Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 1.
Published in final edited form as: Biochim Biophys Acta. 2010 Jun 22;1806(2):258–267. doi: 10.1016/j.bbcan.2010.06.001

Targeting Notch signaling pathway to overcome drug-resistance for cancer therapy

Zhiwei Wang 1,*, Yiwei Li 1, Aamir Ahmad 1, Asfar S Azmi 1, Sanjeev Banerjee 1, Dejuan Kong 1, Fazlul H Sarkar 1,*
PMCID: PMC2955995  NIHMSID: NIHMS217025  PMID: 20600632

Abstract

Chemotherapy is an important therapeutic strategy for cancer treatment and remains the mainstay for the management of human malignancies; however, chemotherapy fails to eliminate all tumor cells because of intrinsic or acquired drug- resistance, which is the most common cause of tumor recurrence. Recently, emerging evidences suggest that Notch signaling pathway is one of the most important signaling pathways in drug-resistant tumor cells. Moreover, down-regulation of Notch pathway could induce drug sensitivity, leading to increased inhibition of cancer cell growth, invasion, and metastasis. This article will provide a brief overview of the published evidences in support of the roles of Notch in drug-resistance, and will further summarize how targeting Notch by “natural agents” could become a novel and safer approach for the improvement of tumor treatment by overcoming drug-resistance.

Keywords: Notch, drug resistance, cancer, EMT

1. Introduction

The Notch signaling pathway is a conserved ligand–receptor signaling pathway that plays critical mechanistic roles in cell proliferation, survival, apoptosis, and differentiation which affects the development and function of many organs [1]. Notch genes encode single-pass transmembrane proteins which can be activated by interacting with a family of its ligands. To date, four Notch receptors have been identified in mammals, including human, such as Notch-1-4. The mammalian canonical ligands are designated as either Delta-like (Delta-like 1, Delta-like 3, and Delta-like 4) or Serrate-like ligands, known as Jagged-1 and Jagged-2 [2]. All four Notch receptors are very similar except subtle differences in their extracellular and cytoplasmic domains. The extracellular domains of Notch contain many repeated copies of an epidermal growth factor (EGF)-like motif, which are involved in ligand interaction. Both Notch-1 and Notch-2 proteins have 36 arranged repeats of EGF-like domain, whereas Notch-3 and Notch-4 contain 34 and 29 EGF-like repeats, respectively [3]. The amino-terminal EGF-like repeats are followed by cysteine-rich Notch Lin12 repeats (N/Lin12) that modulate interactions between the extracellular and the membrane-tethered intracellular domains. The cytoplasmic region of Notch contains a Recombination Signal-Binding Protein 1 for J-kappa (RBP-J)-association molecule (RAM) domain, ankyrin (ANK) repeats, nuclear localization signals (NLS), a trans-activation domain (TAD) and a region rich in proline, glutamine, serine and threonine residues (PEST) sequence. It is well known that ANK repeats are necessary and sufficient for Notch activity. PEST sequence is involved in Notch protein turnover [4] and the cytoplasmic region of conveys the signal to the nucleus. Notch ligands have multiple EGF-like repeats in their extracellular domain and a cysteine-rich region (CR) in Serrate which are absent in Delta. Jagged-1 and Jagged-2 have almost two-fold numbers of EGF-like repeats compared to Delta [4] (Figure-1A).

Figure-1. A, Structure of Notch receptors (1-4) and ligands (Jagged-1, 2, Dll-1, 3, 4).

Figure-1

Both receptors and ligands contain multiple conserved domains. Notch is a single-pass transmembrane receptor. The extracellular domain contains EGF-like repeats and a cysteine-rich region. The intracellular domain contains the RAM domain, NLS, ANK, TAD and PEST domain. Notch ligands have multiple EGF-like repeats in their extracellular domain and a CR in Jagged which are absent in Delta. B, Schematic of Notch signaling. Notch signaling is activated after ligand binding to an adjacent Notch receptor between two neighboring cells. Upon activation, Notch receptors undergo a series of proteolytic cleavages by the metalloprotease, TACE, and γ-secretase complex. The cleavage releases the NICD into the cytoplasm, which can subsequently translocate into the nucleus. In the absence of NICD, transcription of Notch target genes is inhibited by a repressor complex mediated by the CSL. When NICD is in the nucleus, it forms an active transcriptional complex due to displacing the histone deacetylase-corepressor complex and recruiting the protein MAML1 and histone acetyltransferases to the CSL complex, leading to convert it from a transcriptional repressor into a transcription activator complex, leading to activation of Notch target genes. Diagram of putative therapeutic target in the Notch pathway. Notch signaling could be inhibited theoretically at many different levels. It is possible to (1) interfere with Notch-ligand interactions, (2) inhibit receptor activation, (3) promote Notch ubiquitination and degradation, (4) inhibit its translocation to the nuclear compartment and (5) inhibit NICD nuclear complex formation.

ANK: Ankyrin repeat, CR: Cysteine rich region, DSL: Delta-serrate-lag2, EGF: Epidermal growth factor, LNR: Lin12/Notch repeats, NLS: Nuclear localization signals, PEST: Proline, glutamine, serine,threonine, RAM: RBP-J association molecule domain, TAD: Transcriptional activator domain, TM: Transmembrane domain. CSL: C protein binding factor 1/Suppressor of Hairless/Lag-1, NICD: Notch intracellular domain, TACE: tumor necrosis factor-α-converting enzyme, MAML1: mastermind-like 1.

Notch signaling is activated after ligand binding to an adjacent Notch receptor between two neighboring cells. Upon activation, Notch receptors undergo a series of proteolytic cleavages by the metalloprotease, tumor necrosis factor-α-converting enzyme (TACE) and γ-secretase complex (comprised of presenilin-1/2, nicastrin, Pen-2, and Aph-1). The first cleavage is mediated by TACE, which leads to cleave the receptor in the extracellular domain. The released extracellular domain is then trans-endocytosed by the ligand-expressing cell. The second cleavage caused by the γ-secretase complex releases the Notch intracellular domain (NICD) into the cytoplasm, which can subsequently translocate into the nucleus because of the presence of nuclear localization signals located within it [5]. Therefore, inhibiting γ-secretase function would prevent the cleavage of the Notch receptor, blocking Notch signal transduction, and thus γ-secretase inhibitor (GSI) could be useful for the treatment of human malignancies [6]. Consistent with this rationale, GSI are now undergoing clinical trials (see website: clinicaltrials.gov). In the absence of NICD, transcription of Notch target genes is inhibited by a repressor complex mediated by the CSL (C protein binding factor 1/Suppressor of Hairless/Lag-1). When NICD is in the nucleus, it forms an active transcriptional complex due to displacing the histone deacetylase-corepressor complex and recruiting the protein mastermind-like 1 (MAML1) and histone acetyltransferases to the CSL complex, leading to convert it from a transcriptional repressor into a transcription activator complex [2] (Figure-1B). A few Notch target genes have been identified, including Hes (Hairy enhance of split) family, Hey (Hairy/enhancer of spit related with YRPW motif), nuclear factor-kappa B (NF-κB), vascular growth factor receptor (VEGF), mammalian target of rapamycin (mTOR), cyclin D1, c-myc, p21, p27, Akt, etc, all of which have been well documented for their roles in tumor development and progression [7-10].

2. Notch in cancer development and progression

It has been well known that Notch signaling plays important roles in maintaining the balance involved in cell proliferation, survival, apoptosis, and differentiation which affects the development and function of many organs. Therefore, dysfunction of Notch prevents differentiation, ultimately guiding undifferentiated cells toward malignant transformation. Indeed, many observations suggest that alterations in Notch signaling are associated with many human cancers [11-17]. Moreover, Notch receptors and ligands have been found as prognostic markers in human cancers [18; 19].

2.1. Notch functions as oncogene or tumor suppressor

Very interestingly, the function of Notch signaling in tumorigenesis could be either oncogenic or anti-proliferative, and the function could be context dependent. Notch signaling has been shown to be anti-proliferative in a limited number of tumor types, including skin cancer, human hepatocellular carcinoma, medullary thyroid, cervical cancer, and small cell lung cancer [20-24]. For example, Nicolas et al. used a tissue-specific inducible gene-targeting approach to study the physiological role of the Notch-1 receptor in the mouse epidermis and the corneal epithelium of adult mice. They unexpectedly found that ablation of Notch-1 results in epidermal and corneal hyperplasia followed by the development of skin tumors and facilitated chemical-induced skin carcinogenesis through beta-catenin-mediated signaling [24]. Recently, studies have also demonstrated that Notch-1 loss in epidermal keratinocytes promotes tumorigenesis by impairing skin-barrier integrity and creating a wound-like microenvironment in the skin. Using mice with a chimeric pattern of Notch-1 deletion, the authors have found that Notch-1 was insufficient to suppress tumor-promoting effect, and the tumor-promoting effect of Notch-1 loss involves a crosstalk between barrier-defective epidermis and its stroma [25]. More recent findings obtained in melanoma and non-melanoma skin cancers show that Notch signaling has a dual action (either as an oncogene or as a tumor suppressor), depending on the tumor cell type and involving synchronous activation of other intracellular signaling mechanisms [26].

However, most of the studies have shown oncogenic function of Notch in many human carcinomas. Emerging evidence suggest that the Notch signaling network is frequently deregulated in human malignancies with up-regulated expression of Notch receptors and their ligands were found in breast, lung, colon, head and neck, renal carcinoma, acute myeloid, Hodgkin and large-cell lymphomas and pancreatic cancer [8; 9; 15; 27-30]. Notch signaling pathway has also been found to cross-talk with multiple oncogenic signaling pathways, such as NF-κB, Akt, Sonic hedgehog (Shh), mTOR, Ras, Wnt, estrogen receptor (ER), androgen receptor (AR), epidermal growth factor receptor (EGFR) and platelet-derived growth factor (PDGF) [31-36]. Thus it is believed that the cross-talk between Notch and other signaling pathways may play critical roles in tumor aggressiveness. From the literature, Notch may act either as a tumor suppressor or tumor promoter depending on the cell type and tissue context, suggesting the complexity of Notch signaling pathways [26]. The functions of Notch signaling have recently been reviewed [7; 10; 16; 23; 27; 37-40], and thus the readers who are interested in learning more details on the functions of Notch signaling pathway could also consult well-published review articles because the focus of the current article is restricted to overcoming drug-resistance.

