Skip to main content

Impact of RUNX2 on drug-resistant human pancreatic cancer cells with p53 mutations

Abstract

Background

Despite the remarkable advances in the early diagnosis and treatment, overall 5-year survival rate of patients with pancreatic cancer is less than 10%. Gemcitabine (GEM), a cytidine nucleoside analogue and ribonucleotide reductase inhibitor, is a primary option for patients with advanced pancreatic cancer; however, its clinical efficacy is extremely limited. This unfavorable clinical outcome of pancreatic cancer patients is at least in part attributable to their poor response to anti-cancer drugs such as GEM. Thus, it is urgent to understand the precise molecular basis behind the drug-resistant property of pancreatic cancer and also to develop a novel strategy to overcome this deadly disease.

Review

Accumulating evidence strongly suggests that p53 mutations contribute to the acquisition and/or maintenance of drug-resistant property of pancreatic cancer. Indeed, certain p53 mutants render pancreatic cancer cells much more resistant to GEM, implying that p53 mutation is one of the critical determinants of GEM sensitivity. Intriguingly, runt-related transcription factor 2 (RUNX2) is expressed at higher level in numerous human cancers such as pancreatic cancer and osteosarcoma, indicating that, in addition to its pro-osteogenic role, RUNX2 has a pro-oncogenic potential. Moreover, a growing body of evidence implies that a variety of miRNAs suppress malignant phenotypes of pancreatic cancer cells including drug resistance through the down-regulation of RUNX2. Recently, we have found for the first time that forced depletion of RUNX2 significantly increases GEM sensitivity of p53-null as well as p53-mutated pancreatic cancer cells through the stimulation of p53 family TAp63/TAp73-dependent cell death pathway.

Conclusions

Together, it is likely that RUNX2 is one of the promising molecular targets for the treatment of the patients with pancreatic cancer regardless of their p53 status. In this review article, we will discuss how to overcome the serious drug-resistant phenotype of pancreatic cancer.

Peer Review reports

Background

Pancreatic cancer which is highly metastatic to lymph nodes, liver and the other distal sites, is one of the most lethal malignancies among human cancers with 5-year survival rate less than 10%, and its incidence is gradually increasing worldwide [1]. Although 20% of patients receive surgical resection at diagnosis, the remaining 80% of patients are identified as unresectable due to their late diagnosis [2]. Gemcitabine (GEM), a deoxycytidine analogue, has become a standard chemotherapeutic for the treatment of patients with advanced pancreatic cancer [3]. Unfortunately, its clinical efficacy is extremely limited and the extensive efforts to develop the combination regimes with GEM have resulted in only a minor improvement over the conventional therapies [4]. Therefore, it is urgent to develop novel treatment options against pancreatic cancer, and also to understand the precise molecular mechanisms how pancreatic cancer cells could acquire and maintain GEM-resistant phenotype.

As mentioned above, GEM resistance is a critical issue to be adequately addressed for the better treatment of pancreatic cancer patients. Accumulating evidence strongly suggests that the alterations within KRAS, p53, CDKN2A and SMAD4 are frequently detected in pancreatic cancer tissues, and contribute to the genesis and/or maintenance of their advanced phenotypes including GEM resistance [5]. Among these genetic aberrations, p53 mutations (around 75%) appear in the later stages of pancreatic cancer genesis and development [6, 7]. Since p53, which monitors and ensures the genomic integrity, is an essential molecular barrier against carcinogenesis [8, 9], it is possible that loss of function mutation of p53 leads to the accumulation of genetic damage within pancreatic cancer cells, and thus they might acquire GEM-resistant property as well as metastatic potential.

RUNX2 (also called Osf2/Cbfa1, AML-3 or Pebp2αA), a member of RUNX (runt-related transcription factor) family, has been shown to be one of the major determinants of osteoblast differentiation and bone formation [10]. As expected, RUNX2 transactivates number of pro-osteogenic target genes such as collagen type I, bone alkaline phosphatase, osteopontin and osteocalcin [11]. In addition to its pro-osteogenic role, a growing body of evidence strongly suggests that RUNX2 plays a vital role in tumor initiation, progression, invasion and metastasis. From the clinical point of view, the elevated expression level of RUNX2 has been shown to correlate to poor prognosis of patients with pancreatic cancer or with thyroid cancer [12, 13]. In support of these observations, it has been described that RUNX2 regulates numerous genes implicated in carcinogenesis including MMP9 (matrixmetalloproteinase-9), MMP13 (matrixmetalloproteinase-13), VEGF (vascular endothelial growth factor) and survivin [14,15,16]. Furthermore, Pratap et al. found that RUNX2 promotes invasion of bone metastatic cancer cells through the induction of MMP9, and also stimulates the early events of breast cancer progression [17, 18]. Recently, we have described for the first time that siRNA-mediated knockdown of RUNX2 increases adriamycin (ADR) sensitivity of p53-wild-type osteosarcoma cells through the activation of p53 family-dependent cell death pathway [19, 20]. Our subsequent studies revealed that depletion of RUNX2 improves GEM sensitivity of pancreatic cancer cells regardless of their p53 status [21,22,23].

In this review article, we provide a brief overview of the molecular basis behind drug-resistant phenotype of pancreatic cancer cells, and also describe p53 family-dependent cell death pathway in response to DNA damage. Subsequently, we summarize the current understanding of oncogenic potential of RUNX2 and possible involvement of RUNX2 and various miRNAs in pancreatic cancer. Lastly, we discuss how to overcome the serious drug-resistant phenotype of pancreatic cancer.

Main text

Pancreatic cancer

Pancreatic cancer is ranked as the fourth leading cause of cancer-related death in the world (both in industrial countries as well as nonindustrial ones), and is known to exhibit the worst prognosis among cancers (5-year survival rate is less than 10%) [24, 25]. Its mortality rate is nearly equal to its incidence. Up to 80% of pancreatic cancer deaths take place within the first year of diagnosis. Although surgical resection is the only potentially curative approach against pancreatic cancer, greater than 80% of cases are judged as unresectable at the time of diagnosis due to its locally advanced property and/or metastasis. A highly invasive and metastatic nature of the advanced pancreatic cancer is often responsible for its extremely poor clinical outcome. Therefore, it is urgently required to identify the reliable diagnostic and/or prognostic markers. These biomarkers could be helpful to the accurate detection of pancreatic cancer in the early stage, and the prediction of its biological behavior.

Because of the low chance of successful surgery, chemotherapy is the most common approach to extend the survival time of pancreatic cancer patients. For advanced pancreatic cancer patients, a deoxycytidine analogue termed gemcitabine (GEM) (2′,2′-difluorodeoxycytidine) has been considered to be the first choice as a front-line chemotherapy based on the results of the Phase III trial [26, 27]. The cytotoxicity of GEM relies on its ability to promote cancer cell death. Similar to the related nucleoside analog termed cytarabine (Ara-C) [28], GEM is taken up within pancreatic cancer cells through equilibrative nucleoside transporter-1 (hENT1) [29], and subjected to deoxycytidine kinase (dCK)-mediated phosphorylation to become an active form (dFdCTP). dFdCTP is then incorporated at the end of the elongating DNA strand, terminates DNA replication process, and thereby inducing DNA fragmentation (cancer cell death) [30]. In addition to the advanced pancreatic cancer, GEM is also utilized to treat patients with the other serious diseases such as non-small-lung cell, breast, bladder, gastric and ovarian cancers. However, the response rate of GEM monotherapy is low and the improvement of 5-year survival rate is unsatisfied (less than 6 months) [31]. The efficacy of GEM is less than 20% of treated patients, and almost all the patients eventually become resistant to GEM [31].

Subsequently, the extensive studies to improve the unfavorable clinical outcome of the advanced pancreatic cancer patients by GEM-based combination therapies with the other anti-cancer drugs including fluorouracil, irinotecan, pemetrexed, oxaliplatin, exatecan, cisplatin and capecitabine, have been performed. Unfortunately, most of these combination therapies have failed to obtain much better results than GEM monotherapy [32,33,34]. Recently, it has been shown that the combination of GEM with erlotinib (Erlo) or GEM plus nab-paclitaxelm (nab-PTX) has marginal benefits in survival rate of patients with the advanced pancreatic cancer [35, 36]. In support of these observations, it is well known that pancreatic cancer is intrinsically resistant to GEM or acquires GEM resistance. To override GEM-resistant property of pancreatic cancer and improve clinical outcome of the patients, it is critical to understand the precise molecular mechanisms behind its GEM-resistance, and also to develop more efficient GEM-based treatment options for the patients.

Molecular basis underlying GEM-resistance of pancreatic cancer

As mentioned above, drug resistance is a hallmark of pancreatic cancer, and the poor clinical outcome of the patients is partly due to its drug-resistant phenotype. To date, various hypotheses explaining its drug resistance have been postulated. Firstly, it has been generally accepted that its drug resistance is generated by the aberrant overexpression of P-glycoprotein as well as the other transporters, and thereby reducing the accumulation of anti-cancer drugs inside cancer cells [37, 38]. For example, multidrug-resistance 1 (MDR1/ ABCB1/P-glycoprotein) is a transmembrane glycoprotein of 170 kDa and acts as an ATP-dependent drug-efflux pump. O'Driscoll et al. reported that MDR1 is expressed in the majority of pancreatic cancer tissues, indicative of its important contribution to their drug resistance [39]. Consistent with these observations, Song et al. demonstrated that depletion of MDR1 sensitizes GEM-resistant pancreatic cancer Panc-1 cells to GEM [40]. Intriguingly, Zhang et al. found that Ser/Thr kinase PLK1 is expressed at higher level in pancreatic cancer tissues as compared to normal pancreatic ones, and a potent PLK1 inhibitor DMTC acts synergistically with GEM [41]. Recently, Li et al. demonstrated that PLK1 inhibitor GSK461364A enhances the efficacy of GEM [42]. As described [40], PLK1 augmented c-fos-mediated induction of MDR1, suggesting that PLK1 reduces GEM sensitivity of pancreatic cancer cells through up-regulation of MDR1. Another transporter responsible for drug resistance is MDR-related protein MRP1/2 (ABCC1/2). Lee et al. showed that MRP1 and MRP2 are expressed in 84 and 91% of pancreatic cancer cases [43]. Nath et al. found that a significantly higher expression level of MRP1 is closely associated with GEM-resistant phenotype of pancreatic cancer cells [44].

Since the diffusion of hydrophilic GEM through the plasma membrane lipid bilayer is very slow, the efficient cellular uptake of GEM requires the specialized membrane nucleoside transporter protein(s) [45]. Among the nucleoside transporters, it has been shown that human equilibrative nucleoside transporter 1 (hENT1) plays a major role to facilitate the intracellular uptake of GEM across the plasma membrane [46]. Indeed, ENT1-deficient cells were highly resistant to GEM [45], and forced expression of hENT1 enhanced GEM response of pancreatic cancer cells [47]. In accordance with these results, hENT-positive pancreatic cancer patients had a prolonged survival rate after GEM treatment relative to the patients without detectable hENT [48]. Similarly, Nordh et al. described that the expression level of hENT is a reliable predictive indicator for pancreatic cancer patients treated with GEM [49]. Therefore, it is likely that the defect(s) in hENT-mediated intracellular uptake of GEM renders pancreatic cancer cells resistant to GEM.

