Abstract
Resistance to cancer therapies has been a commonly observed phenomenon in clinical practice, which is one of the major causes of treatment failure and poor patient survival. The reduced responsiveness of cancer cells is a multifaceted phenomenon that can arise from genetic, epigenetic, and microenvironmental factors. Various mechanisms have been discovered and extensively studied, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments (TMEs). To overcome cancer resistance, a variety of strategies have been proposed, which are designed to enhance the effectiveness of cancer treatment or reduce drug resistance. These include identifying biomarkers that can predict drug response and resistance, identifying new targets, developing new targeted drugs, combination therapies targeting multiple signaling pathways, and modulating the TME. The present article focuses on the different mechanisms of drug resistance in cancer and the corresponding tackling approaches with recent updates. Perspectives on polytherapy targeting multiple resistance mechanisms, novel nanoparticle delivery systems, and advanced drug design tools for overcoming resistance are also reviewed.
Keywords: cancer therapy, combination therapy, drug resistance, resensitization
The reduced responsiveness of cancer cells can be associated with various mechanisms, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments. Various targeting strategies against different resistance mechanisms have been developed. The purpose of overcoming the drug resistance of cancer cells is to optimize the sensitivity of the therapy. This can be achieved by polytherapy using the combination of at least two drugs; immunotherapy using checkpoint inhibitors or monoclonal antibodies; antibody‐drug conjugates improving the selectivity of cancer treatment; gene technology modifying the epigenetic sequence; targeted therapy targeting the overexpression of drug efflux transporter or vital proteins for the cancer cell apoptosis; and nanoparticle delivery system improving the efficacy of the drug and reducing the side effect.
1. INTRODUCTION
Cancer remains a global health burden, ranking first or second in the leading causes of death before the age of 70 years in more than 60% of the countries worldwide. 1 In 2020, there were an estimated 19.3 million newly diagnosed cases and 10 million cancer deaths globally. 2 Currently, the major therapies for cancer include surgery, radiation therapy, chemotherapy, hormone therapy, targeted therapy, and immunotherapy, either as monotherapy or in combination. 3 , 4 , 5 , 6 Despite the great development and improvement of cancer treatment in recent decades, resistance to cancer therapies has been a commonly observed phenomenon in clinical practice. 7 , 8 Moreover, cancer cells with resistant characteristics often exhibited cross‐resistance to a variety of anticancer drugs that can be structurally irrelevant, namely, the multidrug resistance (MDR) phenomenon. 9 MDR has been a major obstacle impeding therapeutic success and a dominating cause of cancer relapse and cancer‐related death.
Cancer therapeutic resistance can be categorized into intrinsic and acquired resistance based on the timeline of resistance occurrence. The intrinsic resistance, also known as primary resistance, is mediated by the endogenous factors that are present in tumor cells or tissues before therapeutic applications, which provide cancer cells with survival advantages and adaptability to primary therapeutic stress. 10 , 11 , 12 However, acquired drug resistance is developed after receiving cancer treatment, which is generally mediated by the adaptive alterations against the given therapy in initially sensitive tumors, resulting in compromised treatment effectiveness. 13 , 14 , 15 The reduced responsiveness of cancer cells can be associated with various mechanisms, which usually involve the coactions of genetic factors and nongenetic contributors. Genetic factors in tumor cells have been considered critical contributors to therapeutic resistance, such as genetic diversity, acquired mutations of drug targets, amplification of oncogenes in compensatory or bypass pathways, and epigenetic modifications, which can further affect intratumor heterogeneity, tumor cell plasticity, DNA repair, and the susceptibility of tumor cells to cell death pathways, leading to multifactor‐mediated resistance. 16 , 17 , 18 However, resistant cases in the absence of genetic alteration have been extensively recognized in different types of cancers. 19 Phenotype changes may be independent of genotype alteration in resistance mediated by metabolic inactivation of drugs, 20 , 21 reduced intracellular drug concentration by transporters, 22 , 23 , 24 drug compartmentation, 25 and drug‐induced reversible transcriptional or posttranslational regulations on adaptive pathways. In addition to the factors within the tumor cells, the tumor microenvironment (TME) is also considered to be involved in the development of resistance in some cancers 26 (Figure 1).
FIGURE 1.
Cancer resistance mechanisms, including drug inactivation, insufficient intracellular drug concentration, drug target alterations, compensatory pathways activation, DNA repair enhancement, and tumor plasticity. Source: This figure was created with Biorender.com.
Notably, the resistance mechanisms are not mutually exclusive and can act jointly to induce therapeutic irresponsiveness in cancer. Therefore, to develop effective strategies to obviate cancer resistance, it is an urgent need to gain a better understanding of the mechanisms collectively. In this review, we will discuss the discoveries of the diverse mechanisms of cancer resistance, the approaches to improve anticancer efficacy via targeting specific mechanisms, and the progress in developing polytherapy fighting against multiple resistance mechanisms.
2. RESISTANCE MECHANISMS IN CANCER AND COMBATING STRATEGIES
Drug resistance in cancer can arise from genetic, epigenetic, and microenvironmental factors. A better understanding of the molecular mechanisms underlying cancer resistance is necessary to develop effective strategies to overcome it. In the following context, the identified resistance mechanisms will be discussed with reviews of recent research updates, followed by the development of corresponding combating approaches, such as novel drug designs, combination therapies with specific targets, tackling alternative signaling pathways, and the modulation of the TME.
2.1. Metabolism‐associated drug inactivation
The activation and deactivation processes of many chemotherapeutic agents are regulated by drug‐metabolizing enzymes (DMEs). 27 Dysregulation of DMEs and metabolic signaling pathways, which can lead to the detoxification of drugs or failure in the conversion of drugs into active metabolites, is one of the major mechanisms for chemoresistance in cancer. 28
Some anticancer drugs require activation by metabolic enzymes. For example, the biotransformation of irinotecan into the active metabolite SN‐38 is catalyzed by carboxylesterase, 29 and the thymidine phosphorylase is responsible for the activating metabolism of 5‐fluorouracil (5‐FU) into fluorodeoxyuridine monophosphate. 30 Cytarabine (AraC), a nucleoside drug used in patients with acute myeloid leukemia (AML) and non‐Hodgkin's lymphoma, relies on the phosphorylation catalyzed by deoxycytidine kinase (DCK) to become the cytotoxic form cytarabine triphosphate, 31 and the deficiency of DCK has been considered to be associated with AraC resistance in AML. 32 Wu et al. reported that DCK mutations were found in 4 of 10 patients with AML relapse after complete remission and high‐dose AraC post‐remission treatment. 33 In vitro studies using drug‐selected AraC‐resistant AML and lymphoma cell models indicated that mutation or deficient expression and function of DCK could be acquired after receiving AraC therapy, resulting in the reduced cellular response to AraC and possible cross‐resistance to other nucleoside drugs like gemcitabine. 33 , 34 , 35 A potential strategy to overcome DCK deficiency‐mediated nucleoside drug resistance is using nucleoside analog phosphate prodrugs with anticancer efficacy without the demand for DCK phosphorylation, 36 among which NUC‐1031, a gemcitabine phosphoramidate prodrug, has entered clinical trials for the treatment of gemcitabine‐resistant cancers. 37 , 38 Other reported attempts in preclinical evaluations include enhancing DCK activity by etoposide in combination with AraC for the treatment of AML, 39 and bypassing the drug resistance mechanism associated with downregulated DCK expression using second‐generation deoxyadenosine analog clofarabine in acute lymphoblastic leukemia cells. 40
The other aspect of metabolism‐associated anticancer drug inactivation is the detoxification of drugs by metabolizing enzymes. For instance, certain isoforms of aldehyde dehydrogenases (ALDH), including ALDH1A1 and ALDH3A1, have been reported to specifically cause resistance against chemotherapeutic drugs of nitrogen mustard type, such as cyclophosphamide, mafosfamide, and ifosfamide. 27 In contrast, cytochrome P450 (CYP450) in phase I metabolism, and glutathione‐S‐transferase (GST) as well as uridine diphosphoglucuronosyltransferase (UGT) in phase II conjugating biotransformation, are involved in the inactivation of a broader spectrum of anticancer drugs. 41 , 42
The CYP450 enzyme superfamily consists of 57 members that are responsible for the phase I oxidation of most clinically used drugs. 43 Of the CYP450 enzymes, CYP1B1, 2C8, 3A4, and 3A5 have been reported to be correlated to cancer resistance, 44 , 45 with CYP1B1 being mostly studied, which is found to be exclusively overexpressed in various types of cancers but has relatively low expression in normal tissues. 46 Intratumoral CYP1B1 overexpression may contribute to the diminished effectiveness of a diversity of chemotherapeutic drugs, such as paclitaxel and docetaxel, mitoxantrone, flutamide, and gemcitabine. 47 , 48 , 49 , 50 However, whether the resistance against these drugs is contributed by CYP1B1‐catalyzed drug inactivation remains controversial. McFadyen et al. suggested that docetaxel was metabolized by CYP1B1, and the docetaxel resistance in CYP1B1 overexpressing Chinese hamster ovary cancer cells could be reversed by a CYP1 inhibitor. 51 , 52 However, Martinez et al. later demonstrated that CYP1B1 did not directly inactivate docetaxel because the cytotoxicity of docetaxel in MCF‐7 breast cancer cells was not affected by silencing CYP1B1 or adding recombinant CYP1B1. 53 Although it is possible that CYP1B1 may play different roles in regulating drug resistance in different cancer types, the inhibition of CYP1B1 activity has been considered to be a therapeutic target for improving chemotherapy. 54 Although lacking high selectivity, phytochemicals are the most common source of CYP1B1 inhibitors, including stilbene, flavonoids, coumarins, anthraquinones, and alkaloids. 55 , 56 , 57 By modifying the structures of phytochemicals, highly potent and selective inhibitors of CYP1B1 have been developed and under preclinical evaluation, such as TMS ((E)‐2,3′,4,5′‐tetramethoxystilbene) 58 and α‐naphthoflavone derivatives. 59
GSTs are involved in drug detoxification in phase II metabolism and catalyze the glutathione (GSH) conjugation to drugs. 60 Overexpression of GSTs in cancer cells may enhance the detoxification of anticancer drugs. 61 Moreover, after GSH conjugation, the conjugated drugs may become substrates of ATP‐binding cassette (ABC) transporters, particularly the MDR‐associated proteins (MRPs, belongs to ABCC subfamily), and got actively pumped out of the cancer cells. 62 , 63 , 64 Among the GST superfamily, GST alpha 1 (GSTA1), GST Mu 2 (GSTM2), and GST Pi 1 (GSTP1) have been found to be associated with cisplatin resistance in ovarian, lung, and gastric cancers. 65 Notably, the overexpression of GSTP1 has been considered a factor impairing the efficacy of platinum drugs like cisplatin, carboplatin, and oxaliplatin by promoting the formation of platinum‐GSH conjugates. 66 , 67 The I150V polymorphism of GSTP1 with a phenotype of reduced enzymatic capacity has been correlated with better therapeutic outcomes in gastric cancer patients receiving oxaliplatin‐based chemotherapy. 68 , 69 Besides platinum drugs, the sensitivity to doxorubicin and various alkylating agents in cancer may also be affected by the detoxification process mediated by GSTA subclass, GSTP1, and GSTM1 enzymes. 70 , 71 However, additional mechanisms other than GSH conjugation may be involved in GST‐related chemoresistance to doxorubicin and alkylators, such as free radical scavenging and apoptosis suppression via the inhibition of the mitogen‐activated protein kinase (MAPK) pathway. 72 , 73 , 74 Thus, there has been an increasing focus on targeting the GSTs to overcome chemoresistance. Inhibitors of GSTs have been shown effective in treating resistant cancer cells. Ethacrynic acid and analogs are among the earliest investigated GST inhibitors that exhibited resensitizing effects on tumor cells to alkylating agents via covalent binding to GSTs and reducing the enzyme activity. 75 , 76 NBDHEX (6‐(7‐nitro‐2,1,3‐benzoxadiazol‐4‐ylthio)hexanol) is another potent inhibitor against GSTP1, GSTM2, and other GST isoenzymes, 77 which has been found beneficial in combating cisplatin‐resistant osteosarcoma when used in combination with cisplatin. 78 Apart from inhibiting GST activity, NBDHEX can also induce the dissociation of GSTP1 from its complex with c‐Jun N‐terminal kinase or tumor necrosis factor receptor‐associated factor 2 thereby promoting the activation of apoptosis pathways in cancer cells. 79 However, one of the significant roadblocks that GST inhibitors encounter in clinical trials is their insufficient specificity, prompting the development of novel NBDHEX analogs with improved selectivity, among which MC3181 was screened out as a specific GSTP1‐1 inhibitor and tested highly efficient in inhibiting vemurafenib‐resistant melanoma in vitro and in vivo. 80 , 81 Although the GST inhibitors showed promising effects on inducing cell apoptosis via inhibiting GST activities, investigations on combinations with chemotherapeutic drugs are lacking; thus, whether the inhibitors can reduce drug inactivation remains to be determined. On the other hand, utilizing prodrugs like canfosfamide (TLK286) and brostallicin, which are activated by GSTs to become cytotoxic, has entered clinical evaluations as a practical strategy to circumvent the GST‐mediated resistance mechanism. 82 , 83
Similar to GSTs, the UGT enzymes are involved in phase II metabolism, and they catalyze the glucuronidation process. Among the three subfamilies (UGT1A, 2A, and 2B) of the UGTs, UGT1A enzymes, particularly UGT1A1, have been shown to overexpress in tumor tissues and play a role in resistance to anticancer drugs. 20 UGT1A1 is physiologically responsible for facilitating bilirubin elimination by catalyzing the glucuronic acid conjugation of bilirubin. Elevated tumoral UGT1A1 levels can increase the glucuronidation of SN‐38, the active metabolite of anticancer drug irinotecan, resulting in the inactive SN‐38 glucuronide and reduced therapeutic efficacy. 84 UGT1A enzymes have also been reported to be associated with primary resistance to some targeted drugs, such as heat shock protein HSP90 inhibitors ganetespib and luminespib 85 and epidermal growth factor receptor (EGFR) inhibitor erlotinib. 86 Drug‐induced increase of UGT1A expression has been reported in AML patients treated with ribavirin, which resulted in acquired resistance. 87 Apart from substrate drugs, the efficacy of monoclonal antibody anticancer drugs may be affected by UGT1As. For instance, UGT1A6 has been found to be correlated with resistance to programmed cell death 1 (PD‐1) antibody nivolumab in patients with advanced renal clear‐cell cancer. 88 As UGTs are not known to conjugate proteins like antibodies, UGT‐mediated resistance to nivolumab may not be related to directed drug inactivation, but to other mechanisms, such as indirect metabolism regulations by UGTs and other resistance‐correlated signaling regulations involving UGTs. 9 Because of their association with hampered drug response, the expression of UGT1As might be a useful indicator for patient stratifying so as to avoid the application of substrate drugs to patients who are likely to have low response due to high UGT1A levels. Meanwhile, UGTs can be potential pharmacological targets to overcome drug resistance in cancer. However, the use of UGT inhibitors has been limited by toxic side effects, and selectivity remains the major challenge in developing novel inhibitors of UGTs. 89 , 90
Notably, direct detoxification is not the only mechanism of DME‐associated cancer resistance, other involving factors like drug–drug interactions and signaling molecules inherent in cancer cells should be taken into consideration in developing strategies targeting metabolic enzymes in cancer. Regulating the expression and activity of DMEs through regulatory signaling transductions can be beneficial to overcoming cancer resistance not only by means of retaining drug bioavailability but also in other aspects like inhibiting cancer cell proliferation or invasion.
2.2. Reduced drug uptake
Drugs can cross the cell membrane through diffusion, endocytosis, or transporters, which can be affected by the permeability and lipid composition of the plasma membrane, and the functions or expression levels of membrane transporters. Alteration of cell membrane structure can impair drug diffusion across the plasma membrane and the endocytosis process. The plasma membranes from drug‐resistant cancer cells have a different lipid composition compared to those from the parental drug‐sensitive cells: The cholesterol and phospholipid levels are elevated, and the protein/lipid ratio is increased up to 60% in MDR cells compared to sensitive cells. 91 Besides, the slight alkalic pH of the cytoplasm in MDR cancer cells could attenuate the repulsions between the polar groups from membrane lipids by shielding the negative charges, thereby increasing lipid packing and membrane rigidity. 92 Therefore, MDR cells have plasma membranes with a relatively lower fluidity and reduced permeability leading to decreased drug absorption. 92 Chemotherapeutic drugs, including vinblastine, doxorubicin, and cisplatin, are vulnerable to this resistance mechanism. 93 , 94 , 95 Additionally, using model membranes by molecular dynamic simulations, Rivel et al. recently demonstrated that, cancer cell membranes are common with the loss of lipid asymmetry compared to normal cell membranes, which may be a contributor to the slower diffusion of cisplatin into cancer cells. 96 These findings have suggested that lipid composition assessment may be useful for cancer prognosis, and modulating cell membranes can be a potential strategy to ameliorate drug resistance in cancer.
It has been shown that treating cancer cells with short‐chain ceramides can increase cell membrane permeability and fluidity, resulting in an increased uptake of amphiphilic anticancer drugs such as doxorubicin, either in free form or encapsulated form with lipid‐based nanoparticles. 97 , 98 The other way to modulate the lipid composition of the cell membrane is by enhancing sphingomyelinase activity using an agonist, such as daunorubicin, etoposide, and ara‐C, thereby decreasing sphingomyelin levels and increasing ceramide levels, leading to increased membrane fluidity. 99 , 100 Moreover, biomimetic cell membrane‐coated nanoparticle delivery systems are gaining increased recognition. Cancer cell membrane‐based nanoparticles contain surface proteins from cancer cells; therefore, they can reduce side effects by specifically targeting cancer cells through homotypic binding. 101 Fang et al. reported that the uptake of membrane‐coated nanoparticles in human breast cancer MDA‐MB‐435 cells was increased by 20‐fold compared to naked nanoparticles. 102 Many studies have demonstrated the selectivity and biosafety of cancer cell membrane‐based biomimetic nanoparticles using in vitro and in vivo MDR cancer models, 103 , 104 , 105 suggesting a novel drug delivery strategy to improve therapeutic efficacy for MDR cancer.
Intracellular drug concentrations largely depend on the activity of uptake transporters. The main transporters involved in drug uptake are the drug solute carriers (SLCs). The SLC superfamily consists of more than 400 members categorized into 52 families. 106 Chemoresistance‐relevant SLCs include the organic anion transporting proteins (OATPs), organic cation transporters, concentrative nucleoside transporters, equilibrative nucleoside transporters, and copper transporters. 107 , 108 Reduced uptake of chemotherapeutic drugs can be caused by either genetic variants resulting in truncated uptake transporter proteins with reduced or absent function or acquired expression changes in uptake transporters mediated by pharmacological selection pressures. Among them, OATPs (SLCO family) are mostly studied. In particular, amplified expression of OATP1A2, OATP1B1, and OATP1B3 has been found in various cancer tissues, 109 which is considered associated with chemosensitivity because of its role in the uptake of several classes of anticancer drugs, such as taxanes, platinum‐based drugs, camptothecin analogs, methotrexate, and some tyrosine kinase inhibitors (TKIs). 110 , 111 Downregulation of OATP1B3 was reported in a patient‐derived docetaxel‐resistant prostate tumor xenograft model, where the intratumoral concentrations of docetaxel and cabazitaxel were both lower than chemotherapy‐naive tumors. In contrast, prostate tumors with OATP1B3 expression exhibited an increased uptake of both taxanes and higher chemotherapeutic sensitivity. 112 Overexpression of OATP1B3 in prostate cancer may be beneficial to chemotherapy but can be detrimental to hormone therapy because OATP1B3 drives testosterone uptake in cancer cells, leading to the development of resistance to androgen deprivation therapy (ADT). 113 Increased OATP1B3 expression in prostate cancer can be induced by ADT, which provides a clue for sequential therapy design: Upregulated tumor SLCO expression following ADT could potentially enhance the uptake of chemotherapeutic drugs like taxanes and improve treatment response. 114 Interestingly, the genetic variant of OATP transporters may confer resistance to TKIs via the uptake activity instead of enhancing sensitivity.
Haberkorn et al. recently discovered that cancer‐type OATP1B3 protein, a splice variant of liver‐type OATP1B3, is localized in the lysosomal membrane of colorectal cancer (CRC) cells and contributes to the transport of encorafenib and vemurafenib into lysosomes, resulting in decreased drug concentrations in the cytoplasm and reduced drug efficacy. 115 Therefore, it is necessary to identify the genotype of intratumorally expressed SLCs in the process of confirming their roles in drug resistance.
Although enhancing drug uptakes by modulating SLC transporters may improve chemosensitivity, it may not be a practical approach in cancer treatment. Due to the ubiquitous expression of SLCs in many tissues, modulating the function of SLCs could interfere with the transport activities of nutrients and xenobiotics in normal tissues, which can lead to unfavorable side effects and drug–drug interactions. On the other hand, besides the uptake of anticancer drugs, modulating SLCs may also influence the nutrient uptake in cancer cells, risking promoting cancer progression and metastasis. The best strategy to overcome the uptake transporter‐mediated resistance in cancer may be to use drugs or delivery systems that can circumvent the aberrant transporter, either with improved diffusion efficacy or the capability to utilize alternative transporters.
2.3. Increased drug efflux
In addition to reduced drug uptake, drug efflux mediated by the overexpression of ABC transporters is the major mechanism contributing to insufficient intracellular drug concentration. In humans, the ABC transporter superfamily consists of 49 members categorized into 7 subfamilies (ABCA–ABCG) functioning to transport various substrates, including lipids, ions, peptides, and xenobiotics. 116 The involvement of ABC transporters in MDR was first discovered in 1976 when ABCB1, which is also known as P‐glycoprotein (P‐gp) or multidrug resisitance 1 (MDR1) protein, was discovered in mouse MDR cell lines. 117 To date, at least 13 ABC transporters have been found to directly mediate chemoresistance via the efflux of anticancer drugs. 118 These ABC transporters mainly distribute on the plasma membrane and are triggered upon binding to substrate drugs, resulting in an ATP hydrolysis–driven conformational change of the transporter and the extrusion of the substrate drugs. 119 Consequently, the overexpression of these transporters has been correlated with poor chemotherapy response and unfavorable patient prognosis in many different types of cancers. Most studies have focused on the ABC transporters with wide spectrums of substrates, such as ABCB1, ABCG2 (also known as breast cancer resistance protein), and the ABCC subfamily (also known as multidrug resistance‐associated proteins, MRPs) with ABCC1 and ABCC10 as representative members. Many cytotoxic chemotherapeutic drugs can be pumped out of cancer cells by ABCB1 and ABCCs, such as taxanes, vinca alkaloids, and anthracyclines. The spectrum of anticancer drugs ABCG2 confers resistance to largely overlap with that of ABCB1 and ABCCs but with some specificities; anthracenedione (such as mitoxantrone) and camptothecins (such as topotecan and SN‐38) are especially vulnerable to the efflux activity of ABCG2, whereas taxanes and vinca alkaloids are not much affected by ABCG2. 120 Certain clinically used kinase inhibitors, such as imatinib, nilotinib, dasatinib, palbociclib, and some newly developed targeted drugs, such as OTS964 and ARS‐1620, have also been reported as substrates of ABC transporters. 121 , 122 , 123 , 124 Moreover, ABCCs generally have high affinities to GSH‐conjugated or glucuronate‐conjugated forms of drugs; therefore, the efflux activity of ABCC transporters may be regulated by phase II metabolic enzymes. 24 Additionally, for brain tumors and metastatic cancer in the brain, chemosensitivity is also affected by the o expression of ABCB1 and ABCG2 transporters at the luminal membrane of endothelial cells of brain microvessels, which are known to impede anticancer drug delivery across the blood–brain barrier. 125 , 126
Ongoing efforts have been taken to develop modulators of ABC transporters in order to overcome this resistance mechanism. Numerous small molecule compounds have been developed and tested as inhibitors against ABC transporters, particularly ABCB1. There have been three generations of ABCB1 inhibitors; however, these inhibitors have failed to enter clinical use as combination therapy with substrate anticancer drugs due to different drawbacks. The first generation of ABCB1 inhibitors, such as verapamil, erythromycin, and cyclosporine A, mostly lacks sufficient therapeutic efficacy, which results in considerable side effects by the required high dosages. Although the second‐generation inhibitors, such as dexverapamil and valspodar, have exhibited improved potency, they failed to proceed to clinical application because of their unwanted interactions with the CYP450 enzymes leading to unfavorable pharmacokinetic profiles. The third generation, exampled by tariquidar and zosuquidar, is able to overcome the selectivity problem; however, the performance in clinical settings turns out to be unsuccessful due to interpatient variability. 127 In the recent decade, some repurposing targeted anticancer drugs, such as selonsertib, tepotinib, and poziotinib, have been discovered as dual inhibitors for ABCB1 and ABCG2. 128 , 129 , 130 Inhibitors that can antagonize both ABCB1‐ and ABCC1‐mediated resistance have also been reported, exampled by cediranib and CBT‐1 (tetrandrine). 131 , 132 Targeting multiple ABC transporters may be more prospective given that co‐expression of different ABC transporters is common in tumor tissues and multitargeting drugs may be beneficial to avoid compensatory upregulation of another transporter induced by selective inhibition of a single transporter. 133
Silencing the efflux transporters using gene editing technologies such as siRNA and CRISPR/Cas9 system has been increasingly investigated as a novel approach to reducing drug resistance, which can potentially benefit genetic variants with intrinsic ABC transporter‐related resistance. 134 However, many studies conducted gene editing in cell line‐based settings before the step of tumor xenograft model establishment, which is unlikely to represent gene therapy for patients. Cancer‐targeted delivery remains a major challenge in developing gene therapies against ABC transporters. Another trending strategy against ABC transporter‐dependent MDR is to target the regulatory factors of the ABC transporter expression, including transcriptional regulation and epigenetic modifications on the gene, and posttranslational modifications on the protein. Inhibiting the cancer‐specific regulators of ABC transporters may render an advantage to targeting tumor tissues selectively and increasing safety to normal tissues. 127 Dysregulated noncoding RNAs, including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have been found to be associated with the overexpression of ABCB1, ABCG2, and ABCCs in chemoresistant cancers. 135 The effect on suppressing ABC transporters and reversing cancer MDR by nanoparticle‐delivered ncRNAs mimics or inhibitors has been verified from in vitro and in vivo studies. 136 , 137 Another epigenetic factor‐linked overexpression of ABC transporters in cancer is the hypomethylation of the transporter gene promoter, and evidence has shown that agonists of DNA methyl transferases can induce the hypermethylation of the ABCG2 promoter resulting in the lower ABCG2 expression and increased intracellular concentration of substrate drugs. 138 , 139 Alternatively, the expression of ABC transporters can be modulated by targeting the upstream regulatory pathways. For instance, in breast cancer cells, inhibiting the overexpressed receptor tyrosine kinase‐like orphan receptor 1 (ROR1), which is an upstream regulator of ABCB1 transcription via MAPK/extracellular signal‐regulated kinase (ERK) and p53 pathways, can reduce ABCB1‐mediated drug efflux and resensitize breast cancer cells to doxorubicin. 140 Another recent study showed that suppressing mitochondrial respiration using methylation‐controlled J protein mimetics can decrease ATP production thereby decreasing the energy for ABCB1 and ABCG2 efflux activity and overcoming chemoresistance in vitro and in vivo. 23
Indeed, the direct approach to circumvent this resistance mechanism is to avoid using substrate drugs or develop novel anticancer drugs that can bypass the efflux transporters. This may rely on advanced computer‐aided drug designs to modify drug structures of current known and actively reported substrates of ABC transporters.
