Quick viewing(Text Mode)

The RUNX Family in Breast Cancer: Relationships with Estrogen Signaling

The RUNX Family in Breast Cancer: Relationships with Estrogen Signaling

Oncogene (2013) 32, 2121–2130 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc

REVIEW The RUNX family in : relationships with signaling

N-O Chimge1 and B Frenkel2

The three RUNX family members are lineage specific master regulators, which also have important, context-dependent roles in carcinogenesis as either tumor suppressors or oncogenes. Here we review evidence for such roles in breast cancer (BCa). RUNX1, the predominant RUNX family member in breast epithelial cells, has a tumor suppressor role reflected by many somatic found in primary tumor biopsies. The classical tumor suppressor RUNX3 does not consist of such a hot spot, but it too seems to inhibit BCa; it is often inactivated in human BCa tumors and its haploinsufficiency in mice leads to spontaneous BCa development. The tumor suppressor activities of RUNX1 and RUNX3 are mediated in part by antagonism of estrogen signaling, a feature recently attributed to RUNX2 as well. Paradoxically, however RUNX2, a master regulator, has been implicated in various aspects of metastasis in general and bone metastasis in particular. Reciprocating the anti-estrogenic tumor suppressor activity of RUNX , inhibition of RUNX2 by may help explain their context-dependent anti-metastatic roles. Such roles are reserved to non-osseous metastasis, because ERa is associated with increased, not decreased skeletal dissemination of BCa cells. Finally, based on diverse expression patterns in BCa subtypes, the successful use of future RUNX-based therapies will most likely require careful patient selection.

Oncogene (2013) 32, 2121–2130; doi:10.1038/onc.2012.328; published online 8 October 2012 Keywords: RUNX1; RUNX2; RUNX3; tumor suppressor; oncogene; ERa

The mammalian RUNX family of factors comprises diverse origins have been extensively reviewed,21–24 and Table 1 three members, RUNX1, RUNX2 and RUNX3, which have pivotal summarizes reported abnormalities implicating each of the RUNX roles in both normal development and neoplasia.1 Despite their proteins as a tumor suppressor in various cancer types. structural similarities, RUNX proteins have divergent physiological roles; RUNX1 is required for definitive hematopoiesis,2 RUNX2 has fundamental roles in the differentiation of and RUNX1 AS A BCa SUPPRESSOR ,3–5 and RUNX3 is involved in gastrointestinal and 6–8 In addition to their most notable roles in hematopoietic cells, bone neuronal development. Inherited mutations in RUNX1 are the cells and neurons, the three RUNX are also expressed in the cause of familial platelet disorder with associated myeloid normal , and their mRNA levels fluctuate during malignancy,9 and RUNX2 mutations cause the dominant 25 10 cycles of pregnancy, lactation and involution. RUNX1 is the autosomal skeletal disease cleidocranial dysplasia. predominant RUNX family member expressed in human breast In cancer, RUNX act as either tumor suppressors or oncogenes 13,26,27 1,11 epithelial cells (Figure 1a) and the is readily detected depending on cellular context. RUNX1 is a frequent target in both the basal and the luminal cell layers.26 The most of translocations and other mutations in hematopoietic maligna- compelling evidence for its role in inhibiting BCa is the somatic ncies, and RUNX3, located on 1p36, resides mutations recently observed in RUNX1 (and not RUNX2 or RUNX3) in a hotspot found in diverse cancers of epithelial, 28 12 in tumors from the Washington University BRC77 study and in hematopoietic and neuronal origins. Tumor suppressor activity the cohort of BCa patients studied in The Cancer Genome Atlas in breast cancer (BCa) has been attributed to all three RUNX pilot project29 (Figure 1b). Furthermore, RUNX1 was one of 17 proteins,13–15 whereas RUNX2 is increasingly recognized as pro- 30 11,16–20 genes whose expression pattern predicted BCa metastasis; its metastatic in breast (and prostate) cancer progression. Here expression is less abundant in BCa compared to normal breast we review the dual functions of RUNX proteins in BCa and their epithelial cells, and it progressively decreases with increasing relationships with estrogen signaling. BCa tumor aggression.31 Finally, experimental Ras-mediated transformation of basal-like MCF10A cells was associated with loss of RUNX1;31 and RUNX1 silencing in another study with RUNX PROTEINS ARE TUMOR SUPPRESSORS MCF10A cells led to the formation of abnormal, hyperproliferative All three mammalian RUNX family members have been implicated acinar structures in 3D cultures.13 in tumor suppression. The frequent functional inactivation of How RUNX1 suppresses BCa remains to be elucidated. Among RUNX1 and RUNX3 observed in hematopoietic and solid tumors of possible mechanisms, RUNX1 may stimulate E-cadherin to inhibit

1Department of Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA and 2Departments of Orthopaedic Surgery and Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA. Correspondence: Dr N-O Chimge or Dr B Frenkel, Department of Biochemistry and Molecular Biology, University of Southern California, Institute for Genetic Medicine, 2250 Alcazar Street, CSC-240, Los Angeles, CA 90033, USA. E-mail: [email protected] or [email protected] Received 3 May 2012; revised 20 June 2012; accepted 20 June 2012; published online 8 October 2012 RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2122 Table 1. Tumor suppressor activities of the RUNX family in various cancers

RUNX Types of cancer Related mechanisms References

RUNX1 AML-M2 RUNX1-ETO: t(8;21)(q22;q22) 137–140 RUNX1-LRP16: t(11;21)(q13;q22) RUNX1-MTG16: t(16;21)(q24;q22) RUNX1-others Mut 141,142

MDS RUNX1-EVI: t(3;21)(q26.2;q22) 140,143–149 RUNX1-MDS1: t(3;21)(q26.2;q22) RUNX1-EAP: t(3;21)(q26.2;q22) RUNX1-PRDM6: t(1;21)(p36;q22) RUNX1-others Mut 150–156

B-ALL TEL-RUNX1: t(12;21)(p13;q22) 157–160 FPDMM Hemizygous deletion, SNP 9,161 AML-M1 Mut 162 AML-M0 LOH, biallelic RUNX1 mut 162–164 CMML Mut 165,166 Colorectal Mut 167 Gastointestinal tract Mut 168,169 Esophageal squamous-cell carcinoma SNP 170,171 Prostate SNP 172 Breast Mut 28,29

RUNX2 Inhibition of proliferation 173,174 Breast Antagonism of estrogen signaling 15

RUNX3 Gastric Hemizygous deletion, Mut, 8,167,169,175–177 hypermethylation, cytoplasmic mislocalization Bladder Mut, hypermethylation 178–180 Prostate Hypermethylation 181 Lung Hemizygous deletion, hypermethylation 182 Pancreatic Hemizygous deletion, hypermethylation 183 Colorectal Hypermethylation, Mut, cytoplasmic mislocalization 184–187 Brain Hypermethylation 188 Squamous cell carcinoma Cytoplasmic mislocalization 189 Hepatocellular carcinoma Hypermethylation 37,190 Bile duct Hemizygous deletion 183 Renal cell carcinoma Inhibition of proliferation 191 Bone Hypermethylation 192 Breast Hypermethylation, 14,34,35 cytoplasmic mislocalization Abbreviations: AML, acute myeloblastic ; AML-M0, minimally differentiated acute myeloblastic leukemia; AML-M1, acute myeloblastic leukemia without maturation; CMML, chronic myelomonocytic leukemia; FPDMM, familial platelet disorder with associated myeloid malignancy; LOH, loss of heterozygosity; MDS, ; Mut, mutations other than SNPs or translocations; SNP, single-nucleotide polymorphism; t, translocation. Studies of breast cancer are in bold text.

epithelial-to-mesenchymal transition (EMT)32 and it may increased signaling owing to partial loss of ERa cooperate with FOXO signaling to protect ER-negative BCa cells destabilization by RUNX314 (Figure 2a). Expression of RUNX3 from oxidative stress.13 In ER-positive cells, however, we believe decreases during human BCa progression and high RUNX3 RUNX1 attenuates oncogenic effects of estrogens (Figure 2a) levels are associated with favorable prognosis.35 As in other mainly through physical interaction with ERa.33 In fact, all but one malignancies, RUNX3 is frequently inactivated in BCa by RUNX1 somatic mutations found so far are in ERa-positive hypermethylation, genetic deletion, and protein tumors.28,29 mislocalization.34,35,37–42 RUNX3 promoter hypermethylation in response to estradiol has been observed in mammosphere- derived cells, which may contribute to the oncogenic activity of estradiol in breast epithelial cells.43 RUNX3 AS A BCa SUPPRESSOR Evidence suggesting a role for RUNX3 in inhibiting various malignancies is cited in Table 114,34–36 and the literature on the tumor suppressor role of RUNX3 specifically in BCa has been RUNX2 AS A BCa SUPPRESSOR recently reviewed.24 Most notably, RUNX3 hemizygous female RUNX2 is expressed in the developing mammary gland during mice develop spontaneous ductal carcinoma,14 attributable to embryogenesis5 and , and its mRNA levels in glands of

Oncogene (2013) 2121 – 2130 & 2013 Macmillan Publishers Limited RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2123 RUNX1: Chr.21q22.3 Scale: 200 kb hg19 RUNX1 RUNX1 RUNX1

