Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 2/2009

01-07-2009 | Original Article

Enhancement of the in vivo antitumor activity of clofarabine by 1-β-d-[4-thio-arabinofuranosyl]-cytosine

Authors: William B. Parker, Sue C. Shaddix, Karen S. Gilbert, Rodney V. Shepherd, William R. Waud

Published in: Cancer Chemotherapy and Pharmacology | Issue 2/2009

Login to get access

Abstract

Purpose

Clofarabine increases the activation of 1-β-d-arabinofuranosyl cytosine (araC) in tumor cells, and combination of these two drugs has been shown to result in good clinical activity against various hematologic malignancies. 1-β-d-[4-thio-arabinofuranosyl] cytosine (T-araC) is a new cytosine analog that has exhibited excellent activity against a broad spectrum of human solid tumors and leukemia/lymphoma xenografts in mice and is currently being evaluated in patients as a new drug for the treatment of cancer. Since T-araC has a vastly superior preclinical efficacy profile in comparison to araC, we have initiated studies to determine the potential value of clofarabine/T-araC combination therapy.

Methods

In vitro studies have been conducted to determine the effect of clofarabine on the metabolism of T-araC, and in vivo studies have been conducted to determine the effect of the clofarabine/T-araC combination on five human tumor xenografts in mice.

Results

Initial studies with various tumor cells in culture indicated that a 2-h incubation with clofarabine enhanced the metabolism of T-araC 24 h after its removal by threefold in three tumor cell types (HCT-116 colon, K562 leukemia, and RL lymphoma) and by 1.5-fold in two other tumor cell types (MDA-MB-435 breast (melanoma), and HL-60 leukemia). Pretreatment with clofarabine resulted in a slight decrease in metabolism of T-araC in RPMI-8226 myeloma cells (65% of control) and inhibited metabolism of T-araC in CCRF-CEM leukemia cells by 90%. In vivo combination studies were conducted with various human tumor xenografts to determine whether or not the modulations observed in vitro were reflective of the in vivo situation. Clofarabine and T-araC were administered on alternate days for five treatments each (q2dx5) with the administration of T-araC 24 h after each clofarabine treatment. Combination treatment of HCT-116, K562, HL-60, or RL tumors with clofarabine and T-araC resulted in dramatically superior anti-tumor activity than treatment with either agent alone, whereas this combination resulted in antagonism in CCRF-CEM tumors. The in vivo antitumor activity of clofarabine plus T-araC against HCT-116 tumors was much better than the activity seen with clofarabine plus araC.

Conclusions

These studies provide a rationale for clinical trials using this combination in the treatment of acute leukemias as well as solid tumors and suggest that this combination would exhibit greater antitumor activity than that of clofarabine plus araC.
Literature
1.
go back to reference Bonate PL, Arthaud L, Cantrell WR Jr, Stephenson K, Secrist JA III, Weitman S (2006) Discovery and development of clofarabine: a nucleoside analogue for treating cancer. Nat Rev Drug Discov 5:855–863PubMedCrossRef Bonate PL, Arthaud L, Cantrell WR Jr, Stephenson K, Secrist JA III, Weitman S (2006) Discovery and development of clofarabine: a nucleoside analogue for treating cancer. Nat Rev Drug Discov 5:855–863PubMedCrossRef
2.
go back to reference Faderl S, Gandhi V, Keating MJ, Jeha S, Plunkett W, Kantarjian HM (2005) The role of clofarabine in hematologic and solid malignancies—development of a next-generation nucleoside analog. Cancer 103:1985–1995PubMedCrossRef Faderl S, Gandhi V, Keating MJ, Jeha S, Plunkett W, Kantarjian HM (2005) The role of clofarabine in hematologic and solid malignancies—development of a next-generation nucleoside analog. Cancer 103:1985–1995PubMedCrossRef
3.
go back to reference Kantarjian HM, Jeha S, Gandhi V, Wess M, Faderl S (2007) Clofarabine: past, present, and future. Leuk Lymphoma 48:1922–1930PubMedCrossRef Kantarjian HM, Jeha S, Gandhi V, Wess M, Faderl S (2007) Clofarabine: past, present, and future. Leuk Lymphoma 48:1922–1930PubMedCrossRef
4.
go back to reference Cooper T, Ayres M, Nowak B, Gandhi V (2005) Biochemical modulation of cytarabine triphosphate by clofarabine. Cancer Chemother Pharmacol 55:361–368PubMedCrossRef Cooper T, Ayres M, Nowak B, Gandhi V (2005) Biochemical modulation of cytarabine triphosphate by clofarabine. Cancer Chemother Pharmacol 55:361–368PubMedCrossRef
5.
go back to reference Faderl S, Gandhi V, O’Brien S, Bonate P, Cortes J, Estey E, Beran M, Wierda W, Garcia-Manero G, Ferrajoli A, Estrov Z, Giles FJ, Du M, Kwari M, Keating M, Plunkett W, Kantarjian H (2005) Results of a phase 1–2 study of clofarabine in combination with cytarabine (ara-C) in relapsed and refractory acute leukemias. Blood 105:940–947PubMedCrossRef Faderl S, Gandhi V, O’Brien S, Bonate P, Cortes J, Estey E, Beran M, Wierda W, Garcia-Manero G, Ferrajoli A, Estrov Z, Giles FJ, Du M, Kwari M, Keating M, Plunkett W, Kantarjian H (2005) Results of a phase 1–2 study of clofarabine in combination with cytarabine (ara-C) in relapsed and refractory acute leukemias. Blood 105:940–947PubMedCrossRef
6.
go back to reference Faderl S, Verstovsek S, Cortes J, Ravandi F, Beran M, Garcia-Manero G, Ferrajoli A, Estrov Z, O’Brien S, Koller C, Giles FJ, Wierda W, Kwari M, Kantarjian HM (2006) Clofarabine and cytarabine combination as induction therapy for acute myeloid leukemia (AML) in patients 50 years of age or older. Blood 108:45–51PubMedCrossRef Faderl S, Verstovsek S, Cortes J, Ravandi F, Beran M, Garcia-Manero G, Ferrajoli A, Estrov Z, O’Brien S, Koller C, Giles FJ, Wierda W, Kwari M, Kantarjian HM (2006) Clofarabine and cytarabine combination as induction therapy for acute myeloid leukemia (AML) in patients 50 years of age or older. Blood 108:45–51PubMedCrossRef
7.
go back to reference Gidwani P, Ramesh KH, Liu Y, Kolb EA (2008) The combination of clofarabine and cytarabine in pediatric relapsed acute lymphoblastic leukemia: a case report. Chemotherapy 54:120–124PubMedCrossRef Gidwani P, Ramesh KH, Liu Y, Kolb EA (2008) The combination of clofarabine and cytarabine in pediatric relapsed acute lymphoblastic leukemia: a case report. Chemotherapy 54:120–124PubMedCrossRef
8.
go back to reference Tiwari KN, Shortnacy-Fowler AT, Cappellacci L, Parker WB, Waud WR, Montgomery JA, Secrist JA III (2000) Synthesis of 4′-thio-β-d-arabinofuranosyl-cytosine (4′-thio-ara-C) and comparison of its anticancer activity with that of ara-C. Nucleosides Nucleotides Nucleic Acids 19:329–340PubMedCrossRef Tiwari KN, Shortnacy-Fowler AT, Cappellacci L, Parker WB, Waud WR, Montgomery JA, Secrist JA III (2000) Synthesis of 4′-thio-β-d-arabinofuranosyl-cytosine (4′-thio-ara-C) and comparison of its anticancer activity with that of ara-C. Nucleosides Nucleotides Nucleic Acids 19:329–340PubMedCrossRef
