abc transporters in cancer

11
Failure of cancer chemotherapy can occur through increased efflux of chemothera- peutic agents, leading to the reduction of intracellular drug levels and consequent drug insensitivity, often to multiple agents. A well-established cause of multidrug resistance (MDR) involves the increased expression of members of the ATP binding cassette (ABC) transporter superfamily, many of which efflux various chemothera- peutic compounds from cells 1 . The most extensively characterized MDR transporters include ABCB1 (also known as MDR1 or P-glycoprotein), ABCC1 (also known as MRP1) and ABCG2 (also known as BCRP or MXR) (BOX 1). The discovery of ABCB1 and ABCC1 was a major breakthrough in understanding the mechanisms behind MDR and prompted the identification of numerous proteins with related structures and similar transport capabilities (TABLE 1). It is now known that these proteins belong to the ABC transporter superfamily, which in humans comprises at least 48 genes with diverse functions 2 . Although it has been more than three decades since the discovery of ABCB1, and third generation inhibitors of its drug transport function have been devel- oped, clinical trials using these inhibitors have generally produced disappointing results. Although overcoming drug resist- ance through the inhibition of ABC trans- porters remains a priority, additional roles are emerging for these proteins that might be important for cancer initiation and pro- gression. Validating these additional func- tions and elucidating their roles in tumour biology will be central to our understanding of these key molecules and could inform the development of more effective targeted therapies. MDR and the ABC transporter family The ABC transporters are the largest family of transmembrane proteins, with seven sub- families that are designated A to G on the basis of sequence and structural homology, and are responsible for the ATP-dependent movement of a wide variety of xenobiotics (including drugs), lipids and metabolic products across the plasma and intracellular membranes. ABCA family members are predominantly expressed in cells of the cen- tral nervous system, as well as the haemato- poietic system, and are primarily known for their roles in lipid transport and homeostasis, particularly in lipid trafficking between cel- lular compartments 3–5 . In addition to their roles in drug transport (TABLE 1), several members of the ABCB family are responsible for intracellular peptide transport, including an essential role in major histocompatibility complex (MHC) class I antigen presenta- tion 6 . The ABCC (also known as multidrug resistance protein) family contains the greatest number of known drug transporters (TABLE 1), but also includes the cystic fibrosis transmembrane conductance regulator (CFTR; also known as ABCC7), the only ABC family member known to function as a channel and which has an essential role in chloride ion efflux 7 . The ABCD gene sub- family encodes peroxisomal half-transporters, some of which have been linked to neuro- degenerative diseases 8 . Members of the ABCE and ABCF families seem to have no membrane-spanning domain and are involved in mRNA translation 9,10 . For many years cancer researchers have focused on members of the ABC superfamily with drug efflux capacity to determine the reasons behind chemotherapeutic treatment failure. Most of these studies have examined the gene or protein expression of only a small number of transporters in tumour specimens that were obtained from patients at diagnosis, and they have correlated the expression data with clinical characteristics. Although these studies have confirmed the clinical relevance of ABCB1 in several cancers, the clinical rel- evance of other transporters remains uncer- tain 11 . However, ABC transporter inhibition remains an attractive potential adjuvant to chemotherapy (reviewed in REFS 1,12; BOX 1). Clinical trials involving MDR modulators The goal of reversing MDR in the clinic through the pharmacological inhibition of specific ABC transporters has been pursued for almost two decades. Specific ABC trans- porters, most notably ABCB1, ABCC1 and ABCG2, are overexpressed in various can- cers and can efflux a wide range of chemo- therapeutic agents used to treat patients 11,12 , making them attractive therapeutic targets. Despite initial optimism, the results of clinical trials using modulators of multi- drug transporters have been disappointing overall 13 . Initial clinical trials in the 1980s used compounds such as verapamil and cyclosporine A that were already approved for clinical use for conditions other than cancer and were also shown to inhibit ABCB1. Unfortunately, these first genera- tion inhibitors had unacceptable levels of toxicity. Second generation ABCB1 inhibi- tors, exemplified by valspodar (also known as PSC 833), were subsequently developed and had increased potency and decreased toxicity. However, these also generated dis- appointing results in the clinic. For instance, a recent Phase III study in patients with ovarian or peritoneal cancer, who were being treated with paclitaxel and carboplatin as front-line chemotherapy, failed to show any benefit from the inclusion of valspodar 14 . Similar findings have been observed in other cancers 15 and the further development of this compound is unlikely 11 . Although third generation ABCB1 inhibitors have now been developed that improve many of the short- comings of earlier modulators, initial results remain disappointing. For example, a recent Phase II study of women with metastatic or locally recurrent breast cancer found no additional benefit in survival with the inclusion of zosuquidar 16 . Despite considerable efforts, the clinical benefit of modulating drug efflux pumps has yet to be realized, leading some inves- tigators to conclude that the contribution of multidrug transporters to clinical drug resistance might not be particularly impor- tant 17 . However, several complications might THERAPEUTIC RESISTANCE — OPINION ABC transporters in cancer: more than just drug efflux pumps Jamie I. Fletcher, Michelle Haber, Michelle J. Henderson and Murray D. Norris Abstract | Multidrug transporter proteins are best known for their contributions to chemoresistance through the efflux of anticancer drugs from cancer cells. However, a considerable body of evidence also points to their importance in cancer extending beyond drug transport to fundamental roles in tumour biology. Currently, much of the evidence for these additional roles is correlative and definitive studies are needed to confirm causality. We propose that delineating the precise roles of these transporters in tumorigenesis and treatment response will be important for the development of more effective targeted therapies. PERSPECTIVES NATURE REVIEWS | CANCER VOLUME 10 | FEBRUARY 2010 | 147 © 20 Macmillan Publishers Limited. All rights reserved 10

Upload: rodrigo-hernandez

Post on 24-Oct-2014

32 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: ABC Transporters in Cancer

Failure of cancer chemotherapy can occur through increased efflux of chemothera-peutic agents, leading to the reduction of intracellular drug levels and consequent drug insensitivity, often to multiple agents. A well-established cause of multidrug resistance (MDR) involves the increased expression of members of the ATP binding cassette (ABC) transporter superfamily, many of which efflux various chemothera-peutic compounds from cells1. The most extensively characterized MDR transporters include ABCB1 (also known as MDR1 or P-glycoprotein), ABCC1 (also known as MRP1) and ABCG2 (also known as BCRP or MXR) (BOX 1). The discovery of ABCB1 and ABCC1 was a major breakthrough in understanding the mechanisms behind MDR and prompted the identification of numerous proteins with related structures and similar transport capabilities (TABLE 1). It is now known that these proteins belong to the ABC transporter superfamily, which in humans comprises at least 48 genes with diverse functions2. Although it has been more than three decades since the discovery of ABCB1, and third generation inhibitors of its drug transport function have been devel-oped, clinical trials using these inhibitors have generally produced disappointing results. Although overcoming drug resist-ance through the inhibition of ABC trans-porters remains a priority, additional roles are emerging for these proteins that might be important for cancer initiation and pro-gression. Validating these additional func-tions and elucidating their roles in tumour biology will be central to our understanding of these key molecules and could inform the development of more effective targeted therapies.

MDR and the ABC transporter familyThe ABC transporters are the largest family of transmembrane proteins, with seven sub-families that are designated A to G on the basis of sequence and structural homology, and are responsible for the ATP-dependent movement of a wide variety of xenobiotics (including drugs), lipids and metabolic products across the plasma and intracellular membranes. ABCA family members are predominantly expressed in cells of the cen-tral nervous system, as well as the haemato-poietic system, and are primarily known for their roles in lipid transport and homeostasis, particularly in lipid trafficking between cel-lular compartments3–5. In addition to their roles in drug transport (TABLE 1), several members of the ABCB family are responsible for intracellular peptide transport, including an essential role in major histocompatibility complex (MHC) class I antigen presenta-tion6. The ABCC (also known as multidrug resistance protein) family contains the greatest number of known drug transporters (TABLE 1), but also includes the cystic fibrosis transmembrane conductance regulator (CFTR; also known as ABCC7), the only ABC family member known to function as a channel and which has an essential role in chloride ion efflux7. The ABCD gene sub-family encodes peroxisomal half-transporters, some of which have been linked to neuro -degenerative diseases8. Members of the ABCE and ABCF families seem to have no membrane-spanning domain and are involved in mRNA translation9,10.

For many years cancer researchers have focused on members of the ABC superfamily with drug efflux capacity to determine the reasons behind chemotherapeutic treatment failure. Most of these studies have examined

the gene or protein expression of only a small number of transporters in tumour specimens that were obtained from patients at diagnosis, and they have correlated the expression data with clinical characteristics. Although these studies have confirmed the clinical relevance of ABCB1 in several cancers, the clinical rel-evance of other transporters remains uncer-tain11. However, ABC transporter inhibition remains an attractive potential adjuvant to chemotherapy (reviewed in REFS 1,12; BOX 1).

Clinical trials involving MDR modulatorsThe goal of reversing MDR in the clinic through the pharmacological inhibition of specific ABC transporters has been pursued for almost two decades. Specific ABC trans-porters, most notably ABCB1, ABCC1 and ABCG2, are overexpressed in various can-cers and can efflux a wide range of chemo-therapeutic agents used to treat patients11,12, making them attractive therapeutic targets.

Despite initial optimism, the results of clinical trials using modulators of multi-drug transporters have been disappointing overall13. Initial clinical trials in the 1980s used compounds such as verapamil and cyclosporine A that were already approved for clinical use for conditions other than cancer and were also shown to inhibit ABCB1. Unfortunately, these first genera-tion inhibitors had unacceptable levels of toxicity. Second generation ABCB1 inhibi-tors, exemplified by valspodar (also known as PSC 833), were subsequently developed and had increased potency and decreased toxicity. However, these also generated dis-appointing results in the clinic. For instance, a recent Phase III study in patients with ovarian or peritoneal cancer, who were being treated with paclitaxel and carboplatin as front-line chemotherapy, failed to show any benefit from the inclusion of valspodar14. Similar findings have been observed in other cancers15 and the further development of this compound is unlikely11. Although third generation ABCB1 inhibitors have now been developed that improve many of the short-comings of earlier modulators, initial results remain disappointing. For example, a recent Phase II study of women with metastatic or locally recurrent breast cancer found no additional benefit in survival with the inclusion of zosuquidar16.

Despite considerable efforts, the clinical benefit of modulating drug efflux pumps has yet to be realized, leading some inves-tigators to conclude that the contribution of multidrug transporters to clinical drug resistance might not be particularly impor-tant17. However, several complications might

T h e R A p e u T i C R e s i s TA n C e — o p i n i o n

ABC transporters in cancer: more than just drug efflux pumpsJamie I. Fletcher, Michelle Haber, Michelle J. Henderson and Murray D. Norris

Abstract | Multidrug transporter proteins are best known for their contributions to chemoresistance through the efflux of anticancer drugs from cancer cells. However, a considerable body of evidence also points to their importance in cancer extending beyond drug transport to fundamental roles in tumour biology. Currently, much of the evidence for these additional roles is correlative and definitive studies are needed to confirm causality. We propose that delineating the precise roles of these transporters in tumorigenesis and treatment response will be important for the development of more effective targeted therapies.

P e r s P e c t i v e s

NATURE REVIEwS | CanCer VolUME 10 | FEBRUARy 2010 | 147

© 20 Macmillan Publishers Limited. All rights reserved10

Page 2: ABC Transporters in Cancer

have limited the clinical success of ABC transporter inhibitors to date (reviewed in REF. 11). Foremost among these are altera-tions to the pharmacokinetics of anticancer agents, with decreased systemic clearance potentially leading to increased severity and incidence of adverse side effects18. This complication might apply to even the most specific of ABC transporter inhibitors, as genetic disruption of Abcb1 can result in increased drug levels in many tissues19. Such effects need be taken into consideration with any attempt to develop ABC transporter inhibitors for future clinical applications.

Functional redundancy in transporter families and the frequent overexpression of numerous transporter genes can also compli-cate attempts to inhibit transporter function. A recent examination of the role of ABC transporters in conferring drug resistance in the US National Cancer Institute’s panel of tumour cell lines suggested that more than half the family members might have a role in conferring drug resistance, at least in vitro20. In addition, as members of the ABCB and ABCC families have various roles in the immune system6,21–23, another unwanted consequence of transporter inhibition might be impaired anti-tumour immune responses, as has been recently reviewed24. Further exploration of the physiological roles of ABC transporters will not only raise awareness of potential side effects, but might also iden-tify potential new therapeutic targets and approaches (discussed below).

ABC transporter gene expression, tumour progression and clinical outcome. Several observations from clinical studies cor-relate ABC transporters with malignant

progression and a more aggressive pheno-type. Clinical outcome might therefore be influenced by the ABC transporter levels that are found at diagnosis, even in the absence of a correlation between the known drug substrates of a given transporter and the drugs used to treat the cancer in question.

For solid tumours, the grade or degree of differentiation typically reflects the aggressive nature of the tumour, with less differentiated tumours possessing the greatest proliferative potential and a more aggressive phenotype. Various ABC transporters are expressed at higher levels in tumour subtypes or zones that are less differentiated. In primary untreated hepatocellular carcinoma, ABCC1 levels correlate with differentiation grade, tumour size and degree of microvascular invasion25. The same study also found that ABCC1 is expressed in hepatic progenitor cells, but is absent from mature hepatocytes and is strongly upregulated in carcinoma specimens, particularly in tumours classified as having the worst prognosis. Correlation of ABCC1 overexpression with tumour size was also noted in breast cancer26,27, and ABCC4 levels were found to decrease with differentiation of leukaemia cells to more mature leukocytes28. Similarly, in early-stage lung adenocarcinoma, ABCC3 expression was high in tumours but detectable only at low levels in some precursor lesions and in the corresponding normal lung cell type29. Similar trends were observed in pancreatic cancer specimens, which had significantly higher levels of ABCC3 than normal tissue30, with ABCC3 expression correlating with increasing tumour grade in the 31 tumour specimens assessed.

Although ABCB1 is the most well-known drug transporter and seems to be inducible in response to chemotherapeutic treatment, its level of expression also seems to reflect the tumour phenotype in colorectal carcino-mas in which ABCB1 levels correlate with invasion into vessels31. For those tumours expressing ABCB1, the cells at the inva-sive periphery had the highest levels; high ABCB1 levels were also observed in lymph node metastases31. ABCB1 is preferentially expressed in poorly differentiated colon tumours, but expression is undetectable in normal colon tissue32. Similarly, ABCB1 expression was highest in the largest and most aggressive tumours in a range of soft tissue sarcomas33.

Expression of the intracellular lipid trans-porters ABCA2 and ABCA3 was examined in acute myeloid leukaemia (AMl), and although ABCA3 expression levels correlated

with poor chemotherapeutic response ABCA2 expression did not. Interestingly, however, high expression of ABCA2 was found in leukaemia cells of patients with the highest white cell counts at disease presenta-tion34, suggesting that ABCA2 levels could influence the disease pathogenesis.

A study of ABC transporter expression in primary untreated tumours from 72 patients with advanced non-small-cell lung cancer revealed that high levels of both ABCC2 and ABCG2 were predictive of poor prognosis following cisplatin treatment, and patients with tumours that stained positive for ABCG2 tended to respond less well to ther-apy35. Although ABCC2 is a well-established transporter of platinum conjugates, these compounds are not transported by ABCG2, suggesting that ABCG2 could have a role in the response of tumours to chemotherapy that is independent of drug efflux.

The paediatric malignancy neuroblastoma is an embryonic tumour of the neural crest that is often metastatic at presentation36. Poor survival rates are frequently associated with amplification of the MYCN oncogene, and the development of MDR is one of the major causes of treatment failure. Studies quantifying ABCC1 and ABCC4 expression in primary untreated neuroblastoma found that overexpression of either transporter was independently prognostic of event-free sur-vival37,38. The expression levels of a set of genes that included ABCC1 and ABCC4 were exam-ined in a microarray study of an independent cohort of 251 primary neuroblastomas39. Analysis of these data also showed that high levels of ABCC1 and ABCC4 were strongly predictive of adverse clinical outcome (J.I.F., M.H., M.J.H. and M.D.N., unpublished observations), whereas other ABCC sub-family members were not predictive. ABCC1 is known to efflux several drugs that are used for neuroblastoma treatment. However, the relationship between high ABCC4 expression and poor outcome was unexpected as none of the drugs used to treat the patients in either cohort is a known ABCC4 substrate, sug-gesting that ABCC4 might transport other molecules relevant to neuroblastoma development and/or progression.

Together, these studies indicate a pro-pensity for high level ABC transporter gene expression in tumour cells of a more malig-nant, less differentiated nature in multiple pri-mary untreated cancer types; however, these studies are largely correlative to date. one of the few examples in which an in vivo tumour model has been used to address causation comes from disruption of the Abcb1 gene in the ApcMin/+ mouse model, in which two

Glossary

LeukotrienesA class of arachidonic acid-derived lipid mediators involved in inflammation and homeostatic biological functions.

LipidomicsSystems-level analysis and characterization of cellular lipid profiles using technologies such as mass spectrometry and computational analysis.

Reconstituted vesicle systemsArtificial membrane systems mimicking natural lipid bilayers. Used to enable the full activity of membrane proteins, which often require correct orientation and bilayer insertion.

Smouldering inflammationLocalized chronic, and often subclinical, inflammation with few systemic manifestations.

XenobioticsExogenous chemicals, either naturally occurring or synthetic, found in a living organism.

P e r s P e c t i v e s

148 | FEBRUARy 2010 | VolUME 10 www.nature.com/reviews/cancer

© 20 Macmillan Publishers Limited. All rights reserved10

Page 3: ABC Transporters in Cancer

Nature Reviews | Cancer

DetoxificationExtracellular space

Drug resistance

Increased compound efflux

Increasedtransporterlevels

Increasedtransportergeneexpression

Increased compound efflux

Increasedtransporterlevels

Increasedtransportergeneexpression

DrugXenobiotic

Intracellular space ATP ADP

Nucleus

Metabolite

MDR transporter

independent studies have shown a significant reduction in the number of intestinal polyps in the absence of Abcb1 (REFS 40,41). Further studies in mouse tumour models, coupled with a knockdown approach or a transgenic approach, are required to address whether ABC transporters confer a fundamental biological advantage to tumour cells.

ABC transporters and hallmarks of cancerIn their seminal review in 2000, Hanahan and weinberg elucidated six essential acquired capabilities of cancer cells: self-sufficiency in growth signals, insensitivity to anti-growth signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis, and tissue invasion and metas-tasis42. Since then, compelling evidence has led to the proposal of a seventh capability involving cancer-related inflammation43. As outlined below, several observations suggest that ABC transporters might contribute to some of these cancer hallmarks. In addition, signalling lipids with established roles in tumour biology, including prostaglandins, leukotrienes and sphingosine-1-phosphate (S1P), are either known or strongly suspected to be ABC transporter substrates, raising the question of whether ABC transporter- mediated efflux of these molecules is important in cancer.

Apoptosis and proliferation. The balance between programmed cell death and prolif-eration regulates cell numbers. Most, if not all, cancers acquire resistance to apoptosis42, tipping the balance towards the expansion of tumour populations. Although the abil-ity of ABC transporters to efflux cytotoxic drugs affects the resistance of cancer cells to apoptosis in the context of therapy, several reports provide evidence that ABC trans-porters might also promote cell survival independently of cytotoxic drug efflux. For example, the ABCB1 inhibitor valspodar can promote cell cycle arrest and apoptosis in human leukaemia cells as a single agent in vitro44–46, and can reduce engraftment and prolong overall survival in a mouse leukaemic xenograft model46, suggesting a dependence on ABCB1 for viability in these cells. However, another study indi-cates that valspodar can induce apoptosis independently of transporter inhibition in some cell lines47. Furthermore, expression of ABCB1 has been reported to delay apopto-sis in response to apoptotic stimuli in both normal and cancer cells48–51, and ABCB1 inhibition reverses this resistance49,52. Surprisingly, this function was suggested to be independent of active transport53;

however, a mechanistic explanation for these observations remains to be established, and at least one other study suggests that these phenotypic changes might not be directly attributable to ABCB1 (REF. 54).

In neuroblastoma, ABCC1 has also been implicated in promoting cell survival. Knock down of ABCC1 enhances spontaneous cell death in human neuroblastoma cell lines both in vitro55 and in a mouse xenograft model56. Consistent with these observations, expression of ABCC1 in primary untreated patient samples is a powerful independent prognostic factor in this disease37.

Increased proliferation also contributes to the expansion of tumour populations. Knock down of ABCC4 inhibits prolifera-tion of vascular smooth muscle cells both in vitro and in vivo57, and knock down of ABCC1 reduced the mitotic index in neuro-blastoma cell line xenografts56. Similarly, knockdown of ABCB1 by small interfering RNA (siRNA) suppressed proliferation of a colon cancer cell line in vitro and inhibited tumour expansion in a xenograft model58. In late retinal progenitors, proliferation was increased by transduction with a retrovirus that encoded ABCG2 and decreased with siRNA knockdown of ABCG2 (REF. 59).

Roles in cell differentiation and functional significance in stem cells. Pluripotent stem cells are capable of self-renewal and can give rise to the differentiated cells that constitute tissues. A striking feature of both normal and cancer stem cells is their high level of ABC transporter expression compared with their more differentiated progeny60 (TABLE 1). High ABC transporter expression might confer stem cells with enhanced protection from damage by xenobiotic substances, and it has been suggested that mice deficient in various transporters might be more sus-ceptible to mutagenic chemicals60; however, this is yet to be tested experimentally. The expression of ABC transporters in cancer stem cells also has important therapeu-tic implications, as tumours can have a population of inherently drug resistant pluripotent cells that can survive exposure to chemotherapeutic agents and re-establish the tumour, which is analogous to the stem cell-driven recovery of normal tissue after chemotherapy60. However, as for normal stem cells, it is not yet clear whether expres-sion of ABC transporters has a fundamental role in the cancer stem cell phenotype, or occurs as a result of other genetic changes during tumorigenesis.

Box 1 | ABC transporters and multidrug resistance

A major breakthrough in uncovering the mechanisms behind multidrug resistance (MDR) came in 1976, correlating drug resistance with the overexpression of a single protein encoded by the gene ATP binding cassette, subfamily B, member 1 (ABCB1; also known as MDR1). The encoded protein is an ATP-dependent efflux pump and a member of the ABC transporter family151. ABCB1 overexpression is now associated with treatment failure in many cancers, including those of the kidney, liver and colon, as well as lymphoma and leukaemia (reviewed in REF. 152). Some years later, ABCC1 (also known as multidrug resistance protein 1) was identified as having a role in MDR phenotypes of small-cell lung carcinoma cells153. ABCC1 overexpression has been correlated with drug resistance in prostate, lung and breast cancer, as well as in childhood neuroblastoma (reviewed in REFS 11, 154). ABCB1 and ABCC1 efflux overlapping but distinct sets of compounds (TABLE 1). The third major MDR pump to be discovered was ABCG2 (also known as BCRP), which has a remarkably wide range of substrates and has been associated with drug resistance in breast cancer and leukaemia155–157. A key physiological function of ABC transporters is the protection of cells from many toxic insults from either endogenous or exogenous molecules that can enter the cell by diffusion or active uptake (see the figure). The protective mechanism afforded by ABC transporter-mediated extrusion of such toxic substances, whether they are metabolic waste products, naturally occurring substances or drugs, can make tumour cells resistant to the toxic effects of various chemotherapeutic agents155–157.

P e r s P e c t i v e s

NATURE REVIEwS | CanCer VolUME 10 | FEBRUARy 2010 | 149

© 20 Macmillan Publishers Limited. All rights reserved10

Page 4: ABC Transporters in Cancer

whether ABC transporters have any direct role in regulating differentiation remains unclear. Current genetic evidence does not support an absolute requirement for any individual ABC transporter in the maintenance of normal stem cell compart-ments, and mice deficient in Abcg2, Abcb1 or Abcc1 and mice deficient in both Abcb1 and Abcg2 all develop normally19,61–65. Therefore, these genes do not seem to be responsible for stem cell maintenance

or regulation of cellular differentiation (although these studies did not formally investigate the self-renewal capacity of stem cells by the engraftment of second-ary recipients66). However, given the large number of known ABC transporters2 and their high expression levels in stem cells, it also remains conceivable that partial functional redundancy might mask their importance for stem cell maintenance or growth.

In contrast to knockout studies, however, several overexpression studies link ABC transporters with differentiation66. For instance, the transduction of mouse bone marrow cells with a retrovirus encoding ABCB1 leads to dramatic ex vivo stem cell expansion and myeloproliferative dis-order after engraftment67, and enforced expression of ABCG2 in bone marrow cells causes a reduction in mature prog-eny both in vivo and in vitro68. This raises

Table 1 | selected ABC transporters, cancer-related substrates and their expression in tumour stem cell-like populations*

aBC family

Chemotherapy substrates‡ Cancer-related cellular substrates

expression in cancer stem cell-like populations

refs

ABCA

ABCA1 ND S1P and cholesterol ND 140,174

ABCA2 Estramustine and mitoxantrone Cholesterol Lung cancer cell lines and AML 149,175

ABCA3 Anthracyclines Phospholipids Neuroblastoma 176,177

ABCB

ABCB1 Colchicine, anthracyclines, epipo-dophyllotoxins, vinca alkaloids, taxanes, camptothecins, bisantrene, imatinib, mitoxantrone, saquinivir, methotrexate and actinomycin D

PAF AML and lung cancer cell lines 165,175,178

ABCB4 Anthracyclines, vinca alkaloids, taxanes, epipodophyllotoxins and mitoxantrone

ND ND –

ABCB5 Anthracyclines, camptothecins and thiopurines

ND Melanoma 179

ABCB11 Taxanes ND ND –

ABCC

ABCC1§ Anthracyclines, mitoxantrone, vinca alkaloids, imatinib, epipodophyl-lotoxins, camptothecins, colchicine, saquinivir and methotrexate

LTC4, PGA

2, 15d-PGJ

2, PGE

2

and S1PSquamous cell carcinoma lines, lung cancer cell lines, glioma and AML

93,103,104, 121,122,141, 149,175,180,

181

ABCC2§ Vinca alkaloids, cisplatin, taxanes, anthracyclines, methotrexate, epipo-dophyllotoxins, camptothecins, mitoxantrone and saquinivir

LTC4, PGD

2, PGA

1 and PGE

2 ND 93,120

ABCC3§ Methotrexate and epipodophyllotoxins

LTC4 and 15d-PGJ

2 ND 104,124

ABCC4§ Thiopurines, PMEA, methotrexate, AZT and camptothecins

LTB4, LTC

4, PGA

1, PGE

1, PGE

2,

PGF1α, PGF

2α, TXB2, cAMP and

cGMP

ND 57,94–96, 123,159,160,

182,183

ABCC5 Thiopurines, methotrexate, cisplatin, PMEA and AZT

cAMP and cGMP ND 161

ABCC6 Anthracyclines, cisplatin and epipodophyllotoxins

LTC4

ND 117

ABCC10 Vinca alkaloids and taxanes LTC4

ND 119

ABCC11 Thiopurines LTC4, cAMP and cGMP ND 118,162

ABCG

ABCG2 Mitoxantrone, camptothecins, anthracyclins, bisantrene, imatinib, methotrexate, flavopiridol and epipodophyllotoxins

cGMP Lung cancer, AML, oesophageal carcinoma, glioma, neuroblastoma, squamous cell carcinoma cell lines, melanoma, ovarian cancer and nasopharyngeal carcinoma cell lines

149,175,177, 179–181,

184,185

ABC, ATP binding cassette; cAMP, cyclic AMP; cGMP, cyclic GMP; LT, leukotriene; ND, not determined; PAF, platelet activation factor; PG, prostaglandin; S1P, sphingosine- 1-phosphate; TX, thromboxane. *ABC transporters are major mediators of chemoresistance but also efflux endogenous substrates with well-established roles in tumour biology. ‡Reviewed in REFS 11,173. § Several substrates for ABCC1–4 are effluxed as either glutathione or glucuronide conjugates, or in a glutathione-dependent manner.

P e r s P e c t i v e s

150 | FEBRUARy 2010 | VolUME 10 www.nature.com/reviews/cancer

© 20 Macmillan Publishers Limited. All rights reserved10

Page 5: ABC Transporters in Cancer

the possibility that although transporters are not required to maintain an undif-ferentiated state under normal circum-stances, they might favour its retention when upregulated in cancer. Alternatively, the expression of ABC transporters in undifferentiated cells might simply be a consequence of their stem cell-like trans-criptional programmes rather than a causa-tive factor. For several tumour types, the tumour grade, a low degree of differentia-tion and poor patient outcome each cor-relate with the presence of transcriptional programmes closely resembling those seen in embryonic stem cells69,70. Interestingly, such transcriptional programmes can also be induced by the deregulated expression of MYC70. It is therefore essential to deter-mine whether the high expression of ABC transporters in tumours is functionally relevant for their development (aside from drug efflux) or whether they are simply bystanders and a consequence of an altered transcriptional programme in cancer cells, particularly in the case of MYC deregula-tion. ABC transporter-deficient mouse can-cer models are central to answering these questions, as is a better understanding of the transcriptional and post-translational regulation of ABC transporters.

Cell migration, invasion and metastasis. Metastatic disease is the cause of the vast majority of cancer deaths71. Although direct evidence linking ABC transporters to metas-tasis is currently lacking, roles are emerg-ing for these proteins in cell migration and invasion.

It is apparent that several ABC transport-ers have roles in the migration of normal cells. Induced migration of peripheral dendritic cells to lymph nodes is greatly reduced in Abcc1–/– mice22, and Abcc1-deficient dendritic cells also have markedly attenuated chemotactic responses in vitro. Remarkably, exogenous addition of the ABCC1 substrates leukotriene C4 (lTC4) and lTD4 overcame the absence of the transporter in vivo and in vitro22, suggesting a role for ABCC1 in autocrine signalling in mouse dendritic cell migration. other ABC transporters might also have a role, as ABCB1-specific neutralizing antibodies and the inhibitor verapamil prevented the migration of dendritic cells from human skin explants21. However, comparison of these results across species is complicated by differences in the roles of various fam-ily members24. In human dendritic cells, ABCC4 seems to have a more prominent role than ABCC1 in migration, with

ABCC4 knockdown or pharmaco logical inhibition by sildenafil preventing their migration from human skin explants23.

The apparent roles of ABC transporters in dendritic cell migration are also reflected in other normal and cancer cell lines. Reduction of ABCB1 levels by siRNA reduced the migra-tion of MCF-7 breast cancer cells in transwell migration and Matrigel invasion assays72. Using similar assays, a doxorubicin-selected, multidrug-resistant human melanoma line expressing high levels of ABCB1 showed a more invasive phenotype than the parental cell line73. Although it is possible that other genetic changes contributed to this pheno-type, knock down of ABCB1 by siRNA substantially reduced the invasiveness of this cell line in vitro. Expression of ABCB1 in the canine kidney cell line MDCK also stimu-lated increased migration74, and inhibition of ABCB1 reduced migration of the rat brain endothelial cell line RBE4 (REF. 74). Finally, the downregulation of ABCC1 and ABCC4 reduces the migration of neuroblastoma cell lines in wound-closure assays in vitro (J.I.F., M.H., M.J.H. and M.D.N., unpub-lished observations). Although it remains to be determined whether ABC transporters contribute to metastasis in vivo, several stud-ies indicate such a link. For example, in a breast cancer study, ABCC1 expression was higher in metastatic lymph nodes than in the corresponding primary tumour27, and in melanoma, ABCC1 and ABCC4 were more highly expressed in cell lines derived from metastases than in those derived from pri-mary tumours75. More direct lines of investi-gation will be required to formally investigate these links.

signalling lipid substrates and cancerVarious signalling molecules with estab-lished roles in tumour biology are either known or strongly suspected to be ABC transporter substrates, and for several of these, ABC transporters are either the only known or the most prominent efflux mecha-nism. These substrates, outlined in TABLE 1, include prostaglandins, leukotrienes and S1P (discussed below), as well as platelet activat-ing factor, cholesterol metabolites and cyclic nucleotides (BOX 2).

Prostaglandins. In some cancer types, tumour cells support an inflammatory microenvironment that promotes their own proliferation and survival, and which aids angiogenesis and metastasis76. Among the mediators of chronic inflammation are the eicosanoids (prostanoids and leukotrienes), which are synthesised from arachidonic

Box 2 | Additional ABC transporter substrates with relevance to cancer

Cyclic nucleotidesCyclic nucleotides are important secondary messengers downstream of G protein-coupled protein receptors with relevance to cancer biology (reviewed in REF. 158) and are effluxed by ATP binding cassette, subfamily C, member 4 (ABCC4), ABCC5, ABCC11 (REFS 57,96,159–162) and ABCG2 (REF. 163). The biological significance of these observations is currently unclear as affinities are low, raising the possibility that their transport is relevant under circumstances of high cyclic nucleotide levels only164. However, a recent study demonstrated that Abcc4 deficiency led to decreased extracellular and increased intracellular cAMP in culture96, hinting that ABCC4-mediated cyclic nucleotide transport is of physiological significance at least in some systems.

Platelet activating factorPlatelet activating factor (PAF) has been implicated in various tumour-associated functions and signals and is exported by ABCB1 (REF. 165). PAF activates its extracellular G protein-coupled receptor PAFR and induces upregulation of the anti-apoptotic proteins BCL-2 and BCL-X

L

(REF. 166). It has also been shown to contribute to angiogenesis in breast cancer167 and to facilitate metastasis in melanoma168 and colorectal carcinoma169.

CholesterolMany ABCA family members are involved in regulating lipid transport and metabolism3. Cholesterol is an essential component of cellular membranes, and can be rate-limiting for the rapid growth and division of tumour cells. Furthermore, cholesterol is a key constituent of lipid rafts in cellular membranes, which bring together cooperating membrane-bound proteins for efficient activation of phosphorylation cascades170,171. Not surprisingly, several cancers are associated with faulty cholesterol metabolism (reviewed in REF. 4). For example, prostate cancer xenografts propagated in severe combined immunodeficient mice showed a dependency on circulating cholesterol for cell survival and tumour growth171. Alternatively, as intracellular cholesterol levels affect the rate of low density lipoprotein uptake, cholesterol trafficking can influence the availability of intracellular fatty acids, such as arachidonic acid, for the generation of many important signalling intermediates172 (FIG. 1). The question of whether coordinated regulation of ABC transporters (such as those regulating cholesterol metabolism and those effluxing lipid signalling intermediates) might lead to synergistic effects on tumour phenotype is yet to be addressed.

P e r s P e c t i v e s

NATURE REVIEwS | CanCer VolUME 10 | FEBRUARy 2010 | 151

© 20 Macmillan Publishers Limited. All rights reserved10

Page 6: ABC Transporters in Cancer

ABCC1ABCC2ABCC4 ABCC4ABCC2

EP1–EP4 DP FP B-LT1 andB-LT2

CysLT1 andCysLT2

TP IP

Phospholipids

AA

PGH2 LTA4

TXA2 PGI2 PGE2 PGD2 PGF2α LTB4 LTC4

? ?

lyso-PC

PAF

PAFR

PLA2

Cell survival, proliferation, migration, invasion, angiogenesis and inflammation

S1PR1 andS1RR2

ABCA1ABCC1

S1P

Sphingomyelin

Ceramide

Sphingosine

OATPs

Catabolism

PGs and LTs PGsLTs

LTD4LTE4

Nature Reviews | Cancer

SK1 orSK2

ABCB1

ABCC1–4ABCC6ABCC10 ABCC11

ABCC4

CDase

SMase

LPCATPGFSTxS PGIS PGES PGDS LTA4H LTC4S

ALOX5COX

acid through the cyclooxygenase (CoX; also known as prostaglandin endo-peroxide synthase or PTGS) and arachido-nate 5-lipoxygenase (AloX5) pathways, respectively (FIG. 1).

Clinical observations and genetic evi-dence from mouse models demonstrate the importance of prostaglandin-mediated path-ways in cancer, particularly in colorectal can-cer, but also in several other tumour types, including breast, lung and liver cancers77–79. CoX is a crucial component of prosta-glandin synthesis, with the CoX1 isoform constitutively expressed in many tissues and the CoX2 isoform often detected in prema-lignant and malignant tissues, where it is a major therapeutic target77,80,81. Downstream enzymes involved in prostaglandin synthe-sis and the various prostaglandin receptors have also provoked interest as therapeutic targets79,82.

Prostaglandin signalling culminates in autocrine or paracrine activation of extra cellular G protein-coupled recep-tors (GPCRs) (FIG. 1). The consequences of prostaglandin receptor activation include the activation of the PI3K–Akt pathway83, which triggers various responses that drive tumour progression (such as cell prolifera-tion, survival and motility84) and might also contribute to growth factor autonomy79; the activation of β-catenin signalling to promote a progenitor-like phenotype85; the stimula-tion of angiogenesis through the expres-sion of angio genic factors such as vascular endothelial growth factor A (VEGFA)86; and the promotion of metastasis87. Prostaglandin signalling might also contribute to tumour evasion of immunosurveillance by inhibiting dendritic cell differentiation88, and to the recruitment of inflammatory cells that support smouldering inflammation76 and so further enhancing tumour growth.

Although the inhibition of both prosta-glandin synthesis enzymes and receptors has been the subject of numerous studies, little attention has been given to prostaglan-din efflux mechanisms, particularly in the context of cancer. Prostaglandin efflux from cells by simple diffusion is an inefficient process89 and it is offset by carrier-mediated re-uptake by the prostaglandin transporter (PGT)90, as well as other transporters91, and by subsequent intracellular oxidation by 15-hydroxyprostaglandin dehydrogenase (15-PGDH)92. However, several members of the ABCC family efficiently transport pros-taglandins (FIG. 1; TABLE 1) and in cancer cells in which these transporters are highly upreg-ulated, this is likely to be their principal method of efflux. PGE2, the prostaglandin

most closely linked with cancer biology, is a substrate of ABCC1 (REF. 93), ABCC2 (REF. 93) and ABCC4 (REFS 94,95); Abcc4 deficiency results in markedly reduced extra-cellular PGE2 levels in culture and reduced PGE metabolites in vivo96. Interestingly, in comparison to normal mucosa, ABCC4 expression is increased in human colorectal cancer specimens and cell lines, as well as in adenomas from the ApcMin/+ mouse model of spontaneous intestinal tumorigenesis; how-ever, PGT expression is decreased97, raising the possibility that prostaglandin signalling might be enhanced by increased efflux and decreased re-uptake. other prostaglandins are also ABC transporter substrates: PGD2 is effluxed by ABCC2 (REF. 93), and PGF2α can be effluxed by ABCC4 (REFS 94,95).

The importance of ABC transporters for eicosanoid biology could also extend to the regulation of the cyclopentenone prostag-landins 15d-Δ12,14-PGJ2 and PGA2, which are dehydration products of prostaglandins PGD2 and PGE2, respectively. The cyclopen-tenone prostaglandins exhibit potent anti-proliferative and anti-tumorigenic properties, and function intracellularly partly through nuclear receptors such as peroxisome prolifer-ator-activated receptor-γ (PPARγ) rather than through extracellular G protein-coupled pros-tanoid receptors98–102. Glutathione-conjugated 15d-Δ12,14-PGJ2 and PGA2 are substrates for ABCC1 and ABCC3 (REFS 103,104), which might facilitate their clearance from the cell. ABC transporters may therefore deliver pro-tumorigenic prostaglandins to their

Figure 1 | aBC transporters in tumour-promoting lipid signalling pathways. Eicosanoid (pros-taglandin and leukotriene) and platelet activating factor (PAF) synthesis is initiated by the release of arachidonic acid (AA) and lysophosphophatidylcholine (lyso-PC) from phospholipids by the action of phospholipase A2 (PLA2). AA is converted into prostaglandin H

2 (PGH

2) by prostaglandin endoperox-

ide synthase (PTGS; also known as cyclooxygenase (COX)) and then by the action of specific prostag-landin (PG) and thromboxane (Tx) synthases (depending on cell type) to PGE

2, PGD

2, PGF

2α, PGI2 and

thromboxane A2 (TXA

2). These short-lived molecules are effluxed by members of the ATP binding cas-

sette C (ABCC) subfamily, activate G protein-coupled receptors (GPCRs), including EP1–EP

4 (PGE

2

receptors), DP (PGD2 receptor), FP (PGF

2α receptor), IP (PGI2 receptor) and TP (TXA

2 receptor) and can

be subject to re-uptake by members of the solute carrier organic anion transporter (SLC or OATP) family. AA is also converted by 5-lipoxygenase (ALOX5) into leukotriene A

4 (LTA

4), and either hydro-

lysed by LTA4 hydrolase (LTA

4H) to LTB

4 or conjugated with glutathione by LTC

4 synthase (LTC

4S) to

produce LTC4, which can be further extracellularly metabolized to LTD

4 and LTE

4. LTs are effluxed by

ABCC subfamily members and activate GPCRs including the LTB4 receptors B-LT

1 and B-LT

2 and the

cysteinyl leukotriene receptors CysLT1 and CysLT

2. PAF is synthesized from lysophosphatidylcholine

(LPC) by LPC acetyltransferase (LPCAT), effluxed by ABCB1 and binds to the PAF receptor (PAFR). Sphingosine-1-phosphate (S1P) synthesis begins with the production of ceramide from sphingomyelin by sphingomyelinase (SMase). Ceramide is converted by ceramidase (CDase) to sphingosine, which is subsequently phosphorylated by sphingosine kinases (SK1 and SK2) to produce S1P. S1P is in turn effluxed by ABCC1 and ABCA1 and activates the GPCRs S1PR

1–SIPR

5. Activation of the various GPCRs

initiates various autocrine and paracrine effects that are relevant to tumour biology.

P e r s P e c t i v e s

152 | FEBRUARy 2010 | VolUME 10 www.nature.com/reviews/cancer

© 20 Macmillan Publishers Limited. All rights reserved10

Page 7: ABC Transporters in Cancer

extracellular GPCRs, while diverting anti-tumorigenic cyclopentenone prostaglandins from their intracellular targets.

Leukotrienes. like prostaglandin signalling, leukotriene synthesis also culminates in the activation of extracellular GPCRs (FIG. 1), leading to various effects that include the acti-vation of β-catenin to promote cell survival105, the promotion of proliferation106,107, leukocyte invasive behaviour108 and the secretion of pro-tumorigenic factors by neutrophils109. The potential of leukotriene pathways as targets in cancer is illustrated by the upregulation of leukotriene biosynthetic enzymes in vari-ous cancers110–113, the effectiveness of AloX5 inhibition in animal models of pancreatic cancer114, lung cancer110,115 and chronic mye-loid leukaemia (CMl)116, and by the recent demonstration that the absence of Alox5 impairs the function of leukaemic stem cells, preventing the development of leukaemia in a mouse model of BCR–ABl-induced CMl116.

ABCC1 is a major physiological media-tor of lTC4 efflux and is likely to be the transporter primarily responsible for the release of lTC4 from leukotriene-producing cells. Bone marrow-derived mast cells from Abcc1-deficient mice exhibit only limited lTC4 release and instead accumulate lTC4 intracellularly, and Abcc1-deficient mice show a greatly impaired response to leu-kotriene-inducing inflammatory stimuli65. Furthermore, dendritic cell migration from skin to lymph nodes is impaired in Abcc1-deficient mice and restored by exogenous cysteinyl leukotrienes65. Numerous other members of the ABCC subfamily have been implicated in leukotriene transport with ABCC2, ABCC3, ABCC4, ABCC6, ABCC10 and ABCC11 each being able to efflux lTC4 (REFS 117–124), and ABCC4 has also recently been shown to efflux lTB4 (REF. 123) (FIG. 1). The central role of ABCC transporters in leukotriene signalling sug-gests that they could provide alternative or additional therapeutic targets to enzymes involved in leukotriene synthesis and leukotriene receptors.

Sphingosine‑1‑phosphate. S1P synthesis and signalling is outlined in FIG. 1 and has recently been comprehensively reviewed125. Although S1P can function intracellularly through mechanisms that are currently unclear125, its key biological actions are medi-ated extracellularly through five high-affinity GPCRs (reviewed in REFS 126–129).

S1P is potently angiogenic, stimulating the migration of endothelial cells and promoting blood vessel formation130,131. The importance

of S1P for angiogenesis is illustrated by the S1P1 receptor-knockout mice132 and sphin-gosine kinase 1 and sphingosine kinase 2 double knockout mice133, which both exhibit lethal defects in vascular maturation. A central role for S1P in tumour biology, par-ticularly in angiogenesis, was recently dem-onstrated by substantially reduced tumour progression in mouse xenograft and allograft models after treatment with a S1P-specific monoclonal antibody, apparently by the inhi-bition of tumour-associated angiogenesis134. In addition, S1P has been implicated in the proliferation of tumour cells and their eva-sion of programmed cell death127,135,136, and it has been shown to stimulate invasiveness in human glioblastoma lines137. Cells over-expressing sphingosine kinase 1, the enzyme immediately responsible for S1P production, acquire the ability to form tumours in non-obese–severe combined immunodefi-cient (NoD–SCID) mice138. Furthermore, S1P may induce the expression of CoX2 and the production of PGE2 (REF. 139).

Recent evidence demonstrates that Abca1-deficient astrocytes have substantially reduced S1P efflux capacity, indicating that ABCA1 may be crucial for efficient S1P export from these cells140. In addition, on the basis of siRNA-mediated knockdown, ABCC1 has been suggested as a major efflux mechanism for S1P from mast cells141. Multiple export mechanisms seem to exist for S1P, and whether any of these individual mechanisms is of particular relevance to tumour biology remains to be established. Diseases such as neuroblastoma, in which both high ABCC1 levels37,142 and vascular index143 are strongly predictive of poor outcome, might be suitable to address these questions.

Future directionsThe range of cellular phenotypes observed with deletion, knock down and expression of ABC transporters, their ability to export known cancer-promoting substrates and their upregulation in cancer cells raise the possibility that ABC transporters have roles in cancer biology beyond the efflux of cyto-toxic drugs. However, much of the evidence is circumstantial and few studies have directly addressed these roles from a genetic perspec-tive. Further experiments are clearly required to determine the importance of various ABC transporters in mouse models of diseases for which they are predictive. Particularly enticing candidates include the deletion of Abcc4 in the TH-MyCN mouse model of neuroblastoma144 given that ABCC4 is strongly predictive of neuroblastoma outcome38 in a manner that seems to be independent of drug transport;

and the deletion of Abcc4 in mouse models of colon cancer (such as ApcMin/+ or ApcΔ716) given that PGE2, an ABCC4 substrate, is strongly implicated in colon cancer79 and that ABCC4 is upregulated in this disease97.

Although several lipid signalling mole-cules with roles in tumour biology have been identified as substrates of ABC transporters, there are likely to be more to be found. Most of the substrates have been identified from candidate approaches, and in most cases initially tested in reconstituted vesicle systems. To date, no large-scale analyses have been conducted to identify additional substrates of ABC transporters; however, this is likely to be a fruitful area of research. The emerg-ing field of lipidomics should prove to be a key technology for these studies145.

The contributions of ABC transport-ers to cancer hallmarks and the efflux of cancer-related cellular substrates provide an alternative rationale for developing targeted therapies against these proteins. To date, inhibitors of ABC transporters designed to reverse drug resistance have had limited clinical success11,13. Although some of the same complications might still apply, target-ing these alternative functions is different in several respects. Most inhibitors developed to date target ABCB1; however, most known cancer-related cellular ABC transporter sub-strates are effluxed by the ABCC subfamily (TABLE 1). Although several ABCC1 inhibi-tors have been developed or trialled146,147, few target other members of this subfamily. In addition, failure of a given inhibitor to suc-cessfully overcome drug efflux in vivo does not necessarily extend to other substrates, as small molecule inhibitors can have differen-tial, and sometimes opposing effects on the efflux of alternative substrates148.

ConclusionsABC transporters are well known to oncolo-gists and cancer researchers for their capacity to efflux an array of therapeutic compounds, which leads to drug resistance and treatment failure. Recent studies suggest that ABC transporters may have other important roles in tumour biology; however, evidence linking them to various cancer hallmarks and to the efflux of tumour-promoting substrates is so far largely correlative. Targeted studies are now needed to address whether increased ABC transporter expression contributes to cancer progression independently of drug efflux. Studies that broaden our understand-ing of the function of ABC transporters beyond drug efflux could lead to a re- evaluation of their roles in determining clinical outcome. Given the functional

P e r s P e c t i v e s

NATURE REVIEwS | CanCer VolUME 10 | FEBRUARy 2010 | 153

© 20 Macmillan Publishers Limited. All rights reserved10

Page 8: ABC Transporters in Cancer

redundancy observed for this highly conserved superfamily of transporter pro-teins, a combination of inhibitors may be required to effectively block a given transport function. Future studies examining their clin-ical importance should be carefully designed to enable the potential effects of all relevant transporters to be evaluated. Ultimately, this may entail widening the range of transporters screened in such studies. A handful of studies are leading the way in this respect, screening tumour cell lines or small tumour cohorts for expression of the entire ABC transporter family20,34,75,149,150. Initial results suggest important roles for these genes beyond the expected MDR phenotype and argue that these studies should be extended to large tumour cohorts with comprehensive accompanying clinicopathological data.Jamie I. Fletcher, Michelle Haber, Michelle J. Henderson

and Murray D. Norris are at the Children’s Cancer Institute Australia for Medical Research,

Lowry Cancer Research Centre, University of New South Wales, P.O. BOX 151,

Randwick NSW 2031, Australia. Correspondence to M.H. and M.D.N. e-mails: [email protected];

[email protected]

doi:10.1038/nrc2789Published online 15 January 2010

1. Gottesman, M. M., Fojo, T. & Bates, S. E. Multidrug resistance in cancer: role of ATP-dependent transporters. Nature Rev. Cancer 2, 48–58 (2002).

2. Dean, M. The genetics of ATP-binding cassette transporters. Methods Enzymol. 400, 409–429 (2005).

3. Albrecht, C. & Viturro, E. The ABCA subfamily – gene and protein structures, functions and associated hereditary diseases. Pflugers Arch. 453, 581–589 (2007).

4. Mack, J. T., Brown, C. B. & Tew, K. D. ABCA2 as a therapeutic target in cancer and nervous system disorders. Expert Opin. Ther. Targets. 12, 491–504 (2008).

5. Takahashi, K. et al. ABC proteins: key molecules for lipid homeostasis. Med. Mol. Morphol. 38, 2–12 (2005).

6. Herget, M. & Tampe, R. Intracellular peptide transporters in human – compartmentalization of the “peptidome”. Pflugers Arch. 453, 591–600 (2007).

7. Deeley, R. G., Westlake, C. & Cole, S. P. Transmembrane transport of endo- and xenobiotics by mammalian ATP-binding cassette multidrug resistance proteins. Physiol. Rev. 86, 849–899 (2006).

8. Wanders, R. J., Visser, W. F., van Roermund, C. W., Kemp, S. & Waterham, H. R. The peroxisomal ABC transporter family. Pflugers Arch. 453, 719–734 (2007).

9. Chen, Z. Q. et al. The essential vertebrate ABCE1 protein interacts with eukaryotic initiation factors. J. Biol. Chem. 281, 7452–7457 (2006).

10. Kerr, I. D. Sequence analysis of twin ATP binding cassette proteins involved in translational control, antibiotic resistance, and ribonuclease L inhibition. Biochem. Biophys. Res. Commun. 315, 166–173 (2004).

11. Szakacs, G., Paterson, J. K., Ludwig, J. A., Booth-Genthe, C. & Gottesman, M. M. Targeting multidrug resistance in cancer. Nature Rev. Drug Discov. 5, 219–234 (2006).

12. Teodori, E., Dei, S., Martelli, C., Scapecchi, S. & Gualtieri, F. The functions and structure of ABC transporters: implications for the design of new inhibitors of Pgp and MRP1 to control multidrug resistance (MDR). Curr. Drug Targets. 7, 893–909 (2006).

13. Kaye, S. B. Reversal of drug resistance in ovarian cancer: where do we go from here? J. Clin. Oncol. 26, 2616–2618 (2008).

14. Lhomme, C. et al. Phase III study of valspodar (PSC 833) combined with paclitaxel and carboplatin compared with paclitaxel and carboplatin alone in patients with stage IV or suboptimally debulked stage III epithelial ovarian cancer or primary peritoneal cancer. J. Clin. Oncol. 26, 2674–2682 (2008).

15. Greenberg, P. L. et al. Mitoxantrone, etoposide, and cytarabine with or without valspodar in patients with relapsed or refractory acute myeloid leukemia and high-risk myelodysplastic syndrome: a phase III trial (E2995). J. Clin. Oncol. 22, 1078–1086 (2004).

16. Ruff, P. et al. A randomized, placebo-controlled, double-blind phase 2 study of docetaxel compared to docetaxel plus zosuquidar (LY335979) in women with metastatic or locally recurrent breast cancer who have received one prior chemotherapy regimen. Cancer Chemother. Pharmacol. 64, 763–768 (2009).

17. Garraway, L. A. & Chabner, B. MDR1 inhibition: less resistance or less relevance? Eur. J. Cancer 38, 2337–2340 (2002).

18. Relling, M. V. Are the major effects of P-glycoprotein modulators due to altered pharmacokinetics of anticancer drugs? Ther. Drug Monit. 18, 350–356 (1996).

19. Schinkel, A. H. et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell 77, 491–502 (1994).

20. Szakacs, G. et al. Predicting drug sensitivity and resistance: profiling ABC transporter genes in cancer cells. Cancer Cell 6, 129–137 (2004).

21. Randolph, G. J. et al. A physiologic function for p-glycoprotein (MDR-1) during the migration of dendritic cells from skin via afferent lymphatic vessels. Proc. Natl Acad. Sci. USA 95, 6924–6929 (1998).

22. Robbiani, D. F. et al. The leukotriene C4 transporter MRP1 regulates CCL19 (MIP-3β, ELC)-dependent mobilization of dendritic cells to lymph nodes. Cell 103, 757–768 (2000).

23. van de Ven, R. et al. A role for multidrug resistance protein 4 (MRP4; ABCC4) in human dendritic cell migration. Blood 112, 2353–2359 (2008).

24. van de Ven, R., Scheffer, G. L., Scheper, R. J. & de Gruijl, T. D. The ABC of dendritic cell development and function. Trends Immunol. 30, 421–429 (2009).

25. Vander Borght, S. et al. Expression of multidrug resistance-associated protein 1 in hepatocellular carcinoma is associated with a more aggressive tumour phenotype and may reflect a progenitor cell origin. Liver Int. 28, 1370–1380 (2008).

26. Filipits, M. et al. MRP and MDR1 gene expression in primary breast carcinomas. Clin. Cancer Res. 2, 1231–1237 (1996).

27. Zochbauer-Muller, S. et al. P-glycoprotein and MRP1 expression in axillary lymph node metastases of breast cancer patients. Anticancer Res. 21, 119–124 (2001).

28. Oevermann, L. et al. Hematopoietic stem cell differentiation affects expression and function of MRP4 (ABCC4), a transport protein for signaling molecules and drugs. Int. J. Cancer 124, 2303–2311 (2009).

29. Hanada, S. et al. Expression profile of early lung adenocarcinoma: identification of MRP3 as a molecular marker for early progression. J. Pathol. 216, 75–82 (2008).

30. Konig, J. et al. Expression and localization of human multidrug resistance protein (ABCC) family members in pancreatic carcinoma. Int. J. Cancer 115, 359–367 (2005).

31. Weinstein, R. S. et al. Relationship of the expression of the multidrug resistance gene product (P-glycoprotein) in human colon carcinoma to local tumor aggressiveness and lymph node metastasis. Cancer Res. 51, 2720–2726 (1991).

32. Ohtsuki, S. et al. Correlation of induction of ATP binding cassette transporter A5 (ABCA5) and ABCB1 mRNAs with differentiation state of human colon tumor. Biol. Pharm. Bull. 30, 1144–1146 (2007).

33. Oda, Y. et al. ATP-binding cassette superfamily transporter gene expression in human soft tissue sarcomas. Int. J. Cancer 114, 854–862 (2005).

34. Steinbach, D. et al. ABCA3 as a possible cause of drug resistance in childhood acute myeloid leukemia. Clin. Cancer Res. 12, 4357–4363 (2006).

35. Yoh, K. et al. Breast cancer resistance protein impacts clinical outcome in platinum-based chemotherapy for advanced non-small cell lung cancer. Clin. Cancer Res. 10, 1691–1697 (2004).

36. Maris, J. M., Hogarty, M. D., Bagatell, R. & Cohn, S. L. Neuroblastoma. Lancet 369, 2106–2120 (2007).

37. Haber, M. et al. Association of high-level MRP1 expression with poor clinical outcome in a large prospective study of primary neuroblastoma. J. Clin. Oncol. 24, 1546–1553 (2006).

38. Norris, M. D. et al. Expression of multidrug transporter MRP4/ABCC4 is a marker of poor prognosis in neuroblastoma and confers resistance to irinotecan in vitro. Mol. Cancer Ther. 4, 547–553 (2005).

39. Oberthuer, A. et al. Customized oligonucleotide microarray gene expression-based classification of neuroblastoma patients outperforms current clinical risk stratification. J. Clin. Oncol. 24, 5070–5078 (2006).

40. Mochida, Y. et al. The role of P-glycoprotein in intestinal tumorigenesis: disruption of mdr1a suppresses polyp formation in ApcMin/+ mice. Carcinogenesis 24, 1219–1224 (2003).

41. Yamada, T. et al. Suppression of intestinal polyposis in Mdr1-deficient ApcMin/+ mice. Cancer Res. 63, 895–901 (2003).

42. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

43. Mantovani, A. Cancer: inflaming metastasis. Nature 457, 36–37 (2009).

44. Lehne, G. & Rugstad, H. E. Cytotoxic effect of the cyclosporin PSC 833 in multidrug-resistant leukaemia cells with increased expression of P-glycoprotein. Br. J. Cancer 78, 593–600 (1998).

45. Lehne, G., De Angelis, P., den Boer, M. & Rugstad, H. E. Growth inhibition, cytokinesis failure and apoptosis of multidrug-resistant leukemia cells after treatment with P-glycoprotein inhibitory agents. Leukemia 13, 768–778 (1999).

46. Lehne, G. et al. The cyclosporin PSC 833 increases survival and delays engraftment of human multidrug-resistant leukemia cells in xenotransplanted NOD-SCID mice. Leukemia 16, 2388–2394 (2002).

47. Lopes, E. C. et al. Multidrug resistance modulators PSC 833 and CsA show differential capacity to induce apoptosis in lymphoid leukemia cell lines independently of their MDR phenotype. Leuk. Res. 27, 413–423 (2003).

48. Robinson, L. J. et al. Human MDR 1 protein overexpression delays the apoptotic cascade in Chinese hamster ovary fibroblasts. Biochemistry 36, 11169–11178 (1997).

49. Smyth, M. J., Krasovskis, E., Sutton, V. R. & Johnstone, R. W. The drug efflux protein, P-glycoprotein, additionally protects drug-resistant tumor cells from multiple forms of caspase-dependent apoptosis. Proc. Natl Acad. Sci. USA 95, 7024–7029 (1998).

50. Johnstone, R. W., Cretney, E. & Smyth, M. J. P-glycoprotein protects leukemia cells against caspase-dependent, but not caspase-independent, cell death. Blood 93, 1075–1085 (1999).

51. Pallis, M. & Russell, N. P-glycoprotein plays a drug-efflux-independent role in augmenting cell survival in acute myeloblastic leukemia and is associated with modulation of a sphingomyelin-ceramide apoptotic pathway. Blood 95, 2897–2904 (2000).

52. Bezombes, C. et al. Restoration of TNF-α-induced ceramide generation and apoptosis in resistant human leukemia KG1a cells by the P-glycoprotein blocker PSC833. FASEB J. 12, 101–109 (1998).

53. Tainton, K. M. et al. Mutational analysis of P-glycoprotein: suppression of caspase activation in the absence of ATP-dependent drug efflux. Cell Death Differ. 11, 1028–1037 (2004).

54. Aleman, C. et al. P-glycoprotein, expressed in multidrug resistant cells, is not responsible for alterations in membrane fluidity or membrane potential. Cancer Res. 63, 3084–3091 (2003).

55. Peaston, A. E. et al. MRP1 gene expression level regulates the death and differentiation response of neuroblastoma cells. Br. J. Cancer 85, 1564–1571 (2001).

56. Kuss, B. J. et al. In vitro and in vivo downregulation of MRP1 by antisense oligonucleotides: a potential role in neuroblastoma therapy. Int. J. Cancer 98, 128–133 (2002).

57. Sassi, Y. et al. Multidrug resistance-associated protein 4 regulates cAMP-dependent signaling pathways and controls human and rat SMC proliferation. J. Clin. Invest. 118, 2747–2757 (2008).

58. Katoh, S. Y., Ueno, M. & Takakura, N. Involvement of MDR1 function in proliferation of tumour cells. J. Biochem. 143, 517–524 (2008).

59. Bhattacharya, S., Das, A., Mallya, K. & Ahmad, I. Maintenance of retinal stem cells by Abcg2 is regulated by notch signaling. J. Cell Sci. 120, 2652–2662 (2007).

P e r s P e c t i v e s

154 | FEBRUARy 2010 | VolUME 10 www.nature.com/reviews/cancer

© 20 Macmillan Publishers Limited. All rights reserved10

Page 9: ABC Transporters in Cancer

60. Dean, M., Fojo, T. & Bates, S. Tumour stem cells and drug resistance. Nature Rev. Cancer 5, 275–284 (2005).

61. Zhou, S. et al. Bcrp1 gene expression is required for normal numbers of side population stem cells in mice, and confers relative protection to mitoxantrone in hematopoietic cells in vivo. Proc. Natl Acad. Sci. USA 99, 12339–12344 (2002).

62. Uchida, N., Leung, F. Y. & Eaves, C. J. Liver and marrow of adult mdr-1a/1b-/-mice show normal generation, function, and multi-tissue trafficking of primitive hematopoietic cells. Exp. Hematol. 30, 862–869 (2002).

63. Jonker, J. W. et al. The breast cancer resistance protein protects against a major chlorophyll-derived dietary phototoxin and protoporphyria. Proc. Natl Acad. Sci. USA 99, 15649–15654 (2002).

64. Zhou, S., Zong, Y., Lu, T. & Sorrentino, B. P. Hematopoietic cells from mice that are deficient in both Bcrp1/Abcg2 and Mdr1a/1b develop normally but are sensitized to mitoxantrone. Biotechniques 35, 1248–1252 (2003).

65. Wijnholds, J. et al. Increased sensitivity to anticancer drugs and decreased inflammatory response in mice lacking the multidrug resistance-associated protein. Nature Med. 3, 1275–1279 (1997).

66. Raaijmakers, M. H. ATP-binding-cassette transporters in hematopoietic stem cells and their utility as therapeutical targets in acute and chronic myeloid leukemia. Leukemia 21, 2094–2102 (2007).

67. Bunting, K. D., Galipeau, J., Topham, D., Benaim, E. & Sorrentino, B. P. Transduction of murine bone marrow cells with an MDR1 vector enables ex vivo stem cell expansion, but these expanded grafts cause a myeloproliferative syndrome in transplanted mice. Blood 92, 2269–2279 (1998).

68. Zhou, S. et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nature Med. 7, 1028–1034 (2001).

69. Ben-Porath, I. et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nature Genet. 40, 499–507 (2008).

70. Wong, D. J. et al. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell 2, 333–344 (2008).

71. Steeg, P. S. Tumor metastasis: mechanistic insights and clinical challenges. Nature Med. 12, 895–904 (2006).

72. Miletti-Gonzalez, K. E. et al. The CD44 receptor interacts with P-glycoprotein to promote cell migration and invasion in cancer. Cancer Res. 65, 6660–6667 (2005).

73. Colone, M. et al. The multidrug transporter P-glycoprotein: a mediator of melanoma invasion? J. Invest. Dermatol. 128, 957–971 (2008).

74. Barakat, S. et al. Regulation of brain endothelial cells migration and angiogenesis by P-glycoprotein/caveolin-1 interaction. Biochem. Biophys. Res. Commun. 372, 440–446 (2008).

75. Heimerl, S., Bosserhoff, A. K., Langmann, T., Ecker, J. & Schmitz, G. Mapping ATP-binding cassette transporter gene expression profiles in melanocytes and melanoma cells. Melanoma Res. 17, 265–273 (2007).

76. Mantovani, A., Allavena, P., Sica, A. & Balkwill, F. Cancer-related inflammation. Nature 454, 436–444 (2008).

77. Dannenberg, A. J. & Subbaramaiah, K. Targeting cyclooxygenase-2 in human neoplasia: rationale and promise. Cancer Cell 4, 431–436 (2003).

78. Gasparini, G., Longo, R., Sarmiento, R. & Morabito, A. Inhibitors of cyclo-oxygenase 2: a new class of anticancer agents? Lancet Oncol. 4, 605–615 (2003).

79. Greenhough, A. et al. The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 30, 377–386 (2009).

80. Muller, A. J. & Scherle, P. A. Targeting the mechanisms of tumoral immune tolerance with small-molecule inhibitors. Nature Rev. Cancer 6, 613–625 (2006).

81. Fulton, A. M., Ma, X. & Kundu, N. Targeting prostaglandin E EP receptors to inhibit metastasis. Cancer Res. 66, 9794–9797 (2006).

82. Chell, S., Kaidi, A., Williams, A. C. & Paraskeva, C. Mediators of PGE2 synthesis and signalling downstream of COX-2 represent potential targets for the prevention/treatment of colorectal cancer. Biochim. Biophys. Acta 1766, 104–119 (2006).

83. Tessner, T. G., Muhale, F., Riehl, T. E., Anant, S. & Stenson, W. F. Prostaglandin E2 reduces radiation-induced epithelial apoptosis through a mechanism involving AKT activation and bax translocation. J. Clin. Invest. 114, 1676–1685 (2004).

84. Vivanco, I. & Sawyers, C. L. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nature Rev. Cancer 2, 489–501 (2002).

85. Castellone, M. D., Teramoto, H., Williams, B. O., Druey, K. M. & Gutkind, J. S. Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-β-catenin signaling axis. Science 310, 1504–1510 (2005).

86. Tsujii, M. et al. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 93, 705–716 (1998).

87. Gupta, G. P. et al. Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature 446, 765–770 (2007).

88. Sombroek, C. C. et al. Prostanoids play a major role in the primary tumor-induced inhibition of dendritic cell differentiation. J. Immunol. 168, 4333–4343 (2002).

89. Chan, B. S., Satriano, J. A., Pucci, M. & Schuster, V. L. Mechanism of prostaglandin E2 transport across the plasma membrane of HeLa cells and Xenopus oocytes expressing the prostaglandin transporter “PGT”. J. Biol. Chem. 273, 6689–6697 (1998).

90. Nomura, T., Lu, R., Pucci, M. L. & Schuster, V. L. The two-step model of prostaglandin signal termination: in vitro reconstitution with the prostaglandin transporter and prostaglandin 15 dehydrogenase. Mol. Pharmacol. 65, 973–978 (2004).

91. Schuster, V. L. Prostaglandin transport. Prostaglandins Other Lipid Mediat. 68–69, 633–647 (2002).

92. Tai, H. H., Ensor, C. M., Tong, M., Zhou, H. & Yan, F. Prostaglandin catabolizing enzymes. Prostaglandins Other Lipid Mediat. 68–69, 483–493 (2002).

93. de Waart, D. R., Paulusma, C. C., Kunne, C. & Oude Elferink, R. P. Multidrug resistance associated protein 2 mediates transport of prostaglandin E2. Liver Int. 26, 362–368 (2006).

94. Reid, G. et al. The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc. Natl Acad. Sci. USA 100, 9244–9249 (2003).

95. Rius, M., Thon, W. F., Keppler, D. & Nies, A. T. Prostanoid transport by multidrug resistance protein 4 (MRP4/ABCC4) localized in tissues of the human urogenital tract. J. Urol. 174, 2409–2414 (2005).

96. Lin, Z. P. et al. Disruption of cAMP and prostaglandin E2 transport by multidrug resistance protein 4 deficiency alters cAMP-mediated signaling and nociceptive response. Mol. Pharmacol. 73, 243–251 (2008).

97. Holla, V. R., Backlund, M. G., Yang, P., Newman, R. A. & DuBois, R. N. Regulation of prostaglandin transporters in colorectal neoplasia. Cancer Prev. Res. (Phila Pa) 1, 93–99 (2008).

98. Milne, G. L., Musiek, E. S. & Morrow, J. D. The cyclopentenone (A2/J2) isoprostanes—unique, highly reactive products of arachidonate peroxidation. Antioxid. Redox Signal. 7, 210–220 (2005).

99. Musiek, E. S., Milne, G. L., McLaughlin, B. & Morrow, J. D. Cyclopentenone eicosanoids as mediators of neurodegeneration: a pathogenic mechanism of oxidative stress-mediated and cyclooxygenase-mediated neurotoxicity. Brain Pathol. 15, 149–158 (2005).

100. Negishi, M. & Katoh, H. Cyclopentenone prostaglandin receptors. Prostaglandins Other Lipid Mediat. 68–69, 611–617 (2002).

101. Straus, D. S. & Glass, C. K. Cyclopentenone prostaglandins: new insights on biological activities and cellular targets. Med. Res. Rev. 21, 185–210 (2001).

102. Michalik, L., Desvergne, B. & Wahli, W. Peroxisome-proliferator-activated receptors and cancers: complex stories. Nature Rev. Cancer 4, 61–70 (2004).

103. Evers, R. et al. Transport of glutathione prostaglandin A conjugates by the multidrug resistance protein 1. FEBS Lett. 419, 112–116 (1997).

104. Paumi, C. M., Wright, M., Townsend, A. J. & Morrow, C. S. Multidrug resistance protein (MRP) 1 and MRP3 attenuate cytotoxic and transactivating effects of the cyclopentenone prostaglandin, 15-deoxy-Δ12,14prostaglandin J2 in MCF7 breast cancer cells. Biochemistry 42, 5429–5437 (2003).

105. Mezhybovska, M., Wikstrom, K., Ohd, J. F. & Sjolander, A. The inflammatory mediator leukotriene D4 induces β-catenin signaling and its association with antiapoptotic Bcl-2 in intestinal epithelial cells. J. Biol. Chem. 281, 6776–6784 (2006).

106. Nielsen, C. K. et al. A novel localization of the G-protein-coupled CysLT1 receptor in the nucleus of colorectal adenocarcinoma cells. Cancer Res. 65, 732–742 (2005).

107. Ihara, A. et al. Blockade of leukotriene B4 signaling pathway induces apoptosis and suppresses cell proliferation in colon cancer. J. Pharmacol. Sci. 103, 24–32 (2007).

108. Peters-Golden, M. & Henderson, W. R. Jr. Leukotrienes. N. Engl. J. Med. 357, 1841–1854 (2007).

109. Tazzyman, S., Lewis, C. E. & Murdoch, C. Neutrophils: key mediators of tumour angiogenesis. Int. J. Exp. Pathol. 90, 222–231 (2009).

110. Avis, I. M. et al. Growth control of lung cancer by interruption of 5-lipoxygenase-mediated growth factor signaling. J. Clin. Invest. 97, 806–813 (1996).

111. Hong, S. H. et al. Relationship of arachidonic acid metabolizing enzyme expression in epithelial cancer cell lines to the growth effect of selective biochemical inhibitors. Cancer Res. 59, 2223–2228 (1999).

112. Boado, R. J., Pardridge, W. M., Vinters, H. V. & Black, K. L. Differential expression of arachidonate 5-lipoxygenase transcripts in human brain tumors: evidence for the expression of a multitranscript family. Proc. Natl Acad. Sci. USA 89, 9044–9048 (1992).

113. Hennig, R. et al. 5-Lipoxygenase and leukotriene B4 receptor are expressed in human pancreatic cancers but not in pancreatic ducts in normal tissue. Am. J. Pathol. 161, 421–428 (2002).

114. Tong, W. G., Ding, X. Z., Witt, R. C. & Adrian, T. E. Lipoxygenase inhibitors attenuate growth of human pancreatic cancer xenografts and induce apoptosis through the mitochondrial pathway. Mol. Cancer Ther. 1, 929–935 (2002).

115. Gunning, W. T., Kramer, P. M., Steele, V. E. & Pereira, M. A. Chemoprevention by lipoxygenase and leukotriene pathway inhibitors of vinyl carbamate-induced lung tumors in mice. Cancer Res. 62, 4199–4201 (2002).

116. Chen, Y., Hu, Y., Zhang, H., Peng, C. & Li, S. Loss of the Alox5 gene impairs leukemia stem cells and prevents chronic myeloid leukemia. Nature Genet. 41, 783–792 (2009).

117. Belinsky, M. G., Chen, Z. S., Shchaveleva, I., Zeng, H. & Kruh, G. D. Characterization of the drug resistance and transport properties of multidrug resistance protein 6 (MRP6, ABCC6). Cancer Res. 62, 6172–6177 (2002).

118. Chen, Z. S., Guo, Y., Belinsky, M. G., Kotova, E. & Kruh, G. D. Transport of bile acids, sulfated steroids, estradiol 17-β-D-glucuronide, and leukotriene C4 by human multidrug resistance protein 8 (ABCC11). Mol. Pharmacol. 67, 545–557 (2005).

119. Chen, Z. S. et al. Characterization of the transport properties of human multidrug resistance protein 7 (MRP7, ABCC10). Mol. Pharmacol. 63, 351–358 (2003).

120. Cui, Y. et al. Drug resistance and ATP-dependent conjugate transport mediated by the apical multidrug resistance protein, MRP2, permanently expressed in human and canine cells. Mol. Pharmacol. 55, 929–937 (1999).

121. Jedlitschky, G., Leier, I., Buchholz, U., Center, M. & Keppler, D. ATP-dependent transport of glutathione S-conjugates by the multidrug resistance-associated protein. Cancer Res. 54, 4833–4836 (1994).

122. Leier, I. et al. The MRP gene encodes an ATP-dependent export pump for leukotriene C4 and structurally related conjugates. J. Biol. Chem. 269, 27807–27810 (1994).

123. Rius, M., Hummel-Eisenbeiss, J. & Keppler, D. ATP-dependent transport of leukotrienes B4 and C4 by the multidrug resistance protein ABCC4 (MRP4). J. Pharmacol. Exp. Ther. 324, 86–94 (2008).

124. Zeng, H., Liu, G., Rea, P. A. & Kruh, G. D. Transport of amphipathic anions by human multidrug resistance protein 3. Cancer Res. 60, 4779–4784 (2000).

125. Hannun, Y. A. & Obeid, L. M. Principles of bioactive lipid signalling: lessons from sphingolipids. Nature Rev. Mol. Cell Biol. 9, 139–150 (2008).

126. Ishii, I., Fukushima, N., Ye, X. & Chun, J. Lysophospholipid receptors: signaling and biology. Annu. Rev. Biochem. 73, 321–354 (2004).

P e r s P e c t i v e s

NATURE REVIEwS | CanCer VolUME 10 | FEBRUARy 2010 | 155

© 20 Macmillan Publishers Limited. All rights reserved10

Page 10: ABC Transporters in Cancer

127. Spiegel, S. & Milstien, S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nature Rev. Mol. Cell Biol. 4, 397–407 (2003).

128. Kluk, M. J. & Hla, T. Signaling of sphingosine-1-phosphate via the S1P/EDG-family of G-protein-coupled receptors. Biochim. Biophys. Acta 1582, 72–80 (2002).

129. Taha, T. A., Argraves, K. M. & Obeid, L. M. Sphingosine-1-phosphate receptors: receptor specificity versus functional redundancy. Biochim. Biophys. Acta 1682, 48–55 (2004).

130. Lee, M. J. et al. Vascular endothelial cell adherens junction assembly and morphogenesis induced by sphingosine-1-phosphate. Cell 99, 301–312 (1999).

131. Kimura, T. et al. Sphingosine 1-phosphate stimulates proliferation and migration of human endothelial cells possibly through the lipid receptors, Edg-1 and Edg-3. Biochem. J. 348, 71–76 (2000).

132. Liu, Y. et al. Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J. Clin. Invest. 106, 951–961 (2000).

133. Mizugishi, K. et al. Essential role for sphingosine kinases in neural and vascular development. Mol. Cell. Biol. 25, 11113–11121 (2005).

134. Visentin, B. et al. Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell 9, 225–238 (2006).

135. Maceyka, M., Payne, S. G., Milstien, S. & Spiegel, S. Sphingosine kinase, sphingosine-1-phosphate, and apoptosis. Biochim. Biophys. Acta 1585, 193–201 (2002).

136. Radeff-Huang, J., Seasholtz, T. M., Matteo, R. G. & Brown, J. H. G protein mediated signaling pathways in lysophospholipid induced cell proliferation and survival. J. Cell Biochem. 92, 949–966 (2004).

137. Van Brocklyn, J. R., Young, N. & Roof, R. Sphingosine-1-phosphate stimulates motility and invasiveness of human glioblastoma multiforme cells. Cancer Lett. 199, 53–60 (2003).

138. Xia, P. et al. An oncogenic role of sphingosine kinase. Curr. Biol. 10, 1527–1530 (2000).

139. Pettus, B. J. et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-α. FASEB J. 17, 1411–1421 (2003).

140. Sato, K. et al. Critical role of ABCA1 transporter in sphingosine 1-phosphate release from astrocytes. J. Neurochem. 103, 2610–2619 (2007).

141. Mitra, P. et al. Role of ABCC1 in export of sphingosine-1-phosphate from mast cells. Proc. Natl Acad. Sci. USA 103, 16394–16399 (2006).

142. Norris, M. D. et al. Expression of the gene for multidrug-resistance-associated protein and outcome in patients with neuroblastoma. N. Engl. J. Med. 334, 231–238 (1996).

143. Meitar, D., Crawford, S. E., Rademaker, A. W. & Cohn, S. L. Tumor angiogenesis correlates with metastatic disease, N-myc amplification, and poor outcome in human neuroblastoma. J. Clin. Oncol. 14, 405–414 (1996).

144. Weiss, W., Aldape, K., Mohapatra, G., Feuerstein, B. & Bishop, J. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J. 16, 2985–2995 (1997).

145. Wenk, M. R. The emerging field of lipidomics. Nature Rev. Drug Discov. 4, 594–610 (2005).

146. Norman, B. H. et al. Cyclohexyl-linked tricyclic isoxazoles are potent and selective modulators of the multidrug resistance protein (MRP1). Bioorg Med. Chem. Lett. 15, 5526–5530 (2005).

147. O’Connor, R. et al. A phase I clinical and pharmacokinetic study of the multi-drug resistance protein-1 (MRP-1) inhibitor sulindac, in combination with epirubicin in patients with advanced cancer. Cancer Chemother. Pharmacol. 59, 79–87 (2007).

148. Kondratov, R. V., Komarov, P. G., Becker, Y., Ewenson, A. & Gudkov, A. V. Small molecules that dramatically alter multidrug resistance phenotype by modulating the substrate specificity of P-glycoprotein. Proc. Natl Acad. Sci. USA 98, 14078–14083 (2001).

149. de Grouw, E. P. et al. Preferential expression of a high number of ATP binding cassette transporters in both normal and leukemic CD34+CD38- cells. Leukemia 20, 750–754 (2006).

150. Park, S. et al. Gene expression profiling of ATP-binding cassette (ABC) transporters as a predictor of the pathologic response to neoadjuvant chemotherapy in breast cancer patients. Breast Cancer Res. Treat 99, 9–17 (2006).

151. Juliano, R. L. & Ling, V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim. Biophys. Acta 455, 152–162 (1976).

152. Ambudkar, S. V., Kimchi-Sarfaty, C., Sauna, Z. E. & Gottesman, M. M. P-glycoprotein: from genomics to mechanism. Oncogene 22, 7468–7485 (2003).

153. Cole, S. P. et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 258, 1650–1654 (1992).

154. Munoz, M., Henderson, M., Haber, M. & Norris, M. Role of the MRP1/ABCC1 multidrug transporter protein in cancer. IUBMB Life 59, 752–757 (2007).

155. Bunting, K. D. ABC transporters as phenotypic markers and functional regulators of stem cells. Stem Cells 20, 11–20 (2002).

156. Doyle, L. A. et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc. Natl Acad. Sci. USA 95, 15665–15670 (1998).

157. Robey, R. W., Polgar, O., Deeken, J., To, K. W. & Bates, S. E. ABCG2: determining its relevance in clinical drug resistance. Cancer Metastasis Rev. 26, 39–57 (2007).

158. Dorsam, R. T. & Gutkind, J. S. G-protein-coupled receptors and cancer. Nature Rev. Cancer 7, 79–94 (2007).

159. Chen, Z. S., Lee, K. & Kruh, G. D. Transport of cyclic nucleotides and estradiol 17-β-D-glucuronide by multidrug resistance protein 4. Resistance to 6-mercaptopurine and 6-thioguanine. J. Biol. Chem. 276, 33747–33754 (2001).

160. Wielinga, P. R. et al. Characterization of the MRP4- and MRP5-mediated transport of cyclic nucleotides from intact cells. J. Biol. Chem. 278, 17664–17671 (2003).

161. Jedlitschky, G., Burchell, B. & Keppler, D. The multidrug resistance protein 5 functions as an ATP-dependent export pump for cyclic nucleotides. J. Biol. Chem. 275, 30069–30074 (2000).

162. Guo, Y. et al. MRP8, ATP-binding cassette C11 (ABCC11), is a cyclic nucleotide efflux pump and a resistance factor for fluoropyrimidines 2’, 3’-dideoxycytidine and 9’-(2’-phosphonylmethoxyethyl)adenine. J. Biol. Chem. 278, 29509–29514 (2003).

163. de Wolf, C. J. et al. cGMP transport by vesicles from human and mouse erythrocytes. FEBS J. 274, 439–450 (2007).

164. Borst, P. et al. The potential impact of drug transporters on nucleoside-analog-based antiviral chemotherapy. Antiviral Res. 62, 1–7 (2004).

165. Raggers, R. J., Vogels, I. & van Meer, G. Multidrug-resistance P-glycoprotein (MDR1) secretes platelet-activating factor. Biochem. J. 357, 859–865 (2001).

166. Heon Seo, K. et al. Platelet-activating factor induces up-regulation of antiapoptotic factors in a melanoma cell line through nuclear factor-κB activation. Cancer Res. 66, 4681–4686 (2006).

167. Bussolati, B. et al. PAF produced by human breast cancer cells promotes migration and proliferation of tumor cells and neo-angiogenesis. Am. J. Pathol. 157, 1713–1725 (2000).

168. Melnikova, V. O., Mourad-Zeidan, A. A., Lev, D. C. & Bar-Eli, M. Platelet-activating factor mediates MMP-2 expression and activation via phosphorylation of cAMP-response element-binding protein and contributes to melanoma metastasis. J. Biol. Chem. 281, 2911–2922 (2006).

169. Denizot, Y. et al. Platelet-activating factor and liver metastasis of colorectal cancer. Int. J. Cancer 113, 503–505 (2005).

170. Kim, J., Adam, R. M., Solomon, K. R. & Freeman, M. R. Involvement of cholesterol-rich lipid rafts in interleukin-6-induced neuroendocrine differentiation of LNCaP prostate cancer cells. Endocrinology 145, 613–619 (2004).

171. Zhuang, L., Kim, J., Adam, R. M., Solomon, K. R. & Freeman, M. R. Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J. Clin. Invest. 115, 959–968 (2005).

172. Hughes-Fulford, M., Chen, Y. & Tjandrawinata, R. R. Fatty acid regulates gene expression and growth of human prostate cancer PC-3 cells. Carcinogenesis 22, 701–707 (2001).

173. Gillet, J. P., Efferth, T. & Remacle, J. Chemotherapy-induced resistance by ATP-binding cassette transporter genes. Biochim. Biophys. Acta 1775, 237–262 (2007).

174. Oram, J. F., Wolfbauer, G., Vaughan, A. M., Tang, C. & Albers, J. J. Phospholipid transfer protein interacts with and stabilizes ATP-binding cassette transporter A1 and enhances cholesterol efflux from cells. J. Biol. Chem. 278, 52379–52385 (2003).

175. Ho, M. M., Ng, A. V., Lam, S. & Hung., J. Y. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res. 67, 4827–4833 (2007).

176. Ban, N. et al. ABCA3 as a lipid transporter in pulmonary surfactant biogenesis. J. Biol. Chem. 282, 9628–9634 (2007).

177. Hirschmann-Jax, C. et al. A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc. Natl Acad. Sci. USA 101, 14228–14233 (2004).

178. Raaijmakers, M. H. et al. ABCB1 modulation does not circumvent drug extrusion from primitive leukemic progenitor cells and may preferentially target residual normal cells in acute myelogenous leukemia. Clin. Cancer Res. 12, 3452–3458 (2006).

179. Schatton, T. et al. Identification of cells initiating human melanomas. Nature 451, 345–349 (2008).

180. Jin, F. et al. Comparison between cells and cancer stem-like cells isolated from glioblastoma and astrocytoma on expression of anti-apoptotic and multidrug resistance-associated protein genes. Neuroscience 154, 541–550 (2008).

181. Loebinger, M. R. et al. Squamous cell cancers contain a side population of stem-like cells that are made chemosensitive by ABC transporter blockade. Br. J. Cancer 98, 380–387 (2008).

182. Sager, G., Orbo, A., Pettersen, R. H. & Kjorstad, K. E. Export of guanosine 3’, 5’-cyclic monophosphate (cGMP) from human erythrocytes characterized by inside-out membrane vesicles. Scand. J. Clin. Lab. Invest. 56, 289–293 (1996).

183. Van Aubel, R. A., Smeets, P. H., van den Heuvel, J. J. & Russel, F. G. Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate binding sites. Am. J. Physiol. Renal Physiol. 288, F327–333 (2005).

184. Wang, J. et al. c-Myc is required for maintenance of glioma cancer stem cells. PLoS One 3, 3769 (2008).

185. Zhang, S. et al. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 68, 4311–4320 (2008).

AcknowledgementsThe authors were supported by grants from the National Health and Medical Research Council, Australia, Cancer Institute New South Wales, Australia, and Cancer Council New South Wales, Australia.

Competing interests statementThe authors declare no competing financial interests.

DATABAsesentrez Gene: http://www.ncbi.nlm.nih.gov/geneABCC3 | ABCC4National cancer institute Drug Dictionary: http://www.cancer.gov/drugdictionary/valspodar | zosuquidarUniProtKB: http://www.uniprot.orgABCA2 | ABCA3 | ABCB1 | ABCC1 | ABCC2 | ABCG2

FuRTheR inFoRMATionMichelle Haber’s homepage: http://www.ccia.org.au/index.cfm?pagecall=content&contentId=28522Murray D. Norris’s homepage: http://www.ccia.org.au/index.cfm?pagecall=content&contentId=28523ABc transporters: http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=mono_001&part=A137children’s cancer institute Australia for Medical research: http://www.ccia.org.au/

all links are aCtive in the online pdf

P e r s P e c t i v e s

156 | FEBRUARy 2010 | VolUME 10 www.nature.com/reviews/cancer

© 20 Macmillan Publishers Limited. All rights reserved10

Page 11: ABC Transporters in Cancer

Copyright of Nature Reviews Cancer is the property of Nature Publishing Group and its content may not be

copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written

permission. However, users may print, download, or email articles for individual use.