Next Article in Journal
Dexfenfluramine and Pergolide Cause Heart Valve Disease via Valve Metabolic Reprogramming and Ongoing Matrix Remodeling
Next Article in Special Issue
HP1γ Sensitizes Cervical Cancer Cells to Cisplatin through the Suppression of UBE2L3
Previous Article in Journal
Controlling J-Aggregates Formation and Chirality Induction through Demetallation of a Zinc(II) Water Soluble Porphyrin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Association of the Epithelial–Mesenchymal Transition (EMT) with Cisplatin Resistance

1
Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
2
Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
3
Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
4
Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran
5
Kazerun Health Technology Incubator, Shiraz University of Medical Sciences, Shiraz 1433671348, Iran
6
Department of Genetic Science, Tehran Medical Science Branch, Islamic Azad University, Tehran 19168931813, Iran
7
Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 1355576169, Iran
8
Health Informatics Lab, Metropolitan College, Boston University, Boston, MA 02215, USA
9
Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(11), 4002; https://doi.org/10.3390/ijms21114002
Submission received: 13 April 2020 / Revised: 14 May 2020 / Accepted: 26 May 2020 / Published: 3 June 2020
(This article belongs to the Special Issue Cisplatin in Cancer Therapy: Molecular Mechanisms of Action 2.0)

Abstract

:
Therapy resistance is a characteristic of cancer cells that significantly reduces the effectiveness of drugs. Despite the popularity of cisplatin (CP) as a chemotherapeutic agent, which is widely used in the treatment of various types of cancer, resistance of cancer cells to CP chemotherapy has been extensively observed. Among various reported mechanism(s), the epithelial–mesenchymal transition (EMT) process can significantly contribute to chemoresistance by converting the motionless epithelial cells into mobile mesenchymal cells and altering cell–cell adhesion as well as the cellular extracellular matrix, leading to invasion of tumor cells. By analyzing the impact of the different molecular pathways such as microRNAs, long non-coding RNAs, nuclear factor-κB (NF-ĸB), phosphoinositide 3-kinase-related protein kinase (PI3K)/Akt, mammalian target rapamycin (mTOR), and Wnt, which play an important role in resistance exhibited to CP therapy, we first give an introduction about the EMT mechanism and its role in drug resistance. We then focus specifically on the molecular pathways involved in drug resistance and the pharmacological strategies that can be used to mitigate this resistance. Overall, we highlight the various targeted signaling pathways that could be considered in future studies to pave the way for the inhibition of EMT-mediated resistance displayed by tumor cells in response to CP exposure.

1. Introduction

Cancer is still an increasing challenge for public health and is the second leading cause of death worldwide [1]. The newest statistics related to the incidence rate of cancer in the United States demonstrate that each day, 4950 people are diagnosed with cancer and its annual incidence rate is 1,806,590. The common cancers are different in men and women. Prostate, lung, and colorectal cancers are among the most common cancers in males, while breast, lung, and colorectal cancers are prevalent in females. On the other hand, due to enhanced life expectancy, the population of the world is going to be older in the following years. For instance, in United States, the population of aged people is suggested to be 19 million in 2060, which is significantly higher compared to the 6.6 million aged people in 2016 [2]. This is related to the fact that the possibility of cancer development is higher in aged people. In addition, it is less likely to diagnose aged patients with cancer at early and local stages [3]. These statements are in agreement with the fact that dealing with cancer is of the utmost importance in the modern world and it demands extensive research into finding novel treatments for this life threatening disorder [4,5,6,7,8,9,10,11].
Currently, regardless of diagnostic methods, a number of treatment strategies can be applied for cancer therapy. Chemotherapy is a first-line treatment for cancer due to its minimally invasive nature and satisfactory results in clinical trials [12]. However, patients with cancer and physicians have faced a growing difficulty in chemotherapy, known as chemoresistance [13]. Studies have shown that frequent application of chemotherapeutic agents with high doses has led to the emergence of chemoresistance. After the appearance of chemoresistance, research was performed to discover new chemotherapeutic agents to improve chemotherapy efficacy. Very soon, it was found that cancer cells develop resistance to chemotherapeutic agents by switching between molecular pathways and mechanisms to ensure their proliferation and malignancy [14]. Currently, the focus has been directed towards using anti-tumor drugs along with chemotherapeutic agents, in addition to gene therapy [15,16]. Finding an effective regimen for inhibition of chemoresistance in cancer therapy relies on revealing the molecular pathways and mechanisms involved in development of resistance of cancer cells to chemotherapy. To date, thanks to extensive research in the field of chemoresistance, numerous targets have been explored for chemoresistance/chemosensitivity [17,18,19,20].
In the present review, we investigate the role of epithelial–mesenchymal transition (EMT) in the emergence of cisplatin (CP) resistance. CP is a key member of platinum compounds with excellent anti-tumor activity against different cancers such as breast cancer, prostate cancer, bladder cancer, lung cancer, brain tumors, and so on [21,22,23]. The rational reasons for selection of CP are as follows: (1) its anti-tumor activity has been extensively examined, particularly in clinical trials [24,25,26], and (2) the molecular mechanisms and pathways involved in CP resistance/sensitivity have been explored [27,28,29]. Consequently, it has been possible to implicate the process of EMT in drug resistance and directing further studies towards targeting EMT in suppressing resistance developed against CP treatment.

2. Cisplatin in Cancer Therapy

Chemotherapy is still one of the most common modalities used in cancer therapy [30]. A variety of chemical agents, such as alkylating compounds and antimetabolites, have been applied in cancer therapy to suppress the progression and invasion of cancer cells by affecting different hallmarks of cancer cells [31]. More recently, platinum-based drugs have attracted much attention in the treatment of malignant tumors because of their effectiveness in targeting different signaling pathways and mechanisms that control cancer progression [32]. CP is one of the most common types of platinum-based medicines, which has shown great potential in the treatment of several types of cancer, including brain tumors [33,34], lung cancer [35], breast cancer [36], bladder cancer [37], prostate cancer [38], oral cancer [39], and neck cancer [40]. After the discovery of this anti-tumor agent in 1978, its application has undergone a tremendous increase [41,42]. A very good example of its effectiveness has been shown when combined with Clarithromycin to suppress the invasion and proliferation of ovarian cancer cells, which elevates apoptotic cell death by increasing the production of reactive oxygen species (ROS) [43]. A combination of CP and a 5-aminopyrazole derivative lead compound (BC-7) stimulates M-phase arrest and prevents the proliferation of cervical cancer cells [44]. It is worth mentioning that CP and docetaxel (DOX) can be administered prior to surgery to increase the survival resection rate, reduce intraoperative blood loss, and extend the patient’s survival time. Chemotherapy and radiotherapy can be used simultaneously (CP, DOX, and 5-fluorouracil) to effectively treat advanced esophageal cancer [45].
Nanoparticles (NPs) are promising candidates for the targeted administration of CP. The use of multifunctional NPs may also improve the anti-tumor activity of CP [46]. It has been shown that NPs offer the possibility of administering CP together with other anti-tumor agents to increase the number of cells undergoing apoptosis [47]. It has been proposed that CP induces apoptotic cell death through the mitochondrial signaling pathway and activation of the ROS/AMP-activated protein kinase signaling pathway (AMPK). Administration of β-elemene in combination with CP enhances the effect on the ROS/AMPK axis and consequently improves anti-tumor activity [48]. These studies show that CP is effective in inhibiting the growth and proliferation of cancer cells. Overall, CP induces DNA damage in cancer cells and suppresses cancer cell proliferation and viability. The combination of CP with other chemotherapeutic agents has shown great potential in cancer therapy [49,50,51,52,53,54].
However, despite its great potential in the treatment of cancer, the use of CP in cancer therapy has a number of disadvantages [55]. One of these is adverse effects on the body’s organs and systems resulting from increased production of ROS and stimulation of mitochondrial dysfunction [56,57,58,59]. Gastrointestinal disorders, ototoxicity, neurotoxicity and hepatotoxicity are some of the toxic effects of CP [60]. Regardless of the undesirable effects of CP, which can be mitigated by antioxidant compounds [61,62,63], the main difficulty with CP therapy is the resistance developed by cancer cells against this drug. This could be due to the use of dynamic and flexible molecular pathways by cancer cells to improve their proliferation and survival [64,65,66,67]. It has been shown that frequent use of CP at high doses is associated with the reduced effectiveness of CP in suppressing invasion and malignancy of tumor cells [68,69]. A number of reasons have been attributed for this issue and in this paper, we have focussed on the role of EMT in the development of resistance to CP treatment (Figure 1).

3. Cisplatin Resistance

Recently published articles have shed some light on the molecular pathways and mechanisms involved in CP resistance. A number of strategies have been applied to enhance the sensitivity of cancer cells in CP therapy. Autophagy is a degradation process that serves to reduce stress and maintain proper cell function under physiological conditions [70,71,72]. This mechanism has a controversial function in cancer; on one hand autophagy in cancer therapy suppresses the viability and proliferation of cancer cells [73,74]. However, studies have shown that autophagy may be involved in increasing the survival and propagation of cancer cells, and can also mediate resistance to chemotherapy [75,76,77,78]. The long non-coding RNA (lncRNA) SNHG14 is able to induce CP resistance in colorectal cancer cells by inducing autophagy activation via ATG14 upregulation [79]. The autophagy mechanism also prevents the repolarization of tumor-associated macrophages (TAMs) to stimulate chemoresistance against CP therapy [80].
A number of strategies have been used to increase the sensitivity of cancer cells to CP therapy. To suppress autophagy during CP therapy, propofol was administered as an anti-tumor agent [81] in combination with CP to inhibit autophagy by downregulating the MALAT1/microRNA (miR)-30e/ATG5 axis, resulting in sensitivity of gastric cancer cells (GC cells) to CP therapy [82]. Oridonin supplementation can suppress autophagy by beclin-1 downregulation to increase the efficacy of CP therapy in ovarian cancer cells [83]. In addition, upregulation of miR-29c-3p and miR-138-5p inhibits autophagy and sensitizes cancer cells to CP-induced autophagy [84,85]. It is worth mentioning that the involvement of autophagy in CP resistance has been controversial. It was mentioned that stimulation of autophagy may be associated with CP resistance. However, there are a number of studies, which indicate that induction of autophagy can suppress CP resistance. For example, arctigenin sensitizes colon cancer cells to CP therapy by autophagy induction [86]. These studies underline the fact that the autophagy mechanism plays a crucial role in CP resistance. It appears that the involvement of autophagy in CP resistance varies between different cancer cells (context dependent).
In addition to autophagy, miRs may contribute to resistance to CP therapy. miRs are short non-coding RNAs that are able to regulate vital biological mechanisms such as apoptosis and differentiation [87,88,89]. The potential role of miRs in regulating the migration and invasion of cancer cells has been demonstrated [90,91]. In particular, lncRNAs can act as upstream modulators of miRs [92,93]. Thus, there is a close relationship between lncRNAs, miRs, and CP resistance. The newly published articles have also shown the involvement of miRs and lncRNAs in CP resistance. In oral squamous cell carcinoma (OSCC), the expression of miR-132 is downregulated, which mediates the resistance of OSCC to CP therapy [94]. The downregulation of miR-182-5p induces resistance of GC cells to CP therapy [95]. Notably, OSCC reduces the expression of miR-26b to inhibit CP-mediated DNA damage and induce resistance to chemotherapy [96]. LncRNAs are also involved in CP therapy, and by amplification/reduction of their expression it is possible to reduce the viability and proliferation of cancer cells and sensitize them to CP therapy [97,98,99]. Other molecular signaling pathways such as the Wnt signaling pathway [38,100], phosphoinositide 3-kinase-related protein kinase (PI3K) [77], nuclear factor erythroid 2-related factor 2 (Nrf2) [101], and sirtuin 1 (SIRT1) [102] may also be involved in the resistance of cancer cells to CP therapy. These studies show that CP resistance is a common phenomenon that is due to the increased use of CP in cancer therapy. Interestingly, various genetic strategies have been used to modify the expression of genes involved in CP resistance and increase the effectiveness of this chemotherapeutic agent. In addition, pharmacologically stimulating the sensitization of cancer cells by other chemosensitizers can also be important for increasing the efficacy of CP therapy.

4. EMT Process in Healthy and Cancerous Tissues

EMT is a physiological process necessary for embryonic development and is involved in mesoderm formation and neural crest removal [92,103,104,105]. This highly conserved mechanism is able to control morphogenesis in other cells [104,106]. Cancer cells apply EMT mechanism to enhance their migration and invasion, and consequently, ensure their survival and malignancy [107,108,109]. The EMT mechanism is reversible and can be regulated via a variety of molecular signals such as lncRNA, miR, Akt, and PI3K signaling pathways [110,111,112,113,114]. Both normal and cancer epithelial cells can use the EMT mechanism in the way of elevating their migratory ability and diffusion into neighboring tissues [115,116,117]. During EMT mechanisms, stationary epithelial cells are transformed into mobile mesenchymal cells [118,119]. In EMT, a spindle-like morphology is acquired, cell–cell adhesions are disrupted, and the polarity is changed from apical–basal polarity into front-end–back-end polarity [76,120]. Two proteins known as E-cadherin and N-cadherin are suggested to play a significant role in EMT mechanisms [121,122]. The cadherin proteins shape a cadherin–catenin adhesion complex by attachment into β- and α-catenin via their cytoplasmic tails [123].
Increasing evidence demonstrates the possible tumor suppressor function of E-cadherin and its upregulation in normal epithelial cells [123,124]. In non-cancerous epithelial cells, E-cadherin preserves epithelial phenotype and tissue homeostasis via targeting various molecular pathways. The EMT is an important mechanism for physiological processes such as embryogenesis, wound healing, fibrosis, and so on. Thus, modulation of EMT can also be of importance in treatment of non-cancerous diseases [125,126,127]. It has been shown that E-cadherin downregulation may be associated with the development of a number of cancers such as breast cancer [128], lung cancer [129], skin cancer [130], and GC [131]. This can be related to the EMT mechanism. It is held that a decrease in E-cadherin triggers the EMT mechanism, leading to an increased invasion and migration of tumor cells [132]. A variety of studies have revealed that E-cadherin upregulation may be associated with less migratory ability of cancer cells and their sensitivity to cell death that can be attributed to the EMT mechanism inhibition [133,134,135,136]. These studies highlight the fact that during EMT mechanism, a decrease occurs in E-cadherin protein level to ensure the metastasis and invasion of tumor cells. The story for N-cadherin is completely different. The N-cadherin protein is at the minimum level in non-cancerous epithelial cells, whereas its abundance is evident in cancer epithelial cells [137]. The upregulation of N-cadherin in normal epithelial cells indicates an imminent EMT and development of cancer [138]. N-cadherin contributes to the metastasis and dissemination of epithelial cells from other cells during EMT mechanism [139,140]. Thus, it is conspicuous that N-cadherin and E-cadherin proteins undergo upregulation/downregulation during EMT mechanism, respectively [141,142].
The EMT mechanism is involved in the transformation of static epithelial cells into mobile mesenchymal cells. This mechanism is of crucial importance for the metastasis and migration of tumor cells. There are a number of interactions between the molecular pathways and the EMT mechanism to promote the invasion of cancer cells. The Musashi RNA binding protein 2 (MSI2) contributes to cell differentiation and regulates stem cells and asymmetric division [143]. The collected data show that abnormal expression of MSI2 is associated with the development of cancer [144,145,146]. In pancreatic cancer cells, MSI2 triggers the zinc-finger E-box binding homeobox (ZEB)1/ERK/MAPK axis to increase EMT in these cancer cells, resulting in increased migration and invasion [147]. It appears that miR-1228 increases the proliferation and metastasis of ovarian cancer cells by EMT induction [147].
Four and a half LIM domain proteins (FHL) are evolutionary conserved domains involved in the interaction with molecular pathways such as Wnt [148], Ras [149], and tumor growth factor (TGF) [150], to modulate the growth and invasion of cancer cells. FHL proteins have been shown to regulate EMT in cancer cells [151]. By preventing the ubiquitination of Snail 1 and Twist1, FHL3 stimulates EMT to ensure the malignancy of pancreatic cancer cells [152]. Yes-associated protein 1 (YAP1) and tafazzin (TAZ1) are factors involved in the initiation of cancer development [153]. These proteins exert different strategies in the development of cancer and promoting cancer rigidity. In a recently published article, it was found that stimulation of YAP1/TAZ1 translocation is associated with increased viability of colorectal cancer cells (CRCs). YAP1 and TAZ1 are the downstream mediators of the LINCO1413/hnRNP-k/ZEB1 axis and their induction facilitates the EMT mechanism [154].
The ubiquitin-conjugating enzyme E2O (UBE2O) is expressed in tissues such as brain, heart, liver, and muscle [155]. This protein has several vital functions and its association with the EMT mechanism is of interest. In breast cancer cells, UBE2O degrades AMPK2 by enhancing its ubiquitination and paves the way for the activation of the mammalian target rapamycin (mTOR). This axis increases the proliferation and metastasis of breast cancer cells by inducing the EMT mechanism [156]. MiR-4472 is involved in chemoresistance and cancer development [157]. The study of the molecular pathways has shown that the EMT mechanism can be induced by miR-4472 during cancer progression [158]. Overall, stimulation of the EMT mechanism is a positive factor in tumor metastasis and migration, while it is associated with the poor prognosis of cancer patients [159,160].
EMT can contribute to the resistance of tumor cells to chemotherapy. Afatinib is an anti-tumor agent of a second-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, which is commonly used to treat lung cancer [161,162]. Lung cancer cells may use the EMT mechanism to enhance their migration and malignancy and to achieve resistance to afatinib. To provide effective treatment for lung cancer, we should use a combination of afatinib and EMT targets such as histone deacetylase inhibitors [163]. In fact, another study emphasized that downward regulation of Twist1 can be seen as a promising strategy to sensitize lung cancer cells to EMT-induced chemotherapy-induced resistance [164]. Small molecule inhibitors targeting FGFR1 are widely used in cancer treatment. AZD4547 is one such molecule with the ability to reduce the viability and proliferation of cancer cells. EMT induction has significantly reduced the effectiveness of AZD4547 in cancer therapy [165]. In addition, miRs can significantly contribute to the activation of EMT and chemoresistance. Paclitaxel (PTX) is a chemotherapeutic agent discovered in 1963, isolated from the bark of the Pacific yew Taxus brerifolia [166]. This compound has shown great potential in the treatment of various types of cancer [167]. In recent years, resistance to PTX has been a common phenomenon. It is believed that an increase in the expression of miR-181a induces the EMT mechanism and mediates resistance of ovarian cancer cells to PTX therapy [168]. Overall, the studies confirm that the EMT mechanism is not only crucial for the progression and malignancy of cancer cells, but also induces resistance to chemotherapy and reduces apoptotic cell death [169,170,171,172].

5. Cisplatin Induces EMT-Mediated Cancer Chemoresistance

TAMs are one of the main infiltrations of immune cells into the microenvironment of the tumor and they interact with solid tumors since they are involved in the metastasis of cancer cells [173,174,175,176]. Classically activated macrophages (CAMs) and alternatively activated macrophages are two main types of TAMs [177]. In particular, CAMs appear to promote the migration and malignancy of cancer cells such as hepatocellular carcinoma (HCC), ovarian, and oral cancers [178,179,180]. Chemotherapy with CP is associated with an increase in the migration ability of CAMs. The study of molecular markers shows that the induction of CAMs by CP triggers the EMT mechanism. It is held that CP just stimulates CAMs to secrete chemokine ligand 20 (CCL20) without affecting their phenotype [181]. The chemokine ligand 20 (CCL20) is able to recruit T helper cells to maintain the immunosuppressive microenvironment and ensure the progression of the cancer [182,183,184]. The chemokine receptor 6 (CCR6) is a secondary target of CCL20 that induces cancer migration and metastasis [185]. Interestingly, chemotherapy with CP stimulates CAMs to secrete CCL20, then the CCL20/CCR6 axis enhances tumor cell migration and induces the EMT mechanism, thereby leading to EMT-mediated drug resistance [181].
It appears that not a single factor is responsible for the resistance of cancer cells to CP chemotherapy and a number of diverse mechanism(s) may be involved (summarized in Table 1). The ataxia telangiectasia mutated (ATM) is a key member of phosphoinositide 3-kinase-related protein kinase (PI3K) family, which participates in DNA damage response. Endogenous factors such as ROS and exogenous factors including irradiation are able to induce ATM activation. ATM can subsequently trigger cell cycle checkpoint machinery, DNA repair or apoptosis in response to the aforementioned stimuli [186,187]. On the other hand, Schlafen 11 (SLFN11) is an onco-suppressor factor that enhances sensitivity of cancer cells into anti-tumor agents [188]. Both ATM upregulation and SLFN11 downregulation can activate EMT to stimulate tumor cells’ resistance to CP [189]. CP is also able to increase EMT markers such as Snail to reduce the sensitivity of tumor cells and ensure their migration and metastasis [190]. Although high doses of CP over a long period could induce CP resistance, an experiment conducted by Liu and colleagues showed that short and low concentrations of CP via affecting the EMT can also induce resistance in tumor cells [191]. In addition, CP induces EMT via the activation of oncogenic NF-κB signaling pathway [192]. The discovery of the underlying molecular signaling pathway may therefore pave the way for more targeted influencing and increasing the efficacy of CP in chemotherapy.
Autophagy is considered as a highly conserved mechanism when cells have to degrade the additional toxic and aged organelles and macromolecules [193,194]. The role of autophagy in cancer still remains unclear [195,196]. It appears that the involvement of autophagy in cancer progression/inhibition depends on the context and the type of cancer [197,198]. Recently published articles have shown that autophagy can function as a cytoprotective mechanism to improve the viability and proliferation of tumor cells and induce resistance to chemotherapy [199,200,201,202]. The same applies to CP. It was noted that the administration of CP is associated with the induction of autophagy. After activation of autophagy, the migration of cancer cells is upregulated, and the cancer cells are able to acquire EMT (enhanced levels of vimentin). Inhibition of autophagy is believed to interfere with the EMT mechanism and reduce the malignancy and invasion of tumor cells [203]. Therefore, this aspect of autophagy should be considered in chemotherapy.

6. Strategies to Attenuate EMT-Related Cisplatin Resistance

6.1. Cluster of Differentiation Role

CD13 is a transmembrane glycoprotein with the ability to activate tumor angiogenesis and adhesion [204]. The upregulation of CD13 is associated with the poor prognosis of GC [205]. On the other hand, the epithelial membrane protein 3 (EMP3) is a peripheral myelin protein whose overexpression can increase the invasive ability of cancer cells [206,207]. There is a close relationship between CD13, EMP3, EMT, and CP resistance, so that increased expression of CD13 increases the expression of EMP3 to stimulate the PI3K/Akt/NF-κB axis. This leads to induction of the EMT mechanism and resistance of GC cells to apoptosis and inhibitory effects of CP. Ubenimex is an inhibitor of CD13 that suppresses the signaling pathway mediated by CD13 to inhibit CP resistance by downregulating EMT. Mechanistically, ubenimex enhances CpG island hypermethylation for which CD13 inhibition may be needed. Subsequently, a disruption occurs in downstream signaling pathway PI3K/Akt/NF-κB that suppresses both autophagy and EMT to sensitize cancer cells in CP chemotherapy [208].

6.2. The Contribution of Exosomes

Exosomes are small membrane vesicles that are able to promote the progression and metastasis of tumor cells [209,210]. It seems that exosomes are involved in drug resistance through the transmission of lncRNAs [211,212]. Exosomal transfer of lncRNA HOTTIP (an oncogenic lncRNA) induces EMT and proliferation of GC cells, which leads to resistance of GC cells to CP therapy. By sponging miR-218, HOTTIP stimulates the expression of HMGA1 to trigger EMT (E-cadherin downregulation, and N-cadherin, and vimentin upregulation). By downregulating HOTTIP, the sensitivity of GC cells to CP was restored [213].

6.3. Forkhead Box Protein O1 (FOXO1) Signaling Pathway

FOXO1 is a tumor suppressor protein involved in inhibiting the migration and proliferation of cancer cells [214,215]. Prior studies have shown the involvement of FOXO1 in chemosensitivity [216]. In a newly published article by Li and colleagues, the role of FOXO1 in sensitizing nasopharyngeal carcinoma (NPC) cells to CP therapy was investigated mechanistically. In general, FOXO1 upregulates the expression of miR-200b by ZEB1 induction via the GSK-3β/β-catenin/TCF4 axis. This significantly sensitizes cancer cells to CP therapy by suppressing the EMT mechanism. MYH9 downregulation by FOXO1 also contributes to EMT inhibition [217]. This axis is important for the anti-tumor activity of cinobufotalin (CB). This substance is derived from toad venom and has shown a high anti-tumor activity [218]. CB stimulates the sensitivity of NPC cells to CP therapy by activating FOXO1 and subsequently inhibiting MYH9, which leads to a reduction in cancer rigidity and inhibition of EMT (E-cadherin upregulation, and N-cadherin downregulation) [217].

6.4. MicroRNAs

As it was mentioned in Section 3, miRs are involved in regulation of different biological mechanisms due to their capability in affecting various molecular pathways and mechanisms. Abnormal expression of miRs paves the way for the development of cancer [219,220]. MiR-146b is one of the main regulators of the innate and adaptive immune response [221]. The growing evidence has shed some light on the involvement of miR-146b in cancer, infection, and bone regeneration [222,223,224]. It appears that the effect of miR-146b on protein tyrosine phosphatase 1B (PTP1B) is important in CP therapy. PTP1B is involved in the development of cancer, and there are controversial data on its inhibitory or stimulatory effect in cancer [225,226]. In lung adenocarcinoma cells that are resistant to CP chemotherapy, increasing the expression of miR-146b can be considered a promising strategy. The upregulation of miR-146b remarkably reduces the expression of PTP1B. The in vitro and in vivo experiments confirm the reduced EMT concentration as a consequence of PTP1B downregulation by miR-146b, so that the expression of E-cadherin is upregulated, while the expression of N-cadherin decreases. This shows that EMT prevention by miR-146b/PTP1B axis can sensitize lung adenocarcinoma cells to CP therapy [227].
The miR-363 can act as an onco-suppressor miR. Its downregulation is associated with malignant and aggressive behavior of cancer cells [228]. The miR-363 is able to suppress myeloid cell leukemia 1 (Mcl-1), which leads to reduced proliferation and invasion of laryngeal cancer cells [90]. In view of the inhibitory effect of miR-363 on the malignancy of cancer cells, the lncRNA NNT-AS1 decreases the expression of this miR to increase the migration and invasion of GC cells [229]. In ovarian carcinoma cells resistant to CP, the increase in expression of miR-363 has an inverse relationship to screw expression. Downregulation of the Snail suppresses the EMT mechanism, which leads to the sensitivity of ovarian cancer cells to chemotherapy [230].
Inhibition of miR-200c by CD44 contributes to ZEB1 upregulation, leading to increased migration and invasion of cancer cells and CP resistance [231]. MiR-139-5p is an onco-suppressor miR that is able to reduce invasion, migration, and proliferation of tumor cells and to induce apoptotic cell death. The reduced expression of miR-139-56p is also associated with a poor prognosis in cancer patients [232,233,234,235]. In NPC cells, miR-139-5p suppresses EMT in order to significantly reduce the migration and invasion tumor cells, which leads to the sensitivity of tumor cells to CP chemotherapy [236].

6.5. PI3K/Akt Signaling Pathway

It is worth mentioning that a specific population of tumor cells known as cancer stem cells (CSCs) are involved in migration and metastasis [237,238,239,240]. These cells are also involved in cancer recurrences, and studies, both in vivo and in clinical trials, have shown the inadequacy of anti-tumor drugs in completely eliminating CSCs [241]. The relationship between CSCs and the PI3K/Akt/mTOR axis is important in cancer therapy. The Akt signaling pathway is involved in cell survival, metabolism, and differentiation [242,243,244,245,246]. It has been shown that mTOR is able to stimulate Akt in Ser473 [247]. In particular, it is believed that phosphorylation of Akt by mTOR contributes to tumor development, so that phosphorylated Akt inhibits GSK-3β [248,249]. The activity of GSK-3β is crucial for the increase of E-cadherin levels by cytoplasmic translocation of the cochlea [250]. The degradation of GSK-3β by Akt leads to stimulation of EMT [251,252]. This axis is remarkable in providing CP resistance in non-small cell lung carcinoma (NSCLC). Aspirin can be used to interrupt this axis and sensitize cancer cells to CP chemotherapy. Aspirin administration reduces the expression of the mTOR pathway to suppress Akt by dephosphorylation in Ser473. This stabilizes and induces GSK-3β to inhibit Snail and β-catenin nuclear translocation, which leads to inhibition of the EMT mechanism. Suppressed EMT sensitizes NSCLC cells to CP-mediated apoptosis [253].

6.6. Wnt Signaling Pathway

The Wnt signaling pathway is an important molecular signaling pathway with the ability to regulate biological processes such as apoptosis and differentiation [254,255,256,257]. A number of studies have shown that the Wnt signaling pathway contributes to increased invasion and malignancy of tumor cells via EMT induction [258,259,260]. The inhibition of Wnt by onco-suppressor factor Numb suppresses EMT in cancer cells, thereby illustrating the role of Wnt as an upstream mediator of EMT [261]. In addition, EMT induction by Wnt can elevate proliferation of cancer cells, and triggers their resistance into apoptosis [262]. On the other hand, the Wnt signaling pathway can be considered a secondary target of miRs (33a-5p) and lncRNAs (JPX) [263,264]. An increase in resistance to CP therapy has been observed in ovarian cancer cells. It has been proposed that miR-338-3p undergoes downward regulation in ovarian cancer cells and tissues. An increased expression of miR-338-3p sensitizes these tumor cells to CP therapy. Investigation of the molecular signaling pathways has shown that the overexpression of miR-338-3p can attenuate the Wnt2B signaling pathway for inhibiting EMT-mediated CP resistance [265]. Suppression of the Wnt signaling pathway is also important for the sensitization of malignant mesothelioma (MM) cells for CP therapy [266]. Epstein–Barr virus (EBV) infection is responsible for the development of NPC cells. EBV-miR-BART22 contributes to the resistance of NPC cells to chemotherapy. Mechanistically, EBV-miR-BART22 increases the expression of MYH9 by aligning with the PI3K/Akt/c-Jun axis. Then MYH9 induces the degradation of GSK-3β to mediate nuclear translocation of β-catenin, which leads to stimulation of the EMT mechanism and CP resistance. The interruption of this signaling pathway is therefore important for the inhibition of the EMT mechanism and the suppression of CP resistance. CB administration intervenes in the MYH9/GSK-3β/β−catenin axis by activating MAP2K4. This leads to inhibition of the EMT mechanism and sensitization of cancer cells to CP chemotherapy [267].