9.
go back to reference Waud WR, Gilbert KS, Shepherd RV, Montgomery JA, Secrit JA III (2003) Preclinical antitumor activity of 4′-thio-beta-d-arabinofuranosylcytosine (4′-thio-ara-C). Cancer Chemother Pharmacol 51:422–426PubMed Waud WR, Gilbert KS, Shepherd RV, Montgomery JA, Secrit JA III (2003) Preclinical antitumor activity of 4′-thio-beta-d-arabinofuranosylcytosine (4′-thio-ara-C). Cancer Chemother Pharmacol 51:422–426PubMed
10.
go back to reference Waud WR, Shepherd RV, Gilbert KS, Tiwari KN, Secrist JA III (2004) Precinical antitumor activity of 4′-thio-β-d-arabinofuranosylcytosine (4′-thio-ara-C, OSI-7836) in human leukemia and lymphoma xenograft models. Proc Am Assoc Cancer Res 45:714 Waud WR, Shepherd RV, Gilbert KS, Tiwari KN, Secrist JA III (2004) Precinical antitumor activity of 4′-thio-β-d-arabinofuranosylcytosine (4′-thio-ara-C, OSI-7836) in human leukemia and lymphoma xenograft models. Proc Am Assoc Cancer Res 45:714
11.
go back to reference Parker WB, Shaddix SC, Rose LM, Waud WR, Shewach DS, Tiwari KN, Secrist JA III (2000) Metabolism of 4′-thio-β-d-arabinofuranosylcytosine in CEM cells. Biochem Pharmacol 60:1925–1932PubMedCrossRef Parker WB, Shaddix SC, Rose LM, Waud WR, Shewach DS, Tiwari KN, Secrist JA III (2000) Metabolism of 4′-thio-β-d-arabinofuranosylcytosine in CEM cells. Biochem Pharmacol 60:1925–1932PubMedCrossRef
12.
go back to reference Richardson F, Black C, Richardson K, Franks A, Wells E, Karimi S, Sennello G, Hart K, Meyer D, Emerson D, Brown E, LeRay J, Nilsson C, Tomkinson B, Bendele R (2005) Incorporation of OSI-7836 into DNA of Calu-6 and H460 xenograft tumors. Cancer Chemother Pharmacol 55:213–221PubMedCrossRef Richardson F, Black C, Richardson K, Franks A, Wells E, Karimi S, Sennello G, Hart K, Meyer D, Emerson D, Brown E, LeRay J, Nilsson C, Tomkinson B, Bendele R (2005) Incorporation of OSI-7836 into DNA of Calu-6 and H460 xenograft tumors. Cancer Chemother Pharmacol 55:213–221PubMedCrossRef
13.
go back to reference Clarke ML, Damaraju VL, Zhang J, Mowles D, Tackaberry T, Lang T, Smith KM, Young JD, Tomkinson B, Cass CE (2006) The role of human nucleoside transporters in cellular uptake of 4′-thio-beta-d-arabinofuranosylcytosine and beta-d-arabinosylcytosine. Mol Pharmacol 70:303–310PubMed Clarke ML, Damaraju VL, Zhang J, Mowles D, Tackaberry T, Lang T, Smith KM, Young JD, Tomkinson B, Cass CE (2006) The role of human nucleoside transporters in cellular uptake of 4′-thio-beta-d-arabinofuranosylcytosine and beta-d-arabinosylcytosine. Mol Pharmacol 70:303–310PubMed
14.
go back to reference Richardson KA, Vega TP, Richardson FC, Moore CL, Rohloff JC, Tomkinson B, Bendele RA, Kuchta RD (2004) Polymerization of the triphosphates of AraC, 2′,2′-difluorodeoxycytidine (dFdC) and OSI-7836 (T-araC) by human DNA polymerase alpha and DNA primase. Biochem Pharmacol 68:2337–2346PubMedCrossRef Richardson KA, Vega TP, Richardson FC, Moore CL, Rohloff JC, Tomkinson B, Bendele RA, Kuchta RD (2004) Polymerization of the triphosphates of AraC, 2′,2′-difluorodeoxycytidine (dFdC) and OSI-7836 (T-araC) by human DNA polymerase alpha and DNA primase. Biochem Pharmacol 68:2337–2346PubMedCrossRef
15.
go back to reference Someya H, Shaddix SC, Tiwari KN, Secrist JA III, Parker WB (2003) Phosphorylation of 4′-thio-β-d-arabinofuranosylcytosine and its analogs by human deoxycytidine kinase. J Pharmacol Exp Ther 304:1314–1322PubMedCrossRef Someya H, Shaddix SC, Tiwari KN, Secrist JA III, Parker WB (2003) Phosphorylation of 4′-thio-β-d-arabinofuranosylcytosine and its analogs by human deoxycytidine kinase. J Pharmacol Exp Ther 304:1314–1322PubMedCrossRef
16.
go back to reference Someya H, Waud WR, Parker WB (2006) Long intracellular retention of 4′-thio-arabinofuranosylcytosine 5′-triphosphate as a critical factor for the anti-solid tumor activity of 4′-thio-arabinofuranosylcytosine. Cancer Chemother Pharmacol 57:772–780PubMedCrossRef Someya H, Waud WR, Parker WB (2006) Long intracellular retention of 4′-thio-arabinofuranosylcytosine 5′-triphosphate as a critical factor for the anti-solid tumor activity of 4′-thio-arabinofuranosylcytosine. Cancer Chemother Pharmacol 57:772–780PubMedCrossRef
17.
go back to reference Thottassery JV, Westbrook L, Someya H, Parker WB (2006) c-Abl-independent p73 stabilization during gemcitabine- or 4′-thio-beta-d-arabinofuranosylcytosine-induced apoptosis in wild-type and p53-null colorectal cancer cells. Mol Cancer Ther 5:400–410PubMedCrossRef Thottassery JV, Westbrook L, Someya H, Parker WB (2006) c-Abl-independent p73 stabilization during gemcitabine- or 4′-thio-beta-d-arabinofuranosylcytosine-induced apoptosis in wild-type and p53-null colorectal cancer cells. Mol Cancer Ther 5:400–410PubMedCrossRef
18.
go back to reference Goss G, Siu LL, Gauthier I, Chen EX, Oza AM, Goel R, Maroun J, Powers J, Walsh W, Maclean M, Drolet DW, Rusk J, Seymour LK, Investigational New Drug Program of the National Cancer Institute of Canada Clinical Trials Group (2006) A phase I, first in man study of OSI-7836 in patients with advanced refractory solid tumors: IND.147, a study of the Investigational New Drug Program of the National Cancer Institute of Canada Clinical Trials Group. Cancer Chemother Pharmacol 58:703–710PubMedCrossRef Goss G, Siu LL, Gauthier I, Chen EX, Oza AM, Goel R, Maroun J, Powers J, Walsh W, Maclean M, Drolet DW, Rusk J, Seymour LK, Investigational New Drug Program of the National Cancer Institute of Canada Clinical Trials Group (2006) A phase I, first in man study of OSI-7836 in patients with advanced refractory solid tumors: IND.147, a study of the Investigational New Drug Program of the National Cancer Institute of Canada Clinical Trials Group. Cancer Chemother Pharmacol 58:703–710PubMedCrossRef
19.
go back to reference Lee CP, de Jonge MJ, O’Donnell AE, Schothorst KL, Hanwell J, Chick JB, Brooimans RA, Adams LM, Drolet DW, de Bono JS, Kaye SB, Judson IR, Verweij J (2006) A phase I study of a new nucleoside analogue, OSI-7836, using two administration schedules in patients with advanced solid malignancies. Clin Cancer Res 12:2841–2848PubMedCrossRef Lee CP, de Jonge MJ, O’Donnell AE, Schothorst KL, Hanwell J, Chick JB, Brooimans RA, Adams LM, Drolet DW, de Bono JS, Kaye SB, Judson IR, Verweij J (2006) A phase I study of a new nucleoside analogue, OSI-7836, using two administration schedules in patients with advanced solid malignancies. Clin Cancer Res 12:2841–2848PubMedCrossRef
20.
go back to reference Dykes DJ, Abbott BJ, Mayo JG, Harrison SD Jr, Laster WR, Simpson-Herren L, Griswold DP Jr (1992) Development of human tumor xenograft models for in vivo evaluation of new antitumor drugs. Contrib Oncol Basel Karger 42:1–22 Dykes DJ, Abbott BJ, Mayo JG, Harrison SD Jr, Laster WR, Simpson-Herren L, Griswold DP Jr (1992) Development of human tumor xenograft models for in vivo evaluation of new antitumor drugs. Contrib Oncol Basel Karger 42:1–22
21.
go back to reference Gandhi V, Kantarjian H, Faderl S, Bonate P, Du M, Ayres M, Rios MB, Keating MJ, Plunkett W (2003) Pharmacokinetics and pharmacodynamics of plasma clofarabine and cellular clofarabine triphosphate in patients with acute leukemias. Clin Cancer Res 9:6335–6342PubMed Gandhi V, Kantarjian H, Faderl S, Bonate P, Du M, Ayres M, Rios MB, Keating MJ, Plunkett W (2003) Pharmacokinetics and pharmacodynamics of plasma clofarabine and cellular clofarabine triphosphate in patients with acute leukemias. Clin Cancer Res 9:6335–6342PubMed
22.
go back to reference Waud WR, Schmid SM, Montgomery JA, Secrist JA III (2000) Preclinical antitumor activity of 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl) adenine (Cl–F-ara-A). Nucleosides Nucleotides Nucleic Acids 19:447–460PubMedCrossRef Waud WR, Schmid SM, Montgomery JA, Secrist JA III (2000) Preclinical antitumor activity of 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl) adenine (Cl–F-ara-A). Nucleosides Nucleotides Nucleic Acids 19:447–460PubMedCrossRef
23.
go back to reference Parker WB, Shaddix SC, Chang CH, White EL, Rose LM, Brockman RW, Shortnancy AT, Montgomery JA, Secrist JA III, Bennett LL Jr (1991) Effects of 2-chloro-9-(2-deoxy-2-fluoro-β-d-arabinofuranosyl) adenine on K562 cellular metabolism and the inhibition of human ribonucleotide reductase and DNA polymerases by its 5-triphosphate. Cancer Res 51:2386–2394PubMed Parker WB, Shaddix SC, Chang CH, White EL, Rose LM, Brockman RW, Shortnancy AT, Montgomery JA, Secrist JA III, Bennett LL Jr (1991) Effects of 2-chloro-9-(2-deoxy-2-fluoro-β-d-arabinofuranosyl) adenine on K562 cellular metabolism and the inhibition of human ribonucleotide reductase and DNA polymerases by its 5-triphosphate. Cancer Res 51:2386–2394PubMed
24.
go back to reference Xie C, Plunkett W (1995) Metabolism and actions of 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-adenine in human lymphoblastoid cells. Cancer Res 55:2847–2852PubMed Xie C, Plunkett W (1995) Metabolism and actions of 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl)-adenine in human lymphoblastoid cells. Cancer Res 55:2847–2852PubMed
25.
go back to reference Parker WB, Shaddix SC, Rose LM, Shewach DS, Hertel LW, Secrist JA III, Montgomery JA, Bennett LL Jr (1999) Comparison of the mechanism of cytotoxicity of 2-chloro-9-(2-deoxy-2-fluoro-β-d-arabinofuranosyl) adenine, 2-chloro-9-(2-deoxy-2-fluoro-β-d-ribofuranosyl) adenine, and 2-chloro-9-(2-deoxy-2,2-difluoro-β-d-ribofuranosyl) adenine in CEM cells. Mol Pharmacol 55:515–520PubMed Parker WB, Shaddix SC, Rose LM, Shewach DS, Hertel LW, Secrist JA III, Montgomery JA, Bennett LL Jr (1999) Comparison of the mechanism of cytotoxicity of 2-chloro-9-(2-deoxy-2-fluoro-β-d-arabinofuranosyl) adenine, 2-chloro-9-(2-deoxy-2-fluoro-β-d-ribofuranosyl) adenine, and 2-chloro-9-(2-deoxy-2,2-difluoro-β-d-ribofuranosyl) adenine in CEM cells. Mol Pharmacol 55:515–520PubMed
26.
go back to reference Spasokoukotskaja T, Sasvári-Székely M, Keszler G, Albertioni F, Eriksson S, Staub M (1999) Treatment of normal and malignant cells with nucleoside analogues and etoposide enhances deoxycytidine kinase activity. Eur J Cancer 35:1862–1867PubMedCrossRef Spasokoukotskaja T, Sasvári-Székely M, Keszler G, Albertioni F, Eriksson S, Staub M (1999) Treatment of normal and malignant cells with nucleoside analogues and etoposide enhances deoxycytidine kinase activity. Eur J Cancer 35:1862–1867PubMedCrossRef
27.
go back to reference Csapó Z, Sasvári-Székely M, Spasokoukotskaja T, Talianidis I, Eriksson S, Staub M (2001) Activation of deoxycytidine kinase by inhibition of DNA synthesis in human lymphocytes. Biochem Pharmacol 61:191–197PubMedCrossRef Csapó Z, Sasvári-Székely M, Spasokoukotskaja T, Talianidis I, Eriksson S, Staub M (2001) Activation of deoxycytidine kinase by inhibition of DNA synthesis in human lymphocytes. Biochem Pharmacol 61:191–197PubMedCrossRef
28.
go back to reference Keszler G, Spasokoukotskaja T, Sasvári-Székely M, Eriksson S, Staub M (2006) Deoxycytidine kinase is reversibly phosphorylated in normal human lymphocytes. Nucleosides Nucleotides Nucleic Acids 25:1147–1151PubMedCrossRef Keszler G, Spasokoukotskaja T, Sasvári-Székely M, Eriksson S, Staub M (2006) Deoxycytidine kinase is reversibly phosphorylated in normal human lymphocytes. Nucleosides Nucleotides Nucleic Acids 25:1147–1151PubMedCrossRef
29.
go back to reference Qian M, Wang X, Shanmuganathan K, Chu CK, Gallo JM (1994) Pharmacokinetics of the anticancer agent 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl) adenine in rats. Cancer Chemother Pharmacol 33:484–488PubMed Qian M, Wang X, Shanmuganathan K, Chu CK, Gallo JM (1994) Pharmacokinetics of the anticancer agent 2-chloro-9-(2-deoxy-2-fluoro-beta-d-arabinofuranosyl) adenine in rats. Cancer Chemother Pharmacol 33:484–488PubMed
Metadata
Title
Enhancement of the in vivo antitumor activity of clofarabine by 1-β-d-[4-thio-arabinofuranosyl]-cytosine
Authors
William B. Parker
Sue C. Shaddix
Karen S. Gilbert
Rodney V. Shepherd
William R. Waud
Publication date
01-07-2009
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 2/2009
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-008-0862-z

Other articles of this Issue 2/2009

Cancer Chemotherapy and Pharmacology 2/2009 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