Brain Heart Lymph Node Breast HMEC

RUNX2: Chr.6p21 Scale: 200 kb hg19 RUNX2 RUNX2 RUNX2

Brain Heart Lymph Node Breast HMEC

RUNX3: Chr.1p36 Scale: 200 kb hg19 RUNX3 RUNX3

Brain Heart Lymph Node Breast HMEC

frame shift deletion frame shift insertion RUNX1 in-frame deletion missense nonsense F259fs Q266* L288fs L317F 322e7+2 S100C L112fs 117e3+1 P125fs G135D N139S ELRNA143 in-frame del L161P R166Q G168R G168E R169fs R169K 169e4+1 D198G S218fs splice site splice site insertion

010050 150 200 250 300 350 400 450 481 Figure 1. RUNX1, highly expressed in breast epithelial cells, is frequently mutated in BCa. (a) RNA-sequencing data for RUNX1, RUNX2 and RUNX3 in human brain, heart, lymph node and breast tissues and in Human Mammary Epithelial Cells (HMEC) were produced by Wang et al.27 and displayed using the UCSC Genome Browser (http://genome.ucsc.edu). (b) Lollipop plot of RUNX1 somatic mutations identified by The Cancer Genome Atlas project29 and the Washington University BRC77 study.28 These studies indentified only one synonymous mutation in RUNX2 and none in RUNX3. Blue box represents the Runt DNA-binding domain.

5-week-old female mice are highest in the terminal end buds.44 resulted in a global increase of RUNX2 activity.15 Antagonism of Suggesting a physiological role in mammary epithelial cells, estradiol signaling by RUNX2 in vitro was particularly interesting RUNX2 regulates transcription of beta-casein,45 a mammary for a gene set, heretofore R2iEs, which was inhibited by RUNX2 gland-specific gene, and osteopontin,46 which is expressed in (R2i) and stimulated by estradiol (Es). The short-term response these cells during pregnancy and lactation. A growth-suppressive of the R2iEs gene set to letrozole treatment predicted role for RUNX2 was demonstrated in osteoblasts,47–49 and its clinical outcome better than other estradiol-regulated gene sets. tumor suppressor role in hormone carcinogenesis has been Thus, the R2iEs gene set, which is enriched for polo-like - suggested based on the antagonism of ERa- and androgen related mitotic pathway genes,15 represents a critical growth -mediated transcription in breast and regulatory node where the mitogenic property of estrogens cells, respectively.50,51 In the MCF7/Rx2dox BCa cell line, RUNX2 meets the anti-proliferative effect of RUNX2 in BCa cells. Finally, it diminished E2-mediated colony formation in soft agar.15 This was is important to note that RUNX2 can have either a negative attributable to the RUNX2-mediated general attenuation of the or a positive role in BCa initiation depending on cellular transcriptional response to E2, demonstrated by genome-wide context; contrasting its aforementioned tumor suppressor role in mRNA analysis.15 Consistent with this in vitro finding, inhibition the ERa-positive MCF7 BCa cell line,15 RUNX2 overexpression of estrogen signaling by letrozole treatment of BCa patients increased proliferation and induced abnormal acini formation

& 2013 Macmillan Publishers Limited Oncogene (2013) 2121 – 2130 RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2124 similar to RUNX1, it cooperated with c-,75 as well as with CBFb- SMMHC,76 to induce murine hematologic cancers. Amplification TUMOR TUMOR of RUNX2 on human chromosome 6p21 was related to INITIATION PROGRESSION osteosarcoma,77,78 a malignancy arising from osteoblasts, the ER ER cells in which RUNX2 has its most notable developmental role. Oncogenic Protective Furthermore, RUNX2 is overexpressed in carcinomas of the colon,79 pancreas80 and thyroid.81 In animal models of various malignancies including the prostate,82 lung83 and thyroid 84 RUNX RUNX2 cancer, an increase in RUNX2 observed early during cancer tumor suppressor pro-metastatic development likely induces EMT and local invasion. However, as discussed in detail below, most of the published work implicating RUNX2 in promoting cancer relates to prostate cancer and Figure 2. Working model for the reciprocal antagonism between 11,16–20,85–87 ERa and RUNX signaling. (a) During tumor initiation, RUNX proteins BCa. act as tumor suppressors in part by inhibiting ERa.(b) The beneficial The expression of bone-phenotypic genes in metastatic tumors effect of ERa in late-stage disease is attributable in part to repression was initially pursued in the context of prostate cancer, with the of the pro-metastatic properties of RUNX2. idea that the frequent dissemination of prostate cancer cells to bone may rely on osteomimetic mechanisms.88 A similar line of work followed in the context of BCa, which also metastasizes to bone with high frequency.89 Early studies documented ectopic expression in prostate cancer and BCa cells of CXCR4, MMPs, OPN, in the ERa-negative MCF10A cell line.17 Also contrasting its BSP, RANKL20,90–96 and finally RUNX2, the osteoblast master tumor suppressor activity is the pro-metastatic property regulator.97,98 RUNX2 was shown to prevent prostate cancer cell of RUNX2, which will be discussed in detail under the next apoptosis by stimulating the expression of survivin and Bcl2.82,99 subtitle. Increased levels of RUNX2 during prostate and breast tumor Multiple mechanisms could contribute to the antagonism of ERa growth was linked to BMP7-mediated protection of cancer cells by RUNX2. First, RUNX2 decreases ERa mRNA and protein levels in against apoptosis.100–102 Perturbations of RUNX2 in prostate BCa cells;15 this inhibition could occur at the transcriptional level cancer and BCa cells abolished their ability to form osteolytic because it has been shown, albeit in prostate cancer cells, that lesions in mouse xenograft model.16,20,103 RUNX2 is recruited to the ERa proximal promoter.52 Second, Recent studies have implicated RUNX2 in metastasis beyond a because RUNX2 binds ERa’s DNA-binding domain,51 the interaction role in bone homing, including local invasiveness as well as may result not only in decreased association of RUNX2 with its growth of tumors that have already metastasized to the skeleton. target genes,15,51 but also in decreased association of ERa with its Induction of RUNX2 in prostate cancer cells in vitro stimulated targets.15 Consistent with this idea, re-analysis of two recent data expression of numerous genes with known roles in EMT, sets33,53 indicated that genomic ERa targets are more likely to be invasiveness, extracellular matrix degradation, angiogenesis and antagonized by RUNX2 compared with non-genomic targets.15 osteolysis,11,15,17,19,20,86,104,105 with some of these metastatic Two observations suggest that this mechanism is likely specific for phenotypes possibly depending on RUNX2-stimulated protein RUNX2. First, in contrast to the interaction of RUNX2 with the DNA- secretion.52 Thus, whereas RUNX2 initiates an osteoblastic binding domain of ERa,51 the reported interaction with RUNX3 program in mesenchymal pluripotent cells,106 its ectopic involves the hinge region of ERa.24 Second, the C-terminus of expression in cancer cells initiates a transcriptional program RUNX2, which contains its ERa-binding region,51 is not conserved responsible for various aspects of metastasis, including but not in other RUNX proteins.1 Thus, whereas the BCa-suppressor limited to bone homing. activities of both RUNX2 and RUNX3 are at least in part Interestingly, manipulation of RUNX2 in prostate cancer and attributable to inhibition of ERa, the respective molecular BCa cells modulated the expression of several genes implicated mechanisms are likely distinct. in osteoclast differentiation and bone resorption, including OPG, CSF2, SPHK1, PTHrP, IL8 and several MMPs.15,17–20,104,107 Stimulation of bone resorption by prostate cancer cells is not ONCOGENIC PROPERTIES OF THE RUNX FAMILY AND THE unlike the increased resorption leading to the osteolytic lesions EMERGING ROLE OF RUNX2 IN METASTASIS typically associated with BCa bone metastasis, even though the Insertional mutagenesis studies in murine models have assigned co-stimulation of bone formation predominates in most cases of oncogenic properties to each of the three RUNX genes.54–62 prostate cancer bone metastases, resulting in osteoblastic Furthermore, RUNX163 synergized with c-Myc in murine tumors.108 It is the high bone turnover induced by both prostate lymphoma development, and acted as an oncogene in the cancer and BCa cells that elicits a vicious cycle whereby matrix- mouse skin by promoting epithelial cell proliferation.64 In humans, embedded growth factors, normally reserved to secure the 21q region that includes RUNX1 is amplified in a small, sufficient bone formation, are now released at higher rates and aggressive sub set of childhood B-cell lineage acute lymphoblastic hijacked to promote tumor cell growth.108,109 RUNX2-mediated leukemia.65–68 Moreover, RUNX1 was among the most highly tumor cell-driven osteoclastogenesis represents mimicry of up-regulated genes in invasive endometrial carcinoma.69–71 In BCa the osteoclastogenic role that RUNX2 has in the osteoblast RUNX1-mediated tethering of ERa to target genes33 may also lineage.110–115 Confirming the significance of RUNX2 in human contribute to breast hormone carcinogenesis. There is also some BCa aggression, Onodera et al.87 showed that RUNX2 immuno- evidence that RUNX3, a classical tumor suppressor, can act histochemical labeling index in primary tumor biopsies positively to promote human cancers. Possibly related to promoter correlated with disease progression and metastasis. The pro- demethylation, the gene is over-expressed in carcinomas of the metastatic properties of RUNX2 in the MCF7/Rx2dox BCa cell line head and neck,72 skin73 and ovary.74 critically depended on its target gene SNAI2, as knockdown of Whereas most of the cancer-related literature on RUNX genes, SNAI2 abolished RUNX2-driven EMT and invasiveness86 (Figure 3). in particular RUNX1 and RUNX3, concerns their tumor suppressor Conceivably, RUNX2-driven EMT in BCa cells86 recapitulates a role activities, RUNX2 is increasingly recognized in the cancer field for that RUNX2 has during normal branching , its oncogenic properties. RUNX2 was the most frequent target for as suggested by its co-expression with EMT-related genes in viral insertions in murine retroviral mutagenesis screens,61,68 and mammary gland terminal end buds.44

