative Bacteria/genetics , Humans , Mesalamine/administration & dosage , Mesalamine/therapeutic use , Mice , Oxidative Stress/drug effects
15.
Mol Pharm ; 14(2): 345-358, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28009518

ABSTRACT

As an orally administered, locally acting gastrointestinal drug, mesalamine products are designed to achieve high local drug concentration in the gastrointestinal (GI) tract for the treatment of ulcerative colitis. The aim of this study was to directly measure and compare drug dissolution of three mesalamine formulations in human GI tract and to correlate their GI concentration with drug concentration in plasma. Healthy human subjects were orally administered Pentasa, Apriso, or Lialda. GI fluids were aspirated from stomach, duodenum, proximal jejunum, mid jejunum, and distal jejunum regions. Mesalamine (5-ASA) and its primary metabolite acetyl-5-mesalamine (Ac-5-ASA) were measured using LC-MS/MS. GI tract pH was measured from each GI fluid sample, which averaged 1.82, 4.97, 5.67, 6.17, and 6.62 in the stomach, duodenum, proximal jejunum, middle jejunum, and distal jejunum, respectively. For Pentasa, high levels of 5-ASA in solution were observed in the stomach, duodenum, proximal jejunum, mid jejunum, and distal jejunum from 1 to 7 h. Apriso had minimal 5-ASA levels in stomach, low to medium levels of 5-ASA in duodenum and proximal jejunum from 4 to 7 h, and high levels of 5-ASA in distal jejunum from 3 to 7 h. In contrast, Lialda had minimal 5-ASA levels from stomach and early small intestine. A composite appearance rate (CAR) was calculated from the deconvolution of individual plasma concentration to reflect drug release, dissolution, transit, and absorption in the GI tract. Individuals dosed with Pentasa had high levels of CAR from 1 to 10 h; individuals dosed with Apriso had low levels of CAR from 1 to 4 h and high levels of CAR from 5 to 10 h; Lialda showed minimal levels of CAR from 0 to 5 h, then increased to medium levels from 5 to 12 h, and then decreased to further lower levels after 12 h. In the colon region, Pentasa and Apriso showed similar levels of accumulated 5-ASA excreted in the feces, while Lialda showed slightly higher 5-ASA accumulation in feces. However, all three formulations showed similar levels of metabolite Ac-5-ASA in the feces. These results provide direct measurement of drug dissolution in the GI tract, which can serve as a basis for investigation of bioequivalence for locally acting drug products.


Subject(s)
Drug Liberation/physiology , Gastrointestinal Tract/metabolism , Mesalamine/metabolism , Administration, Oral , Adolescent , Adult , Chemistry, Pharmaceutical/methods , Female , Humans , Male , Middle Aged , Solubility , Young Adult
17.
Nat Immunol ; 17(5): 505-513, 2016 05.
Article in English | MEDLINE | ID: mdl-26998764

ABSTRACT

The effect of alterations in intestinal microbiota on microbial metabolites and on disease processes such as graft-versus-host disease (GVHD) is not known. Here we carried out an unbiased analysis to identify previously unidentified alterations in gastrointestinal microbiota-derived short-chain fatty acids (SCFAs) after allogeneic bone marrow transplant (allo-BMT). Alterations in the amount of only one SCFA, butyrate, were observed only in the intestinal tissue. The reduced butyrate in CD326(+) intestinal epithelial cells (IECs) after allo-BMT resulted in decreased histone acetylation, which was restored after local administration of exogenous butyrate. Butyrate restoration improved IEC junctional integrity, decreased apoptosis and mitigated GVHD. Furthermore, alteration of the indigenous microbiota with 17 rationally selected strains of high butyrate-producing Clostridia also decreased GVHD. These data demonstrate a heretofore unrecognized role of microbial metabolites and suggest that local and specific alteration of microbial metabolites has direct salutary effects on GVHD target tissues and can mitigate disease severity.


Subject(s)
Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Graft vs Host Disease/immunology , Intestines/immunology , Metabolome/immunology , Acetylation/drug effects , Animals , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , Butyrates/immunology , Butyrates/metabolism , Butyrates/pharmacology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Female , Gas Chromatography-Mass Spectrometry , Gastrointestinal Microbiome/physiology , Gene Expression/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/microbiology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/immunology , Histone Acetyltransferases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , Histones/immunology , Histones/metabolism , Immunoblotting , Intestines/cytology , Intestines/microbiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
18.
Infect Immun ; 83(3): 934-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534943

