Skip to main content
Cancer Reports logoLink to Cancer Reports
. 2020 Oct 13;5(12):e1291. doi: 10.1002/cnr2.1291

Molecular and cellular paradigms of multidrug resistance in cancer

Foram U Vaidya 1, Abu Sufiyan Chhipa 1, Vinita Mishra 1, Vishal Kumar Gupta 2, Shiv Govind Rawat 2, Ajay Kumar 2, Chandramani Pathak 1,
PMCID: PMC9780431  PMID: 33052041

Abstract

Background

The acquisition of resistance to chemotherapy is a major hurdle in the successful application of cancer therapy. Several anticancer approaches, including chemotherapies, radiotherapy, surgery and targeted therapies are being employed for the treatment of cancer. However, cancer cells reprogram themselves in multiple ways to evade the effect of these therapies, and over a period of time, the drug becomes inactive due to the development of multi‐drug resistance (MDR). MDR is a complex phenomenon where malignant cells become insensitive to anticancer drugs and attain the ability to survive even after several exposures of anticancer drugs. In this review, we have discussed the molecular and cellular paradigms of multidrug resistance in cancer.

Recent Findings

An Extensive research in cancer biology revealed that drug resistance in cancer is the result of perpetuated intracellular and extracellular mechanisms such as drug efflux, drug inactivation, drug target alteration, oncogenic mutations, altered DNA damage repair mechanism, inhibition of programmed cell death signaling, metabolic reprogramming, epithelial mesenchymal transition (EMT), inherent cell heterogeneity, epigenetic changes, redox imbalance, or any combination of these mechanisms. An inevitable cross‐link between inflammation and drug resistance has been discussed. This review provided insight molecular mechanism to understand the vulnerabilities of cancer cells to develop drug resistance.

Conclusion

MDR is an outcome of interplays between multiple intricate pathways responsible for the inactivation of drug and development of resistance. MDR is a major obstacle in regimens of successful application of anti‐cancer therapy. An improved understanding of the molecular mechanism of multi drug resistance and cellular reprogramming can provide a promising opportunity to combat drug resistance in cancer and intensify anti‐cancer therapy for the upcoming future.

Keywords: Multi drug resistance and cancer, Tumor microenvironment, Cellular reprogramming, Cancer metabolism, Programmed cell death, Inflammation

1. INTRODUCTION

Cancer is the second leading cause of death and it is a socio‐economic problem worldwide. 1 In the current scenario, incidences and mortality rates of cancer are increasing gradually. The advancements in cancer therapies, including chemotherapy, surgery, radiation therapy, precise anticancer therapy, immunotherapy, and targeted therapy are still challenging and not satisfactory. In clinical practices, chemotherapy and surgery are most often therapy offered against cancer, but cancer cells become chemo resistant over a short span of treatment with anticancer drugs. The acquisition of resistance to chemotherapy is a major obstacle in successful application of anti‐cancer therapy. It is well accepted that chemotherapy has adverse side effects such as systemic toxicities, immune surveillance and drug resistance. The majority of chemotherapeutic drugs approved by the FDA having lower molecular weight and require a higher concentration for their pharmacological actions. Many anti‐cancer drugs act indiscriminately adjacent to cancerous and healthy cells. 2 , 3 , 4 Many types of cancers show susceptibility toward chemotherapy at initial stage, but after some time start developing resistance because of multiple intrinsic and extrinsic factors such as cellular reprogramming, oncogenic stimulation, drug efflux due to over expression of multi‐drug resistance (MDR) genes and metabolic changes that promote drug inactivation and inhibition, altered DNA damage repair mechanism, evasion of programmed cell death, epithelial mesenchymal transition (EMT), inherent cell heterogeneity, epigenetic changes, metabolic reprogramming or any combination of these mechanisms. 5 The immune surveillance also may impair due to the unpredictable cell death escape strategies acquired by cancer cells. 6 Moreover, the link between cancer and oxidative stress has been extensively studied, which denotes the significant involvement of ROS in the progression of cancer. 7 In addition, the imbalance in redox homeostasis is also behaving as a critical factor in the development of drug resistance in cancer. The oxidative stress plays crucial role in cell survival and therefore, it may confers drug resistance in cancer. 8

Resistance could be restricted to the drug which was used to treat the patient (single‐agent resistance) or execute simultaneous failure against structurally and functionally different anti‐cancer drugs (Multi drug resistance, MDR). 9 Resistance against multiple drugs during cancer therapy has been a “clinician's nightmare,” owing to its capacity to subvert the desired drug response in cancer patients. Therefore, a regimen of cancer therapy is particularly challenged to deal with drug resistance. In this review, we have attempted to shed light on various mechanisms of drug resistance, those were adopted by cancer cells to ensure for their survival.

2. TYPES OF DRUG RESISTANCE

The drug resistance can be categorized as intrinsic or acquired resistance. Intrinsic resistance is the innate resistance, which exists prior to the treatment of chemotherapeutic drug. 10 Intrinsic resistance may be acquired by different mechanisms including (a) inherent genetic mutations in the tumor cells, (b) development of resistant population such as cancer stem cells in heterogeneous tumors, and (c) commencement of intrinsic pathways that are responsible for the detoxification under normal physiological conditions. On the other hand, acquired resistance can develop after receving anti‐cancer therapy.

Acquired resistance can be an outcome of various cellular and molecular responses including: (a) activation of second proto‐oncogene after treatment; Cancer cells can acquire resistance against targeted drugs by the generation of new mutation or alteration in the expression; (b) alterations in drug targets; (c) Drug metabolisms in the tumor; (d) efflux of drugs by transmembrane transporters (ATP binding cassettes, ABCs transporters); (e) Epigenomic alteration due to acetylation, methylation and altered level of microRNAs which creates changes in the downstream or upstream receptors; (f) changes in the tumor microenvironment (TME) after treatment. 10 All these mechanisms of drug resistance can act independently or in combination to favor multidrug resistance in cancer.

Here, we have briefly described the cellular and molecular events, which are majorly involved in the development of drug resistance in cancer (Figure 1).

FIGURE 1.

FIGURE 1

Cellular and molecular events involved in the development of drug resistance in cancer

3. MECHANISMS OF DRUG RESISTANCE

3.1. Drug efflux by the ATP binding cassette (ABC) transporter family

A major factor that governs drug resistance in cancer is the overexpression of ABC transporter proteins that efflux many structurally and functionally distinct substrates via cell membrane by utilizing ATP hydrolysis. 11 These drug efflux transporters decrease the intracellular drug concentration and impede the drug response, which limits successful application of chemotherapy. 12 The current literature revealed that there are 48 ABC transporters have been identified in humans. 13 Many of them are involved in normal tissue protection and mainly expressed in the kidney, pancreas, liver, gastrointestinal (GI) tract, and the endothelium vessels of the testes and brain. 14 13 different types of ABC transporters have been identified those are directly or indirectly involved in multiple drug resistance in cancer. In the recent past, major transporters such as ABCB1 (Permeability glycoprotein /MDR1), ABCC1 (multidrug resistance associated protein‐1, MRP1) and ABCG2 (breast cancer resistance protein (BCRP) are extensively studied for exploring the mechanism of MDR. 15 Physiologically, ABCs transporters have function to remove the xenobiotics and toxic endogenous substances from the cells and organs to maintain their interstitium homeostasis. Cancer cells employ these membrane bound transporters system to acquire drug resistance. 16 The basic domain structures of ABCB1, ABCC1 and ABCG 2 are shown in Figure 2. Sequences and their domain information were retrieved from Uniprot Database 17 and domains were created with Illustrator of Biological Sequences (IBS). 18

FIGURE 2.

FIGURE 2

Domain structural organization of ABCB1, ABCC1, and ABCG2. Sequences and their domain information were retrieved from Uniprot Database and domains were created with Illustrator of Biological Sequences (IBS). TM, Transmembrane region; NBD, Nucleotide Binding Domain

The mechanisms of these ABC transporters are significantly governed by ATP. Drug resistant cancer cells are known to maintain comparatively higher ATP levels than their parental cancer cells. 19 Depletion of ATP inside the cancer cells significantly sensitizes them to chemotherapy. Conversely, higher intracellular concentrations of ATP transform the sensitive cells to drug resistant. 20 Moreover, the extracellular ATP also enhances the expression of ABC transporters, causing an increased rate of drug efflux. 21 The concentration of extracellular ATP promotes TME. 5 This increased ATP in the extracellular space of the cancer cells get internalized through a process termed as “macropinocytosis.” As a result, the remarkable increase in intracellular ATP concentration causes resistance against multiple chemotherapeutic drugs. 22 The major transporters involved in the efflux of the chemotherapeutic drugs are outlined below:

3.1.1. Permeability glycoprotein (P‐gp)/MDR‐1

P‐glycoprotein is a conserved, high molecular weight plasma membrane glycoprotein and first discovered human ABC transporter (encoded by the ABCB1 [adenosine Triphosphate binding cassette, subfamily B, member 1] gene). The ABCB1 gene is located on chromosome 7 in humans, which comprises 1280 amino acids and consisting molecular weight of ~ 170 kDa. 23 It has a transmembrane domain (TMD) and nucleotide‐binding domain (NBD). P‐gp holds two homologous halves each with 6 membrane‐spanning helices, and nucleotide binding domain which is present in the cytosol. The protein comprises of a large flexible drug binding cavity embedded within the membrane‐bound domain. This pocket contains several sub‐sites where the drug can bind through different sets of interactions. Binding of ATP at the NBDs, and subsequent ATP hydrolysis leads to conformational changes in TMDs, results in switching the conformation of transporter from inward to outward of the cells for unidirectional transport. 24 The substrates of P‐gp are usually lipid‐soluble protein, which interacts with the membrane protein before either being pumped out into the extracellular aqueous phase or moved to the extracellular membrane leaflet. The basal expression of P‐gp has been found in several normal tissues, such as liver, intestine, kidney, testes, placenta, and blood brain barrier to provide protection against xenobiotics and toxic substances. 25 The P‐gp transporters are the most commonly summoned transporter proteins and they play a critical role in the development of drug resistance in cancer cells. Several biochemical changes are associated with multidrug resistant cancer cells, where overexpression of P‐gp is the most common phenomenon in many types of cancers. P‐gp holds the central position in multidrug resistant cells by diminishing the intracellular accumulation of chemotherapeutic agents. 26 , 27 The P‐gp plays a vital role in the intestinal transport and efflux, which alters the bioavailability and pharmaceutical effects of orally administered pharmaceutical drugs. In recent past compiling evidence revealed that overexpression of P‐gp is associated with the development of MDR phenomenon in cancer. 26 P‐gp displays broad substrate specificity; therefore, P‐gp overexpressed cells execute cross resistance against multiple cytotoxic drugs, and help to develop multidrug resistance (MDR) in cancer cells. Initially, it was believed that efflux pumps are responsible for inhibiting the intake of conventional genotoxic anticancer drugs such as vinblastine, paclitaxel, and doxorubicin, but, burgeoning reports on P‐gp revealed its influence on around 300 compounds including the newly added “kinase inhibitors” in the list. 28 The P‐gp expression is one of the primary defensive mechanisms adopted by the cancer cells upon exposure to a cytotoxic agent. Moreover, the frequent confrontations of cancer cells with chemotherapeutic agents subsequently induce the expression of P‐gp to efflux and deplete the intracellular drug concentrations. 26 Besides this, the altered cellular signaling of cancer tends to fabricate a favorable environment for P‐gp expression. Tumor hypoxia, Warburg effect and acidosis in TME collectively impose a remarkable advantage to upregulate P‐gp expression in cancer cells for subverting the drug action. 28 Further, oncogenic stimulation, epigenetic alterations and aberrant cell death signaling simultaneously activate the expression of different genes, which promotes cancer cells to acquire drug resistance. A previous report also suggests that MDR cells hold P‐gp expression in the nuclear and mitochondrial membrane to efflux of anticancer drugs from nuclei and mitochondria to the cytosol for accelerating multidrug resistance in cancer. 29

3.1.2. Multidrug resistance protein (MRPs/ABCC1)

Multidrug resistance protein (MRPs/ABCC1) comprises of three hydrophobic TMD containing 17 membrane spanning helices, two NBD and an extra N‐terminal domain having molecular weight ~190 kDa. 30 Similar to P‐gp, MRP1 also belongs to the family of ABC transporters comprising 13 members. MRP member proteins 1‐9 were primarily found to be expressed in the tumor cells and associated with drug resistance against anticancer therapy. 31 MRP contains three TMD and two NBD. The expression of MDR associated protein 1 (MRP1/ABCC1) has been found in non‐P‐gp MDR cells. 32 The MRP1 has a similar function as P‐gp to pump out toxic substances in an ATP‐dependent manner. 33 The prime location of MRP is the proximal tubules and majorly involved in the excretory function of the kidney. MRP1 expresses constitutively in the testes, kidneys, placenta, and pharmacological barriers. However, a considerably higher expression of MRP1 was noticed in a number of tumors including lung, pancreatic, prostate, brain, and breast cancer. 16 MRP1 significantly contributes to the efflux of the number of anticancer agents, including anthracyclines, vinca alkaloids, methotrexate, camptothecins, and epipodophyllotoxins as well as organic anion substrates including compounds which are conjugated with glucuronide, sulfate and glutathione. Apart from these, the expression of MRP1 is also favored under hypoxic conditions. A positive relation between the HIF‐1α and MRP1 expression was observed in colon cancer cells. 34 However, the expression of MRP1 in cancer cells is more likely the result of induction of MDR by multiple factors that are peculiar to cancer cells.

3.1.3. Breast cancer resistance protein (BCRP/ABCG2)

BCRP/ABCG2 is another transporter protein that acquires the function to extrude the toxic substances in the extracellular spaces under normal physiological conditions. It has one TMD and one NBD consisting molecular weight of ~72 kDa. This protein is normally expressed in stem cells and in the apical membranes of the epithelium, which has involvement in the process of drug disposition. 35 It also expressed in liver, placenta, prostate, kidney, luminal surface of the endothelial cells of human brain microvessel, breast and adrenal gland. ABCG2 is also known as mitoxantrone resistance protein (MXR), which is responsible for efflux of the mitoxantrone in carcinoma cells. 36 In addition to P‐gp, the upregulated expression of BCRP is yet another mechanism has been employed by the cancer cells to prevent themselves from the actions of cytotoxic drugs. 35 , 37 BCRP induces the drug resistance against a wide range of anticancer drugs, including the conventionally employed genotoxic agents and novel tyrosine kinase inhibitors. BCRP is a major drug efflux transporter associated with breast cancer but several growing bodies of evidences suggest that it was also found in other cancers such as leukemia and lung cancer. 38 , 39 It can also be considered as a marker of CSCs in some cancers. ABCG2 can efficiently transport a number of chemotherapeutic drugs such as epipodophyllotoxin, mitoxantrone, camptothecins, bisantrene, anthracyclines, and flavopiridol as well as Tyrosine kinase inhibitor including gefitinib and imatinib. 40 , 41 Collectively, these drug efflux transporters have significant role in the development of multiple drug resistance in cancer. We have analyzed the interaction of ABCB1, ABCC1 and ABCG2 with the genes that are also involved in cancer pathology. STRING database 42 was used to identify interacting partners of proteins of interest. These interacting partner proteins were searched in Comparative Toxicogenomics Database (CTD) 43 by their gene name to confirm their involvement in cancer pathology and all of these were found to be involved in cancer pathology (details are not included in this review and can be found in CTD by gene name). Network view and molecular action view (inhibition or activation of interacting partners by the protein of interest) were shown in Figure 3.

