scholarly journals Open-label, dose escalation phase I study in healthy volunteers to evaluate the safety and pharmacokinetics of a human monoclonal antibody to Clostridium difficile toxin A

Vaccine ◽  
2008 ◽  
Vol 26 (27-28) ◽  
pp. 3404-3409 ◽  
Author(s):  
Claribel P. Taylor ◽  
Sanjeev Tummala ◽  
Deborah Molrine ◽  
Lisa Davidson ◽  
Richard J. Farrell ◽  
...  
2016 ◽  
Vol 22 (21) ◽  
pp. 5204-5210 ◽  
Author(s):  
Danny N. Khalil ◽  
Michael A. Postow ◽  
Nageatte Ibrahim ◽  
Dale L. Ludwig ◽  
Jan Cosaert ◽  
...  

ESMO Open ◽  
2017 ◽  
Vol 1 (6) ◽  
pp. e000154 ◽  
Author(s):  
Howard A Burris ◽  
Suzanne Bakewell ◽  
Johanna C Bendell ◽  
Jeffrey Infante ◽  
Suzanne F Jones ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4029-4029 ◽  
Author(s):  
Suzanne Trudel ◽  
P. Leif Bergsagel ◽  
Seema Singhal ◽  
Ruben Niesvizky ◽  
Raymond L. Comenzo ◽  
...  

Abstract Abstract 4029 Introduction: Chromosomal translocation of the FGFR3 oncogene in t(4;14)-positive multiple myeloma (MM) causes FGFR3 overexpression in plasma cells, chemoresistance, and poor prognosis leading to shorter overall survival in MM patients (pts). MFGR1877S is a human monoclonal antibody that targets FGFR3 to prevent ligand binding, receptor-receptor association, and FGFR3 signaling. In preclinical studies, anti-FGFR3 antibody suppresses FGFR3-mediated cell proliferation, and exerts strong anti-tumor activity in mouse xenograft models of both t(4;14)-positive MM and bladder carcinoma. Methods: This Phase I study assessed the safety, tolerability, and biologic activity of MFGR1877S given intravenously, weekly for 3 weeks, followed by every 28-day dosing, to eligible patients with t(4;14)-positive MM. Dose escalation started with single-pt cohorts at 1 and 2 mg/kg, followed by a standard 3+3 dose-escalation scheme at doses ranging from 4–15 mg/kg. Dose escalation decisions were made based on monitoring pts for dose-limiting toxicities (DLT) during the first 22 days on study. FGFR3 expression, safety, pharmacokinetics (PK), and response (EBMT/IMWG criteria) were assessed. Main efficacy outcome measures include serum/urine M protein and free light chain (FLC) quantities. Results: A total of 14 pts (median age 66, range 45–78; 43% female) with a baseline ECOG status of 0–2, and a median number of 5 prior therapies (range 1–10), received a median of 3.5 doses (range 1–7) of MFGR1877S. Adverse events (AEs) deemed related to MFGR1877S were diarrhea, fatigue, and nausea (14% each), and anemia, increased creatinine, bone pain, confusion, decreased appetite, myalgia, and fever (7% each). The only Grade ≥ 3 related AE was fatigue (7%). Six pts experienced 9 serious AEs (SAE), one of which was Grade 2 pyrexia attributed to MFGR1877S that occurred within 24 hours of infusion and required hospitalization. This pt later discontinued due to a second SAE of Grade 2 pyrexia requiring hospitalization not attributed to MFGR1877S. One pt died of intracranial hemorrhage not attributed to MFGR1877S. No Grade 4 SAEs were reported. Other reported SAEs included Grade 3 hypercalcemia, neutropenia, pain in extremity, and musculoskeletal chest pain (1 pt each), Grade 2 ankle fracture, pneumonia, and pyrexia × 2 (1 pt each). No maximum tolerated dose was identified, as no DLTs were observed through the highest dose tested (15 mg/kg). Preliminary PK analysis for limited number of multiple myeloma patients (n=14) by non-linear mixed effect modeling demonstrated a trend of dose proportional increase of exposure (area under the concentration-time curve and maximal concentration) from 2 to 15 mg/kg. Population clearance of MFGR1877S was estimated to be ∼0.62 L/day, suggesting that MFGR1877S appeared to have a slightly faster clearance in MM patients, compared to the typical IgG1 monoclonal antibody clearance in human. The central volume of distribution of MFGR1877S was ∼3.4 L, which approximated human serum volume and appeared similar to the central volume of distribution of other typical IgG1 monoclonal antibodies. While some degree of FGFR3 expression was detected by immunohistochemistry in 10/14 patient bone marrow samples taken at screening, FACS analysis of FGFR3 surface expression on myeloma cells, although largely consistent with IHC measurements, also revealed heterogeneity in the levels of detectable surface FGFR3 expression. Six pts had stable disease as their best response: 2 pts up to 4 cycles, 1 pt up to 3 cycles, and 3 pts up to 1 cycle. Conclusions: MFGR1877S was well-tolerated overall in these patients with multiple relapsed or refractory MM. Although no objective responses were observed, stable disease was observed in 3 pts for 3–4 cycles. Disclosures: Off Label Use: MFGR1877S is a human monoclonal antibody that targets FGFR3 to prevent ligand binding, receptor-receptor association, and FGFR3 signaling. Singhal:Millennium and Celgene: Speakers Bureau. Niesvizky:Onyx, Millemium, Celgene. Speakers bureau: Millenium and Celgene: Consultancy, Research Funding. Comenzo:Millenium, Neotope; Onyx, Osiris, Millenium: Consultancy, Research Funding. Lebovic:Genentech: Speakers Bureau. Choi:Genentech: Employment. Lu:Genentech: Employment. French:Genentech: Employment. Penuel:Genentech: Employment. Ho:Genentech: Employment.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 3020-3020
Author(s):  
Shabina Roohi Ahmed ◽  
Douglas Wilmot Ball ◽  
David Cosgrove ◽  
Angela Scardina ◽  
Emily Petito ◽  
...  