6.7. Long Non-Coding RNAs

The HOXA-AS3 gene is a member of the homeobox (HOX) family located on chromosome 7p15. It has been reported that this gene may be involved in the progression and metastasis of tumor cells [268,269,270]. It seems that the homeobox A3 (HOXA3) has a modulating influence on the EMT mechanism [271]. In NSCLC cells that are resistant to CP chemotherapy, the lncRNA HOXA-AS3 increases to inhibit HOXA3, leading to CP resistance. Inhibition of the lncRNA HOXA-AS3 increases the expression of HOXA3. Over-expressed HOXA3 inhibits EMT by increasing E-cadherin and reducing vimentin, resulting in the sensitivity of NSCLC cells to CP therapy [50]. The same applies to ovarian cancer cells. The LncRNA H19 increases the migration and malignancy of tumor cells by stimulating EMT, which makes these tumor cells resistant to CP therapy. Inhibition of the lncRNA H19 sensitizes ovarian cancer cells to CP-induced apoptosis by EMT inactivation [76]. These studies show that lncRNAs are involved in CP resistance by EMT induction.
The lncRNA urogenital carcinoma antigen 1 (UCA1) is another factor whose role in the resistance of NSCLC cells to CP therapy has been investigated. The lncRNA UCA1 has been considered as an oncogenic factor. It is believed that the lncRNA UCA1 induces the migration and invasion of thyroid cancer cells via miR-497-3p downregulation [272]. On the other hand, the lncRNA UCA1 reduces the expression of miR-654-5p to upregulate the expression of salt-inducible kinase 2 (SIK2), which leads to the development of PTX resistance [273]. In addition, other lncRNAs such as GAS8-AS1 can inhibit the lncRNA UCA1 to suppress the invasion of osteosarcoma cells [274]. The lncRNA UCA1 increases levels of N-cadherin, vimentin, and Snail, while decreasing levels of E-cadherin to induce the EMT mechanism, resulting in resistance of NSCLC cells to CP therapy [275]. Therefore, knock-out or knock-down of the lncRNA UCA1 and forkhead box protein C2 (FOXC2) can be considered as potential strategies for sensitizing NSCLC cells to CP chemotherapy.

6.8. Nuclear Factor-κB (NF-ĸB) Signaling Pathway

Nuclear factor-kappaB (NF-κB) is a transcription factor associated with pathological events including inflammation and cancer [112,273,276,277,278,279,280,281]. The IκB kinase (IKK) is an upstream mediator of NF-κB that is capable of regulating the proliferation and migration of cancer cells [282,283,284,285,286,287,288]. IKK expression is thought to be associated with higher levels of N-cadherin and EMT activation, which subsequently induces resistance of head and neck cancer cells to CP therapy. Suppression of IKK activity can attenuate EMT, which mediates sensitization of head and neck cancer cells to CP therapy [289].

6.9. Other Molecular Signaling Pathways

The forkhead box protein C2 (FOXC2) is one of the most important members of the forkhead box (FOX) family [249]. A large number of recently published articles have demonstrated the role of FOXC2 in improving the proliferation of cancer cells [290,291,292]. The expression of FOXC2 is believed to be associated with drug resistance and increased progression of melanoma cells [293]. This transcription factor significantly increases metastasis and progression of prostate cancer cells via the EMT mechanism [294,295]. Protein kinase C (PKC) also increases the expression of FOXC2 to inhibit p-120 catenin, leading to the migration and invasion of breast cancer cells [296]. The relationship between FOXC2 and EMT is of considerable importance in NSCLC cells. In A549 cells, FOXC2 increases Snail levels by Akt/GSK-3β upregulation. As a consequence of elevated Snail levels, EMT mechanism can be induced to mediate resistance of NSCLC cells to CP therapy [297].
Chrysotobibenzyl is a derivative of Dendrobium pulchellum with inhibitory activities against lung cancer cells [298]. The administration of chrysotobibenzyl is beneficial in alleviating CP resistance. It appears that chrysotobibenzyl inhibits EMT by reducing vimentin, Snail, and Slug to sensitize lung cancer cells to CP-induced apoptosis [299]. These studies show that different molecular signaling pathways may be involved in CP resistance and their identification is of considerable importance for the inhibition of resistance to CP therapy [300].
Vascular endothelial growth factor (VEGF) and programmed death ligand 1 (PD-L1) can contribute to increased migration and malignancy of cancer cells. The novel strategies are based on the inhibition of VEGF and PD-L1 [263,301,302,303]. Bevacizumab and atezolizumab are able to suppress VEGF and PD-L1 and inhibit the progression of cancer cells, respectively [304,305,306,307]. The administration of atezolizumab and bevacizumab can inhibit VEGF and PD-L1 to suppress ovarian cancer malignancy and sensitize them to CP therapy. It appears that the inhibition of VEGF and PD-L1 and the subsequent downregulation of the EMT mechanism are involved in sensitizing ovarian cancer cells to CP therapy [121].
p53 is an onco-suppressor transcription factor, the inactivation and/or mutation of which has been reported in various types of cancer [308,309]. On the contrary, it has been suggested that the mouse double minute 2 homologue (MDM2) is involved in p53 inhibition by mediating its proteosomal degradation [310]. Recently published articles have shown that inhibitors of MDM2 are beneficial in stimulating p53 and suppressing the malignancy of cancer cells [133,311]. In lung cancer cells, zinc finger CCHC-type containing 10 (ZCCHC10) can inhibit MDM2 to stimulate and enhance the expression of p53. Inhibition of p53 degradation leads to upregulation of the epithelial marker E-cadherin and downregulation of Snail and Slug as mesenchymal markers; this leads to suppression of EMT and sensitization of cancer cells to CP therapy [312].
Norcantharidin (NCTD) is a demethylated form of cantharidin isolated from the bladder beetle [313]. This agent has shown significant anti-tumor effects. The administration of NCTD is beneficial in suppressing the invasion of bladder cancer by inhibiting the DNA damage response (DDR) via cdc6 degradation [314]. NCTD is also beneficial in sensitizing breast cancer cells to tamoxifen chemotherapy by upregulating miR-873 and CDK3 inhibition [159]. Due to the significant anti-tumor activity of NCTD, it can sensitize cancer cells to CP chemotherapy. Administration of NCTD is associated with downregulation of YAP1 and its downstream mediators, connective tissue growth factor (CTGF), and cysteine-rich angiogenic inducer 61 (CYR61). This leads to inhibition of EMT and sensitization of NSCLC cells to CP chemotherapy [315].
It is obvious that Twist is an EMT marker that undergoes upregulation during cancer metastasis [314,316,317]. It is worth mentioning that Twist and EMT are able to regulate the resistance of cancer cells to chemotherapy [318,319]. Twist induces EMT by increasing N-cadherin and vimentin levels and decreasing E-cadherin levels [320,321]. Inhibition of Twist is believed to significantly reduce the migration and invasion of cancer cells [316,322]. In ovarian cancer cells, Twist activates the EMT mechanism to mediate CP chemoresistance. Suppression of Twist expression using siRNA-loaded hyaluronic acid-conjugated nanoparticles inhibits the EMT mechanism, which leads to sensitization of ovarian cancer cells to CP chemotherapy [323]. Programmed cell death 4 (PDCD4) is an important regulator of cell proliferation and apoptosis. PDCD4 is an onco-suppressor factor that inhibits cancer metastasis via EMT downregulation [232]. A number of processes such as autophagy can degrade PDCD4 to ensure the proliferation and malignancy of tumor cells [324].
The carcinogenic inhibitor of protein phosphatase 2A (CIP2A) is upregulated in malignant human diseases and is associated with a poor prognosis [69]. CIP2A contributes to increased metastasis of cancer cells by suppressing protein phosphatase 2A (PP2A) [325,326]. CIP2A is able to target molecular signaling pathways such as MEK and ERK in the activation of EMT [327]. There is growing evidence that CIP2A induces phosphorylation of Akt and subsequently mTOR to mediate CP resistance [297,328,329]. Targeting CIP2A/Akt/mTOR is therefore beneficial in preventing resistance to CP. Polyphyllin I (PPI) and polyphyllin VII (PPVII) are two bioactive components of Parisian polyphylla (PRS) that display substantial anti-tumor activities [330,331]. Administration of PPI and PPVII significantly inhibits the CIP2A/Akt/mTOR axis to attenuate EMT, resulting in sensitivity of NSCLC cells to CP chemotherapy [332].
It appears that in CP-resistant bladder cancer cells, increasing the expression of PDCD4 is important. The upregulation of PDCD4 is associated with the inhibition of the JNK/c-Jun signaling pathway to suppress EMT and sensitize bladder cancer cells to CP chemotherapy [333]. Considering the above evidence, a variety of signaling networks are involved in CP resistance by EMT induction, and studies have investigated the ability of various compounds and molecular pathways to inhibit the EMT mechanism that may sensitize cancer cells to chemotherapy.

7. Possible Pathways for Further Targeting

7.1. PI3k/Akt Signaling Pathway

PI3K/Akt signaling pathway regulates apoptosis, differentiation, metastasis, and drug resistance. It has been demonstrated that PI3K/Akt pathway, as an oncogene pathway, can elevate the invasion and migration of cancer cells by targeting EMT [334,335,336]. It has been reported that this pathway is of importance in CP resistance via EMT stimulation. On the other hand, miRs have been suggested to be involved in cancer invasion and EMT regulation [337,338]. In NPC cells resistant to CP chemotherapy, overexpressed miR-205-5p diminishes the expression of tumor suppressor factor, PTEN, by upregulation of PI3K/Akt signaling pathway, leading to the activation of EMT and resistance to CP chemotherapy [339]. The miR-205-5p/PI3K/Akt/PTEN can be targeted in future studies for inhibition of resistance to CP therapy.

7.2. Laminin Subunit Beta-3

The laminin subunit beta-3 (LAMB3) is an oncogenic factor capable of enhancing the proliferation and migration of pancreatic cancer cells via stimulation of PI3K/Akt signaling pathway [338]. It appears that LAMB3 induces EMT mechanism through MET/Akt activation, leading to an increased migration and malignancy of thyroid cancer cells [340]. Accumulating data show the stimulatory effect of LAMB3 on the viability and growth of tumor cells [341]. A similar phenomenon occurs in head and neck squamous cell carcinoma. It has been found that LAMB3 activates EMT mechanism through vimentin and Slug upregulation to reduce the CP efficacy in elimination of cancer cells. Suppressing the expression of LAMB3 diminishes the metastasis of tumor cells and sensitizes them to the cytotoxicity of CP therapy [338].

7.3. ZEB Proteins

The zinc-finger E-box binding homeobox (ZEB) family includes two major components: ZEB1 and ZEB2 [342,343]. The accumulating data demonstrate that ZEB1 and ZEB2 proteins are able to enhance the invasion and migration of cancer cells [344]. It has been shown that these proteins can induce EMT by E-cadherin downregulation and N-cadherin upregulation [345]. So, in respect to the involvement of EMT in resistance to CP, there is a relationship between ZEB proteins and CP resistance. On the other hand, excision repair cross-complementary gene 1 (ERCC1) and ATP-binding cassette subfamily G member 2 (ABCG2) can contribute to the chemoresistance [139,346,347]. It has been reported that UBE2C enhances the expressions of ZEB1 and ZEB2 to upregulate ZBCG2 and ERCC1. This axis results in EMT activation and enhanced invasion and malignancy of NSCLC cells. Then, these cancer cells with high proliferation and migration ability can induce resistance against CP therapy [348].

7.4. Long Non-Coding RNAs

The lncRNA homeobox A11 antisense RNA (HOXA11-AS) is suggested to be involved in elevating the migration and invasion of GC cells by miR-148a downregulation and subsequent activation of Wnt1/β-catenin signaling pathway [349]. In prostate cancer cells, HOXA11-AS induces actinin alpha 4 (ACTN4) expression through miR-518b sponging, leading to enhanced proliferation and migration of tumor cells [156]. Overall, the studies are in agreement with the role of lncRNA HOXA11-AS in tumorigenesis as well as in poor prognosis [96,350]. The relationship between lncRNA HOXA11-AS and CP resistance in lung adenocarcinoma (LUAD) cells is of importance, so that HOXA11-AS enhances the expression of STAT3 though miR-454-3p sponging. The HOXA11-AS/miR-454-3p/STAT3 axis leads to EMT activation and resistance of LUAD cells in CP therapy [351].

7.5. Targeting Drug Transporters

One of the reasons underlying the induction of chemoresistance is the enhanced activity and expression of drug transporters [352]. Accumulating data demonstrate that tumor cells are able to enhance expression of various transporters to reduce accumulation of chemotherapeutic agents and prevent their cellular uptake [353]. In respect to the adverse effects of chemotherapeutic agents, and some other unexpected outcomes, it is about impossible to administer high doses of a certain chemotherapeutic agent. As a consequence, novel strategies should be considered in targeting the drug transporters to sensitize cancer cells to chemotherapeutic agents via enhancing their cellular internalization [354]. The ATP-binding cassette (ABC) transporters are suggested to mediate chemoresistance via exporting chemotherapeutic drugs out of cells [355]. To date, up to 48 ABC transporters have been identified in humans. The P-glycoprotein (P-gp) is a key member of ABC transporters, known as ABCB1 [356]. This transmembrane glycoprotein has a molecular weight of 170 kDa consisting of two membrane spinning domains (MSDs) and two nucleotide binding domains (NBDs) that can be used for ATP binding [357,358]. The P-gp is expressed in a number of barriers including the blood–brain barrier (BBB), blood–testis barrier (BTB), and blood–cerebrospinal fluid barrier to modulate absorption and excretion of xenobiotics [359]. Although normal activity of P-gp is vital in physiological conditions, its upregulation is an increasing impediment in cancer therapy, since cancer cells are able to trigger chemoresistance by enhancing expression of P-gp [360,361]. As mentioned earlier, administration of CP has been correlated with stimulation of EMT. It has been found that in lung cancer cells exposed to CP, an increase in EMT concentration occurred by the TGF-β signaling pathway. The overexpression of EMT paves the road for induction of P-gp that subsequently exports CP out of tumor cells, thereby leading to chemoresistance [362]. Recently, it was reported that CP-resistant cancer cells that underwent EMT demonstrated an increase in N-cadherin and spindle-shaped cells. Although this study did not explore the relationship between EMT and expression of P-gp, it seems that EMT induction substantially enhances migration and metastasis of cancer cells. This stimulation in malignant behavior of cancer cells can facilitate an overexpression of P-gp, and resistance of cancer cells to CP therapy [363].

7.6. Other Molecular Signaling Pathways

Psoriasin is a member of the calcium-binding EF-hand proteins with a molecular weight of 11.4 kDa. This oncogene protein is found in cells or can be secreted outside the cells [364,365,366,367]. In the field of cancer, psoriasin overexpression has been related to the poor prognosis of patients [263]. This is due to the stimulatory impact of psoriasin on the migration, invasion, and malignancy of cancer cells [368]. In vitro experiments on GC cells show that psoriasin upregulation can reduce ERK signaling pathway to activate EMT and partially mediate the resistance of tumor cells to CP chemotherapy [369]. It has been found that diverse signaling cascades contribute to the CP resistance of tumor cells. For instance, downregulation of Raf kinase inhibitor protein (RKIP) by miR-27a mediates the positive impacts on the proliferation and EMT induction in liver cancer cells. The malignant tumor cells can then acquire resistance to CP chemotherapy [369]. Human HLA-F adjacent transcript 10 (FAT10) is considered as an oncogenic factor with the capability of regulation of apoptosis and differentiation [334,370].
Targeting UBE2C can disrupt the aforementioned axis to sensitize NSCLC cells in CP therapy. The eukaryotic initiation factor 5A2 (elF5A2) is involved in modulation of vital biological processes such as proliferation, differentiation, and apoptosis [371,372]. Various studies have shed light on the involvement of elF5A2 in the migration and metastasis of cancer cells [373,374,375,376]. In GC cells, elF5A2 induces CP resistance by EMT stimulation through enhancing vimentin and N-cadherin levels and decreasing E-cadherin and β-catenin levels [377].
Deubiquitinating enzymes (DUBs) can inhibit the degradation of proteins through prevention of ubiquitination [378]. The abnormal expression of DUBs occurs in tumor malignancy and EMT [379,380]. The ubiquitin specific peptidase 37 (USP37) is a kind of DUB containing 979 amino acids with an oncogene role [381,382]. It appears that USP37 can also facilitate G1/S transition [383,384]. A newly published article demonstrated that USP37 stabilizes Snail protein via deubiquitination to enhance the migration and metastasis of lung cancer cells [385]. On the contrary, Hedgehog (Hh) pathway has been found to be involved in the migration of breast cancer cells via EMT mechanism [386]. The USP37 can stimulate Hh signaling pathway through Smo and Gli-1 upregulation. This axis leads to EMT activation to induce CP resistance in breast cancer stem cells [387]. The ATM is involved in resistance of chemotherapy in cancer cells. It has been reported that oncogenic JAK/STAT3 signaling pathway activation can trigger ATM-mediated EMT [388,389,390,391,392,393]. On the other hand, administration of CP induces EMT through ATM upregulation. In addition, PD-L1 can elevate the progression and metastasis of cancer cells through EMT [394,395]. In lung cancer cells, ATM induces JAK/STAT3 signaling pathway through PD-L1 upregulation to activate EMT, thus resulting in resistance of cancer cells in CP therapy [396]. Interestingly, knock-out of ATM can reverse this axis to enhance the sensitivity of cancer cells to CP therapy.
Visfatin released by adipocytes and inflammatory cells is involved in various biological mechanisms such as angiogenesis, inflammation, and cell growth [397,398]. In recent years, much attention has been directed towards visfatin as a biomarker for diagnosis of cancer [399]. Visfatin induces growth differentiation factor 15 (GDF15)/Akt axis to elevate the progression and proliferation of tumor cells [322]. These studies highlight the fact that visfatin can accelerate the progression of tumor cells [400]. Visfatin follows an interesting route in resistance of cancer cells. In osteosarcoma cells, visfatin enhances the expression of Snail through stimulation of hypoxia-inducible factor-α (HIF-α) transcription, while visfatin has no effect on the expression of ZEB1 and elevates its stability via ATM induction. Consequently, induced ZEB1 and Snail can mediate EMT activation, leading to the repression of osteosarcoma cells to CP chemotherapy [401]. Cyclooxygenase-2 (COX-2) is another important protein and its role in cancer has been extensively explored. It is worth mentioning that suppressing COX-2 can inhibit the invasion of cancer cells [402]. In addition, oncogenic factors such as BPTF enhances the expression of COX-2 to stimulate cancer cell growth and proliferation [403]. It has been found that COX-2 induces Akt signaling pathway to activate EMT mechanism. The EMT elevates the malignancy and invasion of NSCLC cells and induces resistance to CP therapy [404]. Overall, inhibition of visfatin and COX-2 can be advantageous in making cancer cells sensitive to CP therapy by promoting EMT downregulation.
Pyruvate dehydrogenase complex (PDC) is involved in cellular respiration through converting pyruvate into acetyl-coA. Pyruvate dehydrogenase kinase 1 (PDK1) is a key member of glycolytic enzymes that disrupts cellular respiration via PDC phosphorylation. This leads to the enhanced level of lactate in cytosol and inhibition of pyruvate oxidation in mitochondria [405]. Accumulating data demonstrate that overexpression of PDK1 can occur in various cancers [406,407]. Suppressing PDK1 has resulted in an increase in ROS generation and induction of apoptosis in cancer cells [39,408,409]. On the contrary, the epidermal growth factor receptor (EGFR) plays a significant role in development of cancer. It appears that in cutaneous squamous cell carcinoma, EGFR activates NF-κB signaling pathway to elevate the progression and malignancy of cancer cells [410]. Furthermore, the capability of EGFR in stimulation of EMT has been shown [411,412]. In the case of CP, PDK1 upregulates the expression of EGFR and inhibition of PDK1/EGFR axis can suppress EMT and malignancy of tumor cells. This results in greater sensitivity of ovarian cancer cells to chemotherapy [251]. In addition to the EGFR, insulin-like growth factor 1 (IGF1-R) contributes to the malignancy and invasion of tumor cells by stimulation of EMT mechanism. For doing this, IGF1-R can affect a variety of signaling cascades including STAT3/Akt and PI3K/Akt [154,413]. In ovarian cancer cells, IGF1-R undergoes upregulation as a consequence of miR-1294 downregulation. The overexpressed IGF1-R enhances the migration of cancer cells through EMT induction. It is worth mentioning that IGF1-R-mediated EMT may be involved in resistance of ovarian cancer cells in response to CP therapy [351]. These studies highlight the fact that elevating the expression of miR-1294 and PDK1 inhibition may be beneficial in sensitizing ovarian cancer cells in CP therapy.
As mentioned earlier, Snail is a transcription factor involved in regulation of EMT. Understanding the link between Snail and eukaryotic translation initiation factor 4E (elF4E) is vital for inhibiting CP resistance in NPC cells. The abnormal expression of elF4E occurs in a variety of cancers [414,415,416,417]. The elF4E upregulation is associated with malignant transformation of normal cells and enhanced invasion of tumor cells [417,418]. Suppressing the nuclear translocation of β-catenin via targeting elF4E can sensitize NPC cells to apoptotic cell death. In NPC cells and tissues, the expression of elF4E undergoes upregulation to provide the nuclear translocation of Snail. Thereafter, EMT mechanism can be activated to enhance the malignancy and invasion of NPC cells and make them resistant to CP chemotherapy [419]. This connection between elF4E and Snail can be considered as a promising candidate for sensitizing tumor cells to CP therapy, and further targeting these proteins, both pharmacologically and genetically, can pave the road for suppressing resistance to CP. It is worth mentioning that in cancer cells resistant to CP therapy, the expression of molecules involved in EMT such as N-cadherin has been found to be elevated. As a result of EMT induction, the morphology of cells can undergo transformation from round to spindle-shaped [363].
With respect to the role of Wnt signaling pathway in EMT induction, this pathway may also confer resistance to CP therapy. B-cell lymphoma 9 (BCL9) is an oncogenic factor and its upregulation is associated with poor prognosis of patients with cancer, low incidence of apoptosis in cancer cells, and increased malignancy [47,97,410]. In NSCLC cells, BCL9 stimulates the nuclear translocation of β-catenin to activate EMT mechanism and induce resistance of tumor cells to CP chemotherapy [351].
In osteosarcoma cells, FAT10 induces the ubiquitin-mediated degradation of YAP1 to elevate the viability and growth of tumor cells [152]. Investigation of the relationship between FAT10 and CP resistance can pave the road for the effective treatment of bladder cancer. It has been reported that overexpression of FAT10 induces EMT mechanism to elevate the malignancy of cancer cells and reduce the efficacy of CP chemotherapy [369]. This was the first study that has examined the relationship between FAT10 and EMT and its role in the development of resistance to CP therapy. However, further studies should focus on inhibition of FAT10, both pharmacologically and genetically. H2A histone family member Z (H2A.Z) is a potential regulator of proliferation, differentiation, DNA replication, chromosome segregation, and cell cycle [420,421,422,423,424]. Thus, targeting H2A.Z may influence the potential of CP in chemotherapy through its impact on the malignancy of cancer cells. It appears that H2A.Z can diminish the sensitivity of cancer cells to CP chemotherapy by induction of EMT and enhancing the progression of tumor cells [425]. Overall, a number of pathways can regulate the EMT process and contribute to chemoresistance (Figure 2).

8. Conclusions and Remarks

In the present review, we analyzed the involvement of the EMT mechanism in the induction of resistance to CP. As mentioned above, CP is an effective anti-tumor agent that negatively affects the viability and proliferation of tumor cells. Recent studies have shown the great potential of cancer cells to develop CP resistance to chemotherapy. It appears that EMT is one of the most important mechanisms associated with resistance to this drug. To fully describe the role of EMT in chemoresistance, we divided the article into three different sections. In the first section we have shown that the administration of CP is associated with EMT and tumor cell resistance. It was reported that both short and long treatment with CP can induce resistance. Autophagy, NF-κB, CAMs, and ATM are induced by CP to induce EMT and reduce the efficacy of chemotherapy, while downregulation of SLFN1 can also mediate resistance. These studies show that CP itself is capable of inducing EMT and promoting resistance in cancer cells and the above mechanisms and signaling pathways may be involved in this process.
It is worth mentioning that lncRNAs such as HOTTIP, HOXA-AS3, H19, and UCA1 are involved in resistance by targeting the EMT mechanism, and studies have focused on restoring the expression of these lncRNAs to increase CP cytotoxicity. Another possible strategy for inhibiting EMT-mediated resistance is miR upregulation/downregulation. In this case, studies have investigated the role of miR-218, -200b, -146b, -338-3p, -363, -139-5p, and so on. Overall, it has been concluded that manipulation of miR expression may pave the way for inhibition of EMT-mediated resistance to CP. In particular, in addition to lncRNAs and miRs, other molecular pathways such as PI3K/Akt, NF-κB, FOXO1, Wnt/catenin, mTOR, etc., can also influence EMT and chemoresistance. In the last section we presented a variety of molecular pathways that contribute to EMT-mediated resistance to CP and which can be specifically investigated in further studies.
Taking all these aspects into account, a number of complex and dynamic molecular pathways that contribute to resistance to therapy has been summarized here. These pathways may synergistically induce EMT and promote chemoresistance. In view of the role of more than one molecular pathway in EMT-mediated resistance, the focus in enhancing CP chemotherapy should be on targeting different oncogenic pathways. Finally, it should be mentioned that the studies were based on in vitro and in vivo findings and the lack of clinical trials shows that there is still a long way to go in developing strategies to suppress EMT-mediated CP resistance. In addition, disseminated tumor cells (DTCs) are also suggested to be viable after several years. They may reactivate and regrow after a long period of time and are considered as a vital reason for the recurrence of cancer. Prior studies have shown that existence of dormant cells ensures metastasis of cancer cells and is correlated with poor prognosis [483,484,485,486]. To date, there is no experiment about the relationship between CP, EMT, and dormant cells. However, with respect to the significant role of dormant cells in metastasis and recurrence, this can be evaluated in further studies.

