Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alaa Ahmad is active.

Publication


Featured researches published by Alaa Ahmad.


The Lancet | 2017

Anti-MAdCAM antibody (PF-00547659) for ulcerative colitis (TURANDOT): a phase 2, randomised, double-blind, placebo-controlled trial

Severine Vermeire; William J. Sandborn; S. Danese; Xavier Hébuterne; Bruce Salzberg; Maria Kłopocka; Dino Tarabar; Tomas Vanasek; Miloš Greguš; Paul Hellstern; Joo Sung Kim; Miles Sparrow; Kenneth J. Gorelick; Michelle Hinz; Alaa Ahmad; Vivek Pradhan; Mina Hassan-Zahraee; Robert Clare; Fabio Cataldi; W. Reinisch

BACKGROUND PF-00547659 is a fully human monoclonal antibody that binds to human mucosal addressin cell adhesion molecule-1 (MAdCAM-1) to selectively reduce lymphocyte homing to the intestinal tract. We aimed to assess the efficacy and safety of PF-00547659 in patients with moderate to severe ulcerative colitis. METHODS This phase 2, randomised, double-blind, placebo-controlled clinical trial recruited patients aged 18-65 years from 105 centres in 21 countries, with a history (≥3 months) of active ulcerative colitis extending more than 15 cm beyond the anal verge (with a total Mayo score ≥6 and a Mayo endoscopic subscore ≥2) who had failed or were intolerant to at least one conventional therapy. Patients were stratified by previous anti-TNFα treatment, and randomly assigned by a computer-generated randomisation schedule to receive a subcutaneous injection of 7·5 mg, 22·5 mg, 75 mg, or 225 mg PF-00547659 or placebo at baseline, then every 4 weeks. Patients, investigators, and sponsors were blinded to the treatment. The primary endpoint was the proportion of patients achieving remission (total Mayo score ≤2 with no individual subscore >1 and rectal bleeding subscore ≤1) at week 12. The efficacy analysis included all patients who received at least one dose of the randomised treatment; the safety analysis was done according to treatment received. All p values were one-sided and multiplicity-adjusted. This study is registered with ClinicalTrials.gov, number NCT01620255. FINDINGS Between Nov 2, 2012, and Feb 4, 2016, we screened 587 patients; 357 were eligible and randomly assigned to receive placebo (n=73) or PF-00547659 at doses of 7·5 mg (n=71), 22·5 mg (n=72), 75 mg (n=71), or 225 mg (n=70). Remission rates at week 12 were significantly greater in three of four active-treatment groups than in the placebo group (2·7% [two of 73]): 7·5 mg (11·3% [eight of 71]), 22·5 mg (16·7% [12 of 72]), 75 mg (15·5% [11 of 71]), and 225 mg (5·7% [four of 70]). These rates corresponded to a stratum-adjusted (anti-TNFα-naive and anti-TNFα-experienced) risk difference versus placebo of 8·0% for 7·5 mg (90% CI 1·9 to 14, p=0·0425), 12·8% for 22·5 mg (5·6 to 19·9, p=0·0099), 11·8% for 75 mg (4·8 to 18·8, p=0·0119), and 2·6% for 225 mg (-1·2 to 6·4, p=0·1803). Four of 73 (5·5%) patients had a serious adverse event in the placebo group, ten of 71 (14·1%) in the 7·5 mg group, one of 70 (1·4%) in the 22·5 mg group, three of 73 (4·1%) in the 75 mg group, and three of 70 (4·3%) in the 225 mg group. No safety signal was observed for the study drug. INTERPRETATION PF-00547659 was safe and well tolerated in this patient population, and better than placebo for induction of remission in patients with moderate to severe ulcerative colitis. The greatest clinical effects were observed with the 22·5 mg and 75 mg doses. FUNDING Pfizer.


Aaps Journal | 2015

Quantitative Prediction of Human Pharmacokinetics for mAbs Exhibiting Target-Mediated Disposition

Aman P. Singh; Wojciech Krzyzanski; Steven W. Martin; Gregory L. Weber; Alison Betts; Alaa Ahmad; Anson K. Abraham; Anup Zutshi; John Lin; Pratap Singh

Prediction of human pharmacokinetics (PK) can be challenging for monoclonal antibodies (mAbs) exhibiting target-mediated drug disposition (TMDD). In this study, we performed a quantitative analysis of a diverse set of six mAbs exhibiting TMDD to explore translational rules that can be utilized to predict human PK. A TMDD model with rapid-binding approximation was utilized to fit PK and PD (i.e., free and/or total target levels) data, and average absolute fold error (AAFE) was calculated for each model parameter. Based on the comparative analysis, translational rules were developed and applied to a test antibody not included in the original analysis. AAFE of less than two-fold was observed between monkey and human for baseline target levels (R0), body-weight (BW) normalized central elimination rate (Kel/BW−0.25) and central volume (Vc/BW1.0). AAFE of less than three-fold was estimated for the binding affinity constant (KD). The other four parameters, i.e., complex turnover rate (Kint), target turnover rate (Kdeg), central to peripheral distribution rate constant (Kpt) and peripheral to central rate constant (Ktp) were poorly correlated between monkey and human. The projected human PK of test antibody based on the translation rules was in good agreement with the observed nonlinear PK. In conclusion, we recommend a TMDD model-based prediction approach that integrates in vitro human biomeasures and in vivo preclinical data using translation rules developed in this study.


Gut | 2018

Phase II evaluation of anti-MAdCAM antibody PF-00547659 in the treatment of Crohn's disease: report of the OPERA study.

William J. Sandborn; Scott D. Lee; Dino Tarabar; Edouard Louis; Maria Kłopocka; Jochen Klaus; Walter Reinisch; Xavier Hébuterne; Dong Il Park; Stefan Schreiber; Satyaprakash Nayak; Alaa Ahmad; Anindita Banerjee; Lisa S. Brown; Fabio Cataldi; Kenneth J. Gorelick; John B. Cheng; Mina Hassan-Zahraee; Robert Clare; Geert R. D'Haens

Objective This phase II, randomised, double-blind, placebo-controlled clinical trial was designed to evaluate the efficacy and safety of PF-00547659, a fully human monoclonal antibody that binds to human mucosal addressin cell adhesion molecule (MAdCAM) to selectively reduce lymphocyte homing to the intestinal tract, in patients with moderate-to-severe Crohn’s disease (CD). Design Eligible adults were aged 18–75 years, with active moderate-to-severe CD (Crohn’s Disease Activity Index (CDAI) 220–450), a history of failure or intolerance to antitumour necrosis factor and/or immunosuppressive agents, high-sensitivity C reactive protein >3.0 mg/L and ulcers on colonoscopy. Patients were randomised to PF-00547659 22.5 mg, 75 mg or 225 mg or placebo. The primary endpoint was CDAI 70-point decrease from baseline (CDAI-70) at week 8 or 12. Results In all, 265 patients were eligible for study entry. Although CDAI-70 response was not significantly different with placebo versus PF-00547659 treatment at weeks 8 or 12, remission rate was greater in patients with higher baseline C reactive protein (>5 mg/L vs >18.8 mg/L, respectively). Soluble MAdCAM decreased significantly from baseline to week 2 in a dose-related manner and remained low during the study in PF-00547659-treated patients. Circulating β7+ CD4+ central memory T-lymphocytes increased at weeks 8 and 12 with PF-00547659 treatment. No safety signal was seen. Conclusions Clinical endpoint differences between PF-00547659 and placebo did not reach statistical significance in patients with moderate-to-severe CD. PF-00547659 was pharmacologically active, as shown by a sustained dose-related decrease in soluble MAdCAM and a dose-related increase in circulating β7+ central memory T cells. Trial registration number NCT01276509; Results.


The Journal of Clinical Pharmacology | 2015

Derivation of Phase 3 dosing for peginterferon lambda‐1a in chronic hepatitis C, Part 2: Exposure–response analyses for efficacy and safety variables

Matthew Hruska; Xiaodong Wang; Phyllis Chan; Alaa Ahmad; Jeremy Freeman; Maria Arantxa Horga; Jan Hillson; Vikram Kansra; Juan‐Carlos Lopez‐Talavera

This is the second of two manuscripts detailing the pharmacodynamic derivation of peginterferon lambda‐1a (Lambda) dosing and treatment durations for Phase 3 studies in hepatitis C virus (HCV) infection, based on Phase 2 data. Herein, we describe the derivation of regression models for 12‐week on‐treatment virologic response and safety outcomes at 120, 180, and 240 μg Lambda with ribavirin. In patients with HCV genotypes 1 or 4, there was a significant (P = 0.024) relationship between undetectable HCV‐RNA at Week 4 and Lambda exposure (AUC or Cmax), with the largest difference between adjacent dose levels between the 180 and 120 μg exposure ranges. Risk of Grade 3–4 aminotransferase or bilirubin elevations relative to a peginterferon alfa‐2a/ribavirin control were related to Lambda exposure for all patients, and the largest increase between adjacent dose levels was seen for 240 versus 180 μg. Anemia and neutropenia events were lower than control across all doses and exposures. Based on these data and those in our previous manuscript, Phase 3 studies will evaluate fixed 180 µg doses of Lambda in combination with ribavirin and a direct‐acting antiviral for 24–48 weeks in HCV genotypes 1 or 4 or 12–24 weeks in genotypes 2 or 3.