FIGURE 3.

FIGURE 3

Interaction of ABCB1, ABCC1 and ABCG2 with the genes that are involved in cancer pathology. Interaction map was created with STRING database. The network and Molecular action view of protein‐protein interactions (by their gene name) were created by STRING version 11.0 with high confidence (0.700) and custom value for numbers of interactions were set to 50. Modes of action are shown in different colors

3.2. Drug inactivation and reduced cellular uptake

Systemic distribution and absorption of the drug directs the cellular function and response in the body. Drugs once entered into the body, undergo biochemical transformation by a variety of drug metabolism enzymes. Many anticancer agents require metabolic activation to execute their mode of action. However, the alteration or mutation in metabolic enzymes leads to drug inactivation. The enzymes, including cytochrome P450 (CYP) system, glutathione‐S‐transferase (GST) superfamily, and uridine diphospho‐glucuronosyltransferase (UGT) superfamily have been found in association with drug activation and inactivation in cancer cells. 44 Cytochrome P450s (CYP) is the member of a superfamily of heme proteins, and it has a significant role in endobiotic biosynthesis, xenobiotic biotransformation, and catabolism of bile acid, fatty acid, human steroid hormones and lipid‐soluble vitamins. There are almost 57 human microsomal CYPs out of which 15 seemed to be involved in drug metabolism. Alteration in CYP may change the metabolic capabilities of these proteins, such as the breakdown of the drug, and a significant increase in its secretion. As an outcome, the intratumoral concentration of drug will decline in patients, and drug becomes inactive. For instance, as Tamoxifen is a chemotherapeutic agent, which has been widely used for the treatment and prevention of breast cancer, but due to mutation or alteration in CYP2D6 gene, the efficacy of drug drastically decreases and drug becomes ineffective in the long term. 45 Moreover, a reduction in cellular drug uptake is also associated with another possible mechanism to develop drug resistance in cancer cells. Generally, cellular uptake of the drug executed via endocytosis or receptor mediated endocytosis, where the defective process may cause drug resistance. 10 Altered expression of Caveolin ‐1 (CAV1) is associated with the grade of cancer progression and invasion. It plays a key role in modulating the interaction between tumor and host by promoting metastasis, tumor growth, drug resistance for cell survival. 46

Further, the therapeutic efficacy of anticancer agents can be restricted by activation of detoxification systems that act as a guard against environmental toxins. In cancer cells, impaired detoxification system renders the ineffective drug response and promotes resistance. The exclusion of drugs by Glutathione S‐transferase is one of the major causative factors to create drug resistance in cancer. 47 Glutathione S‐transferase plays a vital role in multiple cellular processes, including cell proliferation, differentiation and apoptosis. An upregulation in GSH level contributes to drug resistance by multiple ways. A previous study suggests that it can bind or react with drugs, interact with ROS, prevents DNA/protein damage or involve in DNA repair mechanism and create resistance against cisplatin, 48 5‐fluorouracil 49 and doxorubicin. 50 During the treatment of the drug, an upregulated level of GST can modify the balance of kinases and favors the tumor growth. In brief, the binding sites for the transcriptional regulators, including AP‐1, AP‐2, NF‐kB, and Nrf‐2 are present on the promoter regions encoding γGCL and GST. Upon exposure of oxidative stimuli, Nrf‐2 dissociates from its negative regulator Keap1 and translocates to the nucleus. After translocation, it heterodimerizes with Maf proteins and binds with antioxidant responsive element (ARE) sequences. 51 This binding triggers the cytoprotective adaptive response by upregulating detoxification and cytoprotective genes such as GSH‐S‐reductase (GSR), GCLM, GCLC, GST, ferritin, MRP, heme oxygenase‐1 (HO‐1) and phase‐I drug oxidation enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1), which creates favorable TME. 52 The mutation in Nrf‐2 and keap1 in various human cancers promotes the constitutive expression of cytoprotective (prosurvival) genes due to continuous activation of Nrf‐2 and contributes to drug resistance in cancer. 53 UGT is the superfamily of enzymes that catalyze glucuronidation and regulates the formation of inactive hydrophilic glucuronides along with substrates including steroids, xenobiotic, bile acids, cytotoxics, and xenobiotics. The UGT1 and UGT2 genes code multiple functional UGTs in humans and facilitates first line metabolic defense against pathogenic substrates in multiple tissues including breast, skin, gut, placenta and prostate gland. However, in the cancerous state the UGT1A1 transcription and microsomal activity were found downregulated. DNA methylation negatively regulates the expression of UGT1A1 that facilitates the functional activity of irinotecan, which is a topoisomerase I inhibitor. 44 Nevertheless, the expression of UGT1A1 gets increased due to epigenetic changes, which deactivates the drug and enables the resistance against irinotecan as well as other drugs. Collectively, the mechanism of drug inactivation which develops the resistance in cancer cells needs further investigations.

Further, a remarkable influence of penetrability and tissue diffusion of drugs to the tumor site on drug resistance is also evident from several reports. For a drug to be effective, it is imperative for it to reach the target site at a lethal concentration. 54 Cancer cells and TME adopt multiple approaches to restrict the intracellular accumulation of the drug. It has been observed that cancer stem cells survive preferably in low oxygen tension to create a perfusion barrier against the movement of the drug inside cells. Moreover, the overwhelming clonal expansion of cancer tissue limits the presence of adequate vasculature around the newly developed tumor regions. This further limits the availability of the administered drug to the tumor site. Furthermore, the physical barrier to the drug penetration developed due to the presence of extracellular matrix (ECM), also remarkably restricts the movement of drug to the target sites. 55

3.3. Genomic instability and drug resistance

3.3.1. Alteration of drug targets and mutations

Genomic instability has a crucial role in the initiation and progression of cancer. It can arise by different mechanisms such as a mutation in the DNA, chromosomal abnormalities, telomere damage, and DNA repair mechanism, which fosters tumor growth. 56 Recent report highlighted that chromosomal instability underlies neoplastic cell transformation and tumor heterogeneity along with acquired drug resistance, which is tightly associated with drug responses and survival of cancer patients. 57 Genomic instability has been commonly observed in solid tumors and hematological malignancies, where alternation may occur from single nucleotide to chromosomal level. 58 Genomic instability of cancer cells can cause mutations or aberrant expression of drug targets such as genes or proteins, which may be the major causative factors for drug resistance. Alteration in the cellular targets of anti‐cancer agents reduces their therapeutic potential and promotes drug resistance. A compiling report suggests that due to alteration in the estrogen receptor of tumor cells, patients developed resistance against tamoxifen (anti‐estrogen) mediated endocrine therapy in breast cancer. 59 Cancer cells develop the mutations in a variety of genes that may significantly restrict the efficacy of chemotherapeutic drugs. The clinical study suggests that approximately 20‐30% of patients of chronic myelogenous leukemia developed resistance and experience relapse due to the generation of the point mutation in isoleucine (T315I) of the fusion tyrosine kinase protein after the treatment of tyrosine kinase inhibitor (TKI), imatinib. 60 Further, mutations in tumor suppressor gene p53 result in impaired functions and aberrant pro‐apoptotic balance, which further encourage the circumstance of drug resistance. 61 , 62 Report suggests that mutation in p53, induce resistance to cisplatin in non‐small cell lung cancer (NSCLC) cells by up‐regulating the expression of Nrf‐2, which may be incremental in developing MDR. 63 Furthermore, the nuclear stabilization of mutant p53 upon drug treatment also resulted in the development of resistance against gemcitabine in cancer. 64

3.3.2. DNA damage repair

DNA damaging drug have been used for the treatment of cancer for the induction of cell death or mitotic catastrophe. 65 However, the repair in the DNA damage also leads to the development of drug resistance as these drugs exert their effect by DNA damage. There are multiple DNA Damage Response (DDR) pathways including Nucleotide excision repair (NER), Base‐excision repair (BER), non‐homologous end joining (NHEJ), mismatch repair (MMR), Fanconi anemia (FA) pathway, translesion synthesis (TLS) and homologous recombination (HR) which are involved in the repair of single strand breaks (SSB), DNA lesions and double strand breaks (DSB). The deregulated expressions of these pathways in cancer cells increase the ability of cells for DNA damage repair and evasion of apoptosis. 66 The chemotherapeutic agents such as 5‐fluorouracil (5‐FU) and cisplatin induce DNA damage in cancer cells. However, due to the upregulation of the genes associated with the mechanism of DNA repair, the efficacy of anti‐cancer agents gets reduced and cancer cells eventually develop the drug resistant phenomenon. 67 A previous report suggests that loss of p53 and DNA mismatch repair leads to cisplatin induced drug resistance in cancer. 68 Clinical set of data revealed that human colorectal cells which were resistant to 5‐FU, showed upregulated expression of genes responsible for DNA repairs such as RAD23B, FANCG and FEN1. The altered DNA damage repair mechanisms significantly reduced the cell cycle arrest and skip the programmed cell death which leads to the development of drug resistance in cancer. 69

3.3.3. Epigenetic changes and drug resistance

Epigenetics play a pivotal role in the determination of cell fate and pathological provenience. It has seemed that the non‐genetic heterogeneity leads to the formation of tumor‐initiating cells and/or drug‐resistance. Epigenetic changes lead to the impaired gene expression, which persists for multiple cell divisions that eventually develop non‐genetic heterogeneity and drug non‐responsiveness. 70 The epigenetic alterations influence gene transcription by manipulating chromatin packaging and subsequently regulate the accessibility of DNA to sequence‐specific transcription factors. DNA methylation, chromatin remodeling, histone modification, and alterations in non‐coding RNA are associated with epigenetic alteration which is also driving force for the development of chemoresistance in cancer. 71 The molecular mechanisms revealed that aberrant methylation of CpG islands present at or near to the promoter region of the genes leads to the inactivation of gene during tumor development. 72 DNA methylation is linked with the binding of methyl‐binding domain (MBD) proteins followed by the recruitment of histone methyltransferases and histone deacetylases (HDACs) with subsequent events of histone modification, chromatin condensation and finally transcriptional inactivation of the associated genes. 73 The frequency of these kinds of epimutations is significantly higher as compared to genetic mutations. It was reported that during metastasis ~61 infrequent mutations were observed, out of which 15 were reported as driver genes and remaining were mutated passenger genes. 74 As a result, these mutations tend to exhibit a greater impact on the selection of subpopulations, which are associated with tumor progression and development of resistance against chemotherapeutic agents. The growing body of evidence suggests that acquisition of MDR phenomenon in many tumor cells is associated with the demethylation of the MDR1 promoter. Therefore, methylation at this promoter, decreases drug accumulation, controls MDR1 transcription, and increases the drug resistance in cancer cells. Epigenetic alterations also favor the DNA damage repair in cancer cells and develop acquired resistance against methylating chemotherapeutic agent by reactivating the DNA repair enzyme MGMT that promotes the survival of tumor cells. 75 Several reports suggested that methylation and epigenetic silencing in proapoptotic genes, including APAF1 and hMLH1 as well as in tumor suppressor genes such BRCA1 and E‐cadherin, results in development of resistance in cancer cells. 73 , 76 Moreover, the current compiling report also suggested the prominent role of exosomes in epigenetic alterations. 77 Recent report advocates that exosomes are also involved in the progression of the tumor, cell proliferation, and metastasis. Extracellular vesicles directly or indirectly can transfer the proteins and nucleic acids to the recipient cells, which can modulate histone modification, DNA methylation, and RNA post‐transcriptional regulation. 78 Exosomes are largely secreted by fibroblasts and immunocytes in the TME and transferre different cargos and microRNAs (miRNAs). The mechanisms of drug resistance, including drug efflux, alterations in drug metabolism, mutation of drug target, DNA damage repair, altered metabolism, cancer stem cells, and epigenetic changes are also regulated by exosomal miRNAs. 79 Thus, exosomal miRNA also play a vital role in the development of drug resistance.

