scholarly journals A Single Administration of the Cytolytic CD38 Antibody TAK-079 to Healthy Subjects: Tolerability, Pharmacokinetics and Pharmacodynamics

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3249-3249 ◽  
Author(s):  
Eric Robert Fedyk ◽  
Deborah Berg ◽  
Glenda Smithson ◽  
Jose Estevam ◽  
Lachy Mclean ◽  
...  

Abstract Background: A study investigating the safety, efficacy, pharmacokinetics (PK) and pharmacodynamics (PD) of TAK-079 administered subcutaneously as a single agent in participants with relapsed/refractory multiple myeloma (RRMM) is ongoing because this investigational agent demonstrated distinct pharmacologic properties in a first in human study involving healthy volunteers. CD38 is expressed at high levels on subtypes of leukocytes which mediate immune disorders (e.g. plasma cells, activated B and T lymphocytes) and the selective removal of these cells could also restore immune homeostasis in these patients. TAK-079 is a fully human monoclonal antibody which binds to human CD38 protein with high affinity (KD = 0.7 nM) and lyses bound cells by CDC and ADCC in vitro. Methods: A randomized, double-blind, placebo-controlled study was conducted to assess the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of single intravenous (IV) infusion or subcutaneous (SC) injection of TAK-079 in escalating dose cohorts of healthy subjects. A starting single dose of 0.0003 mg/kg IV was selected, based upon modeling of responses in human and monkey model systems. Dose escalation ceased once a prespecified PD objective was achieved (i.e. ≥ 50% reduction in a leukocyte subset within peripheral blood which was sustained for ≥ 1 month). Six subjects were enrolled per dose cohort (4 receiving TAK-079; 2 receiving matching placebo). Subjects were monitored for at least 92 days post-dosing. Results: After IV (n=30) or SC (n=24) administration at all dose levels, TAK-079 was well tolerated and no safety concerns were identified. The maximum doses given were 0.06 mg/kg IV and 0.6 mg/kg SC. There were no serious adverse events (AE), severe AEs, withdrawals due to AEs, or infusion reactions. All AEs were mild or moderate. At the higher doses of TAK-079 administration, transient, mild to moderate increases in cytokine levels coincided with anticipated cell depletion; clinical symptoms primarily included mild pyrexia, headache, and postural hypotension. No remarkable findings for laboratory tests, ECGs, vital signs, or physical examinations were reported. Following a 2-hour IV infusion of 0.06 mg/kg, Cmax was observed at 5 minutes post EOI with a mean Cmax of 100.4 ng/mL. After Cmax was reached, serum concentrations decreased below the limit of quantification within 1 to 4 hours after the end of infusion. Following a single SC injection of 0.6 mg/kg TAK-079, the mean Cmax of 23.0 ng/mL was observed at approximately 24 hours post-dose and decreased gradually to below the limit of quantification by a median of 8 days (range 3-14). The mean AUClast and AUC∞ values were 90.4 and 212 ng*day/mL, respectively. Pharmacodynamic effects included a transient, dose-dependent reduction in NK cells at doses ≥0.003 mg/kg IV, approximately 167-fold less than the lowest dose reported for NK cell reduction by daratumumab administered to RRMM patients IV (≥0.5 mg/kg1). TAK-079 administered IV or SC reduced NK cells to a comparable extent (EC75 = 21 and 23 ng/mL, respectively). Greater than 90% reduction of plasmablasts was also observed in each subject dosed at 0.6 mg/kg SC, which generally peaked 2 days after injection and returned to baseline levels by 29 days. Neutrophil, lymphocyte, monocyte, red blood cell and platelet counts remained within normal ranges for all dose cohorts. Conclusion: TAK-079 may be a potent and convenient second generation anti-CD38 therapeutic, which warrants development for treatment of hematologic malignancies. Therefore, the safety, efficacy, pharmacokinetics, and pharmacodynamics of subcutaneous TAK-079 is being investigated in patients with RRMM in an ongoing study (NCT03439280; Figure 1) The starting dose in the RRMM study is fixed at 45 mg, based on the profile of the equivalent 0.6 mg/kg in healthy subjects. References Blood Adv. 2017 Oct 24;1(23):2105-2114. Disclosures Fedyk: Takeda Pharmaceuticals Inc.: Employment. Berg:Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited: Employment. Smithson:Takeda Pharmaceuticals Inc.: Employment. Estevam:Takeda Pharmaceuticals Inc.: Employment. Mclean:Takeda Pharmaceuticals Inc.: Employment. Allikmets:Takeda Pharmaceuticals Inc.: Employment. Palumbo:Takeda Pharmaceuticals Inc.: Employment.

2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 6047-6047 ◽  
Author(s):  
Jill Gilbert ◽  
Michael J. Schell ◽  
Xiuhua Zhao ◽  
Barbara A. Murphy ◽  
Tawee Tanvetyanon ◽  
...  

6047 Background: For patients with R/M SCCHN, cetuximab, a monoclonal antibody against EGFR, is approved as a single agent and has a survival benefit when combined with chemotherapy. We hypothesized that addition of sorafenib, a multi-kinase inhibitor of targets including VEGFR, to cetuximab may have greater clinical benefit than cetuximab alone. Methods: This trial was designed as a blinded, randomized phase II, placebo-controlled study of cetuximab at 400 mg/m2 IV on day 1 followed by 250 mg/m2 IV weekly plus placebo bid (Arm A) or cetuximab at the same dose and schedule plus sorafenib 400 mg po bid (Arm B), each in 21 day cycles. After 19 patients were enrolled, the trial was amended to remove the placebo (and blinding) due to issues with placebo tablet solubility. Target sample size was 84 patients with 83% power to detect a 2-month increase in PFS, the primary study endpoint. Interim analysis was planned at midpoint, requiring hazard ratio < 1 to proceed to the second stage of study. Serum cytokine and tumor HPV ISH and p16 analyses were performed. Results: Of 56 patients (ages 26-74, 80% male) enrolled, 53 patients received treatment and 41 were evaluable for response. Of the patients who received therapy, 26 received cetuximab only (Arm A). For Arm A, the mean number of cycles delivered was 4.3 (range 1-16) and the mean for Arm B was 3.3 (range 1-11). The most common grade 3/4 AEs were fatigue (2 A, 1 B), hypertension (3 B), infusion reaction (both arms), and diarrhea (2 B). Arm A had 2 PRs and Arm B had 4 PRs. Median OS was 7 mo and 5.9 mo respectively. Median PFS was 3.1 mo for both arms. 24 patients had pre-treatment cytokine measurements. Of the 12 measured cytokines, high TGFB1 level was significantly correlated with inferior PFS (4.6 mo vs 1.6 mo), regardless of arm (p=0.015). 38 patients had tumors available for p16 staining (31 neg and 7 pos). 3 of 7 p16 pos were also HPV ISH pos. The p16 neg patients had significantly improved PFS (3.5 mo vs 1.6 mo) regardless of arm (p=0.032) but no difference in OS (p=1.0). Conclusions: Both arms demonstrated clinical activity although no significant difference was observed. However, a subset of patients with p16 neg tumors or low serum TGFB1 may have a greater benefit with cetuximab-based therapy. Clinical trial information: NCI-2012-02847.


2012 ◽  
Vol 56 (11) ◽  
pp. 5946-5951 ◽  
Author(s):  
Lillian S. L. Ting ◽  
Jens Praestgaard ◽  
Nicole Grunenberg ◽  
Jenny C. Yang ◽  
Jennifer A. Leeds ◽  
...  

ABSTRACTClostridium difficileis the leading cause of hospital-acquired infectious diarrhea. LFF571 is a novel inhibitor of the prokaryotic translation elongation factor Tu and is active against a range of bacterial species, includingC. difficile. This first-in-human study investigated the safety and pharmacokinetics of single and multiple ascending oral doses of LFF571 in healthy subjects. This was a randomized, double-blind, placebo-controlled study. Except for one cohort, LFF571 was given with a high-fat meal to all single-dose cohorts (25 mg, 100 mg, 400 mg, and 1,000 mg). In the multiple-dose cohorts (25 mg, 100 mg, or 200 mg every 6 h for 10 days), LFF571 was given without regard to food. A total of 56 subjects completed the study, with 32 and 25 receiving single and multiple doses, respectively. There were no deaths, no serious adverse events, and no subject withdrawals due to an adverse event. The most common adverse event was diarrhea; gastrointestinal pain or distension was also noted. Diarrhea did not develop more frequently among subjects who received LFF571 than among those who received a placebo. LFF571 had limited systemic exposure and high steady-state fecal concentrations. The highest concentration of LFF571 in serum (3.2 ng/ml) was observed after the last dose in a subject who received 200 mg every 6 h for 10 days. LFF571 was generally safe and well tolerated in single and multiple oral doses in healthy subjects. The minimal serum and high fecal concentrations support the further development of LFF571 for the treatment ofC. difficileinfections.


