Phase I and Pharmacokinetic Study of Thalidomide With Carboplatin in Children With Cancer

2004 ◽  
Vol 22 (21) ◽  
pp. 4394-4400 ◽  
Author(s):  
Murali Chintagumpala ◽  
Susan M. Blaney ◽  
Lisa R. Bomgaars ◽  
Aleksander Aleksic ◽  
John F. Kuttesch ◽  
...  

Purpose Tumor growth and metastasis is believed to depend on the tumor's ability to induce neovascularization. Recent studies have indicated that thalidomide inhibits angiogenesis. We performed a phase I and pharmacokinetic study of thalidomide with carboplatin in children with refractory solid tumors. Patients and Methods Carboplatin was administered as a single intravenous dose once every 21 days at a target area under the concentration-time curve of 6 mg/mL·min. Thalidomide was administered daily by mouth. The initial dose level was 100 mg/m2/d with intrapatient dose escalation to a maximum dose of 300 mg/m2/d. The next cohort of patients started at a dose of 300 mg/m2/d, with intrapatient dose escalation to a maximum dose of 500 mg/m2/d. Standard response and adverse event criteria were used. Serial blood samples for thalidomide pharmacokinetics studies were obtained after the first dose. Results Twenty-two patients received 56 cycles of therapy. The maximum tolerated thalidomide dose was 400 mg/m2/d. The dose-limiting toxicity was somnolence. There were no objective responses. Thalidomide's apparent clearance was 55 ± 16 mL/min/m2 and the terminal half-life was 5.9 ± 2.8 hours. There was no evidence of dose-dependent pharmacokinetics in the narrow range studied. Conclusion Thalidomide at a dose of 400 mg/m2/d can be safely administered to children with solid tumors in combination with carboplatin. Somnolence is the major toxicity. In addition, we have characterized the pharmacokinetic behavior of thalidomide in children. This study can serve as the basis for future investigation of thalidomide as an anticancer agent in children.

2009 ◽  
Vol 66 (3) ◽  
pp. 441-448 ◽  
Author(s):  
E. Gabriela Chiorean ◽  
Srikar Malireddy ◽  
Anne E. Younger ◽  
David R. Jones ◽  
Mary-Jane Waddell ◽  
...  

2014 ◽  
Vol 74 (6) ◽  
pp. 1191-1198 ◽  
Author(s):  
R. C. Brennan ◽  
W. Furman ◽  
S. Mao ◽  
J. Wu ◽  
D. C. Turner ◽  
...  

2011 ◽  
Vol 69 (4) ◽  
pp. 1013-1020 ◽  
Author(s):  
Sukhmani K. Padda ◽  
Yelena Krupitskaya ◽  
Laveena Chhatwani ◽  
George A. Fisher ◽  
Alexander D. Colevas ◽  
...  

2010 ◽  
Vol 101 (10) ◽  
pp. 2193-2199 ◽  
Author(s):  
Toru Mukohara ◽  
Shunji Nagai ◽  
Minori Koshiji ◽  
Kenichi Yoshizawa ◽  
Hironobu Minami

2009 ◽  
Vol 15 (19) ◽  
pp. 6232-6240 ◽  
Author(s):  
George D. Demetri ◽  
Patricia Lo Russo ◽  
Iain R.J. MacPherson ◽  
Ding Wang ◽  
Jeffrey A. Morgan ◽  
...  

2014 ◽  
Vol 32 (3_suppl) ◽  
pp. 554-554 ◽  
Author(s):  
Patricia LoRusso ◽  
J. Randolph Hecht ◽  
Dung Luong Thai ◽  
Michael J. Hawkins ◽  
Hua Dong ◽  
...  

554 Background: Simtuzumab (SIM) is a monoclonal antibody that inhibits lysyl oxidase like molecule 2 (LOXL2), an extracellular matrix enzyme involved in tumor growth and metastasis. Methods: A phase I dose escalation study evaluated the safety and pharmacokinetics (PK) in patients (pts) with advanced solid tumors. Dose escalation occurred at doses up to 20 mg/kg of SIM every 2 weeks by IV infusion followed by a dose expansion of 20 pts at 10 mg/kg of SIM every 2 weeks. PK was evaluated following the first and fourth doses. In a separate Phase IIa study, 11 pts with KRAS mutant colorectal cancer (CRC) were given SIM at 700 mg IV combined with FOLFIRI every 2 weeks in the 2nd line setting. All pts continued until disease progression or unacceptable toxicity. CT or MRI was performed every 8 weeks. Results: In the phase I study, 12 pts (3 per cohort) received SIM at doses of 1, 3, 10 and 20 mg/kg. Tumor types included CRC (5), pancreatic neuroendocrine (PNE, 1), and other tumors (1 each). In the dose expansion, 20 CRC pts (11 KRAS mutant and 9 KRAS wild-type) received SIM at 10 mg/kg. Treatment-emergent AEs (TEAEs) included fatigue and constipation. No DLTs or drug-related SAEs were observed. The mean terminal T1/2 was ~20 days, and exposure was dose proportional between 1 and 20 mg/kg. Decreased tumor size (-56% for PNE and -17%, -8%, -5%, -5%, and -4% for CRC) as measured by the sum of the linear diameter of the target lesions was observed in 6 pts following treatment with SIM alone. In the Phase IIa trial of SIM/FOLFIRI, the most common TEAEs were diarrhea, fatigue, and nausea. Median PFS was 7.8 months. Conclusions: SIM was well tolerated as a single agent and when combined with FOLFIRI and showed promising efficacy in pts with CRC. A randomized, double-blind, placebo controlled phase II study in KRAS mutant CRC pts is ongoing. Clinical trial information: NCT01323933.


Sign in / Sign up

Export Citation Format

Share Document