25
Critical Reviews in Oncology/Hematology 28 (1998) 181 – 205 Recent developments in drug resistance and apoptosis research Martin Clynes a, *, Carmel Daly a , Ro ´ isı ´n NicAmhlaoibh a , Deirdre Cronin a , Cathal Elliott a , Robert O’Connor a , Toni O’Doherty a , Lisa Connolly a , Anthony Howlett b , Kevin Scanlon b a National Cell and Tissue Culture Centre, Dublin City Uni6ersity, Glasne6in, Dublin 9, Ireland b Berlex Biosciences, 15049 San Pablo A6e., P.O. Box 4099, Richmond, CA 94804 -0099, USA Accepted 20 June 1998 Contents 1. Introduction ............................................... 182 2. Apoptosis: a final common pathway for cancer drug cytotoxicity?................ 183 2.1. Introduction ............................................ 183 2.2. JNK Pathway ........................................... 183 2.3. Growth factor receptor and oncogene signalling and resistance to apoptosis ....... 184 2.4. Cell death signaling via Fas and TNF receptors ....................... 185 3. Bcl proteins in the regulation of apoptosis ............................. 185 3.1. Function of Bcl-2/Bcl-X L , Bax and related proteins and their mechanism of action in antagonising apoptosis ...................................... 185 3.2. Western blot analysis of Bcl family members ......................... 186 3.3. Phosphorylation of Bcl-2/Bcl-X L and Bad ........................... 186 3.4. Bcl-X gene structure and alternative splicing ......................... 187 4. Bcl proteins in the regulation of cancer chemotherapeutic drug-induced apoptosis ...... 187 4.1. Bcl-X L , like bcl-2, can protect cancer cells in vitro against chemotherapeutic drug- induced apoptosis; the presence of antagonistic proteins (Bax) is a crucial factor .... 187 4.2. Enhancement of chemosensitivity in vitro and of cancer cell apoptosis in vivo by over- expression of bcl-X S ....................................... 188 4.3. Role of bcl-2 in modulation of telomerase activity ...................... 188 5. Other proteins which may modulate apoptosis ........................... 188 5.1. Bag-1 co-operates with Bcl-2 in promoting cell survival and also activates Raf-1 .... 188 5.2. Bad can heterodimerise with Bcl-X L displacing Bax and promoting cell death. Bad antagonises Bcl-X L rather than Bcl-2 activity ......................... 189 5.3. Galectin............................................... 189 5.4. MCL-1, BFL-1, survivin, 1AP-like protein........................... 189 5.5. Bcl-X L /Bcl-2-interacting proapoptotic proteins ........................ 189 6. Bcl-X expression in vivo ........................................ 190 6.1. Expression of bcl-X in various tissues, and physiology of bcl-x, bcl-2- and bax- deficient mice............................................ 190 6.2. Expression of bcl-2, bcl-X and bax in tumours ........................ 190 * Corresponding author. Tel.: +353 1 7045720; fax: +353 1 7045484; e-mail: [email protected] 1040-8428/98/$ - see front matter © 1998 Elsevier Science Ireland Ltd. All rights reserved. PII S1040-8428(98)00018-3

Recent developments in drug resistance and apoptosis research

Embed Size (px)

Citation preview

Page 1: Recent developments in drug resistance and apoptosis research

Critical Reviews in Oncology/Hematology 28 (1998) 181–205

Recent developments in drug resistance and apoptosis research

Martin Clynes a,*, Carmel Daly a, Roisın NicAmhlaoibh a, Deirdre Cronin a, Cathal Elliott a,Robert O’Connor a, Toni O’Doherty a, Lisa Connolly a, Anthony Howlett b, Kevin Scanlon b

a National Cell and Tissue Culture Centre, Dublin City Uni6ersity, Glasne6in, Dublin 9, Irelandb Berlex Biosciences, 15049 San Pablo A6e., P.O. Box 4099, Richmond, CA 94804-0099, USA

Accepted 20 June 1998

Contents1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182

2. Apoptosis: a final common pathway for cancer drug cytotoxicity?. . . . . . . . . . . . . . . . 1832.1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1832.2. JNK Pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1832.3. Growth factor receptor and oncogene signalling and resistance to apoptosis . . . . . . . 1842.4. Cell death signaling via Fas and TNF receptors . . . . . . . . . . . . . . . . . . . . . . . 185

3. Bcl proteins in the regulation of apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1853.1. Function of Bcl-2/Bcl-XL, Bax and related proteins and their mechanism of action in

antagonising apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1853.2. Western blot analysis of Bcl family members . . . . . . . . . . . . . . . . . . . . . . . . . 1863.3. Phosphorylation of Bcl-2/Bcl-XL and Bad . . . . . . . . . . . . . . . . . . . . . . . . . . . 1863.4. Bcl-X gene structure and alternative splicing . . . . . . . . . . . . . . . . . . . . . . . . . 187

4. Bcl proteins in the regulation of cancer chemotherapeutic drug-induced apoptosis . . . . . . 1874.1. Bcl-XL, like bcl-2, can protect cancer cells in vitro against chemotherapeutic drug-

induced apoptosis; the presence of antagonistic proteins (Bax) is a crucial factor . . . . 1874.2. Enhancement of chemosensitivity in vitro and of cancer cell apoptosis in vivo by over-

expression of bcl-XS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1884.3. Role of bcl-2 in modulation of telomerase activity . . . . . . . . . . . . . . . . . . . . . . 188

5. Other proteins which may modulate apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . 1885.1. Bag-1 co-operates with Bcl-2 in promoting cell survival and also activates Raf-1 . . . . 1885.2. Bad can heterodimerise with Bcl-XL displacing Bax and promoting cell death. Bad

antagonises Bcl-XL rather than Bcl-2 activity . . . . . . . . . . . . . . . . . . . . . . . . . 1895.3. Galectin. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1895.4. MCL-1, BFL-1, survivin, 1AP-like protein. . . . . . . . . . . . . . . . . . . . . . . . . . . 1895.5. Bcl-XL/Bcl-2-interacting proapoptotic proteins . . . . . . . . . . . . . . . . . . . . . . . . 189

6. Bcl-X expression in vivo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1906.1. Expression of bcl-X in various tissues, and physiology of bcl-x, bcl-2- and bax-

deficient mice. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1906.2. Expression of bcl-2, bcl-X and bax in tumours . . . . . . . . . . . . . . . . . . . . . . . . 190

* Corresponding author. Tel.: +353 1 7045720; fax: +353 17045484; e-mail: [email protected]

1040-8428/98/$ - see front matter © 1998 Elsevier Science Ireland Ltd. All rights reserved.PII S 1 0 4 0 - 8 4 2 8 ( 9 8 ) 0 0 0 1 8 - 3

Page 2: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205182

7. Recent developments in understanding the role of MRP in drug-resistance . . . . . . . . . . . 1907.1. MRP expression in cell lines and in human tumours. . . . . . . . . . . . . . . . . . . . . 1907.2. MRP gene structure. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1917.3. MRP protein structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1917.4. MRP transport activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1927.5. Modulation of MRP mediated drug resistance . . . . . . . . . . . . . . . . . . . . . . . . 1927.6. MRP as a member of the ATP-binding cassette superfamily . . . . . . . . . . . . . . . . 193

8. Clinical studies on reversal of multi-drug resistance in cancer . . . . . . . . . . . . . . . . . . 1948.1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1948.2. Problems associated with chemosensitisation . . . . . . . . . . . . . . . . . . . . . . . . . 1948.3. Circumvention of P-170 overexpression . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1948.4. Resistance due to DNA repair mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . 1968.5. New strategies to overcome drug resistance . . . . . . . . . . . . . . . . . . . . . . . . . . 196

8.5.1. Ribozymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1968.5.2. Cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196

8.6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196

9. Conclusion/prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197

Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198

Reviewer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198

Biography. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205

1. Introduction

Chemotherapy can cure many human cancers, butresistance—intrinsic or acquired—is a problem formany patients, especially in solid tumours. Combina-tion chemotherapy was developed in order to circum-vent specific resistance mechanisms, (as well as toallow higher overall doses of anti-tumour chemicalsby combining agents with different side-effects/organtoxicity). Unfortunately, many tumours can developresistance to a wide range of drugs with differentcellular targets.

Many changes have been associated with multipledrug resistance in cell culture systems and in tumoursin vivo [1–3]. Alterations in drug accumulation at thecellular level, and in intracellular drug distribution,seem to play an important role [4,5] and P-glyco-protein/P-170, the product of the mdr-1 gene, hasbeen thoroughly studied at the molecular [6,7] and atthe clinical [8] levels. The more recently describedefflux pump MRP (discussed in detail in Section 7below) may also be important in drug transport andsubcellular distribution. LRP, the major protein of arecently described subcellular organelle, the vault [9]may be associated with multidrug resistance in celllines and in clinical samples [10]. Chauffert et al. [11]have drawn attention to the phenomenon of kineticresistance to chemotherapy, and the proportion ofcells at different stages of the cell cycle in a tumour

cell population may play a significant role in responseof that tumour to drug exposure; altered levels of thecyclin-dependent kinase inhibitor p27-Kip1 may be in-volved in kinetic drug resistance [12]. Altered levels/activity/isoenzyme profile of protein kinases may beimportant in drug resistance at least in part by modu-lating activity of proteins directly involved in resis-tance, such as the drug efflux pumps [13]. Changes inlevels or substrate affinity of Topoisomerases I and IIseem to be important also in MDR, although thedrug menu involved is often different from that ofclassical MDR [14–17]. Alterations in DNA repairand in glutathione-related pathways [18,19] may be ofparticular importance in cross-resistance betweenalkylating agents [20–22].

Multidrug resistance in vivo may not always be dueto alterations at the level of the tumour cell. Thevascularisation of a tumour may be critical, both insupply of drug to the tumour, and to deliver oxygen,since hypoxia can make tumours more resistant todrugs (see [23], for review). Expression of effluxpumps in blood vessels of brain tumours may form a‘blood-brain barrier’ conferring resistance on inher-ently sensitive tumours [24].

This review attempts to cover in some detail twoareas where there have been many recent develop-ments, and which may impact on improved under-standing and treatment of resistant cancers; theseareas are the relationships between drug resistance

Page 3: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 183

and apoptosis, and the biology of MRP. There is alsoa brief discussion on clinical approaches to overcom-ing MDR, since improved treatment of cancer pa-tients is the goal of MDR research, and new scientificadvances should be seen as opportunities to developnew treatment strategies.

2. Apoptosis: a final common pathway for cancer drugcytotoxicity?

2.1. Introduction

Most interest in multidrug resistance (MDR) overthe past decade has centered on drug-efflux pumpssuch as P-170 and MRP, but so far results with P-170 modulators have been somewhat disappointing,and in any event the range of drugs pumped by P-170 and MRP is considerably narrower than themenu of drugs involved in MDR in vivo. Some invitro models for wider range MDR are now available[25]. With this historical background, the idea that afinal cell-killing pathway common to all anticancerdrugs may exist has caused considerable interest.

The discovery of the bcl family of proteins whichcan apparently alter the threshold of recognition ofcell damage as a cell death signal suggests novelmechanisms of MDR and new approaches to thera-peutic intervention.

The bcl-2 (B-cell leukaemia/lymphoma 2) gene wasdiscovered by Tsujimoto and Croce [26]; the bcl-2gene is translocated in many follicular B-celllymphomas from its normal 18q21 position to 14q32where its location adjacent to enhancers in the im-munoglobulin H gene [27] leads to high level expres-sion. Alternative splicing yields two proteins, bcl-2a

(239 amino acids) and bcl-2b (205 amino acids), dif-fering only at their C terminus. Most publicationsrefer to bcl-2 (without specifying a or b) and whereWestern blots are reported, the a form appears to bepredominant. Vaux et al. [28] found that transfectionof immature pre-B cells with bcl-2 expression vectorsprotected against cell death due to IL-3 deprivation,thus indicating a role in antagonizing apoptosis.Transfection [29,30] and antisense [31] experimentsconfirmed an important role for bcl-2 in resistance toapoptosis induced by chemotherapeutic drugs. Bcl-2 iswidely expressed in human tumours, and by inhibitingcell death and thus altering the normal cell deathversus cell division ratio it may allow tumour cells toaccumulate the mutations which will later cause thecells to become invasive and metastatic. More recentresearch indicates a key role for bcl-XL, as well asbcl-2, in apoptosis regulation in follicular lymphoma[32].

Investigation of genes involved in programmed cell

death in the nematode Caenorhabditis elegans hasprovided many important leads to an understandingof apoptosis in mammalian cells, since many of thesegenes have been conserved between these distantly-re-lated species [33]. In order of sequence of action inthe cell death pathway the most important CEDgenes include CED-9 (bcl-2 homologue), CED-4(mammalian bcl-XL and caspases can interact withCED-4 protein) and CED-3 (homologue of caspaseenzymes).

It is now becoming clear that bcl-2 is just one com-ponent of a large and complex family of proteinswhich determine how readily particular cells die inresponse to particular physiological or pharmacologi-cal environments (e.g. growth factor deprivation, drugexposure). Programmed cell death may be particularlyimportant in determining organ shape and size duringdevelopment [34]. Some of the current knowledge inthis area is reviewed here, with particular emphasison how cancer cells can develop resistance to apopto-sis induced by cancer chemotherapeutic drugs.

Programmed cell death or apoptosis is mediated bydegradation of DNA and a number of important cel-lular proteins. This in turn is dependent on activationof certain members of the caspase family of prote-olytic enzymes (e.g. conversion of 32 kDa procaspase-3 to active 17 kDa caspase 3 [35]). This activation ismediated by release of activating factors, includingcytochrome c, from mitochondria (Section 3.1). Thepathways linking cellular damage induced by toxicagents such as anti-cancer drugs to release of mito-chondrial procaspase activators are still poorly under-stood, but some information on them is nowemerging.

2.2. JNK Pathway

The JNK/SAPK (c-Jun N-terminal kinase/stress-ac-tivated protein kinase) pathway may be one impor-tant signalling pathway linking exposure to drugs andactivation of caspases (JNK is a member of the mito-gen-activated protein kinase (MAPK) family). Seimiyaet al. [36] showed that reduction of JNK1 proteinlevels in U937 cells (by using JNK1 antisense, but notsense, oligodeoxynucleotides) reduced the rate ofapoptosis induced by etoposide or camptothecin;there was a corresponding reduction in caspase-3 acti-vation. Caspase-3 does not have an appropriateMAPK family phosphorylation site [37] so that inter-mediaries (perhaps release of mitochondrial media-tors) must be involved. The upstream regulatorslinking DNA damage to JNK activation are not com-pletely defined, but the tyrosine protein kinase c-Ablis a JNK activator. Antisense experiments indicatethat Lyn Kinase, a src family tyrosine kinase may

Page 4: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205184

also have this function [38]) and SH-P1 protein ty-rosine phosphatase may be a negative feedback regu-lator [39,40]. In CML cells, however, use of antisenseoligonucleotides directed against the bcr–abl tran-script enhanced cell killing, via apoptosis, by mafos-famide, the treatment being relatively non-toxic tonormal bone marrow cells [41]; further antisense ex-periments by [42] indicate a role for Grb2 and Crk1proteins in a bcr–abl-mediated signalling pathway.Activation of JNK is achieved by a protein kinasecascade, being activated by a MAPKK (MAP kinasekinase) which in turn is activated by a MAPKKK.Activated JNK phosphorylates the c-Jun transcriptionfactor (and other transcription factors including p53);the phosphorylated c-Jun mediates transcription of avariety of AP-1-dependent genes, including c-Jun it-self. Although the details are obscure, it appears thatthe JNK pathway plays an important role in trans-duction of apoptotic signals following exposure to awide range of chemotherapeutic drugs including vincaalkaloids, anthracyclines, mitomycin C, etoposide,camptothecin and cisplatin [39,43,44]. Fas ligand sig-nalling (Section 2.4) may also activate JNK, via alinker protein, Daxx, which mediates a FADD-inde-pendent Fas-associated (Section 2.4) apoptotic path-way [45]. Seimiya et al. [36] isolated a TPA-resistantvariant of U937 cells, which showed resistance toapoptosis induction by anticancer drugs (includingetoposide and camptothecin) and which showed afailure in drug-induced JNK1 activation, in spite ofDNA damage (measured by DNA-protein crosslink-ing) equivalent to that of parental cells. This observa-tion may point to a new class of drug-resistancemechanism, i.e. a failure of signalling pathways link-ing drug-induced cellular damage to apoptosis.

2.3. Growth factor receptor and oncogene signallingand resistance to apoptosis

The c-erbB2 gene encodes a 185 kDa transmem-brane glycoprotein which is the preferred het-erodimerisation partner for the other, homologous,members of the erb family (EGF Receptor, erbB3and erbB4) after they have bound their ligands [46].The actual ligand for c-erbB2 is still unknown. C-erbB2 gene amplification and overexpression is seenin about a quarter of breast carcinomas and expres-sion seemed to correlate with sensitivity to CMF (cy-clophosphamide, methotrexate and 5-fluorouracil)according to studies by Gusterson et al. [47], Klijn etal. [48] and Tetu and Brisson [49]. Muss et al. [50]found that higher doses of adriamycin were needed,but were effective, in patients expressing high levels ofc-erbB2. Bacus et al. [51] reported that tumours over-expressing c-erbB2 are less responsive to CMF, butdo respond to high doses of CAF (A=adriamycin).

Bottini et al. [52] found no correlation between c-erbB2 expression and CMF response in breast cancerpatients. A recent report on cross-reactivity (using theP-170 monoclonal antibody C219) between P-170 andc-erbB2 could be relevant in assessing studies involv-ing immunohistochemical measurements [53]. Tsai etal. [54] found a correlation between the level of c-erbB2 expression in cell lines and resistance to cellkilling by a range of chemotherapeutic drugs (e.g.adriamycin, melphalan, mitomycin C, VP-16 and cis-platin); they also found that c-erbB2 cDNA transfec-tion conferred resistance to these drugs. Yu et al. [55]found that c-erbB2 protein expression in a panel ofbreast cancer cell lines correlated with levels of resis-tance to taxol and taxotere, and that transfection ofc-erbB2 cDNA conferred resistance on a humanbreast carcinoma line (MDA-MB-435) with low-levelexpression of p185. While increased levels of EGFReceptor have been reported in some multidrug-resis-tant cells, and combination chemotherapy with cis-platin and an anti-EGFR-monoclonal antibodyshowed synergistic activity in a human tumour xeno-graft system [56], downregulation of EGFR with anti-sense oligonucleotides resulted in increased cisplatinresistance in the human breast carcinoma line MDA-MB-468 [57]. It remains unclear why the effect ofoverexpression of these receptors has variable effectson chemotherapy-induced apoptosis and preciselywhat the signalling pathways involved are.