Meanwhile, it has been well documented that tumor suppressor p53 is frequently mutated in pancreatic cancer tissues (around 75%) [50], indicating that mutant p53 contributes to the development of GEM-resistant nature of pancreatic cancer. Notably, Florini et al. detected GEM-mediated stabilization of mutant p53 at protein level in pancreatic cancer cells [51]. We have also observed the similar phenomenon [22]. In the next chapter, we would like to describe p53 and its family members.

p53-dependent cell death pathway

p53 is one of the well-studied tumor suppressor genes. Since p53 is able to protect cells from serious DNA damage and thus prevent carcinogenesis, p53 is called “guardian of the genome” [52]. Under the normal conditions, p53 is kept at an extremely low level. Upon cellular stresses such as DNA damage, oncogene activation, hypoxia and telomere shortening, p53 becomes stabilized at protein level, activated through the sequential post-translational modifications and then triggers a cascade of the molecular events which determines cell fate such as cell cycle arrest, cellular senescence and/or cell death [53]. These chemical modifications including phosphorylation and acetylation, which attenuate MDM2 (murine double minute 2 homolog)-mediated degradation of p53, extend the half-life of p53, and thereby increasing the intracellular p53 level. MDM2 with an E3 ubiquitin protein ligase enzymatic activity, catalyzes poly-ubiquitination of p53 and facilitates its rapid degradation through proteasome [54, 55]. Once activated, p53 functions as a sequence-specific transcription factor to transactivate a large number of its downstream target genes implicated in the regulation of the above-mentioned cellular processes. For example, p21WAF1 and 14-3-3σ are involved in the induction of p53-dependent G1/S and G2/M cell cycle arrest (cell survival), respectively. While, p53-mediated transcriptional activation of BAX, PUMA, NOXA and/or p53AIP1 participates in the proper cell death response. Therefore, p53 stands at the crossroad between cell survival and death, which might rely on the intensity of stress signal and/or extent of cellular damage (Fig. 1).

Fig. 1
figure 1

p53-dependent cell death pathway. Upon DNA damage, p53 becomes activated through ATM-mediated phosphorylation, and transactivates pro-arrest p21WAF1 and/or 14-3-3σ as well as pro-apoptotic BAX, NOXA, PUMA and/or p53AIP1. The accumulation of these small mitochondrial proteins promotes mitochondria dysfunction followed by caspase-3 activation, and then cells undergo cell death

p53 is frequently mutated in around 50% of human cancers [56]. The majority of mutations occur within its central core sequence-specific DNA-binding domain with 6 hot spots at codons such as R175, G245, R248, R249, R273 and R282, and result in the production of conformationally aberrant p53 proteins (mutant p53). p53 hot spot mutations account for 30% of all reported ones. These observations indicate that mutant p53 lacks the sequence-specific transactivation capability. Since pro-apoptotic function of wild-type p53 is tightly linked to its sequence-specific transcriptional activity, mutant p53 fails to suppress tumor initiation as well as progression. Of note, most common p53 mutations not only impair its tumor-suppressor function (loss of function) but also confer novel pro-oncogenic potential on p53 (gain of function), which markedly enhances tumor progression and drug resistance [57]. Knock-in mice expressing mutant p53 (R175H and R273H) displayed an accelerated tumor growth, which was more invasive and metastatic relative to that of p53-deficient mice [58]. In addition to R175H and R273H mutants, Hanel et al. found that R248 mutant mice show gain of function in carcinogenesis [59]. These findings strongly suggest that at least certain p53 mutants exhibit gain of function. In support of this notion, it has been demonstrated that mutant p53 has an ability to transactivate various pro-oncogenic genes such as MYC, PDGFR, HGFR, EGFR and MDR1 [60]. Recently, Zhao et al. described that Pontin with an ATPase activity interacts with mutant p53 and facilitates its transcriptional activity [61]. Intriguingly, overexpression of Pontin is detectable in a variety of cancer tissues, and strongly associated with poor prognosis of the patients [62]. In addition to Pontin, PML and Pin1 have been shown to enhance the transcriptional capability of mutant p53 through the direct interaction [63, 64].

In a sharp contrast to wild-type p53 with an extremely short half-life (around 20 min), mutant p53, which escapes from ubiquitin/proteasome-dependent degradation pathway mediated by MDM2, has an extended half-life. It has been shown that molecular chaperone Hsp90 is associated with mutant p53, prevents its degradation, and thus facilitates its accumulation in cancer cells [65]. The strong immunohistochemical positivity for p53 has been employed as a diagnostic indicator for the presence of a p53 mutation [57]. Since the vast majority of p53 mutations occur within its central core sequence-specific DNA-binding domain, mutant p53 retains an intact COOH-terminal oligomerization domain. Therefore, a large amount of mutant p53 forms a hetero-oligomer with wild-type p53 via its oligomerization domain through which mutant p53 displays a dominant-negative behavior against wild-type p53 [66]. This hetero-oligomerization diminishes the tumor-suppressive function of wild-type p53. Emerging evidence suggests that p53 status of cancer cells is closely associated with their sensitivity to anti-cancer drugs [67,68,69]. Together, the gain of function (GOF) and/or loss of function (LOF) mutations of p53 might be one of the possible molecular mechanisms of the serious drug resistance of cancer cells. Considering that one half of cancer cells express wild-type p53 but not mutant p53, the presence of p53 mutation-independent mechanisms, which could disrupt p53-dependent cell death pathway, should also be kept in mind.

MDM2, a negative regulator of p53

As described above, around half of cancer patients carry wild-type p53, raising a question why the patients harboring wild-type p53 sometimes do not respond to the standard chemotherapy. A number of evidence indicates that, apart from mutant p53, the other cellular factors might directly prohibit wild-type p53 and/or disrupt the upstream or downstream p53-dependent cell death pathway. Among them, MDM2 is one of the major negative regulators of p53. MDM2 deficiency promoted p53-dependent cell death in a variety of cells [70]. p53 is infrequently mutated in glioblastoma; however, wild-type p53 remains dysfunctional due to the overexpression of MDM2 [71]. In addition to the stimulation of proteasomal degradation of p53, MDM2 binds to NH2-terminal transactivation domain I (TD1) of p53, strongly prohibits it from serving as a transcriptional activator, and thereby attenuates p53-mediated cell death in response to DNA damage. Alternatively, MDM2 interacts with p53 and drives its sequestration in the cytoplasm. Subcellular localization of p53 is regulated through its ubiquitination status. It has been shown that MDM2-mediated monoubiquitination of p53 induces its nuclear export and polyubiquitination facilitates its proteasomal degradation [72].

Since MDM2 is one of the downstream target genes of p53, its expression is tightly regulated in a p53-dependent manner. Thus, p53 modulates the intracellular expression level of its own negative regulator MDM2 via a feedback loop [73]. As a result of this regulatory system, the amounts of p53 and MDM2 are maintained at extremely low level under the healthy conditions. In contrast to normal cells, MDM2 gene is sometimes amplified and/or aberrantly overexpressed in number of cancers including pancreatic cancer [74, 75]. The overall frequency of MDM2 gene amplification in human cancers is around 7%; however, cancer tissues such as osteosarcoma (16%), soft tissue tumors (31%), hepatocellular carcinoma (44%) and Hodgkin disease (67%) have the higher frequencies of MDM2 gene amplification [76]. It is worth noting that the elevated level of MDM2 expression is significantly associated with poor clinical outcome of the patients with pancreatic cancer [77, 78]. In a good agreement with these observations, the abnormal overexpression of MDM2 caused by its gene amplification and/or transcriptional activation mediated by p53, disrupts the balance between the intracellular amounts of p53 and MDM2, and then promotes tumor development [79]. Wade et al. described that a 2-fold increase in MDM2 expression level is enough to prohibit p53 activation [80]. As expected, shRNA-mediated knockdown of MDM2 suppressed proliferation rate and tumor growth potential of highly metastatic pancreatic cancer cells [81]. Under their experimental conditions, depletion of MDM2 caused an up- and down-regulation of p53 and MMP9, respectively.

Consistent with those results, Kondo et al. have described that MDM2 plays a vital role in the development of resistance to CDDP (cisplatin) in human glioblastoma cells [82]. Suzuki et al. demonstrated that forced expression of MDM2 overrides wild-type p53 and confers ADR (adriamycin) resistance on breast cancer cells [83]. Meijer et al. reported that a small chemical compound termed Nutlin-3, which binds to MDM2 and inhibits its interaction with p53, preferentially enhances drug-sensitivity of wild-type p53-expressing ovarian cancer cells through the accumulation of wild-type p53 [84]. Collectively, MDM2-mediated dysfunction of p53 is one of the primary molecular mechanisms underlying p53 mutation-independent acquisition of chemo-resistance.

p53 family

Following the identification of p53, two independent p53-related nuclear transcription factors such as p73 and p63 have been discovered (p53 family members) [85, 86]. These p53 family members have a similar exon/intron organization and display a remarkable amino acid sequence similarity especially in their central DNA-binding domains (around 63%). Subsequent studies revealed that both p73 and p63 genes encode multiple variants, which are basically divided into two groups such as transcription-competent TA (TAp73 and TAp63) and transcription-deficient ΔN (ΔNp73 and ΔNp63) isoforms. The alternative splicing events produce various TA isoforms with the distinct COOH-terminal portions as well as the different tranactivation potential. While, the alternative promoter usage gives rise to NH2-terminaly truncated ΔN isoforms, which lack the first TA (transactivation) domain. As expected from their differential transactivation potentials, TA and ΔN isoforms have their own physiological functions. As described [87, 88], TAp73- or TAp63-null mice developed spontaneous tumors, indicating that, like p53, TAp73 and TAp63 act as tumor suppressors. On the other hand, ΔNp73- or ΔNp63-deficient mice displayed complex developmental defects in the nervous system or in the epidermis and limbs, respectively [89, 90].

Like p53, TAp73/TAp63 are induced following genotoxic insults, recruited onto p53-responsive elements within the promoter regions of the overlapping set of p53-target pro-apoptotic genes including BAX, PUMA, NOXA and/or p53AIP1, and then efficiently promote cell death [91]. The expression of TAp73 is regulated at both mRNA and protein levels. Lissy et al. revealed that TAp73 is transactivated by E2F-1 in response to cell death stimulus [91]. Basically similar results were also reported from several independent groups, suggesting that E2F-1 triggers cell death through the activation of TAp73-dependent cell death pathway [92,93,94]. Alternatively, several lines of evidence suggest that Sp1 (specificity protein 1) and Nrf-2 (nuclear factor erythroid 2-related factor 2) stimulate TAp73 transcription [95, 96]. By contrast, Fontemaggi et al. found that TAp73 expression is down-regulated by the transcriptional repressor ZEB-1 during differentiation [97]. While, Rossi et al. found that HECT domain-containing Nedd4-like E3 ubiquitin ligase Itch binds to TAp73/TAp63 and promotes their proteolytic degradation through proteasome [98, 99]. According to their results, the expression level of Itch was significantly reduced in response to DNA damage caused by ADR, VP16 or ADR, implying that DNA damage-mediated reduction of Itch contributes at least in part to the increased stability of TAp73/TAp63. Consistent with these observations, Hansen et al. described that silencing of Itch increases drug sensitivity of cancer cells even in the absence of functional p53 [100]. Notably, it has been shown that TAp73/TAp63 are required for p53-dependent cell death in response to DNA damage, whereas TAp73/TAp63 induce DNA damage-mediated cell death without functional p53 [101]. Since p73/p63 are rarely mutated in human cancer tissues [102], TAp73/TAp63 are expressed as the functional wild-type forms, raising a possibility that, instead of wild-type p53, TAp73/TAp63 might trigger DNA damage-mediated cell death in p53-null and/or p53-mutated cancer cells.

