Immunosuppressors and reversion of multidrug-resistance

Cell Mol Life Sci 2001;58:931–59. [12] Honjo Y, Hrycyna CA, Yan QW, et al. ..... J Cancer Res Clin Oncol 1994;120:422–6. [109] Szakacs G, Annereau JP, ...
286KB taille 4 téléchargements 265 vues
Critical Reviews in Oncology/Hematology 56 (2005) 61–70

Immunosuppressors and reversion of multidrug-resistance Nassera Aouali a , Lahcen Eddabra b , J´erˆome Macadr´e b , Hamid Morjani b,∗ a

Roswell Park Cancer Institute, Department of Cancer Genetique, Elm and Carlton Streets, Buffalo, NY 14263, USA b JE2428 Onco-Pharmacologie, UFR Pharmacie, IFR53, 51096 Reims, France Accepted 10 December 2004

Contents 1. 2.

3.

4.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Immunosuppressors and modulation of P-glycoprotein activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. ABC transporters and substrate specificity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. Agents that modulate P-glycoprotein activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. Cyclosporin A and PSC833 modulate P-glycoprotein transport activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Immunosuppressors and ceramide metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. De novo synthesis of ceramide and resistance to apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Ceramide glycosylation and multidrug-resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

61 62 62 63 64 65 65 66 66 67 67 70

Abstract Drug resistance is the major reason for failure of cancer therapy. When one drug elicits a response in tumour cells resulting in resistance to a large variety of chemically unrelated drugs, this is called multidrug-resistance (MDR). ATP-binding cassette (ABC) transporters contribute to drug resistance via ATP-dependent drug efflux. P-glycoprotein (Pgp) encoded by MDR1 gene, confers resistance to certain anticancer agents. The development of agents able to modulate MDR mediated by Pgp and ABC transporters remained a major goal for the past 10 years. Immunosuppressors, cyclosporin A (CSA) in particular, were shown to modulate Pgp activity in laboratory models and entered very early into clinical trials for reversal of MDR. The proof of reversing activity of CSA was found in phase II studies with myeloma and acute leukaemia. In phase III studies, the results were less convincing regarding the response rate, progression-free survival and overall survival were detected in advanced refractory myeloma. The non-immunosuppressive derivative PSC833 was then extensively studied. This compound shows 10-fold higher potency in reversal of MDR mediated by Pgp. Results from clinical trials with this modulator are still emerging and the notable finding was the need to reduce the dose of anticancer agent used in combination with it. Other effects of CSA and PSC833 on MDR have been described. These two molecules have been shown to have an action on the metabolism of ceramide which stands as second messenger of anticancer agents-induced apoptosis. PSC833 stimulates de novo ceramide synthesis and enhances cell death induced by anticancer agents, such as camptothecins and anthracyclines. In addition, ceramide glycosylation and storage in some cell lines have been described to play a crucial role in resistance to anticancer drugs. CSA is able to inhibit ceramide glucosylation and modulate MDR phenotype. The emergence of other modulators with several ABC protein targets like VX710 are of clinical interest in malignancies expressing several efflux pumps. © 2005 Elsevier Ireland Ltd. All rights reserved. Keywords: Immunosuppressors; Reversion; Multidrug-resistance

1. Introduction ∗

Corresponding author. Tel.: +33 3 26 91 35 65; fax: +33 3 26 91 89 26. E-mail address: [email protected] (H. Morjani).

1040-8428/$ – see front matter © 2005 Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.critrevonc.2004.12.010

Drug resistance is the major reason for failure of cancer therapy. When one drug elicits a response in tumour

62

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

cells resulting in resistance to a large variety of chemically unrelated drugs, this is called multidrug-resistance (MDR). ATP-binding cassette (ABC) transporters contribute to drug resistance via ATP-dependent drug efflux. A diverse arrays of drugs have been identified that sensitize multidrug-resistant cells to chemotherapy. The drugs selected for the initial clinical studies were ones already approved for clinical use and modulators, such as verapamil, immunosuppressors (cyclosporin A), tamoxifen and quinidine were the agents most frequently evaluated. Plasma concentrations equivalent to the concentrations necessary to inhibit drug efflux in vitro were difficult or impossible to achieve with these drugs because of toxicity. Early on, it became clear that reversing the resistance of malignancies like renal cell cancer and colorectal cancer would not be possible, despite high levels of expression of ABC transporters in these tumours. It also became clear that modulators block normal excretory function and delay clearance of chemotherapy. The limitation of the potency of the modulators has been addressed by the development of compounds that are less toxic and more effective as inhibitors. These secondgeneration modulators include the cyclosporin analogue PSC833 and novel agents, such as VX710. In this issue, we propose to review ABC protein-mediated multidrugresistance and the interaction of immunosuppressors with especially P-glycoprotein-mediated drug efflux and with ceramide metabolism (an important second messenger of cell death).

2. Immunosuppressors and modulation of P-glycoprotein activity Clinical resistance to anticancer drugs is the major reason for treatment failure. Generally, tumour cells initially respond well to chemotherapeutic agents. However, repeated drug administration often results in the selection of drugresistant cells and hence in incurable relapses. Cancer cells can develop multiple mechanisms to evade drug toxicity [1,2]. When one drug elicits a response in tumour cells resulting in resistance to a large variety of chemically unrelated drugs, this is called multidrug-resistance. Many molecular mechanisms responsible for MDR have been discovered [3]. One group of mechanisms results in a decreased intracellular concentration of the anticancer drug and hence in a reduction of damage inflicted on the intracellular drug target(s). Another group of mechanisms includes decreased or alternatively increased expression of target molecules or activation of target repair mechanism [4,5]. The altered pharmacology of drugs in MDR cells is mediated by over-expression of membrane proteins belonging to ATP-binding cassette transporter family [http://nutrigene.4t.com/humanabc.htm] (Fig. 1) such as Pglycoprotein (Pgp), encoded by gene MDR1. Pgp is an energy-dependent drug efflux protein in the plasma membrane that reduces the intracellular concentration and cyto-