Oncogene (2013) 2121 – 2130 & 2013 Macmillan Publishers Limited RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2125 ROLE OF RUNX2 AND ESTROGEN SIGNALING IN BCa As mentioned above, the osteomimetic property of prostate METASTASIS and later BCa was initially considered in the context of bone- Estrogen signaling, which promotes BCa initiation, has a homing. However, consistent with the role of RUNX2 in EMT paradoxical anti-metastatic role in advanced disease,116–118 and and invasion, which promote early metastatic events, the this is attributable in part to inhibiting the expression of RUNX2 predominant contribution of RUNX2 to the metastatic process is 86 target genes, including SNAI251,86,119 (Figure 3). Relationships not necessarily bone-specific. Other signaling pathways, such as among RUNX2, SNAI2 and ERa in vivo are demonstrable in gene Src, increase the predilection of a sub set of primary BCa cells to 120 expression profiles of BCa biopsies, where SNAI2 positively bone. These cells then give rise to metastases with a gene correlates with RUNX2 activity, and the RUNX2/SNAI2 axis expression signature that specifically reflects adaptation to 121 negatively correlates with ERa.86 Furthermore, in the study of bone, as opposed to the gene signature of BCa lung 122 Onodera et al., the negative correlation of RUNX2 with disease- metastases, which better reflects general metastatic properties. free survival was much stronger for ERa-negative compared with Because BCa metastases to bone versus other organs are ERa-positive tumors.87 Thus, antagonism between RUNX2 and processes with distinct molecular characteristics, it is not estrogen signaling in BCa has a role in two distinct contexts. First, surprising that they exhibit differential sensitivity to estrogen as discussed earlier, ERa signaling is targeted by RUNX2 (as well signaling. Whereas ERa is negatively correlated with non- 86 as RUNX1 and RUNX3), acting to suppress tumor initiation osseous metastasis, attributable in part to antagonism of the (Figure 2a). Second, the beneficial effect of ERa signaling in RUNX2/SNAI2 axis, it is positively correlated with bone 86,120,123 late-stage disease is attributable in part to inhibition of RUNX2 metastasis (Figure 3). Thus, ERa in bone-seeking tumors activity, now acting to promote metastasis (Figure 2b). not only permits expression of RUNX2 target genes, but in fact appears to promote bone metastasis. The co-existence of RUNX2 Primary and ERa in bone-seeking BCa cells may even result in positive Tumor interaction at regulatory elements of genes that promote bone- homing and growth within this microenvironment. Indeed, although RUNX2 and estrogen signaling are generally antagonis- RUNX2 tic, expression profiling of MCF7/Rx2dox cells also disclosed genes EMT, that are additively or synergistically regulated by the two 86 Invasion pathways. It will be interesting to test the role of these genes SNAI2 in bone metastasis and to compare the in vivo effects of RUNX2 on growth of ERa-positive versus ERa-negative BCa cells in bone  ER versus non-osseous tissues.

RUNX PROTEINS IN BCa SUBTYPES BCa is a heterogeneous disease in terms of cell of origin, clinical SNAI2 RUNX2 progression and drug responsiveness. Based on their intrinsic profiles, BCa tumors can be classified into distinct ER molecular subtypes: luminal-like A, luminal-like B, basal-like, HER2- positive and normal breast-like.124,125 Figure 4 describes the Brain expression/activity of the three RUNX proteins, along with ERa and HER2, in a cohort of 557 tumors compiled previously86 and stratified per subtype based on the PAM-50 algorithm.126 Most Lung obviously, high RUNX2 activity and high RUNX3 expression characterize the ERa-negative basal-like tumors, attributing to the reported negative correlation between these two RUNX Bone Non-osseous members and ERa in BCa cells.14,15,24,43,51 High RUNX2 activity was metastasis metastasis also present in some HER2 enriched tumors, but not in the luminal subtypes, consistent with the positive correlation previously Figure 3. Roles of RUNX2 and ERa in BCa metastasis. According to 87 this working model, the RUNX2/SNAI2 axis promotes early meta- observed between RUNX2 and HER2. Finally, Figure 4 demon- static events (e.g., EMT, invasion, intravasation) and bone metastasis. strates a negative correlation between RUNX1 and RUNX2/3 and a ERa antagonizes RUNX2 during the early metastatic events but strong positive correlation between RUNX2 activity and RUNX3. operates along with the RUNX2/SNAI2 axis during bone metastasis. The interdependent expression of RUNX members in BCa (Figure 4)

Normal- HER2 like Luminal B Luminal A enriched Basal-like

2 HER2

ESR1

0 RUNX1

RUNX3

-2 RUNX2 metagene Standardized intensities Figure 4. RUNX genes in BCa subtypes. Each tumor from a heterogeneous cohort of 557 BCa patients86 was subtyped according to the PAM- 50 algorithm126 (courtesy of Dr Charles Perou). The values for RUNX1, RUNX3, HER2 and ESR1 represent the signal intensity associated with probe sets 210805_x_at,26 204197_s_at, 216836_s_at and 205225_at,120 respectively (Affymetrix, Santa Clara, CA, USA). Because of the ambiguity with the RUNX2 probe sets in the early Affymetrix arrays used to profile mRNA expression, the RUNX2 values are those of a metagene defined as the average normalized expression of the top 100 RUNX2-stimulated genes in MCF7/Rx2dox BCa cells.15

