1
|
Sychterz C, Chen P, Saxena A, Lin H, Chen L, Li Y, Gaohua L, Cheng Y. Model-informed Drug Development (MIDD) Approach to Support Biopharmaceutical Development of Iberdomide. AAPS J 2025; 27:85. [PMID: 40299238 DOI: 10.1208/s12248-025-01071-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
Iberdomide is a BCS III CELMoD™ agent currently under development for treatment of multiple myeloma. Five formulations were used during clinical development, starting with active ingredient in gelatin capsule (AIC), followed by subsequent formulations in gelatin (F1), HPMC capsules (F2 and F3), and finally the intended commercial form (ICF). A food effect study with the Phase I AIC formulation showed no food effect and two relative bioavailability studies bridging from AIC to formulation F1 and F2 showed similar systemic exposure. Modeling and simulation, based on in vitro dissolution, was used to bridge gaps in clinical data to demonstrate lack of a food effect across all formulations including ICF. First, a previously developed population pharmacokinetic (PK) model showed that formulation was not a covariate on iberdomide PK. Then a physiologically-based pharmacokinetic model (PBPK) informed by in vitro biorelevant dissolution was used to mechanistically describe iberdomide absorption. While minor differences between formulations were noted in their in vitro dissolution, PBPK modeling showed lack of a food effect across all five formulations. Sensitivity analysis using the PBPK model demonstrated that iberdomide permeability and total amount of drug released from formulation are the most sensitive parameters in defining the systemic exposure of iberdomide, in agreement with iberdomide's BCS III classification where drug release from drug product, and not solubility, is the rate-limiting step of dissolution. Based on the totality of evidence, which included clinical and in vitro data supplemented by modeling and simulation, no food effect is expected with the ICF.
Collapse
Affiliation(s)
| | - Ping Chen
- Bristol Myers Squibb, Princeton, New Jersey, USA
- Bristol Myers Squibb, 556 Morris Ave, Summit, New Jersey, 07901, USA
| | - Ajay Saxena
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Hongxia Lin
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Lu Chen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Yan Li
- Bristol Myers Squibb, Princeton, New Jersey, USA
- Bristol Myers Squibb, 556 Morris Ave, Summit, New Jersey, 07901, USA
| | - Lu Gaohua
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Yiming Cheng
- Bristol Myers Squibb, Princeton, New Jersey, USA.
- Bristol Myers Squibb, 556 Morris Ave, Summit, New Jersey, 07901, USA.
| |
Collapse
|
2
|
Chen P, Lin H, Xue Y, Thomas M, Wang A, Li Y, Cheng Y. A population pharmacokinetic analysis to evaluate the impact of renal impairment on the pharmacokinetics of iberdomide. J Pharm Sci 2025; 114:1315-1325. [PMID: 39870177 DOI: 10.1016/j.xphs.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Iberdomide, a novel potent cereblon E3 ligase modulator, is under investigation for multiple myeloma. This study assessed how renal impairment (RI) affects iberdomide pharmacokinetics (PK). Twenty-six subjects with varying renal function, including those with severe renal impairment and those requiring intermittent hemodialysis (IHD), received a single oral 1 mg dose of iberdomide. Plasma, urine, and dialysate samples were analyzed to evaluate the PK of iberdomide and its major active metabolite, M12. Data were subsequently pooled with PK data from four other clinical trials involving 354 patients to develop a parent - metabolite population PK model using nonlinear mixed-effects modeling to assess the impact of various degrees of RI on drug exposure. The population PK model effectively described the PK of iberdomide and M12, showing that normal, mild, and moderate RI had no significant impact on iberdomide PK exposure, whereas severe RI reduced total clearance and increased PK exposure of iberdomide and M12. Subjects with kidney failure on IHD had comparable total clearance and PK exposure to those with normal renal function. These findings systematically examined the effects of various degrees of RI on iberdomide PK and provide a basis for informing iberdomide dosing in patients with varying degrees of renal impairment.
