Abstract
Breast cancer is the second most common cancer worldwide and the leading cause of cancer death in women, with incidence rates that continue to rise. The heterogeneity of the disease makes breast cancer exceptionally difficult to treat, particularly for those patients with triple-negative disease. To address the therapeutic complexity of these tumours, new strategies for diagnosis and treatment are urgently required. The ability of lactating and malignant breast cells to uptake and transport iodide has led to the hypothesis that radioiodide therapy could be a potentially viable treatment for many breast cancer patients. Understanding how iodide is transported, and the factors regulating the expression and function of the proteins responsible for iodide transport, is critical for translating this hypothesis into reality. This review covers the three known iodide transporters – the sodium iodide symporter, pendrin and the sodium-coupled monocarboxylate transporter – and their role in iodide transport in breast cells, along with efforts to manipulate them to increase the potential for radioiodide therapy as a treatment for breast cancer.
Introduction
Based on current incidence projections, 3.2 million new cases of breast cancer will be diagnosed each year by 2050 (Hortobagyi et al. 2005). Meanwhile, the heterogeneity observed at both the intra- and inter-tumour levels continues to make the disease challenging to treat, particularly for those patients with metastatic triple-negative disease, where few treatment options are available. The inherent ability of breast cells to uptake iodide opens the possibility for a potential alternative treatment for breast cancer via radioiodide therapy, currently used in the management and diagnostic imaging of thyroid disorders.
Although utilisation of radioiodide treatment for breast cancer has been proposed previously, improved functional insight is required before it can be translated from bench to bedside. Crucially, the transport of iodide into breast cells must be maximal while iodide efflux is simultaneously minimised. Full functional understanding of the three major transporters of iodide in breast cells and how they may be manipulated is required for this proposed treatment to become a reality. This review aims to provide an overview of the transporters sodium iodide symporter (NIS), pendrin and sodium-coupled monocarboxylate transporter (SMCT), focusing on factors relating to their expression and function alongside strategies that would maximise their potential in breast cancer.
Sodium iodide symporter
The NIS is a large (643 amino acids) integral plasma membrane glycoprotein (Fig. 1), the primary role of which is transporting iodide (I−) into cells. The gene, also referred to as solute carrier family 5 member 5 (SLC5A5), was first cloned in 1996 (Dai et al. 1996), although the ability of the thyroid to accumulate iodide was reported as early as 1896 (Baumann 1896). The protein consists of thirteen transmembrane domains, an extracellular N-terminal and a cytosolic C-terminal tail and has been identified to be phosphorylated in vivo and contain three N-linked glycosylation sites at positions 225, 485 and 497 (Fig. 1) (Levy et al. 1998, Vadysirisack et al. 2007).
NIS expression is primarily observed in the thyroid along with salivary glands, gastric mucosa and lactating mammary gland cells, where it is usually located at the basolateral surface of the plasma membrane. In thyroid follicular cells, the cellular concentration of iodide is 20–50 times that of extracellular levels, so use of the inverse Na+ electrochemical gradient, maintained by the Na+/K+ ATPase, allows NIS to couple the transport of one iodide anion and two sodium cations into cells. The NIS-mediated transport of I− into thyroid follicular cells is the first and rate-limiting step of the biosynthesis of the thyroid hormones triiodothyronine (T3) and throxyine (T4) (Spitzweg et al. 2001). In the thyroid, the expression of NIS is principally regulated by the thyroid-stimulating hormone (TSH) (Kogai et al. 1997). When TSH binds to the TSH receptor (TSHR), adenylate cyclase is activated, leading to increased intracellular cAMP levels (Takasu et al. 1978), which further activates the transcription factors, cAMP response element-binding protein (CREB) and Pax8 (Poleev et al. 1997). The NIS upstream enhancer (NUE), which is fundamental in the initiation of NIS transcription, contains binding sites for both Pax8 and CREB, which stimulate NIS transcription upon transcription factor binding (Ohno et al. 1999).
Only two putative binding partners of NIS have been reported, both of which have been implicated with breast cancer; pituitary tumour transforming gene (PTTG) binding factor (PBF) (Smith et al. 2009) and leukaemia-associated RhoA guanine exchange factor (LARG) (Lacoste et al. 2012). PBF is a small glycoprotein that shares no significant homology to other human proteins but is widely conserved through a large range of species (Chien & Pei 2000). The upregulation of PBF has been reported in a number of carcinomas, including thyroid, breast and colorectal (McCabe et al. 2003, Stratford et al. 2005, Watkins et al. 2010, Read et al. 2014). PBF has been reported to repress radioiodide uptake in thyroid cancer cells by binding to NIS, which leads to NIS internalisation and inhibition of function (Smith et al. 2009). This interaction is modulated by the phosphorylation of PBF; abrogation of residue Y174 restores plasma membrane NIS and radioiodide uptake (Smith et al. 2013). PBF has also been shown to repress NIS transcription at the promoter level, reducing NIS levels and therefore radioiodide uptake (Boelaert et al. 2007). LARG is a guanine nucleotide exchange factor for the RhoA GTPase that plays a major role in the reorganisation of the cytoskeleton and cell adhesion. In multiple cancer cell lines, including breast, the interaction between LARG and NIS led to the activation of RhoA, increasing cell invasion and migration. Interaction between NIS and LARG was established to be ion-independent and occur intracellularly, suggesting that the mislocalisation of NIS in many cancers may increase migration of these cells (Lacoste et al. 2012), correlating with the observation of NIS at the leading edge of metastatic cells from breast cancer patients (Lacoste et al. 2012).
The natural ability of the thyroid to uptake iodide is central to the diagnosis and treatment of hyperthyroidism, Grave's disease and thyroid cancer. This mechanism was exploited as early as 1946 in the treatment of thyroid diseases, including thyroid carcinoma (Seidlin et al. 1946). In all, 68–80% of thyroid cancers and their metastases retain functional NIS activity and therefore the ability to accumulate iodide (Castro et al. 2001). This uptake has allowed nuclear imaging of the disease using radioiodide 123, 124 and 125 (123I, 124I and 125I) and ablation of malignant tissue using the β-emitting radioiodide-131 (131I) (Spitzweg et al. 2001). Ironically, 131I represents a significant public health hazard, implicated in open-air atomic bomb testing in the 1950s, the Chernobyl disaster in 1986 and the Fukushima explosion of 2011. 131I is a major uranium and plutonium fission product, comprising nearly 3% of the total products of fission. Its mechanism is via β decay; iodine-131 is notable for causing mutation and death in cells that it penetrates and other cells up to several millimetres away (the bystander effect). Therapeutic doses therefore differ from atomic fallout in that higher doses kill thyroid cells, whereas lower doses resulting from nuclear contamination initiate mutations, which may then drive tumourigenesis.
Along with iodide accumulation in the thyroid being vital for hormone production, it is also essential that breast-feeding mothers are able to accumulate iodide in their milk. Iodide is an essential constituent of breast milk, with infants who are deficient in iodide having increased risks of impaired neurological development and increased mortality (Cao et al. 1994, DeLong et al. 1997). This reflects the requirement for thyroid hormones in infants under 1 year of age being higher, by body weight, than at any other time in their lifespan (Delange 1998).
NIS expression in the lactating breast
The ability of humans to accumulate iodide in milk was an observation first identified in 1952 (Honour et al. 1952), with NIS being identified as the responsible transporter in 2000 (Cho et al. 2000, Tazebay et al. 2000). The NIS present in the lactating breast was observed to be functional, with distinct basolateral plasma membrane staining in alveolar cells (Cho et al. 2000). In rats, NIS identified in lactating breast was observed to be ∼75 kDa in size compared to the 100 kDa form previously observed in rat thyroids. However, the unglycosylated forms observed in both tissues were found at ∼50 kDa, suggesting differential glycosylation in thyroidal and lactating breast NIS (Tazebay et al. 2000).