Blocking of ‘survival signals’ (e.g. use of antibodieswhich block the interaction of b1 integrin with ECM)in normal but not in malignant breast cells [58] mayalter the balance of ‘death’ and ‘survival’ signals inthe intracellular signal transduction pathways, result-ing in cell death. It has recently been shown that thesurvival/anti-apoptotic signal delivered by IL-3 oper-ates via sequential activation of phosphoinositide 3-kinase and Akt (a serine-threonine protein kinase).Activated Akt can phosphorylate and thus inactivateBAD, a pro-apoptotic protein [59]. It remains to beestablished if this is a common pathway also relevantto other survival signals such as those mediated viaintegrins. Rodeck et al. [60] have shown that block-ade of the EGF receptor or forced suspension culture(preventing cell-substratum adhesion) downregulatesbcl-XL rendering the cells more susceptible to apopto-sis.

The STAT-1 (Signal transducer and activator oftranscription) deficient cells are resistant to TNF-ainduced apoptosis, and have low expression of cas-pases, both properties being restored by transfectionof STAT-1a cDNA [61]. STAT-1 may act as a tran-scriptional activator of caspases [62]. These discover-ies may point to a new potential mechanism by whichcells could become multidrug resistant.

Page 5: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 185

Oncogenes such as c-fos, H-ras and K-ras are im-portant modulators of drug resistance, partly becauseof their role in controlling expression levels of mR-NAs for efflux pumps, repair enzymes and enzymesfor detoxification, as demonstrated by ribozyme ex-periments [63–66]; their role in regulation of apop-totic pathways deserves further study.

2.4. Cell death signaling 6ia Fas and TNF receptors

The Fas/Fas L signal transduction system forapoptosis induction has been reviewed by Nagata[67]. Briefly, Fas (APO-1/CD95) is a cell-surface re-ceptor which, following ligand (Fas L) binding andoligomerisation, induces a cell death pathway involv-ing caspase activation. Other receptor-ligand path-ways (TNF-1 receptor/TNF-a) with similar activitiesexist. Fas and similar receptors contain a homologous80 amino acid cytoplasmic region, called the ‘deathdomain (DD)’ which is essential for transduction ofcell killing signals. Yeast two-hybrid experiments ledto identification of FADD (Fas-associating proteinwith death domain), also called MORT1; and anotherprotein called TRADD (TNFR-1 associated deathdomain protein). FADD contains a C-terminal DDwhich binds to the Fas DD; it also contains an N-terminal ‘death effector domain (DED)’ (also calledthe MORT1 domain). DED, on activation, interactswith and activates Caspase 8 (also called FLICE(FADD-like ICE) or MACH (MORT1-associatedCED-3 homologue)), which contains two DED do-mains. Activated Caspase 8 appears to have substratespecificity similar to that of Caspase 3. (TRADD mayact by direct interaction with FADD, since it has noDeath Effector domain itself, although it also inter-acts with other proteins (RIP) which may recruit cas-pases by other pathways).

Unlike Fas, other members of the TNF receptorfamily can activate the transcription factor NF-kB,via the TRAF protein (TNF receptor-associated fac-tor), thus, in certain cellular/metabolic situations-re-sulting in an overall survival or growth signal. Thismay represent a safety cut-off mechanism to preventinappropriate induction of apoptosis. Bcl-2 and Bcl-XL can inhibit Fas-induced apoptosis [67], but not inall cell systems [68,69]; for further discussion relevantto this, see Section 4.1 and Section 5.1.

Production of Fas L (in the absence of Fas orelements of its transduction pathway) could rendertumours resistant to immune attack by Fas positivelymphocytes. For example, many human lung cancersexpress Fas ligand [70].

Tumours such as prostatic carcinomas [71] havefunctional Fas-mediated cell death pathways. Treat-ment of a number of tumour cell types includingcolon carcinoma [72] and leukaemia [73] with clini-

cally relevant concentrations of anti-cancer drugs in-creases levels of Fas, which could be relevant in vivoby making cells more sensitive to cell killing, e.g. byFas L from immune lymphocytes. There is also recentevidence that drug treatment can induce expression ofFas L [74]. The importance of these observations inchemotherapy (and resistance mechanisms) remains tobe established by further investigations.

3. Bcl proteins in the regulation of apoptosis

3.1. Function of Bcl-2/Bcl-XL, Bax and relatedproteins and their mechanism of action in antagonisingapoptosis

It appears that members of the CED-3 subfamilyof caspases (caspases 2, 3, 9 and 10) and caspase 8(FLICE) are among the most important mediators ofthe proteolytic events determining commitment of acell to apoptosis [75,76]. For example, when U937cells are treated with anticancer agents, caspase 3 isconverted from an inactive 32 kDa precursor to anactive 17 kDa fragment resulting in cleavage of spe-cific cellular substrates including actin [35] and polyADP-ribose polymerase [77]. Selective inhibitors ofcaspase activity (Z-Asp-CH2-DCB) completely blockchemotherapeutic drug-induced apoptosis in cell linessuch as U937 and Jurkat, even under conditionswhere significant DNA damage occurs [78–80]. Itshould be noted, however, that in certain systems,caspase inhibition does not block apoptosis [81]. Re-lease of cytochrome c from mitochondria into the cy-tosol appears to be one of the key steps in caspaseactivation; cytochrome c has been designated asApaf-2 (apoptosis activating factor 2) based on activ-ity in cell-free extracts. Another possibly importantfactor released from mitochondria is AIF (apoptosis-inducing factor) which is a caspase-inhibitor-sensitiveprotease released following mitochondrial depolarisa-tion and permeability transition; this release can beinhibited by bcl-2 [82,83]. Bcl-2 or Bcl-XL inhibit re-lease of cytochrome c from mitochondria followingapoptosis-inducing stimuli, and Kharbanda et al. [84]have demonstrated direct interaction between bcl-XL

and cytochrome c. Zou et al. [85] have recently re-ported the cloning of Apaf-1, a 130 kD protein,which may be the mammalian homologue of CED-4.Apaf-1 binds to cytochrome c and to procaspase 9,and this complex may be responsible for activation ofcaspase 3 [86].

Caspase 3 may activate DNA fragmentation factor(DFF) which initiates DNA fragmentation, long con-sidered a key event in apoptosis [87].

Reed [88] has recently reviewed the mechanisms bywhich Bcl-2 and Bcl-XL may antagonise apoptosis.

Page 6: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205186

These proteins are localised in the outer mitochon-drial membrane, endoplasmic reticulum and nuclearenvelope, and may have cellular functions in additionto modulation of apoptosis. Most emphasis in theliterature to date, however, has been on mitochon-drial activity relating to apoptosis. Bcl-2/bcl-XL createion channels in membranes; Bax may also be able todo this, and possibly by heterodimerization with Bcl-2or Bcl-XL it may alter the properties of channels inthe mitochondrial membrane. Mitochondrial mem-brane depolarisation, with formation of megaporesand calcium efflux, is generally observed during apop-tosis, but there is disagreement on whether this is aconsequence of caspase activation, or a trigger forrelease of cytochrome c and AIF. Hacker and Vaux[89] and Reed [88] have reviewed the actions of pro-apoptotic (Bax, Bcl-XS, Bad, Bak, Bik, Bok) andanti-apoptotic (bcl-XL, bcl-2, Bag-1, Bfl-1, Mcl-1) bclfamily members.

The actual mechanism by which bcl-2/bcl-XL andBax, possibly as homodimers, may form channelswhich facilitate or block apoptosis is not fully under-stood. In one model, heterodimerisation would inhibitformation of functional channels. This hypothesiswould explain some surprising results where overex-pression of Bcl-2 sometimes promotes cell death, andBax or Bak can in some cellular backgrounds unex-pectedly protect against apoptosis. Another model en-visages Bax channels facilitating cytochrome ctransport across the outer mitochondrial membrane,with bcl-2/bcl-XL interfering with this activity [88,90].

Bcl family members (whether pro- or anti-apop-totic) contain several (or all) of 4 bcl-2 homologydomains (BH1 to 4) and in most cases a C-terminaltransmembrane anchor (TM). The different domainsmay correspond to different functions, including inter-action with other proteins. Immunoprecipitation stud-ies and use of yeast-two-hybrid systems haveidentified many proteins which can bind to bcl-2 orbcl-XL (reviewed by Reed [88]). These include theprotein kinase Raf-1, the protein phosphatase cal-cineurin, the GTPases R-Ras and H-Ras, the p53-binding protein p53-BP2, the prion protein, caspasessuch as FLICE (caspase 8) and ICE (caspase 1) anda number of other proteins including CED-4, BAC-1,Nip-1, Nip-2 and Nip-3. Bax does not appear to in-teract with these proteins. These interactions may in-volve sequestering proteins from the cytosol, so thatit is suggested that bcl-2/bcl-XL have a dual function,(membrane channel component and adaptor/dockingprotein). Recently, evidence has been presented to in-dicate that caspase cleavage may convert bcl-2 to aform which has Bax-like apoptosis-promoting func-tions; this could contribute to the rapid collapse ofcells in the latter stages of apoptosis [91]. Other re-cent results indicate that regulation of mitochondrial

mediator release is not the only anti-apoptotic rolewhich bcl-XL and related proteins can play (Section5.5). The various cellular locations of bcl-2/bcl-XL

may be relevant in this regard.Reactive oxygen species (ROS) may be involved in

terminal stages of apoptosis in some situations; forexample, in apoptosis of neuronal cells followingNGF withdrawal, cell death can be prevented byROS traps and antioxidants; ROS production can beblocked by actinomycin D or cycloheximide (whichcan also block apoptosis induction in this system), orby caspase inhibitors [92].

3.2. Western blot analysis of Bcl family members

Krajewska et al. [93] used anti-peptide rabbit anti-seram to examine expression of bcl family membersin tissue from human colorectal tumours. For Bcl-2,the main band was at 26 kDa, with a second band(possibly the phosphorylated inactive form) at 30 kD,in some samples. Bcl-XL migrated as a doublet band(28–30 kD) and a 19 kDa bcl-XS band was also seenin most samples. Bcl-XL levels were elevated in tu-mour versus adjacent normal tissues, but no differ-ence was apparent in bcl-Xs levels. The doubletbcl-XL had also been noted by Krajewski et al. [94],in normal human tissues; they suggested proteolyticprocessing, covalent modifications, initiation of trans-lation from a downstream AUG or CUG in the bcl-XL open reading frame, or additional alternativelyspliced forms of bcl-X.

Bax-a gave a single 21 kDa M.W. band, and Bak asingle 25 kDa band.

Many antibodies available commercially for detec-tion of apoptosis-related proteins give variable resultsand show multiple unexplained bands.

3.3. Phosphorylation of Bcl-2/Bcl-XL and Bad

It is unclear whether or not Bcl-XL activity is regu-lated by phosphorylation, although the doublet bandobserved for the dimeric form on Western blots sug-gests that this is a possibility, and the general similar-ity with Bcl-2 is a further indication that this mayoccur, since phosphorylation of bcl-2 is well docu-mented. Haldar et al. [95], using the phosphatase in-hibitor okadaic acid, demonstrated that Bcl-2 can bephosphorylated in lymphoid cells, and that phospho-rylation inhibits the ability of Bcl-2 to bind to Baxand to modulate apoptosis. Haldar et al. [96,97] fur-ther showed that taxotere and to a lesser extent taxol,cause induction of Bcl-2 phosphorylation in tumourcell types including breast, prostate, leukaemia andlymphoma (possibly as a result of effects on micro-tubules). Phosphorylation of Bad is discussed in Sec-tion 5.2.

Page 7: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 187

3.4. Bcl-X gene structure and alternati6e splicing

The promoter region and genomic organisation ofthe mouse bcl-X gene have been analysed in detail[98]. Two major alternatively spliced mRNAs, withopposing actions on cell death, bcl-XL and bcl-XS,have been described. Bcl-XS lacks the BH1 and BH2domains of bcl-2 and as a negative regulator of apop-tosis it is dominant over bcl-2 and bcl-XL, possiblyby heterodimersing with them. Two additional formsof bcl-X mRNA, bcl-Xb and bcl-XDTM have been de-scribed in rodents, but their function is unknown.Shiraiwa et al. [99] have suggested that rat bcl-Xb (anunspliced form of bcl-X mRNA) may, like bcl-XS,promote apoptosis. A fifth novel alternatively splicedform containing a unique 47 amino acid C-terminus,and termed bcl-X gamma, appears to play a role ininhibiting activation-induced apoptosis in T-cells. Ithas also been found in thymocytes, where interactionof the T-cell receptor with host MHC molecules mayinvolve a cellular pathway affected by bcl-X gamma[100].

A number of alternatively spliced Bax transcriptshave also been described [101].

Pecci et al. [102] found that in a rat endometrialcell line which expresses functional progesterone re-ceptors, cell survival/prevention of apoptosis is depen-dent on the presence of progesterone (underconditions of low serum and absence of glucocorti-coids). Apoptosis could be induced by progesteroneantagonists. In this cell line, progesterone administra-tion increased overall bcl-X mRNA levels, and alsoshifted the ratio of spliced transcripts in favour ofbcl-XL as opposed to bcl-XS. Administration ofprogesterone antagonists increased the relativeamount of bcl-XS, without altering overall bcl-XmRNA levels. The authors speculate that modulationof the bcl-XL: bcl-XS ratio may be involved in degen-eration of the endometrial wall lining at the end ofthe menstrual cycle, when progesterone levels (fromthe ovary) drop.

Tilly et al. [103] found that, in equine systems, cho-rionic gonadotrophin-mediated inhibition of apoptosisin ovarian granulosa cells may be linked to a shift inthe ratio between bcl-2/bcl-XL and bax/bcl-XS mRNAlevels, although in this system it appeared that alter-ation in bax levels was the most important determi-nant.

Chang et al. [104] reported that treatment of a hu-man gastric cancer line with translation/transcriptioninhibitors upregulated bcl-XS and induced apoptosis;this could be inhibited by dexamethasone treatmentwhich suppressed the chemically-induced upregulationof bcl-XS mRNA, and also increased the bcl-XL

mRNA level as well as enhancing overall bcl-XmRNA stability.

4. Bcl proteins in the regulation of cancerchemotherapeutic drug-induced apoptosis

4.1. Bcl-XL, like bcl-2, can protect cancer cells in 6itroagainst chemotherapeutic drug-induced apoptosis; thepresence of antagonistic proteins (Bax) is a crucialfactor

Transfection with bcl-XL cDNA has been shown toprotect several cell types in vitro against apoptosisinduced by a wide range of chemotherapeutic drugs.Minn et al. [105] transfected human bcl-XL cDNA intothe murine IL-3-dependent prolymphocytic cell lineFL5.12; they found increased resistance to the anti-cancer drugs bleomycin, cisplatin, etoposide and vin-cristine, as well as the protein synthesis inhibitorhygromycin B and mycophenolic acid (which inhibitsguanine biosynthesis). Reversible cell cycle arrest wasnot prevented by bcl-XL overexpression; the protectionappeared, instead, to involve direct protection againstundergoing apoptosis. Vincristine-treated bcl-XL over-expressing cells became polyploid after drug removal,and Minn et al. [105] suggest a role for bcl-XL protec-tion against apoptosis in accumulation of chromosomalabnormalities in tumours.

Dole et al. [106] examined a range of neuroblastomacell lines with different levels of bcl-2 and bcl-XL ex-pression; they also transfected bcl-XL cDNA into oneof the lines which expressed very low endogenous levelsof bcl-XL. The transfected lines displayed enhancedresistance to apoptosis induced by cisplatinum or by anactivated form of cyclophosphamide (4-hydroperoxy-cyclophosphamide); apoptosis induced by the topoiso-merase II inhibitor VP-16 was, however, slightlydelayed, but not prevented.

Transfection with bcl-2 or bcl-XL cDNA conferredresistance to taxol in human leukaemia cell lines HL-60(acute myeloid leukaemia) and 697 (pre-B humanleukaemia), [107,108]. Bcl-XL cDNA transfection alsoconferred resistance to cycloheximide-induced apopto-sis in mouse myeloma cells [109].

In vitro experiments to downregulate bcl-2 expres-sion using antisense oligodeoxynucleotides in small-celllung cancer cells [110] and a hammerhead ribozyme inprostatic carcinoma cells [111] have been successful,and represent a possible prelude to gene therapy appli-cations. Kitada et al. [31] found enhanced sensitivity tomethotrexate and ara C following bcl-2 antisense treat-ment of non-Hodgkins leukaemia cells. Webb et al.[112] described the first trial of bcl-2 antisense oligos inhumans. Nine patients who had bcl-2 positive relapsednon-Hodgkin’s lymphoma received an 18-base phos-phothioated sequence over a 2-week period. No side-ef-fects were noted, and tumour response was recorded.

Bcl-2 and bcl-XL appear to act similarly in antagonis-ing apoptosis induced by cancer chemotherapeutic

Page 8: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205188

drugs and other chemicals. Ibrado et al. [113] foundthat overexpression of bcl-2 or bcl-XL in the humanacute myelogenous leukaemia line HL-60 inhibited acti-vation of one of the caspases believed to be involved inapoptosis (Caspase 3/CPP32/Yama/apopain); in thiscase, the apoptosis-inducing stimulus was cytosine ara-binoside. This group later [114] reported that the in-hibitory effect of bcl-XL may involve blockingcytochrome c release from mitochondria, and thus pre-venting caspase 3 activation. Similarly, Decaudin et al.[115] found that, in human leukaemia cell lines, overex-pression of bcl-2 or bcl-XL protected against chemi-cally-(VP-16, adriamycin, cytosine arabinoside,ceramide) induced disruption of mitochondrialtransmembrane potential (which may be a key step inprocaspase activation via release of mitochondrial me-diators such as cytochrome c). Decaudin et al., alsofound that while overexpression of the protease in-hibitor cytokine response modifier A, blocked Fas/Apo-1/CD95-mediated effects on mitochondrialtransmembrane potential and nuclear apoptosis, it hadno effect on the corresponding chemotherapy-inducedevents.

Simonian et al. [116] generated clones of FL5.12murine lymphoid cells with equivalent levels of bcl-2and bcl-XL expression (using constructs coding for theflag epitope, to facilitate monitoring of equal expressionlevels). Their assays involved continuous exposure totoxic agents, followed by viability assessment usingtrypan blue exclusion (which would, of course, identifycells only at a late stage in cell damage, when theplasma membrane becomes generally permeable). Withthis assay, their results indicated that bcl-XL providedsomewhat better protection than did bcl-2, againstchemotherapeutic agents that these authors classified asbeing specific for M phase (vincristine, vinblastine,VP-16, VM-26) and for S-phase (methotrexate, 5-FU,hydroxyurea) or which are non-phase specific (cis-platin). Bcl-XL and bcl-2 provided similar levels ofprotection against irradiation-induced apoptosis. (Irra-diation, like cisplatin, was classified as not cell cyclephase specific). On the other hand, Tu et al. [117] foundthat drug treatment of myeloma cells caused up-regula-tion of bcl-2 but not of bcl-X.

Huang et al. [68] have presented evidence that bcl-2,bcl-XL and adenovirus protein EIBI9 kDa are function-ally equivalent, can interact with Bax and Bak, and canblock cell death induced by many cytotoxic agents; inthe systems they examined, however, these proteinswere ineffective against death signals from the Fas orTNF receptors although in other systems Fas-inducedapoptosis can be blocked by bcl-2 [30]. Susin et al. [69]have shown that caspases, (which could be activateddirectly rather than via mitochondrial disruption inFas-activated pathways in certain situations or celltypes) can cause permeability transition and AIF re-

lease in isolated mitochondria. This is not inhibited bybcl-2, in contrast to permeability transition induced byceramide or peroxidants, and provides a partial modelfor understanding bcl-2-independent Fas-induced apop-tosis (but for other possibilities, see Section 5.1).

4.2. Enhancement of chemosensiti6ity in 6itro and ofcancer cell apoptosis in 6i6o by o6erexpression ofbcl-XS

Bcl-XS is the translation product of the short (alter-natively spliced) form of bcl-X mRNA. It exerts aneffect on apoptosis which is antagonistic to bcl-XL andbcl-2, serving as a dominant negative inhibitor of theiractivity. Bcl-XS cDNA transfectants of the humanbreast carcinoma line MCF-7 (which overexpresses bcl-XL) show a 5–10-fold increase to apoptosis induced invitro by VP-16 or taxol [118] Bcl- XS cDNA transfec-tion also enhanced sensitivity to adriamycin, vincristine,VP-16 and 5-FU in a low-level adriamycin pulse-se-lected resistant variant of the human lung carcinomacell line DLKP (R. NicAmhlaoibh and M. Clynes, inpreparation).

Transient adenovirus-vector mediated bcl-XS cDNAinfection induced apoptosis of MCF-7 cells in vitro,and also when injected intratumorally into MCF-7derived tumours in nude mice [119].

4.3. Role of bcl-2 in modulation of telomerase acti6ity

Telomerase activity is involved in maintaining telom-eres at the ends of chromosomes; loss of telomeres mayplay an important role in cellular senescence/differenti-ation, and telomerase activity is high in reproductivecells and most cancer cells. Mandal and Kumar [120]found that over-expression of Bcl-2 in human cervicalcarcinoma HeLa cells and in the human colorectalcarcinoma line DiFi led to a significant increase intelomerase activity. They also showed that IL-2 depri-vation of the cytotoxic T-cell line, CTLL-2, causeddown-regulation of both Bcl-2 and of telomerase activ-ity.

5. Other proteins which may modulate apoptosis

5.1. Bag-1 co-operates with Bcl-2 in promoting cellsur6i6al and also acti6ates Raf-1

Takayama et al. [121] used an interesting approach tosearching for new proteins which might interact withBcl-2. They screened a mouse embryo phage expressionlibrary with recombinant bcl-2, and then screened witha human-specific anti-Bcl-2 monoclonal antibody. Thisresulted in discovery of Bag-1 (Bcl-2-associated athano-gene 1, apparently named from the Greek

Page 9: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 189

athanos, anti-death). Bag-1 is not homologous to theBcl-2 family, but co-transfection of Bag-1 and Bcl-2into the human lymphoid line, Jurkat, enhanced protec-tion against apoptosis induced by staurosporine, anti-Fas antibody and cytolytic T-cells (in comparison tocells transfected with either cDNA alone). Takayama etal. [121] suggested that some pathways previously de-scribed as bcl-2/XL independent may not be indepen-dent, but may require co-expression of Bag-1 (Section4.1). Investigations using yeast two-hybrid systems andco-immunoprecipitation indicate that Bag-1 also inter-acts with Raf-1 (a protein kinase), although it shares nohomology with the previously-described Raf-1 activat-ing proteins Ras and 14-3-3 [122]. Overexpression ofeither Bcl-2 or Bag-1 (or, more potently, both together)blocks apoptosis caused by NGF withdrawal in PC12cells [92]. Bag-1 can occur, in human cells, in 36 and 50kDa isoforms (32 and 50 kDa in mice), due to alterna-tive transcription initiation sites. The lower M.W. formseems to be cytosolic, with the 50 kD form beinglocated in nuclei and mitochondria. Bag-1 is virtuallyidentical to RAP46, which interacts with activatedsteroid hormone receptors; it also binds to hepatocytegrowth factor receptors and PDGF receptor, enhancingtheir anti-apoptotic activity [123].

5.2. Bad can heterodimerise with Bcl-XL displacingBax and promoting cell death. Bad antagonises Bcl-XL

rather than Bcl-2 acti6ity

Bad [124] was discovered by using yeast two-hybridsystems and by expression library cloning. Bad antago-nises cell death inhibition by bcl-XL. It contains theBH1 and BH2 domains, but lacks the hydrophobicC-terminal signal anchor sequence which other familymembers use for membrane integration. Yang et al.[124] suggest that Bad in itself has little effect on theapoptotic pathway, but that it promotes cell death bydisplacing Bax from Bcl-XL: Bax heterodimers (whichpromote cell survival). They cite evidence that Bcl-2and bcl-XL which are mutated so that they no longerinteract with Bax, also lose their ability to block celldeath. They also suggest an analogy with the Myc,Max, Mad regulatory network [125], in which, forexample, while Myc:Max heterodimers are transcrip-tional activators, Max:Max or Mad:Max are believedto be transcriptional repressors [126].

Bcl-2 can sequester the protein kinase Raf-1 fromcytosol to the mitochondrial membrane, resulting inphosphorylation of BAD (which can in its unphospho-rylated form heterodimerise with Bcl-2 or Bcl-XL); thephosphorylated BAD dissociates from bcl-2 or bcl-XL

and moves into the cytosol in a complex with theRaf-1-interacting protein 14-3-3, and no longer inter-feres with bcl-2/bcl-XL activity [88]. BAD is also phos-phorylated by active Akt protein kinase in a PI

3-kinase-dependent, IL-3 stimulated pathway [59], asmentioned in Section 2.3.

5.3. Galectin

Galectin 3 is a beta-galactoside binding proteinwhich has recently been shown [127] to inhibit cisplatininduced apoptosis, without altering expression of Bcl-2,Bcl-XL or Bax. This protein contains the highly con-served NWGR amino acid sequence in the BH1 domainand mutations in this region abolish its ability to pro-tect against apoptosis.

5.4. MCL-1, BFL-1, sur6i6in, IAP-like protein

Mcl-1 [128,129], and bfl-1 [130] are bcl-2 homologueswhich suppress apoptosis. Their importance remains tobe established. A human gene, termed survivin, withhomology to the baculovirus IAP gene has been clonedrecently by [131]; it is widely expressed in tumours andin foetal tissues, but not in terminally differentiatedcells. It can (on transfection) antagonise IL-3-depriva-tion-induced apoptosis in lymphocytes, and may havean anti-apoptotic role in vivo. Another human IAP-likeprotein (hILP) may inhibit apoptosis at a step down-stream of mitochondrial events [132].

5.5. Bcl-XL/Bcl-2-interacting proapoptotic proteins

Recently-discovered proteins which may be impor-tant in bcl-XL activity include:� MRIT, (a caspase homologue-named from the San-

skrit word for death, which is one up on the Greekscholars-whose C-terminal lacks the caspase catalyticconcensus sequence, but which contains a DED,Section 2.4); MRIT, like CED-4, interacts with Bcl-XL and with caspases including FLICE and possiblycaspase 3 [133];

� p28 Bap 31, an integral endoplasmic reticulumprotein which interacts with Bcl-2, Bcl-XL and pro-caspase 8 (PRO-FLICE), this interaction beingblocked by Bax. Bcl-2/bcl-XL may inhibit a caspase-activation function of this protein [134];

� Harakiri, which interacts with Bcl-2 or Bcl-XL (butnot with Bax or Bak), and promotes apoptosis in abcl-2/bcl-XL inhibitable fashion [135];

� Bim is a pro-apoptotic protein, containing the BH3domain common to most of the bcl family; it existsin three isoforms (possibly due to alternative splic-ing) and its pro-apoptotic activity is antagonised bybinding of bcl-2, bcl-XL or bcl-w [136].The discovery of these proteins illustrates the com-

plexity of the cell’s apoptotic machinery, and indicatesthat release of mitochondrial mediators is only part ofthe overall picture; furthermore, the results of Li et al.[137] support an anti-apoptotic role of bcl-XL indepen-dent of cytochrome c release.

Page 10: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205190

6. Bcl-X expression in vivo

6.1. Expression of bcl-X in 6arious tissues, andphysiology of bcl-x, bcl-2- and bax-deficient mice

Krajewski et al. [94] used an anti-peptide polyclonalantiserum which detects both Bcl-XL and Bcl-XS toexamine, by immunohistochemistry, expression of Bcl-X in normal tissues in vivo. High level expression wasobserved in specific subsets of the haemopoietic system(including cortical lymphocytes, activated lymphocytes,and several bone marrow cell types), reproductive tis-sues, and a variety of epithelial cells. In general, expres-sion patterns of Bcl-X were different from that of Bcl-2,indicating that they may play different, non-redundantbut possibly similar roles in different tissues and atdifferent stages of differentiation; the differences infunction are not, however, clear at the moment.

Bcl-X-deficient mice [138] died around embryonicday 13, and widespread apoptosis was observed inpostmitotic immature neurons of developing brain,spinal cord and dorsal root ganglia. The lifespan ofimmature lymphocytes was also shortened. Bcl-2-knockout mice (lacking both Bcl-2a and Bcl-2b

proteins) were less severely affected; they had appar-ently normal embryonic development, but tended to dieprematurely and showed (to a variable degree) growthretardation, and exhibited abnormalities in kidney,spleen, thymus, lymphocytes, hair pigmentation andsmall intestine [139].

Bax-deficient mice were viable, displaying hyperplasiain thymocytes, B cells and ovaries, but abnormalities ofmale reproductive organs and associated sterility; theabnormalities in testes included hypoplasia and in-creased apoptosis. Obviously, the functions of genessuch as bax in the whole animal cannot be reduced tosimple positive or negative effects on apoptosis rates[140].

6.2. Expression of bcl-2, bcl-X and bax in tumours

As already discussed, bcl-XL appears to be widelyexpressed in normal tissues, [94]. Any cancer therapydirected against bcl-XL must take account of possibleside-effects on normal tissues, unless it can be targetedto the tumour tissue. Previous experience with modula-tors of P-170 indicates the types of problem which canarise from inhibition of a target tumour protein whichis also expressed in normal tissue [141–143].

There is little information available in the literatureabout expression of bcl-XL in tumours, and little clearevidence on correlation of bcl-XL expression with resis-tance to chemotherapy. This is in contrast with a fairlyextensive literature on bcl-2 (which of course was dis-covered much earlier). There is some evidence thatbcl-XL expression may be correlated with chemoresis-

tance in multiple myeloma [144] and that bcl-2, bcl-XL

and Bax may not be relevant to nitrogen mustard drugresistance in B-CLL [145].

Bcl-2 may be overexpressed in more than half of allhuman cancers (see [146] for review) and may be associ-ated with poorer prognosis in some lymphomas andleukaemias, and in prostate adenocarcinoma. Increasedexpression of bcl-2 may be associated with drug resis-tance in squamous cell lung carcinoma [147] and bcl-2is widely expressed in breast cancer [148,149]. Reed’sgroup [93] used polyclonal antibodies to specific peptidesequences to examine expression of bcl-XL and relatedproteins in primary colorectal adenocarcinomas andalso in adenomatous colonic polyps. Their results sug-gested a tendency for Bcl-XL expression to increase,and for Bcl-2 and Mcl-1 to decrease as differentiationstate decreased. Expression of the anti-apoptoticprotein Bak was also reduced in both adenomas andcarcinomas, while Bax expression did not show majorchanges.

The ratio between Bcl-XL (and related anti-apoptoticproteins) and Bax (or similar pro-apoptotic proteins)seems to be important in determining sensitivity tochemotherapy. Reduced expression of Bax (by im-munocytochemistry) is associated with poor responserates to chemotherapy in breast cancer [150]. The bcl-2/bax ratio showed good correlation with chemoresis-tance in acute myeloid leukaemia patients [151]. HighBax expression [152] relative to Bcl-2 may contribute tothe inherent chemosensitivity of testicular tumours.

It should be realised that resistance is often multi-functional [153], that tumour cell populations can bevery heterogenous with respect to resistance level andmechanism [154] and that some resistance mechanismsmay be dominant over others; therefore measurementof a single biochemical determinant, even one appar-ently so central as bcl-2/bcl-XL, may be unable topredict sensitivity to chemotherapy-induced apoptosisin patients. For example, bcl-2 expression per se wasnot a useful predictive marker of chemosensitivity innon-Hodgkins lymphoma [155] or chronic lymphocyticleukaemia [156,157] and bcl-2 was surprisingly reportedto be a good predictor of response in breast carcinomawhen analysed on its own [158,159].

7. Recent developments in understanding the role ofMRP in drug-resistance

7.1. MRP expression in cell lines and in humantumours

Since its initial discovery in the drug-resistant smallcell lung cancer cell line H69AR [160], MRP has beenshown to be present in multidrug resistant cell linesfrom a variety of tumor types. These tumors

Page 11: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 191

include leukaemias [161,162], fibrosarcoma [163], smallcell and non-small cell lung carcinomas [164–166],breast carcinoma [167,168], melanoma [169] and bladdercarcinoma [170,171]. There have also been reports ofco-expression of P-glycoprotein (Pgp) and MRP in anumber of drug-selected cell lines [172,173]. Cell linesoverexpressing MRP are typically cross-resistant to an-thracyclines, epipodophyllotoxins and some vinca alka-loids [174–176]. The resistance profiles of MRP and Pgpoverexpressing cell lines exhibit similarities, but theseprofiles differ, notably with regard to Taxol resistance[177] (see Section 7.4). The explanation for a cell devel-oping MRP rather than Pgp-mediated resistance duringdrug exposure remains unclear, but it is believed thatoverexpression of MRP may confer initial low levelresistance, while Pgp overexpression develops as higherlevels of resistance are required for survival [165,172].

The role of MRP expression in drug-resistance inhuman cancers in vivo remains uncertain, althoughpreliminary evidence suggests a correlation in neuroblas-toma [178] retinoblastoma [179] primary breast car-cinoma [167] and non-small cell lung cancer [180]. MRPis present in many normal tissues, but Flens et al. [181]reported that immunostaining was predominantly cyto-plasmic in normal cells, but predominantly plasmamembrane-located in malignant tissue. Althoughderived from data with cell lines rather than tumourtissue, the data presented by Berger et al. [169] stronglysupports a role for MRP in the intrinsic drug resistanceof melanoma.

7.2. MRP gene structure

The MRP gene has been localised to band p13.13-13.12 on chromosome 16 [160]. Sequence analysis per-formed by Zhu et al. [182] of a 2.2 kb 5%-flankingsequence of genomic MRP from HL60-ADR cells sug-gested the presence of regulatory elements in this region.These elements included AP-1, AP-2, SP-1, ERE (estro-gen response element), GRE (glucocorticoid responseelement) and CRE (cyclic AMP response element) sites.

Zhu and Center [182] discovered that the most signifi-cant region of promoter activity may be present in ahighly GC-rich region of 194 nucleotides. This regiondoes not contain a TATA box for facilitating site-spe-cific initiation of transcription. It contains multiple startsites for transcription, as is found in the case of Pgp[183]. The MRP gene has been shown to be amplified ina number of drug- selected cell lines [184,185]. In othercases MRP mRNA levels have been shown to be in-creased in the absence of gene amplification[186,187,160] and this is believed to be due to alterationsin transcriptional control of the MRP gene. The MRPgene encodes a message of 6.5 kb which has the potentialfor synthesis of the 1531 amino acid MRP protein.Transfection studies using full length MRP cDNA have

shown that MRP can confer vincristine and VP-16resistance but not cisplatin resistance to drug sensitiveHeLa cells [176] (see Section 7.4).

7.3. MRP protein structure

A number of models have been proposed [170] for thesecondary structure of the MRP protein. Hipfner et al.[188] demonstrated that MRP has an extracytosolic NH2

terminus and provided evidence to suggest that in theNH2 terminal domain, the membrane-spanning domainof MRP (MSD-1) contains five transmembrane (TM)segments. MSD-2 was predicted as possessing six TMsegments and MSD-3 was predicted as possessing eitherfour or six TM regions. Kast and Gros [189] alsoprovided evidence to suggest that MRP possesses 5 and6 TM regions in MSD-1 and MSD-2, respectively.Bakos et al. [190] presented a membrane topology modelof MRP which predicted that MSD-1 contained four orfive TM segments and that both MSD-2 and MSD-3each contained six TM segments. Other members of theABC (ATP-binding cassette) transporter family, such asthe pancreatic b-cell sulfonylurea receptor (SUR), theyeast cadmium resistance factor YCF1 and the MOAT(multispecific organic anion transporter) protein mayalso have topologies similar to MRP [191]. MRP may belocalised in the endoplasmic reticulum [192], post-Golgicytoplasmic vesicles and in the plasma membrane ofresistant cells [193].

The amino acid sequence of MRP contains a numberof sites known to be relevant for ATP binding andpost-translational modification [188,190,170]. It hasbeen demonstrated that the unmodified MRP proteinhas an apparent mass of 170 kDa and is processed intoa mature 190 kDa form by the addition of N-linkedoligosaccharides [191,193]. Human MRP is now be-lieved to contain 14 potential sites for N-linked glycosy-lation. Only three of these sites (Asn19, Asn23 andAsn1006) are believed to be glycosylated [188]. The effectsof glycosylation on MRP activity are not fully known.It has been demonstrated that tunicamycin-induced inhi-bition of glycosylation has little effect on the cellulardrug accumulation characteristics of MRP-transfectedcells [193,192].

The specific location and functional role of the phos-phorylated residues on MRP are currently unclear. InCFTR (cystic fibrosis transmembrane conductance reg-ulator) and Pgp (P-glycoprotein), phosphorylation oc-curs predominately in the linker region [194,195]. Thisis also believed to be the case for phosphorylation ofthe MRP protein, but comparison of the linker regionamino acid sequences of human and murine MRP haveshown that there is only a low level of conservation ofpotential PKC phosphorylation sites between these twohomologs. Human and murine MRP proteins share avery high proportion of conserved sequences

Page 12: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205192

[174,191,196]. MRP is phosphorylated on serineresidues only and the phosphate groups are containedin nine tryptic peptides [197]. The phosphorylation ofthese amino acids was shown to be blocked by com-pounds such as staurosporine and this inhibition ofphosphorylation may result in an increase in drugaccumulation in MRP overexpressing cells [197]. Theprotein kinases involved in the phosphorylation ofMRP have not been conclusively identified. There arepotential sites for phosphorylation of MRP by proteinkinase C, cyclic AMP/cyclic GMP-dependent proteinkinases and tyrosine kinases [193].

7.4. MRP transport acti6ity

The MRP transporter has the ability to transportcysteinyl leukotrienes such as leukotriene C4 (LtC4) andglutathione conjugates such as glutathione disulphide(GSSG), [198–200]. A wide variety of organic anionsincluding anionic conjugates of bile salts and steroidhormones have the potential to serve as physiologicalsubstrates of MRP [201]. Transport of heavy metaloxyanions by MRP was demonstrated by Cole et al.[177].

A relationship exists between MRP protein expres-sion and drug resistance in various MRP expressing celllines but the mechanism by which MRP confers resis-tance to chemotherapeutic drugs is currently unclear.MRP has the ability to transport glutathione conju-gates and so if a cytotoxic drug became conjugated toglutathione then this drug conjugate could serve as apotential substrate for transport by MRP. Glutathioneconjugation is not known to occur for natural productdrugs [202]. In addition, MRP transfectants do notexhibit increased resistance to alkylating agents, a classof drugs for which glutathione conjugation is known tooccur [175,177]. Although MRP has been indirectlyshown to reduce cellular accumulation of drugs such asadriamycin and daunorubicin [203,166], only a limitednumber of reports have described the direct transportof cytotoxic drugs by MRP. Studies carried out by Paulet al. [204] demonstrated that MRP isolated fromMRP-transfected NIH/3T3 fibroblasts could serve asan ATP-dependent pump for a variety of naturalproduct, unconjugated cytotoxic drugs including an-thracyclines, vincristine and etoposide. Paul et al. [205]demonstrated that MRP isolated from the HL60-ADRcell line actively transported daunorubicin, VP-16 andvincristine but not vinblastine or Taxol. Stride et al.[174] and Loe et al. [206] demonstrated that transportof vincristine by MRP occurred only in the presence ofglutathione.

The analysis of drug resistance profiles of MRP-over-expressing, drug-selected cell lines has generated infor-mation on the potential drug transporting abilities ofMRP [207,208]. This information is complicated by the

fact that many of these cell lines were obtained by astepwise selection in drug, which facilitates the develop-ment of multiple collateral resistance mechanisms. Thismay result in the simultaneous expression of numerousdrug resistance mechanisms which ultimately results indifficulties in interpreting the contribution of any indi-vidual mechanism to drug resistance levels [172,173].Studies carried out with MRP-transfected cell lines mayserve as more useful models for definition of the profileof drugs potentially transported by MRP. A studycarried out by Breuninger et al. [175] using MRPcDNA-transfected NIH/3T3 fibroblasts demonstratedthat these transfectants displayed increased resistanceto several lipophilic drugs including adriamycin,daunorubicin, VP-16, actinomycin D and vincristine,but negligible levels of resistance to vinblastine andTaxol, but were more sensitive to cisplatin (Table 1).The absence of high level resistance to Taxol in MRP-transfected cells is significant as this drug is a highaffinity substrate for transport by Pgp. In a similarstudy carried out by Cole et al. [177] it was found thatMRP-transfected cell populations possessed a moderatelevel resistance to adriamycin, daunorubicin, epirubicin,vincristine and VP-16. These cells exhibited only lowlevel resistance to Taxol, vinblastine and colchicine. Inaddition, these transfectants were not resistant to mi-toxantrone or cisplatin. The MRP-transfected cellswere also resistant to a selection of heavy metal anionsincluding arsenite, arsenate and trivalent and pentava-lent antimonials, but were not resistant to cadmiumchloride [177].

7.5. Modulation of MRP mediated drug resistance

The chemosensitisers verapamil and cyclosporin Aare significantly less effective at reversing drug resis-tance in MRP overexpressing cell lines, than is the casein Pgp overexpressing cell lines, [175,209,166]. It was

Table 1Summary of drug resistances of two MRP transfected cell lines

Fold resistanceaCytotoxic drug Fold resistanceb

5.6 9.4Adriamycin5.1Daunorubicin 7.8

Epirubicin N.D.c9.0Mitoxantrone 1.2 1.5

3.8Vincristine 8.44.3Vinblastine 1.2

11.411.6VP-16N.D.c1.1Colchicine

2.0Taxol 1.30.9 0.9cisplatin

2.3N.DcActinomycin D

a IC50 of MRP transfectant/IC50 of control transfectant ([177]).b IC50 of MRP transfectant/IC50 of control transfectant ([175]).c N.D.=Not done.

Page 13: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 193

shown by McGrath et al. [210] that MRP was notlabelled with a photoaffinity analogue of verapamil inthe MRP-overexpressing HL60-ADR cell line. Thissuggests that verapamil does not directly interact withMRP. In the case of Pgp, verapamil is known to binddirectly to the protein and interfere with transport inthis manner [211]. A number of compounds have beenidentified as having the ability to efficiently modulateMRP activity. The use of these compounds as modula-tors of MRP has been reviewed by Twentyman andVersantvoort [212]. The leukotriene D4 receptor antag-onist MK571 has been extensively characterised byGekeler et al. [213] in selected Pgp and MRP expressingcell lines. A complete reversal of vincristine resistancewas achieved by co-incubation with a non-toxic level ofMK571 in an MRP-expressing cell line. This compoundwas found to have no effect on the level of toxicityachieved in a Pgp-overexpressing cell line. Gekeler et al.[214] showed that the specific bisindolylmaleimideprotein kinase C inhibitor GF109203X totally reversedvincristine resistance in one MRP-overexpressing cellline. This compound only partially reversed adriamycinand vincristine resistance in a second MRP-overex-pressing cell line. The effect of this compound on drugresistance of a Pgp-overexpressing cell line was notexamined in this study [214]. Duffy et al. [215] demon-strated that a sub-set of NSAIDs including in-domethacin, sulindac and tolmetin enhanced drugtoxicity (in MRP-expressing cell lines) of drugs such asadriamycin, vincristine and VP-16 which are MRP sub-strates (but not of non-MRP-substrate drugs), probablyas a result of interference with MRP activity. Draper etal. [216] reported similar effects with indomethacin. Invivo efficacy of MRP-substrate drug /NSAID combina-tions has also been demonstrated in transplantablemurine tumours and human lung tumour xenografts(R. O’Connor, E. Moran and M. Clynes, unpublished).Consideration must obviously be given also to possiblesensitisation of normal tissues to anticancer drugs, ifMRP is inhibited; the studies of Wijnholds et al. [217]showing hypersensitivity to VP-16 in mrp−/− miceand of Lorico et al. [218] showing increased drugsensitivity in double-MRP-knockout embryonic stemcells, are relevant in this regard.

7.6. MRP as a member of the ATP-binding cassettesuperfamily

Members of the ATP-binding cassette (ABC) super-family contain at least one hydrophobic transmem-brane region and a cytoplasmic nucleotide bindingdomain (NBD). These proteins are involved in energydependent transport of a variety of substrates acrossmembranes. The ATP binding NBDs contain conservedresidues (Walker A and B motifs) spaced by 90–120amino acids (Walker C motif), [219]. A total of 21 new

genes from the human ABC superfamily were relativelyrecently identified, bringing the total number of charac-terised human ABC genes to 33 [220].

A number of human ABC proteins have been wellcharacterised and these include proteins involved indrug transport (Pgp and MRP) and peptide transport(TAP), as well as proteins such as CFTR (cystic fibrosistransmembrane conductance regulator) and SUR (sul-fonylurea receptor), [221–223]. MRP exhibits signifi-cant sequence homology to CFTR (19% amino acididentity), rat SUR (29% amino acid identity), YORI(33% amino acid identity), a yeast protein with theability to confer oligomycin resistance, ItpgpA (32%amino acid identity), a Leishmania protein involved inresistance to oxyanions and YCF1 (43% amino acididentity), a yeast protein involved in cadmium resis-tance [170].

As already described, MRP has the ability to trans-port a wide variety of hydrophobic anionic compounds.The multispecific organic anion transporter (MOAT) inliver canicular membranes and other tissues has similartransport abilities [224–226]. This suggested that theMRP-1 gene may have been responsible for cMOATprotein synthesis. The extremely low expression ofMRP-1 protein in the liver [227,160] and the predomi-nantly basolateral localisation of MRP-1 in the hepato-cyte membrane [228], indicated that the cMOATprotein was not a product of the MRP-1 gene.Paulusma et al. [229] provided evidence that MRP-1and cMOAT are encoded by two different genesnamely MRP-1 and MRP-2.

It has been shown that increased levels of GST(glutathione S-transferase) are involved in resistance toalkylating agents [230]. Ishikawa and Ali-Osman [231]demonstrated that GST-mediated cisplatin resistanceinvolves two steps. The initial step involves the forma-tion of a glutathione (GSH) S-conjugate and this isthen followed by the subsequent removal of the toxicconjugate from the cell by a glutathione conjugatepump (GS-X pump). Conjugation of cisplatin andGSH can occur non-enzymatically [231] but exportfrom the cell requires a GS-X pump [232].

The activity of a GS-X pump was shown by Ishikawaet al. [233] to be enhanced in cisplatin-resistant humanpromyelocyte leukemia HL60/R-CP cells. Muller et al.[200] found no increased GS-conjugate transport activ-ity in a cisplatin resistant lung cancer cell line, butfound a substantial increase in GS-conjugate transportactivity in the MRP-overexpressing GLC4/ADR cellline relative to the sensitive parental cells. Muller et al.[200] and Leier et al. [234] demonstrated that overex-pression of the MRP protein in cells resulted in in-creased ATP dependent GS-conjugate transport. Thecisplatin resistance conferred by the GS-X pump inHL60/R-CP cells was dependent on intracellular levelsof GSH in this cell line. The sensitivity of the vast

Page 14: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205194

majority of MRP overexpressing cell lines to cisplatinwas explained by Ishikawa et al. [187] as being a resultof insufficient intracellular levels of GSH in these cells.Indeed, not only the function of MRP-1, but alsoregulation of its expression may be closely correlatedwith glutathione metabolism. Gamma-glutamylcysteinesynthetase is coordinately upregulated with MRP-1 fol-lowing exposure to heavy metals, cisplatin or the bi-functional nitrosourea, ACNU; post-transcriptionalregulation may be involved [235,236]. In colorectalcarcinoma, coordinated overexpression of MRP-1 andgamma-glutamylcysteine synthetase is frequently ob-served [237].

Database screening identified three new homologuesof the MRP-1 and MRP-2 genes. The MRP-3 protein,like cMOAT is predominantly expressed in the liver,with MRP-4 expressed at very low levels in a numberof different tissues and MRP-5 expressed in almost alltissues analysed [207]. Preliminary investigations havealso been carried out to investigate the role of theseMRP homologues in drug resistance. MRP-4 was notover-expressed in any of the drug resistant cell linestested by Kool et al. [207]. The MRP-3 and MRP-5transporters were over-expressed in a limited number ofcell lines. Kool et al. [207] demonstrated that cMOATwas over-expressed in a number of drug resistant celllines and that mRNA levels correlated with cisplatinbut not adriamycin resistance. HepG2 cells transfectedwith cMOAT antisense cDNA were shown by Koike etal. [238] to exhibit increased sensitivity to cisplatin,vincristine and adriamycin. Further investigations arenecessary in order to investigate the exact contributionof each of the homologues of MRP to drug resistancein MRP expressing cells.

8. Clinical studies on reversal of multi-drug resistancein cancer

8.1. Introduction

The resistance of tumour cells to cytotoxicchemotherapy may be intrinsic or acquired. Tumoursare termed intrinsically resistant when they fail to re-spond to the initial chemotherapy schedule. Tumoursmay also acquire resistance to subsequent cytotoxictreatment presumably by mutation and/or gene induc-tion, and/or due to the survival of a resistant tumourfraction from the initially responsive tumour [239].Certain cancers are generally associated with acquiredor intrinsic resistance; for example, leukaemias havebeen shown to display acquired P-170-mediated resis-tance, whereas others, e.g. of colorectal origin, typicallyexpress P-170 and/or MRP constitutively without anyprior therapy. Attempts have been made to use currentknowledge of drug-resistance mechanisms to circum-

vent resistance and thereby extend patient survival.Agents which inhibit cancer resistance mechanisms aretermed resistance modulators, chemosensitising agents,resistance circumvention agents. As described below,the experience to date of clinical trials using inhibitorsof P-glycoprotein underlines the need for new pharma-cological agents and new cellular targets such as thosedescribed earlier in this review.

8.2. Problems associated with chemosensitisation

Resistance mechanisms seen in cancer cells are de-pendent on the ability of tumour cells to use innateprotective or adaptive responses which may also beexpressed by normal cells. Therefore, any inhibition ofthese mechanisms in resistant cancer may augment thetoxicity of cytotoxic therapy in normal tissues [240]. Todate, there are no significant reports of agents withparticular affinity for cancer cells versus normal tissue.Many modulators interfere with the normal pharma-cokinetic variables of standard chemotherapeuticagents [241]. Commonly, the peak plasma level (Cmax),area under curve (AUC) and elimination half life (t1/2)are all elevated, thus increasing the toxic burden on therest of the body and necessitating dose rescheduling ormore intensive patient monitoring [240,242]. For exam-ple Lum et al. [243] showed that cyclosporin signifi-cantly increased the AUC, and decreased the CL (totalrate of clearance) values of etoposide when co-adminis-tered. Additionally, many of the earlier clinical investi-gations specifically in the area of P-gp modulation usedsensitisers with relatively weak inhibitory efficiency.

8.3. Circum6ention of P-170 o6erexpression

One of the most studied mechanisms of resistance isthe 170 kDa protein pump termed P-glycoprotein (P-gp). This pump utilises the energy of ATP to activelyefflux a variety of agents including a broad spectrum ofcancer chemotherapeutic drugs [244]. The net effect ofthis efflux mechanism is to reduce the time a cell isexposed to the agent and to reduce its maximum intra-cellular concentration. Since most cytotoxic cancertherapies are given at or near the maximum tolerabledose, and have little selectivity for cancer cells, the dosegiven cannot be increased to compensate without be-coming toxic for the patient. P-gp can pump a widevariety of structurally dissimilar agents including manyof the more effective, first-line anti-cancer drugs suchas adriamycin and vinca alkaloids [240]. If cells alreadyexpress a high level of P-gp activity they will be intrin-sically resistant to the cytotoxic effects of these agents.Likewise, if P-gp expression is induced in a cancer cellby initial cytotoxic exposure, all of the other substrateagents will be less effective if given subsequently [240].Because P-gp was the earliest resistance

Page 15: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 195

Table 2Selected reports of clinical trials involving P-gp inhibition

Authors Toxicity findingsClinical protocol drugs and tumour type

[247] 24 Patients, Cyclosporin A and epidoxorubicin, Colorectal Increased incidence of leucocytopenia; no significant effect onoutcomecancer

[276] High incidence of serious cardiotoxicity; no objective responsesEight patients; verapamil and adriamycin; ovarian cancerSeven paediatric patients; verapamil, etoposide and vin-[248] Cardiac and haematological toxicty evident; partial responses

but no improvement on overall outcomeblastine; various refractory malagnancies20 Paediatric patients; verapamil and etoposide; various re-[276] Lower toxicity; six partial responsesfractory malagnancies16 Female patients; trifluoperazine and vinblastine; refractory[251] Significant CNS toxicity; similar response rate to controlbreast cancer

[252] 59 Patients; verapamil and VAD (vincristine, doxorubicin and No increased toxicity; no beneficial effect versus controldexamethasone); resistant melanoma18 Patients; verapamil and CVAD (cyclophosphamide vin- Significant but manageable toxicity; high (72%) response rate[277]cristine adriamycin and dexamethasone); drug refractorylymphoma16 Patients; cyclosporine A and teniposide; metastatic renal[278] Haematopoietic toxicity; one minor responsecell carcinoma

mechanism studied, it is also the first mechanism to betargeted by clinical strategies aimed at returning normaldrug sensitivity to the cancer cell. More recently, avariety of other cellular pump resistance mechanismshave been described. These include the multiple drugresistance protein (MRP), which is an ATP dependentpump with a similar, but not identical, substrate menuto that of Pgp (Section 7).

The intense investigation of P-gp has led to theidentification of a broad range of chemicals that modu-late its activity. They have been classed into sevenbroad categories: (1) calcium channel blockers; (2) Im-munosupressive agents; (3) vinca alkaloid analogues;(4) steroidal agents; (5) cyclosporin analogues; (6)calmodulin antagonists; and (7) a miscellaneous collec-tion of hydrophobic agents [245,246]. In many cases theinhibitory action of these agents is more an experimen-tal curiosity than a useful effect, since the concentra-tions at which MDR modulation is evident are greaterthan the obtainable therapeutic levels. Many of theagents also have significant therapeutic effect or toxicityin their own right that precludes their clinical use asP-gp modulators.

Several clinical studies of P-gp modulators in combi-nation with chemotherapeutic regimes have been re-ported or are underway [247–252] (Table 2). There arelimited claims for measurable increases in the anti-cancer effect using P-gp modulation in a chemothera-peutic combination to treat some haematologicalmalignancies. However, overall the results have beendisappointing and the studies have not mirrored thesuccess seen with MDR modulation in vitro. The rea-sons for this poor therapeutic activity are not simplydue to problems with the direct inhibition of drugefflux; pharmacokinetic and other considerations alsolimit therapeutic use.

As one might anticipate from the broad spectrum ofagents that affect P-gp activity, several forms of modu-lation are thought to occur. Most agents appear toblock the P-gp pump competitively [253]. However,differences are seen in the pump affinity for varyingcytotoxic agents under the modulation of particularsensitisers. This suggests the possibility of distinctbinding sites for certain P-gp substrates [254–256]. Theamount of drug required for P-gp modulation variesvery significantly among the list of agents. Some in-hibitors such as the antibiotics erythromycin and tetra-cycline are active only under in vitro experimentalconditions as the drug concentrations required aregreatly in excess of those attainable in the body [246].The ability to obtain physiologically relevant levels ofthe modulator significantly limits the number of agentsavailable. It has been suggested that this problem canbe overcome by combining modulators with non-over-lapping toxicities [254], but this does not seem to havebeen investigated clinically. These concentrations mustalso be sustainable for a suitable length of time so theP-gp-mediated drug efflux can be blocked over theduration of chemotherapy.

Historically, the screening for P-gp modulators hastended to use existing pharmacological agents. As aresult, most of these agents have strong pharmacologi-cal actions distinct from their activity on the effluxpump. In many cases these actions reduce the overalldose or the duration of that dose which can be givenwithout very significant side effects. The two mostcommonly used modulators, verapamil and cyclosporingenerally cause the side effects of cardiac abnormali-ties, circulatory problems, constipation, headache [249]and potent immunosupression, nephrotoxicity, muscu-loskeletal pain, nausea and vomiting, respectively [247].

Page 16: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205196

As with any pharmaceutical, P-gp modulators canhave toxic effects. The presence of additional toxicchemotherapeutic agents increases the susceptibility tothese toxic effects. P-gp is expressed in normal tissuesincluding the liver, kidney and brain capillary endothe-lial cells (the blood brain barrier) and has an activerole in protecting the body from the effects of xenobi-otics [257,258]. The non-selective body-wide nature ofthe P-gp inhibition caused by these agents renders thebody more sensitive to the direct toxicity of the anti-cancer drugs being administered. Additional cytotoxicdose refinement may be required due to inhibited drugelimination mechanisms and direct modification of theusual pharmacological profile of the anti-cancer agent.

Several studies have shown that the correlation be-tween P-gp and tumour response is quite variable be-tween patients and different forms of cancer [259]. Thisfinding may be partly explained by the observationthat the relative activity of P-gp can vary betweentumour cells [260,261].

Additionally, since several resistance mechanismsmay be operational at the same time in a given tu-mour, the ideal approach would be to identify thepresence or absence of the P-gp protein and to quan-tify the level of its impact on the resistance of atumour before deciding on a particular chemothera-peutic regime [262].

Agents such as verapamil and cyclosporin which arepotent in vitro and which can reach sufficientchemosensitising concentrations in vivo have markedpharmacological effects as already described. There-fore, more recent approaches have utilised analogueswith similar or greater potency while largely lackingthe additional pharmacological actions regarded as sideeffects in this application of therapy. Several differentanalogues of verapamil have been produced and testedin vitro. Most notably the R-isomer of verapamil hasbeen brought to the stage of clinical testing as it largelylacks the calcium channel blocking capacity of theS-isomer while still efficiently blocking P-gp efflux[263]. A more potent P-gp modulating cyclosporinderivative, PSC 833, with reduced immunotoxicity, isalso now in clinical trial stage [264,240]. There is noclear evidence so far that either of these drugs willmake a very significant contribution to treating pa-tients with resistant tumours.

8.4. Resistance due to DNA repair mechanisms

There are a few reports of attempts to overcomesome forms of resistance due to the overexpression ofDNA repair mechanisms. Willson et al. [265], reportedthe results of a clinical trial using streptozotocin todeplete the level of the resistance enzyme O6-alkylgua-nine-DNA alkyltransferase (AT) in resistant colon andrectal cancer. However, although the dose of modula-

tor administered depleted AT levels, increased toxicitywas evident without significant increases in therapeuticresponse.

8.5. New strategies to o6ercome drug resistance

8.5.1. RibozymesA paper by Holm et al. [266], indicated that cultures

of resistant cells could be rendered more sensitive bytransfecting with an MDR-1-specific ribozyme, whichselectively destroyed the mRNA for P-gp in cancercells in vitro. Other authors (including our own group)have shown similar results; however, clinical use of thistechnology still awaits a suitable delivery system for invivo use [267]. The use of antisense agents and ri-bozymes for MDR modulators has been reviewed re-cently by Byrne et al. [268].

8.5.2. CytokinesRecent in vitro data indicates that certain cytokines

may also have activity as resistance modulating agentsboth directly inhibiting the mechanisms and indirectlyby affecting the expression of certain genes leading tothe emergence of resistance to chemotherapy [269,270].However, clinical application of this finding is likely tobe difficult due to the inherent biological actions ofthese agents in a variety of other cellular systems in thebody.

8.6. Conclusion

Based on the clinical findings with MDR modulationto date we can draft a set of specifications for an agentthat would be therapeutically most useful to modulateMDR. The ideal agent should inhibit the resistancemechanism at a low, and readily physiologicallyachievable, concentration. It should display some levelof selectivity for cancerous tissue over normal tissue tomaintain the protective mechanisms in non-cancerouscells while producing a more toxic effect in the tumour.If selectivity is achievable, the ideal agent should po-tently and irreversibly inhibit the resistance mechanismin the tumour cell for the duration of chemotherapy orlonger. The agent should be able to permeate throughall cells in a tumour at sufficient concentration to beeffective in modulating resistance. The agent shouldhave no significant effect on the pharmacokinetics ofthe anti-cancer drugs either directly or through inter-fering with the normal cytotoxic elimination processes.On its own this novel chemosensitiser should be rela-tively non-toxic and free from other pharmaceuticalactivity. It should be available as a pure chemical withwell-described, consistent pharmacokinetics. The toxic-ity profile of this agent should be distinct from that ofthe chemotherapeutic partner so there would not be a

Page 17: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 197

need to reduce the dose of anti-cancer agent. It shouldalso not increase the toxicity of the chemotherapy inthe rest of the body, allowing the maximum chemother-apeutic dose to be used. In particular liver, kidney,cardiac, nervous and immune functions should not berendered more sensitive. Combination therapy withsuch an ideal agent should be considered as a first linetreatment to stem the emergence of acquired resistanceafter the initial dose of chemotherapy.

The initial trials of chemosensitising agents werelargely unsuccessful due to a complex set of factors.These included the use of modulators with low potency,drug-induced alterations in the pharmacokinetics of thecancer therapy, overt toxicity and other pharmacologi-cal effects of the modulators and the multifunctionalnature of cancer resistance. However, research in thisfield continues with clinical trials of more potent P-gpmodulating agents and a collection of promising invitro ideas which await extrapolation into the patient.

9. Conclusion/prospects

The inherent genetic instability of cancer cells conferson them the ability to generate variants exhibiting atremendous variety of resistance mechanisms to toxicagents [27]. Furthermore, the mechanisms by whichnormal cells protect themselves against toxins can berapidly recruited by cancer cells to protect them againstdrugs. In spite of these problems, chemotherapy curesmany cancers, and it is reasonable to hope that as weunderstand in more detail the mechanisms of resistance,we should be able to devise strategies so that more andmore cancer patients can be cured.

In spite of the significant expansion in our knowledgeof the P-170 efflux pump, clinical trials involving use ofP-170 modulators have produced disappointing results,due at least in part to enhanced toxicity to normalP-170-expressing tissues [272]. Many unanswered ques-tions remain-the circumventing agents may have af-fected other cellular activities; perhaps more specificP-170 inhibitors will be better? Could modulators bedirected to the tumour cells, and not to normal tissues?Or, must we use drugs not affected by P-170, in Pgp-positive tumours?

It is possible that inhibitors of other pumps (e.g.MRP [273,274,275]) may have fewer side-effects, butdata is as yet insufficient to reach any conclusions. Thefrequency of MRP-positive tumours (and in which P-170 does not play a significant role) remains unclear.The role of more recently-discovered proteins (such asLRP) in MDR is still not established; they could con-ceivably be future targets for modulation.

If it turns out to be the case that many/all anticancerdrugs converge on a (few) final death/apoptosis path-way(s), steps in these pathways will be promising

targets for new chemotherapeutic agents. These stepscould, unfortunately, also represent mechanisms forcancer cells to develop resistance to a very wide rangeof drugs; this may already be part of the clinicalproblem, where the multidrug resistance observed inpatients often seems to be wider than can be explainedby known MDR mechanisms (P-170, MRP, Topoiso-merase II). On the positive side, alternative redundantpathways may represent an opportunity for new combi-nation chemotherapy approaches to overcome resis-tance and the closer the targets are to the final celldeath committment steps (caspase activation?) thefewer downstream chances exist for additional resis-tance mechanisms. Obviously, once again, if ways couldbe found to selectively stimulate apoptosis in tumourcells versus normal tissue (either by physical targettingor by exploiting any differentially expressed pathwaysor proteins), the prospects for cure could be enhancedsignificantly.

There may also be a subset of tumours for whichmolecular approaches alone will not overcome resis-tance, and in which therapies must address drug pene-tration/vascularisation/ oxygenation.

In principle, based on our knowledge to date, atumour could exhibit multiple drug resistance due to acombination of all of the following:1. Poor drug supply/oxygenation2. Over-expression of drug efflux and sequestration

mechanisms (P-170, MRP)3. Altered multidrug targets (e.g. topoisomerase)4. Resistance to apoptosis (e.g. by overexpression of

anti-apoptotic proteins such as bcl-2 or bcl-XL)5. Enhanced degradation or impaired activation of

drugs6. Altered cell cycle properties

Tackling such a worst-case scenario could be virtu-ally impossible, but if chemotherapy can be made moreeffective at the first phase, the tumour should not havetime to co-express these multiple mechanisms; manyrounds of different types of chemo- and radio-therapymight encourage development of such super-resistanttumours.

The realistic target of MDR research may thereforebe to identify the range of mechanisms operating indifferent kinds of pre-treatment tumours; to developreliable diagnostic and therapeutic strategies associatedwith these mechanisms; and to start optimized treat-ment, based on the resistance mechanisms expressed,immediately. The disappointing progress to date inclinical application of MDR research reflects the inher-ent complexity of the problem, and should not discour-age researchers from pursuing the valuable andachievable goal of rational mechanism-based therapy ofmulti-drug resistant cancer.

Page 18: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205198

Acknowledgements

We are grateful to Yvonne Reilly for patientlyretyping the many versions of this manuscript.

Reviewer

This paper was reviewed by Toshihisa Ishikawa,Ph.D., Manager, Medicinal Biology Laboratory, PfizerInc. Central Research.

References

[1] Roninson IB, editor. Molecular and Cellular Biology of Multi-drug Resistance in Cancer Cells. New York: Plenum, 1991.

[2] Clynes M, editor. Multiple Drug Resistance in Cancer: CellularMolecular and Clinical Approaches, Dordrecht: Kluwer, 1994.

[3] Teicher BA, editor. Drug Resistance in Oncology. New York:Marcel Dekker, 1993.

[4] Arancia, G., Calcabrini, A., Meschini, S., Molinari, A. Intracel-lular Distribution of anthracyclines in Drug Resistant Cells.Cytotechnology 1988 (in press).

[5] Cleary I, Doherty G, Moran E, Clynes M. The multidrug-resis-tant human lung tumor cell line DLKP-A10 expresses novel drugaccumulation a and sequestration systems. Biochem Pharmacol1997;53:1493–502.

[6] Croop J. P-Glycoprotein Structure and evolutionary homologies.Cytotechnology 1998 (in press).

[7] Germann U, Chambers TC. Molecular Analysis of the MultidrugTransporter, P-Glycoprotein. Cytotechnology 1998 (in press).

[8] Lehnert M. Clinical multidrug resistance in cancer: a multifacto-rial problem. Eur J Cancer 1996;32A:912–20.

[9] Kickhoefer VA, Rome LH. The sequence of a cDNA encodingthe major vault protein from Rattus nor6egicus. Gene1994;151:257–60.

[10] Izquierdo MA, Scheffer GL, Schroeijers AB, Scheper RJ. Vault-related resistance to anticancer drugs determined by the expres-sion of the major vault protein LRP. Cytotechnology 1998 (inpress).

[11] Chauffert B, Dimanche-Boitrel MT, Garrido C, Martin M,Martin F, Solary E. New Insights into the kinetic resistance toanticancer agents. Cytotechnology 1998 (in press).

[12] St. Croix B, Florenes V, Rak JW, Flanagan M, Bhattacharya N,Slingerland JM, Kerbel RS. Impact of the cyclin-dependentkinase inhibitor p27-Kip1 on resistance of tumour cells toanticancer agents. Nat Med 1996;2:1204–10.

[13] Rumsby MG, Drew L, Warr JR. Protein kinases and multidrugresistance. Cytotechnology 1998, (in press).

[14] Hofmann GA, Mattern MR. Topoisomerase II in multiple drugresistance. Cytotechnology 1993;12:137–54.

[15] Beck WT, Chen M, Khelifa T, Kusumoto H, Mo YY, Wang Q.Cytotoxic signalling pathways and G2/M arrest as novelchemotherapeutic targets revealed by topoisomerase inhibitors.Anti-Cancer Drugs 1996;7:75–82.

[16] Pessina A. Topoisomerase I in multiple drug resistance. Cyto-technology 1993;12:127–36.

[17] Pessina A, Parchment RE. Topoisomerase I inhibitors and drugresistance. Cytotechnology 1998 (in press).

[18] Moscow JA, Dixon KH. Glutathione-related enzymes, glu-tathione and multidrug resistance. Cytotechnology 1993;12:155–70.

[19] O’Brien ML, Tew KD. Glutathione and related enzymes inmultidrug rresistance. Eur J Cancer 1996;32A:967–78.

[20] Buschfort C, Muller MR, Seeber S, Rajewsky MR, Thomale J.DNA excision repair profiles of normal and leukemic humanlymphocytes: functional analysis at the single-cell level. CancerRes 1997;57:651–8.

[21] D’Incalci M, Damia G. Mechanisms of resistance to alkylatingagents. Cytotechnology 1998 (in press).

[22] Reed E. Nucleotide Excision Repair and Anticancer chemother-apy. Cytotechnology 1998 (in press).

[23] Mattern J, Volm M. Role of oxygenation and vascularisation inDrug Resistance. Cytotechnology 1998 (in press).

[24] Takamiya Y, Abe Y, Tanaka Y, Tsugu A, Kazuno M, OshikaY, Maruo K, Ohnishi Y, Sato O, Yamazaki H, Kijima H,Ueyama Y, Tamaoki N, Nakamura M. Murine P-glycoproteinon stromal vessels mediates multidrug resistance in intracerebralglioma xenografts. Br J Cancer 1997;76:445–50.

[25] Marks DC, Su GM, Davey RA, Davey MW. Extended multidrugresistance in haemopoietic cells. Br J Haematol 1996;95:587–95.

[26] Tsujimoto Y, Croce CM. Analysis of the structure, transcriptsand protein products of bcl-2, the gene involved in humanfollicular lymphoma. Proc Natl Acad Sci USA 1986;83:5214–8.

[27] Craig RW. The BCL-2 gene family. Sem Cancer Biol 1995;6:35–43.

[28] Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemtopoi-etic cell survival and cooperates with c-myc to immortalize pre-Bcells. Nature 1988;335:440–2.

[29] Dole M, Nunez G, Merchant AK, Maybaum J, Rode CK, BlochCA, Castle VP. bcl-2 inhibits chemotherapy-induced apoptosisin neuroblastoma. Cancer Res 1994;54:3252–9.

[30] Weller M, Malipiero U, Aguzzi A, Reed JC, Fontana A.Proto-oncogene bcl-2 gene transfer abrogates Fas/APO-1 anti-body-mediated apoptosis of human malignant glioma cells andconfers resistance to chemotherapeutic drugs and therapeuticirradiation. J Clin Invest 1995;95:2633–43.

[31] Kitada S, Takayama S, De Riel K, Tanaka S, Reed JC. Reversalof chemoresistance of lymphoma cells by antisense-mediatedreduction of bcl-2 gene expression. Antisense Res Dev 1994;4:71–9.

[32] Ghia P, Boussiotis VA, Schultze JL, Cardosa AA, Dorfman DM,Gribben JG, Freedman AS, Nadler LM. Unbalanced Expressionof Bcl-2 family proteins in follicular lymphoma: contribution ofCD40 signalling in promoting survival. Blood 1998;91:244–51.

[33] Stewart BW. Mechanisms of apoptosis; integration of genetic,biochemical and cellular indicators. J Natl Cancer Inst1994;86:1286–96.

[34] Jacobson MD, Weil M, Raff MC. Programmed cell death inanimal development. Cell 1997;88:347–54.

[35] Mashima T, Naito M, Nuguchi K, Miller DK, Nicholson DW,Tsuruo T. Actin cleavage by CPP32/Apopain during the devel-opment of apoptosis. Oncogene 1997;14:1007–12.

[36] Seimiya H, Mashima T, Toho M, Tsuruo T. C-Jun NH2-terminalkinase-mediated activation of interleukin-1-beta-converting en-zyme/CED-3 like protease during anticancer drug-induced apop-tosis. J Biol Chem 1997;272:4631–6.

[37] Davis RJ. The mitogen-activated protein kinase signal transduc-tion pathway. J Biol Chem 1993;268:14553–6.

[38] Liu X, Gupta AK, Corry PM, Lee YJ. Hypoglycemia inducedc-Jun phosphorylation is mediated by c-Jun N-terminal kinase1 and Lyn kinase in drug-resistant human breast carcinomaMCF-7/ADR cells. J Biol Chem 1997;272:11690–3.

[39] Kharbanda S, Ren R, Pandey P, Shafman TD, Feller SM,Weichselbaum RR, Kufe DW. Activation of the C-Ab1 ty-rosine kinase in the stress response to DNA damaging agents.Nature 1995;376:785–8.

[40] Kharbanda S, Bharti A, Pei D, Wang J, Pandey P, Ren R,Weichselbaum R, Walsh CT, Kufe D. The stress response to

Page 19: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 199

ionizing radiation involves C-Ab1-dependent phosphorylationof SHPTP1. Proc Natl Acad Sci USA 1996;93:6898–901.

[41] Skorski T, Nieborowska-Skorska M, Wiodarski P, Perrotti D,Hoser G, Kawiak J, Majewski M, Christensen L, Iozzo RV,Calabretta B. Treatment of Philadelphia leukemia in severecombined immunodeficient mice by combination of cyclosphos-phamide and bcr/abl antisense oligodeozynucleotides. J NatlCancer Inst 1997;89:124–33.

[42] Tari AM, Arlinghaus R, Lopez-Berstein G. Inhibition of Grb2and Crk1 proteins results in growth inhibition of Philadelphiachromosome positive leukemic cells. Biochem Biophys ResCommun 1997;235:383–8.

[43] Saleem A, Datta R, Yuan ZM, Kharbanda S, Kufe D. Involve-ment of stress-activated protein kinase in the cellular responsesto 1-beta-D-arabinofuranosylcytosine and other DNA damag-ing agents. Cell Growth Differ 1995;6:1651–8.

[44] Osborn MT, Chambers TC. Role of the stress-activated c-JunNH2-terminal protein kinase pathway in the cellular responseto adriamycin and other chemotherapeutic drugs. J Biol Chem1996;271:30950–5.

[45] Yang X, Khosravi-Far R, Chang HY, Baltimore D. Daxx, anovel fax-binding protein that activates JNK and apoptosis.Cell 1997;89:1067–76.

[46] Graus-Porta D, Beerli RR, Daly JM, Hynes NE. ErbB-2, thepreferred heterodimerisation partner of all ErbB receptors, is amediator of lateral signalling. EMBO J 1997;16:1647–55.

[47] Gusterson BA, Gelber RD, Goldhirsch A, Price KN, Save-Rudenstam C, Anbazhagan R, Styles J, Grigolato P, GolouhR, Reed R, Martinez-Tello F, Tiltman A, Torhurst J, GrigolatoP, Bettelheim R, Neville AM, Bunci K, Castiglione M, CollinsJ, Lindtner J, Senn H, Ludwig IBCSG. Prognostic importanceof C-erbB-2 expression in breast cancer. J Clin Oncol1992;10:1049–56.

[48] Klijn JG, Berns EM, Bontenbal M, Foekens J. Cell Biologicalfactors associated with the response of breast cancer to systemictreatment. Cancer Treat Rev 1993;19:45–63.

[49] Tetu B, Brisson J. Prognostic significance of HER-2/neu onco-protein expression in node-positive breast cancer. The influenceof the pattern of immunostaining and adjuvant therapy. Cancer1994;73:2359–65.

[50] Muss HB, Thor AD, Berry DA, Kute T, Liu ET, Koerner F,Cirrincione CT, Budman DR, Wood WC, Barcos M, Hender-son IC. C-erbB2 expression and response to adjuvant therapyin women with node-positive early breast cancer. New Engl JMed 1994;330:1260–6.

[51] Bacus SS, Zelnik CR, Plowman G, Yarden Y. Expression ofthe erbB-2 family of growth factor receptors and their ligandsin breast cancer. Am J Clin Pathol 1994;102(Suppl. 1):13–23.

[52] Bottini A, Berruti A, Bersiga A, Brunelli A, Brizzi MP, Di-Marco B, Cirillo F, Bolsi G, Bertoli G, Alquati P, Dogliotti L.Effect of neoadjuvant chemotherapy on Ki67 labelling index,c-erbB2 expression and steroid hormone receptor status inhuman breast tumour. Anticancer Res 1996;16:3105–10.

[53] Liu B, Sun D, X1A W, Hung MC, Yu D. Cross-reactivity ofC219 Anti-P-170-mdr-1 antibody with p185-c-erbB2 in breastcancer cells: cautions on evaluating P-170-mdr-1. J Natl CancerInst 1997;89:1524–9.

[54] Tsai CM, Yu D, Chang K-T, Wu L-H, Perng RP, Ibrahim NK,Hung MC. Enhanced chemoresistance by elevation of p185-neulevels in HER-2/neu transfected human lung cancer cells. J NatlCancer Inst 1995;87:682–4.

[55] Yu D, Liu B, Tan M, Li J, Wang SS, Hung M-C. Overexpres-sion of C-erbB2/neu in breast cancer cells confers increasedresistance to Taxol via mdr-1-independent mechanisms. Onco-gene 1996;13:1359–65.

[56] Mendelsohn J, Fan Z. Epidermal growth factor receptor familyand chemosensitization. J Natl Cancer Inst 1997;89:341–3.

[57] Dixit M, Yang JL, Poirier MC, Price JO, Andrews PA, ArteagaCL. Abrogation of cisplatin-induced programmed cell death inhuman breast cancer cells by epidermal growth factor antisenseRNA. J Natl Cancer Inst 1997;89:365–73.

[58] Howlett AR, Bailey N, Damsky C, Peterson OW, Bissel MJ.Cellular growth and survival are mediated by B1 integrins innormal human breast epithelium but not in breast carcinoma. JCell Sci 1995;108:1945–57.

[59] del Peso L, Gonzalez-Garcia M, Page C, Herrera R, Nunez G.Inter-leukin-3-induced phosphorylation of BAD through theprotein kinase Akt. Science 1997;278:687–9.

[60] Rodeck U, Jost M, DuHadaway J, Kari C, Jensen PJ, Risse B,Ewert DL. Regulation of Bcl-XL expression in human kerati-nocytes by cell-substratum adhesion and the epidermal growthfactor receptor. Proc Natl Acad Sci USA 1997;94:5067–72.

[61] Kumar A, Commane M, Flickinger TW, Horvath CM, StarkGR. Defective TNF-a-induced apoptosis in STAT-1-Null cellsdue to low constitutive levels of caspases. Science1997;278:1630–2.

[62] Hoey T. A new player in cell death. Science 1997;278:1578–9.[63] Scanlon KJ, Jiao L, Funato T, Wang W, Tone T, Rosi JJ,

Kashani-Sabet M. Ribozyme-mediated cleavage of c-fosmRNA reduces gene expression of DNA synthesis enzymes andmetallo-thionein. Proc Natl Acad Sci USA 1991;88:10591–5.

[64] Scanlon KJ, Ishida H, Kashani-Sabet M. Ribozyme-mediatedreversal of the multidrug resistant phenotype. Proc Natl AcadSci USA 1994;91:11123–7.

[65] Kashani-Sabet M, Funato T, Florenes VA, Fodstad O, ScanlonKJ. Suppression of the neoplastic phenotype in vivo by ananti-ras ribozyme. Cancer Res 1994;54:900–2.

[66] Bouffard DY, Ohkawa T, Kijlma H, Irie A, Suzuki T, CurcioLD, Holm PS, Sassani A, Scanlon KJ. Oligonucleotide modula-tion of multidrug resistance. Eur J Cancer 1996;32A:1010–8.

[67] Nagata S. Apoptosis by death factor. Cell 1997;88:355–65.[68] Huang DC, Cory S, Strasser A. Bcl-2, bcl-XL and adenovirus

protein EIB19 kD are functionally equivalent in their ability toinhibit cell death. Oncogene 1997;14:405–14.

[69] Susin SA, Zamzami N, Castedo M, Daugas E, Wang HG,Geley S, Fassy F, Reed JC, Kroemer G. The central execu-tioner of apoptosis: multiple connections between protease acti-vation and mitochrondria in Fas/APO-1/CD95-andceramide-induced apoptosis. J Exp Med 1997;186:25–37.

[70] Niehans GA, Brunner T, Frizelle SF, Liston JC, Salerno CT,Knapp DJ, Green DR, Kratzke RA. Human lung carcinomasexpress fas ligand. Cancer Res 1997;57:1007–12.

[71] Rokhlin OW, Bishop GA, Hostager BS, Waldschmidt TJ,Sidorenko SP, Pavloff N, Kiefer MC, Umansky SR, GloverRA, Cohen MB. Fas-mediated apoptosis in human prostaticcarcinoma cell lines. Cancer Res 1997;57:1758–68.

[72] Micheau O, Solary E, Hammann A, Martin F, Dimanche-Boitrel M-T. Sensitisation of cancer cells treated with cytotoxicdrugs to Fas-mediated cytotoxicity. J Natl Cancer Inst1997;89:783–9.

[73] Villunger A, Egle A, Kos M, Hartmann BL, Geley S, Kofler R,Greil R. Drug-induced apoptosis is associated with enhancedFas (Apo-1/CD95) ligand expression but occurs indpendentlyof Fas (Apo-1/CD95) signalling in human T-acute lymphaticleukaemia cells. Cancer Res 1997;57:3331–4.

[74] Debatin KM. Cytotoxic drugs, programmed cell death, and theimmune system: defining new roles in an old play. J NatlCancer Inst 1997;89:750–1.

[75] Nicholson DW, Thornberry NA. Caspases: killer proteases.Trends Biochem Sci 1997;22:299–306.

[76] Villa P, Kaufmann S, Earnshaw W. Caspases and caspaseinhibitors. Trends Biochem Sci 1997;22:388–93.

Page 20: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205200

[77] Gu Y, Sarnecki C, Aldape RA, Livingston DJ, Su MS. Cleav-age of Poly (ADP-ribase) polymerase by interleukin-1-betaconverting enzyme and its homologs TX and Nedd-2. J BiolChem 1995;270:18715–8.

[78] Mashima T, Naito M, Kataoka S, Kawa H, Tsuruo T. Aspar-tate-based inhibitor of interleukin-1-beta converting enzymeprevents antitumor agent-induced apoptosis in human myeloidleukemia U937 cells. Biochem Biophys Res Commun1995;209:907–15.

[79] Naito M, Nagashima K, Mashima T, Tsuruo T. Phos-phatidylserine externalisation is a downstream event of inter-leukin-1-beta converting enzyme family protease activationduring apoptosis. Blood 1997;89:2060–6.

[80] Longthorne VL, Williams GT. Caspase activity is required forcommitment to Fas-mediated apoptosis. EMBO J1997;16:3805–12.

[81] McCarthy NJ, Whyte MK, Gilbert CS, Evan GI. Inhibition ofCed-3/ICE-related proteases does not prevent cell death in-duced by oncogenes, DNA damage or the Bcl-2 homologueBak. J Cell Biol 1997;136:215–27.

[82] Susin SQ, Zamzami N, Castedo M, Hirsch T, Marchetti P,Macho A, Daugas E, Geustens M, Kroemer G. Bcl-2 inhibitsthe mitochondrial release of an apoptogenic protease. J ExpMed 1996;184:1331–41.

[83] Golstein P. Controlling cell death. Science 1997;275:1081–2.[84] Kharbanda S, Pandey P, Schofield L, Israels S, Roncinske R,

Yoshida K, Bharti A, Yuan ZM, Saxena S, Weichselbaum R,Nalin C, Kufe D. Role for Bcl-XL as an inhibitor of cytosoliccytochrome c accumulation in DNA-damage-induced apopto-sis. Proc Natl Acad Sci 1997;94:6939–42.

[85] Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, ahuman protein homologous to C. elegans CED-4, participatesin cytochrome c-dependent activation of caspase 3. Cell1997;90:405–13.

[86] Jacobson MD. Programmed cell death; a missing link is found.Trends Cell Biol 1997;7:467–9.

[87] Liu X, Zou H, Slaughter C, Wang X. DFF, a heterodimericprotein that functions downstream of Caspase 3 to triggerDNA framentation during apoptosis. Cell 1997;89:175–84.

[88] Reed JC. Double identity for proteins of the Bcl-2 family.Nature 1997;387:773–6.

[89] Hacker G, Vaux DL. A sticky business. Curr Biol 1995;5:622–4.

[90] Antonsson B, Conti F, Ciavatta A, Montessuit S, Lewis S,Martinou I, Bernasconi L, Bernard A, Mermod J, Mazzei G,Maundrell K, Gambale F, Sadoul R, Martinou J. Inhibition ofBax channel-forming activity by Bcl-2. Science 1997;277:370–2.

[91] Cheng EH, Kirsch DG, Clem RJ, Ravi R, Kastan MB, Bedi A,Veno K, Hardwick JM. Conversion of Bcl-2 to Bax-like deatheffector by caspases. Science 1997;278:1966–8.

[92] Schulz JB, Bremen D, Reed JC, Lommatzsch J, Takayama S,Wullner U, Loschmann PA, Klockgether T, Weller M. Cooper-ative interception of neuronal apoptosis by BCL-2 and BAG-1expression; prevention of caspase activation and reduced pro-duction of reactive oxygen species. J Neurochem 1997;69:2075–86.

[93] Krajewska M, Moss SF, Krajewski S, Song K, Holt PR, ReedJC. Elevated expression of Bcl-X and reduced Bak in primarycolorectal adenocarcinomas. Cancer Res 1996;56:2422–7.

[94] Krajewski S, Krajewska M, Shabaik A, Wang H, Irie S, FongL, Reed JC. Immunohistochemical analysis of in vivo patternsof bcl-X expression. Cancer Res 1994;54:5501–7.

[95] Haldar S, Jena N, Croce CM. Inactivation of Bcl-2 by phos-phorylation. Proc Natl Acad Sci 1995;92:4507–11.

[96] Haldar S, Chintapalli J, Croce CM. Taxon induces bcl-2 phos-phorylation and death of prostate cancer cells. Cancer Res1996;56:1253–5.

[97] Haldar S, Basu R, Croce CM. Bcl-2 is the guardian of micro-tubule integrity. Cancer Res 1997;57:229–33.

[98] Grillot D, Gonzalez-Garcia M, Ekhterae D, Duan L, InoharaN, Ohta S, Seldin M, Nunez G. Genomic organisation, pro-moter region analysis and chromosome location of the mousebcl-X Gene. J Immunol 1997;158:4750–7.

[99] Shiraiwa N, Inohara N, Okada S, Yuzaki M, Shoji S, Ohta S.An additional form of rat Bcl-X, Bcl-Xb, generated by anunspliced RNA, promotes apoptosis in promyeloid cells. J BiolChem 1996;271:13258–65.

[100] Yang XF, Weber GF, Cantor H. A novel Bcl-X isoformconnected to the T-cell receptor regulates apoptosis in T-cells.Immunity 1997;7:629–39.

[101] Apte SS, Mattei MG, Olsen BR. Mapping of the human BAXgene to chromosome 19q13.3–q13.4 and isolation of a novelalternatively spliced transcript, BAXd. Genomics 1995;26:592–4.

[102] Pecci A, Scholz A, Pelster D, Beato M. Progestins preventapoptosis in a rat endometrial cell line and increase the ratio ofbcl-XL to bcl-XS. J Biol Chem 1997;272:11791–8.

[103] Tilly J, Tilly K, Kenton M, Johnson AL. Expression of mem-bers of the bcl-2 gene family in the immature rat ovary: equinechorionic gonadotropin-mediated inhibition of granulosa cellapoptosis is associated with decreased Bax and constitutivebcl-2 and bcl-XL mRNA levels. Endocrinology 1995;136:232–41.

[104] Chang T, Hung M, Jiang S, Chu J, Chu L, Tsai L. Dexam-ethasone suppresses apoptosis in a human gastric cancer cellline through modulation of bcl-X gene expression. FEBS Lett1997;415:11–5.

[105] Minn AJ, Rudin CM, Boise LH, Thompson CB. Expression ofbcl-XL can confer a multidrug resistance phenotype. Blood1995;86:1903–10.

[106] Dole M, Jasty R, Cooper M, Thompson C, Nunez G, CastleVP. Bcl-XL is expressed in neuroblastoma cells and modulateschemotherapy-induced apoptosis. Cancer Res 1995;55:2576–82.

[107] Tang C, Willingham MC, Reed JC, Miyashita T, Ray S,Ponnathpur V, Huang Y, Mahoney ME, Bullock G, Bhalla K.High levels of p26BCL-2 oncoprotein retard taxol-inducedapoptosis in human pre-B leukaemia cells. Leukaemia1994;8:1960–9.

[108] Huang Y, Ibrado AM, Reed JC, Bullock G, Ray S, Tang C,Bhalla K. Co-expression of several molecular mechanisms ofmulti-drug resistance and their significance for paclitaxel cyto-toxicity in human AML HL-60 cells. Leukemia 1997;11:153–257.

[109] Gauthier ER, Piche L, Lemieux G, Lemieux R. Role of bcl-XL

in the control of apoptosis in murine myeloma cells. CancerRes 1996;56:1451–6.

[110] Ziegler A, Luedke GH, Fabbro D, Altmann KH, Stahel RA,Zangemeister-Wittke U. Induction of apoptosis in small-celllung cancer cells by an antisense oligodeoxynucleotide targetingthe Bcl-2 coding sequence. J Natl Cancer Inst 1997;89:1027–36.

[111] Dorai T, Olsson C, Katz A, Buttyan R. Development of ahammerhead ribozyme against bcl-2. I. Preliminary evaluationof a prostatic cancer. The Prostate 1997;32:246–58.

[112] Webb A, Cunningham D, Cotter F, Clarke PA, diStefano F,Ross P, Corbo M, Dziewanawska Z. Bcl-2 antisense therapy inpatients with non-Hodkin’s lymphoma. Lancet 1997;349:1137–41.

[113] Ibrado AM, Huang Y, Fang G, Liu L, Bhalla K. Overexpres-sion of bcl-2 or bcl-XL inhibits Ara C-induced Cpp32/yamaprotease activity and apoptosis of human AML HL-60 cells.Cancer Res 1996;56:4743–8.

[114] Kim CN, Wang X, Huang Y, Ibrado AM, Liu L, Fang G,Bhalla K. Overexpression of bcl-XL inhibits Ara C-inducedmitochrondrial loss of cytochrome c and other perturbations

Page 21: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 201

that activate the molecular cascade of apoptosis. Cancer Res1997;57:3115–20.

[115] Decaudin D, Geley S, Hirsch T, Castedo M, Marchetti P,Macho A, Kofler R, Kroemer C. Bcl-2 and bcl-XL antago-nize the mitochondrial dysfunction preceding nuclear apopto-sis induced by chemotherapeutic agents. Cancer Res1997;57:62–7.

[116] Simonian PL, Grillot AM, Nunez G. Bcl-2 and bcl-XL candifferentially block chemotherapy-induced cell death. Blood1997;90:1208–16.

[117] Tu Y, Xu FH, Liu J, Vescio R, Berenson J, Fady C, Licht-enstein A. Upregulated expression of Bcl-2 in multiplemyeloma cells induced by exposure to doxorubicin, etoposideand hydrogen peroxide. Blood 1996;88:1805–12.

[118] Sumantran VN, Ealovega MW, Nunez G, Clarke MC, WichaMS. Overexpression of Bcl-XS sensitizes MCF-7 cells tochemotherapy-induced apoptosis. Cancer Res 1995;55:2507–10.

[119] Ealovega M, McGinnis P, Sumantran V, Clarke M, WichaM. Bcl-XS gene therapy induces apoptosis of human mam-mary tumors in nude mice. Cancer Res 1996;56:1965–9.

[120] Mandal M, Kumar R. Bcl-2 modulates telomerase activity. JBiol Chem 1997;272:14183–97.

[121] Takayama S, Sato T, Krajewski S, Kochel K, Irie S, MillanJA, Reed JC. Cloning and functional analysis of BAG-1: Anovel Bcl-2-binding protein with anti-cell death activity. Cell1995;80:279–84.

[122] Wang HG, Takayama S, Rapp UR, Reed JC. Bcl-2 interact-ing protein, BAG-1, binds to and activates the kinase Raf-1.Proc Natl Acad Sci USA 1996;93:7063–8.

[123] Packham G, Brimmell M, Cleveland SL. Mammalian cellsexpress two differently localised Bag-1 isoforms generated byalternative translation initiation. Biochem J 1997;328:807–13.

[124] Yang E, Zha J, Jockel J, Boise LH, Thompson CB, Ko-rsmeyer SJ. Bad, a heterodimeric partner for Bcl-XL and Bcl-2, displaces Bax and promotes cell death. Cell1995;80:285–91.

[125] Yin XM, Oltvai ZN, Korsmeyer SJ. BH1 and BH2 domainsof bcl-2 are required for inhibition of apoptosis and het-erodimerisation with Bax. Nature 1994;369:321–3.

[126] Ryan KM, Birnie GD. Myc oncogenes: the enigmatic family.Biochem J 1996;314:713–21.

[127] Akahani S, Nangia-Makker P, Inohara H, Kim HR, Raz A.Galectin-3: a novel antiapoptotic molecule with a functionalBH1 (NWGR) domain of Bcl-2 family. Cancer Res1997;57:5272–6.

[128] Reynolds JE, Li J, Craig RW, Eastman A. Bcl-2 and MCL-1expression in CHO cells inhibits intracellular acidification andapoptosis induced by staurosporine. Exp Cell Res1996;225:430–6.

[129] Yang T, Buchan HL, Townsend KJ, Craig RW. MCL-1, amember of the BCL-2 family, is induced rapidly in responseto signals for cell differentiation or death, but not to signalsfor cell proliferation. J Cell Physiol 1996;166:523–36.

[130] D’sa-Eipper C, Subramanian T, Chinnadurai G. Bfl-1, a bcl-2homologue, suppresses p53-induced apoptosis and exhibitspotent cooperative transforming activity. Cancer Res1996;56:3879–82.

[131] Ambrosini G, Adida C, Altieri DC. A novel anti apoptosisgene, survivin, expressed in cancer and lymphoma. Nat Med1997;3:917–21.

[132] Duckett CS, Ki F, Wang Y, Tomaselli KJ, Thompson CB,Armstrong RC. Human IAP-like protein regulates pro-grammed cell death downstream of Bcl-XL and cytochrome c.Mol Cell Biol 1998;18:608–15.

[133] Han DK, Chaudhary PM, Wright ME, Friedman C, TraskBJ, Riedel RT, Baskin DG, Schwartz SM, Hood L. MRIT, a

novel death-effector domain-containing protein, interacts withcaspases and bcl-XL and initiates cell death. Proc Natl AcadSci USA 1997;94:11333–8.

[134] Ng FW, Nguyen M, Kwan T, Branton PE, Nicholson DW,Cromlish JA, Shore GC. P28 Bap 31, a Bcl-2/Bcl-XL-andprocaspase 8-associated protein in the endoplasmic reticulum.J Cell Biol 1997;139:327–38.

[135] Inohara N, Ding L, Chen S, Nunez G. Harakiri, a novelregulator of cell death, encodes a protein that activates apop-tosis and interacts selectively with survival-promoting proteinsBcl-2 and Bcl-XL. EMBO J 1997;16:1686–1694.

[136] O’Connor L, Strasser A, O’Reilly LA, Hausmann G, AdamsJM, Cory S, Huang DC. Bim, a novel member of the bcl-family that promotes apoptosis. EMBO J 1998;17:384–95.

[137] Li F, Srinivasan A, Wang Y, Armstrong RC, Tomaselli KJ,Fritz LC. Cell-specific induction of apoptosis by microinjec-tion of cytochrome c. Bcl-XL has activity independent of cy-tochrome c release. J Biol Chem 1997;272:30299–305.

[138] Motoyama N, Wang F, Roth KA, Sawa H, Nakayama KI,Nakayama K, Negishi I, Senju S, Zhang Q, Fujii S, Loh D.Massive cell death of immature haematopoietic cells and neu-rons in Bcl-X-deficient mice. Science 1995;267:1506–10.

[139] Kamada S, Shimono A, Shinjo Y, Tsujimura T, TakahashiT, Noda T, Kitamura Y, Kondoh H, Tsujimoto Y. Bcl-2deficiency in mice leads to pleiotropic abnormalities: acceler-ated lymphoid cell death in thymus and spleen, polycystickidney, hair hypopigmentation and distorted small intestine.Cancer Res 1995;55:354–9.

[140] Knudson CM, Tung KS, Tourtellotte WG, Brown GA, Ko-rsmeyer SJ. Bax-deficient mice with lymphoid hyperplasia andmale germ cell death. Science 1995;270:96–9.

[141] Ferry DR, Traunecker H, Kerr DJ. Clinical trials of P-glyco-protein reversal in solid tumours. Eur J Cancer1996;32A:1070–81.

[142] Fisher GA, Lum BL, Hausdorff J, Slkic BI. Pharmacologicalconsiderations in the modulation of multidrug resistance. EurJ Cancer 1996;32A:1082–8.

[143] Sonneveld P. Reversal of multidrug resistance in acutemyeloid leukaemia and other haematological malignancies.Eur J Cancer 1996;32A:1062–9.

[144] Tu Y, Renner S, Xu F, Fleishman A, Taylor J, Weisz J,Vescio R, Rettig M, Berenson J, Krajewski S, Reed JC,Lichtenstein A. Bcl-X expression in multiple myeloma; possi-ble indicator of chemoresistance. Cancer Res 1998;58:256–62.

[145] Christodoulopoulos G, Fotouchi N, Krajewski S, Reed JC,Alaoui-Jamali M, Panasci L. Relationship between nitrogenmustard drug resistance in B-cell chronic lymphocyticleukemia (B-CLL) and protein expression of Bcl-2, Bax, Bcl-X and P53. Cancer Lett 1997;121:59–67.

[146] Reed JC. Promise and problems of Bcl-2 antisense therapy. JNatl Cancer Inst 1997;89:988–90.

[147] Volm M, Mattern J. Increased expression of Bcl-2 in drug-re-sistant squamous cell lung carcinomas. Int J Oncol1995;7:1333–8.

[148] Alsabeh R, Wilson C, Ahn C, Vasef M, Battifora H. Expres-sion of bcl-2 by breast cancer: a possible diagnostic applica-tion. Mod Pathol 1996;9:439–44.

[149] Papadimitriou CS, Costopoulos JS, Christoforidou BP, Kot-sianti AJ, Karkavelas GS, Hytiroglou PM, Koufogiannis DJ,Nenopoulou HE. Expression of Bcl-2 protein in human pri-mary breast carcinomas and its correlation with multifocalityhistopathological types and prognosis. Eur J Cancer1997;33:1275–80.

[150] Krajewski S, Blomqvist C, Franssila K, Krajewska M, Wase-nius VM, Niskanen E, Nordling S, Reed JC. Reduced expres-sion of proapoptotic gene Bax is associated with poor responserates in combination chemotherapy and shorter survival in

Page 22: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205202

women with metastatic breast adenocarcinoma. Cancer Res1995;55:4471–8.

[151] Stoetzer OJ, Nussler V, Darsow M, Gullis E, Pelka-FleischerR, Scheel U, Wilmanns W. Association of bcl-2, bax, bcl-XL

and IL-1-beta converting enzyme expression with initial re-sponse to chemotherapy in acute myeloid leukaemia.Leukaemia 1996;10(Suppl. 3):S18–22.

[152] Chresta CM, Masters JRW, Hickman JA. Hypersensitivity ofhuman testicular tumors to etoposide-induced apoptosis is as-sociated with functional p53 and a high Bax:Bcl-2 ratio. Can-cer Res 1996;56:1834–41.

[153] Clynes M, Heenan M, Hall K. Human cell lines as modelsfor multidrug resistance in solid tumours. Cytotechnology1993;12:231–56.

[154] Heenan M, O’Driscoll L, Cleary I, Connolly L, Clynes M.Isolation from a human MDR lung cell line of multipleclonal subpopulations which exhibit significantly differentdrug resistance. Int J Cancer 1997;7:907–15.

[155] Wilson WH, Teroya-Feldstein J, Fest T, Harris C, SteinbergSM, Jaffe ES, Raffeld M. Relationship of p53, bcl-2, andtumor proliferation to clinical drug resistance in non-Hodgkin’s lymphoma. Blood 1997;89:601–9.

[156] Begleiter A, Mowat M, Israels LG, Johnston JB. Chlorambu-cil in chronic lymphocytic leukaemia: mechanism of action.Leuk Lymphoma 1996;23:187–201.

[157] Morabito F, Filangeri M, Callea I, Sculli G, Callea V,Francchiolla NS, Neri A, Brugiatelli M. Bcl-2 protein expres-sion and p53 gene mutation in chronic lymphocyticleukaemia; correlation with in vitro sensitivity to chlorambu-cil and purine analogs. Haematologica 1997;82:16–20.

[158] Hurlimann J, Larringo B, Vala DLM. Bcl-2 protein in inva-sive ductal breast carcinoma. Virchows Archiv 1995;426:163–8.

[159] Hellemans P, Van Dam PA, Weyler J, Van Oosterom AT,Buytaert P, Van Merck E. Prognostic Value of bcl-2 expres-sion in invasive breast carcinoma. Br J Cancer 1995;72:354–60.

[160] Cole SPC, Bhardwaj G, Gerlach JH, Mackie JE, Grant CE,Almquist KC, Stewart AJ, Kurz EU, Duncan AMV, DeeleyRG. Overexpression of a transporter gene in a multidrug-re-sistant human lung cancer cell line. Science 1992;258:1650–4.

[161] Germann U, Ford P, Shlyakhter D, Mason V, Harding M.Chemosensitization of VX-710, verapamil, cyclosporin A,MS-209 and GF120918 in multidrug resistant HL60/ADRcells expressing the multidrug resistance-associated proteinMRP. Anti-Cancer Drugs 1997;8:141–55.

[162] Hart S, Ganeshaguru K, Hoffbrand H, Prentice H, Mehta A.Expression of the multidrug resistance associated protein(MRP) in acute leukaemia. Leukemia 1994;8:2163–8.

[163] Slovak M, Ho J, Bhardwaj G, Kurz E, Deeley R, Cole S.Localisation of a novel multidrug resistance associated genein the HT1080/DR4 and H69AR human tumor cell lines.Cancer Res 1993;53:3221–5.

[164] Narasaki F, Oka M, Fukuda M, Ikeda K, Terashi K, Naka-mura T, Soda H, Kohno S. Multidrug resistance associatedprotein gene expression and drug sensitivity in human lungcancer cells. Anticancer Res 1997;17:3493–7.

[165] Berger W, Elbling L, Hauptmann E, Micksche M. Expressionof the multidrug resistance associated protein (MRP) andchemoresistance of human non-small cell lung cancer cells.Int J Cancer 1997;73:84–93.

[166] Coley H, Workman P, Twentyman P. Retention of activityby selected anthracyclines in a multidrug resistant humanlarge cell lung carcinoma line without P-glycoprotein hyperex-pression. Br J Cancer 1991;63:351–7.

[167] Nooter K, Brutel de la Riviere G, Look M, van WingerdenK, Henzen-Logmans S, Scheper R, Flens M, Klijn J, Stoter

G, Foekens J. The prognostic significance of the multidrugresistance-associated protein (MRP) in promary breast cancer.Br J Cancer 1997;76:486–93.

[168] Stein U, Walther W, Lemm M, Naundorf H, Fichtner I.Development and characterisation of novel human multidrugresistant mammary carcinoma lines in vitro and in vivo. Int JCancer 1997;72:885–91.

[169] Berger W, Hauptmann E, Eibling L, Vetterlein M,Kokoschka EM, Micksche M. Possible role of the multidrugresistance-associated protein (MRP) in chemoresistance of hu-man melanoma cells. Int J Cancer 1997;71:108–15.

[170] Loe D, Deeley R, Cole S. Biology of the multidrug resistanceassociated protein MRP. Eur J Cancer 1996;32A:945–57.

[171] Naito S, Hasegawa S, Yokomizo A, Koga H, Kotoh S,Kuwano M, Kumazawa J. Non-P-glycoprotein mediatedatypical multidrug resistance in a human bladder cancer cellline. Jpn J Cancer Res 1995;86:1112–8.

[172] Brock I, Hipfnerm D, Nielson B, Jensen P, Deeley R, ColeS, Sehested M. Sequential co-expression of the multidrug re-sistance genes MRP and mdr1 and their products in VP-16selected H69 small cell lung cancer cells. Cancer Res1995;55:459–62.

[173] Hasegawa S, Abe T, Naito S, Kotoh S, Kumazawa J,Hipfner D, Deeley R, Cole S, Kuwano M. Expression ofmultidrug resistance associated protein (MRP), MDR1 andDNA topoisomerase II in human multidrug resistant bladdercancer cell lines. Br J Cancer 1995;71:907–13.

[174] Stride B, Grant C, Loe D, Hipfner D, Cole S, Deeley R.Pharmacological characterisation of the murine and humanorthologs of multidrug resistance protein in transfected hu-man embryonic kidney cells. Mol Pharmacol 1997;52:344–53.

[175] Breuninger L, Paul S, Gaughan K, Miki T, Chan A,Aaronson S, Kruh G. Expression of multidrug resistance as-sociated protein in NIH/3T3 cells confers multidrug resistanceassociated with increased drug efflux and altered intracellulardrug distribution. Cancer Res 1995;55:5342–7.

[176] Grant C, Valdimarsson G, Hipfner D, Almquist K, Cole S,Deeley R. Overexpression of multidrug resistance associatedprotein (MRP) increases resistance to natural product drugs.Can Res 1994;54:357–61.

[177] Cole SPC, Sparks KE, Fraser K, Loe DW, Grant CE,Wilson GM, Deeley RG. Pharmacological characterisation ofmultidrug resistant MRP-transfected human tumor cells. CanRes 1994;54:5902–10.

[178] Norris MD, Bordow JB, Marshall GM, Haber PS, CohnMD, Haber M. Expression of the gene for multidrug resis-tance associated protein and outcome in patients with neu-roblastoma. New Engl J Med 1996;334:231–8.

[179] Chan HSL, Lu Y, Grogan TM, Haddad G, Hipfner DR,Cole SPC, Deeley RG, Ling V, Gallie BL. MRP Expressionin Retinoblastoma correlates with the rare failure ofchemotherapy despite cyclosporine for reversal of P-Glyco-protein. Cancer Res 1997;57:2325–30.

[180] Ota E, Abe Y, Oshika Y, Ozeki Y, Iwasaki M, Inove H,Yamazaki H, Ueyama Y, Takag K, Ogata T, Tamaoki N,Nakamura M. Expression of the MRP gene in non-small-celllung cancer. Br J Cancer 1995;72:550–4.

[181] Flens MJ, Zaman GJ, Van der Valk P, Izquierdo MA,Schroeijers AB, Scheffer GL, Van der Groep P, deHaas M,Meijer CJLM, Scheper RJ. Tissue distribution of the multi-drug resistance protein. Am J Pathol 1996;148:1237–47.

[182] Zhu Q, Center M. Cloning and sequence analysis of the pro-moter region of the MRP gene of HL60 cells isolated forresistance to adriamycin. Can Res 1994;54:4488–92.

[183] Ueda K, Pastan I, Gottesman M. Isolation and sequence of thepromoter region of the human multidrug resistance (P-glyco-protein) gene. J Biol Chem 1987;262:17432–6.

Page 23: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 203

[184] Eijdems E, De Haas M, Coco-Martin J, Ottenheim C, ZamanG, Dauwerse H, Breuning M, Twentyman P, Borst P, Baas F.Mechanisms of MRP over-expression in four human lung can-cer cell lines and analysis of the MRP amplicon. Int J Cancer1995;60:676–84.

[185] Slapak C, Fracasso P M, Martell R, Toppmeyer D, Lecerf J,Levy S. Overexpression of the multidrug resistance associatedprotein in vincristine but not doxorubicin selected multidrugresistant murine erythroleukemia cells. Cancer Res1994;54:5607–13.

[186] Giaccone G, van Ark-Otte J, Rubio G, Gazdar A, Broxter-man H, Dingemans A, Flens M, Scheper R, Pinedo H. MRPis frequently expressed in human lung cancer cell lines innon-small cell lung cancer and in normal lungs. Int J Cancer1996;66:760–7.

[187] Ishikawa T, Bao J, Yamane Y, Akimaru K, Frindrich K,Wright C, Kuo M. Co-ordinated induction of MRP/GS-Xpump and g-glutamylcysteine synthase by heavy metals inhuman leukemia cells. J Biol Chem 1996;271:14981–8.

[188] Hipfner D, Almquist K, Leslie E, Gerlach J, Grant C, DeeleyR, Cole S. Membrane topology of the multidrug resistanceprotein (MRP). J Biol Chem 1997;272:23623–30.

[189] Kast C, Gros P. Topology mapping of the amino terminalhalf of multidrug resistance associated protein by epitope in-sertion and immunofluorescence. J Biol Chem1997;272:26479–87.

[190] Bakos E, Hegedus T, Hollo Z, Welker E, Tusnady G, ZamanG, Flens M, Varadi A, Sarkadi B. Membrane topology andglycosylation of the human multidrug resistance associatedprotein. J Biol Chem 1996;271:12322–6.

[191] Lautier D, Canitrot Y, Deeley R, Cole S. Multidrug resistancemediated by the multidrug resistance protein gene. BiochemPharmacol 1996;52:967–77.

[192] Krishnamachary N, Center M. The MRP gene associated witha non- P-glycoprotein multidrug resistance encodes a 190 kDamembrane bound glycoprotein. Cancer Res 1993;53:3658–61.

[193] Almquist K, Loe D, Hipfner D, Mackie J, Cole S, Deeley R.Characterisation of the 190 kDa multidrug resistance proteinin drug selected and transfected human tumor cells. CancerRes 1995;55:102–10.

[194] Szczypka M, Wemmie J, Moye-Rowley W, Thiele D. A yeastmetal resistance protein similar to human cystic fibrosistransmembrane conductance regulator (CFTR) and multidrugresistance associated protein. J Biol Chem 1994;269:22853–7.

[195] Chambers T, Pohl J, Raynor R, Kuo J. Identification ofspecific sites in human P-glycoprotein phosphorylated byprotein kinase-C. J Biol Chem 1993;268:4592–5.

[196] Paul S, Belinsky M, Shen H, Kruh G. Structure and in vitrosubstrate specificity of the murine multidrug resistance-associ-ated protein. Biochemistry 1996;35:13647–55.

[197] Ma L, Krishnamachary N, Center M. Phosphorylation of themultidrug resistance associated protein gene encoded proteinP190. Biochemistry 1995;34:3338–43.

[198] Jedlitschky G, Lier I, Buchholz U, Barnouin K, Kurz G,Keppler D. Transport of glutathione, glucuronate and sulfateconjugates by the MRP gene-encoded conjugate export pump.Can Res 1996;56:988–94.

[199] Leier I, Jedlitschky G, Buchholz U, Center M, Cole S, DeeleyR, Keppler D. ATP dependent glutathione disulphide trans-port mediated by the MRP gene encoded conjugate exportpump. Biochem J 1996;313:433–7.

[200] Muller M, Meijer C, Zaman G, Borst P, Scheper R, MulderN, de Vries E, Jansen P. Overexpression of the gene encodingthe multidrug resistance associated protein results in increasedATP dependent glutathione S conjugate transport. Proc NatlAcad Sci USA 1994;91:13033–7.

[201] Loe D, Almquist K, Cole S, Deeley R. ATP-dependent 17-b-

estradiol 17-(b-D-glucuronide) transport by multidrug resis-tance protein (MRP). Inhibition by cholestatic steroids. J BiolChem 1996;271:9683–9.

[202] Tew K. Glutathione-associated enzymes in anticancer drugresistance. Cancer Res 1994;54:4313–20.

[203] Versantvoort CHM, Broxterman HJ, Bagrij T, Scheper RJ,Twentyman PR. Regulation by glutathione of drug transportin multidrug resistant human lung tumor cell lines overex-pressing multidrug resistance associated protein. Br J Cancer1995;72:82–9.

[204] Paul S, Breuninger LM, Tew KD, Shen H, Kruh GD. ATP-dependent uptake of natural product cytotoxic drugs by mem-brane vesicles establishes MRP as a broad specificitytransporter. Proc Natl Acad Sci 1996;93:6929–34.

[205] Paul S, Breuninger L, Kruh G. ATP-dependent transport oflipophilic drugs by membrane vesicles prepared from MRP-overexpressing HL60/ADR cells. Biochemistry 1996;35:14003–11.

[206] Loe D, Almquist K, Deeley R, Cole S. Multidrug resistanceprotein (MRP) mediated transport of leukotriene C4 andchemotherapeutic agents in membrane vesicles. J Biol Chem1996;271(16):9675–82.

[207] Kool M, de Haas M, Scheffer G, Scheper R, van Eijk M,Juijn J, Baas F, Borst P. Analysis of expression of cMOAT(MRP2), MRP3, MRP4 and MRP5, homologues of the multi-drug resistance associated protein gene (MRP1) in humancancer cell lines. Cancer Res 1997;57:3537–47.

[208] Versantvoort C, Broxterman H, Pinedo H, de Vries E, FellerN, Kuiper C, Lankelma J. Energy dependent processes in-volved in reduced drug accumulation in multidrug resistanthuman lung cancer cell lines without P-glycoprotein expres-sion. Can Res 1992;52:17–23.

[209] Zaman G, Flens M, Van Leusden M, De Haas M, Pinedo H,Mulder S, Lankelma J, Baas F, Broxterman H, Borst P. Thehuman multidrug resistance-associated protein MRP is aplasma membrane efflux pump. Proc Natl Acad Sci1994;91:8822–6.

[210] McGrath T, Latoud C, Arnold S, Safa A, Felsted R, CenterM. Mechanisms of multidrug resistance in HL60 cells: analy-sis of resistance associated membrane proteins and levels ofmdr gene expression. Biochem Pharmacol 1989;38:3611–9.

[211] Safa A. Photoaffinity labelling of the multidrug resistancerelated P-glycoprotein with photoactive analogs of verapamil.Proc Natl Acad Sci 1988;85:7181–91.

[212] Twentyman PR, Versantvoort CHM. Experimental modula-tion of MRP (multidrug resistance associated protein) medi-ated resistance. Eur J Cancer 1996;32A:1002–9.

[213] Gekeler V, Ise W, Sanders K H, Ulrich W R, Beck J. Theleukotriene LtD4 receptor antagonist MK571 specifically mod-ulates MRP associated multidrug resistance. Biochem BiophysRes Commun 1995;208:345–52.

[214] Gekeler V, Boer R, Ise W, Sanders K H, Schachtele C, BeckJ. The specific bisindolylmaleimide PKC inhibitor GF109203X efficiently modulates MRP associated multiple drugresistance. Biochem Biophys Res Commun 1995;206:119–26.

[215] Duffy C, Elliott C, O’Connor R, Heenan M, Coyle S, ClearyI, Kavanagh K, Verhaegen S, O’Loughlin C, NicAmhlaoibhR, Clynes M. Enhancement of chemotherapeutic drug toxicityto human tumor cells in vitro by a sub-set of non-steroidalanti-inflammatory drugs (NSAIDs). Eur J Cancer 1998 (inpress).

[216] Draper MP, Martell RL, Levy SB. Indomethacin-mediatedreversal of multidrug resistance and drug efflux in human andmurine cell lines overexpressing MRP, but not P-glycoprotein.Br J Cancer 1997;75:810–5.

[217] Wijnholds J, Evers R, van Leusden MR, Mol CA, Zaman GJ,Mayer U, Beijnen JH, van der Valk M, Krimpenfort P, Borst

Page 24: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205204

P. Increased sensitivity to anticancer drugs and decreased infl-ammatory response in mice lacking the multidrug resistance-as-sociated protein. Nature Med 1997;3:1275–9.

[218] Lorico A, Rappa G, Flavell RA, Sartorelli AC. Double knock-out of the MRP gene leads to an increased drug sensitivity invitro. Cancer Res 1996;56:5351–5.

[219] Higgins C. ABC transporters: from microorganisms to man.Annu Rev Cell Biol 1992;8:67–113.

[220] Allikmets R, Gerrard B, Hutchinson A, Dean M. Characterisa-tion of the human ABC superfamily: isolation and mapping of21 new genes using the expressed sequence tags database. HumMol Genet 1996;5:1649–55.

[221] Mohamed A, Ferguson D, Seibert F, Cai H, Kartner N,Grinstein S, Riordan J, Lukas G. Functional expression andapical localisation of the cystic fibrosis transmembrane conduc-tance regulator in MDCK I cells. Biochem J 1997;322:259–65.

[222] Izquierdo M, Neefles J, Mathari A, Flens M, Scheffer G,Scheper R. Overexpression of the ABC transporter TAP inmultidrug resistant human cancer cell lines. Br J Cancer1996;74:1961–7.

[223] Nelson D, Byran J, Wechsler S, Clement J, Aguilar-Byran L.The high affinity sulfonylurea receptor: distribution, glycosyla-tion, purification and immunoprecipitation of two forms fromendocrine and neuroendocrine cell lines. Biochemistry1996;35:14793–9.

[224] Taniguchi K, Wada M, Kohno K, Nakamura T, Kawabe T,Kawakami M, Kagotani K, Okumura J, Akiyama S, KuwanoM. A human canalicular multispecific organic anion transporter(cMOAT) gene is expressed in cisplatin-resistant human cancercell lines with decreased drug accumulation. Cancer Res1996;56:4124–9.

[225] Muller M, de Vries G, Jansen P. Role of multidrug resistanceprotein (MRP) in glutathione S-conjugate transport in mam-malian cells. J Hepatol 1996;24:100–8.

[226] Mayer R, Kartenbeck J, Bucher M, Jedlitschky G, Leier I,Keppler D. Expression of the MRP gene encoded conjugateexport pump in liver and its selective absence from the canicu-lar membrane in transport deficient mutant hepatocytes. J CellBiol 1995;131:137–50.

[227] Zaman GJR, Versantvoort CHM, Smit JJM, Eijdems WHM,de Haas M, Smith AJ, Broxterman HJ, Mulder NH, de VriesEGE, Baas F, Borst P. Analysis of the expression of MRP, thegene for a new putative transmembrane drug transporter, inhuman multidrug resistant lung cancer cell lines. Can Res1993;53:1747–50.

[228] Evers R, Zaman G, van Deemter L, Jansen H, Calafat J,Oomen L, Oude Elfernik R, Borst P, Schinkel A. Basolaterallocalization and export activity of the human multidrug resis-tance-associated protein in polarized pig kidney cells. J ClinInvest 1996;97:1211–8.

[229] Paulusma C, Bosma P, Zaman J, Bakker C, Otter M, SchefferG, Scheper R, Borst P, Elferink R. Congenital jaundice in ratswith a mutation in a multidrug resistance associated proteingene. Science 1996;271:1126–8.

[230] Puchalski R, Fahl W. Expression of recombinant glutathioneS-transferase pi, Ya or Yb1 confers resistance to alkylatingagents. Proc Natl Acad Sci 1990;87:2443–7.

[231] Ishikawa T, Ali-Osman F. Glutathione associated cis-di-amminedichloroplatinum (II) metabolism and ATP dependentefflux from leukemia cells. J Biol Chem 1993;268:20116–25.

[232] Ishikawa T. The ATP-dependent glutathione S-conjugate ex-port pump. Trends Biochem Sci 1992;17:463–8.

[233] Ishikawa T, Wright C, Ishizuka H. GS-X pump is functionallyoverexpresesed in cis-diamminedichloroplatinum (II) resistanthuman leukemia HL-60 cells and down regulated by cell differ-entiation. J Biol Chem 1994;269:29085–93.

[234] Leier I, Jedlitschky G, Buchholz U, Keppler D. Characterisa-tion of the ATP dependent leukotriene C4 export carrier inmastocytoma cells. Eur J Biochem 1994;220:599–606.

[235] Gomi A, Shinoda S, Masuzawa T, Ishikawa T, Kuo MT.Transient co-induction of MRP/GS-X Pump and g-Glutamyl-cysteine Synthetase by 1-(4-Amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea in human glioma cells.Cancer Res 1997;57:5292–9.

[236] Gomi A, Masuzawa T, Ishikawa T, Kuo MT. Posttranscrip-tional regulation of MRP/GS-X pump and g-Glutamylcysteinesynthetase expression by 1-(4-Amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloreothyl)-3-nitrosourea and by cycloheximide inhuman glioma cells. Biochem Biophys Res Commn1997;239:51–6.

[237] Kuo MT, Bao J-J, Curley SA, Ikeguchi M, Johnston DA,Ishikawa T. Frequent coordinated overexpression of the MRP/GS-X pump and g-glutamylcystein synthetase genes in humancolorectal cancers. Cancer Res 1996;56:3642–4.

[238] Koike K, Kawabe T, Tanaka T, Toh S, Uchiumi T, Wada M,Akiyama S, Ono M, Kuwano M. A canicular multispecificorganic anion transporter (cMOAT) antisence cDNA enhancesdrug sensitivity in human hepatic cancer cells. Can Res1997;57:5475–9.

[239] Lehnert M, Dalton WS, Roe D, Emerson S, Salmon SE.Synergistic inhibition by verapamil and quinine of P-glyco-protein-mediated multidrug resistance in a human myeloma cellline model. Blood 1991;77:348–54.

[240] Sikic BI, Fisher GA, Lum BL, Halsey J, Beketic-Oreskovic L,Chen G. Modulation and prevention of multidrug resistance byinhibitors of P-glycoprotein. Cancer Chemother Pharmacol1997;40:s13–9.

[241] Ford JM. Modulators of multidrug resistance. preclinical stud-ies. Hematol Oncol Clin North Am 1995;9:337–61.

[242] Wilson WH, Jamis-Dow C, Bryant G, Balis FM, Klecker RW,Bates SE, Chabner BA, Steinberg SM, Kohler DR, Wittes RE.Phase I and pharmacokinetic study of the multidrug resistancemodulator dexverapamil with EPOCH chemotherapy. J ClinOncol 1995;13:1985–94.

[243] Lum BL, Kaubisch S, Yahanda AM, Adler KM, Jew L, EhsanMN, Brophy NA, Halsey J, Gosland MP, Sikic BI. Alterationof etoposide pharmacokinetics and pharmacodynamics by cy-clsporine in a phase I trial to modulate multidrug resistance. JClin Oncol 1992;10:1635–42.

[244] Horio M, Gottesman MM, Pastan I. ATP-dependent transportof vinblastine in vesicles from human multidrug-resistant cells.Proc Natl Acad Sci 1988;85:3580–4.

[245] Ford JM, Hait WN. Pharmacology of drugs that alter multi-drug resistance in cancer. Pharmacol Rev 1990;42:156–99.

[246] Ford JM, Hait WN. Pharmacological circumvention of multi-drug resistance. Cytotechnology 1993;12:171–212.

[247] Verweij J, Herweijer H, Oosterom R, van der Burg ME,Planting AS, Seynaeve C, Stoter G, Nooter K. A phaseII studyof epidoxorubicin in colorectal cancer and the use of cy-closporin-A in an attempt to reverse multidrug resistance. Br JCancer 1991;64:361–4.

[248] Cairo MS, Siegel S, Anas N, Sender L. Clinical trial of contin-uous infusion verapamil, bolus vinblastine, and continuousinfusion VP-16 in drug resistant pediatric tumors. Cancer Res1989;49:1063–6.

[249] Ozols RF, Cunnion RE, Klecker RW Jr, Hamilton TC,Ostchega Y, Parrillo JE, Young RC. Verapamil and adriamycinin the treatment of drug-resistant ovarian cancer patients. JClin Oncol 1987;5:641–47.

[250] Fisher GA, Lum BL, Hausdorff J, Sikic BI. Pharmacologicalconsiderations in the modulation of multidrug resistance. Eur JCancer 1996;32A:1082–88.

Page 25: Recent developments in drug resistance and apoptosis research

M. Clynes et al. / Critical Re6iews in Oncology/Hematology 28 (1998) 181–205 205

[251] Murren JR, Durivage HJ, Buzaid AC, Reiss M, Flynn SD,Carter D, Hait WN. Trifluoperazine as a modulator of multi-drug resistance in refractory breast cancer. Cancer ChemotherPharmacol 1996;38:65–70.

[252] Dalton WS, Crowley JJ, Salmon SS, Grogan TM, LaufmanLR, Weiss GR, Bonnet JD. A phase III randomised study oforal verapamil as a chemosensitizer to reverse drug resistance inpatients with refractory melanoma. Cancer 1995;75:815–20.

[253] Cano-Gauci DF, Riordan JR. Action of calcium antagonists onmultidrug resistant cells. Biochem Pharmacol 1987;36:2115–23.

[254] Ford JM, Bruggemann EP, Pastan I, Gottesman MM, HaitWN. Cellular and biochemical characterization of thioxanthenefor reversal of multidrug resistance in human and murine celllines. Cancer Res 1990;50:1748–56.

[255] Tsuruo T, Sorio R, Gigante M, Corona G, Galligioni E,Boiocchi M. Circumvention of vincristine and adriamycin resis-tance in vitro and in vivo by calcium influx blockers. CancerRes 1983;43:2905–10.

[256] Ambudkar SV, Lelong IH, Zhang J, Cardarelli CO, GottesmanMM, Pastan I. Partial purification and reconstitution of thehuman multidrug-resistance pump: characterization of thedrug-stimulatable ATP hydrolysis. Proc Natl Acad Sci1992;89:8472–6.

[257] Schinkel AH, Mol CA, Wagenaar E, van Deemter L, Smit JJ,Borst P. Multidrug resistance and the role of P-glycoproteinknockout mice. Eur J Cancer 1995;31A:1295–8.

[258] Ferguson LR, Baguley BC. Multidrug resistance and mutageni-sis. Mutat Res 1993;285:79–90.

[259] Goldstein LJ, Bruggemann EP, Pastan I, Gottesman MM, HaitWN. Multidrug resistance in human cancer. Crit Rev OncolHematol 1992;12:243–53.

[260] Bailly J, Muller C, Jaffrezou JP, Demur C, Gassar G, BordierC, Laurent G. Lack of correlation between expression andfunction of P-glycoprotein in acute myeloid leukemia cell lines.Leukemia 1995;9:799–807.

[261] Beck WT, Danks MK. Isolation and Properties of Multidrug-Resistant Cell Lines: Characteristics of Multidrug Resistance inHuman Tumor Cells. In: Roninson IB, editor. Molecular andCellular Biology of Multidrug Resistance in Tumour CellsPlenum Press, NewYork, 1991:3–55.

[262] Luker GD, Lubbert M, Martens C, Bross KJ, Fiebig HH,Mertelsmann R, Herrmann F. Modulation of the multidrugresistance P-glycoprotein: detection with technetium-99m-ses-tamibi in vivo. J Nucl Med 1997;38:369–72.

[263] Wilson WH. Reversal of multidrug resistance with R-verapamiland analysis of mdr-1 expression in patients with lymphomarefractory to EPOCH chemotherapy. Proc Am Assoc CancerRes 1993;34:1266.

[264] Keller DJ, Altermatt HJ, Donatsch P, Zihlmann H, Laissue JA,Hiestand PC. Pharmacologic interactions between the resis-tance-modifying cyclosporin SDZ PSC 833 and etoposide (VP16-213) enhance in vivo cytostatic activity and toxicity. Int JCancer 1992;51:433–8.

[265] Willson JKV, Herweijer H, Oosterom R, van der Burg ME,Planting AS, Seynaeve C, Stoter G, Nooter K. Modulation afO6-alkylguanine alkyltransferase-directed DNA repair inmetastatic colon cancers. J Clin Oncol 1995;13:2301–8.

[266] Holm PS, Scanlon KJ, Dietel M. Reversion of multidrugresistance in the P-glycoprotein-positive human pancreatic cellline (EPP85-181RDB) by introduction of a hammerhead ri-bozyme. Br J Cancer 1994;70:239–43.

[267] Daly C, Coyle S, McBride S, O’Driscoll L, Daly N, Scanlon K,Clynes M. Mdr1 ribozyme mediated reversal of the multi-drugresistant phenotype in human lung cell lines. Cytotechnology1996;19:199–205.

[268] Byrne D, Daly C, Howlett A, Scanlon K, Clynes M. Antisense

and ribozyme strategies to reverse drug resistance. Cytotechnol-ogy 1998 (in press).

[269] Licht T, Lubbert M, Martens C, Bross KJ, Fiebig HH, Mertels-mann R, Herrmann F. Modulation of vindesine and doxoru-bicin resistance in multidrug-resistant pleural mesotheliomacells by tumour necrosis factor-alpha. Cytokines Mol Ther1995;1:123–32.

[270] Stein U, Walther W, Shoemaker RH. Modulation of mdr1expression by cytokines in human colon carcinoma cells: anapproach for reversal of multidrug resistance. Br J Cancer1996;74:1384–91.

[271] Clynes M. Cellular models for multidrug resistance in cancer.In Vitro Cell Dev Biol 1993;29A:171–9.

[272] Beck WT, Grogan TM, Willman CL, Cordon-Cardo C,Parham DM, Kuttesch JF, Andreeff M, Bates SE, Berard CW,Boyet JM, Brophy NA, Broxterman HJ, Chan HSL, DaltonWS, Dietel M, Fojo AT, Gascoyne RD, Head D, Houghton PJ,Srivastava DK, Lehnert M, Leith CP, Paietta E, Pavelic ZP,Rimsza L, Roninson IB, Sikic BI, Twentyman PR, Warnke R,Weinstein R. Methods to detect P-glycoprotein-associated mul-tidrug resistance in patient’s tumors: consensus recommenda-tions. Cancer Res 1996;56:3010–20.

[273] Heijn M, Hooijberg J, Scheffer G, Szabo G, Westerhoff H,Lankelma J. Anthracyclines modulate multidrug resistanceprotein (MRP) mediated organic anion transport. BiochimBiophys Acta 1997;1326:12–22.

[274] Kaye SB. Clinical drug resistance: the role of factors other thanP-glycoprotein. Am J Med 1995;99(6A):40s–44.

[275] Shen H, Paul S, Breninger L, Ciaccio P, Laing N, Helt M, TewK, Kruh G. Cellular and in vitro transport of glutathioneconjugates by MRP. Biochemistry 1996;35:5719–25.

[276] Cowie FJ, Pinkerton CR, Phillips M, Dick G, Judson I, Mc-Carthy PT, Flanagan RJ. Continuous-infusion verapamil withetoposide in relapsed or resistant paediatric cancers. Br J Can-cer 1995;71:871–81.

[277] Miller JP, Grogan TM, Dalton WS, Spier CM, Scheper RJ,Salmon SE. P-glycoprotein expression in malignant lymphomaand reversal of clinical drug resistance with chemotherapy plushigh dose verapamil. J Clin Oncol 1991;9:17–24.

[278] Toffoli G, Sorio R, Gigante M, Corona G, Galligioni E,Boiocchi M. Cyclosporin A as a multidrug-resistant modulatorin patients with renal cell carcinoma treated with teniposide. BrJ Cancer 1997;75:715–21.

Biography

Martin Clynes is Professor of Biotechnology atDublin City University, and Director of BioResearchIreland’s National Cell and Tissue Culture Centre(NCTCC). A major focus of the research programmesat NCTCC is to discover how cancer cells becomeresistant to chemotherapy, and how this clinical prob-lem could be overcome. Carmel Daly, Roisin Nic Amh-laoibh, Robert O’Connor, Cathal Elliott and ToniO’Doherty (Research Scientists) and Deirdre Croninand Lisa Connolly (Postgraduate Students) are workingin this research programme. Kevin Scanlon has pio-neered studies on oncogene cDNA transfection andribozyme technology to investigate drug resistance incancer cells, and is currently Head of Cancer Researchat Berlex Biosciences; Anthony Howlett is Senior Scien-tist in the cancer Research Department at Berlex..