2.4. Alterations of the drug targets and activation of compensatory pathways
The leading limitation of targeted cancer therapies is that cancer cells can be intrinsically irresponsive or acquire resistance after a period of treatment because of either the mutation of target molecules (“on‐target” mechanism) or tumor cells gaining survival advantages from a new mechanism independent of the target (“off‐target” mechanism), such as the activation of downstream signaling pathways or compensatory pathways 141 , 142 (Figure 2).
FIGURE 2.
Alteration of drug target and activation of compensating pathways. Cancer resistance associated with alterations in the drug target site or modifications in the structure of the target. The reactivation of the downstream pathway bypasses the other unblocked pathway enabling drug resistance. Activation of compensatory signaling pathways to resist cell death leading to drug resistance. Source: This figure was created with Biorender.com.
The primary approach to overcome “on‐target” mechanism is to develop new generations of targeted drugs against drug‐resistant tumors. For instance, the first‐ and second‐generation EGFR–TKIs, such as gefitinib, erlotinib, and dacomitinib, are less effective in treating patients with additional EGFR mutation T790M. 143 This has led to structural modification and the development of the third‐generation EGFR–TKIs therapy. Osimertinib (AZD9291), a third‐generation EGFR–TKI, presented a superior clinical response and outcome in EGFR‐mutated non‐small cell lung cancer (NSCLC). However, the rapidly acquired resistance to osimertinib conferred by EGFR C797S mutation has been observed. 144 Besides, some patients who exhibit primary resistance remain unresponsive to third‐generation and other newly developed EGFR–TKIs. Complete remission is rare, and all patients eventually develop resistance, suggesting that both primary and acquired resistance mechanisms reduce the efficacy of the drug. 145 , 146 To reverse these types of resistance, more recently, the fourth‐generation EGFR inhibitors, which can inhibit both T790M and C797S signaling, have been introduced into clinical evaluation. 143 So far, EAI045 is the first allosteric TKI developed for this purpose. The C797S mutation is unlikely to impair the efficacy of EAI045 because its allosteric binding pocket is not affected by this cysteine residue. EGFR receptor dimerization invalidates drug‐mediated inhibition alone. The activity against T790M and C797S can be restored by a combination regimen with cetuximab, an antibody against EGFR dimerization. 147 Other fourth‐generation EGFR‐TKIs, such as JND3229 and JBJ‐04‐125‐02, were recently found to be active in EGFR C797S‐T790M‐L858R signal transduction in vitro and in vivo. 148 , 149
For the first‐generation tropomyosin receptor kinase (TRK) kinase inhibitors, such as entrectinib and larotrectinib, the secondary mutations occurring at the ATP binding pocket of the TRK kinase domain, including G667C and G595R mutations in NTRK1 gene, and G696A and G623R mutations in NTRK3 gene, are common acquired‐resistance mechanisms. 150 , 151 In addition, second‐generation TRK inhibitors like selitrectinib have been developed to overcome this acquired resistance.
The multitargeted TKI crizotinib was approved by the Food and Drug Administration (FDA) in 2011 to treat patients with advanced NSCLC harboring ALK rearrangements. 152 However, despite a high response rate of 60% in ALK‐rearranged NSCLC, most patients develop resistance to crizotinib, typically within 1–2 years. Studies of ALK‐rearranged lung cancers with acquired resistance to crizotinib have identified ALK fusion gene amplification and secondary ALK tyrosine kinase domain mutations in about one third of cases. 153 , 154 To date, seven different acquired resistance mutations have been identified among crizotinib‐resistant patients. The most frequently identified secondary mutations are L1196M and G1269A. In addition to these mutations, the 1151T‐ins, L1152R, C1156Y, G1202R, and S1206Y mutations have also been detected in crizotinib‐resistant cancers. 155 , 156 , 157 In approximately one third of crizotinib‐resistant tumors, there is evidence of activation of bypass signaling tracts such as EGFR. 157 In the remaining one third of crizotinib‐resistant tumors, the resistance mechanisms remain to be identified. Next‐generation ALK inhibitors with improved potency and selectivity compared with crizotinib have been developed to overcome crizotinib resistance in the clinic. The ability of several ALK TKIs (TAE684, AP26113, ASP3026, and CH5424802) to inhibit ALK activity was evaluated in models harboring different ALK secondary mutations. 153 , 158 These studies revealed variable sensitivity to these ALK inhibitors depending on the specific resistance mutation present. For example, the gatekeeper L1196M mutation was sensitive to TAE684, AP26113, and ASP3026, whereas 1151T‐ins conferred resistance to all next‐generation ALK TKIs. Ceritinib, an ATP‐competitive, potent, and selective next‐generation ALK inhibitor, has shown favorable selectivity to ALK. 159 In clinical studies involving ALK‐positive NSCLC patients, ceritinib has exhibited marked antitumor activity has been observed in both crizotinib‐relapsed and crizotinib‐naive patients. 160 , 161 On the basis of this impressive clinical activity, ceritinib received FDA approval on April 29, 2014.
Despite increasing successes in efforts to target oncogenic driver amplifications or mutations, several of the most formidable oncogenes and tumor suppressor genes remain undruggable, including RAS (KRAS, NRAS, and HRAS) and RAF (ARAF, BRAF, and CRAF). Effective targeting of KRAS signaling has been tough to realize in patients. 162 , 163 Blocking the localization of KRAS at the plasma membrane, a vital element for its activation, has been ineffective because multiple compensatory pathways modulate this process. Similarly, targeting effector signaling downstream of KRAS has not achieved remarkable clinical benefits on the account of paradoxical signaling activation generated by the inhibitor or because of on‐target toxicity limiting the maximum tolerated dose in patients. 164 , 165 Tumor cells with KRAS or BRAFV600E mutations are addicted to a downstream ERK signaling cascade for their growth, viability, and other malignant properties. 166 The adaption of selective inhibitor target KRASG12C only briefly inhibits KRAS within 1–3 days of treatment, and initial signaling suppression is accompanied by the re‐storage of active KRAS and reactivation of ERK1/2 signaling pathway. 167 , 168 Accumulation of active KRAS illustrates that compensatory activation of upstream receptor tyrosine kinases (RTKs), including EGFR, is to a large extent responsible for the adaptive alterations noted during KRASG12C inhibitor treatment. In RASMut tumors, RAF inhibitors (RAFis) are ineffective because they drive paradoxical ERK1/2 pathway activation and adventitious tumor progression. 169 However, MAPK kinase (MEK) inhibitors (MEKis) do not present the same limitation as RAFis, but the relief of feedback inhibition and pathway reactivation restricts MEKi monotherapy in RASMut‐driven tumors. 166 , 170 By the amplification of KRASG13D or BRAFV600E, CRC cells acquire resistance to the MEKi. 171 Furthermore, MEKi resistance driven by BRAFV600E amplification is thoroughly reversible upon prolonged drug withdrawal as BRAFV600E amplification brings about a selective disadvantage in the absence of MEKi, and MEKi withdrawal drives ERK1/2 activation beyond a key point that is optimal for cell proliferation and viability. 172 Remarkably, MEKi resistance driven by KRASG13D amplification is not reversible. Upon MEKi withdrawal, CRC cells experience an ERK1/2‐dependent epithelial‐to‐mesenchymal transition (EMT) and emerge resistance to frequently used chemotherapeutics instead of exhibiting growth defects. 172 Consequently, the appearance of MEKi resistance, drug‐addiction, and the possible options of intermittent dosing schedules, may depend on the nature of the amplified oncogenes (e.g., KRASG13D BRAFV600E), further highlighting the hardships of targeting RAS and RAF mutant tumors. Mutations of tumor suppressor can also result in resistance to targeted therapies, for example, mutations in phosphatase and tensin homolog deleted on chromosome 10 (PTEN) can activate phosphatidylinositol‐3‐kinase (PI3K) β signaling in response to PI3Kα inhibitors, 173 as well as reverse mutations in breast cancer 1 (BRCA1) or BRCA2 in response to poly(ADP‐ribose) polymerase (PARP) inhibitors. 174
In NSCLC, the majority of all EGFR mutations are EGFRL858R in exon 21 and activating EGFR exon 19 deletions, and tumor cells harboring the specific activating mutations exhibit high sensitivity to EGFR–TKIs. 175 , 176 These constitutively activate mutant EGFR oncoproteins signaling by regulating MAPK and PI3K/AKT/mTOR signaling cascades to promote tumorigenesis. 177 In a series of EGFR–TKI‐resistant tumor samples, 1% of patients exhibited resistance to early‐generation EGFR–TKIs via the acquisition of BRAFV600E or BRAFG469A. 178 Increased mTOR level was related to EGFR–TKI resistance in clinical samples, 179 and in mouse models, the application of rapamycin (mTOR inhibitor) presented the positive progression of EGFRmut lung tumors. 180 EGFR–TKIs in combination with PI3K/AKT/mTOR signaling pathway inhibitors have proved improved tolerability and efficacy. Another pattern of acquired resistance to kinase inhibition is the amplification of upstream genes, which can exemplify by mesenchymal–epithelial transition (MET) amplification leading to resistance to EGFR–TKIs. 181
The clinical value of proto‐oncogene 1 (ROS1)‐directed TKIs was first prospectively explored in NSCLC patients, and in 2016, the FDA and EMA approved crizotinib for the treatment of advanced‐stage ROS1‐rearranged NSCLC. 182 Afterward, new ROS1–TKIs got into clinical trial, leading to the FDA and Ministry of Health, Labor and Welfare of Japan approvals of entrectinib for the treatment of ROS1‐aberrant NSCLC. So far, all developed ROS1–TKIs are multikinase inhibitors, which can also target MET (e.g., crizotinib and cabozantinib), ALK (e.g., ceritinib and crizotinib), TRK (e.g., entrectinib and repotrectinib), and several other kinases (e.g., SRC, EGFR, JAK2, and FLT3) with equivalent or lower potency. 183 , 184 , 185 Various compensatory pathways involved in TKI therapy of ROS1‐rearranged tumors progression, such as KRAS, NRAS, BRAF, HER2, EGFR, MEK, and MET. For example, mutations of KRASG12D and BRAFV600E have emerged with crizotinib treatment in the clinical testing, 186 and NRASQ61K has occurred while entrectinib treatment. 187 Upregulated HER2 phosphorylation has been identified in patient‐derived, crizotinib‐resistant CD74–ROS1‐positive CUTO23 (NSCLC cell line), and afatinib combined applied recovered crizotinib sensitivity in CUTO23. 188 Usually, ROS1 fusions de novo barely co‐occur with EGFR mutations in clinic, 189 but ROS1 fusions come into sight as resistance mechanisms to EGFR–TKI therapy in EGFR‐mutant NSCLC, 190 suggesting feedback or potential signaling reciprocity among ROS1 and EGFR. In patients treated with ROS1–TKIs, activating PIK3CA mutations have also been reported. 188 , 191 In ROS1 fusion‐positive NSCLCs patients treated with ROS1–TKIs, 11% were found to have concurrent MEK/MAPK alterations. 192 Subsequently, it was proved that the deletion of MEK1 or MEKK1 in cells conferred resistance to ROS1–TKIs and combined ROS1 and MEKis inhibited cell growth. 192 Until 2020, MET amplification was found to act as an important participator of ROS1–TKI (e.g., lorlatinib) resistance. 141 ROS1 kinase domain mutations that trigger the resistance of ROS1 fusion‐positive tumors to ROS1–TKIs have been identified by various preclinical and clinical studies. 188 Some of these mutations are paralogous to ALK resistance mutations that emerge with targeted therapy for ALK fusions, 193 among which ROS1G2032R is the most common resistance substitution observed in patients treated with crizotinib. 194 Considering its clinical benefits but its lack of activity against ROS1G2032R, lorlatinib can rebuild enduring disease control in ROS1‐rearranged tumors with the limited spectrum of resistance mutations. 195 However, FDA has conferred repotrectinib as the Fast Track Designation agent in patients who previously experienced one ROS1–TKI and platinum‐doublet chemotherapy. Up to now, next‐generation ROS1–TKIs have not been approved for the treatment of ROS1 rearrangement‐positive tumors.
2.5. Enhanced DNA repair
The DNA damage response (DDR) is a mechanism for repairing drug‐induced DNA damage. Mutation or dysregulation of certain genes and DDR mechanisms are common in many types of cancer and can influence the sensitivity to DNA‐damaging chemotherapy drugs, such as cisplatin and 5‐FU. 196 , 197 The nucleotide excision repair (NER) responsible for repairing DNA damage repairing and mismatch repair (MMR) responsible for maintaining genomic integrity are known to be related to 5‐FU resistance. It is demonstrated by Liu et al. that the acquired upregulation of the excision repair cross‐complementing 1 (ERCC1), a component of the NER system, was induced by 5‐FU treatment in gastric cancer cells, which can subsequently weaken the anticancer activity of 5‐FU. Further mechanism studies revealed that 5‐FU‐induced ERCC1 overexpression may be regulated by ERK 1/2 and p38 signaling‐mediated activation of the transcription factor c‐jun/activator protein‐1, which provides a clue to overcome this resistance mechanism using ERK inhibitors or p38 kinase inhibitors. 198 In contrast, the role of MMR in 5‐FU resistance remains uncertain. It was recently reported by Oliver et al. that both MMR‐proficient and MMR‐deficient CRC cells exhibited 5‐FU resistance after a period of treatment, suggesting that 5‐FU chemoresistance in CRC cells may be independent of MMR status. Nevertheless, MMR genes human homolog 1 (hMLH1), a component of MMR, was upregulated in an MMR‐deficient cell line, which indicated a potential involvement in 5‐FU sensitivity. 199
MMR deficient–associated microsatellite instability has been linked to cisplatin resistance in clinical germ cell tumor sample, 200 whereas in ovarian cancer, cisplatin resistance is generally related to homologous recombination repair (HRR) for DNA damage. The BRCA1 and BRCA2 tumor suppressor genes are critical for the HRR of DNA double‐strand breaks by the HRR pathway; therefore, BRCA1/BRCA2‐mutated ovarian cancers are usually more sensitive to cisplatin. 201 However, acquired secondary mutations of BRCA1/2 induced by long‐term cisplatin exposure can reconstruct the function of BRCA1/2 and enhance DNA repair, resulting in resistance to cisplatin 200 as well as inhibitors of DNA repair poly (ADP‐ribose) polymerase (PARP). 202 Recently, another DNA repair–related protein, actin‐like protein ACTL6A, has been reported to promote DNA lesion induced by cisplatin damage through the SWI/SNF chromatin remodeling complex in several types of cancers, suggesting a novel cisplatin‐resistance mechanism. As ACTL6A is also a component of NuA4/TIP60 histone acetylase, treatment with a histone deacetylase inhibitor can be useful to attenuate this resistance mechanism and resensitize tumor cells to cisplatin. 203
In addition to gene mutations that activate DNA repair genes, epigenetic regulations, including DNA methylation, histone modifications, and miRNAs, can all affect the expression of DNA repair genes, contributing to drug resistance.
DNA methylation can suppress gene expression. In the context of DNA repair–associated drug resistance, DNA methylation has been implicated in the regulation of key DNA repair genes, such as O 6‐methylguanine‐DNA methyltransferase (MGMT), BRCA1/2, and MLH1. MGMT promoter methylation is associated with a loss of MGMT protein expression and activity in the tumor and has been shown to correlate with a better outcome of therapy in several studies. 204 , 205 Consequently, the methylation status of the MGMT promoter has emerged as a useful biomarker for predicting the treatment response of glioblastoma (GBM) patients to methylating chemotherapeutic agents like temozolomide. 206 In breast cancer, the hypermethylation of the BRCA1 promoter has been associated with decreased BRCA1 expression and sensitivity to DNA‐damaging drugs and PARP inhibitors. 207 In contrast, the hypermethylation of the MLH1 promoter has been associated with decreased MLH1 expression and DNA repair capacity in cancer cells, which can increase the accumulation of somatic mutation in cancer cells and induce drug resistance. 208 These findings highlight the complex interplay among DNA methylation, DNA repair, and cancer resistance and suggest that targeting DNA methylation may be a promising strategy for overcoming drug resistance in cancer.
Histone modifications also play an important role in DNA repair and cancer resistance. A recent study demonstrated that, in high‐grade serous ovarian carcinoma cell lines and patient‐derived xenograft models, the upregulation of histone methyltransferases EHMT1 and EHMT2 are responsible for the trimethylation of histone H3 lysine 9 (H3K9me3) and contribute to PARP inhibitor resistance through inhibiting the expression of the tumor suppressor gene CDKN1A, which is involved in the regulation of the cell cycle and DNA repair. 209 However, another study showed that the loss of the histone methyltransferase, enhancer of zeste homolog 2 (EZH2), led to decrease in global H3K27 methylation, resulting in the upregulation of genes associated with cisplatin resistance and the downregulation of genes involved in DDR. This study also showed that treatment with EZH2 inhibitors could reverse chemotherapy resistance in TGCTs, indicating the potential for epigenetic therapies in treating drug‐resistant tumors. 210
MiRNAs and LncRNAs have been implicated in the regulation of multiple pathways, including DNA repair. For example, miR‐15a and miR‐16 are found to downregulate the expression of B‐lymphoma Moloney murine leukemia virus insertion region‐1 protein (BMI1), a protein involved in the ubiquitin‐mediated DNA repair pathway, thereby decreasing DNA repair capacity and increased sensitivity of breast cancer cells to doxorubicin treatment. 211 Upregulation of LncRNA HOTAIR has been observed in breast cancer cells following radiation therapy, which promotes DNA repair and radioresistance by interacting with the EZH2 protein. 212 These findings have implicated that the specific miRNAs and LncRNAs could serve as biomarkers for predicting therapeutic response in cancer patients and could be potential target to overcome drug resistance.
2.6. Inhibition of cell death
Cancerous cells are known to highly regulate the apoptotic pathways, and apoptosis plays different roles in tumor: eliminates infected cells from the human body, assists the functioning of the immune system, as well as contributes to the maintenance of homeostasis. 213 Generally, those signals induce the activation of effector caspases that mediate intracellular signaling, resulting in DNA fragmentation. Caspase mutations occurring in tumor therapy chemoresistance have been widely reported. For example, caspase‐8 mutations are mainly detected in gastric cancers, and the procaspase‐8Q482H mutation abrogates apoptosis by leading to the dimerization attenuation of the procaspase‐8 protein monomers, resulting in resistance to chemotherapy. 214 The intrinsic apoptosis pathway by mitochondrial damage is activated through numerous exogenous or endogenous stimuli that induce DNA damage. 215 The activation of this pathway involves the activation or inhibition of proteins from the B‐cell lymphoma 2 (Bcl‐2) family, and multiple drugs are known to induce apoptosis of cancerous cells through this mechanism. For example, small molecule RAF kinase inhibitor sorafenib induces apoptosis of AML through the downregulation of Mcl‐1 (an anti‐apoptotic protein) and the upregulation of BIM (an activator of the intrinsic pathway). 216 Previous studies (in vitro and in vivo) have suggested that upregulating of Bcl‐2 in breast cancer cells promotes metastasis to the lung in mice by the EMT process and triggers resistance to chemotherapy (PD168393, a specific inhibitor of EGFR; AG490, an inhibitor of JAK2). 217 , 218 In the treatment of hematological malignancies, the aim to integrate the application of Bcl‐2 inhibitors has led to further research and development of Bcl‐2 selective and dual Bcl‐2 and Bcl‐xL inhibitors. Similar to ABT‐199 (the first‐ever Bcl‐2 inhibitor known as Venetoclax), Navitoclax (ABT‐263) is one of the most studied dual inhibitors; it acts as a BH3‐mimetic, permitting the release of BH3‐only proteins and further MOMP by Bax/Bak activation. 219 One clinical trial detected the effect of ABT‐263 in 118 chronic lymphocytic leukemia (CLL) patients and reported partial responses in 34.6% of patients in phase I trials. 220 Rituximab, a monoclonal antibody targeting the protein CD‐20 was combined with ABT‐263 and showed a statistically significant overall response rate of 70% in untreated patients with CLL compared to rituximab alone. 221 However, because of the character of Bcl‐xL in platelet survival, a main concern with ABT‐263 is the negative effect: inducing thrombocytopenia in a dose‐dependent manner. Furthermore, the additional concern is regarding the emergence of tumor cell resistance to this agent, through the upregulation of Mcl‐1 phosphorylation and further sequestration of the proapoptotic protein Bim. 222 Overall, the clinical use of ABT‐263 is under restriction, and further studies are necessary before its approval.
Autophagy is a conserved and tightly regulated catabolic self‐degrading mechanism whereby cells keep homeostasis and respond to stress through recycling damaged cellular organelles, proteins, and other cellular components. 223 In tumors, autophagy is a double‐edged sword, and its anti‐ or pro‐tumorigenic character relies on the oncogenic context and stage of tumorigenesis. 224 In the early stages of tumorigenesis, autophagy exhibits tumor suppression function by removing aggregated and misfolded proteins, damaged organelles, limiting cell growth, necrosis, and chronic inflammation. 225 Upon advanced periods of tumor progression, autophagy helps to keep tumor cell survival and increases cellular resistance by conferring stress tolerance by providing the recycling substrate to promote cancer cell survival and contributes to cancer progression and drug resistance. 226 For example, in ovarian cancer, 227 leukemia, 228 hepatocellular carcinoma (HCC), 229 and CRC, 230 the increased levels of basal autophagy contribute to tumor cell growth and increase aggressiveness. Another study showed that autophagy was activated in mice with Myc‐induced lymphomas when treated by chemotherapy; autophagy inhibition sensitizes tumor cells to cell death induced by chemotherapeutic agents, suggesting that autophagy induction leads to a self‐defense effect in this case. 231 Several barriers result from TKI resistances that emerge in a later period of tumor attribute to the mutation in the kinase domain or function of basal autophagy as housekeeping as well as the regulator of RTK activity, which challenges effective treatment against cancer targeted therapy. Therefore, the combination of autophagy modulators with TKIs has been considered in cancer therapy. For example, in NSCLC, erlotinib and gefitinib can induce a high level of autophagy as a drug resistance and cytoprotective mechanism that was accompanied by the suppression of the PI3K/AKT/mTOR signaling cascade. In addition, autophagy inhibition by the pharmacological inhibitor, chloroquine, and siRNAs targeting autophagy‐related gene 5 (ATG5) and ATG7, enhances the erlotinib and gefitinib cytotoxicity. 232 Gastrointestinal stromal tumors (GISTs), in which the activation of CD117, stem cell growth factor receptor, or platelet‐derived growth factor receptor‐α mutations comes up, rarely answer to only‐imatinib mesylate (IM) treatment. IM triggers autophagy as a survival mechanism in quiescent GIST cells that result in acquired resistance and ineffective treatment. Nevertheless, the combination therapy by which IM was used with siRNAs targeting ATGs potentiates GIST cytotoxicity. 233 Another study revealed that IM induces autophagy in chronic myeloid leukemia by induction of ER stress in a different way from apoptosis induction and the combination therapy with autophagy inhibition using either siRNA targeting ATGs or pharmacological inhibitors promotes cell death. Moreover, other TKIs like dasatinib or nilotinib, combined with autophagy suppression treatment, exhibited similar effects. 234
Ferroptosis is an intracellular iron‐dependent mechanism of cell death that is distinct from apoptosis, necrosis, and autophagy. 235 Extensive preclinical and clinical studies have concentrated on conquering drug resistance, and inducing ferroptosis has been proven to reverse drug resistance. According to the theoretical framework for initiating the process of ferroptosis, there are three diverse pathways to reverse chemotherapy resistance: the canonical glutathione peroxidase 4 (GPX4)‐regulated pathway, iron metabolism pathway, and lipid metabolism pathway. 236 Genetic inhibition of GPX4 can induce tumor cell ferroptosis and suppress tumor growth in vivo. 237 In addition, the inactivation of GPX4 results in the accumulation of phospholipid hydroperoxides to induce cell membrane damage and ferroptotic death. 238 In GBM, androgen receptor ubiquitination induced by the curcumin analog was found to inhibit GPX4, generating ferroptosis and reversing temozolomide resistance. 239 In CRC, elevated kinesin family member 20A (KIF20A) expression was associated with oxaliplatin resistance, cellular ferroptosis may be induced by disturbing the KIF20A/NUAK1/GPX4 signaling pathway to reverse the resistance of CRC to oxaliplatin. 240 Cystine/glutamate antiporter (xCT) system, as an essential component of the GPX4‐regulated pathway, has also been revealed to be capable of triggering ferroptosis. A study reported that erastin and sulfasalazine (xCT system inhibitors) could induce head‐and‐neck cancer cell ferroptosis and overcome cisplatin resistance. 241 Another study showed that in gastric cancer, inducing ferroptosis by restraining Nrf2/Keap1/xCT signaling, can sensitize cisplatin‐resistant cells to cisplatin treatment. 242 Ferroptosis is defined as an iron‐catalyzed form of regulated necrosis. 243 Upregulation of cellular labile iron pool (LIP) leads to increase vulnerability to ferroptosis. Du et al. reported that tumor cell ferroptosis contributed to catastrophic LIP accumulation after dihydroartemisinin treatment in pancreatic ductal adenocarcinoma, and dihydroartemisinin treatment could overcome cisplatin resistance by triggering ferroptosis. 243 Divalent metal transporter 1 (DMT1) and secreted glycoprotein lipocalin‐2 are both key proteins in regulating iron homeostasis. Turcu et al. found that elevated cellular LIP caused by DMT1 inhibition induced ferroptosis, thereby wiping out breast cancer stem cells (CSCs) and reversing MDR. 244 A recent report by Chaudhary et al. indicated that targeting lipocalin‐2 overcame 5‐FU resistance in CRC by elevating intracellular iron levels, which in turn resulted in tumor cell ferroptosis. 245 The lipid metabolism pathway is also tightly associated with cell vulnerability to ferroptosis. Acyl–CoA synthetase long‐chain family member 4 (ACSL4), an important participant in ferroptosis execution, is involved in producing phospholipid hydroperoxides in an enzymatic manner. 246 In pancreatic cancer, the activation of ACSL4 conquered gemcitabine resistance by inducing ferroptosis. 247
Recently, Tsvetkov et al. revealed a previously uncharacterized cell death mechanism termed cuproptosis. 248 Cuproptosis a mechanism distinct from all other known manners of regulated cell death, such as apoptosis, pyroptosis, necroptosis, and ferroptosis. Cuproptosis is regulated by protein lipoylation in the citric acid (TCA) cycle, where lipoylation is necessary for enzymatic function. 249 , 250 Their research illuminates the connection between the sensitivity to copper‐mediated cell death and mitochondrial metabolism: respiring. TCA‐cycle active cells have upregulated levels of lipoylated TCA enzymes (particularly, the pyruvate dehydrogenase complex), and the lipoyl moiety acts as a direct copper binder, inducing the aggregation of lipoylated proteins and destabilization of Fe–S cluster proteins, ultimately leading to proteotoxic stress and cell death. 248 Genetic variation in copper homeostasis leads to serious disease, and copper chelators and copper ionophores have been proposed as anticancer agents. 251 , 252 , 253 , 254 For genetic disorders of copper homeostasis like Menke's disease and Wilson's disease, copper chelation is an effective treatment. 255 However, in tumors, copper ionophores like elesclomol have been tested in clinical trials, but neither the benefit of a biomarker of the appropriate patient population nor an understanding of the drug's mechanism of action is considered in such testing. For example, a phase III combination clinical trial of elesclomol in melanoma patients indicated the absence of efficacy in this unselected population, yet a post hoc analysis of samples with low plasma lactate dehydrogenase (LDH) levels displayed evidence of antitumor activity. 256 Low LDH represents a higher cellular dependency on mitochondrial metabolism, in accordance with Tsvetkov's finding that cells more reliant on mitochondrial respiration are nearly 1000‐fold more sensitive to copper ionophores than cells going through glycolysis. Considering the distinct mechanism of cuproptosis from other forms of cell death, deeply understanding how cuproptosis is initiated, progressed, and ultimately executed may exhibit great significance for specific therapeutic interventions and workable combination therapies. 257
2.7. Regulation from tumor microenvironment
How tumor cells react to therapy not merely relies on the genomic aberrations they harbor but also is mediated by the TME. 258 TME is a complicated element of tumors and is quite heterogeneous. 259 The significant character of the TME in changing tumor activity has been stated by plentiful studies uncovering how the TME can influence the malignant behavior and therapeutic resistance of the tumor cells 260 (Figure 3). The TME contains a multitudinous cellular and acellular milieu; diverse stromal and immune cells are recruited to develop and maintain such self‐sustained circumstances. 261 The extracellular matrix (ECM), including proteins like laminins, fibronectin, proteoglycans, vitronectin, tenascin‐C, and collagen, devotes to the majority component of the TME and is vital for the maintenance of TME and the induction of cellular adhesion, migration, invasion, and metastasis. 262 Moreover, the composition and organization of ECM can also influence the sensitivity to drug therapy. For example, it has been reported that ECM proteins (laminin, fibronectin, and vitronectin) mediated cell adhesion‐mediated drug resistance (cilengitide, an integrin inhibitor, and/or carmustine, an alkylating chemotherapy) in glioblastoma (GBM). Enrichment of the above proteins in the TME facilitates GBM cell proliferation via integrin αv‐mediated FAK/paxillin/AKT signaling cascade and suppresses p53‐involved tumor apoptosis. 263 In breast cancer, cancer‐associated fibroblasts (CAFs) promote drug resistance by increasing hyaluronan production. 264 Another study showed that the inhibition of the β1 integrin activity by monoclonal antibody AIIB2 markedly promotes radiotherapy efficacy and elevates sensitivity to HER2‐targeting agents of breast cancer cells. 265 , 266 In melanoma, CAFs promote cell metastasis and drug resistance by upregulating the level of matrix metalloproteinase 1 (MMP1) and MMP2. 267 As a significant aspect of the TME, the cell‐to‐ECM interaction and cellular crosstalk induce the release of soluble factors (like, angiogenic factors and chemokines) in charge of ECM remodeling and immune evasion, which further bring about therapy resistance. For example, the interaction between α4β1 integrin on cancer cells and fibronectin induce drug resistance in AML and CLL through the PI3K/AKT/BCL2 signaling pathway. 268 , 269 Stromal cell–derived factor 1 (SDF1) can interplay with CXC motif chemokine receptor type 4 (CXCR4) and activate AKT and ERK1/2 signaling pathways, resulting in antiapoptotic effects and contributing to tumor cell survival in CLL cells. 270 In AML cells, CXCR4 activation induced by SDF1‐mediated leads to resistance to cytarabine through reducing miRNA let‐7a and facilitating transcriptional activation of MYC and Bcl‐xl. 271 In CRC, CXCR4 is upregulated in chemoresistant tumor cells, and lymph node–derived stromal cells enhance resistance to oxaliplatin and 5‐FU via an SDF1/CXCR4 dependent manner. 272
FIGURE 3.
Adaptive mechanisms of cancer cell survival and cancer resistance driven by tumor microenvironment (TME). The TME is important for cancer resistance; the cancer cells within the TME can undergo a series of adaptive changes, such as various cellular components can complement the growth signal of cancer cells, combining with angiogenesis to promote cell survival and resistance. The immunosuppression caused by the TME prevents immune cells from killing cancer cells. The induction of the TGF‐β signaling and the release of prostaglandin E2 (PGE2) resulting in further augmentation of self‐renewal and plasticity of cancer stem cells (CSCs). Source: This figure was created with Biorender.com.
Exosomes, intraluminal vesicles of multivesicular bodies with a diameter of 30–100 nm, are ubiquitously present in most body fluids like blood, cerebrospinal fluids, urine, saliva, and lymphatic fluid. 273 , 274 Exosome contents not only reflect the composition of the donor cell but also mirror a regulated sorting mechanism. 275 A complicated of multifarious proteins, including ECM proteins, enzymes, transcription factors, receptors, lipids, and nucleic acids (DNA, mRNA, and miRNA) inside and on the surface of the exosomes constitute their content. 276 , 277 Extensive evidence has shown that exosome‐mediated factors can promote tumorigenesis, metastasis, and therapeutic resistance of cancer cells via intercellular communication within TME. 273 , 278 , 279 Tumor‐derived exosomes (TDEs) involve in the initiation, development, and progression of diverse tumor courses, including angiogenesis, TME remodeling, metastasis, and therapy resistance. 273 , 279 , 280 TDEs induce the differentiation of various kinds of TME cells to CAFs that are the main cell population of TME in the majority of tumors; thus, exosomes play a vital role in ECM remodeling and TME reprogramming. 281 , 282 Exosomes derived from CAFs contain different molecules such as miRNAs and growth factors that have distinct influences on the target cells of TME. For example, the gemcitabine therapy of CAFs in pancreatic cancer stimulates the expression of miR‐146a and snail and prolonged exosome secretion, thereby promoting epithelial cell proliferation. 283 Drug‐resistant tumor cells can pack the chemotherapeutic drugs in exosomes and shuttle therapeutic agents out of cancer cells. 283 Besides, the delivery of exosomal cargo containing proteins, mRNA, and miRNA to cancerous cells is related to therapeutic resistance. 284 , 285 For instance, GBM cell–derived exosomes, which comprise MET‐protein and tyrosine phosphatase receptor type Z (PTPRZ1) fusion proteins, obtain temozolomide resistance via EMT. 286 TDEs from gemcitabine‐resistant pancreatic cancer cells induce chemoresistance by trapping MRP5 and P‐gp or letting gemcitabine to flow back to TME. 287 Exosomes derived from MCF7WT breast cancer cells contain P‐gp and ubiquitin C‐terminal hydrolase‐L1 proteins that are able to induce doxorubicin resistance by elevating the level of P‐gp. 288 TDEs promote platinum resistance by upregulating EMT markers and changing TGF‐β/SMAD signaling pathway in ovarian cancer cells, and exosomes from epithelial ovarian cancer A2780 platinum‐resistant cells attain resistance via promoting EMT. 289 Exosome‐transmitted miR‐567 reverses trastuzumab resistance in breast tumor cells by inhibiting ATG5. 290 In MDA‐MB‐231 and MCF‐7 breast cancer cells, exosome‐mediated miR‐155 induces chemoresistance via upregulating EMT markers and targeting CCAAT/enhancer‐binding protein β (C/EBP‐β), TGF‐β, and forkhead box O 3α (FOXO‐3α) mRNA. 291 In HER2‐positive breast cancer, lncRNA‐SNHG14 induces exosome‐mediated trastuzumab resistance by targeting the apoptosis regulator Bcl‐2/BAX signaling. 292 In NSCLC, exosome‐mediated miR‐21 delivery induces the upregulation of p‐AKT level, thereby resulting in increased gefitinib resistance. 293 Exosomes containing miR‐32‐5p generate MDR in HCC cells by inhibiting PTEN, activating PI3K/AKT signaling cascade and promoting angiogenesis and EMT process. 294 HCC‐derived exosomes containing miR‐221 induce sorafenib resistance through modulating apoptosis inhibition and caspase‐3 activity. 295 Considering that exosomes are involved in various pathophysiological conditions, understanding the molecular mechanisms underlying exosome biogenesis and chemoresistance will benefit in developing novel therapeutics targeting exosome‐mediated tumorigenesis, progression, and chemoresistance.
One of the important functions of TDEs is to induce tumor vascular development. Angiogenesis, a multistep course by which cancers form new vasculature, is essential for tumor progression. 296 Since Judah Folkman revealed the significant character of vascular networks for the proliferation and progression of solid tumors, establishing angiogenesis as a therapeutic target has become a focal aim. 297 Up to now, antiangiogenic therapy has emerged as significant targeted therapeutic and diverse antiangiogenic agents are currently used in combination with other chemotherapeutic drugs for the treatment of various malignant tumors. Nowadays, over 11 antiangiogenic drugs have been approved by the U.S. FDA, including bevacizumab, aflibercept, sorafenib, ramucirumab, sunitinib, pazopanib, axitinib, vandetanib, Lenvatinib, and regorafenib. 298 However, the treatment is in doubt on the grounds of sustainable efficacy, side effects, off‐target toxicities, and therapy resistance. 299 , 300 Clinical and experimental studies have illustrated that cancers employ compensatory/alternative/bypass angiogenic pathways and other adaptive mechanisms for their sustained growth, proliferation, and metastasis, after undergoing a therapy episode(s) with antiangiogenic drugs. That is, various compensatory signaling pathways driving tumor growth and metastasis invariably become the underlying cause of tumor refractoriness. 301 Several preclinical and clinical studies have linked poor performance and resistance to antiangiogenic agents to the activation of a range of compensatory angiogenic signaling pathways and various angiogenic factors that support the angiogenic bypass mechanism. 302 The vascular endothelial growth factor (VEGF)/VEGF receptor signaling pathway is the most promising angiogenic target due to its pivotal role in angiogenesis and tumor growth. 303 , 304 Previous studies have suggested that revascularization appears even after blocking VEGF signaling pathways because of the activation of redundant angiogenic pathways.
Serial evidence accumulated in recent years linked the function of various compensatory/alternative/bypass angiogenic canonical mechanisms sustaining the progression of cancers while exposed to antiangiogenic drugs. 299 , 300 , 305 The current experimental, clinical, and epidemiological data has definitely outlined at least four potential different mechanisms, which can be considered for an explanation of evasive resistance to antiangiogenic therapies. The first mechanism refers to the activation and/or upregulation of compensatory pro‐angiogenic signaling pathways within the cancer. 306 The second mechanism is mainly driven by myeloid/stromal cells, which compensate for the requirement of the VEGF‐mediated pathway, thereby promoting tumor angiogenesis. 305 The third manner is attributed to the dual role of pericytes; first, in establishing the increased pericyte coverage of the tumor vasculature, and second, their potential angiogenic attributes, both serving as escape mechanisms from VEGF‐mediated angiogenesis. 307 The fourth way is associated with remodeling and accessing normal vasculature for the invasion and metastasis of tumors in lieu of obligate neovascularization. 306
As an important component of TME, the immune system plays a critical role in regulating the response of cancer cells to therapies, particularly immunotherapies. Immune‐targeted therapies approved for cancer include monoclonal antibodies against immune checkpoints like cytotoxic T‐lymphocyte‐associated protein 4 (CTLA4), PD‐1, and programmed cell death ligand 1 (PD‐L1), which aim to modulate the antitumor T‐cell immune response. The resistance to immune‐checkpoint inhibitors (ICIs) has been attributed to genomic and nongenomic mechanisms that are influenced by the tumor–host–microenvironment relationship. 308 One mechanism is the upregulation of alternative immune checkpoint pathways. Koyama et al. revealed that, in mouse xenograft models with lung adenocarcinoma, other immune checkpoint proteins, including T‐cell immunoglobulin mucin 3, lymphocyte activation gene 3, and CTLA‐4, were upregulated after PD‐1 blockade therapy. The increased expression of alternative immune checkpoints in PD‐1 antibody‐bound T cells can inhibit T cell activity and confer resistance to PD‐1 blockade. 309 Other mechanisms include the presence of immunosuppressive cells, such as regulatory T cells (Tregs) or myeloid‐derived suppressor cells (MDSCs), which can inhibit T cell function. 310 Besides, the presence of stromal cells in TME, such as CAFs, has also been found to be associated with suppressive CD8+ T cell infiltration and insensitivity to αPD‐L1 antibody plus αCTLA‐4 antibody dual immune checkpoint blockade. 311
Combination treatments appear to be the promising strategy to overcome resistance to immune checkpoint inhibitors. Combinations of ICIs with radiation, chemotherapy, targeted drug, or tumor vaccines have been proposed and tested. Radiation can trigger an adaptive mechanism to increase the expression of PD‐L1 on tumor cells. 312 Additionally, radiation may increase the depth and duration of immune responses by promoting a more diverse adaptive antitumor immune response. As reported by Victor et al., the combination of radiation with dual checkpoint blockade (anti‐CTLA‐4 and anti‐PD‐1) can enhance tumor control and survival in a preclinical model of melanoma, where radiation prime immune response by increasing T cell receptor repertoire of intratumoral T cells and expanding peripheral clones. 313 Combination with chemotherapy aims to assist ICI treatment by releasing neoantigens or modulating the TME via depleting Tregs and MDSCs. 314 However, it has been reported that the efficacy of this combination therapy is model (cancer type)‐dependent and regimen‐type‐dependent. Combination with chemotherapeutic drugs is not always able to tune the TME or enhanced antitumor efficacy. Even with the effect of depleting MDSCs, the combination may not be beneficial to enhance treatment responsiveness. 314 Additional studies combining single agents with ICIs are still required to better understand potential interactions between drugs and the heterogeneity of TME. Compared to the less predictable ICI–chemotherapy combination, the combination of TME‐targeted drugs with immune checkpoint blockade is considered a relatively logical approach to enhance the stimulation of antitumor immune response. TME‐targeted therapies, such as angiogenesis inhibitors, stromal cell depletion agents, and immunomodulatory agents, can modulate the TME and enhance the immune response against the tumor. Combining these agents with ICIs can lead to synergistic effects of blocking inhibitory signals in T cells and activating the immune response, thereby improving treatment outcomes. 315 For example, the VEGF inhibitor bevacizumab can augment intratumoral CD8+ T cell infiltration through vascular normalization and endothelial cell activation, thereby potentiating PD‐L1 checkpoint inhibition with PD‐L1 inhibitor atezolizumab. 316 This combination has shown promising anticancer efficacy and has been granted FDA approval for the treatment of patients with advanced unresectable or metastatic HCC. 317
Combining oncolytic vaccines with ICIs is an emerging approach to overcome ICI resistance. Oncolytic vaccines are a type of oncolytic virus that are engineered to express tumor‐specific antigens, which can stimulate an immune response against the cancer cells. 318 As ICI‐resistant tumors are with a suppressed immune microenvironment, the application of an oncolytic virus can reshape TME and assist ICIs to boost the immune system leading to a stronger antitumor response. 319 This combination therapy has shown promising results in preclinical and clinical studies in a variety of cancers, including melanoma, breast cancer, and bladder cancer. 320 , 321 , 322
Recent studies have shown that bacteria can reside within tumors and play a role in controlling cancer response to therapy by modulating the TME. 323 For example, Pseudomonas aeruginosa and Escherichia coli are two types of bacteria found to colonize tumors in various types of cancer, including lung, breast, and pancreatic cancer. 324 , 325 , 326 These bacteria can metabolize chemotherapy drugs such as gemcitabine, reducing their concentration within the tumor and rendering them less effective. 327 , 328 Moreover, certain commensal bacteria or pathogens within the TME may interact with immune cells and alter the production of cytokines and chemokines, which are involved in the immune response to cancer. 329 , 330 The influence of gut microbiota on cancer resistance has also gained extensive attention. A recent study found that in mice and patients with castration‐resistant prostate cancer, an adaptive change in the gut commensal microbiota was observed after ADT: There was an expansion of commensal microbiota species that can convert androgen precursors to produce androgens, which were absorbed into the systemic circulation thus inducing castration or endocrine therapy resistance. 331 This type of resistance could be delayed by applying antibiotics to reduce gut microbiota or could be reversed by fecal microbiota transplantation hormone‐sensitive prostate cancer patients, 331 implicating two possible ways to overcome bacterial‐mediated resistance. Although using antibiotics can directly eradicate the bacteria within the TME, this approach may have limitations due to the risk of antibiotic resistance. 328 Another approach is to develop drug delivery systems specifically targeting the tumor cells while avoiding bacterial‐mediated metabolism. 332 , 333 For example, nanotechnology‐based drug delivery systems can be engineered to release drugs only in the tumor cells, minimizing the exposure of the drug to the bacteria. 323 , 332 The presence of bacteria within the TME and possible influences from gut microbiota are thought to be significant factors contributing to treatment resistance in cancer. Understanding the complex interactions between bacteria and tumors is essential for developing effective cancer treatments to overcome this resistance.
2.8. Tumor plasticity
CSCs are a small population of cells within a tumor that possess stem cell–like properties and are thought to be responsible for tumor initiation, growth, and metastasis. 334 , 335 These cells have been identified in various cancers, including breast, colon, and GBM. CSCs are less sensitive to chemotherapy and radiation therapy due to their ability to activate DNA damage repair and antiapoptotic signaling pathways, which may lead to incomplete tumor eradication and recurrence after treatment. Based on these findings, the researchers put forward the “CSC theory” and found evidence of the existence of CSCs in some cancer tissues. 336 , 337 CSCs can resist therapy mainly because of the overexpression of MDR transporters that mediate drug efflux, the more active DNA repair capacity, and the tendency to form new microvascular for the tumor. 338 These characteristics enable CSCs to tolerate treatment, maintain the tumor with nutrients and oxygen, and rapidly repopulate the tumor. This mechanism is similar to that of normal tissue stem cells in response to trauma, which may explain that the bladder CSCs actively contribute to the cause of chemotherapy resistance after several cycles of chemotherapy. 339 , 340 Targeting this trauma response of CSCs can become a new therapeutic intervention. CSCs between different treatment cycles actively regenerate and respond to chemotherapy‐induced injury or apoptosis, much as normal tissue stem cells respond to trauma‐induced damage. Dying cells release a metabolite called prostaglandin E2 (PGE2) that stimulates proliferation and causes CSC to repopulate cancers shrunk by chemotherapy. 341 , 342 In normal cells, this is an integral part of the wound repair process, where PGE2 induces the regeneration of tissue stem cells. Ironically, PGE2 induces more CSC regeneration between chemotherapy cycles in cancers. An essential characteristic of CSC is maintaining self‐renewal ability, and the mechanism may be one key factor in promoting cancer development and metastasis. 343 , 344 CSC theory helps people understand the occurrence and development of cancers. In addition, CSCs can activate signaling pathways that promote cell survival and proliferation, such as the Wnt/β‐catenin and Notch pathways. 345 , 346 , 347 Therefore, understanding the mechanisms underlying CSC‐mediated resistance to therapy is essential for developing effective cancer treatments. It provides a new perspective on cancer development, metastasis, drug resistance, and recurrence. Leukemia therapy targeting LSC (leukemia stem cells) has achieved good clinical efficacy, and beneficial research has also been carried out in solid cancers. 348
EMT is one of the essential mechanisms of cancer metastasis. The latest research suggests that cancer cells with dual characteristics of EMT and stem cells are the key to cancer metastasis and drug resistance. 349 There is a direct link between EMT and CSCs, but the molecular mechanism is still unclear. EMT phenomenon refers to a reversible process in which relatively stable epithelial cells lose cell polarity and intercellular adhesion and transform into spindle‐shaped mesenchymal cells with migration ability. It is ubiquitous in epithelial cancer cells. Its essential feature is the loss of the expression of epithelial markers, such as E‐cadherin, β‐catenin, tight junction protein, and epithelial cell adhesion molecules on the cell membrane of cancer cells, 350 , 351 , 352 , 353 and meanwhile obtaining a mesenchymal phenotype with the increased expression of vimentin, N‐cadherin, fibronectin, and β1 and β3 integrins. 354 , 355 , 356 The mechanism of EMT is mainly due to changes in the epithelial cells themselves or the surrounding microenvironment, leading to the activation of a series of signal transduction pathways, and related transcription factors in the nucleus play a regulatory role. 357 Precise intracellular signaling mechanisms regulate different degrees of epithelial cell transformation. Various extracellular signals activate different nuclear transcription factors by binding to specific receptors on the cell surface. The common feature of these transcription factors is that they can recognize the DNA binding sequence of the E‐box motif on the target gene promoter, thereby regulating the expression of the target gene and initiating EMT. 358 Loss of E‐cadherin expression is currently considered the most prominent feature of EMT. Decreased E‐cadherin levels can lead to decreased cell adhesion and make cells easily invade and metastasize. 351 Cancer cells lose some of the characteristics of epithelial cells and acquire the characteristics of mesenchymal cells through EMT, so cancer cells can obtain stronger invasion and migration capabilities. 359 , 360
Snail is involved in triggering EMT during the progression of epithelial cancers and is a crucial point in the occurrence of EMT. Snail expression correlates with the reduced loss of E‐cadherin and acts as a direct repressor of E‐cadherin transcription. 361 Twist is another essential transcription factor that regulates EMT. Its mechanism of inducing EMT is to directly or indirectly bind the E‐cadherin promoter through the E‐box motif, thereby inhibiting the expression of E‐cadherin. 362 Snail/Twist1 knockout breast cancer model demonstrates that chemotherapy resistance is associated with EMT. 363 Studies have shown that EMT may be involved in drug resistance of cancer cells, such as lung, pancreatic, and breast cancer. 364 , 365 , 366 Numerous studies have shown that cancer cells develop resistance to carboplatin or paclitaxel by acquiring mesenchymal cell phenotype, indicating that EMT may be the instigator of chemotherapy resistance. 367 , 368 The latest research shows that cancers may have a type of partial EMT, which means the cancer cells have both epithelial and mesenchymal cell phenotypes, and these cancer cells are more aggressive than the others. 368 EMT has been described as a significant cause of EGFRi failure in EGFR‐mutated NSCLC. 369 Cellular transdifferentiation, a phenotypic shift from adenocarcinoma to squamous cell carcinoma or neuroendocrine carcinoma, occurs in 3%–14% of EGFR‐mutant NSCLC patients, and approximately 17% of prostate cancer failed with abiraterone/enzalutamide therapy. 370
From the discussion above, it is evident that targeting EMT processes or cellular plasticity has great potential to circumvent drug resistance. However, only a few compounds adesigned to inhibit the EMT process are currently in clinical trials. Inhibitors targeting Notch, TGF‐β, and Wnt signaling pathways are also promising candidates for inhibiting EMT and cellular plasticity. 371 TGF‐β heterogeneity in the cancer microenvironment creates rapidly dividing CSCs that accelerate cancer growth and others that invade surrounding healthy tissue and evade treatment. 372 , 373 For example, PF‐03446962 and galunisertib are antagonists designed to inhibit TGF‐β receptors, currently in phase I clinical studies in solid cancers (NCT00557856, NCT02423343). 374 Both PF‐03446962 and galunisertib inhibit the EMT program, thereby preventing cancer development. In addition, Wnt inhibitors such as ETC‐1922159 and OMP‐54F28 have been reported to inhibit the EMT program and are currently in phase I clinical trials in cancer (NCT02521844, NCT01608867). 375
3. PERSPECTIVES IN OVERCOMING CANCER RESISTANCE
Cancer chemotherapy is still a required treatment method for most cancer patients. However, cancer cells are prone to MDR, a significant problem limiting the efficacy of current chemotherapeutic drugs lacking selectivity. Many cancer patients have a remarkable curative effect in the early stage of chemotherapy. However, the drug resistance of cancer cells increases with the prolongation of treatment time, eventually leading to treatment failure. Anticancer strategies are constantly evolving, and targeted therapy drugs have encountered increasing resistance after the initial excitement, including primary and acquired resistance. Strategies to address drug resistance through combination therapy have made some progress, although considerable challenges have been faced, and the full potential of combination therapy has not yet been realized.