The regulation of NIS expression in mammary gland tissue was found to be different from that of the thyroid with NIS, being expressed towards the end of gestation and throughout suckling; however, NIS levels are markedly reduced within 24 h of weaning (Tazebay et al. 2000). With the hormones oxytocin and prolactin being heavily associated with lactation, investigations focused on their potential involvement in the regulation of lactating mammary NIS.
As hypothesised, when mice were given doses of oxytocin, they displayed increased accumulation of radioiodide in their milk and elevated NIS expression was observed in mammary tissue (Tazebay et al. 2000). This was also confirmed in rats, with an oxytocin antagonist significantly decreasing radioiodide uptake compared to control-treated animals (Cho et al. 2000). Prolactin on the other hand, produced varying results. Treatment of mice with prolactin, alone or in combination with oxytocin, did not stimulate NIS expression and was incapable of increasing iodide accumulation in murine milk. However, in rats, treatment with prolactin increased NIS mRNA levels, and bromocriptine, which inhibits prolactin release from the pituitary gland, decreased radioiodide uptake in mammary glands (Cho et al. 2000). Both the Tazebay and Cho studies agree that oxytocin and prolactin do not act synergistically, with prolactin actually acting as an antagonist to oxytocin's stimulation of NIS. It was postulated that this was due to prolactin's inhibitory effect on steroidogenesis (Dorrington & Gore-Langton 1981), which potentially could decrease estrogen levels below a threshold preventing NIS stimulation by concomitantly administered oxytocin. This hypothesis was supported in ovariectomised mice that, when treated with high levels of exogenous estrogen, prolactin and oxytocin, displayed NIS levels above those without estrogen (Tazebay et al. 2000).
NIS expression is thus vital in lactating breasts for the accumulation of iodide, with gene expression being induced by hormones produced by the mother whilst breast-feeding, whereas ‘normal’ non-lactating breast tissue does not display detectable NIS expression.
NIS in breast cancer
The ability of breast carcinomas to uptake radioiodide was identified as early as 1974, when tumour biopsy tissues were observed to uptake more radioiodide than their normal breast tissue counterparts (Eskin et al. 1974). This early study was replicated 2 years later, with a murine study observing high levels of radioiodide uptake within mammary tumours (Thorpe 1976). However, the identification of NIS expression in mammary tumours did not occur until nearly 25 years later, when NIS mRNA was initially detected in six out of seven human breast cancer samples (Kilbane et al. 2000), with the identification of NIS protein by immunohistochemistry (IHC) following shortly after (Tazebay et al. 2000). Of the total, 87% of breast cancer tumours were reported positive for NIS expression, compared to 0% of the normal non-lactating breast tissues (Tazebay et al. 2000). Expression of NIS does not appear to fluctuate between breast cancer tumour types, with tumours such as ductal carcinoma in situ and invasive carcinoma having similar NIS levels (Tazebay et al. 2000, Wapnir et al. 2003). The expression of NIS also appeared to be independent of breast cancer receptor status and has not been correlated to expression of TSHR, ER or PR in breast cancer (Oh et al. 2005), although a more recent study did identify a correlation between ER and NIS expression (Chatterjee et al. 2013).
Despite NIS expression being identified within breast cancers, only high levels of NIS mRNA expression correlated to measurable NIS functionality, with 17% of NIS-positive tumours being capable of 99mTc-pertechnetate uptake (Moon et al. 2001). The low percentage of tumours capable of radioiodide uptake was confirmed in a later study with only two out of eight of NIS-positive tumours (25%) displaying detectable 123I− uptake (Wapnir et al. 2004). The disparity between NIS expression levels and the observed radioiodide uptake led to the hypothesis that NIS may be mislocalised within breast cancer cells. Tazebay et al. (2000) initially reported a mixture of cell surface and intracellular staining in cancerous tissue compared to solely basolateral plasma membrane staining observed in lactating breast. Strikingly, a later study identified that only 27% of NIS-positive breast cancers actually have any plasma membrane staining (Beyer et al. 2009).
To further investigate NIS expression in breast cancer, 14 genetically engineered mice models of breast cancer were investigated for NIS expression. Mice expressing the oncogene PvMT, neu, the human chorionic gonadotrophin (hCG) subunit and Cox2 in mammary glands all strongly immunostained for NIS protein. PvMT, neu and Cox2 are capable of activating the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) pathway while hCG and Cox2 can induce cAMP. An investigation into the individual pathways showed that PI3K activation led to increased expression of NIS protein and radioiodide uptake in MCF7 cells, whereas increased cAMP increased NIS promoter activity and mRNA but did not have an effect on radioiodide uptake (Knostman et al. 2004). Further investigation into the PI3K pathway suggested that although PI3K activation was capable of increasing NIS expression, it led to an underglycosylated form of NIS and interrupted cell surface trafficking of NIS. Further to this, PI3K levels were positively correlated with NIS expression levels within patient tumours (Knostman et al. 2007).
Along with primary breast tumours, the use of radioiodide for locating, monitoring and treating metastases has also been suggested. One study reported that metastatic disease has lower NIS expression, with only 33% of metastases having detectable NIS (Wapnir et al. 2004). However, an immunohistochemical study of 28 brain metastases from primary breast cancer tumours reported that 21 (75%) of the tumours were NIS-positive. The NIS-positive tumours were all ER/PR-negative with a mixture of positive and negative HER2 staining. As reported in primary breast tumour, only 24% of the metastasised tumours had plasma membrane staining for NIS, with the rest being primarily intracellular. Although only about a quarter of tumours displayed plasma membrane staining, the observations are promising, as brain metastases, due to the blood brain barrier being impermeable to many chemotherapy reagents, are poor prognostic indicators, with patients having to rely solely on surgery and/or external radiation (Renier et al. 2010).
NIS regulation in breast cancer
As previously discussed, the expression of NIS is under the regulation of TSH in the thyroid. However, this is not the case in breast cancer. It is important to understand what regulates NIS expression in breast cancer to allow potential exploitation of the mechanism for therapeutic purposes. As oxytocin and prolactin are involved in the induction of NIS in lactating mammary glands it was logical to investigate whether they have a role in the expression of NIS in breast cancer. In 3D histocultures of breast cancer tumours, individual treatment with oxytocin and prolactin induced NIS mRNA in a dose-dependent manner. However, a combination of the two hormones was not capable of inducing further NIS expression (Cho et al. 2000). Prolactin increases the expression of genes containing gamma-interferon activation sequences (GAS) in their promoter regions, through the activation of the Jak2/Stat5 cascade in breast cells (Burdon et al. 1994) (Fig. 2). A GAS sequence has subsequently been identified in the promoter of human NIS (Cho et al. 2000). There are conflicting reports as to whether prolactin can increase NIS expression in MCF7s, with Kogai et al. (2000) not identifying any stimulation compared to Arturi et al. (2005) who observed increased NIS mRNA and iodide-trapping after prolactin treatment. This discrepancy could be due to a variety of technical considerations (Arturi et al. 2005). The oxytocin receptor is present in 50–90% of breast cancers (Bussolati et al. 1996, Ito et al. 1996, Sapino et al. 1998) and is a G protein-coupled receptor capable of activating the Gs-cAMP-protein kinase A (PKA) pathway (Olins & Bremel 1984) (Fig. 2). This is the same pathway activated by TSH stimulation in thyrocytes.
Another factor identified to be involved in NIS induction in breast cancers is insulin, along with insulin-like growth factor 1 (IGF1) and IGF2. Insulin and IGF1/2 stimulated NIS expression at both mRNA and protein levels, and increased 125I uptake (Arturi et al. 2005). Binding of a ligand to the IGF1 receptor is known to act on insulin receptor substrate 1 (IRS1), which leads to the activation of PI3K (Dupont & Le Roith 2001) (Fig. 2), a pathway associated with NIS expression in breast cells.
Retinoids have been described to be useful in a treatment of a variety of cancers (reviewed in Connolly et al. (2013)) with systemic retinoids being FDA-approved for treatment of cutaneous T cell lymphoma (Duvic et al. 2001) and acute promyelocytic leukaemia (APL) (Tallman et al. 1997). Kogai et al. hypothesised that retinoic acid (RA) may be capable of inducing NIS expression in MCF7 cells, a cell line that expresses both RA receptors (RARs) and retinoid X receptors (RXRs). In MCF7s, all trans-RA (ATRA) increased 125I uptake in a dose dependant manner that could be inhibited by potassium perchlorate suggesting that uptake was mediated specifically via NIS. Treatment with ATRA increased NIS expression at both the mRNA and protein level, with subsequent 131I treatment reducing colony formation in MCF7 cells. MCF12A cells (derived from normal breast tissue) and ER-negative MDA-MB231 cells did not show such a marked response to ATRA treatment, with NIS protein expression being only slightly stimulated in MDA-MB231s and treatment having no effect on iodide uptake in either cell line (Kogai et al. 2000). This disparity between cell lines is most likely due to MDA-MB231 cells being RA ‘resistant’ due to decreased expression of RARα and RARβ compared to MCF7 cells (Liu et al. 1996). Subsequently, mice bearing MCF7 xenograft tumours were utilised to establish whether systemic retinoids were capable of the same NIS induction witnessed in vitro. Tumour-bearing mice treated with a time release pellet containing ATRA for 5 days prior to 125I treatment accumulated 15 times more radioiodide than control treated mice and displayed increased NIS expression. ATRA was also capable of increasing iodide uptake approximately twofold in a transgenic breast cancer mouse model (murine mammary tumour virus-polyoma virus middle T antigen (MMTV-PyVT)-transgenic mice) (Kogai et al. 2004). Further studies established that RARβ/γ stimulation by retinoids led to the most pronounced increase in radioiodide uptake, whereas stimulation of RARα, RARγ and RXR only lead to a modest increase in uptake. Dexamethasone (Dex) was discovered to synergistically increase NIS expression and radioiodide uptake alongside ATRA in MCF7 cells. Dex treatment alone increased NIS mRNA levels slightly but when used in conjunction with ATRA, NIS mRNA increased over 70-fold, with Dex increasing the stability of NIS mRNA (Kogai et al. 2005). These findings were later confirmed with MCF7 cells treated with ATRA and Dex in combination having significantly decreased cell survival after 131I treatment compared to ATRA alone treated cells (Unterholzner et al. 2006).
ATRA and other retinoids are capable of binding to RARs that heterodimerise with RXR. The heterodimer then binds to RA response elements (RAREs) on target genes and stimulates transcription. Investigation into the NIS promoter in MCF7 cells revealed there were no putative full RAREs. However, several DR2 element sequences with typical half-sites were identified between the first and thirteenth introns of NIS (Kogai et al. 2008). These DR2 sites were unresponsive to ATRA, although a DR5 element located in the promoter region of NIS was responsive to ATRA in MCF7 cells (Kogai et al. 2008) (Fig. 2). Binding of RARα to the intronic DR2 elements was later reported within 30 min of treatment of MCF7 cells with ATRA (Alotaibi et al. 2010), suggesting these DR2 elements have a potential role in the induction of NIS (Kogai & Brent 2012). However, the introns of murine NIS do not contain DR2 elements yet are still capable of ATRA induced NIS expression in transgenic mice models (Kogai et al. 2004), suggesting other non-traditional mechanisms of induction may be apparent.
One potential mechanism is through homeobox protein Nkx2.5, which is induced following ATRA treatment. Use of Nkx2.5 mutants suggested that Nkx2.5 is critical for ATRA's induction of NIS and radioiodide uptake (Dentice et al. 2004) (Fig. 2). A range of inhibitors was utilised to try to identify which pathways are critical for ATRA mediated induction of NIS expression and identified that the activities of IGF receptor, PI3K, JAK, and p38 MAPK were important (Kogai et al. 2008). It is established that natural retinoids are capable of stimulating the IGF receptors (Kang et al. 1997) and that activated RAR/RXR heterodimers can interact with IRS1 (del Rincon et al. 2003). It was hypothesised that RAR, through its interaction with IRS1, can activate the PI3K pathway leading to NIS expression (Kogai et al. 2008). Activated RAR/RXR heterodimers have also been shown to directly activate PI3K with p85, a sub-unit of PI3K, observed to bind the heterodimers (Fig. 2). Silencing/inhibition of p85 led to a reduced NIS expression after ATRA treatment (Ohashi et al. 2009) suggesting that the PI3K pathway plays an important role in ATRA mediated NIS induction.
The p38 MAPK pathway was also associated with ATRA's induction of NIS (Kogai et al. 2008) and ATRA stimulated RAR/RXR heterodimers have been observed to activate the p38-Rac1 pathway (Alsayed et al. 2001). Inhibition of p38 was noted to reduce NIS at both the basal level and with ATRA treatment (Kogai et al. 2008) and the use of inhibitors and siRNA demonstrated that Rac1, MAPK kinase 3B (MAPKK3B) and p38β were all required for the full induction of NIS by ATRA in MCF7 cells (Kogai et al. 2012). It is hypothesised that ATRA stimulates dimerisation of RAR/RXR, which interacts with the small GTPase Rac1, leading to MAPKK3B phosphorylating p38. The phosphorylation of p38 leads to the induction of NIS transcription through unknown factors in breast cells (Kogai et al. 2012) (Fig. 2).
Although the use of prolactin, oxytocin, insulin and retinoids has been identified to induce NIS expression within breast cancer tissues, the mechanisms are not fully elucidated and the complications of these systemic treatments are not fully overcome. Thus, efforts have been made to try to enhance NIS levels and radioiodide uptake by exogenously expressing NIS.
NIS gene therapy in breast cancer
Along with the induction of endogenous NIS, another method of increasing NIS expression in breast cancer cells is via exogenous expression. Retroviral expression of NIS in murine and human tumour cells induced uptake of radioiodide in xenograft models, with in vitro uptake of 131I being sufficient to kill cells (Mandell et al. 1999). To identify whether NIS could be exogenously targeted to pre-existing tumours, xenograft SiHa and MCF7 tumours were established in murine models before NIS adenoviral plaque-forming units (PFU) were injected directly into the tumour. The injected tumours were observed to be capable of 25-fold higher radioiodide uptake than their control counterparts. However, after treatment with 131I, there was no difference in tumour size between control and NIS-transfected tumours, suggesting that the tumour retention time of radioiodide was not sufficient to inhibit cell growth or ablate tumour cells (Boland et al. 2000).
To further increase NIS expression in tissues, NIS can be exogenously expressed within cells under promoters specific to certain cell types. LNCaP prostate cancer cells were stably transfected with NIS under the control of the prostate specific antigen (PSA) promoter. Xenografts of these cells were transplanted into mice and treated with a single i.p. injection containing 3 mCi 131I. Mice with tumours expressing NIS had a significantly reduced tumour size compared to control tumours (Spitzweg et al. 2000). Subsequently, breast tumours were investigated using NIS under control of the MUC1 promoter. MUC1 is a glycoprotein usually observed in haematopoietic cells, although expression is observed in ∼90% of breast cancers and has been correlated with poor survival and increased metastasis (Gendler 2001). MUC1-positive T47D cells displayed a 58-fold increase in radioiodide uptake after adenoviral transduction with NIS compared to control transfected cells, whereas MUC1-negative MDA-MB231 cells showed no increase in radioiodide uptake after transduction. In vivo studies with T47D xenograft tumours established in mice were injected at multiple sites with adenoviral NIS and treated with an i.p. injection of 3 mCi of 131I. NIS expressing tumours were reduced in size by ∼83% whilst control tumours continued to increase in size (Dwyer et al. 2005). Use of adenoviral expression of NIS under the control of an estrogen receptor responsive promoter (4ERE) was also successful in increasing radioiodide uptake and inhibited tumour growth after 131I treatment in ER-positive breast cancer cells.
It has previously been reported that one of the limitations of gene therapy in a preclinical and clinical setting is the difficulty in detecting gene expression following administration. For clinical settings in particular, a non-invasive and reproducible detection method is required to establish location, magnitude and kinetics of gene expression (Tjuvajev et al. 1995). In this context, NIS is advantageous as it can be easily and non-invasively detected using gamma scintigraphy with 123I or 99TcO4− or positron emission tomography (PET) with 124I and 76Br to image where it has successfully been expressed. Studies have also been conducted with the use of NIS as a reporter gene for the transfection of tumour cells with p53 and manganese superoxide dismutase (MnSOD) (Niu et al. 2006).
Use of RA was also capable of increasing adenoviral NIS expression when NIS was under control of the CMV promoter in MCF7 cells (Lim et al. 2007). The CMV promoter contains RAREs (Angulo et al. 1996), which can be bound by stimulated nuclear RARs endogenously present in MCF7 cells (Titcomb et al. 1994). The use of the human telomerase reverse transcriptase promoter also proved effective for increasing iodide uptake and ablation in breast cancer cells (Riesco-Eizaguirre et al. 2011).
Along with the use of retro- and adenoviruses, oncolytic viruses have been used as a form of delivery system for exogenous targeting of proteins. GLV-h153, an oncolytic vaccinia virus carrying hNIS, was injected into mice xenograft tumours, leading to increased visualisation of the tumours with 124I. Subsequent treatment with 131I also suppressed tumour growth compared to control (Gholami et al. 2014).
Although NIS gene therapy appears effective, an efficient and optimal method of delivery of the gene to the tumour site has not been established. Use of superparamagnetic iron oxide (SPIO)-labelled AC133+ progenitor cells (APCs) transduced with adenoviral hNIS in mice with MDA-MB231 xenograft tumours increased 99mTm activity at the site of the tumour. The labelled APCs were administered to the mice intravenously and images taken using MRI and single photon emission computer tomography (SPECT) showed accumulation of the labelled cells at the site of the tumour along with NIS expression (Rad et al. 2009). The use of mesenchymal stem cells (MSCs), which have been shown to migrate specifically to tumours in vivo, has been suggested for gene therapy (Spaeth et al. 2008, Dwyer et al. 2010). MSCs adenovirally infected with NIS were intravenously injected into mice bearing MDA-MB231 xenograft tumours. Three days after i.v. injection of the MSCs, uptake of 99mTc was detected at the site of the tumour using SPECT and remained detectable 14 days after injection. NIS expression was detected in tumour tissue, and at day 14 mice treated with 131I had a significant reduction in breast xenograft tumour size compared to control mice (Dwyer et al. 2011).
Pendrin
Pendrin, coded by the solute carrier family 26 member 4 gene, (SLC26A4) PDS, is a sodium-independent chloride/iodide transporter (Everett et al. 1997) and is found to be abundantly expressed in the thyroid and the inner ear. Like NIS, pendrin is a large (780 amino acids) transmembrane protein that spans the plasma membrane multiple times (Everett et al. 1997). Initial studies found that in the thyroid, pendrin was capable of mediating the transport of iodide and chloride (Scott et al. 1999), with pendrin being located on the apical membrane of thyrocytes (Bidart et al. 2000) mediating iodide efflux (Yoshida et al. 2002). Supporting these in vitro data, patients suffering Pendred syndrome, a disease where the gene-encoding pendrin (PDS) contains an inactivating mutation, display altered iodine organification (Sheffield et al. 1996). However, more recently there have been questions as to pendrin's role in iodide transport as patients with biallelic PDS mutations, with sufficient iodide intake, displayed no altered thyroidal phenotype (Sato et al. 2001) and pendrin knockout mice did not develop goitres and had normal thyroid function tests (Everett et al. 2001).
In lactating mammary glands, an iodide/anion exchanger that was sensitive to 4,4′-diisothiocyano-2,2′-stilbenedisulfonic acid (DIDS) was discovered (Shennan 2001) and was later identified as pendrin (Rillema & Hill 2003a). Akin to NIS, there is little to no expression of pendrin in normal mammary glands, but it is induced in lactating breast. Pendrin expression was shown to be stimulated by prolactin, increasing levels of iodide uptake, which were returned to normal levels in the presence of several pendrin inhibitors, including DIDS. Although the data were not shown, Rillema and Hill claimed lactating tissue provided the highest expression of pendrin in immunohistochemical studies, with the staining being specific to alveolar epithelial cells with none detected in stromal cells. However, it was then stated that there was no specific localisation of pendrin to the apical or basolateral surface of the alveolar epithelial cells, and so no comment could be made on the localisation of functional pendrin that was observed (Rillema & Hill 2003a).
Following identification of pendrin in the lactating breast, subsequent studies were performed to identify potential pendrin expression in breast cancer. Pendrin expression was confirmed in MCF7 cells by western blotting. Radioiodide uptake in MCF7 cells was shown to be decreased in cells treated with DIDS but not sodium perchlorate or ouabain. This decrease was shown to be sodium-independent, suggesting that pendrin was responsible for iodide uptake, not NIS as previously observed. From these findings the authors went on to suggest that differences between batches of MCF7 cells or culturing conditions could potentially result in variable expression of iodide transporters (Rillema & Hill 2003b). A later study also found that in the mammary glands of rats treated with the carcinogen, N-methyl-N-nitrosourea (MNU), pendrin expression was increased when cells were treated with iodine (I2) (Garcia-Solis et al. 2005).
Although pendrin expression has been reported in MCF7s and rat breast tumours, further work is required to elucidate whether there is widespread pendrin expression within breast cancer. Currently there is little evidence to suggest that pendrin is a major player in breast cancer, let alone iodide transport within the disease.
Sodium-coupled monocarboxylate transporter
The SMCT, also referred to as apical iodide transporter (AIT), is encoded by the SLC5A8 gene and is usually located on the apical membrane of thyrocytes, transporting iodide from thyrocytes to colloid lumen (Rodriguez et al. 2002). SMCT is a ∼69 kDa protein that shares 46% homology with NIS and is downregulated in many carcinomas, including thyroid cancer (Lacroix et al. 2004, Porra et al. 2005), and has thus been suggested to be a tumour suppressor gene (Porra et al. 2005). SMCT is abundantly expressed in the colon (Li et al. 2003) and functions as a Na+/short-chain fatty acid co-transporter (Miyauchi et al. 2004). High expression levels of SMCT have been reported during mammary gland involution and in normal breast epithelial cells (Gupta et al. 2006). However, SMCT is significantly downregulated in breast cancer cell lines and in primary breast tumours, with MCF7 cells reported to have no expression (Thangaraju et al. 2006).
Due to its lack of expression in breast cancer, this transporter plays no reported role in the iodide transport of breast cancer cells. Even though SMCT is hailed as a tumour suppressor, given its role in iodide efflux within the thyroid, expression of SMCT within breast cancer could be counterintuitive for the proposed radioiodide therapy. Although studies have reported a lack of expression of SMCT in breast cancer, it may be important to further investigate the role of SMCT in iodide transport within normal breast tissue. Observations from these studies could be important in identifying changes in iodide transport in breast cells progressing to cancer, where SMCT expression is switched off.
Conclusion
Understanding the roles of the three known iodide transporters within breast cancer is of the utmost important for translating the proposed use of radioiodide therapy from the bench to the bedside. From the evidence discussed, the major iodide transporter in breast cancer is NIS, with pendrin potentially having a more minor role. Although NIS is expressed in the majority of breast cancers and metastases, the inhibitory issue for radioiodide therapy is the intracellular localisation of NIS, rendering it non-functional. There is an obvious need for new studies to discern mechanisms of increasing functional NIS localisation. The two prospective ways of doing this are by increasing total NIS levels or by manipulating cells to alter the trafficking of NIS.
The use of retinoids to increase endogenous NIS expression has proved promising in vitro, but concerns over the systemic effects associated with retinoid use need to be carefully considered. While exogenous expression of NIS in cells has proved successful in vivo, delivery methods for gene targeting are not fully optimised and further investigation and trials are required before this is a viable solution. Although increasing total NIS levels is often championed, there are potential problems that could be associated with this method of increasing iodide transport. An increase of total NIS may lead to a serendipitous increase of plasma membrane NIS, but it also may be associated with a rise in intracellular NIS levels, which has previously been correlated to increased migration and invasiveness through its interaction with LARG (Lacoste et al. 2012). Alternative studies into manipulating the subcellular location of NIS, using established and potentially novel binding partners should also be heavily considered. PBF has been observed to decrease plasma membrane expression of NIS in thyroid cancer (Smith et al. 2009), so a similar mechanism may exist within breast cancer, with the potential for inhibition of phosphorylation of PBF being capable of restoring plasma membrane expression.
Overall, the potential for the use of radioiodide treatment in breast cancer is one that should be continued to be investigated. The regulation and subcellular location of NIS appears to be unique in breast cells, so further investigations within normal, lactating and malignant tissues are required. Although NIS appears to be the protein responsible for the majority of iodide transport within breast cancer, SMCT and pendrin should not be overlooked and their roles within the disease should continue to be elucidated.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.
Funding
This review did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.
References
Alotaibi H, Yaman E, Salvatore D, Di Dato V, Telkoparan P, Di Lauro R & Tazebay UH 2010 Intronic elements in the Na+/I− symporter gene (NIS) interact with retinoic acid receptors and mediate initiation of transcription. Nucleic Acids Research 38 3172–3185. (doi:10.1093/nar/gkq023)
Alsayed Y, Uddin S, Mahmud N, Lekmine F, Kalvakolanu DV, Minucci S, Bokoch G & Platanias LC 2001 Activation of Rac1 and the p38 mitogen-activated protein kinase pathway in response to all-trans-retinoic acid. Journal of Biological Chemistry 276 4012–4019. (doi:10.1074/jbc.M007431200)
Angulo A, Suto C, Heyman RA & Ghazal P 1996 Characterization of the sequences of the human cytomegalovirus enhancer that mediate differential regulation by natural and synthetic retinoids. Molecular Endocrinology 10 781–793. (doi:10.1210/mend.10.7.8813719)
Arturi F, Ferretti E, Presta I, Mattei T, Scipioni A, Scarpelli D, Bruno R, Lacroix L, Tosi E & Gulino A et al. 2005 Regulation of iodide uptake and sodium/iodide symporter expression in the MCF-7 human breast cancer cell line. Journal of Clinical Endocrinology and Metabolism 90 2321–2326. (doi:10.1210/jc.2004-1562)
Baumann E 1896 Uber das Thyrojodin. Münchener Medizinische Wochenschrift 43 4.
Beyer SJ, Jimenez RE, Shapiro CL, Cho JY & Jhiang SM 2009 Do cell surface trafficking impairments account for variable cell surface sodium iodide symporter levels in breast cancer? Breast Cancer Research and Treatment 115 205–212. (doi:10.1007/s10549-008-0059-5)
Bidart JM, Mian C, Lazar V, Russo D, Filetti S, Caillou B & Schlumberger M 2000 Expression of pendrin and the Pendred syndrome (PDS) gene in human thyroid tissues. Journal of Clinical Endocrinology and Metabolism 85 2028–2033. (doi:10.1210/jcem.85.5.6519)
Boelaert K, Smith VE, Stratford AL, Kogai T, Tannahill LA, Watkinson JC, Eggo MC, Franklyn JA & McCabe CJ 2007 PTTG and PBF repress the human sodium iodide symporter. Oncogene 26 4344–4356. (doi:10.1038/sj.onc.1210221)
Boland A, Ricard M, Opolon P, Bidart JM, Yeh P, Filetti S, Schlumberger M & Perricaudet M 2000 Adenovirus-mediated transfer of the thyroid sodium/iodide symporter gene into tumors for a targeted radiotherapy. Cancer Research 60 3484–3492.
Burdon TG, Demmer J, Clark AJ & Watson CJ 1994 The mammary factor MPBF is a prolactin-induced transcriptional regulator which binds to STAT factor recognition sites. FEBS Letters 350 177–182. (doi:10.1016/0014-5793(94)00757-8)
Bussolati G, Cassoni P, Ghisolfi G, Negro F & Sapino A 1996 Immunolocalization and gene expression of oxytocin receptors in carcinomas and non-neoplastic tissues of the breast. American Journal of Pathology 148 1895–1903.
Cao XY, Jiang XM, Dou ZH, Rakeman MA, Zhang ML, O'Donnell K, Ma T, Amette K, DeLong N & DeLong GR 1994 Timing of vulnerability of the brain to iodine deficiency in endemic cretinism. New England Journal of Medicine 331 1739–1744. (doi:10.1056/NEJM199412293312603)
Castro MR, Bergert ER, Goellner JR, Hay ID & Morris JC 2001 Immunohistochemical analysis of sodium iodide symporter expression in metastatic differentiated thyroid cancer: correlation with radioiodine uptake. Journal of Clinical Endocrinology and Metabolism 86 5627–5632. (doi:10.1210/jcem.86.11.8048)
Chatterjee S, Malhotra R, Varghese F, Bukhari AB, Patil A, Budrukkar A, Parmar V, Gupta S & De A 2013 Quantitative immunohistochemical analysis reveals association between sodium iodide symporter and estrogen receptor expression in breast cancer. PLoS ONE 8 e54055. (doi:10.1371/journal.pone.0054055)
Chien W & Pei L 2000 A novel binding factor facilitates nuclear translocation and transcriptional activation function of the pituitary tumor-transforming gene product. Journal of Biological Chemistry 275 19422–19427. (doi:10.1074/jbc.M910105199)
Cho JY, Leveille R, Kao R, Rousset B, Parlow AF, Burak WE Jr, Mazzaferri EL & Jhiang SM 2000 Hormonal regulation of radioiodide uptake activity and Na+/I− symporter expression in mammary glands. Journal of Clinical Endocrinology and Metabolism 85 2936–2943. (doi:10.1210/jcem.85.8.6727)
Connolly RM, Nguyen NK & Sukumar S 2013 Molecular pathways: current role and future directions of the retinoic acid pathway in cancer prevention and treatment. Clinical Cancer Research 19 1651–1659. (doi:10.1158/1078-0432.CCR-12-3175)
Dai G, Levy O & Carrasco N 1996 Cloning and characterization of the thyroid iodide transporter. Nature 379 458–460. (doi:10.1038/379458a0)
Delange F 1998 Screening for congenital hypothyroidism used as an indicator of the degree of iodine deficiency and of its control. Thyroid 8 1185–1192. (doi:10.1089/thy.1998.8.1185)
DeLong GR, Leslie PW, Wang SH, Jiang XM, Zhang ML, Rakeman M, Jiang JY, Ma T & Cao XY 1997 Effect on infant mortality of iodination of irrigation water in a severely iodine-deficient area of China. Lancet 350 771–773. (doi:10.1016/S0140-6736(96)12365-5)
Dentice M, Luongo C, Elefante A, Romino R, Ambrosio R, Vitale M, Rossi G, Fenzi G & Salvatore D 2004 Transcription factor Nkx-2.5 induces sodium/iodide symporter gene expression and participates in retinoic acid- and lactation-induced transcription in mammary cells. Molecular and Cellular Biology 24 7863–7877. (doi:10.1128/MCB.24.18.7863-7877.2004)
Dorrington J & Gore-Langton RE 1981 Prolactin inhibits oestrogen synthesis in the ovary. Nature 290 600–602. (doi:10.1038/290600a0)
Dupont J & Le Roith D 2001 Insulin-like growth factor 1 and oestradiol promote cell proliferation of MCF-7 breast cancer cells: new insights into their synergistic effects. Molecular Pathology 54 149–154. (doi:10.1136/mp.54.3.149)
Duvic M, Hymes K, Heald P, Breneman D, Martin AG, Myskowski P, Crowley C, Yocum RC & Bexarotene Worldwide Study Group 2001 Bexarotene is effective and safe for treatment of refractory advanced-stage cutaneous T-cell lymphoma: multinational phase II–III trial results. Journal of Clinical Oncology 19 2456–2471.
Dwyer RM, Bergert ER, O'Connor MK, Gendler SJ & Morris JC 2005 In vivo radioiodide imaging and treatment of breast cancer xenografts after MUC1-driven expression of the sodium iodide symporter. Clinical Cancer Research 11 1483–1489. (doi:10.1158/1078-0432.CCR-04-1636)
Dwyer RM, Khan S, Barry FP, O'Brien T & Kerin MJ 2010 Advances in mesenchymal stem cell-mediated gene therapy for cancer. Stem Cell Research & Therapy 1 25. (doi:10.1186/scrt25)
Dwyer RM, Ryan J, Havelin RJ, Morris JC, Miller BW, Liu Z, Flavin R, O'Flatharta C, Foley MJ & Barrett HH et al. 2011 Mesenchymal Stem Cell-mediated delivery of the sodium iodide symporter supports radionuclide imaging and treatment of breast cancer. Stem Cells 29 1149–1157. (doi:10.1002/stem.665)
Eskin BA, Parker JA, Bassett JG & George DL 1974 Human breast uptake of radioactive iodine. Obstetrics and Gynecology 44 398–402.
Everett LA, Glaser B, Beck JC, Idol JR, Buchs A, Heyman M, Adawi F, Hazani E, Nassir E & Baxevanis AD et al. 1997 Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nature Genetics 17 411–422. (doi:10.1038/ng1297-411)
Everett LA, Belyantseva IA, Noben-Trauth K, Cantos R, Chen A, Thakkar SI, Hoogstraten-Miller SL, Kachar B, Wu DK & Green ED 2001 Targeted disruption of mouse PDS provides insight about the inner-ear defects encountered in Pendred syndrome. Human Molecular Genetics 10 153–161. (doi:10.1093/hmg/10.2.153)
Garcia-Solis P, Alfaro Y, Anguiano B, Delgado G, Guzman RC, Nandi S, Diaz-Munoz M, Vazquez-Martinez O & Aceves C 2005 Inhibition of N-methyl-N-nitrosourea-induced mammary carcinogenesis by molecular iodine (I2) but not by iodide (I−) treatment evidence that I2 prevents cancer promotion. Molecular and Cellular Endocrinology 236 49–57. (doi:10.1016/j.mce.2005.03.001)
Gendler SJ 2001 MUC1, the renaissance molecule. Journal of Mammary Gland Biology and Neoplasia 6 339–353. (doi:10.1023/A:1011379725811)
Gholami S, Chen CH, Lou E, Belin LJ, Fujisawa S, Longo VA, Chen NG, Gonen M, Zanzonico PB & Szalay AA et al. 2014 Vaccinia virus GLV-1h153 in combination with 131I shows increased efficiency in treating triple-negative breast cancer. FASEB Journal 28 676–682. (doi:10.1096/fj.13-237222)
Gupta N, Martin PM, Prasad PD & Ganapathy V 2006 SLC5A8 (SMCT1)-mediated transport of butyrate forms the basis for the tumor suppressive function of the transporter. Life Sciences 78 2419–2425. (doi:10.1016/j.lfs.2005.10.028)
Honour AJ, Myant NB & Rowlands EN 1952 Secretion of radioiodine in digestive juices and milk in man. Clinical Science 11 449–462.
Hortobagyi GN, de la Garza Salazar J, Pritchard K, Amadori D, Haidinger R, Hudis CA, Khaled H, Liu MC, Martin M & Namer M et al. 2005 The global breast cancer burden: variations in epidemiology and survival. Clinical Breast Cancer 6 391–401. (doi:10.3816/CBC.2005.n.043)
Ito Y, Kobayashi T, Kimura T, Matsuura N, Wakasugi E, Takeda T, Shimano T, Kubota Y, Nobunaga T & Makino Y et al. 1996 Investigation of the oxytocin receptor expression in human breast cancer tissue using newly established monoclonal antibodies. Endocrinology 137 773–779. (doi:10.1210/endo.137.2.8593829)
Kang JX, Li Y & Leaf A 1997 Mannose-6-phosphate/insulin-like growth factor-II receptor is a receptor for retinoic acid. PNAS 94 13671–13676. (doi:10.1073/pnas.94.25.13671)
Kilbane MT, Ajjan RA, Weetman AP, Dwyer R, McDermott EW, O'Higgins NJ & Smyth PP 2000 Tissue iodine content and serum-mediated 125I uptake-blocking activity in breast cancer. Journal of Clinical Endocrinology and Metabolism 85 1245–1250. (doi:10.1210/jcem.85.3.6442)
Knostman KA, Cho JY, Ryu KY, Lin X, McCubrey JA, Hla T, Liu CH, Di Carlo E, Keri R & Zhang M et al. 2004 Signaling through 3′,5′-cyclic adenosine monophosphate and phosphoinositide-3 kinase induces sodium/iodide symporter expression in breast cancer. Journal of Clinical Endocrinology and Metabolism 89 5196–5203. (doi:10.1210/jc.2004-0907)
Knostman KA, McCubrey JA, Morrison CD, Zhang Z, Capen CC & Jhiang SM 2007 PI3K activation is associated with intracellular sodium/iodide symporter protein expression in breast cancer. BMC Cancer 7 137. (doi:10.1186/1471-2407-7-137)
Kogai T & Brent GA 2012 The sodium iodide symporter (NIS): regulation and approaches to targeting for cancer therapeutics. Pharmacology & Therapeutics 135 355–370. (doi:10.1016/j.pharmthera.2012.06.007)
Kogai T, Endo T, Saito T, Miyazaki A, Kawaguchi A & Onaya T 1997 Regulation by thyroid-stimulating hormone of sodium/iodide symporter gene expression and protein levels in FRTL-5 cells. Endocrinology 138 2227–2232. (doi:10.1210/endo.138.6.5189)
Kogai T, Schultz JJ, Johnson LS, Huang M & Brent GA 2000 Retinoic acid induces sodium/iodide symporter gene expression and radioiodide uptake in the MCF-7 breast cancer cell line. PNAS 97 8519–8524. (doi:10.1073/pnas.140217197)
Kogai T, Kanamoto Y, Che LH, Taki K, Moatamed F, Schultz JJ & Brent GA 2004 Systemic retinoic acid treatment induces sodium/iodide symporter expression and radioiodide uptake in mouse breast cancer models. Cancer Research 64 415–422. (doi:10.1158/0008-5472.CAN-03-2285)
Kogai T, Kanamoto Y, Li AI, Che LH, Ohashi E, Taki K, Chandraratna RA, Saito T & Brent GA 2005 Differential regulation of sodium/iodide symporter gene expression by nuclear receptor ligands in MCF-7 breast cancer cells. Endocrinology 146 3059–3069. (doi:10.1210/en.2004-1334)
Kogai T, Ohashi E, Jacobs MS, Sajid-Crockett S, Fisher ML, Kanamoto Y & Brent GA 2008 Retinoic acid stimulation of the sodium/iodide symporter in MCF-7 breast cancer cells is mediated by the insulin growth factor-I/phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase signaling pathways. Journal of Clinical Endocrinology and Metabolism 93 1884–1892. (doi:10.1210/jc.2007-1627)
Kogai T, Liu YY, Mody K, Shamsian DV & Brent GA 2012 Regulation of sodium iodide symporter gene expression by Rac1/p38β mitogen-activated protein kinase signaling pathway in MCF-7 breast cancer cells. Journal of Biological Chemistry 287 3292–3300. (doi:10.1074/jbc.M111.315523)
Lacoste C, Herve J, Bou Nader M, Dos Santos A, Moniaux N, Valogne Y, Montjean R, Dorseuil O, Samuel D & Cassio D et al. 2012 Iodide transporter NIS regulates cancer cell motility and invasiveness by interacting with the Rho guanine nucleotide exchange factor LARG. Cancer Research 72 5505–5515. (doi:10.1158/0008-5472.CAN-12-0516)
Lacroix L, Pourcher T, Magnon C, Bellon N, Talbot M, Intaraphairot T, Caillou B, Schlumberger M & Bidart JM 2004 Expression of the apical iodide transporter in human thyroid tissues: a comparison study with other iodide transporters. Journal of Clinical Endocrinology and Metabolism 89 1423–1428. (doi:10.1210/jc.2003-030542)
Levy O, De la Vieja A, Ginter CS, Riedel C, Dai G & Carrasco N 1998 N-linked glycosylation of the thyroid Na+/I− symporter (NIS). Implications for its secondary structure model. Journal of Biological Chemistry 273 22657–22663. (doi:10.1074/jbc.273.35.22657)
Li H, Myeroff L, Smiraglia D, Romero MF, Pretlow TP, Kasturi L, Lutterbaugh J, Rerko RM, Casey G & Issa JP et al. 2003 SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers. PNAS 100 8412–8417. (doi:10.1073/pnas.1430846100)
Lim SJ, Paeng JC, Kim SJ, Kim SY, Lee H & Moon DH 2007 Enhanced expression of adenovirus-mediated sodium iodide symporter gene in MCF-7 breast cancer cells with retinoic acid treatment. Journal of Nuclear Medicine 48 398–404. (doi:10.1.1.498.8174)
Liu Y, Lee MO, Wang HG, Li Y, Hashimoto Y, Klaus M, Reed JC & Zhang X 1996 Retinoic acid receptor β mediates the growth-inhibitory effect of retinoic acid by promoting apoptosis in human breast cancer cells. Molecular and Cellular Biology 16 1138–1149.
Mandell RB, Mandell LZ & Link CJ Jr 1999 Radioisotope concentrator gene therapy using the sodium/iodide symporter gene. Cancer Research 59 661–668.
McCabe CJ, Khaira JS, Boelaert K, Heaney AP, Tannahill LA, Hussain S, Mitchell R, Olliff J, Sheppard MC & Franklyn JA et al. 2003 Expression of pituitary tumour transforming gene (PTTG) and fibroblast growth factor-2 (FGF-2) in human pituitary adenomas: relationships to clinical tumour behaviour. Clinical Endocrinology 58 141–150. (doi:10.1046/j.1365-2265.2003.01598.x)
Miyauchi S, Gopal E, Fei YJ & Ganapathy V 2004 Functional identification of SLC5A8, a tumor suppressor down-regulated in colon cancer, as a Na(+)-coupled transporter for short-chain fatty acids. Journal of Biological Chemistry 279 13293–13296. (doi:10.1074/jbc.C400059200)
Moon DH, Lee SJ, Park KY, Park KK, Ahn SH, Pai MS, Chang H, Lee HK & Ahn IM 2001 Correlation between 99mTc-pertechnetate uptakes and expressions of human sodium iodide symporter gene in breast tumor tissues. Nuclear Medicine and Biology 28 829–834. (doi:10.1016/S0969-8051(01)00243-8)
Niu G, Anderson RD, Madsen MT, Graham MM, Oberley LW & Domann FE 2006 Dual-expressing adenoviral vectors encoding the sodium iodide symporter for use in noninvasive radiological imaging of therapeutic gene transfer. Nuclear Medicine and Biology 33 391–398. (doi:10.1016/j.nucmedbio.2006.01.003)
Oh HJ, Chung JK, Kang JH, Kang WJ, Noh DY, Park IA, Jeong JM, Lee DS & Lee MC 2005 The relationship between expression of the sodium/iodide symporter gene and the status of hormonal receptors in human breast cancer tissue. Cancer Research and Treatment 37 247–250. (doi:10.4143/crt.2005.37.4.247)
Ohashi E, Kogai T, Kagechika H & Brent GA 2009 Activation of the PI3 kinase pathway by retinoic acid mediates sodium/iodide symporter induction and iodide transport in MCF-7 breast cancer cells. Cancer Research 69 3443–3450. (doi:10.1158/0008-5472.CAN-08-3234)
Ohno M, Zannini M, Levy O, Carrasco N & di Lauro R 1999 The paired-domain transcription factor Pax8 binds to the upstream enhancer of the rat sodium/iodide symporter gene and participates in both thyroid-specific and cyclic-AMP-dependent transcription. Molecular and Cellular Biology 19 2051–2060.
Olins GM & Bremel RD 1984 Oxytocin-stimulated myosin phosphorylation in mammary myoepithelial cells: roles of calcium ions and cyclic nucleotides. Endocrinology 114 1617–1626. (doi:10.1210/endo-114-5-1617)
Poleev A, Okladnova O, Musti AM, Schneider S, Royer-Pokora B & Plachov D 1997 Determination of functional domains of the human transcription factor PAX8 responsible for its nuclear localization and transactivating potential. European Journal of Biochemistry/FEBS 247 860–869. (doi:10.1111/j.1432-1033.1997.00860.x)
Porra V, Ferraro-Peyret C, Durand C, Selmi-Ruby S, Giroud H, Berger-Dutrieux N, Decaussin M, Peix JL, Bournaud C & Orgiazzi J et al. 2005 Silencing of the tumor suppressor gene SLC5A8 is associated with BRAF mutations in classical papillary thyroid carcinomas. Journal of Clinical Endocrinology and Metabolism 90 3028–3035. (doi:10.1210/jc.2004-1394)
Rad AM, Iskander AS, Janic B, Knight RA, Arbab AS & Soltanian-Zadeh H 2009 AC133+ progenitor cells as gene delivery vehicle and cellular probe in subcutaneous tumor models: a preliminary study. BMC Biotechnology 9 28. (doi:10.1186/1472-6750-9-28)
Read ML, Seed RI, Modasia B, Kwan PP, Sharma N, Smith VE, Watkins RJ, Bansal S, Gagliano T & Stratford AL et al. The proto-oncogene PBF binds p53 and is associated with prognostic features in colorectal cancer Molecular Carcinogenesis 2014 [in press] doi:10.1002/mc.22254)
Renier C, Vogel H, Offor O, Yao C & Wapnir I 2010 Breast cancer brain metastases express the sodium iodide symporter. Journal of Neuro-Oncology 96 331–336. (doi:10.1007/s11060-009-9971-8)
Riesco-Eizaguirre G, De la Vieja A, Rodriguez I, Miranda S, Martin-Duque P, Vassaux G & Santisteban P 2011 Telomerase-driven expression of the sodium iodide symporter (NIS) for in vivo radioiodide treatment of cancer: a new broad-spectrum NIS-mediated antitumor approach. Journal of Clinical Endocrinology and Metabolism 96 E1435–E1443. (doi:10.1210/jc.2010-2373)
Rillema JA & Hill MA 2003a Prolactin regulation of the pendrin-iodide transporter in the mammary gland. American Journal of Physiology. Endocrinology and Metabolism 284 E25–E28. (doi:10.1152/ajpendo.00383.2002)
Rillema JA & Hill MA 2003b Pendrin transporter carries out iodide uptake into MCF-7 human mammary cancer cells. Experimental Biology and Medicine 228 1078–1082.
del Rincon SV, Rousseau C, Samanta R & Miller WH Jr 2003 Retinoic acid-induced growth arrest of MCF-7 cells involves the selective regulation of the IRS-1/PI 3-kinase/AKT pathway. Oncogene 22 3353–3360. (doi:10.1038/sj.onc.1206485)
Rodriguez AM, Perron B, Lacroix L, Caillou B, Leblanc G, Schlumberger M, Bidart JM & Pourcher T 2002 Identification and characterization of a putative human iodide transporter located at the apical membrane of thyrocytes. Journal of Clinical Endocrinology and Metabolism 87 3500–3503. (doi:10.1210/jcem.87.7.8797)
Sapino A, Cassoni P, Stella A & Bussolati G 1998 Oxytocin receptor within the breast: biological function and distribution. Anticancer Research 18 2181–2186.
Sato E, Nakashima T, Miura Y, Furuhashi A, Nakayama A, Mori N, Murakami H, Naganawa S & Tadokoro M 2001 Phenotypes associated with replacement of His by Arg in the Pendred syndrome gene. European Journal of Endocrinology/European Federation of Endocrine Societies 145 697–703. (doi:10.1530/eje.0.1450697)
Scott DA, Wang R, Kreman TM, Sheffield VC & Karniski LP 1999 The Pendred syndrome gene encodes a chloride-iodide transport protein. Nature Genetics 21 440–443. (doi:10.1038/7783)
Seidlin SM, Marinelli LD & Oshry E 1946 Radioactive iodine therapy; effect on functioning metastases of adenocarcinoma of the thyroid. Journal of the American Medical Association 132 838–847. (doi:10.1001/jama.1946.02870490016004)
Sheffield VC, Kraiem Z, Beck JC, Nishimura D, Stone EM, Salameh M, Sadeh O & Glaser B 1996 Pendred syndrome maps to chromosome 7q21-34 and is caused by an intrinsic defect in thyroid iodine organification. Nature Genetics 12 424–426. (doi:10.1038/ng0496-424)
Shennan DB 2001 Iodide transport in lactating rat mammary tissue via a pathway independent from the Na+/I− cotransporter: evidence for sulfate/iodide exchange. Biochemical and Biophysical Research Communications 280 1359–1363. (doi:10.1006/bbrc.2001.4278)
Smith VE, Read ML, Turnell AS, Watkins RJ, Watkinson JC, Lewy GD, Fong JC, James SR, Eggo MC & Boelaert K et al. 2009 A novel mechanism of sodium iodide symporter repression in differentiated thyroid cancer. Journal of Cell Science 122 3393–3402. (doi:10.1242/jcs.045427)
Smith V, Sharma N, Watkins R, Read M, Ryan G, Kwan P, Martin A, Watkinson J, Boelaert K & Franklyn J et al. 2013 Manipulation of PBF/PTTG1IP phosphorylation status; a potential new therapeutic strategy for improving radioiodine uptake in thyroid and other tumors. Journal of Clinical Endocrinology and Metabolism 98 2876–2886. (doi:10.1210/jc.2012-3640)
Spaeth E, Klopp A, Dembinski J, Andreeff M & Marini F 2008 Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Therapy 15 730–738. (doi:10.1038/gt.2008.39)
Spitzweg C, O'Connor MK, Bergert ER, Tindall DJ, Young CY & Morris JC 2000 Treatment of prostate cancer by radioiodine therapy after tissue-specific expression of the sodium iodide symporter. Cancer Research 60 6526–6530.
Spitzweg C, Harrington KJ, Pinke LA, Vile RG & Morris JC 2001 Clinical review 132: the sodium iodide symporter and its potential role in cancer therapy. Journal of Clinical Endocrinology and Metabolism 86 3327–3335. (doi:10.1210/jcem.86.7.7641)
Stratford AL, Boelaert K, Tannahill LA, Kim DS, Warfield A, Eggo MC, Gittoes NJ, Young LS, Franklyn JA & McCabe CJ 2005 Pituitary tumor transforming gene binding factor: a novel transforming gene in thyroid tumorigenesis. Journal of Clinical Endocrinology and Metabolism 90 4341–4349. (doi:10.1210/jc.2005-0523)
Takasu N, Charrier B, Mauchamp J & Lissitzky S 1978 Modulation of adenylate cyclase/cyclic AMP response by thyrotropin and prostaglandin E2 in cultured thyroid cells. 2. Positive regulation. European Journal of Biochemistry/FEBS 90 139–146. (doi:10.1111/j.1432-1033.1978.tb12584.x)
Tallman MS, Andersen JW, Schiffer CA, Appelbaum FR, Feusner JH, Ogden A, Shepherd L, Willman C, Bloomfield CD & Rowe JM et al. 1997 All-trans-retinoic acid in acute promyelocytic leukemia. New England Journal of Medicine 337 1021–1028. (doi:10.1056/NEJM199710093371501)
Tazebay UH, Wapnir IL, Levy O, Dohan O, Zuckier LS, Zhao QH, Deng HF, Amenta PS, Fineberg S & Pestell RG et al. 2000 The mammary gland iodide transporter is expressed during lactation and in breast cancer. Nature Medicine 6 871–878. (doi:10.1038/78630)
Thangaraju M, Gopal E, Martin PM, Ananth S, Smith SB, Prasad PD, Sterneck E & Ganapathy V 2006 SLC5A8 triggers tumor cell apoptosis through pyruvate-dependent inhibition of histone deacetylases. Cancer Research 66 11560–11564. (doi:10.1158/0008-5472.CAN-06-1950)
Thorpe SM 1976 Increased uptake of iodide by hormone-responsive compared to hormone-independent mammary tumors in GR mice. International Journal of Cancer. Journal International du Cancer 18 345–350. (doi:10.1002/ijc.2910180312)
Titcomb MW, Gottardis MM, Pike JW & Allegretto EA 1994 Sensitive and specific detection of retinoid receptor subtype proteins in cultured cell and tumor extracts. Molecular Endocrinology 8 870–877. (doi:10.1210/mend.8.7.7984149)
Tjuvajev JG, Stockhammer G, Desai R, Uehara H, Watanabe K, Gansbacher B & Blasberg RG 1995 Imaging the expression of transfected genes in vivo. Cancer Research 55 6126–6132.
Unterholzner S, Willhauck MJ, Cengic N, Schutz M, Goke B, Morris JC & Spitzweg C 2006 Dexamethasone stimulation of retinoic acid-induced sodium iodide symporter expression and cytotoxicity of 131-I in breast cancer cells. Journal of Clinical Endocrinology and Metabolism 91 69–78. (doi:10.1210/jc.2005-0779)
Vadysirisack DD, Chen ES, Zhang Z, Tsai MD, Chang GD & Jhiang SM 2007 Identification of in vivo phosphorylation sites and their functional significance in the sodium iodide symporter. Journal of Biological Chemistry 282 36820–36828. (doi:10.1074/jbc.M706817200)
Wapnir IL, van de Rijn M, Nowels K, Amenta PS, Walton K, Montgomery K, Greco RS, Dohan O & Carrasco N 2003 Immunohistochemical profile of the sodium/iodide symporter in thyroid, breast, and other carcinomas using high density tissue microarrays and conventional sections. Journal of Clinical Endocrinology and Metabolism 88 1880–1888. (doi:10.1210/jc.2002-021544)
Wapnir IL, Goris M, Yudd A, Dohan O, Adelman D, Nowels K & Carrasco N 2004 The Na+/I− symporter mediates iodide uptake in breast cancer metastases and can be selectively down-regulated in the thyroid. Clinical Cancer Research 10 4294–4302. (doi:10.1158/1078-0432.CCR-04-0074)
Watkins RJ, Read ML, Smith VE, Sharma N, Reynolds GM, Buckley L, Doig C, Campbell MJ, Lewy G & Eggo MC et al. 2010 Pituitary tumor transforming gene binding factor: a new gene in breast cancer. Cancer Research 70 3739–3749. (doi:10.1158/0008-5472.CAN-09-3531)
Yoshida A, Taniguchi S, Hisatome I, Royaux IE, Green ED, Kohn LD & Suzuki K 2002 Pendrin is an iodide-specific apical porter responsible for iodide efflux from thyroid cells. Journal of Clinical Endocrinology and Metabolism 87 3356–3361. (doi:10.1210/jcem.87.7.8679)