Unlike TAp73/TAp63, ΔNp73/ΔNp63 lack the acidic NH2-terminal transactivation domain. As expected, ΔNp73/ΔNp63 fail to specifically transactivate p53-target gene promoters, although they are capable to bind to p53-responsve elements within them. Intriguingly, it has been described that ΔNp73/ΔNp63 exert their own transcriptional activities, which is dependent on two additional transactivation domains located between their COOH-terminal oligomerization domain and SAM domain, and close to the Pro-rich domain [103]. Therefore, ΔNp73/ΔNp63 have an ability to activate their own downstream target gene promoters [104]. For example, Wu et al. found that ΔNp63 activates pro-metastatic Hsp70 gene transcription [105]. Soldevilla et al. reported that ABCB1 and HMGB1 are the putative downstream target genes of ΔNp73 [106]. In addition to the transactivation of pro-oncogenic genes, ΔNp73/ΔNp63 act as the dominant-negative inhibitors against TAp73/TAp63 and wild-type p53 through the formation of the inactive complexes with them or the competition for promoter binding sites [107]. Indeed, ΔNp73 is overexpressed in a variety of cancers including lung, breast, brain, thymus, colon, ovary, skin and prostate cancers [108, 109], and this up-regulation of ΔNp73 has been shown to tightly link to poor prognosis of cancer patients [110]. Leung et al. demonstrated that ΔNp73 contributes to the development of CDDP resistance of ovarian cancer cells through the activation of pro-oncogenic AKT signaling pathway [111]. For ΔNp63, its overexpression was detectable in head and neck, lung, esophagus, bladder, liver and tongue cancers [112, 113], and the increased expression of ΔNp63 has been considered to be unfavorable clinical indicator of patients with melanoma [114]. Rocco et al. found that ΔNp63 attenuates TAp73-dependent cell death pathway, and acts as the major determinant of CDDP sensitivity of head and neck cancer [115]. Together, it is likely that, although mutant p53 and ΔNp73/ΔNp63 have a strong dominant-negative potential against TAp73/TAp63, the response to anti-cancer drugs of cancer cells lacking wild-type p53 might be determined at least in part by functional TAp73/TAp63 (Fig. 2). With these in mind, we have to discuss especially how to override the negative effect of mutant p53 on TAp73/TAp63.

Fig. 2
figure 2

Functional interplay among p53 family members. Mutant p53 inhibits pro-apoptotic wild-type p53, TAp73 and TAp63 through the direct interaction, and thus contributes to the acquisition and/or maintenance of the serious drug-resistant phenotype of malignant tumors

Pro-oncogenic RUNX2

Runt-domain containing transcription factor 2 (also called Osf2/Cbfa1, AML-3 or Pebp2αA) is a member of RUNX family, which is identified by an evolutionary conserved DNA-binding/protein-protein interaction domain called runt-homology domain. In contrast to the other RUNX family members such as RUNX1 and RUNX3 whose mutations are tightly linked to the promotion of leukemia and gastric cancer, respectively [116, 117], the initial studies strongly suggest that RUNX2 acts as a master regulator of osteoblast differentiation and bone development. In support of this notion, RUNX2-deficient mice died shortly after birth due to a complete lack of bone formation with arrest of osteoblast differentiation [118, 119]. During bone development, RUNX2 facilitates the differentiation of mesenchymal stem cells into the osteoblast lineage [120]. As expected, RUNX2 transactivates its downstream target gene promoters implicated in these cellular and developmental processes including collagen type I, bone alkaline phosphatase, osteopontin and osteocalcin. Of note, Liu et al. demonstrated that RUNX2 is a prerequisite for the early stage of osteoblast differentiation, whereas its overexpression attenuates the subsequent osteoblast maturation [121]. Thus, it is indicative that RUNX2 is under the strict transcriptional and/or post-translational regulation during the cellular differentiation as well as development.

Recently, the possible involvement of RUNX2 in tumor initiation/progression has been increasingly recognized depending on the cellular context. In addition to osteogenesis-related genes, RUNX2 has an ability to transactivate its downstream target genes involved in tumor progression, invasion and metastasis such as MMP9, MMP13, VEGF, survivin, IL-8 and TGFβR [14,15,16, 122,123,124]. The extensive expression studies demonstrated that the expression level of RUNX2 is aberrantly elevated in numerous cancer tissues as compared to their corresponding normal ones including pancreatic cancer, breast cancer, prostate cancer and osteosarcoma [125]. Of note, overexpression of RUNX2 has been shown to cause a poor response to chemotherapy of certain cancer cells. For example, RUNX2 gene is sometimes amplified, overexpressed in osteosarcomas and thus has been employed as a reliable marker for the estimation of their chemo-resistance [126, 127]. Roos et al. have revealed that loss of RUNX2 expression increases ADR sensitivity of osteosarcoma cells [128]. Similarly, RUNX2 was aberrantly overexpressed in breast cancer cells and promoted their progression [17, 18, 129]. Consistent with these observations, El-Gendi and Mostafa have described that the expression level of RUNX2 is a potential prognostic indicator for breast cancer [130]. As expected, targeting of RUNX2 suppressed breast cancer progression and bone metastasis [131]. Additionally, it has been shown that the abnormally higher RUNX2 expression level in prostate cancer tissues is positively associated with their stage and aggressiveness [132]. For pancreatic cancer, Kayed et al. found that the increased expression level of RUNX2 correlates to unfavorable prognosis of the patients with this serious disease [12]. Given those findings, it is suggestive that RUNX2 is a potential diagnostic marker and/or therapeutic target of the advanced tumors such as pancreatic cancer.

Growth of solid tumors including pancreatic cancer often depends on angiogenesis with the continuous blood vessel formation, and thus the inhibition of tumor angiogenesis is a potential strategy for cancer treatment [133]. Indeed, numerous studies have shown that the attenuation of tumor angiogenesis prohibits pancreatic cancer growth and metastasis [134,135,136]. Among RUNX2-target gene products as described above, VEGF is the most potent angiogenic cytokine [137]. Through the binding to its receptors (VEGFR-1 and VEGFR-2) whose expression is predominantly restricted to endothelial cells in blood vessels, VEGF triggers a variety of downstream survival and migration pathways, and then induces angiogenesis. For transcriptional regulation of VEGF, a large body of evidence suggests that VEGF is transcriptionally activated by hypoxia-inducible factor (HIF). Hypoxia is a major stimulator of angiogenesis. HIF is a heterodimeric transcription factor composed of an oxygen-sensitive alpha subunit (HIF-1α or HIF-2α) and a constitutively expressed beta subunit (HIF-1β). Under normoxic conditions, HIF-1α and HIF-2α are subjected to a rapid hydroxylation at their proline residues, followed by proteasomal degradation mediated by an E3 ubiquitin ligase VHL (von Hippel-Lindau). By contrast, HIF-1α as well as HIF-2α is stabilized at protein level due to the strong inhibition of its hydroxylation and ubiquitination under hypoxic conditions, forms an active HIF transcriptional complex with its binding partner HIF-1β in cell nucleus and regulates its downstream target gene expression. Since HIF-1β is not affected by oxygen concentration and is expressed in excess, the protein levels of HIF-1α and HIF-2α are responsible for HIF transcriptional activity [138, 139]. As expected from those observations, HIF-1α was highly expressed in several solid tumors, and forced suppression of HIF-1α impaired tumor angiogenesis, growth and metastasis [140, 141].

In addition to HIF transcription complex, Zelzer et al. demonstrated for the first time that RUNX2 directly binds to VEGF promoter region, and stimulates its transcription [142]. According to their results, targeting RUNX2 resulted in a significant loss of VEGF expression in mice, and overexpression of RUNX2 in cultured murine fibroblasts caused an obvious increase in VEGF mRNA level. Alternatively, Lee et al. found that RUNX2 has an ability to increase the protein stability and transcriptional activity of HIF-1α by competing with VHL to block its ubiquitination [143]. From their results, ectopic expression of RUNX2 promoted the nuclear access of HIF-1α, elevated the secretion of VEGF, and augmented the in vitro and in vivo angiogenesis, indicative that RUNX2 acts as a potent inducer of angiogenesis through the enhancement of HIF-1α-dependent transactivation of VEGF. Intriguingly, Rhaman et al have reported that VEGF enhances the protein stability of RUNX2 by blocking its degradation, indicating the presence of a positive feedback loop between VEGF and RUNX2, which might synergistically modulate angiogenesis [144]. In a sharp contrast to RUNX2, it has been shown that RUNX3 is recruited onto the putative RUNX3-binding sites of VEGF promoter, trans-represses its transcription, and suppresses gastric cancer angiogenesis [145]. Peng et al. described that RUNX1 physically interacts with HIF-1α and prohibits its transcriptional activity [146]. Under their experimental conditions, forced expression of RUNX1 resulted in a marked decrease in VEGF mRNA level. These observations raise a possibility that RUNX family members might differentially regulate angiogenesis. Since, among RUNX family proteins, RUNX2 strongly stimulates VEGF-dependent angiogenesis, RUNX2 might be an attractive molecular target for therapies, which seek to repress malignant progression of solid tumors such as pancreatic cancer.

Functional interplay between p53 family and RUNX2

Previously, we have found that tumor-suppressive RUNX3 enhances pro-apoptotic activity of p53 in osteosarcoma-derived U2OS cells exposed to ADR through the stimulation of ATM-dependent phosphorylation of p53 at Ser-15 [147]. Subsequent studies revealed that another RUNX family member RUNX1 facilitates p300-mediated acetylation of p53 at Lys-373/382 and thus augments p53-induced cell death of U2OS cells in response to ADR [148]. We then asked whether there could exist a functional interaction between p53 and the remaining RUNX family member RUNX2. In a sharp contrast to RUNX1 and RUNX3, we have found for the first time that RUNX2 strongly prohibits p53/TAp73-mediated cell death of U2OS cells following ADR exposure. Based on our prior results, RUNX2 was associated with histone deacetylase HDAC6 as well as p53 and impaired its transcriptional and pro-apoptotic activities. In addition to p53, RUNX2 repressed TAp73 transcription and also bound to TAp73 to diminish its pro-apoptotic activity. These observations were in accordance with the recent findings of Roos et al. showing that shRNA-mediated knockdown of RUNX2 increases ADR sensitivity of osteosarcoma cells [128].

As described above, U2OS cells bearing wild-type p53 undergo cell death following DNA damage primarily in a p53-dependent manner. Therefore, it is of interest to ask whether RUNX2 could be also involved in poor drug response of p53-null or p53-mutated cancer cells. To this end, we have employed pancreatic cancer cells in which p53 is frequently mutated. Firstly, Sugimoto et al. revealed that depletion of RUNX2 improves GEM sensitivity of p53-negative pancreatic cancer AsPC-1 cells through the stimulation of TAp63-dependent cell death pathway [21]. Secondary, Nakamura et al. demonstrated that GEM sensitivity of p53-mutated pancreatic cancer MiaPaCa-2 cells is increased by RUNX2 depletion-mediated up-regulation of TAp73 [22]. Recently, we have found that RUNX2 suppresses TAp63 expression and also impairs its pro-apoptotic activity in p53-mutated pancreatic cancer Panc-1 cells [23]. These observations imply that RUNX2 is implicated in poor response to GEM of pancreatic cancer cells lacking functional p53, and TAp73/TAp63 might potentiate GEM-induced cell death of RUNX2-knocked down pancreatic cancer cells instead of wild-type p53 (Fig. 3). Previously, Flores et al. described that p53 requires TAp73 and/or TAp63 for DNA damage-induced cell death, whereas TAp73 or TAp63 is capable to promote cell death in response to DNA damage without functional p53 [101]. Thus, it is possible that RUNX2 gene silencing-mediated up-regulation of TAp63 augments GEM-induced cell death of p53-null AsPC-1 cells. For p53-mutated MiaPaCa-2 and Panc-1 cells, it has been shown that mutant p53 acts as a strong dominant-negative inhibitor against TAp73 and TAp63 [149].

Fig. 3
figure 3

RUNX2 prohibits pro-apoptotic TAp63 in p53-mutated pancreatic cancer cells. RUNX2 collaborates with mutant p53 to inhibitpro-apoptotic TAp63 in pancreatic cancer Panc-1 cells exposed to GEM. In addition to the direct interaction, RUNX2 trans-represses TAp63 transcription

The question is how RUNX2 depletion could partially override the negative effect of mutant p53 on TAp73/TAp63. Recently, Wang et al. reported that, in contrast to RUNX2, low ATF3 (activating transcription factor 3) expression level is significantly associated with poor survival of prostate cancer patients, indicating that ATF3 might serve as a tumor suppressor against prostate cancer [150]. In line with these findings, ATF3 deficiency promoted prostate cancer development in PTEN-knockout mice [151]. Of note, Wei et al. found that ATF3 binds to COOH-terminal portion of mutant p53 and diminishes its pro-oncogenic activity [152]. Forced expression of ATF3 sensitized mutant p53-expressing cancer cells to CDDP or VP16. Further analysis demonstrated that ATF3 disrupts the physical interaction between mutant p53 and TAp63, and thereby facilitates the reactivation of TAp63. In addition to TAp63 reactivation, ATF3 impaired pro-oncogenic activity of mutant p53 through the direct binding. Therefore, it is suggestive that RUNX2 depletion might enhance ATF3 expression and/or activity and thereby augment TAp63-mediated cell death even in the presence of a large amount of mutant p53. However, Gokulnath et al. described that ATF3 is efficiently recruited onto RUNX2 promoter and stimulates its transcription in bone metastatic breast cancer cells, indicating that RUNX2 is a direct downstream target gene of ATF3 [153]. Further studies should be necessary to verify the functional significance of the interaction among RUNX2, ATF3, mutant p53 and TAp63 in the acquisition and/or maintenance of drug-resistant phenotype of pancreatic cancer cells.

Implication of microRNA-mediated down-regulation of RUNX2 in pancreatic cancer

MicroRNAs (miRNAs) are a family of small (20-25 nucleotides in length) and single-stranded non-coding RNAs, which bind to the 3’-untranslated region of their target mRNAs in a sequence-specific manner, and repress their expressions through mRNA degradation and/or translation inhibition [154, 155]. An individual miRNA has a capability to regulate numerous distinct mRNAs. Intriguingly, miRNAs act as either oncogenes or tumor suppressor genes, which might be dependent on their target genes. It has been described that the dysregulated expression of certain miRNAs is closely associated with proliferation rate, invasion potential and chemo-sensitivity of pancreatic cancer cells [156]. For example, Lu et al. found that miR-301a whose expression level is specifically elevated in pancreatic cancer tissues, contributes to the persistent activation of NF-κB-mediated pro-oncogenic signaling pathway [157]. By contrast, Liang et al. demonstrated that miR-33a is capable to increase the sensitivity of pancreatic cancer cells to GEM through the down-regulation of pro-oncogenic AKT/Gsk-3β/β-catenin signaling pathway [158]. Ji et al. reported that miR-34 family members have tumor-suppressive function downstream of p53, and their restoration renders p53-mutated pancreatic cancer cells 2-3-fold more sensitive to GEM [159].

Meanwhile, Huang et al. found the inverse relationship between the expression levels of miR-204/miR-211 and RUNX2 during adipocyte differentiation, and also demonstrated for the first time that miR-204/miR-211 bind to the 3’-untranslated region of RUNX2, and attenuate its expression, indicating that miR-204/miR-211 act as the negative regulators of RUNX2 [160]. In support of their observations, Wu et al. described that miR-30 family members prohibit BMP-2-induced osteoblast differentiation by targeting RUNX2 [161]. It has been shown that the additional miRNAs (miR-23a, miR-34c, miR-133a, miR-135a, miR-205 and miR-217) also attenuate osteogenesis by targeting RUNX2 [162, 163]. In addition to adipocyte and osteoblast differentiation, Saini et al. revealed that ectopic expression of miR-203 impairs the development of metastasis originated from prostate cancer in association with the down-regulation of pro-metastatic genes such as ZEB2, survivin and RUNX2 [164]. van der Deen et al. described that p53-mediated stimulation of miR-34c expression causes a massive decrease in RUNX2 and reduces the metastatic potential of osteosarcoma cells [165]. Considering that the aberrant overexpression of RUNX2 correlates to resistance to chemotherapy [166], it is indicative that miR-34c contributes to the improvement of chemo-sensitivity of drug-resistant osteosarcoma cells through the down-regulation of RUNX2. Moreover, Li et al. found that the lower expression level of miR-23b is associated with worse prognosis of ovarian cancer patients, and miR-23b-induced repression of RUNX2 slow downs ovarian cancer cell proliferation [167]. Recently, Taipaleenmäki et al. showed that malignant phenotypes of breast cancer cells are significantly suppressed by miR-135/miR-203-caused direct reduction of RUNX2. Together, these observations suggest that RUNX2-targeting miRNAs effectively suppress the progression and/or metastasis of various types of aggressive tumors including pancreatic cancer.

Notably, miR-203, which prohibits prostate cancer cell metastasis, has also been shown to reduce migration/invasion capacity of pancreatic cancer cells [168]. Chen et al. reported that miR-204 is highly expressed in normal pancreatic ductal tissues relative to pancreatic cancer tissues, and promotes pancreatic cancer cell death [169]. miRNA profiling studies revealed that miR-205 is down-regulated in GEM-resistant pancreatic cancer cells and metastatic pancreatic cancer tissues [170]. Zhao et al. found that the expression level of miR-217 is significantly lower in pancreatic cancer tissues as compared to that in normal ones, and exogenous miR-217 reduces tumor growth in mouse xenograft models [171]. As mentioned above, these miRNAs such as miR-203, miR-204, miR-205 and miR-217 negatively regulate RUNX2 expression, raising a possibility that miRNA-induced down-regulation of RUNX2 contributes to the suppression of malignant properties of pancreatic cancer cells such as drug resistance (Fig. 4).

Fig. 4
figure 4

RUNX2 is a potential therapeutic target for pancreatic cancer. siRNA- and/or synthetic microRNA-mediated down-regulation of pro-oncogenic RUNX2 augments TAp73/TAp63-dependent cell death pathway, and enhances chemo-sensitivity of pancreatic cancer cells

Conclusions

Since the patients with pancreatic cancer show the worst prognosis despite the extensive therapy, it is urgent to develop a novel strategy to enable its early detection and increase its drug sensitivity. For this purpose, the precise understanding of the biology of pancreatic cancer and also molecular mechanisms how pancreatic cancer cells could acquire and maintain this serious drug-resistant phenotype should be required. An increasing body of evidence has demonstrated that RUNX2 is aberrantly overexpressed in numerous cancer tissues including pancreatic cancer relative to their corresponding normal ones, and its depletion suppresses their malignant phenotypes such as migration, invasion, metastasis and drug resistance. Consistent with these observations, we have demonstrated that siRNA-mediated knockdown of RUNX2 improves GEM sensitivity of various pancreatic cancer cells regardless of their p53 status. According to our results, RUNX2 gene silencing increased GEM sensitivity of p53-null pancreatic cancer AsPC-1 cells [21] as well as p53-mutated pancreatic cancer MiaPaCa-2 and Panc-1 cells [22, 23], raising a possibility that RUNX2 depletion improves GEM sensitivity of pancreatic cancer cells without functional p53. In a sharp contrast to p53, TAp73/TAp63 is rarely mutated in cancer tissues. Of note, it has been shown that pro-apoptotic p53 family TAp73/TAp63 has an ability to promote DNA damage-mediated cell death in the absence of functional p53. Mutant p53 acts as a strong dominant-negative inhibitor against p53 family members; however, RUNX2 depletion-mediated up-regulation of TAp73 and/or TAp63 resulted in an increase in GEM sensitivity of p53-mutated pancreatic cancer cells, which might be at least in part due to the disruption of the intracellular balance between the amounts of mutant p53 and TAp73/TAp63. Furthermore, miRNAs targeting RUNX2 suppress malignant phenotypes of pancreatic cancer cells, suggesting that the delivery of chemically stable synthetic miRNAs to pancreatic cancer tissues is an attractive strategy to treat advanced pancreatic cancer patients. Although it is unknown whether miRNA-mediated down-regulation of RUNX2 could lead to the potentiation of TAp73/TAp63-dependent cell death pathway, RUNX2 silencing-mediated restoration of TAp73/TAp63 and down-regulation of its pro-oncogenic downstream target genes might represent a promising approach to override the serious drug-resistant phenotype of pancreatic cancer with p53 mutation.

Abbreviations

ADR:

Adriamycin

ATF3:

Activating transcription factor 3

ATM:

Ataxia telangiectasia mutated

GEM:

Gemcitabine

HDAC:

Histone deacetylase

hENT1:

Equilibrative nucleoside transporter-1

HIF:

Hypoxia-inducible factor

MDM2:

Murine double minute 2

MDR1:

Multidrug-resistance 1

miRNA:

microRNA

MMP9:

Matrixmetalloproteinase-9

MMP13:

Matrixmetalloproteinase-13

Nrf-2:

Nuclear factor erythroid 2-related factor 2

PLK1:

Polo-like kinase 1

RUNX2:

Runt-related transcription factor 2

siRNA:

Small interfering RNA

shRNA:

Small hairpin RNA

TA:

Transactivation

TD1:

Transactivation domain I

VEGF:

Vascular endothelial growth factor

VHL:

Von Hippel-Lindau

References

  1. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378:607–20.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363:1049–57.

    Article  CAS  PubMed  Google Scholar 

  3. Abbruzzese JL. New applications of gemcitabine and future directions in the management of pancreatic cancer. Cancer. 2002;15:941–5.

    Article  CAS  Google Scholar 

  4. Van CE, Verslype C, Grusenmeyer PA. Lessons learned in the management of advanced pancreatic cancer. J Clin Oncol. 2007;25:1949–52.

    Article  Google Scholar 

  5. Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2006;15:1218–49.