& 2013 Macmillan Publishers Limited Oncogene (2013) 2121 – 2130 RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2126 is in line with the inverse relationships observed between RUNX1 review. This work was supported by NIH grant R01 DK071122 from the National and RUNX3 mRNA in B cells127,128 and RUNX1 and RUNX2 mRNAs Institute of Diabetes and Digestive and Kidney Diseases to BF, who holds the J Harold during skeletal development.129 These relationships are attributable and Edna L LaBriola Chair in Genetic Orthopaedic Research at USC. in part to the high similarities among the DNA-binding domains of the three RUNX proteins1 and the consequential similarities among REFERENCES their DNA binding elements52,130,131 and their transcriptomes.132 1 Blyth K, Cameron ER, Neil JC. The RUNX genes: gain or loss of function in cancer. Also likely having a role in the observed relationships between Nat Rev Cancer 2005; 5: 376–387. expression of different RUNX proteins are the RUNX-binding 2 Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. AML1 the target of elements in the RUNX gene promoters52,133,134 and their subsequent multiple chromosomal translocations in human leukemia, is essential for normal cross-regulation.127,128 fetal liver hematopoiesis. Cell 1996; 84: 321–330. 3 Komori T. Regulation of bone development and extracellular matrix protein genes by RUNX2. Cell Tissue Res 2010; 339: 189–195. 4 Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K et al. Targeted CONCLUDING REMARKS disruption of Cbfa1 results in a complete lack of bone formation owing to RUNX proteins have unique and often opposing roles in BCa. The maturational arrest of osteoblasts. Cell 1997; 89: 755–764. global reciprocal inhibitory effects between estrogen and RUNX2 5 Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR et al. Cbfa1, a signaling, and the clinical relevance of this antagonism15,86 candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast provide novel insight into hormone-driven carcinogenesis. They differentiation and bone development. Cell 1997; 89: 765–771. 6 Fukamachi H, Ito K. Growth regulation of gastric epithelial cells by Runx3. may ultimately contribute to a better understanding of how RUNX Oncogene 2004; 23: 4330–4335. genes switch their function from tumor suppressors to oncogenes. 7 Levanon D, Bettoun D, Harris-Cerruti C, Woolf E, Negreanu V, Eilam R et al. The They may also shed light on the contribution of estrogen signaling Runx3 regulates development and survival of TrkC dorsal to bone, but not other metastatic processes. So far, RUNX1 and root ganglia neurons. EMBO J 2002; 21: 3454–3463. RUNX3 were shown to exhibit only tumor suppressor activities 8 Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ et al. Causal relationship in BCa,13,24 whereas RUNX2 was capable of acting either as a between the loss of RUNX3 expression and gastric cancer. Cell 2002; 109: tumor suppressor or as an oncogene depending on cellular 113–124. context.15,17,86 Given the solid evidence for a tumor suppressor 9 Song WJ, Sullivan MG, Legare RD, Hutchings S, Tan X, Kufrin D et al. Hap- role for RUNX3 in BCa, its preferential expression in the basal-like, loinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nat Genet 1999; 23: 166–175. typically aggressive BCa subtype, may be explained by the low 10 Mundlos S. Cleidocranial dysplasia: clinical and molecular genetics. J Med Genet expression of ERa and/or RUNX1 (Figure 4). It remains to be 1999; 36: 177–182. investigated whether RUNX3 and/or RUNX1 share with RUNX2 the 11 Pratap J, Lian JB, Stein GS. Metastatic bone disease: role of transcription factors potential of driving BCa metastasis. Functional differences and future targets. Bone 2011; 48: 30–36. observed between the three RUNX members in BCa likely reflect 12 Bagchi A, Mills AA. The quest for the 1p36 tumor suppressor. Cancer Res 2008; various mechanisms regulating their own expression, as well as 68: 2551–2556. dissimilarities between their respective C and N termini, which 13 Wang L, Brugge JS, Janes KA. Intersection of FOXO- and RUNX1-mediated gene dictate formation of complexes and activation of genes that may expression programs in single breast epithelial cells during morphogenesis and be specific for individual RUNX proteins. Among many additional tumor progression. Proc Natl Acad Sci USA 2011; 108: E803–E812. 14 Huang B, Qu Z, Ong CW, Tsang YH, Xiao G, Shapiro D et al. RUNX3 acts as a open questions, it remains to be seen how functional redundancy, tumor suppressor in breast cancer by targeting alpha. auto- and cross-regulatory effects, competition, cooperation and/ Oncogene 2012; 31: 527–534. 135 or heterodimerization of RUNX proteins may contribute to 15 Chimge NO, Baniwal SK, Luo J, Coetzee S, Khalid O, Berman BP et al. Opposing defining their transcriptomes and cellular functions. effects of Runx2 and estradiol on breast cancer cell proliferation: in vitro iden- While improving our understanding of the roles of RUNX tification of reciprocally regulated gene signature related to clinical letrozole proteins in BCa, efforts are warranted to translate this knowledge responsiveness. Clin Cancer Res 2012; 18: 901–911. into improvement of BCa patient care. Successful development 16 Javed A, Barnes GL, Pratap J, Antkowiak T, Gerstenfeld LC, van Wijnen AJ et al. of RUNX-based therapeutic approaches will unlikely target Impaired intranuclear trafficking of Runx2 (AML3/CBFA1) transcription factors in features shared by all RUNX proteins, such as the RUNT domain breast cancer cells inhibits osteolysis in vivo. Proc Natl Acad Sci USA 2005; 102: 136 1454–1459. or the common co-activator CBFb. Rather, RUNX-based future 17 Pratap J, Imbalzano KM, Underwood JM, Cohet N, Gokul K, Akech J et al. Ectopic therapeutics will likely target mechanisms specific for individual expression in mammary epithelial cells disrupts formation of normal acini RUNX proteins in specific BCa subtypes. For example, the design structure: implications for breast cancer progression. Cancer Res 2009; 69: of RUNX-based anti-metastatic drugs should probably target 6807–6814. RUNX2, sparing the tumor suppressor activities of RUNX1 and 18 Pratap J, Wixted JJ, Gaur T, Zaidi SK, Dobson J, Gokul KD et al. Runx2 tran- RUNX3. During the development of such drugs, special attention scriptional activation of Indian Hedgehog and a downstream bone metastatic should be paid to the potential loss of the inhibition of estrogen pathway in breast cancer cells. Cancer Res 2008; 68: 7795–7802. signaling in ERa-positive tumors. On the other hand, the design of 19 Baniwal SK, Khalid O, Gabet Y, Shah RR, Purcell DJ, Mav D et al. Runx2 tran- future selective estrogen receptor modulators (SERMs) should take scriptome of prostate cancer cells: insights into invasiveness and bone metas- tasis. Mol Cancer 2010; 9: 258. into consideration their effects on RUNX proteins, with the ideal 20 Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA et al. Runx2 SERM possessing both anti-ER and anti-RUNX2, but not anti- association with progression of prostate cancer in patients: mechanisms med- RUNX1 or anti-RUNX3 properties. iating bone osteolysis and osteoblastic metastatic lesions. Oncogene 2010; 29: 811–821. 21 Chuang LS, Ito Y. RUNX3 is multifunctional in carcinogenesis of multiple solid CONFLICT OF INTEREST tumors. Oncogene 2010; 29: 2605–2615. 22 Mangan JK, Speck NA. RUNX1 mutations in clonal myeloid disorders: from The authors declare no conflict of interest. conventional cytogenetics to next generation sequencing. A story 40 years in the making. Crit Rev Oncog 2011; 16: 77–91. 23 De Braekeleer E, Ferec C, De Braekeleer M. RUNX1 translocations in malignant ACKNOWLEDGEMENTS hemopathies. Anticancer Res 2009; 29: 1031–1037. We thank Gerhard A Coetzee, Debu Tripathy, Gillian H Little (University of Southern 24 Chen LF. Tumor suppressor function of RUNX3 in breast cancer. J Cell Biochem California) and Andre van Wijnen (University of Massachusetts Medical School) for 2012; 113: 1470–1477. carefully reading the manuscript and providing critical comments. We are particularly 25 Blyth K, Vaillant F, Jenkins A, McDonald L, Pringle MA, Huser C et al. Runx2 thankful to Charles Perou (University of North Carolina), Dan Koboldt and Matthew in normal tissues and cancer cells: a developing story. Blood Cells Mol Dis 2010; Ellis (Washington University), who also shared, analyzed and edited data for this 45: 117–123.

Oncogene (2013) 2121 – 2130 & 2013 Macmillan Publishers Limited RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2127 26 Janes KA. RUNX1 and its understudied role in breast cancer. 2011; 10: 51 Khalid O, Baniwal SK, Purcell DJ, Leclerc N, Gabet Y, Stallcup MR et al. Modulation 3461–3465. of Runx2 activity by estrogen receptor-alpha: implications for osteoporosis and 27 Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C et al. Alternative breast cancer. Endocrinology 2008; 149: 5984–5995. isoform regulation in human tissue transcriptomes. Nature 2008; 456: 470–476. 52 Little GH, Noushmehr H, Baniwal SK, Berman BP, Coetzee GA, Frenkel B. 28 Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW et al. Whole genome analysis Genome-wide Runx2 occupancy in prostate cancer cells suggests a role in informs breast cancer response to aromatase inhibition. Nature 2012; 486: regulating secretion. Nucleic Acids Res 2012; 40: 3538–3547. 353–360. 53 Cicatiello L, Mutarelli M, Grober OM, Paris O, Ferraro L, Ravo M et al. Estrogen 29 Network TCGA. Comprehensive molecular portraits of human breast tumors. receptor alpha controls a gene network in luminal-like breast cancer cells Nature (in press). comprising multiple transcription factors and microRNAs. Am J Pathol 2010; 176: 30 Ramaswamy S, Ross KN, Lander ES, Golub TR. A molecular signature of metas- 2113–2130. tasis in primary solid tumors. Nat Genet 2003; 33: 49–54. 54 Stewart M, MacKay N, Cameron ER, Neil JC. The common retroviral insertion 31 Kadota M, Yang HH, Gomez B, Sato M, Clifford RJ, Meerzaman D et al. Deli- Dsi1 maps 30 kilobases upstream of the P1 promoter of the murine Runx3/ neating genetic alterations for tumor progression in the MCF10A series of breast Cbfa3/Aml2 gene. J Virol 2002; 76: 4364–4369. cancer cell lines. PLoS One 2010; 5: e9201. 55 Miething C, Grundler R, Mugler C, Brero S, Hoepfl J, Geigl J et al. Retroviral 32 Liu YN, Lee WW, Wang CY, Chao TH, Chen Y, Chen JH. Regulatory insertional mutagenesis identifies RUNX genes involved in chronic myeloid mechanisms controlling human E-cadherin gene expression. Oncogene 2005; 24: leukemia disease persistence under imatinib treatment. Proc Natl Acad Sci USA 8277–8290. 2007; 104: 4594–4599. 33 Stender JD, Kim K, Charn TH, Komm B, Chang KC, Kraus WL et al. Genome-wide 56 Dupuy AJ, Akagi K, Largaespada DA, Copeland NG, Jenkins NA. Mammalian analysis of DNA binding and tethering mechanisms mutagenesis using a highly mobile somatic Sleeping Beauty transposon system. identifies Runx1 as a novel tethering factor in receptor-mediated transcriptional Nature 2005; 436: 221–226. activation. Mol Cell Biol 2010; 30: 3943–3955. 57 Rahrmann EP, Collier LS, Knutson TP, Doyal ME, Kuslak SL, Green LE et al. 34 Lau QC, Raja E, Salto-Tellez M, Liu Q, Ito K, Inoue M et al. RUNX3 is frequently Identification of PDE4D as a proliferation promoting factor in prostate cancer inactivated by dual mechanisms of protein mislocalization and promoter using a sleeping beauty transposon-based somatic mutagenesis screen. Cancer hypermethylation in breast cancer. Cancer Res 2006; 66: 6512–6520. Res 2009; 69: 4388–4397. 35 Jiang Y, Tong D, Lou G, Zhang Y, Geng J. Expression of RUNX3 gene, methylation 58 Li J, Shen H, Himmel KL, Dupuy AJ, Largaespada DA, Nakamura T et al. Leu- status and clinicopathological significance in breast cancer and breast cancer kaemia disease genes: large-scale cloning and pathway predictions. Nat Genet cell lines. Pathobiology 2008; 75: 244–251. 1999; 23: 348–353. 36 Subramaniam MM, Chan JY, Soong R, Ito K, Ito Y, Yeoh KG et al. RUNX3 inacti- 59 Mikkers H, Allen J, Knipscheer P, Romeijn L, Hart A, Vink E et al. High-throughput vation by frequent promoter hypermethylation and protein mislocalization retroviral tagging to identify components of specific signaling pathways in constitute an early event in breast cancer progression. Breast Cancer Res Treat cancer. Nat Genet 2002; 32: 153–159. 2009; 113: 113–121. 60 Suzuki T, Shen H, Akagi K, Morse HC, Malley JD, Naiman DQ et al. New genes 37 Kim TY, Lee HJ, Hwang KS, Lee M, Kim JW, Bang YJ et al. Methylation of RUNX3 in involved in cancer identified by retroviral tagging. Nat Genet 2002; 32: 166–174. various types of human cancers and premalignant stages of gastric carcinoma. 61 Stewart M, Terry A, Hu M, O0Hara M, Blyth K, Baxter E et al. Proviral insertions Lab Invest 2004; 84: 479–484. induce the expression of bone-specific isoforms of PEBP2alphaA (CBFA1): 38 Suzuki M, Shigematsu H, Shames DS, Sunaga N, Takahashi T, Shivapurkar N et al. evidence for a new myc collaborating oncogene. Proc Natl Acad Sci USA 1997; DNA methylation-associated inactivation of TGFbeta-related genes DRM/Grem- 94: 8646–8651. lin, RUNX3, and HPP1 in human cancers. Br J Cancer 2005; 93: 1029–1037. 62 Vaillant F, Blyth K, Terry A, Bell M, Cameron ER, Neil J et al. A full-length Cbfa1 39 Hwang KT, Han W, Bae JY, Hwang SE, Shin HJ, Lee JE et al. Downregulation of the gene product perturbs T-cell development and promotes lymphomagenesis in RUNX3 gene by promoter hypermethylation and hemizygous deletion in breast synergy with myc. Oncogene 1999; 18: 7124–7134. cancer. JKorean Med Sci 2007; 22(Suppl): S24–S31. 63 Blyth K, Slater N, Hanlon L, Bell M, Mackay N, Stewart M et al. Runx1 promotes 40 Tan SH, Ida H, Lau QC, Goh BC, Chieng WS, Loh M et al. Detection of promoter B-cell survival and lymphoma development. Blood Cells Mol Dis 2009; 43: 12–19. hypermethylation in serum samples of cancer patients by methylation-specific 64 Hoi CS, Lee SE, Lu SY, McDermitt DJ, Osorio KM, Piskun CM et al. Runx1 directly polymerase chain reaction for tumour suppressor genes including RUNX3. Oncol promotes proliferation of hair follicle stem cells and epithelial tumor formation Rep 2007; 18: 1225–1230. in mouse skin. Mol Cell Biol 2010; 30: 2518–2536. 41 Park SY, Kwon HJ, Choi Y, Lee HE, Kim SW, Kim JH et al. Distinct patterns of 65 Niini T, Kanerva J, Vettenranta K, Saarinen-Pihkala UM, Knuutila S. AML1 gene promoter CpG island methylation of breast cancer subtypes are associated with amplification: a novel finding in childhood acute lymphoblastic leukemia. stem cell phenotypes. Mod Pathol 2012; 25: 185–196. Haematologica 2000; 85: 362–366. 42 Park SY, Kwon HJ, Lee HE, Ryu HS, Kim SW, Kim JH et al. Promoter CpG island 66 Harewood L, Robinson H, Harris R, Al-Obaidi MJ, Jalali GR, Martineau M et al. hypermethylation during breast cancer progression. Virchows Arch 2011; 458: Amplification of AML1 on a duplicated in acute lymphoblastic 73–84. leukemia: a study of 20 cases. Leukemia 2003; 17: 547–553. 43 Cheng AS, Culhane AC, Chan MW, Venkataramu CR, Ehrich M, Nasir A et al. 67 Robinson HM, Broadfield ZJ, Cheung KL, Harewood L, Harris RL, Jalali GR et al. Epithelial progeny of estrogen-exposed breast progenitor cells display a cancer- Amplification of AML1 in acute lymphoblastic leukemia is associated with a poor like methylome. Cancer Res 2008; 68: 1786–1796. outcome. Leukemia 2003; 17: 2249–2250. 44 Kouros-Mehr H, Werb Z. Candidate regulators of mammary branching mor- 68 Cameron ER, Blyth K, Hanlon L, Kilbey A, Mackay N, Stewart M et al. The Runx phogenesis identified by genome-wide transcript analysis. Dev Dyn 2006; 235: genes as dominant oncogenes. Blood Cells Mol Dis 2003; 30: 194–200. 3404–3412. 69 Planaguma J, Diaz-Fuertes M, Gil-Moreno A, Abal M, Monge M, Garcia A et al. 45 Shore P. A role for Runx2 in normal mammary gland and breast cancer bone A differential gene expression profile reveals overexpression of RUNX1/AML1 metastasis. J Cell Biochem 2005; 96: 484–489. in invasive endometrioid carcinoma. Cancer Res 2004; 64: 8846–8853. 46 Inman CK, Shore P. The osteoblast transcription factor Runx2 is expressed in 70 Doll A, Gonzalez M, Abal M, Llaurado M, Rigau M, Colas E et al. An orthotopic mammary epithelial cells and mediates osteopontin expression. J Biol Chem mouse model demonstrates a role for RUNX1 in distant 2003; 278: 48684–48689. metastasis. Int J Cancer 2009; 125: 257–263. 47 Pratap J, Galindo M, Zaidi SK, Vradii D, Bhat BM, Robinson JA et al. Cell growth 71 Planaguma J, Liljestrom M, Alameda F, Butzow R, Virtanen I, Reventos J et al. regulatory role of Runx2 during proliferative expansion of preosteoblasts. Cancer Matrix metalloproteinase-2 and matrix metalloproteinase-9 codistribute with Res 2003; 63: 5357–5362. transcription factors RUNX1/AML1 and ETV5/ERM at the invasive front of 48 Galindo M, Pratap J, Young DW, Hovhannisyan H, Im HJ, Choi JY et al. The bone- endometrial and ovarian carcinoma. Hum Pathol 2011; 42: 57–67. specific expression of Runx2 oscillates during the cell cycle to support a 72 Tsunematsu T, Kudo Y, Iizuka S, Ogawa I, Fujita T, Kurihara H et al. RUNX3 has an G1-related antiproliferative function in osteoblasts. J Biol Chem 2005; 280: oncogenic role in head and neck cancer. PLoS One 2009; 4: e5892. 20274–20285. 73 Salto-Tellez M, Peh BK, Ito K, Tan SH, Chong PY, Han HC et al. RUNX3 protein is 49 Young DW, Hassan MQ, Pratap J, Galindo M, Zaidi SK, Lee SH et al. Mitotic overexpressed in human basal cell carcinomas. Oncogene 2006; 25: 7646–7649. occupancy and lineage-specific transcriptional control of rRNA genes by Runx2. 74 Nevadunsky NS, Barbieri JS, Kwong J, Merritt MA, Welch WR, Berkowitz RS et al. Nature 2007; 445: 442–446. RUNX3 protein is overexpressed in human epithelial ovarian cancer. Gynecol 50 Baniwal SK, Khalid O, Sir D, Buchanan G, Coetzee GA, Frenkel B. Repression of Oncol 2009; 112: 325–330. Runx2 by (AR) in osteoblasts and prostate cancer cells: AR 75 Blyth K, Vaillant F, Hanlon L, Mackay N, Bell M, Jenkins A et al. Runx2 and MYC binds Runx2 and abrogates its recruitment to DNA. Mol Endocrinol 2009; 23: collaborate in lymphoma development by suppressing apoptotic and growth 1203–1214. arrest pathways in vivo. Cancer Res 2006; 66: 2195–2201.