ABSTRACT

Clostridium difficile infection (CDI) following antibiotic therapy is a major public health threat. While antibiotic disruption of the indigenous microbiota underlies the majority of cases of CDI, the early dynamics of infection in the disturbed intestinal ecosystem are poorly characterized. This study defines the dynamics of infection with C. difficile strain VPI 10463 throughout the gastrointestinal (GI) tract using a murine model of infection. After inducing susceptibility to C. difficile colonization via antibiotic administration, we followed the dynamics of spore germination, colonization, sporulation, toxin activity, and disease progression throughout the GI tract. C. difficile spores were able to germinate within 6 h postchallenge, resulting in the establishment of vegetative bacteria in the distal GI tract. Spores and cytotoxin activity were detected by 24 h postchallenge, and histopathologic colitis developed by 30 h. Within 36 h, all infected mice succumbed to infection. We correlated the establishment of infection with changes in the microbiota and bile acid profile of the small and large intestines. Antibiotic administration resulted in significant changes to the microbiota in the small and large intestines, as well as a significant shift in the abundance of primary and secondary bile acids. Ex vivo analysis suggested the small intestine as the site of spore germination. This study provides an integrated understanding of the timing and location of the events surrounding C. difficile colonization and identifies potential targets for the development of new therapeutic strategies.


Subject(s)
Clostridioides difficile/pathogenicity , Clostridium Infections/pathology , Colitis/pathology , Gastrointestinal Tract/pathology , Animals , Anti-Bacterial Agents/adverse effects , Bile Acids and Salts/chemistry , Clostridioides difficile/growth & development , Clostridioides difficile/metabolism , Clostridium Infections/etiology , Clostridium Infections/microbiology , Clostridium Infections/mortality , Colitis/etiology , Colitis/microbiology , Colitis/mortality , Disease Progression , Enterotoxins/biosynthesis , Enterotoxins/metabolism , Feces/microbiology , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Spores, Bacterial/growth & development , Spores, Bacterial/metabolism , Spores, Bacterial/pathogenicity , Survival Analysis , Time Factors
19.
Nat Commun ; 5: 3114, 2014.
Article in English | MEDLINE | ID: mdl-24445449

ABSTRACT

Antibiotics can have significant and long-lasting effects on the gastrointestinal tract microbiota, reducing colonization resistance against pathogens including Clostridium difficile. Here we show that antibiotic treatment induces substantial changes in the gut microbial community and in the metabolome of mice susceptible to C. difficile infection. Levels of secondary bile acids, glucose, free fatty acids and dipeptides decrease, whereas those of primary bile acids and sugar alcohols increase, reflecting the modified metabolic activity of the altered gut microbiome. In vitro and ex vivo analyses demonstrate that C. difficile can exploit specific metabolites that become more abundant in the mouse gut after antibiotics, including the primary bile acid taurocholate for germination, and carbon sources such as mannitol, fructose, sorbitol, raffinose and stachyose for growth. Our results indicate that antibiotic-mediated alteration of the gut microbiome converts the global metabolic profile to one that favours C. difficile germination and growth.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/physiology , Clostridium Infections/metabolism , Clostridium Infections/microbiology , Gastrointestinal Tract/microbiology , Metabolome/drug effects , Microbiota/drug effects , Animals , Clostridioides difficile/drug effects , Clostridioides difficile/growth & development , Disease Susceptibility/metabolism , Disease Susceptibility/microbiology , Female , Male , Metabolomics , Mice, Inbred C57BL
20.
Curr Opin Gastroenterol ; 29(6): 628-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24100717

ABSTRACT

PURPOSE OF REVIEW: The use of faecal microbiota transplantation (FMT) as treatment for recurrent Clostridium difficile infection (CDI) has increased rapidly over the past few years. In this review, we highlight clinical studies of FMT for treatment of recurrent CDI and discuss the safety, standardization and future of this treatment option. The major risk factor for CDI is prior antibiotic use, which results in an altered state of the gut microbiota characterized by decreased microbial diversity. This altered gut microbiota increases the patient's susceptibility to CDI. In patients with recurrent CDI, the microbiota remains in a state with decreased diversity, and FMT from a healthy individual restores the gut microbiota and subsequently colonization resistance against the pathogen. RECENT FINDINGS: Recent studies have shown the success rate for FMT as treatment for recurrent CDI being greater than 90%. Standardized, frozen preparations of faeces can be used, which increases the availability of faeces for FMT and decreases the cost of screening individual donors. In addition, there have been recent advances in identifying a defined microbial community isolated from faeces that can restore colonization resistance against C. difficile. SUMMARY: The use of FMT is a successful treatment for recurrent CDI when primary treatment options have failed. However, more work needs to define potential long-term consequences of this treatment and understand how specific members of the gut microbiota can restore colonization resistance against C. difficile.


Subject(s)
Biological Therapy/methods , Enterocolitis, Pseudomembranous/therapy , Feces/microbiology , Microbiota , Biological Therapy/adverse effects , Biological Therapy/standards , Enterocolitis, Pseudomembranous/microbiology , Humans , Intestines/microbiology , Randomized Controlled Trials as Topic , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...