According to statistics, in the early 1990s, the main reasons for the failure of new drug development were concentrated in poor pharmacokinetics and limited biological activity. 376 , 377 With the introduction of absorption, distribution, metabolism, and excretion for predictive analysis and research applications, the clinical failure rate dropped from 40% to 10% in 2000. 378 With the accumulation of basic research on targets and signaling pathways and the advent of the era of genome sequences, the success rate of clinical development of targeted drugs is also increasing. For example, the familiar TKIs have become one of the most popular research directions. In recent years, with a series of significant breakthroughs in technological applications, such as the application of high‐throughput genomics to discover new targets, 379 , 380 the identification of molecular biomarkers, 381 , 382 and the progress of statistical methods in biological and chemoinformatics, 383 , 384 the progress and the success rate of research and development have been greatly improved. However, new problems are still emerging, with lacking clinical efficacy and increased toxicity still the main directions of failure in the second research and development phase.
Nanodrugs represent a promising approach to overcoming resistance to cancer. These drugs are engineered to be nanoscale size and have unique physicochemical properties that enable them to target cancer cells while minimizing off‐target effects. 385 Nanodrugs can be designed to overcome resistance by delivering drugs to cancer cells in more targeted and controlled manner and bypassing resistance mechanisms such as efflux pumps and DNA repair pathways. 386 , 387 , 388 One key advantage of nanodrugs is their ability to improve the pharmacokinetics and pharmacodynamics of drugs. 386 By encapsulating drugs within nanocarriers, the drug concentration within the tumor can be increased, improving drug efficacy. 332 Additionally, nanocarriers can protect the drug from degradation and clearance, leading to a longer half‐life and a sustained drug release. 386 , 389 Nanodrugs can also be engineered to overcome specific resistance mechanisms. For example, nanodrugs can target CSCs by incorporating specific ligands that bind to stem cell markers. 390 Nanodrugs can also be engineered to overcome bacterial‐mediated resistance by releasing drugs only within the TME and avoiding bacterial exposure. 328 By improving drug delivery, targeting, and overcoming specific resistance mechanisms, nanodrugs can potentially improve the efficacy of cancer treatment and patient outcomes. 391 However, further research is needed to optimize the design and delivery of nanodrugs and to ensure their safety and efficacy in clinical use.
Polytherapy can make cancer cells less likely to develop compensatory resistance mechanisms than a single or sequential drug regimen. There are many problems in the reversal of MDR with combination drugs, including clinical toxicity and pharmacokinetic interactions. The therapeutic range of each drug may be narrow when administered in combination, and there may be an overlap in toxicity. 392 In some cases, in addition to the expected overlapping toxicities, combination therapy has some characteristic toxicities. 393 , 394 We cannot accurately predict toxicity from preclinical models, which adds to the challenge of optimizing the toxicity–potency balance of drug combinations. Meanwhile, the pharmacokinetic interaction is the question of how to combine two or more drugs. 395 , 396 For example, lapatinib is a substrate and moderate inhibitor of CYP450‐3A4 and substantially reduces the clearance of drugs such as other CYP450‐3A4 substrates. 397 In the phase I clinical trial of lapatinib and pazopanib, investigators compared the historical pharmacokinetic parameters of the two drugs and concluded that the drug combination did not alter drug exposure. 398 However, in a more detailed phase II pharmacokinetic analysis, the combination indicated a significant drug–drug interaction contributing to the reduced efficacy of lapatinib in glioma patients. 399 , 400 In addition, the frequent use of antiepileptic drugs in this patient population also reduced pazopanib exposure, resulting in poorer patient outcomes. 401 This highlights the importance of detailed pharmacokinetic assessment for the combined evaluation of anticancer drugs. Combination therapies approved or in clinical trials in the recent 5 years are summarized in Table 1.
TABLE 1.
Cancer combination therapies approved or in clinical trials in the recent 5 years.
Combination therapy | Target cancer type | Approval date/Clinical trials process |
---|---|---|
Tremelimumab + durvalumab + platinum‐based chemotherapy | Adult patients with metastatic NSCLC with no sensitizing EGFR mutation or ALK genomic cancer aberrations | November 10, 2022 (NCT03164616) |
Brentuximab vedotin + doxorubicin + vincristine + etoposide + prednisone + cyclophosphamide | Pediatric patients 2 years of age and older with previously untreated high risk cHL | November 10, 2022 (NCT03755804) |
Cemiplimab‐rwlc + platinum‐based chemotherapy | Adult patients with advanced NSCLC with no EGFR, ALK, or ROS1 aberrations | November 8, 2022 (NCT03409614) |
Tremelimumab + durvalumab | Adult patients with uHCC | October 21, 2022 (NCT03298451) |
Durvalumab + gemcitabine + cisplatin | Adult patients with locally advanced or metastatic BTC | September 2, 2022 (NCT03875235) |
Darolutamide + docetaxel | Adult patients with mHSPC | August 5, 2022 (NCT02799602) |
Dabrafenib + trametinib | Adult and pediatric patients ≥6 years of age with unresectable or metastatic solid cancers with BRAF V600E mutation | Accelerated Approval June 22, 2022 (NCT02034110, NCT02465060, NCT02124772, original projected completion: October 21, 2028) |
Nivolumab + fluoropyrimidine‐ and platinum‐based chemotherapy | Patients with advanced or metastatic ESCC | May 27, 2022 (NCT03143153) |
Nivolumab + ipilimumab | Patients with advanced or metastatic ESCC | May 27, 2022 (NCT03143153) |
Ivosidenib + azacitidine (azacitidine for injection) | Newly diagnosed AML with a susceptible IDH1 mutation | May 25, 2022 (NCT03173248) |
Nivolumab + relatlimab‐rmbw | Adult and pediatric patients 12 years of age or older with unresectable or metastatic melanoma | March 18, 2022 (NCT03470922) |
Abatacept + a calcineurin inhibitor + methotrexate | Adults and pediatric patients 2 years of age and older undergoing HSCT | December 15, 2022 (NCT 01743131) |
Rituximab + chemotherapy | Pediatric patients (≥6 months to <18 years) with previously untreated, advanced stage, CD20‐positive DLBCL, BL, BLL, or mature B‐AL | December 2, 2022 (NCT01516580) |
Pembrolizumab + chemotherapy, with or without bevacizumab | Patients with persistent, recurrent, or metastatic cervical cancer whose cancers express PD‐L1 | October 13, 2021 (NCT03635567) |
Lenvatinib + pembrolizumab | Adult patients with advanced RCC | August 10, 2021 (NCT02811861) |
Pembrolizumab + chemotherapy as neoadjuvant treatment, and then continued as a single agent as adjuvant treatment after surgery | High‐risk, early‐stage, TNBC | July 26, 2021 (NCT03036488) |
Pembrolizumab + lenvatinib | Patients with advanced endometrial carcinoma | July 21, 2021 (NCT03517449) |
Daratumumab + hyaluronidase‐fihj + pomalidomide + dexamethasone | Adult patients with multiple myeloma | July 9, 2022 (NCT03180736) |
Pembrolizumab + trastuzumab + fluoropyrimidine‐ and platinum‐containing chemotherapy | Patients with locally advanced unresectable or metastatic HER2‐positive gastric or GEJ adenocarcinoma | Accelerated Approval May 5, 2021 (NCT03615326, original projected completion: September 30, 2024) |
Nivolumab + fluoropyrimidine‐ and platinum‐containing chemotherapy | Advanced or metastatic gastric cancer, GEJ, and esophageal adenocarcinoma | April 16, 2021 (NCT02872116) |
Isatuximab‐irfc + carfilzomib + dexamethasone | Adult patients with relapsed or refractory multiple myeloma | March 31, 2021 (NCT03275285) |
Pembrolizumab + platinum and fluoropyrimidine‐based chemotherapy | Patients with metastatic or locally advanced GEJ carcinoma | March 22, 2021 (NCT03189719) |
Melphalan flufenamide + dexamethasone | Adult patients with relapsed or refractory multiple myeloma | Accelerated Approval February 26, 2021 (NCT02963493, original projected completion: February 28, 2022) |
Nivolumab + cabozantinib | Patients with advanced RCC | January 22, 2021 (NCT03141177) |
Selinexor + bortezomib + dexamethasone | Adult patients with multiple myeloma | December 18, 2020 (NCT03110562) |
Margetuximab‐cmkb + chemotherapy | Adult patients with metastatic HER2‐positive breast cancer | December 16, 2020 (NCT02492711) |
Naxitamab + granulocyte‐macrophage colony‐stimulating factor | Pediatric patients 1 year of age and older and adult patients with relapsed or refractory high‐risk neuroblastoma in the bone or bone marrow | Accelerated Approval November 25, 2020 (NCT 03363373, original projected completion: September 30, 2027) |
Pembrolizumab + chemotherapy | Patients with locally recurrent unresectable or metastatic TNBC | November 13, 2020 (NCT02819518) |
Venetoclax + azacitidine + decitabine | Newly diagnosed AML in adults 75 years or older | October 16, 2020 (NCT02993523) |
Nivolumab + ipilimumab | Adult patients with unresectable malignant pleural mesothelioma | October 2, 2020 (NCT02899299) |
Carfilzomib + daratumumab + dexamethasone | Adult patients with relapsed or refractory multiple myeloma | August 20,2020 (NCT03158688) |
Tafasitamab‐cxix + lenalidomide | Adult patients with relapsed or refractory DLBCL | Accelerated Approval July 31, 2020 (NCT02399085, original projected completion: December 31, 2025) |
Atezolizumab + cobimetinib + vemurafenib | Patients with BRAF V600 mutation‐positive unresectable or metastatic melanoma | July 30, 2020 (NCT02908672) |
Oral decitabine + cedazuridine | Adult patients with MDS | July 7, 2020 (NCT02103478) |
Pertuzumab + trastuzumab + hyaluronidase–zzxf | Patients with HER2‐positive, locally advanced, inflammatory, or early stage breast cancer | June 29, 2020 (NCT03493854) |
Ramucirumab + erlotinib | Metastatic NSCLC with EGFR exon 19 deletions or exon 21 mutations | May 29, 2020 (NCT02411448) |
Atezolizumab + bevacizumab | Patients with unresectable or metastatic hepatocellular carcinoma | May 29, 2020 (NCT03434379) |
Nivolumab + ipilimumab + 2 cycles of platinum‐doublet chemotherapy | Patients with metastatic or recurrent NSCLC | May 26, 2020 (NCT03215706) |
Olaparib + bevacizumab | Adult patients with advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer | May 8, 2020 (NCT03737643) |
Ibrutinib + rituximab | Adult patients with CLL or SLL | April 21, 2020 (NCT02048813) |
Tucatinib + trastuzumab + capecitabine | Adult patients with advanced unresectable or metastatic HER2‐positive breast cancer | April 17, 2020 (NCT02614794) |
Encorafenib + cetuximab | Adult patients with metastatic CRC with a BRAF V600E mutation | April 8, 2020 (NCT02928224) |
Durvalumab + etoposide + either carboplatin or cisplatin | Patients with ES‐SCLC | March 27, 2020 (NCT03043872) |
Nivolumab + ipilimumab | Patients with HCC | Accelerated Approval March 10, 2020 (NCT01658878, original projected completion: July 31, 2024) |
Isatuximab‐irfc + pomalidomide + dexamethasone | Adult patients with multiple myeloma | March 2, 2020 (NCT02990338) |
Neratinib + capecitabine | Adult patients with advanced or metastatic HER2‐positive breast cancer | February 25, 2020 (NCT01808573) |
Atezolizumab + paclitaxel protein‐bound + carboplatin | Adult patients with metastatic NSCLC | December 3, 2019 (NCT02367781) |
Daratumumab + bortezomib + thalidomide + dexamethasone | Adult patients with multiple myeloma in newly diagnosed | September 26, 2019 (NCT02541383) |
Pembrolizumab + lenvatinib | Patients with advanced endometrial carcinoma | September 17, 2019 (NCT02501096) |
Selinexor + dexamethasone | Adult patients with RRMM | July 3, 2019 (NCT02336815) |
Daratumumab + lenalidomide + dexamethasone | Patients with newly diagnosed multiple myeloma | June 27, 2019 (NCT02252172) |
Polatuzumab vedotin‐piiq + bendamustine + a rituximab product | Adult patients with relapsed or refractory DLBCL | Accelerated Approval June 10, 2019 (NCT02257567, original projected completion: June 30, 2024) |
Lenalidomide + a rituximab product | Previously treated FL and previously treated MZL | May 28, 2019 (NCT01996865) |
Alpelisib + fulvestrant | Postmenopausal women, and men, with HR‐positive, HER2‐negative, PIK3CA‐mutated, advanced or metastatic breast cancer | May 24, 2019 (NCT02437318) |
Avelumab + axitinib | Patients with advanced RCC | May 14, 2019 (NCT02684006) |
Atezolizumab + carboplatin + etoposide | Adult patients with ES‐SCLC | March 18, 2019 (NCT02763579) |
Glasdegib + low‐dose cytarabine | Newly diagnosed AML in patients who are 75 years old or older | November 21, 2018 (NCT01546038) |
Brentuximab vedotin + chemotherapy | Previously untreated sALCL or other PTCL | November 16, 2018 (NCT01777152) |
Pembrolizumab + carboplatin + either paclitaxel or nab‐paclitaxel | Metastatic squamous NSCLC | October 30, 2018 (NCT02775435) |
Pembrolizumab + pemetrexed + platinum | Patients with metastatic, non‐squamous NSCLC | August 20, 2018 (NCT02578680) |
Ribociclib + an aromatase inhibitor | Pre/perimenopausal women with HR‐positive, HER2‐negative advanced or metastatic breast cancer | July 18, 2018 (NCT02278120) |
Ribociclib + fulvestrant | Postmenopausal women with HR‐positive, HER2‐negative advanced or metastatic breast cancer | July 18, 2018 (NCT02278120) |
Ipilimumab + nivolumab | Patients 12 years of age and older with metastatic CRC | Accelerated Approval July 10, 2018 (NCT02060188, original projected completion: July 31, 2024) |
Encorafenib + binimetinib | Patients with unresectable or metastatic melanoma with a BRAF V600E or V600K mutation | June 27, 2018 (NCT01909453) |
Bevacizumab + carboplatin + paclitaxel | Patients with epithelial ovarian, fallopian tube, or primary peritoneal cancer | June 13, 2018 (NCT00262847) |
Dabrafenib + trametinib for the adjuvant treatment | Patients with melanoma with BRAF V600E or V600K mutations | April 30, 2018 (NCT01682083) |
Nivolumab + ipilimumab | Previously untreated advanced renal cell carcinoma | April 16, 2018 (NCT02231749) |
Abiraterone acetate tablets + prednisone | Metastatic high‐risk castration‐sensitive prostate cancer | February 7, 2018 (NCT01715285) |
Pertuzumab + trastuzumab + chemotherapy | Patients with HER2‐positive early breast cancer | December 20, 2017 (NCT01358877) |
Dabrafenib + trametinib | Patients with metastatic NSCLC with BRAF V600E mutation | June 22, 2017 (NCT01336634) |
Abbreviations: ALK, anaplastic lymphoma kinase; AML, acute myeloid leukemia; B‐AL, B‐cell acute leukemia; BL, Burkitt's lymphoma; BLL, Burkitt‐like lymphoma; BRAF, B‐raf proto‐oncogene; BTC, biliary tract cancer; cHL, classic Hodgkin lymphoma; CLL, chronic lymphocytic leukemia; CRC, colorectal cancer; DLBCL, diffuse large B cell lymphoma; EGFR, epidermal growth factor receptor; ESCC, esophageal squamous cell carcinoma; ES‐SCLC, extensive‐stage small cell lung cancer; GEJ, gastroesophageal junction adenocarcinoma; HER2, human epidermal growth factor receptor 2; HSCT, hematopoietic stem cell transplantation; IDH1, isocitrate dehydrogenase 1; MDS, myelodysplastic syndromes; mHSPC, metastatic hormone‐sensitive prostate cancer; MZL, marginal zone lymphoma; NSCLC, non‐small cell lung cancer; PD‐L1, programmed cell death ligand 1; PIK3CA, phosphatidylinositol‐4,5‐bisphosphate 3‐kinase catalytic subunit alpha; PTCL, peripheral T‐cell lymphoma; RCC, renal cell carcinoma; ROS1, proto‐oncogene 1; RRMM, relapsed or refractory multiple myeloma; sALCL, systemic anaplastic large cell lymphoma; SLL, small lymphocytic lymphoma; TNBC, triple‐negative breast cancer; uHCC, unresectable hepatocellular carcinoma.
Sources: Data from ClinicalTrials.gov.
Antibodies, cytotoxic drugs, and conjugates bridge ADC (antibody–drug–conjugates) drugs. The antibody will specifically recognize and guide the drug to the lesion. The conjugate can generally be broken in the pH environment of the lesion site, thereby releasing the cytotoxin with a therapeutic effect. Cytotoxins mainly target the DNA and tubulin of cancer cells, block the proliferation of tumor cells, and induce apoptosis. 401 , 402 After the second new ADC drug Adcetris (brentuximab vedotin) was approved by the FDA in 2011, 10 ADC drugs have been approved so far, 7 of which have been approved in the last 3–4 years. ADC drugs can improve the selectivity of tumor treatment and can better deal with the drug resistance of targeted monoclonal antibodies. 403 , 404 It is considered one of the essential directions for developing monoclonal antibodies (especially in the field of tumor‐targeted therapy) in the next decade. The tissue specificity and cytotoxicity of the new generation of ADCs have been improved compared with the previous generation products, allowing them to show fantastic activity in treating refractory cancers. 405 , 406 The currently available evidence shows that the potency of ADCs is based on complex and delicate interactions among antibodies, conjugates, various components of the cytotoxin, and the tumor and its microenvironment. 407 Combining ADCs with other antibody therapies, such as ADCs with the anti‐VEGF monoclonal antibody bevacizumab, has also shown activity in preclinical models. 408 This may be due to bevacizumab's enhanced drug delivery efficiency by altering tumor vascular innervation.
Some researchers proposed to reverse chemotherapy resistance by targeting inhibitor of apoptosis (IAP) inhibitors. 409 The IAP protein family plays an essential role in controlling programmed cell death, and the expression level of IAP in cancer cells is significantly increased. Therefore, by directly or indirectly regulating the expression of apoptotic proteins on the extrinsic apoptotic pathway (transmembrane apoptotic pathway) 410 and the intrinsic apoptotic pathway (mitochondrial apoptotic pathway), 411 the chemosensitivity of cell apoptosis can be improved and obtained a potential response. However, the current combination of this method and chemotherapy only targets a small number of tumor cells, and there are cases where tumor cells are resistant to IAP inhibitors. 412
The technologies for reversing tumor MDR at the gene level mainly include antisense nucleic acid technology, 413 ribozyme technology, 414 and RNA interference (RNAi) technology. 415 , 416 Drugs that regulate miRNA expression (such as miR‐125, −20, −24) may have specific clinical application prospects in reversing tumor MDR. 417 , 418 Although many constructed‐cell line banks exist, the genetic characterization of these cells and human tumor samples varies enormously. 419 Researchers are attempting to create patient‐derived cell culture models and then test these models on pharmacogenomics platforms to rapidly discover multiple effective drug combinations. 418 , 419 These models can better reflect the biological complexity of human drug‐resistant tumor cells than constructed cell models. However, the combination therapy results still need to be verified by randomized clinical trials.
The study of functional drug delivery systems that can reverse tumor MDR will have broad application prospects in improving the efficacy of chemotherapy drugs and reducing side effects. 395 Due to tumors’ heterogeneity and drug resistance, it is usually challenging to achieve an excellent therapeutic effect using one drug alone. Therefore, people have been devoting themselves to designing nanocarriers that can be loaded with multiple anticancer drugs and improve the therapeutic effect through synergistic therapeutic effects. 414 , 420 Combining chemotherapeutics and MDR‐reversing agents using a drug delivery system has been a promising strategy for reversing MDR in recent years. 421 , 422 , 423 Common nano‐drug carriers that have been reported to be used for the co‐delivery of drugs include liposomes, nanoparticles, micelles, nano‐emulsions, and nanogels. 424 Combining MDR drug reversal agents, RNAi/DNA, targeted drugs with nanocarrier drug delivery systems, through nonspecific internalization, reduce the efflux of drugs by ABC transporters in tumor cells, increases the uptake of drugs, and through RNAi/DNA Delivery, active targeting, and increased responsiveness to physiological stimuli can reverse tumor cell MDR. 425 , 426 , 427 Combination types of drugs delivered in combination include chemotherapeutics and chemotherapeutics, 428 , 429 chemotherapeutics and MDR reversal agents, 424 , 430 , 431 chemotherapy drugs and siRNA, 432 , 433 , 434 chemotherapy drugs and monoclonal antibodies. 425 , 435 Among them, combining chemotherapy and other drugs is the most common type of combination administration. 423 The rapid development of nano‐drug delivery carriers provides a good carrier platform for improving the therapeutic effect of drugs and overcoming the MDR problem of tumors. Combining drug administration with nano‐drug carriers can enhance the effect of reversing MDR through various forms and achieve a substantial combined effect by co‐delivering different types of drugs. Nano‐drug carrier‐mediated co‐administration is a promising strategy for reversing tumor MDR. Currently, there is more and more research on reversing MDR of tumors by combined administration of nano‐drug carriers. 419 , 436 With the clinical application of nanocarrier drug delivery system, this technology shows great potential in reversing tumor MDR.
Tumor immunotherapy can improve tumor patients’ immune function, kill or inhibit tumor cells, and reverse tumor MDR with little adverse reactions. There are many studies on tumor immunohistology, enzymology, and cytokines, 437 , 438 but most are still experimental research. Among them, the systems biology approach involves the analysis of large amounts of data, including genetics, transcriptomics, proteomics, or factors affecting posttranslational regulation. Some systems’ biology approaches study the importance of proteins in intracellular signaling networks and explore the possibility of reversing tumor drug resistance based on the differential effects of drug targets on intracellular associations. 439
The new technologies, for example, the novel computational method, can be used for predicting polytherapy switching strategies to overcome tumor heterogeneity and evolution. In addition, the widely used gene silencing tools, including shRNA and CRISPR, could help researchers propose and predict effective drug effects both in vitro and in vivo. Finally, the clinical trial designs that reflect drug toxicity and utilize intermittent doses and adaptive trial designs that give dynamic combinations after the emergence of resistance will significantly accelerate the development of more effective therapeutic combinations to reverse the MDR in cancer cells (Figure 4).
FIGURE 4.
Overcoming drug resistance in cancer. The purpose of overcoming the drug resistance of cancer cells is to optimize the sensitivity of the therapy. This can be achieved by polytherapy using the combination of at least two drugs; immunotherapy using checkpoint inhibitors or monoclonal antibodies; antibody–drug–conjugates improving the selectivity of cancer treatment; gene technology modifying the epigenetic sequence; targeted therapy targeting the overexpression of drug efflux transporter or vital proteins for the cancer cell apoptosis; and nanoparticle delivery system improving the efficacy of the drug and reducing the side effect. Source: This figure was created with Biorender.com.
4. CONCLUSION
As summarized in Table 2, MDR in cancer is a multifactorial phenomenon that results in drug inactivation, efflux, target alteration, cancer cell death inhibition, DNA damage repair, cellular heterogeneity, and more. Tumor drug resistance has become a significant problem in oncology, affecting the treatment effect and prognosis of cancer patients, and may lead to tumor progression or even recurrence. Therefore, it becomes crucial to understand the causes and underlying mechanisms of cancer drug resistance, which will facilitate the development of various therapies or combinations for treating different cancers. Combination therapy is considered the most important treatment option for personalized medicine and overcoming MDR. Simultaneous use of two or more treatment methods can effectively overcome MDR, avoid clinical toxicity, and improve patients’ survival rate and quality of life. The specificity of cancer cells is a promising research and therapeutic area, which will help to develop tumor‐targeted therapies with low toxicity to normal cells and higher tumor specificity.
TABLE 2.
Typical resistance mechanisms in cancer and combating strategies.
Resistance mechanisms | Combating strategies | Typical example |
---|---|---|
Drug inactivation | Drug replacement or combination with enzyme enhancer | Nucleoside analog phosphate prodrugs could bypass DCK deficiency‐mediated AraC resistance in AML without the demand for DCK phosphorylation 36 or combination with etoposide‐enhancing DCK activity 39 |
Drug detoxification | Combination with metabolism enzyme inhibitor | GSTP1‐1 inhibitor MC3181 could be used to reverse the vemurafenib resistance in melanoma 80 , 81 |
Reduced drug uptake | Modulation of the cell membrane lipid composition to increase permeability and fluidity | Cancer cell membrane–based biomimetic nanoparticle delivery systems could improve therapeutic efficacy for various MDR cancers 103 , 104 , 105 |
Increased drug efflux | Drug replacement, combination with ABC transporter inhibitor, or silence of the efflux transporters | Targeted anticancer drugs, such as tepotinib and poziotinib, could inhibit ABCB1 and ABCG2 to reverse the resistance to ABCB1 and ABCG2 substrates 128 , 129 , 130 |
Mutation of drug targets | Development of new generation or multitarget anticancer drug | EAI045 is the first allosteric EGFR TKI designed to overcome the acquired resistance mediated by T790M and C797S mutations 147 |
DNA damage repair | Combination with DNA repair inhibitor | ERK or p38 kinase inhibitors could overcome the resistance mediated by 5‐FU‐induced ERCC1 overexpression 198 |
Blocked apoptosis | Combination with apoptosis inducer | Navitoclax could trigger the resistance in CLL by inhibiting Bcl‐2 to induce apoptosis 219 |
Autophagy induction | Combination with autophagy modulators | Imatinib combined with siRNAs targeting ATGs could potentiate the cytotoxicity and reverse the acquired resistance in both GIST and CML 233 |
Cell death inhibition | Ferroptosis and cuproptosis induction | Cellular ferroptosis and cuproptosis could sensitize chemotherapy resistance in various cancers 239 , 240 , 241 , 242 |
Tumor microenvironment | Combination with inhibitors targeting the specific protein function in TME | Inhibition of the β1 integrin activity by monoclonal antibody AIIB2 could promote radiotherapy efficacy and elevates sensitivity to HER2‐targeting agents of breast cancer cells 265 , 266 |
Cancer stem cell | Development of novel drugs targeting cancer stem cells | PF‐03446962 is designed to inhibit TGF‐β receptors resulting in the antagonization of CSC division 374 |
Epithelial‐to‐mesenchymal transition | Development of novel drugs targeting the EMT process | Inhibitors targeting Notch, TGF‐β, and Wnt signaling pathways are promising candidates for inhibiting EMT 371 |
Immunotherapy and immune responses | Combination with radiation, chemotherapy, targeted drug, or tumor vaccines | VEGF inhibitor bevacizumab can augment intratumoral CD8+ T cell infiltration and endothelial cell activation, thereby potentiating PD‐L1 checkpoint inhibition with PD‐L1 inhibitor atezolizumab 316 , 317 |
Abbreviations: 5‐FU, 5‐fluorouracil; ABC, ATP‐binding cassette; ATG, autophagy‐related gene; Bcl‐2, B‐cell lymphoma 2; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; CSC, cancer stem cell; DCK, deoxycytidine kinase; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal transition; ERCC, excision repair cross‐complementing 1; ERK, extracellular signal‐regulated kinase; GIST, gastrointestinal stromal tumors; GSTP1, glutathione‐S‐transferase Pi 1; HER2, human epidermal growth factor receptor 2; MDR, multidrug resistance; PD‐L1, programmed cell death ligand 1; TGF‐β, transforming growth factor beta; TKI, tyrosine kinase inhibitor; TME, tumor microenvironment; VEGF, vascular endothelial growth factor.
AUTHOR CONTRIBUTIONS
Zhe‐Sheng Chen, Yihang Pan, and Leli Zeng designed the review. Zi‐Ning Lei, Qin Tian, and Qiu‐Xu Teng did the literature search and wrote the manuscript. Zi‐Ning Lei and Qiu‐Xu Teng prepared the table and figures. Zhe‐Sheng Chen, Yihang Pan, and John N. D. Wurpel reviewed and revised the manuscript. All authors listed have made a substantial contribution to the work. All authors have read and approved the article.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest. Author Zhe‐Sheng Chen is the Editorial Board Member of MedComm. Author Zhe‐Sheng Chen was not involved in the journal's review of, or decisions related to, this manuscript.
EtHICS STATEMENT
Not applicable.
ACKNOWLEDGMENTS
This work is funded by the National Natural Science Foundation of China (32270815), the National Key Research and Development Program of China #2018YFA0902801, the 100 Top Talents Program of Sun Yat‐sen University (ZSQYBRJH0001), and Guangdong Basic and Applied Basic Research Foundation #2021A1515010117. Z.N.L. and Q.X.T. appreciate the scholarship from the Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University. We would like to acknowledge BioRender (https://biorender.com/) for the platform where the figures were generated.
Lei Z‐N, Tian Q, Teng Q‐X, et al. Understanding and targeting resistance mechanisms in cancer. Molecular Oral Microbiology. 2023;4:e265. 10.1002/mco2.265
Contributor Information
Leli Zeng, Email: zenglli6@mail.sysu.edu.cn.
Yihang Pan, Email: panyih@mail.sysu.edu.cn.
Zhe‐Sheng Chen, Email: chenz@stjohns.edu.
DATA AVAILABILITY STATEMENT
Not applicable.
REFERENCES
- 1. Bray F, Laversanne M, Weiderpass E, Soerjomataram I. The ever‐increasing importance of cancer as a leading cause of premature death worldwide. Cancer. 2021;127(16):3029‐3030. [DOI] [PubMed] [Google Scholar]
- 2. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. [DOI] [PubMed] [Google Scholar]
- 3. Awan A, Esfahani K. Endocrine therapy for breast cancer in the primary care setting. Curr Oncol. 2018;25(4):285‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Kruger S, Ilmer M, Kobold S, et al. Advances in cancer immunotherapy 2019 – latest trends. J Exp Clin Cancer Res. 2019;38(1):268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Kareliotis G, Tremi I, Kaitatzi M, et al. Combined radiation strategies for novel and enhanced cancer treatment. Int J Radiat Biol. 2020;96(9):1087‐1103. [DOI] [PubMed] [Google Scholar]
- 6. Bedard PL, Hyman DM, Davids MS, Siu LL. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet. 2020;395(10229):1078‐1088. [DOI] [PubMed] [Google Scholar]
- 7. Szostakowska M, Trebinska‐Stryjewska A, Grzybowska EA, Fabisiewicz A. Resistance to endocrine therapy in breast cancer: molecular mechanisms and future goals. Breast Cancer Res Treat. 2019;173(3):489‐497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bukowski K, Kciuk M, Kontek R. Mechanisms of multidrug resistance in cancer chemotherapy. Int J Mol Sci. 2020;21(9):3233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bueschbell B, Caniceiro AB, Suzano PMS, Machuqueiro M, Rosario‐Ferreira N, Moreira IS. Network biology and artificial intelligence drive the understanding of the multidrug resistance phenotype in cancer. Drug Resist Updat. 2022;60:100811. [DOI] [PubMed] [Google Scholar]
- 10. Turajlic S, Furney SJ, Stamp G, et al. Whole‐genome sequencing reveals complex mechanisms of intrinsic resistance to BRAF inhibition. Ann Oncol. 2014;25(5):959‐967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ma Y, Wang L, Neitzel LR, et al. The MAPK pathway regulates intrinsic resistance to BET inhibitors in colorectal cancer. Clin Cancer Res. 2017;23(8):2027‐2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Kalbasi A, Ribas A. Tumour‐intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol. 2020;20(1):25‐39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wicki A, Mandala M, Massi D, et al. Acquired resistance to clinical cancer therapy: a twist in physiological signaling. Physiol Rev. 2016;96(3):805‐829. [DOI] [PubMed] [Google Scholar]
- 14. Saleh R, Elkord E. Acquired Resistance to Cancer Immunotherapy: Role of Tumor‐Mediated Immunosuppression. Elsevier; 2020:13‐27. [DOI] [PubMed] [Google Scholar]
- 15. Meador CB, Hata AN. Acquired resistance to targeted therapies in NSCLC: updates and evolving insights. Pharmacol Ther. 2020;210:107522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hu X, Zhang Z. Understanding the genetic mechanisms of cancer drug resistance using genomic approaches. Trends Genet. 2016;32(2):127‐137. [DOI] [PubMed] [Google Scholar]
- 17. Kara A, Ozgur A, Nalbantoglu S, Karadag A. DNA repair pathways and their roles in drug resistance for lung adenocarcinoma. Mol Biol Rep. 2021;48(4):3813‐3825. [DOI] [PubMed] [Google Scholar]
- 18. Damia G, Broggini M. Platinum resistance in ovarian cancer: role of DNA repair. Cancers (Basel). 2019;11(1):119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Marine J‐C, Dawson S‐J, Dawson MA. Non‐genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer. 2020;20(12):743‐756. [DOI] [PubMed] [Google Scholar]
- 20. Pathania S, Bhatia R, Baldi A, Singh R, Rawal RK. Drug metabolizing enzymes and their inhibitors’ role in cancer resistance. Biomed Pharmacother. 2018;105:53‐65. [DOI] [PubMed] [Google Scholar]
- 21. Allain EP, Rouleau M, Levesque E, Guillemette C. Emerging roles for UDP‐glucuronosyltransferases in drug resistance and cancer progression. Br J Cancer. 2020;122(9):1277‐1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Buss I, Hamacher A, Sarin N, Kassack MU, Kalayda GV. Relevance of copper transporter 1 and organic cation transporters 1–3 for oxaliplatin uptake and drug resistance in colorectal cancer cells. Metallomics. 2018;10(3):414‐425. [DOI] [PubMed] [Google Scholar]
- 23. Giddings EL, Champagne DP, Wu M‐H, et al. Mitochondrial ATP fuels ABC transporter‐mediated drug efflux in cancer chemoresistance. Nat Commun. 2021;12(1):2804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Wang J‐Q, Yang Y, Cai C‐Y, et al. Multidrug resistance proteins (MRPs): structure, function and the overcoming of cancer multidrug resistance. Drug Resist Updat. 2021;54:100743. [DOI] [PubMed] [Google Scholar]
- 25. Zhitomirsky B, Assaraf YG. Lysosomes as mediators of drug resistance in cancer. Drug Resist Updat. 2016;24:23‐33. [DOI] [PubMed] [Google Scholar]
- 26. Wu P, Gao W, Su M, et al. Adaptive mechanisms of tumor therapy resistance driven by tumor microenvironment. Front Cell Dev Biol. 2021;9:641469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Verma H, Singh Bahia M, Choudhary S, Kumar Singh P, Silakari O. Drug metabolizing enzymes‐associated chemo resistance and strategies to overcome it. Drug Metab Rev. 2019;51(2):196‐223. [DOI] [PubMed] [Google Scholar]
- 28. Rahman M, Hasan MR. Cancer metabolism and drug resistance. Metabolites. 2015;5(4):571‐600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Xu Y, Villalona‐Calero MA. Irinotecan: mechanisms of tumor resistance and novel strategies for modulating its activity. Ann Oncol. 2002;13(12):1841‐1851. [DOI] [PubMed] [Google Scholar]
- 30. Miura K, Kinouchi M, Ishida K, et al. 5‐FU metabolism in cancer and orally‐administrable 5‐FU drugs. Cancers (Basel). 2010;2(3):1717‐1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol. 2002;3(7):415‐424. [DOI] [PubMed] [Google Scholar]
- 32. Medina‐Sanson A, Ramirez‐Pacheco A, Moreno‐Guerrero SS, Dorantes‐Acosta EM, Sanchez‐Preza M, Reyes‐Lopez A. Role of genetic polymorphisms of deoxycytidine kinase and cytidine deaminase to predict risk of death in children with acute myeloid leukemia. Biomed Res Int. 2015;2015:309491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wu B, Mao ZJ, Wang Z, et al. Deoxycytidine kinase (DCK) mutations in human acute myeloid leukemia resistant to cytarabine. Acta Haematol. 2021;144(5):534‐541. [DOI] [PubMed] [Google Scholar]
- 34. Song JH, Kim SH, Kweon SH, et al. Defective expression of deoxycytidine kinase in cytarabine‐resistant acute myeloid leukemia cells. Int J Oncol. 2009;34(4):1165‐1171. [DOI] [PubMed] [Google Scholar]
- 35. Klanova M, Lorkova L, Vit O, et al. Downregulation of deoxycytidine kinase in cytarabine‐resistant mantle cell lymphoma cells confers cross‐resistance to nucleoside analogs gemcitabine, fludarabine and cladribine, but not to other classes of anti‐lymphoma agents. Mol Cancer. 2014;13:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Cahard D, McGuigan C, Balzarini J. Aryloxy phosphoramidate triesters as pro‐tides. Mini Rev Med Chem. 2004;4(4):371‐381. [DOI] [PubMed] [Google Scholar]
- 37. Slusarczyk M, Lopez MH, Balzarini J, et al. Application of ProTide technology to gemcitabine: a successful approach to overcome the key cancer resistance mechanisms leads to a new agent (NUC‐1031) in clinical development. J Med Chem. 2014;57(4):1531‐1542. [DOI] [PubMed] [Google Scholar]
- 38. Knox JJ, McNamara MG, Goyal L, et al. Phase III study of NUC‐1031 + cisplatin vs gemcitabine + cisplatin for first‐line treatment of patients with advanced biliary tract cancer (NuTide:121). J Clin Oncol. 2021;39(3 suppl):TPS351‐TPS351. [Google Scholar]
- 39. Spasokoukotskaja T, Sasvari‐Szekely M, Keszler G, Albertioni F, Eriksson S, Staub M. Treatment of normal and malignant cells with nucleoside analogues and etoposide enhances deoxycytidine kinase activity. Eur J Cancer. 1999;35(13):1862‐1867. [DOI] [PubMed] [Google Scholar]
- 40. Huang M, Inukai T, Miyake K, et al. Clofarabine exerts antileukemic activity against cytarabine‐resistant B‐cell precursor acute lymphoblastic leukemia with low deoxycytidine kinase expression. Cancer Med. 2018;7(4):1297‐1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Riddick DS, Lee C, Ramji S, et al. Cancer chemotherapy and drug metabolism. Drug Metab Dispos. 2005;33(8):1083‐1096. [DOI] [PubMed] [Google Scholar]
- 42. Kaur G, Gupta SK, Singh P, Ali V, Kumar V, Verma M. Drug‐metabolizing enzymes: role in drug resistance in cancer. Clin Transl Oncol. 2020;22(10):1667‐1680. [DOI] [PubMed] [Google Scholar]
- 43. Esteves F, Rueff J, Kranendonk M. The central role of cytochrome P450 in xenobiotic metabolism—a brief review on a fascinating enzyme family. J Xenobiot. 2021;11(3):94‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Hofman J, Vagiannis D, Chen S, Guo L. Roles of CYP3A4, CYP3A5 and CYP2C8 drug‐metabolizing enzymes in cellular cytostatic resistance. Chem Biol Interact. 2021;340:109448. [DOI] [PubMed] [Google Scholar]
- 45. Garcia‐Martin E, Pizarro RM, Martinez C, et al. Acquired resistance to the anticancer drug paclitaxel is associated with induction of cytochrome P450 2C8. Pharmacogenomics. 2006;7(4):575‐585. [DOI] [PubMed] [Google Scholar]
- 46. Morvan VL, Richard E, Cadars M, et al. Cytochrome P450 1B1 polymorphism drives cancer cell stemness and patient outcome in head‐and‐neck carcinoma. Br J Cancer. 2020;123(5):772‐784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Yada E, Kasajima R, Niida A, et al. Possible role of cytochrome P450 1B1 in the mechanism of gemcitabine resistance in pancreatic cancer. Biomedicines. 2021;9(10):1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. McFadyen MCE, Murray GI. Cytochrome P450 1B1: a novel anticancer therapeutic target. Future Oncol. 2005;1(2):259‐263. [DOI] [PubMed] [Google Scholar]
- 49. Pastina I, Giovannetti E, Chioni A, et al. Cytochrome 450 1B1 (CYP1B1) polymorphisms associated with response to docetaxel in castration‐resistant prostate cancer (CRPC) patients. BMC Cancer. 2010;10(1):511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Norouzi‐Barough L, Sarookhani MR, Sharifi M, Moghbelinejad S, Jangjoo S, Salehi R. Molecular mechanisms of drug resistance in ovarian cancer. J Cell Physiol. 2018;233(6):4546‐4562. [DOI] [PubMed] [Google Scholar]
- 51. McFadyen MC, McLeod HL, Jackson FC, Melvin WT, Doehmer J, Murray GI. Cytochrome P450 CYP1B1 protein expression: a novel mechanism of anticancer drug resistance. Biochem Pharmacol. 2001;62(2):207‐212. [DOI] [PubMed] [Google Scholar]
- 52. Rochat B, Morsman JM, Murray GI, Figg WD, McLeod HL. Human CYP1B1 and anticancer agent metabolism: mechanism for tumor‐specific drug inactivation? J Pharmacol Exp Ther. 2001;296(2):537‐541. [PubMed] [Google Scholar]
- 53. Martinez VG, O'Connor R, Liang Y, Clynes M. CYP1B1 expression is induced by docetaxel: effect on cell viability and drug resistance. Br J Cancer. 2008;98(3):564‐570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Carrera AN, Grant MKO, Zordoky BN. CYP1B1 as a therapeutic target in cardio‐oncology. Clin Sci (Lond). 2020;134(21):2897‐2927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Dutour R, Poirier D. Inhibitors of cytochrome P450 (CYP) 1B1. Eur J Med Chem. 2017;135:296‐306. [DOI] [PubMed] [Google Scholar]
- 56. Zhu Z, Mu Y, Qi C, et al. CYP1B1 enhances the resistance of epithelial ovarian cancer cells to paclitaxel in vivo and in vitro. Int J Mol Med. 2015;35(2):340‐348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Mikstacka R, Dutkiewicz Z. New perspectives of CYP1B1 inhibitors in the light of molecular studies. Processes. 2021;9(5):817. [Google Scholar]
- 58. Lin H, Hu B, He X, et al. Overcoming taxol‐resistance in A549 cells: a comprehensive strategy of targeting P‐gp transporter, AKT/ERK pathways, and cytochrome P450 enzyme CYP1B1 by 4‐hydroxyemodin. Biochem Pharmacol. 2020;171:113733. [DOI] [PubMed] [Google Scholar]
- 59. Cui J, Meng Q, Zhang X, Cui Q, Zhou W, Li S. Design and synthesis of new alpha‐naphthoflavones as cytochrome P450 (CYP) 1B1 inhibitors to overcome docetaxel‐resistance associated with CYP1B1 overexpression. J Med Chem. 2015;58(8):3534‐3547. [DOI] [PubMed] [Google Scholar]
- 60. Chatterjee A, Gupta S. The multifaceted role of glutathione S‐transferases in cancer. Cancer Lett. 2018;433:33‐42. [DOI] [PubMed] [Google Scholar]
- 61. Townsend DM, Tew KD. The role of glutathione‐S‐transferase in anti‐cancer drug resistance. Oncogene. 2003;22(47):7369‐7375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Smitherman PK, Townsend AJ, Kute TE, Morrow CS. Role of multidrug resistance protein 2 (MRP2, ABCC2) in alkylating agent detoxification: MRP2 potentiates glutathione S‐transferase A1‐1‐mediated resistance to chlorambucil cytotoxicity. J Pharmacol Exp Ther. 2004;308(1):260‐267. [DOI] [PubMed] [Google Scholar]
- 63. Cole SPC, Deeley RG. Transport of glutathione and glutathione conjugates by MRP1. Trends Pharmacol Sci. 2006;27(8):438‐446. [DOI] [PubMed] [Google Scholar]
- 64. Brechbuhl HM, Gould N, Kachadourian R, Riekhof WR, Voelker DR, Day BJ. Glutathione transport is a unique function of the ATP‐binding cassette protein ABCG2. J Biol Chem. 2010;285(22):16582‐16587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Zou M, Hu X, Xu B, et al. Glutathione S‑transferase isozyme alpha 1 is predominantly involved in the cisplatin resistance of common types of solid cancer. Oncol Rep. 2019;41(2):989‐998. [DOI] [PubMed] [Google Scholar]
- 66. Sawers L, Ferguson MJ, Ihrig BR, et al. Glutathione S‐transferase P1 (GSTP1) directly influences platinum drug chemosensitivity in ovarian tumour cell lines. Br J Cancer. 2014;111(6):1150‐1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Mathieu A, Remmelink M, D'Haene N, et al. Development of a chemoresistant orthotopic human nonsmall cell lung carcinoma model in nude mice: analyses of tumor heterogeneity in relation to the immunohistochemical levels of expression of cyclooxygenase‐2, ornithine decarboxylase, lung‐related resistance protein, prostaglandin E synthetase, and glutathione‐S‐transferase‐alpha (GST)‐alpha, GST‐mu, and GST‐pi. Cancer. 2004;101(8):1908‐1918. [DOI] [PubMed] [Google Scholar]
- 68. Li Q‐F, Yao R‐Y, Liu K‐w, Lv H‐Y, Jiang T, Liang J. Genetic polymorphism of GSTP1: prediction of clinical outcome to oxaliplatin/5‐FU‐based chemotherapy in advanced gastric cancer. J Korean Med Sci. 2010;25(6):846‐852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Huang Z‐H, Hua D, Du X. Polymorphisms in p53, GSTP1 and XRCC1 predict relapse and survival of gastric cancer patients treated with oxaliplatin‐based adjuvant chemotherapy. Cancer Chemother Pharmacol. 2009;64(5):1001‐1007. [DOI] [PubMed] [Google Scholar]
- 70. Wang Z, Liang S, Lian X, et al. Identification of proteins responsible for adriamycin resistance in breast cancer cells using proteomics analysis. Sci Rep. 2015;5(1):9301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Pljesa‐Ercegovac M, Savic‐Radojevic A, Matic M, et al. Glutathione transferases: potential targets to overcome chemoresistance in solid tumors. Int J Mol Sci. 2018;19(12):3785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Saleh EM, El‐Awady RA, Abdel Alim MA, Abdel Wahab AHA. Altered expression of proliferation‐inducing and proliferation‐inhibiting genes might contribute to acquired doxorubicin resistance in breast cancer cells. Cell Biochem Biophys. 2009;55(2):95‐105. [DOI] [PubMed] [Google Scholar]
- 73. Tew KD, Townsend DM. Regulatory functions of glutathione S‐transferase P1‐1 unrelated to detoxification. Drug Metab Rev. 2011;43(2):179‐193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Attaoua C, Vincent L‐A, Abdel Jaoued A, et al. Differential involvement of glutathione S‐transferase mu 1 and multidrug resistance protein 1 in melanoma acquired resistance to vinca alkaloids. Fundam Clin Pharmacol. 2015;29(1):62‐71. [DOI] [PubMed] [Google Scholar]
- 75. Hansson J, Berhane K, Castro VM, Jungnelius U, Mannervik B, Ringborg U. Sensitization of human melanoma cells to the cytotoxic effect of melphalan by the glutathione transferase inhibitor ethacrynic acid. Cancer Res. 1991;51(1):94‐98. [PubMed] [Google Scholar]
- 76. Tew KD, Bomber AM, Hoffman SJ. Ethacrynic acid and piriprost as enhancers of cytotoxicity in drug resistant and sensitive cell lines. Cancer Res. 1988;48(13):3622‐3625. [PubMed] [Google Scholar]
- 77. Federici L, Lo Sterzo C, Pezzola S, et al. Structural basis for the binding of the anticancer compound 6‐(7‐nitro‐2,1,3‐benzoxadiazol‐4‐ylthio)hexanol to human glutathione s‐transferases. Cancer Res. 2009;69(20):8025‐8034. [DOI] [PubMed] [Google Scholar]
- 78. Pasello M, Michelacci F, Scionti I, et al. Overcoming glutathione S‐transferase P1‐related cisplatin resistance in osteosarcoma. Cancer Res. 2008;68(16):6661‐6668. [DOI] [PubMed] [Google Scholar]
- 79. Sau A, Filomeni G, Pezzola S, et al. Targeting GSTP1‐1 induces JNK activation and leads to apoptosis in cisplatin‐sensitive and ‐resistant human osteosarcoma cell lines. Mol Biosyst. 2012;8(4):994‐1006. [DOI] [PubMed] [Google Scholar]
- 80. De Luca A, Carpanese D, Rapanotti MC, et al. The nitrobenzoxadiazole derivative MC3181 blocks melanoma invasion and metastasis. Oncotarget. 2017;8(9):15520‐15538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Di Paolo V, Fulci C, Rotili D, et al. Characterization of water‐soluble esters of nitrobenzoxadiazole‐based GSTP1‐1 inhibitors for cancer treatment. Biochem Pharmacol. 2020;178:114060. [DOI] [PubMed] [Google Scholar]
- 82. Dourado DFAR, Fernandes PA, Ramos MJ, Mannervik B. Mechanism of glutathione transferase P1‐1‐catalyzed activation of the prodrug canfosfamide (TLK286, TELCYTA). Biochemistry. 2013;52(45):8069‐8078. [DOI] [PubMed] [Google Scholar]
- 83. Gelderblom H, Blay JY, Seddon BM, et al. Brostallicin versus doxorubicin as first‐line chemotherapy in patients with advanced or metastatic soft tissue sarcoma: an European organisation for research and treatment of cancer soft tissue and bone sarcoma group randomised phase II and pharmacogenetic study. Eur J Cancer. 2014;50(2):388‐396. [DOI] [PubMed] [Google Scholar]
- 84. Raynal C, Pascussi J‐M, Leguelinel G, et al. Pregnane X receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN‐38 glucuronidation. Mol Cancer. 2010;9:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Landmann H, Proia DA, He S, et al. UDP glucuronosyltransferase 1A expression levels determine the response of colorectal cancer cells to the heat shock protein 90 inhibitor ganetespib. Cell Death Dis. 2014;5(9):e1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. López‐Ayllón BD, de Castro‐Carpeño J, Rodriguez C, et al. Biomarkers of erlotinib response in non‐small cell lung cancer tumors that do not harbor the more common epidermal growth factor receptor mutations. Int J Clin Exp Pathol. 2015;8(3):2888. [PMC free article] [PubMed] [Google Scholar]
- 87. Zahreddine HA, Culjkovic‐Kraljacic B, Assouline S, et al. The sonic hedgehog factor GLI1 imparts drug resistance through inducible glucuronidation. Nature. 2014;511(7507):90‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Ascierto ML, McMiller TL, Berger AE, et al. The intratumoral balance between metabolic and immunologic gene expression is associated with anti‐PD‐1 response in patients with renal cell carcinoma. Cancer Immunol Res. 2016;4(9):726‐733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Miners JO, Chau N, Rowland A, et al. Inhibition of human UDP‐glucuronosyltransferase enzymes by lapatinib, pazopanib, regorafenib and sorafenib: implications for hyperbilirubinemia. Biochem Pharmacol. 2017;129:85‐95. [DOI] [PubMed] [Google Scholar]
- 90. Osborne MJ, Coutinho de Oliveira L, Volpon L, Zahreddine HA, Borden KLB. Overcoming drug resistance through the development of selective inhibitors of UDP‐glucuronosyltransferase enzymes. J Mol Biol. 2019;431(2):258‐272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Peetla C, Vijayaraghavalu S, Labhasetwar V. Biophysics of cell membrane lipids in cancer drug resistance: implications for drug transport and drug delivery with nanoparticles. Adv Drug Deliv Rev. 2013;65(13‐14):1686‐1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Zalba S, Ten Hagen TLM. Cell membrane modulation as adjuvant in cancer therapy. Cancer Treat Rev. 2017;52:48‐57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. May GL, Wright LC, Dyne M, Mackinnon WB, Fox RM, Mountford CE. Plasma membrane lipid composition of vinblastine sensitive and resistant human leukaemic lymphoblasts. Int J Cancer. 1988;42(5):728‐733. [DOI] [PubMed] [Google Scholar]
- 94. Peetla C, Bhave R, Vijayaraghavalu S, Stine A, Kooijman E, Labhasetwar V. Drug resistance in breast cancer cells: biophysical characterization of and doxorubicin interactions with membrane lipids. Mol Pharm. 2010;7(6):2334‐2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Todor IN, Lukyanova NY, Chekhun VF. The lipid content of cisplatin‐ and doxorubicin‐resistant MCF‐7 human breast cancer cells. Exp Oncol. 2012;34(2):97‐100. [PubMed] [Google Scholar]
- 96. Rivel T, Ramseyer C, Yesylevskyy S. The asymmetry of plasma membranes and their cholesterol content influence the uptake of cisplatin. Sci Rep. 2019;9(1):5627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. van Lummel M, van Blitterswijk WJ, Vink SR, et al. Enriching lipid nanovesicles with short‐chain glucosylceramide improves doxorubicin delivery and efficacy in solid tumors. FASEB J. 2011;25(1):280‐289. [DOI] [PubMed] [Google Scholar]
- 98. Pedrosa LRC, van Hell A, Suss R, et al. Improving intracellular doxorubicin delivery through nanoliposomes equipped with selective tumor cell membrane permeabilizing short‐chain sphingolipids. Pharm Res. 2013;30(7):1883‐1895. [DOI] [PubMed] [Google Scholar]
- 99. Adada M, Luberto C, Canals D. Inhibitors of the sphingomyelin cycle: sphingomyelin synthases and sphingomyelinases. Chem Phys Lipids. 2016;197:45‐59. [DOI] [PubMed] [Google Scholar]
- 100. Vijayaraghavalu S, Peetla C, Lu S, Labhasetwar V. Epigenetic modulation of the biophysical properties of drug‐resistant cell lipids to restore drug transport and endocytic functions. Mol Pharm. 2012;9(9):2730‐2742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Bose RJ, Paulmurugan R, Moon J, Lee S‐H, Park H. Cell membrane‐coated nanocarriers: the emerging targeted delivery system for cancer theranostics. Drug Discov Today. 2018;23(4):891‐899. [DOI] [PubMed] [Google Scholar]
- 102. Fang RH, Hu C‐MJ, Luk BT, et al. Cancer cell membrane‐coated nanoparticles for anticancer vaccination and drug delivery. Nano Lett. 2014;14(4):2181‐2188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Wu P, Yin D, Liu J, et al. Cell membrane based biomimetic nanocomposites for targeted therapy of drug resistant EGFR‐mutated lung cancer. Nanoscale. 2019;11(41):19520‐19528. [DOI] [PubMed] [Google Scholar]
- 104. Gao Y, Zhu Y, Xu X, et al. Surface PEGylated cancer cell membrane‐coated nanoparticles for codelivery of curcumin and doxorubicin for the treatment of multidrug resistant esophageal carcinoma. Front Cell Dev Biol. 2021;9:688070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Lu K, Li Z, Hu Q, Sun J, Chen M. CRPC membrane‐camouflaged, biomimetic nanosystem for overcoming castration‐resistant prostate cancer by cellular vehicle‐aided tumor targeting. Int J Mol Sci. 2022;23(7):3623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Perland E, Fredriksson R. Classification systems of secondary active transporters. Trends Pharmacol Sci. 2017;38(3):305‐315. [DOI] [PubMed] [Google Scholar]
- 107. Al‐Abdulla R, Perez‐Silva L, Abete L, Romero MR, Briz O, Marin JJG. Unraveling ‘The Cancer Genome Atlas’ information on the role of SLC transporters in anticancer drug uptake. Expert Rev Clin Pharmacol. 2019;12(4):329‐341. [DOI] [PubMed] [Google Scholar]
- 108. Wu Z, Xu J, Liang C, et al. Emerging roles of the solute carrier family in pancreatic cancer. Clin Transl Med. 2021;11(3):e356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Buxhofer‐Ausch V, Secky L, Wlcek K, et al. Tumor‐specific expression of organic anion‐transporting polypeptides: transporters as novel targets for cancer therapy. J Drug Deliv. 2013;2013:863539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Sun R, Ying Y, Tang Z, et al. The emerging role of the SLCO1B3 protein in cancer resistance. Protein Pept Lett. 2020;27(1):17‐29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Zimmerman EI, Hu S, Roberts JL, et al. Contribution of OATP1B1 and OATP1B3 to the disposition of sorafenib and sorafenib‐glucuronide. Clin Cancer Res. 2013;19(6):1458‐1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. de Morree ES, Bottcher R, van Soest RJ, et al. Loss of SLCO1B3 drives taxane resistance in prostate cancer. Br J Cancer. 2016;115(6):674‐681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Sissung TM, Ley AM, Strope JD, et al. Differential expression of OATP1B3 mediates unconjugated testosterone influx. Mol Cancer Res. 2017;15(8):1096‐1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Alsinnawi M, Zhang A, Bianchi‐Frias D, et al. Association of prostate cancer SLCO gene expression with Gleason grade and alterations following androgen deprivation therapy. Prostate Cancer Prostatic Dis. 2019;22(4):560‐568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Haberkorn B, Oswald S, Kehl N, et al. Cancer‐type organic anion transporting polypeptide 1B3 (Ct‐OATP1B3) is localized in lysosomes and mediates resistance against kinase inhibitors. Mol Pharmacol. 2022;102(6):248‐258. [DOI] [PubMed] [Google Scholar]
- 116. Falguieres T. ABC transporters in human diseases: future directions and therapeutic perspectives. Int J Mol Sci. 2022;23(8):4250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta. 1976;455(1):152‐162. [DOI] [PubMed] [Google Scholar]
- 118. Wang J‐Q, Wu Z‐X, Yang Y, et al. ATP‐binding cassette (ABC) transporters in cancer: a review of recent updates. J Evid Based Med. 2021;14(3):232‐256. [DOI] [PubMed] [Google Scholar]
- 119. Thomas C, Tampe R. Structural and mechanistic principles of ABC transporters. Annu Rev Biochem. 2020;89:605‐636. [DOI] [PubMed] [Google Scholar]
- 120. Chen XY. Natural product as substrates of ABC transporters: a review. Recent Pat Anti‐Cancer Drug Discov. 2021;16(2):222‐238. [DOI] [PubMed] [Google Scholar]
- 121. Anreddy N, Gupta P, Kathawala RJ, Patel A, Wurpel JND, Chen Z‐S. Tyrosine kinase inhibitors as reversal agents for ABC transporter mediated drug resistance. Molecules. 2014;19(9):13848‐13877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Dong X‐D, Zhang M, Cai C‐Y, et al. Overexpression of ABCB1 associated with the resistance to the KRAS‐G12C specific inhibitor ARS‐1620 in cancer cells. Front Pharmacol. 2022;13:843829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Yang Y, Teng Q‐X, Wu Z‐X, et al. PBK/TOPK inhibitor OTS964 resistance is mediated by ABCB1‐dependent transport function in cancer: in vitro and in vivo study. Mol Cancer. 2022;21(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Fu H, Wu Z‐X, Lei Z‐N, et al. The resistance of cancer cells to palbociclib, a cyclin‐dependent kinase 4/6 inhibitor, is mediated by the ABCB1 transporter. Front Pharmacol. 2022;13:861642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Parrish KE, Pokorny J, Mittapalli RK, Bakken K, Sarkaria JN, Elmquist WF. Efflux transporters at the blood‐brain barrier limit delivery and efficacy of cyclin‐dependent kinase 4/6 inhibitor palbociclib (PD‐0332991) in an orthotopic brain tumor model. J Pharmacol Exp Ther. 2015;355(2):264‐271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Mittapalli RK, Chung AH, Parrish KE, et al. ABCG2 and ABCB1 limit the efficacy of dasatinib in a PDGF‐B‐driven brainstem glioma model. Mol Cancer Ther. 2016;15(5):819‐829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Engle K, Kumar G. Cancer multidrug‐resistance reversal by ABCB1 inhibition: a recent update. Eur J Med Chem. 2022;239:114542. [DOI] [PubMed] [Google Scholar]
- 128. Ji N, Yang Y, Cai C‐Y, et al. Selonsertib (GS‐4997), an ASK1 inhibitor, antagonizes multidrug resistance in ABCB1‐ and ABCG2‐overexpressing cancer cells. Cancer Lett. 2019;440‐441:82‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Wu Z‐X, Teng Q‐X, Yang Y, et al. MET inhibitor tepotinib antagonizes multidrug resistance mediated by ABCG2 transporter: in vitro and in vivo study. Acta Pharm Sin B. 2022;12(5):2609‐2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Zhang Y, Wu Z‐X, Yang Y, et al. Poziotinib inhibits the efflux activity of the ABCB1 and ABCG2 transporters and the expression of the ABCG2 transporter protein in multidrug resistant colon cancer cells. Cancers (Basel). 2020;12(11):3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Fanelli M, Hattinger CM, Vella S, et al. Targeting ABCB1 and ABCC1 with their specific inhibitor CBT‐1(R) can overcome drug resistance in osteosarcoma. Curr Cancer Drug Targets. 2016;16(3):261‐274. [DOI] [PubMed] [Google Scholar]
- 132. Tao L‐Y, Liang Y‐J, Wang F, et al. Cediranib (recentin, AZD2171) reverses ABCB1‐ and ABCC1‐mediated multidrug resistance by inhibition of their transport function. Cancer Chemother Pharmacol. 2009;64(5):961‐969. [DOI] [PubMed] [Google Scholar]
- 133. Stefan SM. Multi‐target ABC transporter modulators: what next and where to go? Future Med Chem. 2019;11(18):2353‐2358. [DOI] [PubMed] [Google Scholar]
- 134. Tiash S, Chowdhury EH. siRNAs targeting multidrug transporter genes sensitise breast tumour to doxorubicin in a syngeneic mouse model. J Drug Target. 2019;27(3):325‐337. [DOI] [PubMed] [Google Scholar]
- 135. Wang Y, Wang Y, Qin Z, et al. The role of non‐coding RNAs in ABC transporters regulation and their clinical implications of multidrug resistance in cancer. Expert Opin Drug Metab Toxicol. 2021;17(3):291‐306. [DOI] [PubMed] [Google Scholar]
- 136. Yi H, Liu L, Sheng N, et al. Synergistic therapy of doxorubicin and miR‐129‐5p with self‐cross‐linked bioreducible polypeptide nanoparticles reverses multidrug resistance in cancer cells. Biomacromolecules. 2016;17(5):1737‐1747. [DOI] [PubMed] [Google Scholar]
- 137. Yan L, Ding B, Liu H, et al. Inhibition of SMYD2 suppresses tumor progression by down‐regulating microRNA‐125b and attenuates multi‐drug resistance in renal cell carcinoma. Theranostics. 2019;9(26):8377‐8391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. X‐k Wang, J‐h He, J‐h Xu, et al. Afatinib enhances the efficacy of conventional chemotherapeutic agents by eradicating cancer stem‐like cells. Cancer Res. 2014;74(16):4431‐4445. [DOI] [PubMed] [Google Scholar]
- 139. Martin V, Sanchez‐Sanchez AM, Herrera F, et al. Melatonin‐induced methylation of the ABCG2/BCRP promoter as a novel mechanism to overcome multidrug resistance in brain tumour stem cells. Br J Cancer. 2013;108(10):2005‐2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Fultang N, Illendula A, Lin J, Pandey MK, Klase Z, Peethambaran B. ROR1 regulates chemoresistance in breast cancer via modulation of drug efflux pump ABCB1. Sci Rep. 2020;10(1):1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Liu W‐J, Du Y, Wen R, Yang M, Xu J. Drug resistance to targeted therapeutic strategies in non‐small cell lung cancer. Pharmacol Ther. 2020;206:107438. [DOI] [PubMed] [Google Scholar]
- 142. Sarmento‐Ribeiro AB, Scorilas A, Goncalves AC, Efferth T, Trougakos IP. The emergence of drug resistance to targeted cancer therapies: clinical evidence. Drug Resist Updat. 2019;47:100646. [DOI] [PubMed] [Google Scholar]
- 143. Di Noia V, D'Aveni A, D'Argento E, et al. Treating disease progression with osimertinib in EGFR‐mutated non‐small‐cell lung cancer: novel targeted agents and combination strategies. ESMO Open. 2021;6(6):100280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E, Tiseo M. Resistance mechanisms to osimertinib in EGFR‐mutated non‐small cell lung cancer. Br J Cancer. 2019;121(9):725‐737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Piotrowska Z, Niederst MJ, Karlovich CA, et al. Heterogeneity underlies the emergence of EGFRT790 wild‐type clones following treatment of T790M‐positive cancers with a third‐generation EGFR inhibitor. Cancer Discov. 2015;5(7):713‐722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Thress KS, Paweletz CP, Felip E, et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non‐small cell lung cancer harboring EGFR T790M. Nat Med. 2015;21(6):560‐562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Jia Y, Yun C‐H, Park E, et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant‐selective allosteric inhibitors. Nature. 2016;534(7605):129‐132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Lu X, Zhang T, Zhu S‐J, et al. Discovery of JND3229 as a new EGFR(C797S) mutant inhibitor with in vivo monodrug efficacy. ACS Med Chem Lett. 2018;9(11):1123‐1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. To C, Jang J, Chen T, et al. Single and dual targeting of mutant EGFR with an allosteric inhibitor. Cancer Discov. 2019;9(7):926‐943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion‐positive cancers in adults and children. N Engl J Med. 2018;378(8):731‐739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Drilon A, Li G, Dogan S, et al. What hides behind the MASC: clinical response and acquired resistance to entrectinib after ETV6‐NTRK3 identification in a mammary analogue secretory carcinoma (MASC). Ann Oncol. 2016;27(5):920‐926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Kwak EL, Bang Y‐J, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non‐small‐cell lung cancer. N Engl J Med. 2010;363(18):1693‐1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired crizotinib resistance in ALK‐rearranged lung cancers. Sci Transl Med. 2012;4(120):120ra17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Gainor JF, Varghese AM, Ou S‐HI, et al. ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non‐small cell lung cancer. Clin Cancer Res. 2013;19(15):4273‐4281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Doebele RC, Pilling AB, Aisner DL, et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non‐small cell lung cancer. Clin Cancer Res. 2012;18(5):1472‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Sasaki T, Okuda K, Zheng W, et al. The neuroblastoma‐associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK‐translocated cancers. Cancer Res. 2010;70(24):10038‐10043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Sasaki T, Koivunen J, Ogino A, et al. A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res. 2011;71(18):6051‐6060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Katayama R, Khan TM, Benes C, et al. Therapeutic strategies to overcome crizotinib resistance in non‐small cell lung cancers harboring the fusion oncogene EML4‐ALK. Proc Natl Acad Sci USA. 2011;108(18):7535‐7540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Marsilje TH, Pei W, Chen B, et al. Synthesis, structure‐activity relationships, and in vivo efficacy of the novel potent and selective anaplastic lymphoma kinase (ALK) inhibitor 5‐chloro‐N2‐(2‐isopropoxy‐5‐methyl‐4‐(piperidin‐4‐yl)phenyl)‐N4‐(2‐(isopropylsulfonyl)phenyl)pyrimidine‐2,4‐diamine (LDK378) currently in phase 1 and phase 2 clinical trials. J Med Chem. 2013;56(14):5675‐5690. [DOI] [PubMed] [Google Scholar]
- 160. Friboulet L, Li N, Katayama R, et al. The ALK inhibitor ceritinib overcomes crizotinib resistance in non‐small cell lung cancer. Cancer Discov. 2014;4(6):662‐673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Shaw AT, Kim D‐W, Mehra R, et al. Ceritinib in ALK‐rearranged non‐small‐cell lung cancer. N Engl J Med. 2014;370(13):1189‐1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Ostrem JML, Shokat KM. Direct small‐molecule inhibitors of KRAS: from structural insights to mechanism‐based design. Nat Rev Drug Discov. 2016;15(11):771‐785. [DOI] [PubMed] [Google Scholar]
- 163. Papke B, Der CJ. Drugging RAS: know the enemy. Science. 2017;355(6330):1158‐1163. [DOI] [PubMed] [Google Scholar]
- 164. Karoulia Z, Gavathiotis E, Poulikakos PI. New perspectives for targeting RAF kinase in human cancer. Nat Rev Cancer. 2017;17(11):676‐691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF inhibitors. Nat Med. 2013;19(11):1401‐1409. [DOI] [PubMed] [Google Scholar]
- 166. Caunt CJ, Sale MJ, Smith PD, Cook SJ. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer. 2015;15(10):577‐592. [DOI] [PubMed] [Google Scholar]
- 167. Ryan MB, Fece de la Cruz F, Phat S, et al. Vertical pathway inhibition overcomes adaptive feedback resistance to KRAS(G12C) inhibition. Clin Cancer Res. 2020;26(7):1633‐1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Xue JY, Zhao Y, Aronowitz J, et al. Rapid non‐uniform adaptation to conformation‐specific KRAS(G12C) inhibition. Nature. 2020;577(7790):421‐425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild‐type BRAF. Nature. 2010;464(7287):427‐430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Lito P, Saborowski A, Yue J, et al. Disruption of CRAF‐mediated MEK activation is required for effective MEK inhibition in KRAS mutant tumors. Cancer Cell. 2014;25(5):697‐710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Little AS, Balmanno K, Sale MJ, et al. Amplification of the driving oncogene, KRAS or BRAF, underpins acquired resistance to MEK1/2 inhibitors in colorectal cancer cells. Sci Signal. 2011;4(166):ra17. [DOI] [PubMed] [Google Scholar]
- 172. Sale MJ, Balmanno K, Saxena J, et al. MEK1/2 inhibitor withdrawal reverses acquired resistance driven by BRAF(V600E) amplification whereas KRAS(G13D) amplification promotes EMT‐chemoresistance. Nat Commun. 2019;10(1):2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Juric D, Castel P, Griffith M, et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature. 2015;518(7538):240‐244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Patch A‐M, Christie EL, Etemadmoghadam D, et al. Corrigendum: whole‐genome characterization of chemoresistant ovarian cancer. Nature. 2015;527(7578):398. [DOI] [PubMed] [Google Scholar]
- 175. Lee CK, Wu Y‐L, Ding PN, et al. Impact of specific epidermal growth factor receptor (EGFR) mutations and clinical characteristics on outcomes after treatment with EGFR tyrosine kinase inhibitors versus chemotherapy in EGFR‐mutant lung cancer: a meta‐analysis. J Clin Oncol. 2015;33(17):1958‐1965. [DOI] [PubMed] [Google Scholar]
- 176. Kancha RK, von Bubnoff N, Peschel C, Duyster J. Functional analysis of epidermal growth factor receptor (EGFR) mutations and potential implications for EGFR targeted therapy. Clin Cancer Res. 2009;15(2):460‐467. [DOI] [PubMed] [Google Scholar]
- 177. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127‐137. [DOI] [PubMed] [Google Scholar]
- 178. Ohashi K, Sequist LV, Arcila ME, et al. Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF gene mutations but lack mutations in KRAS, NRAS, or MEK1. Proc Natl Acad Sci USA. 2012;109(31):E2127‐E2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Karachaliou N, Codony‐Servat J, Teixido C, et al. BIM and mTOR expression levels predict outcome to erlotinib in EGFR‐mutant non‐small‐cell lung cancer. Sci Rep. 2015;5(1):17499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Kawabata S, Mercado‐Matos JR, Hollander MC, et al. Rapamycin prevents the development and progression of mutant epidermal growth factor receptor lung tumors with the acquired resistance mutation T790M. Cell Rep. 2014;7(6):1824‐1832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316(5827):1039‐1043. [DOI] [PubMed] [Google Scholar]
- 182. Shaw AT, Ou S‐HI, Bang Y‐J, et al. Crizotinib in ROS1‐rearranged non‐small‐cell lung cancer. N Engl J Med. 2014;371(21):1963‐1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Drilon A, Ou S‐HI, Cho BC, et al. Repotrectinib (TPX‐0005) is a next‐generation ROS1/TRK/ALK inhibitor that potently inhibits ROS1/TRK/ALK solvent‐ front mutations. Cancer Discov. 2018;8(10):1227‐1236. [DOI] [PubMed] [Google Scholar]
- 184. Katayama R, Gong B, Togashi N, et al. The new‐generation selective ROS1/NTRK inhibitor DS‐6051b overcomes crizotinib resistant ROS1‐G2032R mutation in preclinical models. Nat Commun. 2019;10(1):3604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Drilon A, Siena S, Ou S‐HI, et al. Safety and antitumor activity of the multitargeted pan‐TRK, ROS1, and ALK inhibitor entrectinib: combined results from two phase I trials (ALKA‐372‐001 and STARTRK‐1). Cancer Discov. 2017;7(4):400‐409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Watanabe J, Furuya N, Fujiwara Y. Appearance of a BRAF mutation conferring resistance to crizotinib in non‐small cell lung cancer harboring oncogenic ROS1 fusion. J Thorac Oncol. 2018;13(4):e66‐e69. [DOI] [PubMed] [Google Scholar]
- 187. Zhu Y‐C, Lin X‐P, Li X‐F, et al. Concurrent ROS1 gene rearrangement and KRAS mutation in lung adenocarcinoma: a case report and literature review. Thorac Cancer. 2018;9(1):159‐163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. McCoach CE, Le AT, Gowan K, et al. Resistance mechanisms to targeted therapies in ROS1(+) and ALK(+) non‐small cell lung cancer. Clin Cancer Res. 2018;24(14):3334‐3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. He Y, Sheng W, Hu W, et al. Different types of ROS1 fusion partners yield comparable efficacy to crizotinib. Oncol Res. 2019;27(8):901‐910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Zhu VW, Klempner SJ, Ou S‐HI. Receptor tyrosine kinase fusions as an actionable resistance mechanism to EGFR TKIs in EGFR‐mutant non‐small‐cell lung cancer. Trends Cancer. 2019;5(11):677‐692. [DOI] [PubMed] [Google Scholar]
- 191. Dagogo‐Jack I, Rooney M, Nagy RJ, et al. Molecular analysis of plasma from patients with ROS1‐positive NSCLC. J Thorac Oncol. 2019;14(5):816‐824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Sato H, Schoenfeld AJ, Siau E, et al. MAPK pathway alterations correlate with poor survival and drive resistance to therapy in patients with lung cancers driven by ROS1 fusions. Clin Cancer Res. 2020;26(12):2932‐2945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Cocco E, Scaltriti M, Drilon A. NTRK fusion‐positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 2018;15(12):731‐747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Gainor JF, Tseng D, Yoda S, et al. Patterns of metastatic spread and mechanisms of resistance to crizotinib in ROS1‐positive non‐small‐cell lung cancer. JCO Precis Oncol. 2017;2017:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Sehgal K, Piper‐Vallillo AJ, Viray H, Khan AM, Rangachari D, Costa DB. Cases of ROS1‐rearranged lung cancer: when to use crizotinib, entrectinib, lorlatinib, and beyond? Precis Cancer Med. 2020;3:17‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Li L‐Y, Guan Y‐d, Chen X‐S, Yang J‐M, Cheng Y. DNA repair pathways in cancer therapy and resistance. Front Pharmacol. 2020;11:629266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Sethy C, Kundu CN. 5‐Fluorouracil (5‐FU) resistance and the new strategy to enhance the sensitivity against cancer: implication of DNA repair inhibition. Biomed Pharmacother. 2021;137:111285. [DOI] [PubMed] [Google Scholar]
- 198. Liu J‐L, Huang W‐S, Lee K‐C, Tung S‐Y, Chen C‐N, Chang S‐F. Effect of 5‐fluorouracil on excision repair cross‐complementing 1 expression and consequent cytotoxicity regulation in human gastric cancer cells. J Cell Biochem. 2018;119(10):8472‐8480. [DOI] [PubMed] [Google Scholar]
- 199. Oliver JA, Ortiz R, Jimenez‐Luna C, et al. MMR‐proficient and MMR‐deficient colorectal cancer cells: 5‐fluorouracil treatment response and correlation to CD133 and MGMT expression. J Biosci. 2020;45(1):121. [PubMed] [Google Scholar]
- 200. Honecker F, Wermann H, Mayer F, et al. Microsatellite instability, mismatch repair deficiency, and BRAF mutation in treatment‐resistant germ cell tumors. J Clin Oncol. 2009;27(13):2129‐2136. [DOI] [PubMed] [Google Scholar]
- 201. Song M, Cui M, Liu K. Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem. 2022;232:114205. [DOI] [PubMed] [Google Scholar]
- 202. Bouwman P, Jonkers J. Molecular pathways: how can BRCA‐mutated tumors become resistant to PARP inhibitors? Clin Cancer Res. 2014;20(3):540‐547. [DOI] [PubMed] [Google Scholar]
- 203. Xiao Y, Lin F‐T, Lin W‐C. ACTL6A promotes repair of cisplatin‐induced DNA damage, a new mechanism of platinum resistance in cancer. Proc Natl Acad Sci USA. 2021;118(3):e2015808118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Weller M, Tabatabai G, Kastner B, et al. MGMT promoter methylation is a strong prognostic biomarker for benefit from dose‐intensified temozolomide rechallenge in progressive glioblastoma: the DIRECTOR trial. Clin Cancer Res. 2015;21(9):2057‐2064. [DOI] [PubMed] [Google Scholar]
- 205. Abe H, Natsumeda M, Okada M, et al. MGMT expression contributes to temozolomide resistance in H3K27M‐mutant diffuse midline gliomas. Front Oncol. 2019;9:1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Brandner S, McAleenan A, Kelly C, et al. MGMT promoter methylation testing to predict overall survival in people with glioblastoma treated with temozolomide: a comprehensive meta‐analysis based on a cochrane systematic review. Neuro Oncol. 2021;23(9):1457‐1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Jacot W, Lopez‐Crapez E, Mollevi C, et al. BRCA1 promoter hypermethylation is associated with good prognosis and chemosensitivity in triple‐negative breast cancer. Cancers (Basel). 2020;12(4):828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Mazloumi Z, Farahzadi R, Rafat A, et al. Effect of aberrant DNA methylation on cancer stem cell properties. Exp Mol Pathol. 2022;125:104757. [DOI] [PubMed] [Google Scholar]
- 209. Watson ZL, Yamamoto TM, McMellen A, et al. Histone methyltransferases EHMT1 and EHMT2 (GLP/G9A) maintain PARP inhibitor resistance in high‐grade serous ovarian carcinoma. Clin Epigenetics. 2019;11(1):165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Singh R, Fazal Z, Corbet AK, et al. Epigenetic remodeling through downregulation of polycomb repressive complex 2 mediates chemotherapy resistance in testicular germ cell tumors. Cancers (Basel). 2019;11(6):796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Patel N, Garikapati KR, Pandita RK, et al. Erratum: miR‐15a/miR‐16 down‐regulates BMI1, impacting Ub‐H2A mediated DNA repair and breast cancer cell sensitivity to doxorubicin. Sci Rep. 2017;7(1):12932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Qian L, Fei Q, Zhang H, et al. lncRNA HOTAIR promotes DNA repair and radioresistance of breast cancer via EZH2. DNA Cell Biol. 2020;39(12):2166‐2173. [DOI] [PubMed] [Google Scholar]
- 213. Edinger AL, Thompson CB. Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol. 2004;16(6):663‐669. [DOI] [PubMed] [Google Scholar]
- 214. Li M, Le W, Zhang X‐M, Zhang Y‐J, Jiang J, Liu P‐Y. The M476W/Q482H mutation of procaspase‐8 restored caspase‐8‐mediated apoptosis. Biochem Biophys Res Commun. 2019;514(3):653‐658. [DOI] [PubMed] [Google Scholar]
- 215. Roos WP, Thomas AD, Kaina B. DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer. 2016;16(1):20‐33. [DOI] [PubMed] [Google Scholar]
- 216. Zhang W, Konopleva M, Ruvolo VR, et al. Sorafenib induces apoptosis of AML cells via Bim‐mediated activation of the intrinsic apoptotic pathway. Leukemia. 2008;22(4):808‐818. [DOI] [PubMed] [Google Scholar]
- 217. Zuo J, Ishikawa T, Boutros S, Xiao Z, Humtsoe JO, Kramer RH. Bcl‐2 overexpression induces a partial epithelial to mesenchymal transition and promotes squamous carcinoma cell invasion and metastasis. Mol Cancer Res. 2010;8(2):170‐182. [DOI] [PubMed] [Google Scholar]
- 218. Real PJ, Sierra A, De Juan A, Segovia JC, Lopez‐Vega JM, Fernandez‐Luna JL. Resistance to chemotherapy via Stat3‐dependent overexpression of Bcl‐2 in metastatic breast cancer cells. Oncogene. 2002;21(50):7611‐7618. [DOI] [PubMed] [Google Scholar]
- 219. Ritter V, Krautter F, Klein D, Jendrossek V, Rudner J. Bcl‐2/Bcl‐xL inhibitor ABT‐263 overcomes hypoxia‐driven radioresistence and improves radiotherapy. Cell Death Dis. 2021;12(7):694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Roberts AW, Seymour JF, Brown JR, et al. Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease. J Clin Oncol. 2012;30(5):488‐496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Kipps TJ, Eradat H, Grosicki S, et al. A phase 2 study of the BH3 mimetic BCL2 inhibitor navitoclax (ABT‐263) with or without rituximab, in previously untreated B‐cell chronic lymphocytic leukemia. Leuk Lymphoma. 2015;56(10):2826‐2833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Bose P, Gandhi V, Konopleva M. Pathways and mechanisms of venetoclax resistance. Leuk Lymphoma. 2017;58(9):1‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Salimi‐Jeda A, Ghabeshi S, Gol Mohammad Pour Z, et al. Autophagy modulation and cancer combination therapy: a smart approach in cancer therapy. Cancer Treat Res Commun. 2022;30:100512. [DOI] [PubMed] [Google Scholar]
- 224. Liu X‐L, Ding J, Meng L‐H. Oncogene‐induced senescence: a double edged sword in cancer. Acta Pharmacol Sin. 2018;39(10):1553‐1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Lorente J, Velandia C, Leal JA, et al. The interplay between autophagy and tumorigenesis: exploiting autophagy as a means of anticancer therapy. Biol Rev Camb Philos Soc. 2018;93(1):152‐165. [DOI] [PubMed] [Google Scholar]
- 226. Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications. Mol Cancer Ther. 2011;10(9):1533‐1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Wang J, Wu GS. Role of autophagy in cisplatin resistance in ovarian cancer cells. J Biol Chem. 2014;289(24):17163‐17173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Castro I, Sampaio‐Marques B, Ludovico P. Targeting metabolic reprogramming in acute myeloid leukemia. Cells. 2019;8(9):967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Wong MM‐T, Chan H‐Y, Aziz NA, et al. Interplay of autophagy and cancer stem cells in hepatocellular carcinoma. Mol Biol Rep. 2021;48(4):3695‐3717. [DOI] [PubMed] [Google Scholar]
- 230. Wen Y‐A, Xing X, Harris JW, et al. Adipocytes activate mitochondrial fatty acid oxidation and autophagy to promote tumor growth in colon cancer. Cell Death Dis. 2017;8(2):e2593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Amaravadi RK, Yu D, Lum JJ, et al. Autophagy inhibition enhances therapy‐induced apoptosis in a Myc‐induced model of lymphoma. J Clin Invest. 2007;117(2):326‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Han W, Pan H, Chen Y, et al. EGFR tyrosine kinase inhibitors activate autophagy as a cytoprotective response in human lung cancer cells. PLoS One. 2011;6(6):e18691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Gupta A, Roy S, Lazar AJF, et al. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST). Proc Natl Acad Sci USA. 2010;107(32):14333‐14338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Bellodi C, Lidonnici MR, Hamilton A, et al. Targeting autophagy potentiates tyrosine kinase inhibitor‐induced cell death in Philadelphia chromosome‐positive cells, including primary CML stem cells. J Clin Invest. 2009;119(5):1109‐1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron‐dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060‐1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Zhang C, Liu X, Jin S, Chen Y, Guo R. Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer. 2022;21(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1‐2):317‐331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Forcina GC, Dixon SJ. GPX4 at the crossroads of lipid homeostasis and ferroptosis. Proteomics. 2019;19(18):e1800311. [DOI] [PubMed] [Google Scholar]
- 239. Chen T‐C, Chuang J‐Y, Ko C‐Y, et al. AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide‐resistant glioblastoma through disrupting GPX4‐Mediated redox homeostasis. Redox Biol. 2020;30:101413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Yang C, Zhang Y, Lin S, Liu Y, Li W. Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging (Albany NY). 2021;13(10):13515‐13534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Roh J‐L, Kim EH, Jang HJ, Park JY, Shin D. Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer. Cancer Lett. 2016;381(1):96‐103. [DOI] [PubMed] [Google Scholar]
- 242. Fu D, Wang C, Yu L, Yu R. Induction of ferroptosis by ATF3 elevation alleviates cisplatin resistance in gastric cancer by restraining Nrf2/Keap1/xCT signaling. Cell Mol Biol Lett. 2021;26(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Du J, Wang X, Li Y, et al. DHA exhibits synergistic therapeutic efficacy with cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via modulation of iron metabolism. Cell Death Dis. 2021;12(7):705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Turcu AL, Versini A, Khene N, et al. DMT1 inhibitors kill cancer stem cells by blocking lysosomal iron translocation. Chemistry. 2020;26(33):7369‐7373. [DOI] [PubMed] [Google Scholar]
- 245. Chaudhary N, Choudhary BS, Shah SG, et al. Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer. Int J Cancer. 2021;149(7):1495‐1511. [DOI] [PubMed] [Google Scholar]
- 246. Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13(1):91‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Ye Z, Hu Q, Zhuo Q, et al. Abrogation of ARF6 promotes RSL3‐induced ferroptosis and mitigates gemcitabine resistance in pancreatic cancer cells. Am J Cancer Res. 2020;10(4):1182‐1193. [PMC free article] [PubMed] [Google Scholar]
- 248. Tsvetkov P, Coy S, Petrova B, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375(6586):1254‐1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Mayr JA, Feichtinger RG, Tort F, Ribes A, Sperl W. Lipoic acid biosynthesis defects. J Inherit Metab Dis. 2014;37(4):553‐563. [DOI] [PubMed] [Google Scholar]
- 250. Solmonson A, DeBerardinis RJ. Lipoic acid metabolism and mitochondrial redox regulation. J Biol Chem. 2018;293(20):7522‐7530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Gaggelli E, Kozlowski H, Valensin D, Valensin G. Copper homeostasis and neurodegenerative disorders (Alzheimer's, prion, and Parkinson's diseases and amyotrophic lateral sclerosis). Chem Rev. 2006;106(6):1995‐2044. [DOI] [PubMed] [Google Scholar]
- 252. Bandmann O, Weiss KH, Kaler SG. Wilson's disease and other neurological copper disorders. Lancet Neurol. 2015;14(1):103‐113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Tsang T, Posimo JM, Gudiel AA, Cicchini M, Feldser DM, Brady DC. Copper is an essential regulator of the autophagic kinases ULK1/2 to drive lung adenocarcinoma. Nat Cell Biol. 2020;22(4):412‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Cui L, Gouw AM, LaGory EL, et al. Mitochondrial copper depletion suppresses triple‐negative breast cancer in mice. Nat Biotechnol. 2021;39(3):357‐367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Aggarwal A, Bhatt M. Advances in treatment of Wilson disease. Tremor Other Hyperkinet Mov (NY). 2018;8:525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. O'Day SJ, Eggermont AMM, Chiarion‐Sileni V, et al. Final results of phase III SYMMETRY study: randomized, double‐blind trial of elesclomol plus paclitaxel versus paclitaxel alone as treatment for chemotherapy‐naive patients with advanced melanoma. J Clin Oncol. 2013;31(9):1211‐1218. [DOI] [PubMed] [Google Scholar]
- 257. Tang D, Chen X, Kroemer G. Cuproptosis: a copper‐triggered modality of mitochondrial cell death. Cell Res. 2022;32(5):417‐418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 2019;575(7782):299‐309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Lambrechts D, Wauters E, Boeckx B, et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 2018;24(8):1277‐1289. [DOI] [PubMed] [Google Scholar]
- 260. Burgos‐Panadero R, Lucantoni F, Gamero‐Sandemetrio E, de la Cruz‐Merino L, Alvaro T, Noguera R. The tumour microenvironment as an integrated framework to understand cancer biology. Cancer Lett. 2019;461:112‐122. [DOI] [PubMed] [Google Scholar]
- 261. Leroy J, Jamali F, Forbes L, et al. Laparoscopic total mesorectal excision (TME) for rectal cancer surgery: long‐term outcomes. Surg Endosc. 2004;18(2):281‐289. [DOI] [PubMed] [Google Scholar]
- 262. Henke E, Nandigama R, Ergun S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci. 2019;6:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Yu Q, Xiao W, Sun S, Sohrabi A, Liang J, Seidlits SK. Extracellular matrix proteins confer cell adhesion‐mediated drug resistance through integrin α v in glioblastoma cells. Front Cell Dev Biol. 2021;9:616580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Misra S, Ghatak S, Zoltan‐Jones A, Toole BP. Regulation of multidrug resistance in cancer cells by hyaluronan. J Biol Chem. 2003;278(28):25285‐25288. [DOI] [PubMed] [Google Scholar]
- 265. Park CC, Zhang HJ, Yao ES, Park CJ, Bissell MJ. Beta1 integrin inhibition dramatically enhances radiotherapy efficacy in human breast cancer xenografts. Cancer Res. 2008;68(11):4398‐4405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2‐targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res Treat. 2010;122(1):35‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Zigrino P, Nischt R, Mauch C. The disintegrin‐like and cysteine‐rich domains of ADAM‐9 mediate interactions between melanoma cells and fibroblasts. J Biol Chem. 2011;286(8):6801‐6807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. de la Fuente MT, Casanova B, Moyano JV, et al. Engagement of alpha4beta1 integrin by fibronectin induces in vitro resistance of B chronic lymphocytic leukemia cells to fludarabine. J Leukoc Biol. 2002;71(3):495‐502. [PubMed] [Google Scholar]
- 269. Matsunaga T, Takemoto N, Sato T, et al. CORRIGENDUM: interaction between leukemic‐cell VLA‐4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia. Nat Med. 2005;11(5):578. [DOI] [PubMed] [Google Scholar]
- 270. Nishio M, Endo T, Tsukada N, et al. Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF‐1alpha. Blood. 2005;106(3):1012‐1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Chen Y, Jacamo R, Konopleva M, Garzon R, Croce C, Andreeff M. CXCR4 downregulation of let‐7a drives chemoresistance in acute myeloid leukemia. J Clin Invest. 2013;123(6):2395‐2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Margolin DA, Silinsky J, Grimes C, et al. Lymph node stromal cells enhance drug‐resistant colon cancer cell tumor formation through SDF‐1alpha/CXCR4 paracrine signaling. Neoplasia. 2011;13(9):874‐886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32(3‐4):623‐642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255‐289. [DOI] [PubMed] [Google Scholar]
- 275. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654‐659. [DOI] [PubMed] [Google Scholar]
- 276. Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteomics. 2010;73(10):1907‐1920. [DOI] [PubMed] [Google Scholar]
- 277. D'Asti E, Garnier D, Lee TH, Montermini L, Meehan B, Rak J. Oncogenic extracellular vesicles in brain tumor progression. Front Physiol. 2012;3:294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Zhang H‐G, Grizzle WE. Exosomes: a novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am J Pathol. 2014;184(1):28‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19(11):1423‐1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280. Deep G, Panigrahi GK. Hypoxia‐induced signaling promotes prostate cancer progression: exosomes role as messenger of hypoxic response in tumor microenvironment. Crit Rev Oncog. 2015;20(5‐6):419‐434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Webber J, Steadman R, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010;70(23):9621‐9630. [DOI] [PubMed] [Google Scholar]
- 282. Zhang Q, Peng C. Cancer‐associated fibroblasts regulate the biological behavior of cancer cells and stroma in gastric cancer. Oncol Lett. 2018;15(1):691‐698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE, Hill R. Cancer‐associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 2017;36(13):1770‐1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Shedden K, Xie XT, Chandaroy P, Chang YT, Rosania GR. Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Cancer Res. 2003;63(15):4331‐4337. [PubMed] [Google Scholar]
- 285. Corrado C, Raimondo S, Chiesi A, Ciccia F, De Leo G, Alessandro R. Exosomes as intercellular signaling organelles involved in health and disease: basic science and clinical applications. Int J Mol Sci. 2013;14(3):5338‐5366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Maleki S, Jabalee J, Garnis C. The role of extracellular vesicles in mediating resistance to anticancer therapies. Int J Mol Sci. 2021;22(8):4166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Muralidharan‐Chari V, Kohan HG, Asimakopoulos AG, et al. Microvesicle removal of anticancer drugs contributes to drug resistance in human pancreatic cancer cells. Oncotarget. 2016;7(31):50365‐50379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Ning K, Wang T, Sun X, et al. UCH‐L1‐containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J Surg Oncol. 2017;115(8):932‐940. [DOI] [PubMed] [Google Scholar]
- 289. Crow J, Atay S, Banskota S, Artale B, Schmitt S, Godwin AK. Exosomes as mediators of platinum resistance in ovarian cancer. Oncotarget. 2017;8(7):11917‐11936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Han M, Hu J, Lu P, et al. Exosome‐transmitted miR‐567 reverses trastuzumab resistance by inhibiting ATG5 in breast cancer. Cell Death Dis. 2020;11(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 291. Santos JC, Lima NdS, Sarian LO, Matheu A, Ribeiro ML, Derchain SFM. Exosome‐mediated breast cancer chemoresistance via miR‐155 transfer. Sci Rep. 2018;8(1):829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Dong H, Wang W, Chen R, et al. Exosome‐mediated transfer of lncRNA‑SNHG14 promotes trastuzumab chemoresistance in breast cancer. Int J Oncol. 2018;53(3):1013‐1026. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 293. Jing C, Cao H, Qin X, et al. Exosome‐mediated gefitinib resistance in lung cancer HCC827 cells via delivery of miR‐21. Oncol Lett. 2018;15(6):9811‐9817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Fu X, Liu M, Qu S, et al. Exosomal microRNA‐32‐5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J Exp Clin Cancer Res. 2018;37(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 295. Fornari F, Pollutri D, Patrizi C, et al. In hepatocellular carcinoma miR‐221 modulates sorafenib resistance through inhibition of caspase‐3‐mediated apoptosis. Clin Cancer Res. 2017;23(14):3953‐3965. [DOI] [PubMed] [Google Scholar]
- 296. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 2005;23(5):1011‐1027. [DOI] [PubMed] [Google Scholar]
- 297. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182‐1186. [DOI] [PubMed] [Google Scholar]
- 298. Liang H, Wang M. Prospect of immunotherapy combined with anti‐angiogenic agents in patients with advanced non‐small cell lung cancer. Cancer Manag Res. 2019;11:7707‐7719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Gacche RN. Compensatory angiogenesis and tumor refractoriness. Oncogenesis. 2015;4(6):e153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet. 2016;388(10043):518‐529. [DOI] [PubMed] [Google Scholar]
- 301. Gacche RN, Assaraf YG. Redundant angiogenic signaling and tumor drug resistance. Drug Resist Updat. 2018;36:47‐76. [DOI] [PubMed] [Google Scholar]
- 302. Ribatti D. The chick embryo chorioallantoic membrane (CAM). A multifaceted experimental model. Mech Dev. 2016;141:70‐77. [DOI] [PubMed] [Google Scholar]
- 303. Lohela M, Bry M, Tammela T, Alitalo K. VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol. 2009;21(2):154‐165. [DOI] [PubMed] [Google Scholar]
- 304. Ellis LM, Hicklin DJ. Pathways mediating resistance to vascular endothelial growth factor‐targeted therapy. Clin Cancer Res. 2008;14(20):6371‐6375. [DOI] [PubMed] [Google Scholar]
- 305. Crawford Y, Kasman I, Yu L, et al. PDGF‐C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti‐VEGF treatment. Cancer Cell. 2009;15(1):21‐34. [DOI] [PubMed] [Google Scholar]
- 306. Vasudev NS, Reynolds AR. Anti‐angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis. 2014;17(3):471‐494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Bergers G, Song S. The role of pericytes in blood‐vessel formation and maintenance. Neuro Oncol. 2005;7(4):452‐464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Aldea M, Andre F, Marabelle A, Dogan S, Barlesi F, Soria J‐C. Overcoming resistance to tumor‐targeted and immune‐targeted therapies. Cancer Discov. 2021;11(4):874‐899. [DOI] [PubMed] [Google Scholar]
- 309. Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to therapeutic PD‐1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7(1):10501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Saleh R, Elkord E. Treg‐mediated acquired resistance to immune checkpoint inhibitors. Cancer Lett. 2019;457:168‐179. [DOI] [PubMed] [Google Scholar]
- 311. Jenkins L, Jungwirth U, Avgustinova A, et al. Cancer‐associated fibroblasts suppress CD8+ T‐cell infiltration and confer resistance to immune‐checkpoint blockade. Cancer Res. 2022;82(16):2904‐2917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312. Deng L, Liang H, Burnette B, et al. Irradiation and anti‐PD‐L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124(2):687‐695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313. Twyman‐Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non‐redundant immune mechanisms in cancer. Nature. 2015;520(7547):373‐377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314. Galluzzi L, Humeau J, Buque A, Zitvogel L, Kroemer G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol. 2020;17(12):725‐741. [DOI] [PubMed] [Google Scholar]
- 315. Tang T, Huang X, Zhang G, Hong Z, Bai X, Liang T. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct Target Ther. 2021;6(1):72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Wallin JJ, Bendell JC, Funke R, et al. Atezolizumab in combination with bevacizumab enhances antigen‐specific T‐cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7(1):12624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317. Casak SJ, Donoghue M, Fashoyin‐Aje L, et al. FDA approval summary: atezolizumab plus bevacizumab for the treatment of patients with advanced unresectable or metastatic hepatocellular carcinoma. Clin Cancer Res. 2021;27(7):1836‐1841. [DOI] [PubMed] [Google Scholar]
- 318. Harrington K, Freeman DJ, Kelly B, Harper J, Soria J‐C. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discov. 2019;18(9):689‐706. [DOI] [PubMed] [Google Scholar]
- 319. Martin NT, Bell JC. Oncolytic virus combination therapy: killing one bird with two stones. Mol Ther. 2018;26(6):1414‐1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320. Bourgeois‐Daigneault M‐C, Roy DG, Aitken AS, et al. Neoadjuvant oncolytic virotherapy before surgery sensitizes triple‐negative breast cancer to immune checkpoint therapy. Sci Transl Med. 2018;10(422):eaao1641. [DOI] [PubMed] [Google Scholar]
- 321. Li R, Zhang J, Gilbert SM, Conejo‐Garcia J, Mule JJ. Using oncolytic viruses to ignite the tumour immune microenvironment in bladder cancer. Nat Rev Urol. 2021;18(9):543‐555. [DOI] [PubMed] [Google Scholar]
- 322. Ribas A, Dummer R, Puzanov I, et al. Oncolytic virotherapy promotes intratumoral t cell infiltration and improves anti‐PD‐1 immunotherapy. Cell. 2017;170(6):1109‐1119.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323. Zhang X, Chen X, Guo Y, et al. Dual gate‐controlled therapeutics for overcoming bacterium‐induced drug resistance and potentiating cancer immunotherapy. Angew Chem Int Ed Engl. 2021;60(25):14013‐14021. [DOI] [PubMed] [Google Scholar]
- 324. Arthur JC, Perez‐Chanona E, Muhlbauer M, et al. Intestinal inflammation targets cancer‐inducing activity of the microbiota. Science. 2012;338(6103):120‐123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325. Dejea CM, Fathi P, Craig JM, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359(6375):592‐597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326. Zheng D‐W, Dong X, Pan P, et al. Phage‐guided modulation of the gut microbiota of mouse models of colorectal cancer augments their responses to chemotherapy. Nat Biomed Eng. 2019;3(9):717‐728. [DOI] [PubMed] [Google Scholar]
- 327. Han H, Hou Y, Chen X, et al. Metformin‐induced stromal depletion to enhance the penetration of gemcitabine‐loaded magnetic nanoparticles for pancreatic cancer targeted therapy. J Am Chem Soc. 2020;142(10):4944‐4954. [DOI] [PubMed] [Google Scholar]
- 328. Geller LT, Barzily‐Rokni M, Danino T, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357(6356):1156‐1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329. Dong X, Pan P, Zheng D‐W, Bao P, Zeng X, Zhang X‐Z. Bioinorganic hybrid bacteriophage for modulation of intestinal microbiota to remodel tumor‐immune microenvironment against colorectal cancer. Sci Adv. 2020;6(20):eaba1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967‐970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331. Pernigoni N, Zagato E, Calcinotto A, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374(6564):216‐224. [DOI] [PubMed] [Google Scholar]
- 332. Zhu Y‐X, Jia H‐R, Duan Q‐Y, Wu F‐G. Nanomedicines for combating multidrug resistance of cancer. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2021;13(5):e1715. [DOI] [PubMed] [Google Scholar]
- 333. Xiong M‐H, Bao Y, Du X‐J, et al. Differential anticancer drug delivery with a nanogel sensitive to bacteria‐accumulated tumor artificial environment. ACS Nano. 2013;7(12):10636‐10645. [DOI] [PubMed] [Google Scholar]
- 334. Lawson DA, Bhakta NR, Kessenbrock K, et al. Single‐cell analysis reveals a stem‐cell program in human metastatic breast cancer cells. Nature. 2015;526(7571):131‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335. Mani SA, Guo W, Liao M‐J, et al. The epithelial‐mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704‐715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336. Shimokawa M, Ohta Y, Nishikori S, et al. Visualization and targeting of LGR5(+) human colon cancer stem cells. Nature. 2017;545(7653):187‐192. [DOI] [PubMed] [Google Scholar]
- 337. Soliman AA, Elzarkaa AA, Malik E. Epithelial ovarian cancer and cancer stem cells. Adv Exp Med Biol. 2021;1330:21‐32. [DOI] [PubMed] [Google Scholar]
- 338. Zhou H‐M, Zhang J‐G, Zhang X, Li Q. Targeting cancer stem cells for reversing therapy resistance: mechanism, signaling, and prospective agents. Signal Transduct Target Ther. 2021;6(1):62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Wang H, Mei Y, Luo C, et al. Single‐cell analyses reveal mechanisms of cancer stem cell maintenance and epithelial‐mesenchymal transition in recurrent bladder cancer. Clin Cancer Res. 2021;27(22):6265‐6278. [DOI] [PubMed] [Google Scholar]
- 340. Shi Z‐D, Hao L, Han X‐X, et al. Targeting HNRNPU to overcome cisplatin resistance in bladder cancer. Mol Cancer. 2022;21(1):37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341. He X, Smith SE, Chen S, et al. Tumor‐initiating stem cell shapes its microenvironment into an immunosuppressive barrier and pro‐tumorigenic niche. Cell Rep. 2021;36(10):109674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Wang D, Fu L, Sun H, Guo L, DuBois RN. Prostaglandin E2 promotes colorectal cancer stem cell expansion and metastasis in mice. Gastroenterology. 2015;149(7):1884‐1895.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Sun L, Huang C, Zhu M, et al. Gastric cancer mesenchymal stem cells regulate PD‐L1‐CTCF enhancing cancer stem cell‐like properties and tumorigenesis. Theranostics. 2020;10(26):11950‐11962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344. Wei F, Zhang T, Deng S‐C, et al. PD‐L1 promotes colorectal cancer stem cell expansion by activating HMGA1‐dependent signaling pathways. Cancer Lett. 2019;450:1‐13. [DOI] [PubMed] [Google Scholar]
- 345. Ouadah Y, Rojas ER, Riordan DP, Capostagno S, Kuo CS, Krasnow MA. Rare pulmonary neuroendocrine cells are stem cells regulated by Rb, p53, and notch. Cell. 2019;179(2):403‐416.e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346. Novellasdemunt L, Kucharska A, Jamieson C, et al. NEDD4 and NEDD4L regulate Wnt signalling and intestinal stem cell priming by degrading LGR5 receptor. EMBO J. 2020;39(3):e102771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347. Zhang L, He X, Liu X, et al. Single‐cell transcriptomics in medulloblastoma reveals tumor‐initiating progenitors and oncogenic cascades during tumorigenesis and relapse. Cancer Cell. 2019;36(3):302‐318.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348. Deslauriers AG, Kotini AG, Papapetrou EP. Modeling leukemia stem cells with patient‐derived induced pluripotent stem cells. Methods Mol Biol. 2021;2185:411‐422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 349. Chaffer CL, San Juan BP, Lim E, Weinberg RA. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016;35(4):645‐654. [DOI] [PubMed] [Google Scholar]
- 350. Na T‐Y, Schecterson L, Mendonsa AM, Gumbiner BM. The functional activity of E‐cadherin controls tumor cell metastasis at multiple steps. Proc Natl Acad Sci USA. 2020;117(11):5931‐5937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351. Han LL, Jia L, Wu F, Huang C. Sirtuin6 (SIRT6) promotes the EMT of hepatocellular carcinoma by stimulating autophagic degradation of E‐cadherin. Mol Cancer Res. 2019;17(11):2267‐2280. [DOI] [PubMed] [Google Scholar]
- 352. Zhang J, Cai H, Sun L, et al. LGR5, a novel functional glioma stem cell marker, promotes EMT by activating the Wnt/beta‐catenin pathway and predicts poor survival of glioma patients. J Exp Clin Cancer Res. 2018;37(1):225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353. Tang Q, Chen J, Di Z, et al. TM4SF1 promotes EMT and cancer stemness via the Wnt/beta‐catenin/SOX2 pathway in colorectal cancer. J Exp Clin Cancer Res. 2020;39(1):232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354. Zhang N, Hua X, Tu H, Li J, Zhang Z, Max C. Isorhapontigenin (ISO) inhibits EMT through FOXO3A/METTL14/VIMENTIN pathway in bladder cancer cells. Cancer Lett. 2021;520:400‐408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355. Tian H, Lian R, Li Y, et al. AKT‐induced lncRNA VAL promotes EMT‐independent metastasis through diminishing Trim16‐dependent Vimentin degradation. Nat Commun. 2020;11(1):5127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356. Luo J, Yao J‐F, Deng X‐F, et al. 14, 15‐EET induces breast cancer cell EMT and cisplatin resistance by up‐regulating integrin alphavbeta3 and activating FAK/PI3K/AKT signaling. J Exp Clin Cancer Res. 2018;37(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 357. Kim TW, Lee YS, Yun NH, et al. MicroRNA‐17‐5p regulates EMT by targeting Vimentin in colorectal cancer. Br J Cancer. 2020;123(7):1123‐1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358. Sinha S, Pattnaik S, Aradhyam GK. Molecular evolution guided functional analyses reveals nucleobindin‐1 as a canonical E‐box binding protein promoting epithelial‐to‐mesenchymal transition (EMT). Biochim Biophys Acta: Proteins Proteomics. 2019;1867(9):765‐775. [DOI] [PubMed] [Google Scholar]
- 359. Liang S‐Q, Marti TM, Dorn P, et al. Blocking the epithelial‐to‐mesenchymal transition pathway abrogates resistance to anti‐folate chemotherapy in lung cancer. Cell Death Dis. 2015;6(7):e1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360. Torzilli PA, Bourne JW, Cigler T, Vincent CT. A new paradigm for mechanobiological mechanisms in tumor metastasis. Semin Cancer Biol. 2012;22(5‐6):385‐395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361. Zhu Y, Tan J, Xie H, Wang J, Meng X, Wang R. HIF‐1alpha regulates EMT via the Snail and beta‐catenin pathways in paraquat poisoning‐induced early pulmonary fibrosis. J Cell Mol Med. 2016;20(4):688‐697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362. Zhang P, Sun Y, Ma L. ZEB1: at the crossroads of epithelial‐mesenchymal transition, metastasis and therapy resistance. Cell Cycle. 2015;14(4):481‐487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 363. Xu Y, Lee D‐K, Feng Z, et al. Breast tumor cell‐specific knockout of Twist1 inhibits cancer cell plasticity, dissemination, and lung metastasis in mice. Proc Natl Acad Sci USA. 2017;114(43):11494‐11499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364. Zhang W, Feng M, Zheng G, et al. Chemoresistance to 5‐fluorouracil induces epithelial‐mesenchymal transition via up‐regulation of Snail in MCF7 human breast cancer cells. Biochem Biophys Res Commun. 2012;417(2):679‐685. [DOI] [PubMed] [Google Scholar]
- 365. Wang H, Zhang G, Zhang H, et al. Acquisition of epithelial‐mesenchymal transition phenotype and cancer stem cell‐like properties in cisplatin‐resistant lung cancer cells through AKT/beta‐catenin/Snail signaling pathway. Eur J Pharmacol. 2014;723:156‐166. [DOI] [PubMed] [Google Scholar]
- 366. Namba T, Kodama R, Moritomo S, Hoshino T, Mizushima T. Zidovudine, an anti‐viral drug, resensitizes gemcitabine‐resistant pancreatic cancer cells to gemcitabine by inhibition of the Akt‐GSK3beta‐Snail pathway. Cell Death Dis. 2015;6(6):e1795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367. Fischer KR, Durrans A, Lee S, et al. Epithelial‐to‐mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 368. Jin H, He Y, Zhao P, et al. Targeting lipid metabolism to overcome EMT‐associated drug resistance via integrin beta3/FAK pathway and tumor‐associated macrophage repolarization using legumain‐activatable delivery. Theranostics. 2019;9(1):265‐278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369. Raoof S, Mulford IJ, Frisco‐Cabanos H, et al. Targeting FGFR overcomes EMT‐mediated resistance in EGFR mutant non‐small cell lung cancer. Oncogene. 2019;38(37):6399‐6413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 370. Sheng W, Chen C, Dong M, et al. Calreticulin promotes EGF‐induced EMT in pancreatic cancer cells via Integrin/EGFR‐ERK/MAPK signaling pathway. Cell Death Dis. 2017;8(10):e3147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 371. Wu Q, Ma J, Wei J, Meng W, Wang Y, Shi M. lncRNA SNHG11 promotes gastric cancer progression by activating the Wnt/beta‐catenin pathway and oncogenic autophagy. Mol Ther. 2021;29(3):1258‐1278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 372. Cheng F, Shen Y, Mohanasundaram P, et al. Vimentin coordinates fibroblast proliferation and keratinocyte differentiation in wound healing via TGF‐beta‐Slug signaling. Proc Natl Acad Sci USA. 2016;113(30):E4320‐E4327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373. Kariya Y, Oyama M, Suzuki T, Kariya Y. Alphavbeta3 integrin induces partial EMT independent of TGF‐beta signaling. Commun Biol. 2021;4(1):490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 374. Gachpazan M, Kashani H, Hassanian SM, et al. Therapeutic potential of targeting transforming growth factor‐beta in colorectal cancer: rational and progress. Curr Pharm Des. 2019;25(38):4085‐4089. [DOI] [PubMed] [Google Scholar]
- 375. Katoh M, Katoh M. Molecular genetics and targeted therapy of WNT‐related human diseases (Review). Int J Mol Med. 2017;40(3):587‐606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376. Alavijeh MS, Palmer AM. The pivotal role of drug metabolism and pharmacokinetics in the discovery and development of new medicines. Curr Opin Investig Drugs. 2004;7(8):755‐763. [PubMed] [Google Scholar]
- 377. Palmer AM. New horizons in drug metabolism, pharmacokinetics and drug discovery. Drug News Perspect. 2003;16(1):57‐62. [PubMed] [Google Scholar]
- 378. Sacks LV, Shamsuddin HH, Yasinskaya YI, Bouri K, Lanthier ML, Sherman RE. Scientific and regulatory reasons for delay and denial of FDA approval of initial applications for new drugs, 2000–2012. JAMA. 2014;311(4):378‐384. [DOI] [PubMed] [Google Scholar]
- 379. Massard C, Michiels S, Ferte C, et al. High‐throughput genomics and clinical outcome in hard‐to‐treat advanced cancers: results of the MOSCATO 01 trial. Cancer Discov. 2017;7(6):586‐595. [DOI] [PubMed] [Google Scholar]
- 380. Bertucci F, Goncalves A, Guille A, et al. Prospective high‐throughput genome profiling of advanced cancers: results of the PERMED‐01 clinical trial. Genome Med. 2021;13(1):87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 381. Dellett M, O'Hagan KA, Colyer HAA, Mills KI. Identification of gene networks associated with acute myeloid leukemia by comparative molecular methylation and expression profiling. Biomarkers Cancer. 2010;2:43‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 382. Qu X, Davison J, Du L, et al. Identification of differentially methylated markers among cytogenetic risk groups of acute myeloid leukemia. Epigenetics. 2015;10(6):526‐535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383. Valerio LG Jr, Choudhuri S. Chemoinformatics and chemical genomics: potential utility of in silico methods. J Appl Toxicol. 2012;32(11):880‐889. [DOI] [PubMed] [Google Scholar]
- 384. Tratnyek PG, Bylaska EJ, Weber EJ. In silico environmental chemical science: properties and processes from statistical and computational modelling. Environ Sci Process Impacts. 2017;19(3):188‐202. [DOI] [PubMed] [Google Scholar]
- 385. Wolfram J, Ferrari M. Clinical cancer nanomedicine. Nano Today. 2019;25:85‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 386. Deirram N, Zhang C, Kermaniyan SS, Johnston APR, Such GK. pH‐responsive polymer nanoparticles for drug delivery. Macromol Rapid Commun. 2019;40(10):e1800917. [DOI] [PubMed] [Google Scholar]
- 387. Qiao Y, Wan J, Zhou L, et al. Stimuli‐responsive nanotherapeutics for precision drug delivery and cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11(1):e1527. [DOI] [PubMed] [Google Scholar]
- 388. Xiao X, Wang K, Zong Q, Tu Y, Dong Y, Yuan Y. Polyprodrug with glutathione depletion and cascade drug activation for multi‐drug resistance reversal. Biomaterials. 2021;270:120649. [DOI] [PubMed] [Google Scholar]
- 389. Chen Z, Wang Z, Gu Z. Bioinspired and biomimetic nanomedicines. ACC Chem Res. 2019;52(5):1255‐1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390. Shen S, Xu X, Lin S, et al. A nanotherapeutic strategy to overcome chemotherapeutic resistance of cancer stem‐like cells. Nat Nanotechnol. 2021;16(1):104‐113. [DOI] [PubMed] [Google Scholar]
- 391. Guo Y, Zhang X, Sun W, et al. Metal–phenolic network‐based nanocomplexes that evoke ferroptosis by apoptosis: promoted nuclear drug influx and reversed drug resistance of cancer. Chem Mater. 2019;31(24):10071‐10084. [Google Scholar]
- 392. Toki T, Iwasaki T, Ishii M. Topiramate blood levels during polytherapy for epilepsy in children. Am J Ther. 2019;26(1):e18‐e24. [DOI] [PubMed] [Google Scholar]
- 393. Lyons TG. Targeted therapies for triple‐negative breast cancer. Curr Treat Options Oncol. 2019;20(11):82. [DOI] [PubMed] [Google Scholar]
- 394. Owen CN, Shoushtari AN, Chauhan D, et al. Management of early melanoma recurrence despite adjuvant anti‐PD‐1 antibody therapy. Ann Oncol. 2020;31(8):1075‐1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 395. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev. 2013;65(1):36‐48. [DOI] [PubMed] [Google Scholar]
- 396. Murakami T. Absorption sites of orally administered drugs in the small intestine. Expert Opin Drug Discov. 2017;12(12):1219‐1232. [DOI] [PubMed] [Google Scholar]
- 397. Meira Menezes T, Assis C, Lacerda Cintra AJ, et al. Binding mechanism between acetylcholinesterase and drugs pazopanib and lapatinib: biochemical and biophysical studies. ACS Chem Neurosci. 2021;12(24):4500‐4511. [DOI] [PubMed] [Google Scholar]
- 398. de Jonge MJA, Hamberg P, Verweij J, et al. Phase I and pharmacokinetic study of pazopanib and lapatinib combination therapy in patients with advanced solid tumors. Invest New Drugs. 2013;31(3):751‐759. [DOI] [PubMed] [Google Scholar]
- 399. Monk BJ, Mas Lopez L, Zarba JJ, et al. Phase II, open‐label study of pazopanib or lapatinib monotherapy compared with pazopanib plus lapatinib combination therapy in patients with advanced and recurrent cervical cancer. J Clin Oncol. 2010;28(22):3562‐3569. [DOI] [PubMed] [Google Scholar]
- 400. Olaussen KA, Commo F, Tailler M, et al. Synergistic proapoptotic effects of the two tyrosine kinase inhibitors pazopanib and lapatinib on multiple carcinoma cell lines. Oncogene. 2009;28(48):4249‐4260. [DOI] [PubMed] [Google Scholar]
- 401. Tavallai S, Hamed HA, Grant S, Poklepovic A, Dent P. Pazopanib and HDAC inhibitors interact to kill sarcoma cells. Cancer Biol Ther. 2014;15(5):578‐585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 402. Dean AQ, Luo S, Twomey JD, Zhang B. Targeting cancer with antibody‐drug conjugates: promises and challenges. mAbs. 2021;13(1):1951427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 403. Tsujii S, Serada S, Fujimoto M, et al. Glypican‐1 is a novel target for stroma and tumor cell dual‐targeting antibody‐drug conjugates in pancreatic cancer. Mol Cancer Ther. 2021;20(12):2495‐2505. [DOI] [PubMed] [Google Scholar]
- 404. Salomon PL, Singh R. Sensitive ELISA method for the measurement of catabolites of antibody‐drug conjugates (ADCs) in target cancer cells. Mol Pharm. 2015;12(6):1752‐1761. [DOI] [PubMed] [Google Scholar]
- 405. Glatt DM, Beckford Vera DR, Prabhu SS, et al. Synthesis and characterization of cetuximab‐docetaxel and panitumumab‐docetaxel antibody‐drug conjugates for EGFR‐overexpressing cancer therapy. Mol Pharm. 2018;15(11):5089‐5102. [DOI] [PubMed] [Google Scholar]
- 406. Theunissen J‐W, Cai AG, Bhatti MM, et al. Treating tissue factor‐positive cancers with antibody‐drug conjugates that do not affect blood clotting. Mol Cancer Ther. 2018;17(11):2412‐2426. [DOI] [PubMed] [Google Scholar]
- 407. Chudasama V, Maruani A, Caddick S. Recent advances in the construction of antibody‐drug conjugates. Nat Chem. 2016;8(2):114‐119. [DOI] [PubMed] [Google Scholar]
- 408. O'Malley DM, Matulonis UA, Birrer MJ, et al. Phase Ib study of mirvetuximab soravtansine, a folate receptor alpha (FRalpha)‐targeting antibody‐drug conjugate (ADC), in combination with bevacizumab in patients with platinum‐resistant ovarian cancer. Gynecol Oncol. 2020;157(2):379‐385. [DOI] [PubMed] [Google Scholar]
- 409. Rathore R, McCallum JE, Varghese E, Florea A‐M, Busselberg D. Overcoming chemotherapy drug resistance by targeting inhibitors of apoptosis proteins (IAPs). Apoptosis. 2017;22(7):898‐919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 410. Hunter AM, LaCasse EC, Korneluk RG. The inhibitors of apoptosis (IAPs) as cancer targets. Apoptosis. 2007;12(9):1543‐1568. [DOI] [PubMed] [Google Scholar]
- 411. Brunckhorst MK, Lerner D, Wang S, Yu Q. AT‐406, an orally active antagonist of multiple inhibitor of apoptosis proteins, inhibits progression of human ovarian cancer. Cancer Biol Ther. 2012;13(9):804‐811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 412. van Roosmalen IAM, Quax WJ, Kruyt FAE. Two death‐inducing human TRAIL receptors to target in cancer: similar or distinct regulation and function? Biochem Pharmacol. 2014;91(4):447‐456. [DOI] [PubMed] [Google Scholar]
- 413. Obeid MA, Aljabali AAA, Rezigue M, et al. Use of nanoparticles in delivery of nucleic acids for melanoma treatment. Methods Mol Biol. 2021;2265:591‐620. [DOI] [PubMed] [Google Scholar]
- 414. Majidinia M, Mirza‐Aghazadeh‐Attari M, Rahimi M, et al. Overcoming multidrug resistance in cancer: recent progress in nanotechnology and new horizons. IUBMB Life. 2020;72(5):855‐871. [DOI] [PubMed] [Google Scholar]
- 415. Ye DM, Ye SC, Yu SQ, et al. Drug‐resistance reversal in colorectal cancer cells by destruction of flotillins, the key lipid rafts proteins. Neoplasma. 2019;66(4):576‐583. [DOI] [PubMed] [Google Scholar]
- 416. Wu S, Wen F, Li Y, et al. PIK3CA and PIK3CB silencing by RNAi reverse MDR and inhibit tumorigenic properties in human colorectal carcinoma. Tumour Biol. 2016;37(7):8799‐8809. [DOI] [PubMed] [Google Scholar]
- 417. Koshkin PA, Chistiakov DA, Nikitin AG, et al. Analysis of expression of microRNAs and genes involved in the control of key signaling mechanisms that support or inhibit development of brain tumors of different grades. Clin Chim Acta. 2014;430:55‐62. [DOI] [PubMed] [Google Scholar]
- 418. Piedra‐Carrasco N, Miguel L, Fabrega A, et al. Effectiveness of a double‐carbapenem regimen in a KPC‐producing Klebsiella pneumoniae infection in an immunocompromised patient. Microb Drug Resist. 2018;24(2):199‐202. [DOI] [PubMed] [Google Scholar]
- 419. Neufeld L, Yeini E, Reisman N, et al. Microengineered perfusable 3D‐bioprinted glioblastoma model for in vivo mimicry of tumor microenvironment. Sci Adv. 2021;7(34):eabi9119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420. Aghamiri S, Mehrjardi KF, Shabani S, Keshavarz‐Fathi M, Kargar S, Rezaei N. Nanoparticle‐siRNA: a potential strategy for ovarian cancer therapy? Nanomedicine (Lond). 2019;14(15):2083‐2100. [DOI] [PubMed] [Google Scholar]
- 421. Jia L, Li Z, Shen J, et al. Multifunctional mesoporous silica nanoparticles mediated co‐delivery of paclitaxel and tetrandrine for overcoming multidrug resistance. Int J Pharm. 2015;489(1‐2):318‐330. [DOI] [PubMed] [Google Scholar]
- 422. Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev. 2013;65(13‐14):1866‐1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 423. Saraswathy M, Gong S. Different strategies to overcome multidrug resistance in cancer. Biotechnol Adv. 2013;31(8):1397‐1407. [DOI] [PubMed] [Google Scholar]
- 424. Hu C‐MJ, Zhang L. Nanoparticle‐based combination therapy toward overcoming drug resistance in cancer. Biochem Pharmacol. 2012;83(8):1104‐1111. [DOI] [PubMed] [Google Scholar]
- 425. Tang Y, Soroush F, Tong Z, Kiani MF, Wang B. Targeted multidrug delivery system to overcome chemoresistance in breast cancer. Int J Nanomedicine. 2017;12:671‐681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 426. Liu Q, Li J, Pu G, Zhang F, Liu H, Zhang Y. Co‐delivery of baicalein and doxorubicin by hyaluronic acid decorated nanostructured lipid carriers for breast cancer therapy. Drug Deliv. 2016;23(4):1364‐1368. [DOI] [PubMed] [Google Scholar]
- 427. Zou W, Sarisozen C, Torchilin VP. The reversal of multidrug resistance in ovarian carcinoma cells by co‐application of tariquidar and paclitaxel in transferrin‐targeted polymeric micelles. J Drug Target. 2017;25(3):225‐234. [DOI] [PubMed] [Google Scholar]
- 428. Chen Y, Zhang W, Huang Y, Gao F, Sha X, Fang X. Pluronic‐based functional polymeric mixed micelles for co‐delivery of doxorubicin and paclitaxel to multidrug resistant tumor. Int J Pharm. 2015;488(1‐2):44‐58. [DOI] [PubMed] [Google Scholar]
- 429. Akbar MU, Zia KM, Nazir A, Iqbal J, Ejaz SA, Akash MSH. Pluronic‐based mixed polymeric micelles enhance the therapeutic potential of curcumin. AAPS PharmSciTech. 2018;19(6):2719‐2739. [DOI] [PubMed] [Google Scholar]
- 430. Sivak L, Subr V, Tomala J, et al. Overcoming multidrug resistance via simultaneous delivery of cytostatic drug and P‐glycoprotein inhibitor to cancer cells by HPMA copolymer conjugate. Biomaterials. 2017;115:65‐80. [DOI] [PubMed] [Google Scholar]
- 431. Meng L, Xia X, Yang Y, et al. Co‐encapsulation of paclitaxel and baicalein in nanoemulsions to overcome multidrug resistance via oxidative stress augmentation and P‐glycoprotein inhibition. Int J Pharm. 2016;513(1‐2):8‐16. [DOI] [PubMed] [Google Scholar]
- 432. Xiao B, Ma L, Merlin D. Nanoparticle‐mediated co‐delivery of chemotherapeutic agent and siRNA for combination cancer therapy. Expert Opin Drug Deliv. 2017;14(1):65‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 433. Zhang C‐G, Zhu W‐J, Liu Y, et al. Novel polymer micelle mediated co‐delivery of doxorubicin and P‐glycoprotein siRNA for reversal of multidrug resistance and synergistic tumor therapy. Sci Rep. 2016;6:23859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 434. Reddy TL, Garikapati KR, Reddy SG, et al. Simultaneous delivery of paclitaxel and Bcl‐2 siRNA via pH‐sensitive liposomal nanocarrier for the synergistic treatment of melanoma. Sci Rep. 2016;6:35223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 435. Wang H, Li F, Du C, Wang H, Mahato RI, Huang Y. Doxorubicin and lapatinib combination nanomedicine for treating resistant breast cancer. Mol Pharm. 2014;11(8):2600‐2611. [DOI] [PubMed] [Google Scholar]
- 436. Liu X, Wu Z, Guo C, et al. Hypoxia responsive nano‐drug delivery system based on angelica polysaccharide for liver cancer therapy. Drug Deliv. 2022;29(1):138‐148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 437. Deb B, George IA, Sharma J, Kumar P. Phosphoproteomics profiling to identify altered signaling pathways and kinase‐targeted cancer therapies. Methods Mol Biol. 2020;2051:241‐264. [DOI] [PubMed] [Google Scholar]
- 438. Lim YP. Mining the tumor phosphoproteome for cancer markers. Clin Cancer Res. 2005;11(9):3163‐3169. [DOI] [PubMed] [Google Scholar]
- 439. Miryala SK, Anbarasu A, Ramaiah S. Systems biology studies in Pseudomonas aeruginosa PA01 to understand their role in biofilm formation and multidrug efflux pumps. Microb Pathog. 2019;136:103668. [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
Not applicable.