scholarly journals Preliminary Results of a Phase 1 Study in Healthy Subjects Administered Inclacumab, a Fully Human IgG4 Anti-P-Selectin Monoclonal Antibody in Development for Treatment of Sickle Cell Disease

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 977-977
Author(s):  
Christina Mayer ◽  
Daniel Scott Cooper ◽  
Andrew Redfern ◽  
Xin Geng ◽  
Jianxia Shi ◽  
...  

Abstract Background: Inclacumab, a fully human IgG4 anti-P-selectin monoclonal antibody, is being developed for the reduction of vaso-occlusive crises (VOCs) in patients with sickle cell disease (SCD). P-selectin-mediated platelet-leukocyte aggregate (PLA) formation has been shown to contribute to vaso-occlusion. Safety and pharmacology of inclacumab have previously been well characterized in over 700 subjects (healthy volunteers and patients with cardiovascular disease), at doses up to 20 mg/kg every 4 weeks for up to 9 months. The current Phase 1 study was initiated to evaluate the safety and pharmacology of inclacumab at doses of 20 mg/kg and 40 mg/kg in healthy subjects in support of a target Phase 3 dose of 30 mg/kg administered every 12 weeks to patients with SCD. Methods: Healthy adult subjects over 18 years of age without significant current or prior health conditions received a single intravenous (IV) dose of 20 mg/kg inclacumab infused over approximately one hour (Cohort 1). Following a review of safety, a second cohort received a single IV dose of 40 mg/kg infused over approximately one hour (Cohort 2). The total study duration and sample collection period was 29 weeks. Final safety and preliminary pharmacokinetics (PK), anti-drug antibody (ADA), and ex vivo thrombin receptor-activating peptide (TRAP)-activated PLA formation data are reported. Results: Fifteen subjects received a single dose of inclacumab 20 mg/kg (n=6) or 40 mg/kg (n=9). Fourteen subjects completed the study. Median age was 42 years (range 22 - 52 years); median body weight was 73.6 kg (range 63.7 - 89.3 kg). Through the pre-specified 72-hour post-infusion safety assessment period in both cohorts, no treatment-emergent adverse events (AEs) > grade 1 (mild) nor dose-limiting toxicities were reported. During the duration of the study, there were no serious AEs, infusion-related reactions, or hypersensitivity reactions. Additionally, no clinically significant changes in vital signs, laboratory findings, or ECGs were observed. The most common AEs were headache, myalgia, and contact dermatitis. The only events assessed by the investigator as potentially related to inclacumab were headache and dizziness, which were experienced by one subject (20 mg/kg) and occurred 4 hours following the end of infusion. In healthy subjects, inclacumab demonstrated dose-proportional PK over the dose range tested; PK parameter estimates were consistent with those reported for monoclonal antibodies. Geometric mean C max following single doses of 20 and 40 mg/kg were 402 and 970 µg/mL, respectively. Mean TRAP-activated predose PLA formation was 33 - 39% across cohorts and decreased to 9-14% at 2 hours following end of infusion. PLA inhibition was sustained through at least 12 weeks for both the 20 and 40 mg/kg doses. Two subjects in the 40 mg/kg cohort were ADA-positive on Week 12 and thereafter; a preliminary analysis demonstrated no apparent impact on PK or safety in these subjects. Conclusions: Inclacumab displayed a well-tolerated safety profile for up to 29 weeks following a single dose of 20 or 40 mg/kg in healthy subjects. Durable inhibition of TRAP-activated PLA formation was observed through at least 12 weeks, consistent with prior observations. Overall, the results support a Phase 3 dose of 30 mg/kg every 12 weeks in patients with SCD-related VOCs. Funding: This study was supported by Global Blood Therapeutics. Disclosures Mayer: Global Blood Therapeutics: Consultancy. Redfern: Linear Clinical Research: Current Employment; Novartis: Other: Advisory Board; Pfizer: Other: Advisory Board; Roche: Other: Advisory Board; Eisai: Other: Advisory Board; Astra Zeneca: Other: Advisory Board. Geng: Global Blood Therapeutics: Current Employment. Shi: Global Blood Therapeutics: Current Employment. van Zutphen-van Geffen: Global Blood Therapeutics: Consultancy. Kuan: Global Blood Therapeutics: Consultancy. Koeck: Global Blood Therapeutics: Consultancy. Kastrissios: Global Blood Therapeutics: Consultancy. Patel: Global Blood Therapeutics: Consultancy. Davis: Global Blood Therapeutics: Current Employment. Yue: Global Blood Therapeutics: Current Employment.

PLoS ONE ◽  
2016 ◽  
Vol 11 (4) ◽  
pp. e0152895 ◽  
Author(s):  
Jennifer F. Doss ◽  
Jude C. Jonassaint ◽  
Melanie E. Garrett ◽  
Allison E. Ashley-Koch ◽  
Marilyn J. Telen ◽  
...  

2013 ◽  
Vol 75 (6) ◽  
pp. 1433-1444 ◽  
Author(s):  
Joseph A. Jakubowski ◽  
Chunmei Zhou ◽  
David S. Small ◽  
Kenneth J. Winters ◽  
D. Richard Lachno ◽  
...  

PLoS Medicine ◽  
2017 ◽  
Vol 14 (9) ◽  
pp. e1002382 ◽  
Author(s):  
Robert Molokie ◽  
Donald Lavelle ◽  
Michel Gowhari ◽  
Michael Pacini ◽  
Lani Krauz ◽  
...  

2014 ◽  
Vol 133 (2) ◽  
pp. 190-195 ◽  
Author(s):  
Joseph A. Jakubowski ◽  
Chunmei Zhou ◽  
Stipo Jurcevic ◽  
Kenneth J. Winters ◽  
D. Richard Lachno ◽  
...  

2021 ◽  
Vol 27 (3) ◽  
pp. S12-S13
Author(s):  
Mark C. Walters ◽  
Alexis A. Thompson ◽  
Markus Y. Mapara ◽  
Janet L. Kwiatkowski ◽  
Lakshmanan Krishnamurti ◽  
...  

2020 ◽  
Vol 26 (3) ◽  
pp. S1-S2
Author(s):  
Mark C. Walters ◽  
Julie Kanter ◽  
Janet L. Kwiatkowski ◽  
Lakshmanan Krishnamurti ◽  
Markus Y. Mapara ◽  
...  

Blood ◽  
2015 ◽  
Vol 125 (23) ◽  
pp. 3668-3669 ◽  
Author(s):  
Maureen M. Okam ◽  
Erica B. Esrick ◽  
Elyse Mandell ◽  
Federico Campigotto ◽  
Donna S. Neuberg ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2240-2240 ◽  
Author(s):  
Evgeny Ozhegov ◽  
Ramprasad Srinivasan ◽  
Vladimir Bogdanov

Abstract Abstract 2240 Background and Rationale: Vasoocclusive crises are a major hallmark of sickle cell disease (SCD) pathobiology; experimental evidence suggests that SCD vasoocclusion can be triggered by the increased adhesion of white blood cells, including monocytes, to the microvascular endothelium. Pro-coagulant activity of Tissue Factor, the trigger of blood coagulation, is heightened in the blood of patients with SCD. We recently reported that, compared to full length Tissue Factor (flTF), alternatively spliced Tissue Factor (asTF) acts as a very potent inducer of cell adhesion molecules E-selectin, VCAM-1, and ICAM-1 on microvascular endothelial cells, thereby raising the possibility that asTF may promote monocyte adhesion to the endothelium in vivo (Srinivasan et al, J Thromb Haemost 2011). Analogously to flTF, asTF is continuously present in circulation. Currently, no asTF-specific assay exists that can reliably detect asTF protein in plasma, and no data is available on the levels of asTF in the plasma of patients with SCD. We sought to develop monoclonal antibodies suitable for asTF-specific enzyme-linked immunosorbent assay (ELISA), to evaluate the levels of plasma asTF in SCD patients and age/gender matched healthy subjects. Methods: Two rabbit monoclonal antibodies were raised and characterized: i) antibody RabMab-95 recognizing amino acid residues 81–95 of mature asTF; ii) antibody RabMab-1 recognizing the last 11 amino acid residues of the asTF's unique C-terminus. By western blotting, both RabMab's recognized a) recombinant asTF produced in E. coli, b) eukaryotic asTF expressed in HEK293 cells using an inducible promoter system, and c) native asTF constitutively expressed in human pancreatic adenocarcinoma cell lines, with high specificity and sensitivity. In a sandwich ELISA of platelet poor plasma (PPP) samples, RabMab-95 was used as the capture antibody and horseradish peroxidase-conjugated RabMab-1 as the detection antibody; conventional blocking, sample incubation, and substrate development techniques were used. In addition, levels of flTF in PPP samples were assessed using ZYMUTEST Tissue Factor kit (RK035A, HYPHEN BioMed). Results: The SCD cohort comprised 16 pediatric and adult patients (10 females and 6 males, average age: 28.25±11.3 years); in the healthy subject cohort (n=17, 10 females and 7 males), the average age was 26.6±6.7 years. 14 out of 16 SCD patients had detectable levels of asTF, ranging from 25 pg/mL to 38,350 pg/mL (average: 5,323±9,934 pg/mL); in contrast, only 2 out of 17 healthy subjects had detectable levels of asTF: one PPP sample had 650 pg/ml and the other, 1,883 pg/mL (p=0.0397, SCD vs healthy subjects). The adult (>20 y.o., n= 10, average age: 35.2±7.8 years) and the pediatric (≤20 y.o., n=6, average age: 16.7±3.6 years) SCD sub-cohorts had average asTF values of 8,319±11,738 pg/mL and 329±446 pg/mL, respectively; while the difference between the adult SCD sub-cohort and the age-matched healthy subject sub-cohort was statistically significant (p=0.0337, adult SCD vs age-matched healthy subjects), there was a trend toward statistical significance in the pediatric asTF sub-cohort when compared to age-matched healthy subjects (p=0.1004, pediatric SCD vs age-matched healthy subjects). The levels of flTF in SCD plasma ranged from less than 1 pg/mL to 105 pg/mL (8 out of 16 patients), and did not correlate with asTF levels. Conclusions: We have developed a monoclonal ELISA for specific detection of asTF in human PPP. Our findings indicate that adult as well as pediatric SCD patients have heightened levels of asTF protein in circulation. Importantly, in ∼50% of SCD patients the levels of plasma asTF were in the range vastly exceeding the levels previously reported for any form of blood borne TF, likely sufficient to trigger a physiologically significant increase in leukocyte adhesion to the endothelium. Examination of circulating asTF levels in larger cohorts of pediatric and adult patients with SCD is thus highly warranted. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document