Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Transl Med ; 11(516)2019 10 30.
Article in English | MEDLINE | ID: mdl-31666405

ABSTRACT

Canine studies of spontaneous osteoarthritis (OA) pain add valuable data supporting drug treatment mechanisms that may translate to humans. A multicenter, randomized, double-blind, placebo- and active-controlled study was conducted in client-owned dogs with moderate OA pain to evaluate efficacy of LYA, an inhibitor of microsomal prostaglandin E synthase-1 (mPGES1), an EP4 antagonist (LYB), and carprofen, versus placebo. Of 255 dogs screened, 163 were randomized (placebo/LYA/LYB/carprofen: n = 43/39/42/39) and 158 completed treatment. Efficacy versus placebo was assessed using Bayesian mixed-effect model for repeated measure analyses of the Canine Brief Pain Inventory (CBPI) pain interference score (PIS; primary endpoint), pain severity score, and overall impression, as well as the Liverpool Osteoarthritis in Dogs (LOAD) mobility score. The posterior probability that the difference to placebo was <0 at week 2 was 80% for LYA and 54% for LYB for CBPI PIS (both <95% predefined threshold). For secondary endpoints, the posterior probability that the difference to placebo was <0 at week 2 ranged from 89 to 96% for LYA and from 56 to 89% for LYB. The posterior probabilities comparing carprofen to placebo groups were ≥90% for all efficacy endpoints. The proportion of dogs with one or more adverse event was not significantly different from placebo (32.6%) for LYA (35.9%) or carprofen (25.6%), but the rate for LYB (59.5%) was higher versus placebo (P = 0.017). LYA treatment demonstrated consistent improvement in all efficacy measures, suggesting that inhibition of mPGES1 may be an effective treatment for chronic pain associated with OA.


Subject(s)
Osteoarthritis/drug therapy , Osteoarthritis/veterinary , Prostaglandin-E Synthases/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Animals , Dogs , Endpoint Determination , Female , Follow-Up Studies , Male , Placebos , Probability , Prostaglandin-E Synthases/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Treatment Outcome
2.
Br J Clin Pharmacol ; 84(1): 179-188, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28865237

ABSTRACT

AIMS: LY3031207, a novel microsomal prostaglandin E synthase 1 inhibitor, was evaluated in a multiple ascending dose study after nonclinical toxicology studies and a single ascending dose study demonstrated an acceptable toxicity, safety and tolerability profile. METHODS: Healthy subjects were randomized to receive LY3031207 (25, 75 and 275 mg), placebo or celecoxib (400 mg) once daily for 28 days. The safety, tolerability and pharmacokinetic and pharmacodynamic profiles of LY3031207 were evaluated. RESULTS: The study was terminated when two subjects experienced drug-induced liver injury (DILI) after they had received 225 mg LY3031207 for 19 days. Liver biopsy from these subjects revealed acute liver injury with eosinophilic infiltration. Four additional DILI cases were identified after LY3031207 dosing had been stopped. All six DILI cases shared unique presentations of hepatocellular injury with hypersensitivity features and demonstrated a steep dose-dependent trend. Prompt discontinuation of the study drug and supportive medical care resulted in full recovery. Metabolites from metabolic activation of the imidazole ring were observed in plasma and urine samples from all subjects randomized to LY3031207 dosing. CONCLUSIONS: This study emphasized the importance of careful safety monitoring and serious adverse events management in phase I trials. Metabolic activation of the imidazole ring may be involved in the development of hepatotoxicity of LY3031207.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug Hypersensitivity/etiology , Enzyme Inhibitors/adverse effects , Imidazoles/adverse effects , Prostaglandin-E Synthases/antagonists & inhibitors , Administration, Oral , Adult , Area Under Curve , Celecoxib/adverse effects , Chemical and Drug Induced Liver Injury/pathology , Cyclooxygenase 2 Inhibitors/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Drug Hypersensitivity/pathology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Female , Half-Life , Healthy Volunteers , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Middle Aged , Pain/drug therapy , Withholding Treatment
3.
Clin Transl Sci ; 11(1): 46-53, 2018 01.
Article in English | MEDLINE | ID: mdl-28857461

ABSTRACT

Safety, tolerability, and pharmacology profiles of LY3127760, an EP4 antagonist, were explored in healthy subjects in a subject/investigator-blind, parallel-group, multiple-ascending dose study. Cohorts consisted of 13 patients randomized to LY3127760, celecoxib (400 mg), or placebo (9:2:2 ratio) for 28 days. LY3127760 was well tolerated; the most commonly observed adverse events were gastrointestinal, similar to celecoxib. LY3127760 increased release of ex vivo tumor necrosis factor alpha after lipopolysaccharide/prostaglandin E2 stimulation when compared with placebo, suggesting a dose-dependent blockade of the EP4 receptor. Compared with placebo, 24-h urinary excretion of prostaglandin E metabolite was modestly increased; prostacyclin metabolite was inhibited; and thromboxane A2 metabolite was unchanged. Effects on sodium and potassium excretion were similar to those of celecoxib. We conclude that LY3127760 demonstrated similar effects on prostacyclin synthesis and renal sodium retention as celecoxib. These data support exploration of LY3127760 at daily doses of 60 mg to 600 mg in phase II trials. This trial's registration number: NCT01968070.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthralgia/drug therapy , Celecoxib/pharmacology , Organic Chemicals/pharmacology , Osteoarthritis/drug therapy , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Administration, Oral , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthralgia/etiology , Celecoxib/therapeutic use , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , Organic Chemicals/chemistry , Osteoarthritis/complications , Placebos , Young Adult
4.
Br J Clin Pharmacol ; 81(5): 908-17, 2016 May.
Article in English | MEDLINE | ID: mdl-26648084

ABSTRACT

AIMS: Two phase 1 studies evaluated the pharmacokinetics (PK), safety and biological activity of tabalumab, a human monoclonal antibody against B-cell activating factor (BAFF), administered intravenously (i.v.) or subcutaneously (s.c.) in subjects with rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE). METHODS: In study A, subjects with RA (n = 23) or SLE (n = 6) received a single i.v. dose of tabalumab (RA 0.01, 0.04, 0.125, 0.5, 2.0, and 8.0 mg kg(-1) and SLE 0.125 or 2.0 mg kg(-1) ) or placebo. In study B, subjects with RA received a single tabalumab dose i.v. (10 mg) (n = 12) or s.c. (20 mg) (n = 12). Serum tabalumab and CD20+ B cells were evaluated and safety was assessed throughout both studies. RESULTS: Tabalumab PK were non-linear across the 0.01 to 8.0 mg kg(-1) dose range. Clearance (CL) decreased from 2.9 to 0.1 l day(-1) and terminal half-life (t1/2 ) increased from about 1.6 to 25 days. Subjects with RA or SLE had similar PK. After s.c. dosing, tabalumab time to maximal concentration (tmax ) was 5.5 days. Absolute bioavailability (F) was approximately 62%. Following tabalumab dosing, CD20+ B cells transiently increased from baseline followed by a progressive decrease below baseline. CONCLUSION: A single tabalumab dose administered i.v. or s.c. was well tolerated and had non-linear CL over the dose range investigated in subjects with RA and SLE. The non-linearity likely reflects target-mediated CL due to binding to BAFF. Tabalumab showed biological activity based on changes in peripheral CD20+ lymphocyte numbers in both subjects with RA and SLE.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Arthritis, Rheumatoid/drug therapy , B-Cell Activating Factor/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antigens, CD20/immunology , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , Female , Humans , Injections, Intravenous , Injections, Subcutaneous , Male , Middle Aged
5.
Lancet Diabetes Endocrinol ; 3(12): 948-57, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26516121

ABSTRACT

BACKGROUND: Myostatin inhibits skeletal muscle growth. The humanised monoclonal antibody LY2495655 (LY) binds and neutralises myostatin. We aimed to test whether LY increases appendicular lean body mass (aLBM) and improves physical performance in older individuals who have had recent falls and low muscle strength and power. METHODS: In this proof-of-concept, randomised, placebo-controlled, double-blind, parallel, multicentre, phase 2 study, we recruited patients aged 75 years or older who had fallen in the past year from 21 investigator sites across Argentina, Australia, France, Germany, Sweden, and the USA. Eligible patients had low performance on hand grip strength and chair rise tests, tested with the procedure described by Guralnik and colleagues. Participants were stratified by country, age, hand grip strength, and performance on the chair rise test, and were randomly assigned (1:1) by a computer-generated random sequence to receive subcutaneous injections of placebo or 315 mg LY at weeks 0 (randomisation visit), 4, 8, 12, 16, and 20, followed by 16 weeks observation. The primary outcome was change in aLBM from baseline to 24 weeks. We measured physical performance as secondary outcomes (four-step stair climbing time, usual gait speed, and time to rise five times from a chair without arms, or with arms for participants unable to do it without arms) and exploratory outcomes (12-step stair climbing test, 6-min walking distance, fast gait speed, hand grip strength, and isometric leg extension strength). Efficacy analyses included all randomly assigned patients who received at least one dose and had a baseline and at least one subsequent measure. The primary analysis and all other tests of treatment effect (except physical performance tests) were done at a two-sided alpha level of 0·05. Tests of treatment effect on physical performance tests were done at a pre-specified two-sided alpha level of 0·1. This trial is registered with ClinicalTrials.gov, number NCT01604408. FINDINGS: Between June 19, 2012, and Dec 12, 2013, we screened 365 patients. 99 were randomly assigned to receive placebo and 102 to receive LY. Treatment was completed in 85 (86%) of patients given placebo and in 82 (80%) given LY. At 24 weeks, the least-squares mean change in aLBM was -0·123 kg (95% CI -0·287 to 0·040) in the placebo group and 0·303 kg (0·135 to 0·470) in the LY group, a difference of 0·43 kg (95% CI 0·192 to 0·660; p<0·0001). Stair climbing time (four-step and 12-step tests), chair rise with arms, and fast gait speed improved significantly from baseline to week 24 with differences between LY and placebo of respectively -0·46 s (p=0·093), -1·28 s (p=0·011), -4·15 s (p=0·054), and 0·05 m/s (p=0·088). No effect was detected for other performance-based measures. Injection site reactions were recorded in nine (9%) patients given placebo and in 31 (30%) patients given LY (p<0·0001), and were generally mild, and led to treatment discontinuation in two patients given LY. INTERPRETATION: Our findings show LY treatment increases lean mass and might improve functional measures of muscle power. Although additional studies are needed to confirm these results, our data suggest LY should be tested for its potential ability to reduce the risk of falls or physical dependency in older weak fallers. FUNDING: Eli Lilly and Company.


Subject(s)
Accidental Falls , Antibodies, Monoclonal, Humanized/therapeutic use , Muscle Weakness/drug therapy , Muscle, Skeletal/drug effects , Myostatin/antagonists & inhibitors , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Double-Blind Method , Female , Gait/drug effects , Hand Strength , Humans , Injections, Subcutaneous , Male , Myostatin/immunology , Treatment Outcome
6.
J Bone Miner Res ; 30(9): 1717-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25707611

ABSTRACT

Administration of blosozumab, a humanized monoclonal antibody that binds sclerostin, increases bone formation and bone mineral density (BMD) in postmenopausal women with low BMD. To evaluate the effect of discontinuing blosozumab, we studied women enrolled in a 1-year randomized, placebo-controlled phase 2 trial for an additional year after they completed treatment. Of the 120 women initially enrolled in the study, 106 women completed treatment and continued into follow-up; 88 women completed 1 year of follow-up. At the beginning of follow-up, groups remained balanced for age, race, and body mass index, but lumbar spine and total hip BMD were increased in prior blosozumab groups, reflecting an anabolic treatment effect. At the end of follow-up, 1 year after discontinuing treatment, lumbar spine BMD remained significantly greater than placebo in women initially treated with blosozumab 270 mg every 2 weeks (Q2W) and blosozumab 180 mg Q2W (6.9% and 3.6% above baseline, respectively). Total hip BMD also declined after discontinuation of treatment but at 1 year after treatment remained significantly greater than placebo in women initially treated with blosozumab 270 mg Q2W and blosozumab 180 mg Q2W (3.9% and 2.6% above baseline, respectively). During follow-up, median serum P1NP was not consistently different between the prior blosozumab groups and placebo. A similar pattern was apparent for median serum C-terminal telopeptide of type 1 collagen (CTx) levels, with more variability. Mean serum total sclerostin concentration increased with blosozumab, indicating target engagement, and declined to baseline after discontinuation. There were no adverse events considered related to prior treatment with blosozumab. Anti-drug antibodies generally declined in patients who had detectable levels during prior treatment. These findings support the continued study of blosozumab as an anabolic therapy for treatment of osteoporosis.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Bone Density , Bone Morphogenetic Proteins/blood , Osteoporosis, Postmenopausal/drug therapy , Postmenopause , Adaptor Proteins, Signal Transducing , Aged , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized/blood , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/blood , Bone Morphogenetic Proteins/immunology , Bone Resorption , Double-Blind Method , Drug Administration Schedule , Female , Follow-Up Studies , Genetic Markers/immunology , Humans , Middle Aged , Osteogenesis , Steroids/chemistry , Time Factors
7.
J Bone Miner Res ; 30(2): 216-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25196993

ABSTRACT

Sclerostin, a SOST protein secreted by osteocytes, negatively regulates formation of mineralized bone matrix and bone mass. We report the results of a randomized, double-blind, placebo-controlled multicenter phase 2 clinical trial of blosozumab, a humanized monoclonal antibody targeted against sclerostin, in postmenopausal women with low bone mineral density (BMD). Postmenopausal women with a lumbar spine T-score -2.0 to -3.5, inclusive, were randomized to subcutaneous blosozumab 180 mg every 4 weeks (Q4W), 180 mg every 2 weeks (Q2W), 270 mg Q2W, or matching placebo for 1 year, with calcium and vitamin D. Serial measurements of spine and hip BMD and biochemical markers of bone turnover were performed. Overall, 120 women were enrolled in the study (mean age 65.8 years, mean lumbar spine T-score -2.8). Blosozumab treatment resulted in statistically significant dose-related increases in spine, femoral neck, and total hip BMD as compared with placebo. In the highest dose group, BMD increases from baseline reached 17.7% at the spine, and 6.2% at the total hip. Biochemical markers of bone formation increased rapidly during blosozumab treatment, and trended toward pretreatment levels by study end. However, bone specific alkaline phosphatase remained higher than placebo at study end in the highest-dose group. CTx, a biochemical marker of bone resorption, decreased early in blosozumab treatment to a concentration less than that of the placebo group by 2 weeks, and remained reduced throughout blosozumab treatment. Mild injection site reactions were reported more frequently with blosozumab than placebo. In conclusion, treatment of postmenopausal women with an antibody targeted against sclerostin resulted in substantial increases in spine and hip BMD. These results support further study of blosozumab as a potential anabolic therapy for osteoporosis.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Bone Density/drug effects , Bone Morphogenetic Proteins/immunology , Genetic Markers/immunology , Postmenopause , Adaptor Proteins, Signal Transducing , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/metabolism , Bone Remodeling/drug effects , Double-Blind Method , Female , Humans , Osteoporosis, Postmenopausal/drug therapy , Placebos , Postmenopause/drug effects , Treatment Outcome
8.
Clin Pharmacol Drug Dev ; 4(4): 305-14, 2015 07.
Article in English | MEDLINE | ID: mdl-27136911

ABSTRACT

BACKGROUND: LY500307 is a highly selective estrogen receptor ß (ERß) agonist, which loses its selectivity at high dose and leads to undesirable suppression of total testosterone (TT) concentration. The objective of the present analysis was to define the LY500307 dose with minimal effect on TT METHODS: LY500307 and TT concentrations were obtained from a single ascending-dose study in a total of 30 healthy male subjects. LY500307 (in the range of 0.5 to 500 mg) or placebo was administered orally as a single dose on 2 occasions with a 3-week washout period. A population pharmacokinetics/pharmacodynamics (PK/PD) model that integrated Fourier series in an indirect response model was developed to describe the circadian rhythm of TT and the exposure-response relationship of LY500307 on TT. RESULTS: The maximum TT suppression (Emax ) was approximately 28.6%. The potency (EC50 ) of LY500307 on TT suppression was approximately 1.69 ng/mL with a 95%CI of 0.871 to 4.44 ng/mL. This model could provide inferences on LY500307 dose levels that would result in various magnitudes of TT suppression. CONCLUSIONS: Population PK/PD modeling is a highly sensitive tool to detect exposure-response relationships on top of the complicated and highly variable circadian rhythm of TT.


Subject(s)
Benzopyrans/administration & dosage , Benzopyrans/pharmacokinetics , Estrogen Receptor beta/agonists , Selective Estrogen Receptor Modulators/administration & dosage , Selective Estrogen Receptor Modulators/pharmacokinetics , Testosterone/blood , Administration, Oral , Adult , Benzopyrans/adverse effects , Benzopyrans/blood , Biomarkers/blood , Circadian Rhythm , Cross-Over Studies , Drug Administration Schedule , Estrogen Receptor beta/metabolism , Fourier Analysis , Healthy Volunteers , Humans , London , Male , Middle Aged , Models, Biological , Nonlinear Dynamics , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/blood , Single-Blind Method , Young Adult
9.
J Bone Miner Res ; 29(4): 935-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23996473

ABSTRACT

Two clinical studies were conducted to assess the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple doses (intravenous [iv] and subcutaneous [sc]) of blosozumab in postmenopausal women, including prior/current bisphosphonate (BP) users. In these phase 1, randomized, subject- and investigator-blind, placebo-controlled studies, subjects received escalating doses of blosozumab: single iv doses up to 750 mg, single sc doses of 150 mg, multiple iv doses up to 750 mg every 2 weeks (Q2W) for 8 weeks, multiple sc doses up to 270 mg Q2W for 8 weeks, or placebo. Six subjects were randomized to each dose in the single-dose study (12 to placebo) and up to 12 subjects to each arm in the multiple-dose study. Blosozumab was well tolerated with no safety concerns identified after single or multiple administrations up to 750 mg. Dose-dependent responses were observed in sclerostin, N-terminal propeptide of procollagen type 1, bone-specific alkaline phosphatase, osteocalcin, C-terminal fragment of type 1 collagen, and bone mineral density (BMD) after single and multiple (up to 5) administrations of blosozumab. There was up to a 3.41% (p=0.002) and up to a 7.71% (p<0.001) change from baseline in lumbar spine BMD at day 85 after single or multiple administrations of blosozumab, respectively. Prior BP use did not appear to have a clear impact on the effects of single doses of blosozumab when considering bone biomarker and BMD responses. Antibodies to blosozumab were detected by a screening assay, but no patterns with regard to dose or route of administration and no clear impact on blosozumab exposure or PD responses were identified. In summary, blosozumab was well tolerated and exhibited anabolic effects on bone. These findings support further investigation of blosozumab as a potential anabolic therapy for osteoporosis.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Bone Morphogenetic Proteins/immunology , Genetic Markers/immunology , Postmenopause , Adaptor Proteins, Signal Transducing , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Bone Density , Female , Humans , Middle Aged
10.
J Pharm Sci ; 102(9): 2898-908, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23508847

ABSTRACT

Over the last two decades, there has been a simultaneous explosion in the levels of activity and capability in both monoclonal antibody (mAb) drug development and in the use of quantitative pharmacologic models to facilitate drug development. Both of these topics are currently areas of great interest to academia, the pharmaceutical and biotechnology industries, and to regulatory authorities. In this article, we summarize convergence of these two areas and discuss some of the current and historical applications of the use of mathematical-model-based techniques to facilitate the discovery and development of mAb therapeutics. We also consider some of the current issues and limitations in model-based antibody discovery/development and highlight areas of further opportunity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Drug Discovery/methods , Animals , Computational Biology , Humans , Models, Biological
11.
AAPS J ; 12(4): 586-91, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20625863

ABSTRACT

Suramin, at non-cytotoxic doses, reverses chemoresistance and enhances the activity of mitomycin C (MMC) in mice bearing human bladder xenograft tumors. The present study evaluated the pharmacokinetics of the intravesical suramin and MMC, alone or in combination, in dogs. Animals received either high dose suramin (20 mg/ml), low dose suramin (6 mg/ml), MMC (2 mg/ml), or combination of low dose suramin and MMC, instilled for 2 h. The dosing volume was 20 ml. All groups showed dilution of drug levels over time due to continued urine production. For single agent suramin, the results showed (a) 5% to 10% penetration into bladder tissues, (b) minimal and clinically insignificant systemic absorption (i.e., undetectable at low dose or a peak concentration that was 6,000× lower than urine concentrations), and (c) disproportionally higher drug penetration and concentrations in bladder tissues at the higher dose. Results for single agent MMC are consistent with our earlier observations. The co-administration of MMC did not alter the plasma, urine, or tissue pharmacokinetics of suramin. Adding suramin did not alter plasma or tissue pharmacokinetics of MMC, but lowered the MMC concentrations in urine by about 20%. This may be in part due to accelerated MMC degradation by co-incubation of suramin or due to variations in urine production rate (because animals were allowed for water during treatment). Suramin readily penetrates the urothelium and into deeper bladder tissues, indicating its potential utility in intravesical therapy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Mitomycin/pharmacokinetics , Suramin/pharmacokinetics , Urinary Bladder/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/urine , Dogs , Female , Male , Mitomycin/administration & dosage , Mitomycin/urine , Suramin/administration & dosage , Suramin/urine
12.
Clin Cancer Res ; 13(4): 1278-87, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17317840

ABSTRACT

PURPOSE: To evaluate a computational approach that incorporates experimental data in preclinical models to depict doxorubicin human tissue pharmacokinetics. EXPERIMENTAL DESIGN: Beagle dogs were given 2 mg/kg doxorubicin as i.v. bolus, 4-h infusion, or 96-h infusion. Concentrations in plasma, prostate (target tissue), heart (toxicity), and major tissues for disposition were determined and modeled. Model parameters were obtained after the bolus injection with model validation based on the 4-h and 96-h infusion data. Clinical pharmacokinetic data and scale-up gave doxorubicin profiles in human prostate and heart. RESULTS: In agreement with in vitro results, tissues were best modeled with two compartments, one rapidly and one slowly equilibrating. The developed tissue distribution model predicted concentrations for all three administration regimens well, with an average deviation of 34% (median, 29%). Interspecies scale-up to humans showed that the change from a bolus injection to a slow, 96-h infusion (a) had different effects on the drug partition and accumulation in heart and prostate, and (b) lowered the peak concentration in the plasma by approximately 100-fold but had relatively little effect on maximal heart concentration ( approximately 33% lower). The simulated drug exposure in a human prostate was above the exposure required to inhibit tumor proliferation but was 30 to 50 times below that needed for cell death. CONCLUSION: The present study shows a computation-based paradigm for translating in vitro and in vivo preclinical data and to estimate and compare the drug delivery and pharmacokinetics in target tissues after different treatment schedules.


Subject(s)
Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Delivery Systems , Models, Biological , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Computer Simulation , Dogs , Drug Administration Schedule , Humans , Male
13.
Clin Cancer Res ; 11(11): 4204-11, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930358

ABSTRACT

PURPOSE: The present study evaluated the tissue distribution and targeting advantage of intraprostatic chemotherapy. EXPERIMENTAL DESIGN: We studied the delivery and spatial distribution of a fluorescent drug, doxorubicin, in the prostate of beagle dogs, after intraprostatic or i.v. administration. Drug concentrations were measured using high-performance liquid chromatography and confocal fluorescence microscopy. RESULTS: I.v. and intraprostatic injections yielded qualitatively and quantitatively different doxorubicin distribution in the prostate. A relatively homogeneous distribution was found after i.v. administration, whereas intraprostatic injection yielded a highly heterogeneous distribution with >10-fold higher concentrations localized in a cone-shaped glandular lobule bound by fibromuscular stroma, compared with other parts of the prostate. Compared with i.v. injection, intraprostatic injection yielded, on average, approximately 100-fold higher tissue-to-plasma concentration ratio, ranging from 963-fold near the injection site to 19-fold in the contralateral half of the prostate. The drug distribution within the prostate further suggests an important role for acinar flow in intraprostatic drug transport. CONCLUSIONS: Intraprostatic administration represents a viable option to deliver high drug concentrations within the prostate. The results further suggest the fibromuscular stroma separating the prostatic lobules as a major barrier to drug transport and convective flow as an important drug transport mechanism in the prostate.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Prostate/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Biological Transport , Dogs , Doxorubicin/administration & dosage , Doxorubicin/blood , Infusions, Intravenous , Lymph Nodes/metabolism , Male , Microscopy, Fluorescence , Models, Biological , Prostate/drug effects , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...