2021 ◽  
Vol 27 (Supplement_1) ◽  
pp. S1-S1
Author(s):  
Stephen Hanauer ◽  
Terry O’Reilly ◽  
Robert Lester ◽  
Neal Slatkin ◽  
Jimin Lee ◽  
...  

Abstract Objective To evaluate the safety profile of amiselimod, a selective sphingosine 1-phosphate receptor modulator which has been shown to regulate lymphocyte trafficking and is in development for the treatment of inflammatory bowel disease. Methods A randomized, double-blind, multiple-dose, placebo-controlled, parallel study with a nested crossover design evaluated the safety and tolerability profile of amiselimod. Healthy adults were randomized in a 2:1:1 ratio during a 28-day treatment period accordingly: a single dose of placebo followed by oral amiselimod (upwardly titrated in doses ranging from 0.4 to 1.6 mg to achieve 0.4 mg and 0.8 mg steady-state exposure; a single dose of oral moxifloxacin 400 mg followed by placebo; or placebo followed by a single dose of moxifloxacin 400 mg. The safety population included all subjects who received at least one dose of treatment. Adverse events (AE) and serious AEs were collected. Treatment-emergent AEs were defined as an AE that was starting or worsening at the time of or after study drug administration. Changes in clinical laboratory parameters (including lymphocyte counts), physical examinations, vital signs, and electrocardiogram parameters (including heart rate, PR, QRS, and QT intervals) were recorded. Subjects were permitted to withdraw if lymphocyte counts were ≤ 0.2 x 109/L. Results The safety population included 190 subjects of which 95 received amiselimod and 95 were in the combined moxifloxacin group. Subjects were 40% female, 83% white, and the mean (standard deviation) age was 39.0 (8.8) years. The discontinuation rate was 8% (n=8) in the amiselimod group and 4% (n=4) in the moxifloxacin group. Three subjects who received amiselimod discontinued because they met the stopping criteria for low lymphocyte counts. One subject experienced an amiselimod-related serious AE of atrial fibrillation on day 26 (after receiving amiselimod 1.6 mg for 3 of the preceding 4 days) that required hospitalization, cardioversion, and led to discontinuation. No deaths were reported. All other AEs were mild to moderate in severity. Decreased white blood counts were the most commonly reported TEAE, followed by headache and constipation (Table). Reductions in white blood counts returned to normal range after study discontinuation without sequelae. Decreased neutrophils, lymphocytes and hemoglobin, and increased creatine kinase, alanine aminotransferase, and aspartate aminotransferase were reported, all of which resolved without sequelae. The mean absolute lymphocyte count for amiselimod exhibited a gradual decrease from predose (1.681 thou/uL) to a nadir of 0.424 thou/uL on day 27 (Figure). Changes to vital signs, physical examinations, and ECG parameters were within normal limits. Conclusions Upwardly titrated doses of amiselimod ranging from 0.4 to 1.6 mg were generally well tolerated in healthy subjects.


1972 ◽  
Vol 71 (1) ◽  
pp. 24-36 ◽  
Author(s):  
Ariel Gordin ◽  
Pirkko Saarinen

ABSTRACT An account is given of a methodological study of the double-antibody radioimmunoassay of human TSH, using highly purified labelled human TSH as tracer. It was shown that conventional paper electrophoresis was not adequate for studying the purity of labelled human TSH. When polyvinylchloride (Pevikon®) electrophoresis was used, four subfractions could still be separated, even though, on paper electrophoresis, the material seemed to be homogeneous. Only two of the four Pevikon fractions were immunoreactive. Purification of labelled human TSH by Pevikon electrophoresis also improved the sensitivity of the assay. Specific activities of about 100 mCi/mg gave the highest initial binding capacity, produced least damage to the labelled hormone and showed the best stability of the tracer without influencing the sensitivity of the method. In different storage conditions, labelled human TSH was found to be most stable at −20°C and diluted 1/100. Only in pregnancy did the addition of HCG seem necessary. The mean TSH value in healthy subjects was 3.6 ± 1.4 μU/ml (mean±sd) with a range from 1.6 μU/ml to 8.8 μU/ml.


Sign in / Sign up

Export Citation Format

Share Document