Michael A. Tortorici
CSL Behring
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Michael A. Tortorici.
The Lancet Respiratory Medicine | 2017
Noel G. McElvaney; Jonathan Burdon; Mark Holmes; Allan R. Glanville; Peter Wark; Philip J. Thompson; Paul Hernandez; Jan Chlumsky; Helmut Teschler; Joachim H. Ficker; Niels Seersholm; Alan Altraja; Riitta Mäkitaro; Joanna Chorostowska-Wynimko; Marek Sanak; Paul I Stoicescu; Eeva Piitulainen; Oliver Vit; Marion Wencker; Michael A. Tortorici; Michael Fries; Jonathan Edelman; Kenneth R. Chapman
BACKGROUND Purified α1 proteinase inhibitor (A1PI) slowed emphysema progression in patients with severe α1 antitrypsin deficiency in a randomised controlled trial (RAPID-RCT), which was followed by an open-label extension trial (RAPID-OLE). The aim was to investigate the prolonged treatment effect of A1PI on the progression of emphysema as assessed by the loss of lung density in relation to RAPID-RCT. METHODS Patients who had received either A1PI treatment (Zemaira or Respreeza; early-start group) or placebo (delayed-start group) in the RAPID-RCT trial were included in this 2-year open-label extension trial (RAPID-OLE). Patients from 22 hospitals in 11 countries outside of the USA received 60 mg/kg per week A1PI. The primary endpoint was annual rate of adjusted 15th percentile lung density loss measured using CT in the intention-to-treat population with a mixed-effects regression model. This trial is registered with ClinicalTrials.gov, number NCT00670007. FINDINGS Between March 1, 2006, and Oct 13, 2010, 140 patients from RAPID-RCT entered RAPID-OLE: 76 from the early-start group and 64 from the delayed-start group. Between day 1 and month 24 (RAPID-RCT), the rate of lung density loss in RAPID-OLE patients was lower in the early-start group (-1·51 g/L per year [SE 0·25] at total lung capacity [TLC]; -1·55 g/L per year [0·24] at TLC plus functional residual capacity [FRC]; and -1·60 g/L per year [0·26] at FRC) than in the delayed-start group (-2·26 g/L per year [0·27] at TLC; -2·16 g/L per year [0·26] at TLC plus FRC, and -2·05 g/L per year [0·28] at FRC). Between months 24 and 48, the rate of lung density loss was reduced in delayed-start patients (from -2·26 g/L per year to -1·26 g/L per year), but no significant difference was seen in the rate in early-start patients during this time period (-1·51 g/L per year to -1·63 g/L per year), thus in early-start patients the efficacy was sustained to month 48. INTERPRETATION RAPID-OLE supports the continued efficacy of A1PI in slowing disease progression during 4 years of treatment. Lost lung density was never recovered, highlighting the importance of early intervention with A1PI treatment. FUNDING CSL Behring.
The Journal of Clinical Pharmacology | 2015
Michael A. Tortorici; David L. Cutler; Anasuya Hazra; Thomas D. Nolin; Karen Rowland-Yeo; Karthik Venkatakrishnan
Chronic kidney disease (CKD) has been shown to alter the pharmacokinetics of drugs that are eliminated not only via the renal pathway but also by nonrenal clearance and transport. Dosing recommendations in subjects with CKD have historically come from small pharmacokinetic (PK) studies, which have been insulated from the broader clinical development strategy. Opportunities for prospective strategic integration of both preclinical and clinical data on drug clearance mechanisms, model‐based approaches, and clinical knowledge of therapeutic index are therefore often missed in designing and analyzing the results of PK studies in subjects with CKD, and eventually providing dosing recommendations. These considerations are valuable in designing informative PK studies in subjects with CKD, as well as for guiding kidney function‐related inclusion/exclusion criteria in the broader clinical program and ultimately defining dosing guidelines that optimize benefit‐risk balance for these special patient populations based on all available data. This paper offers points to consider for drug developers to increase adoption of a contemporary multidisciplinary approach, which includes key considerations on study design and conduct, methodologies for analysis (population PK and physiologically based PK modeling), and a roadmap to interpret the effect of kidney function on the overall benefit‐risk profile of drugs in development.
Expert Review of Clinical Pharmacology | 2014
Michael A. Tortorici; Thomas D. Nolin
A key regulatory requirement pertaining to drug development is characterization of the role of kidney function in drug disposition and response, along with provision of corresponding renal dose adjustment recommendations. Traditionally, this information has been derived from Phase I pharmacokinetic studies in which regulatory guidance exists for pharmaceutical manufacturers on the design, conduct, analysis, and interpretation of data. Categorization and stratification of subjects into kidney function groups and dosing recommendations have historically been based on creatinine clearance estimates using the Cockcroft–Gault equation. As new estimating equations have emerged, the choice of equation for assessment of kidney function has become an area of debate. This review highlights these equations and provides recent examples of the use of quantitative models, incorporating efficacy and safety to make rational dose recommendations in subjects with impaired kidney function.