3.4. Evasion of programmed cell death and drug resistance

Cancer cells ensure their overwhelming proliferative potential by evading the programmed cell death. Dysregulation of apoptosis is a characteristic feature and one of the hallmarks of cancer. 80 In the recent past, several reports advocate that inhibition of apoptosis and altered gene expression, mutation of apoptotic and anti‐apoptotic genes may contribute to drug resistance. Overexpression of several anti‐apoptotic genes and proteins such as Bcl‐2 family, decoy receptors (such as TRAIL‐R3/DcR1 and TRAIL‐R4/DcR2), cFLIP and inhibitor of apoptotic proteins (IAPs) have been found to be associated with resistance against chemotherapy. 81 , 82 Compelling evidence revealed that upregulation of BCL2, BCL‐XL, and MCL‐1, is associated with chemotherapy induced drug resistance in cancer. 83 Moreover, overexpression of death receptor such as TRAIL‐R1, TRAIL‐R2, and FAS has been found to associate with chemotherapy resistance. 84 , 85 The programmed cell death mechanism is intricately regulated by complex signaling mechanism. The cell death or survival signaling stimulated by intracellular or extracellular stimuli, targets various transcription factors such as NF‐kB, HIF‐1, c‐MYC, AP‐1 and STAT‐3 to mediate cellular response and fate of cells. 86 Chemotherapeutic drugs may induce cell death by distinct mechanisms including apoptosis, autophagy, and necroptosis. Apoptosis suppresses the inflammation but usually evaded by the immune cells, whereas necrotic cell death may cause inflammation and activate survival signaling by nuclear translocation of NF‐kB and secretion of pro‐inflammatory cytokines. This process promotes the TME and cell survival mechanisms. An activation of NF‐κB in response to drug exposure is also an approach adopted by sensitive cells to subvert the drug action. Activation and subsequent translocation of NF‐κB to nucleus activates the transcription factors, which are also responsible for the induction of chemoresistance in cancer cells. The molecular mechanisms of cell signaling are intricate to the drug response. Chemotherapeutic drugs instigate cell death through cytotoxic response by up‐regulating reactive oxygen species (ROS), changes in the mitochondrial membrane permeability, DNA damage, activation of tumor suppressor genes, and proteins as well as alteration of immune cells. 86 Cancer cells acquired several molecular changes for their survival. For instance, mutations in p53 gene alter the anticancer response of a chemotherapeutic agent that relies on the p53 mediated apoptosis in cancer cells. 87 Survivin is an anti‐apoptotic protein, which expresses in higher level in resistant cancer cells, apparently due to the down regulation of tumor suppressor genes. The overexpressed survivin in cancer cells promote evasion of cell death and favor anti‐cancer drug resistance. 88 In addition, cancer cells develop resistance against cisplatin due to DNA repair mechanism as the mode of action of cisplatin, relies on the DNA damage. Wip1, a protein that negatively regulates the ATM pathway of DNA damage was found in the resistant cells. Following this, the knockdown of Wip1 in oral squamous carcinoma cells (SCC) sensitized the cisplatin resistant cells. 89 The expression of P‐gp also interferes with the apoptotic signaling in cancer cells, thus providing “two‐way” protection to cancer cells from cell death. There is an inverse relationship between the expression patterns of P‐gp and TNF related apoptosis inducing ligand (TRAIL). It has been reported that P‐gp expression in cancer cells limits the action of TRAIL and therefore inhibits the apoptosis in transformed cells. 90

Autophagy is also another way of programmed cell death, which activates under stress condition. Autophagy has been defined as a lysosomal mediated degradation pathway that helps to degrade damaged organelles and cellular components to maintain homoeostasis. 91 At normal physiological condition, autophagy function as tumor suppression, but defective autophagy is associated with cell proliferation in cancer. The insight molecular mechanism of autophagy revealed that cells have an innate capacity to restore their energy balance during nutrients deprivation condition. Indeed, an upregulated autophagic flux can favor cell survival via activation of pro‐death signals. 92 Cancer cells also gain energy from the dead cells for their survival. The autophagic cell death during nutrient depreciation or stress condition developed from the cytotoxic drug may contribute to drug resistance during cancer therapy. However, the role of autophagy in cancer therapy is still controversial. Recent report highlighted that autophagy is a frequently confronted phenomenon during chemotherapy and has proven to be protective against the drug treatment in cancer. Autophagy is a widely recognized accomplice that drives a cancer cell toward MDR. 57 Supportively, inhibition of autophagy re‐sensitizes the resistant cells against chemotherapeutic agents. The upregulation of autophagy function as a constructive factor for developing drug resistance against chemotherapy, radiation therapy and even targeted therapies. Moreover, autophagy mediated MDR is regulated through a diverse signaling pathways that work in an intervention, context and type of cancer dependent manner. 93 For instance, resistance to doxorubicin, methotrexate and cisplatin in osteosarcoma cells is mediated through the activation of HSP90AA1 gene that regulates the activation of autophagy through PI3K/Akt/mTOR pathway. 94 A multifunctional protein, p62 is also reported to play a critical role in autophagy mediated drug resistance. 95 Similarly, another study revealed that triple resistant HEp‐2 cells were found to deplete p62 levels with simultaneous increases in Nrf‐2 (an antioxidant protein) and autophagy. Interestingly, cells with reduced p62 accompanied by an increased Nrf‐2 and autophagic flux were resistant to oxidative stress induced autophagy. 96 However, one contradictory report is also available which suggests that the overexpression of p62 in human hepatocarcinoma cells (HCC) is positively associated with Sorafenib resistance due to drug and cancer type dependent functions of p62. 97 In addition, IL‐6 mediated autophagy is yet another mechanism through which some cancer cells acquire MDR. Transglutaminase (TG2) mediated constitutive activation of NF‐kB initiates IL‐6 activation and subsequent autophagic response in resistant cells. Existing report advocates that IL‐6 mediated autophagy follows a positive loop mechanism for its consistent activation through the release of ATG5, an autophagic protein involved in autophagosome formation. 98 Autophagy also poses a significant impact on the response of cancer cells toward the radiotherapy or ionization therapy. Moreover, resistance to radiation therapy in breast cancer was found to be mediated through autophagy. 99 In another study, resistance to radiation therapy in cancer cells was reported to be mediated through Liver kinase B1 (LKB1), a tumor suppressor protein that activated autophagy via AMP‐ activated protein kinase (AMPK) with concurrent inhibition of apoptosis in resistant cells. 100 Collectively, inhibition of autophagy with simultaneous activation of apoptosis following the exposure of cancer cells to different interventions can be a lucrative approach to curb the transformation of sensitive cells to resistant phenotype on repeated exposure to chemotherapies.

Necrosis is an accidental cell death. It has been considered as caspase‐independent programmed cell death and termed as Necroptosis, which is morphologically analogous to necrosis and mechanically resembles to apoptosis. 101 The key necrotic component HMGB1 is known to be released from necrotic cells and triggers activation of the inflammatory signaling cascade to constitute TME. 102 High mobility group box 1 (HMGB1) is highly conserved chromatin associated nuclear protein that plays an important role in maintaining homeostasis of the cells. It translocates in between cytoplasm and nucleus and mainly resides in the nucleus to orchestrate the various nuclear events. HMGB1 is a critical regulator of cell death and survival signaling and also known as an alarming molecule, released by stressed cells which are undergoing necrosis and acts as endogenous danger signals to promote and exacerbate the inflammatory response that leads to the progression of cancer. 103 A previous study highlighted that HMGB1 releases after chemotherapy and promotes cell survival and drug resistance in cancer. 104 Mechanistically, necrosis triggers the release of danger‐associated molecular pattern molecules (DAMPs) that activate inflammasome components to secret the pro‐inflammatory cytokines, that is, IL‐1β, IL‐18, and TNF‐α, which build up inflammatory TME that aid resistance against anti‐cancer therapy. Chronic inflammatory responses have long been observed to be associated with various types of cancer and play decisive roles at different stages of cancer development. The release of danger signaling molecule HMGB1 can activate immune cells, including dendritic cells (DCs), via Toll‐like receptors (TLRs), RAGE, NF‐kB signaling for cell survival. 105 Thus, these reports suggest that defective programmed cell death signaling has a closed link with initiation and progression of cancer to acquire drug resistance.

3.5. Immunotherapy, Immune responses and drug resistance

Chemotherapy attributes an immunological response. Cell death, survival and drug resistance are intricately associated with immune response, and cell functions. Cancer cells hijack normal function and response of immune cell and direct the signals in their own favor. In recent years, immunotherapy has shown emerging interest and challenges for treating cancer patients. The advancement of cancer immunotherapy has considerably changed the paradigm of cancer therapy. Immunotherapy aims to restore or boost the immune response that is typically subverted by cancer cells through multiple mechanisms. Therefore, immunotherapy is predicated to underly the long‐term effects of conventional or targeted therapies. Tumor induces an immunosuppressive response, which counteracts the effective response of immunotherapy. Tumor microenvironment and infiltrating immune cells generate immunosuppressive response, which restrict immunotherapy and anti‐tumor immune response. Dendritic cells (DCs) are the most potent Antigen presenting cells (APCs) for initiating immune responses. Tumor‐derived pro‐inflammatory cytokines and other factors that is, VEGF and CSF1 interfere with DCs maturation and restricting the migration to the tumor‐draining lymphoid organs and stimulate the oncogenic immune response to other immune cells for invasion and migration. 106 Thus, the modulation of immunological response favors tumor growth and drug resistance. The commonly employed immunotherapy approaches include the checkpoint inhibitors (anti‐CTLA‐4, anti‐PD‐1), monoclonal antibodies, tumor infiltrating lymphocytes and chimeric antigenic receptor (CAR‐T) influences immunogenic cell death. 107 , 108 But, still immunotherapy is also challenging as cancer emerges to develop resistance. Recent report revealed that resistance to immunotherapy also occurs as either primary or acquired resistance similar to the drug resistance mechanisms developed against conventional chemotherapeutic drugs. 109 The resistance to immunotherapy is largely governed by the tumor intrinsic (absence of antigenic proteins or antigen presentation, T‐cells instability), and tumor extrinsic factors (presence of inhibitory immune checkpoints or immunosuppressive cells, deficiency of T‐ cells). 110 Some commonly encountered pathways that prevent the immunotherapy response include the activation of MAPK and Wnt/β‐catenin pathways, abrogation/alteration of interferon‐gamma (IFN‐γ) signaling, reduced T‐cell response, and tumor antigen expression. 109

The activation of MAPK signaling results in the increased expression of VEGF and IL‐8, which restricts T cell recruitment and function. 110 Also, the stabilization of β‐catenin and subsequent activation of Wnt signaling results in reduced response to checkpoint inhibitors. The increased expression of β‐catenin negatively regulates CCL4, a chemokine protein that is known to attract the dendritic cells. 111 Continuous activation to interferon‐gamma (IFN‐γ) signaling due to consistent tumor specific T‐cell activation helps immune response escape mechanism in cancer cells apparently by inhibiting the expression of molecules associated with downstream IFN‐γ signaling. 112 Recent clinical investigation showed that resistance to anti‐CTLA4 molecule ipilimumab showed considerable mutations in Interferon‐gamma (IFN‐γ) receptors and interferon regulatory factor 1 (IRF1). 113 Another tumor intrinsic factor known as innate anti PD‐1 resistance signature (IPRES), is expressed in various types of cancer, which are irresponsive to anti‐PD‐1 therapy. 114 In addition to the tumor intrinsic mechanisms, several extra‐tumoral factors such as Treg cells, M2 macrophages, and myeloid derived suppressor cells (MDSCs) act as the extrinsic causes in determining the resistance against the cancer immunotherapy. Tregs are inhibitory cells that suppress the action of effector T cells (Teff) either through direct interaction with Teff cells or through the secretion of inhibitory cytokines (IL‐8, IL‐10, TGF‐β). Furthermore, the instances of acquired resistance to cancer immunotherapy are also reported extensively, where patients develop resistance in the later stage of therapy. Several regulating mechanisms are reported that govern the consequence of acquired drug resistance against the immunotherapy in cancer due to an altered response of antigen presenting machinery. For instance, patients responding to tumor infiltrating lymphocytes (TIL), tend to lose their sensitivity to the therapy due to the loss of a component of HLA class 1 molecules known as B2M that is required for the HLA class 1 folding and transport to the cell surface. 111 Undoubtedly, a loss of HLA class function would considerably affect the T‐cell recognition process. The underlying molecular mechanisms of intrinsic and acquired resistance to cancer immunotherapy are largely needed to explore in the near future. Further, in‐depth knowledge of the molecular mechanism of immunogenic cell death influenced immunotherapy could be beneficial for cancer therapeutics.

4. CELLULAR REPROGRAMMING AND DRUG RESISTANCE

4.1. Cancer stem cells, Epithelial to mesenchymal transition and drug resistance

In multicellular organisms, stem cells play a fundamental role in the maintenance of tissue homeostasis, and, therefore, potentiate to develop daughter cells with the self‐renewal capacity. 115 Cancer stem cells (CSCs) are the key drivers for the progression of tumor and the development of drug resistance. 116 CSCs tend to display the potential for self‐renewal, differentiation with tumorigenicity, which are the key factors for the chemotherapeutic failure that eventually leads to tumor reoccurrence and metastasis. Cancer stem cells are also known as tumor initiating cells (TCIs), which reside in the specific microenvironment termed as “niche” which is filled with mesenchymal, endothelial, and immune cells. These are the neighboring cells of the CSCs and endorse the signaling pathways which are essential for the maintenance and survival of CSCs. These surrounding molecules are major causative factors for the development of endogenous drug resistance in CSCs. 117 Indeed, a recent report suggests the presence of a few subpopulation of quiescent, highly tumorigenic and pluripotent cancer stem cells (CSCs) along with population of stem cells. The “cancer stem cell theory” advocates that the sensitivity of chemotherapy relies upon several intrinsic and extrinsic resistance properties of CSCs malignant progenitors, which promote drug relapse. 36 An alteration in the Wnt/β‐catenin, Notch and Hedgehog, are the characteristics feature of cancer stem cells. A previous report advocated that the upregulation of Wnt/β‐catenin signaling results in the dedifferentiation of tumor cells and also generates fundic gland polyps in gastric epithelial cells. 118 Moreover, Wnt/β‐catenin also promotes the expression of stemness markers (ALDH and CD44) and drug resistant marker (ABCC4 and ABCG2). Collectively, these will result in the development of cisplatin induced resistance in head and neck squamous cell carcinoma. 119 Further, upregulation of the Hedgehog pathway favors the self‐renewal tendency of CSCs by regulating the expression of multiple genes such as SOX2, BMI1 and OCT4 in glioma, lung squamous cell carcinoma, breast cancer and colon cancer. 120 , 121 , 122 An alteration in Notch signaling pathways also regulate the expression of TWIST and SLUG in oral squamous cell carcinoma and trigger the self‐renewal tendency in breast CSCs. 123 Dysregulation of these signaling pathways of stem cells promotes EMT (Epithelial to mesenchymal transition) in cancer cells. Interestingly, cancer cells attain CSCs phenotype by EMT, where epithelial cells lose their polarity, change the morphology, become elongated and inhibit the cadherin, which ultimately results in the upregulation of N‐cadherin and downregulation of E‐cadherin. 124 EMT has an essential role in the developmental process of the neural tube and mesoderm formation as well as in the process of wound healing which favors the tumor growth and reoccurrence. 125 EMT regulates several transcriptional factors which are also common in CSCs such as SLUG, SNAIL, TWIST, ZEB1/2, HIFs, Notch, Wnt/β‐catenin, Hedgehog, Wnt/β‐catenin and signaling pathway of TGF‐ β. A previous report suggests that, NFκB/Twist signal axis induces EMT and increases mesenchymal properties of cancer cells upon stimulation of TNF α. 126 These cells will acquire the ability of mammosphere formation and increases the subpopulation of CD44high/CD24low, which is widely known as CSCs. 127 Moreover, in the case of prostate cancer, it was observed that the cells with EMT phenotype also upregulated the expression of prostate CSC markers such as NANOG, LIN28B, SOX2, NOTCH1, and OCT4. 128 Apart from these signaling pathways, telomerase reactivation also contributes to favor self‐renewal capacity of tumor cells to promote CSCs. In normal physiology, telomere shorting leads to chromosomal instability and fusion collectively known as DNA damage response (DDR), which ultimately results in cellular senescence. In the case of cancer, instead of shortening, an expansion in the terminal repeats at the 3' end of telomerase by an alternative lengthening of the telomeres (ALT) pathway, extend the long term self‐renewal capacity of CSCs. 129 In many drug resistant tumors, an activated telomerase were found which made tumors more difficult to treat and invasive. 130 CSCs population found to overexpress CD133+ which develops resistance towards chemotherapy and radiation in number of cancers including colon cancer, glioblastoma and non‐small cell lung cancer. 131 Moreover, CSCs tend to skip cell senescence and apoptosis that eventually lead to the development of resistance against multiple drugs, specifically docetaxel, cisplatin, cetuximab, and paclitaxel.

Moreover, mesenchymal stromal cells are also one of the major factors responsible for chemotherapeutic drug resistance in the number of cancers. MSCs are elongated spindle shaped adherent cells, which can be isolated from various types of tissue origins such as adipose and bone marrow. MSCs are multipotent, which differentiate into various types of cells. Cell expressing CD105+, CD73+, CD90+, and negative to CD45−, CD34− CD14−, CD19−, CD3−, HLA DR− are considered as MSCs. Accumulating evidence suggests that MSCs are able to stimulate tumor growth and promote chemoresistance through direct interactions with tumor cells. 132 Moreover, MSCs can release various factors including cytokines, growth factors, exosomes, and fatty acids which promote metastasis and drug resistance in cancer. 133 It has been noticed that IL‐6 and IL‐8 secreted by MSCs, protect cancer cells against platinum‐based chemotherapeutics. 134 Additionally, it was found that MSCs secret polyunsaturated fatty acids (PUFAs) such as 12‐oxo‐5,8,10‐heptadecatrienoic acid (KHT) and hexadeca‐4,7,10,13‐tetraenoic acid (16:4(n‐3)) in response to platinum‐based chemotherapy that may be responsible for drug resistance to platinum‐based therapies in colon cancer, lung cancer, and breast cancer. 135 Similarly, MSCs secreted CXCL1 and IL‐8 induce the doxorubicin resistance in triple negative breast cancer (TNBC) through the up‐regulation of ABCG2 which also known as breast cancer resistance protein (BCRP). 136 , 137 Moreover, NO produced by TA‐MSCs, and elevated release of IL‐1beta by the tumor cells shown to reduce the sensitivity of etoposide in pancreatic tumor cells. 138 Therefore, it could be considered that EMT, CSCs and MSCs may contribute in the development of multi drug resistance in cancer cells.

4.2. Cancer associated fibroblasts (CAF) and drug resistance

Cancer‐associated fibroblasts (CAFs) are a critical component of the TME. It has diverse functions including tissue remolding, matrix deposition, interactions with immune cells and intensive cross‐talk with cancer cells. 139 CAFs tend to show phenotypic and functional heterogeneity, based on their source and the type of stimulation. Apart from playing crucial roles in the tumor development, CAFs are also responsible for the development of MDR during anti‐cancer therapy. 140 Upon receiving the stimulation from tissue derived factors such as fibroblast growth factors monocyte chemotactic protein 1 (MCP1), platelet‐derived growth factor (PDGF), tissue inhibitor of metalloproteinase 1 (TIMP‐1) and tumor transforming growth factor β (TGF‐β), normal fibroblasts transform into cancer associated fibroblasts (CAFs) and exert their role in pathological consequences. The involvement of CAF in drug resistant is emerging evidence where it was noticed that the inhibition of CAF reversed the drug resistance and improved the therapeutic efficacy. Recent report suggested that the administration of 5‐FU (as a metronomic agent) in combination with taxol attenuated the tumor growth by overcoming drug resistance through the downregulation of P‐gp and simultaneous targeting of CAF. 141 Moreover, an emerging report suggests that upon the treatment of conditioned medium filled with breast cancer associated fibroblast, the human triple negative breast cancer cells (MDA‐MB‐231) attain the resistance against doxorubicin due to the release of HMGB1 that led to sustained autophagy in treated cells. 142 Taken together, the wide range of approaches governed by different CAFs to cause drug resistance in cancer cells (along with the transporter proteins) require further attention to overcome the chemo resistance.

4.3. Tumor microenvironment (TME) and drug resistance

The interaction between the drug resistant cells and TME remarkably modulates the efficacy of efflux pumps and other ECM components. 143 The acidic pH of TME considerably depletes the uptake of drug that is weakly acidic through “ion trapping,” a phenomenon that is generally observed in case of therapeutic agents carrying large permeability differences between their ionized and unionized form. Concurrently, a low acidic pH of the TME also promotes the efflux of drugs through P‐gp. A long term exposure to acidic pH also promotes the expression of proteins responsible for resistance, such as heat shock protein 27 (HSP27). 144 In addition, the different components of ECM in TME are also known to generate chemoresistance. For example, Type I collagen (a ECM constituent) is implicated in the development of chemoresistance to oxaliplatin. Moreover, the stiffening of tumor stroma is an important contributor to epithelial to mesenchymal transition (EMT) and resistance to paclitaxel. Further, the stromal cells encourage drug resistance in the surrounding cells through a process of cell‐cell interaction termed as “trogocytosis” by utilizing the integrin receptors present on cancer cells and their binding with the ligands generated by the stromal cells. Ultimately, the receptor‐ ligand interaction activates the intracellular pathways including mTOR, NF‐κB, AKT, and STAT3 signaling to sustain the mechanisms of drug resistance. 144 , 145 Recent report advocate that matrix cells in the TME exchange the communication network with cancer cells through exosomes, which play critical roles in evasion and metastasis. 146 Exosomes are tiny bilayered molecules secreted by both cancer cells and several stromal cells in the TME which participate in endocrine, paracrine, and autocrine signaling. Exosomes potentiate to convey the resistant trait to recipient cells. 147 A study suggests that exosomes mediated transfer of various non‐coding RNAs (ncRNAs), including long non coding RNAs (lncRNAs) and microRNAs (miRNAs) is a possible mechanism for procuring drug resistance in cancer cells by inducing genetic and epigenetic mutations. A previous report revealed that drug‐sensitive cells produce less extracellular microvesicles such as exosomes and microvesicles compared to drug‐resistant cancer cells and exosomal proteins can be used as a biomarker in cancer diagnostics. 148 It has been found that exosomal transfer of lncRNA‐ROR and urothelial carcinoma‐associated 1 (UCA1) induce chemoresistance in Hepatocellular carcinoma and ER‐positive MCF‐7 cells respectively. 149 , 150 It was reported that exosomes secreted by HER2‐overexpressing tumor cell lines SKBR3 and BT474 can express full‐length human epidermal growth factor 2 molecules (EGF‐2) and manifest MDR effect by hampering the activity of Trastuzumab in breast cancer. 151 In addition, circulating exosome‐associated miRNAs were found to be responsible for bortezomib resistance in multiple myeloma. 152 Moreover, tumor associated mesenchymal stromal cells (TA‐MSCs) derived exosomes also promote drug resistance. 153 These available reports strongly suggest that exosomes secreted from tumor stromal cells, confer drug resistance against anti‐cancer therapy.

4.4. Oxidative stress and drug resistance

Oxidative stress is the result of an imbalance between the generation of free oxygen radicals and their elimination through the antioxidant defense system. It is generally produced by disruption of the respiratory chain and aberrant mitochondrial function leading to the generation of ROS. 154 ROS provoke DNA mutation, genome instability, and cell proliferation, which required initially for tumor development and progression. 155 However, excess ROS also induce apoptosis and ceases the tumor progression. 156 Therefore, ROS play a dual role in the development and treatment of cancer through regulating several transcription factors such as NF‐κB, AP‐1, p53, HIF‐1α, PPAR‐γ, Wnt/β‐catenin, & Nrf2 and enzymes such as superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase, catalase, and nonenzymatic antioxidants including glutathione (GSH), vitamins C and D. Further, several reports suggest that anticancer drugs such as cisplatin, doxorubicin, vincristine and vinblastine exert their anticancer efficacy through the generation of ROS. 156 , 157 , 158 However, prolonged use of these drugs also promotes resistance through the reduction in ROS production. 159 Generally, ROS trigger mitochondrial dysfunction and apoptosis, but tumor cells have several survival mechanisms acquired by genetic alterations that promote tumor survival. Further, ROS also lead to mutagenesis through promoting instability of genetic material and continuous mutations, which results in tumor heterogeneity. 160 Next, chronic hypoxia is an indispensable need of a cancer cell. The perpetuated hypoxic conditions in transformed cells encourage the expression of different oncogenes to ensure cancer proliferation. Hypoxic conditions favor chemoresistance in different types of cancer. 161 Anticancer drugs that rely on excessive ROS production to cause DNA damage and subsequent cell death depends upon the presence of oxygen inside the cells to generate sufficient ROS. The limited supply of oxygen, therefore, enables the cancer cells to escape from death by maintaining the ROS concentration below the lethal threshold. 161 Under such conditions, the failure of anticancer drugs that exert their anticancer effect through free radical generation is undoubtedly perceivable. Cells employ several other alternative pathways to restrict their undesired proliferation. Cell senescence is a stable form of cell cycle arrest that is activated under stressful conditions in a cellular environment. 162 The induction of senescence is a mechanism employed by various anticancer agents to halt the cancer progression. 161 Importantly, the prevalence of hypoxia in cancer cells substantially limits the drug induced senescence in cancer cells, thus, apparently developing resistance against such interventions. 163 The transcription of multidrug resistance genes in response to hypoxia is a widely acknowledged phenomenon that occurs in cancer cells. The hypoxia mediated activation of HIF1α subunit of HIF acts as a transcription factor that initiates the expression of drug resistance genes including, MDR‐1 and BCRP. 161 Cancer cells also manage to evade drug action by sustaining autophagy. Autophagy induction in cancer cells has been observed as a mechanism of resistance against multiple anticancer drugs. 57 Importantly, the HIF1 axis considerably regulates the autophagy mediated drug resistance. 161 Moreover, the paradoxical role of anti‐oxidant defense system in the perpetuation of MDR gene (P‐gp) expression was also inferable from the study, where the selective upregulation of P‐gp expression in HEP‐G2 cells upon treatment with anti‐oxidant enzyme catalase was regulated through the JNK signaling pathway. A possible mechanism has been proposed in such a case, where activation of JNK signaling was suggested as a result of reduced intracellular ROS concentration. 164 However, JNK dependent activation of P‐gp is also advocated to be independent of ROS levels. Specifically, the hypoxia mediated activation of JNK is observed to be independent of concurrently increased ROS levels in cancer cells. 165 In addition, upregulated expression of P‐gp was found in colorectal cancer cells and suggested that COX‐2 mediated activation of P‐gp expression was associated with JNK dependent pathway. 166 Moreover, a previous report advocate that activation of MDR gene (P‐gp) is associated with other kinases including the cAMP dependent protein kinase, Protein kinase C and P13K. 26 Therefore, impertinent of ROS and redox signaling in context to multidrug resistance in cancer is remain elusive and needed further investigations to explore the mechanism of drug resistance in cancer.

4.5. Cancer metabolism and drug resistance

Metabolic reprogramming is one of the key features and hallmarks of cancer cells. Cancer cells adapted TME through chronically elevated oxidative stress and metabolic reprogramming to ascertain its energy demands. Almost a century ago, Otto Warburg made a revolutionary remark on cancer and explained reprogrammed metabolism in cancer. The bizarre behavior of cancer cells allows them to shift toward the less energy efficient aerobic glycolytic pathway and sidelining an energy efficient oxidative phosphorylation pathway for its energy production. The phenomenon is popularly known as the “Warburg effect,” which still remains a large enigma in cancer biology. However, today, this reprogrammed metabolism is not only constrained to glucose metabolism but also extended to lipid and glutamine metabolism. A recent understanding of cancer cell metabolism has brought the knowledge in the way that cancer cells generate oxidative stress and TME to ensure continuous synthesis of amino acids and proteins even in the presence of a chronically low level of ATP inside the cells. 167 Compiling reports suggested the prominent roles of lactate, produced as a result of aerobic glycolysis, in cancer progression. In line with these findings, it can be inferred that lactate is a deliberately produced product of cancer cells and is one of the reasons behind the shifting of cell metabolism toward the glycolytic pathway. 168 , 169 Pyruvate kinase isoform 2 (PKM2) is a glycolytic enzyme, that is commonly upregulated in many human cancers. PKM2 functions to regulate the glycolytic flux and hinders oxidative phosphorylation in cancer cells. PKM2 has been found to play a critical role in gene transcription and cell cycle progression along with metabolism reprogramming. The number of non‐metabolic roles of PKM2 has been reported including the regulation of programmed cell death and drug resistance in cancer cells. 170 Further, 2‐Deoxyglucose (2‐DG) is a glycolytic inhibitor that regulates various signaling pathways. Normal cells and tissues during radio‐ and chemo‐sensitization of the tumor were found to be protected by 2‐deoxy‐D‐glucose. 171 It was well evident that many oncogenes, tumor suppressor genes and proteins influence signal transduction pathways and metabolism, that is, HIF1, MYC, p53, and Bcl2 family proteins. The interplay between drug resistant genes such as MDR1 (P‐gp), MRP1 & BCRP and tumor metabolism genes such as HIF1‐α, LDHA, HK II, and c‐Myc has been shown in the tumor progression. 172 A Study by Wartenberg et al, has shown that P‐gp expression was downregulated with the inhibition of glycolysis. Inhibition of glycolysis also reduces the production of ATP, which is required for the P‐gp‐ATPase activity. The possible underlying mechanisms include decreased expression of HIF1‐α regulated glycolytic enzymes such as LDHA, PDHA1, and HK1. 173 Altered tumor metabolism and upregulated expression of P‐ gp have revealed many secrets of drug metabolism and chemoresistance. Moreover, the development of hypoxia in cancer cells plays a critical role in altered tumor metabolism and acidic microenvironment, which has been attributed to the induction of P‐gp expression. 174 Reprogramming of cancer cell metabolism promotes drug resistance that attributes major obstacles in cancer therapy. 175 Therefore, additional studies are required to investigate the cross‐talk between cancer metabolism and chemo resistant to overcome the drug resistance in cancer.

4.5.1. Reverse pH gradient

Tumor acidosis has been recently recognized as one of the emerging hallmarks of cancer. 176 It is the outcome of an accumulation of metabolic acids due to the high rate of metabolic demands by cancer cells. Lactic acid and carbonic acid have been noticed as one of the major driving forces for the creation of acidic TME. 177 , 178 Cancer cells extrude these harmful metabolic acids to the extracellular environment in order to protect themselves from intracellular acidification‐induced apoptosis and thus causes reverse pH gradient, that is, lower pH (pHe 5.6 ‐ 6.8) of the extracellular region and higher pH (pHi 7.2‐7.5) of the intracellular region. 177 , 179 Reverse pH gradient provides several benefits to the cancer cells and helps in exaggerating proliferation, inhibition of apoptosis, and invasion and metastasis. 178 , 180 Further, reverse pH gradient has also been reported in providing the chemoresistance property to the cancerous cells by elevating the expression of multidrug resistance proteins, affecting distribution and uptake of chemotherapeutic drugs. 180 Previous reports demonstrated the crucial role of reverse pH gradient in the uptake of weakly acidic or weakly basic chemotherapeutic drugs by the tumor cells due to their protonated or unprotonated forms. 181 , 182 Indeed lower extracellular pH influences reduced cytotoxic response of many anti‐cancer drugs like paclitaxel, mitoxantrone, and topotecan against murine mammary carcinoma cells and human bladder carcinoma cells. 183 Further, in support, it has been reported that an increased therapeutic efficacy of a weak basic chemotherapeutic drug doxorubicin against MCF‐7 xenografts in vivo was the influence of elevating the extracellular pH maintained through sodium bicarbonate‐supplemented water orally. 182 Later on the implication of acidosis in the promotion of chemoresistance has been shown due to increased p‐glycoprotein (P‐gp) activity. Authors have demonstrated that in vitro and in vivo extracellular acidification (pH 6.6) caused daunorubicin resistance in rat prostate cancer via increasing the P‐gp activity through activation of p38. 184 In addition, recent report has shown that several anticancer drugs such as doxorubicin, vincristine, and vinblastine have lower efficacy in the acidic extracellular environment due to protonation as these drugs are mildly basic in nature. 144 It has been assumed that the acidic environment promotes the expression of P‐gp and efflux of drugs. A previous report revealed that expression of P gp increased linearly with a decrease in pH of TME through activation of p38/MAPK pathway. 185 Interestingly, current study suggests that alkaline intracellular pH of tumor cells not only inhibits the accumulation of chemotherapeutic drugs (such as weakly basic chemotherapeutic drugs) but also interferes in the binding of their targets such as tubulin and DNA. 186 However, these effects were reversed upon acidic shifts. 186 Moreover, alkaline pHi‐mediated chemoresistance also depends on the elevated expression of ABCB1 or P‐gp, which enhances the rate of efflux depending upon the protonation of drug. 187 These proteins effectively transport the neutral or positively charged drugs from the intracellular environment to extracellular milleu through binding to the transporter site of the membrane. Further, the expression of lactate dehydrogenase A (LDH‐A), an enzyme that catalyzes pyruvate to lactate and supports the acidification of TME, has been reported in the development of chemoresistance in several cancers including breast cancer, and colon cancer. 188 , 189 It has been shown that inhibition of LDHA by siRNA and its inhibitor, oxamate, promoted the sensitivity of taxol against the taxol resistant breast cancer cells 189 . Tumor acidosis is the result of the orchestrated expression of several pH regulators such as NHE1, CAIX, CAXII and V‐ATPase. A previous report revealed that expression of NHE1 has been found to be increased in several cancers such as breast, colon, glioma, and leukemia and imparts tumor acidosis. The over expression of NHE1 is associated with inhibition of apoptosis and promoting resistance to cytarabine in acute myeloid leukemia. 190 There are ample reports suggesting the implication of reverse pH gradient in the promotion of chemoresistance in cancer treatment. However, the role of pH regulators and sensors in the chemoresistance has not been explored much. Therefore, further studies are needed to decipher the cross‐talk between reverse pH gradient, pH regulators such as NHE1, CAIX, CAXII, and V‐ATPase and chemoresistant genes such as MDR1, MRP1, and BCRP in drug resistance.

4.6. Inflammation and drug resistance

Inflammation is an innate immune response of our body against harmful stimuli such as tissue injury or invading pathogens. It is a multi‐step process that initiates upon the activation of immune cells, which subsequently release pro‐inflammatory mediators and activates several inflammatory cells to exclude the pathogens or foreign cells. The perpetuation of an inflammatory milieu in TME contributes to progress tumor growth and development. 191 Inflammation plays a significant role in all the major events of tumor development including angiogenesis, evasion from cell death, cancer migration or acquiring resistance against the administered interventions. 192 , 193 Inflammation has been considered as the seventh hallmark of cancer. 171 Here, with relevance to the present discussion, we will attempt to shed the light on the connection between inflammation and MDR in cancer. Activation of immune cells and immunological response promotes secretion of pro‐inflammatory cytokines and inflammatory signaling cascades, which may promote the development of drug resistance. Briefly, at the site of malignant growth, frequent accumulation of inflammatory mediators and inflammatory cells generates the local inflammatory TME which regulates the expression of drug resistant proteins in cancer cells and significantly alters the cellular response of chemotherapeutic agents. 194 Similarly, overexpression of multidrug resistance associated protein 1 (MRP1) was observed in inflamed intestine of patients with ulcerative colitis and Crohn's disease. 195 Moreover, exposure of chemotherapeutic agent to the tumor cells also generates the inflammatory response and promotes metastasis and drug resistance. 196 , 197 Previous reports demonstrated the close link between Nuclear Factor (NF)‐kappa B (NF‐kB) activation, production of cytokines, and drug resistance in cancer. 198 , 199 The Nuclear Factor (NF)‐kappa B is the transcription factor, involved in prosurvival mechanisms by initiating inflammatory pathways. However, NF‐kB signaling pathway also gets activated by exposure of multiple chemotherapeutic agents including, paclitaxel, cisplatin, doxorubicin, and docetaxel, which subsequently leads to the development of drug resistance in tumors by growth factor receptor stimulation, PI3K/AKT pathway, MAP kinase/ERK pathway, Janus Kinase/Signal Transducers and Activators of Transcription pathway, DNA repair mechanisms, and deregulating apoptotic mechanisms. 200 , 201 NF‐kB signaling induces drug resistance in cancer cells by multiple mechanisms such as by growth factor receptor stimulation, PI3K/AKT pathway, MAP kinase/ERK pathway, Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) pathway, DNA repair mechanisms and deregulating apoptotic mechanisms. A previous study suggested that upon treatment on A549 cells, cisplatin phosphorylates EGFR and activates PI3K/AKT/NF‐κB pathway, which results in the development of cisplatin resistance in NSCLC. 202 In addition, the constitutive activation of NF‐kB, up regulate the expression of Snail (transcription factor involved in EMT) in prostate cancer. An elevated level of Snail via NF‐kB, inhibits the expression of metastasis suppressor gene Raf kinase inhibitor protein (RKIP), and protects the cancer cells against chemotherapy induced apoptosis. 203 Apart from these, NF‐kB can also be activated by Tumor necrosis factor (TNF) receptor signaling in cancer cells, which is associated with chemoresistance. For example, the exogenous addition of TNF‐α in breast cancer cells, up regulates the expression of NF‐kB which enhances the survival of cancer cells and develops resistance against ionizing radiation. 204

Moreover, inflammatory cytokines are associated with multiple physiological processes such as cell migration, 205 angiogenesis, 206 apoptosis, 207 and inflammation, which involves tumor development, tumorigenesis and metastasis. 208 Emerging evidences suggest that cancer cells and their stroma secrete the cytokines, which plays a significant role in various drug resistance mechanisms. 209 , 210 According to the report, prostate cancer cells developed resistance against the enzalutamide, which is an antagonist of androgen, due to IL‐6 mediated activation of signal transducer and activator of transcription 3 (STAT3) and its target genes. 211 Further, it was found that IL‐6 produced in an autocrine manner, induces the multidrug resistance in breast cancer cells. 212 Moreover, an elevated expression of IL‐6 and IL‐8 can also induce drug resistance against the inhibitor of Notch signaling axis in the xenograft model. 213 Autocrine motility factor (AMF) is another cytokine secreted from cancer cells involved in drug resistance in fibrosarcoma cells. The secretion of AMF in large amounts resulted in resistance to mitomycine C by degrading Apaf‐1 and caspase‐9 expression, the key proteins accountable for the execution of intrinsic apoptosis. 214 Several chemokines such as CXCR1/CXCR2, CC chemokine subfamily are associated with drug resistance in cancer. A recent report revealed that CXCR2 and CXCL8 expression level were found higher in dacarbazine induced drug resistant melanoma cells and is suggested marker of drug resistance. 215 Further, CC chemokine subfamily significantly involved in the pro‐tumorigenic functions and drug‐resistance in cancer cells. 216 Moreover, other inflammatory molecules can also fuel the drug resistance in cancer cells, such as Cyclooxygenase (COX) ‐1 and COX‐2. Cyclooxygenase (COX) isoenzymes function to mediate the synthesis of prostaglandins (PGs) from arachidonic acid. COX‐1 and COX‐2 are the most extensively studied isoforms of COX. 217 Importantly, an increased expression of COX‐2 in tumor cells is also positively associated with the ability of cancer cells to acquire drug efflux mechanisms. The report suggested that the expression of P‐gp in the breast tumor is directly related to the expression of COX‐2. 218 It was further hypothesized that the manifold increase in COX‐2 in breast tumors results in the production of prostaglandins that activates the downstream PKC/c‐Jun (JNK) signaling axis to initiate the P‐gp expression. 218 In addition, supportive evidence also suggested the remarkable contribution of COX‐2 in the activation of P‐gp expression via JNK signaling in colorectal cancer cells. 166 Seemingly, the COX‐2 facilitated expression of drug efflux proteins is not restricted only to the P‐gp transporter. COX‐2 also involved in the development of drug resistance by upregulating the expression of MRP and BCRP in cancer cells. 219 Therefore, exploring the molecular mechanism behind inflammation and cancer can be harnessed to overcome MDR in cancer.

5. CONCLUSION

Cancer cells acquire drug resistance against chemotherapeutic drugs, cause major failure of anti‐cancer therapy. The mechanisms of drug resistance including drug efflux, alterations in drug metabolism, drug inactivation and reduced cellular uptake, mutation of drug target, DNA damage repair, genomic instability epigenetic changes, evasion of programmed cell death, and alteration in cellular reprogramming including Epithelial to mesenchymal transition, cancer stem cells, TME, oxidative Stress, altered energy metabolism, compromised immune response contributes to the development of resistance against anti‐cancer therapy. Multidrug resistance (MDR) is an outcome of intricate relationship between multiple intricate pathways responsible for the inactivation of drug, cellular reprogramming and genes resposible for development of drug resistance. MDR is a major obstacle in regimens of successful cancer therapy. In the recent past, several attempts have been made to overcome the MDR in cancer but still do not meet with success. An improved understanding of the molecular mechanism of MDR and cellular reprogramming can provide a promising opportunity to combat drug resistance in cancer and intensify cancer therapy for the upcoming future. Subsequently, identification of novel anti‐cancer drug candidates and molecular targets can be harnessed to overcome multidrug resistance in cancer.

ETHICAL STATEMENT

Not Applicable.

CONFLICT OF INTEREST

Authors are not having any conflict of interest.

AUTHORS' CONTRIBUTIONS

All authors had full access to the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Conceptualization, F.V. and C.P.; Software, V.M.; Validation, F.V., C.P. and A.K.; Investigation, C.P.; Formal Analysis, F.V. and C.P.; Resources, Data curation, F.V., A.S.C., C.P.; Writing—Original Draft, F.V., A.S.C., V.K.G., S.G.R., A.K.; Writing —Review & Editing, C.P.; Visualization, F.V. and C.P.; Supervision, C.P.; Project Administration, C.P.; Funding Acquisition, C.P.

ACKNOWLEDGMENT

SERB, Department of Science & Technology, New Delhi, Government of India is acknowledged for providing research grant (EMR/2016/002574) to Chandramani Pathak and Indian Council of Medical Research, New Delhi for providing Senior Research Fellowship award to Foram U. Vaidya. Indian Institute of Advanced Research, Gandhinagar, Puri foundation for education in India is gratefully acknowledged for providing infrastructure and support.

Vaidya FU, Sufiyan Chhipa A, Mishra V, et al. Molecular and cellular paradigms of multidrug resistance in cancer. Cancer Reports. 2022;5(12):e1291. 10.1002/cnr2.1291

Funding information Science and Engineering Research Board, Grant/Award Number: EMR/2016/002574 dated 23/02/2017

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

REFERENCES

  • 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA: Cancer J. Clin. 2019;69(1):7‐34. [DOI] [PubMed] [Google Scholar]
  • 2. Beheshti M. Different strategies to overcome multidrug resistance in cancer Research review. Int Pharm Acta. 2018;1(1):88‐89. [Google Scholar]
  • 3. Batrakova EV, Kabanov AV. Pluronic block copolymers: evolution of drug delivery concept from inert nanocarriers to biological response modifiers. J. Control Release. 2008;130(2):98‐106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Maeda H, Khatami M. Analyses of repeated failures in cancer therapy for solid tumors: poor tumor‐selective drug delivery, low therapeutic efficacy and unsustainable costs. Clin Transl Med. 2018;7(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019;2:141‐160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Mansoori B, Mohammadi A, Davudian S, Shirjang S, Baradaran B. The different mechanisms of cancer drug resistance: a brief review. Adv Pharm Bull. 2017;7(3):339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Vaidya FU, Chhipa AS, Sagar N, Pathak C. Oxidative stress and inflammation can fuel cancer. In: Maurya P, Dua K (Eds.), Role of Oxidative Stress in Pathophysiology of Diseases. Singapore: Springer; 2020:229‐258. [Google Scholar]
  • 8. Liu Y, Li Q, Zhou L, et al. Cancer drug resistance: redox resetting renders a way. Oncotarget. 2016;7(27):42740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Gottesman MM, Lavi O, Hall MD, Gillet J‐P. Toward a better understanding of the complexity of cancer drug resistance. Annu Rev Pharmacol Toxicol. 2016;56:85‐102. [DOI] [PubMed] [Google Scholar]
  • 10. Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med. 2002;53(1):615‐627. [DOI] [PubMed] [Google Scholar]
  • 11. Kathawala RJ, Gupta P, Ashby CR Jr, Chen Z‐S. The modulation of ABC transporter‐mediated multidrug resistance in cancer: a review of the past decade. Drug Resist Updat. 2015;18:1‐17. [DOI] [PubMed] [Google Scholar]
  • 12. Wilkens S. Structure and mechanism of ABC transporters. F1000prime Rep. 2015;7:14‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Dean M, Hamon Y, Chimini G. The human ATP‐binding cassette (ABC) transporter superfamily. J Lipid Res. 2001;42(7):1007‐1017. [PubMed] [Google Scholar]
  • 14. Amawi H, Sim H‐M, Tiwari AK, Ambudkar SV, Shukla S. ABC transporter‐mediated multidrug‐resistant cancer. In: Liu X, Pan G (Eds.), Drug Transporters in Drug Disposition, Effects and Toxicity. 1141. Singapore: Springer; 2019:549‐580. [DOI] [PubMed] [Google Scholar]
  • 15. Xue X, Liang X‐J. Overcoming drug efflux‐based multidrug resistance in cancer with nanotechnology. Chin J Cancer. 2012;31(2):100‐109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Lu JF, Pokharel D, Bebawy M. MRP1 and its role in anticancer drug resistance. Drug Metab Rev. 2015;47(4):406‐419. [DOI] [PubMed] [Google Scholar]
  • 17. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 2017;45(D1):D158‐d169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Liu W, Xie Y, Ma J, et al. IBS: an illustrator for the presentation and visualization of biological sequences. Bioinformatics. 2015;31(20):3359‐3361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Wang X, Li Y, Qian Y, et al. Extracellular ATP, as an energy and phosphorylating molecule, induces different types of drug resistances in cancer cells through ATP internalization and intracellular ATP level increase. Oncotarget. 2017;8(50):87860‐87877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Schneider V, Krieger ML, Bendas G, Jaehde U, Kalayda GV. Contribution of intracellular ATP to cisplatin resistance of tumor cells. J Biol Inorg Chem. 2013;18(2):165‐174. [DOI] [PubMed] [Google Scholar]
  • 21. Qian Y, Wang X, Liu Y, et al. Extracellular ATP is internalized by macropinocytosis and induces intracellular ATP increase and drug resistance in cancer cells. Cancer Lett. 2014;351(2):242‐251. [DOI] [PubMed] [Google Scholar]
  • 22. Yin Y, Li W, Deng M, et al. Extracellular high mobility group box chromosomal protein 1 promotes drug resistance by increasing the expression of P‐glycoprotein expression in gastric adenocarcinoma cells. Mol Med Rep. 2014;9(4):1439‐1443. [DOI] [PubMed] [Google Scholar]
  • 23. Ambudkar SV, Dey S, Hrycyna CA, Ramachandra M, Pastan I, Gottesman MM. Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol. 1999;39(1):361‐398. [DOI] [PubMed] [Google Scholar]
  • 24. Ward AB, Szewczyk P, Grimard V, et al. Structures of P‐glycoprotein reveal its conformational flexibility and an epitope on the nucleotide‐binding domain. Proc Nal Acad Sci USA. 2013;110(33):13386‐13391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Cascorbi I. P‐glycoprotein: tissue distribution, substrates, and functional consequences of genetic variations. In: Formm M, Kim R. (Eds.), Drug Transporters. 201. Berlin, Heidelberg: Springer; 2011:261‐283. [DOI] [PubMed] [Google Scholar]
  • 26. Sui H, Fan Z, Li Q. Signal transduction pathways and transcriptional mechanisms of ABCB1/Pgp‐mediated multiple drug resistance in human cancer cells. J Int Med Res. 2012;40(2):426‐435. [DOI] [PubMed] [Google Scholar]
  • 27. Szakács G, Paterson JK, Ludwig JA, Booth‐Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5(3):219‐234. [DOI] [PubMed] [Google Scholar]
  • 28. Callaghan R, Luk F, Bebawy M. Inhibition of the multidrug resistance P‐glycoprotein: time for a change of strategy? Drug Metab Dispos. 2014;42(4):623‐631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Zhang J, Zhou F, Wu X, et al. Cellular pharmacokinetic mechanisms of adriamycin resistance and its modulation by 20(S)‐ginsenoside Rh2 in MCF‐7/Adr cells. Br J Pharmacol. 2012;165(1):120‐134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Ozben T. Mechanisms and strategies to overcome multiple drug resistance in cancer. FEBS Lett. 2006;580(12):2903‐2909. [DOI] [PubMed] [Google Scholar]
  • 31. Sodani K, Patel A, Kathawala RJ, Chen Z‐S. Multidrug resistance associated proteins in multidrug resistance. Chin J Cancer. 2012;31(2):58‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kruh GD, Belinsky MG. The MRP family of drug efflux pumps. Oncogene. 2003;22(47):7537‐7552. [DOI] [PubMed] [Google Scholar]
  • 33. Stavrovskaya A. Cellular mechanisms of multidrug resistance of tumor cells. Biochemistry. 2000;65(1):95‐106. [PubMed] [Google Scholar]
  • 34. Lv Y, Zhao S, Han J, Zheng L, Yang Z, Zhao L. Hypoxia‐inducible factor‐1α induces multidrug resistance protein in colon cancer. OncoTargets Ther. 2015;8:1941‐1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Staud F, Pavek P. Breast cancer resistance protein (BCRP/ABCG2). Int J Biochem Cell Biol. 2005;37(4):720‐725. [DOI] [PubMed] [Google Scholar]
  • 36. McIntosh K, Balch C, Tiwari AK. Tackling multidrug resistance mediated by efflux transporters in tumor‐initiating cells. Expert Opin Drug Metab Toxicol. 2016;12(6):633‐644. [DOI] [PubMed] [Google Scholar]
  • 37. Nakanishi T, Ross DD. Breast cancer resistance protein (BCRP/ABCG2): its role in multidrug resistance and regulation of its gene expression. Chin J Cancer. 2012;31(2):73‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Mao Q, Unadkat JD. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport—an update. AAPS J. 2015;17(1):65‐82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Horsey AJ, Cox MH, Sarwat S, Kerr ID. The multidrug transporter ABCG2: still more questions than answers. Biochem Soc Trans. 2016;44(3):824‐830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Sharom FJ. ABC multidrug transporters: structure, function and role in chemoresistance. Pharmacogenomics. 2008;9(1):105‐127. [DOI] [PubMed] [Google Scholar]
  • 41. Stacy AE, Jansson PJ, Richardson DR. Molecular pharmacology of ABCG2 and its role in chemoresistance. Mol Pharmacol. 2013;84(5):655‐669. [DOI] [PubMed] [Google Scholar]
  • 42. Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein‐protein association networks with increased coverage, supporting functional discovery in genome‐wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607‐d613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Mattingly CJ, Rosenstein MC, Colby GT, Forrest JN Jr, Boyer JL. The Comparative Toxicogenomics Database (CTD): a resource for comparative toxicological studies. J Exp Zool A Comp Exp Biol. 2006;305(9):689‐692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Michael M, Doherty MM. Tumoral drug metabolism: overview and its implications for cancer therapy. J Clin Oncol. 2005;23(1):205‐229. [DOI] [PubMed] [Google Scholar]
  • 45. Higgins MJ, Rae JM, Flockhart DA, Hayes DF, Stearns V. Pharmacogenetics of tamoxifen: who should undergo CYP2D6 genetic testing? J Natl Compr Canc Netw. 2009;7(2):203‐213. [DOI] [PubMed] [Google Scholar]
  • 46. Ketteler J, Klein D. Caveolin‐1, cancer and therapy resistance. Int J Cancer. 2018;143(9):2092‐2104. [DOI] [PubMed] [Google Scholar]
  • 47. Estrela JM, Ortega A, Obrador E. Glutathione in cancer biology and therapy. Crit Rev Clin Lab Sci. 2006;43(2):143‐181. [DOI] [PubMed] [Google Scholar]
  • 48. Godwin AK, Meister A, O'Dwyer PJ, Huang CS, Hamilton TC, Anderson ME. High resistance to cisplatin in human ovarian cancer cell lines is associated with marked increase of glutathione synthesis. Proc Natl Acad Sci USA. 1992;89(7):3070‐3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Lewis AD, Hayes JD, Wolf CR. Glutathione and glutathione‐dependent enzymes in ovarian adenocarcinoma cell lines derived from a patient before and after the onset of drug resistance: intrinsic differences and cell cycle effects. Carcinogenesis. 1988;9(7):1283‐1287. [DOI] [PubMed] [Google Scholar]
  • 50. Hochwald SN, Rose DM, Brennan MF, Burt ME. Elevation of glutathione and related enzyme activities in high‐grade and metastatic extremity soft tissue sarcoma. Ann Surg Oncol. 1997;4(4):303‐309. [DOI] [PubMed] [Google Scholar]
  • 51. Dhakshinamoorthy S, Jaiswal AK. Small maf (MafG and MafK) proteins negatively regulate antioxidant response element‐mediated expression and antioxidant induction of the NAD(P)H:quinone oxidoreductase1 gene. J Biol Chem. 2000;275(51):40134‐40141. [DOI] [PubMed] [Google Scholar]
  • 52. Rushworth SA, MacEwan DJ, O'Connell MA. Lipopolysaccharide‐induced expression of NAD(P)H: quinone oxidoreductase 1 and heme oxygenase‐1 protects against excessive inflammatory responses in human monocytes. J Immunol. 2008;181(10):6730‐6737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Shibata T, Kokubu A, Gotoh M, et al. Genetic alteration of Keap1 confers constitutive Nrf2 activation and resistance to chemotherapy in gallbladder cancer. Gastroenterology. 2008;135(4):1358‐1368. e4. [DOI] [PubMed] [Google Scholar]
  • 54. Tannock IF, Lee CM, Tunggal JK, Cowan DSM, Egorin MJ. Limited penetration of anticancer drugs through tumor tissue: a potential cause of resistance of solid tumors to chemotherapy. Clin Cancer Res. 2002;8(3):878‐884. [PubMed] [Google Scholar]
  • 55. Alfarouk KO, Stock C‐M, Taylor S, et al. Resistance to cancer chemotherapy: failure in drug response from ADME to P‐gp. Cancer Cell Int. 2015;15(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Ferguson LR, Chen H, Collins AR, et al. Genomic instability in human cancer: Molecular insights and opportunities for therapeutic attack and prevention through diet and nutrition. Semin Cancer Biol. 2015;35:S5‐S24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Li Y‐J, Lei Y‐H, Yao N, et al. Autophagy and multidrug resistance in cancer. Chin J Cancer. 2017;36(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. McGranahan N, Burrell RA, Endesfelder D, Novelli M, Swanton C. Cancer chromosomal instability: therapeutic and diagnostic challenges: ‘Exploring aneuploidy: the significance of chromosomal imbalance’ review series. EMBO Rep. 2012;13(6):528‐538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Luqmani Y. Mechanisms of drug resistance in cancer chemotherapy. Med Princ Pract. 2005;14(Suppl 1):35‐48. [DOI] [PubMed] [Google Scholar]
  • 60. Quintás‐Cardama A, Kantarjian HM, Cortes JE. Mechanisms of primary and secondary resistance to imatinib in chronic myeloid leukemia. Cancer Control. 2009;16(2):122‐131. [DOI] [PubMed] [Google Scholar]
  • 61. Hientz K, Mohr A, Bhakta‐Guha D, Efferth T. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget. 2017;8(5):8921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Marin JJ, Romero MR, Martinez‐Becerra P, Herraez E, Briz O. Overview of the molecular bases of resistance to chemotherapy in liver and gastrointestinal tumours. Curr Mol Med. 2009;9(9):1108‐1129. [DOI] [PubMed] [Google Scholar]
  • 63. Tung MC, Lin PL, Wang YC, et al. Mutant p53 confers chemoresistance in non‐small cell lung cancer by upregulating Nrf2. Oncotarget. 2015;6(39):41692‐41705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Fiorini C, Cordani M, Padroni C, Blandino G, Di Agostino S, Donadelli M. Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta. 2015;1853(1):89‐100. [DOI] [PubMed] [Google Scholar]
  • 65. Swift LH, Golsteyn RM. Genotoxic anti‐cancer agents and their relationship to DNA damage, mitosis, and checkpoint adaptation in proliferating cancer cells. Int J Mol Sci. 2014;15(3):3403‐3431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Assaraf YG, Brozovic A, Gonçalves AC, et al. The multi‐factorial nature of clinical multidrug resistance in cancer. Drug Resist Updat. 2019;46:100645. [DOI] [PubMed] [Google Scholar]
  • 67. Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA. DNA repair pathways as targets for cancer therapy. Nat Rev Cancer. 2008;8(3):193‐204. [DOI] [PubMed] [Google Scholar]
  • 68. Lin X, Howell SB. DNA mismatch repair and p53 function are major determinants of the rate of development of cisplatin resistance. Mol Cancer Ther. 2006;5(5):1239‐1247. [DOI] [PubMed] [Google Scholar]
  • 69. De Angelis PM, Svendsrud DH, Kravik KL, Stokke T. Cellular response to 5‐fluorouracil (5‐FU) in 5‐FU‐resistant colon cancer cell lines during treatment and recovery. Mol Cancer. 2006;5(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Wilting RH, Dannenberg J‐H. Epigenetic mechanisms in tumorigenesis, tumor cell heterogeneity and drug resistance. Drug Resist Updat. 2012;15(1‐2):21‐38. [DOI] [PubMed] [Google Scholar]
  • 71. Zeller C, Brown R. Therapeutic modulation of epigenetic drivers of drug resistance in ovarian cancer. Therapeutic advances in medical oncology. 2010;2(5):319‐329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Glasspool R, Teodoridis JM, Brown R. Epigenetics as a mechanism driving polygenic clinical drug resistance. Br J Cancer. 2006;94(8):1087‐1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Teodoridis JM, Strathdee G, Brown R. Epigenetic silencing mediated by CpG island methylation: potential as a therapeutic target and as a biomarker. Drug Resist Updat. 2004;7(4‐5):267‐278. [DOI] [PubMed] [Google Scholar]
  • 74. Balch C, Ramapuram JB, Tiwari AK. The epigenomics of embryonic pathway signaling in colorectal cancer. Front Pharmacol. 2017;8:267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Shah K, Rawal RM. Genetic and epigenetic modulation of drug resistance in cancer: challenges and opportunities. Curr Drug Metab. 2019;20(14):1114‐1131. [DOI] [PubMed] [Google Scholar]
  • 76. Esteller M, Garcia‐Foncillas J, Andion E, et al. Inactivation of the DNA‐repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343(19):1350‐1354. [DOI] [PubMed] [Google Scholar]
  • 77. Behbahani GD, Khani S, Hosseini HM, Abbaszadeh‐Goudarzi K, Nazeri S. The role of exosomes contents on genetic and epigenetic alterations of recipient cancer cells. Iran J Basic Med Sci. 2016;19(10):1031. [PMC free article] [PubMed] [Google Scholar]
  • 78. Qian Z, Shen Q, Yang X, Qiu Y, Zhang W. The role of extracellular vesicles: an epigenetic view of the cancer microenvironment. BioMed Res Int. 2015;2015:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Guo Q‐r, Wang H, Yan Y‐D, et al. The role of exosomal microRNA in cancer drug resistance. Front Oncol. 2020;10:472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57‐70. [DOI] [PubMed] [Google Scholar]
  • 81. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714‐726. [DOI] [PubMed] [Google Scholar]
  • 82. Ranjan K, Pathak C. FADD regulates NF‐κB activation and promotes ubiquitination of cFLIP L to induce apoptosis. Sci Rep. 2016;6(1):1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Campbell KJ, Tait SW. Targeting BCL‐2 regulated apoptosis in cancer. Open Biol. 2018;8(5):180002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Flørenes VA, Mælandsmo GM, Forus A, Andreassen A, Myklebost O, Fodstad O. MDM2 gene amplification and transcript levels in human sarcomas: relationship to TP53 gene status. J Natl Cancer Inst. 1994;86(17):1297‐1302. [DOI] [PubMed] [Google Scholar]
  • 85. Dong HP, Kleinberg L, Silins I, et al. Death receptor expression is associated with poor response to chemotherapy and shorter survival in metastatic ovarian carcinoma. Cancer. 2008;112(1):84‐93. [DOI] [PubMed] [Google Scholar]
  • 86. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2012;2(98):1‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Chen L, Zeng Y, Zhou S‐F. Role of apoptosis in cancer resistance to chemotherapy. In: Yusuf T, (Ed.) Current Understanding of Apoptosis‐Programmed Cell Death. London, UK: IntechOpen Limited; 2018:125‐136. [Google Scholar]
  • 88. Scanlon M, Shajahan A, Wang A, Clarke R. Caveolin‐1 and survivin in drug resistance in breast cancer cells. Paper presented at: AACR Annual Meeting; April 12‐16, 2008:3207‐3207.
  • 89. Wang L, Mosel AJ, Oakley GG, Peng A. Deficient DNA damage signaling leads to chemoresistance to cisplatin in oral cancer. Mol Cancer Ther. 2012;11(11):2401‐2409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Souza PS, Madigan JP, Gillet J‐P, et al. Expression of the multidrug transporter P‐glycoprotein is inversely related to that of apoptosis‐associated endogenous TRAIL. Exp Cell Res. 2015;336(2):318‐328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Mizushima N. Autophagy: process and function. Genes Dev. 2007;21(22):2861‐2873. [DOI] [PubMed] [Google Scholar]
  • 92. Li X, Zhou Y, Li Y, et al. Autophagy: a novel mechanism of chemoresistance in cancers. Biomed Pharmacother. 2019;119:109415. [DOI] [PubMed] [Google Scholar]
  • 93. Sui X, Chen R, Wang Z, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis. 2013;4(10):e838‐e838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Xiao X, Wang W, Li Y, et al. HSP90AA1‐mediated autophagy promotes drug resistance in osteosarcoma. J Exp Clin Cancer Res. 2018;37(1):201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Wang J, Garbutt C, Ma H, et al. Expression and role of autophagy‐associated p62 (SQSTM1) in multidrug resistant ovarian cancer. Gynecol Oncol. 2018;150(1):143‐150. [DOI] [PubMed] [Google Scholar]
  • 96. Battista RA, Resnati M, Facchi C, et al. Autophagy mediates epithelial cancer chemoresistance by reducing p62/SQSTM1 accumulation. PloS One. 2018;13(8):e0201621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Pan J, Lu C, Jun W, Wu Y, Shi X, Ding Y. The up‐regulation of P62 levels is associated with resistance of sorafenib in hepatocarcinoma cells. Int J Clin Exp Pathol. 2019;12(7):2622. [PMC free article] [PubMed] [Google Scholar]
  • 98. Zhang H, McCarty N. Tampering with cancer chemoresistance by targeting the TGM2‐IL6‐autophagy regulatory network. Autophagy. 2017;13(3):627‐628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Liang DH, El‐Zein R, Dave B. Autophagy inhibition to increase radiosensitization in breast cancer. J Nucl Med Radiat Ther. 2015;6(5):1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. He Q, Li J, Dong F, Cai C, Zou X. LKB1 promotes radioresistance in esophageal cancer cells exposed to radiation, by suppression of apoptosis and activation of autophagy via the AMPK pathway. Mol Med Rep. 2017;16(2):2205‐2210. [DOI] [PubMed] [Google Scholar]
  • 101. Gong Y, Fan Z, Luo G, et al. The role of necroptosis in cancer biology and therapy. Mol Cancer. 2019;18(1):100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418(6894):191‐195. [DOI] [PubMed] [Google Scholar]
  • 103. Martinotti S, Patrone M, Ranzato E. Emerging roles for HMGB1 protein in immunity, inflammation, and cancer. ImmunoTargets Ther. 2015;4:101‐109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Inoue H, Tani K. Multimodal immunogenic cancer cell death as a consequence of anticancer cytotoxic treatments. Cell Death Differ. 2014;21(1):39‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Yang H, Tracey KJ. Targeting HMGB1 in inflammation. Biochim Biophys Acta. 2010;1799(1‐2):149‐156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Pinzon‐Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005;83(5):451‐461. [DOI] [PubMed] [Google Scholar]
  • 107. Zhang H, Chen J. Current status and future directions of cancer immunotherapy. J Cancer. 2018;9(10):1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Liu M, Guo F. Recent updates on cancer immunotherapy. Precis Clin Med. 2018;1(2):65‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Rodriguez‐Pascual J, Ayuso‐Sacido A, Belda‐Iniesta C. Drug resistance in cancer immunotherapy: new strategies to improve checkpoint inhibitor therapies. Cancer Drug Resist. 2019;2:980‐993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Sharma P, Hu‐Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168(4):707‐723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Spranger S, Bao R, Gajewski TF. Melanoma‐intrinsic β‐catenin signalling prevents anti‐tumour immunity. Nature. 2015;523(7559):231‐235. [DOI] [PubMed] [Google Scholar]
  • 112. Darnell JE, Kerr IM, Stark GR. Jak‐STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264(5164):1415‐1421. [DOI] [PubMed] [Google Scholar]
  • 113. Gao J, Shi LZ, Zhao H, et al. Loss of IFN‐γ pathway genes in tumor cells as a mechanism of resistance to anti‐CTLA‐4 therapy. Cell. 2016;167(2):397‐404. e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of response to anti‐PD‐1 therapy in metastatic melanoma. Cell. 2016;165(1):35‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861‐872. [DOI] [PubMed] [Google Scholar]
  • 116. Ayob AZ, Ramasamy TS. Cancer stem cells as key drivers of tumour progression. J Biomed Sci. 2018;25(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kise K, Kinugasa‐Katayama Y, Takakura N. Tumor microenvironment for cancer stem cells. Adv. Drug Delivery Rev. 2016;99:197‐205. [DOI] [PubMed] [Google Scholar]
  • 118. Scheel C, Eaton EN, Li SH‐J, et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell. 2011;145(6):926‐940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Warrier S, Bhuvanalakshmi G, Arfuso F, Rajan G, Millward M, Dharmarajan A. Cancer stem‐like cells from head and neck cancers are chemosensitized by the Wnt antagonist, sFRP4, by inducing apoptosis, decreasing stemness, drug resistance and epithelial to mesenchymal transition. Cancer Gene Ther. 2014;21(9):381‐388. [DOI] [PubMed] [Google Scholar]
  • 120. Hoffmann W. Self‐renewal of the gastric epithelium from stem and progenitor cells. Front Biosci (Schol Ed). 2013;5:720‐731. [DOI] [PubMed] [Google Scholar]
  • 121. Justilien V, Walsh MP, Ali SA, Thompson EA, Murray NR, Fields AP. The PRKCI and SOX2 oncogenes are coamplified and cooperate to activate Hedgehog signaling in lung squamous cell carcinoma. Cancer Cell. 2014;25(2):139‐151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Park I‐k, Qian D, Kiel M, et al. Bmi‐1 is required for maintenance of adult self‐renewing haematopoietic stem cells. Nature. 2003;423(6937):302‐305. [DOI] [PubMed] [Google Scholar]
  • 123. Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial‐to‐mesenchymal transition. Breast Cancer Res. 2011;13(3):211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Thiery JP, Sleeman JP. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131‐142. [DOI] [PubMed] [Google Scholar]
  • 125. Shook D, Keller R. Mechanisms, mechanics and function of epithelial‐mesenchymal transitions in early development. Mech Dev. 2003;120(11):1351‐1383. [DOI] [PubMed] [Google Scholar]
  • 126. Li CW, Xia W, Huo L, et al. Epithelial‐mesenchymal transition induced by TNF‐α requires NF‐κB‐mediated transcriptional upregulation of Twist1. Cancer Res. 2012;72(5):1290‐1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Eun K, Ham SW, Kim H. Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting. BMB Rep. 2017;50(3):117‐125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Kong D, Banerjee S, Ahmad A, et al. Epithelial to mesenchymal transition is mechanistically linked with stem cell signatures in prostate cancer cells. PLoS One. 2010;5(8):e12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Allsopp RC, Morin GB, DePinho R, Harley CB, Weissman IL. Telomerase is required to slow telomere shortening and extend replicative lifespan of HSCs during serial transplantation. Blood. 2003;102(2):517‐520. [DOI] [PubMed] [Google Scholar]
  • 130. Makki J, Myint O, Wynn AA, Samsudin AT, John DV. Expression distribution of cancer stem cells, epithelial to mesenchymal transition, and telomerase activity in breast cancer and their association with clinicopathologic characteristics. Clin Med Insights Pathol. 2015;8:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Bao S, Wu Q, McLendon RE, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756‐760. [DOI] [PubMed] [Google Scholar]
  • 132. Rafii A, Mirshahi P, Poupot M, et al. Oncologic trogocytosis of an original stromal cells induces chemoresistance of ovarian tumours. PLoS One. 2008;3(12):e3894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25(4):198‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Skolekova S, Matuskova M, Bohac M, et al. Cisplatin‐induced mesenchymal stromal cells‐mediated mechanism contributing to decreased antitumor effect in breast cancer cells. Cell Commun Signal. 2016;14(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Roodhart JM, Daenen LG, Stigter EC, et al. Mesenchymal stem cells induce resistance to chemotherapy through the release of platinum‐induced fatty acids. Cancer Cell. 2011;20(3):370‐383. [DOI] [PubMed] [Google Scholar]
  • 136. Yeh WL, Tsai CF, Chen DR. Peri‐foci adipose‐derived stem cells promote chemoresistance in breast cancer. Stem Cell Res Ther. 2017;8(1):177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Chen DR, Lu DY, Lin HY, Yeh W‐L. Mesenchymal stem cell‐induced doxorubicin resistance in triple negative breast cancer. BioMed Res Int. 2014;2014:532161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Muerkoster S, Wegehenkel K, Arlt A, et al. Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin‐1beta. Cancer Res. 2004;64(4):1331‐1337. [DOI] [PubMed] [Google Scholar]
  • 139. Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer‐associated fibroblasts. Nat Rev Cancer. 2020;20:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Liu T, Han C, Wang S, et al. Cancer‐associated fibroblasts: an emerging target of anti‐cancer immunotherapy. J Hematol Oncol. 2019;12(1):1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Ma Y, Wang Y, Xu Z, Wang Y, Fallon JK, Liu F. Extreme low dose of 5‐fluorouracil reverses MDR in cancer by sensitizing cancer associated fibroblasts and down‐regulating P‐gp. PLOS one. 2017;12(6):e0180023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Amornsupak K, Insawang T, Thuwajit P, O‐Charoenrat P, Eccles SA, Thuwajit C. Cancer‐associated fibroblasts induce high mobility group box 1 and contribute to resistance to doxorubicin in breast cancer cells. BMC cancer. 2014;14(1):1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Tezcan O, Ojha T, Storm G, Kiessling F, Lammers T. Targeting cellular and microenvironmental multidrug resistance. Expert Opin Drug Deliv. 2016;13(9):1199‐1202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Jo Y, Choi N, Kim K, Koo H‐J, Choi J, Kim HN. Chemoresistance of cancer cells: requirements of tumor microenvironment‐mimicking in vitro models in anti‐cancer drug development. Theranostics. 2018;8(19):5259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Qu Y, Dou B, Tan H, Feng Y, Wang N, Wang D. Tumor microenvironment‐driven non‐cell‐autonomous resistance to antineoplastic treatment. Mol Cancer. 2019;18(1):69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in cancer: small particle, big player. J Hematol Oncol. 2015;8:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Sousa D, Lima RT, Vasconcelos MH. Intercellular transfer of cancer drug resistance traits by extracellular vesicles. Trends Mol Med. 2015;21(10):595‐608. [DOI] [PubMed] [Google Scholar]
  • 148. Lopes‐Rodrigues V, Di Luca A, Sousa D, et al. Data supporting the shedding of larger extracellular vesicles by multidrug resistant tumour cells. Data Brief. 2016;6:1023‐1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Takahashi K, Yan IK, Kogure T, Haga H, Patel T. Extracellular vesicle‐mediated transfer of long non‐coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio. 2014;4:458‐467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Xu CG, Yang MF, Ren YQ, Wu C‐H, Wang L‐Q. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. Eur Rev Med Pharmacol Sci. 2016;20(20):4362‐4368. [PubMed] [Google Scholar]
  • 151. Ciravolo V, Huber V, Ghedini GC, et al. Potential role of HER2‐overexpressing exosomes in countering trastuzumab‐based therapy. J Cell Physiol. 2012;227(2):658‐667. [DOI] [PubMed] [Google Scholar]
  • 152. Zhang L, Pan L, Xiang B, et al. Potential role of exosome‐associated microRNA panels and in vivo environment to predict drug resistance for patients with multiple myeloma. Oncotarget. 2016;7(21):30876‐30891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Ono M, Kosaka N, Tominaga N, et al. Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Sci Signal. 2014;7(332):ra63. [DOI] [PubMed] [Google Scholar]
  • 154. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417(1):1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Liou GY, Storz P. Reactive oxygen species in cancer. Free Radic Res. 2010;44(5):479‐496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Yang H, Villani RM, Wang H, et al. The role of cellular reactive oxygen species in cancer chemotherapy. J Exp Clin Cancer Res. 2018;37(1):266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49(11):1603‐1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Yu W, Chen Y, Dubrulle J, et al. Cisplatin generates oxidative stress which is accompanied by rapid shifts in central carbon metabolism. Sci Rep. 2018;8(1):4306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Maiti AK. Gene network analysis of oxidative stress‐mediated drug sensitivity in resistant ovarian carcinoma cells. Pharmacogenomics J. 2010;10(2):94‐104. [DOI] [PubMed] [Google Scholar]
  • 160. Sallmyr A, Fan J, Datta K, et al. Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: implications for poor prognosis in AML. Blood. 2008;111(6):3173‐3182. [DOI] [PubMed] [Google Scholar]
  • 161. Minassian LM, Cotechini T, Huitema E, Graham CH. Hypoxia‐induced resistance to chemotherapy in cancer. In: Gilkes D, ed. Hypoxia and Cancer Metastasis. Switzerland AG: Springer Nature; 2019:123‐139. [DOI] [PubMed] [Google Scholar]
  • 162. Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin Invest. 2018;128(4):1238‐1246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Sullivan R, Paré GC, Frederiksen LJ, Semenza GL, Graham CH. Hypoxia‐induced resistance to anticancer drugs is associated with decreased senescence and requires hypoxia‐inducible factor‐1 activity. Mol Cancer Ther. 2008;7(7):1961‐1973. [DOI] [PubMed] [Google Scholar]
  • 164. Li L, Xu J, Min T, Huang W. Up‐regulation of P‐glycoprotein expression by catalase via JNK activation in HepG2 cells. Redox Rep. 2006;11(4):173‐178. [DOI] [PubMed] [Google Scholar]
  • 165. Comerford KM, Cummins EP, Taylor CT. c‐Jun NH2‐terminal kinase activation contributes to hypoxia‐inducible factor 1α–dependent P‐glycoprotein expression in hypoxia. Cancer Res. 2004;64(24):9057‐9061. [DOI] [PubMed] [Google Scholar]
  • 166. Sui H, Zhou S, Wang Y, et al. COX‐2 contributes to P‐glycoprotein‐mediated multidrug resistance via phosphorylation of c‐Jun at Ser63/73 in colorectal cancer. Carcinogenesis. 2011;32(5):667‐675. [DOI] [PubMed] [Google Scholar]
  • 167. Sreedhar A, Zhao Y. Dysregulated metabolic enzymes and metabolic reprogramming in cancer cells. Biomed Rep. 2018;8(1):3‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Goodwin ML, Pennington Z, Westbroek EM, Cottrill E, Karim Ahmed A, Sciubba DM. Lactate and cancer: a "lactatic" perspective on spinal tumor metabolism (part 1). Ann Transl Med. 2019;7(10):220‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Dhup S, Dadhich RK, Porporato PE, Sonveaux P. Multiple biological activities of lactic acid in cancer: influences on tumor growth, angiogenesis and metastasis. Curr Pharm Des. 2012;18(10):1319‐1330. [DOI] [PubMed] [Google Scholar]
  • 170. Amin S, Yang P, Li Z. Pyruvate kinase M2: a multifarious enzyme in non‐canonical localization to promote cancer progression. Biochim Biophys Acta. 2019;1871(2):331‐341. [DOI] [PubMed] [Google Scholar]
  • 171. Farooque A, Afrin F, Adhikari JS, Dwarakanath BS. Protection of normal cells and tissues during radio‐ and chemosensitization of tumors by 2‐deoxy‐D‐glucose. J Cancer Res Ther. 2009;5(Suppl 1):S32‐S35. [DOI] [PubMed] [Google Scholar]
  • 172. Housman G, Byler S, Heerboth S, et al. Drug resistance in cancer: an overview. Cancers (Basel). 2014;6(3):1769‐1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Wartenberg M, Richter M, Datchev A, et al. Glycolytic pyruvate regulates P‐Glycoprotein expression in multicellular tumor spheroids via modulation of the intracellular redox state. J Cell Biochem. 2010;109(2):434‐446. [DOI] [PubMed] [Google Scholar]
  • 174. Xie J, Li DW, Chen XW, Wang F, Dong P. Expression and significance of hypoxia‐inducible factor‐1α and MDR1/P‐glycoprotein in laryngeal carcinoma tissue and hypoxic Hep‐2 cells. Oncol Lett. 2013;6(1):232‐238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Rahman M, Hasan MR. Cancer metabolism and drug resistance. Metabolites. 2015;5(4):571‐600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Gupta VK, Jaiswara P, Kumar A. The emerging role of miRNAs in tumor acidosis. J Cell Biol Cell Metab. 2018;5:015. [Google Scholar]
  • 177. Chiche J, Brahimi‐Horn MC, Pouyssegur J. Tumour hypoxia induces a metabolic shift causing acidosis: a common feature in cancer. J Cell Mol Med. 2010;14(4):771‐794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Corbet C, Feron O. Tumour acidosis: from the passenger to the driver's seat. Nat Rev Cancer. 2017;17(10):577‐593. [DOI] [PubMed] [Google Scholar]
  • 179. Webb BA, Chimenti M, Jacobson MP, Barber DL. Dysregulated pH: a perfect storm for cancer progression. Nat Rev Cancer. 2011;11(9):671‐677. [DOI] [PubMed] [Google Scholar]
  • 180. Wojtkowiak JW, Verduzco D, Schramm KJ, Gillies RJ. Drug resistance and cellular adaptation to tumor acidic pH microenvironment. Mol Pharm. 2011;8(6):2032‐2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Gerweck LE, Seetharaman K. Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer. Cancer Res. 1996;56(6):1194‐1198. [PubMed] [Google Scholar]
  • 182. Raghunand N, He X, van Sluis R, et al. Enhancement of chemotherapy by manipulation of tumour pH. Br J Cancer. 1999;80(7):1005‐1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Vukovic V, Tannock IF. Influence of low pH on cytotoxicity of paclitaxel, mitoxantrone and topotecan. Br J Cancer. 1997;75(8):1167‐1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Sauvant C, Nowak M, Wirth C, et al. Acidosis induces multi‐drug resistance in rat prostate cancer cells (AT1) in vitro and in vivo by increasing the activity of the p‐glycoprotein via activation of p38. Int J Cancer. 2008;123(11):2532‐2542. [DOI] [PubMed] [Google Scholar]
  • 185. Thews O, Dillenburg W, Fellner M, et al. Activation of P‐glycoprotein (Pgp)‐mediated drug efflux by extracellular acidosis: in vivo imaging with 68Ga‐labelled PET tracer. Eur J Nucl Med Mol Imaging. 2010;37(10):1935‐1942. [DOI] [PubMed] [Google Scholar]
  • 186. Simon S, Roy D, Schindler M. Intracellular pH and the control of multidrug resistance. Proc Natl Acad Sci U S A. 1994;91(3):1128‐1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Roepe PD, Wei LY, Cruz J, Carlson D. Lower electrical membrane potential and altered pHi homeostasis in multidrug‐resistant (MDR) cells: further characterization of a series of MDR cell lines expressing different levels of P‐glycoprotein. Biochemistry. 1993;32(41):11042‐11056. [DOI] [PubMed] [Google Scholar]
  • 188. Li X, Zhao H, Zhou X, Song L. Inhibition of lactate dehydrogenase A by microRNA‐34a resensitizes colon cancer cells to 5‐fluorouracil. Mol Med Rep. 2015;11(1):577‐582. [DOI] [PubMed] [Google Scholar]
  • 189. Zhou M, Zhao Y, Ding Y, et al. Warburg effect in chemosensitivity: targeting lactate dehydrogenase‐A re‐sensitizes taxol‐resistant cancer cells to taxol. Mol Cancer. 2010;9:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Hyun SY, Kim YK, Jang JE, Kim Y, Kim YR, Cheong J‐W, Shim KY, Min YH. Inhibition of NHE1 Induced Apoptosis in Cytarabine Resistant Leukemia Cell Lines and Primary Leukemia Cells from AML Patients, Which Showed Increased Intracellular pH and NHE1 Activity. Blood. 2014;124(21):3614‐3614. 10.1182/blood.v124.21.3614.3614. [DOI] [Google Scholar]
  • 191. Lu H, Ouyang W, Huang C. Inflammation, a key event in cancer development. Mol Cancer Res. 2006;4(4):221‐233. [DOI] [PubMed] [Google Scholar]
  • 192. Kobayashi H, Lin PC. Angiogenesis links chronic inflammation with cancer. Methods Mol Biol. 2009;511:185‐191. [DOI] [PubMed] [Google Scholar]
  • 193. Liu J, Lin PC, Zhou BP. Inflammation fuels tumor progress and metastasis. Curr. Pharm. Des. 2015;21(21):3032‐3040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Hartmann G, Vassileva V, Piquette‐Miller M. Impact of endotoxin‐induced changes in P‐glycoprotein expression on disposition of doxorubicin in mice. Drug Metab Dispos. 2005;33(6):820‐828. [DOI] [PubMed] [Google Scholar]
  • 195. Blokzijl H, Van Steenpaal A, Vander Borght S, et al. Up‐regulation and cytoprotective role of epithelial multidrug resistance‐associated protein 1 in inflammatory bowel disease. J Biol Chem. 2008;283(51):35630‐35637. [DOI] [PubMed] [Google Scholar]
  • 196. Niiya M, Niiya K, Kiguchi T, et al. Induction of TNF‐α, uPA, IL‐8 and MCP‐1 by doxorubicin in human lung carcinoma cells. Cancer Chemother Pharmacol. 2003;52(5):391‐398. [DOI] [PubMed] [Google Scholar]
  • 197. De Larco JE, Wuertz BR, Manivel JC, Furcht LT. Progression and enhancement of metastatic potential after exposure of tumor cells to chemotherapeutic agents. Cancer Res. 2001;61(7):2857‐2861. [PubMed] [Google Scholar]
  • 198. Jones VS, Huang R‐Y, Chen L‐P, Chen Z‐S, Fu L, Huang R‐P. Cytokines in cancer drug resistance: cues to new therapeutic strategies. Biochim Biophys Acta. 2016;1865(2):255‐265. [DOI] [PubMed] [Google Scholar]
  • 199. Chen R, Alvero A, Silasi D, et al. Regulation of IKKβ by miR‐199a affects NF‐κB activity in ovarian cancer cells. Oncogene. 2008;27(34):4712‐4723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Nakanishi C, Toi M. Nuclear factor‐κB inhibitors as sensitizers to anticancer drugs. Nat Rev Cancer. 2005;5(4):297‐309. [DOI] [PubMed] [Google Scholar]
  • 201. Li F, Sethi G. Targeting transcription factor NF‐κB to overcome chemoresistance and radioresistance in cancer therapy. Biochim Biophys Acta. 2010;1805(2):167‐180. [DOI] [PubMed] [Google Scholar]
  • 202. Kuroda H, Takeno M, Murakami S, Miyazawa N, Kaneko T, Ishigatsubo Y. Inhibition of heme oxygenase‐1 with an epidermal growth factor receptor inhibitor and cisplatin decreases proliferation of lung cancer A549 cells. Lung Cancer. 2010;67(1):31‐36. [DOI] [PubMed] [Google Scholar]
  • 203. Baritaki S, Yeung K, Palladino M, Berenson J, Bonavida B. Pivotal roles of snail inhibition and RKIP induction by the proteasome inhibitor NPI‐0052 in tumor cell chemoimmunosensitization. Cancer Res. 2009;69(21):8376‐8385. [DOI] [PubMed] [Google Scholar]
  • 204. Braunstein S, Formenti SC, Schneider RJ. Acquisition of stable inducible up‐regulation of nuclear factor‐κB by tumor necrosis factor exposure confers increased radiation resistance without increased transformation in breast cancer cells. Mol. Cancer Res. 2008;6(1):78‐88. [DOI] [PubMed] [Google Scholar]
  • 205. Moser B, Wolf M, Walz A, Loetscher P. Chemokines: multiple levels of leukocyte migration control⋆. Trends Immunol. 2004;25(2):75‐84. [DOI] [PubMed] [Google Scholar]
  • 206. Belperio JA, Keane MP, Arenberg DA, et al. CXC chemokines in angiogenesis. J. Leukocyte Biol. 2000;68(1):1‐8. [PubMed] [Google Scholar]
  • 207. Janes KA, Albeck JG, Gaudet S, Sorger PK, Lauffenburger DA, Yaffe MB. A systems model of signaling identifies a molecular basis set for cytokine‐induced apoptosis. Science. 2005;310(5754):1646‐1653. [DOI] [PubMed] [Google Scholar]
  • 208. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005;352(16):1685‐1695. [DOI] [PubMed] [Google Scholar]
  • 209. de Visser KE, Jonkers J. Towards understanding the role of cancer‐associated inflammation in chemoresistance. Curr. Pharm. Des. 2009;15(16):1844‐1853. [DOI] [PubMed] [Google Scholar]
  • 210. McMillin DW, Negri JM, Mitsiades CS. The role of tumour–stromal interactions in modifying drug response: challenges and opportunities. Nat Rev Drug Discov. 2013;12(3):217‐228. [DOI] [PubMed] [Google Scholar]
  • 211. Liu C, Zhu Y, Lou W, Cui Y, Evans CP, Gao AC. Inhibition of constitutively active Stat3 reverses enzalutamide resistance in LNCaP derivative prostate cancer cells. Prostate. 2014;74(2):201‐209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Conze D, Weiss L, Regen PS, et al. Autocrine production of interleukin 6 causes multidrug resistance in breast cancer cells. Cancer Res. 2001;61(24):8851‐8858. [PubMed] [Google Scholar]
  • 213. He W, Luistro L, Carvajal D, et al. High tumor levels of IL6 and IL8 abrogate preclinical efficacy of the γ‐secretase inhibitor, RO4929097. Mol Oncol. 2011;5(3):292‐301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Haga A, Funasaka T, Niinaka Y, Raz A, Nagase H. Autocrine motility factor signaling induces tumor apoptotic resistance by regulations Apaf‐1 and Caspase‐9 apoptosome expression. Int J Cancer. 2003;107(5):707‐714. [DOI] [PubMed] [Google Scholar]
  • 215. Wu S, Saxena S, Varney ML, Singh RK. CXCR1/2 chemokine network regulates melanoma resistance to chemotherapies mediated by NF‐κB. Curr Mol Med. 2017;17(6):436‐449. [DOI] [PubMed] [Google Scholar]
  • 216. Reyes ME, de La Fuente M, Hermoso M, Ili CG, Brebi P. Role of CC chemokines subfamily in the platinum drugs resistance promotion in cancer. Front Immunol. 2020;11:901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Mirzaei SA, Dinmohammadi F, Alizadeh A, Elahian F. Inflammatory pathway interactions and cancer multidrug resistance regulation. Life Sci. 2019;235:116825. [DOI] [PubMed] [Google Scholar]
  • 218. Ratnasinghe D, Daschner PJ, Anver MR, et al. Cyclooxygenase‐2, P‐glycoprotein‐170 and drug resistance; is chemoprevention against multidrug resistance possible? Anticancer Res. 2001;21(3c):2141‐2147. [PubMed] [Google Scholar]
  • 219. Maeng HJ, Lee WJ, Jin QR, Chang J‐E, Shim W‐S. Upregulation of COX‐2 in the lung cancer promotes overexpression of multidrug resistance protein 4 (MRP4) via PGE2‐dependent pathway. Eur J Pharm Sci. 2014;62:189‐196. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.


Articles from Cancer Reports are provided here courtesy of Wiley

RESOURCES