2.2. Notch as diagnostic and prognostic markers in human cancers

Notch signaling pathway has been shown to play a role in cancer patient survival. Patients with tumors expressing high levels of Jagged-1 or Notch-1 had a significantly poorer overall survival compared with patients expressing low levels of these genes. Jagged-1 was also found to be highly expressed in metastatic prostate cancer compared to localized prostate cancer or benign prostatic tissues [41]. Furthermore, high Jagged-1 expression in a subset of clinically localized tumors was significantly associated with recurrence, suggesting that Jagged-1 may be a useful marker in distinguishing indolent vs. aggressive prostate carcinomas [41]. Recently, high level co-expression of Jagged-1 and Notch-1 has been observed in human breast cancer and the expression was found to be associated with poor overall survival. Moreover, Jagged-1 expression is associated with a basal phenotype and recurrence in lymph node-negative breast cancer [42-44]. Very recently, it was found that Notch-1 and Notch-4 receptors could serve as prognostic markers in breast cancer [18]. Shi et al. also found that the Notch family expression pattern in papillary bladder transitional cell carcinoma which was different from that in invasive bladder transitional cell carcinoma. Therefore, the expression of Notch-1 and Jagged-1 could potentially be a useful marker for survival of patients diagnosed with papillary bladder transitional cell carcinoma [45]. More recently, it was reported that patients with cervical carcinomas positive for nuclear Notch-3 expression had significantly shorter overall survival than their peers whose tumors did not express nuclear Notch-3, suggesting that Notch-3 could be a prognostic marker in cervical carcinomas [46]. Further research toward exploration of the Notch signal as diagnostic and prognostic markers in human cancers requires in-depth investigations.

3. The role of Notch in drug-resistance

Recently, Notch pathway has been reported to be involved in drug-resistance. More importantly, the studies have demonstrated that Notch regulates the formation of cancer stem cells (CSCs) and contributes to the acquisition of the epithelial-mesenchymal transition (EMT) phenotype, which are critically associated with drug-resistance [40; 47]. Experimental evidence also revealed that Notch was involved in anticancer drug-resistance, indicating that targeting Notch could be a novel therapeutic approach for the treatment of cancer by overcoming drug-resistance of cancer cells, which may lead to the elimination of CSCs or EMT type cells which are typically drug-resistant, and are believed to be the “root cause” of tumor recurrence. Therefore, in the following sections, we have attempted to summarize the state-of-our-knowledge on the functional role of Notch signaling pathway in drug-resistance, and approaches by which one could overcome drug-resistance for the successful treatment of most human malignancies.

3.1 Drug-resistance

Chemotherapy is an important therapeutic strategy for cancer treatment and remains the mainstay for the management of human malignancies; however, chemotherapy fails to eliminate all tumor cells because of intrinsic or acquired drug-resistance, which is the most common cause of tumor recurrence. Human cancers are generally initially responsive to standard chemotherapies; however; response is almost inevitably followed by the development of drug-resistant phenotype [48], which leads to tumor recurrence and metastasis. The mechanisms responsible for drug-resistance are complex and still poorly understood. It may be due to either the specific nature and genetic background of the cancer cell itself, or the genetic changes that follow toxic chemotherapy [49]. Drug-resistance to therapy is classified by two categories: intrinsic (de novo) and acquired. Intrinsic resistance would make the therapy ineffective because prior to receiving the therapy, the cancer cells have already resistant to anti-cancer drugs due to multiple mechanisms. Acquired resistance develops during the treatment, although the tumor cells were not initially resistant to anti-cancer drugs. The most common reasons for the acquisition of resistance to anti-cancer drugs are due to expression of one or more energy-dependent transporters that detect and eject anti-cancer drugs from cells, insensitivity to drug-induced apoptosis and the induction of drug-detoxification mechanisms [50]. For example, the ATP-binding cassette (ABC) drug transporters have been shown to protect tumor cells from chemotherapeutic agents. ABC transporters eject toxic drugs from cancer cells, leading to reducing the effect of drug’s ability to kill the cancer cells. There are three ABC protein members that have been identified, which are ABCB1 (PGP, P-glycoprotein), ABCG2 (BCRP, breast cancer resistant protein) and ABCC1 (MRP1, multidrug resistance associated protein) [49; 50].

Studies over the past years have shown that a number of genes are involved in chemotherapy drug-resistance. These genes include: K-ras, TOP1 (topoisomerase 1), ERCC1 (excision repair cross complementation 1), LRP (lung resistance-related protein), COX-2, cyclin D1, Bcl-2, Survivin, etc [50-53]. Recently, many signaling pathways have been found to be involved in drug-resistance such as PTEN, Akt, mTOR, NF-κB, EGFR, FGFR (fibroblast growth factor receptor), Raf/MEK/ERK, MAPK (mitogen-activated protein kinase), IGF (insulin-like growth factor), and Notch signaling pathway [54-61]. The main roles of these pathways (except Notch signaling pathway) in drug-resistance have recently been reviewed [53-60]. Therefore, in this review article, we will focus our discussion on describing the role of Notch in drug-resistance and summarize approaches by which one could overcome drug-resistance.

3.2 Notch regulates EMT in drug-resistance

Recent studies have shown that EMT is associated with drug-resistance and cancer cell metastasis. It is now widely accepted that epithelial cells can acquire mesenchymal phenotype by a fundamental yet complex processes. The processes of EMT is a unique process by which epithelial cells undergo remarkable morphologic changes characterized by a transition from epithelial cobblestone phenotype to elongated fibroblastic phenotype (mesenchymal phenotype) leading to increased motility and invasion [62]. During the acquisition of EMT characteristics, cells lose epithelial cell-cell junction, actin cytoskeleton reorganization and the expression of proteins that promote cell-cell contact such as E-cadherin and γ-catenin, and gains in the expression of mesenchymal markers such as vimentin, fibronectin, α-smooth muscle actin (SMA), N-cadherin as well as increased activity of matrix metalloproteinases (MMPs) like MMP-2, MMP-3 and MMP-9, leading to an invasive phenotype [63]. Indeed, increasing evidence has shown the relationship between drug-resistance and the existence of EMT phenotype. Fox instance, epithelial but not mesenchymal gene signature has been associated with sensitivity to the EGFR inhibitor erlotinib mediated growth inhibition in lung cancer cells [64]. These results were confirmed in other types of tumors like head and neck squamous cell carcinoma and hepatocellular carcinoma as well as for the treatment of cancer with other EGFR inhibitors such as gefitinib and cetuximad [65; 66]. The processes of EMT has also been shown to be important on conferring drug-resistance characteristics to cancer cells against conventional therapeutics including taxol, vincristine and oxaliplatin [67]. Consistent with these observations, recent studies has also shown the link between EMT and gemcitabine-resistant pancreatic cancer cells with increased invasive capacities, oxaliplatin-resistant colorectal cancer cells, lapatinib-resistant breast cancer, and paclitaxel-resistant ovarian carcinoma cells [68-71]. Therefore, the discovery of precise mechanisms that governs the acquisition of EMT phenotype in cancer cells would likely be useful for devising targeted therapeutic approaches in combination with conventional therapeutics for the treatment of human malignancies.

Notch signaling pathway has been reported to be involved with the acquisition of EMT in drug-resistant cancer cells. Our recently published data showed that pancreatic cancer cells that are gemcitabine-resistant (GR) have acquired EMT phenotype as evidenced by elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as zinc-finger E-box binding homeobox 1 (ZEB1) and vimentin [70; 72]. We also found that Notch-2 and Jagged-1 are highly up-regulated in GR cells. Moreover, down-regulation of Notch signaling by siRNA approach led to partial reversal of the EMT phenotype, resulting in the mesenchymal-to-epithelial transition (MET), which was associated with decreased expression of vimentin, ZEB1, Slug, Snail, and NF-κB [72]. These results provide molecular evidence indicating that the activation of Notch signaling is mechanistically linked with chemoresistance phenotype, which is consistent with the acquisition of EMT phenotype by pancreatic cancer cells, and further suggesting that the inactivation of Notch signaling by novel strategies could be a potential targeted therapeutic approach for overcoming chemoresistance toward the prevention of tumor progression and/or treatment of human cancer for which current conventional therapeutic strategies are highly disappointing.

3.3 Notch regulates cancer stem cell in drug-resistance

Current cancer therapeutic strategies based on tumor regression may target and kill differentiated tumor cells, which constitute the bulk of the tumor, while sparing the rare cancer stem cell population. Cancer stem cells (CSCs) constitute a small subset of cancer cells that are a reservoir of self-sustaining cells with the exclusive ability to self-renew capacity leading to the maintenance of the tumor mass. The CSCs have been identified and isolated from tumors of the hematopoietic system, breast, lung, prostate, colon, brain, head and neck, and pancreas [73]. The CSCs are able to self-renew, differentiate, and regenerate to phenotypic cells of the original tumor when implanted into the severe combined immunodeficient mouse. Recently, CSCs have been believed to play critical roles in drug-resistance and cancer metastasis especially because CSCs express drug transporters and DNA repair systems, which allow CSCs to resist the killing effects of the drug. For instance, ABC drug transporters have been shown to protect CSCs from chemotherapeutic agents [74; 75]. Another mechanism is that CSCs accumulate mutations over time as a consequence of a long-term exposure to drug, which confer drug-resistance phenotype acquired by the daughter cancer cells [76]. Thus, the molecular knowledge of drug-resistance and metastasis with respect to CSCs in human cancer is considered very important, and the gain of such knowledge is likely to be helpful not only in the discovery of newer drugs but also in the design of novel therapeutic strategies for the treatment of human cancer with better treatment outcome.