The Journal of Clinical Pharmacology | 2015

Derivation of Phase 3 dosing for peginterferon lambda‐1a in chronic hepatitis C, Part 1: Modeling optimal treatment duration and sustained virologic response rates

Xiaodong Wang; Matthew Hruska; Phyllis Chan; Alaa Ahmad; Jeremy Freeman; Maria Arantxa Horga; Jan Hillson; Vikram Kansra; Juan‐Carlos Lopez‐Talavera

Peginterferon lambda‐1a (Lambda) is under clinical development for the treatment of chronic hepatitis B and C virus (HBV, HCV, respectively) infection. This is the first of two manuscripts detailing the pharmacodynamic derivation of Lambda dosing and treatment durations for Phase 3 studies in HCV, based on Phase 2 data. We describe here the derivation of a population model of Lambda exposure; the adaptation of a previously published viral dynamic model for Lambda treatment and host genotype, and its use to simulate sustained virologic responses (SVR). Lambda population pharmacokinetics was described by a one‐compartment model with first‐order absorption, and 33.0 L per day clearance with 47% interindividual (36% intra‐individual) variability. Weight explained a negligible proportion of the variability. Based on SVR predictions, optimum treatment durations were 48 weeks for HCV genotypes 1 or 4 (SVR estimates for 120, 180, and 240 μg Lambda: 58%, 54%, 47%, respectively) and 24 weeks for genotypes 2 or 3 (75%, 72%, 67%). SVR predictions for 240 μg were lower due to dropout predictions. The SVR model established the optimum treatment duration for Phase 3 studies but did not differentiate between 120 and 180 μg dosing. A companion manuscript describes dose selection based on exposure–response/safety modeling.


Journal of Crohns & Colitis | 2018

Effect of PF-00547659 on Central Nervous System Immune Surveillance and Circulating β7+ T Cells in Crohn’s Disease: Report of the TOSCA Study

Geert D’Haens; Severine Vermeire; Harald Vogelsang; Matthieu Allez; Pierre Desreumaux; André Van Gossum; William J. Sandborn; Daniel C. Baumgart; Richard M. Ransohoff; Gail M. Comer; Alaa Ahmad; Fabio Cataldi; John B. Cheng; Robert Clare; Kenneth J. Gorelick; Annamarie Kaminski; Vivek Pradhan; Sunday Rivers; Matthew Sikpi; Yanhua Zhang; Mina Hassan-Zahraee; W. Reinisch; Olaf Stüve

Abstract Background and Aims Progressive multifocal leukoencephalopathy [PML], a brain infection associated with anti-integrin drugs that inhibit lymphocyte translocation from bloodstream to tissue, can be fatal. Decreased central nervous system [CNS] immune surveillance leading to this infection has been reported in patients with multiple sclerosis or Crohn’s disease treated with anti-integrin antibody natalizumab. PF-00547659 is an investigational human monoclonal antibody for inflammatory bowel disease, targeted against α4β7-mucosal addressin cell-adhesion molecule-1 [the integrin ligand selectively expressed in the gut]. We hypothesised that this selective agent would not affect central nervous system immune surveillance. Methods Cerebrospinal fluid from five healthy volunteers, and from 10 patients with Crohn’s disease previously treated with immunosuppressants, was evaluated to assess the feasibility of the study. Subsequently, 39 patients with active Crohn’s disease and previous immunosuppression were evaluated over 12 weeks of PF-00547659-induction therapy. We measured total lymphocytes, T cell subsets in cerebrospinal fluid, and circulating β7+ memory cells. Disease activity was assessed using the Harvey–Bradshaw Index. Results Patients treated with PF-00547659 had no reduction of cerebrospinal fluid lymphocytes, T-lymphocyte subsets, or CD4:CD8 ratio, whereas circulating β7+ memory cells increased significantly. A total of 28/35 [80%] patients had a clinical response and 27/34 [79%] had disease remission. Treatment-related adverse events, none serious, were reported in 23/49 [47%] patients. Conclusions In patients with active Crohn’s disease, natalizumab therapy increases the risk for PML, and the increased risk is thought to be associated with iatrogenic leukopenia within the CNS. PML under PF-00547659 may be a lesser concern, as this agent did not reduce lymphocytes or T cell subsets in the cerebrospinal fluid.


Journal of Crohns & Colitis | 2018

Anti-MAdCAM Antibody Increases ß7+ T Cells and CCR9 Gene Expression in the Peripheral Blood of Patients With Crohn’s Disease

Mina Hassan-Zahraee; Anindita Banerjee; John B. Cheng; Weidong Zhang; Alaa Ahmad; Karen Page; David von Schack; Baohong Zhang; Steven W. Martin; Satyaprakash Nayak; Padma Reddy; Li Xi; Hendrik Neubert; Mireia Fernandez Ocana; Ken Gorelick; Robert Clare; Michael Vincent; Fabio Cataldi; Kenneth E. Hung

Abstract Objective To define pharmacodynamic biomarkers in the peripheral blood of patients with Crohn’s disease [CD] after treatment with PF-00547659, an anti-human mucosal addressin cell adhesion molecule-1 [MAdCAM-1] monoclonal antibody. Methods In this Phase 2, randomised, double-blind, controlled study [OPERA], blood samples were analysed from patients with moderate to severe active CD who received placebo or 22.5 mg, 75 mg, or 225 mg of PF-00547659 subcutaneously at baseline and at Weeks 4 and 8, with follow-up at Week 12. Soluble MAdCAM [sMAdCAM] was measured by mass spectrometry, β7-expressing T cells by flow cytometry, and gene transcriptome by RNA sequencing. Results A slight increase in sMAdCAM was measured in the placebo group from baseline to Week 12 [6%], compared with significant decreases in all PF-00547659 groups [–87% to –98%]. A slight increase from baseline to Week 12 was observed in frequency and molecules of equivalent soluble fluorochrome for β7+ central memory T cells in the placebo group [4%], versus statistically significant increases in the active treatment groups [48% to 81%]. Similar trends were seen for β7+ effector memory T cells [placebo, 8%; PF-00547659, 84–138%] and β7+ naïve T cells [8%; 13–50%]. CCR9 gene expression had statistically significant up-regulation [p = 1.09e-06; false discovery rate < 0.1] with PF-00547659 treatment, and was associated with an increase in β7+ T cells. Conclusions Results of the OPERA study demonstrate positive pharmacology and dose-dependent changes in pharmacodynamic biomarker measurements in blood, including changes in cellular composition of lymphocytes and corresponding CCR9 gene expression changes.


Gastroenterology | 2015

825 Anti-MAdCAM-1 Antibody (PF-00547659) for Active Refractory Crohn's Disease: Results of the OPERA Study

William J. Sandborn; Scott D. Lee; Dino Tarabar; Edouard Louis; Maria Kłopocka; Jochen Klaus; Walter Reinisch; Xavier Hébuterne; Dong Il Park; Stefan Schreiber; Kenneth J. Gorelick; Mina Hassan-Zahraee; Lisa S. Brown; Alaa Ahmad; John B. Cheng; Fabio Cataldi; Geert R. D'Haens


Gastroenterology | 2015

901a A Randomized, Multicenter Double-Blind, Placebo-Controlled Study of the Safety and Efficacy of Anti-MAdCAM Antibody PF-00547659 (PF) in Patients With Moderate to Severe Ulcerative Colitis: Results of the TURANDOT Study

Walter Reinisch; William J. Sandborn; Silvio Danese; Fabio Cataldi; Xavier Hébuterne; Bruce Salzberg; Maria Kłopocka; Dino Tarabar; Tomas Vanasek; Miloš Greguš; Paul Hellstern; Joo Sung Kim; Miles Sparrow; Kenneth J. Gorelick; Michelle Hinz; Alaa Ahmad; Mina Hassan-Zahraee; Vivek Pradhan; Severine Vermeire


D'Haens, G., Lee, S., Tarabar, D., Louis, E., Klopocka, M., Park, D. I., Claus, J., Hebuterne, X., Reinisch, W., Schreiber, Stefan, Gorelick, K. J., Cheng, J., Hassan-Zahraee, M., Brown, L., Ahmad, A., Banerjee, A., Cataldi, F. and Sandborn, W. (2015) Anti-MAdCAM-1 Antibody (PF-00547659) for Active Refractory Crohn's Disease: Results of the OPERA study Journal of Crohns & Colitis, 9 . S14-S14. | 2015

Anti-MAdCAM-1 Antibody (PF-00547659) for Active Refractory Crohn's Disease: Results of the OPERA study

G. D'Haens; Sang Joon Lee; Dino Tarabar; Edouard Louis; Maria Kłopocka; Dong Il Park; J. Claus; Xavier Hébuterne; W. Reinisch; Stefan Schreiber; Kenneth J. Gorelick; John B. Cheng; Mina Hassan-Zahraee; Lisa S. Brown; Alaa Ahmad; Anindita Banerjee; Fabio Cataldi; W. Sandborn

Collaboration


Dive into the Alaa Ahmad's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xavier Hébuterne

University of Nice Sophia Antipolis

View shared research outputs
Top Co-Authors

Avatar

Maria Kłopocka

Nicolaus Copernicus University in Toruń

View shared research outputs
Top Co-Authors

Avatar

Dino Tarabar

Military Medical Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge