Skip to main content
Top
Published in: Investigational New Drugs 4/2013

01-08-2013 | PHASE I STUDIES

Phase I study of oral CP-4126, a gemcitabine derivative, in patients with advanced solid tumors

Authors: F. E. Stuurman, E. E. Voest, A. Awada, P. O. Witteveen, T. Bergeland, P.-A. Hals, W. Rasch, J. H. M. Schellens, A. Hendlisz

Published in: Investigational New Drugs | Issue 4/2013

Login to get access

Summary

CP-4126 is a gemcitabine (2′,2′-difluorodeoxycytidine; dFdC) 5′ elaidic acid ester. The purpose of this dose-escalating study was to assess safety, pharmacokinetics (PK) and preliminary antitumor activity of the oral formulation and to determine the recommended dose (RD) for phase II studies. The study had a two-step design: a non-randomized dose-escalating step I with oral CP-4126 alone, followed by a randomized, cross-over step II that compared oral CP-4126 with dFdC i.v.. CP-4126 was given on days 1,8,15 in a 4-week schedule with increasing doses until the RD was established. 26 patients with different solid tumours were enrolled in step I at seven dose levels (100–3,000 mg/day). The most frequent drug-related AEs were fatigue and dysgeusia, the majority being grade 1–2. One patient experienced a dose limiting toxicity after one dose of CP-4126 at 1,300 mg/day (ASAT grade 3). PK of CP-4126 could not be determined. The metabolites dFdC and dFdU obeyed dose-dependent pharmacokinetics. Exposures to dFdC were about ten-fold lower compared to exposures after comparable doses of dFdC i.v.. Nine patients reached stable disease as best response, whereby in one patient with vaginal carcinoma a 25 % reduction of tumor volume was reached. This study demonstrates that CP-4126 can be safely administered orally to patients up to 3,000 mg/day in a d1,8,15 q4w schedule with a tolerable safety profile. CP-4126 acts as a prodrug for dFdC when given orally, but because of the poor absorption and the rapid pre-systemic metabolism the study was terminated early and no RD could be determined.
Literature
1.
go back to reference Noble S, Goa KL (1997) Gemcitabine. A review of its pharmacology and clinical potential in non-small cell lung cancer and pancreatic cancer. Drugs 54(3):447–472PubMedCrossRef Noble S, Goa KL (1997) Gemcitabine. A review of its pharmacology and clinical potential in non-small cell lung cancer and pancreatic cancer. Drugs 54(3):447–472PubMedCrossRef
2.
go back to reference Mackey JR, Mani RS, Selner M, Mowles D, Young JD, Belt JA, Crawford CR, Cass CE (1998) Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res 58(19):4349–4357PubMed Mackey JR, Mani RS, Selner M, Mowles D, Young JD, Belt JA, Crawford CR, Cass CE (1998) Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res 58(19):4349–4357PubMed
3.
go back to reference Bergman AM, Pinedo HM, Peters GJ (2002) Determinants of resistance to 2′,2′-difluorodeoxycytidine (gemcitabine). Drug Resist Updat 5(1):19–33PubMedCrossRef Bergman AM, Pinedo HM, Peters GJ (2002) Determinants of resistance to 2′,2′-difluorodeoxycytidine (gemcitabine). Drug Resist Updat 5(1):19–33PubMedCrossRef
4.
go back to reference Burke T, Lee S, Ferguson PJ, Hammond JR (1998) Interaction of 2′,2′-difluorodeoxycytidine (gemcitabine) and formycin B with the Na + -dependent and -independent nucleoside transporters of Ehrlich ascites tumor cells. J Pharmacol Exp Ther 286(3):1333–1340PubMed Burke T, Lee S, Ferguson PJ, Hammond JR (1998) Interaction of 2′,2′-difluorodeoxycytidine (gemcitabine) and formycin B with the Na + -dependent and -independent nucleoside transporters of Ehrlich ascites tumor cells. J Pharmacol Exp Ther 286(3):1333–1340PubMed