    Article  CAS  Google Scholar 

  6. Boschman CR, Stryker S, Reddy JK, Rao MS. Expression of p53 protein in precursor lesions and adenocarcinoma of human pancreas. Am J Pathol. 1994;145:1291–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Maitra A, Adsay NV, Argani P, Iacobuzio-Donahue C, De Marzo A, Cameron JL, Yeo CJ, Hruban RH. Multicomponent analysis of the pancreatic adenocarcinoma progression model using a pancreatic intraepithelial neoplasia tissue microarray. Mod Pathol. 2003;16:902–12.

    Article  PubMed  Google Scholar 

  8. Vousden KH, Lu X. Live or let die: the cell's response to p53. Nat Rev Cancer. 2002;2:594–604.

    Article  CAS  PubMed  Google Scholar 

  9. Muller PA, Vousden KH. p53 mutations in cancer. Nat Cell Biol. 2013;15:2–8.

    Article  CAS  PubMed  Google Scholar 

  10. Karsenty G, Kronenberg HM, Settembre C. Genetic control of bone formation. Annu Rev Cell Dev Biol. 2009;25:629–48.

    Article  CAS  PubMed  Google Scholar 

  11. Cohen MM Jr. Perspectives on RUNX genes: an update. Am J Med Genet Part A. 2009. 149A: 2629-2646.

  12. Kayed H, Jiang X, Keleg S, Jesnowski R, Giese T, Berger MR, Esposito I, Löhr M, Friess H, Kleeff J. Regulation and functional role of the Runt-related transcription factor-2 in pancreatic cancer. Br J Cancer. 2007;97:1106–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Endo T, Ohta K, Kobayashi T. Expression and function of Cbfa-1/Runx2 in thyroid papillary carcinoma cells. J Clin Endocr Metab. 2008;93:2409–12.

    Article  CAS  PubMed  Google Scholar 

  14. Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA, van Wijnen AJ, Stein JL, Languino LR, Altieri DC, Pratap J, Keller E, Stein GS, Lian JB. Oncogene. 2010;29:811–21.

    Article  CAS  PubMed  Google Scholar 

  15. Mendoza-Villanueva D, Deng W, Lopez-Camacho C, Shore P. The Runx transcriptional co-activator, CBFbeta, is essential for invasion of breast cancer cells. Mol Cancer. 2010;9:171.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Lim M, Zhong C, Yang S, Bell AM, Cohen MB, Roy-Burman P. RUNX2 regulates survivin expression in prostate cancer cells. Lab Invest. 2010;90:222–33.

    Article  CAS  PubMed  Google Scholar 

  17. Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS, Lian JB. The RUNX2 osteogenic transcription factor regulates matrix metalloproteinase 9 in bone metastatic cancer cells and controls cell invasion. Mol Cell Biol. 2005;25:8581–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pratap J, Imbalzano KM, Underwood JM, Cohet N, Gokul K, Akech J, van Wijnen AJ, Stein JL, Imbalzano AN, Nickerson JA, Lian JB, Stein GS. Ectopic RUNX2 expression in mammary epithelial cells disrupts formation of normal acini structure: implications for breast cancer progression. Cancer Res. 2009;69:6807–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ozaki T, Wu D, Sugimoto H, Nagase H, Nakagawara A. Runt-related transcription factor 2 (RUNX2) inhibits p53-dependent apoptosis through the collaboration with HDAC6 in response to DNA damage. Cell Death Dis. 2013;4:e610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ozaki T, Sugimoto H, Nakamura M, Hiraoka K, Yoda H, Sang M, Fujiwara K, Nagase H. Runt-related transcription factor 2 attenuates the transcriptional activity as well as DNA damage-mediated induction of pro-apoptotic TAp73 to regulate chemosensitivity. FEBS J. 2015;282:114–28.

    Article  CAS  PubMed  Google Scholar 

  21. Sugimoto H, Nakamura M, Yoda H, Hiraoka K, Shinohara K, Sang M, Fujiwara K, Shimozato O, Nagase H, Ozaki T. Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death. Cell Death Discov. 2015;1:15010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nakamura M, Sugimoto H, Ogata T, Hiraoka K, Yoda H, Sang M, Sang M, Zhu Y, Yu M, Shimozato O, Ozaki T. Improvement of gemcitabine sensitivity of p53-mutated pancreatic cancer MiaPaCa-2 cells by RUNX2 depletion-mediated augmentation of TAp73-dependent cell death. Oncogenesis. 2016;5:e233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ozaki T, Nakamura M, Ogata T, Sang M, Yoda H, Hiraoka K, Sang M, Shimozato O. Depletion of pro-oncogenic RUNX2 enhances gemcitabine (GEM) sensitivity of p53-mutated pancreatic cancer Panc-1 cells through the induction of pro-apoptotic TAp63. Oncotarget. 2016;7:71937–50.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Sporn MB. The war on cancer. Lancet. 1996;347:1377–81.

    Article  CAS  PubMed  Google Scholar 

  25. Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med. 2013;19:1450–64.

    Article  CAS  PubMed  Google Scholar 

  26. Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, Von Hoff DD. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13.

    Article  CAS  PubMed  Google Scholar 

  27. Louvet C, Labianca R, Hammel P, Lledo G, Zampino MG, Andre T, Zaniboni A, Ducreux M, Aitini E, Taieb J, Faroux R, Lepere C, de GA. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial. J Clin Oncol. 2005;23:3509–16.

    Article  CAS  PubMed  Google Scholar 

  28. Li ZR, Campbell J, Rustum YM. Effect of 3-deazauridine on the metabolism, toxicity, and antitumor activity of azacitidine in mice bearing L1210 leukemia sensitive and resistant to cytarabine. Cancer Treat Rep. 1983;67:547–54.

    CAS  PubMed  Google Scholar 

  29. Farrell JJ, Elsaleh H, Garcia M, Lai R, Ammar A, Regine WF, Abrams R, Benson AB, Macdonald J, Cass CE, Dicker AP, Mackey JR. Human equilibrative nucleoside transporter 1 levels predict response to gemcitabine in patients with pancreatic cancer. Gastroenterol. 2009;136:187–95.

    Article  Google Scholar 

  30. Plunkett W, Huang P, Xu YZ, Heinemann V, Grunewald R, Gandhi V. Gemcitabine: metabolism, mechanisms of action, and self-potentiation. Sem. Oncol. 1995;22:3–10.

    CAS  Google Scholar 

  31. Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, Schramm H, Fahlke J, Zuelke C, Burkart C, Gutberlet K, Kettner E, Schmalenberg H, Weigang-Koehler K, Bechstein WO, Niedergethmann M, Schmidt-Wolf I, Roll L, Doerken B, Riess H. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297:267–77.

    Article  CAS  PubMed  Google Scholar 

  32. Berlin JD, Catalano P, Thomas JP, Kugler JW, Haller DG, Benson AB 3rd. Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol. 2002;20:3270–5.

    Article  CAS  PubMed  Google Scholar 

  33. Rocha Lima CM, Green MR, Rotche R, Miller WH Jr, Jeffrey GM, Cisar LA, Morganti A, Orlando N, Gruia G, Miller LL. Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J Clin Oncol. 2004;22:3776–83.

    Article  CAS  PubMed  Google Scholar 

  34. Heinemann V, Quietzsch D, Gieseler F, Gonnermann M, Schönekäs H, Rost A, Neuhaus H, Haag C, Clemens M, Heinrich B, Vehling-Kaiser U, Fuchs M, Fleckenstein D, Gesierich W, Uthgenannt D, Einsele H, Holstege A, Hinke A, Schalhorn A, Wilkowski R. Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol. 2006;24:3946–52.

    Article  CAS  PubMed  Google Scholar 

  35. Wang Y, Hu GF, Zhang QQ, Tang N, Guo J, Liu LY, Han X, Wang X, Wang ZH. Efficacy and safety of gemcitabine plus erlotinib for locally advanced or metastatic pancreatic cancer: a systematic review and meta-analysis. Drug Des Dev Ther. 2016;10:1961–72.

    Article  Google Scholar 

  36. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. New Engl J Med. 2013;369:1691–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58.

    Article  CAS  PubMed  Google Scholar 

  38. Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer. 2010;10:147–56.

    Article  CAS  PubMed  Google Scholar 

  39. O'Driscoll L, Walsh N, Larkin A, Ballot J, Ooi WS, Gullo G, O'Connor R, Clynes M, Crown J, Kennedy S. MDR1/P-glycoprotein and MRP-1 drug efflux pumps in pancreatic carcinoma. Anticancer Res. 2007;27:2115–20.

    PubMed  Google Scholar 

  40. Song B, Liu XS, Rice SJ, Kuang S, Elzey BD, Konieczny SF, Ratliff TL, Hazbun T, Chiorean EG, Liu X. Plk1 phosphorylation of orc2 and hbo1 contributes to gemcitabine resistance in pancreatic cancer. Mol Cancer Ther. 2013;12:58–68.

    Article  CAS  PubMed  Google Scholar 

  41. Zhang C, Sun X, Ren Y, Lou Y, Zhou J, Liu M, Li D. Validation of Polo-like kinase 1 as a therapeutic target in pancreatic cancer cells. Cancer Biol Ther. 2012;13:1214–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li J, Wang R, Schweickert PG, Karki A, Yang Y, Kong Y, Ahmad N, Konieczny SF, Liu X. Plk1 inhibition enhances the efficacy of gemcitabine in human pancreatic cancer. Cell Cycle. 2016;15:711–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Lee SH, Kim H, Hwang JH, Lee HS, Cho JY, Yoon YS, Han HS. Breast cancer resistance protein expression is associated with early recurrence and decreased survival in resectable pancreatic cancer patients. Pathol Int. 2012;62:167–75.

    Article  PubMed  Google Scholar 

  44. Nath S, Daneshvar K, Roy LD, Grover P, Kidiyoor A, Mosley L, Sahraei M, Mukherjee P. MUC1 induces drug resistance in pancreatic cancer cells via upregulation of multidrug resistance genes. Oncogenesis. 2013;2:e51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mackey JR, Mani RS, Selner M, Mowles D, Young JD, Belt JA, Crawford CR, Cass CE. Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res. 1998;58:4349–57.

    CAS  PubMed  Google Scholar 

  46. Mackey JR, Yao SY, Smith KM, Karpinski E, Baldwin SA, Cass CE, Young JD. Gemcitabine transport in xenopus oocytes expressing recombinant plasma membrane mammalian nucleoside transporters. J Nat Cancer Inst. 1999;91:1876–81.

    Article  CAS  PubMed  Google Scholar 

  47. Pérez-Torras S, García-Manteiga J, Mercadé E, Casado FJ, Carbó N, Pastor-Anglada M, Mazo A. Adenoviral-mediated overexpression of human equilibrative nucleoside transporter 1 (hENT1) enhances gemcitabine response in human pancreatic cancer. Biochem Pharm. 2008;76:322–9.

    Article  PubMed  CAS  Google Scholar 

  48. Spratlin J, Sangha R, Glubrecht D, Dabbagh L, Young JD, Dumontet C, Cass C, Lai R, Mackey JR. The absence of human equilibrative nucleoside transporter 1 is associated with reduced survival in patients with gemcitabine-treated pancreas adenocarcinoma. Clin Cancer Res. 2004;10:6956–61.

