Role for Drug Transporters in Chemotherapy-Induced Peripheral Neuropathy

Tore B Stage, Shuiying Hu, Alex Sparreboom, Deanna L Kroetz, Tore B Stage, Shuiying Hu, Alex Sparreboom, Deanna L Kroetz

Abstract

Chemotherapy-induced peripheral neuropathy (CIPN) is a common and dose-limiting toxicity to widely used chemotherapeutics. Although the exact molecular mechanism of chemotherapy-induced peripheral neuropathy remains elusive, there is consensus that it is caused by damage to the peripheral nervous system leading to sensory symptoms. Recently developed methodologies have provided evidence of expression of drug transporters in the peripheral nervous system. In this literature review, we explore the role for drug transporters in CIPN. First, we assessed the transport of chemotherapeutics that cause CIPN (taxanes, platins, vincristine, bortezomib, epothilones, and thalidomide). Second, we cross-referenced the transporters implicated in genetic or functional studies with CIPN with their expression in the peripheral nervous system. Several drug transporters are involved in the transport of chemotherapeutics that cause peripheral neuropathy and particularly efflux transporters, such as ABCB1 and ABCC1, are expressed in the peripheral nervous system. Previous literature has linked genetic variants in efflux transporters to higher risk of peripheral neuropathy with the taxanes paclitaxel and docetaxel and the vinca alkaloid vincristine. We propose that this might be due to accumulation of the chemotherapeutics in the peripheral nervous system due to reduced neuronal efflux capacity. Thus, concomitant administration of efflux transporter inhibitors may lead to higher risk of adverse events of drugs that cause CIPN. This might prove valuable in drug development where screening new drugs for neurotoxicity might also require drug transporter consideration. There are ongoing efforts targeting drug transporters in the peripheral nervous system to reduce intraneuronal concentrations of chemotherapeutics that cause CIPN, which might ultimately protect against this dose-limiting adverse event.

Trial registration: ClinicalTrials.gov NCT04205903 NCT04164069.

Conflict of interest statement

A provisional patent application related to chemotherapy‐induced peripheral neuropathy has been filed by University of Southern Denmark with Tore Bjerregaard Stage listed as inventor. All other authors declared no competing interest for this work.

© 2020 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Drug transporters regulate influx and efflux of chemotherapeutics in the peripheral nervous system likely modulating toxicity and peripheral neuropathy.

