The global prevalence and incidence of metabolic dysfunction-associated fatty liver disease (MAFLD), including its progressive form metabolic dysfunction-associated steatohepatitis (MASH), are steadily rising, making them the most common chronic liver diseases worldwide. However, therapeutic options for MAFLD are currently limited. Glucolipid dysregulation drives MAFLD pathogenesis through intertwined glucose metabolic imbalance and lipid accumulation. Patients with type 2 diabetes mellitus (T2DM) are particularly prone to developing MASH and are at a higher risk of progressing to cirrhosis and hepatocellular carcinoma. The coexistence of MAFLD and T2DM correlates with clinical prognosis and elevates the risk of extrahepatic complications. Given the close association between MAFLD and T2DM, glucagon-like peptide-1 receptor agonists (GLP-1RAs), which have been approved for the treatment of T2DM and obesity, were the first to be investigated in patients with MAFLD/MASH. Recently, beyond GLP-1RAs, novel combination agents integrating glucose-dependent insulinotropic peptide receptor (GIPR) and/or glucagon receptor (GCR) agonists have also been explored. A large number of phase II randomized clinical trials have demonstrated significant improvements in body weight, insulin resistance, and liver parameters. Thus, GLP-1RAs and dual/triple agonists are promising for MAFLD/MASH, especially in those with obesity or T2DM. This study explores mechanisms and clinical evidence of incretin-based therapies for MAFLD by targeting its core pathogenesis-glucolipid disorders. With growing evidence, it also forecasts the broad clinical prospects on MAFLD treatment.
{"title":"Advances in Incretin-Based Therapies for MAFLD: Mechanisms and Clinical Evidence.","authors":"Wenqi Dong, Haiming Zhang, Shaowei Mu, Shuyi Shi, Junli Zhang, Keshu Xu","doi":"10.1002/cpt.70131","DOIUrl":"https://doi.org/10.1002/cpt.70131","url":null,"abstract":"<p><p>The global prevalence and incidence of metabolic dysfunction-associated fatty liver disease (MAFLD), including its progressive form metabolic dysfunction-associated steatohepatitis (MASH), are steadily rising, making them the most common chronic liver diseases worldwide. However, therapeutic options for MAFLD are currently limited. Glucolipid dysregulation drives MAFLD pathogenesis through intertwined glucose metabolic imbalance and lipid accumulation. Patients with type 2 diabetes mellitus (T2DM) are particularly prone to developing MASH and are at a higher risk of progressing to cirrhosis and hepatocellular carcinoma. The coexistence of MAFLD and T2DM correlates with clinical prognosis and elevates the risk of extrahepatic complications. Given the close association between MAFLD and T2DM, glucagon-like peptide-1 receptor agonists (GLP-1RAs), which have been approved for the treatment of T2DM and obesity, were the first to be investigated in patients with MAFLD/MASH. Recently, beyond GLP-1RAs, novel combination agents integrating glucose-dependent insulinotropic peptide receptor (GIPR) and/or glucagon receptor (GCR) agonists have also been explored. A large number of phase II randomized clinical trials have demonstrated significant improvements in body weight, insulin resistance, and liver parameters. Thus, GLP-1RAs and dual/triple agonists are promising for MAFLD/MASH, especially in those with obesity or T2DM. This study explores mechanisms and clinical evidence of incretin-based therapies for MAFLD by targeting its core pathogenesis-glucolipid disorders. With growing evidence, it also forecasts the broad clinical prospects on MAFLD treatment.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145533921","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This study investigated real-world edoxaban concentrations across different dosing regimens, with a specific focus on patients who met the inclusion criteria of low-dose edoxaban for the Elder Care Atrial Fibrillation (ELDERCARE-AF) trial. Patients with atrial fibrillation receiving edoxaban were enrolled to measure trough concentrations. The levels were compared with the expected therapeutic range (12-43 ng/mL) to define low or high concentrations. Data on ischemic stroke and systemic thromboembolism (SSE), and major bleeding, were collected prospectively. In total, 402 patients (77.2 ± 10.6 years; 52.2% men) were included. The SSE incidence was 1.22 (0.56-2.31) per 100 person years and was significantly associated with low edoxaban concentration (aHR, 4.13 [1.08-15.82]). Among patients meeting the ELDERCARE-AF criteria (n = 160), the use of the 15-mg regimen (ELDER-15 mg, n = 46) vs. the 30-mg regimen (ELDER-30 mg, n = 114) (aOR, 7.65 [2.92-20.09]), higher creatinine clearance (CrCL) (aOR, 1.05 [1.01-1.08]) and lower CHA2DS2-VA score (aOR, 0.69 [0.51, 0.95]) were significantly associated with low concentrations. In the ELDER-15 mg group, patients with CrCL > 50 mL/min had more than 50% probability of exhibiting low drug concentrations. The incidence of SSE was numerically higher in the ELDER-15 mg group than the ELDER-30 mg group (2.34 [0.06-13.02] vs. 1.55 [0.32-4.52]), while major bleeding rates were similar (4.67 [0.57-16.89] vs. 5.15 [2.47-9.48]). In conclusion, low edoxaban concentrations have been associated with thromboembolic events. Among ELDERCARE-AF-eligible patients, the 15-mg regimen results in underexposure to edoxaban, particularly when the CrCL exceeded 50 mL/min.
{"title":"Real-World Edoxaban Concentrations in Older Patients Receiving Reduced-Dose Regimens.","authors":"Sung-Chun Tang, Shin-Yi Lin, Chih-Hao Chen, Yen-Bin Liu, Li-Ting Ho, Ching-Hua Kuo, Yu-Fong Peng, Chih-Fen Huang, Jiann-Shing Jeng","doi":"10.1002/cpt.70129","DOIUrl":"https://doi.org/10.1002/cpt.70129","url":null,"abstract":"<p><p>This study investigated real-world edoxaban concentrations across different dosing regimens, with a specific focus on patients who met the inclusion criteria of low-dose edoxaban for the Elder Care Atrial Fibrillation (ELDERCARE-AF) trial. Patients with atrial fibrillation receiving edoxaban were enrolled to measure trough concentrations. The levels were compared with the expected therapeutic range (12-43 ng/mL) to define low or high concentrations. Data on ischemic stroke and systemic thromboembolism (SSE), and major bleeding, were collected prospectively. In total, 402 patients (77.2 ± 10.6 years; 52.2% men) were included. The SSE incidence was 1.22 (0.56-2.31) per 100 person years and was significantly associated with low edoxaban concentration (aHR, 4.13 [1.08-15.82]). Among patients meeting the ELDERCARE-AF criteria (n = 160), the use of the 15-mg regimen (ELDER-15 mg, n = 46) vs. the 30-mg regimen (ELDER-30 mg, n = 114) (aOR, 7.65 [2.92-20.09]), higher creatinine clearance (CrCL) (aOR, 1.05 [1.01-1.08]) and lower CHA<sub>2</sub>DS<sub>2</sub>-VA score (aOR, 0.69 [0.51, 0.95]) were significantly associated with low concentrations. In the ELDER-15 mg group, patients with CrCL > 50 mL/min had more than 50% probability of exhibiting low drug concentrations. The incidence of SSE was numerically higher in the ELDER-15 mg group than the ELDER-30 mg group (2.34 [0.06-13.02] vs. 1.55 [0.32-4.52]), while major bleeding rates were similar (4.67 [0.57-16.89] vs. 5.15 [2.47-9.48]). In conclusion, low edoxaban concentrations have been associated with thromboembolic events. Among ELDERCARE-AF-eligible patients, the 15-mg regimen results in underexposure to edoxaban, particularly when the CrCL exceeded 50 mL/min.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145533916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marella Cornelia Elizabeth van Ruissen, Menno van Diemen, Liza Botros, Ingrid Tomljanovic, Hind Achbo, Dimitra Eleftheriou, Paul Willem Schenk, Naomi Bertine Klarenbeek, Derek William Gilroy, Manon Aleida Adriana Jansen, Matthijs Moerland
Vascular leakage and its associated phenomena vasodilation and endothelial activation are pathophysiological features of various diseases. Multiple drug candidates targeting these phenomena are in development, necessitating translational models to demonstrate proof-of-pharmacology and proof-of-mechanism in early-phase clinical trials. This single-center experimental study evaluated the intradermal lipopolysaccharide (id LPS) challenge model as a tool to induce and characterize vascular leakage in healthy participants. Eight participants (male:female = 4:4) received id LPS in the volar forearms, followed by serial pharmacodynamic assessments, including imaging and suction blister induction up to 9 hours after injection. Id LPS administration resulted in significant increases in skin perfusion (P < 0.0001), erythema (P = 0.0013), and skin volume (P = 0.0008), indicating initial stages of inflammation and fluid extravasation. Blister fluid analysis revealed elevated extravascular concentrations of albumin (P = 0.0011), total protein (P < 0.0001), and neutrophils (P = 0.0342), supporting the presence of vascular leakage. Moreover, the expression of endothelial activation markers VCAM-1 (P = 0.0015), ICAM-1 (P = 0.0004), ITGB1 (P = 0.01), and E-selectin (P = 0.0218) increased significantly. Disruption of endothelial cell-cell integrity was supported by increased expression of VE-cadherin (P = 0.0002) in blister fluid. These findings support the applicability of the id LPS model for the induction of vascular leakage in humans. This model holds potential as a translational tool for evaluating the pharmacodynamic responses of vascular leakage-targeting drugs in early clinical development.
{"title":"Breaking Barriers: Characterization of the Intradermal Lipopolysaccharide Challenge as an In Vivo Model for Controlled Induction of Vascular Leakage in Healthy Volunteers.","authors":"Marella Cornelia Elizabeth van Ruissen, Menno van Diemen, Liza Botros, Ingrid Tomljanovic, Hind Achbo, Dimitra Eleftheriou, Paul Willem Schenk, Naomi Bertine Klarenbeek, Derek William Gilroy, Manon Aleida Adriana Jansen, Matthijs Moerland","doi":"10.1002/cpt.70126","DOIUrl":"https://doi.org/10.1002/cpt.70126","url":null,"abstract":"<p><p>Vascular leakage and its associated phenomena vasodilation and endothelial activation are pathophysiological features of various diseases. Multiple drug candidates targeting these phenomena are in development, necessitating translational models to demonstrate proof-of-pharmacology and proof-of-mechanism in early-phase clinical trials. This single-center experimental study evaluated the intradermal lipopolysaccharide (id LPS) challenge model as a tool to induce and characterize vascular leakage in healthy participants. Eight participants (male:female = 4:4) received id LPS in the volar forearms, followed by serial pharmacodynamic assessments, including imaging and suction blister induction up to 9 hours after injection. Id LPS administration resulted in significant increases in skin perfusion (P < 0.0001), erythema (P = 0.0013), and skin volume (P = 0.0008), indicating initial stages of inflammation and fluid extravasation. Blister fluid analysis revealed elevated extravascular concentrations of albumin (P = 0.0011), total protein (P < 0.0001), and neutrophils (P = 0.0342), supporting the presence of vascular leakage. Moreover, the expression of endothelial activation markers VCAM-1 (P = 0.0015), ICAM-1 (P = 0.0004), ITGB1 (P = 0.01), and E-selectin (P = 0.0218) increased significantly. Disruption of endothelial cell-cell integrity was supported by increased expression of VE-cadherin (P = 0.0002) in blister fluid. These findings support the applicability of the id LPS model for the induction of vascular leakage in humans. This model holds potential as a translational tool for evaluating the pharmacodynamic responses of vascular leakage-targeting drugs in early clinical development.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145522559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The short-term efficacy of allopurinol add-on thiopurine therapy has been well documented; however, the long-term effects and dose recommendations are poorly detailed. This study aimed to elucidate the long-term implications of this combination therapy and to develop initial dose applying pharmacokinetic modeling. Forty-two pediatric patients with acute lymphoblastic leukemia, treated with 6-mercaptopurine (6-MP) and allopurinol, were enrolled in this prospective before-after pharmacokinetic study. Metabolite levels (6-thioguanine nucleotides (6-TGNs), methyl mercaptopurine nucleotides (6-MMPN), DNA-thioguanine (DNA-TG)), thiopurine methyltransferase (TPMT) activity were measured pre- and post-combination. Another retrospective cohort of 40 patients receiving 6-MP monotherapy was taken as controls. Compared with the control group, the combination therapy showed a similar myelosuppression effect ((P = 0.060, adjusted Hazard Rates, aHR = 0.94 (0.89-1.00)), while markedly reducing the cumulative hazard of severe neutropenia (P = 0.009, aHR = 0.49 (0.28-0.83)) and hepatotoxicity (P < 0.001, aHR = 0.54 (0.40-0.73)). Allopurinol combination led to a fourfold reduction in TPMT activity, the 6-MMPN:6-TGNs ratios, and 6-MMPN: DNA-TG ratios. This metabolic adjustment improved control of white blood cell (WBC) counts, neutrophil counts (ANC), and aminotransferase levels. LOESS regression estimates indicated significant fluctuations in WBC, ANC, and 6-MP/allopurinol dosage ratios following 3 months of combination therapy (P < 0.001), reflecting the need for close monitoring and frequent dose adjustments. Pharmacokinetic analysis further reinforces the benefits of allopurinol add-on 6-MP strategy, and suggested that, for patients with normal or high TPMT activity, an initial 6-MP dose of 20-30 mg/m2/day is recommended for those with BSA ≤ 1 m2, and 15-20 mg/m2/day for those with BSA > 1 m2, when co-administered with allopurinol (50 mg/m2/day).
{"title":"Allopurinol Add-on 6-Mercaptopurine Strategy Improves Efficacy and Reduces Toxicity in Pediatric Patients With Acute Lymphoblastic Leukemia.","authors":"Yanping Guan, Xiaoli Zhang, Sumyuet Chan, Shan Su, Qiaolan Xuan, Ting Yang, Xuequn Luo, Zhong Zuo, Dunhua Zhou, Min Huang, Xueding Wang, Libin Huang","doi":"10.1002/cpt.70123","DOIUrl":"https://doi.org/10.1002/cpt.70123","url":null,"abstract":"<p><p>The short-term efficacy of allopurinol add-on thiopurine therapy has been well documented; however, the long-term effects and dose recommendations are poorly detailed. This study aimed to elucidate the long-term implications of this combination therapy and to develop initial dose applying pharmacokinetic modeling. Forty-two pediatric patients with acute lymphoblastic leukemia, treated with 6-mercaptopurine (6-MP) and allopurinol, were enrolled in this prospective before-after pharmacokinetic study. Metabolite levels (6-thioguanine nucleotides (6-TGNs), methyl mercaptopurine nucleotides (6-MMPN), DNA-thioguanine (DNA-TG)), thiopurine methyltransferase (TPMT) activity were measured pre- and post-combination. Another retrospective cohort of 40 patients receiving 6-MP monotherapy was taken as controls. Compared with the control group, the combination therapy showed a similar myelosuppression effect ((P = 0.060, adjusted Hazard Rates, aHR = 0.94 (0.89-1.00)), while markedly reducing the cumulative hazard of severe neutropenia (P = 0.009, aHR = 0.49 (0.28-0.83)) and hepatotoxicity (P < 0.001, aHR = 0.54 (0.40-0.73)). Allopurinol combination led to a fourfold reduction in TPMT activity, the 6-MMPN:6-TGNs ratios, and 6-MMPN: DNA-TG ratios. This metabolic adjustment improved control of white blood cell (WBC) counts, neutrophil counts (ANC), and aminotransferase levels. LOESS regression estimates indicated significant fluctuations in WBC, ANC, and 6-MP/allopurinol dosage ratios following 3 months of combination therapy (P < 0.001), reflecting the need for close monitoring and frequent dose adjustments. Pharmacokinetic analysis further reinforces the benefits of allopurinol add-on 6-MP strategy, and suggested that, for patients with normal or high TPMT activity, an initial 6-MP dose of 20-30 mg/m<sup>2</sup>/day is recommended for those with BSA ≤ 1 m<sup>2</sup>, and 15-20 mg/m<sup>2</sup>/day for those with BSA > 1 m<sup>2</sup>, when co-administered with allopurinol (50 mg/m<sup>2</sup>/day).</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145522612","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Physiology Based Pharmacokinetic (PBPK) modeling is an established essential tool for predicting and/or analyzing drug-drug interactions (DDI). Uncertainty and variability associated with in vitro determined DDI-related parameters have often been considered a limitation for predicting PBPK-DDIs. Sensitivity analysis (SA) around DDI input parameters using PBPK analysis is often applied for assessing the relevance of clinical DDI predictions/prioritization/study designs. This perspective aims to explore and advocate practical approaches for precipitant (inhibitor/inducer) PBPK-DDI SA for optimal clinically relevant evaluations.
{"title":"Strategy for Identifying Rational Sensitivity Analysis Using PBPK Modeling for Precipitant Drug-Drug Interaction Predictions.","authors":"Kunal S Taskar, Pradeep Sharma, Karen Rowland Yeo","doi":"10.1002/cpt.70127","DOIUrl":"https://doi.org/10.1002/cpt.70127","url":null,"abstract":"<p><p>Physiology Based Pharmacokinetic (PBPK) modeling is an established essential tool for predicting and/or analyzing drug-drug interactions (DDI). Uncertainty and variability associated with in vitro determined DDI-related parameters have often been considered a limitation for predicting PBPK-DDIs. Sensitivity analysis (SA) around DDI input parameters using PBPK analysis is often applied for assessing the relevance of clinical DDI predictions/prioritization/study designs. This perspective aims to explore and advocate practical approaches for precipitant (inhibitor/inducer) PBPK-DDI SA for optimal clinically relevant evaluations.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145501262","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maddalena Centanni, Afroditi Nanou, Leon W M M Terstappen, Kees J A Punt, Frank A W Coumans, Mats O Karlsson, Lena E Friberg
Circulating tumor cells (CTCs) and tumor-derived extracellular vesicles (tdEVs) are promising biomarkers for predicting survival and informing treatment decisions in metastatic colorectal cancer (mCRC); yet their clinical application remains limited. In this study, we analyzed CellSearch image archives from 446 patients with mCRC treated in the CAIRO2 study to evaluate the predictive value of CTCs and tdEVs and explore their utility in guiding personalized therapy. Using pharmacodynamic modeling, we examined longitudinal changes in manually and automatically classified CTC and tdEV counts, assessing their relationship with tumor size dynamics and overall survival. Automated tdEV counts demonstrated the strongest association with survival, followed by automated CTCs, which outperformed manually assessed counts. The combination of automated tdEVs and manual CTCs further improved predictive performance. Simulations predict transitioning high-risk patients (CTC/tdEV score >1.75) to second-line FOLFIRI at week 4 or 10 improves median survival from 15.1 months (12.8-18.4 CI) to 22.7 months (17.1-35.8 CI), and from 13.3 months (10.5-21.7 CI) to 23.3 months (17.8-31.2 CI), respectively. Additionally, biomarker monitoring demonstrated reduced cost (€3,913 vs. €7,802) and environmental burden (10 kg vs. 167 kg CO2e per patient). These findings suggest that tdEVs, alone or in combination with CTCs, may help optimize treatment timing and outcomes in mCRC. The integration of CTCs and tdEVs into clinical practice could offer a personalized, cost-effective, and more sustainable alternative to routine imaging in managing advanced colorectal cancer.
{"title":"Longitudinal Analysis of Manually and Automatically Classified Circulating Tumor Biomarkers and their Prediction of Survival in Metastatic Colorectal Cancer.","authors":"Maddalena Centanni, Afroditi Nanou, Leon W M M Terstappen, Kees J A Punt, Frank A W Coumans, Mats O Karlsson, Lena E Friberg","doi":"10.1002/cpt.70128","DOIUrl":"https://doi.org/10.1002/cpt.70128","url":null,"abstract":"<p><p>Circulating tumor cells (CTCs) and tumor-derived extracellular vesicles (tdEVs) are promising biomarkers for predicting survival and informing treatment decisions in metastatic colorectal cancer (mCRC); yet their clinical application remains limited. In this study, we analyzed CellSearch image archives from 446 patients with mCRC treated in the CAIRO2 study to evaluate the predictive value of CTCs and tdEVs and explore their utility in guiding personalized therapy. Using pharmacodynamic modeling, we examined longitudinal changes in manually and automatically classified CTC and tdEV counts, assessing their relationship with tumor size dynamics and overall survival. Automated tdEV counts demonstrated the strongest association with survival, followed by automated CTCs, which outperformed manually assessed counts. The combination of automated tdEVs and manual CTCs further improved predictive performance. Simulations predict transitioning high-risk patients (CTC/tdEV score >1.75) to second-line FOLFIRI at week 4 or 10 improves median survival from 15.1 months (12.8-18.4 CI) to 22.7 months (17.1-35.8 CI), and from 13.3 months (10.5-21.7 CI) to 23.3 months (17.8-31.2 CI), respectively. Additionally, biomarker monitoring demonstrated reduced cost (€3,913 vs. €7,802) and environmental burden (10 kg vs. 167 kg CO<sub>2</sub>e per patient). These findings suggest that tdEVs, alone or in combination with CTCs, may help optimize treatment timing and outcomes in mCRC. The integration of CTCs and tdEVs into clinical practice could offer a personalized, cost-effective, and more sustainable alternative to routine imaging in managing advanced colorectal cancer.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145501211","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Namam Ali, Stephan van Erp, Cornelis Kramers, Cornelis J Tack, Bastiaan E de Galan
Polypharmacy is increasingly recognized as a relevant issue in diabetes care, but its prevalence and clinical relevance in individuals with type 1 diabetes remain underexplored. This study aimed to determine the prevalence of polypharmacy and to identify associated clinical and psychological factors. Participants were recruited from a tertiary diabetes outpatient clinic between February 2020 and April 2021. Polypharmacy was defined as the concurrent use of five or more medications, including insulin. Clinical, sensor-based, and psychosocial data were collected. Logistic regression was used to identify variables independently associated with polypharmacy. A total of 484 individuals with type 1 diabetes were included (mean age 51.3 ± 15.9 years; 51.2% male; median diabetes duration 30 [IQR 16-40] years; mean HbA1c 60.3 ± 11.6 mmol/mol). Polypharmacy was present in 175 (36.2%) participants. Individuals with polypharmacy were more often female, were older, and had longer diabetes duration, higher BMI, higher HbA1c, more complications, and higher rates of hospital admission. They also were more likely to have impaired awareness of hypoglycemia and reported higher levels of fear of hypoglycemia with no differences in hyperglycemia-related worry or behavior or diabetes-related emotional distress. Polypharmacy affects over one-third of individuals with type 1 diabetes and is associated with poorer health status and a greater hypoglycemia-related burden. Future studies should investigate whether targeted medication review and psychological interventions may alleviate some of the burden in this high-risk group.
{"title":"The Prevalence and Implications of Polypharmacy in Individuals With Type 1 Diabetes.","authors":"Namam Ali, Stephan van Erp, Cornelis Kramers, Cornelis J Tack, Bastiaan E de Galan","doi":"10.1002/cpt.70130","DOIUrl":"https://doi.org/10.1002/cpt.70130","url":null,"abstract":"<p><p>Polypharmacy is increasingly recognized as a relevant issue in diabetes care, but its prevalence and clinical relevance in individuals with type 1 diabetes remain underexplored. This study aimed to determine the prevalence of polypharmacy and to identify associated clinical and psychological factors. Participants were recruited from a tertiary diabetes outpatient clinic between February 2020 and April 2021. Polypharmacy was defined as the concurrent use of five or more medications, including insulin. Clinical, sensor-based, and psychosocial data were collected. Logistic regression was used to identify variables independently associated with polypharmacy. A total of 484 individuals with type 1 diabetes were included (mean age 51.3 ± 15.9 years; 51.2% male; median diabetes duration 30 [IQR 16-40] years; mean HbA<sub>1c</sub> 60.3 ± 11.6 mmol/mol). Polypharmacy was present in 175 (36.2%) participants. Individuals with polypharmacy were more often female, were older, and had longer diabetes duration, higher BMI, higher HbA<sub>1c</sub>, more complications, and higher rates of hospital admission. They also were more likely to have impaired awareness of hypoglycemia and reported higher levels of fear of hypoglycemia with no differences in hyperglycemia-related worry or behavior or diabetes-related emotional distress. Polypharmacy affects over one-third of individuals with type 1 diabetes and is associated with poorer health status and a greater hypoglycemia-related burden. Future studies should investigate whether targeted medication review and psychological interventions may alleviate some of the burden in this high-risk group.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145493891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Eva-Maria A Wansing, Sebastian G Wicha, Peter Bannas, Alexander Heitkamp, Adrin Dadkhah, Nicolaus M Kröger, Isabel Molwitz, Claudia Langebrake
Knowledge of the glomerular filtration rate (GFR) is mandatory when dosing renally eliminated drugs such as vancomycin. In clinical practice, different biomarkers and various equations are used to estimate GFR (eGFR), resulting in varying estimates. These variations may be explained by nonrenal factors, such as muscle status or glucocorticoid administration. This study aimed to evaluate the performance of different eGFR equations in terms of accuracy and precision compared to renal vancomycin clearance, including subgroup analyses for nonrenal confounders. We retrospectively analyzed data from 121 adult allogeneic hematopoietic stem cell transplant (allo-HSCT) patients. All patients received vancomycin treatment including trough concentration therapeutic drug monitoring. The eGFR was calculated using eight equations and compared to the renal vancomycin clearance that was calculated using a pharmacokinetic model and served as the reference. Individual muscle status was determined by computed tomography scans. Median renal vancomycin clearance was 49 mL/minute/1.73 m2 (range 24-96). All eight eGFR equations overestimated renal vancomycin clearance. The six (partially) creatinine-based equations were significantly less accurate (bias: 24.0-62.8 mL/minute/1.73 m2) than both cystatin C-based equations (bias: 6.3-9.5 mL/minute/1.73 m2). This decreased accuracy for creatinine-based eGFR was more pronounced in patients with reduced muscle status or glucocorticoid medication. All CKD-EPI equations and the Hoek equation were more precise with an IQR of the difference to renal vancomycin clearance ≤22.5 mL/minute/1.73 m2 compared to ≥35.5 mL/minute/1.73 m2 (Cockcroft-Gault, MDRD). In conclusion, cystatin C-based eGFR equations are preferable to creatinine-based approaches for vancomycin dosing in allo-HSCT patients.
{"title":"Cystatin C-based eGFR better predicts renal vancomycin clearance than creatinine-based eGFR in patients with allogeneic stem cell transplantation.","authors":"Eva-Maria A Wansing, Sebastian G Wicha, Peter Bannas, Alexander Heitkamp, Adrin Dadkhah, Nicolaus M Kröger, Isabel Molwitz, Claudia Langebrake","doi":"10.1002/cpt.70125","DOIUrl":"https://doi.org/10.1002/cpt.70125","url":null,"abstract":"<p><p>Knowledge of the glomerular filtration rate (GFR) is mandatory when dosing renally eliminated drugs such as vancomycin. In clinical practice, different biomarkers and various equations are used to estimate GFR (eGFR), resulting in varying estimates. These variations may be explained by nonrenal factors, such as muscle status or glucocorticoid administration. This study aimed to evaluate the performance of different eGFR equations in terms of accuracy and precision compared to renal vancomycin clearance, including subgroup analyses for nonrenal confounders. We retrospectively analyzed data from 121 adult allogeneic hematopoietic stem cell transplant (allo-HSCT) patients. All patients received vancomycin treatment including trough concentration therapeutic drug monitoring. The eGFR was calculated using eight equations and compared to the renal vancomycin clearance that was calculated using a pharmacokinetic model and served as the reference. Individual muscle status was determined by computed tomography scans. Median renal vancomycin clearance was 49 mL/minute/1.73 m<sup>2</sup> (range 24-96). All eight eGFR equations overestimated renal vancomycin clearance. The six (partially) creatinine-based equations were significantly less accurate (bias: 24.0-62.8 mL/minute/1.73 m<sup>2</sup>) than both cystatin C-based equations (bias: 6.3-9.5 mL/minute/1.73 m<sup>2</sup>). This decreased accuracy for creatinine-based eGFR was more pronounced in patients with reduced muscle status or glucocorticoid medication. All CKD-EPI equations and the Hoek equation were more precise with an IQR of the difference to renal vancomycin clearance ≤22.5 mL/minute/1.73 m<sup>2</sup> compared to ≥35.5 mL/minute/1.73 m<sup>2</sup> (Cockcroft-Gault, MDRD). In conclusion, cystatin C-based eGFR equations are preferable to creatinine-based approaches for vancomycin dosing in allo-HSCT patients.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145493935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Joseph Piscitelli, Micaela B Reddy, Lance Wollenberg, Laurence Del Frari, Jason Gong, Kyle Matschke, Jason H Williams
A clinical drug-drug interaction (DDI) study was designed to evaluate the effect of single and multiple oral doses of encorafenib on the single oral dose pharmacokinetics (PK) of the cytochrome P450 (CYP) enzyme probe substrates, losartan (CYP2C9), midazolam (CYP3A4), caffeine (CYP1A2), omeprazole (CYP2C19), and dextromethorphan (CYP2D6) administered as a cocktail (Inje). This study was conducted, post-approval, in patients with BRAF V600-mutant advanced solid tumors, and aimed to address the remaining uncertainty in the DDI potential of encorafenib as a perpetrator of these CYP enzymes. Study participants received the cocktail on Days -7, 1, and 14 and continuous doses of encorafenib (450 mg q.d.) and binimetinib (45 mg b.i.d.) starting on Day 1. PK sampling and urine collection were conducted from 0 to 8 hours on cocktail administration days. PK parameters were calculated for each participant using noncompartmental analysis of concentration-time data or amount excreted for urine parameters. At steady-state encorafenib plasma concentrations, midazolam plasma Cmax and AUClast decreased by 74% and 82%, respectively. No clinically significant DDIs were observed at encorafenib steady-state concentrations with the other probe substrates of interest. The results from this clinical study indicate that encorafenib is a strong inducer of CYP3A (≥ 80% decrease in midazolam area under the curve (AUC)) at steady state. Based on these results regarding co-administration with encorafenib, sensitive substrates of CYP3A should be avoided or dose adjusted based on the recommendations of their approved product labeling. This information has been included in the updated prescribing information for encorafenib.
{"title":"An Evaluation of the Drug Interaction Potential of Encorafenib in Combination With Binimetinib Using the Inje Cocktail in Patients With Cancer.","authors":"Joseph Piscitelli, Micaela B Reddy, Lance Wollenberg, Laurence Del Frari, Jason Gong, Kyle Matschke, Jason H Williams","doi":"10.1002/cpt.70117","DOIUrl":"https://doi.org/10.1002/cpt.70117","url":null,"abstract":"<p><p>A clinical drug-drug interaction (DDI) study was designed to evaluate the effect of single and multiple oral doses of encorafenib on the single oral dose pharmacokinetics (PK) of the cytochrome P450 (CYP) enzyme probe substrates, losartan (CYP2C9), midazolam (CYP3A4), caffeine (CYP1A2), omeprazole (CYP2C19), and dextromethorphan (CYP2D6) administered as a cocktail (Inje). This study was conducted, post-approval, in patients with BRAF V600-mutant advanced solid tumors, and aimed to address the remaining uncertainty in the DDI potential of encorafenib as a perpetrator of these CYP enzymes. Study participants received the cocktail on Days -7, 1, and 14 and continuous doses of encorafenib (450 mg q.d.) and binimetinib (45 mg b.i.d.) starting on Day 1. PK sampling and urine collection were conducted from 0 to 8 hours on cocktail administration days. PK parameters were calculated for each participant using noncompartmental analysis of concentration-time data or amount excreted for urine parameters. At steady-state encorafenib plasma concentrations, midazolam plasma C<sub>max</sub> and AUC<sub>last</sub> decreased by 74% and 82%, respectively. No clinically significant DDIs were observed at encorafenib steady-state concentrations with the other probe substrates of interest. The results from this clinical study indicate that encorafenib is a strong inducer of CYP3A (≥ 80% decrease in midazolam area under the curve (AUC)) at steady state. Based on these results regarding co-administration with encorafenib, sensitive substrates of CYP3A should be avoided or dose adjusted based on the recommendations of their approved product labeling. This information has been included in the updated prescribing information for encorafenib.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145487208","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Conor J O'Hanlon, Jonas Denck, Elif Ozkirimli, Stefanie Bendels, Candice Jamois, Clarisse Chavanne, Dirk Fey, Kimmo Porkka, Oscar Brück, Ken Wang
Accurately labeling outcomes in real-world data for machine learning is challenging due to data sparsity and imbalances. This study developed and evaluated a pharmacokinetic-pharmacodynamic (PKPD)-informed labeling strategy to enhance the risk prediction of docetaxel-induced neutropenia. Machine learning models were trained on real-world data from 4,248 patients using two approaches for comparison. The "naive" labeling method used only neutrophil observations, while the "PKPD-informed" method used simulations from a semi-mechanistic model to determine the neutrophil nadir for each treatment cycle. Three machine learning models (logistic regression, XGBoost, TabPFN) were trained with baseline laboratory data to predict severe neutropenia (neutrophil count <0.1 cells × 109/L) prior to the first docetaxel dose. The PKPD labeling approach enabled the labeling of 3.4 times more patient instances (7,719 vs. 2,283) than the naive method. Across all machine learning architectures, models trained with PKPD-informed labels demonstrated significantly superior predictive performance (AUC-ROC and AUC-PR) compared to those trained with naive labels. This advantage was maintained even when training set sizes were matched. PKPD-informed labeling overcomes limitations of sparse real-world data, increasing both the quantity and apparent quality of labels for machine learning model training. This methodology enhances the performance of machine learning models for predicting severe neutropenia and represents a robust, generalizable framework for improving clinical outcome prediction.
{"title":"Enhancing Severe Neutropenia Prediction: PKPD-Informed Labeling for Machine Learning Models Trained on Real-World Data.","authors":"Conor J O'Hanlon, Jonas Denck, Elif Ozkirimli, Stefanie Bendels, Candice Jamois, Clarisse Chavanne, Dirk Fey, Kimmo Porkka, Oscar Brück, Ken Wang","doi":"10.1002/cpt.70116","DOIUrl":"https://doi.org/10.1002/cpt.70116","url":null,"abstract":"<p><p>Accurately labeling outcomes in real-world data for machine learning is challenging due to data sparsity and imbalances. This study developed and evaluated a pharmacokinetic-pharmacodynamic (PKPD)-informed labeling strategy to enhance the risk prediction of docetaxel-induced neutropenia. Machine learning models were trained on real-world data from 4,248 patients using two approaches for comparison. The \"naive\" labeling method used only neutrophil observations, while the \"PKPD-informed\" method used simulations from a semi-mechanistic model to determine the neutrophil nadir for each treatment cycle. Three machine learning models (logistic regression, XGBoost, TabPFN) were trained with baseline laboratory data to predict severe neutropenia (neutrophil count <0.1 cells × 10<sup>9</sup>/L) prior to the first docetaxel dose. The PKPD labeling approach enabled the labeling of 3.4 times more patient instances (7,719 vs. 2,283) than the naive method. Across all machine learning architectures, models trained with PKPD-informed labels demonstrated significantly superior predictive performance (AUC-ROC and AUC-PR) compared to those trained with naive labels. This advantage was maintained even when training set sizes were matched. PKPD-informed labeling overcomes limitations of sparse real-world data, increasing both the quantity and apparent quality of labels for machine learning model training. This methodology enhances the performance of machine learning models for predicting severe neutropenia and represents a robust, generalizable framework for improving clinical outcome prediction.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145480355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}