3020 Background: Significant crosstalk exists between the PI3K and Raf/MEK/ERK pathways and treatment of select cell lines with a MEK inhibitor and an IGF1-R inhibitor has been shown to cause greater inhibition of growth than either agent alone. IMC-A12 (I) is a recombinant human monoclonal antibody directed IGFI-R; it blocks interaction between IGF-1R and its ligands, IGF-I and -II. AZD6244 (A) is a highly selective, non-competitive MEK ½ inhibitor. Methods: The study is a phase I, dose-escalation with a standard 3+3 design. Eligible patients had advanced solid tumor, good end organ function and performance status with exclusion for poorly controlled diabetes or growth hormone abnormalities. Part 1: open label, dose escalation. Pts were treated with A at 50 mg BID, then 75 mg BID; I at 12mg/kg q2wk held constant. DLTs were defined as grade (G) 3 or 4 toxicities during cycle 1. Part 2: expansion cohort of 15 pts for correlative endpoints and PK studies; serial tumor biopsies will be evaluated for changes in signaling in the IGF pathway, including p-ERK, p-Akt and RAS. Mutational analysis of RAS and RAF will be done to correlate with PD endpoints and disease response. Results: Part 1: 16 patients accrued with 6 treated at DL1 and 10 treated at DL2. Multiple tumor types were represented, including 4 colon, 2 thyroid, 2 pancreatic, and 1 breast. At 75 mg of A, there was 1 DLT of visual changes. G3 AEs included nausea/vomiting (n=2, 12%), visual changes/retinopathy (n=2, 12%), MRSA skin infection (n=1, 6%), (n=1, 6%), hyperglycemia (n=1, 6%), and stroke (n=1, 6%). Other common AEs included acneform rash (n=10, 63%), nausea/vomiting (n=6, 37%), anorexia (n=4, 25%), and diarrhea (n=3, 19%). Of 10 evaluable patients in Part 1, 1 had a partial response and 4 patients had stable disease (40%). Sustained responses of 10 and 14 months were seen in the DTC patients. 3 patients are currently enrolled in the expansion cohort. The RP2D is 50 mg of A, due to the visual changes seen with 75 mg, and 12mg/kg of I. Conclusions: A at 50 mg QD combines safely with I 12mg/kg. An expansion cohort is underway to determine if this combination warrants further investigation.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e14525-e14525 ◽  
Author(s):  
C. K. Kollmannsberger ◽  
H. Hurwitz ◽  
G. Vlahovic ◽  
C. Maroun ◽  
J. Dumouchel ◽  
...  

e14525 Background: MGCD265 is a novel multitargeted receptor tyrosine kinase (RTK) inhibitor that targets the mesenchymal epithelial transition (c-Met) and the vascular endothelial growth factor (VEGF) receptors (VEGFR1, VEGFR2, and VEGFR3). Additional RTK targets include Tie-2, and Ron. Those kinases are known to be involved in tumor development and angiogenesis. The objective of this Phase I study is to evaluate the safety, pharmacokinetics (PK) and pharmacodynamics (PD) of daily administration of MGCD265 in patients with solid tumors. Methods: This is a multicenter, open-label, dose escalation study of oral MGCD265 administered as a continuous 21-day cycle. Cohorts of 3–4 patients were enrolled per dose level, initially with dose doubling between cohorts, followed by smaller increments once grade 2 drug-related toxicity is observed. Dose limiting toxicity (DLT) was defined as: grade 4 neutropenia; grade 4 thrombocytopenia; any > grade 3 nonhematologic toxicity; severe/sustained hypertension; or any toxic effect leading to a patient missing > 4 doses of MGCD265. Treatment would continue until disease progression or toxicity. Results: Ten patients with advanced solid tumors have been treated. Characteristics: age range 25–75; gender: 8 M/2F; ECOG: 0 (1 patient); 1 (9 patients). At dose levels of 24, 48 and 96 mg/m2, no DLTs nor grade 2 or greater drug-related AEs during cycle 1 have been reported. Eight patients received treatment for 2 cycles or more. Preliminary PK profile after the first dose of administration shows a dose dependent increase in AUC and Cmax with an approximate mean half-life of 23 hours (see Table below). At the 96 mg/m2 dose, exposure was in the range of the lower end of the efficacious exposure in certain xenograft models. PD markers including plasma HGF and VEGF and shed/soluble receptors s-Met and s-VEGFR2 have been evaluated. Conclusions: Daily oral administration of MGCD265 was found to be well tolerated at doses of 24, 48, and 96 mg/m2. Further dose escalation is underway. [Table: see text] [Table: see text]


Sign in / Sign up

Export Citation Format

Share Document