    Article  CAS  PubMed  Google Scholar 

  49. Nordh S, Ansari D, Andersson R. hENT1 expression is predictive of gemcitabine outcome in pancreatic cancer: A systematic review. World J Gastroenterol. 2014;20:8482–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Nigro JM, Baker SJ, Preisinger AC, Jessup JM, Hostetter R, Cleary K, Bigner SH, Davidson N, Baylin S, Devilee P, Glover T, Collins FS, Weston A, Modali R, Harris CC, Vogelstein B. Mutations in the p53 gene occur in diverse human tumour types. Nature. 1989;342:705–8.

    Article  CAS  PubMed  Google Scholar 

  51. Fiorini C, Cordani M, Padroni C, Blandino G, Di Agostino S, Donadelli M. Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta. 2015;1853:89–100.

    Article  CAS  PubMed  Google Scholar 

  52. Lane DP. Cancer p53 guardian of the genome. Nature. 1992;358:15–6.

    Article  CAS  PubMed  Google Scholar 

  53. Vogelstein B, Lane DP, Levine AJ. Surfing the p53 network. Nature. 2000;408:307–10.

    Article  CAS  PubMed  Google Scholar 

  54. Kubbutat MH, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature. 1997;387:299–303.

    Article  CAS  PubMed  Google Scholar 

  55. Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387:296–9.

    Article  CAS  PubMed  Google Scholar 

  56. Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 mutations in human cancers. Science. 1991;253:49–53.

    Article  CAS  PubMed  Google Scholar 

  57. Brosh R, Rotter V. When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer. 2009;9:701–13.

    Article  CAS  PubMed  Google Scholar 

  58. Liu DP, Song H, Xu Y. A common gain of function of p53 cancer mutants in inducing genetic instability. Oncogene. 2010;29:949–56.

    Article  CAS  PubMed  Google Scholar 

  59. Hanel W, Marchenko N, Xu S, Yu SX, Weng W, Moll U. Two hot spot mutant p53 mouse models display differential gain of function in tumorigenesis. Cell Death Differ. 2013;20:898–909.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Strano S, Dell'Orso S, Di Agostino S, Fontemaggi G, Sacchi A, Blandino G. Mutant p53: an oncogenic transcription factor. Oncogene. 2007;26:2212–9.

    Article  CAS  PubMed  Google Scholar 

  61. Zhao Y, Zhang C, Yue X, Li X, Liu J, Yu H, Belyi VA, Yang Q, Feng Z, Hu W. Pontin, a new mutant p53-binding protein, promotes gain-of-function of mutant p53. Cell Death Differ. 2015;22:1824–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lauscher JC, Elezkurtaj S, Dullat S, Lipka S, Gröne J, Buhr HJ, Huber O, Kruschewski M. Increased Pontin expression is a potential predictor for outcome in sporadic colorectal carcinoma. Oncol Rep. 2012;28:1619–24.

    Article  PubMed  Google Scholar 

  63. Haupt S, di Agostino S, Mizrahi I, Alsheich-Bartok O, Voorhoeve M, Damalas A, Blandino G, Haupt Y. Promyelocytic leukemia protein is required for gain of function by mutant p53. Cancer Res. 2009;69:4818–26.

    Article  CAS  PubMed  Google Scholar 

  64. Girardini JE, Napoli M, Piazza S, Rustighi A, Marotta C, Radaelli E, Capaci V, Jordan L, Quinlan P, Thompson A, Mano M, Rosato A, Crook T, Scanziani E, Means AR, Lozano G, Schneider C, Del Sal GA. Pin1/mutant p53 axis promotes aggressiveness in breast cancer. Cancer Cell. 2011;20:79–91.

    Article  CAS  PubMed  Google Scholar 

  65. Muller P, Hrstka R, Coomber D, Lane DP, Vojtesek B. Chaperone-dependent stabilization and degradation of p53 mutants. Oncogene. 2008;27:3371–83.

    Article  CAS  PubMed  Google Scholar 

  66. Blagosklonny MV. p53 from complexity to simplicity: mutant p53 stabilization, gain-of-function, and dominant-negative effect. FASEB J. 2000;14:1901–7.

    Article  CAS  PubMed  Google Scholar 

  67. Fan S, el-Deiry WS, Bae I, Freeman J, Jondle D, Bhatia K, Fornace AJ Jr, Magrath I, Kohn KW, O'Connor PM. p53 gene mutations are associated with decreased sensitivity of human lymphoma cells to DNA damaging agents. Cancer Res. 1994;54:5824–30.

    CAS  PubMed  Google Scholar 

  68. Lai SL, Perng RP, Hwang J. p53 gene status modulates the chemosensitivity of non-small cell lung cancer cells. J Biomed Sci. 2000;7:64–70.

    Article  CAS  PubMed  Google Scholar 

  69. Huang Y, Sadee W. Membrane transporters and channels in chemoresistance and -sensitivity of tumor cells. Cancer Lett. 2006;239:168–82.

    Article  CAS  PubMed  Google Scholar 

  70. Montes de Oca Luna R, Wagner DS, Lozano G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature. 1995;378:203–6.

    Article  CAS  PubMed  Google Scholar 

  71. Ohgaki H, Kleihues P. Genetic alterations and signaling pathways in the evolution of gliomas. Cancer Sci. 2009;100:2235–41.

    Article  CAS  PubMed  Google Scholar 

  72. Li M, Brooks CL, Wu-Baer F, Chen D, Baer R, Gu W. Mono- versus polyubiquitination: differential control of p53 fate by Mdm2. Science. 2003;302:1972–5.

    Article  CAS  PubMed  Google Scholar 

  73. Meek DW, Hupp TR. The regulation of MDM2 by multisite phosphorylation—opportunities for molecular-based intervention to target tumours? Sem Cancer Biol. 2010;20(1):9–28.

    Article  CAS  Google Scholar 

  74. Rayburn E, Zhang R, He J, Wang H. MDM2 and human malignancies: expression, clinical pathology, prognostic markers, and implications for chemotherapy. Curr Cancer Drug Targets. 2005;5:27–41.

    Article  CAS  PubMed  Google Scholar 

  75. Rayburn ER, Ezell SJ, Zhang R. Recent advances in validating MDM2 as a cancer target. Anticancer Agents Med Chem. 2009;9:882–903.

    Article  CAS  PubMed  Google Scholar 

  76. Toledo F, Wahl GM. Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer. 2006;6:909–23.

    Article  CAS  PubMed  Google Scholar 

  77. Grochola LF, Taubert H, Greither T, Bhanot U, Udelnow A, Würl P. Elevated transcript levels from the MDM2 P1 promoter and low p53 transcript levels are associated with poor prognosis in human pancreatic ductal adenocarcinoma. Pancreas. 2011;40:265–70.

    Article  CAS  PubMed  Google Scholar 

  78. Sheng W, Dong M, Chen C, Wang Z, Li Y, Wang K, Li Y, Zhou J. Cooperation of Musashi-2, Numb, MDM2, and P53 in drug resistance and malignant biology of pancreatic cancer. FASEB J. in press

  79. Onel K, Cordon-Cardo C. MDM2 and prognosis. Mol Cancer Res. 2004;2:1–8.

    CAS  PubMed  Google Scholar 

  80. Wade M, Wang YV, Wahl GM. The p53 orchestra: Mdm2 and Mdmx set the tone. Trends Cell Biol. 2010;20:299–309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Shi W, Meng Z, Chen Z, Hua Y, Gao H, Wang P, Lin J, Zhou Z, Luo J, Liu L. RNA interference against MDM2 suppresses tumor growth and metastasis in pancreatic carcinoma SW1990HM cells. Mol Cell Biochem. 2014;387:1–8.

    Article  CAS  PubMed  Google Scholar 

  82. Kondo S, Barnett GH, Hara H, Morimura T, Takeuchi J. MDM2 protein confers the resistance of a human glioblastoma cell line to cisplatin-induced apoptosis. Oncogene. 1995;10:2001–6.

    CAS  PubMed  Google Scholar 

  83. Suzuki A, Toi M, Yamamoto Y, Saji S, Muta M, Tominaga T. Role of MDM2 overexpression in doxorubicin resistance of breast carcinoma. Jpn J Cancer Res. 1998;89:221–7.

    Article  CAS  PubMed  Google Scholar 

  84. Meijer A, Kruyt FA, van der Zee AG, Hollema H, Le P, ten Hoor KA, Groothuis GM, Quax WJ, de Vries EG, de Jong S. Nutlin-3 preferentially sensitises wild-type p53-expressing cancer cells to DR5-selective TRAIL over rhTRAIL. Br J Cancer. 2013;109:2685–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, Valent A, Minty A, Chalon P, Lelias JM, Dumont X, Ferrara P, McKeon F, Caput D. Monoallelically expressed gene related to p53 at 1p36, a region frequently deleted in neuroblastoma and other human cancers. Cell. 1997;90:809–19.

    Article  CAS  PubMed  Google Scholar 

  86. Yang A, Kaghad M, Wang Y, Gillett E, Fleming MD, Dötsch V, Andrews NC, Caput D, McKeon F. p63, a p53 homolog at 3q27-29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities. Mol Cell. 1998;2:305–16.

    Article  CAS  PubMed  Google Scholar 

  87. Tomasini R, Tsuchihara K, Wilhelm M, Fujitani M, Rufini A, Cheung CC, Khan F, Itie-Youten A, Wakeham A, Tsao MS, Iovanna JL, Squire J, Jurisica I, Kaplan D, Melino G, Jurisicova A, Mak TW. TAp73 knockout shows genomic instability with infertility and tumor suppressor functions. Genes Dev. 2008;22:2677–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Su X, Chakravarti D, Cho MS, Liu L, Gi YJ, Lin YL, Leung ML, El-Naggar A, Creighton CJ, Suraokar MB, Wistuba I, Flores ER. TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs. Nature. 2010;467:986–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Wilhelm MT, Rufini A, Wetzel MK, Tsuchihara K, Inoue S, Tomasini R, Itie-Youten A, Wakeham A, Arsenian-Henriksson M, Melino G, Kaplan DR, Miller FD, Mak TW. Isoform-specific p73 knockout mice reveal a novel role for delta Np73 in the DNA damage response pathway. Genes Dev. 2010;24:549–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Chakravarti D, Su X, Cho MS, Bui NH, Coarfa C, Venkatanarayan A, Benham AL, Flores González RE, Alana J, Xiao W, Leung ML, Vin H, Chan IL, Aquino A, Müller N, Wang H, Cooney AJ, Parker-Thornburg J, Tsai KY, Gunaratne PH, Flores ER. Induced multipotency in adult keratinocytes through down-regulation of ΔNp63 or DGCR8. Proc Natl Acad Sci USA. 2014;111:E572–1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Lissy NA, Davis PK, Irwin M, Kaelin WG, Dowdy SFA. common E2F-1 and p73 pathway mediates cell death induced by TCR activation. Nature. 2000;407:642–5.

    Article  CAS  PubMed  Google Scholar 

  92. Irwin M, Marin MC, Phillips AC, Seelan RS, Smith DI, Liu W, Flores ER, Tsai KY, Jacks T, Vousden KH, Kaelin WG Jr. Role for the p53 homologue p73 in E2F-1-induced apoptosis. Nature. 2000;407:645–8.

    Article  CAS  PubMed  Google Scholar 

  93. Stiewe T, Pützer BM. Role of the p53-homologue p73 in E2F1-induced apoptosis. Nat Genet. 2000;26:464–9.

    Article  CAS  PubMed  Google Scholar 

  94. Zaika A, Irwin M, Sansome C, Moll UM. Oncogenes induce and activate endogenous p73 protein. J Biol Chem. 2001;276:11310–6.

    Article  CAS  PubMed  Google Scholar 

  95. Logotheti S, Michalopoulos I, Sideridou M, Daskalos A, Kossida S, Spandidos DA, Field JK, Vojtesek B, Liloglou T, Gorgoulis V, Zoumpourlis V. Sp1 binds to the external promoter of the p73 gene and induces the expression of TAp73gamma in lung cancer. FEBS J. 2010;277:3014–27.

    Article  CAS  PubMed  Google Scholar 

  96. Lai J, Nie W, Zhang W, Wang Y, Xie R, Wang Y, Gu J, Xu J, Song W, Yang F, Huang G, Cao P, Guan X. Transcriptional regulation of the p73 gene by Nrf-2 and promoter CpG methylation in human breast cancer. Oncotarget. 2014;5:6909–22.

    PubMed  PubMed Central  Google Scholar 

  97. Fontemaggi G, Gurtner A, Strano S, Higashi Y, Sacchi A, Piaggio G, Blandino G. The transcriptional repressor ZEB regulates p73 expression at the crossroad between proliferation and differentiation. Mol Cell Biol. 2001;21:8461–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Rossi M, De Laurenzi V, Munarriz E, Green DR, Liu YC, Vousden KH, Cesareni G, Melino G. The ubiquitin-protein ligase Itch regulates p73 stability. EMBO J. 2005;24:836–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Rossi M, Aqeilan RI, Neale M, Candi E, Salomoni P, Knight RA, Croce CM, Melino G. The E3 ubiquitin ligase Itch controls the protein stability of p63. Proc Natl Acad Sci U S A. 2006;103:12753–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hansen TM, Rossi M, Roperch JP, Ansell K, Simpson K, Taylor D, Mathon N, Knight RA, Melino G. Itch inhibition regulates chemosensitivity in vitro. Biochem Biophys Res Commun. 2007;36:33–6.

  101. Flores ER, Tsai KY, Crowley D, Sengupta S, Yang A, McKeon F, Jacks T. p63 and p73 are required for p53-dependent apoptosis in response to DNA damage. Nature. 2002;416:560–4.

    Article  CAS  PubMed  Google Scholar 

  102. Melino G, Lu X, Gasco M, Crook T, Knight RA. Functional regulation of p73 and p63: development and cancer. Trends Biochem Sci. 2003;28:663–70.

    Article  CAS  PubMed  Google Scholar 

  103. Helton ES, Zhu J, Chen X. The unique NH2-terminally deleted (DeltaN) residues, the PXXP motif, and the PPXY motif are required for the transcriptional activity of the DeltaN variant of p63. J Biol Chem. 2006;281:2533–42.

    Article  CAS  PubMed  Google Scholar 

  104. Ghioni P, Bolognese F, Duijf PH, Van Bokhoven H, Mantovani R, Guerrini L. Complex transcriptional effects of p63 isoforms: identification of novel activation and repression domains. Mol Cell Biol. 2002;22:8659–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wu G, Osada M, Guo Z, Fomenkov A, Begum S, Zhao M, Upadhyay S, Xing M, Wu F, Moon C, Westra WH, Koch WM, Mantovani R, Califano JA, Ratovitski E, Sidransky D, Trink B. DeltaNp63alpha up-regulates the Hsp70 gene in human cancer. Cancer Res. 2005;65:758–66.

    CAS  PubMed  Google Scholar 

  106. Soldevilla B, Díaz R, Silva J, Campos-Martín Y, Muñoz C, García V, García JM, Peña C, Herrera M, Rodriguez M, Gómez I, Mohamed N, Marques MM, Bonilla F, Domínguez G. Prognostic impact of ΔTAp73 isoform levels and their target genes in colon cancer patients. Clin Cancer Res. 2011;17:6029–39.

    Article  CAS  PubMed  Google Scholar 

  107. Melino G, De Laurenzi V, Vousden KH. p73: Friend or foe in tumorigenesis. Nat Rev Cancer. 2002;2:605–15.

    Article  CAS  PubMed  Google Scholar 

  108. Oswald C, Stiewe T. In good times and bad: p73 in cancer. Cell Cycle. 2008;7:1726–31.

    Article  CAS  PubMed  Google Scholar 

  109. Vilgelm AE, Hong SM, Washington MK, Wei J, Chen H, El-Rifai W, Zaika A. Characterization of DeltaNp73 expression and regulation in gastric and esophageal tumors. Oncogene. 2010;29:5861–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Müller M, Schleithoff ES, Stremmel W, Melino G, Krammer PH, Schilling T. One, two, three--p53, p63, p73 and chemosensitivity. Drug Resist Updat. 2006;9:288–306.

    Article  PubMed  CAS  Google Scholar 

  111. Leung TH, Wong SC, Chan KK, Chan DW, Cheung AN, Ngan HY. The interaction between C35 and ΔNp73 promotes chemo-resistance in ovarian cancer cells. Br J Cancer. 2013;109:965–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Ramsey MR, Wilson C, Ory B, Rothenberg SM, Faquin W, Mills AA, Ellisen LW. FGFR2 signaling underlies p63 oncogenic function in squamous cell carcinoma. J Clin Invest. 2013;123:3525–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Ram Kumar RM, Betz MM, Robl B, Born W, Fuchs B. ΔNp63α enhances the oncogenic phenotype of osteosarcoma cells by inducing the expression of GLI2. BMC Cancer. 2014;14:559.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Matin RN, Chikh A, Chong SL, Mesher D, Graf M, Sanza’ P, Senatore V, Scatolini M, Moretti F, Leigh IM, Proby CM, Costanzo A, Chiorino G, Cerio R, Harwood CA, Bergamaschi D. p63 is an alternative p53 repressor in melanoma that confers chemoresistance and a poor prognosis. J Exp Med. 2013;210:581–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Rocco JW, Leong CO, Kuperwasser N, DeYoung MP, Ellisen LW. p63 mediates survival in squamous cell carcinoma by suppression of p73-dependent apoptosis. Cancer Cell. 2006;9:45–56.

    Article  CAS  PubMed  Google Scholar 

  116. Song WJ, Sullivan MG, Legare RD, Hutchings S, Tan X, Kufrin D, Ratajczak J, Resende IC, Haworth C, Hock R, Loh M, Felix C, Roy DC, Busque L, Kurnit D, Willman C, Gewirtz AM, Speck NA, Bushweller JH, Li FP, Gardiner K, Poncz M, Maris JM, Gilliland DG. Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nat Genet. 1999;23:166–75.

    Article  CAS  PubMed  Google Scholar 

  117. Li QL, Ito K, Sakakura C, Fukamachi H, Ki I, Chi XZ, Lee KY, Nomura S, Lee CW, Han SB, Kim HM, Kim WJ, Yamamoto H, Yamashita N, Yano T, Ikeda T, Itohara S, Inazawa J, Abe T, Hagiwara A, Yamagishi H, Ooe A, Kaneda A, Sugimura T, Ushijima T, Bae SC, Ito Y. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell. 2002;109:113–24.

    Article  CAS  PubMed  Google Scholar 

  118. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M, Sato M, Okamoto R, Kitamura Y, Yoshiki S, Kishimoto T. Cell. 1997;89:755–64.

    Article  CAS  PubMed  Google Scholar 

  119. Otto F, Thornell AP, Crompton T, Denzel A, Gilmour KC, Rosewell IR, Stamp GW, Beddington RS, Mundlos S, Olsen BR, Selby PB, Owen MJ. Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell. 1997;89:765–71.

    Article  CAS  PubMed  Google Scholar 

  120. Komori T. Regulation of bone development and extracellular matrix protein genes by RUNX2. Cell Tissue Res. 2010;339:189–95.

    Article  CAS  PubMed  Google Scholar 

  121. Liu W, Toyosawa S, Furuichi T, Kanatani N, Yoshida C, Liu Y, Himeno M, Narai S, Yamaguchi A, Komori T. Overexpression of Cbfa1 in osteoblasts inhibits osteoblast maturation and causes osteopenia with multiple fractures. J Cell Biol. 2001;155:157–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Blyth K, Cameron ER, Neil JC. The RUNX genes: gain or loss of function in cancer. Nat Rev. 2005;5:376–87.

    Article  CAS  Google Scholar 

  123. Pratap J, Lian JB, Javed A, Barnes GL, van Wijnen AJ, Stein JL, Stein GS. Regulatory roles of Runx2 in metastatic tumor and cancer cell interactions with bone. Cancer Metastasis Rev. 2006;25:589–600.

    Article  CAS  PubMed  Google Scholar 

  124. Chua CW, Chiu YT, Yuen HF, Chan KW, Man K, Wang X, Ling MT, Wong YC. Suppression of androgen-independent prostate cancer cell aggressiveness by FTY720: validating Runx2 as a potential antimetastatic drug screening platform. Clin Cancer Res. 2009;15:4322–35.

    Article  CAS  PubMed  Google Scholar 

  125. Ito Y, Bae SC, Chuang LS. The RUNX family: developmental regulators in cancer. Nat Rev Cancer. 2015;15:81–95.

    Article  CAS  PubMed  Google Scholar 

  126. Man TK, Lu XY, Jaeweon K, Perlaky L, Harris CP, Shah S, Ladanyi M, Gorlick R, Lau CC, Rao PH. Genome-wide array comparative genomic hybridization analysis reveals distinct amplifications in osteosarcoma. BMC Cancer. 2004;4:45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Sadikovic B, Thorner P, Chilton-Macneill S, Martin JW, Cervigne NK, Squire J, Zielenska M. Expression analysis of genes associated with human osteosarcoma tumors shows correlation of RUNX2 overexpression with poor response to chemotherapy. BMC Cancer. 2010;10:202.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Roos A, Satterfield L, Zhao S, Fuja D, Shuck R, Hicks MJ, Donehower LA, Yustein JT. Loss of Runx2 sensitises osteosarcoma to chemotherapy-induced apoptosis. Br J Cancer. 2015;113:1289–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Pratap J, Wixted JJ, Gaur T, Zaidi SK, Dobson J, Gokul KD, Hussain S, van Wijnen AJ, Stein JL, Stein GS, Lian JB. Runx2 transcriptional activation of Indian Hedgehog and a downstream bone metastatic pathway in breast cancer cells. Cancer Res. 2008;68:7795–802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. El-Gendi SM, Mostafa MF. Runx2 Expression as a Potential Prognostic Marker in Invasive Ductal Breast Carcinoma. Pathol Oncol Res. 2016;22:461–70.

    Article  CAS  PubMed  Google Scholar 

  131. Taipaleenmäki H, Browne G, Akech J, Zustin J, van Wijnen AJ, Stein JL, Hesse E, Stein GS, Lian JB. Targeting of Runx2 by miRNA-135 and miRNA-203 impairs progression of breast cancer and metastatic bone disease. Cancer Res. 2015;75:1433–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Dutta A, Li J, Lu H, Akech J, Pratap J, Wang T, Zerlanko BJ, TJ FG, Jiang Z, Birbe R, Wixted J, Violette SM, Stein JL, Stein GS, Lian JB, Languino LR. Integrin αvβ6 promotes an osteolytic program in cancer cells by upregulating MMP2. Cancer Res. 2014;74:1598–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70.