Collapse
Affiliation(s)
- Ping Chen
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Hongxia Lin
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yongjun Xue
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Mark Thomas
- Early Clinical Development ICN, Bristol Myers Squibb, Princeton, NJ, USA
| | - Alice Wang
- Early Clinical Development ICN, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yan Li
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yiming Cheng
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA.
| |
Collapse
|
3
|
Xue Y, Wang L, Huo R, Chen M, Melo B, Dingley K, Gaudy A, Shen JX. 1β-Hydroxydeoxycholic Acid as an Endogenous Biomarker in Human Plasma for Assessment of CYP3A Clinical Drug-Drug Interaction Potential. Drug Metab Dispos 2024; 52:966-974. [PMID: 38991779 DOI: 10.1124/dmd.124.001680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
4β-Hydroxycholesterol (4β-HC) in plasma has been used as a biomarker to assess CYP3A drug-drug interaction (DDI) potential during drug development. However, due to the long half-life and narrow dynamic range of 4β-HC, its use has been limited to the identification of CYP3A inducers, but not CYP3A inhibitors. The formation of 1β-hydroxydeoxycholic acid (1β-OH DCA) from deoxycholic acid (DCA) is mediated by CYP3A, thus 1β-OH DCA can potentially serve as an alternative to 4β-HC for assessment of CYP3A DDI potential. To study this feasibility, we developed a sensitive liquid chromatography-tandem mass spectrometry method for the simultaneous quantitation of 1β-OH DCA and its glycine and taurine conjugates in human plasma with the lower limit of quantitation of 50 pg/ml, which enabled the quantitation of basal levels and further reduction. The method was applied to a DDI study to assess how 1β-OH DCA and its glycine and taurine conjugates would respond to CYP3A induction or inhibition. Rifampin induction resulted in an increase of 1β-OH DCA and its conjugates in plasma, with 6.8-, 7.8-, 8.3-, and 10.3-fold increases of area under the curve from the time of dosing to the last measurable concentration (AUCLST), area under the curve from the time of dosing to 24 hours (AUC24h), C max, and mean concentrations for total 1β-OH DCA (total of all three forms), respectively. Importantly, inhibition with itraconazole resulted in notable reduction of these biomarkers, with 84%, 85%, 82%, and 81% reductions of AUCLST, AUC24h, C max, and mean concentrations for total 1β-OH DCA, respectively. These preliminary data demonstrate for the first time that total 1β-OH DCA in plasma has the potential to serve as a biomarker for CYP3A DDI assessment in early clinical development and may provide key advantages over 4β-HC. SIGNIFICANCE STATEMENT: The authors have reported the use of total 1β-hydroxydeoxycholic acid (1β-OH DCA) (sum of 1β-OH DCA and its glycine and taurine conjugates) plasma exposure as a biomarker for CYP3A activity. Itraconazole inhibition led to an 81%-85% decrease of total 1β-OH DCA plasma exposures, whereas rifampin induction led to a 6.8- to 10.3-fold increase of total 1β-OH DCA plasma exposures. Using 1β-OH DCA exposures in plasma also provides the benefit of allowing pharmacokinetic and biomarker assessment using the same matrix.
Collapse
Affiliation(s)
- Yongjun Xue
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Linna Wang
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Runlan Huo
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Mu Chen
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Brian Melo
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Karen Dingley
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Allison Gaudy
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| | - Jim X Shen
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol-Myers Squibb, Princeton, New Jersey (Y.X., L.W., R.H., B.M., A.G., J.X.S.); Regeneron Pharmaceuticals Inc., Tarrytown, New York (M.C.); and Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (K.D.)
| |
Collapse
|
4
|
Cheng Y, Wang X, Liu L, Silva J, Thomas M, Li Y. A Phase I, Open-Label, Mass Balance Study of [ 14C]-Iberdomide in Healthy Subjects. Eur J Drug Metab Pharmacokinet 2024; 49:355-365. [PMID: 38521893 DOI: 10.1007/s13318-024-00886-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Iberdomide is a novel potent cereblon modulator (CELMoD®) agent, which is currently under clinical development for hematological malignancies. A human mass balance study was conducted to characterize the biotransformation and excretion pathways of iberdomide. METHOD After a single dose of radiolabelled [14C]-iberdomide (1 mg) in six healthy subjects. Blood, urine, and fecal samples were collected for pharmacokinetics, mass balance, and clinical laboratory assessments. RESULTS Results showed that a single oral dose of 1 mg iberdomide was generally well tolerated in healthy subjects. The recovery of [14C]-iberdomide-derived radioactivity in humans was 45.9% in urine and 42.6% in feces. Based on exposure (area under the concentration-time curve [AUC0-24]), iberdomide and M12 (metabolites) accounted for approximately 59% and 14% of circulating total radioactivity (TRA) exposure, respectively. Of the 88.5% TRA excreted, approximately 27% was excreted as unchanged iberdomide and 62% as metabolites, with similar amounts of excreted metabolites in the urine (16%) and feces (11%). CONCLUSION Biotransformation of iberdomide in humans included multiple oxidations of the morpholino moiety as well as glutarimide ring hydrolysis of parent and oxidized metabolites and a combination of these pathways. Iberdomide was the predominant component in human plasma, with metabolite M12 being the most prominent circulating metabolite. In excreta, similar iberdomide-derived radioactivity was found in urine and feces. TRIAL REGISTRATION NUMBER NCT03294603.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Xiaomin Wang
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Liangang Liu
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Jose Silva
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Michael Thomas
- Clinical Scientist Early Clinical Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yan Li
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA.
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, 556 Morris Ave, Summit, NJ, 07901, USA.
| |
Collapse
|
5
|
Cheng Y, Gaudy A, Liu L, Ye Y, Thomas M, Xue Y, Zhou S, Li Y. Exposure-Response Analysis to Assess the Concentration-QTc Relationship of Iberdomide. Clin Pharmacol Drug Dev 2023; 12:819-825. [PMID: 37079714 DOI: 10.1002/cpdd.1254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/26/2023] [Indexed: 04/22/2023]
Abstract
Iberdomide is an orally available cereblon-modulating agent being developed for the treatment of hematologic malignancies and autoimmune-mediated diseases. To assess the potential concentration-QTc relationship in humans and to ascertain or exclude a potential QT effect by iberdomide, a plasma concentration and ΔQTcF (change from baseline of corrected QT interval using the Fridericia formula) model of iberdomide was developed. Iberdomide concentration and paired high-quality, intensive electrocardiogram signal from a single-ascending-dose study in healthy subjects (N = 56) were included in the analysis. The primary analysis was based on a linear mixed-effect model with ΔQTcF as the dependent variable; iberdomide plasma concentration and baseline QTcF as continuous covariates; treatment (active or placebo) and time as a categorical factor; and a random intercept per subject. The predicted change from baseline and placebo corrected (ΔΔQTcF) at the observed geometric mean maximum plasma concentration and 2-sided 90% confidence intervals at different dose levels were calculated. The upper bound of the 90% confidence interval of the model-predicted ΔΔQTcF effect at maximum concentration from the supratherapeutic dose of 6 mg (2.54 milliseconds) is <10-millisecond threshold, suggesting that iberdomide does not have a clinically relevant QT prolongation liability.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Allison Gaudy
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Liangang Liu
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Ying Ye
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Michael Thomas
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Yongjun Xue
- Nonclinical Research & Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Simon Zhou
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Yan Li
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| |
Collapse
|
6
|
Cheng Y, Ye Y, Gaudy A, Ghosh A, Xue Y, Wang A, Zhou S, Li Y. A Phase 1, Multicenter, Open-Label Study to Evaluate the Pharmacokinetics of Iberdomide in Subjects with Mild, Moderate, or Severe Hepatic Impairment Compared with Healthy Subjects. Clin Pharmacol 2023; 15:9-19. [PMID: 36880014 PMCID: PMC9985425 DOI: 10.2147/cpaa.s397826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
Introduction Iberdomide, a novel cereblon modulator (CELMoD®), is currently under clinical investigation for hematology indications. To evaluate the influence of hepatic impairment on the pharmacokinetics (PK) of iberdomide and its major active metabolite M12, a phase 1, multicenter, open-label study was conducted in healthy subjects and subjects with mild, moderate, and severe hepatic impairment. Methods Forty subjects were enrolled in the study and divided into five groups based on hepatic function. 1 mg iberdomide was administered and plasma samples were collected to evaluate the pharmacokinetics of iberdomide and M12. Results After a single dose of iberdomide (1 mg), mean iberdomide Cmax (maximum observed concentration) and AUC (area under the concentration-time curve) exposure were generally comparable between hepatic impairment (HI) subjects (severe, moderate and mild) and their respective matched normal controls. Mean Cmax and AUC exposure of the metabolite M12 were generally comparable between mild HI and matched normal subjects. However, mean Cmax of the M12 was 30% and 65% lower and AUC was 57% and 63% lower in moderate and severe HI subjects as compared to their respective matched normal controls. However, given the relatively low M12 exposure as compared to its parent drug, the observed differences were not considered clinically meaningful. Conclusion In summary, 1 mg single oral dose of iberdomide was generally well-tolerated. HI (mild, moderate or severe) had no clinically relevant impact on iberdomide PK and therefore, no dose adjustment is warranted.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Ying Ye
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Allison Gaudy
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Atalanta Ghosh
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yongjun Xue
- Nonclinical Research & Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Alice Wang
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Simon Zhou
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yan Li
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
7
|
Fuchs O. Targeting cereblon in hematologic malignancies. Blood Rev 2023; 57:100994. [PMID: 35933246 DOI: 10.1016/j.blre.2022.100994] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
The protein cereblon (CRBN) is a substrate receptor of the cullin 4-really interesting new gene (RING) E3 ubiquitin ligase complex CRL4CRBN. Targeting CRBN mediates selective protein ubiquitination and subsequent degradation via the proteasome. This review describes novel thalidomide analogs, immunomodulatory drugs, also known as CRBN E3 ubiquitin ligase modulators or molecular glues (avadomide, iberdomide, CC-885, CC-90009, BTX-1188, CC-92480, CC-99282, CFT7455, and CC-91633), and CRBN-based proteolysis targeting chimeras (PROTACs) with increased efficacy and potent activity for application in hematologic malignancies. Both types of CRBN-binding drugs, molecular glues, and PROTACs stimulate the interaction between CRBN and its neosubstrates, recruiting target disease-promoting proteins and the E3 ubiquitin ligase CRL4CRBN. Proteins that are traditionally difficult to target (transcription factors and oncoproteins) can be polyubiquitinated and degraded in this way. The competition of CRBN neosubstrates with endogenous CRBN-interacting proteins and the pharmacology and rational combination therapies of and mechanisms of resistance to CRL4CRBN modulators or CRBN-based PROTACs are described.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12800 Praha 2, Czech Republic.
| |
Collapse
|
8
|
Deng K, Zou Y, Zou C, Wang H, Xiang Y, Yang X, Yang S, Cui C, Yang G, Huang J. Study on pharmacokinetic interactions between SHR2554 and itraconazole in healthy subjects: A single-center, open-label phase I trial. Cancer Med 2022; 12:1431-1440. [PMID: 35841331 PMCID: PMC9883540 DOI: 10.1002/cam4.5028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND SHR2554, a novel oral Enhancer of Zeste Homolog 2 inhibitor, shows broad-spectrum anti-tumor efficacy in preclinical studies. As SHR2554 is mainly metabolized by CYP3A4, it is helpful to conduct research on the effects of itraconazole, a strong inhibitor of CYP3A4-metabolizing enzymes, on the pharmacokinetic characteristics and safety of SHR2554. METHODS We conducted a single-center, open-label pharmacokinetic study of itraconazole on SHR2554 in 18 healthy Chinese subjects. Subjects were orally administrated SHR2554 50 mg on Day 1, itraconazole 200 mg Quaque Die (QD) from Days 4 to 7, SHR2554 50 mg co-administrated with itraconazole 200 mg on Day 8, and itraconazole 200 mg QD from Days 9 to 12. Then, 4 ml of venous blood was collected at predetermined time points. Plasma SHR2554 concentrations were analyzed using a validated high-performance liquid chromatography tandem mass spectrometry method. Pharmacokinetic parameters were calculated using Phoenix WinNonlin v8.1. RESULTS The Cmax of SHR2554 alone and in combination was 10.197 ± 7.0262 ng·ml-1 versus 70.538 ± 25.0219 ng·ml-1 , AUC0-∞ was 50.99 ± 19.358 h·ng·ml-1 versus 641.53 ± 319.538 h·ng·ml-1 , and AUC0-t was 28.70 ± 18.913 h·ng·ml-1 versus 612.13 ± 315.720 h·ng·ml-1 . Co-administration of SHR2554 and itraconazole caused 7.73-, 12.47-, and 23.75-fold adjusted geometric mean ratios increases in SHR2554 Cmax , AUC0-∞ and AUC0-t respectively. The co-administration regimen was well tolerated and had a good safety profile. CONCLUSIONS Compared with a single dose of SHR2554 50 mg, the exposure of SHR2554 in vivo was significantly affected by the combined administration of itraconazole.
Collapse
Affiliation(s)
- Kunhong Deng
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Yi Zou
- School of Mathematics and StatisticsCentral South UniversityChangshaChina
| | - Chan Zou
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Hong Wang
- School of Mathematics and StatisticsCentral South UniversityChangshaChina
| | - Yuxia Xiang
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina,Research Center of Drug Clinical Evaluation of Central South UniversityChangshaChina
| | - Xiaoyan Yang
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Shuang Yang
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Chang Cui
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina
| | - Guoping Yang
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina,Research Center of Drug Clinical Evaluation of Central South UniversityChangshaChina,Department of PharmacyThe Third Xiangya Hospital, Central South UniversityChangshaChina,XiangYa School of Pharmaceutical SciencesCentral South UniversityChangshaChina,National‐Local Joint Engineering Laboratory of Drug Clinical Evaluation TechnologyChangshaChina
| | - Jie Huang
- Center of Clinical PharmacologyThe Third Xiangya Hospital, Central South UniversityChangshaChina,Research Center of Drug Clinical Evaluation of Central South UniversityChangshaChina
| |
Collapse
|
9
|
Kably B, Launay M, Derobertmasure A, Lefeuvre S, Dannaoui E, Billaud EM. Antifungal Drugs TDM: Trends and Update. Ther Drug Monit 2022; 44:166-197. [PMID: 34923544 DOI: 10.1097/ftd.0000000000000952] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/09/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE The increasing burden of invasive fungal infections results in growing challenges to antifungal (AF) therapeutic drug monitoring (TDM). This review aims to provide an overview of recent advances in AF TDM. METHODS We conducted a PubMed search for articles during 2016-2020 using "TDM" or "pharmacokinetics" or "drug-drug-interaction" with "antifungal," consolidated for each AF. Selection was limited to English language articles with human data on drug exposure. RESULTS More than 1000 articles matched the search terms. We selected 566 publications. The latest findings tend to confirm previous observations in real-life clinical settings. The pharmacokinetic variability related to special populations is not specific but must be considered. AF benefit-to-risk ratio, drug-drug interaction (DDI) profiles, and minimal inhibitory concentrations for pathogens must be known to manage at-risk situations and patients. Itraconazole has replaced ketoconazole in healthy volunteers DDI studies. Physiologically based pharmacokinetic modeling is widely used to assess metabolic azole DDI. AF prophylactic use was studied more for Aspergillus spp. and Mucorales in oncohematology and solid organ transplantation than for Candida (already studied). Emergence of central nervous system infection and severe infections in immunocompetent individuals both merit special attention. TDM is more challenging for azoles than amphotericin B and echinocandins. Fewer TDM requirements exist for fluconazole and isavuconazole (ISZ); however, ISZ is frequently used in clinical situations in which TDM is recommended. Voriconazole remains the most challenging of the AF, with toxicity limiting high-dose treatments. Moreover, alternative treatments (posaconazole tablets, ISZ) are now available. CONCLUSIONS TDM seems to be crucial for curative and/or long-term maintenance treatment in highly variable patients. TDM poses fewer cost issues than the drugs themselves or subsequent treatment issues. The integration of clinical pharmacology into multidisciplinary management is now increasingly seen as a part of patient care.