Author Contributions

M.A. and R.M. provided the concept. M.A. and T.J contributed to organization of manuscript. M.A., A.Z., K.H., M.K., R.M., T.J. and G.S. participated in writing and revision of the manuscript. All authors have read and agreed to the published version of this manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

EMTEpithelial–mesenchymal transition
CPCisplatin
ROSReactive oxygen species
DOXDocetaxel
NPsNanoparticles
AMPKAMP-activated protein kinase
lncRNALong non-coding RNAs
TAMsTumor associated macrophages
GCGastric cancer
OSCCOral squamous cell carcinoma
Nrf2Nuclear factor erythroid 2-related factor 2
SIRT1Sirtuin 1
MSI2Musashi RNA-binding protein 2
FHLFour-and-half LIM domain
TGFTumor growth factor
YAP1Yes-associated protein 1
TAZ1Tazaffin
CRCColorectal cancer
UBE2OUbiquitin-conjugating enzyme E2O
mTORMammalian target of rapamycin
TKITyrosine kinase inhibitor
PTXPaclitaxel
CamsClassically activated macrophages
HCCHepatocellular carcinoma
CCL20Chemokine ligand 20
CCR6Chemokine receptor 6
EMP3Epithelial membrane protein 3
NPCNasopharyngeal carcinoma
EBVEpstein-Barr virus
PTP1BProtein tyrosine phosphatase 1B
CSCsCancer stem cells
NSCLCNon-small cell lung cancer
CIP2ACancerous inhibitor of protein phosphatase 2A
PP2AProtein phosphatase 2A
PPIPolyphyllin I
PPVIIPolyphyllin VII
PRSParis polyphylla
MMMalignant mesothelioma
HOXA3Homeobox A3
FOXC2Forkhead box protein C2
FOXForkhead box
PKCαProtein kinase Cα
UCA1Urogenital carcinoma antigen 1
SIK2Salt inducible kinase 2
VEGFVascular endothelial growth factor
PD-L1Programmed death-ligand 1
MDM2Mouse double minute 2 homolog
ZCCHC10Zinc finger CCHC-type containing 10
NF-κBNuclear factor-κB
IKKβInhibitor of NF-κB kinase subunit β
Mcl-1Myeloid cell leukemia 1
NCTDNorcantharidin
DDRDNA damage response
CTGFConnective tissue growth factor
CYR61Cysteine rich angiogenic factor 61
PDCD4Programmed cell death 4
elF4EEukaryotic translation initiation factor 4E
MDR1Multidrug resistance protein 1
LAMB3Laminin subunit β-3;
ZEBZinc-finger E-box binding homeobox
ERCC1Excision repair cross-complementary gene 1
ABCG2ATP-binding cassette subfamily G member 2
elF5A2Eukaryotic initiation factor 5A2
DUBsDeubiquitinating enzymes
USP37Ubiquitin specific peptidase 37
HhHedgehog
ATMAtaxia telangiectasia mutated
GDF15Growth differentiation factor 15
HIF-α,Hypoxia inducible factor-α
COX-2Cyclooxygenase-2
PDCPyruvate dehydrogenase complex
PDK1Pyruvate dehydrogenase kinase 1
EGFREpidermal growth factor receptor
IGF1RInsulin-like growth factor 1
HOXA11-ASHomeobox A11 antisense RNA
ACTN4Actinin alpha 4
LUADLung adenocarcinoma
BCL9B-cell lymphoma 9
RKIPRaf kinase inhibitor protein
FAT10HLA-F adjacent transcript 10
H2A ZH2A histone family member Z
ABCATP-binding cassette
P-gpP-glycoprotein
MSDsMembrane spinning domains
NBDsNucleotide binding domains
BBBBlood–brain barrier
BTBBlood–testis barrier