    Article  CAS  PubMed  Google Scholar 

  134. Ishikawa T, Chen J, Wang J, Okada F, Sugiyama T, Kobayashi T, Shindo M, Higashino F, Katoh H, Asaka M, Kondo T, Hosokawa M, Kobayashi M. Adrenomedullin antagonist suppresses in vivo growth of human pancreatic cancer cells in SCID mice by suppressing angiogenesis. Oncogene. 2003;22:1238–42.

    Article  CAS  PubMed  Google Scholar 

  135. Wei D, Wang L, He Y, Xiong HQ, Abbruzzese JL, Xie K. Celecoxib inhibits vascular endothelial growth factor expression in and reduces angiogenesis and metastasis of human pancreatic cancer via suppression of Sp1 transcription factor activity. Cancer Res. 2004;64:2030–8.

    Article  CAS  PubMed  Google Scholar 

  136. Maruyama Y, Ono M, Kawahara A, Yokoyama T, Basaki Y, Kage M, Aoyagi S, Kinoshita H, Kuwano M. Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis. Cancer Res. 2006;66:6233–42.

    Article  CAS  PubMed  Google Scholar 

  137. Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, Yancopoulos GD, Wiegand SJ. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science. 1999;284:1994–8.

    Article  CAS  PubMed  Google Scholar 

  138. Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA. 1995;92:5510–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Kaelin WG Jr, Ratcliffe PJ. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell. 2008;30:393–402.

    Article  CAS  PubMed  Google Scholar 

  140. Maxwell PH, Dachs GU, Gleadle JM, Nicholls LG, Harris AL, Stratford IJ, Hankinson O, Pugh CW, Ratcliffe PJ. Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc Natl Acad Sci USA. 1997;94:8104–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, Buechler P, Isaacs WB, Semenza GL, Simons JW. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59:5830–5.

    CAS  PubMed  Google Scholar 

  142. Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai N, Soker S, Olsen BR. Tissue specific regulation of VEGF expression during bone development requires Cbfa1/Runx2. Mech Dev. 2001;106:97–106.

    Article  CAS  PubMed  Google Scholar 

  143. Lee SH, Che X, Jeong JH, Choi JY, Lee YJ, Lee YH, Bae SC, Lee YM. Runx2 protein stabilizes hypoxia-inducible factor-1α through competition with von Hippel-Lindau protein (pVHL) and stimulates angiogenesis in growth plate hypertrophic chondrocytes. J Biol Chem. 2012;287:14760–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Rahman SU, Lee MS, Baek JH, Ryoo HM, Woo KM. The prolyl hydroxylase inhibitor dimethyloxalylglycine enhances dentin sialophoshoprotein expression through VEGF-induced Runx2 stabilization. PLoS One. 2014;9:e112078.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Peng Z, Wei D, Wang L, Tang H, Zhang J, Le X, Jia Z, Li Q, Xie K. RUNX3 inhibits the expression of vascular endothelial growth factor and reduces the angiogenesis, growth, and metastasis of human gastric cancer. Clin Cancer Res. 2006;12:6386–94.

    Article  CAS  PubMed  Google Scholar 

  146. Peng ZG, Zhou MY, Huang Y, Qiu JH, Wang LS, Liao SH, Dong S, Chen GQ. Physical and functional interaction of Runt-related protein 1 with hypoxia-inducible factor-1alpha. Oncogene. 2008;27:839–47.

    Article  CAS  PubMed  Google Scholar 

  147. Yamada C, Ozaki T, Ando K, Suenaga Y, Inoue K, Ito Y, Okoshi R, Kageyama H, Kimura H, Miyazaki M, Nakagawara A. RUNX3 modulates DNA damage-mediated phosphorylation of tumor suppressor p53 at Ser-15 and acts as a co-activator for p53. J Biol Chem. 2010;285:16693–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Wu D, Ozaki T, Yoshihara Y, Kubo N, Nakagawara A. Runt-related transcription factor 1 (RUNX1) stimulates tumor suppressor p53 protein in response to DNA damage through complex formation and acetylation. J Biol Chem. 2013;288:1353–64.

    Article  CAS  PubMed  Google Scholar 

  149. Gaiddon C, Lokshin M, Ahn J, Zhang T, Prives C. A subset of tumor-derived mutant forms of p53 down-regulate p63 and p73 through a direct interaction with the p53 core domain. Mol Cell Biol. 2001;21:1874–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Wang Z, Kim J, Teng Y, Ding HF, Zhang J, Hai T, Cowell JK, Yan C. Loss of ATF3 promotes hormone-induced prostate carcinogenesis and the emergence of CK5+CK8+ epithelial cells. Oncogene. 2016;35:3555–64.

    Article  CAS  PubMed  Google Scholar 

  151. Wang Z, Xu D, Ding HF, Kim J, Zhang J, Hai T, Yan C. Loss of ATF3 promotes Akt activation and prostate cancer development in a Pten knockout mouse model. Oncogene. 2016;34:4975–84.

    Article  CAS  Google Scholar 

  152. Wei S, Wang H, Lu C, Malmut S, Zhang J, Ren S, Yu G, Wang W, Tang DD, Yan C. The activating transcription factor 3 protein suppresses the oncogenic function of mutant p53 proteins. J Biol Chem. 2014;289:8947–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Gokulnath M, Partridge NC, Selvamurugan N. Runx2, a target gene for activating transcription factor-3 in human breast cancer cells. Tumour Biol. 2015;36:1923–31.

    Article  CAS  PubMed  Google Scholar 

  154. Chitwood DH, Timmermans MC. Small RNAs are on the move. Nature. 2010;467:415–9.

    Article  CAS  PubMed  Google Scholar 

  155. Kosik KS. MicroRNAs and cellular phenotype. Cell. 2010;143:21–6.

    Article  CAS  PubMed  Google Scholar 

  156. Sun T, Kong X, Du Y, Li Z. Aberrant microRNAs in pancreatic cancer: Researches and clinical implications. Gastroenterol Res Pract. 2014;2014:386561.

    PubMed  PubMed Central  Google Scholar 

  157. Lu Z, Li Y, Takwi A, Li B, Zhang J, Conklin DJ, Young KH, Martin R, Li Y. miR-301a as an NF-κB activator in pancreatic cancer cells. EMBO J. 2011;30:57–67.

    Article  CAS  PubMed  Google Scholar 

  158. Liang C, Yu XJ, Guo XZ, Sun MH, Wang Z, Song Y, Ni QX, Li HY, Mukaida N, Li YY. MicroRNA-33a-mediated downregulation of Pim-3 kinase expression renders human pancreatic cancer cells sensitivity to gemcitabine. Oncotarget. 2015;6:14440–55.

    PubMed  PubMed Central  Google Scholar 

  159. Ji Q, Hao X, Zhang M, Tang W, Yang M, Li L, Xiang D, Desano JT, Bommer GT, Fan D, Fearon ER, Lawrence TS, Xu L. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One. 2009;4:e6816.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  160. Huang J, Zhao L, Xing L, Chen D. MicroRNA-204 regulates Runx2 protein expression and mesenchymal progenitor cell differentiation. Stem Cells. 2010;28:357–64.

    PubMed  PubMed Central  Google Scholar 

  161. Wu T, Zhou H, Hong Y, Li J, Jiang X, Huang H. miR-30 family members negatively regulate osteoblast differentiation. J Biol Chem. 2012;287:7503–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Li Z, Hassan MQ, Volinia S, van Wijnen AJ, Stein JL, Croce CM, Lian JB, Stein GS. A microRNA signature for a BMP2-induced osteoblast lineage commitment program. Proc Natl Acad Sci USA. 2008;105:13906–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Zhang Y, Xie RL, Croce CM, Stein JL, Lian JB, van Wijnen AJ, Stein GS. A program of microRNAs controls osteogenic lineage progression by targeting transcription factor Runx2. Proc Natl Acad Sci USA. 2011; 108: 9863–9868.

  164. Saini S, Majid S, Yamamura S, Tabatabai L, Suh SO, Shahryari V, Chen Y, Deng G, Tanaka Y, Dahiya R. Regulatory Role of mir-203 in Prostate Cancer Progression and Metastasis. Clin Cancer Res. 2011;17:5287–98.

    Article  CAS  PubMed  Google Scholar 

  165. van der Deen M, Taipaleenmäki H, Zhang Y, Teplyuk NM, Gupta A, Cinghu S, Shogren K, Maran A, Yaszemski MJ, Ling L, Cool SM, Leong DT, Dierkes C, Zustin J, Salto-Tellez M, Ito Y, Bae SC, Zielenska M, Squire JA, Lian JB, Stein JL, Zambetti GP, Jones SN, Galindo M, Hesse E, Stein GS, van Wijnen AJ. MicroRNA-34c inversely couples the biological functions of the runt-related transcription factor RUNX2 and the tumor suppressor p53 in osteosarcoma. J Biol Chem. 2013;288:21307–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Martin JW, Zielenska M, Stein GS, van Wijnen AJ, Squire JA. The role of RUNX2 in osteosarcoma oncogenesis. Sarcoma. 2011;2011:282745.

    Article  CAS  PubMed  Google Scholar 

  167. Li W, Liu Z, Chen L, Zhou L, Yao Y. MicroRNA-23b is an independent prognostic marker and suppresses ovarian cancer progression by targeting runt-related transcription factor-2. FEBS Lett. 2014;588:1608–15.

    Article  CAS  PubMed  Google Scholar 

  168. Miao L, Xiong X, Lin Y, Cheng Y, Lu J, Zhang J, Cheng N. miR-203 inhibits tumor cell migration and invasion via caveolin-1 in pancreatic cancer cells. Oncol Lett. 2014;7:658–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Chen Z, Sangwan V, Banerjee S, Mackenzie T, Dudeja V, Li X, Wang H, Vickers SM, Saluja AK. miR-204 mediated loss of Myeloid cell leukemia-1 results in pancreatic cancer cell death. Mol Cancer. 2013;12:105.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Singh S, Chitkara D, Kumar V, Behrman SW, Mahato RI. miRNA profiling in pancreatic cancer and restoration of chemosensitivity. Cancer Lett. 2013;334:211–20.

    Article  CAS  PubMed  Google Scholar 

  171. Zhao WG, Yu SN, Lu ZH, Ma YH, Gu YM, Chen J. The miR-217 microRNA functions as a potential tumor suppressor in pancreatic ductal adenocarcinoma by targeting KRAS. Carcinogenesis. 2010;31:1726–33.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Dr. Hiroki Nagase (Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute) for his valuable discussion.

Funding

This work was supported in part by JSPS (MEXT) KAKENHI Grant Number 23501278. The funding body was not involved in the study design, data collection, analysis and interpretation of data, or in writing of this review article.

Availability of data and materials

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

TO provided direction throughout the preparation of this manuscript. TO, DY and MY wrote the manuscript. DS, YZ, YB and MS reviewed and made significant revisions on the manuscript. All authors approved the final manuscript.

Corresponding author

Correspondence to Toshinori Ozaki.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ozaki, T., Yu, M., Yin, D. et al. Impact of RUNX2 on drug-resistant human pancreatic cancer cells with p53 mutations. BMC Cancer 18, 309 (2018). https://doi.org/10.1186/s12885-018-4217-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-018-4217-9

Keywords