The Journal of Clinical Pharmacology | 2016
James F. Burris; Michael A. Tortorici; Maja Mandic; Michael Neely; Michael D. Reed
Differences in physiology related to young or old age and/or organ system impairment alter the absorption, distribution, metabolism, and excretion of many medications and consequently their effectiveness and toxicity. This module discusses common alterations in medication use and dosage that are required in the pediatric age group, in the elderly, and in patients with renal or hepatic disease.
CPT: Pharmacometrics & Systems Pharmacology | 2018
Ying Zhang; Michael A. Tortorici; Dipti Pawaskar; Ingo Pragst; Thomas Machnig; Matthew Hutmacher; Bruce L. Zuraw; Marco Cicardi; Timothy J. Craig; Hilary Longhurst; Jagdev Sidhu
Subcutaneous C1‐inhibitor (HAEGARDA, CSL Behring), is a US Food and Drug Administration (FDA)‐approved, highly concentrated formulation of a plasma‐derived C1‐esterase inhibitor (C1‐INH), which, in the phase III Clinical Studies for Optimal Management in Preventing Angioedema with Low‐Volume Subcutaneous C1‐inhibitor Replacement Therapy (COMPACT) trial, reduced the incidence of hereditary angioedema (HAE) attacks when given prophylactically. Data from the COMPACT trial were used to develop a repeated time‐to‐event model to characterize the timing and frequency of HAE attacks as a function of C1‐INH activity, and then develop an exposure–response model to assess the relationship between C1‐INH functional activity levels (C1‐INH(f)) and the risk of an attack. The C1‐INH(f) values of 33.1%, 40.3%, and 63.1% were predicted to correspond with 50%, 70%, and 90% reductions in the HAE attack risk, respectively, relative to no therapy. Based on trough C1‐INH(f) values for the 40 IU/kg (40.2%) and 60 IU/kg (48.0%) C1‐INH (SC) doses, the model predicted that 50% and 67% of the population, respectively, would see at least a 70% decrease in the risk of an attack.
Arteriosclerosis, Thrombosis, and Vascular Biology | 2018
Andreas Gille; Denise D’Andrea; Michael A. Tortorici; Gunter Hartel; Samuel D. Wright
Objective— CSL112 (apolipoprotein A-I [apoA-I; human]) is a novel formulation of apoA-I in development for reduction of early recurrent cardiovascular events after acute myocardial infarction. Cholesterol efflux capacity (CEC) is a marker of high-density lipoprotein (HDL) function that is strongly correlated with incident cardiovascular disease. Impaired CEC has been observed in patients with coronary heart disease. Here, we determined whether infused apoA-I improves CEC when administered to patients with stable atherosclerotic disease versus healthy volunteers. Approach and Results— Measurements of apoA-I, HDL unesterified cholesterol, HDL esterified cholesterol, pre–&bgr;1-HDL, and CEC were determined in samples from patients with stable atherosclerotic disease before and after intravenous administration of CSL112. These measures were compared with 2 prior studies in healthy volunteers for differences in CEC at baseline and after CSL112 infusion. Patients with stable atherosclerotic disease exhibited significantly lower ATP-binding cassette transporter 1–mediated CEC at baseline (P<0.0001) despite slightly higher apoA-I levels when compared with healthy individuals (2 phase 1 studies pooled; P⩽0.05), suggesting impaired HDL function. However, no differences were observed in apoA-I pharmacokinetics or in pre–&bgr;1-HDL (P=0.5) or CEC (P=0.1) after infusion of CSL112. Similar elevation in CEC was observed in patients with low or high baseline HDL function (based on tertiles of apoA-I–normalized CEC; P=0.1242). These observations were extended and confirmed using cholesterol esterification as an additional measure. Conclusions— CSL112 shows comparable, strong, and immediate effects on CEC despite underlying cardiovascular disease. CSL112 is, therefore, a promising novel therapy for lowering the burden of atherosclerosis and reducing the risk of recurrent cardiovascular events.
Clinical pharmacology in drug development | 2018
Michael A. Tortorici; Danielle Duffy; Rebecca Evans; John Feaster; Andreas Gille; Timothy G. K. Mant; Samuel D. Wright; Denise D'Andrea
CSL112 (Apolipoprotein A‐I [human]) is an intravenous preparation of apolipoprotein A‐I (apoA‐I), formulated with phosphatidylcholine (PC) and stabilized with sucrose, in development to prevent early recurrent cardiovascular events following acute myocardial infarction (AMI). This phase 1 study was designed to determine if moderate renal impairment (RI) influenced the pharmacokinetics (PK) and safety of CSL112. Thirty‐two subjects, 16 with moderate RI (estimated glomerular filtration rate [eGFR] ≥ 30 and < 60 mL/min/1.73 m2) and 16 age‐, sex‐, and weight‐matched subjects with normal renal function (eGFR ≥ 90 mL/min/1.73 m2) were randomized 3:1 to receive a single infusion of CSL112 2 g (n = 6) or placebo (n = 2), or CSL112 6 g (n = 6) or placebo (n = 2). PK sampling was at prespecified times from 48 hours prior to 144 hours following infusions, with final safety assessments at 90 days. Renal and hepatic safety, and adverse events (AEs) were monitored throughout the study. Plasma apoA‐I and PC PK profiles were similar between renal function cohorts at both doses. For CSL112 6 g mean ± SD apoA‐I AUC0‐last was 7670 ± 1900 and 9170 ± 2910 mg·h/dL in normal renal function and moderate RI subjects, respectively. Renal apoA‐I clearance was <1% of CSL112 dose. In moderate RI, sucrose clearance was slower; however, approximately 70% was excreted within 48 hours in both renal function cohorts. No CSL112‐related serious AEs or clinically significant renal or hepatic safety changes were observed. Dose adjustment of CSL112 is not required in subjects with moderate RI, supporting its further investigation in AMI patients with moderate RI.
Clinical & Experimental Allergy | 2018
Dipti Pawaskar; Michael A. Tortorici; Bruce L. Zuraw; Timothy J. Craig; Marco Cicardi; Hilary Longhurst; H. Henry Li; William R. Lumry; Inmaculada Martinez-Saguer; Joshua J. Jacobs; Jonathan A. Bernstein; Marc A. Riedl; Constance H. Katelaris; Paul K. Keith; Annette Feussner; Jagdev Sidhu
Long‐term prophylaxis with subcutaneous (SC) administration of a highly concentrated plasma‐derived C1‐esterase inhibitor (C1‐INH) formulation was recently approved by the Food and Drug Administration for hereditary angioedema (HAE) attack prevention.
Journal of the American College of Cardiology | 2017
Andreas Gille; Danielle Duffy; Michael A. Tortorici; Samuel D. Wright; Larry Deckelbaum; Denise D'Andrea
Background: CSL112 is a novel formulation of plasma-derived apolipoprotein A-I (apoA-I) that is in development for cardiovascular (CV) event reduction following acute coronary syndrome (ACS) by enhancement of cholesterol efflux capacity (CEC). Completed clinical trials have demonstrated favourable
British Journal of Clinical Pharmacology | 2017
Michael A. Tortorici; James A. Rogers; Oliver Vit; Martin Bexon; Robert A. Sandhaus; Jonathan Burdon; Joanna Chorostowska-Wynimko; Philip J. Thompson; James M. Stocks; Noel G. McElvaney; Kenneth R. Chapman; Jonathan Edelman
Aims Early‐onset emphysema attributed to α‐1 antitrypsin deficiency (AATD) is frequently overlooked and undertreated. RAPID‐RCT/RAPID‐OLE, the largest clinical trials of purified human α‐1 proteinase inhibitor (A1‐PI; 60 mg kg–1 week–1) therapy completed to date, demonstrated for the first time that A1‐PI is clinically effective in slowing lung tissue loss in AATD. A posthoc pharmacometric analysis was undertaken to further explore dose, exposure and response. Methods A disease progression model was constructed, utilizing observed A1‐PI exposure and lung density decline rates (measured by computed tomography) from RAPID‐RCT/RAPID‐OLE, to predict effects of population variability and higher doses on A1‐PI exposure and clinical response. Dose–exposure and exposure–response relationships were characterized using nonlinear and linear mixed effects models, respectively. The dose–exposure model predicts summary exposures and not individual concentration kinetics; covariates included baseline serum A1‐PI, forced expiratory volume in 1 s and body weight. The exposure–response model relates A1‐PI exposure to lung density decline rate at varying exposure levels. Results A dose of 60 mg kg–1 week–1 achieved trough serum levels >11 μmol l–1 (putative ‘protective threshold’) in ≥98% patients. Dose–exposure–response simulations revealed increasing separation between A1‐PI and placebo in the proportions of patients achieving higher reductions in lung density decline rate; improvements in decline rates ≥0.5 g l–1 year–1 occurred more often in patients receiving A1‐PI: 63 vs. 12%. Conclusion Weight‐based A1‐PI dosing reliably raises serum levels above the 11 μmol l–1 threshold. However, our exposure–response simulations question whether this is the maximal, clinically effective threshold for A1‐PI therapy in AATD. The model suggested higher doses of A1‐PI would yield greater clinical effects.