5.
go back to reference Mini E, Nobili S, Caciagli B, Landini I, Mazzei T (2006) Cellular pharmacology of gemcitabine. Ann Oncol 17(Suppl 5):7–12CrossRef Mini E, Nobili S, Caciagli B, Landini I, Mazzei T (2006) Cellular pharmacology of gemcitabine. Ann Oncol 17(Suppl 5):7–12CrossRef
6.
go back to reference Heinemann V, Xu YZ, Chubb S, Sen A, Hertel LW, Grindey GB, Plunkett W (1990) Inhibition of ribonucleotide reduction in CCRF-CEM cells by 2′,2′-difluorodeoxycytidine. Mol Pharmacol 38(4):567–572PubMed Heinemann V, Xu YZ, Chubb S, Sen A, Hertel LW, Grindey GB, Plunkett W (1990) Inhibition of ribonucleotide reduction in CCRF-CEM cells by 2′,2′-difluorodeoxycytidine. Mol Pharmacol 38(4):567–572PubMed
7.
go back to reference Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W (1991) Action of 2′,2′-difluorodeoxycytidine on DNA synthesis. Cancer Res 51(22):6110–6117PubMed Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W (1991) Action of 2′,2′-difluorodeoxycytidine on DNA synthesis. Cancer Res 51(22):6110–6117PubMed
8.
go back to reference Heinemann V, Xu YZ, Chubb S, Sen A, Hertel LW, Grindey GB, Plunkett W (1992) Cellular elimination of 2′,2′-difluorodeoxycytidine 5′-triphosphate: a mechanism of self-potentiation. Cancer Res 52(3):533–539PubMed Heinemann V, Xu YZ, Chubb S, Sen A, Hertel LW, Grindey GB, Plunkett W (1992) Cellular elimination of 2′,2′-difluorodeoxycytidine 5′-triphosphate: a mechanism of self-potentiation. Cancer Res 52(3):533–539PubMed
9.
go back to reference Shipley LA, Brown TJ, Cornpropst JD, Hamilton M, Daniels WD, Culp HW (2006) Metabolism and disposition of gemcitabine, and oncolytic deoxycytidine analog, in mice, rats, and dogs. Drug Metab Dispos 20(6):849–855 Shipley LA, Brown TJ, Cornpropst JD, Hamilton M, Daniels WD, Culp HW (2006) Metabolism and disposition of gemcitabine, and oncolytic deoxycytidine analog, in mice, rats, and dogs. Drug Metab Dispos 20(6):849–855
10.
go back to reference Plunkett W, Huang P, Searcy CE, Gandhi V (1996) Gemcitabine: preclinical pharmacology and mechanisms of action. Semin Oncol 23(5 Suppl 10):3–15PubMed Plunkett W, Huang P, Searcy CE, Gandhi V (1996) Gemcitabine: preclinical pharmacology and mechanisms of action. Semin Oncol 23(5 Suppl 10):3–15PubMed
11.
go back to reference Banna GL, Collovà E, Gebbia V, Lipari H, Giuffrida P, Cavallaro S, Condorelli R, Buscarino C, Tralongo P, Ferraù F (2010) Anticancer oral therapy: emerging related issues. Cancer Treat Rev 36(8):595–605PubMedCrossRef Banna GL, Collovà E, Gebbia V, Lipari H, Giuffrida P, Cavallaro S, Condorelli R, Buscarino C, Tralongo P, Ferraù F (2010) Anticancer oral therapy: emerging related issues. Cancer Treat Rev 36(8):595–605PubMedCrossRef
12.
go back to reference Kerbel RS, Kamen B (2004) The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer 4(6):423–436PubMedCrossRef Kerbel RS, Kamen B (2004) The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer 4(6):423–436PubMedCrossRef
13.
go back to reference André N, Padovani L, Pasquier E (2011) Metronomic scheduling of anticancer treatment: the next generation of multitarget therapy? Future Oncol 7(3):385–394PubMedCrossRef André N, Padovani L, Pasquier E (2011) Metronomic scheduling of anticancer treatment: the next generation of multitarget therapy? Future Oncol 7(3):385–394PubMedCrossRef
14.
go back to reference Cham KKY, Baker JHE, Takhar KS et al (2010) Metronomic gemcitabine suppresses tumour growth, improves perfusion, and reduces hypoxia in human pancreatic ductal adenocarcinoma. Br J Cancer 103(1):52–60PubMedCrossRef Cham KKY, Baker JHE, Takhar KS et al (2010) Metronomic gemcitabine suppresses tumour growth, improves perfusion, and reduces hypoxia in human pancreatic ductal adenocarcinoma. Br J Cancer 103(1):52–60PubMedCrossRef
15.
go back to reference Laquente B, Lacasa C, Ginestà MM, Casanovas O, Figueras A, Galán M, Ribas IG, Germà JR, Capellà G, Viñals F (2008) Antiangiogenic effect of gemcitabine following metronomic administration in a pancreas cancer model. Mol Cancer Ther 7(3):638–647PubMedCrossRef Laquente B, Lacasa C, Ginestà MM, Casanovas O, Figueras A, Galán M, Ribas IG, Germà JR, Capellà G, Viñals F (2008) Antiangiogenic effect of gemcitabine following metronomic administration in a pancreas cancer model. Mol Cancer Ther 7(3):638–647PubMedCrossRef
16.
go back to reference Veltkamp SA, Jansen RS, Callies S et al (2008) Oral administration of gemcitabine in patients with refractory tumors: a clinical and pharmacologic study. Clin Cancer Res 14(11):3477–3486PubMedCrossRef Veltkamp SA, Jansen RS, Callies S et al (2008) Oral administration of gemcitabine in patients with refractory tumors: a clinical and pharmacologic study. Clin Cancer Res 14(11):3477–3486PubMedCrossRef
17.
go back to reference Bender DM, Bao J, Dantzig AH et al (2009) Synthesis, crystallization, and biological evaluation of an orally active prodrug of gemcitabine. J Med Chem 52(22):6958–6961PubMedCrossRef Bender DM, Bao J, Dantzig AH et al (2009) Synthesis, crystallization, and biological evaluation of an orally active prodrug of gemcitabine. J Med Chem 52(22):6958–6961PubMedCrossRef
18.
go back to reference Koolen SLW, Witteveen PO, Jansen RS et al (2011) Phase I study of Oral gemcitabine prodrug (LY2334737) alone and in combination with erlotinib in patients with advanced solid tumors. Clin Cancer Res 17(18):6071–6082PubMedCrossRef Koolen SLW, Witteveen PO, Jansen RS et al (2011) Phase I study of Oral gemcitabine prodrug (LY2334737) alone and in combination with erlotinib in patients with advanced solid tumors. Clin Cancer Res 17(18):6071–6082PubMedCrossRef
19.
go back to reference Adema AD, Bijnsdorp IV, Sandvold ML, Verheul HM, Peters GJ (2009) Innovations and opportunities to improve conventional (deoxy)nucleoside and fluoropyrimidine analogs in cancer. Curr Med Chem 16(35):4632–4643PubMedCrossRef Adema AD, Bijnsdorp IV, Sandvold ML, Verheul HM, Peters GJ (2009) Innovations and opportunities to improve conventional (deoxy)nucleoside and fluoropyrimidine analogs in cancer. Curr Med Chem 16(35):4632–4643PubMedCrossRef
20.
go back to reference Bergman AM, Adema AD, Balzarini J et al (2011) Antiproliferative activity, mechanism of action and oral antitumor activity of CP-4126, a fatty acid derivative of gemcitabine, in in vitro and in vivo tumor models. Invest New Drugs 29(3):456–466PubMedCrossRef Bergman AM, Adema AD, Balzarini J et al (2011) Antiproliferative activity, mechanism of action and oral antitumor activity of CP-4126, a fatty acid derivative of gemcitabine, in in vitro and in vivo tumor models. Invest New Drugs 29(3):456–466PubMedCrossRef
21.
go back to reference Adema AD, Smid K, Losekoot N, Honeywell RJ, Verheul HM, Myhren F, Sandvold ML, Peters GJ (2012) Metabolism and accumulation of the lipophilic deoxynucleoside analogs elacytarabine and CP-4126. Invest New Drugs 30(5):1908–1916PubMedCrossRef Adema AD, Smid K, Losekoot N, Honeywell RJ, Verheul HM, Myhren F, Sandvold ML, Peters GJ (2012) Metabolism and accumulation of the lipophilic deoxynucleoside analogs elacytarabine and CP-4126. Invest New Drugs 30(5):1908–1916PubMedCrossRef
22.
go back to reference Eisenhauer E, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45(2):228–247PubMedCrossRef Eisenhauer E, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45(2):228–247PubMedCrossRef
23.
go back to reference Lilly E Gemzar 200 mg powder for solution for infusion, Gemzar 1000 mg powder for solution for infusion. Summary of Product Characteristics (SPC) Lilly E Gemzar 200 mg powder for solution for infusion, Gemzar 1000 mg powder for solution for infusion. Summary of Product Characteristics (SPC)
24.
go back to reference Fukami T, Yokoi T (2012) The emerging role of human esterases. Drug Metab Pharmacokinet 27(5):466–477PubMedCrossRef Fukami T, Yokoi T (2012) The emerging role of human esterases. Drug Metab Pharmacokinet 27(5):466–477PubMedCrossRef
25.
go back to reference Abbruzzese JL, Grunewald R, Weeks EA, D G, Adams T, Nowak B, Mineishi S, Tarassoff P, Satterlee W, Raber MN (1991) A phase I clinical, plasma, and cellular pharmacology study of gemcitabine. J Clin Oncol 9(3):491–498PubMed Abbruzzese JL, Grunewald R, Weeks EA, D G, Adams T, Nowak B, Mineishi S, Tarassoff P, Satterlee W, Raber MN (1991) A phase I clinical, plasma, and cellular pharmacology study of gemcitabine. J Clin Oncol 9(3):491–498PubMed
26.
go back to reference Peters GJ, Clavel M, Noordhuis P, Geyssen GJ, Laan AC, Guastalla J, Edzes HT, Vermorken JB (2007) Clinical phase I and pharmacology study of gemcitabine (2′, 2′-difluorodeoxycytidine) administered in a two-weekly schedule. J Chemother 19(2):212–221PubMed Peters GJ, Clavel M, Noordhuis P, Geyssen GJ, Laan AC, Guastalla J, Edzes HT, Vermorken JB (2007) Clinical phase I and pharmacology study of gemcitabine (2′, 2′-difluorodeoxycytidine) administered in a two-weekly schedule. J Chemother 19(2):212–221PubMed
27.
go back to reference Stuurman FE, Lolkema MPJ, Soetekouw PMMB, Rosing H, Mergui-Roelvink M, Beijnen JH, Voest EE, Van Tinteren H, Huitema ADR, Schellens JHM (2012) 581 an adaptive, 2-stage, phase 1 comparative pharmacokinetic and cardiac safety study of two intravenous formulations of CO-101 in patients with advanced solid tumors. Eur J Cancer Supp 48(supp 6):178 Stuurman FE, Lolkema MPJ, Soetekouw PMMB, Rosing H, Mergui-Roelvink M, Beijnen JH, Voest EE, Van Tinteren H, Huitema ADR, Schellens JHM (2012) 581 an adaptive, 2-stage, phase 1 comparative pharmacokinetic and cardiac safety study of two intravenous formulations of CO-101 in patients with advanced solid tumors. Eur J Cancer Supp 48(supp 6):178
28.
go back to reference Veltkamp S, Pluim D, Van Eijndhoven MAJ, Bolijn MJ, Ong FHG, Govindarajan R, Unadkat JD, Beijnen JH, Schellens JHM (2008) New insights into the pharmacology and cytotoxicity of gemcitabine and 2′,2′-difluorodeoxyuridine. Mol Cancer Ther 7(8):2415–2425PubMedCrossRef Veltkamp S, Pluim D, Van Eijndhoven MAJ, Bolijn MJ, Ong FHG, Govindarajan R, Unadkat JD, Beijnen JH, Schellens JHM (2008) New insights into the pharmacology and cytotoxicity of gemcitabine and 2′,2′-difluorodeoxyuridine. Mol Cancer Ther 7(8):2415–2425PubMedCrossRef
Metadata
Title
Phase I study of oral CP-4126, a gemcitabine derivative, in patients with advanced solid tumors
Authors
F. E. Stuurman
E. E. Voest
A. Awada
P. O. Witteveen
T. Bergeland
P.-A. Hals
W. Rasch
J. H. M. Schellens
A. Hendlisz
Publication date
01-08-2013
Publisher
Springer US
Published in
Investigational New Drugs / Issue 4/2013
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-013-9925-z

Other articles of this Issue 4/2013

Investigational New Drugs 4/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine