Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
F S Rep ; 4(2): 206-212, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37398623

ABSTRACT

Objective: Evaluate the efficacy and safety of elagolix, a GnRH antagonist, to treat polycystic ovarian syndrome (PCOS). Design: A phase 2, multicenter, double-blind, randomized, placebo-controlled trial. Setting: Outpatient and academic medical centers. Patients: One hundred fourteen women with PCOS (aged 18-35 years, body mass index 18.5-38 kg/m2). Interventions: Patients were randomized 2:2:2:2:2:3 to elagolix (25 mg twice daily, 50 mg once daily, 75 mg twice daily, 150 mg once daily, and 300 mg twice daily) or placebo. Main Outcome Measures: The primary endpoint was menstrual cycle normalization (defined as 2 menstrual cycles 21-35 days in length during the 4-month treatment period). The secondary endpoint was change from baseline to week 1 in the area under the luteinizing hormone (LH) serum concentration-time curve (AUC). Additional endpoints included change from baseline in serum hormone levels. Results: No significant improvement in restoring normal menstrual cycles was observed in treated subjects; 3 of 114 patients met the primary endpoint. Six patients experienced progesterone elevations indicative of ovulation. The LH levels decreased from baseline to week 16, and LH AUC was significantly reduced from baseline to week 1 in all elagolix treatment groups (P<.1 vs placebo). Follicle-stimulating hormone (FSH) levels generally remained stable through week 16, with no significant differences in FSH AUCs. Serum estradiol and testosterone concentrations were consistently reduced from baseline in all elagolix dose groups compared with placebo. Adverse event rates were similar across treatment groups. Conclusions: Elagolix treatment did not normalize the ovulatory cycle in patients with PCOS. Clinical Trial Registration Number: NCT03951077.

2.
Pharmacol Res Perspect ; 11(1): e01036, 2023 02.
Article in English | MEDLINE | ID: mdl-36537346

ABSTRACT

ABBV-47D11 is a neutralizing monoclonal antibody that targets a mutationally conserved hydrophobic pocket distal to the ACE2 binding site of SARS-CoV-2. This first-in-human safety, pharmacokinetics, and antiviral pharmacodynamic assessment in patients with COVID-19 provide an initial evaluation of this antibody that may allow further development. This multicenter, randomized, double-blind, and placebo-controlled single ascending dose study of ABBV-47D11 (180, 600, or 2400 mg) as an intravenous infusion, was in hospitalized and non-hospitalized (confined) adults with mild to moderate COVID-19. Primary outcomes were grade 3 or higher study drug-related adverse events and infusion-related reactions. Secondary outcomes were pharmacokinetic parameters and concentration-time profiles to Day 29, immunogenicity (anti-drug antibodies), and antiviral activity (change in RT-PCR viral load) from baseline to Days 15 and 29. ABBV-47D11 single doses up to 2400 mg were safe and tolerated and no safety signals were identified. The pharmacokinetics of ABBV-47D11 were linear and showed dose-proportional increases in serum concentrations with ascending doses. The exploratory anti-SARS-CoV-2 activity revealed a reduction of viral load at and above the 600 mg dose of ABBV-47D11 regardless of patient demographics and baseline characteristics, however; because of the high inter-individual variability and small sample size a statistical significance was not reached. There is potential for anti-SARS-CoV-2 activity with ABBV-47D11 doses of 600 mg or higher, which could be evaluated in future clinical trials designed and powered to assess viral load reductions and clinical benefit.