References

    1. Hershman, D.L. et al. Association between patient reported outcomes and quantitative sensory tests for measuring long‐term neurotoxicity in breast cancer survivors treated with adjuvant paclitaxel chemotherapy. Breast Cancer Res. Treat. 125, 767–774 (2011).
    1. Bennett, B.K. , Park, S.B. , Lin, C.S.‐Y. , Friedlander, M.L. , Kiernan, M.C. & Goldstein, D. Impact of oxaliplatin‐induced neuropathy: a patient perspective. Support Care Cancer 20, 2959–2967 (2012).
    1. Hanai, A. et al. Effects of cryotherapy on objective and subjective symptoms of paclitaxel‐induced neuropathy: prospective self‐controlled trial. J. Natl. Cancer Inst. 110, 141–148 (2018).
    1. Chon, H.J. et al. Docetaxel versus paclitaxel combined with 5‐FU and leucovorin in advanced gastric cancer: combined analysis of two phase II trials. Cancer Res. Treat. 41, 196–204 (2009).
    1. Argyriou, A.A. , Bruna, J. , Marmiroli, P. & Cavaletti, G. Chemotherapy‐induced peripheral neurotoxicity (CIPN): an update. Crit. Rev. Oncol. Hematol. 82, 51–77 (2012).
    1. Cashman, C.R. & Höke, A. Mechanisms of distal axonal degeneration in peripheral neuropathies. Neurosci. Lett. 2, 33–50 (2015).
    1. Flatters, S.J.L. , Dougherty, P.M. & Colvin, L.A. Clinical and preclinical perspectives on chemotherapy‐induced peripheral neuropathy (CIPN): a narrative review. Br. J. Anaesth. 119, 737–749 (2017).
    1. Gréen, H. et al. Pharmacogenetic studies of paclitaxel in the treatment of ovarian cancer. Basic Clin. Pharmacol. Toxicol. 104, 130–137 (2009).
    1. Lagas, J.S. et al. Multidrug resistance protein 2 is an important determinant of paclitaxel pharmacokinetics. Clin. Cancer Res. 12(20 Pt 1), 6125–6132 (2006).
    1. Allen, D.T. & Kiernan, J.A. Permeation of proteins from the blood into peripheral nerves and ganglia. Neuroscience 59, 755–764 (1994) <>.
    1. Filipski, K.K. , Mathijssen, R.H. , Mikkelsen, T.S. , Schinkel, A.H. & Sparreboom, A. Contribution of organic cation transporter 2 (OCT2) to cisplatin‐induced nephrotoxicity. Clin. Pharmacol. Ther. 86, 396–402 (2009).
    1. More, S.S. , Akil, O. , Ianculescu, A.G. , Geier, E.G. , Lustig, L.R. & Giacomini, K.M. Role of the copper transporter, CTR1, in platinum‐induced ototoxicity. J. Neurosci. 30, 9500–9509 (2010).
    1. Huang, K.M. , Hu, S. & Sparreboom, A. Drug transporters and anthracycline‐induced cardiotoxicity. Pharmacogenomics 19, 883–888 (2018).
    1. Kocer, B. , Sucak, G. , Kuruoglu, R. , Aki, Z. , Haznedar, R. & Erdogmus, N.I. Clinical and electrophysiological evaluation of patients with thalidomide‐induced neuropathy. Acta Neurol. Belg. 109, 120–126 (2009).
    1. Fellner, S. et al. Transport of paclitaxel (Taxol) across the blood‐brain barrier in vitro and in vivo. J. Clin. Invest. 110, 1309–1318 (2002).
    1. Brooks, T.A. et al. Taxane‐based reversal agents modulate drug resistance mediated by P‐glycoprotein, multidrug resistance protein, and breast cancer resistance protein. Mol. Cancer Ther. 2, 1195–1205 (2003).
    1. Allen, J.D. , Brinkhuis, R.F. , van Deemter, L. , Wijnholds, J. & Schinkel, A.H. Extensive contribution of the multidrug transporters P‐glycoprotein and Mrp1 to basal drug resistance. Cancer Res. 60, 5761–5766 (2000).
    1. Woo, J.S. , Lee, C.H. , Shim, C.K. & Hwang, S.‐J. Enhanced oral bioavailability of paclitaxel by coadministration of the P‐glycoprotein inhibitor KR30031. Pharm. Res. 20, 24–30 (2003).
    1. Huisman, M.T. , Chhatta, A.A. , van Tellingen, O. , Beijnen, J.H. & Schinkel, A.H. MRP2 (ABCC2) transports taxanes and confers paclitaxel resistance and both processes are stimulated by probenecid. Int. J. Cancer 116, 824–829 (2005).
    1. Hopper‐Borge, E.A. et al. Contribution of Abcc10 (Mrp7) to in vivo paclitaxel resistance as assessed in Abcc10−/− mice. Cancer Res. 71, 3649–3657 (2011).
    1. Hopper‐Borge, E. , Chen, Z.‐S. , Shchaveleva, I. , Belinsky, M.G. & Kruh, G.D. Analysis of the drug resistance profile of multidrug resistance protein 7 (ABCC10): resistance to docetaxel. Cancer Res. 64, 4927–4930 (2004).
    1. Smith, N.F. , Acharya, M.R. , Desai, N. , Figg, W.D. & Sparreboom, A. Identification of OATP1B3 as a high‐affinity hepatocellular transporter of paclitaxel. Cancer Biol. Ther. 4, 815–818 (2005).
    1. Nieuweboer, A.J.M. et al. Influence of drug formulation on OATP1B‐mediated transport of paclitaxel. Cancer Res. 74, 3137–3145 (2014).
    1. Mori, D. et al. Alteration in the plasma concentrations of endogenous OATP1B‐biomarkers in non‐small cell lung cancer patients treated with paclitaxel. Drug Metab. Dispos. 48, 387–394 (2020).
    1. Stage, T.B. , Bergmann, T.K. & Kroetz, D.L. Clinical pharmacokinetics of paclitaxel monotherapy: an updated literature review. Clin. Pharmacokinet. 57, 7–19 (2017).
    1. Kobayashi, Y. , Ohshiro, N. , Sakai, R. , Ohbayashi, M. , Kohyama, N. & Yamamoto, T. Transport mechanism and substrate specificity of human organic anion transporter 2 (hOat2 [SLC22A7]). J. Pharm. Pharmacol. 57, 573–578 (2005).
    1. Sissung, T.M. et al. ABCB1 genetic variation influences the toxicity and clinical outcome of patients with androgen‐independent prostate cancer treated with docetaxel. Clin. Cancer Res. 14, 4543–4549 (2008).
    1. Zhu, Y. et al. Inhibition of ABCB1 expression overcomes acquired docetaxel resistance in prostate cancer. Mol. Cancer Ther. 12, 1829–1836 (2013).
    1. Baker, S.D. et al. Pharmacogenetic pathway analysis of docetaxel elimination. Clin. Pharmacol. Ther. 85, 155–163 (2009).
    1. Lee, H.H. , Leake, B.F. , Teft, W. , Tirona, R.G. , Kim, R.B. & Ho, R.H. Contribution of hepatic organic anion‐transporting polypeptides to docetaxel uptake and clearance. Mol. Cancer Ther. 14, 994–1003 (2015).
    1. de Graan, A.‐J.M. et al. Influence of polymorphic OATP1B‐type carriers on the disposition of docetaxel. Clin. Cancer Res. 18, 4433–4440 (2012).
    1. Iusuf, D. et al. Human OATP1B1, OATP1B3 and OATP1A2 can mediate the in vivo uptake and clearance of docetaxel. Int. J. Cancer 136, 225–233 (2015).
    1. Leblanc, A.F. et al. OATP1B2 deficiency protects against paclitaxel‐induced neurotoxicity. J. Clin. Invest. 128, 816–825 (2018).
    1. Ishida, S. , Lee, J. , Thiele, D.J. & Herskowitz, I. Uptake of the anticancer drug cisplatin mediated by the copper transporter Ctr1 in yeast and mammals. Proc. Natl. Acad. Sci. USA 99, 14298–14302 (2002).
    1. Holzer, A.K. , Manorek, G.H. & Howell, S.B. Contribution of the major copper influx transporter CTR1 to the cellular accumulation of cisplatin, carboplatin, and oxaliplatin. Mol. Pharmacol. 70, 1390–1394 (2006).
    1. Ivy, K.D. & Kaplan, J.H. A re‐evaluation of the role of hCTR1, the human high‐affinity copper transporter, in platinum‐drug entry into human cells. Mol. Pharmacol. 83, 1237–1246 (2013).
    1. Pabla, N. , Murphy, R.F. , Liu, K. & Dong, Z. The copper transporter Ctr1 contributes to cisplatin uptake by renal tubular cells during cisplatin nephrotoxicity. Am. J. Physiol. Renal Physiol. 296, F505–F511 (2009).
    1. Ip, V. , Liu, J.J. , Mercer, J.F.B. & McKeage, M.J. Differential expression of ATP7A, ATP7B and CTR1 in adult rat dorsal root ganglion tissue. Mol. Pain 13, 53 (2010).
    1. Jong, N.N. , Nakanishi, T. , Liu, J.J. , Tamai, I. & McKeage, M.J. Oxaliplatin transport mediated by organic cation/carnitine transporters OCTN1 and OCTN2 in overexpressing human embryonic kidney 293 cells and rat dorsal root ganglion neurons. J. Pharmacol. Exp. Ther. 338, 537–547 (2011).
    1. Tschirka, J. , Kreisor, M. , Betz, J. & Gründemann, D. Substrate selectivity check of the ergothioneine transporter. Drug Metab. Dispos. Biol. Fate Chem. 46, 779–785 (2018).
    1. Fujita, S. , Hirota, T. , Sakiyama, R. , Baba, M. & Ieiri, I. Identification of drug transporters contributing to oxaliplatin‐induced peripheral neuropathy. J. Neurochem. 148, 373–385 (2018).
    1. Nishida, K. , et al. Ergothioneine ameliorates oxaliplatin‐induced peripheral neuropathy in rats. Life Sci. 15, 516–524 (2018).
    1. Burger, H. et al. Differential transport of platinum compounds by the human organic cation transporter hOCT2 (hSLC22A2). Br. J. Pharmacol. 159, 898–908 (2010).
    1. Sprowl, J.A. et al. Oxaliplatin‐induced neurotoxicity is dependent on the organic cation transporter OCT2. Proc. Natl. Acad. Sci. USA 110, 11199–11204 (2013).
    1. Lancaster, C.S. , Sprowl, J.A. , Walker, A.L. , Hu, S. , Gibson, A.A. & Sparreboom, A. Modulation of OATP1B‐type transporter function alters cellular uptake and disposition of platinum chemotherapeutics. Mol. Cancer Ther. 12, 1537–1544 (2013).
    1. Horio, M. et al. Transepithelial transport of drugs by the multidrug transporter in cultured Madin‐Darby canine kidney cell epithelia. J. Biol. Chem. 264, 14880–14884 (1989).
    1. Takara, K. et al. Cytotoxic effects of 27 anticancer drugs in HeLa and MDR1‐overexpressing derivative cell lines. Biol. Pharm. Bull. 25, 771–778 (2002).
    1. Bertrand, Y. , Capdeville, R. , Balduck, N. & Philippe, N. Cyclosporin A used to reverse drug resistance increases vincristine neurotoxicity. Am. J. Hematol. 40, 158–159 (1992).
    1. Johnson, D.R. , Finch, R.A. , Lin, Z.P. , Zeiss, C.J. & Sartorelli, A.C. The pharmacological phenotype of combined multidrug‐resistance mdr1a/1b‐ and mrp1‐deficient mice. Cancer Res. 61, 1469–1476 (2001).
    1. Chen, Z.S. et al. Effect of multidrug resistance‐reversing agents on transporting activity of human canalicular multispecific organic anion transporter. Mol. Pharmacol. 56, 1219–1228 (1999).
    1. Marada, V.V. , Flörl, S. , Kühne, A. , Burckhardt, G. & Hagos, Y. Interaction of human organic anion transporter polypeptides 1B1 and 1B3 with antineoplastic compounds. Eur. J. Med. Chem. 92, 723–731 (2015).
    1. Nicolaï, J. et al. Role of the OATP transporter family and a Benzbromarone‐SensitiveEfflux transporter in the hepatocellular disposition of vincristine. Pharm. Res. 34, 2336–2348 (2017).
    1. O’Connor, R. et al. The interaction of bortezomib with multidrug transporters: implications for therapeutic applications in advanced multiple myeloma and other neoplasias. Cancer Chemother. Pharmacol. 71, 1357–1368 (2013).
    1. Buda, G. et al. Polymorphisms in the multiple drug resistance protein 1 and in P‐glycoprotein 1 are associated with time to event outcomes in patients with advanced multiple myeloma treated with bortezomib and pegylated liposomal doxorubicin. Ann. Hematol. 89, 1133–1140 (2010).
    1. Jandial, D.D. , Farshchi‐Heydari, S. , Larson, C.A. , Elliott, G.I. , Wrasidlo, W.J. & Howell, S.B. Enhanced delivery of cisplatin to intraperitoneal ovarian carcinomas mediated by the effects of bortezomib on the human copper transporter 1. Clin. Cancer Res. 15, 553–560 (2009).
    1. Clemens, J. et al. Cellular uptake kinetics of bortezomib in relation to efficacy in myeloma cells and the influence of drug transporters. Cancer Chemother. Pharmacol. 75, 281–291 (2015).
    1. Shen, H. , Lee, F.Y. & Gan, J. Ixabepilone, a novel microtubule‐targeting agent for breast cancer, is a substrate for P‐glycoprotein (P‐gp/MDR1/ABCB1) but not breast cancer resistance protein (BCRP/ABCG2). J. Pharmacol. Exp. Ther. 337, 423–432 (2011).
    1. Flegel, C. et al. RNA‐seq analysis of human trigeminal and dorsal root ganglia with a focus on chemoreceptors. PLoS One 10, e0128951 (2015) <>.
    1. Ray, P. et al. Comparative transcriptome profiling of the human and mouse dorsal root ganglia: an RNA‐seq‐based resource for pain and sensory neuroscience research. Pain 159, 1325–1345 (2018).
    1. Schwaid, A.G. , Krasowka‐Zoladek, A. , Chi, A. & Cornella‐Taracido, I. Comparison of the rat and human dorsal root ganglion proteome. Sci. Rep. 8, 13469 (2018).
    1. Sparreboom, A. et al. Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P‐glycoprotein in the intestine. Proc. Natl. Acad. Sci. USA 94, 2031–2035 (1997).
    1. Lhommé, C. et al. Phase III study of valspodar (PSC 833) combined with paclitaxel and carboplatin compared with paclitaxel and carboplatin alone in patients with stage IV or suboptimally debulked stage III epithelial ovarian cancer or primary peritoneal cancer. J. Clin. Oncol. 26, 2674–2682 (2008).
    1. Fracasso, P.M. et al. Phase I study of paclitaxel in combination with a multidrug resistance modulator, PSC 833 (Valspodar), in refractory malignancies. J. Clin. Oncol. 18, 1124–1134 (2000).
    1. Fracasso, P.M. et al. Phase II study of paclitaxel and valspodar (PSC 833) in refractory ovarian carcinoma: a gynecologic oncology group study. J. Clin. Oncol. 19, 2975–2982 (2001).
    1. Noda‐Narita, S. et al. Peripheral neuropathy from paclitaxel: risk prediction by serum microRNAs. BMJ Support Palliat Care. .
    1. Stage, T.B. et al. P‐glycoprotein inhibition exacerbates paclitaxel neurotoxicity in neurons and cancer patients. Clin. Pharmacol. Ther. .
    1. Kilari, D. et al. Copper transporter‐CTR1 expression and pathological outcomes in platinum‐treated muscle‐invasive bladder cancer patients. Anticancer Res. 36, 495–501 (2016).
    1. Mani, S. et al. Activation of the steroid and xenobiotic receptor (human pregnane X receptor) by nontaxane microtubule‐stabilizing agents. Clin. Cancer Res. 11, 6359–6369 (2005).
    1. Geick, A. , Eichelbaum, M. & Burk, O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J. Biol. Chem. 276, 14581–14587 (2001).
    1. Feurstein, D. , Kleinteich, J. , Heussner, A.H. , Stemmer, K. & Dietrich, D.R. Investigation of microcystin congener‐dependent uptake into primary murine neurons. Environ. Health Perspect. 118, 1370–1375 (2010).
    1. Abraham, J.E. et al. Replication of genetic polymorphisms reported to be associated with taxane‐related sensory neuropathy in patients with early breast cancer treated with Paclitaxel. Clin. Cancer Res. 20, 2466–2475 (2014).
    1. Kus, T. et al. Polymorphism of CYP3A4 and ABCB1 genes increase the risk of neuropathy in breast cancer patients treated with paclitaxel and docetaxel. OncoTargets Ther. 9, 5073–5080 (2016).
    1. Tanabe, Y. et al. Paclitaxel‐induced sensory peripheral neuropathy is associated with an ABCB1 single nucleotide polymorphism and older age in Japanese. Cancer Chemother. Pharmacol. 79, 1179–1186 (2017).
    1. Custodio, A. et al. Pharmacogenetic predictors of severe peripheral neuropathy in colon cancer patients treated with oxaliplatin‐based adjuvant chemotherapy: a GEMCAD group study. Ann. Oncol. 25, 398–403 (2014).
    1. Broyl, A. et al. Mechanisms of peripheral neuropathy associated with bortezomib and vincristine in patients with newly diagnosed multiple myeloma: a prospective analysis of data from the HOVON‐65/GMMG‐HD4 trial. Lancet Oncol. 11, 1057–1065 (2010).
    1. Lopez‐Lopez, E. et al. Vincristine pharmacokinetics pathway and neurotoxicity during early phases of treatment in pediatric acute lymphoblastic leukemia. Pharmacogenomics 17, 731–741 (2016).
    1. Wright, G.E.B. et al. Pharmacogenomics of vincristine‐induced peripheral neuropathy implicates pharmacokinetic and inherited neuropathy genes. Clin. Pharmacol. Ther. 105, 402–410 (2018).
    1. Ceppi, F. et al. Polymorphisms of the vincristine pathway and response to treatment in children with childhood acute lymphoblastic leukemia. Pharmacogenomics 15, 1105–1116 (2014).

Source: PubMed

3
Abonnere