Collapse
Affiliation(s)
- Benjamin Kably
- Laboratoire de Pharmacologie-Toxicologie, Hôpital Européen Georges Pompidou, AP-HP Centre
- Faculté de Médecine, Université de Paris, Paris, France
| | - Manon Launay
- Laboratoire de Pharmacologie-Toxicologie-Gaz du sang, Hôpital Nord-CHU Saint Etienne, Saint-Etienne
| | - Audrey Derobertmasure
- Laboratoire de Pharmacologie-Toxicologie, Hôpital Européen Georges Pompidou, AP-HP Centre
| | - Sandrine Lefeuvre
- Laboratoire de Toxicologie et Pharmacocinétique, CHU de Poitiers, Poitiers; and
| | - Eric Dannaoui
- Faculté de Médecine, Université de Paris, Paris, France
- Unité de Parasitologie-Mycologie, Laboratoire de Microbiologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Eliane M Billaud
- Laboratoire de Pharmacologie-Toxicologie, Hôpital Européen Georges Pompidou, AP-HP Centre
- Faculté de Médecine, Université de Paris, Paris, France
| |
Collapse
|
10
|
Kong NR, Liu H, Che J, Jones LH. Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition. ACS Med Chem Lett 2021; 12:1861-1865. [PMID: 34795877 DOI: 10.1021/acsmedchemlett.1c00475] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide engage cereblon and mediate a protein interface with neosubstrates such as zinc finger transcription factors promoting their polyubiquitination and degradation. The IMiDs have garnered considerable excitement in drug discovery, leading to exploration of targeted protein degradation strategies. Although the molecular modes-of-action of the IMiDs and related degraders have been the subject of intense research, their pharmacokinetics and disposition have been relatively understudied. Here, we assess the effects of physicochemistry of the IMiDs, the phthalimide EM-12, and the candidate drug CC-220 (iberdomide) on lipophilicity, solubility, metabolism, permeability, intracellular bioavailability, and cell-based potency. The insights yielded in this study will enable the rational property-based design and development of targeted protein degraders in the future.
Collapse
Affiliation(s)
- Nikki R. Kong
- Center for Protein Degradation, Dana-Farber Cancer Institute, 360 Longwood Avenue, Boston, Massachusetts 02215, United States
| | - Hu Liu
- Center for Protein Degradation, Dana-Farber Cancer Institute, 360 Longwood Avenue, Boston, Massachusetts 02215, United States
| | - Jianwei Che
- Center for Protein Degradation, Dana-Farber Cancer Institute, 360 Longwood Avenue, Boston, Massachusetts 02215, United States
| | - Lyn H. Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute, 360 Longwood Avenue, Boston, Massachusetts 02215, United States
| |
Collapse
|
11
|
Wang S, Li Z, Gao S. Key regulators of sensitivity to immunomodulatory drugs in cancer treatment. Biomark Res 2021; 9:43. [PMID: 34090534 PMCID: PMC8180172 DOI: 10.1186/s40364-021-00297-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) include thalidomide, lenalidomide, and pomalidomide, which have shown significant efficacy in the treatment of multiple myeloma (MM), myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)) and other hematological malignancies. IMiDs hijack the CRL4CRBN ubiquitin ligase to target cellular proteins for ubiquitination and degradation, which is responsible for their clinical activity in MM and MDS with del(5q). However, intrinsic and acquired resistance frequently limit the efficacy of IMiDs. Recently, many efforts have been made to explore key regulators of IMiD sensitivity, resulting in great advances in the understanding of the regulatory networks related to this class of drugs. In this review, we describe the mechanism of IMiDs in cancer treatment and summarize the key regulators of IMiD sensitivity. Furthermore, we introduce genome-wide CRISPR-Cas9 screenings, through which the regulatory networks of IMiD sensitivity could be identified.
Collapse
Affiliation(s)
- Shichao Wang
- The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfu Front Street, 450052, Zhengzhou, China.
| | - Zhiyue Li
- The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| | - Shaobing Gao
- The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China.
| |
Collapse
|