& 2013 Macmillan Publishers Limited Oncogene (2013) 2121 – 2130 RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2128 76 Kuo YH, Zaidi SK, Gornostaeva S, Komori T, Stein GS, Castilla LH. Runx2 induces tumor cells depend on cell phenotype and the tumor microenvironment. in cooperation with Cbfbeta-SMMHC in mice. Blood Neoplasia 2010; 12: 192–205. 2009; 113: 3323–3332. 101 Yang S, Lim M, Pham LK, Kendall SE, Reddi AH, Altieri DC et al. Bone morpho- 77 Lau CC, Harris CP, Lu XY, Perlaky L, Gogineni S, Chintagumpala M et al. Frequent genetic protein 7 protects prostate cancer cells from stress-induced apoptosis amplification and rearrangement of chromosomal bands 6p12- and 17p11.2 via both Smad and c-Jun NH2-terminal kinase pathways. Cancer Res 2006; 66: in osteosarcoma. Genes Cancer 2004; 39: 11–21. 4285–4290. 78 Forus A, Weghuis DO, Smeets D, Fodstad O, Myklebost O, Geurts van Kessel A. 102 Alarmo EL, Parssinen J, Ketolainen JM, Savinainen K, Karhu R, Kallioniemi A. Comparative genomic hybridization analysis of human sarcomas: II. Identifica- BMP7 influences proliferation, migration, and invasion of breast cancer cells. tion of novel amplicons at 6p and 17p in . Genes Chromosomes Cancer Lett 2009; 275: 35–43. Cancer 1995; 14: 15–21. 103 Barnes GL, Hebert KE, Kamal M, Javed A, Einhorn TA, Lian JB et al. Fidelity of 79 Sase T, Suzuki T, Miura K, Shiiba K, Sato I, Nakamura Y et al. Runt-related tran- Runx2 activity in breast cancer cells is required for the generation of metastases- scription factor 2 (RUNX2) in human colon carcinoma: a potent prognostic factor associated osteolytic disease. Cancer Res 2004; 64: 4506–4513. associated with estrogen receptor. Int J Cancer (e-pub ahead of print 7 March 104 Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS et al. The Runx2 2012; doi:10.1002/ijc.27525). osteogenic transcription factor regulates matrix metalloproteinase 9 in bone 80 Kayed H, Jiang X, Keleg S, Jesnowski R, Giese T, Berger MR et al. Regulation and metastatic cancer cells and controls cell invasion. Mol Cell Biol 2005; 25: functional role of the Runt-related transcription factor-2 in pancreatic cancer. 8581–8591. Br J Cancer 2007; 97: 1106–1115. 105 Barnes GL, Javed A, Waller SM, Kamal MH, Hebert KE, Hassan MQ et al. Osteo- 81 Endo T, Ohta K, Kobayashi T. Expression and function of Cbfa-1/Runx2 in thyroid blast-related transcription factors Runx2 (Cbfa1/AML3) and MSX2 mediate the papillary carcinoma cells. J Clin Endocrinol Metab 2008; 93: 2409–2412. expression of bone sialoprotein in human metastatic breast cancer cells. Cancer 82 Lim M, Zhong C, Yang S, Bell AM, Cohen MB, Roy-Burman P. Runx2 regulates Res 2003; 63: 2631–2637. survivin expression in prostate cancer cells. Lab Invest 2010; 90: 222–233. 106 Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: a transcriptional 83 Hsu YL, Huang MS, Yang CJ, Hung JY, Wu LY, Kuo PL. Lung tumor-associated activator of osteoblast differentiation. Cell 1997; 89: 747–754. osteoblast-derived bone morphogenetic protein-2 increased epithelial-to- 107 Mendoza-Villanueva D, Zeef L, Shore P. Metastatic breast cancer cells inhibit mesenchymal transition of cancer by Runx2/Snail signaling pathway. J Biol Chem osteoblast differentiation through the Runx2/CBFbeta-dependent expression of 2011; 286: 37335–37346. the Wnt antagonist, sclerostin. Breast Cancer Res 2011; 13: R106. 84 Vasko V, Espinosa AV, Scouten W, He H, Auer H, Liyanarachchi S et al. Gene 108 Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nat expression and functional evidence of epithelial-to-mesenchymal transition in Rev Cancer 2011; 11: 411–425. papillary thyroid carcinoma invasion. Proc Natl Acad Sci US A 2007; 104: 2803–2808. 109 Roodman GD. Mechanisms of bone metastasis. NEngJMed2004; 350: 85 Leong DT, Lim J, Goh X, Pratap J, Pereira BP, Kwok HS et al. Cancer-related 1655–1664. ectopic expression of the bone-related transcription factor RUNX2 in non-oss- 110 Baniwal SK, Shah PK, Shi Y, Haduong JH, Declerck YA, Gabet Y et al. Runx2 eous metastatic tumor cells is linked to cell proliferation and motility. Breast promotes both osteoblastogenesis and novel osteoclastogenic signals in ST2 Cancer Res 2010; 12: R89. mesenchymal progenitor cells. Osteoporos Int 2012; 23: 1399–1413. 86 Chimge NO, Baniwal SK, Little GH, Chen YB, Kahn M, Tripathy D et al. Regulation 111 Liu W, Toyosawa S, Furuichi T, Kanatani N, Yoshida C, Liu Y et al. Overexpression of breast cancer metastasis by Runx2 and estrogen signaling: role of SNAI2. of Cbfa1 in osteoblasts inhibits osteoblast maturation and causes osteopenia Breast Cancer Res 2011; 13: R127. with multiple fractures. J Cell Biol 2001; 155: 157–166. 87 Onodera Y, Miki Y, Suzuki T, Takagi K, Akahira J, Sakyu T et al. Runx2 in human 112 Geoffroy V, Kneissel M, Fournier B, Boyde A, Matthias P. High bone resorption in breast carcinoma: its potential roles in cancer progression. Cancer Sci 2010; 101: adult aging transgenic mice overexpressing cbfa1/runx2 in cells of the osteo- 2670–2675. blastic lineage. Mol Cell Biol 2002; 22: 6222–6233. 88 Koeneman KS, Yeung F, Chung LW. Osteomimetic properties of prostate cancer 113 Frenkel B, Hong A, Baniwal SK, Coetzee GA, Ohlsson C, Khalid O et al. Regulation cells: a hypothesis supporting the predilection of prostate cancer metastasis and of adult bone turnover by sex . J Cell Physiol 2010; 224: 305–310. growth in the bone environment. Prostate 1999; 39: 246–261. 114 Maruyama Z, Yoshida CA, Furuichi T, Amizuka N, Ito M, Fukuyama R et al. Runx2 89 Coleman RE. Clinical features of metastatic bone disease and risk of skeletal determines bone maturity and turnover rate in postnatal bone development morbidity. Clin Cancer Res 2006; 12: 6243–6249. and is involved in bone loss in estrogen deficiency. Dev Dyn 2007; 236: 90 Carlinfante G, Vassiliou D, Svensson O, Wendel M, Heinegard D, Andersson G. 1876–1890. Differential expression of osteopontin and bone sialoprotein in bone metastasis 115 He N, Xiao Z, Yin T, Stubbs J, Li L, Quarles LD. Inducible expression of Runx2 of breast and prostate carcinoma. Clin Exp Metastasis 2003; 20: 437–444. results in multiorgan abnormalities in mice. J Cell Biochem 2011; 112: 653–665. 91 Thalmann GN, Sikes RA, Devoll RE, Kiefer JA, Markwalder R, Klima I et al. 116 Platet N, Cunat S, Chalbos D, Rochefort H, Garcia M. Unliganded and liganded Osteopontin: possible role in prostate cancer progression. Clin Cancer Res 1999; estrogen receptors protect against cancer invasion via different mechanisms. 5: 2271–2277. Mol Endocrinol 2000; 14: 999–1009. 92 Chen G, Sircar K, Aprikian A, Potti A, Goltzman D, Rabbani SA. Expression of 117 Ellis MJ, Gao F, Dehdashti F, Jeffe DB, Marcom PK, Carey LA et al. Lower-dose vs RANKL/RANK/OPG in primary and metastatic human prostate cancer as markers high-dose oral estradiol therapy of -positive, aromatase inhi- of disease stage and functional regulation. Cancer 2006; 107: 289–298. bitor-resistant advanced breast cancer: a phase 2 randomized study. Jama 2009; 93 Patani N, Jouhra F, Jiang W, Mokbel K. Osteopontin expression profiles predict 302: 774–780. pathological and clinical outcome in breast cancer. Anticancer Res 2008; 28: 118 Maynadier M, Ramirez JM, Cathiard AM, Platet N, Gras D, Gleizes M et al. 4105–4110. Unliganded estrogen receptor alpha inhibits breast cancer cell growth through 94 Cross SS, Harrison RF, Balasubramanian SP, Lippitt JM, Evans CA, Reed MW et al. interaction with a cyclin-dependent kinase inhibitor (p21(WAF1)). Faseb J 2008; Expression of receptor activator of nuclear factor kappabeta ligand (RANKL) and 22: 671–681. tumour necrosis factor related, apoptosis inducing ligand (TRAIL) in breast 119 Ye Y, Xiao Y, Wang W, Yearsley K, Gao JX, Shetuni B et al. ERalpha cancer, and their relations with osteoprotegerin, oestrogen receptor, and clin- signaling through slug regulates E-cadherin and EMT. Oncogene 2010; 29: icopathological variables. J Clin Pathol 2006; 59: 716–720. 1451–1462. 95 Bellahcene A, Merville MP, Castronovo V. Expression of bone sialoprotein, a bone 120 Zhang XH, Wang Q, Gerald W, Hudis CA, Norton L, Smid M et al. Latent bone matrix protein, in human breast cancer. Cancer Res 1994; 54: 2823–2826. metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell 96 Tsoli E, Tsantoulis PK, Papalambros A, Perunovic B, England D, Rawlands DA et al. 2009; 16: 67–78. Simultaneous evaluation of maspin and CXCR4 in patients with breast cancer. 121 Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon-Cardo C et al. J Clin Pathol 2007; 60: 261–266. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 97 Brubaker KD, Vessella RL, Brown LG, Corey E. Prostate cancer expression of runt- 2003; 3: 537–549. domain transcription factor Runx2, a key regulator of osteoblast differentiation 122 Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD et al. Genes that mediate and function. Prostate 2003; 56: 13–22. breast cancer metastasis to lung. Nature 2005; 436: 518–524. 98 DasK,LeongDT,GuptaA,ShenL,PuttiT,SteinGSet al. Positive association between 123 Hess KR, Pusztai L, Buzdar AU, Hortobagyi GN. Estrogen receptors and distinct nuclear Runx2 and oestrogen- receptor gene expression characterises patterns of breast cancer relapse. Breast Cancer Res Treat 2003; 78: 105–118. a biological subtype of breast cancer. Eur J Cancer 2009; 45: 2239–2248. 124 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA et al. Molecular 99 Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat portraits of human breast tumours. Nature 2000; 406: 747–752. Rev Cancer 2008; 8: 61–70. 125 Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A et al. Repeated 100 Morrissey C, Brown LG, Pitts TE, Vessella RL, Corey E. Bone morphogenetic observation of breast tumor subtypes in independent gene expression data sets. protein 7 is expressed in prostate cancer metastases and its effects on prostate Proc Natl Acad Sci USA 2003; 100: 8418–8423.

Oncogene (2013) 2121 – 2130 & 2013 Macmillan Publishers Limited RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2129 126 Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T et al. Supervised 151 Harada H, Harada Y, Tanaka H, Kimura A, Inaba T. Implications of somatic risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 2009; 27: mutations in the AML1 gene in radiation-associated and therapy-related mye- 1160–1167. lodysplastic syndrome/acute myeloid leukemia. Blood 2003; 101: 673–680. 127 Spender LC, Whiteman HJ, Karstegl CE, Farrell PJ. Transcriptional cross-regulation 152 Harada H, Harada Y, Niimi H, Kyo T, Kimura A, Inaba T. High incidence of of RUNX1 by RUNX3 in human B cells. Oncogene 2005; 24: 1873–1881. somatic mutations in the AML1/RUNX1 gene in myelodysplastic syndrome and 128 Brady G, Whiteman HJ, Spender LC, Farrell PJ. Downregulation of RUNX1 by low blast percentage myeloid leukemia with myelodysplasia. Blood 2004; 103: RUNX3 requires the RUNX3 VWRPY sequence and is essential for Epstein-Barr 2316–2324. virus-driven B-cell proliferation. J Virol 2009; 83: 6909–6916. 153 Steensma DP, Gibbons RJ, Mesa RA, Tefferi A, Higgs DR. Somatic point 129 Smith N, Dong Y, Lian JB, Pratap J, Kingsley PD, van Wijnen AJ et al. Overlapping mutations in RUNX1/CBFA2/AML1 are common in high-risk myelodysplastic expression of Runx1(Cbfa2) and Runx2(Cbfa1) transcription factors supports syndrome, but not in myelofibrosis with myeloid metaplasia. Eur J Haematol cooperative induction of skeletal development. J Cell Physiol 2005; 203: 133–143. 2005; 74: 47–53. 130 Pencovich N, Jaschek R, Tanay A, Groner Y. Dynamic combinatorial interactions 154 Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G et al. of RUNX1 and cooperating partners regulates megakaryocytic differentiation in Clinical effect of point mutations in myelodysplastic syndromes. N Eng J Med cell line models. Blood 2011; 117: e1–14. 2011; 364: 2496–2506. 131 Bae SC, Ito Y. Regulation mechanisms for the heterodimeric transcription factor, 155 Dicker F, Haferlach C, Sundermann J, Wendland N, Weiss T, Kern W et al. PEBP2/CBF. Histol Histopatho 1999; 14: 1213–1221. Mutation analysis for RUNX1, MLL-PTD, FLT3-ITD, NPM1 and NRAS in 269 132 Wotton S, Terry A, Kilbey A, Jenkins A, Herzyk P, Cameron E et al. Gene array patients with MDS or secondary AML. Leukemia 2010; 24: 1528–1532. analysis reveals a common Runx transcriptional programme controlling cell 156 Kohlmann A, Grossmann V, Klein HU, Schindela S, Weiss T, Kazak B et al. Next- adhesion and survival. Oncogene 2008; 27: 5856–5866. generation sequencing technology reveals a characteristic pattern of molecular 133 van der Deen M, Akech J, Lapointe D, Gupta S, Young DW, Montecino MA et al. mutations in 72.8% of chronic myelomonocytic leukemia by detecting frequent Genomic promoter occupancy of runt-related transcription factor RUNX2 in alterations in TET2, CBL, RAS, and RUNX1. J Clin Oncol 2010; 28: 3858–3865. osteosarcoma cells identifies genes involved in cell adhesion and motility. J Biol 157 Romana SP, Poirel H, Leconiat M, Flexor MA, Mauchauffe M, Jonveaux P et al. Chem 2012; 287: 4503–4517. High frequency of t(12;21) in childhood B-lineage acute lymphoblastic leukemia. 134 Ducy P, Starbuck M, Priemel M, Shen J, Pinero G, Geoffroy V et al. A Cbfa1- Blood 1995; 86: 4263–4269. dependent genetic pathway controls bone formation beyond embryonic 158 Douet-Guilbert N, Morel F, Le Bris MJ, Herry A, Le Calvez G, Marion V et al. A development. Genes Dev 1999; 13: 1025–1036. fluorescence in situ hybridization study of TEL-AML1 fusion gene in B-cell acute 135 Li D, Sinha KK, Hay MA, Rinaldi CR, Saunthararajah Y, Nucifora G. RUNX1-RUNX1 lymphoblastic leukemia (1984-2001). Cancer Genet Cytogenet 2003; 144: homodimerization modulates RUNX1 activity and function. J Biol Chem 2007; 143–147. 282: 13542–13551. 159 Golub TR, Barker GF, Bohlander SK, Hiebert SW, Ward DC, Bray-Ward P et al. 136 Mendoza-Villanueva D, Deng W, Lopez-Camacho C, Shore P. The Runx tran- Fusion of the TEL gene on 12p13 to the AML1 gene on 21q22 in acute lym- scriptional co-activator, CBFbeta, is essential for invasion of breast cancer cells. phoblastic leukemia. Proc Natl Acad Sci USA 1995; 92: 4917–4921. Mol Cancer 2010; 9: 171. 160 Hiebert SW, Sun W, Davis JN, Golub T, Shurtleff S, Buijs A et al. The t(12;21) 137 Rowley JD. Identificaton of a translocation with quinacrine fluorescence in a translocation converts AML-1B from an activator to a repressor of transcription. patient with acute leukemia. Ann Genet 1973; 16: 109–112. Mol Cell Biol 1996; 16: 1349–1355. 138 Miyoshi H, Shimizu K, Kozu T, Maseki N, Kaneko Y, Ohki M. t(8;21) breakpoints on 161 Michaud J, Wu F, Osato M, Cottles GM, Yanagida M, Asou N et al. In vitro analyses chromosome 21 in acute myeloid leukemia are clustered within a limited region of known and novel RUNX1/AML1 mutations in dominant familial platelet dis- of a single gene, AML1. Proc Natl Acad Sci USA 1991; 88: 10431–10434. order with predisposition to acute myelogenous leukemia: implications for 139 Lam K, Zhang DE. RUNX1 and RUNX1-ETO: roles in hematopoiesis and leuke- mechanisms of pathogenesis. Blood 2002; 99: 1364–1372. mogenesis. Front Biosci 2012; 17: 1120–1139. 162 Tang JL, Hou HA, Chen CY, Liu CY, Chou WC, Tseng MH et al. AML1/RUNX1 140 De Braekeleer E, Douet-Guilbert N, Le Bris MJ, Morel F, Ferec C, De Braekeleer M. mutations in 470 adult patients with de novo acute myeloid leukemia: prog- RUNX1-MTG16 fusion gene in acute myeloblastic leukemia with nostic implication and interaction with other gene alterations. Blood 2009; 114: t(16;21)(q24;q22): case report and review of the literature. Cancer Genet 5352–5361. Cytogenet 2008; 185: 47–50. 163 Silva FP, Morolli B, Storlazzi CT, Anelli L, Wessels H, Bezrookove V et al. Identi- 141 Gaidzik VI, Bullinger L, Schlenk RF, Zimmermann AS, Rock J, Paschka P et al. fication of RUNX1/AML1 as a classical tumor suppressor gene. Oncogene 2003; RUNX1 mutations in acute myeloid leukemia: results from a comprehensive 22: 538–547. genetic and clinical analysis from the AML study group. J Clin Oncol 2011; 29: 164 Osato M, Asou N, Abdalla E, Hoshino K, Yamasaki H, Okubo T et al. Biallelic and 1364–1372. heterozygous point mutations in the runt domain of the AML1/PEBP2alphaB 142 Radtke I, Mullighan CG, Ishii M, Su X, Cheng J, Ma J et al. Genomic analysis gene associated with myeloblastic . Blood 1999; 93: 1817–1824. reveals few genetic alterations in pediatric acute myeloid leukemia. Proc Natl 165 Gelsi-Boyer V, Trouplin V, Adelaide J, Aceto N, Remy V, Pinson S et al. Genome Acad Sci USA 2009; 106: 12944–12949. profiling of chronic myelomonocytic leukemia: frequent alterations of RAS and 143 Nucifora G, Begy CR, Kobayashi H, Roulston D, Claxton D, Pedersen-Bjergaard J RUNX1 genes. BMC Cancer 2008; 8: 299. et al. Consistent intergenic splicing and production of multiple transcripts 166 Kuo MC, Liang DC, Huang CF, Shih YS, Wu JH, Lin TL et al. RUNX1 mutations are between AML1 at 21q22 and unrelated genes at 3q26 in (3;21)(q26;q22) frequent in chronic myelomonocytic leukemia and mutations at the C-terminal translocations. Proc Natl Acad Sci USA 1994; 91: 4004–4008. region might predict acute myeloid leukemia transformation. Leukemia 2009; 23: 144 Mitani K, Ogawa S, Tanaka T, Miyoshi H, Kurokawa M, Mano H et al. Generation of 1426–1431. the AML1-EVI-1 fusion gene in the t(3;21)(q26;q22) causes blastic crisis in chronic 167 Slattery ML, Lundgreen A, Herrick JS, Caan BJ, Potter JD, Wolff RK. Associations myelocytic leukemia. EMBO J 1994; 13: 504–510. between genetic variation in RUNX1, RUNX2, RUNX3, MAPK1 and eIF4E and 145 Nucifora G, Rowley JD. AML1 and the 8;21 and 3;21 translocations in acute and riskof colon and rectal cancer: additional support for a TGF-beta-signaling chronic myeloid leukemia. Blood 1995; 86: 1–14. pathway. Carcinogenesis 2011; 32: 318–326. 146 Nucifora G, Begy CR, Erickson P, Drabkin HA, Rowley JD. The 3;21 translocation in 168 Fijneman RJ, Anderson RA, Richards E, Liu J, Tijssen M, Meijer GA et al. Runx1 is a myelodysplasia results in a fusion transcript between the AML1 gene and the tumor suppressor gene in the mouse gastrointestinal tract. Cancer Sci 2012; 103: gene for EAP, a highly conserved protein associated with the Epstein-Barr virus 593–599. small RNA EBER 1. Proc Natl Acad Sci USA 1993; 90: 7784–7788. 169 Sakakura C, Hagiwara A, Miyagawa K, Nakashima S, Yoshikawa T, Kin S et al. 147 Nucifora G, Birn DJ, Espinosa 3rd R, Erickson P, LeBeau MM, Roulston D et al. Frequent downregulation of the runt domain transcription factors RUNX1, Involvement of the AML1 gene in the t(3;21) in therapy-related leukemia and in RUNX3 and their cofactor CBFB in gastric cancer. Int J Cancer 2005; 113: chronic myeloid leukemia in blast crisis. Blood 1993; 81: 2728–2734. 221–228. 148 Hromas R, Shopnick R, Jumean HG, Bowers C, Varella-Garcia M, Richkind K. 170 Wu C, Hu Z, He Z, Jia W, Wang F, Zhou Y et al. Genome-wide association study A novel syndrome of radiation-associated acute myeloid leukemia involving identifies three new susceptibility loci for esophageal squamous-cell carcinoma AML1 gene translocations. Blood 2000; 95: 4011–4013. in Chinese populations. Nat Genet 2011; 43: 679–684. 149 Roulston D, Espinosa 3rd R, Nucifora G, Larson RA, Le Beau MM, Rowley JD. 171 Boonstra JJ, van Marion R, Douben HJ, Lanchbury JS, Timms KM, Abkevich V CBFA2(AML1) translocations with novel partner chromosomes in myeloid leu- et al. Mapping of homozygous deletions in verified esophageal adenocarcinoma kemias: association with prior therapy. Blood 1998; 92: 2879–2885. cell lines and xenografts. Genes Chromosomes Cancer 2012; 51: 272–282. 150 Imai Y, Kurokawa M, Izutsu K, Hangaishi A, Takeuchi K, Maki K et al. Mutations of 172 Huang SP, Lan YH, Lu TL, Pao JB, Chang TY, Lee HZ et al. Clinical significance of the AML1 gene in myelodysplastic syndrome and their functional implications in runt-related transcription factor 1 polymorphism in prostate cancer. BJU Int leukemogenesis. Blood 2000; 96: 3154–3160. 2011; 107: 486–492.

& 2013 Macmillan Publishers Limited Oncogene (2013) 2121 – 2130 RUNX1,2,3 in breast cancer N-O Chimge and B Frenkel 2130 173 Thomas DM, Johnson SA, Sims NA, Trivett MK, Slavin JL, Rubin BP et al. Terminal 183 Wada M, Yazumi S, Takaishi S, Hasegawa K, Sawada M, Tanaka H et al. Frequent osteoblast differentiation, mediated by runx2 and p27KIP1, is disrupted in loss of RUNX3 gene expression in human bile duct and pancreatic cancer cell osteosarcoma. J Cell Biol 2004; 167: 925–934. lines. Oncogene 2004; 23: 2401–2407. 174 Martin JW, Zielenska M, Stein GS, van Wijnen AJ, Squire JA. The role of RUNX2 in 184 Ahlquist T, Lind GE, Costa VL, Meling GI, Vatn M, Hoff GS et al. Gene methylation osteosarcoma oncogenesis. Sarcoma 2011; 2011: 282745. profiles of normal mucosa, and benign and malignant colorectal tumors identify 175 Ito K, Liu Q, Salto-Tellez M, Yano T, Tada K, Ida H et al. RUNX3, a novel tumor early onset markers. Mol Cancer 2008; 7:94. suppressor, is frequently inactivated in gastric cancer by protein mislocalization. 185 Subramaniam MM, Chan JY, Soong R, Ito K, Yeoh KG, Wong R et al. RUNX3 Cancer Res 2005; 65: 7743–7750. inactivation in colorectal polyps arising through different pathways of colonic 176 Lee SH, Kim J, Kim WH, Lee YM. Hypoxic silencing of tumor suppressor carcinogenesis. Am J Gastroenterol 2009; 104: 426–436. RUNX3 by histone modification in gastric cancer cells. Oncogene 2009; 28: 186 Soong R, Shah N, Peh BK, Chong PY, Ng SS, Zeps N et al. The expression of 184–194. RUNX3 in colorectal cancer is associated with disease stage and patient out- 177 Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K et al. Frequent loss come. Br J Cancer 2009; 100: 676–679. of RUNX3 expression by promoter hypermethylation in gastric carcinoma. 187 Ito K, Lim AC, Salto-Tellez M, Motoda L, Osato M, Chuang LS et al. RUNX3 Pathobiology 2004; 71: 137–143. attenuates beta-catenin/T cell factors in intestinal tumorigenesis. Cancer Cell 178 Kim WJ, Kim EJ, Jeong P, Quan C, Kim J, Li QL et al. RUNX3 inactivation by point 2008; 14: 226–237. mutations and aberrant DNA methylation in bladder tumors. Cancer Res 2005; 188 Mueller W, Nutt CL, Ehrich M, Riemenschneider MJ, von Deimling A, van den 65: 9347–9354. Boom D et al. Downregulation of RUNX3 and TES by hypermethylation in glio- 179 Wolff EM, Liang G, Cortez CC, Tsai YC, Castelao JE, Cortessis VK et al. RUNX3 blastoma. Oncogene 2007; 26: 583–593. methylation reveals that bladder tumors are older in patients with a history of 189 Gao F, Huang C, Lin M, Wang Z, Shen J, Zhang H et al. Frequent inactivation of smoking. Cancer Res 2008; 68: 6208–6214. RUNX3 by promoter hypermethylation and protein mislocalization in oral 180 Yan C, Kim YW, Ha YS, Kim IY, Kim YJ, Yun SJ et al. RUNX3 methylation as a squamous cell carcinomas. J Cancer Res Clinical Oncol 2009; 135: 739–747. predictor for disease progression in patients with non-muscle-invasive bladder 190 Shiraha H, Nishina S, Yamamoto K. Loss of runt-related transcription factor 3 cancer. J Surg Oncol 2012; 105: 425–430. causes development and progression of hepatocellular carcinoma. JCell 181 Richiardi L, Fiano V, Vizzini L, De Marco L, Delsedime L, Akre O et al. Promoter Biochem 2011; 112: 745–749. methylation in APC, RUNX3, and GSTP1 and mortality in prostate cancer 191 He L, Zhao X, Wang H, Zhang P, Guo C, Huang C et al. RUNX3 Mediates Sup- patients. J Clin Oncol 2009; 27: 3161–3168. pression of Tumor Growth and Metastasis of Human CCRCC by Regulating Cyclin 182 Sato K, Tomizawa Y, Iijima H, Saito R, Ishizuka T, Nakajima T et al. Related Proteins and TIMP-1. PLoS One 2012; 7:3. Epigenetic inactivation of the RUNX3 gene in lung cancer. Oncol Rep 2006; 15: 192 Han YX, Liang DY. The role of the tumor suppressor RUNX3 in giant cell tumor of 129–135. the bone. Int J Oncol 2012; 40: 673–678.

Oncogene (2013) 2121 – 2130 & 2013 Macmillan Publishers Limited