Emerging evidence are clearly showing that Notch signaling plays critical roles in both stem cells and progenitor cells, suggesting that abnormal Notch signaling may contribute to carcinogenesis by deregulating the self-renewal of normal stem cells. For example, Phillips et al. have reported that CSCs can be identified by phenotypic markers and their fate is controlled by the Notch pathway in breast cancer [77]. Recombinant human erythropoietin receptor increased the numbers of stem cells and self-renewing capacity in a Notch-dependent fashion by induction of Jagged-1. Inhibitors of the Notch pathway blocked this effect, suggesting the mechanistic role of Notch signaling in the maintenance of cancer stem-like cell phenotype [77]. Farnie et al. also provided evidence for breast cancer stem cells, and their studies have consistently shown that stem-like cells and breast cancer initiating populations can be enriched using cell surface markers CD44+/CD24− and, as such, these cells showed up-regulated genes including Notch that are known to contribute to cancer stem-like cells characteristics [78]. It has also been reported that glioma stem cells have elevated chemo-resistance because of the high expression levels of drug-transporter proteins such as ABCG2. Furthermore, ABCG2 expression is also associated with proliferation, and the ABCG2 positive cells preferentially express several “stemness” genes such as Notch-1 [79]. Therefore, eradication of CSCs is an important goal for curing cancer, and thus the Notch pathway is considered an attractive target for treatment of cancer because targeting Notch will not only kill differentiated cancer cells but could also kill CSCs by overcoming drug-resistance.

3.4 Notch cross-talks with miRNAs in drug-resistance

Recently evidences suggest that microRNAs (miRNAs) play important roles in the regulation of drug-resistance [80]. It is well known that the miRNAs elicit their regulatory effects in post-transcriptional regulation of genes by binding to the 3′ untranslated region (3′UTR) of target messenger RNA (mRNA). Either perfect or near perfect complimentary base pairing results in the degradation of the mRNA, while partial base pairing leads to translational inhibition to functional proteins [81]. Very interestingly, some miRNAs are thought to have oncogenic activity while others have tumor suppressor activity. Oncogenic miRNAs are up-regulated in cancer and contribute to its pathology through various mechanisms such as targeting tumor suppressor genes. In contrast to the oncogenic miRNAs, other miRNAs are considered to have tumor suppressor activity and are down-regulated in cancer [82]. Recent studies have suggested altered expression of specific miRNAs in drug-resistant tumor cells. For example, the expression of three miRNAs (miR-192, miR-424 and miR-98) was significantly up-regulated while the expression of three other miRNAs (miR-194, miR-200b and miR-212) was down-regulated in docetaxel-resistant NSCLC cells [83]. Recently, Song et al. found that the expression of miR-140 was associated with chemo-sensitivity to 5-fluorouracil (5-FU) and methotrexate in osteosarcoma. Specifically, blocking endogenous miR-140 sensitized resistant cancer cells to 5-FU treatment, whereas overexpression of miR-140 made tumor cells more resistant to 5-FU, suggesting that miR-140 could be a novel target to develop therapeutic strategy to overcome drug-resistance [84]. Increasing evidence clearly implicating the role of miRNAs in drug-resistance, and in a recent review article, we have summarized the implication of miRNAs in drug-resistance for designing novel cancer therapy [80]. Here, we will discuss further how miRNAs could crosstalk with Notch pathway leading to drug-resistance and how and what novel agents could be useful to overcome such a drug-resistance phenotype of cancer cells.

One miRNA, namely miR-34, has been found to participate in Notch pathways regulation, and has been reported to be involved drug-resistance [85]. The miR-34 family is composed of three processed miRNAs: miR-34a is encoded by its own transcript, whereas miR-34b and miR-34c share a common primary transcript [86]. The expression of miR-34a has been found to be lower or undetectable in pancreatic cancer, osteosarcoma, breast cancer and non-small cell lung cancer, suggesting that miR-34a could function as a tumor suppressor gene [86]. Recently, Li et al. reported that transfection of miR-34a to glioma cells down-regulated the protein expression of Notch-1, Notch-2, and CDK6 [87]. More recently, Ji et al reported that human gastric cancer cells with miR-34 restoration reduced the expression of target gene Notch [88]. In parallel, the same group reported that Notch-1 and Notch-2 are downstream genes of miR-34 in pancreatic cancer cells because restoration of miR-34 expression in the pancreatic cancer cells down-regulated the expression of Notch-1 and Notch-2. Moreover, they reported that pancreatic cancer stem cells are enriched with tumor-initiating cells or CSCs with high levels of Notch-1/2 and loss of miR-34 [89], suggesting that miR-34 may be involved in pancreatic cancer stem cell self-renewal mediated by Notch signaling. More recently, Fujita et al. demonstrated that miR-34a is down-regulated in drug-resistant prostate cancer cells, and ectopic over-expression of miR-34a resulted in growth inhibition and attenuated chemoresistance to the anti-cancer drug camptothecin [85]. Very recently, another study determined that miR-34a was down-regulated in doxorubicin and verapamil resistance MCF-7 breast cancer cells [90]. Collectively, these reports clearly suggest the role of miR-34 in drug-resistance, which is in part mediated through the regulation of Notch signaling; however, further in-depth research is needed in order to fully understanding how miR-34 regulates the Notch pathway in drug-resistant cells and finding novel avenues by which one could up-regulate miR-34 would be highly innovative for designing novel treatment strategies for eliminating tumor cells that are the root cause of tumor recurrence and metastasis.

Another miRNA, miR-1, was markedly reduced in primary human hepatocellular carcinoma, prostate cancer, head and neck, and lung cancer [91; 92]. Recently, miR-1 was also found to alter sensitivity of cancer cells to therapeutic agents. Nasser et al. reported that ectopic miR-1 expression sensitize lung cancer cells to anti-cancer drug doxorubicin, suggesting that up-regulation of miR-1 has potential as a target for therapy against lung cancers [92]. It has been reported that Notch ligand Dll-1 protein levels are negatively regulated by miR-1 [93]. In parallel, miR-1 directly targets the Notch ligand delta for repression [94], suggesting that miR-1 may regulate drug-resistance in part via regulating the Notch signaling pathway. Recently, the alteration of miR-200 family was also found in drug-resistant cells. The miR-200 family has five members: miR-200a, miR-200b, miR-200c, miR-141 and miR-429. The expression of miR-200b was significantly down-regulated in docetaxel-resistant NSCLC cells [83]. Recently, many studies have shown that the miR-200 family regulates EMT which is associated with drug-resistance. One study discovered that miR-200 expression regulates EMT in bladder cancer cells and reverses resistance to EGFR therapy [95]. Another recent study reported that miR-200c restored microtubule-binding chemotherapeutic agents in breast and ovarian cancer cells [96]. We also found that miR-200a, miR-200b, and miR-200c were down-regulated in gemcitabine-resistant pancreatic cancer cells, which show the acquisition of EMT phenotype. Furthermore, we have shown that re-expression of miR-200 family resulted in the down-regulation of ZEB1, slug, E-cadherin, and vimentin, and increased cell sensitivity to gemcitabine [97]. In addition, we found that Notch-1 could be one of miR-200b targets because over-expression of miR-200 family significantly inhibited Notch-1 expression in gemcitabine-resistant pancreatic cancer cells and prostate cancer cells (unpublished data), suggesting that re-expression of miR-200 could increase drug-sensitivity, which indeed could be mediated through the regulation of Notch signaling pathway. Thus, it is our belief that more and more miRNAs will be discovered, whose re-expression will make drug-resistant cells drug-sensitive, and such strategy could be useful in eliminating cancer cells with propensity of recurrence and metastasis.

3.5. Notch pathway in specific chemoresistance

Chemotherapy is critically important for cancer therapy; however, chemotherapy fails to eliminate all tumor cells due to chemoresistance either the de novo or acquired chemo-resistance. Currently, chemo-resistance is still the most common cause of tumor progression. Many cellular pathways have been found to be involved in drug-resistance. Recent studies have demonstrated that Notch pathway plays a critical role in anti-cancer drug-resistance as documented in the previous paragraphs. Here, we will further discuss the roles of Notch pathway in chemoresistance and a comprehensive list of Notch pathway that is involved in chemoresistance is presented in Table 1.

Table 1. A comprehensive list of Notch pathway involved in chemoresistance.

Drug Targeted genes Cell or tissue Reference
Cisplatin Notch-1 was highly expressed in cisplatin
resistance cells
Head and neck squamous
cell, colorectal and
ovarian cancer cells
[98-101]
Doxorubicin Notch-3 was up-regulated and contributed
to the anti-cancer drug doxorubicin
resistance through regulating p53
expression and DNA damage.
GSI improved the cytotoxicity of the
doxorubicin
Hepatocellular carcinoma
cells, myeloma cells
[114; 116]
Erlotinib GSI enhance the EGFR tyrosine kinase
inhibitor erlotinib anti-tumor activity
Lung cancer [112]
5-fluorouracil 5-FU induced NICD protein and activated
Hes-1
Colon cancer cells [108]
Gemcitabine Inhibition of Notch-3 enhances sensitivity
to gemcitabine.
Pancreatic cancer cells [72; 102]
Notch-2 and Jagged-1 are highly up-
regulated in gemcitabine-resistant.
Gefitinib Over-expression of Notch-1 contributes to
the gefitinib resistance
Breast cancer cells [111; 113]
Melphalan GSI improved the cytotoxicity of the
melphalan
Myeloma cells [116]
Mitoxantrone Activation of Notch-1 resulted in the
protection from mitoxantrone-induced
apoptosis
Myeloma cells and
malignant lymphoid cell
lines
[115]
Oxaliplatin oxaliplatin induced NICD protein and
activated Hes-1
Colon cancer cells [108]
GSI sensitized cells to oxaliplatin
Paclitaxel Taxol enhanced the expression of Notch
downstream gene CBF1.
Erythroleukemia,
cervical, colorectal,
ovarian cancer cell
[105-107]
GSIs are useful for the chemotherapeutic
treatment of taxol-resistant cancer cells
Tamoxifen Down-regulation of Notch-1 or GSI
potentiated the effects of tamoxifen
Breast cancer cells [27]
Taxol GSIs enhance taxol-induced mitotic arrest
and apoptosis of colon cancer cells
erythroleukemia cells,
cervical , colon, ovarian
cancer cells
[105-107]
Taxetere Down-regulation of Notch-1 signaling
increased chemosensitivity to taxotere
Breast and prostate
cancer cells
[104]
Trastuzumab Notch-1 signaling regulates ErbB-2
transcription.
Breast cancer cells [61; 109; 110]
Down-regulation of Notch-1 increased
efficacy of trastuzumab and restored
sensitivity in resistant cells

3.5.1 The role of Notch in anti-cisplatin resistance

Cisplatin is the most important chemotherapeutic agent for the treatment of human carcinoma including lung, ovarian, bladder and testicular cancers. However, acquired resistance to cisplatin therapy is still a critical problem in the clinical management of cancer patients. Recent studies have shown that Notch may play a role in the mechanisms of cisplatin resistance. One such study by Zhang et al. demonstrated that the positive rate of Notch-1 was significantly higher in head and neck squamous cell carcinoma (HNSCC) than in normal squamous epithelium, and it was negatively correlated with cisplatin-sensitivity [98]. Moreover, Notch-1 was highly expressed in cisplatin resistance HNSCC patients [99]. Further, cisplatin resistance of HNSCC was decreased after inhibition of Notch signaling [99]. In addition, combination of GSI and cisplatin elicits a striking induction of colorectal cancer cell death [100]. Human ovarian cancer-initiating cells enhanced chemoresistance to cisplatin and up-regulation of Notch-1 compared with parental tumor cells [101]. These results support the notion that inactivation of Notch pathway could be a novel strategy for patients who is likely respond to such chemotherapy.

