Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 4/2014

01-04-2014 | Short Communication

The interactions of lenalidomide with human uptake and efflux transporters and UDP-glucuronosyltransferase 1A1: lack of potential for drug–drug interactions

Authors: Zeen Tong, Usha Yerramilli, Sekhar Surapaneni, Gondi Kumar

Published in: Cancer Chemotherapy and Pharmacology | Issue 4/2014

Login to get access

Abstract

Purpose

Lenalidomide is an immunomodulatory agent used for the treatment of myelodysplastic syndromes and multiple myeloma. Renal clearance of lenalidomide is the predominant elimination route and is approximately twofold greater than the glomerular filtration rate (GFR), suggesting the potential contribution of an active secretory mechanism. In vitro studies were conducted to examine whether lenalidomide is a substrate of drug transporters, namely P-glycoprotein (P-gp), human breast cancer resistance protein (BCRP), multidrug resistance proteins (MRP1, MRP2, MRP3), organic anion transporters (OAT1, OAT3), organic cation transporters (OCT1 and OCT2), human organic cation transporter novel 1 and 2 (OCTN1 and OCTN2), multidrug and toxin extrusion (MATE1) and organic anion transporting polypeptide (OATP1B1). Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). In addition, inhibition of UDP-glucuronosyltransferase 1A1 (UGT1A1) variants by lenalidomide was also assessed.

Method

Cells or vesicles expressing each of the human transporters were used for uptake and inhibition studies, with appropriate probe substrates and known inhibitors.

Results

Results of these studies indicate that the lenalidomide is not a substrate for the transporters examined, except that it is weak substrate of P-gp. None of the transporters studied were inhibited by lenalidomide. Lenalidomide is not an inhibitor of UGT1A1*1/*1 or its polymorphic variants UGT1A1*1/*28 and UGT1A1*28/*28.

Conclusions

Drug interactions are unlikely to occur when lenalidomide is co-administered with substrates or inhibitors of these transporters. In addition, lenalidomide is unlikely to cause interactions when co-administered with substrates of UGT1A1.
Literature
1.
go back to reference Zhu YX, Kortuem KM, Stewark AK (2013) Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk Lymphoma 54(4):683–687PubMedCentralPubMedCrossRef Zhu YX, Kortuem KM, Stewark AK (2013) Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk Lymphoma 54(4):683–687PubMedCentralPubMedCrossRef
2.
go back to reference Chen N, Wen L, Lau H, Surapaneni S, Kumar G (2012) Pharmacokinetics, metabolism and excretion of [14C]-lenalidomide following oral administration in healthy male subjects. Cancer Chemother Pharmacol 69:789–797PubMedCentralPubMedCrossRef Chen N, Wen L, Lau H, Surapaneni S, Kumar G (2012) Pharmacokinetics, metabolism and excretion of [14C]-lenalidomide following oral administration in healthy male subjects. Cancer Chemother Pharmacol 69:789–797PubMedCentralPubMedCrossRef
3.
go back to reference Kumar G, Lau H, Laskin OL (2009) Lenalidomide: in vitro evaluation of the metabolism and assessment of cytochrome P450 inhibition and induction. Cancer Chemother Pharmacol 63:1171–1175PubMedCrossRef Kumar G, Lau H, Laskin OL (2009) Lenalidomide: in vitro evaluation of the metabolism and assessment of cytochrome P450 inhibition and induction. Cancer Chemother Pharmacol 63:1171–1175PubMedCrossRef
4.
go back to reference Chen N, Lau H, Kong L, Kumar G, Zeldis JB, Knight R, Laskin OL (2007) Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis. J Clin Pharmacol 47:1466–1475PubMedCrossRef Chen N, Lau H, Kong L, Kumar G, Zeldis JB, Knight R, Laskin OL (2007) Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis. J Clin Pharmacol 47:1466–1475PubMedCrossRef
5.
go back to reference Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, Dahlin A, Evers R, Fischer V, Hillgren KM, Hoffmaster KA, Ishikawa T, Keppler D, Kim RB, Lee CA, Niemi M, JPolli JW, Sugiyama Y, Swaan PW, Ware JA, Wright SH, Yee SW, Zamek-Gliszczynski MJ, Zhang L (2010) Membrane transporters in drug development. Nat Rev Drug Discov 9:215–236PubMedCrossRef Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, Dahlin A, Evers R, Fischer V, Hillgren KM, Hoffmaster KA, Ishikawa T, Keppler D, Kim RB, Lee CA, Niemi M, JPolli JW, Sugiyama Y, Swaan PW, Ware JA, Wright SH, Yee SW, Zamek-Gliszczynski MJ, Zhang L (2010) Membrane transporters in drug development. Nat Rev Drug Discov 9:215–236PubMedCrossRef
8.
go back to reference Zamek-Gliszczynski MJ, Hoffmaster KA, Tweedie DJ, Giacomini KM, Hillgren KM (2012) Highlights from the International Transporter Consortium second workshop. Clin Pharmacol Ther 92:553–556PubMedCrossRef Zamek-Gliszczynski MJ, Hoffmaster KA, Tweedie DJ, Giacomini KM, Hillgren KM (2012) Highlights from the International Transporter Consortium second workshop. Clin Pharmacol Ther 92:553–556PubMedCrossRef
9.
go back to reference Hillgren KM, Keppler D, Zur A, Giacomini KM, Stieger B, Cass CE, Zhang L (2013) Emerging transporters of clinical importance: an update from the international transporter consortium. Clin Pharmacol Ther 94(1):52–63PubMedCrossRef Hillgren KM, Keppler D, Zur A, Giacomini KM, Stieger B, Cass CE, Zhang L (2013) Emerging transporters of clinical importance: an update from the international transporter consortium. Clin Pharmacol Ther 94(1):52–63PubMedCrossRef
10.
go back to reference Simmondsen KA, Kolesar JM (2012) Lenalidomide-induced elavated bilirubin. J Oncol Pharm Pract 18(4):402–405CrossRef Simmondsen KA, Kolesar JM (2012) Lenalidomide-induced elavated bilirubin. J Oncol Pharm Pract 18(4):402–405CrossRef
11.
go back to reference Zhang D, Cui D, Gambardella J, Ma L, Barros A, Wang L, Fu Y, Rahematpura S, Nielsen J, Donegan M, Zhang H, Humphreys WG (2007) Characterization of the UDP glucuronosyltransferase activity of human liver microsomes genotyped for the UGT1A1*28 polymorphism. Drug Metab Dispos 35(12):2270–2280PubMedCrossRef Zhang D, Cui D, Gambardella J, Ma L, Barros A, Wang L, Fu Y, Rahematpura S, Nielsen J, Donegan M, Zhang H, Humphreys WG (2007) Characterization of the UDP glucuronosyltransferase activity of human liver microsomes genotyped for the UGT1A1*28 polymorphism. Drug Metab Dispos 35(12):2270–2280PubMedCrossRef
12.
go back to reference Riedmaier AE, Nies AT, Schaeffeler E, Schwab M (2012) Organic anion transporters and their implications in pharmacotherapy. Pharmacol Rev 64:421–449CrossRef Riedmaier AE, Nies AT, Schaeffeler E, Schwab M (2012) Organic anion transporters and their implications in pharmacotherapy. Pharmacol Rev 64:421–449CrossRef
13.
go back to reference Koepsell H (2013) The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 34:413–435PubMedCrossRef Koepsell H (2013) The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 34:413–435PubMedCrossRef
14.
go back to reference Hofmeister CC, Yang X, Pichiorri F, Chen P, Rozewski DM, Johnson AJ, Lee S, Liu Z, Garr CL, Hade EM, Ji J, Schaaf LJ, Benson DM Jr, Kraut EH, Hicks WJ, Chan KK, Chen CS, Farag SS, Grever MR, Byrd JC, Phelps MA (2011) Phase I Trial of lenalidomide and CCI-779 in patients with relapsed multiple myeloma: evidence for lenalidomide-CCI-779 Interaction via P-glycoprotein. J Clin Oncol 29(25):3427–3434PubMedCentralPubMedCrossRef Hofmeister CC, Yang X, Pichiorri F, Chen P, Rozewski DM, Johnson AJ, Lee S, Liu Z, Garr CL, Hade EM, Ji J, Schaaf LJ, Benson DM Jr, Kraut EH, Hicks WJ, Chan KK, Chen CS, Farag SS, Grever MR, Byrd JC, Phelps MA (2011) Phase I Trial of lenalidomide and CCI-779 in patients with relapsed multiple myeloma: evidence for lenalidomide-CCI-779 Interaction via P-glycoprotein. J Clin Oncol 29(25):3427–3434PubMedCentralPubMedCrossRef
15.
go back to reference Takahashi N, Miura M, Kameoka Y, Abumiya M, Sawada K (2012) Drug interaction between lenalidomide and itraconazole. Am J Hematol 87(3):338–339PubMedCrossRef Takahashi N, Miura M, Kameoka Y, Abumiya M, Sawada K (2012) Drug interaction between lenalidomide and itraconazole. Am J Hematol 87(3):338–339PubMedCrossRef
16.
go back to reference Chen N, Kasserra C, Kumar G, Palmisano M (2012) Evidence does not support clinically significant lenalidomide-CCI-779 interaction via P-glycoprotein. J Clin Oncol 30(3):340–341PubMedCrossRef Chen N, Kasserra C, Kumar G, Palmisano M (2012) Evidence does not support clinically significant lenalidomide-CCI-779 interaction via P-glycoprotein. J Clin Oncol 30(3):340–341PubMedCrossRef
17.
go back to reference Fenner KS, Troutman MD, Kempshall S, Cook JA, Ware JA, Smith DA, Lee CA (2009) Drug–drug interactions mediated through P-glycoprotein: clinical relevance and in vitro-in vivo correlation using digoxin as a probe drug. Clin Pharmacol Ther 85(2):173–181PubMedCrossRef Fenner KS, Troutman MD, Kempshall S, Cook JA, Ware JA, Smith DA, Lee CA (2009) Drug–drug interactions mediated through P-glycoprotein: clinical relevance and in vitro-in vivo correlation using digoxin as a probe drug. Clin Pharmacol Ther 85(2):173–181PubMedCrossRef
18.
go back to reference Lankisch TO, Moebius U, Wehmeier M, Behrens G, Manns MP, Schmidt RE, Strassburg CP (2006) Gilbert’s disease and atazanavir: from phenotype to UDP-glucuronosyltransferase haplotype. Hepatology 44(5):1324–1332PubMedCrossRef Lankisch TO, Moebius U, Wehmeier M, Behrens G, Manns MP, Schmidt RE, Strassburg CP (2006) Gilbert’s disease and atazanavir: from phenotype to UDP-glucuronosyltransferase haplotype. Hepatology 44(5):1324–1332PubMedCrossRef
19.
go back to reference Meza-Junco J, Chu QS, Christensen O, Rajagopalan P, Das S, Stefanyschyn R, Sawyer MB (2009) UGT1A1 polymorphism and hyperbilirubinemia in a patient who received sorafenib. Cancer Chemother Pharmacol 65(1):1–4PubMedCrossRef Meza-Junco J, Chu QS, Christensen O, Rajagopalan P, Das S, Stefanyschyn R, Sawyer MB (2009) UGT1A1 polymorphism and hyperbilirubinemia in a patient who received sorafenib. Cancer Chemother Pharmacol 65(1):1–4PubMedCrossRef
20.
go back to reference Culley CL, Kiang TK, Gilchrist SE, Ensom MH (2013) Effect of the UGT1A1*28 allele on unconjugated hyperbilirubinemia in HIV-positive patients receiving atazanavir: a systematic review. Ann Pharmacother 47(4):561–572PubMedCrossRef Culley CL, Kiang TK, Gilchrist SE, Ensom MH (2013) Effect of the UGT1A1*28 allele on unconjugated hyperbilirubinemia in HIV-positive patients receiving atazanavir: a systematic review. Ann Pharmacother 47(4):561–572PubMedCrossRef
Metadata
Title
The interactions of lenalidomide with human uptake and efflux transporters and UDP-glucuronosyltransferase 1A1: lack of potential for drug–drug interactions
Authors
Zeen Tong
Usha Yerramilli
Sekhar Surapaneni
Gondi Kumar
Publication date
01-04-2014
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 4/2014
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-014-2415-y

Other articles of this Issue 4/2014

Cancer Chemotherapy and Pharmacology 4/2014 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