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA A Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. DeSantis, C.E.; Miller, K.D.; Dale, W.; Mohile, S.G.; Cohen, H.J.; Leach, C.R.; Goding Sauer, A.; Jemal, A.; Siegel, R.L. Cancer statistics for adults aged 85 years and older, 2019. CA A Cancer J. Clin. 2019, 69, 452–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Miller, K.D.; Nogueira, L.; Mariotto, A.B.; Rowland, J.H.; Yabroff, K.R.; Alfano, C.M.; Jemal, A.; Kramer, J.L.; Siegel, R.L. Cancer treatment and survivorship statistics, 2019. CA A Cancer J. Clin. 2019, 69, 363–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dai, X.; Zhang, J.; Arfuso, F.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Kumar, A.P.; Ahn, K.S.; Sethi, G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp. Biol. Med. (Maywood) 2015, 240, 760–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Shanmugam, M.K.; Warrier, S.; Kumar, A.P.; Sethi, G.; Arfuso, F. Potential Role of Natural Compounds as Anti-Angiogenic Agents in Cancer. Curr. Vasc. Pharmacol. 2017, 15, 503–519. [Google Scholar] [CrossRef] [PubMed]
  6. Najafi, M.; Mortezaee, K.; Majidpoor, J. Stromal reprogramming: A target for tumor therapy. Life Sci. 2019, 239, 117049. [Google Scholar] [CrossRef] [PubMed]
  7. Prasannan, R.; Kalesh, K.A.; Shanmugam, M.K.; Nachiyappan, A.; Ramachandran, L.; Nguyen, A.H.; Kumar, A.P.; Lakshmanan, M.; Ahn, K.S.; Sethi, G. Key cell signaling pathways modulated by zerumbone: role in the prevention and treatment of cancer. Biochem. Pharmacol. 2012, 84, 1268–1276. [Google Scholar] [CrossRef] [Green Version]
  8. Mortezaee, K.; Potes, Y.; Mirtavoos-Mahyari, H.; Motevaseli, E.; Shabeeb, D.; Musa, A.E.; Najafi, M.; Farhood, B. Boosting immune system against cancer by melatonin: A mechanistic viewpoint. Life Sci. 2019, 238, 116960. [Google Scholar] [CrossRef]
  9. Mortezaee, K.; Najafi, M.; Farhood, B.; Ahmadi, A.; Shabeeb, D.; Musa, A.E. NF-κB targeting for overcoming tumor resistance and normal tissues toxicity. J. Cell. Physiol. 2019, 234, 17187–17204. [Google Scholar] [CrossRef]
  10. Shanmugam, M.K.; Manu, K.A.; Ong, T.H.; Ramachandran, L.; Surana, R.; Bist, P.; Lim, L.H.; Kumar, A.P.; Hui, K.M.; Sethi, G. Inhibition of CXCR4/CXCL12 signaling axis by ursolic acid leads to suppression of metastasis in transgenic adenocarcinoma of mouse prostate model. Int. J. Cancer 2011, 129, 1552–1563. [Google Scholar] [CrossRef]
  11. Ramachandran, L.; Manu, K.A.; Shanmugam, M.K.; Li, F.; Siveen, K.S.; Vali, S.; Kapoor, S.; Abbasi, T.; Surana, R.; Smoot, D.T.; et al. Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor γ activation pathway in gastric cancer. J. Biol. Chem. 2012, 287, 38028–38040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Li, K.; Xie, W.; Gao, L.; Huang, G.; Zhou, J.; Mei, B.; Chen, J. Impact of neoadjuvant chemotherapy on survival prognosis and pathological downstaging in patients presenting with high-risk upper tract urothelial carcinoma: A protocol for systematic review and meta analysis. Medicine 2020, 99, e20184. [Google Scholar] [CrossRef] [PubMed]
  13. Minami, K.; Ueda, N.; Ishimoto, K.; Tsujiuchi, T. Lysophosphatidic acid receptor-2 (LPA2)-mediated signaling enhances chemoresistance in melanoma cells treated with anticancer drugs. Mol. Cell. Biochem. 2020. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Z.; Qiu, N.; Yin, J.; Zhang, J.; Liu, H.; Guo, W.; Liu, M.; Liu, T.; Chen, D.; Luo, K.; et al. SRGN crosstalks with YAP to maintain chemoresistance and stemness in breast cancer cells by modulating HDAC2 expression. Theranostics 2020, 10, 4290–4307. [Google Scholar] [CrossRef]
  15. Zhang, J.; Chen, G.; Gao, Y.; Liang, H. HOTAIR/miR-125 axis-mediated Hexokinase 2 expression promotes chemoresistance in human glioblastoma. J. Cell. Mol. Med. 2020. [Google Scholar] [CrossRef] [Green Version]
  16. Paciello, F.; Rita Fetoni, A.; Mezzogori, D.; Rolesi, R.; Di Pino, A.; Paludetti, G.; Grassi, C.; Troiani, D. The dual role of curcumin and ferulic acid in counteracting chemoresistance and cisplatin-induced ototoxicity. Sci. Rep. 2020, 10, 1063. [Google Scholar] [CrossRef] [Green Version]
  17. Zou, Z.; Li, X.; Sun, Y.; Li, L.; Zhang, Q.; Zhu, L.; Zhong, Z.; Wang, M.; Wang, Q.; Liu, Z.; et al. NOS1 expression promotes proliferation and invasion and enhances chemoresistance in ovarian cancer. Oncol. Lett. 2020, 19, 2989–2995. [Google Scholar] [CrossRef] [Green Version]
  18. Dou, N.; Hu, Q.; Li, L.; Wu, Q.; Li, Y.; Gao, Y. USP32 promotes tumorigenesis and chemoresistance in gastric carcinoma via upregulation of SMAD2. Int. J. Biol. Sci. 2020, 16, 1648–1657. [Google Scholar] [CrossRef] [Green Version]
  19. Xi, Y.; Yuan, P.; Li, T.; Zhang, M.; Liu, M.F.; Li, B. hENT1 reverses chemoresistance by regulating glycolysis in pancreatic cancer. Cancer Lett. 2020, 479, 112–122. [Google Scholar] [CrossRef]
  20. Tang, L.; Chen, Y.; Chen, H.; Jiang, P.; Yan, L.; Mo, D.; Tang, X.; Yan, F. DCST1-AS1 Promotes TGF-beta-Induced Epithelial-Mesenchymal Transition and Enhances Chemoresistance in Triple-Negative Breast Cancer Cells via ANXA1. Front. Oncol. 2020, 10, 280. [Google Scholar] [CrossRef]
  21. Lin, G.; Zhao, R.; Wang, Y.; Han, J.; Gu, Y.; Pan, Y.; Ren, C.; Ren, S.; Xu, C. Dynamic analysis of N-glycomic and transcriptomic changes in the development of ovarian cancer cell line A2780 to its three cisplatin-resistant variants. Ann. Transl. Med. 2020, 8, 289. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, J.; Quan, L.N.; Meng, Q.; Wang, H.Y.; Wang, J.; Yu, P.; Fu, J.T.; Li, Y.J.; Chen, J.; Cheng, H.; et al. miR-548e Sponged by ZFAS1 Regulates Metastasis and Cisplatin Resistance of OC by Targeting CXCR4 and let-7a/BCL-XL/S Signaling Axis. Mol. Ther. Nucleic Acids 2020, 20, 621–638. [Google Scholar] [CrossRef] [PubMed]
  23. Hu, W.C.; Teo, W.H.; Huang, T.F.; Lee, T.C.; Lo, J.F. Combinatorial Low Dose Arsenic Trioxide and Cisplatin Exacerbates Autophagy via AMPK/STAT3 Signaling on Targeting Head and Neck Cancer Initiating Cells. Front. Oncol. 2020, 10, 463. [Google Scholar] [CrossRef] [PubMed]
  24. Feldmann, D.P.; Heyza, J.; Zimmermann, C.M.; Patrick, S.M.; Merkel, O.M. Nanoparticle-Mediated Gene Silencing for Sensitization of Lung Cancer to Cisplatin Therapy. Molecules 2020, 25, 1994. [Google Scholar] [CrossRef]
  25. Chen, T.W.; Jan, I.S.; Chang, D.Y.; Lin, C.H.; Chen, I.C.; Chen, H.M.; Cheng, A.L.; Lu, Y.S. Systemic treatment of breast cancer with leptomeningeal metastases using bevacizumab, etoposide and cisplatin (BEEP regimen) significantly improves overall survival. J. Neuro-Oncol. 2020. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, M.W.; Ryu, H.; Song, I.C.; Yun, H.J.; Jo, D.Y.; Ko, Y.B.; Lee, H.J. Efficacy of cisplatin combined with topotecan in patients with advanced or recurrent ovarian cancer as second- or higher-line palliative chemotherapy. Medicine 2020, 99, e19931. [Google Scholar] [CrossRef]
  27. Li, W.H.; Zhang, L.; Wu, Y.H. CDKN3 regulates cisplatin resistance to colorectal cancer through TIPE1. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3614–3623. [Google Scholar] [CrossRef]
  28. Murakami, M.; Izumi, H.; Kurita, T.; Koi, C.; Morimoto, Y.; Yoshino, K. UBE2L6 is Involved in Cisplatin Resistance by Regulating the Transcription of ABCB6. Anti-Cancer Agents Med. Chem. 2020. [Google Scholar] [CrossRef]
  29. Jin, L.; Zhang, N.; Zhang, Q.; Ding, G.; Yang, Z.; Zhang, Z. Serum microRNAs as potential new biomarkers for cisplatin resistance in gastric cancer patients. PeerJ 2020, 8, e8943. [Google Scholar] [CrossRef] [Green Version]
  30. Kasikova, L.; Hensler, M.; Truxova, I.; Skapa, P.; Laco, J.; Belicova, L.; Praznovec, I.; Vosahlikova, S.; Halaska, M.J.; Brtnicky, T.; et al. Calreticulin exposure correlates with robust adaptive antitumor immunity and favorable prognosis in ovarian carcinoma patients. J. Immunother. Cancer 2019, 7, 312. [Google Scholar] [CrossRef]
  31. Jain, A.; Jahagirdar, D.; Nilendu, P.; Sharma, N.K. Molecular approaches to potentiate cisplatin responsiveness in carcinoma therapeutics. Expert Rev. Anticancer Ther. 2017, 17, 815–825. [Google Scholar] [CrossRef] [PubMed]
  32. Rezaee, R.; Momtazi, A.A.; Monemi, A.; Sahebkar, A. Curcumin: A potentially powerful tool to reverse cisplatin-induced toxicity. Pharmacol. Res. 2017, 117, 218–227. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, C.; Nance, E.A.; Mastorakos, P.; Chisholm, J.; Berry, S.; Eberhart, C.; Tyler, B.; Brem, H.; Suk, J.S.; Hanes, J. Convection enhanced delivery of cisplatin-loaded brain penetrating nanoparticles cures malignant glioma in rats. J. Controll. Release 2017, 263, 112–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wang, M.; Wu, Q.; Fang, M.; Huang, W.; Zhu, H. miR-152-3p Sensitizes Glioblastoma Cells Towards Cisplatin Via Regulation Of SOS1. OncoTargets Ther. 2019, 12, 9513–9525. [Google Scholar] [CrossRef] [Green Version]
  35. Wu, Z.; Gong, Q.; Yu, Y.; Zhu, J.; Li, W. Knockdown of circ-ABCB10 promotes sensitivity of lung cancer cells to cisplatin via miR-556-3p/AK4 axis. BMC Pulm. Med. 2020, 20, 10. [Google Scholar] [CrossRef] [Green Version]
  36. Balog, J.A.; Hackler, L., Jr.; Kovacs, A.K.; Neuperger, P.; Alfoldi, R.; Nagy, L.I.; Puskas, L.G.; Szebeni, G.J. Single Cell Mass Cytometry Revealed the Immunomodulatory Effect of Cisplatin Via Downregulation of Splenic CD44+, IL-17A+ MDSCs and Promotion of Circulating IFN-gamma+ Myeloid Cells in the 4T1 Metastatic Breast Cancer Model. Int. J. Mol. Sci. 2019, 21, 170. [Google Scholar] [CrossRef] [Green Version]
  37. Zhao, W.; Li, W.; Jin, X.; Niu, T.; Cao, Y.; Zhou, P.; Zheng, M. Silencing long non-coding RNA NEAT1 enhances the suppression of cell growth, invasion, and apoptosis of bladder cancer cells under cisplatin chemotherapy. Int. J. Clin. Exp. Pathol. 2019, 12, 549–558. [Google Scholar]
  38. Jiang, H.; Xiong, W.; Chen, L.; Lv, Z.; Yang, C.; Li, Y. Knockdown of the long noncoding RNA HOTTIP inhibits cell proliferation and enhances cell sensitivity to cisplatin by suppressing the Wnt/beta-catenin pathway in prostate cancer. J. Cell. Biochem. 2019, 120, 8965–8974. [Google Scholar] [CrossRef]
  39. Zang, H.; Qian, G.; Arbiser, J.; Owonikoko, T.K.; Ramalingam, S.S.; Fan, S.; Sun, S.Y. Overcoming acquired resistance of EGFR-mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib, with the natural product honokiol. Mol. Oncol. 2020. [Google Scholar] [CrossRef] [Green Version]
  40. Bostan, M.; Petrica-Matei, G.G.; Ion, G.; Radu, N.; Mihaila, M.; Hainarosie, R.; Brasoveanu, L.I.; Roman, V.; Constantin, C.; Neagu, M.T. Cisplatin effect on head and neck squamous cell carcinoma cells is modulated by ERK1/2 protein kinases. Exp. Ther. Med. 2019, 18, 5041–5051. [Google Scholar] [CrossRef] [Green Version]
  41. Achkar, I.W.; Abdulrahman, N.; Al-Sulaiti, H.; Joseph, J.M.; Uddin, S.; Mraiche, F. Cisplatin based therapy: The role of the mitogen activated protein kinase signaling pathway. J. Transl. Med. 2018, 16, 96. [Google Scholar] [CrossRef] [PubMed]
  42. Fadejeva, I.; Olschewski, H.; Hrzenjak, A. MicroRNAs as regulators of cisplatin-resistance in non-small cell lung carcinomas. Oncotarget 2017, 8, 115754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zhou, B.; Xia, M.; Wang, B.; Thapa, N.; Gan, L.; Sun, C.; Guo, E.; Huang, J.; Lu, Y.; Cai, H. Clarithromycin synergizes with cisplatin to inhibit ovarian cancer growth in vitro and in vivo. J. Ovarian Res. 2019, 12, 107. [Google Scholar] [CrossRef] [PubMed]
  44. Swanepoel, B.; Nitulescu, G.M.; Olaru, O.T.; Venables, L.; van de Venter, M. Anti-Cancer Activity of a 5-Aminopyrazole Derivative Lead Compound (BC-7) and Potential Synergistic Cytotoxicity with Cisplatin against Human Cervical Cancer Cells. Int. J. Mol. Sci. 2019, 20, 5559. [Google Scholar] [CrossRef] [Green Version]
  45. Hashimoto, M.; Shirakawa, Y.; Maeda, N.; Tanabe, S.; Noma, K.; Sakurama, K.; Katsui, K.; Nishizaki, M.; Fujiwara, T. Induction chemoradiotherapy including docetaxel, cisplatin, and 5-fluorouracil for locally advanced esophageal cancer. Esophagus Off. J. Jpn. Esophageal Soc. 2020. [Google Scholar] [CrossRef]
  46. Khafaji, M.; Zamani, M.; Vossoughi, M.; Iraji Zad, A. Doxorubicin/Cisplatin-Loaded Superparamagnetic Nanoparticles As A Stimuli-Responsive Co-Delivery System For Chemo-Photothermal Therapy. Int. J. Nanomed. 2019, 14, 8769–8786. [Google Scholar] [CrossRef] [Green Version]
  47. Jiang, M.; Kang, Y.; Sewastianik, T.; Wang, J.; Tanton, H.; Alder, K.; Dennis, P.; Xin, Y.; Wang, Z.; Liu, R.; et al. BCL9 provides multi-cellular communication properties in colorectal cancer by interacting with paraspeckle proteins. Nat. Commun. 2020, 11, 19. [Google Scholar] [CrossRef] [Green Version]
  48. Gan, D.; He, W.; Yin, H.; Gou, X. beta-elemene enhances cisplatin-induced apoptosis in bladder cancer cells through the ROS-AMPK signaling pathway. Oncol. Lett. 2020, 19, 291–300. [Google Scholar] [CrossRef]
  49. Heeren, A.M.; van Luijk, I.F.; Lakeman, J.; Pocorni, N.; Kole, J.; de Menezes, R.X.; Kenter, G.G.; Bosse, T.; de Kroon, C.D.; Jordanova, E.S. Neoadjuvant cisplatin and paclitaxel modulate tumor-infiltrating T cells in patients with cervical cancer. Cancer Immunol. Immunother. CII 2019, 68, 1759–1767. [Google Scholar] [CrossRef] [Green Version]
  50. Lin, S.; Zhang, R.; An, X.; Li, Z.; Fang, C.; Pan, B.; Chen, W.; Xu, G.; Han, W. LncRNA HOXA-AS3 confers cisplatin resistance by interacting with HOXA3 in non-small-cell lung carcinoma cells. Oncogenesis 2019, 8, 60. [Google Scholar] [CrossRef]
  51. Nan, Y. Lung carcinoma therapy using epidermal growth factor receptortargeted lipid polymeric nanoparticles coloaded with cisplatin and doxorubicin. Oncol. Rep. 2019, 42, 2087–2096. [Google Scholar] [CrossRef] [PubMed]
  52. Roy, S.; Roy, S.; Kar, M.; Chakraborty, A.; Kumar, A.; Delogu, F.; Asthana, S.; Hande, M.P.; Banerjee, B. Combined treatment with cisplatin and the tankyrase inhibitor XAV-939 increases cytotoxicity, abrogates cancer-stem-like cell phenotype and increases chemosensitivity of head-and-neck squamous-cell carcinoma cells. Mutat. Res. 2019, 846, 503084. [Google Scholar] [CrossRef] [PubMed]
  53. Sato, K.; Hayashi, Y.; Watanabe, K.; Yoshimi, R.; Hibi, H. Concurrent chemoradiotherapy with intravenous cisplatin and docetaxel for advanced oral cancer. Nagoya J. Med Sci. 2019, 81, 407–414. [Google Scholar] [CrossRef] [PubMed]
  54. Xue, T.; Wang, L.; Li, Y.; Song, H.; Chu, H.; Yang, H.; Guo, A.; Jiao, J. SiRNA-Mediated RRM2 Gene Silencing Combined with Cisplatin in the Treatment of Epithelial Ovarian Cancer In Vivo: An Experimental Study of Nude Mice. Int. J. Med. Sci. 2019, 16, 1510–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wang, W.; Shanmugam, M.K.; Xiang, P.; Yam, T.Y.A.; Kumar, V.; Chew, W.S.; Chang, J.K.; Ali, M.Z.B.; Reolo, M.J.Y.; Peh, Y.X.; et al. Sphingosine 1-Phosphate Receptor 2 Induces Otoprotective Responses to Cisplatin Treatment. Cancers 2020, 12, 211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Bayurova, E.; Jansons, J.; Skrastina, D.; Smirnova, O.; Mezale, D.; Kostyusheva, A.; Kostyushev, D.; Petkov, S.; Podschwadt, P.; Valuev-Elliston, V.; et al. HIV-1 Reverse Transcriptase Promotes Tumor Growth and Metastasis Formation via ROS-Dependent Upregulation of Twist. Oxidative Med. Cell. Longev. 2019, 2019, 6016278. [Google Scholar] [CrossRef] [Green Version]
  57. Jang, H.S.; Noh, M.R.; Jung, E.M.; Kim, W.Y.; Southekal, S.; Guda, C.; Foster, K.W.; Oupicky, D.; Ferrer, F.A.; Padanilam, B.J. Proximal tubule cyclophilin D regulates fatty acid oxidation in cisplatin-induced acute kidney injury. Kidney Int. 2019. [Google Scholar] [CrossRef]
  58. Li, Q.; Liang, X.; Yang, Y.; Zeng, X.; Zhong, X.; Huang, C. Panax notoginseng saponins ameliorate cisplatin-induced mitochondrial injury via the HIF-1alpha/mitochondria/ROS pathway. FEBS Open Bio 2020, 10, 118–126. [Google Scholar] [CrossRef]
  59. Dai, X.; Wang, L.; Deivasigamni, A.; Looi, C.Y.; Karthikeyan, C.; Trivedi, P.; Chinnathambi, A.; Alharbi, S.A.; Arfuso, F.; Dharmarajan, A.; et al. A novel benzimidazole derivative, MBIC inhibits tumor growth and promotes apoptosis via activation of ROS-dependent JNK signaling pathway in hepatocellular carcinoma. Oncotarget 2017, 8, 12831–12842. [Google Scholar] [CrossRef] [Green Version]
  60. Hanigan, M.H.; Devarajan, P. Cisplatin nephrotoxicity: Molecular mechanisms. Cancer Ther. 2003, 1, 47. [Google Scholar]
  61. Tan, R.Z.; Liu, J.; Zhang, Y.Y.; Wang, H.L.; Li, J.C.; Liu, Y.H.; Zhong, X.; Zhang, Y.W.; Yan, Y.; Lan, H.Y.; et al. Curcumin relieved cisplatin-induced kidney inflammation through inhibiting Mincle-maintained M1 macrophage phenotype. Phytomedicine 2019, 52, 284–294. [Google Scholar] [CrossRef] [PubMed]
  62. Said, R.S.; Mantawy, E.M.; El-Demerdash, E. Mechanistic perspective of protective effects of resveratrol against cisplatin-induced ovarian injury in rats: Emphasis on anti-inflammatory and anti-apoptotic effects. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 1225–1238. [Google Scholar] [CrossRef] [PubMed]
  63. Domitrovic, R.; Cvijanovic, O.; Pernjak-Pugel, E.; Skoda, M.; Mikelic, L.; Crncevic-Orlic, Z. Berberine exerts nephroprotective effect against cisplatin-induced kidney damage through inhibition of oxidative/nitrosative stress, inflammation, autophagy and apoptosis. Food Chem. Toxicol. 2013, 62, 397–406. [Google Scholar] [CrossRef] [PubMed]
  64. Mortezaee, K. CXCL12/CXCR4 axis in the microenvironment of solid tumors: A critical mediator of metastasis. Life Sci. 2020, 249, 117534. [Google Scholar] [CrossRef] [PubMed]
  65. Mortezaee, K. Hypoxia induces core-to-edge transition of progressive tumoral cells: A critical review on differential yet corroborative roles for HIF-1α and HIF-2α. Life Sci. 2020, 242, 117145. [Google Scholar] [CrossRef] [PubMed]
  66. Bahrami, A.; Bianconi, V.; Pirro, M.; Orafai, H.M.; Sahebkar, A. The role of TFEB in tumor cell autophagy: Diagnostic and therapeutic opportunities. Life Sci. 2020, 244, 117341. [Google Scholar] [CrossRef]
  67. Meng, D.; Li, Z.; Ma, X.; Wu, L.; Fu, L.; Qin, G. ETV5 overexpression contributes to tumor growth and progression of thyroid cancer through PIK3CA. Life Sci. 2020, 253, 117693. [Google Scholar] [CrossRef]
  68. Gu, Y.; Fei, Z.; Zhu, R. miR-21 modulates cisplatin resistance of gastric cancer cells by inhibiting autophagy via the PI3K/Akt/mTOR pathway. Anti-cancer Drugs 2020. [Google Scholar] [CrossRef]
  69. Yin, J.; Chen, D.; Luo, K.; Lu, M.; Gu, Y.; Zeng, S.; Chen, X.; Song, Y.; Zhang, Z.; Zheng, G.; et al. Cip2a/miR-301a feedback loop promotes cell proliferation and invasion of triple-negative breast cancer. J. Cancer 2019, 10, 5964–5974. [Google Scholar] [CrossRef]
  70. Hazari, Y.; Bravo-San Pedro, J.M.; Hetz, C.; Galluzzi, L.; Kroemer, G. Autophagy in hepatic adaptation to stress. J. Hepatol. 2020, 72, 183–196. [Google Scholar] [CrossRef]
  71. Galluzzi, L.; Green, D.R. Autophagy-Independent Functions of the Autophagy Machinery. Cell 2019, 177, 1682–1699. [Google Scholar] [CrossRef] [PubMed]
  72. Jamali, Z.; Taheri-Anganeh, M.; Shabaninejad, Z.; Keshavarzi, A.; Taghizadeh, H.; Razavi, Z.S.; Mottaghi, R.; Abolhassan, M.; Movahedpour, A.; Mirzaei, H. Autophagy regulation by microRNAs: Novel insights into osteosarcoma therapy. IUBMB Life 2020, n/a. [Google Scholar] [CrossRef] [PubMed]
  73. Song, S.; Lee, J.Y.; Ermolenko, L.; Mazumder, A.; Ji, S.; Ryu, H.; Kim, H.; Kim, D.W.; Lee, J.W.; Dicato, M.; et al. Tetrahydrobenzimidazole TMQ0153 triggers apoptosis, autophagy and necroptosis crosstalk in chronic myeloid leukemia. Cell Death Dis. 2020, 11, 109. [Google Scholar] [CrossRef] [PubMed]
  74. Sala de Oyanguren, F.J.; Rainey, N.E.; Moustapha, A.; Saric, A.; Sureau, F.; O’Connor, J.E.; Petit, P.X. Highlighting Curcumin-Induced Crosstalk between Autophagy and Apoptosis as Supported by Its Specific Subcellular Localization. Cells 2020, 9, 361. [Google Scholar] [CrossRef] [Green Version]
  75. Yu, T.; Guo, F.; Yu, Y.; Sun, T.; Ma, D.; Han, J.; Qian, Y.; Kryczek, I.; Sun, D.; Nagarsheth, N. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell 2017, 170, 548–563. e516. [Google Scholar] [CrossRef] [Green Version]
  76. Wu, Y.; Zhou, Y.; He, J.; Sun, H.; Jin, Z. Long non-coding RNA H19 mediates ovarian cancer cell cisplatin-resistance and migration during EMT. Int. J. Clin. Exp. Pathol. 2019, 12, 2506–2515. [Google Scholar]
  77. Liu, Y.; Hu, Q.; Wang, X. AFAP1-AS1 induces cisplatin resistance in non-small cell lung cancer through PI3K/AKT pathway. Oncol. Lett. 2020, 19, 1024–1030. [Google Scholar] [CrossRef] [Green Version]
  78. Miyamoto, I.; Kasamatsu, A.; Yamatoji, M.; Nakashima, D.; Saito, K.; Higo, M.; Endo-Sakamoto, Y.; Shiiba, M.; Tanzawa, H.; Uzawa, K. Kinesin family member 14 in human oral cancer: A potential biomarker for tumoral growth. Biochem. Biophys. Rep. 2015, 3, 26–31. [Google Scholar] [CrossRef] [Green Version]
  79. Han, Y.; Zhou, S.; Wang, X.; Mao, E.; Huang, L. SNHG14 stimulates cell autophagy to facilitate cisplatin resistance of colorectal cancer by regulating miR-186/ATG14 axis. Biomed. Pharmacother. Biomed. Pharmacother. 2020, 121, 109580. [Google Scholar] [CrossRef]
  80. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  81. Liu, F.; Qiu, F.; Fu, M.; Chen, H.; Wang, H. Propofol Reduces Epithelial to Mesenchymal Transition, Invasion and Migration of Gastric Cancer Cells through the MicroRNA-195-5p/Snail Axis. Med. Sci. Monit. 2020, 26, e920981. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, Y.F.; Li, C.S.; Zhou, Y.; Lu, X.H. Propofol facilitates cisplatin sensitivity via lncRNA MALAT1/miR-30e/ATG5 axis through suppressing autophagy in gastric cancer. Life Sci. 2020, 244, 117280. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, Y.; Xia, H. Oridonin elevates sensitivity of ovarian carcinoma cells to cisplatin via suppressing cisplatin-mediated autophagy. Life Sci. 2019, 233, 116709. [Google Scholar] [CrossRef] [PubMed]
  84. Hu, Z.; Cai, M.; Zhang, Y.; Tao, L.; Guo, R. miR-29c-3p inhibits autophagy and cisplatin resistance in ovarian cancer by regulating FOXP1/ATG14 pathway. Cell Cycle 2020, 19, 193–206. [Google Scholar] [CrossRef]
  85. Pan, X.; Chen, Y.; Shen, Y.; Tantai, J. Knockdown of TRIM65 inhibits autophagy and cisplatin resistance in A549/DDP cells by regulating miR-138-5p/ATG7. Cell Death Dis. 2019, 10, 429. [Google Scholar] [CrossRef]
  86. Wang, Y.; Lina, L.; Xu, L.; Yang, Z.; Qian, Z.; Zhou, J.; Suoni, L. Arctigenin enhances the sensitivity of cisplatin resistant colorectal cancer cell by activating autophagy. Biochem. Biophys. Res. Commun. 2019, 520, 20–26. [Google Scholar] [CrossRef]
  87. Liu, J.; Huang, Y.; Wang, H.; Wu, D. MiR-106a-5p promotes 5-FU resistance and the metastasis of colorectal cancer by targeting TGFbetaR2. Int. J. Clin. Exp. Pathol. 2018, 11, 5622–5634. [Google Scholar]
  88. Pourhanifeh, M.H.; Mahjoubin-Tehran, M.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; Mirzaei, H.; Asemi, Z. MicroRNAs and exosomes: Small molecules with big actions in multiple myeloma pathogenesis. IUBMB Life 2020, 72, 314–333. [Google Scholar] [CrossRef]
  89. Naeli, P.; Yousefi, F.; Ghasemi, Y.; Savardashtaki, A.; Mirzaei, H. The Role of MicroRNAs in Lung Cancer: Implications for Diagnosis and Therapy. Curr. Mol. Med. 2020, 20, 90–101. [Google Scholar] [CrossRef]
  90. Feng, W.T.; Yao, R.; Xu, L.J.; Zhong, X.M.; Liu, H.; Sun, Y.; Zhou, L.L. Effect of miR-363 on the proliferation, invasion and apoptosis of laryngeal cancer by targeting Mcl-1. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4564–4572. [Google Scholar] [CrossRef]
  91. Xu, C.; Du, Z.; Ren, S.; Liang, X.; Li, H. MiR-129-5p sensitization of lung cancer cells to etoposide-induced apoptosis by reducing YWHAB. J. Cancer 2020, 11, 858–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Cheng, J.-T.; Wang, L.; Wang, H.; Tang, F.-R.; Cai, W.-Q.; Sethi, G.; Xin, H.-W.; Ma, Z. Insights into Biological Role of LncRNAs in Epithelial-Mesenchymal Transition. Cells 2019, 8, 1178. [Google Scholar] [CrossRef] [Green Version]
  93. Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Mohammadi, S.; Fathullahzadeh, S.; Mirzaei, H.R.; Namdar, A.; Savardashtaki, A.; Mirzaei, H. Long Non-Coding RNAs As Epigenetic Regulators in Cancer. Curr. Pharm. Design 2019, 25, 3563–3577. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, L.; Zhu, Q.; Lu, L.; Liu, Y. MiR-132 inhibits migration and invasion and increases chemosensitivity of cisplatin-resistant oral squamous cell carcinoma cells via targeting TGF-beta1. Bioengineered 2020, 11, 91–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Huang, X.X.; Zhang, Q.; Hu, H.; Jin, Y.; Zeng, A.L.; Xia, Y.B.; Xu, L. A novel circular RNA circFN1 enhances cisplatin resistance in gastric cancer via sponging miR-182-5p. J. Cell. Biochem. 2020. [Google Scholar] [CrossRef] [PubMed]
  96. Gao, M.; Li, H.; Bi, Y.; Zhang, Z.; Wang, S.; Li, J.; Yang, Z.; Lv, X.; Zhou, B.; Yin, Z. The Polymorphisms of lncRNA HOXA11-AS and the risk of Lung Cancer in Northeastern Chinese population. J. Cancer 2020, 11, 592–598. [Google Scholar] [CrossRef] [Green Version]
  97. Wang, J.; Zheng, M.; Zhu, L.; Deng, L.; Li, X.; Gao, L.; Wang, C.; Wang, H.; Liu, J.; Lin, B. Low BCL9 expression inhibited ovarian epithelial malignant tumor progression by decreasing proliferation, migration, and increasing apoptosis to cancer cells. Cancer Cell Int. 2019, 19, 330. [Google Scholar] [CrossRef]
  98. Cheng, Y.; Shen, X.; Zheng, M.; Zou, G.; Shen, Y. Knockdown Of lncRNA NCK-AS1 Regulates Cisplatin Resistance Through Modulating miR-137 In Osteosarcoma Cells. OncoTargets Ther. 2019, 12, 11057–11068. [Google Scholar] [CrossRef] [Green Version]
  99. Xu, D.; Yang, F.; Wu, K.; Kai, Z.; Xinxing, X.; An, Y.; Xu, F.; Xun, J.; Lv, X.; Zhang, X.; et al. Lost miR-141 and upregulated TM4SF1 expressions associate with poor prognosis of pancreatic cancer: Regulation of EMT and angiogenesis by miR-141 and TM4SF1 via AKT. Cancer Biol. Ther. 2020, 1–10. [Google Scholar] [CrossRef]
  100. Liu, Y.; Liu, C.; Tan, T.; Li, S.; Tang, S.; Chen, X. Sinomenine sensitizes human gastric cancer cells to cisplatin through negative regulation of PI3K/AKT/Wnt signaling pathway. Anti-cancer Drugs 2019, 30, 983–990. [Google Scholar] [CrossRef]
  101. Silva, M.M.; Rocha, C.R.R.; Kinker, G.S.; Pelegrini, A.L.; Menck, C.F.M. The balance between NRF2/GSH antioxidant mediated pathway and DNA repair modulates cisplatin resistance in lung cancer cells. Sci. Rep. 2019, 9, 17639. [Google Scholar] [CrossRef] [PubMed]
  102. Asaka, R.; Miyamoto, T.; Yamada, Y.; Ando, H.; Mvunta, D.H.; Kobara, H.; Shiozawa, T. Sirtuin 1 promotes the growth and cisplatin resistance of endometrial carcinoma cells: A novel therapeutic target. Lab. Investig. J. Tech. Methods Pathol. 2015, 95, 1363–1373. [Google Scholar] [CrossRef] [PubMed]
  103. Loh, C.-Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Roshan, M.K.; Soltani, A.; Soleimani, A.; Kahkhaie, K.R.; Afshari, A.R.; Soukhtanloo, M. Role of AKT and mTOR signaling pathways in the induction of epithelial-mesenchymal transition (EMT) process. Biochimie 2019, 165, 229–234. [Google Scholar] [CrossRef] [PubMed]
  105. Kang, P.J.; Son, D.; Ko, T.H.; Hong, W.; Yun, W.; Jang, J.; Choi, J.I.; Song, G.; Lee, J.; Kim, I.Y.; et al. mRNA-Driven Generation of Transgene-Free Neural Stem Cells from Human Urine-Derived Cells. Cells 2019, 8, 1043. [Google Scholar] [CrossRef] [Green Version]
  106. Tentler, D.; Lomert, E.; Novitskaya, K.; Barlev, N.A. Role of ACTN4 in Tumorigenesis, Metastasis, and EMT. Cells 2019, 8, 1427. [Google Scholar] [CrossRef] [Green Version]
  107. Teeuwssen, M.; Fodde, R. Wnt Signaling in Ovarian Cancer Stemness, EMT, and Therapy Resistance. J. Clin. Med. 2019, 8, 1658. [Google Scholar] [CrossRef] [Green Version]
  108. Zhu, X.; Chen, L.; Liu, L.; Niu, X. EMT-Mediated Acquired EGFR-TKI Resistance in NSCLC: Mechanisms and Strategies. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
  109. Ayyar, B.V.; Arora, S.; O’Kennedy, R. Coming-of-Age of Antibodies in Cancer Therapeutics. Trends Pharmacol. Sci. 2016, 37, 1009–1028. [Google Scholar] [CrossRef]
  110. Yan, W.; Wu, Q.; Yao, W.; Li, Y.; Liu, Y.; Yuan, J.; Han, R.; Yang, J.; Ji, X.; Ni, C. MiR-503 modulates epithelial-mesenchymal transition in silica-induced pulmonary fibrosis by targeting PI3K p85 and is sponged by lncRNA MALAT1. Sci. Rep. 2017, 7, 1–15. [Google Scholar] [CrossRef] [Green Version]
  111. Xiao, H.; Zhu, Q.; Zhou, J. Long non-coding RNA MALAT1 interaction with miR-429 regulates the proliferation and EMT of lung adenocarcinoma cells through RhoA. Int. J. Clin. Exp. Pathol. 2019, 12, 419–430. [Google Scholar] [PubMed]
  112. Cai, H.; Yan, L.; Liu, N.; Xu, M.; Cai, H. IFI16 promotes cervical cancer progression by upregulating PD-L1 in immunomicroenvironment through STING-TBK1-NF-kB pathway. Biomed. Pharmacother. Biomed. Pharmacother. 2020, 123, 109790. [Google Scholar] [CrossRef] [PubMed]
  113. Qu, T.; Zhao, Y.; Chen, Y.; Jin, S.; Fang, Y.; Jin, X.; Sun, L.; Ma, Y. Down-regulated MAC30 expression inhibits breast cancer cell invasion and EMT by suppressing Wnt/beta-catenin and PI3K/Akt signaling pathways. Int. J. Clin. Exp. Pathol. 2019, 12, 1888–1896. [Google Scholar] [PubMed]
  114. Baek, S.H.; Ko, J.H.; Lee, J.H.; Kim, C.; Lee, H.; Nam, D.; Lee, J.; Lee, S.G.; Yang, W.M.; Um, J.Y.; et al. Ginkgolic Acid Inhibits Invasion and Migration and TGF-beta-Induced EMT of Lung Cancer Cells Through PI3K/Akt/mTOR Inactivation. J. Cell. Physiol. 2017, 232, 346–354. [Google Scholar] [CrossRef]
  115. Zhang, H.; Li, M.; Xu, X. MicroRNA-204 attenuates the migration and invasion of pancreatic cancer cells by targeting ZEB1/EMT axis. Int. J. Clin. Exp. Pathol. 2018, 11, 3802–3811. [Google Scholar]
  116. Yang, L.; Yu, Y.; Xiong, Z.; Chen, H.; Tan, B.; Hu, H. Downregulation of SEMA4C Inhibit Epithelial-Mesenchymal Transition (EMT) and the Invasion and Metastasis of Cervical Cancer Cells via Inhibiting Transforming Growth Factor-beta 1 (TGF-beta1)-Induced Hela cells p38 Mitogen-Activated Protein Kinase (MAPK) Activation. Med. Sci. Monit. 2020, 26, e918123. [Google Scholar] [CrossRef]
  117. Yang, M.H.; Lee, J.H.; Ko, J.H.; Jung, S.H.; Sethi, G.; Ahn, K.S. Brassinin Represses Invasive Potential of Lung Carcinoma Cells through Deactivation of PI3K/Akt/mTOR Signaling Cascade. Molecules 2019, 24, 1584. [Google Scholar] [CrossRef] [Green Version]
  118. Bunz, F. EMT and Back Again: Visualizing the Dynamic Phenotypes of Metastasis. Cancer Res. 2020, 80, 153–155. [Google Scholar] [CrossRef]
  119. Ko, J.H.; Nam, D.; Um, J.Y.; Jung, S.H.; Sethi, G.; Ahn, K.S. Bergamottin Suppresses Metastasis of Lung Cancer Cells through Abrogation of Diverse Oncogenic Signaling Cascades and Epithelial-to-Mesenchymal Transition. Molecules 2018, 23, 1601. [Google Scholar] [CrossRef] [Green Version]
  120. Zhu, H.; Zhang, Y.; Geng, Y.; Lu, W.; Yin, J.; Li, Z.; Huang, L.; Liu, H.; Xu, N. IGFBP2 promotes the EMT of colorectal cancer cells by regulating E-cadherin expression. Int. J. Clin. Exp. Pathol. 2019, 12, 2559–2565. [Google Scholar]
  121. Wu, Z.; Xue, S.; Zheng, B.; Ye, R.; Xu, G.; Zhang, S.; Zeng, T.; Zheng, W.; Chen, C. Expression and significance of c-kit and epithelial-mesenchymal transition (EMT) molecules in thymic epithelial tumors (TETs). J. Thorac. Dis. 2019, 11, 4602–4612. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, R.; Wang, K.; Feng, Z.; Zhang, M.Y.; Wu, J.; Geng, J.J.; Chen, Z.N. CD147 deficiency in T cells prevents thymic involution by inhibiting the EMT process in TECs in the presence of TGFbeta. Cell. Mol. Immunol. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Seddiki, R.; Narayana, G.H.N.S.; Strale, P.-O.; Balcioglu, H.E.; Peyret, G.; Yao, M.; Le, A.P.; Teck Lim, C.; Yan, J.; Ladoux, B. Force-dependent binding of vinculin to α-catenin regulates cell–cell contact stability and collective cell behavior. Mol. Biol. Cell 2018, 29, 380–388. [Google Scholar] [CrossRef] [PubMed]
  124. Xu, S.T.; Ma, Y.C.; Wang, C.H.; Xu, Y.; Gu, G.J. Prognostic and clinicopathologic significance of AEG-1/MTDH and E-cadherin expression in human gallbladder carcinoma. Int. J. Clin. Exp. Pathol. 2018, 11, 6025–6031. [Google Scholar]
  125. Haensel, D.; Dai, X. Epithelial-to-mesenchymal transition in cutaneous wound healing: Where we are and where we are heading. Dev. Dyn. 2018, 247, 473–480. [Google Scholar] [CrossRef] [Green Version]
  126. Kim, D.H.; Xing, T.; Yang, Z.; Dudek, R.; Lu, Q.; Chen, Y.-H. Epithelial mesenchymal transition in embryonic development, tissue repair and cancer: A comprehensive overview. J. Clin. Med. 2018, 7, 1. [Google Scholar] [CrossRef] [Green Version]
  127. Rout-Pitt, N.; Farrow, N.; Parsons, D.; Donnelley, M. Epithelial mesenchymal transition (EMT): A universal process in lung diseases with implications for cystic fibrosis pathophysiology. Respir. Res. 2018, 19, 136. [Google Scholar] [CrossRef]
  128. Kassouf, T.; Larive, R.M.; Morel, A.; Urbach, S.; Bettache, N.; Marcial Medina, M.C.; Merezegue, F.; Freiss, G.; Peter, M.; Boissiere-Michot, F.; et al. The Syk Kinase Promotes Mammary Epithelial Integrity and Inhibits Breast Cancer Invasion by Stabilizing the E-Cadherin/Catenin Complex. Cancers 2019, 11, 1974. [Google Scholar] [CrossRef] [Green Version]
  129. Shu, J.; Wang, L.; Han, F.; Chen, Y.; Wang, S.; Luo, F. BTBD7 Downregulates E-Cadherin and Promotes Epithelial-Mesenchymal Transition in Lung Cancer. BioMed Res. Int. 2019, 2019, 5937635. [Google Scholar] [CrossRef] [Green Version]
  130. Dehli, J.; Karlsson, C.; Bizelli-Silveira, C.; Jiang, X.; Kraft, D.; Foss, M. E-cadherin mediated cell-biomaterial interaction reduces migration of keratinocytes in-vitro. Colloids Surf. B Biointerfaces 2019, 180, 326–333. [Google Scholar] [CrossRef]
  131. Sun, S.; Gong, Q. The expressions and prognostic implications of Twist and E-cadherin in adenocarcinomas of the gastroesophageal junction and proximal gastric carcinoma. Medicine 2019, 98, e18449. [Google Scholar] [CrossRef] [PubMed]
  132. Dai, J.; He, H.; Lin, D.; Wang, C.; Zhu, Y.; Xu, D. Up-regulation of E-cadherin by saRNA inhibits the migration and invasion of renal carcinoma cells. Int. J. Clin. Exp. Pathol. 2018, 11, 5792–5800. [Google Scholar] [PubMed]
  133. Fang, D.D.; Tang, Q.; Kong, Y.; Wang, Q.; Gu, J.; Fang, X.; Zou, P.; Rong, T.; Wang, J.; Yang, D.; et al. MDM2 inhibitor APG-115 synergizes with PD-1 blockade through enhancing antitumor immunity in the tumor microenvironment. J. Immunother. Cancer 2019, 7, 327. [Google Scholar] [CrossRef]
  134. Miro, C.; Di Cicco, E.; Ambrosio, R.; Mancino, G.; Di Girolamo, D.; Cicatiello, A.G.; Sagliocchi, S.; Nappi, A.; De Stefano, M.A.; Luongo, C. Thyroid hormone induces progression and invasiveness of squamous cell carcinomas by promoting a ZEB-1/E-cadherin switch. Nat. Commun. 2019, 10, 1–13. [Google Scholar] [CrossRef]
  135. Manshouri, R.; Coyaud, E.; Kundu, S.T.; Peng, D.H.; Stratton, S.A.; Alton, K.; Bajaj, R.; Fradette, J.J.; Minelli, R.; Peoples, M.D.; et al. ZEB1/NuRD complex suppresses TBC1D2b to stimulate E-cadherin internalization and promote metastasis in lung cancer. Nat. Commun. 2019, 10, 5125. [Google Scholar] [CrossRef]
  136. Liu, B.; Li, X.; Li, C.; Xu, R.; Sun, X. miR-25 mediates metastasis and epithelial-mesenchymal-transition in human esophageal squamous cell carcinoma via regulation of E-cadherin signaling. Bioengineered 2019, 10, 679–688. [Google Scholar] [CrossRef] [Green Version]
  137. Masuda, T.; Ueo, H.; Kai, Y.; Noda, M.; Hu, Q.; Sato, K.; Fujii, A.; Hayashi, N.; Tsuruda, Y.; Otsu, H.; et al. N-Cadherin mRNA Levels in Peripheral Blood Could Be a Potential Indicator of New Metastases in Breast Cancer: A Pilot Study. Int. J. Mol. Sci. 2020, 21, 511. [Google Scholar] [CrossRef] [Green Version]
  138. Po, J.W.; Roohullah, A.; Lynch, D.; DeFazio, A.; Harrison, M.; Harnett, P.R.; Kennedy, C.; de Souza, P.; Becker, T.M. Improved ovarian cancer EMT-CTC isolation by immunomagnetic targeting of epithelial EpCAM and mesenchymal N-cadherin. J. Circ. Biomark. 2018, 7. [Google Scholar] [CrossRef] [Green Version]
  139. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef]
  140. Zhu, G.J.; Song, P.P.; Zhou, H.; Shen, X.H.; Wang, J.G.; Ma, X.F.; Gu, Y.J.; Liu, D.D.; Feng, A.N.; Qian, X.Y.; et al. Role of epithelial-mesenchymal transition markers E-cadherin, N-cadherin, beta-catenin and ZEB2 in laryngeal squamous cell carcinoma. Oncol. Lett. 2018, 15, 3472–3481. [Google Scholar] [CrossRef] [Green Version]
  141. Azimi, I.; Petersen, R.M.; Thompson, E.W.; Roberts-Thomson, S.J.; Monteith, G.R. Hypoxia-induced reactive oxygen species mediate N-cadherin and SERPINE1 expression, EGFR signalling and motility in MDA-MB-468 breast cancer cells. Sci. Rep. 2017, 7, 15140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Dobritoiu, M.; Stepan, A.E.; Margaritescu, C.; Simionescu, C.E.; Vere, C.C.; Schenker, M.; Crisan, A.E. Immunoexpression of E-cadherin, P-cadherin and fibronectin in gastric carcinomas. Rom. J. Morphol. Embryol. Rev. Roum. Morphol. Embryol. 2019, 60, 573–579. [Google Scholar]
  143. Zhan, Y.; Chen, Z.; Li, Y.; He, A.; He, S.; Gong, Y.; Li, X.; Zhou, L. Long non-coding RNA DANCR promotes malignant phenotypes of bladder cancer cells by modulating the miR-149/MSI2 axis as a ceRNA. J. Exp. Clin. Cancer Res. CR 2018, 37, 273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wang, Z.L.; Wang, C.; Liu, W.; Ai, Z.L. Emerging roles of the long non-coding RNA 01296/microRNA-143-3p/MSI2 axis in development of thyroid cancer. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef]
  145. Yeh, Y.; Guo, Q.; Connelly, Z.; Cheng, S.; Yang, S.; Prieto-Dominguez, N.; Yu, X. Wnt/Beta-Catenin Signaling and Prostate Cancer Therapy Resistance. Adv. Exp. Med. Biol. 2019, 1210, 351–378. [Google Scholar] [CrossRef]
  146. Duggimpudi, S.; Kloetgen, A.; Maney, S.K.; Munch, P.C.; Hezaveh, K.; Shaykhalishahi, H.; Hoyer, W.; McHardy, A.C.; Lang, P.A.; Borkhardt, A.; et al. Transcriptome-wide analysis uncovers the targets of the RNA-binding protein MSI2 and effects of MSI2’s RNA-binding activity on IL-6 signaling. J. Biol. Chem. 2018, 293, 15359–15369. [Google Scholar] [CrossRef] [Green Version]
  147. Sheng, W.; Shi, X.; Lin, Y.; Tang, J.; Jia, C.; Cao, R.; Sun, J.; Wang, G.; Zhou, L.; Dong, M. Musashi2 promotes EGF-induced EMT in pancreatic cancer via ZEB1-ERK/MAPK signaling. J. Exp. Clin. Cancer Res. CR 2020, 39, 16. [Google Scholar] [CrossRef] [Green Version]
  148. Brun, J.; Dieudonne, F.X.; Marty, C.; Muller, J.; Schule, R.; Patino-Garcia, A.; Lecanda, F.; Fromigue, O.; Marie, P.J. FHL2 silencing reduces Wnt signaling and osteosarcoma tumorigenesis in vitro and in vivo. PLoS ONE 2013, 8, e55034. [Google Scholar] [CrossRef] [Green Version]
  149. Zienert, E.; Eke, I.; Aust, D.; Cordes, N. LIM-only protein FHL2 critically determines survival and radioresistance of pancreatic cancer cells. Cancer Lett. 2015, 364, 17–24. [Google Scholar] [CrossRef]
  150. Ding, L.; Wang, Z.; Yan, J.; Yang, X.; Liu, A.; Qiu, W.; Zhu, J.; Han, J.; Zhang, H.; Lin, J.; et al. Human four-and-a-half LIM family members suppress tumor cell growth through a TGF-beta-like signaling pathway. J. Clin. Investig. 2009, 119, 349–361. [Google Scholar] [CrossRef] [Green Version]
  151. Zhao, J.L.; Liang, S.Q.; Fu, W.; Zhu, B.K.; Li, S.Z.; Han, H.; Qin, H.Y. The LIM domain protein FHL1C interacts with tight junction protein ZO-1 contributing to the epithelial-mesenchymal transition (EMT) of a breast adenocarcinoma cell line. Gene 2014, 542, 182–189. [Google Scholar] [CrossRef] [PubMed]
  152. Yi, X.; Deng, X.; Zhao, Y.; Deng, B.; Deng, J.; Fan, H.; Du, Y.; Hao, L. Ubiquitin-like protein FAT10 promotes osteosarcoma growth by modifying the ubiquitination and degradation of YAP1. Exp. Cell Res. 2020, 387, 111804. [Google Scholar] [CrossRef] [PubMed]
  153. Zanconato, F.; Cordenonsi, M.; Piccolo, S. YAP/TAZ at the Roots of Cancer. Cancer Cell 2016, 29, 783–803. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, X.-H.; Wu, H.-Y.; Gao, J.; Wang, X.-H.; Gao, T.-H.; Zhang, S.-F. IGF1R facilitates epithelial-mesenchymal transition and cancer stem cell properties in neuroblastoma via the STAT3/AKT axis. Cancer Manag. Res. 2019, 11, 5459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Yokota, T.; Nagai, H.; Harada, H.; Mine, N.; Terada, Y.; Fujiwara, H.; Yabe, A.; Miyazaki, K.; Emi, M. Identification, tissue expression, and chromosomal position of a novel gene encoding human ubiquitin-conjugating enzyme E2-230k. Gene 2001, 267, 95–100. [Google Scholar] [CrossRef]
  156. Xing, Z.; Li, S.; Liu, Z.; Zhang, C.; Bai, Z. CTCF-induced upregulation of HOXA11-AS facilitates cell proliferation and migration by targeting miR-518b/ACTN4 axis in prostate cancer. Prostate 2020. [Google Scholar] [CrossRef]
  157. Georgakilas, A.G.; Tsantoulis, P.; Kotsinas, A.; Michalopoulos, I.; Townsend, P.; Gorgoulis, V.G. Are common fragile sites merely structural domains or highly organized “functional” units susceptible to oncogenic stress? Cell. Mol. Life Sci. 2014, 71, 4519–4544. [Google Scholar] [CrossRef]
  158. Zhang, X.; Zhang, B.; Zhang, P.; Lian, L.; Li, L.; Qiu, Z.; Qian, K.; Chen, A.; Liu, Q.; Jiang, Y.; et al. Norcantharidin regulates ERalpha signaling and tamoxifen resistance via targeting miR-873/CDK3 in breast cancer cells. PLoS ONE 2019, 14, e0217181. [Google Scholar] [CrossRef] [Green Version]
  159. Zhang, L.; Chen, Y.; Li, F.; Bao, L.; Liu, W. Atezolizumab and Bevacizumab attenuate cisplatin resistant ovarian cancer cells progression synergistically via suppressing epithelial-mesenchymal transition. Front. Immunol. 2019, 10, 867. [Google Scholar] [CrossRef] [Green Version]
  160. Liang, H.; Yu, M.; Yang, R.; Zhang, L.; Zhang, L.; Zhu, D.; Luo, H.; Hong, Y.; Yu, T.; Sun, J. A PTAL-miR-101-FN1 axis promotes EMT and invasion-metastasis in serous ovarian cancer. Mol. Ther. Oncolytics 2020, 16, 53–62. [Google Scholar] [CrossRef]
  161. Montanuy, H.; Martinez-Barriocanal, A.; Casado, J.A.; Rovirosa, L.; Ramirez, M.J.; Nieto, R.; Carrascoso-Rubio, C.; Riera, P.; Gonzalez, A.; Lerma, E.; et al. Gefitinib and afatinib show potential efficacy for Fanconi anemia-related head and neck cancer. Clin. Cancer Res. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Chih-Hsin Yang, J.; Schuler, M.; Popat, S.; Miura, S.; Heeke, S.; Park, K.; Marten, A.; Kim, E.S. Afatinib for the Treatment of Non-Small Cell Lung Cancer Harboring Uncommon EGFR Mutations: A Database of 693 Cases. J. Thorac. Oncol. 2020. [Google Scholar] [CrossRef] [Green Version]
  163. Poh, M.E.; Liam, C.K.; Rajadurai, P.; Chai, C.S. Epithelial-to-mesenchymal transition (EMT) causing acquired resistance to afatinib in a patient with epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma. J. Thorac. Dis. 2018, 10, E560–E563. [Google Scholar] [CrossRef] [PubMed]
  164. Guo, X.F.; Wang, A.Y.; Liu, J. HIFs-MiR-33a-Twsit1 axis can regulate invasiveness of hepatocellular cancer cells. Eur. Rev. Med Pharmacol. Sci. 2016, 20, 3011–3016. [Google Scholar] [PubMed]
  165. Ryan, M.R.; Sohl, C.D.; Luo, B.; Anderson, K.S. The FGFR1 V561M Gatekeeper Mutation Drives AZD4547 Resistance through STAT3 Activation and EMT. Mol. Cancer Res. MCR 2019, 17, 532–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Rowinsky, E.K.; Donehower, R.C. Paclitaxel (taxol). N. Engl. J. Med. 1995, 332, 1004–1014. [Google Scholar] [CrossRef] [PubMed]
  167. Zhu, L.; Chen, L. Progress in research on paclitaxel and tumor immunotherapy. Cell. Mol. Biol. Lett. 2019, 24, 40. [Google Scholar] [CrossRef] [Green Version]
  168. Li, L.; Xu, Q.; Dong, Y.; Li, G.; Yang, L.; Wang, L.; Li, H. MiR-181a upregulation is associated with epithelial-to-mesenchymal transition (EMT) and multidrug resistance (MDR) of ovarian cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2004–2010. [Google Scholar]
  169. Jing, L.; Bo, W.; Yourong, F.; Tian, W.; Shixuan, W.; Mingfu, W. Sema4C mediates EMT inducing chemotherapeutic resistance of miR-31-3p in cervical cancer cells. Sci. Rep. 2019, 9, 17727. [Google Scholar] [CrossRef]
  170. Wang, L.; Zhang, F.; Cui, J.Y.; Chen, L.; Chen, Y.T.; Liu, B.W. CAFs enhance paclitaxel resistance by inducing EMT through the IL6/JAK2/STAT3 pathway. Oncol. Rep. 2018, 39, 2081–2090. [Google Scholar] [CrossRef] [Green Version]
  171. Shi, Y.; Zhang, J.; Liu, M.; Huang, Y.; Yin, L. SMAD3 inducing the transcription of STYK1 to promote the EMT process and improve the tolerance of ovarian carcinoma cells to paclitaxel. J. Cell. Biochem. 2019, 120, 10796–10811. [Google Scholar] [CrossRef]
  172. Sale, M.J.; Balmanno, K.; Saxena, J.; Ozono, E.; Wojdyla, K.; McIntyre, R.E.; Gilley, R.; Woroniuk, A.; Howarth, K.D.; Hughes, G.; et al. MEK1/2 inhibitor withdrawal reverses acquired resistance driven by BRAF(V600E) amplification whereas KRAS(G13D) amplification promotes EMT-chemoresistance. Nat. Commun. 2019, 10, 2030. [Google Scholar] [CrossRef] [Green Version]
  173. Chang, Y.S.; Jalgaonkar, S.P.; Middleton, J.D.; Hai, T. Stress-inducible gene Atf3 in the noncancer host cells contributes to chemotherapy-exacerbated breast cancer metastasis. Proc. Natl. Acad. Sci. USA 2017, 114, E7159–E7168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. De Palma, M.; Lewis, C.E. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 2013, 23, 277–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Karagiannis, G.S.; Pastoriza, J.M.; Wang, Y.; Harney, A.S.; Entenberg, D.; Pignatelli, J.; Sharma, V.P.; Xue, E.A.; Cheng, E.; D’Alfonso, T.M.; et al. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Ruffell, B.; Coussens, L.M. Macrophages and therapeutic resistance in cancer. Cancer Cell 2015, 27, 462–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Gordon, S.; Pluddemann, A.; Martinez Estrada, F. Macrophage heterogeneity in tissues: Phenotypic diversity and functions. Immunol. Rev. 2014, 262, 36–55. [Google Scholar] [CrossRef] [Green Version]
  178. Wang, H.; Wang, X.; Li, X.; Fan, Y.; Li, G.; Guo, C.; Zhu, F.; Zhang, L.; Shi, Y. CD68+HLA-DR+ M1-like macrophages promote motility of HCC cells via NF-κB/FAK pathway. Cancer Lett. 2014, 345, 91–99. [Google Scholar] [CrossRef]
  179. Cho, U.; Kim, B.; Kim, S.; Han, Y.; Song, Y.S. Pro-inflammatory M1 macrophage enhances metastatic potential of ovarian cancer cells through NF-κB activation. Mol. Carcinog. 2018, 57, 235–242. [Google Scholar] [CrossRef]
  180. Xiao, M.; Zhang, J.; Chen, W.; Chen, W. M1-like tumor-associated macrophages activated by exosome-transferred THBS1 promote malignant migration in oral squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 143. [Google Scholar] [CrossRef] [Green Version]
  181. Liu, W.; Wang, W.; Wang, X.; Xu, C.; Zhang, N.; Di, W. Cisplatin-stimulated macrophages promote ovarian cancer migration via the CCL20-CCR6 axis. Cancer Lett. 2020, 472, 59–69. [Google Scholar] [CrossRef] [PubMed]
  182. Chen, K.J.; Lin, S.Z.; Zhou, L.; Xie, H.Y.; Zhou, W.H.; Taki-Eldin, A.; Zheng, S.S. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS ONE 2011, 6, e24671. [Google Scholar] [CrossRef]
  183. Kryczek, I.; Lin, Y.; Nagarsheth, N.; Peng, D.; Zhao, L.; Zhao, E.; Vatan, L.; Szeliga, W.; Dou, Y.; Owens, S.; et al. IL-22+CD4+ T Cells Promote Colorectal Cancer Stemness via STAT3 Transcription Factor Activation and Induction of the Methyltransferase DOT1L. Immunity 2014, 40, 772–784. [Google Scholar] [CrossRef] [Green Version]
  184. Walch-Ruckheim, B.; Mavrova, R.; Henning, M.; Vicinus, B.; Kim, Y.J.; Bohle, R.M.; Juhasz-Boss, I.; Solomayer, E.F.; Smola, S. Stromal Fibroblasts Induce CCL20 through IL6/C/EBPbeta to Support the Recruitment of Th17 Cells during Cervical Cancer Progression. Cancer Res. 2015, 75, 5248–5259. [Google Scholar] [CrossRef] [Green Version]
  185. Ranasinghe, R.; Eri, R. Modulation of the CCR6-CCL20 Axis: A Potential Therapeutic Target in Inflammation and Cancer. Medicina 2018, 54, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Bernstein, J.L.; Group, W.S.C.; Concannon, P. ATM, radiation, and the risk of second primary breast cancer. Int. J. Radiat. Biol. 2017, 93, 1121–1127. [Google Scholar] [CrossRef] [PubMed]
  187. Nanda, N.; Roberts, N.J. ATM Serine/Threonine Kinase and its Role in Pancreatic Risk. Genes (Basel) 2020, 11, 108. [Google Scholar] [CrossRef] [Green Version]
  188. Murai, J.; Thomas, A.; Miettinen, M.; Pommier, Y. Schlafen 11 (SLFN11), a restriction factor for replicative stress induced by DNA-targeting anti-cancer therapies. Pharmacol. Ther. 2019, 201, 94–102. [Google Scholar] [CrossRef]
  189. Allison Stewart, C.; Tong, P.; Cardnell, R.J.; Sen, T.; Li, L.; Gay, C.M.; Masrorpour, F.; Fan, Y.; Bara, R.O.; Feng, Y.; et al. Dynamic variations in epithelial-to-mesenchymal transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget 2017, 8, 28575–28587. [Google Scholar] [CrossRef] [Green Version]
  190. Fang, S.; Yu, L.; Mei, H.; Yang, J.; Gao, T.; Cheng, A.; Guo, W.; Xia, K.; Liu, G. Cisplatin promotes mesenchymal-like characteristics in osteosarcoma through Snail. Oncol. Lett. 2016, 12, 5007–5014. [Google Scholar] [CrossRef]
  191. Liu, Y.Q.; Zhang, G.A.; Zhang, B.C.; Wang, Y.; Liu, Z.; Jiao, Y.L.; Liu, N.; Zhao, Y.R. Short low concentration cisplatin treatment leads to an epithelial mesenchymal transition-like response in DU145 prostate cancer cells. Asian Pac. J. Cancer Prev. APJCP 2015, 16, 1025–1028. [Google Scholar] [CrossRef] [PubMed]
  192. Miow, Q.H.; Tan, T.Z.; Ye, J.; Lau, J.A.; Yokomizo, T.; Thiery, J.P.; Mori, S. Epithelial-mesenchymal status renders differential responses to cisplatin in ovarian cancer. Oncogene 2015, 34, 1899–1907. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, J.; Yang, M.; Kang, R.; Klionsky, D.J.; Tang, D. Autophagic degradation of the circadian clock regulator promotes ferroptosis. Autophagy 2019, 15, 2033–2035. [Google Scholar] [CrossRef] [PubMed]
  194. Liu, X.; Jin, M.; Yao, Z.; Bernard, A.; Klionsky, D.J. Bidirectional roles of Dhh1 in regulating autophagy. Autophagy 2019, 15, 1838–1839. [Google Scholar] [CrossRef] [PubMed]
  195. Klionsky, D.J. Autophagy participates in, well, just about everything. Cell Death Differ. 2020. [Google Scholar] [CrossRef] [Green Version]
  196. Yang, Y.; Klionsky, D.J. Autophagy and disease: Unanswered questions. Cell Death Differ. 2020. [Google Scholar] [CrossRef]
  197. Tyutyunyk-Massey, L.; Gewirtz, D.A. Roles of autophagy in breast cancer treatment: Target, bystander or benefactor. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
  198. Towers, C.G.; Fitzwalter, B.E.; Regan, D.; Goodspeed, A.; Morgan, M.J.; Liu, C.-W.; Gustafson, D.L.; Thorburn, A. Cancer Cells Upregulate NRF2 Signaling to Adapt to Autophagy Inhibition. Dev. Cell 2019, 50, 690–703.e696. [Google Scholar] [CrossRef]
  199. Kocaturk, N.M.; Akkoc, Y.; Kig, C.; Bayraktar, O.; Gozuacik, D.; Kutlu, O. Autophagy as a molecular target for cancer treatment. Eur. J. Pharm. Sci. 2019, 134, 116–137. [Google Scholar] [CrossRef]
  200. Quan, Y.; Lei, H.; Wahafu, W.; Liu, Y.; Ping, H.; Zhang, X. Inhibition of autophagy enhances the anticancer effect of enzalutamide on bladder cancer. Biomed. Pharmacother. 2019, 120, 109490. [Google Scholar] [CrossRef]
  201. Lei, Y.; Tang, L.; Hu, J.; Wang, S.; Liu, Y.; Yang, M.; Zhang, J.; Tang, B. Inhibition of MGMT-mediated autophagy suppression decreases cisplatin chemosensitivity in gastric cancer. Biomed. Pharmacother. 2020, 125, 109896. [Google Scholar] [CrossRef] [PubMed]
  202. Long, M.; McWilliams, T.G. Monitoring autophagy in cancer: From bench to bedside. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef] [PubMed]
  203. Su, Z.; Li, G.; Liu, C.; Ren, S.; Deng, T.; Zhang, S.; Tian, Y.; Liu, Y.; Qiu, Y. Autophagy inhibition impairs the epithelial-mesenchymal transition and enhances cisplatin sensitivity in nasopharyngeal carcinoma. Oncol. Lett. 2017, 13, 4147–4154. [Google Scholar] [CrossRef] [Green Version]
  204. Mina-Osorio, P. The moonlighting enzyme CD13: Old and new functions to target. Trends Mol. Med. 2008, 14, 361–371. [Google Scholar] [CrossRef]
  205. Carl-McGrath, S.; Lendeckel, U.; Ebert, M.; Wolter, A.-B.; Roessner, A.; Röcken, C. The ectopeptidases CD10, CD13, CD26, and CD143 are upregulated in gastric cancer. Int. J. Oncol. 2004, 25, 1223–1232. [Google Scholar] [CrossRef]
  206. Ben-Porath, I.; Kozak, C.A.; Benvenisty, N. Chromosomal Mapping ofTmp (Emp1), Xmp (Emp2), andYmp (Emp3), Genes Encoding Membrane Proteins Related toPmp22. Genomics 1998, 49, 443–447. [Google Scholar] [CrossRef]
  207. Mikata, R.; Yokosuka, O.; Fukai, K.; Imazeki, F.; Arai, M.; Tada, M.; Kurihara, T.; Zhang, K.; Kanda, T.; Saisho, H. Analysis of genes upregulated by the demethylating agent 5-aza-2′-deoxycytidine in gastric cancer cell lines. Int. J. Cancer 2006, 119, 1616–1622. [Google Scholar] [CrossRef]
  208. Guo, Q.; Jing, F.J.; Xu, W.; Li, X.; Li, X.; Sun, J.L.; Xing, X.M.; Zhou, C.K.; Jing, F.B. Ubenimex induces autophagy inhibition and EMT suppression to overcome cisplatin resistance in GC cells by perturbing the CD13/EMP3/PI3K/AKT/NF-kappaB axis. Aging 2019, 12, 80. [Google Scholar] [CrossRef]
  209. Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
  210. Kahlert, C.; Kalluri, R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J. Mol. Med. 2013, 91, 431–437. [Google Scholar] [CrossRef] [Green Version]
  211. Qu, L.; Ding, J.; Chen, C.; Wu, Z.-J.; Liu, B.; Gao, Y.; Chen, W.; Liu, F.; Sun, W.; Li, X.-F. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell 2016, 29, 653–668. [Google Scholar] [CrossRef] [PubMed]
  212. Xu, C.; Yang, M.; Ren, Y.; Wu, C.; Wang, L. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4362–4368. [Google Scholar] [PubMed]
  213. Wang, J.; Lv, B.; Su, Y.; Wang, X.; Bu, J.; Yao, L. Exosome-Mediated Transfer of lncRNA HOTTIP Promotes Cisplatin Resistance in Gastric Cancer Cells by Regulating HMGA1/miR-218 Axis. OncoTargets Ther. 2019, 12, 11325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Zhu, G.; Wang, Z.; Mijiti, M.; Du, G.; Li, Y.; Dangmurenjiafu, G. MiR-28-5p promotes human glioblastoma cell growth through inactivation of FOXO1. Int. J. Clin. Exp. Pathol. 2019, 12, 2972. [Google Scholar]
  215. Xie, C.; Guo, Y.; Lou, S. LncRNA ANCR Promotes Invasion and Migration of Gastric Cancer by Regulating FoxO1 Expression to Inhibit Macrophage M1 Polarization. Dig. Dis. Sci. 2020, 1–10. [Google Scholar] [CrossRef]
  216. Zhao, M.; Luo, R.; Liu, Y.; Gao, L.; Fu, Z.; Fu, Q.; Luo, X.; Chen, Y.; Deng, X.; Liang, Z. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR–p-PI3K/AKT-c-JUN. Nat. Commun. 2016, 7, 11309. [Google Scholar] [CrossRef] [Green Version]
  217. Li, Y.; Liu, X.; Lin, X.; Zhao, M.; Xiao, Y.; Liu, C.; Liang, Z.; Lin, Z.; Yi, R.; Tang, Z.; et al. Chemical compound cinobufotalin potently induces FOXO1-stimulated cisplatin sensitivity by antagonizing its binding partner MYH9. Signal Transduct. Target. Ther. 2019, 4, 48. [Google Scholar] [CrossRef]
  218. Zhang, F.; Yin, Y.; Xu, T. Cinobufotalin injection combined with chemotherapy for the treatment of advanced NSCLC in China: A PRISMA-compliant meta-analysis of 29 randomized controlled trials. Medicine 2019, 98, e16969. [Google Scholar] [CrossRef]
  219. Mirzaei, H.; Momeni, F.; Saadatpour, L.; Sahebkar, A.; Goodarzi, M.; Masoudifar, A.; Kouhpayeh, S.; Salehi, H.; Mirzaei, H.R.; Jaafari, M.R. MicroRNA: Relevance to stroke diagnosis, prognosis, and therapy. J. Cell. Physiol. 2018, 233, 856–865. [Google Scholar] [CrossRef]
  220. Moridikia, A.; Mirzaei, H.; Sahebkar, A.; Salimian, J. MicroRNAs: Potential candidates for diagnosis and treatment of colorectal cancer. J. Cell. Physiol. 2018, 233, 901–913. [Google Scholar] [CrossRef]
  221. Hermann, H.; Runnel, T.; Aab, A.; Baurecht, H.; Rodriguez, E.; Magilnick, N.; Urgard, E.; Šahmatova, L.; Prans, E.; Maslovskaja, J. miR-146b probably assists miRNA-146a in the suppression of keratinocyte proliferation and inflammatory responses in psoriasis. J. Investig. Dermatol. 2017, 137, 1945–1954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Deng, X.; Wu, B.; Xiao, K.; Kang, J.; Xie, J.; Zhang, X.; Fan, Y. MiR-146b-5p promotes metastasis and induces epithelial-mesenchymal transition in thyroid cancer by targeting ZNRF3. Cell. Physiol. Biochem. 2015, 35, 71–82. [Google Scholar] [CrossRef] [PubMed]
  223. Balducci, E.; Leroyer, A.S.; Lacroix, R.; Robert, S.; Todorova, D.; Simoncini, S.; Lyonnet, L.; Chareyre, C.; Zaegel-Faucher, O.; Micallef, J. Microvesicles from T cells overexpress miR-146b-5p in HIV-1 infection and repress endothelial activation. J. Extracell. Vesicles 2018, 7, 71–72. [Google Scholar]
  224. Cho, S.; Lee, H.-M.; Yu, I.-S.; Choi, Y.S.; Huang, H.-Y.; Hashemifar, S.S.; Lin, L.-L.; Chen, M.-C.; Afanasiev, N.D.; Khan, A.A. Differential cell-intrinsic regulations of germinal center B and T cells by miR-146a and miR-146b. Nat. Commun. 2018, 9, 2757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Labbé, D.P.; Tremblay, M.L. PTP1B: From Metabolism to Cancer. In Protein Tyrosine Phosphatases in Cancer; Springer: Berlin/Heidelberg, Germany, 2016; pp. 169–199. [Google Scholar]
  226. Wang, N.; She, J.; Liu, W.; Shi, J.; Yang, Q.; Shi, B.; Hou, P. Frequent amplification of PTP1B is associated with poor survival of gastric cancer patients. Cell Cycle 2015, 14, 732–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Han, Q.; Cheng, P.; Yang, H.; Liang, H.; Lin, F. miR-146b Reverses epithelial-mesenchymal transition via targeting PTP1B in cisplatin-resistance human lung adenocarcinoma cells. J. Cell. Biochem. 2019. [Google Scholar] [CrossRef]
  228. Bian, W.G.; Zhou, X.N.; Song, S.; Chen, H.T.; Shen, Y.; Chen, P. Reduced miR-363-3p expression in non-small cell lung cancer is associated with gemcitabine resistance via targeting of CUL4A. Eur. Rev. Med Pharmacol. Sci. 2019, 23, 649–659. [Google Scholar] [CrossRef]
  229. Wang, X.; Ren, M.; Li, Y.; Hu, J.; Lu, G.; Ma, W.; Guo, D.; Lu, X.; He, S. Long noncoding RNA NNT-AS1 promotes gastric cancer proliferation and invasion by regulating microRNA-363 expression. J. Cell. Biochem. 2019, 120, 5704–5712. [Google Scholar] [CrossRef]
  230. Cao, L.; Wan, Q.; Li, F.; Tang, C.E. MiR-363 inhibits cisplatin chemoresistance of epithelial ovarian cancer by regulating snail-induced epithelial-mesenchymal transition. BMB Rep. 2018, 51, 456. [Google Scholar] [CrossRef] [Green Version]
  231. Miyazaki, H.; Takahashi, R.U.; Prieto-Vila, M.; Kawamura, Y.; Kondo, S.; Shirota, T.; Ochiya, T. CD44 exerts a functional role during EMT induction in cisplatin-resistant head and neck cancer cells. Oncotarget 2018, 9, 10029–10041. [Google Scholar] [CrossRef] [Green Version]
  232. Wang, F.; Wu, H.; Fan, M.; Yu, R.; Zhang, Y.; Liu, J.; Zhou, X.; Cai, Y.; Huang, S.; Hu, Z.; et al. Sodium butyrate inhibits migration and induces AMPK-mTOR pathway-dependent autophagy and ROS-mediated apoptosis via the miR-139-5p/Bmi-1 axis in human bladder cancer cells. FASEB J. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Liu, X.; Li, Y.; Wen, J.; Qi, T.; Wang, Y. Long non-coding RNA TTN-AS1 promotes tumorigenesis of ovarian cancer through modulating the miR-139-5p/ROCK2 axis. Biomed. Pharmacother. Biomed. Pharmacother. 2020, 125, 109882. [Google Scholar] [CrossRef] [PubMed]
  234. Qin, H.; Wen, D.Y.; Que, Q.; Zhou, C.Y.; Wang, X.D.; Peng, Y.T.; He, Y.; Yang, H.; Liao, B.M. Reduced expression of microRNA-139-5p in hepatocellular carcinoma results in a poor outcome: An exploration the roles of microRNA-139-5p in tumorigenesis, advancement and prognosis at the molecular biological level using an integrated meta-analysis and bioinformatic investigation. Oncol. Lett. 2019, 18, 6704–6724. [Google Scholar] [CrossRef] [PubMed]
  235. Huang, N.; Guo, W.; Ren, K.; Li, W.; Jiang, Y.; Sun, J.; Dai, W.; Zhao, W. LncRNA AFAP1-AS1 Supresses miR-139-5p and Promotes Cell Proliferation and Chemotherapy Resistance of Non-small Cell Lung Cancer by Competitively Upregulating RRM2. Front. Oncol. 2019, 9, 1103. [Google Scholar] [CrossRef] [Green Version]
  236. Shao, Q.; Zhang, P.; Ma, Y.; Lu, Z.; Meng, J.; Li, H.; Wang, X.; Chen, D.; Zhang, M.; Han, Y.; et al. MicroRNA-139-5p affects cisplatin sensitivity in human nasopharyngeal carcinoma cells by regulating the epithelial-to-mesenchymal transition. Gene 2018, 652, 48–58. [Google Scholar] [CrossRef]
  237. Nieto, M.A. Epithelial plasticity: A common theme in embryonic and cancer cells. Science 2013, 342, 1234850. [Google Scholar] [CrossRef] [Green Version]
  238. Meacham, C.E.; Morrison, S.J. Tumour heterogeneity and cancer cell plasticity. Nature 2013, 501, 328. [Google Scholar] [CrossRef] [Green Version]
  239. Najafi, M.; Mortezaee, K.; Majidpoor, J. Cancer stem cell (CSC) resistance drivers. Life Sci. 2019, 234, 116781. [Google Scholar] [CrossRef]
  240. Najafi, M.; Farhood, B.; Mortezaee, K. Cancer stem cells (CSCs) in cancer progression and therapy. J. Cell. Physiol. 2019, 234, 8381–8395. [Google Scholar] [CrossRef]
  241. Saha, S.; Mukherjee, S.; Khan, P.; Kajal, K.; Mazumdar, M.; Manna, A.; Mukherjee, S.; De, S.; Jana, D.; Sarkar, D.K. Aspirin suppresses the acquisition of chemoresistance in breast cancer by disrupting an NFκB–IL6 signaling axis responsible for the generation of cancer stem cells. Cancer Res. 2016, 76, 2000–2012. [Google Scholar] [CrossRef] [Green Version]
  242. Moore, S.F.; Hunter, R.W.; Hers, I. mTORC2 protein-mediated protein kinase B (Akt) serine 473 phosphorylation is not required for Akt1 activity in human platelets. J. Biol. Chem. 2011, 286, 24553–24560. [Google Scholar] [CrossRef] [Green Version]
  243. Singh, S.S.; Yap, W.N.; Arfuso, F.; Kar, S.; Wang, C.; Cai, W.; Dharmarajan, A.M.; Sethi, G.; Kumar, A.P. Targeting the PI3K/Akt signaling pathway in gastric carcinoma: A reality for personalized medicine? World J. Gastroenterol. 2015, 21, 12261–12273. [Google Scholar] [CrossRef] [PubMed]
  244. Siveen, K.S.; Ahn, K.S.; Ong, T.H.; Shanmugam, M.K.; Li, F.; Yap, W.N.; Kumar, A.P.; Fong, C.W.; Tergaonkar, V.; Hui, K.M.; et al. Y-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model. Oncotarget 2014, 5, 1897–1911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Ong, P.S.; Wang, L.Z.; Dai, X.; Tseng, S.H.; Loo, S.J.; Sethi, G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front. Pharmacol. 2016, 7, 395. [Google Scholar] [CrossRef] [PubMed]
  246. Lee, J.H.; Kim, C.; Um, J.Y.; Sethi, G.; Ahn, K.S. Casticin-Induced Inhibition of Cell Growth and Survival Are Mediated through the Dual Modulation of Akt/mTOR Signaling Cascade. Cancers 2019, 11, 254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Tu, Y.; Ji, C.; Yang, B.; Yang, Z.; Gu, H.; Lu, C.-C.; Wang, R.; Su, Z.-L.; Chen, B.; Sun, W.-L. DNA-dependent protein kinase catalytic subunit (DNA-PKcs)-SIN1 association mediates ultraviolet B (UVB)-induced Akt Ser-473 phosphorylation and skin cell survival. Mol. Cancer 2013, 12, 172. [Google Scholar] [CrossRef] [Green Version]
  248. Sunayama, J.; Matsuda, K.I.; Sato, A.; Tachibana, K.; Suzuki, K.; Narita, Y.; Shibui, S.; Sakurada, K.; Kayama, T.; Tomiyama, A. Crosstalk between the PI3K/mTOR and MEK/ERK pathways involved in the maintenance of self-renewal and tumorigenicity of glioblastoma stem-like cells. Stem Cells 2010, 28, 1930–1939. [Google Scholar] [CrossRef] [PubMed]
  249. Zheng, C.-H.; Wang, J.-B.; Lin, M.-Q.; Zhang, P.-Y.; Liu, L.-C.; Lin, J.-X.; Lu, J.; Chen, Q.-Y.; Cao, L.-L.; Lin, M. CDK5RAP3 suppresses Wnt/β-catenin signaling by inhibiting AKT phosphorylation in gastric cancer. J. Exp. Clin. Cancer Res. 2018, 37, 59. [Google Scholar] [CrossRef] [Green Version]
  250. Zheng, H.; Li, W.; Wang, Y.; Liu, Z.; Cai, Y.; Xie, T.; Shi, M.; Wang, Z.; Jiang, B. Glycogen synthase kinase-3 beta regulates Snail and β-catenin expression during Fas-induced epithelial–mesenchymal transition in gastrointestinal cancer. Eur. J. Cancer 2013, 49, 2734–2746. [Google Scholar] [CrossRef]
  251. Yuan, L.; Zhang, K.; Zhou, M.M.; Wasan, H.S.; Tao, F.F.; Yan, Q.Y.; Feng, G.; Tang, Y.S.; Shen, M.H.; Ma, S.L.; et al. Jiedu Sangen Decoction Reverses Epithelial-to-mesenchymal Transition and Inhibits Invasion and Metastasis of Colon Cancer via AKT/GSK-3beta Signaling Pathway. J. Cancer 2019, 10, 6439–6456. [Google Scholar] [CrossRef]
  252. Shin, J.H.; Kim, K.M.; Jeong, J.U.; Shin, J.M.; Kang, J.H.; Bang, K.; Kim, J.H. Nrf2-Heme Oxygenase-1 Attenuates High-Glucose-Induced Epithelial-to-Mesenchymal Transition of Renal Tubule Cells by Inhibiting ROS-Mediated PI3K/Akt/GSK-3beta Signaling. J. Diabetes Res. 2019, 2019, 2510105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Khan, P.; Bhattacharya, A.; Sengupta, D.; Banerjee, S.; Adhikary, A.; Das, T. Aspirin enhances cisplatin sensitivity of resistant non-small cell lung carcinoma stem-like cells by targeting mTOR-Akt axis to repress migration. Sci. Rep. 2019, 9, 1–15. [Google Scholar] [CrossRef] [PubMed]
  254. Bordonaro, M. Hypothesis: Retinoblastoma protein inactivation mediates effects of histone deacetylase inhibitor-induced Wnt hyperactivation in colorectal cancer cells. J. Cancer 2020, 11, 668–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Jarman, E.J.; Boulter, L. Targeting the Wnt signalling pathway: The challenge of reducing scarring without affecting repair. Expert Opin. Investig. Drugs 2020. [Google Scholar] [CrossRef]
  256. Ong, M.S.; Cai, W.; Yuan, Y.; Leong, H.C.; Tan, T.Z.; Mohammad, A.; You, M.L.; Arfuso, F.; Goh, B.C.; Warrier, S.; et al. ’Lnc’-ing Wnt in female reproductive cancers: Therapeutic potential of long non-coding RNAs in Wnt signalling. Br. J. Pharmacol. 2017, 174, 4684–4700. [Google Scholar] [CrossRef] [Green Version]
  257. Bhuvanalakshmi, G.; Basappa; Rangappa, K.S.; Dharmarajan, A.; Sethi, G.; Kumar, A.P.; Warrier, S. Breast Cancer Stem-Like Cells Are Inhibited by Diosgenin, a Steroidal Saponin, by the Attenuation of the Wnt beta-Catenin Signaling via the Wnt Antagonist Secreted Frizzled Related Protein-4. Front. Pharmacol. 2017, 8, 124. [Google Scholar] [CrossRef] [Green Version]
  258. Yi, Z.; Pu, Y.; Gou, R.; Chen, Y.; Ren, X.; Liu, W.; Dong, P. Silencing of RIPK4 inhibits epithelialmesenchymal transition by inactivating the Wnt/betacatenin signaling pathway in osteosarcoma. Mol. Med. Rep. 2020. [Google Scholar] [CrossRef] [Green Version]
  259. Lu, Y.; Sun, W.; Zhang, L.; Li, J. Silencing Of MAGI1 Promotes The Proliferation And Inhibits Apoptosis Of Glioma Cells Via The Wnt/beta-Catenin And PTEN/AKT Signaling Pathways. OncoTargets Ther. 2019, 12, 9639–9650. [Google Scholar] [CrossRef] [Green Version]
  260. Peng, Y.; Xu, Y.; Yang, G.; Li, S.; Rui, Z. Knockdown Of Long Non-Coding RNA TP73-AS1 Inhibited Cell Proliferation And Metastasis Through Wnt/beta-Catenin Pathway In Lung Adenocarcinoma. OncoTargets Ther. 2019, 12, 9599–9610. [Google Scholar] [CrossRef] [Green Version]
  261. Cheng, C.; Huang, Z.; Zhou, R.; An, H.; Cao, G.; Ye, J.; Huang, C.; Wu, D. Numb negatively regulates the epithelial-to-mesenchymal transition in colorectal cancer through the Wnt signalling pathway. Am. J. Physiol. Gastrointest. Liver Physiol. 2020. [Google Scholar] [CrossRef]
  262. Wu, W.; Guo, L.; Liang, Z.; Liu, Y.; Yao, Z. Lnc-SNHG16/miR-128 axis modulates malignant phenotype through WNT/beta-catenin pathway in cervical cancer cells. J. Cancer 2020, 11, 2201–2212. [Google Scholar] [CrossRef] [PubMed]
  263. Liu, J.; Zhao, Z.; Sun, Z.; Liu, C.; Cheng, X.; Ruge, F.; Yang, Y.; Jiang, W.G.; Ye, L. Increased expression of Psoriasin is correlated with poor prognosis of bladder transitional cell carcinoma by promoting invasion and proliferation. Oncol. Rep. 2020, 43, 562–570. [Google Scholar] [CrossRef] [PubMed]
  264. Pan, J.; Fang, S.; Tian, H.; Zhou, C.; Zhao, X.; Tian, H.; He, J.; Shen, W.; Meng, X.; Jin, X.; et al. lncRNA JPX/miR-33a-5p/Twist1 axis regulates tumorigenesis and metastasis of lung cancer by activating Wnt/beta-catenin signaling. Mol. Cancer 2020, 19, 9. [Google Scholar] [CrossRef] [PubMed]
  265. Niu, Q.; Liu, Z.; Gao, J.; Wang, Q. MiR-338-3p Enhances Ovarian Cancer Cell Sensitivity to Cisplatin by Downregulating WNT2B. Yonsei Med. J. 2019, 60, 1146–1156. [Google Scholar] [CrossRef]
  266. Worthmuller, J.; Salicio, V.; Oberson, A.; Blum, W.; Schwaller, B. Modulation of Calretinin Expression in Human Mesothelioma Cells Reveals the Implication of the FAK and Wnt Signaling Pathways in Conferring Chemoresistance towards Cisplatin. Int. J. Mol. Sci. 2019, 20, 5391. [Google Scholar] [CrossRef] [Green Version]
  267. Liu, Y.; Jiang, Q.; Liu, X.; Lin, X.; Li, Y.; Tang, Z.; Liu, C.; Zhao, M.; Li, X.; Liu, Z. Cinobufotalin Powerfully Reversed EBV-miR-BART22-Induced Cisplatin Resistance via Stimulating MAP2K4 to Antagonize Non-Muscle Myosin Heavy Chain IIA/Glycogen Synthase 3β/β-Catenin Signaling Pathway. EBioMedicine 2019, 48, 386–404. [Google Scholar] [CrossRef] [Green Version]
  268. Ma, L.H.; Grove, C.L.; Baker, R. Development of oculomotor circuitry independent of hox3 genes. Nat. Commun. 2014, 5, 4221. [Google Scholar] [CrossRef] [Green Version]
  269. Sun, H.; Chen, J.; Qian, W.; Kang, J.; Wang, J.; Jiang, L.; Qiao, L.; Chen, W.; Zhang, J. Integrated long non-coding RNA analyses identify novel regulators of epithelial-mesenchymal transition in the mouse model of pulmonary fibrosis. J. Cell. Mol. Med. 2016, 20, 1234–1246. [Google Scholar] [CrossRef] [Green Version]
  270. Sun, J.; Gu, X.; Wu, N.; Zhang, P.; Liu, Y.; Jiang, S. Human antigen R enhances the epithelial-mesenchymal transition via regulation of ZEB-1 in the human airway epithelium. Respir. Res. 2018, 19, 109. [Google Scholar] [CrossRef] [Green Version]
  271. Tong, Y.; Wang, M.; Dai, Y.; Bao, D.; Zhang, J.; Pan, H. LncRNA HOXA-AS3 Sponges miR-29c to Facilitate Cell Proliferation, Metastasis, and EMT Process and Activate the MEK/ERK Signaling Pathway in Hepatocellular Carcinoma. Hum. Gene Ther. Clin. Dev. 2019, 30, 129–141. [Google Scholar] [CrossRef]
  272. Gao, H.; Yang, J.Y.; Tong, L.X.; Jin, H.; Liu, C.Z. Long noncoding RNA UCA1 promotes proliferation and metastasis of thyroid cancer cells by sponging miR-497-3p. Eur. Rev. Med Pharmacol. Sci. 2020, 24, 728–734. [Google Scholar] [CrossRef] [PubMed]
  273. Jayaganesh, R.; Pugalendhi, P.; Murali, R. Effect of citronellol on NF-kB inflammatory signaling molecules in chemical carcinogen-induced mammary cancer in the rat model. J. Biochem. Mol. Toxicol. 2020, e22441. [Google Scholar] [CrossRef] [PubMed]
  274. Zha, Z.; Han, Q.; Liu, W.; Huo, S. lncRNA GAS8-AS1 downregulates lncRNA UCA1 to inhibit osteosarcoma cell migration and invasion. J. Orthop. Surg. Res. 2020, 15, 38. [Google Scholar] [CrossRef] [PubMed]
  275. Liu, X.; Huang, Z.; Qian, W.; Zhang, Q.; Sun, J. Silence of lncRNA UCA1 rescues drug resistance of cisplatin to non-small-cell lung cancer cells. J. Cell. Biochem. 2019, 120, 9243–9249. [Google Scholar] [CrossRef]
  276. Puar, Y.R.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; Sethi, G.; Tergaonkar, V. Evidence for the Involvement of the Master Transcription Factor NF-kappaB in Cancer Initiation and Progression. Biomedicines 2018, 6, 82. [Google Scholar] [CrossRef] [Green Version]
  277. Shin, E.M.; Hay, H.S.; Lee, M.H.; Goh, J.N.; Tan, T.Z.; Sen, Y.P.; Lim, S.W.; Yousef, E.M.; Ong, H.T.; Thike, A.A.; et al. DEAD-box helicase DP103 defines metastatic potential of human breast cancers. J. Clin. Investig. 2014, 124, 3807–3824. [Google Scholar] [CrossRef]
  278. Sethi, G.; Ahn, K.S.; Sung, B.; Aggarwal, B.B. Pinitol targets nuclear factor-κB activation pathway leading to inhibition of gene products associated with proliferation, apoptosis, invasion, and angiogenesis. Mol. Cancer Ther. 2008, 7, 1604–1614. [Google Scholar] [CrossRef] [Green Version]
  279. Sethi, G.; Sung, B.; Kunnumakkara, A.B.; Aggarwal, B.B. Targeting TNF for Treatment of Cancer and Autoimmunity. Adv. Exp. Med. Biol. 2009, 647, 37–51. [Google Scholar] [CrossRef]
  280. Ahn, K.S.; Sethi, G.; Jain, A.K.; Jaiswal, A.K.; Aggarwal, B.B. Genetic deletion of NAD(P)H:quinone oxidoreductase 1 abrogates activation of nuclear factor-kappaB, IkappaBalpha kinase, c-Jun N-terminal kinase, Akt, p38, and p44/42 mitogen-activated protein kinases and potentiates apoptosis. J. Biol. Chem. 2006, 281, 19798–19808. [Google Scholar] [CrossRef] [Green Version]
  281. Siveen, K.S.; Mustafa, N.; Li, F.; Kannaiyan, R.; Ahn, K.S.; Kumar, A.P.; Chng, W.J.; Sethi, G. Thymoquinone overcomes chemoresistance and enhances the anticancer effects of bortezomib through abrogation of NF-kappaB regulated gene products in multiple myeloma xenograft mouse model. Oncotarget 2014, 5, 634–648. [Google Scholar] [CrossRef] [Green Version]
  282. Nottingham, L.K.; Yan, C.H.; Yang, X.; Si, H.; Coupar, J.; Bian, Y.; Cheng, T.-F.; Allen, C.; Arun, P.; Gius, D. Aberrant IKKα and IKKβ cooperatively activate NF-κB and induce EGFR/AP1 signaling to promote survival and migration of head and neck cancer. Oncogene 2014, 33, 1135–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Psyrri, A.; Seiwert, T.Y.; Jimeno, A. Molecular pathways in head and neck cancer: EGFR, PI3K, and more. Am. Soc. Clin. Oncol. Educ. Book 2013, 33, 246–255. [Google Scholar] [CrossRef] [PubMed]
  284. Shostak, K.; Chariot, A. EGFR and NF-κB: Partners in cancer. Trends Mol. Med. 2015, 21, 385–393. [Google Scholar] [CrossRef] [PubMed]
  285. Wang, F.; Arun, P.; Friedman, J.; Chen, Z.; Van Waes, C. Current and potential inflammation targeted therapies in head and neck cancer. Curr. Opin. Pharmacol. 2009, 9, 389–395. [Google Scholar] [CrossRef] [Green Version]
  286. Manu, K.A.; Shanmugam, M.K.; Li, F.; Chen, L.; Siveen, K.S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J. Mol. Med. 2014, 92, 267–276. [Google Scholar] [CrossRef] [Green Version]
  287. Li, F.; Shanmugam, M.K.; Chen, L.; Chatterjee, S.; Basha, J.; Kumar, A.P.; Kundu, T.K.; Sethi, G. Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev. Res. 2013, 6, 843–854. [Google Scholar] [CrossRef] [Green Version]
  288. Manu, K.A.; Shanmugam, M.K.; Ramachandran, L.; Li, F.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Arfuso, F.; Kumar, A.P.; et al. Isorhamnetin augments the anti-tumor effect of capecitabine through the negative regulation of NF-kappaB signaling cascade in gastric cancer. Cancer Lett. 2015, 363, 28–36. [Google Scholar] [CrossRef]
  289. Liao, J.; Yang, Z.; Carter-Cooper, B.; Chang, E.T.; Choi, E.Y.; Kallakury, B.; Liu, X.; Lapidus, R.G.; Cullen, K.J.; Dan, H. Suppression of migration, invasion, and metastasis of cisplatin-resistant head and neck squamous cell carcinoma through IKKβ inhibition. Clin. Exp. Metastasis 2020, 37, 283–292. [Google Scholar] [CrossRef]
  290. He, Y.; Xie, H.; Yu, P.; Jiang, S.; Wei, L. FOXC2 promotes epithelial-mesenchymal transition and cisplatin resistance of non-small cell lung cancer cells. Cancer Chemother. Pharmacol. 2018, 82, 1049–1059. [Google Scholar] [CrossRef]
  291. Soleimani, F.; Hajjari, M.; Mohammad Soltani, B.; Behmanesh, M. Up-Regulation of FOXC2 and FOXQ1 Is Associated with The Progression of Gastric-Type Adenocarcinoma. Cell J. 2017, 19, 66–71. [Google Scholar] [CrossRef]
  292. Song, L.; Tang, H.; Liao, W.; Luo, X.; Li, Y.; Chen, T.; Zhang, X. FOXC2 positively regulates YAP signaling and promotes the glycolysis of nasopharyngeal carcinoma. Exp. Cell Res. 2017, 357, 17–24. [Google Scholar] [CrossRef] [PubMed]
  293. Hargadon, K.M.; Gyorffy, B.; Strong, E.W.; Tarnai, B.D.; Thompson, J.C.; Bushhouse, D.Z.; Johnson, C.E.; Williams, C.J. The FOXC2 Transcription Factor Promotes Melanoma Outgrowth and Regulates Expression of Genes Associated With Drug Resistance and Interferon Responsiveness. Cancer Genom. Proteom. 2019, 16, 491–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Borretzen, A.; Gravdal, K.; Haukaas, S.A.; Beisland, C.; Akslen, L.A.; Halvorsen, O.J. FOXC2 expression and epithelial-mesenchymal phenotypes are associated with castration resistance, metastasis and survival in prostate cancer. J. Pathol. Clin. Res. 2019, 5, 272–286. [Google Scholar] [CrossRef] [PubMed]
  295. Sarkar, P.L.; Lee, W.; Williams, E.D.; Lubik, A.A.; Stylianou, N.; Shokoohmand, A.; Lehman, M.L.; Hollier, B.G.; Gunter, J.H.; Nelson, C.C. Insulin Enhances Migration and Invasion in Prostate Cancer Cells by Up-Regulation of FOXC2. Front. Endocrinol. 2019, 10, 481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Pham, T.N.D.; Perez White, B.E.; Zhao, H.; Mortazavi, F.; Tonetti, D.A. Protein kinase C alpha enhances migration of breast cancer cells through FOXC2-mediated repression of p120-catenin. BMC Cancer 2017, 17, 832. [Google Scholar] [CrossRef]
  297. Gao, F.; Wang, X.; Chen, S.; Xu, T.; Wang, X.; Shen, Y.; Dong, F.; Zhong, S.; Shen, Z. CIP2A depletion potentiates the chemosensitivity of cisplatin by inducing increased apoptosis in bladder cancer cells. Oncol. Rep. 2018, 40, 2445–2454. [Google Scholar] [CrossRef]
  298. Chanvorachote, P.; Chunhacha, P. Caveolin-1 regulates endothelial adhesion of lung cancer cells via reactive oxygen species-dependent mechanism. PLoS ONE 2013, 8, e57466. [Google Scholar] [CrossRef]
  299. Petpiroon, N.; Bhummaphan, N.; Tungsukruthai, S.; Pinkhien, T.; Maiuthed, A.; Sritularak, B.; Chanvorachote, P. Chrysotobibenzyl inhibition of lung cancer cell migration through Caveolin-1-dependent mediation of the integrin switch and the sensitization of lung cancer cells to cisplatin-mediated apoptosis. Phytomedicine 2019, 58, 152888. [Google Scholar] [CrossRef]
  300. Milone, M.R.; Lombardi, R.; Roca, M.S.; Bruzzese, F.; Addi, L.; Pucci, B.; Budillon, A. Novel pathways involved in cisplatin resistance identified by a proteomics approach in non-small-cell lung cancer cells. J. Cell. Physiol. 2019, 234, 9077–9092. [Google Scholar] [CrossRef]
  301. Sato, N.; Kumasawa, K.; Yamashita, M.; Miyake, T.; Nakamura, H.; Kimura, T. Therapeutic potential of combination therapy of soluble VEGF receptor 1 and conventional chemotherapy for ovarian cancer growth. J. Obstet. Gynaecol. Res. 2020. [Google Scholar] [CrossRef]
  302. Alghzzawy, Z.M.; Elmaghraby, T.K.; El-Hamid Hagag, S.A.; Awwad, M.H. Combretastatin A-4 disodium phosphate and low dose gamma irradiation suppress hepatocellular carcinoma by downregulating ROCK1 and VEGF gene expression. Mol. Biol. Rep. 2020. [Google Scholar] [CrossRef] [PubMed]
  303. Bailly, C. Regulation of PD-L1 expression on cancer cells with ROS-modulating drugs. Life Sci. 2020, 117403. [Google Scholar] [CrossRef] [PubMed]
  304. Gadducci, A.; Lanfredini, N.; Sergiampietri, C. Antiangiogenic agents in gynecological cancer: State of art and perspectives of clinical research. Crit. Rev. Oncol. Hematol. 2015, 96, 113–128. [Google Scholar] [CrossRef] [PubMed]
  305. Cereda, V.; Formica, V.; Roselli, M. Issues and promises of bevacizumab in prostate cancer treatment. Expert Opin. Biol. Ther. 2018, 18, 707–717. [Google Scholar] [CrossRef]
  306. Schmidt, E.V. Developing combination strategies using PD-1 checkpoint inhibitors to treat cancer. Semin. Immunopathol. 2019, 41, 21–30. [Google Scholar] [CrossRef] [Green Version]
  307. Balar, A.V.; Weber, J.S. PD-1 and PD-L1 antibodies in cancer: Current status and future directions. Cancer Immunol. Immunother. 2017, 66, 551–564. [Google Scholar] [CrossRef]
  308. Kastenhuber, E.R.; Lowe, S.W. Putting p53 in context. Cell 2017, 170, 1062–1078. [Google Scholar] [CrossRef] [Green Version]
  309. Wasylishen, A.R.; Lozano, G. Attenuating the p53 pathway in human cancers: Many means to the same end. Cold Spring Harb. Perspect. Med. 2016, 6, a026211. [Google Scholar] [CrossRef]
  310. Honda, R.; Tanaka, H.; Yasuda, H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett. 1997, 420, 25–27. [Google Scholar] [CrossRef] [Green Version]
  311. Kim, M.; Laramy, J.K.; Gampa, G.; Parrish, K.E.; Brundage, R.; Sarkaria, J.N.; Elmquist, W.F. Brain Distributional Kinetics of a Novel MDM2 Inhibitor SAR405838: Implications for Use in Brain Tumor Therapy. Drug Metab. Dispos. 2019, 47, 1403–1414. [Google Scholar] [CrossRef]
  312. Ning, Y.; Hui, N.; Qing, B.; Zhuo, Y.; Sun, W.; Du, Y.; Liu, S.; Liu, K.; Zhou, J. ZCCHC10 suppresses lung cancer progression and cisplatin resistance by attenuating MDM2-mediated p53 ubiquitination and degradation. Cell Death Dis. 2019, 10, 1–12. [Google Scholar] [CrossRef] [PubMed]
  313. Wang, G.-S. Medical uses of mylabris in ancient China and recent studies. J. Ethnopharmacol. 1989, 26, 147–162. [Google Scholar] [CrossRef]
  314. Shi, X.; Chen, S.; Zhang, Y.; Xie, W.; Hu, Z.; Li, H.; Li, J.; Zhou, Z.; Tan, W. Norcantharidin inhibits the DDR of bladder cancer stem-like cells through cdc6 degradation. OncoTargets Ther. 2019, 12, 4403–4413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Jin, D.; Wu, Y.; Shao, C.; Gao, Y.; Wang, D.; Guo, J. Norcantharidin reverses cisplatin resistance and inhibits the epithelial mesenchymal transition of human nonsmall lung cancer cells by regulating the YAP pathway. Oncol. Rep. 2018, 40, 609–620. [Google Scholar] [CrossRef]
  316. Yeo, C.; Han, D.S.; Lee, H.J.; Lee, E.O. Epigallocatechin-3-Gallate Suppresses Vasculogenic Mimicry through Inhibiting the Twist/VE-Cadherin/AKT Pathway in Human Prostate Cancer PC-3 Cells. Int. J. Mol. Sci. 2020, 21, 439. [Google Scholar] [CrossRef] [Green Version]
  317. Nagaishi, M.; Fujii, Y.; Sugiura, Y.; Takano, I.; Takigawa, T.; Yokoo, H.; Suzuki, K. Increased Twist and ZEB2 expression in a cutaneous metastasis of high-grade glioma. Neuropathology 2019. [Google Scholar] [CrossRef]
  318. Wang, L.; Tan, R.Z.; Zhang, Z.X.; Yin, R.; Zhang, Y.L.; Cui, W.J.; He, T. Association between Twist and multidrug resistance gene-associated proteins in Taxol((R))-resistant MCF-7 cells and a 293 cell model of Twist overexpression. Oncol. Lett. 2018, 15, 1058–1066. [Google Scholar] [CrossRef]
  319. Lai, Y.J.; Yu, W.N.; Kuo, S.C.; Ho, C.T.; Hung, C.M.; Way, T.D.; Chen, C.T. CSC-3436 inhibits TWIST-induced epithelial-mesenchymal transition via the suppression of Twist/Bmi1/Akt pathway in head and neck squamous cell carcinoma. J. Cell. Physiol. 2019, 234, 9118–9129. [Google Scholar] [CrossRef]
  320. Cheong, C.M.; Mrozik, K.M.; Hewett, D.R.; Bell, E.; Panagopoulos, V.; Noll, J.E.; Licht, J.D.; Gronthos, S.; Zannettino, A.C.W.; Vandyke, K. Twist-1 is upregulated by NSD2 and contributes to tumour dissemination and an epithelial-mesenchymal transition-like gene expression signature in t(4;14)-positive multiple myeloma. Cancer Lett. 2020. [Google Scholar] [CrossRef]
  321. Cho, Y.A.; Kim, E.K.; Cho, B.C.; Koh, Y.W.; Yoon, S.O. Twist and Snail/Slug Expression in Oropharyngeal Squamous Cell Carcinoma in Correlation With Lymph Node Metastasis. Anticancer Res. 2019, 39, 6307–6316. [Google Scholar] [CrossRef]
  322. Lin, C.R.; Chu, T.M.; Luo, A.; Huang, S.J.; Chou, H.Y.; Lu, M.W.; Wu, J.L. Omega-3 polyunsaturated fatty acids suppress metastatic features of human cholangiocarcinoma cells by suppressing twist. J. Nutr. Biochem. 2019, 74, 108245. [Google Scholar] [CrossRef] [PubMed]
  323. Shahin, S.A.; Wang, R.; Simargi, S.I.; Contreras, A.; Echavarria, L.P.; Qu, L.; Wen, W.; Dellinger, T.; Unternaehrer, J.; Tamanoi, F. Hyaluronic acid conjugated nanoparticle delivery of siRNA against TWIST reduces tumor burden and enhances sensitivity to cisplatin in ovarian cancer. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1381–1394. [Google Scholar] [CrossRef] [PubMed]
  324. Manirujjaman, M.; Ozaki, I.; Murata, Y.; Guo, J.; Xia, J.; Nishioka, K.; Perveen, R.; Takahashi, H.; Anzai, K.; Matsuhashi, S. Degradation of the Tumor Suppressor PDCD4 Is Impaired by the Suppression of p62/SQSTM1 and Autophagy. Cells 2020, 9, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. De, P.; Carlson, J.; Leyland-Jones, B.; Dey, N. Oncogenic nexus of cancerous inhibitor of protein phosphatase 2A (CIP2A): An oncoprotein with many hands. Oncotarget 2014, 5, 4581. [Google Scholar] [CrossRef] [Green Version]
  326. Junttila, M.R.; Puustinen, P.; Niemelä, M.; Ahola, R.; Arnold, H.; Böttzauw, T.; Ala-aho, R.; Nielsen, C.; Ivaska, J.; Taya, Y. CIP2A inhibits PP2A in human malignancies. Cell 2007, 130, 51–62. [Google Scholar] [CrossRef] [Green Version]
