Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
1.
Drug Dev Res ; 85(3): e22191, 2024 May.
Article in English | MEDLINE | ID: mdl-38685610

ABSTRACT

Psoriasis is a chronic inflammatory and proliferative skin disease that causes pathological skin changes and has a substantial impact on the quality of patient life. Apremilast was approved by the US Food and Drug Administration as an oral medication for psoriasis and is beneficial in mild to moderate conditions for chronic usage. However, 5%-7% of withdrawals were reported due to severe side effects. To address the issue, a localized drug delivery strategy via the topical route may be a viable approach. However, poor physicochemical properties make it vulnerable to passing through the skin, requiring a specialized drug delivery system to demonstrate its full potential via a topical route like lecithin organogel. The formulation was optimized by screening the suitable lecithin type and non-polar solvents based on the gel formation ability of lecithin and the solubility of apremilast in the solvent. The pseudo-ternary diagram was used to optimize the water content required to form the gel. The optimized gel was found to be shear thinning characterized for rheological parameters, in-vitro diffusion studies, and in-vitro skin distribution studies. Preclinical studies in Imiquimod-induced mice showed a better reduction in severity index, cytokine levels, and epidermal hyperplasia from the lecithin organogel group compared to the apremilast oral administration and marketed standard topical gel group. Based on these results, lecithin organogel can be considered a promising approach to deliver molecules like apremilast by topical route in psoriatic-like conditions.


Subject(s)
Drug Delivery Systems , Gels , Lecithins , Psoriasis , Thalidomide , Thalidomide/analogs & derivatives , Psoriasis/drug therapy , Lecithins/chemistry , Animals , Mice , Thalidomide/administration & dosage , Thalidomide/chemistry , Thalidomide/pharmacokinetics , Skin Absorption/drug effects , Skin/metabolism , Skin/drug effects , Administration, Cutaneous , Administration, Topical , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Drug Evaluation, Preclinical , Imiquimod/administration & dosage , Male
2.
Invest New Drugs ; 41(1): 60-69, 2023 02.
Article in English | MEDLINE | ID: mdl-36441437

ABSTRACT

OBJECTIVE: Imnovid® is an immunomodulatory drug with antineoplastic activity. The aim of this study was to evaluate the bioequivalence and safety of the generic drug pomalidomide (Chia Tai Tianqing Pharmaceutical Group Co., Ltd) and its originator product Imnovid® (Celgene Europe Ltd) in the fasting and fed states, respectively. METHODS: The research consisted of two parts: one with a dose of 1 mg and the other with a dose of 4 mg. 48 healthy subjects were included in each study and were divided into two groups (fasting group and fed group) at a 1:1 ratio to administrate study drugs orally. The plasma drug concentrations were determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS: The 90% CI of GMR for main pharmacokinetic (PK) parameters (Cmax, AUC0 - t and AUC0-∞) met the requirements of bioequivalence standards. The incidence and severity of AEs associated with pomalidomide and Imnovid® were similar. CONCLUSION: The results proved the PK parameters of pomalidomide and Imnovid® were similar and bioequivalent. Both drugs showed safety profile well.


Subject(s)
Thalidomide , Therapeutic Equivalency , Humans , Area Under Curve , Capsules , Chromatography, Liquid , Cross-Over Studies , East Asian People , Fasting , Healthy Volunteers , Tablets , Tandem Mass Spectrometry , Thalidomide/analogs & derivatives , Thalidomide/pharmacokinetics
3.
J Toxicol Sci ; 46(7): 311-317, 2021.
Article in English | MEDLINE | ID: mdl-34193768

ABSTRACT

The approved drug thalidomide is teratogenic in humans, nonhuman primates, and rabbits but not in rodents. The extensive biotransformation of 5'-hydroxythalidomide after oral administration of thalidomide (250 mg/kg) in rats was investigated in detail using liquid chromatography-tandem mass spectrometry. Probable metabolites 5'-hydroxythalidomide sulfate and glucuronide were extensively formed, with approximately tenfold and onefold peak areas, respectively, to the primary 5'-hydroxythalidomide measured using authentic standards. As a minor metabolite, 5-hydroxythalidomide was also detected. The output of simplified physiologically based pharmacokinetic rat models was consistent with the observed in vivo data under a metabolic ratio of 0.05 for the hepatic intrinsic clearance of thalidomide to unconjugated 5'-hydroxythalidomide. The aggregate of unconjugated and sulfate/glucuronide conjugated 5'-hydroxythalidomide forms appear to be the predominant metabolites in rats. Two hours after oral administration of thalidomide (100 mg/kg) to chimeric mice humanized with four different batches of genotyped human hepatocytes, the plasma concentration ratios of 5-hydroxythalidomide to 5'-hydroxythalidomide were correlated with replacement indexes of human liver cells previously transplanted in immunodeficient mice. These results indicate that rodent livers mediate thalidomide primary oxidation, leading to extensive deactivation in vivo to unconjugated/conjugated 5'-hydroxythalidomide and suggest that thalidomide activation might be dependent on the humanized livers in mice transplanted with human hepatocytes.


Subject(s)
Hepatocytes/drug effects , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Teratogens/pharmacokinetics , Teratogens/toxicity , Thalidomide/pharmacokinetics , Thalidomide/toxicity , Animals , Humans , Male , Metabolic Networks and Pathways , Mice , Models, Animal , Rats , Species Specificity , Teratogens/metabolism , Thalidomide/analogs & derivatives , Thalidomide/metabolism
4.
Clin Transl Sci ; 14(4): 1505-1511, 2021 07.
Article in English | MEDLINE | ID: mdl-33932093

ABSTRACT

We performed a two-part study to evaluate the pharmacokinetics, safety, and tolerability of oral apremilast, a phosphodiesterase 4 inhibitor indicated for the treatment of psoriasis, in healthy Korean adult men. In part 1, there were 12 subjects who randomly received a single oral dose of apremilast at 20, 30, or 40 mg in each of 3 periods in a crossover fashion. In part 2, there were 16 subjects who randomly received 30 mg of apremilast or its matching placebo in a ratio of 3:1 twice daily for 14 days. Apremilast was rapidly absorbed (maximum concentration: ~2-3 h postdose), and eliminated according to a monoexponential pattern with a terminal-phase elimination half-life of 8-9 h. The exposure to apremilast increased in a dose-proportional manner and accumulation was 1.6-fold at steady-state. Apremilast was well-tolerated after a single oral administration and multiple oral administrations in Korean adult men; all of the treatment-emergent adverse events were mild and recovered without sequelae. In conclusion, apremilast was safe and well-tolerated in healthy Korean adult men when administered single oral doses of 20, 30, or 40 mg or when administered multiple oral doses of 30 mg b.i.d. for 14 days. Overall exposures increased in an approximate dose proportional manner in healthy Korean adult men.


Subject(s)
Phosphodiesterase 4 Inhibitors/pharmacokinetics , Thalidomide/analogs & derivatives , Administration, Oral , Adolescent , Adult , Area Under Curve , Asian People , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/adverse effects , Psoriasis/drug therapy , Republic of Korea , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/pharmacokinetics , Young Adult
5.
AAPS PharmSciTech ; 22(4): 142, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33893566

ABSTRACT

Apremilast (APST) is an effective inhibitor of phosphodieasterase 4 (PDE4) which is the first oral drug for the treatment of adult patients with active psoriatic arthritis. However, Apremilast's low solubility restricts its dissolution and bioavailability. In this study, APST solid dispersion with D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and Poly(1-vinylpyrrolidone-co-vinyl acetate) (PVPVA) was developed to improve the dissolution and bioavailability of APST by spray drying. A series of TPGS were synthesized to elucidate the effect of the ratio of monoester to diester on solubilizing capacity. X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), and Fourier transform infrared spectrophotometry (FT-IR) were used to characterize the solid dispersion, and the results showed that APST was amorphous in solid dispersion. In vitro dissolution study showed that the dissolution rate of solid dispersion in phosphate buffered saline (pH 6.8) was remarkably increased, reaching a release of 90% within 10 min. Moreover, in vivo pharmacokinetics study revealed that the bioavailability of solid dispersion in rats had significant improvement. In particular, its Cmax and AUClast were nearly 22- and 12.9-fold greater as compared to APST form B, respectively. In conclusion, APST solid dispersion with TPGS and PVPVA is an alternative drug delivery system to improve the solubility and oral bioavailability of APST.


Subject(s)
Dosage Forms , Phosphodiesterase 4 Inhibitors/chemistry , Povidone/analogs & derivatives , Thalidomide/analogs & derivatives , Vitamin E/chemistry , Animals , Biological Availability , Calorimetry, Differential Scanning , Phosphodiesterase 4 Inhibitors/pharmacokinetics , Povidone/chemistry , Powder Diffraction , Rats , Solubility , Spectroscopy, Fourier Transform Infrared , Thalidomide/chemistry , Thalidomide/pharmacokinetics
6.
Crit Rev Anal Chem ; 51(3): 258-267, 2021.
Article in English | MEDLINE | ID: mdl-32024370

ABSTRACT

Apremilast is an anti-inflammatory agent. It has been a flourishing molecule in the field of dermatology. In the year 2014, Apremilast got its approval for treatment of psoriatic arthritis. Presently it is known to treat a number of other conditions, including atopic dermatitis and plaque psoriasis. Apremilast a phthalimide derivative, is non-hygroscopic in nature. It is practically insoluble in water. Apremilast acts by inhibiting the activity of phosphodiesterase 4 (PDE4), an intracellular enzyme. Analytical method plays a key role to understand the physio-chemical properties of a drug molecule. Because of poor solubility and low permeability, analytical method development and formulation becomes challenging. Till date, there are no standard test methods available to analyze Apremilast. So, a critical review of the analytical techniques of Apremilast was carried out. The literature search was done by screening the papers reporting analytical techniques of Apremilast from year 2014 to 2019. Methodologies particularly UV spectroscopy, HPTLC, HPLC, X-ray diffraction, NMR, LC-MS were collected and reviewed. Interminable efforts are made by the researchers to develop simple, accurate, robust and cost-effective methods of analysis. In pharmaceutical research, this information will aid in the development of new delivery systems. The review will prove beneficial and advantageous pre-formulation studies and will guide the formulation development.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/analysis , Chemistry Techniques, Analytical/methods , Drug Monitoring/methods , Thalidomide/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arthritis, Psoriatic/drug therapy , Chemistry Techniques, Analytical/instrumentation , Chromatography/instrumentation , Chromatography/methods , Drug Monitoring/instrumentation , Humans , Spectrum Analysis/instrumentation , Spectrum Analysis/methods , Thalidomide/analysis , Thalidomide/pharmacokinetics
7.
Pediatr Blood Cancer ; 68(2): e28756, 2021 02.
Article in English | MEDLINE | ID: mdl-33025730

ABSTRACT

BACKGROUND: Central nervous system (CNS) malignancies are the most common solid tumors among children, and novel therapies are needed to help improve survival. Pomalidomide is an immunomodulatory agent that displays antiangiogenic and cytotoxic activity, making it an appropriate candidate to explore in pediatric CNS tumors. METHODS: A phase 1 first in pediatric trial of pomalidomide was conducted in children with recurrent, progressive, and refractory CNS tumors. The primary objective was to determine the maximum tolerated dose (MTD) and/or recommended phase 2 dose (RP2D) when given orally once daily for 21 consecutive days of a 28-day cycle. Once the MTD was established, 12 additional patients were enrolled on expansion cohorts based on age and steroid use. RESULTS: Twenty-nine children were enrolled and 25 were evaluable for dose-limiting toxicity (DLT). The MTD was 2.6 mg/m2 (dose level 2). Four DLTs were observed in three patients at dose level 3 (3.4 mg/m2 ) includeding grade 3 diarrhea, grade 3 thrombocytopenia, grade 3 lung infection, and grade 4 neutropenia. The most common adverse events were grade 1 and 2 myelosuppression. One patient with an oligodendroglioma had stable disease for nine cycles, and a second patient with an anaplastic pleomorphic xanthoastrocytoma achieved a sustained partial response. Immunologic analyses suggested that pomalidomide triggers immunomodulation. CONCLUSIONS: The MTD of pomalidomide is 2.6 mg/m2 . It was well tolerated, and immune correlates showed a serum immune response. These data led to an industry-sponsored phase 2 trial of pomalidomide monotherapy in children with recurrent brain tumors (NCT03257631).


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Thalidomide/analogs & derivatives , Adolescent , Angiogenesis Inhibitors/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Immunomodulation/drug effects , Male , Thalidomide/pharmacokinetics , Thalidomide/therapeutic use , Young Adult
8.
Blood ; 137(7): 896-907, 2021 02 18.
Article in English | MEDLINE | ID: mdl-32976576

ABSTRACT

Steroid-refractory chronic graft-versus-host disease (cGVHD) is a therapeutic challenge. Sclerotic skin manifestations are especially difficult to treat. We conducted a randomized phase 2 clinical trial (#NCT01688466) to determine the safety, efficacy, and preferred dose of pomalidomide in persons with moderate to severe cGVHD unresponsive to corticosteroids and/or subsequent lines of therapy. Thirty-four subjects were randomized to receive pomalidomide 0.5 mg per day orally (n = 17; low-dose cohort) or 2 mg per day at a starting dose of 0.5 mg per day increasing to 2 mg per day over 6 weeks (n = 17; high-dose cohort). The primary endpoint was overall response rate (ORR) at 6 months according to the 2005 National Institutes of Health cGVHD Response Criteria. Thirty-two patients had severe sclerotic skin and received a median of 5 (range, 2-10) previous systemic therapies. ORR was 47% (95% confidence interval, 30-65) in the intention-to-treat analyses. All were partial responses, with no difference in ORR between the cohorts. ORR was 67% (45%-84%) in the 24 evaluable subjects at 6 months. Nine had improvement in National Institutes of Health joint/fascia scores (P = .018). Median change from the baseline in body surface area involvement of skin cGVHD was -7.5% (-10% to 35%; P = .002). The most frequent adverse events were lymphopenia, infection, and fatigue. Eight subjects in the high-dose cohort had dose decreases because of adverse events. There was 1 death in the low-dose cohort from bacterial pneumonia. Our data indicate antifibrotic effects of pomalidomide and possible association with increases in concentrations of blood regulatory T-cell and interleukin-2. Pomalidomide 0.5 mg per day is a safe and effective therapy for advanced corticosteroid-refractory cGVHD.


Subject(s)
Graft vs Host Disease/drug therapy , Immunologic Factors/therapeutic use , Salvage Therapy/methods , Thalidomide/analogs & derivatives , Adolescent , Adult , Aged , Allografts , Disease Susceptibility , Dose-Response Relationship, Drug , Drug Resistance , Fatigue/etiology , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Infections , Joints/pathology , Kaplan-Meier Estimate , Lymphocyte Count , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Lymphopenia/etiology , Male , Middle Aged , Quality of Life , Skin/pathology , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/pharmacokinetics , Thalidomide/therapeutic use , Young Adult
9.
J Vet Sci ; 21(5): e60, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33016014

ABSTRACT

BACKGROUND: Tumor-associated neoangiogenesis is a crucial target for antitumor therapies. Thalidomide (TAL) is a promising anti-neoangiogenetic drug that has recently been used in the treatment of several malignancies in dogs. OBJECTIVES: The aim of the study was to assess the pharmacokinetics of TAL after single oral administration in dogs. Additionally, the influence of feeding on the pharmacokinetic profile of TAL in dogs has been preliminarily investigated. METHODS: Six healthy adult female Labradors were enrolled according to a randomized single-dose, 2-treatment, 2-phase, paired 2 × 2 cross-over study design. The dogs were administered a single 400 mg capsule of TAL in fasted and fed conditions. Blood was collected from 15 min to 48 h after dosing, and TAL quantified in plasma by a validated high-performance liquid chromatography method. The pharmacokinetics of TAL were analyzed using a non-compartmental approach. RESULTS: TAL concentration was quantifiable up to 10 h and 24 h after fasted and fed conditions, respectively. Cmax (fasted, 1.34 ± 0.12 µg/mL; fed, 2.47 ± 0.19 µg/mL) and Tmax (fasted, 3 h; fed, 10 h) differed substantially between the 2 groups. AUC and t1/2λz were significantly higher in fed (42.46 ± 6.64 mg × h/L; 17.14 ± 4.68 h) compared to fasted (12.38 ± 1.13 mg × h/L; 6.55 ± 1.25 h) dogs. The relative oral bioavailability of TAL for the fasted group was low (36.92% ± 3.28%). CONCLUSIONS: Feeding affects the pharmacokinetics of oral TAL in dogs, showing a delayed, but higher absorption with different rate of elimination. These findings are of importance in clinical veterinary settings, and represent a starting point for further related studies.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Fasting , Thalidomide/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Chromatography, High Pressure Liquid/veterinary , Cross-Over Studies , Dogs , Female , Half-Life
10.
J Med Chem ; 63(23): 14562-14575, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33058680

ABSTRACT

We report compounds 5 (CG416) and 6 (CG428) as two first-in-class tropomyosin receptor kinase (TRK) degraders that target the intracellular kinase domain of TRK. Degraders 5 and 6 reduced levels of the tropomyosin 3 (TPM3)-TRKA fusion protein in KM12 colorectal carcinoma cells and inhibited downstream PLCγ1 signaling at sub-nanomolar concentrations. Both degraders also degraded human wild-type TRKA with similar potency. Interestingly, both degraders, especially 6, showed selectivity for the degradation of endogenous TPM3-TRKA over ectopically expressed ATP/GTP binding protein-like 4 (AGBL4)-TRKB or ETS variant transcription factor 6 (ETV6)-TRKC fusion proteins in KM12 cells. Global proteomic profiling assays demonstrated that 5 is highly selective for the intended target. TPM3-TRKA protein degradation induced by 5 and 6 was further confirmed to be mediated through cereblon and the ubiquitin-proteasome system. Compared with the parental TRK kinase inhibitor, both degraders exhibited higher potency for inhibiting growth of KM12 cells. Moreover, both 5 and 6 showed good plasma exposure levels in mice. Therefore, 5 and 6 are valuable chemical tool compounds for investigating the in vivo function of TRK fusion during tumorigenesis. Our study also paves the way for pharmacological degradation of TRK.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Drug Design , Drug Discovery , Humans , Male , Mice, Inbred ICR , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Proteolysis/drug effects , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/metabolism , Structure-Activity Relationship , Thalidomide/pharmacokinetics , Ubiquitin-Protein Ligases/metabolism
11.
J Clin Pharmacol ; 60(8): 1061-1075, 2020 08.
Article in English | MEDLINE | ID: mdl-32168381

ABSTRACT

Multiple myeloma is an incurable progressive neoplastic disease that accounts for 10% of all hematologic malignancies. Even though significant progress has been made in the treatment of newly diagnosed multiple myeloma, the disease follows a relapsing course in the majority of patients, and there is a need for more effective therapeutic options for the treatment of relapsed or refractory multiple myeloma. CC-4047-MM-005 and CC-4047-MM-007 were phase 1 and 3 studies to evaluate the novel combination of pomalidomide, bortezomib, and low-dose dexamethasone for the treatment of patients with relapsed or refractory multiple myeloma who have already received lenalidomide-based treatments early. This analysis was performed to characterize the population pharmacokinetics (PK) of pomalidomide from the combination treatment and to examine exposure-response relationships. Our analysis showed that pomalidomide concentration-time profiles from the combination treatment were adequately described with a 1-compartment PK model, with first-order absorption and elimination and pomalidomide exhibiting linear and time-invariant PK with moderate variability from the combination treatment. Except for the body surface area, none of the tested covariates had an effect on pomalidomide PK. Although body surface area was identified as a statistically significant covariate of pomalidomide PK, the impact was not deemed clinically relevant. A flat exposure-response curve was observed, consistent with a near-saturated drug effect at the tested exposure range suggesting an appropriately recommended clinical dose of 4 mg of pomalidomide for the combination treatment. Finally, pomalidomide exposure was not associated with higher probabilities of dose interruption during cycle 1 or dose reduction during the treatment period.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Immunologic Factors/pharmacokinetics , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/blood , Area Under Curve , Bayes Theorem , Bortezomib/administration & dosage , Clinical Trials, Phase I as Topic , Clinical Trials, Phase III as Topic , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/blood , Kaplan-Meier Estimate , Male , Middle Aged , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Progression-Free Survival , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/blood , Thalidomide/pharmacokinetics
12.
J Dig Dis ; 21(2): 98-103, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31916702

ABSTRACT

OBJECTIVE: To explore the relationship between hepatic cytochrome P450 2C19 (CYP2C19) gene polymorphisms and the effectiveness and safety of thalidomide in the treatment of patients with immune-related bowel disease (IRBD). METHODS: CYP2C19 variants in 79 patients treated with thalidomide were analyzed using a polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The clinical response and adverse events of the thalidomide treatment were recorded. The potential influences of the CYP2C19 genotype polymorphisms on the clinical efficacy and adverse events of thalidomide were then investigated. RESULTS: Altogether 79 patients with IRBD (70 with Crohn's disease, three with ulcerative colitis and six with Behcet's disease) receiving thalidomide therapy were recruited from January 2013 to February 2015 in a tertiary IBD center in China. Overall, 21.5% (17/79) of these patients had CYP2C19 poor metabolizers genotype (PM). The overall response rate and the incidence of adverse events of CYP2C19 extensive metabolizers genotype were not significantly different from that of the PM when IRBD patients were treated with thalidomide (P = 0.517 and 0.816, respectively). CONCLUSION: CYP2C19 polymorphisms do not seem to be associated with efficacy of thalidomide and the incidence of adverse events in treating IRBD.


Subject(s)
Cytochrome P-450 CYP2C19/drug effects , Immunosuppressive Agents/pharmacokinetics , Intestinal Diseases/drug therapy , Pharmacogenomic Variants/drug effects , Polymorphism, Restriction Fragment Length/drug effects , Thalidomide/pharmacokinetics , Adult , Behcet Syndrome/drug therapy , Behcet Syndrome/genetics , Behcet Syndrome/immunology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Crohn Disease/drug therapy , Crohn Disease/genetics , Crohn Disease/immunology , Female , Genotype , Humans , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Male , Retrospective Studies , Treatment Outcome , Young Adult
13.
J Am Acad Dermatol ; 82(2): 389-397, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31408686

ABSTRACT

BACKGROUND: No oral systemic treatments are approved for pediatric patients with psoriasis. OBJECTIVE: To evaluate the pharmacokinetics and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in pediatric patients with psoriasis. METHODS: This phase 2, multicenter, open-label study enrolled pediatric patients with moderate to severe plaque psoriasis. Patients received apremilast twice daily without titration for 2 weeks (group 1 [age, 12-17 years; weight, ≥35 kg]: apremilast 20 or 30 mg; group 2 [age, 6-11 years; weight, ≥15 kg]: apremilast 20 mg), followed by a 48-week extension. Primary endpoints were pharmacokinetics and safety. Other endpoints were taste/acceptability and change from baseline in score on the Psoriasis Area and Severity Index. RESULTS: A total of 42 enrolled patients (21 adolescents [age, 12-17 years] and 21 children [age, 6-11 years]) received apremilast. Pharmacokinetics modeling and noncompartmental analyses showed that weight-based dosing with apremilast 20 mg twice daily in children or apremilast 20 or 30 mg twice daily in adolescents provides exposure (area under the concentration-time curve from time 0 to 12 hours after the dose) that is comparable to that achieved with apremilast 30 mg twice daily in adults. The safety profile was generally similar to that in adults. Most study participants liked the taste of the tablet. Improvements from baseline in mean Psoriasis Area and Severity Index score were 68% for adolescents (overall) and 79% for children. LIMITATIONS: No children weighing less than 20 kg were enrolled. CONCLUSIONS: This first-time-in-children phase 2 study supports weight-based apremilast dosing for future phase 3 studies of pediatric plaque psoriasis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Phosphodiesterase 4 Inhibitors/pharmacokinetics , Phosphodiesterase 4 Inhibitors/therapeutic use , Psoriasis/drug therapy , Thalidomide/analogs & derivatives , Adolescent , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Child , Humans , Phosphodiesterase 4 Inhibitors/adverse effects , Severity of Illness Index , Thalidomide/adverse effects , Thalidomide/pharmacokinetics , Thalidomide/therapeutic use
14.
J Clin Pharmacol ; 60(1): 67-74, 2020 01.
Article in English | MEDLINE | ID: mdl-31392755

ABSTRACT

A population pharmacokinetic (PPK) model to describe the pharmacokinetics of thalidomide in different patient populations was developed using data pooled from healthy subjects and patients with Hansen's disease, human immunodeficiency virus (HIV), and multiple myeloma (MM). The analysis data set had a total of 164 evaluable subjects who received various doses (50 to 400 mg) of oral thalidomide in single- and/or multiple-dose regimens. The plasma thalidomide concentrations were adequately described by a linear 1-compartment PPK model with first-order absorption and first-order elimination. Inclusion of MM as a covariate on apparent clearance (CL/F) accounted for 4.4% of the interindividual variability (IIV) of CL/F. Body weight as a covariate on CL/F and apparent volume of distribution (V/F) also improved model fitting slightly, accounting for 7.2% and 20% of IIV, respectively. Although inclusion of body weight and MM as covariates of CL/F and body weight on V/F improved the goodness of fit of the model in a statistically significant manner, the impact of this difference in CL/F is not considered clinically relevant. Other factors such as age, sex, race, creatinine clearance, and alanine transaminase had no effect on thalidomide pharmacokinetics. MM, HIV, and Hansen's disease have no clinically relevant effect on thalidomide disposition relative to healthy volunteers.


Subject(s)
HIV Infections/metabolism , Immunosuppressive Agents/pharmacokinetics , Leprosy/metabolism , Multiple Myeloma/metabolism , Thalidomide/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Drug Administration Schedule , Female , HIV Infections/drug therapy , Healthy Volunteers , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Immunosuppressive Agents/therapeutic use , Leprosy/drug therapy , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Multiple Myeloma/drug therapy , Thalidomide/administration & dosage , Thalidomide/blood , Thalidomide/therapeutic use , Young Adult
15.
Eur J Pharm Sci ; 140: 105068, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31518681

ABSTRACT

Maternofoetal physiologically-based pharmacokinetic models integrating multi-compartmental maternal and foetal units were developed using Simbiology® to estimate prenatal drug exposure. Processes governing drug disposition were described using differential equations with key system and drug-specific parameters. Transplacental drug transfer was modelled as bidirectional passive diffusion and benchmarked against those for thalidomide as a control. Model-predictions for pharmacokinetic parameters during pregnancy were within acceptable ranges for qualification (two-fold difference of clinically-observed values). Predicted foetal exposure to thalidomide was higher than efavirenz, with median (range) foetal-to-maternal plasma ratios of 4.55 (3.06-9.57) for 400 mg thalidomide versus 0.89 (0.73-1.05) for 400 mg efavirenz at third trimester. Model-predictions indicated foetal exposure consistently above 300% of maternal plasma concentration for thalidomide throughout pregnancy, while exposure to efavirenz increased from under 20% at second trimester to above 100% at third trimester. Further qualification of this approach as a tool in evaluating drug exposure and safety during pregnancy is warranted.


Subject(s)
Benzoxazines/pharmacokinetics , Computer Simulation , Fetus/drug effects , Models, Biological , Thalidomide/pharmacokinetics , Alkynes , Benzoxazines/adverse effects , Cyclopropanes , Cytochrome P-450 Enzyme System/metabolism , Female , Humans , Intestinal Absorption , Permeability , Placenta/metabolism , Pregnancy , Thalidomide/adverse effects
16.
Clin Lymphoma Myeloma Leuk ; 19(9): 570-578.e1, 2019 09.
Article in English | MEDLINE | ID: mdl-31326409

ABSTRACT

INTRODUCTION: This phase Ib study evaluated oprozomib, an oral proteasome inhibitor, plus pomalidomide-dexamethasone in relapsed/refractory multiple myeloma (RRMM). PATIENTS AND METHODS: Patients received oprozomib once-daily on days 1 to 5 and 15 to 19 (5/14 schedule; 150 mg/day starting dose) or on 2 consecutive days weekly (2/7 schedule; 210 mg/day starting dose) of 28-day cycles, pomalidomide on days 1 to 21 (4 mg/day starting dose), and dexamethasone 20 mg on 2 consecutive days weekly. A 3 + 3 dose-escalation schema was used to determine the maximum tolerated dose. RESULTS: Thirty-one patients were treated (5/14, n = 4; 2/7, n = 27). Oprozomib maximum tolerated dose was not defined. The 2/7 schedule (oprozomib 210 mg/day, pomalidomide 4 mg/day) was selected for dose expansion based on overall safety (n = 17). In this group, the most common adverse events (AEs) were gastrointestinal (diarrhea [88.2%], nausea [58.8%], and vomiting [58.8%]); grade ≥ 3 gastrointestinal AEs were uncommon. The most common grade ≥ 3 AEs were hematologic (anemia [47.1%], neutropenia [35.3%], and thrombocytopenia [29.4%]). One dose-limiting toxicity (gastric hemorrhage) occurred; 3 patients discontinued owing to AEs. The overall response rate was 70.6%. CONCLUSION: Safety and pharmacokinetic profiles were concerns with the oprozomib formulation used in this study and need to be improved. Oprozomib-pomalidomide-dexamethasone (2/7 schedule) had encouraging efficacy, supporting an ongoing phase Ib study evaluating new oprozomib formulations for this combination in relapsed/refractory multiple myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Neoplasm Recurrence, Local , Neoplasm Staging , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Research Design , Retreatment , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/pharmacokinetics
17.
Eur J Pharm Sci ; 136: 104937, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31128208

ABSTRACT

Thalidomide (TLD) is used to treat erythema nodosum leprosum (ENL), multiple myeloma, aphthous ulceration and wasting syndrome in HIV patients. The API can be found in two crystalline habits known as α-TLD and ß-TLD. The saturation solubility (Cs) and the dissolution profiles under non-sink and sink conditions of both polymorphs were assessed. In addition, mini-capsules containing α-TLD or ß-TLD without excipients were orally given (10 mg/kg) to Wistar rats. An intravenous (i.v.) dose was also administrated (5 mg/kg). The Cs values for α-TLD and ß-TLD were not significantly different (α = 56.2 ±â€¯0.5 µg·mL-1; ß = 55.2 ±â€¯0.2 µg·mL-1). However, the dissolution profile of α-TLD presented the fastest rate and the largest extension of drug dissolution than that from ß-TLD (80% in 4 h versus 55% in 4 h). The α-TLD provided a more favorable pharmacokinetic than the ß-TLD (maximum plasma concentration - Cmax: 5.4 ±â€¯0.90 µg·mL-1versus 2.6 ±â€¯0.2 µg·mL-1; area under the curve of the concentration-time profile from time zero to infinity - AUC0-∞: 44.3 ±â€¯8.8 µg·h·mL-1versus 33.9 ±â€¯4.7 µg·h·mL-1; absolute bioavailability - F: 92.2 ±â€¯18.5% versus 70.5 ±â€¯9.9%, respectively). Drug suppliers and pharmaceutical companies should strictly control the technological processes involved in the TLD API synthesis as well as in the production of the pharmaceutical dosage form in order to guarantee the inter-batch homogeneity and therefore, product compliance.


Subject(s)
Thalidomide/chemistry , Thalidomide/pharmacokinetics , Animals , Area Under Curve , Biological Availability , Capsules/chemistry , Capsules/pharmacokinetics , Drug Liberation/drug effects , Excipients/chemistry , HIV Infections/drug therapy , HIV Infections/metabolism , Male , Rats , Rats, Wistar , Solubility/drug effects
18.
Int J Nanomedicine ; 14: 1587-1595, 2019.
Article in English | MEDLINE | ID: mdl-30880967

ABSTRACT

BACKGROUND: Apremilast (APM) is a novel, orally administered small molecule drug approved for treatment of psoriasis or psoriatic arthritis. Due to its low solubility and permeability, it is classified as a class IV drug according to BCS classification. Dose titration is recommended during APM treatment due to its tolerability and twice-daily dosing regimen issues. MATERIALS AND METHODS: In this study, three different APM-loaded PLGA nanoparticles (F1-F3) were prepared by single emulsion and evaporation method. Based on particle size, PDI, zeta potential (ZP), entrapment efficiency (%EE), drug loading (%DL), and spectral characterization, the nanoparticles (F3) were optimized. The F3 nanoparticles were further evaluated for in vitro release and in vivo pharmacokinetic studies in rats. RESULTS: The optimized nanoparticles (F3) had particles size 307.3±8.5 nm with a low PDI value 0.317, ZP of -43.4±2.6 mV, EE of 61.1±1.9% and DL of 1.9±0.1%. The in vitro release profile showed a sustained release pattern of F3 nanoparticles of APM. The pharmacokinetic results showed 2.25 times increase in bio-availability of F3 nanoparticles compared to normal APM suspension. Moreover, significant increase in half-life and mean residence time confirms long-term retention of F3 nanoparticles. CONCLUSION: Bioavailability enhancement along-with long-term retention of the APM-loaded PLGA nanoparticles might be helpful for the once-daily regimen treatment.


Subject(s)
Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Thalidomide/analogs & derivatives , Animals , Calorimetry, Differential Scanning , Delayed-Action Preparations , Drug Carriers , Drug Liberation , Kinetics , Male , Nanoparticles/ultrastructure , Particle Size , Rats, Wistar , Spectroscopy, Fourier Transform Infrared , Static Electricity , Thalidomide/administration & dosage , Thalidomide/blood , Thalidomide/pharmacokinetics , Thalidomide/pharmacology , X-Ray Diffraction
19.
Proc Natl Acad Sci U S A ; 116(7): 2539-2544, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683719

ABSTRACT

Current systems for modulating the abundance of proteins of interest in living cells are powerful tools for studying protein function but differ in terms of their complexity and ease of use. Moreover, no one system is ideal for all applications, and the best system for a given protein of interest must often be determined empirically. The thalidomide-like molecules (collectively called the IMiDs) bind to the ubiquitously expressed cereblon ubiquitin ligase complex and alter its substrate specificity such that it targets the IKZF1 and IKZF3 lymphocyte transcription factors for destruction. Here, we mapped the minimal IMiD-responsive IKZF3 degron and show that this peptidic degron can be used to target heterologous proteins for destruction with IMiDs in a time- and dose-dependent manner in cultured cells grown ex vivo or in vivo.


Subject(s)
Peptides/metabolism , Proteins/metabolism , Thalidomide/analogs & derivatives , Animals , Blood-Brain Barrier , Ikaros Transcription Factor/metabolism , Mice , Proteolysis , Thalidomide/pharmacokinetics , Thalidomide/pharmacology , Trans-Activators/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...