Fig. 1. Structures of Pgp, MRP1 and BCRP proteins. The structures of three categories of ABC transporter. Pgp has 12 transmembrane domains (TM) and two ATP-binding sites (NBD). MRP1 is similar to Pgp in that they possess two NBD. MRP1 also contains an additional domain that is composed of five transmembrane segments at the amino-terminal end (TMD0 ), giving to this protein a total of 17 transmembrane domains. TMD0 is connected to the core by a cytoplasmic linker (L0 ). The “half-transporter” BCRP contains six transmembrane domains and one NBD on the amino-terminal side. This half-transporter is thought to homodimerize or heterodimerize to function.

toxic effects of chemotherapeutic drugs [6]. Other pumps have been discovered and are also responsible for drug efflux. One of these pumps is the so-called multidrug-resistance associated protein (MRP1) [7]. MRP1 shares many substrates with Pgp, but overall has slightly different substrate specificity. Moreover, the mechanism of drug efflux by MRP1 is different from that of Pgp. In fact, MRP1 is involved in the ATP-dependent transport of glutathione-S-conjugates or cotransport of the drug and glutathione [8]. Other MRP1-related proteins have been described [9]. Among these proteins, MRP2, MRP3 and MRP5 seemed to induce resistance to anticancer drugs. More recently, a novel ABC protein has been discovered called breast cancer resistance protein or BCRP [10,11]. It is important to note that these proteins are also expressed in some normal cells such as hepatocytes. 2.1. ABC transporters and substrate specificity The three major MDR proteins are highly promiscuous transporters, they share the ability of recognizing and translocating a large number of structurally diverse, mainly hydrophobic compounds. In addition to their overlapping substrate specificity, each transporter can handle unique compounds. Pgp is a transporter for large hydrophobic, either uncharged or slightly positively charged compounds while the MRP family primarily transports hydrophobic anionic conjugates and extrudes hydrophobic uncharged drugs. The MRPl-related uncharged drug transport is linked to the trans-

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

Fig. 2. ABC transporters-substrate anticancer agents. Abbreviations: TAX, taxotere; TXL, taxol; COL, colchicine; MYT C, mitomycin C; ACT D, actinomycin D; VLB, vinblastine; VCR, vincristine; VP16, etoposide; MX, mitoxantrone; TPT, topotecan; SN38, active metabolite of irinotecan; MTX, methotrexate; ATH, (anthracyclines: doxorubicin, daunorucin, epirubicin); CARM, carmustine; CHLB, chlorambucil; LEUC, leucovorine and Cis-Pt, Cis-platin.

port or allosteric effect of cellular-free reduced glutathione [8]. The exact spectrum of the BCRP (MXR) transported substrates has not yet been explored in detail and these studies are complicated by the variable substrate-mutants of BCRP observed in the most recent studies [12]. Some of the key molecules are presented in Fig. 2 and are unfortunately also MDR substrates for the patients. Drug resistance was first documented experimentally in mouse leukaemia cells that acquired resistance to 4-amino-N10 -methyl-pteroylglutamic acid [13]. In 1973, Dano discovered active outward transport of daunorubicin by drug-resistant cells that were cross-resistant to other chemotherapeutic agents, such as vinca-alkaloids (vincristine and vinblastine) and other anthracyclines (doxorubicin) [14]. Other authors studying MDR phenotype noted a constant over-expression of a 170 kDa membrane protein termed P-glycoprotein [6]. The gene encoding Pgp was cloned and identified as MDR1 [15]. Riordan and Ling purified also this protein from plasma membrane vesicles of Chinese hamster ovary cell mutants with reduced colchicine permeability [16]. More later, taxotere and taxol, other molecule from the cytoskeleton poisons group, have been identified as exclusive substrates of Pgp [17,18] (Fig. 2). 2.2. Agents that modulate P-glycoprotein activity Fig. 3 shows the MDR modulators used experimentally or in clinical trials. As early as 1987, results from the first clinical trial of MDR (mediated by Pgp) reversal

63

Fig. 3. ABC transporters modulators. Abbreviations: VLP, verapamil; CSA, cyclosporin A; PRO, probenecid; SULF, sulfinpyrazone and FTC, fumitremorgine C.

in ovarian cancer with verapamil (VPL) in combination with doxorubicin (DOX) were published [19]. Because of significant cardiac toxicity, the study was discontinued. In myeloma and non-Hodgkin’s lymphoma, VPL was clearly shown to be active on resistance to chemotherapy protocols with anthracyclines and/or vinkaalcaloids [20,21]. Quinidine entered very early in a randomized trial in breast cancer, aimed at reversing anthracycline resistance [22]. Quinine is much less toxic than quinidine and can be used at higher doses. In a phase II study, Solary et al. showed that its association with mitoxantrone and cytarabine could improve the response rate of acute leukaemias with poor prognosis [23]. Bennis et al. showed that quinine probably modulates resistance without any increase in intracellular accumulation of DOX and concluded that quinine was able to alter nucleocytoplasmic distribution since the target of the anthracycline is the nucleus [24]. More later, Belhoussine et al. showed that modulation of resistance by quinine was not accompanied by an increase in nuclear accumulation of pirarubicin [25]. Biricodar (VX710), from the third-generation drugs, has been shown to reverse MDR in vitro and in vivo by acting on both Pgp and MRP1 (Fig. 3) [26,27]. This modulator was studied in two phase I trials in combination with DOX [28]. S9788 has a selective and high effect on Pgp transport activity in vitro and in vivo [29] but has not been further developed [30]. Elacridar (GG918) is active on Pgp and another ABC protein BCRP (MXR) (Figs. 1 and 3). This compound is not active on MRP1. Zosuquidar (LY335979) is also an active inhibitor of Pgp in vitro and in vivo [31]. Phase I results were recently reported showing some risk of neurotoxicity at high dosage and no pharmacokinetic interaction with doxorubicin

64

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

[32]. For more details, a large panel of Pgp inhibitors are discussed in a review by Robert and Jarry [33]. 2.3. Cyclosporin A and PSC833 modulate P-glycoprotein transport activity Cyclosporin A (CSA) and PSC833 (Fig. 4) belong to the group of MDR modulators that are able to inhibit the Pgp associated ATPase activity. The best characterized is PSC833, which at nanomolar concentrations inhibits the ATPase and transport function of Pgp. Concerning the transport of CSA and PSC833, data are not clear. Several works favor the view that CSA and PSC833 are transport substrates for Pgp [34–36] and other data do not [37]. The last report considers that PSC833 is indeed a substrate of Pgp, but a slow one. That is, when PSC833 competes with a substrate, it will win out due to its higher affinity (a larger interaction surface) [38]. But since its transport rate is slower, it will slow down the turnover rate, which will then be reflected in the decrease in ATPase activity. So, PSC833 seems to be a partial antagonist, since it does not completely block Pgp function, but just slows it down due to the bulkiness of this molecule acting as an “obstructive” substrate, slowing down the Pgp machinery. Studies performed in vitro on samples from leukaemia patients showed that CSA and PSC833 were appropriate inhibitors that can be used for MDR diagnosis. In fact,

Fig. 4. Structures of cyclosporin A and PSC833.

Merlin et al. have shown that in the Pgp-positive samples, a significant increase in cellular daunorubicin accumulation is observed in the presence of CSA [39]. More later, the same group demonstrated the influence of PSC833 on daunorubicin intracellular accumulation in bone marrow specimens from patients with acute myeloid leukaemia [40]. Legrand et al. have demonstrated also that there was a good correlation between Pgp expression and the in vitro modulatory effect of CSA on calcein-AM uptake in sample patients with acute myeloid leukaemia [41]. A simultaneous functional study of calcein-AM uptake and calcein efflux in the presence of, respectively, CSA and probenecid (a specific inhibitor of MRP1, Fig. 3) have shown that it was possible to discriminate the Pgp and MRP1 transport activities [42]. CSA entered early into trials of reversal of multidrugresistance. The proof of reversing activity of CSA was found in phase II studies with myeloma [43] and acute leukaemia [44]. Phase III studies were conducted in haematological malignancies and no effect of CSA on the overall response rate and progression-free survival in myeloma patients [45], whereas among several studies, only one showed a positive effect of CSA in acute myeloblastic leukaemia [46]. A widely tested second-generation compound is PSC833 (valsopodar), a derivative of cyclosporin D that is 10 times more potent than CSA [47]. PSC833 was the first molecule from cyclosporins group without immunosuppressive properties [48] and showed modulation of MDR in vivo with a lower renal toxicity when compared with CSA [49]. During phase I studies, an important effect of this compound on the pharmacokinetics of the associated drugs was shown, the anticancer drugs being etoposide (VP16) [50], DOX [51], mitoxantrone (MIT) [52] and taxol [53]. In most cases, either a doubling of the time-plasma concentration area under the curve or an important increase in elimination half-life was found. Therefore, it was not possible to separate the pharmacokinetic effects of PSC833 from its pharmacodynamic effects. Those results were in general disappointing, particularly in acute myeloblastic leukaemia trials and then suggested that it was he needed to reduce the dose of anticancer agent used in combination with it and dose reduction ranged from 25% for etoposide to 66% for taxol [54,55]. Those dose reductions were required to prevent toxicities of the anticancer agent in combined therapy and have compromised drug concentration in the tumour even with complete inhibition of Pgp. One treatment that has not been fully explored is that of prevention of the emergence of resistance through the use of Pgp inhibitors. In the laboratory, PSC833 reduced the mutation rate for DOX-selected resistance in sarcoma cells by 10-fold, thus reducing the development of resistant clones via the MDR mechanism. In those sarcoma cells, treated with PSC833, resistance was mediated by an alternative pathway with reduced expression of topoisom´erase II␣, the target enzyme for anthracyclines [56]. Another study examined six agents for their ability to prevent vincristine resistance in a rhabdomyosarcoma cell line [57]. MDR modulators and

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

particularly PSC833 prevented the development of resistance, suggesting the role of the use of Pgp inhibitors prior to cytotoxic therapy.

3. Immunosuppressors and ceramide metabolism 3.1. De novo synthesis of ceramide and resistance to apoptosis Sphingolipid are a large family of lipids that are implicated in signal transduction process and ceramide is the most studied (Fig. 5) [58–60]. Ceramide synthesis results from sphingomyelin hydrolysis [58] or by de novo synthesis (Fig. 5). De novo synthesis of ceramide is initiated at the cytosolic surface of the endoplasmic reticulum (RE) membrane by condensation of l-serine and palmitoyl coenzyme A [61,62]. This NADPH-dependent reaction gives 3-ketosphinganine [63], which is reduced to dihydrosphingosine. The next step is an acylation reaction. The enzyme acyltransferase transfers a long-chain fatty acid to the amino group of the molecule and generates dihydroceramide. The

65

introduction of a trans-4,5 double bond by a desaturase converts the dihydroceramide to ceramide (Fig. 5) [64]. A number of cytotoxic agents have been shown to activate de novo synthesis of ceramide. Daunorubicine, an inhibitor of topoisomerase II from anthracycline family is able to promote ceramide formation and apoptosis by ceramide synthase activation in P388 and U937 cells [65] and in human myeloid leukaemia cells [66]. Vinca-alkaloids which target tubulin polymerisation increases cellular ceramide. In fact, vincristine treatment of ALL-697 leukaemia cells induces apoptosis after an increase in ceramide level [67]. In CCRFCEM leukaemia cells, vincristine treatment induces apoptosis and an elevation of ceramide [68]. Similar data have been obtained in human epidermoid carcinoma cells KB-344 cells [69]. Taxol, which is able to inhibit the microtubules depolymerisation, induces apoptosis in a number of cell lines including leukaemia and breast cancer. It induces ceramide production and acts synergistically with administration of exogenous ceramide [70,71]. Etoposide, which inhibits topoisomerase II, also induces an increase in ceramide level [72]. Etoposide is able to increase ceramide level via activation of serine palmitoyl-

Fig. 5. Ceramide metabolism pathways. PSC833 and cyclosporin A are able to inhibit de novo synthesis and glucosylation of ceramide, respectively.

66

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

transferase [73,74]. Campthotecin, a topoisomerase I inhibitor, increases the ceramide level in 4B1 mouse fibroblasts [75] and HT-29 colon cancer cells [76]. In LNCaP prostate cancer cells, camptothecin treatment induces an elevation of ceramide and apoptosis [77]. The synthetic retinoids are also to induce an increase in cellular ceramide level and apoptosis. In PCC7-Mzl stem cells, retinoic acid is able increase ceramide level by stimulating the ceramide de novo synthesis [78]. N-(4Hydroxyphenyl)retinamide (4-HPR) induces an elevation of ceramide and apoptosis in leukaemia cells [79]. In LNCaP prostate cancer cells, the effect of 4-HPR on ceramide synthesis was shown to be through activation of serine palmitoyltransferase [80,81]. Gemcitabine is able to activate the de novo ceramide synthesis of ceramide in A549 cells [82]. Moreover, it has been shown that ∆ (9)-tetrahydrocannabinol (THC) and other cannabinoids induce ceramide elevation and apoptosis in cancer cells by activating the serine palmitoyltransferase in glioma cells [83,84]. PSC833, in addition, at this action on Pgp is able to increase the ceramide level by a Pgp-independent mechanism [85,86]. In fact, PSC833 induces a ceramide generation associated with a decrease in cell survival in drug-resistant MCF7 cells [85]. Moreover, in KB-V-1 human epidermoid carcinoma cells, PSC833 activates ceramide synthesis and increases vinblastine sensitivity [69]. The PSC833 effect on ceramide generation have been also demonstrated in human ovarian carcinoma cells SKOV-3 [87]. Moreover, cells whose ceramide glycosylation is enhanced, are resistant to PSC833 effect [88]. This can be explained by the fact that ceramide generated by PSC833 is converted to glucosylceramide [89]. Co-treatment with PSC833 and fumonisin B1 (a ceramide synthase inhibitor) indicates that PSC833 activates the de novo synthesis of ceramide in cancer cells [69,85,88]. Recently, a study on MDA-MB 468 breast cancer cells has shown that generation of ceramide by PCS833 results from activation of serine palmitoyltransferase enzyme [90]. In addition, PSC833 is able to restore an sphingomyelinceramide pathway stimulation. In fact, the amount of hydrolysable sphingomyelin is another candidate for regulating ceramide production and sphingomyelin is distributed on both inner and outer leaflets of the plasma membrane [91]. Bezombes et al. have reported that the KGla cells, which are inherently resistant to TNF␣ and do not produce ceramide upon cytokine stimulation, can be sensitized by the use PSC833 [92]. The authors suggested that resistance to TNF␣-mediated apoptosis of these cells is linked to the disposability of the sphingomyelin pool, and a role for Pglycoprotein in sphingomyelin transverse plasma membrane asymmetry which can be affected in this case by PCS833. 3.2. Ceramide glycosylation and multidrug-resistance Once generated, ceramide can accumulate in the cell or may be converted into a variety of metabolites. In fact,

ceramide is a precursor of both glycosphingolipids and sphingomyelin (Fig. 5). The transfer of phosphocholine group of phosphatidylcholine to ceramide generates sphingomyelin. The glucosylceramide (GlcCer) synthesis results from transfer of glucose of UDP-glucose to ceramide by a glucosyltransferase (Fig. 5). The GlcCer is a metabolic precursor of lactosylceramide (LacCer) and gangliosides. Early reports already indicate that the lipid composition of drug-resistant cells is different from that of sensitive cells [93]. Lavie et al. have shown that MDR cells, which overexpress Pgp, display an elevation of GlcCer when compared to the drug-sensitive counterparts [94–96]; Kok et al. have reported that colchicine-resistant colon carcinoma cells overexpressing MRP1 have elevated GlcCer level [97]. It has then been suggested to consider GlcCer level as a diagnosis marker for drug resistance in tumours [98]. The increase in GlcCer level has been explained by a higher activity of GlcCer synthase (GCS) [95] and/or uncoupling of GlcCer conversion to LacCer [99]. Several arguments are in favor of a role for GCS in MDR, including the observation that over-expression of GCS confers resistance to chemotherapy [95,100] and on the other hand, transfection of multidrug-resistant cells with a GCS antisense oligonucleotide attenuates resistance [101]. However, a recent study indicates that the absence of functional GCS does not sensitize melanoma cells to anticancer drugs [102]. The inhibition of ceramide glycosylation increases sensitivity of MDR cells to chemotherapeutic agents [69]. Moreover, the use of specific glucosylceramide synthase inhibitors like 1-phenyl-2-palmitoylamino-3-morpholino-1propanolol (PPMP) decreases the GlcCer level in resistant cells and partially restores sensitivity to chemotherapeutic agents [96,103–105]. Tamoxifen, that is used in first for treatment of estrogen receptor positive breast cancer [106], is able to inhibit the GlcCer synthesis in MCF7/ADR which are oestrogen receptor negative and hence modulate their resistance to chemotherapy [107,103]. This has been also shown in other cell lines [96,108]. However, this mechanism is not well known [103]. Cyclosporin A is able to block the glucosylation of ceramide. In fact, a study on MCF7/ADR cells has shown that cyclosporin A is a potent inhibitor of glucosylceramide synthase [88]. The effect of cyclosporin A on ceramide glycosylation is accompanied by a circumvention of MDR phenotype [103]. The same data have been reported by Morjani et al. [96]. However, the mechanism by which cyclosporin A inhibits ceramide glycosylation is inhibited is not clear.

4. Conclusion A number of lessons have been learned from the evolution of the field of multidrug-resistance. A very nice and recent study have well demonstrated that prediction of drug sensitivity and resistance can be accessed by profiling ABC transporter genes in cancer cells [109]. However, the reversal

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

of multidrug-resistance has not yet reached the level of routine clinical applications. The future of the potential therapeutic area remains uncertain. This is not for lack of molecules, since hundreds of compounds have been selected or designed with comprehensive studies on structure–activity relationships in several chemical families. Rather, the reason for this failure originates from the inadequate design of clinical trials. The pharmacological and toxicological properties of multidrug-resistance modulators should have been taken into consideration with those of anticancer drugs in terms of the benefit/risk ratio of combination. The pharmacokinetic interaction of cyclosporin A or PSC833 with cytotoxic drugs has considerably rendered difficult the clinical development of these drugs as multidrug-resistance modulators. In fact, cyclosporin A and PSC833 appear to be the only ones to present this interaction that might well be related to the specific inhibition of another ABC protein that P-glycoprotein involved in the biliary elimination of drugs. Another property of PSC833 has been reported in a recent work by Lehne et al. who have shown in vivo that a simple inhibition of P-glycoprotein by PSC833 may lead to direct elimination of multidrug-resistant cells due to the fact that this membrane protein is involved in malignancy as well as in drug resistance [110]. Concerning ABC transporters, one emerging idea is the use of inhibitors in enhancing the oral bioavailability of anticancer drugs. Reviewers Dr. Marc Diederich, Fondation Recherche sur le Cancer et les Maladies du Sang, Laboratoire de Biologie Mol´eculaire et Cellulaire, du Cancer (LBMCC), Hˆopital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg. Prof. Jean-Louis Merlin, Laboratoire de Recherche en Oncologie, Centre Alexis Vautrin, Av. de Bourgogne, F54511 Vandoeuvre-Les Nancy Cedex, France. Prof. Xavier Ronot, Directeur du Laboratoire de Dynamique Cellulaire de l’EPHE, UMR CNRS 5525, In3S, Universit´e Joseph Fourier, F-38706 La Tronche Cedex, France.

References [1] Gottesman MM. How cancer cells evade chemotherapy: sixteenth Richard and Hinda Rosenthal Foundation Award Lecture. Cancer Res 1993;53:747–54. [2] Volm M, Kastel M, Mattern J, Efferth T. Expression of resistance factors (P-glycoprotein, glutathione S-transferase-␲, and topoisomerase II) and their interrelationship to proto-oncogene products in renal cell carcinomas. Cancer 1993;71:3981–7. [3] Nooter K, Stoter G. Molecular mechanisms of multidrug resistance in cancer chemotherapy. Pathol Res Pract 1996;192:768–80. [4] Friche E, Danks MK, Schmidt CA, Beck WT. Decreased DNA topoisomerase II in daunorubicin-resistant Ehrlich ascites tumor cells. Cancer Res 1991;51:4213–8. [5] Tiirikainen MI, Elonen E, Syrjala MT, Jansson SE, Krusius T. Flow cytometric analysis of glutathione-S-transferase-pi in acute leukemia. Leukemia 1994;8:978–84.

67

[6] Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biophys Biochim Acta 1976;455:152–62. [7] Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 1992;258:1650–4. [8] Leier I, Jedlitschky G, Buchholz U, Cole SPC, Deeley RG, Keppler D. The MRP gene encodes an ATP-dependent export pump for leukotriene C4 and structurally related conjugates. J Biol Chem 1994;269:27807–10. [9] Borst P, Evers R, Kool M, Wijnholds J. The multidrug resistance protein family. Biochim Biophys Acta 1999;1461:347–57. [10] Doyle LA, Yang W, Abruzzo LV, et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci USA 1998;95:15665–70. [11] Litman T, Druley TE, Stein WD, Bates SE. From MDR to MXR: new understanding of multidrug resistance systems, their properties and clinical significance. Cell Mol Life Sci 2001;58:931–59. [12] Honjo Y, Hrycyna CA, Yan QW, et al. Acquired mutations in the MXR/BCRP/ABCP gene alter substrate specificity in MXR/BCRP/ABCP-overexpressing cells. Cancer Res 2001;61:6635–9. [13] Burchenal JH, Robinson E, Johnston SF, et al. The induction of resistance to 4-amino-N10-methyl-pteroylglutaminic acid in a strain of transmitted mouse leukemia. Science 1950;111:116–7. [14] Dano K. Active outward transport of daunomycin in resistant Ehrlich ascites tumor cells. Biochim Biophys Acta 1973;323:466–83. [15] Ueda K, Clark DP, Chen CJ, et al. The human multidrug resistance (mdr1) gene. cDNA cloning and transcription initiation. J Biol Chem 1987;262:505–8. [16] Riordan JR, Ling V. Purification of P-glycoprotein from plasma membrane vesicles of Chinese hamster ovary cell mutants with reduced colchicine permeability. J Biol Chem 1979;254:12701–5. [17] Riou JF, Naudin A, Lavelle F. Effects of Taxotere on rnurine and human tumor cell lines. Biochem Biophys Res Commun 1992;87(August (1)):164–70. [18] Greenberger LM, Lothstein L, Williams SS, Horwitz SB. Distinct P-glycoprotein precursors are overproduced in independently isolated drug-resistant cell lines. Proc Natl Acad Sci USA 1988;85:3762–6. [19] Ozols RF, Cunnion RE, Klecker Jr RW, et al. Verapamil and adriamycin in the treatment of drug-resistant ovarian cancer patients. J Clin Oncol 1987;5:641–7. [20] Dalton WS, Grogan TM, Meltzer PS, et al. Drug-resistance in multiple myeloma and non-Hodgkin’s lymphoma: detection of Pglycoprotein and potential circumvention by addition of verapamil to chemotherapy. J Clin Oncol 1989;7:1578–9. [21] Salmon SE, Dalton WS, Grogan TM, et al. Multidrug-resistant myeloma: laboratory and clinical effects of verapamil as a chemosensitizer. Blood 1991;78:44–50. [22] Wishart GC, Bissett D, Paul J, et al. Quinidine as a resistance modulator of epirubicin in advanced breast cancer: mature results of a placebo-controlled randomized trial. J Clin Oncol 1994;12:1771–7. [23] Solary E, Witz B, Caillot D, et al. Combination of quinine as a potential reversing agent with mitoxantrone and cytarabine for the treatment of acute leukemias: a randomized multicenter study. Blood 1996;88(August (4)):1198–205. [24] Bennis S, Ichas F, Robert J. Differential effects of verapamil and quinine on the reversal of doxorubicin resistance in a human leukemia cell line. Int J Cancer 1995;62:283–90. [25] Belhoussine R, Morjani H, Lahlil R, Manfait M. Evidence for reversal of multidrug resistance by quinine in LR73 cells without alteration of nuclear pirarubicin uptake and down-regulation of mdrl gene, expression. Int J Cancer 1997;73:600–6. [26] Germann UA, Shlyakhter D, Mason VS, et al. Cellular and biochemical characterization of VX-710 as a chemosensitizer: reversal

68

[27]

[28]

[29]

[30]

[31]

[32]

[33] [34]

[35]

[36]

[37] [38]

[39]

[40]

[41]

[42]

[43]

[44]

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70 of P-glycoprotein-mediated multidrug resistance in vitro. Anticancer Drugs 1997;8:125–40. Germann UA, Ford PJ, Shlyakhter D, Mason VS, Harding MW. Chemosensitization and drug accumulation effects of VX710, verapamil, cyclosporin A, MS-209 and GF120918 in multidrug resistant HL60/ADR cells expressing the multidrug resistance-associated protein MRP. Anticancer Drugs 1997;8:141– 55. Peck RA, Hewett J, Harding MW, et al. Phase I and pharmacokinetic study of the novel MDR1 and MRP1 inhibitor biricodar administered alone and in combination with doxorubicin. J Clin Oncol 2001;19:3130–41. Pierre A, Dunn TA, Kraus-Berthier L, et al. In vitro and in vivo circumvention of multidrug resistance by Sender 9788, a novel triazinoaminopiperidine derivative. Invest New Drugs 1992;10: 137–48. Punt CJ, Voest EE, Tueni E, et al. Phase IB study of doxorubicin in combination with the multidrug resistance reversing agent S9788 in advanced colorectal and renal cell cancer. Br J Cancer 1997;76:1376–81. Dantzig AH, Law KL, Cao J, Starling JJ. Reversal of multidrug resistance by the P-glycoprotein modulator, LY335979, from the bench to the clinic. Curr Med Chem 2001;8:39–50. Rubin EH, de Alwis DP, Pouliquen I, et al. A phase I trial of a potent P-glycoprotein inhibitor, Zosuquidar.3HCl trihydrochloride (LY335979), administered orally in combination with doxorubicin in patients with advanced malignancies. Clin Cancer Res 2002;8:3710–7. Robert J, Jarry C. Multidrug resistance reversal agents. J Med Chem 2003;46:4805–17. Saeki T, Ueda K, Tanigawara Y, Hori R, Komano T. Human Pglycoprotein transports cyclosporin A and FK506. J Biol Chem 1993;268:6077–80. Shirai A, Naito M, Tatsuta T, et al. Transport of cyclosporin A across the brain capillary endothelial cell monolayer by Pglycoprotein. Biochim Biophys Acta 1994;1222:400–4. Schramm U, Fricker G, Wenger R, Miller DS. P-glycoproteinmediated secretion of a fluorescent cyclosporin analogue by teleost renal proximal tubules. Am J Physiol 1995;268:F46–52. Stein WD. Kinetics of the multidrug transporter (P-glycoprotein) and its reversal. Physiol Rev 1997;77:545–90. Litman T, Zeuthen T, Skovsgaard T, Stein WD. Structure–activity relationships of P-glycoprotein interacting drugs: kinetic characterization of their effects on ATPase activity. Biochim Biophys Acta 1997;1361:159–68. Merlin JL, Guerci A, Marchal S, et al. Comparative evaluation of S9788, verapamil, and cyclosporine A in K562 human leukemia cell lines and in P-glycoprotein-expressing samples from patients with hematologic malignancies. Blood 1994;84:262–9. Merlin JL, Guerci AP, Marchal S, et al. Influence of SDZ-PSC833 on daunorubicin intracellular accumulation in bone marrow specimens from patients with acute myeloid leukaemia. Br J Haematol 1998;103:480–7. Legrand O, Simonin G, Perrot JY, Zittoun R, Marie JP. Pgp and MRP activities using calcein-AM are prognostic factors in adult acute myeloid leukemia patients. Blood 1998;91:4480–8. Legrand O, Simonin G, Beauchamp-Nicoud A, Zittoun R, Marie JP. Simultaneous activity of MRP1 and Pgp is correlated with in vitro resistance to daunorubicin and with in vivo resistance in adult acute myeloid leukemia. Blood 1998;94:1046–56. Sonneveld P, Durie BG, Lokhorst HM, et al. Modulation of multidrug-resistant multiple myeloma by cyclosporine. The Leukaemia Group of the EORTC and the HO VON. Lancet 1992;340:255–9. List AF, Spier C, Greer J, et al. Phase I/II trial of cyclosporine as a chemotherapy-resistance modifier in acute leukemia. J Clin Oncol 1993;11:1652–60.

[45] Sonneveld P, Suciu S, Weijermans P, et al. Cyclosporin A combined with vincristine, doxorubicin and dexamethasone (VAD)compared with VAD alone in patients with advanced refractory multiple myeloma: an EORTC-HOVON randomized phase HI study (06914). Br J Haematol 2001;115:895–902. [46] List AF, Kopecky KJ, Willman CL, et al. Benefit of cyclosporine modulation of drug resistance in patients with poor-risk acute myeloid leukemia: a Southwest Oncology Group study. Blood 2001;98:3212–20. [47] Twentyman PR. Cyclosporins as drug resistance modifiers. Biochem Pharmacol 1992;43:109–17. [48] Twentyman PR, Bleehen NM. Resistance modification by PSC833, a novel non-immunosuppressive cyclosporin. Eur J Cancer 1991;27:1639–42. [49] Boesch D, Gaveriaux C, Jachez B, Pourtier-Manzanedo A, Bollinger P, Loor F. In vivo circumvention of P-glycoproteinmediated multidrug resistance of tumor cells with SDZ PSC 833. Cancer Res 1991;51:4226–33. [50] Boote DJ, Dennis IF, Twentyman PR, et al. Phase I study of etoposide with SDZ PSC 833 as a modulator of multidrug resistance in patients with cancer. J Clin Oncol 1996;14:610–8. [51] Giaccone G, Linn SC, Welink J, et al. A dose-finding and pharmacokinetic study of reversal of multidrug resistance with SDZ PSC 833 in combination with doxorubicin in patients with solid tumors. Clin Cancer Res 1997;3:2005–15. [52] Advani R, Saba HI, Tallman MS, et al. Treatment of refractory and relapsed acute myelogenous leukemia with combination chemotherapy plus the multidrug resistance modulator PSC 833 (Valspodar). Blood 1999;93:787–95. [53] Fracasso PM, Westervelt P, Fears CL, et al. Phase I study of paclitaxel in combination with a multidrug resistance modulator, PSC 833 (Valspodar), in refractory malignancies. J Clin Oncol 2000;18:1124–34. [54] Leonard GD, Fojo T, Bates SE. The role of ABC transporters in clinical practice. Oncologist 2003;8:411–24. [55] Bates SE. Solving the problems of multidrug-resistance: ABC transporters in clinical oncology. In: Holland IB, Cole SP, Kuchler K, et al., editors. ABC proteins: from bacteria to man. London: Elsevier Science; 2002. p. 359–91. [56] Beketic-Oreskovic L, Duran GE, Chen G, Dumontet C, Sikic BI. Decreased mutation rate for cellular resistance to doxorubicin and suppression of mdrl gene activation by the cyclosporin PSC 833. J Natl Cancer Inst 1995;87:1593–602. [57] Cocker HA, Tiffin N, Pritchard-Jones K, Pinkerton CR, Kelland LR. In vitro prevention of the emergence of multidrug resistance in a pediatric rhabdomyosarcoma cell line. Clin Cancer Res 2001;7:3193–8. [58] Levade T, Jaffrezou JP. Signalling sphingomyelinases: which, where, how and why? Biochim Biophys Acta 1999;1438:1–17. [59] Sietsma H, Veldman RJ, Kok JW. The involvement of sphingolipids in multidrug resistance. J Membr Biol 2001;181:153–62. [60] Perry DK. The role of de novo ceramide synthesis in chemotherapy-induced apoptosis. Ann N Y Acad Sci 2000;905:91–6. [61] Mandon EC, Ehses I, Rother J, van Echten G, Sandhoff K. Subcellular localization and membrane topology of serine palmitoyltransferase, 3-dehydrosphinganine reductase, and sphinganine N-acyltransferase in mouse liver. J Biol Chem 1992;267: 11144–8. [62] Michel C, van Echten-Deckert G. Conversion of dihydroceramide to ceramide occurs at the cytosolic face of the endoplasmic reticulum. FEBS Lett 1997;416:153–5. [63] Hannun YA, Luberto C, Argraves KM. Enzymes of sphingolipid metabolism: from modular to integrative signaling. Biochemistry 2001;40:4893–903. [64] Michel C, van Echten-Deckert G, Rother J, Sandhoff K, Wang E, Merrill Jr AH. Characterization of ceramide synthesis. A dihydroceramide desaturase introduces the 4,5-trans-double bond

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

[65]

[66]

[67]

[68]

[69]

[70]

[71]

[72]

[73]

[74] [75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

of sphingosine at the level of dihydroceramide. J Biol Chem 1997;272:22432–7. Bose R, Verheij M, Haimovitz-Friedman A, Scotto K, Fuks Z, Kolesnick R. Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mechanism for generating death signals. Cell 1995;82:405–14. Turnbull KJ, Brown BL, Dobson PR. Caspase-3-like activity is necessary but not sufficient for daunorubicin-induced apoptosis in Jurkat human lymphoblastic leukemia cells. Leukemia 1999;13:1056–61. Zhang J, Alter N, Reed JC, Borner C, Obeid LM, Hannun YA. Bcl2 interrupts the ceramide-mediated pathway of cell death. Proc Natl Acad Sci USA 1996;93:5325–8. Olshefski RS, Ladisch S. Glucosylceramide synthase inhibition enhances vincristine-induced cytotoxicity. Int J Cancer 2001;93:131–8. Cabot MC, Giuliano AE, Han TY, Liu YY. SDZ PSC 833, the cyclosporine A analogue and multidrug resistance modulator, activates ceramide synthesis and increases vinblastine sensitivity in drug-sensitive and drug-resistant cancer cells. Cancer Res 1999;59:880–5. Myrick D, Blackinton D, Klostergaard J, et al. Paclitaxel-induced apoptosis in Jurkat, a leukemic T cell line, is enhanced by ceramide. Leuk Res 1999;23:569–78. Mehta S, Blackinton D, Omar I, et al. Combined cytotoxic action of paclitaxel and ceramide against the human Tul38 head and neck squamous carcinoma cell line. Cancer Chemother Pharmacol 2000;46:85–92. Tepper AD, de Vries E, van Blitterswijk WJ, Borst J. Ordering of ceramide formation, caspase activation, and mitochondrial changes during CD95- and DNA damage-induced apoptosis. J Clin Invest 1999;103:971–8. Perry DK, Carton J, Shah AK, Meredith F, Uhlinger DJ, Hannun YA. Serine palmitoyltransferase regulates de novo ceramide generation during etoposide-induced apoptosis. J Biol Chem 2000;275:9078–84. Radin NS. The development of aggressive cancer: a possible role for sphingolipids. Cancer Invest 2002;20:779–86. Suzuki A, Iwasaki M, Kato M, Wagai N. Sequential operation of ceramide synthesis and ICE cascade in CPT-11-initiated apoptotic death signaling. Exp Cell Res 1997;233:41–7. Chauvier D, Morjani H, Manfait M. Ceramide involvement in homocamptothecin- and camptothecin-induced cytotoxicity and apoptosis in colon HT29 cells. Int J Oncol 2002;20:855–63. Akao Y, Kusakabe S, Banno Y, et al. Ceramide accumulation is independent of camptothecin-induced apoptosis in prostate cancer LNCaP cells. Biochem Biophys Res Commun 2002;294: 363–70. Herget T, Esdar C, Oehrlein SA, et al. Production of ceramides causes apoptosis during early neural differentiation in vitro. J Biol Chem 2000;275:30344–54. Maurer BJ, Metelitsa LS, Seeger RC, Cabot MC, Reynolds CP. Increase of ceramide and induction of mixed apoptosis/necrosis by N-(4-hydroxyphenyl)-retinamide in neuroblastoma cell lines. J Natl Cancer Inst 1999;91:1138–46. Wang H, Charles AG, Frankel AJ, Cabot MC. Increasing intracellular ceramide: an approach that enhances the cytotoxic response in prostate cancer cells. Urology 2003;61:1047–52. Reynolds CP, Maurer BJ, Kolesnick RN. Ceramide synthesis and metabolism as a target for cancer therapy. Cancer Lett 2004;206:169–80. Chalfant CE, Rathman K, Pinkerman RL, et al. De novo ceramide regulates the alternative splicing of caspase 9 and Bcl-x in A549 lung adenocarcinoma cells. Dependence on protein phosphatase-1. J Biol Chem 2002;277:12587–95. Gomez Del Pulgar T, De Ceballos ML, Guzman M, Veiasco G. Cannabinoids protect astrocytes from ceramide-induced apoptosis

[84]

[85]

[86]

[87]

[88]

[89]

[90]

[91]

[92]

[93]

[94]

[95]

[96]

[97]

[98]

[99]

[100] [101]

[102]

69

through the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem 2002;277:36527–33. Gomez del Pulgar T, Veiasco G, Sanchez C, Haro A, Guzman M. De novo-synthesized ceramide is involved in cannabinoid-induced apoptosis. Biochem J 2002;363:183–8. Cabot MC, Han TY, Giuliano AE. The multidrug resistance modulator SDZ PSC 833 is a potent activator of cellular ceramide formation. FEBS Lett 1998;431:185–8. Goulding CW, Giuliano AE, Cabot MC. SDZ PSC 833 the drug resistance modulator activates cellular ceramide formation by a pathway independent of P-glycoprotein. Cancer Lett 2000;149:143–51. Senchenkov A, Litvak DA, Cabot MC. Targeting ceramide metabolism—a strategy for overcoming drug resistance. J Natl Cancer Inst 2001;93:347–57. Lucci A, Han TY, Liu YY, Giuliano AE, Cabot MC. Multidrug resistance modulators and doxorubicin synergize to elevate ceramide levels and elicit apoptosis in drug-resistant cancer cells. Cancer 1999;86:300–11. Lucci A, Han TY, Liu YY, Giuliano AE, Cabot MC. Modification of ceramide metabolism increases cancer cell sensitivity to cytotoxics. Int J Oncol 1999;15:541–6. Wang H, Giuliano AE, Cabot MC. Enhanced de novo ceramide generation through activation of serine palmitoyltransferase by the P-glycoprotein antagonist SDZ PSC 833 in breast cancer cells. Mol Cancer Ther 2002;1:719–26. Linardic CM, Hannun YA. Identification of a distinct pool of sphingomyelin involved in the sphingomyelin cycle. J Biol Chem 1994;269:23530–7. Bezombes C, Maestre N, Laurent G, Levade T, Bettaieb A, Jaffrezou JP. Restoration of TNF-alpha-induced ceramide generation and apoptosis in resistant human leukemia KGla cells by the Pglycoprotein blocker PSC833. FASEB J 1998;12:101–9. Peterson RH, Meyers MB, Spengler BA, Biedler JL. Alteration of plasma membrane glycopeptides and gangliosides of Chinese hamster cells accompanying development of resistance to daunorubicin and vincristine. Cancer Res 1983;43:222–8. Lavie Y, Cao H, Bursten SL, Giuliano AE, Cabot MC. Accumulation of glucosylceramides in multidrug-resistant cancer cells. J Biol Chem 1996;271:19530–6. Liu YY, Han TY, Giuliano AE, Cabot MC. Expression of glucosylceramide synthase, converting ceramide to glucosylceramide, confers adriamycin resistance in human breast cancer cells. J Biol Chem 1999;274:1140–6. Morjani H, Aouali N, Belhoussine R, Veldman RJ, Levade T, Manfait M. Elevation of glucosylceramide in multidrug-resistant cancer cells and accumulation in cytoplasmic droplets. Int J Cancer 2001;94:157–65. Kok JW, Veldman RJ, Klappe K, Koning H, Filipeanu CM, Muller M. Differential expression of sphingolipids in MRP1 overexpressing HT29 cells. Int J Cancer 2000;87:172–8. Lucci A, Cho WI, Han TY, Giuliano AE, Morton DL, Cabot MC. Glucosylceramide: a marker for multiple-drug resistant cancers. Anticancer Res 1998;18:475–80. Veldman RJ, Klappe K, Hinrichs J, et al. Altered sphingolipid metabolism in multidrug-resistant ovarian cancer cells is due to uncoupling of glycolipid biosynthesis in the Golgi apparatus. FASEB J 2002;16:1111–3. Liu YY, Han TY, Giuliano AE, Cabot MC. Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J 2001;15:719–30. Liu YY, Han TY, Giuliano AE, Hansen N, Cabot MC. Uncoupling ceramide glycosylation by transfection of glucosylceramide synthase antisense reverses adriamycin resistance. J Biol Chem 2000;275:7138–43. Veldman RJ, Mita A, Cuvillier O, et al. The absence of functional glucosylceramide synthase does not sensitize melanoma cells for anticancer drugs. FASEB J 2003;17:1144–6.

70

N. Aouali et al. / Critical Reviews in Oncology/Hematology 56 (2005) 61–70

[103] Lavie Y, Cao H, Volner A, et al. Agents that reverse multidrug resistance, tamoxifen, verapamil, and cyclosporin A, block glycosphingolipid metabolism by inhibiting ceramide glycosylation in human cancer cells. J Biol Chem 1997;272:1682–7. [104] Lucci A, Giuliano AE, Han TY, et al. Ceramide toxicity and metabolism differ in wild-type and multidrug-resistant cancer cells. Int J Oncol 1999;15:535–40. [105] Nicholson KM, Quinn DM, Kellett GL, Warr JR. Preferential killing of multidrug-resistant KB cells by inhibitors of glucosylceramide synthase. Br J Cancer 1999;81:423–30. [106] Coezy E, Borgna JL, Rochefort H. Tamoxifen and metabolites in MCF7 cells: correlation between binding to estrogen receptor and inhibition of cell growth. Cancer Res 1982;42:317–23. [107] Cabot MC, Giuliano AE, Volner A, Han TY. Tamoxifen retards glycosphingolipid metabolism in human cancer cells. FEBS Lett 1996;394:129–31. [108] Pommerenke E, Mattern J, Volm M. Modulation of doxorubicintoxicity by tamoxifen in multidrug-resistant tumor cells in vitro and in vivo. J Cancer Res Clin Oncol 1994;120:422–6. [109] Szakacs G, Annereau JP, Lababidi S, et al. Predicting drug sensitivity and resistance: profiling ABC transporter genes in cancer cells. Cancer Cell 2004;6:129–37.

[110] Lehne G, Sorensen DR, Tjonnfjord GE, et al. The cyclosporin PSC 833 increases survival and delays engraftment of human multidrug-resistant leukemia cells in xenotransplanted NOD-SCID mice. Leukemia 2002;16:2388–94.

Biography Hamid Morjani is 41 years old and obtained his Ph.D. degree in Biophysics in 1993. He became lecturer researcher in the University of Reims, France in 1993, where he is currently working in the JE2428 Onco-Pharmacology Group. He is a member of the American Association for Cancer Research and the Federation of European Biochemical Societies. He has authored 40 articles, mainly in the field of “multidrug-resistance to anticancer drugs” and especially on “mechanisms of resistance to topoisomerase I and II inhibitors, and telomerase inhibitors belonging to the G-quadruplex ligand group”.