Subject(s)
COVID-19 , Adult , Humans , SARS-CoV-2 , Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents , Antibodies, Neutralizing
4.
Br J Clin Pharmacol ; 85(10): 2351-2359, 2019 10.
Article in English | MEDLINE | ID: mdl-31269278

ABSTRACT

AIMS: Patients with Alzheimer's disease (AD), the most common form of dementia, have reduced P-glycoprotein (P-gp) function at the blood-brain barrier. However, the effect of AD on P-gp function in peripheral organs, and the impact on medication efficacy and toxicity is unknown. In this study, clinical chart review and physiologically based pharmacokinetic (PBPK) modelling were employed to determine whether disease-associated changes in P-gp could be assessed from clinically measured digoxin concentrations in patients without and with dementia. METHODS: A retrospective chart review was conducted to compare digoxin dose and concentrations between cohorts. A PBPK model was developed to simulate changes in digoxin concentrations at single and multiple 62.5 and 125 µg/d doses due to reduced P-gp function in peripheral organs. RESULTS: Digoxin concentrations were similar between the nondementia (n = 75) and dementia (n = 72) cohorts (mean ± standard deviation; 0.64 ± 0.31 and 0.60 ± 0.34 ng/mL, respectively; -0.06 to 0.15, 95% confidence interval of difference). PBPK simulations showed that reduced P-gp function resulted in a significant increase in digoxin exposure (AUC), but not in Cmax . For example, when a 2-fold reduction in P-gp function was simulated in older people following multiple 125 µg/d digoxin doses, the AUC over the last dosing interval was increased compared to baseline (24.29 ± 3.94 vs 17.04 ± 3.46 ng/mL*h; 4.52 to 9.98); however, Cmax was similar (1.38 ± 0.20 vs 0.99 ± 0.18 ng/mL; -2.33 to 3.13). CONCLUSION: Clinically measured digoxin concentrations were not statistically different in patients with dementia. Based on PBPK simulations, digoxin AUC may need to be evaluated to adequately assess the impact of reduced P-gp function in peripheral organs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cardiotonic Agents/administration & dosage , Dementia/complications , Digoxin/administration & dosage , Models, Biological , Aged , Aged, 80 and over , Alzheimer Disease/complications , Area Under Curve , Blood-Brain Barrier/metabolism , Cardiotonic Agents/pharmacokinetics , Computer Simulation , Digoxin/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Male , Retrospective Studies
5.
J Pharm Sci ; 108(7): 2484-2489, 2019 07.
Article in English | MEDLINE | ID: mdl-30825461

ABSTRACT

It is well established that the expression and function of drug transporters at the blood-brain barrier are altered in Alzheimer's disease (AD). However, we recently demonstrated in a mouse model of AD that the expression of key drug transporters and metabolizing enzymes was modified in peripheral organs, such as the small intestine and liver, suggesting that systemic drug absorption may be altered in AD. The purpose of this study was to determine whether the expression of drug transporters in the kidneys differed between 8- to 9-month-old wild-type mice and APPswe/PSEN1dE9 (APP/PS1) transgenic mice, a mouse model of familial AD, using a quantitative targeted absolute proteomics approach. The protein expression of the drug transporters-multidrug resistance-associated protein 2, organic anion transporter 3, and organic cation transporter 2-was upregulated 1.6-, 1.3-, and 1.4-fold, respectively, in kidneys from APP/PS1 mice relative to wild-type mice. These results suggest that in addition to modified oral absorption of certain drugs, it is possible that the renal excretion of drugs that are multidrug resistance-associated protein 2, organic anion transporter 3, and organic cation transporter 2 substrates could be altered in AD. These changes could affect the interpretation of studies conducted during drug development using this mouse model of AD and potentially impact dosage regimens of such drugs prescribed in this patient population.


Subject(s)
Alzheimer Disease/metabolism , Kidney/metabolism , Membrane Transport Proteins/metabolism , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Disease Models, Animal , Female , Liver/metabolism , Mice, Transgenic , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters/metabolism
6.
Mol Pharm ; 16(3): 1406-1411, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30608704

ABSTRACT

The bile salt export pump (BSEP) is the primary canalicular transporter responsible for the secretion of bile acids from hepatocytes into bile canaliculi, and inhibition of this transporter has been associated with drug-induced liver injury (DILI). A common variant (rs2287622; p.V444A) in the gene encoding BSEP has been associated with an increased risk of cholestatic DILI. Although p.444V BSEP (reference) and p.444A BSEP (variant) do not differ in their transport kinetics of taurocholic acid (TCA), transport of the more abundant glycocholic acid (GCA) has not been investigated. Importantly, differences in the susceptibility of p.444V and p.444A BSEP to inhibition by drugs causing cholestatic DILI have not been investigated. To address these issues, the transport kinetics of GCA were evaluated by incubating membrane vesicles expressing either p.444V or p.444A BSEP with GCA over a range of concentrations (1, 10, 25, 50, and 100 µM). The abilities of commonly used cholestatic medications to inhibit the transport of TCA and GCA by the reference and variant proteins were compared. Resulting data indicated that GCA transport kinetics for reference and variant BSEP followed Michaelis-Menten kinetics and were not statistically different [ Vmax values of 1132 ± 246 and 959 ± 256 pmol min-1 (mg of protein)-1, respectively, and Km values of 32.7 ± 18.2 and 45.7 ± 25.5 µM, respectively]. There were no statistically significant differences between the reference and variant BSEP in the inhibition of TCA or GCA transport by the cholestatic drugs tested. In conclusion, differential inhibition of TCA or GCA transport cannot account for an association between the variant BSEP and the risk for cholestatic DILI due to the drugs tested.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , Bile Acids and Salts/metabolism , Cholagogues and Choleretics/therapeutic use , Cholestasis/drug therapy , Polymorphism, Single Nucleotide , ATP Binding Cassette Transporter, Subfamily B, Member 11/antagonists & inhibitors , Animals , Baculoviridae , Chemical and Drug Induced Liver Injury/metabolism , Cholagogues and Choleretics/pharmacology , Dipyridamole/pharmacology , Erythromycin/pharmacology , Glycocholic Acid/antagonists & inhibitors , Glycocholic Acid/metabolism , Ketoconazole/pharmacology , Kinetics , Membrane Transport Proteins/metabolism , Sf9 Cells , Signal Transduction/drug effects , Spodoptera/virology , Taurocholic Acid/antagonists & inhibitors , Taurocholic Acid/metabolism , Transport Vesicles/metabolism
7.
Mol Pharm ; 15(9): 4073-4083, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30074800

ABSTRACT

Drug transporter expression and function at the blood-brain barrier is altered in Alzheimer's disease (AD). However, the impact of AD on the expression of transporters and metabolizing enzymes in peripheral tissues has received little attention. The current study evaluated the expression of drug transporters and metabolizing enzymes in the small intestine and liver from 8- to 9-month-old female wild-type (WT) and APPswe/PSEN 1dE9 (APP/PS1) transgenic mice, a widely used AD model, using a quantitative targeted absolute proteomics (QTAP) approach. Furthermore, the general morphological appearance of the liver was assessed by immunohistochemistry, and lipid content was visualized using Oil Red O staining. The small intestines of APP/PS1 mice exhibited a significant 2.3-fold increase in multidrug resistance-associated protein 2 (Mrp2), a 1.9-fold decrease in monocarboxylate transporter 1 (Mct1), and a 3.6-fold increase in UDP-glucuronosyltransferase (Ugt) 2b5 relative to those from WT mice based on QTAP analysis. While the liver from APP/PS1 mice exhibited no changes in drug transporter expression, there was a 1.3-fold elevation in cytochrome P450 (Cyp) 51a1 and a 1.2-fold reduction in Cyp2c29 protein expression, and this was associated with morphological alterations including accumulation of hepatocyte lipids. These studies are the first to demonstrate that the protein expression of transporters and metabolizing enzymes important in oral drug absorption are modified in a mouse model of familial AD, which may lead to altered disposition of some orally administered drugs in AD.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Intestine, Small/enzymology , Intestine, Small/metabolism , Liver/enzymology , Liver/metabolism , Animals , Blood-Brain Barrier/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2/metabolism , Disease Models, Animal , Female , Hepatocytes/enzymology , Hepatocytes/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Presenilin-1/metabolism , Proteomics
8.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G597-G609, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29420067

ABSTRACT

The heteromeric steroid transporter organic solute transporter α/ß (OSTα/ß, SLC51A/B) was discovered over a decade ago, but its physiological significance in the liver remains uncertain. A major challenge has been the lack of suitable models expressing OSTα/ß. Based on observations first reported here that hepatic OSTα/ß is upregulated in nonalcoholic steatohepatitis, the aim of this research was to develop an in vitro model to evaluate OSTα/ß function and interaction with drugs and bile acids. OSTα/ß expression in human liver tissue was analyzed by quantitative RT-PCR, Western blotting, and immunofluorescence. Radiolabeled compounds were used to determine OSTα/ß-mediated transport in the established in vitro model. The effect of bile acids and drugs, including those associated with cholestatic drug-induced liver injury, on OSTα/ß-mediated transport was evaluated. Expression of OSTα/ß was elevated in the liver of patients with nonalcoholic steatohepatitis and primary biliary cholangitis, whereas hepatocyte expression of OSTα/ß was low in control liver tissue. Studies in the novel cell-based system showed rapid and linear OSTα/ß-mediated transport for all tested compounds: dehydroepiandrosterone sulfate, digoxin, estrone sulfate, and taurocholate. The interaction study with 26 compounds revealed novel OSTα/ß inhibitors: a biomarker for cholestasis, glycochenodeoxycholic acid; the major metabolite of troglitazone, troglitazone sulfate; and a macrocyclic antibiotic, fidaxomicin. Additionally, some drugs (e.g., digoxin) consistently stimulated taurocholate uptake in OSTα/ß-overexpressing cells. Our findings demonstrate that OSTα/ß is an important transporter in liver disease and imply a role for this transporter in bile acid-bile acid and drug-bile acid interactions, as well as cholestatic drug-induced liver injury. NEW & NOTEWORTHY The organic solute transporter OSTα/ß is highly expressed in hepatocytes of liver tissue obtained from patients with nonalcoholic steatohepatitis and primary biliary cholangitis. OSTα/ß substrates exhibit rapid, linear, and concentration-driven transport in an OSTα/ß-overexpressing cell line. Drugs associated with hepatotoxicity modulate OSTα/ß-mediated taurocholate transport. These data suggest that hepatic OSTα/ß plays an essential role in patients with cholestasis and may have important clinical implications for bile acid and drug disposition.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Liver Cirrhosis, Biliary/metabolism , Membrane Transport Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Taurocholic Acid/metabolism , Bile Acids and Salts/metabolism , Biological Transport/physiology , Biomarkers/metabolism , Cell Line , Cholestasis/metabolism , Female , Glycochenodeoxycholic Acid/metabolism , Humans , Liver/metabolism , Male , Middle Aged
9.
Clin Pharmacol Ther ; 2017 Dec 22.
Article in English | MEDLINE | ID: mdl-29271075

ABSTRACT

The expression of hepatic transporters, including organic anion transporting polypeptides (OATPs) and multidrug resistance-associated proteins (MRPs), is altered in nonalcoholic steatohepatitis (NASH); however, functional data in humans are lacking. In this study, 99m Tc-mebrofenin (MEB) was used to evaluate OATP1B1/1B3 and MRP2 function in NASH patients. Healthy subjects (n = 14) and NASH patients (n = 7) were administered MEB (∼2.5 mCi). A population pharmacokinetic model was developed to describe systemic and hepatic MEB disposition. Study subjects were genotyped for SLCO1B1 variants. NASH increased systemic and hepatic exposure (median ± 2 SE, healthy vs. NASH) to MEB (AUC0-300,blood : 1,780 ± 242 vs. 2,440 ± 775 µCi*min/L, P = 0.006; AUC0-180,liver : 277 ± 36.9 vs. 433 ± 40.3 kcounts*min/sec, P < 0.0001) due to decreased biliary clearance (0.035 ± 0.008 vs. 0.017 ± 0.002 L/min, P = 0.0005) and decreased Vcentral (11.1 ± 0.57 vs. 6.32 ± 1.02 L, P < 0.0001). MEB hepatic CLuptake was reduced in NASH and also in healthy subjects with SLCO1B1 *15/*15 and *1A/*15 genotypes. The pharmacokinetics of drugs that are OATP1B1/1B3 and MRP2 substrates may be substantially altered in NASH.

10.
Eur J Pharm Sci ; 103: 52-59, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28238947

ABSTRACT

INTRODUCTION: Multidrug resistance-associated protein 3 (MRP3), an efflux transporter on the hepatic basolateral membrane, may function as a compensatory mechanism to prevent the accumulation of anionic substrates (e.g., bile acids) in hepatocytes. Inhibition of MRP3 may disrupt bile acid homeostasis and is one hypothesized risk factor for the development of drug-induced liver injury (DILI). Therefore, identifying potential MRP3 inhibitors could help mitigate the occurrence of DILI. METHODS: Bayesian models were developed using MRP3 transporter inhibition data for 86 structurally diverse drugs. The compounds were split into training and test sets of 57 and 29 compounds, respectively, and six models were generated based on distinct inhibition thresholds and molecular fingerprint methods. The six Bayesian models were validated against the test set and the model with the highest accuracy was utilized for a virtual screen of 1470 FDA-approved drugs from DrugBank. Compounds that were predicted to be inhibitors were selected for in vitro validation. The ability of these compounds to inhibit MRP3 transport at a concentration of 100µM was measured in membrane vesicles derived from stably transfected MRP3-over-expressing HEK-293 cells with [3H]-estradiol-17ß-d-glucuronide (E217G; 10µM; 5min uptake) as the probe substrate. RESULTS: A predictive Bayesian model was developed with a sensitivity of 73% and specificity of 71% against the test set used to evaluate the six models. The area under the Receiver Operating Characteristic (ROC) curve was 0.710 against the test set. The final selected model was based on compounds that inhibited substrate transport by at least 50% compared to the negative control, and functional-class fingerprints (FCFP) with a circular diameter of six atoms, in addition to one-dimensional physicochemical properties. The in vitro screening of predicted inhibitors and non-inhibitors resulted in similar model performance with a sensitivity of 64% and specificity of 70%. The strongest inhibitors of MRP3-mediated E217G transport were fidaxomicin, suramin, and dronedarone. Kinetic assessment revealed that fidaxomicin was the most potent of these inhibitors (IC50=1.83±0.46µM). Suramin and dronedarone exhibited IC50 values of 3.33±0.41 and 47.44±4.41µM, respectively. CONCLUSION: Bayesian models are a useful screening approach to identify potential inhibitors of transport proteins. Novel MRP3 inhibitors were identified by virtual screening using the selected Bayesian model, and MRP3 inhibition was confirmed by an in vitro transporter inhibition assay. Information generated using this modeling approach may be valuable in predicting the potential for DILI and/or MRP3-mediated drug-drug interactions.


Subject(s)
Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Aminoglycosides/pharmacology , Amiodarone/analogs & derivatives , Amiodarone/pharmacology , Bayes Theorem , Biological Transport , Cell Survival , Chemical and Drug Induced Liver Injury/drug therapy , Databases, Chemical , Dronedarone , Estradiol/analogs & derivatives , Estradiol/metabolism , Fidaxomicin , HEK293 Cells , Humans , Models, Molecular , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , Quantitative Structure-Activity Relationship , Suramin/pharmacology
11.
Eur J Pharmacol ; 715(1-3): 328-36, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23712005

ABSTRACT

The use of clonidine as a primary and adjuvant analgesic is well-documented. It is known that imidazoline and α2-adrenoceptors are involved in clonidine antinociception. Clonidine also produces antihypertensive actions mediated through the central nervous system. We have reported that centhaquin, a centrally-acting anti-hypertensive drug produces its hypotensive effect through a mechanism of action similar to clonidine. Centhaquin has also been shown to possess significant antinociceptive activity. Centhaquin antinociception is partially blocked by yohimbine, idazoxan, and naloxone; however, the involvement of specific adrenoceptor subtypes (α2A, α2B, or α2C) in centhaquin antinociception is unknown. The present study was conducted to determine antinociceptive properties of centhaquin citrate, a water soluble salt of centhaquin, and involvement of α2A-, α2B-, or α2C-adrenoceptors in centhaquin citrate antinociception in mice. BRL-44408 (α2A-adrenoceptor antagonist), imiloxan (α2B-adrenoceptor antagonist) and JP-1302 (α2C-adrenoceptor antagonist) were used to determine the involvement of α2A-, α2B-, or α2C-adrenoceptors, respectively. Antinociceptive (tail-flick and hot-plate) latencies were determined in male Swiss-Webster mice treated with centhaquin citrate alone and in combination with BRL-44408, imiloxan, or JP-1302. Centhaquin citrate produced significant antinociception in mice (P<0.05) which was unaffected by JP-1302 (P>0.05) but blocked by BRL-44408 (tail-flick test: 49.75% decrease, P<0.05; hot-plate test: 49.12% decrease, P<0.05) and imiloxan (tail-flick test: 46.98% decrease, P<0.05; hot-plate test: 46.42% decrease, P<0.05). This is the first report demonstrating centhaquin citrate antinociception and its blockade by BRL-44408 and imiloxan. We conclude that α2A and α2B but not α2C adrenoceptors are involved in centhaquin antinociception in mice.


Subject(s)
Analgesics/pharmacology , Piperazines/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Mice , Nociception/drug effects
12.
Pharmacol Biochem Behav ; 103(3): 550-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23103903

ABSTRACT

The potentiation of oxycodone antinociception by BMS182874 (endothelin-A (ET(A)) receptor antagonist) and agmatine (imidazoline receptor/α(2)-adrenoceptor agonist) is well-documented. It is also known that imidazoline receptors but not α(2)-adrenoceptors are involved in potentiation of oxycodone antinociception by agmatine and BMS182874 in mice. However, the involvement of specific imidazoline receptor subtypes (I(1), I(2), or both) in this interaction is not clearly understood. The present study was conducted to determine the involvement of imidazoline I(1) and I(2) receptors in agmatine- and BMS182874-induced potentiation of oxycodone antinociception in mice. Antinociceptive (tail flick and hot-plate) latencies were determined in male Swiss Webster mice treated with oxycodone, agmatine, BMS182874, and combined administration of oxycodone with agmatine or BMS182874. Efaroxan (imidazoline I(1) receptor antagonist) and BU224 (imidazoline I(2) receptor antagonist) were used to determine the involvement of I(1) and I(2) imidazoline receptors, respectively. Oxycodone produced significant antinociceptive response in mice which was not affected by efaroxan but was blocked by BU224. Agmatine-induced potentiation of oxycodone antinociception was blocked by BU224 but not by efaroxan. Similarly, BMS182874-induced potentiation of oxycodone antinociception was blocked by BU224 but not by efaroxan. This is the first report demonstrating that BMS182874- or agmatine-induced enhancement of oxycodone antinociception is blocked by BU224 but not by efaroxan. We conclude that imidazoline I(2) receptors but not imidazoline I(1) receptors are involved in BMS182874- and agmatine-induced potentiation of oxycodone antinociception in mice.


Subject(s)
Agmatine/pharmacology , Analgesics, Opioid/pharmacology , Dansyl Compounds/pharmacology , Imidazoline Receptors/metabolism , Oxycodone/pharmacology , Agmatine/antagonists & inhibitors , Analgesics, Opioid/antagonists & inhibitors , Animals , Benzofurans/pharmacology , Drug Synergism , Imidazoles/pharmacology , Imidazoline Receptors/agonists , Imidazoline Receptors/antagonists & inhibitors , Male , Mice , Oxycodone/antagonists & inhibitors , Pain Measurement
SELECTION OF CITATIONS
SEARCH DETAIL
...