3.5.2 The role of Notch in anti-gemcitabine resistance

Gemcitabine monotherapy (2′,2′-difluorodeoxycytidine), a deoxycytidine analogue, or its combination with other agents has become standard chemotherapy for the treatment of advanced human cancers. However, the effect of gemcitabine on survival has been disappointing, which could be due to many factors including intrinsic drug-resistance or acquired drug-resistance. For example, gemcitabine showed only about 5% partial response rate and imparts a progression-free survival interval ranging from 0.9 to 4.2 months in pancreatic cancer. This disappointing outcome strongly suggests that a better understanding of the mechanism by which chemo-resistance arises is likely to lead to novel therapeutic strategies for the successful treatment of cancer patients. Recently, Notch signaling pathway was found to play a critical role in gemcitabine-resistant cancer cells. Yao et al. demonstrated that Notch-3 siRNA suppressed Notch-3 expression, and increased gemcitabine-induced, caspase-mediated apoptosis in pancreatic cancer. Moreover, inhibition of Notch-3 enhances sensitivity to gemcitabine in pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway [102]. We also found that Notch-2 and Jagged-1 are highly up-regulated in gemcitabine-resistant pancreatic cancer cells. Moreover, down-regulation of Notch signaling by siRNA approach led to partial reversal of the EMT phenotype, which was associated with increased gemcitabine sensitivity [72].

3.5.3 The role of Notch in anti-taxotere resistance

Taxotere (Docetaxel), a member of the taxane family, has shown high efficacy in the treatment of a wide spectrum of solid tumors including prostate, breast, and gastric cancer [103]. It has been found that taxotere inhibits cell growth and induces apoptosis with down-regulation of some genes for cell proliferation, transcription factors, and oncogenesis, and up-regulation of some genes related to the induction of apoptosis and cell cycle arrest in tumor cells, suggesting pleiotropic effects of taxotere on tumor cells. Clinical trials have shown that the combination chemotherapy using taxotere with other agents improves survival in cancer patients [103]. However, the effect of taxoetere is also disappointed due to drug-resistant. Recently, we found that taxotere-resistant DU145 prostate cells have high expression of Notch-1 (unpublished data), suggesting that Notch pathway is involved in taxotere-resistance. Another group also reported that down-regulation of Notch-1 signaling increased chemosensitivity to taxotere and doxorubicin in breast cancer [104], indicating that Notch signaling may be a promising target for overcoming taxotere-resistant in breast cancer and other cancers.

3.5.4 The role of Notch in anti-taxol resistance

Taxol (Paclitaxel) is another anti-cancer chemotherapy drug. It is used for the treatment of breast, ovarian, lung, bladder, prostate, melanoma, esophageal, as well as other types of solid tumors. It has been reported that taxol could enhance the expression of Notch downstream gene CBF1 in erythroleukemia K562 cells and cervical carcinoma HeLa cells [105]. Recently, it was found that GSIs could enhance taxol-induced mitotic arrest and apoptosis of colon cancer cells both in vitro and in vivo, suggesting that GSIs could be useful for the treatment of taxol-resistant colorectal cancers [106]. More recently, Mine et al. reported that targeting Notch-1 was significant for novel treatments to eliminate taxol-resistant ovarian cancer stem cells [107]. These limited emerging evidences suggests that overcoming taxol-resistance could be achieved by inactivation of Notch signaling which would become a rational approach for the treatment of most human malignancies.

3.5.5 The role of Notch in anti-tamoxifen resistance

Tamoxifen is a well known anti-cancer drug for the treatment of breast cancer. Certain types of breast cancer require estrogen to grow and tamoxifen blocks the actions of estrogen. Tamoxifen works by blocking the effect of estrogen, resulting in inhibiting gene transcription and tumor growth that are activated by estrogen. Resistance to tamoxifen is often seen in tumor cells that become estrogen independent, thus tamoxifen can not inhibit tumor growth. In a recent study, Rizzo et al found that down-regulation of Notch-1 by siRNA or GSI potentiated the effects of tamoxifen in breast cancer cells. Moreover, GSI in combination with tamoxifen caused regression of breast cancer cell growth in mice [27]. These data indicate that the combinations of tamoxifen and Notch inhibitors may be effective in ERα (+) breast cancer, and such a combination treatment could eliminate the emergence of Tamoxifen-resistance, which certainly would improve the treatment outcome of patients diagnosed with breast cancer.

3.5.6 The role of Notch in anti-oxaliplatin resistance

Oxaliplatin is a platinum-compound chemotherapy drug that acts as an alkylating agent. Oxaliplatin is used to treat colorectal cancer, and it is often given in combination with other anticancer drugs (5-fluorouracil and leucovorin). It has been shown that Notch-1 is up-regulated in colon cancer. Further, oxaliplatin or 5-FU could induce NICD protein and activated Hes-1 though an increase in the activity and expression of gamma-secretase complex. Therefore, GSI could sensitize cells to chemotherapy, which has been demonstrated showing synergistic activity with oxaliplatin and 5-FU [108]. The authors have summarized that colon cancer cells with up-regulated expression of Notch-1 could function as a protective mechanism in response to chemotherapy [108], further suggesting that combining GSIs with chemotherapy may be a novel strategy for overcoming chemoresistance in colon cancer.

3.5.7 The role of Notch in anti-trastuzumab resistance

Trastuzumab is the humanized, monoclonal antibody that directed against ErbB-2. It has shown efficacy causing improved overall survival for breast cancer patients. However, resistance to trastuzumab remains a major concern, specifically in women with metastatic breast cancer. It has been found that Notch-1 could contribute to trastuzumab resistance in breast cancer [61]. Notch-1 signaling regulates ErbB-2 transcription in ErbB-2-overexpressing breast carcinoma tumor-initiating cells, therefore affecting their self-renewal properties [109]. Trastuzumab increased the Notch-1 activity and its target gene expression. The expression of Notch-1, Hey-1, and Hes-5 was highly expressed in trastuzumab-resistant BT474 compared to trastuzumab-sensitive BT474 [110]. Moreover, down-regulation of Notch-1 increased efficacy of trastuzumab in BT474 sensitive cells and restored sensitivity in resistant cells. Furthermore, the growth of both trastuzumab sensitive and resistant cells was completely inhibited by combining trastuzumab plus Notch-1 siRNA. The Notch-1 siRNA or a GSI re-sensitized trastuzumab-resistant BT474 cells to trastuzumab [110], suggesting that Notch-1 might play a novel role in resistance to trastuzumab, which could be prevented or reversed by inhibiting Notch-1.

3.5.8 The role of Notch in other chemoresistance drugs

Notch signaling pathway was also found in many other chemo-resistant cancer cells [111-113]. For example, Notch-3 was up-regulated and contributed to the anti-cancer drug doxorubicin resistance through regulating p53 expression and DNA damage in human hepatocellular carcinoma (HCC) cell lines, suggesting that Notch-3 silencing in combination with chemotherapeutics could conceivably provide a novel strategy for HCC treatment [114]. Another study determined that Notch-1 signaling was involved in bone marrow stroma-mediated de novo melphalan and mitoxantrone resistance of myeloma [115]. Moreover, GSI significantly improved the cytotoxicity of the chemotherapeutic drugs doxorubicin and melphalan in myeloma cells, demonstrating that inhibition of Notch signaling prevents bone marrow mediated drug-resistance and sensitizes to chemotherapy [116]. There are currently more and more studies being done to uncover the resistance mechanism by Notch signaling pathway.

4. Targeting Notch to increase drug-sensitivity

Notch signaling has been reported to be involved in drug-resistance as documented in the previous paragraphs. Therefore, targeting Notch pathway for cancer therapy is a novel strategy for optimizing treatment outcome of conventional chemotherapy. Strategies to regulate Notch expression in cancers could be at many different levels. It is possible to interfere with Notch-ligand interactions, receptor activation, mono-ubiquitination, NICD nuclear complex formation and inhibition of its translocation to the nuclear compartment (Figure 1B). Notch signaling is activated via the activity of γ-secretase which became a target in cancer therapy. Several forms of γ-secretase inhibitors (GSIs) have been tested for anti-tumor effects. The GSI inhibits cell growth and could induce apoptosis in many human cancer cells, such as hepatoma cells, breast cancer cells, pancreatic cancer cells, and myeloma cells [1; 7]. Recently, it was found that inhibition of Notch signaling with GSI sensitized cells to chemotherapy and was synergistic with oxaliplatin and 5-FU, suggesting that combining GSI with chemotherapy may represent a novel approach for treating metastatic colon cancers as indicated above [108]. Recently, Gu et al reported that cisplatin resistance of HNSCC was decreased by inhibition of Notch signaling, suggesting that inactivation of Notch-1 could help HNSCC response to chemotherapy [99]. Very recently, Song et al. evaluated the effects of Notch-1 silencing on cisplatin induced cytotoxicity in CaSki cervival cancer cells. They found that Notch-1 knockdown by siRNA significantly potentiated cisplatin-induced cytotoxicity, lowering the IC50 value of cisplatin in CaSki cells by almost two orders of magnitude [28]. Collectively, all the published data suggest that targeting Notch pathway could increase drug-sensitivity in human cancers; however, one of the major challenges is to eliminate unwanted toxicity associated with the GSI, especially the cytotoxicity in the gastrointestinal tract. Shih et al. reported the possible mechanisms underlying the unwanted cytotoxicity of GSI [39]. Notch signaling pathway is known to widely participate in cellular physiology in normal tissues, therefore, it is plausible that inactivation of γ-secretase may lead to the dysfunction of vital organs. Moreover, GSI do not exclusively target the Notch signaling pathways because γ-secretase has many substrates in addition to Notch receptors, such as several Notch ligands, v-erb-a erythroblastic leukemia viral oncogene homolog 4 (ErbB4), CD44, etc. Further, GSI may target proteases other than γ-secretase. Therefore, GSI may have widespread adverse effects in vivo because proteases participate in a wide array of cellular functions [39].

In order to overcome such limitations, recent studies have shown that “natural agents”, which are typically non-toxic to humans, including isoflavone, resveratrol, curcumin, withaferin-A, and others could inhibit the Notch-1 expression. The studies from our laboratory have shown that genistein and curcumin down regulated the transcription and translation of Notch-1 and its downstream genes, Hes-1, cyclin D1, Bcl-XLand NF-κB. Over-expression of Notch-1 by Notch-1 cDNA transfection abrogated genistein- and curcumin-induced apoptosis to a certain degree. Therefore, we strongly believe that down-regulation of Notch-1 by genistein and curcumin is mechanistically linked to cell proliferation and apoptotic processes [9; 117-119]. In addition, studies from other laboratories have shown that resveratrol could induce apoptosis by inhibiting the Notch pathway mediated by p53 and PI3K/Akt in T-ALL [120]. Moreover, one Chinese herb antitumor B was also found to inhibit Notch expression in a mouse lung tumor model [121]. Recently, it was reported that withaferin-A could inhibit Notch-1 signaling and thereby down-regulates pro-survival pathways, such as Akt/NF-κB/Bcl-2, in colon cancer cells [122]. Furthermore, recent studies have shown that natural agents could alter the expression of specific miRNAs that could regulate Notch signaling pathway. We found that re-expression of miR-200 by pre-miR-200 transfection or treatment of GR pancreatic cancer cells with isoflavone resulted in the up-regulation of miR-200, leading to increased sensitivity of GR cells to gemcitabine. Isoflavone also induced the expression of let-7, which could be linked to the treatment effects [97]. Considering the relatively non-toxic nature of natural agents, targeting the Notch pathway by these natural agents combined with conventional chemotherapy could be a novel and safer approach for achieving better treatment outcome; however, further in-depth preclinical and clinical studies are warranted in order to appreciate the value of natural agents in overcoming drug-resistance to eliminate cancer cells that are the root cause of tumor recurrence and metastasis.

5. Conclusion and overall perspectives

In this review article we attempted to summarize the role of Notch pathway in drug-resistance; however we could not cite all the published studies, and thus we sincerely apologize to those whose work has not been cited here due to space limitations. In conclusion, recent studies demonstrate that Notch signaling pathway may play critical roles in the regulation of anti-cancer drug-sensitivity and resistance. Since Notch signaling pathway has been found to be involved in EMT and CSCs and deregulated expression of miRNAs, suggesting that up-regulation and down-regulation of specific miRNA that are intimately associated with Notch signaling could become a novel approach for overcoming drug-resistance (Figure-2). As such, high expression of Notch pathway can reduce response to anti-cancer agents such as cispltin, doxorubicin, 5-fluorouracil, gemcitabine, tamoxifen, etc., and thus, down-regulation of Notch signaling by multiple approaches appears to be a novel strategy for increasing drug-sensitivity of cancer cells to conventional chemotherapeutics. To that end, natural agents such as genistein, curcumin, resveratrol, and others could be very useful for the inhibition of Notch signaling pathway, which could lead to the inhibition of cancer growth, induction of apoptosis, reversal of EMT phenotype, elimination of drug-resistant CSCs, and thereby increasing drug-sensitivity, which would be useful for treatment of cancer patients with better treatment outcome. In summary, our findings together with those reported in the literature are becoming an exciting area for further in-depth research toward targeted inactivation of Notch signaling proteins, especially by natural agents, as a novel therapeutic approach for increasing the drug-sensitivity, and thereby improving the treatment outcome of cancer patients, which is believed to be due to eliminating the cancer cells that are the root cause of tumor recurrence and metastasis.

Figure-2.

Figure-2

The role of Notch signaling pathway in the progression of cancer, and during the acquisition of EMT phenotype, and the formation of cancer stem cells, leading to drug resistance. Natural agents and γ-secretase inhibitors could be useful for targeting Notch signaling pathway proteins, which could enhance the sensitivity of chemotherapeutic drugs.

Acknowledgements

The authors’ work cited in this review was funded by grants from the National Cancer Institute, NIH (5R01CA131151, 5R01CA083695, 1R01CA132794, 1R01CA101870) to F.H.S. and Department of Defense Postdoctoral Training Award W81XWH-08-1-0196 (Zhiwei Wang) and also partly supported by a subcontract award (F.H.S.) from the University of Texas MD Anderson Cancer Center through a SPORE grant (5P20-CA101936) on pancreatic cancer awarded to James Abbruzzese. We also sincerely thank both Puschelberg and Guido foundation for their generous contributions to our research.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Reference List

  • [1].Miele L. Notch signaling. Clin.Cancer Res. 2006;12:1074–1079. doi: 10.1158/1078-0432.CCR-05-2570. [DOI] [PubMed] [Google Scholar]
  • [2].Miele L, Osborne B. Arbiter of differentiation and death: Notch signaling meets apoptosis. J.Cell Physiol. 1999;181:393–409. doi: 10.1002/(SICI)1097-4652(199912)181:3<393::AID-JCP3>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • [3].Weinmaster G. The ins and outs of notch signaling. Mol.Cell Neurosci. 1997;9:91–102. doi: 10.1006/mcne.1997.0612. [DOI] [PubMed] [Google Scholar]
  • [4].Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137:216–233. doi: 10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Fortini ME. Notch signaling: the core pathway and its posttranslational regulation. Dev.Cell. 2009;16:633–647. doi: 10.1016/j.devcel.2009.03.010. [DOI] [PubMed] [Google Scholar]
  • [6].Miele L, Miao H, Nickoloff BJ. Notch signaling as a novel cancer therapeutic target. Curr.Cancer Drug Targets. 2006;6:313–323. doi: 10.2174/156800906777441771. [DOI] [PubMed] [Google Scholar]
  • [7].Rizzo P, Osipo C, Foreman K, Golde T, Osborne B, Miele L. Rational targeting of Notch signaling in cancer. Oncogene. 2008;27:5124–5131. doi: 10.1038/onc.2008.226. [DOI] [PubMed] [Google Scholar]
  • [8].Wang Z, Banerjee S, Li Y, Rahman KM, Zhang Y, Sarkar FH. Down-regulation of Notch-1 inhibits invasion by inactivation of nuclear factor-{kappa}B, vascular endothelial growth factor, and matrix metalloproteinase-9 in pancreatic cancer cells. Cancer Res. 2006;66:2778–2784. doi: 10.1158/0008-5472.CAN-05-4281. [DOI] [PubMed] [Google Scholar]
  • [9].Wang Z, Zhang Y, Li Y, Banerjee S, Liao J, Sarkar FH. Down-regulation of Notch-1 contributes to cell growth inhibition and apoptosis in pancreatic cancer cells. Mol.Cancer Ther. 2006;5:483–493. doi: 10.1158/1535-7163.MCT-05-0299. [DOI] [PubMed] [Google Scholar]
  • [10].Wang Z, Li Y, Banerjee S, Sarkar FH. Exploitation of the Notch signaling pathway as a novel target for cancer therapy. Anticancer Res. 2008;28:3621–3630. [PubMed] [Google Scholar]
  • [11].Weng AP, Lau A. Notch signaling in T-cell acute lymphoblastic leukemia. Future.Oncol. 2005;1:511–519. doi: 10.2217/14796694.1.4.511. [DOI] [PubMed] [Google Scholar]
  • [12].Sjolund J, Manetopoulos C, Stockhausen MT, Axelson H. The Notch pathway in cancer: differentiation gone awry. Eur.J.Cancer. 2005;41:2620–2629. doi: 10.1016/j.ejca.2005.06.025. [DOI] [PubMed] [Google Scholar]
  • [13].Li JL, Harris AL. Notch signaling from tumor cells: a new mechanism of angiogenesis. Cancer Cell. 2005;8:1–3. doi: 10.1016/j.ccr.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • [14].Bray SJ. Notch signalling: a simple pathway becomes complex. Nat.Rev.Mol.Cell Biol. 2006;7:678–689. doi: 10.1038/nrm2009. [DOI] [PubMed] [Google Scholar]
  • [15].Real PJ, Ferrando AA. Notch inhibition and glucocorticoid therapy in T-cell acute lymphoblastic leukemia. Leukemia. 2009;23:1374–1377. doi: 10.1038/leu.2009.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Qiao L, Wong BC. Role of Notch signaling in colorectal cancer. Carcinogenesis. 2009;30:1979–1986. doi: 10.1093/carcin/bgp236. [DOI] [PubMed] [Google Scholar]
  • [17].Sandy AR, Maillard I. Notch signaling in the hematopoietic system. Expert.Opin.Biol.Ther. 2009;9:1383–1398. doi: 10.1517/14712590903260777. [DOI] [PubMed] [Google Scholar]
  • [18].Yao K, Rizzo P, Rajan P, Albain K, Rychlik K, Sha S, Miele L. Notch-1 and Notch-4 Receptors as Prognostic Markers in Breast Cancer. Int.J.Surg.Pathol. 2010 doi: 10.1177/1066896910362080. [DOI] [PubMed] [Google Scholar]
  • [19].Jubb AM, Soilleux EJ, Turley H, Steers G, Parker A, Low I, Blades J, Li JL, Allen P, Leek R, Noguera-Troise I, Gatter KC, Thurston G, Harris AL. Expression of vascular notch ligand delta-like 4 and inflammatory markers in breast cancer. Am.J.Pathol. 2010;176:2019–2028. doi: 10.2353/ajpath.2010.090908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Wang M, Xue L, Cao Q, Lin Y, Ding Y, Yang P, Che L. Expression of Notch1, Jagged1 and beta-catenin and their clinicopathological significance in hepatocellular carcinoma. Neoplasma. 2009;56:533–541. doi: 10.4149/neo_2009_06_533. [DOI] [PubMed] [Google Scholar]
  • [21].Gao J, Chen Y, Wu KC, Liu J, Zhao YQ, Pan YL, Du R, Zheng GR, Xiong YM, Xu HL, Fan DM. RUNX3 directly interacts with intracellular domain of Notch1 and suppresses Notch signaling in hepatocellular carcinoma cells. Exp.Cell Res. 2010;316:149–157. doi: 10.1016/j.yexcr.2009.09.025. [DOI] [PubMed] [Google Scholar]
  • [22].Wang C, Qi R, Li N, Wang Z, An H, Zhang Q, Yu Y, Cao X. Notch1 signaling sensitizes tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in human hepatocellular carcinoma cells by inhibiting Akt/Hdm2-mediated p53 degradation and up-regulating p53-dependent DR5 expression. J.Biol.Chem. 2009;284:16183–16190. doi: 10.1074/jbc.M109.002105. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [23].Dotto GP. Notch tumor suppressor function. Oncogene. 2008;27:5115–5123. doi: 10.1038/onc.2008.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Nicolas M, Wolfer A, Raj K, Kummer JA, Mill P, van NM, Hui CC, Clevers H, Dotto GP, Radtke F. Notch1 functions as a tumor suppressor in mouse skin. Nat.Genet. 2003;33:416–421. doi: 10.1038/ng1099. [DOI] [PubMed] [Google Scholar]
  • [25].Demehri S, Turkoz A, Kopan R. Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment. Cancer Cell. 2009;16:55–66. doi: 10.1016/j.ccr.2009.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Panelos J, Massi D. Emerging role of Notch signaling in epidermal differentiation and skin cancer. Cancer Biol.Ther. 2009;8:1986–1993. doi: 10.4161/cbt.8.21.9921. [DOI] [PubMed] [Google Scholar]
  • [27].Rizzo P, Miao H, D’Souza G, Osipo C, Song LL, Yun J, Zhao H, Mascarenhas J, Wyatt D, Antico G, Hao L, Yao K, Rajan P, Hicks C, Siziopikou K, Selvaggi S, Bashir A, Bhandari D, Marchese A, Lendahl U, Qin JZ, Tonetti DA, Albain K, Nickoloff BJ, Miele L. Cross-talk between notch and the estrogen receptor in breast cancer suggests novel therapeutic approaches. Cancer Res. 2008;68:5226–5235. doi: 10.1158/0008-5472.CAN-07-5744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Song LL, Peng Y, Yun J, Rizzo P, Chaturvedi V, Weijzen S, Kast WM, Stone PJ, Santos L, Loredo A, Lendahl U, Sonenshein G, Osborne B, Qin JZ, Pannuti A, Nickoloff BJ, Miele L. Notch-1 associates with IKKalpha and regulates IKK activity in cervical cancer cells. Oncogene. 2008;27:5833–5844. doi: 10.1038/onc.2008.190. [DOI] [PubMed] [Google Scholar]
  • [29].Wang Z, Li Y, Banerjee S, Kong D, Ahmad A, Nogueira V, Hay N, Sarkar FH. Down-regulation of Notch-1 and Jagged-1 inhibits prostate cancer cell growth, migration and invasion, and induces apoptosis via inactivation of Akt, mTOR, and NF-kappaB signaling pathways. J.Cell Biochem. 2010;109:726–736. doi: 10.1002/jcb.22451. [DOI] [PubMed] [Google Scholar]
  • [30].Zhang Y, Wang Z, Ahmed F, Banerjee S, Li Y, Sarkar FH. Down-regulation of Jagged-1 induces cell growth inhibition and S phase arrest in prostate cancer cells. Int.J.Cancer. 2006 doi: 10.1002/ijc.22077. [DOI] [PubMed] [Google Scholar]
  • [31].De La OJ, Murtaugh LC. Notch and Kras in pancreatic cancer: at the crossroads of mutation, differentiation and signaling. Cell Cycle. 2009;8:1860–1864. doi: 10.4161/cc.8.12.8744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Osipo C, Golde TE, Osborne BA, Miele LA. Off the beaten pathway: the complex cross talk between Notch and NF-kappaB. Lab Invest. 2008;88:11–17. doi: 10.1038/labinvest.3700700. [DOI] [PubMed] [Google Scholar]
  • [33].Sundaram MV. The love-hate relationship between Ras and Notch. Genes Dev. 2005;19:1825–1839. doi: 10.1101/gad.1330605. [DOI] [PubMed] [Google Scholar]
  • [34].Wang Z, Sengupta R, Banerjee S, Li Y, Zhang Y, Rahman KM, Aboukameel A, Mohammad R, Majumdar AP, Abbruzzese JL, Sarkar FH. Epidermal growth factor receptor-related protein inhibits cell growth and invasion in pancreatic cancer. Cancer Res. 2006;66:7653–7660. doi: 10.1158/0008-5472.CAN-06-1019. [DOI] [PubMed] [Google Scholar]
  • [35].Wang Z, Kong D, Banerjee S, Li Y, Adsay NV, Abbruzzese J, Sarkar FH. Down-regulation of platelet-derived growth factor-D inhibits cell growth and angiogenesis through inactivation of Notch-1 and nuclear factor-kappaB signaling. Cancer Res. 2007;67:11377–11385. doi: 10.1158/0008-5472.CAN-07-2803. [DOI] [PubMed] [Google Scholar]
  • [36].Weijzen S, Rizzo P, Braid M, Vaishnav R, Jonkheer SM, Zlobin A, Osborne BA, Gottipati S, Aster JC, Hahn WC, Rudolf M, Siziopikou K, Kast WM, Miele L. Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat.Med. 2002;8:979–986. doi: 10.1038/nm754. [DOI] [PubMed] [Google Scholar]
  • [37].De La OJ, Murtaugh LC. Notch signaling: where pancreatic cancer and differentiation meet? Gastroenterology. 2009;136:1499–1502. doi: 10.1053/j.gastro.2009.03.022. [DOI] [PubMed] [Google Scholar]
  • [38].Koch U, Radtke F. Notch and cancer: a double-edged sword. Cell Mol.Life Sci. 2007;64:2746–2762. doi: 10.1007/s00018-007-7164-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Shih I, Wang TL. Notch signaling, gamma-secretase inhibitors, and cancer therapy. Cancer Res. 2007;67:1879–1882. doi: 10.1158/0008-5472.CAN-06-3958. [DOI] [PubMed] [Google Scholar]
  • [40].Wang Z, Li Y, Kong D, Ahmad A, Banerjee S, Sarkar FH. Cross-talk between miRNA and Notch signaling pathways in tumor development and progression. Cancer Lett. 2010;292:141–148. doi: 10.1016/j.canlet.2009.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Santagata S, Demichelis F, Riva A, Varambally S, Hofer MD, Kutok JL, Kim R, Tang J, Montie JE, Chinnaiyan AM, Rubin MA, Aster JC. JAGGED1 expression is associated with prostate cancer metastasis and recurrence. Cancer Res. 2004;64:6854–6857. doi: 10.1158/0008-5472.CAN-04-2500. [DOI] [PubMed] [Google Scholar]
  • [42].Reedijk M, Odorcic S, Chang L, Zhang H, Miller N, McCready DR, Lockwood G, Egan SE. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res. 2005;65:8530–8537. doi: 10.1158/0008-5472.CAN-05-1069. [DOI] [PubMed] [Google Scholar]
  • [43].Reedijk M, Pinnaduwage D, Dickson BC, Mulligan AM, Zhang H, Bull SB, O’Malley FP, Egan SE, Andrulis IL. JAG1 expression is associated with a basal phenotype and recurrence in lymph node-negative breast cancer. Breast Cancer Res.Treat. 2007 doi: 10.1007/s10549-007-9805-3. [DOI] [PubMed] [Google Scholar]
  • [44].Dickson BC, Mulligan AM, Zhang H, Lockwood G, O’Malley FP, Egan SE, Reedijk M. High-level JAG1 mRNA and protein predict poor outcome in breast cancer. Mod.Pathol. 2007;20:685–693. doi: 10.1038/modpathol.3800785. [DOI] [PubMed] [Google Scholar]
  • [45].Shi TP, Xu H, Wei JF, Ai X, Ma X, Wang BJ, Ju ZH, Zhang GX, Wang C, Wu ZQ, Zhang X. Association of low expression of notch-1 and jagged-1 in human papillary bladder cancer and shorter survival. J.Urol. 2008;180:361–366. doi: 10.1016/j.juro.2008.02.037. [DOI] [PubMed] [Google Scholar]
  • [46].Yeasmin S, Nakayama K, Rahman MT, Rahman M, Ishikawa M, Iida K, Ohtuski Y, Kobayashi H, Nakayama S, Miyazaki K. Expression of nuclear Notch3 in cervical squamous cell carcinomas and its association with adverse clinical outcomes. Gynecol.Oncol. 2010 doi: 10.1016/j.ygyno.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • [47].Wang Z, Li Y, Banerjee S, Sarkar FH. Emerging role of Notch in stem cells and cancer. Cancer Lett. 2009;279:8–12. doi: 10.1016/j.canlet.2008.09.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Broxterman HJ, Gotink KJ, Verheul HM. Understanding the causes of multidrug resistance in cancer: a comparison of doxorubicin and sunitinib. Drug Resist.Updat. 2009;12:114–126. doi: 10.1016/j.drup.2009.07.001. [DOI] [PubMed] [Google Scholar]
  • [49].Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat.Rev.Drug Discov. 2006;5:219–234. doi: 10.1038/nrd1984. [DOI] [PubMed] [Google Scholar]
  • [50].Gottesman MM. Mechanisms of cancer drug resistance. Annu.Rev.Med. 2002;53:615–627. doi: 10.1146/annurev.med.53.082901.103929. [DOI] [PubMed] [Google Scholar]
  • [51].Bardelli A, Siena S. Molecular mechanisms of resistance to cetuximab and panitumumab in colorectal cancer. J.Clin.Oncol. 2010;28:1254–1261. doi: 10.1200/JCO.2009.24.6116. [DOI] [PubMed] [Google Scholar]
  • [52].Liu B, Qu L, Tao H. Cyclo-oxygenase 2 up-regulates the effect of multidrug resistance. Cell Biol.Int. 2010;34:21–25. doi: 10.1042/CBI20090129. [DOI] [PubMed] [Google Scholar]
  • [53].Lopez-Chavez A, Carter CA, Giaccone G. The role of KRAS mutations in resistance to EGFR inhibition in the treatment of cancer. Curr.Opin.Investig.Drugs. 2009;10:1305–1314. [PubMed] [Google Scholar]
  • [54].LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist.Updat. 2008;11:32–50. doi: 10.1016/j.drup.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Hendrickson AW, Haluska P. Resistance pathways relevant to insulin-like growth factor-1 receptor-targeted therapy. Curr.Opin.Investig.Drugs. 2009;10:1032–1040. [PubMed] [Google Scholar]
  • [56].Kono SA, Marshall ME, Ware KE, Heasley LE. The fibroblast growth factor receptor signaling pathway as a mediator of intrinsic resistance to EGFR-specific tyrosine kinase inhibitors in non-small cell lung cancer. Drug Resist.Updat. 2009;12:95–102. doi: 10.1016/j.drup.2009.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Hopper-Borge EA, Nasto RE, Ratushny V, Weiner LM, Golemis EA, Astsaturov I. Mechanisms of tumor resistance to EGFR-targeted therapies. Expert.Opin.Ther.Targets. 2009;13:339–362. doi: 10.1517/14712590902735795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Haagenson KK, Wu GS. The role of MAP kinases and MAP kinase phosphatase-1 in resistance to breast cancer treatment. Cancer Metastasis Rev. 2010;29:143–149. doi: 10.1007/s10555-010-9208-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Lin Y, Bai L, Chen W, Xu S. The NF-kappaB activation pathways, emerging molecular targets for cancer prevention and therapy. Expert.Opin.Ther.Targets. 2010;14:45–55. doi: 10.1517/14728220903431069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Jiang BH, Liu LZ. Role of mTOR in anticancer drug resistance: perspectives for improved drug treatment. Drug Resist.Updat. 2008;11:63–76. doi: 10.1016/j.drup.2008.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Mehta K, Osipo C. Trastuzumab resistance: role for Notch signaling. Scientific World Journal. 2009;9:1438–1448. doi: 10.1100/tsw.2009.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–890. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
  • [63].Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat.Rev.Mol.Cell Biol. 2006;7:131–142. doi: 10.1038/nrm1835. [DOI] [PubMed] [Google Scholar]
  • [64].Yauch RL, Januario T, Eberhard DA, Cavet G, Zhu W, Fu L, Pham TQ, Soriano R, Stinson J, Seshagiri S, Modrusan Z, Lin CY, O’Neill V, Amler LC. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin.Cancer Res. 2005;11:8686–8698. doi: 10.1158/1078-0432.CCR-05-1492. [DOI] [PubMed] [Google Scholar]
  • [65].Voulgari A, Pintzas A. Epithelial-mesenchymal transition in cancer metastasis: mechanisms, markers and strategies to overcome drug resistance in the clinic. Biochim.Biophys.Acta. 2009;1796:75–90. doi: 10.1016/j.bbcan.2009.03.002. [DOI] [PubMed] [Google Scholar]
  • [66].Frederick BA, Helfrich BA, Coldren CD, Zheng D, Chan D, Bunn PA, Jr., Raben D. Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma. Mol.Cancer Ther. 2007;6:1683–1691. doi: 10.1158/1535-7163.MCT-07-0138. [DOI] [PubMed] [Google Scholar]
  • [67].Sabbah M, Emami S, Redeuilh G, Julien S, Prevost G, Zimber A, Ouelaa R, Bracke M, De WO, Gespach C. Molecular signature and therapeutic perspective of the epithelial-to-mesenchymal transitions in epithelial cancers. Drug Resist.Updat. 2008;11:123–151. doi: 10.1016/j.drup.2008.07.001. [DOI] [PubMed] [Google Scholar]
  • [68].Kajiyama H, Shibata K, Terauchi M, Yamashita M, Ino K, Nawa A, Kikkawa F. Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int.J.Oncol. 2007;31:277–283. [PubMed] [Google Scholar]
  • [69].Konecny GE, Venkatesan N, Yang G, Dering J, Ginther C, Finn R, Rahmeh M, Fejzo MS, Toft D, Jiang SW, Slamon DJ, Podratz KC. Activity of lapatinib a novel HER2 and EGFR dual kinase inhibitor in human endometrial cancer cells. Br.J.Cancer. 2008;98:1076–1084. doi: 10.1038/sj.bjc.6604278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Shah AN, Summy JM, Zhang J, Park SI, Parikh NU, Gallick GE. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann.Surg.Oncol. 2007;14:3629–3637. doi: 10.1245/s10434-007-9583-5. [DOI] [PubMed] [Google Scholar]
  • [71].Yang AD, Fan F, Camp ER, van BG, Liu W, Somcio R, Gray MJ, Cheng H, Hoff PM, Ellis LM. Chronic oxaliplatin resistance induces epithelial-to-mesenchymal transition in colorectal cancer cell lines. Clin.Cancer Res. 2006;12:4147–4153. doi: 10.1158/1078-0432.CCR-06-0038. [DOI] [PubMed] [Google Scholar]
  • [72].Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, Ali S, Abbruzzese JL, Gallick GE, Sarkar FH. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69:2400–2407. doi: 10.1158/0008-5472.CAN-08-4312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Tang C, Ang BT, Pervaiz S. Cancer stem cell: target for anti-cancer therapy. FASEB J. 2007;21:3777–3785. doi: 10.1096/fj.07-8560rev. [DOI] [PubMed] [Google Scholar]
  • [74].Styczynski J, Drewa T. Leukemic stem cells: from metabolic pathways and signaling to a new concept of drug resistance targeting. Acta Biochim.Pol. 2007;54:717–726. [PubMed] [Google Scholar]
  • [75].Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, Lagutina I, Grosveld GC, Osawa M, Nakauchi H, Sorrentino BP. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat.Med. 2001;7:1028–1034. doi: 10.1038/nm0901-1028. [DOI] [PubMed] [Google Scholar]
  • [76].Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–111. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • [77].Phillips TM, Kim K, Vlashi E, McBride WH, Pajonk F. Effects of recombinant erythropoietin on breast cancer-initiating cells. Neoplasia. 2007;9:1122–1129. doi: 10.1593/neo.07694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Farnie G, Clarke RB. Mammary stem cells and breast cancer--role of Notch signalling Stem. Cell Rev. 2007;3:169–175. doi: 10.1007/s12015-007-0023-5. [DOI] [PubMed] [Google Scholar]
  • [79].Patrawala L, Calhoun T, Schneider-Broussard R, Zhou J, Claypool K, Tang DG. Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and A. Cancer Res. 2005;65:6207–6219. doi: 10.1158/0008-5472.CAN-05-0592. [DOI] [PubMed] [Google Scholar]
  • [80].Sarkar FH, Li Y, Wang Z, Kong D, Ali S. Implication of microRNAs in drug resistance for designing novel cancer therapy. Drug Resist.Updat. 2010 doi: 10.1016/j.drup.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Croce CM, Calin GA. miRNAs, cancer, and stem cell division. Cell. 2005;122:6–7. doi: 10.1016/j.cell.2005.06.036. [DOI] [PubMed] [Google Scholar]
  • [82].Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat.Rev.Genet. 2009;10:704–714. doi: 10.1038/nrg2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Rui W, Bing F, Hai-Zhu S, Wei D, Long-Bang C. Identification of microRNA profiles in docetaxel-resistant human non-small cell lung carcinoma cells (SPC-A1) J.Cell Mol.Med. 2010;14:206–214. doi: 10.1111/j.1582-4934.2009.00964.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Song B, Wang Y, Xi Y, Kudo K, Bruheim S, Botchkina GI, Gavin E, Wan Y, Formentini A, Kornmann M, Fodstad O, Ju J. Mechanism of chemoresistance mediated by miR-140 in human osteosarcoma and colon cancer cells. Oncogene. 2009;28:4065–4074. doi: 10.1038/onc.2009.274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Fujita Y, Kojima K, Hamada N, Ohhashi R, Akao Y, Nozawa Y, Deguchi T, Ito M. Effects of miR-34a on cell growth and chemoresistance in prostate cancer PC3 cells. Biochem.Biophys.Res.Commun. 2008;377:114–119. doi: 10.1016/j.bbrc.2008.09.086. [DOI] [PubMed] [Google Scholar]
  • [86].Hermeking H. The miR-34 family in cancer and apoptosis Cell. Death.Differ. 2010;17:193–199. doi: 10.1038/cdd.2009.56. [DOI] [PubMed] [Google Scholar]
  • [87].Li Y, Guessous F, Zhang Y, Dipierro C, Kefas B, Johnson E, Marcinkiewicz L, Jiang J, Yang Y, Schmittgen TD, Lopes B, Schiff D, Purow B, Abounader R. MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res. 2009 doi: 10.1158/0008-5472.CAN-09-0529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Ji Q, Hao X, Meng Y, Zhang M, Desano J, Fan D, Xu L. Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres. BMC.Cancer. 2008;8:266. doi: 10.1186/1471-2407-8-266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].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. doi: 10.1371/journal.pone.0006816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Chen GQ, Zhao ZW, Zhou HY, Liu YJ, Yang HJ. Systematic analysis of microRNA involved in resistance of the MCF-7 human breast cancer cell to doxorubicin. Med.Oncol. 2010;27:406–415. doi: 10.1007/s12032-009-9225-9. [DOI] [PubMed] [Google Scholar]
  • [91].Datta J, Kutay H, Nasser MW, Nuovo GJ, Wang B, Majumder S, Liu CG, Volinia S, Croce CM, Schmittgen TD, Ghoshal K, Jacob ST. Methylation mediated silencing of MicroRNA-1 gene and its role in hepatocellular carcinogenesis. Cancer Res. 2008;68:5049–5058. doi: 10.1158/0008-5472.CAN-07-6655. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [92].Nasser MW, Datta J, Nuovo G, Kutay H, Motiwala T, Majumder S, Wang B, Suster S, Jacob ST, Ghoshal K. Down-regulation of micro-RNA-1 (miR-1) in lung cancer. Suppression of tumorigenic property of lung cancer cells and their sensitization to doxorubicin-induced apoptosis by miR-1. J.Biol.Chem. 2008;283:33394–33405. doi: 10.1074/jbc.M804788200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [93].Ivey KN, Muth A, Arnold J, King FW, Yeh RF, Fish JE, Hsiao EC, Schwartz RJ, Conklin BR, Bernstein HS, Srivastava D. MicroRNA regulation of cell lineages in mouse and human embryonic stem cells. Cell Stem Cell. 2008;2:219–229. doi: 10.1016/j.stem.2008.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Kwon C, Han Z, Olson EN, Srivastava D. MicroRNA1 influences cardiac differentiation in Drosophila and regulates Notch signaling. Proc.Natl.Acad.Sci.U.S.A. 2005;102:18986–18991. doi: 10.1073/pnas.0509535102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Adam L, Zhong M, Choi W, Qi W, Nicoloso M, Arora A, Calin G, Wang H, Siefker-Radtke A, McConkey D, Bar-Eli M, Dinney C. miR-200 expression regulates epithelial-to-mesenchymal transition in bladder cancer cells and reverses resistance to epidermal growth factor receptor therapy. Clin.Cancer Res. 2009;15:5060–5072. doi: 10.1158/1078-0432.CCR-08-2245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Cochrane DR, Spoelstra NS, Howe EN, Nordeen SK, Richer JK. MicroRNA-200c mitigates invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic agents. Mol.Cancer Ther. 2009 doi: 10.1158/1535-7163.MCT-08-1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Li Y, Vandenboom TG, Kong D, Wang Z, Ali S, Philip PA, Sarkar FH. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009;69:6704–6712. doi: 10.1158/0008-5472.CAN-09-1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Zhang ZP, Sun YL, Fu L, Gu F, Zhang L, Hao XS. Correlation of Notch1 expression and activation to cisplatin-sensitivity of head and neck squamous cell carcinoma. Ai.Zheng. 2009;28:100–103. [PubMed] [Google Scholar]
  • [99].Gu F, Ma Y, Zhang Z, Zhao J, Kobayashi H, Zhang L, Fu L. Expression of Stat3 and Notch1 is associated with cisplatin resistance in head and neck squamous cell carcinoma. Oncol.Rep. 2010;23:671–676. doi: 10.3892/or_00000683. [DOI] [PubMed] [Google Scholar]
  • [100].Aleksic T, Feller SM. Gamma-secretase inhibition combined with platinum compounds enhances cell death in a large subset of colorectal cancer cells. Cell Commun.Signal. 2008;6:8. doi: 10.1186/1478-811X-6-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH, Nephew KP. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008;68:4311–4320. doi: 10.1158/0008-5472.CAN-08-0364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Yao J, Qian C. Inhibition of Notch3 enhances sensitivity to gemcitabine in pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway. Med.Oncol. 2009 doi: 10.1007/s12032-009-9326-5. [DOI] [PubMed] [Google Scholar]
  • [103].Chiuri VE, Silvestris N, Lorusso V, Tinelli A. Efficacy and safety of the combination of docetaxel (Taxotere) with targeted therapies in the treatment of solid malignancies. Curr.Drug Targets. 2009;10:982–1000. doi: 10.2174/138945009789578007. [DOI] [PubMed] [Google Scholar]
  • [104].Zang S, Chen F, Dai J, Guo D, Tse W, Qu X, Ma D, Ji C. RNAi-mediated knockdown of Notch-1 leads to cell growth inhibition and enhanced chemosensitivity in human breast cancer. Oncol.Rep. 2010;23:893–899. doi: 10.3892/or_00000712. [DOI] [PubMed] [Google Scholar]
  • [105].Yeh TS, Hsieh RH, Shen SC, Wang SH, Tseng MJ, Shih CM, Lin JJ. Nuclear betaII-tubulin associates with the activated notch receptor to modulate notch signaling. Cancer Res. 2004;64:8334–8340. doi: 10.1158/0008-5472.CAN-04-2197. [DOI] [PubMed] [Google Scholar]
  • [106].Akiyoshi T, Nakamura M, Yanai K, Nagai S, Wada J, Koga K, Nakashima H, Sato N, Tanaka M, Katano M. Gamma-secretase inhibitors enhance taxane-induced mitotic arrest and apoptosis in colon cancer cells. Gastroenterology. 2008;134:131–144. doi: 10.1053/j.gastro.2007.10.008. [DOI] [PubMed] [Google Scholar]
  • [107].Mine T, Matsueda S, Gao H, Li Y, Wong KK, Peoples GE, Ferrone S, Ioannides CG. Created Gli-1 duplex short-RNA (i-Gli-RNA) eliminates CD44 Hi progenitors of taxol-resistant ovarian cancer cells. Oncol.Rep. 2010;23:1537–1543. doi: 10.3892/or_00000793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Meng RD, Shelton CC, Li YM, Qin LX, Notterman D, Paty PB, Schwartz GK. gamma-Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Cancer Res. 2009;69:573–582. doi: 10.1158/0008-5472.CAN-08-2088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Magnifico A, Albano L, Campaner S, Delia D, Castiglioni F, Gasparini P, Sozzi G, Fontanella E, Menard S, Tagliabue E. Tumor-initiating cells of HER2-positive carcinoma cell lines express the highest oncoprotein levels and are sensitive to trastuzumab. Clin.Cancer Res. 2009;15:2010–2021. doi: 10.1158/1078-0432.CCR-08-1327. [DOI] [PubMed] [Google Scholar]
  • [110].Osipo C, Patel P, Rizzo P, Clementz AG, Hao L, Golde TE, Miele L. ErbB-2 inhibition activates Notch-1 and sensitizes breast cancer cells to a gamma-secretase inhibitor. Oncogene. 2008;27:5019–5032. doi: 10.1038/onc.2008.149. [DOI] [PubMed] [Google Scholar]
  • [111].Dai J, Ma D, Zang S, Guo D, Qu X, Ye J, Ji C. Cross-talk between Notch and EGFR signaling in human breast cancer cells. Cancer Invest. 2009;27:533–540. doi: 10.1080/07357900802563036. [DOI] [PubMed] [Google Scholar]
  • [112].Konishi J, Kawaguchi KS, Vo H, Haruki N, Gonzalez A, Carbone DP, Dang TP. Gamma-secretase inhibitor prevents Notch3 activation and reduces proliferation in human lung cancers. Cancer Res. 2007;67:8051–8057. doi: 10.1158/0008-5472.CAN-07-1022. [DOI] [PubMed] [Google Scholar]
  • [113].Piechocki MP, Yoo GH, Dibbley SK, Lonardo F. Breast cancer expressing the activated HER2/neu is sensitive to gefitinib in vitro and in vivo and acquires resistance through a novel point mutation in the HER2/neu. Cancer Res. 2007;67:6825–6843. doi: 10.1158/0008-5472.CAN-07-0765. [DOI] [PubMed] [Google Scholar]
  • [114].Giovannini C, Gramantieri L, Chieco P, Minguzzi M, Lago F, Pianetti S, Ramazzotti E, Marcu KB, Bolondi L. Selective ablation of Notch3 in HCC enhances doxorubicin’s death promoting effect by a p53 dependent mechanism. J.Hepatol. 2009;50:969–979. doi: 10.1016/j.jhep.2008.12.032. [DOI] [PubMed] [Google Scholar]
  • [115].Nefedova Y, Cheng P, Alsina M, Dalton WS, Gabrilovich DI. Involvement of Notch-1 signaling in bone marrow stroma-mediated de novo drug resistance of myeloma and other malignant lymphoid cell lines. Blood. 2004;103:3503–3510. doi: 10.1182/blood-2003-07-2340. [DOI] [PubMed] [Google Scholar]
  • [116].Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008;111:2220–2229. doi: 10.1182/blood-2007-07-102632. [DOI] [PubMed] [Google Scholar]
  • [117].Wang Z, Zhang Y, Banerjee S, Li Y, Sarkar FH. Inhibition of nuclear factor kappab activity by genistein is mediated via Notch-1 signaling pathway in pancreatic cancer cells. Int.J.Cancer. 2006;118:1930–1936. doi: 10.1002/ijc.21589. [DOI] [PubMed] [Google Scholar]
  • [118].Wang Z, Zhang Y, Banerjee S, Li Y, Sarkar FH. Notch-1 down-regulation by curcumin is associated with the inhibition of cell growth and the induction of apoptosis in pancreatic cancer cells. Cancer. 2006;106:2503–2513. doi: 10.1002/cncr.21904. [DOI] [PubMed] [Google Scholar]
  • [119].Wang Z, Desmoulin S, Banerjee S, Kong D, Li Y, Deraniyagala RL, Abbruzzese J, Sarkar FH. Synergistic effects of multiple natural products in pancreatic cancer cells. Life Sci. 2008;83:293–300. doi: 10.1016/j.lfs.2008.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Cecchinato V, Chiaramonte R, Nizzardo M, Cristofaro B, Basile A, Sherbet GV, Comi P. Resveratrol-induced apoptosis in human T-cell acute lymphoblastic leukaemia MOLT-4 cells. Biochem.Pharmacol. 2007;74:1568–1574. doi: 10.1016/j.bcp.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • [121].Zhang Z, Wang Y, Yao R, Li J, Yan Y, La RM, Lemon WL, Grubbs CJ, Lubet RA, You M. Cancer chemopreventive activity of a mixture of Chinese herbs (antitumor B) in mouse lung tumor models. Oncogene. 2004;23:3841–3850. doi: 10.1038/sj.onc.1207496. [DOI] [PubMed] [Google Scholar]
  • [122].Koduru S, Kumar R, Srinivasan S, Evers MB, Damodaran C. Notch-1 inhibition by Withaferin-A: a therapeutic target against colon carcinogenesis. Mol.Cancer Ther. 2010;9:202–210. doi: 10.1158/1535-7163.MCT-09-0771. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES