Anees M. Dauki, Nishanthan Rajakumaraswamy, Torsten Trowe, Winnie Weng, Kai-Wen Lin, Emon Elboudjwarej, Ann Ran-Ran Qin, Christian Schwabe, Michelle R. Kuhne, Ahmed A. Othman
Conventional dendritic cells subtype 1 (cDC1) play a vital role in the priming and expansion of tumor-specific CD8+ T cells and their recruitment to tumor microenvironment. However, cDC1s are often underrepresented in the microenvironment. Systemic administration of Fms-like tyrosine kinase 3 ligand, a hematopoietic growth factor that binds to FLT3 on myeloid and lymphoid progenitor cells, leads to cDC1 expansion in the periphery and recruitment into the microenvironment. FLT3 pathway stimulation using GS-3583, a novel FLT3 agonistic Fc fusion protein, has the potential to promote T-cell mediated antitumor activity. This was a first-in-human, placebo-controlled study of GS-3583 in healthy participants to evaluate the safety, pharmacokinetics (PK), and pharmacodynamic (PD) of escalating single doses (75–2000 μg) of GS-3583. Each dose cohort enrolled 8–12 healthy participants who received GS-3583 or placebo as single IV infusion at 3:1 ratio. As part of the PD evaluation, the changes in the number of cDC1 cells were investigated. GS-3583 was well-tolerated in healthy participants up to the highest evaluated dose (2000 μg). There have been no serious or grade III or higher adverse events. PK analysis suggested a dose-dependent increase in GS-3583 exposure with target-mediated disposition characteristics at low doses. PD analysis shows that administration of GS-3583 resulted in transient, dose-dependent increases in cDC1 cells that returned to baseline within 3 weeks of drug administration. The pharmacokinetics and pharmacodynamics of GS-3583 following single dosing were characterized in this study which enabled subsequent phase Ib assessments in patients with advanced solid tumors.
{"title":"Pharmacokinetics, pharmacodynamics, and safety of GS-3583, a FLT3 agonist Fc fusion protein, from single-ascending-dose phase I study in healthy participants","authors":"Anees M. Dauki, Nishanthan Rajakumaraswamy, Torsten Trowe, Winnie Weng, Kai-Wen Lin, Emon Elboudjwarej, Ann Ran-Ran Qin, Christian Schwabe, Michelle R. Kuhne, Ahmed A. Othman","doi":"10.1111/cts.70011","DOIUrl":"10.1111/cts.70011","url":null,"abstract":"<p>Conventional dendritic cells subtype 1 (cDC1) play a vital role in the priming and expansion of tumor-specific CD8+ T cells and their recruitment to tumor microenvironment. However, cDC1s are often underrepresented in the microenvironment. Systemic administration of Fms-like tyrosine kinase 3 ligand, a hematopoietic growth factor that binds to FLT3 on myeloid and lymphoid progenitor cells, leads to cDC1 expansion in the periphery and recruitment into the microenvironment. FLT3 pathway stimulation using GS-3583, a novel FLT3 agonistic Fc fusion protein, has the potential to promote T-cell mediated antitumor activity. This was a first-in-human, placebo-controlled study of GS-3583 in healthy participants to evaluate the safety, pharmacokinetics (PK), and pharmacodynamic (PD) of escalating single doses (75–2000 μg) of GS-3583. Each dose cohort enrolled 8–12 healthy participants who received GS-3583 or placebo as single IV infusion at 3:1 ratio. As part of the PD evaluation, the changes in the number of cDC1 cells were investigated. GS-3583 was well-tolerated in healthy participants up to the highest evaluated dose (2000 μg). There have been no serious or grade III or higher adverse events. PK analysis suggested a dose-dependent increase in GS-3583 exposure with target-mediated disposition characteristics at low doses. PD analysis shows that administration of GS-3583 resulted in transient, dose-dependent increases in cDC1 cells that returned to baseline within 3 weeks of drug administration. The pharmacokinetics and pharmacodynamics of GS-3583 following single dosing were characterized in this study which enabled subsequent phase Ib assessments in patients with advanced solid tumors.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70011","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ezhilpavai Mohanan, Guofang Shen, Suping Ren, Hsuan-Hao Fan, Kao Tang Ying Moua, Aleksandra Karolak, Russell C. Rockne, Ryotaro Nakamura, David A. Horne, Christopher G. Kanakry, Donald E. Mager, Jeannine S. McCune
Dose optimization of sirolimus may further improve outcomes in allogeneic hematopoietic cell transplant (HCT) patients receiving post-transplantation cyclophosphamide (PTCy) to prevent graft-versus-host disease (GVHD). Sirolimus exposure–response association studies in HCT patients (i.e., the association of trough concentration with clinical outcomes) have been conflicting. Sirolimus has important effects on T-cells, including conventional (Tcons) and regulatory T-cells (Tregs), both of which have been implicated in the mechanisms by which PTCy prevents GVHD, but there is an absence of validated biomarkers of sirolimus effects on these cell subsets. Considering the paucity of existing biomarkers and the complexities of the immune system, we conducted a literature review to inform a quantitative systems pharmacology (QSP) model of GVHD. The published literature presented multiple challenges. The sirolimus pharmacokinetic models insufficiently describe sirolimus distribution to relevant physiological compartments. Despite multiple publications describing sirolimus effects on Tcons and Tregs in preclinical and human ex vivo models, consistent parameters relating sirolimus concentrations to circulating Tcons and Tregs could not be found. Each aspect presents a challenge in building a QSP model of sirolimus and its temporal effects on T-cell subsets and GVHD prevention. To optimize GVHD prevention regimens, phase I studies and systematic studies of immunosuppressant concentration–effect association are needed for QSP modeling.
西罗莫司剂量的优化可进一步改善接受移植后环磷酰胺(PTCy)治疗以预防移植物抗宿主病(GVHD)的异基因造血细胞移植(HCT)患者的预后。对 HCT 患者进行的西罗莫司暴露-反应关联研究(即谷浓度与临床结果的关联)一直存在矛盾。西罗莫司对 T 细胞有重要影响,包括常规 T 细胞(Tcons)和调节性 T 细胞(Tregs),这两种细胞都与 PTCy 预防 GVHD 的机制有关,但目前还没有西罗莫司对这些细胞亚群影响的有效生物标志物。考虑到现有生物标志物的匮乏和免疫系统的复杂性,我们进行了一次文献综述,为GVHD的定量系统药理学(QSP)模型提供信息。已发表的文献提出了多重挑战。西罗莫司药代动力学模型没有充分描述西罗莫司在相关生理区域的分布。尽管有多篇文献描述了西罗莫司在临床前和人体体外模型中对Tcons和Tregs的影响,但无法找到西罗莫司浓度与循环中Tcons和Tregs相关的一致参数。在建立西罗莫司及其对T细胞亚群和GVHD预防的时间效应的QSP模型时,每个方面都是一个挑战。为了优化 GVHD 预防方案,需要对免疫抑制剂浓度效应关联进行 I 期研究和系统研究,以建立 QSP 模型。
{"title":"Challenges with sirolimus experimental data to inform QSP model of post-transplantation cyclophosphamide regimens","authors":"Ezhilpavai Mohanan, Guofang Shen, Suping Ren, Hsuan-Hao Fan, Kao Tang Ying Moua, Aleksandra Karolak, Russell C. Rockne, Ryotaro Nakamura, David A. Horne, Christopher G. Kanakry, Donald E. Mager, Jeannine S. McCune","doi":"10.1111/cts.70014","DOIUrl":"10.1111/cts.70014","url":null,"abstract":"<p>Dose optimization of sirolimus may further improve outcomes in allogeneic hematopoietic cell transplant (HCT) patients receiving post-transplantation cyclophosphamide (PTCy) to prevent graft-versus-host disease (GVHD). Sirolimus exposure–response association studies in HCT patients (i.e., the association of trough concentration with clinical outcomes) have been conflicting. Sirolimus has important effects on T-cells, including conventional (Tcons) and regulatory T-cells (Tregs), both of which have been implicated in the mechanisms by which PTCy prevents GVHD, but there is an absence of validated biomarkers of sirolimus effects on these cell subsets. Considering the paucity of existing biomarkers and the complexities of the immune system, we conducted a literature review to inform a quantitative systems pharmacology (QSP) model of GVHD. The published literature presented multiple challenges. The sirolimus pharmacokinetic models insufficiently describe sirolimus distribution to relevant physiological compartments. Despite multiple publications describing sirolimus effects on Tcons and Tregs in preclinical and human ex vivo models, consistent parameters relating sirolimus concentrations to circulating Tcons and Tregs could not be found. Each aspect presents a challenge in building a QSP model of sirolimus and its temporal effects on T-cell subsets and GVHD prevention. To optimize GVHD prevention regimens, phase I studies and systematic studies of immunosuppressant concentration–effect association are needed for QSP modeling.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70014","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142005735","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Florencia A. Tettamanti, Holly Kimko, Shringi Sharma, Giovanni Di Veroli
Association between measurable residual disease (MRD) and survival outcomes in chronic lymphocytic leukemia (CLL) has often been reported. However, limited quantitative analyses over large datasets have been undertaken to establish the predictive power of MRD. Here, we provide a comprehensive assessment of published MRD data to explore the utility of MRD in the prediction of progression-free survival (PFS). We undertook two independent analyses, which leveraged available published data to address two complimentary questions. In the first, data from eight clinical trials was modeled via a meta-regression approach, showing that median PFS can be predicted from undetectable MRD rates at 3–6 months of post-treatment. The resulting model can be used to predict the probability of technical success of a planned clinical trial in chemotherapy. In the second, we investigated the evidence for predicting PFS from competing MRD metrics, for example baseline value and instantaneous MRD value, via a joint modeling approach. Using data from four small studies, we found strong evidence that including MRD metrics in joint models improves predictions of PFS compared with not including them. This analysis suggests that incorporating MRD is likely to better inform individual progression predictions. It is therefore proposed that systematic MRD collection should be accompanied by modeling to generate algorithms that inform patients' progression.
{"title":"Predicting progression-free survival from measurable residual disease in chronic lymphocytic leukemia","authors":"Florencia A. Tettamanti, Holly Kimko, Shringi Sharma, Giovanni Di Veroli","doi":"10.1111/cts.13905","DOIUrl":"10.1111/cts.13905","url":null,"abstract":"<p>Association between measurable residual disease (MRD) and survival outcomes in chronic lymphocytic leukemia (CLL) has often been reported. However, limited quantitative analyses over large datasets have been undertaken to establish the predictive power of MRD. Here, we provide a comprehensive assessment of published MRD data to explore the utility of MRD in the prediction of progression-free survival (PFS). We undertook two independent analyses, which leveraged available published data to address two complimentary questions. In the first, data from eight clinical trials was modeled via a meta-regression approach, showing that median PFS can be predicted from undetectable MRD rates at 3–6 months of post-treatment. The resulting model can be used to predict the probability of technical success of a planned clinical trial in chemotherapy. In the second, we investigated the evidence for predicting PFS from competing MRD metrics, for example baseline value and instantaneous MRD value, via a joint modeling approach. Using data from four small studies, we found strong evidence that including MRD metrics in joint models improves predictions of PFS compared with not including them. This analysis suggests that incorporating MRD is likely to better inform individual progression predictions. It is therefore proposed that systematic MRD collection should be accompanied by modeling to generate algorithms that inform patients' progression.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.13905","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009911","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hidetoshi Shimizu, Michael A. Tortorici, Yoshiyasu Ohta, Kei Ogawa, Sheikh Mohammed Ashfaq Rahman, Aya Fujii, Yuki Hiraga, Mizue Kawai, Kanami Sugimoto-Kawabata, Mattheus (Thijs) van Iersel, Jan Jaap van Lier, Stephen Djedjos, B. T. Slingsby, David M. Rodman
Dysregulation of the mineralocorticoid hormone aldosterone is an increasingly prevalent cause of hypertension. Aldosterone synthase (CYP11B2) shares 93% homology to 11β-hydroxylase (CYP11B1), which produces cortisol. Lorundrostat, a highly selective inhibitor of CYP11B2, is a potential safe and effective treatment for aldosterone-dependent, uncontrolled hypertension, including treatment-resistant hypertension. Lorundrostat showed highly selective inhibition of CYP11B2 in vitro, with 374-fold selectivity for CYP11B2 vs. CYP11B1. A first-in-human study of single ascending doses ranging from 5 to 800 mg and multiple ascending doses ranging from 40 to 360 mg once daily was conducted in healthy participants. After single- and multiple-dose administration, lorundrostat plasma levels peaked 1–3 h after administration with a t1/2 of 10–12 h. Plasma aldosterone decreased up to 40% with single 100-mg to 200-mg doses and up to 70% with single 400 to 800-mg doses. Plasma aldosterone returned to baseline within 16 h after single 100-mg doses and multiple once-daily 120-mg doses. Lorundrostat demonstrated a favorable safety profile in healthy participants. Dose- and exposure-dependent inhibition of renal tubular sodium reabsorption was observed across a clinically relevant dose range with no suppression of basal or cosyntropin-stimulated cortisol production and only a modest increase in mean serum potassium.
{"title":"First-in-human study evaluating safety, pharmacokinetics, and pharmacodynamics of lorundrostat, a novel and highly selective aldosterone synthase inhibitor","authors":"Hidetoshi Shimizu, Michael A. Tortorici, Yoshiyasu Ohta, Kei Ogawa, Sheikh Mohammed Ashfaq Rahman, Aya Fujii, Yuki Hiraga, Mizue Kawai, Kanami Sugimoto-Kawabata, Mattheus (Thijs) van Iersel, Jan Jaap van Lier, Stephen Djedjos, B. T. Slingsby, David M. Rodman","doi":"10.1111/cts.70000","DOIUrl":"https://doi.org/10.1111/cts.70000","url":null,"abstract":"<p>Dysregulation of the mineralocorticoid hormone aldosterone is an increasingly prevalent cause of hypertension. Aldosterone synthase (CYP11B2) shares 93% homology to 11β-hydroxylase (CYP11B1), which produces cortisol. Lorundrostat, a highly selective inhibitor of CYP11B2, is a potential safe and effective treatment for aldosterone-dependent, uncontrolled hypertension, including treatment-resistant hypertension. Lorundrostat showed highly selective inhibition of CYP11B2 in vitro, with 374-fold selectivity for CYP11B2 vs. CYP11B1. A first-in-human study of single ascending doses ranging from 5 to 800 mg and multiple ascending doses ranging from 40 to 360 mg once daily was conducted in healthy participants. After single- and multiple-dose administration, lorundrostat plasma levels peaked 1–3 h after administration with a <i>t</i><sub>1/2</sub> of 10–12 h. Plasma aldosterone decreased up to 40% with single 100-mg to 200-mg doses and up to 70% with single 400 to 800-mg doses. Plasma aldosterone returned to baseline within 16 h after single 100-mg doses and multiple once-daily 120-mg doses. Lorundrostat demonstrated a favorable safety profile in healthy participants. Dose- and exposure-dependent inhibition of renal tubular sodium reabsorption was observed across a clinically relevant dose range with no suppression of basal or cosyntropin-stimulated cortisol production and only a modest increase in mean serum potassium.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70000","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141994267","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Larisa H. Cavallari, Craig R. Lee, Francesco Franchi, Ellen C. Keeley, Joseph S. Rossi, Cameron D. Thomas, Yan Gong, Caitrin W. McDonough, Petr Starostik, Maryam J. Al Saeed, Latonya Been, Natasha Kulick, Jean Malave, Ian R. Mulrenin, Anh B. Nguyen, Joshua N. Terrell, Grace Tillotson, Amber L. Beitelshees, Almut G. Winterstein, George A. Stouffer, Dominick J. Angiolillo
Dual antiplatelet therapy (DAPT) with aspirin and a P2Y12 receptor inhibitor (clopidogrel, prasugrel, or ticagrelor) is indicated after percutaneous coronary intervention (PCI) to reduce the risk of atherothrombotic events. Approximately 30% of the US population has a CYP2C19 no-function allele that reduces the effectiveness of clopidogrel, but not prasugrel or ticagrelor, after PCI. We have shown improved outcomes with the integration of CYP2C19 genotyping into clinical care to guide the selection of prasugrel or ticagrelor in CYP2C19 no-function allele carriers. However, the influence of patient-specific demographic, clinical, and other genetic factors on outcomes with genotype-guided DAPT has not been defined. In addition, the impact of genotype-guided de-escalation from prasugrel or ticagrelor to clopidogrel in patients without a CYP2C19 no-function allele has not been investigated in a diverse, real-world clinical setting. The Precision Antiplatelet Therapy after Percutaneous Coronary Intervention (Precision PCI) Registry is a multicenter US registry of patients who underwent PCI and clinical CYP2C19 testing. The registry is enrolling a diverse population, assessing atherothrombotic and bleeding events over 12 months, collecting DNA samples, and conducting platelet function testing in a subset of patients. The registry aims to define the influence of African ancestry and other patient-specific factors on clinical outcomes with CYP2C19-guided DAPT, evaluate the safety and effectiveness of CYP2C19-guided DAPT de-escalation following PCI in a real-world setting, and identify additional genetic influences of clopidogrel response after PCI, with the ultimate goal of establishing optimal strategies for individualized antiplatelet therapy that improves outcomes in a diverse, real-world population.
{"title":"Precision Antiplatelet Therapy after Percutaneous Coronary Intervention (Precision PCI) Registry – Informing optimal antiplatelet strategies","authors":"Larisa H. Cavallari, Craig R. Lee, Francesco Franchi, Ellen C. Keeley, Joseph S. Rossi, Cameron D. Thomas, Yan Gong, Caitrin W. McDonough, Petr Starostik, Maryam J. Al Saeed, Latonya Been, Natasha Kulick, Jean Malave, Ian R. Mulrenin, Anh B. Nguyen, Joshua N. Terrell, Grace Tillotson, Amber L. Beitelshees, Almut G. Winterstein, George A. Stouffer, Dominick J. Angiolillo","doi":"10.1111/cts.70004","DOIUrl":"10.1111/cts.70004","url":null,"abstract":"<p>Dual antiplatelet therapy (DAPT) with aspirin and a P2Y<sub>12</sub> receptor inhibitor (clopidogrel, prasugrel, or ticagrelor) is indicated after percutaneous coronary intervention (PCI) to reduce the risk of atherothrombotic events. Approximately 30% of the US population has a <i>CYP2C19</i> no-function allele that reduces the effectiveness of clopidogrel, but not prasugrel or ticagrelor, after PCI. We have shown improved outcomes with the integration of <i>CYP2C19</i> genotyping into clinical care to guide the selection of prasugrel or ticagrelor in <i>CYP2C19</i> no-function allele carriers. However, the influence of patient-specific demographic, clinical, and other genetic factors on outcomes with genotype-guided DAPT has not been defined. In addition, the impact of genotype-guided de-escalation from prasugrel or ticagrelor to clopidogrel in patients without a <i>CYP2C19</i> no-function allele has not been investigated in a diverse, real-world clinical setting. The Precision Antiplatelet Therapy after Percutaneous Coronary Intervention (Precision PCI) Registry is a multicenter US registry of patients who underwent PCI and clinical <i>CYP2C19</i> testing. The registry is enrolling a diverse population, assessing atherothrombotic and bleeding events over 12 months, collecting DNA samples, and conducting platelet function testing in a subset of patients. The registry aims to define the influence of African ancestry and other patient-specific factors on clinical outcomes with <i>CYP2C19</i>-guided DAPT, evaluate the safety and effectiveness of <i>CYP2C19-</i>guided DAPT de-escalation following PCI in a real-world setting, and identify additional genetic influences of clopidogrel response after PCI, with the ultimate goal of establishing optimal strategies for individualized antiplatelet therapy that improves outcomes in a diverse, real-world population.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70004","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141989330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nicolas Hohmann, Anna S. Friedrichs, Jürgen Burhenne, Antje Blank, Gerd Mikus, Walter E. Haefeli
The dose dependence of the effect of enzyme inducers and the effect of the combined administration of two inducers that exert their effect via the same induction pathway (pregnane X receptor) have not been well studied. Using oral midazolam microdoses (30 μg), we have investigated CYP3A4 induction by St. John's wort (SJW) in 11 healthy volunteers using low (300 mg/day containing 7.48 mg hyperforin), therapeutic (900 mg/day), and supratherapeutic doses of SJW (1800 mg/day) for 14 days. SJW was then co-administered with rifampin (600 mg/day) for a further 7 days to evaluate the effect of the combined administration of two inducers. In addition, intravenous midazolam microdoses (10 μg) were administered before SJW, at SJW 1800 mg/day, and during administration of the two inducers to assess the hepatic contribution to total induction (semi-simultaneous administration). Administration of SJW increased oral midazolam clearance 1.96-fold (300 mg/day), 3.86-fold (900 mg/day), and 5.62-fold (1800 mg/day), and 17.5-fold after the addition of rifampin. Concurrently, the clearance of intravenous midazolam increased 2.05-fold (1800 mg/day) and 2.93-fold (SJW + rifampin). These results show that rifampin significantly enhances the induction of the highest SJW doses both hepatically and overall and suggest that these metabolic effects occur predominantly in the gut. These findings also suggest that in drug interactions involving strong and moderate enzyme inducers, the perpetrator effects of the strong inducer are decisive for the interaction.
{"title":"Dose-dependent induction of CYP3A activity by St. John's wort alone and in combination with rifampin","authors":"Nicolas Hohmann, Anna S. Friedrichs, Jürgen Burhenne, Antje Blank, Gerd Mikus, Walter E. Haefeli","doi":"10.1111/cts.70007","DOIUrl":"https://doi.org/10.1111/cts.70007","url":null,"abstract":"<p>The dose dependence of the effect of enzyme inducers and the effect of the combined administration of two inducers that exert their effect via the same induction pathway (pregnane X receptor) have not been well studied. Using oral midazolam microdoses (30 μg), we have investigated CYP3A4 induction by St. John's wort (SJW) in 11 healthy volunteers using low (300 mg/day containing 7.48 mg hyperforin), therapeutic (900 mg/day), and supratherapeutic doses of SJW (1800 mg/day) for 14 days. SJW was then co-administered with rifampin (600 mg/day) for a further 7 days to evaluate the effect of the combined administration of two inducers. In addition, intravenous midazolam microdoses (10 μg) were administered before SJW, at SJW 1800 mg/day, and during administration of the two inducers to assess the hepatic contribution to total induction (semi-simultaneous administration). Administration of SJW increased oral midazolam clearance 1.96-fold (300 mg/day), 3.86-fold (900 mg/day), and 5.62-fold (1800 mg/day), and 17.5-fold after the addition of rifampin. Concurrently, the clearance of intravenous midazolam increased 2.05-fold (1800 mg/day) and 2.93-fold (SJW + rifampin). These results show that rifampin significantly enhances the induction of the highest SJW doses both hepatically and overall and suggest that these metabolic effects occur predominantly in the gut. These findings also suggest that in drug interactions involving strong and moderate enzyme inducers, the perpetrator effects of the strong inducer are decisive for the interaction.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70007","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141994235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>We read with great interest the article recently published by Koroknai et al.<span><sup>1</sup></span> entitled “Expression pattern of osteopontin isoforms in malignant melanoma cell lines,” in which the expression of five OPN splice variants (OPN-SV) has been described in melanoma cell lines, providing new insights into this previously underexplored issue regarding osteopontin in melanoma research.</p><p>The authors found that the five OPN-SV presented higher expression levels in melanoma metastasis (MM) compared with melanoma primary (MP)-derived cell lines, although they did not reach statistical significance. They also observed that MM cell lines significantly express higher levels of all five OPN-SV in relation to those derived from primary superficial spreading melanoma (SSM), and between SSM and nodular melanoma (NM)-derived cell lines. These findings corroborate previous ones from the same group, in which <i>OPNa</i>, <i>OPNb</i>, and <i>OPNc</i> are expressed at higher levels in metastatic tumor tissues compared with primary lesions.<span><sup>2</sup></span> However, the recently described <i>OPN4</i> and <i>OPN5</i> variants presented low expression levels in both metastatic and primary tumor tissue samples, as opposed to data found in melanoma cell lines. The authors emphasized the association of <i>OPNc</i> at higher levels with invasive behavior. Consistent with these data, Jambor et al.<span><sup>2</sup></span> reported a significant positive correlation between <i>OPNc</i> expression and the presence of metastasis.</p><p>Building upon these findings, we further explored these data by analyzing skin cutaneous melanoma (SKCM) samples using the TSVdb database (http://www.tsvdb.com/). We found that metastatic SKCM samples significantly express higher levels of these five OPN-SV compared with primary melanoma tumors (Figure 1). Our data in tumor samples corroborate data regarding melanoma-derived cell lines for the five OPN-SV reported by Koroknai et al., in contrast to previous data showing that <i>OPN4</i> and <i>OPN5</i> isoforms are downregulated in melanoma subtypes.<span><sup>2</sup></span></p><p>Altogether, these data provide evidence that not only <i>OPNa</i>, <i>OPNb</i>, and <i>OPNc</i> are overexpressed in metastatic melanoma tissues in comparison with primary tumors, but also <i>OPN4</i> and <i>OPN5</i>, further evidencing that all five OPN-SV could be associated with an unfavorable prognosis in melanoma. Further research should elucidate how each OPN-SV could contribute to a worse prognosis in melanoma cancer, as reported for total osteopontin.<span><sup>3</sup></span> The involvement of each OPN-SV in melanoma progression is key to a better understanding of this disease and for the development of new therapeutic approaches, considering their specific expression patterns and roles in each step of tumor progression.<span><sup>4</sup></span></p><p>Funding for the works described was provided by the FAPERJ, CNPq, CAPES, and
{"title":"Expression and prognostic potential of osteopontin splice variants in malignant melanoma","authors":"Gabriela Ribeiro Silva, Luciana Bueno Ferreira, Etel Rodrigues Pereira Gimba","doi":"10.1111/cts.70002","DOIUrl":"https://doi.org/10.1111/cts.70002","url":null,"abstract":"<p>We read with great interest the article recently published by Koroknai et al.<span><sup>1</sup></span> entitled “Expression pattern of osteopontin isoforms in malignant melanoma cell lines,” in which the expression of five OPN splice variants (OPN-SV) has been described in melanoma cell lines, providing new insights into this previously underexplored issue regarding osteopontin in melanoma research.</p><p>The authors found that the five OPN-SV presented higher expression levels in melanoma metastasis (MM) compared with melanoma primary (MP)-derived cell lines, although they did not reach statistical significance. They also observed that MM cell lines significantly express higher levels of all five OPN-SV in relation to those derived from primary superficial spreading melanoma (SSM), and between SSM and nodular melanoma (NM)-derived cell lines. These findings corroborate previous ones from the same group, in which <i>OPNa</i>, <i>OPNb</i>, and <i>OPNc</i> are expressed at higher levels in metastatic tumor tissues compared with primary lesions.<span><sup>2</sup></span> However, the recently described <i>OPN4</i> and <i>OPN5</i> variants presented low expression levels in both metastatic and primary tumor tissue samples, as opposed to data found in melanoma cell lines. The authors emphasized the association of <i>OPNc</i> at higher levels with invasive behavior. Consistent with these data, Jambor et al.<span><sup>2</sup></span> reported a significant positive correlation between <i>OPNc</i> expression and the presence of metastasis.</p><p>Building upon these findings, we further explored these data by analyzing skin cutaneous melanoma (SKCM) samples using the TSVdb database (http://www.tsvdb.com/). We found that metastatic SKCM samples significantly express higher levels of these five OPN-SV compared with primary melanoma tumors (Figure 1). Our data in tumor samples corroborate data regarding melanoma-derived cell lines for the five OPN-SV reported by Koroknai et al., in contrast to previous data showing that <i>OPN4</i> and <i>OPN5</i> isoforms are downregulated in melanoma subtypes.<span><sup>2</sup></span></p><p>Altogether, these data provide evidence that not only <i>OPNa</i>, <i>OPNb</i>, and <i>OPNc</i> are overexpressed in metastatic melanoma tissues in comparison with primary tumors, but also <i>OPN4</i> and <i>OPN5</i>, further evidencing that all five OPN-SV could be associated with an unfavorable prognosis in melanoma. Further research should elucidate how each OPN-SV could contribute to a worse prognosis in melanoma cancer, as reported for total osteopontin.<span><sup>3</sup></span> The involvement of each OPN-SV in melanoma progression is key to a better understanding of this disease and for the development of new therapeutic approaches, considering their specific expression patterns and roles in each step of tumor progression.<span><sup>4</sup></span></p><p>Funding for the works described was provided by the FAPERJ, CNPq, CAPES, and","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141994234","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jean De Dieu Ndayishimiye, Mari Cayabyab, Glenda Hoffecker, Victoria Wittner, Penn Medicine Biobank, Sony Tuteja
We read with great interest the findings published in your journal by Muldoon et al. of real-world implementation of DPYD and UGT1A1 pharmacogenetic testing in a community-based cancer center1 and would like to share some comments regarding the need for specifically reporting outcomes by tumor type.
Most existing studies focus on preemptive DPYD genotyping for fluoropyrimidines in gastrointestinal cancers, rarely are outcomes reported for breast cancer. Previous studies evaluating the impact of DPYD on outcomes enrolled a small proportion of patients with breast cancer, including Muldoon, at 6%,1 and another recent study at 12%.2 Because of the small number of patients, the outcomes are not typically reported separately by tumor type, it is challenging to determine the true incidence of treatment-related toxicities experienced by DPYD variant carriers in this patient population.
Preemptive DPYD testing for patients treated with fluoropyrimidine is still controversial among US oncologists, and the hesitation is even greater in breast cancer clinicians due to limited data and a lack of endorsements from the NCCN and ASCO.3 To fill this gap in knowledge, we evaluated capecitabine-related toxicities in 62 patients with breast cancer that were enrolled in an institutional biobank.4 Serious treatment-related adverse events (TRAEs) were defined as chemotherapy-related events necessitating treatment in the hospital, emergency department, or oncology evaluation center (i.e., oncology urgent care).
One of the three variant carriers experienced severe diarrhea. This TRAE began during Week 1 of Cycle 1 of capecitabine initiation and resulted in 14 days of hospitalization. The patient was dose reduced from 2000 mg twice daily to 1000 mg in the morning and 1500 mg in the evening for cycle two. However, despite the dose reduction and loperamide administration, the patient still could not tolerate the treatment. The patient skipped several doses due to worsening diarrhea, which resulted in capecitabine being later discontinued.
Patients with breast cancer treated with capecitabine are classically monitored for reversible Hand-Foot Syndrome.5 However, other side effects such as diarrhea and mucositis still occur and can result in hospitalization or treatment discontinuation as demonstrated in this case study. It is critical to focus on bridging the current knowledge and practice gap around the use of DPYD genotyping and preemptive dose reduction in patients on capecitabine for breast cancer, thereby optimizing efficacy and minimizing harm in this population.
No funding was received for this work.
The authors declared no competing interests for this work.
{"title":"A call for reporting of tumor-specific outcomes in studies of DPYD genotyping","authors":"Jean De Dieu Ndayishimiye, Mari Cayabyab, Glenda Hoffecker, Victoria Wittner, Penn Medicine Biobank, Sony Tuteja","doi":"10.1111/cts.70003","DOIUrl":"10.1111/cts.70003","url":null,"abstract":"<p>We read with great interest the findings published in your journal by Muldoon et al. of real-world implementation of DPYD and UGT1A1 pharmacogenetic testing in a community-based cancer center<span><sup>1</sup></span> and would like to share some comments regarding the need for specifically reporting outcomes by tumor type.</p><p>Most existing studies focus on preemptive DPYD genotyping for fluoropyrimidines in gastrointestinal cancers, rarely are outcomes reported for breast cancer. Previous studies evaluating the impact of DPYD on outcomes enrolled a small proportion of patients with breast cancer, including Muldoon, at 6%,<span><sup>1</sup></span> and another recent study at 12%.<span><sup>2</sup></span> Because of the small number of patients, the outcomes are not typically reported separately by tumor type, it is challenging to determine the true incidence of treatment-related toxicities experienced by DPYD variant carriers in this patient population.</p><p>Preemptive DPYD testing for patients treated with fluoropyrimidine is still controversial among US oncologists, and the hesitation is even greater in breast cancer clinicians due to limited data and a lack of endorsements from the NCCN and ASCO.<span><sup>3</sup></span> To fill this gap in knowledge, we evaluated capecitabine-related toxicities in 62 patients with breast cancer that were enrolled in an institutional biobank.<span><sup>4</sup></span> Serious treatment-related adverse events (TRAEs) were defined as chemotherapy-related events necessitating treatment in the hospital, emergency department, or oncology evaluation center (i.e., oncology urgent care).</p><p>One of the three variant carriers experienced severe diarrhea. This TRAE began during Week 1 of Cycle 1 of capecitabine initiation and resulted in 14 days of hospitalization. The patient was dose reduced from 2000 mg twice daily to 1000 mg in the morning and 1500 mg in the evening for cycle two. However, despite the dose reduction and loperamide administration, the patient still could not tolerate the treatment. The patient skipped several doses due to worsening diarrhea, which resulted in capecitabine being later discontinued.</p><p>Patients with breast cancer treated with capecitabine are classically monitored for reversible Hand-Foot Syndrome.<span><sup>5</sup></span> However, other side effects such as diarrhea and mucositis still occur and can result in hospitalization or treatment discontinuation as demonstrated in this case study. It is critical to focus on bridging the current knowledge and practice gap around the use of DPYD genotyping and preemptive dose reduction in patients on capecitabine for breast cancer, thereby optimizing efficacy and minimizing harm in this population.</p><p>No funding was received for this work.</p><p>The authors declared no competing interests for this work.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.70003","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141989329","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ferroptosis is a novel, iron-dependent regulatory cell death mainly caused by an imbalance between the production and degradation of intracellular reactive oxygen species (ROS). Recently, ferroptosis induction has been considered a potential therapeutic approach for hepatocellular carcinoma (HCC). Fibroblast growth factor 21 (FGF21) is a new modulator of ferroptosis; however, the regulatory role of FGF21 in HCC ferroptosis has not been investigated. In this study, we explored the role of FGF21 and its underlying molecular mechanism in the ferroptotic death of HCC cells. We identified Major vault protein (MVP) as a target of FGF21 and revealed that knockdown of MVP inhibited the lipid peroxidation levels of HCC cells by decreasing NADPH oxidase 4 (NOX4, a major source of ROS) transcription, thereby attenuating the effect of FGF21-mediated ferroptosis. On the other hand, MVP overexpression showed the opposite results. Mechanistically, MVP binds to IRF1 and thus interferes with the interaction between IRF1 and the YAP1 promoter, leading to an increase in NOX4 transcription. Importantly, forced expression of IRF1 or downregulation of YAP1 partially reversed the effect of MVP overexpression on HCC ferroptosis. Furthermore, the results in xenograft tumor models suggested that overexpression of MVP can efficiently increase the level of lipid peroxidation in vivo. Taken together, these results provide new insights into the regulatory mechanism of ferroptosis in HCC.
{"title":"MVP enhances FGF21-induced ferroptosis in hepatocellular carcinoma by increasing lipid peroxidation through regulation of NOX4","authors":"Jinkun Xia, Boqi Fu, Zhe Wang, Gaolin Wen, Quanshui Gu, Dayu Chen, Haozhen Ren","doi":"10.1111/cts.13910","DOIUrl":"10.1111/cts.13910","url":null,"abstract":"<p>Ferroptosis is a novel, iron-dependent regulatory cell death mainly caused by an imbalance between the production and degradation of intracellular reactive oxygen species (ROS). Recently, ferroptosis induction has been considered a potential therapeutic approach for hepatocellular carcinoma (HCC). Fibroblast growth factor 21 (FGF21) is a new modulator of ferroptosis; however, the regulatory role of FGF21 in HCC ferroptosis has not been investigated. In this study, we explored the role of FGF21 and its underlying molecular mechanism in the ferroptotic death of HCC cells. We identified Major vault protein (MVP) as a target of FGF21 and revealed that knockdown of MVP inhibited the lipid peroxidation levels of HCC cells by decreasing NADPH oxidase 4 (NOX4, a major source of ROS) transcription, thereby attenuating the effect of FGF21-mediated ferroptosis. On the other hand, MVP overexpression showed the opposite results. Mechanistically, MVP binds to IRF1 and thus interferes with the interaction between IRF1 and the YAP1 promoter, leading to an increase in NOX4 transcription. Importantly, forced expression of IRF1 or downregulation of YAP1 partially reversed the effect of MVP overexpression on HCC ferroptosis. Furthermore, the results in xenograft tumor models suggested that overexpression of MVP can efficiently increase the level of lipid peroxidation in vivo. Taken together, these results provide new insights into the regulatory mechanism of ferroptosis in HCC.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cts.13910","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141983810","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Andreas Halman, Sebastian Lunke, Simon Sadedin, Claire Moore, Rachel Conyers
Pharmacogenomics (PGx) investigates the influence of genetics on drug responses, enabling tailored treatments for personalized healthcare. This study assessed the accuracy of genotyping six genes using whole genome sequencing with four different computational tools and various sequencing depths. The effects of using different reference genomes (GRCh38 and GRCh37) and sequence aligners (BWA-MEM and Bowtie2) were also explored. The results showed generally minor variations in tool performance across most genes; however, more notable discrepancies were observed in the analysis of the complex CYP2D6 gene. Cyrius, a CYP2D6-specific tool, demonstrated the most robust performance, achieving the highest concordance rates for CYP2D6 in all instances, comparable to the consensus approach in most cases. There were rather small differences between the samples with 20× coverage depth and those with higher depth, but the decreased performance was more evident at lower depths, particularly at 5×. Additionally, variations in CYP2D6 results were observed when samples were aligned to different reference genomes using the same method, or to the same genome using different aligners, which led to reporting incorrect rare star alleles in several cases. These findings inform the selection of optimal PGx tools and methodologies as well as suggest that employing a consensus approach with two or more tools might be preferable for certain genes and tool combinations, especially at lower sequencing depths, to ensure accurate results. Additionally, we show how the upstream alignment can affect the performance of tools, an important factor to take into account.
{"title":"Benchmarking pharmacogenomics genotyping tools: Performance analysis on short-read sequencing samples and depth-dependent evaluation","authors":"Andreas Halman, Sebastian Lunke, Simon Sadedin, Claire Moore, Rachel Conyers","doi":"10.1111/cts.13911","DOIUrl":"10.1111/cts.13911","url":null,"abstract":"<p>Pharmacogenomics (PGx) investigates the influence of genetics on drug responses, enabling tailored treatments for personalized healthcare. This study assessed the accuracy of genotyping six genes using whole genome sequencing with four different computational tools and various sequencing depths. The effects of using different reference genomes (GRCh38 and GRCh37) and sequence aligners (BWA-MEM and Bowtie2) were also explored. The results showed generally minor variations in tool performance across most genes; however, more notable discrepancies were observed in the analysis of the complex <i>CYP2D6</i> gene. Cyrius, a <i>CYP2D6</i>-specific tool, demonstrated the most robust performance, achieving the highest concordance rates for <i>CYP2D6</i> in all instances, comparable to the consensus approach in most cases. There were rather small differences between the samples with 20× coverage depth and those with higher depth, but the decreased performance was more evident at lower depths, particularly at 5×. Additionally, variations in <i>CYP2D6</i> results were observed when samples were aligned to different reference genomes using the same method, or to the same genome using different aligners, which led to reporting incorrect rare star alleles in several cases. These findings inform the selection of optimal PGx tools and methodologies as well as suggest that employing a consensus approach with two or more tools might be preferable for certain genes and tool combinations, especially at lower sequencing depths, to ensure accurate results. Additionally, we show how the upstream alignment can affect the performance of tools, an important factor to take into account.</p>","PeriodicalId":50610,"journal":{"name":"Cts-Clinical and Translational Science","volume":"17 8","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11315677/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141914451","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}