  327. Wu, Y.; Gu, T.-T.; Zheng, P.-S. CIP2A cooperates with H-Ras to promote epithelial–mesenchymal transition in cervical-cancer progression. Cancer Lett. 2015, 356, 646–655. [Google Scholar] [CrossRef]
  328. Xu, P.; Yao, J.; He, J.; Zhao, L.; Wang, X.; Li, Z.; Qian, J. CIP2A down regulation enhances the sensitivity of pancreatic cancer cells to gemcitabine. Oncotarget 2016, 7, 14831. [Google Scholar] [CrossRef] [Green Version]
  329. Liu, L.-Z.; Zhou, X.-D.; Qian, G.; Shi, X.; Fang, J.; Jiang, B.-H. AKT1 amplification regulates cisplatin resistance in human lung cancer cells through the mammalian target of rapamycin/p70S6K1 pathway. Cancer Res. 2007, 67, 6325–6332. [Google Scholar] [CrossRef] [Green Version]
  330. Tian, Y.; Gong, G.Y.; Ma, L.L.; Wang, Z.Q.; Song, D.; Fang, M.Y. Anti-cancer effects of Polyphyllin I: An update in 5 years. Chem. Biol. Interact. 2019, 316, 108936. [Google Scholar] [CrossRef]
  331. Cui, J.; Man, S.; Cui, N.; Yang, L.; Guo, Q.; Ma, L.; Gao, W. The synergistic anticancer effect of formosanin C and polyphyllin VII based on caspase-mediated cleavage of Beclin1 inhibiting autophagy and promoting apoptosis. Cell Prolif. 2019, 52, e12520. [Google Scholar] [CrossRef] [Green Version]
  332. Feng, F.; Cheng, P.; Wang, C.; Wang, Y.; Wang, W. Polyphyllin I and VII potentiate the chemosensitivity of A549/DDP cells to cisplatin by enhancing apoptosis, reversing EMT and suppressing the CIP2A/AKT/mTOR signaling axis. Oncol. Lett. 2019, 18, 5428–5436. [Google Scholar] [CrossRef] [PubMed]
  333. Liu, J.; Zhai, R.; Zhao, J.; Kong, F.; Wang, J.; Jiang, W.; Xin, Q.; Xue, X.; Luan, Y. Programmed cell death 4 overexpression enhances sensitivity to cisplatin via the JNK/c-Jun signaling pathway in bladder cancer. Int. J. Oncol. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Liu, X.; Chen, L.; Ge, J.; Yan, C.; Huang, Z.; Hu, J.; Wen, C.; Li, M.; Huang, D.; Qiu, Y. The ubiquitin-like protein FAT10 stabilizes eEF1A1 expression to promote tumor proliferation in a complex manner. Cancer Res. 2016, 76, 4897–4907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Shi, D.-M.; Bian, X.-Y.; Qin, C.-D.; Wu, W.-Z. miR-106b-5p promotes stem cell-like properties of hepatocellular carcinoma cells by targeting PTEN via PI3K/Akt pathway. OncoTargets Ther. 2018, 11, 571. [Google Scholar] [CrossRef] [Green Version]
  336. Talesa, V.N.; Ferri, I.; Bellezza, G.; Love, H.D.; Sidoni, A.; Antognelli, C. Glyoxalase 2 is involved in human prostate cancer progression as part of a mechanism driven by PTEN/PI3K/AKT/mTOR signaling with involvement of PKM2 and ERα. Prostate 2017, 77, 196–210. [Google Scholar] [CrossRef]
  337. Zhao, X.; Dai, L.; Yue, Q.; Wang, H.; Wang, X.U.; Li, Y.; Chen, R. MiR-195 inhibits migration, invasion and epithelial-mesenchymal transition (EMT) of endometrial carcinoma cells by targeting SOX4. J. Biosci. 2019, 44, 146. [Google Scholar] [CrossRef]
  338. Liu, L.; Jung, S.N.; Oh, C.; Lee, K.; Won, H.R.; Chang, J.W.; Kim, J.M.; Koo, B.S. LAMB3 is associated with disease progression and cisplatin cytotoxic sensitivity in head and neck squamous cell carcinoma. Eur. J. Surg. Oncol. 2019, 45, 359–365. [Google Scholar] [CrossRef]
  339. Zhang, P.; Lu, X.; Shi, Z.; Li, X.; Zhang, Y.; Zhao, S.; Liu, H. miR-205-5p regulates epithelial-mesenchymal transition by targeting PTEN via PI3K/AKT signaling pathway in cisplatin-resistant nasopharyngeal carcinoma cells. Gene 2019, 710, 103–113. [Google Scholar] [CrossRef]
  340. Jung, S.N.; Lim, H.S.; Liu, L.; Chang, J.W.; Lim, Y.C.; Rha, K.S.; Koo, B.S. LAMB3 mediates metastatic tumor behavior in papillary thyroid cancer by regulating c-MET/Akt signals. Sci. Rep. 2018, 8, 2718. [Google Scholar] [CrossRef] [Green Version]
  341. Wang, Y.; Jin, Y.; Bhandari, A.; Yao, Z.; Yang, F.; Pan, Y.; Zheng, Z.; Lv, S.; Wang, O. Upregulated LAMB3 increases proliferation and metastasis in thyroid cancer. OncoTargets Ther. 2018, 11, 37–46. [Google Scholar] [CrossRef] [Green Version]
  342. Shi, D.; Li, Y.; Fan, L.; Zhao, Q.; Tan, B.; Cui, G. Upregulation Of miR-153 Inhibits Triple-Negative Breast Cancer Progression By Targeting ZEB2-Mediated EMT And Contributes To Better Prognosis. OncoTargets Ther. 2019, 12, 9611–9625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Sarkar, A.; Rahaman, A.; Biswas, I.; Mukherjee, G.; Chatterjee, S.; Bhattacharjee, S.; Mandal, D.P. TGFbeta mediated LINC00273 upregulation sponges mir200a-3p and promotes invasion and metastasis by activating ZEB1. J. Cell. Physiol. 2020. [Google Scholar] [CrossRef] [PubMed]
  344. Duan, Y.; Zhang, X.; Yang, L.; Dong, X.; Zheng, Z.; Cheng, Y.; Chen, H.; Lan, B.; Li, D.; Zhou, J. Disruptor of telomeric silencing 1-like (DOT1L) is involved in breast cancer metastasis via transcriptional regulation of MALAT1 and ZEB2. J. Genet. Genom. Yi Chuan Xue Bao 2019, 46, 591–594. [Google Scholar] [CrossRef] [PubMed]
  345. Yao, X.; Sun, S.; Zhou, X.; Zhang, Q.; Guo, W.; Zhang, L. Clinicopathological significance of ZEB-1 and E-cadherin proteins in patients with oral cavity squamous cell carcinoma. OncoTargets Ther. 2017, 10, 781. [Google Scholar] [CrossRef] [Green Version]
  346. Cleophas, M.; Joosten, L.; Stamp, L.K.; Dalbeth, N.; Woodward, O.M.; Merriman, T.R. ABCG2 polymorphisms in gout: Insights into disease susceptibility and treatment approaches. Pharm. Pers. Med. 2017, 10, 129. [Google Scholar] [CrossRef] [Green Version]
  347. Hamilton, G.; Rath, B. Pharmacogenetics of platinum-based chemotherapy in non-small cell lung cancer: Predictive validity of polymorphisms of ERCC1. Expert Opin. Drug Metab. Toxicol. 2018, 14, 17–24. [Google Scholar] [CrossRef]
  348. Wu, Y.; Jin, D.; Wang, X.; Du, J.; Di, W.; An, J.; Shao, C.; Guo, J. UBE2C induces cisplatin resistance via ZEB1/2-dependent upregulation of ABCG2 and ERCC1 in NSCLC cells. J. Oncol. 2019, 2019, 8607859. [Google Scholar] [CrossRef] [Green Version]
  349. Guo, T.; Yuan, X.; Liu, D.F.; Peng, S.H.; Xu, A.M. LncRNA HOXA11-AS promotes migration and invasion through modulating miR-148a/WNT1/beta-catenin pathway in gastric cancer. Neoplasma 2020. [Google Scholar] [CrossRef]
  350. Bai, Y.; Lang, L.; Zhao, W.; Niu, R. Long Non-Coding RNA HOXA11-AS Promotes Non-Small Cell Lung Cancer Tumorigenesis Through microRNA-148a-3p/DNMT1 Regulatory Axis. OncoTargets Ther. 2019, 12, 11195–11206. [Google Scholar] [CrossRef] [Green Version]
  351. Zhang, Y.; Zhang, Q.; Chen, H.; Wang, C. BCL9 promotes epithelial mesenchymal transition and invasion in cisplatin resistant NSCLC cells via β-catenin pathway. Life Sci. 2018, 208, 284–294. [Google Scholar] [CrossRef]
  352. Chen, N.; Kong, Y.; Wu, Y.; Gao, Q.; Fu, J.; Sun, X.; Geng, Q. CAC1 knockdown reverses drug resistance through the downregulation of P-gp and MRP-1 expression in colorectal cancer. PLoS ONE 2019, 14, e0222035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. Liu, H.Y.; Duan, G.L.; Xu, R.Y.; Li, X.R.; Xiao, L.; Zhao, L.; Ma, Z.X.; Xu, X.W.; Qiu, L.J.; Zhu, Z.M.; et al. DJ-1 overexpression confers the multidrug resistance phenotype to SGC7901cells by upregulating P-gp and Bcl-2. Biochem. Biophys. Res. Commun. 2019, 519, 73–80. [Google Scholar] [CrossRef] [PubMed]
  354. Sachs, J.; Dohl, K.; Weber, A.; Bonus, M.; Ehlers, F.; Fleischer, E.; Klinger, A.; Gohlke, H.; Pietruszka, J.; Schmitt, L.; et al. Novel 3,4-Dihydroisocoumarins Inhibit Human P-gp and BCRP in Multidrug Resistant Tumors and Demonstrate Substrate Inhibition of Yeast Pdr5. Front. Pharmacol. 2019, 10, 400. [Google Scholar] [CrossRef] [PubMed]
  355. Gomes, B.C.; Honrado, M.; Armada, A.; Viveiros, M.; Rueff, J.; Rodrigues, A.S. ABC Efflux Transporters and the Circuitry of miRNAs: Kinetics of Expression in Cancer Drug Resistance. Int. J. Mol. Sci. 2020, 21, 2985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  356. Leopoldo, M.; Nardulli, P.; Contino, M.; Leonetti, F.; Luurtsema, G.; Colabufo, N.A. An updated patent review on P-glycoprotein inhibitors (2011–2018). Expert Opin. Ther. Pat. 2019, 29, 455–461. [Google Scholar] [CrossRef]
  357. Ayrton, A.; Morgan, P. Role of transport proteins in drug absorption, distribution and excretion. Xenobiotica 2001, 31, 469–497. [Google Scholar] [CrossRef]
  358. Dean, M.; Hamon, Y.; Chimini, G. The human ATP-binding cassette (ABC) transporter superfamily. J. Lipid Res. 2001, 42, 1007–1017. [Google Scholar] [CrossRef]
  359. Giacomini, K.M.; Huang, S.-M.; Tweedie, D.J.; Benet, L.Z.; Brouwer, K.L.; Chu, X.; Dahlin, A.; Evers, R.; Fischer, V.; Hillgren, K.M. Membrane transporters in drug development. Nat. Rev. Drug Discov. 2010, 9, 215. [Google Scholar]
  360. Lin, H.; Hu, B.; He, X.; Mao, J.; Wang, Y.; Wang, J.; Zhang, T.; Zheng, J.; Peng, Y.; Zhang, F. Overcoming Taxol-resistance in A549 cells: A comprehensive strategy of targeting P-gp transporter, AKT/ERK pathways, and cytochrome P450 enzyme CYP1B1 by 4-hydroxyemodin. Biochem. Pharmacol. 2020, 171, 113733. [Google Scholar] [CrossRef]
  361. Zhang, E.; Liu, J.; Shi, L.; Guo, X.; Liang, Z.; Zuo, J.; Xu, H.; Wang, H.; Shu, X.; Huang, S.; et al. 7-O-geranylquercetin contributes to reverse P-gp-mediated adriamycin resistance in breast cancer. Life Sci. 2019, 238, 116938. [Google Scholar] [CrossRef]
  362. Wang, J.; Chen, Y.; Xiang, F.; Li, M.; Li, H.; Chi, J.; Ren, K. Suppression of TGF-beta1 enhances chemosensitivity of cisplatin-resistant lung cancer cells through the inhibition of drug-resistant proteins. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1505–1512. [Google Scholar] [CrossRef] [PubMed]
  363. Choi, H.S.; Kim, Y.-K.; Yun, P.-Y. Upregulation of MDR-and EMT-Related Molecules in Cisplatin-Resistant Human Oral Squamous Cell Carcinoma Cell Lines. Int. J. Mol. Sci. 2019, 20, 3034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  364. Enerbäck, C.; Porter, D.A.; Seth, P.; Sgroi, D.; Gaudet, J.; Weremowicz, S.; Morton, C.C.; Schnitt, S.; Pitts, R.L.; Stampl, J. Psoriasin expression in mammary epithelial cells in vitro and in vivo. Cancer Res. 2002, 62, 43–47. [Google Scholar] [PubMed]
  365. Jinquan, T.; Vorum, H.; Larsen, C.G.; Madsen, P.; Rasmussen, H.H.; Gesser, B.; Etzerodt, M.; Honoré, B.; Celis, J.E.; Thestrup-Pedersen, K. Psoriasin: A novel chemotactic protein. J. Investig. Dermatol. 1996, 107, 5–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  366. Madsen, P.; Rasmussen, H.H.; Leffers, H.; Honoré, B.; Dejgaard, K.; Olsen, E.; Kiil, J.; Walbum, E.; Andersen, A.H.; Basse, B. Molecular cloning, occurrence, and expression of a novel partially secreted protein “psoriasin” that is highly up-regulated in psoriatic skin. J. Investig. Dermatol. 1991, 97, 701–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  367. Watson, P.H.; Leygue, E.R.; Murphy, L.C. Psoriasin (S100A7). Int. J. Biochem. Cell Biol. 1998, 30, 567–571. [Google Scholar] [CrossRef]
  368. Liu, Y.; Bunston, C.; Hodson, N.; Resaul, J.; Sun, P.-H.; Cai, S.; Chen, G.; Gu, Y.; Satherley, L.K.; Bosanquet, D.C. Psoriasin promotes invasion, aggregation and survival of pancreatic cancer cells; association with disease progression. Int. J. Oncol. 2017, 50, 1491–1500. [Google Scholar] [CrossRef] [Green Version]
  369. Li, W.; Yu, Z.; Ma, B. The increase of miR-27a affects the role of cisplatin on proliferation and migration capacities of liver cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5490–5498. [Google Scholar]
  370. Ren, J.; Kan, A.; Leong, S.H.; Ooi, L.L.; Jeang, K.-T.; Chong, S.S.; Kon, O.L.; Lee, C.G. FAT10 plays a role in the regulation of chromosomal stability. J. Biol. Chem. 2006, 281, 11413–11421. [Google Scholar] [CrossRef] [Green Version]
  371. Guan, X.-Y.; Sham, J.S.; Tang, T.C.; Fang, Y.; Huo, K.-K.; Yang, J.-M. Isolation of a novel candidate oncogene within a frequently amplified region at 3q26 in ovarian cancer. Cancer Res. 2001, 61, 3806–3809. [Google Scholar]
  372. Guan, X.-Y.; Fung, J.M.; Ma, N.-F.; Lau, S.-H.; Tai, L.-S.; Xie, D.; Zhang, Y.; Hu, L.; Wu, Q.-L.; Fang, Y. Oncogenic role of eIF-5A2 in the development of ovarian cancer. Cancer Res. 2004, 64, 4197–4200. [Google Scholar] [CrossRef] [Green Version]
  373. Yang, G.-F.; Xie, D.; Liu, J.-H.; Luo, J.-H.; Li, L.-J.; Hua, W.-F.; Wu, H.-M.; Kung, H.-F.; Zeng, Y.-X.; Guan, X.-Y. Expression and amplification of eIF-5A2 in human epithelial ovarian tumors and overexpression of EIF-5A2 is a new independent predictor of outcome in patients with ovarian carcinoma. Gynecol. Oncol. 2009, 112, 314–318. [Google Scholar] [CrossRef] [PubMed]
  374. Tang, D.J.; Dong, S.S.; Ma, N.F.; Xie, D.; Chen, L.; Fu, L.; Lau, S.H.; Li, Y.; Li, Y.; Guan, X.Y. Overexpression of eukaryotic initiation factor 5A2 enhances cell motility and promotes tumor metastasis in hepatocellular carcinoma. Hepatology 2010, 51, 1255–1263. [Google Scholar] [CrossRef] [PubMed]
  375. Xie, D.; Ma, N.-F.; Pan, Z.-Z.; Wu, H.-X.; Liu, Y.-D.; Wu, G.-Q.; Kung, H.-F.; Guan, X.-Y. Overexpression of EIF-5A2 is associated with metastasis of human colorectal carcinoma. Hum. Pathol. 2008, 39, 80–86. [Google Scholar] [CrossRef]
  376. Huang, P.-Y.; Zeng, T.-T.; Ban, X.; Li, M.-Q.; Zhang, B.-Z.; Zhu, Y.-H.; Hua, W.-F.; Mai, H.-Q.; Zhang, L.; Guan, X.-Y. Expression of EIF5A2 associates with poor survival of nasopharyngeal carcinoma patients treated with induction chemotherapy. BMC Cancer 2016, 16, 669. [Google Scholar] [CrossRef] [Green Version]
  377. Sun, J.; Xu, Z.; Lv, H.; Wang, Y.; Wang, L.; Ni, Y.; Wang, X.; Hu, C.; Chen, S.; Teng, F.; et al. eIF5A2 regulates the resistance of gastric cancer cells to cisplatin via induction of EMT. Am. J. Transl. Res. 2018, 10, 4269–4279. [Google Scholar]
  378. Amerik, A.Y.; Hochstrasser, M. Mechanism and function of deubiquitinating enzymes. Biochim. Biophys. Acta BBA Mol. Cell Res. 2004, 1695, 189–207. [Google Scholar] [CrossRef] [Green Version]
  379. Alhosin, M.; Omran, Z.; Zamzami, M.A.; Al-Malki, A.L.; Choudhry, H.; Mousli, M.; Bronner, C. Signalling pathways in UHRF1-dependent regulation of tumor suppressor genes in cancer. J. Exp. Clin. Cancer Res. 2016, 35, 174. [Google Scholar] [CrossRef] [Green Version]
  380. He, M.; Zhou, Z.; Wu, G.; Chen, Q.; Wan, Y. Emerging role of DUBs in tumor metastasis and apoptosis: Therapeutic implication. Pharmacol. Ther. 2017, 177, 96–107. [Google Scholar] [CrossRef] [Green Version]
  381. Zhang, Q.; Wang, X.; Chen, S.; Qin, X. Predictive value of deubiquitination enzymes USP37 in the prognosis of breast cancer. Zhonghua Yi Xue Za Zhi 2016, 96, 944–948. [Google Scholar]
  382. Tanno, H.; Shigematsu, T.; Nishikawa, S.; Hayakawa, A.; Denda, K.; Tanaka, T.; Komada, M. Ubiquitin-interacting motifs confer full catalytic activity, but not ubiquitin chain substrate specificity, to deubiquitinating enzyme USP37. J. Biol. Chem. 2014, 289, 2415–2423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  383. Huang, X.; Summers, M.K.; Pham, V.; Lill, J.R.; Liu, J.; Lee, G.; Kirkpatrick, D.S.; Jackson, P.K.; Fang, G.; Dixit, V.M. Deubiquitinase USP37 is activated by CDK2 to antagonize APCCDH1 and promote S phase entry. Mol. Cell 2011, 42, 511–523. [Google Scholar] [CrossRef] [PubMed]
  384. Burrows, A.C.; Prokop, J.; Summers, M.K. Skp1-Cul1-F-box ubiquitin ligase (SCFβTrCP)-mediated destruction of the ubiquitin-specific protease USP37 during G2-phase promotes mitotic entry. J. Biol. Chem. 2012, 287, 39021–39029. [Google Scholar] [CrossRef] [Green Version]
  385. Cai, J.; Li, M.; Wang, X.; Li, L.; Li, Q.; Hou, Z.; Jia, H.; Liu, S. USP37 promotes lung cancer cell migration by stabilizing Snail protein via deubiquitination. Front. Genet. 2020, 10, 1324. [Google Scholar] [CrossRef] [PubMed]
  386. Kasper, M.; Jaks, V.; Fiaschi, M.; Toftgård, R. Hedgehog signalling in breast cancer. Carcinogenesis 2009, 30, 903–911. [Google Scholar] [CrossRef] [Green Version]
  387. Qin, T.; Li, B.; Feng, X.; Fan, S.; Liu, L.; Liu, D.; Mao, J.; Lu, Y.; Yang, J.; Yu, X. Abnormally elevated USP37 expression in breast cancer stem cells regulates stemness, epithelial-mesenchymal transition and cisplatin sensitivity. J. Exp. Clin. Cancer Res. 2018, 37, 287. [Google Scholar] [CrossRef] [Green Version]
  388. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.; Choi, H.; El Rayes, T.; Ryu, S.; Troeger, J. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef]
  389. Yu, Y.-C.; Yang, P.-M.; Chuah, Q.-Y.; Huang, Y.-H.; Peng, C.-W.; Lee, Y.-J.; Chiu, S.-J. Radiation-induced senescence in securin-deficient cancer cells promotes cell invasion involving the IL-6/STAT3 and PDGF-BB/PDGFR pathways. Sci. Rep. 2013, 3, 1675. [Google Scholar] [CrossRef] [Green Version]
  390. Wong, A.L.A.; Hirpara, J.L.; Pervaiz, S.; Eu, J.Q.; Sethi, G.; Goh, B.C. Do STAT3 inhibitors have potential in the future for cancer therapy? Expert Opin. Investig. Drugs 2017, 26, 883–887. [Google Scholar] [CrossRef] [Green Version]
  391. Tan, S.M.; Li, F.; Rajendran, P.; Kumar, A.P.; Hui, K.M.; Sethi, G. Identification of beta-escin as a novel inhibitor of signal transducer and activator of transcription 3/Janus-activated kinase 2 signaling pathway that suppresses proliferation and induces apoptosis in human hepatocellular carcinoma cells. J. Pharmacol. Exp. Ther. 2010, 334, 285–293. [Google Scholar] [CrossRef]
  392. Mohan, C.D.; Bharathkumar, H.; Bulusu, K.C.; Pandey, V.; Rangappa, S.; Fuchs, J.E.; Shanmugam, M.K.; Dai, X.; Li, F.; Deivasigamani, A.; et al. Development of a novel azaspirane that targets the Janus kinase-signal transducer and activator of transcription (STAT) pathway in hepatocellular carcinoma in vitro and in vivo. J. Biol. Chem. 2014, 289, 34296–34307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  393. Rajendran, P.; Li, F.; Shanmugam, M.K.; Vali, S.; Abbasi, T.; Kapoor, S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J. Cell. Physiol. 2012, 227, 2184–2195. [Google Scholar] [CrossRef] [PubMed]
  394. Kim, S.; Koh, J.; Kim, M.-Y.; Kwon, D.; Go, H.; Kim, Y.A.; Jeon, Y.K.; Chung, D.H. PD-L1 expression is associated with epithelial-to-mesenchymal transition in adenocarcinoma of the lung. Hum. Pathol. 2016, 58, 7–14. [Google Scholar] [CrossRef] [PubMed]
  395. Wang, Y.; Wang, H.; Zhao, Q.; Xia, Y.; Hu, X.; Guo, J. PD-L1 induces epithelial-to-mesenchymal transition via activating SREBP-1c in renal cell carcinoma. Med. Oncol. 2015, 32, 212. [Google Scholar] [CrossRef]
  396. Shen, M.; Xu, Z.; Xu, W.; Jiang, K.; Zhang, F.; Ding, Q.; Xu, Z.; Chen, Y. Inhibition of ATM reverses EMT and decreases metastatic potential of cisplatin-resistant lung cancer cells through JAK/STAT3/PD-L1 pathway. J. Exp. Clin. Cancer Res. 2019, 38, 149. [Google Scholar] [CrossRef] [Green Version]
  397. Romacho, T.; Sánchez-Ferrer, C.F.; Peiró, C. Visfatin/Nampt: An adipokine with cardiovascular impact. Mediat. Inflamm. 2013, 2013, 946427. [Google Scholar] [CrossRef]
  398. Carbone, F.; Liberale, L.; Bonaventura, A.; Vecchiè, A.; Casula, M.; Cea, M.; Monacelli, F.; Caffa, I.; Bruzzone, S.; Montecucco, F. Regulation and function of extracellular nicotinamide phosphoribosyltransferase/visfatin. Compr. Physiol. 2011, 7, 603–621. [Google Scholar]
  399. Mohammadi, M.; Moradi, A.; Farhadi, J.; Akbari, A.; Pourmandi, S.; Mehrad-Majd, H. Prognostic value of visfatin in various human malignancies: A systematic review and meta-analysis. Cytokine 2020, 127, 154964. [Google Scholar] [CrossRef]
  400. Mohammadi, M.; Mianabadi, F.; Mehrad-Majd, H. Circulating visfatin levels and cancers risk: A systematic review and meta-analysis. J. Cell. Physiol. 2019, 234, 5011–5022. [Google Scholar] [CrossRef]
  401. Wang, D.; Qian, G.; Wang, J.; Wang, T.; Zhang, L.; Yang, P.; Lin, F. Visfatin is involved in the cisplatin resistance of osteosarcoma cells via upregulation of Snail and Zeb1. Cancer Boil. Ther. 2019, 20, 999–1006. [Google Scholar] [CrossRef]
  402. Wang, J.; Han, Y.; Wang, M.; Zhao, Q.; Chen, X.; Liu, X. Natural triterpenoid saponin Momordin Ic suppresses HepG2 cell invasion via COX-2 inhibition and PPARgamma activation. Toxicol. Vitro 2020, 65, 104784. [Google Scholar] [CrossRef] [PubMed]
  403. Dai, M.; Hu, S.; Liu, C.F.; Jiang, L.; Yu, W.; Li, Z.L.; Guo, W.; Tang, R.; Dong, C.Y.; Wu, T.H.; et al. BPTF cooperates with p50 NF-kappaB to promote COX-2 expression and tumor cell growth in lung cancer. Am. J. Transl. Res. 2019, 11, 7398–7409. [Google Scholar] [PubMed]
  404. Jiang, G.B.; Fang, H.Y.; Tao, D.Y.; Chen, X.P.; Cao, F.L. COX-2 potentiates cisplatin resistance of non-small cell lung cancer cells by promoting EMT in an AKT signaling pathway-dependent manner. Eur. Rev. Med Pharmacol. Sci. 2019, 23, 3838–3846. [Google Scholar] [CrossRef] [PubMed]
  405. Patel, M.S.; Nemeria, N.S.; Furey, W.; Jordan, F. The pyruvate dehydrogenase complexes: Structure-based function and regulation. J. Boil. Chem. 2014, 289, 16615–16623. [Google Scholar] [CrossRef] [Green Version]
  406. Zhang, W.; Su, J.; Xu, H.; Yu, S.; Liu, Y.; Zhang, Y.; Sun, L.; Yue, Y.; Zhou, X. Dicumarol inhibits PDK1 and targets multiple malignant behaviors of ovarian cancer cells. PLoS ONE 2017, 12, e0179672. [Google Scholar] [CrossRef] [Green Version]
  407. Liu, T.; Yin, H. PDK1 promotes tumor cell proliferation and migration by enhancing the Warburg effect in non-small cell lung cancer. Oncol. Rep. 2017, 37, 193–200. [Google Scholar] [CrossRef] [Green Version]
  408. Zhang, S.-L.; Hu, X.; Zhang, W.; Yao, H.; Tam, K.Y. Development of pyruvate dehydrogenase kinase inhibitors in medicinal chemistry with particular emphasis as anticancer agents. Drug Discov. Today 2015, 20, 1112–1119. [Google Scholar] [CrossRef]
  409. Sugiyama, E.; Togashi, Y.; Takeuchi, Y.; Shinya, S.; Tada, Y.; Kataoka, K.; Tane, K.; Sato, E.; Ishii, G.; Goto, K.; et al. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR-mutated non-small cell lung cancer. Sci. Immunol. 2020, 5. [Google Scholar] [CrossRef]
  410. Huang, L.; Jiang, X.; Kang, P.; Wang, Z.; Leng, K.; Ji, D.; Xu, Y.; Wang, H.; Cui, Y. Long non-coding RNA NNT-AS1 functions as an oncogenic gene through modulating miR-485/BCL9 in cholangiocarcinoma. Cancer Manag. Res. 2019, 11, 7739–7749. [Google Scholar] [CrossRef] [Green Version]
  411. Cao, Y.; Shen, T.; Zhang, C.; Zhang, Q.H.; Zhang, Z.Q. MiR-125a-5p inhibits EMT of ovarian cancer cells by regulating TAZ/EGFR signaling pathway. Eur. Rev. Med Pharmacol. Sci. 2019, 23, 8249–8256. [Google Scholar] [CrossRef]
  412. Xing, Y.; Jing, H.; Zhang, Y.; Suo, J.; Qian, M. MicroRNA-141-3p affected proliferation, chemosensitivity, migration and invasion of colorectal cancer cells by targeting EGFR. Int. J. Biochem. Cell Boil. 2020, 118, 105643. [Google Scholar] [CrossRef] [PubMed]
  413. Chiu, Y.-J.; Hour, M.-J.; Jin, Y.-A.; Lu, C.-C.; Tsai, F.-J.; Chen, T.-L.; Ma, H.; Juan, Y.-N.; Yang, J.-S. Disruption of IGF-1R signaling by a novel quinazoline derivative, HMJ-30, inhibits invasiveness and reverses epithelial-mesenchymal transition in osteosarcoma U-2 OS cells. Int. J. Oncol. 2018, 52, 1465–1478. [Google Scholar] [CrossRef] [PubMed]
  414. Ernst, B.P.; Mikstas, C.; Stoever, T.; Stauber, R.; Strieth, S. Association of eIF4E and SPARC expression with lymphangiogenesis and lymph node metastasis in hypopharyngeal cancer. Anticancer Res. 2018, 38, 699–706. [Google Scholar] [PubMed]
  415. Liang, S.; Guo, R.; Zhang, Z.; Liu, D.; Xu, H.; Xu, Z.; Wang, X.; Yang, L. Upregulation of the eIF4E signaling pathway contributes to the progression of gastric cancer, and targeting eIF4E by perifosine inhibits cell growth. Oncol. Rep. 2013, 29, 2422–2430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  416. Oblinger, J.L.; Burns, S.S.; Huang, J.; Pan, L.; Ren, Y.; Shen, R.; Kinghorn, A.D.; Welling, D.B.; Chang, L.-S. Overexpression of eIF4F components in meningiomas and suppression of meningioma cell growth by inhibiting translation initiation. Exp. Neurol. 2018, 299, 299–307. [Google Scholar] [CrossRef] [Green Version]
  417. Wu, M.; Liu, Y.; Di, X.; Kang, H.; Zeng, H.; Zhao, Y.; Cai, K.; Pang, T.; Wang, S.; Yao, Y. EIF4E over-expresses and enhances cell proliferation and cell cycle progression in nasopharyngeal carcinoma. Med. Oncol. 2013, 30, 400. [Google Scholar] [CrossRef]
  418. Zhao, Y.; Pang, T.Y.; Wang, Y.; Wang, S.; Kang, H.X.; Ding, W.B.; Yong, W.W.; Bie, Y.H.; Cheng, X.G.; Zeng, C. LMP 1 stimulates the transcription of e IF 4E to promote the proliferation, migration and invasion of human nasopharyngeal carcinoma. FEBS J. 2014, 281, 3004–3018. [Google Scholar] [CrossRef]
  419. Yao, Y.; Pang, T.; Cheng, Y.; Yong, W.; Kang, H.; Zhao, Y.; Wang, S.; Hu, X. Positive Correlative over-Expression between eIF4E and Snail in Nasopharyngeal Carcinoma Promotes its Metastasis and Resistance to Cisplatin. Pathol. Oncol. Res. 2019, 1–11. [Google Scholar] [CrossRef]
  420. Giaimo, B.D.; Ferrante, F.; Herchenröther, A.; Hake, S.B.; Borggrefe, T. The histone variant H2A.Z in gene regulation. Epigenet. Chromatin 2019, 12, 37. [Google Scholar] [CrossRef] [Green Version]
  421. Rispal, J.; Baron, L.; Beaulieu, J.F.; Chevillard-Briet, M.; Trouche, D.; Escaffit, F. The H2A.Z histone variant integrates Wnt signaling in intestinal epithelial homeostasis. Nat. Commun. 2019, 10, 1827. [Google Scholar] [CrossRef] [Green Version]
  422. Tyagi, M.; Cheema, M.S.; Dryhurst, D.; Eskiw, C.H.; Ausio, J. Metformin alters H2A.Z dynamics and regulates androgen dependent prostate cancer progression. Oncotarget 2018, 9, 37054–37068. [Google Scholar] [CrossRef] [PubMed]
  423. Ito, S.; Kayukawa, N.; Ueda, T.; Taniguchi, H.; Morioka, Y.; Hongo, F.; Ukimura, O. MRGBP promotes AR-mediated transactivation of KLK3 and TMPRSS2 via acetylation of histone H2A.Z in prostate cancer cells. Biochim. Biophys. Acta Gene Regul. Mech. 2018. [Google Scholar] [CrossRef] [PubMed]
  424. Hsu, C.C.; Shi, J.; Yuan, C.; Zhao, D.; Jiang, S.; Lyu, J.; Wang, X.; Li, H.; Wen, H.; Li, W.; et al. Recognition of histone acetylation by the GAS41 YEATS domain promotes H2A.Z deposition in non-small cell lung cancer. Genes Dev. 2018, 32, 58–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  425. Yang, B.; Tong, R.; Liu, H.; Wu, J.; Chen, D.; Xue, Z.; Ding, C.; Zhou, L.; Xie, H.; Wu, J. H2A. Z regulates tumorigenesis, metastasis and sensitivity to cisplatin in intrahepatic cholangiocarcinoma. Int. J. Oncol. 2018, 52, 1235–1245. [Google Scholar] [PubMed]
  426. Wangpaichitr, M.; Kandemir, H.; Li, Y.Y.; Wu, C.; Nguyen, D.; Feun, L.G.; Kuo, M.T.; Savaraj, N. Relationship of Metabolic Alterations and PD-L1 Expression in Cisplatin Resistant Lung Cancer. Cell Dev. Biol. 2017, 6. [Google Scholar] [CrossRef]
  427. Xie, S.L.; Fan, S.; Zhang, S.Y.; Chen, W.X.; Li, Q.X.; Pan, G.K.; Zhang, H.Q.; Wang, W.W.; Weng, B.; Zhang, Z.; et al. SOX8 regulates cancer stem-like properties and cisplatin-induced EMT in tongue squamous cell carcinoma by acting on the Wnt/beta-catenin pathway. Int. J. Cancer 2018, 142, 1252–1265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  428. Wang, J.; Tian, L.; Khan, M.N.; Zhang, L.; Chen, Q.; Zhao, Y.; Yan, Q.; Fu, L.; Liu, J. Ginsenoside Rg3 sensitizes hypoxic lung cancer cells to cisplatin via blocking of NF-kappaB mediated epithelial-mesenchymal transition and stemness. Cancer Lett. 2018, 415, 73–85. [Google Scholar] [CrossRef]
  429. Hou, L.; Hou, X.; Wang, L.; Li, Z.; Xin, B.; Chen, J.; Gao, X.; Mu, H. PD98059 impairs the cisplatin-resistance of ovarian cancer cells by suppressing ERK pathway and epithelial mesenchymal transition process. Cancer Biomark. Sect. A Dis. Markers 2017, 21, 187–194. [Google Scholar] [CrossRef]
  430. Islam, S.S.; Al-Sharif, I.; Sultan, A.; Al-Mazrou, A.; Remmal, A.; Aboussekhra, A. Eugenol potentiates cisplatin anti-cancer activity through inhibition of ALDH-positive breast cancer stem cells and the NF-kappaB signaling pathway. Mol. Carcinog. 2018, 57, 333–346. [Google Scholar] [CrossRef]
  431. Mei, Y.; Peng, C.; Liu, Y.B.; Wang, J.; Zhou, H.H. Silencing RIF1 decreases cell growth, migration and increases cisplatin sensitivity of human cervical cancer cells. Oncotarget 2017, 8, 107044–107051. [Google Scholar] [CrossRef] [Green Version]
  432. Luo, J.; Yao, J.F.; Deng, X.F.; Zheng, X.D.; Jia, M.; Wang, Y.Q.; Huang, Y.; Zhu, J.H. 14, 15-EET induces breast cancer cell EMT and cisplatin resistance by up-regulating integrin alphavbeta3 and activating FAK/PI3K/AKT signaling. J. Exp. Clin. Cancer Res. 2018, 37, 23. [Google Scholar] [CrossRef] [PubMed]
  433. Xu, Y.; Miao, C.; Jin, C.; Qiu, C.; Li, Y.; Sun, X.; Gao, M.; Lu, N.; Kong, B. SUSD2 promotes cancer metastasis and confers cisplatin resistance in high grade serous ovarian cancer. Exp. Cell Res. 2018, 363, 160–170. [Google Scholar] [CrossRef]
  434. Kim, S.K.; Park, J.A.; Zhang, D.; Cho, S.H.; Yi, H.; Cho, S.M.; Chang, B.J.; Kim, J.S.; Shim, J.H.; Abd El-Aty, A.M.; et al. Sustainability of CD24 expression, cell proliferation and migration, cisplatin-resistance, and caspase-3 expression during mesenchymal-epithelial transition induced by the removal of TGF-beta1 in A549 lung cancer cells. Oncol. Lett. 2017, 14, 2410–2416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  435. Liu, Y.; Han, S.; Li, Y.; Liu, Y.; Zhang, D.; Li, Y.; Zhang, J. MicroRNA-20a contributes to cisplatin-resistance and migration of OVCAR3 ovarian cancer cell line. Oncol. Lett. 2017, 14, 1780–1786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  436. Wu, D.M.; Liu, T.; Deng, S.H.; Han, R.; Xu, Y. SLC39A4 expression is associated with enhanced cell migration, cisplatin resistance, and poor survival in non-small cell lung cancer. Sci. Rep. 2017, 7, 7211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  437. Li, S.; Zhang, X.; Zhang, R.; Liang, Z.; Liao, W.; Du, Z.; Gao, C.; Liu, F.; Fan, Y.; Hong, H. Hippo pathway contributes to cisplatin resistant-induced EMT in nasopharyngeal carcinoma cells. Cell Cycle 2017, 16, 1601–1610. [Google Scholar] [CrossRef] [PubMed]
  438. Fortunato, A. The role of hERG1 ion channels in epithelial-mesenchymal transition and the capacity of riluzole to reduce cisplatin resistance in colorectal cancer cells. Cell. Oncol. 2017, 40, 367–378. [Google Scholar] [CrossRef]
  439. Liu, S.L.; Lin, H.X.; Lin, C.Y.; Sun, X.Q.; Ye, L.P.; Qiu, F.; Wen, W.; Hua, X.; Wu, X.Q.; Li, J.; et al. TIMELESS confers cisplatin resistance in nasopharyngeal carcinoma by activating the Wnt/beta-catenin signaling pathway and promoting the epithelial mesenchymal transition. Cancer Lett. 2017, 402, 117–130. [Google Scholar] [CrossRef]
  440. Han, X.; Zhen, S.; Ye, Z.; Lu, J.; Wang, L.; Li, P.; Li, J.; Zheng, X.; Li, H.; Chen, W.; et al. A Feedback Loop Between miR-30a/c-5p and DNMT1 Mediates Cisplatin Resistance in Ovarian Cancer Cells. Cell. Physiol. Biochem. 2017, 41, 973–986. [Google Scholar] [CrossRef]
  441. Xiong, Y.; Sun, F.; Dong, P.; Watari, H.; Yue, J.; Yu, M.F.; Lan, C.Y.; Wang, Y.; Ma, Z.B. iASPP induces EMT and cisplatin resistance in human cervical cancer through miR-20a-FBXL5/BTG3 signaling. J. Exp. Clin. Cancer Res. 2017, 36, 48. [Google Scholar] [CrossRef]
  442. Feng, S.; Yang, G.; Yang, H.; Liang, Z.; Zhang, R.; Fan, Y.; Zhang, G. NEDD4 is involved in acquisition of epithelial-mesenchymal transition in cisplatin-resistant nasopharyngeal carcinoma cells. Cell Cycle 2017, 16, 869–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  443. Wang, X.; Guo, H.; Yao, B.; Helms, J. miR-15b inhibits cancer-initiating cell phenotypes and chemoresistance of cisplatin by targeting TRIM14 in oral tongue squamous cell cancer. Oncol. Rep. 2017, 37, 2720–2726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  444. Yu, M.; Qi, B.; Xiaoxiang, W.; Xu, J.; Liu, X. Baicalein increases cisplatin sensitivity of A549 lung adenocarcinoma cells via PI3K/Akt/NF-kappaB pathway. Biomed. Pharmacother. 2017, 90, 677–685. [Google Scholar] [CrossRef] [PubMed]
  445. Lin, X.J.; He, C.L.; Sun, T.; Duan, X.J.; Sun, Y.; Xiong, S.J. hsa-miR-485-5p reverses epithelial to mesenchymal transition and promotes cisplatin-induced cell death by targeting PAK1 in oral tongue squamous cell carcinoma. Int. J. Mol. Med. 2017, 40, 83–89. [Google Scholar] [CrossRef] [Green Version]
  446. Ge, L.; Li, D.S.; Chen, F.; Feng, J.D.; Li, B.; Wang, T.J. TAZ overexpression is associated with epithelial-mesenchymal transition in cisplatin-resistant gastric cancer cells. Int. J. Oncol. 2017, 51, 307–315. [Google Scholar] [CrossRef] [Green Version]
  447. Li, M.; Zheng, C.; Xu, H.; He, W.; Ruan, Y.; Ma, J.; Zheng, J.; Ye, C.; Li, W. Inhibition of AMPK-related kinase 5 (ARK5) enhances cisplatin cytotoxicity in non-small cell lung cancer cells through regulation of epithelial-mesenchymal transition. Am. J. Transl. Res. 2017, 9, 1708–1719. [Google Scholar]
  448. Kuang, P.; Chen, Z.; Wang, J.; Liu, Z.; Wang, J.; Gao, J.; Shen, L. Characterization of Aurora A and Its Impact on the Effect of Cisplatin-Based Chemotherapy in Patients with Non-Small Cell Lung Cancer. Transl. Oncol. 2017, 10, 367–377. [Google Scholar] [CrossRef]
  449. Bugide, S.; Gonugunta, V.K.; Penugurti, V.; Malisetty, V.L.; Vadlamudi, R.K.; Manavathi, B. HPIP promotes epithelial-mesenchymal transition and cisplatin resistance in ovarian cancer cells through PI3K/AKT pathway activation. Cell. Oncol. 2017, 40, 133–144. [Google Scholar] [CrossRef]
  450. Roberts, C.M.; Tran, M.A.; Pitruzzello, M.C.; Wen, W.; Loeza, J.; Dellinger, T.H.; Mor, G.; Glackin, C.A. TWIST1 drives cisplatin resistance and cell survival in an ovarian cancer model, via upregulation of GAS6, L1CAM, and Akt signalling. Sci. Rep. 2016, 6, 37652. [Google Scholar] [CrossRef] [Green Version]
  451. Ye, Z.; Yin, S.; Su, Z.; Bai, M.; Zhang, H.; Hei, Z.; Cai, S. Downregulation of miR-101 contributes to epithelial-mesenchymal transition in cisplatin resistance of NSCLC cells by targeting ROCK2. Oncotarget 2016, 7, 37524–37535. [Google Scholar] [CrossRef]
  452. Yang, L.; Zhang, F.; Wang, X.; Tsai, Y.; Chuang, K.H.; Keng, P.C.; Lee, S.O.; Chen, Y. A FASN-TGF-beta1-FASN regulatory loop contributes to high EMT/metastatic potential of cisplatin-resistant non-small cell lung cancer. Oncotarget 2016, 7, 55543. [Google Scholar] [CrossRef] [Green Version]
  453. Song, J.; Li, Y. miR-25-3p reverses epithelial-mesenchymal transition via targeting Sema4C in cisplatin-resistance cervical cancer cells. Cancer Sci. 2017, 108, 23–31. [Google Scholar] [CrossRef]
  454. Li, C.; Ding, H.; Tian, J.; Wu, L.; Wang, Y.; Xing, Y.; Chen, M. Forkhead Box Protein C2 Promotes Epithelial-Mesenchymal Transition, Migration and Invasion in Cisplatin-Resistant Human Ovarian Cancer Cell Line (SKOV3/CDDP). Cell. Physiol. Biochem. 2016, 39, 1098–1110. [Google Scholar] [CrossRef] [Green Version]
  455. Liu, S.; Yang, H.; Chen, Y.; He, B.; Chen, Q. Kruppel-Like Factor 4 Enhances Sensitivity of Cisplatin to Lung Cancer Cells and Inhibits Regulating Epithelial-to-Mesenchymal Transition. Oncol. Res. 2016, 24, 81–87. [Google Scholar] [CrossRef]
  456. Zhen, Y.; Fang, W.; Zhao, M.; Luo, R.; Liu, Y.; Fu, Q.; Chen, Y.; Cheng, C.; Zhang, Y.; Liu, Z. miR-374a-CCND1-pPI3K/AKT-c-JUN feedback loop modulated by PDCD4 suppresses cell growth, metastasis, and sensitizes nasopharyngeal carcinoma to cisplatin. Oncogene 2017, 36, 275–285. [Google Scholar] [CrossRef]
  457. Piskareva, O.; Harvey, H.; Nolan, J.; Conlon, R.; Alcock, L.; Buckley, P.; Dowling, P.; Henry, M.; O’Sullivan, F.; Bray, I.; et al. The development of cisplatin resistance in neuroblastoma is accompanied by epithelial to mesenchymal transition in vitro. Cancer Lett. 2015, 364, 142–155. [Google Scholar] [CrossRef] [Green Version]
  458. Zhu, X.; Shen, H.; Yin, X.; Long, L.; Xie, C.; Liu, Y.; Hui, L.; Lin, X.; Fang, Y.; Cao, Y.; et al. miR-186 regulation of Twist1 and ovarian cancer sensitivity to cisplatin. Oncogene 2016, 35, 323–332. [Google Scholar] [CrossRef]
  459. Gu, J.; Liang, Y.; Qiao, L.; Lu, Y.; Hu, X.; Luo, D.; Li, N.; Zhang, L.; Chen, Y.; Du, J.; et al. URI expression in cervical cancer cells is associated with higher invasion capacity and resistance to cisplatin. Am. J. Cancer Res. 2015, 5, 1353–1367. [Google Scholar]
  460. Boac, B.M.; Xiong, Y.; Marchion, D.C.; Abbasi, F.; Bush, S.H.; Ramirez, I.J.; Khulpateea, B.R.; Clair McClung, E.; Berry, A.L.; Bou Zgheib, N.; et al. Micro-RNAs associated with the evolution of ovarian cancer cisplatin resistance. Gynecol. Oncol. 2016, 140, 259–263. [Google Scholar] [CrossRef] [Green Version]
  461. Liu, J.; Chen, X.; Ward, T.; Pegram, M.; Shen, K. Combined niclosamide with cisplatin inhibits epithelial-mesenchymal transition and tumor growth in cisplatin-resistant triple-negative breast cancer. Tumor Boil. 2016, 37, 9825–9835. [Google Scholar] [CrossRef]
  462. Huang, D.; Duan, H.; Huang, H.; Tong, X.; Han, Y.; Ru, G.; Qu, L.; Shou, C.; Zhao, Z. Cisplatin resistance in gastric cancer cells is associated with HER2 upregulation-induced epithelial-mesenchymal transition. Sci. Rep. 2016, 6, 20502. [Google Scholar] [CrossRef] [Green Version]
  463. Tieche, C.C.; Peng, R.W.; Dorn, P.; Froment, L.; Schmid, R.A.; Marti, T.M. Prolonged pemetrexed pretreatment augments persistence of cisplatin-induced DNA damage and eliminates resistant lung cancer stem-like cells associated with EMT. BMC Cancer 2016, 16, 125. [Google Scholar] [CrossRef] [Green Version]
  464. Ge, X.; Liu, X.; Lin, F.; Li, P.; Liu, K.; Geng, R.; Dai, C.; Lin, Y.; Tang, W.; Wu, Z.; et al. MicroRNA-421 regulated by HIF-1alpha promotes metastasis, inhibits apoptosis, and induces cisplatin resistance by targeting E-cadherin and caspase-3 in gastric cancer. Oncotarget 2016, 7, 24466–24482. [Google Scholar] [CrossRef]
  465. Chen, Q.Y.; Jiao, D.M.; Wang, J.; Hu, H.; Tang, X.; Chen, J.; Mou, H.; Lu, W. miR-206 regulates cisplatin resistance and EMT in human lung adenocarcinoma cells partly by targeting MET. Oncotarget 2016, 7, 24510–24526. [Google Scholar] [CrossRef] [Green Version]
  466. Zhang, P.; Hong, H.; Sun, X.; Jiang, H.; Ma, S.; Zhao, S.; Zhang, M.; Wang, Z.; Jiang, C.; Liu, H. MicroRNA-10b regulates epithelial-mesenchymal transition by modulating KLF4/Notch1/E-cadherin in cisplatin-resistant nasopharyngeal carcinoma cells. Am. J. Cancer Res. 2016, 6, 141–156. [Google Scholar]
  467. Wang, L.L.; Zhang, X.H.; Zhang, X.; Chu, J.K. MiR-30a increases cisplatin sensitivity of gastric cancer cells through suppressing epithelial-to-mesenchymal transition (EMT). Eur. Rev. Med Pharmacol. Sci. 2016, 20, 1733–1739. [Google Scholar]
  468. Xu, G.; Yu, H.; Shi, X.; Sun, L.; Zhou, Q.; Zheng, D.; Shi, H.; Li, N.; Zhang, X.; Shao, G. Cisplatin sensitivity is enhanced in non-small cell lung cancer cells by regulating epithelial-mesenchymal transition through inhibition of eukaryotic translation initiation factor 5A2. BMC Pulm. Med. 2014, 14, 174. [Google Scholar] [CrossRef] [Green Version]
  469. Zhang, F.; Yang, B.; Shi, S.; Jiang, X. RNA interference (RNAi) mediated stable knockdown of protein casein kinase 2-alpha (CK2alpha) inhibits migration and invasion and enhances cisplatin-induced apoptosis in HEp-2 laryngeal carcinoma cells. Acta Histochem. 2014, 116, 1000–1006. [Google Scholar] [CrossRef]
  470. Yu, M.; Zhang, C.; Li, L.; Dong, S.; Zhang, N.; Tong, X. Cx43 reverses the resistance of A549 lung adenocarcinoma cells to cisplatin by inhibiting EMT. Oncol. Rep. 2014, 31, 2751–2758. [Google Scholar] [CrossRef]
  471. Jiang, Z.; Yin, J.; Fu, W.; Mo, Y.; Pan, Y.; Dai, L.; Huang, H.; Li, S.; Zhao, J. MiRNA 17 family regulates cisplatin-resistant and metastasis by targeting TGFbetaR2 in NSCLC. PLoS ONE 2014, 9, e94639. [Google Scholar] [CrossRef] [Green Version]
  472. Sobral, L.M.; Sousa, L.O.; Coletta, R.D.; Cabral, H.; Greene, L.J.; Tajara, E.H.; Gutkind, J.S.; Curti, C.; Leopoldino, A.M. Stable SET knockdown in head and neck squamous cell carcinoma promotes cell invasion and the mesenchymal-like phenotype in vitro, as well as necrosis, cisplatin sensitivity and lymph node metastasis in xenograft tumor models. Mol. Cancer 2014, 13, 32. [Google Scholar] [CrossRef] [Green Version]
  473. Baribeau, S.; Chaudhry, P.; Parent, S.; Asselin, E. Resveratrol inhibits cisplatin-induced epithelial-to-mesenchymal transition in ovarian cancer cell lines. PLoS ONE 2014, 9, e86987. [Google Scholar] [CrossRef] [Green Version]
  474. Wolf, M.A.; Claudio, P.P. Benzyl isothiocyanate inhibits HNSCC cell migration and invasion, and sensitizes HNSCC cells to cisplatin. Nutr. Cancer 2014, 66, 285–294. [Google Scholar] [CrossRef] [Green Version]
  475. Yu, H.G.; Wei, W.; Xia, L.H.; Han, W.L.; Zhao, P.; Wu, S.J.; Li, W.D.; Chen, W. FBW7 upregulation enhances cisplatin cytotoxicity in non- small cell lung cancer cells. Asian Pac. J. Cancer Prev. 2013, 14, 6321–6326. [Google Scholar] [CrossRef] [Green Version]
  476. Pennati, M.; Lopergolo, A.; Profumo, V.; De Cesare, M.; Sbarra, S.; Valdagni, R.; Zaffaroni, N.; Gandellini, P.; Folini, M. miR-205 impairs the autophagic flux and enhances cisplatin cytotoxicity in castration-resistant prostate cancer cells. Biochem. Pharmacol. 2014, 87, 579–597. [Google Scholar] [CrossRef]
  477. Liu, H.; Gu, Y.; Yin, J.; Zheng, G.; Wang, C.; Zhang, Z.; Deng, M.; Liu, J.; Jia, X.; He, Z. SET-mediated NDRG1 inhibition is involved in acquisition of epithelial-to-mesenchymal transition phenotype and cisplatin resistance in human lung cancer cell. Cell. Signal. 2014, 26, 2710–2720. [Google Scholar] [CrossRef]
  478. Xiong, X.; Arvizo, R.R.; Saha, S.; Robertson, D.J.; McMeekin, S.; Bhattacharya, R.; Mukherjee, P. Sensitization of ovarian cancer cells to cisplatin by gold nanoparticles. Oncotarget 2014, 5, 6453–6465. [Google Scholar] [CrossRef]
  479. Fan, G.L.; Yao, Y.; Yao, L.; Li, Y. PDCD5 transfection increases cisplatin sensitivity and decreases invasion in hepatic cancer cells. Oncol. Lett. 2015, 9, 411–417. [Google Scholar] [CrossRef] [Green Version]
  480. Liu, W.H.; Chen, M.T.; Wang, M.L.; Lee, Y.Y.; Chiou, G.Y.; Chien, C.S.; Huang, P.I.; Chen, Y.W.; Huang, M.C.; Chiou, S.H.; et al. Cisplatin-selected resistance is associated with increased motility and stem-like properties via activation of STAT3/Snail axis in atypical teratoid/rhabdoid tumor cells. Oncotarget 2015, 6, 1750–1768. [Google Scholar] [CrossRef] [Green Version]
  481. Zhao, Z.; Zhang, L.; Yao, Q.; Tao, Z. miR-15b regulates cisplatin resistance and metastasis by targeting PEBP4 in human lung adenocarcinoma cells. Cancer Gene Ther. 2015, 22, 108–114. [Google Scholar] [CrossRef]
  482. Chen, D.J.; Chen, W.; Jiang, H.; Yang, H.; Wang, Y.C.; Chen, J.H. Downregulation of DOCK1 sensitizes bladder cancer cells to cisplatin through preventing epithelial-mesenchymal transition. Drug Des. Dev. Ther. 2016, 10, 2845–2853. [Google Scholar] [CrossRef] [Green Version]
  483. Quiles, J.L.; Sánchez-González, C.; Vera-Ramirez, L.; Giampieri, F.; Navarro-Hortal, M.D.; Xiao, J.; Llopis, J.; Battino, M.; Varela-Lopez, A. Reductive Stress, Bioactive Compounds, Redox-Active Metals and Dormant Tumor Cell Biology to Develop Redox-Based Tools for the Treatment of Cancer. Antioxid. Redox Signal. 2020. [Google Scholar] [CrossRef]
  484. Flores-Guzmán, F.; Utikal, J.; Umansky, V. Dormant tumor cells interact with memory CD8+ T cells in RET transgenic mouse melanoma model. Cancer Lett. 2020, 474, 74–81. [Google Scholar] [CrossRef]
  485. Barney, L.E.; Hall, C.L.; Schwartz, A.D.; Parks, A.N.; Sparages, C.; Galarza, S.; Platt, M.O.; Mercurio, A.M.; Peyton, S.R. Tumor cell–organized fibronectin maintenance of a dormant breast cancer population. Sci. Adv. 2020, 6, eaaz4157. [Google Scholar] [CrossRef] [Green Version]
  486. Hen, O.; Barkan, D. Dormant disseminated tumor cells and cancer stem/progenitor-like cells: Similarities and opportunities. Semin. Cancer Boil. 2020, 60, 157–165. [Google Scholar] [CrossRef]
Figure 1. The involvement of different molecular pathways in anticancer actions of cisplatin (CP).
Figure 1. The involvement of different molecular pathways in anticancer actions of cisplatin (CP).
Ijms 21 04002 g001
Figure 2. A summary of selected molecular pathways regulating epithelial–mesenchymal transition (EMT) and their possible involvement in resistance to CP therapy. mTOR, mammalian target of rapamycin; Akt, protein kinase B; GSK-3β, glycogensynthase kinase-3β; miR, microRNA; PTP1B, protein tyrosine phosphatase 1B; HOXA3, homeobox A3; Mdm2, mouse double minute 2 homolog; IKKβ, Inhibitor of NF-κB kinase subunit β; ZEB, zinc-finger E-box binding homeobox; lncRNA, long non-coding RNA; UCA1, urogenital carcinoma antigen 1; elF5A2, eukaryotic initiation factor 5A2.
Figure 2. A summary of selected molecular pathways regulating epithelial–mesenchymal transition (EMT) and their possible involvement in resistance to CP therapy. mTOR, mammalian target of rapamycin; Akt, protein kinase B; GSK-3β, glycogensynthase kinase-3β; miR, microRNA; PTP1B, protein tyrosine phosphatase 1B; HOXA3, homeobox A3; Mdm2, mouse double minute 2 homolog; IKKβ, Inhibitor of NF-κB kinase subunit β; ZEB, zinc-finger E-box binding homeobox; lncRNA, long non-coding RNA; UCA1, urogenital carcinoma antigen 1; elF5A2, eukaryotic initiation factor 5A2.
Ijms 21 04002 g002
Table 1. The involvement of diverse molecular pathways in EMT-mediated resistance to CP therapy.
Table 1. The involvement of diverse molecular pathways in EMT-mediated resistance to CP therapy.
Cancer TypeSignaling AxisEffect on EMTResultsRefs
Lung cancerEMT/PD-L1/PD-1-The CP-resistant lung cancer cells activate EMT to induce the expression of PD-L1/PD-1, ensuring their survival and resistance to CP therapy. Anti-PD-1 or anti-PD-L1 therapies may lead to the sensitivity of cancer cells in CP chemotherapy[426]
Lung cancerTGF-β1/EMTInductionTGF-β1 is able to stimulate EMT via enhancing N-cadherin and vimentin and decreasing E-cadherin. In addition, drug-resistant proteins such as ERCC1 and p-glycoprotein undergo upregulation. Inhibition of TGF-β1 enhances the sensitivity of cancer cells in CP therapy[362]
Squamous cell carcinomaSOX8/Wnt-β-catenin/EMTInductionSOX8 induces EMT through Frizzled-7-mediated Wnt signaling pathway to diminish the efficacy of CP chemotherapy[427]
Lung cancerMF-κB/EMTInductionThe administration of ginsenoside Rg3 sensitizes cancer cells in CP chemotherapy by inhibition of NF-κB-induced EMT[428]
Ovarian cancer--PD98059 induces EMT mechanism to reduce the sensitivity of cancer cells to CP chemotherapy[429]
Breast cancerNF-κB/EMTInductionEugenol inhibits EMT through NF-κB downregulation, leading to the enhanced anti-tumor activity of CP [430]
Cervical cancerRIF1/EMTInductionRIF1 enhances the malignancy of cancer cells through EMT induction. Suppressing RIF1 sensitizes cancer cells in CP chemotherapy[431]
Breast cancerαvβ3 integrin/EMT
FAK/PI3K/Akt/EMT
Induction14, 15-EET trigger EMT through activation of FAK/PI3K/Akt and αvβ3 integrin to ensure the resistance of cancer cells in CP chemotherapy[432]
Ovarian cancerNotch3/SUSD2/EMTInductionThe downstream mediator of Notch3, SUSD4 induces EMT to trigger the resistance of cancer cells in CP chemotherapy [433]
Lung cancerTGF-β1/EMTInductionInhibition of TGF-β1 is associated with EMT downregulation and consequently, the sensitivity of cancer cells in CP chemotherapy [434]
Ovarian cancerMiR-20a/EMTInductionThe miR-20a induces CP resistance in cancer cells through EMT induction[435]
Non-small cell lung cancerSLC39A4/EMTInductionThe inhibition of SLC39A4 suppresses EMT and sensitizes cancer cells in CP chemotherapy[436]
Nasopharyngeal carcinomaHippo/TAZ/EMTInductionThe TAZ activation as a key gene of Hippo pathway induces EMT and triggers the resistance of cancer cells in CP chemotherapy[437]
Colorectal cancerhERG1/EMTInductionThe hERG1 ion channels induce EMT to diminish the sensitivity of cancer cells in CP chemotherapy[438]
Nasopharyngeal carcinomaTIMELESS/EMTInductionSuppressing TIMELESS expression inhibits EMT to enhance the efficacy of CP chemotherapy[439]
Ovarian cancerMiR-30a/c-5p/DNMT1/EMTInhibitionThe overexpressed miR-30a/c-5p inhibits EMT through DNMT1 downregulation, leading to the sensitivity of cancer cells in CP chemotherapy[440]
Cervical canceriASPP/miR-20a/FBXL5/BTG3/EMTInductionThe iASPP inhibits FBXL5/BTG3 axis through miR-20a upregulation. As a consequence, EMT mechanism occurs to ensure the resistance of cancer cells in CP chemotherapy[441]
Nasopharyngeal carcinomaNEDD4/EMTInductionKnock-out of NEDD4 inhibits EMT and induces mesenchymal–epithelial transition, resulting in sensitivity of cancer cells in CP chemotherapy[442]
Oral tongue squamous cell cancerMiR-15b/TRIM14/EMTInhibitionThe upregulated miR-15b suppresses EMT via TRIM14 downregulation to induce mesenchymal –epithelial transition and sensitize cancer cells in CP chemotherapy[443]
Lung adenocarcinomaPI3K/Akt/NF-κB/EMTInductionThe administration of baicalein inhibits the PI3K/Akt/NF-κB-mediated EMT to sensitize cancer cells in CP chemotherapy and reduce their malignancy and migration[444]
Oral tongue squamous cell carcinomahas-miR-485-5p/PAK1/ERCC1-YAP/EMTInhibitionThe downstream mediators of PAK1 including ERCC1 and YAP are involved in induction of EMT and resistance of cancer cells in CP chemotherapy. The hsa-miR-485-5p disrupts the aforementioned axis to suppresses EMT and malignancy, and restore the sensitivity to CP treatment[445]
Gastric cancerTAZ/EMTInductionThe overexpression of TAZ stimulates EMT to ensure the malignancy and invasion of cancer cells, leading to resistance in CP therapy[446]
Non-small cell lung cancerARK5/EMTInductionThe ARK5 triggers EMT to enhance the malignancy and metastasis of cancer cells. The silencing of ARK5 sensitizes cancer cells in CP chemotherapy through EMT inhibition[447]
Non-small cell lung cancerAurora A/EMTInductionThe downregulation of Aurora A inhibits EMT, leading to the sensitivity of cancer cells in CP chemotherapy[448]
Ovarian cancerHPIP/PI3K/Akt/EMTInductionHPIP elevates the malignancy and invasion of cancer cells by EMT induction via PI3K/Akt axis. This enhanced malignancy leads to the resistance in CP chemotherapy[449]
Ovarian cancerEMT/Akt-The EMT induces Akt activation to drive the resistance in CP chemotherapy[450]
Non-small cell lung cancerMiR-101/ROCK2/EMTInhibitionThe miR-101 has a reverse relationship with ROCK2 to inhibit EMT and promote the sensitivity of cancer cells in CP chemotherapy[451]
Non-small cell lung cancerFASN/TGF-β1/FASNInductionA positive loop between FASN and TGF-β1 induces EMT and triggers CP resistance[452]
Cervical cancerMiR-25-3p/Sema4C/EMTInhibitionBy inhibition of Sema4C, miR-25-3p suppresses EMT to inhibit cancer metastasis and malignancy and sensitize them in CP chemotherapy[453]
Ovarian cancerFOXC2/ERK/EMT
FOXC2/Akt/GSK-3β/EMT
InductionThe FOXC2 activates EMT mechanism via ERK and Akt/GSK-3β signaling pathways to induce the resistance of cancer cells in CP chemotherapy[454]
Lung cancerKLF4/EMTInhibitionThe upregulation of KLF4 inhibits EMT mechanism through enhancing Slug, Twist and vimentin levels, and reducing E-cadherin levels, resulting in decreased malignancy and induction of apoptosis by CP chemotherapy[455]
Nasopharyngeal carcinomaMiR-374a/PDCD4/CCND1/
PI3K/Akt/c-Jun/EMT
InhibitionThe miR-374a downregulates the expression of CCND1 by induction of tumor suppressor PDCD4. Then, a decrease occurs in PI3K/Akt/c-Jun signaling pathway to inhibit EMT, leading to the sensitivity of cancer cells in CP chemotherapy[456]
NeuroblastomaMYH9
ACTN4
ROCK1
Induction
Induction
Inhibition
Overexpressed MYH9 and ACTN4, and decreased expression of ROCK1, are involved in EMT and resistance of cancer cells in CP chemotherapy[457]
Ovarian cancerMiR-186/Twist1/EMTInhibitionThe miR-186 inhibits EMT through Twist1 downregulation to drive the sensitivity of cancer cells in CP chemotherapy[458]
Cervical cancerURI/EMTInductionThe silencing of URI inhibits EMT to sensitize cancer cells in CP chemotherapy[459]
Ovarian cancerMiR-496
MiR-485-5p
MiR-152
Let-7g
InductionThese miRs are associated with induction of EMT and resistance of cancer cells in CP chemotherapy[460]
Triple negative breast cancer--The administration of niclosamide inhibits EMT to enhance the induction of apoptotic cell death[461]
Gastric cancerHER2/Snail/EMTInductionThe HER2 activates Snail/EMT axis to reduce the efficacy of CP chemotherapy[462]
Lung cancer--Pemetrexed pretreatment inhibits EMT to sensitize cancer cells in CP chemotherapy[463]
Gastric cancerHIF-α/miR-421/EMTInductionThe HIF-α-mediated miR-421 upregulation reduces E-cadherin levels, leading to the EMT stimulation and CP resistance [464]
Lung adenocarcinomaMiR-206/MET/PI3K/AKT/
mTOR/EMT
InhibitionThe inactivation of MET/PI3K/Akt/mTOR axis by miR-206 inhibits EMT and drives the sensitivity of cancer cells in CP chemotherapy[465]
Nasopharyngeal carcinomaMiR-10b/KLF4/Notch1/E-cadherinInductionThe miR-10b upregulates the expression of Notch1 via KLF4 inhibition to reduce E-cadherin levels, leading to the stimulation of EMT and CP resistance[466]
Gastric cancerMiR-30a/EMTInhibitionThe overexpressed miR-30a reduces EMT to sensitize cancer cells in CP chemotherapy[467]
Non-small cell lung cancerelF5A2/EMTInductionGC7 inhibits EMT through elF5A2 downregulation, resulting in sensitivity of cancer cells in CP chemotherapy[468]
Laryngeal carcinoma cellsCK2α/EMTInductionSilencing of CK2α suppresses EMT to inhibit malignancy and resistance of cancer cells in CP chemotherapy[469]
Lung adenocarcinomaCx43/EMTInhibitionCx43 reduces the malignancy and invasion of cancer cells through EMT inhibition, leading to the sensitivity in CP chemotherapy[470]
Non-small cell lung cancerMiR-17
MiR-20a
MiR-20b
InhibitionThese miRs are able to suppress EMT and sensitizing cancer cells in CP chemotherapy[471]
Head and neck squamous cell carcinomaSET/EMTInductionAs an oncogenic factor, SET induces EMT to reduce the sensitivity of cancer cells in CP chemotherapy[472]
Ovarian cancerERK/Snail/EMTInductionThe administration of resveratrol diminishes the expression of ERK to inhibit Snail and EMT, leading to the sensitivity of cancer cells in CP chemotherapy[473]
Head and neck squamous cell carcinoma--Benzyl isothiocyanate suppresses CP resistance through EMT downregulation [474]
Non-small cell lung cancerFBW7/EMTInhibitionThe overexpression of FBW7 inhibits EMT to enhance the sensitivity of cancer cells in CP chemotherapy[475]
Prostate cancerMiR-205/autophagy/EMTInhibitionThe overexpression of miR-205 inhibits autophagy to suppress EMT and sensitize cancer cells in CP chemotherapy[476]
Lung cancerSET/NDRG1/EMTInductionThe inhibition of NDRG by SET triggers EMT, leading to the resistance of cancer cells in CP chemotherapy[477]
Ovarian cancerAkt/EMT
NF-κB/EMT
InductionGold nanoparticles are able to inhibit Akt and NF-κB signaling pathways to suppress EMT and CP resistance[478]
Hepatic cancerPDCD5/TGF-β/EMTInhibitionBy downregulation of TGF-β, PDCD5 inhibits EMT to sensitize cancer cells in CP chemotherapy[479]
Atypical teratoid/rhabdoid tumorSTAT3/Snail/EMTInductionInhibition of STAT3/Snail axis suppresses EMT and partially sensitizes cancer cells in CP chemotherapy[480]
Lung adenocarcinomaMiR-15b/PEBP4/EMTInductionBy downregulation of PEBP4, miR-15b triggers EMT mechanism to mediate the resistance of cancer cells in CP chemotherapy[481]
Bladder cancerDOCK1/EMTInductionSilencing DOCK1 expression is associated with sensitivity of cancer cells to CP via EMT inhibition[482]

Share and Cite

MDPI and ACS Style

Ashrafizadeh, M.; Zarrabi, A.; Hushmandi, K.; Kalantari, M.; Mohammadinejad, R.; Javaheri, T.; Sethi, G. Association of the Epithelial–Mesenchymal Transition (EMT) with Cisplatin Resistance. Int. J. Mol. Sci. 2020, 21, 4002. https://doi.org/10.3390/ijms21114002

AMA Style

Ashrafizadeh M, Zarrabi A, Hushmandi K, Kalantari M, Mohammadinejad R, Javaheri T, Sethi G. Association of the Epithelial–Mesenchymal Transition (EMT) with Cisplatin Resistance. International Journal of Molecular Sciences. 2020; 21(11):4002. https://doi.org/10.3390/ijms21114002

Chicago/Turabian Style

Ashrafizadeh, Milad, Ali Zarrabi, Kiavash Hushmandi, Mahshad Kalantari, Reza Mohammadinejad, Tahereh Javaheri, and Gautam Sethi. 2020. "Association of the Epithelial–Mesenchymal Transition (EMT) with Cisplatin Resistance" International Journal of Molecular Sciences 21, no. 11: 4002. https://doi.org/10.3390/ijms21114002

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop