Next Article in Journal
Advances on the Study of Diurnal Flower-Opening Times of Rice
Next Article in Special Issue
Signaling Pathways Leading to mTOR Activation Downstream Cytokine Receptors in Lymphocytes in Health and Disease
Previous Article in Journal
CC Chemokine 2 Promotes Ovarian Cancer Progression through the MEK/ERK/MAP3K19 Signaling Pathway
Previous Article in Special Issue
mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics

CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10651; https://doi.org/10.3390/ijms241310651
Submission received: 26 May 2023 / Revised: 21 June 2023 / Accepted: 23 June 2023 / Published: 26 June 2023
(This article belongs to the Special Issue Regulation of mTOR Signaling in Human Diseases)

Abstract

:
Cisplatin (cis-diamminedichloroplatinum I) is a platinum-based drug, the mainstay of anticancer treatment for numerous solid tumors. Since its approval by the FDA in 1978, the drug has continued to be used for the treatment of half of epithelial cancers. However, resistance to cisplatin represents a major obstacle during anticancer therapy. Here, we review recent findings on how the mTORC1 pathway and autophagy can influence cisplatin sensitivity and resistance and how these data can be applicable for the development of new therapeutic strategies.

1. Biochemical Mechanisms of Cisplatin Cytotoxicity

Cisplatin is a first line drug for many cancers, especially for lung, ovarian, head and neck, testicular and colorectal cancers (Figure 1). During anticancer therapy, cisplatin is injected intravenously. A high concentration of chloride ions (~100 mM) in the bloodstream suppresses the hydrolysis of the drug and maintains it in a neutral state. The binding of cisplatin to the plasma proteins, mainly to albumin, results in the deactivation of up to 95% of the injected drug.
Cisplatin enters the cells by passive diffusion through plasma membrane or by active transport via copper influx transporter 1 (CTR1) [1] and volume-regulated anion channel (VRAC) [2] (Figure 2). The majority of cytoplasmic cisplatin accumulates in vesicles that contain copper efflux transporters ATP7A or ATP7B rather than being diffusely localized throughout the cytoplasm. These intracellular secretory vesicles can further traffic and fuse with the plasma membrane, releasing cisplatin by exocytosis as a free drug, as a conjugate or as a complex with cellular proteins [3]. Copper transporters can be used by malignant cells to detoxify cisplatin, thereby promoting tumor resistance to chemotherapy [4].
Once in the cytoplasm, cisplatin undergoes aquation due to much lower chloride concentrations (~4–20 mM) than in the bloodstream. The displacement of one or two chloride ions by water molecules results in the generation of a highly electrophilic molecule that can interact with nucleic acids, phospholipids and proteins.
DNA is a primary target of cisplatin. Aquated cisplatin induces DNA damage by the forming of intrastrand and interstrand DNA cross-links through the preferential binding to the N7 position of guanine [5]. DNA lesions that are not resolved by DNA repair pathways block the production of DNA, mRNA and proteins and activate several transduction pathways, which finally lead to necrosis or apoptosis. Many cancers have defective DNA repair pathways; therefore, while normal cells can cope with the harm caused by cisplatin, cancer cells will die.
It is estimated that only ~1% of cisplatin interacts with nuclear DNA [6], the rest interacts with the mitochondrial DNA (mtDNA), lipids and sulfur donors (thiols as well as cysteines, methionines of cytoplasmic, mitochondrial and membrane proteins) [7,8,9]. The binding of aquated cisplatin to cytoplasmic molecules, including reduced glutathione (GSH) and metallothioneins (MTs), results in the accumulation of reactive oxygen species (ROS) that also triggers mitochondrial outer membrane permeabilization (MOMP) and DNA damage [8]. Cisplatin forms 4–6 times more DNA adducts with mtDNA than with gDNA. One of the reasons for the preference for mtDNA is due to a higher initial binding, because of the absence of histones in mitochondria. Another important reason is that nucleotide excision repair (NER), a major pathway for removing cisplatin damage in genomic DNA, does not function in mitochondria [10], which leads to the accumulation of cisplatin–DNA adducts. Increased mitochondrial biogenesis and dynamic alteration are essential factors in developing drug resistance [8,11].
Cisplatin does not show its highest potential in anticancer treatment because of side effects and drug resistance. The major adverse events that arise from cisplatin therapy are nephro-, hepato-, neuro- and gastrointestinal toxicities [12,13]. To overcome these limitations, great efforts have been made to search for cisplatin analogues which are better tolerated by patients and/or show anticancer activity in cisplatin-resistant tumors. The most widely used cisplatin derivatives are carboplatin and oxaliplatin (Figure 3). Carboplatin shows less toxicity than cisplatin at equimolar concentrations. It is used as a first-line treatment for patients with advanced ovarian cancer and advanced small-cell and non-small-cell lung cancer, while it shows a lower activity than cisplatin in germ cell tumors, bladder and head and neck cancers [14]. While the adducts formed by cisplatin and carboplatin are identical, the rate of formation of adducts is 10 times slower and 20- to 40- times higher concentrations of carboplatin are needed to obtain the same number of adducts [15]. Oxaliplatin forms different types of adducts than cisplatin and carboplatin, resulting in an altered DNA damage response. Oxaliplatin is used in combination with 5-fluorouracil for the treatment of metastatic colorectal cancer, which is insensitive to treatment with cisplatin and carboplatin [16].
Drug resistance is a serious problem in the treatment with platinum-based drugs. Cisplatin resistance could be both intrinsic (occurs from the beginning of treatment) and acquired (initially sensitive cells develop resistance to the drug over time). One of the major determinants of resistance is the type of cancer [17]. For example, small-cell lung cancer (SCLC; 10–15% of all lung cancers) is very sensitive to cisplatin, while non-small-cell lung cancer (NSCLC; about 80% to 85% of lung cancers) is quite resistant. Many cancers will eventually develop resistance against the drug with one notable exception, testicular germ cell cancer, which demonstrates a durable complete remission of ~80% of the patients treated with cisplatin.
Resistance to cisplatin depends on both the inner adaptive mechanisms of cancer cells and the tumor microenvironment, where hypoxic conditions increase the tolerance of cancer cells to the drug [18,19] (Figure 2). Among intercellular adaptive factors, the most important are: (1) a reduced drug accumulation due to either a decreased influx or an increased efflux; (2) an increase in DNA repair and changes in DNA damage response (DDR); (3) an alteration of apoptosis; (4) changes in signaling pathways, notably the mTORC1 pathway. These factors were conveniently classified by Galluzi et al. as pretarget, on-target, post-target and off-target, respectively [20]. There are numerous reviews addressing pre-, on- and post-target mechanisms [20,21,22,23,24], although relatively few are devoted to the changes in signaling pathways [25,26,27]. Here, we describe recent findings that connect platinum-based resistance mechanisms with major metabolic signaling networks—the mTORC1 pathway and autophagy. Metabolic remodeling is one of the main characteristics of tumors. Changes in glucose, amino acid and lipid metabolism, all these processes being under the control of the mTORC1 pathway, affect cisplatin resistance in tumor cells [28].

2. Molecular Basis of the mTORC1 Pathway and Autophagy

Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that belongs to the phosphatidylinositol 3-kinase PI3K-related family (PIKK). As a part of two structurally and functionally different complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), mTOR maintains the balance between anabolism and catabolism in response to nutritional or environmental conditions via the phosphorylation of its multiple substrates [29]. Among nearly 60 direct targets of mTORC1, the most known and well-characterized are p70S6 Kinase 1 (S6K1) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), involved in protein translation; transcription factor EB (TFEB), important for lysosome biogenesis and lipid metabolism; and Unc-51-like autophagy activating kinase 1 (ULK1), a member of the autophagy initiation complex.
The mTORC1 pathway is subjected to a tight regulation, allowing its activation, when growth factors, energy, and nutrients are sufficient [30,31] (Figure 4). In order to have an adequate and timely response to extra and intracellular inputs, mTORC1 responds to upstream signals through two different sets of the small GTPases–RHEB (Ras homologue enriched in brain) and RAGs (RAG guanosine triphosphatases). The activity of these GTPases depends on their effectors, GTPase-activating proteins (GAPs), which stimulate GTP hydrolysis, and guanine nucleotide exchange factors (GEFs).
The major site of mTORC1 activation is the lysosomal surface, where mTORC1 is recruited and induced in a RAG-GTPase-dependent manner when amino acids are abundant [32,33]. Four RAGs exist as heterodimers (i.e., RAGA (or RAGB) with RAGC (or RAGD)). In the presence of amino acids, RAGA/B is loaded with GTP while RAGC/D is bound to GDP. RAGs interact with the pentameric RAGULATOR complex, which exerts a GEF activity towards RAGA or RAGB [34,35]. Active RAGULATOR-RAG stimulates the recruitment of mTORC1 to the lysosomal membrane where it is fully activated by RHEB, loaded with GTP [36]. RHEB is under the control of another signaling node, the tuberous sclerosis (TSC) complex, which acts as a GAP to inhibit RHEB. TSC is a nexus of multiple physiological stimuli (e.g., energy status, growth factors, DNA damage) that signal to mTORC1 through the PI3K-AKT network [37]. mTORC1 is a well-recognized downstream effector of the PI3K/AKT signaling pathway, which responds to three main inputs: growth factors, hormones, chemokines and cytokines. RAG GTPases regulate the recruitment of TSC to the lysosome and its ability to interact with and inhibit RHEB in response to amino acid starvation, growth factors removal and to other stresses that inhibit mTORC1 [38,39,40]. Both RAGs and RHEB are necessary for mTORC1 activation at the lysosome [40]. When amino acids are scarce, GEFs and GAPs (e.g., GAP activity toward RAGs complex 1 (GATOR1) [41]) transform RAGs in their inactive form, which further leads to mTORC1 suppression.
mTORC1 plays a central role as a negative regulator of autophagy, a catabolic process by which cytosol and organelles are sequestered within double-membrane-bound vesicles that deliver their contents to the lysosome for degradation and recycling [42]. mTORC1 is active and autophagy is suppressed under optimal growth conditions, e.g., a sufficient quantity of amino acids and glucose. In order to inhibit autophagy initiation, active mTORC1 associates with a ULK1 protein complex and phosphorylates its members, ULK1 and autophagy-related protein 13 (ATG13) [43]. This phosphorylation prevents the kinase activity of the ULK1 complex. mTORC1 also regulates autophagy at the transcriptional level by modulating the localization of TFEB, which regulates the expression of autophagy and lysosomal genes. mTORC1 phosphorylates TFEB, thereby inhibiting TFEB activity when nutrients are present [44].
Finally, mTORC1 functioning is also important during DNA damage. The interaction between the DNA damage response and the mTORC1 pathway is especially relevant in the context of cisplatin treatment and can be considered as an outstanding example of how genotoxic stress and metabolic pathways can be triggered to overcome the disease [45]. Ideally, during anticancer therapy, the DDR and mTORC1 pathways should be repressed in cancer cells to provide a prodeath scenario, while in the healthy cells, these pathways should be active to repair damage and maintain proliferation to enable survival. Accordingly, one of the main causes of cancer therapy resistance is sustained DNA repair associated with hyperactivated mTORC1 signaling.

3. mTORC1 Pathway and Cisplatin Resistance

In normal physiological conditions, the PI3K/AKT/mTOR pathway undergoes stringent regulation to ensure the proper activity and balance necessary for healthy homeostasis. In the context of numerous cancers, this pathway exhibits a persistent state of activation. Thus, the aberrant activation of the pathway has been detected in ~70% of ovarian and breast cancers [46,47] and in ~90% of head and neck cancers and lung adenocarcinoma [26,48]. The mechanisms behind this activation include the amplification of the mutations of genes encoding PI3K subunits, AKT, inactivating mutations in TSC genes, or, conversely, activating mutations in MTOR [49]. Interestingly, the mutation frequency of the MTOR gene itself depends on the cancer type and is generally quite low (around 2–5%), especially in comparison with p53, which is mutated in more than 25% of cancers [50,51,52]. Cancer cells with a subset of mTOR-activating mutations are hypersensitive to the mTOR inhibitor rapamycin [51].
Tumors with acquired resistance to cisplatin often have a constitutive activation of mTORC1 signaling [53]. mTORC1 can influence cisplatin resistance in many ways—at the transcriptional and translational levels and by responding to various cues, such as amino acids, energy, and DNA damage (Figure 5). One of the most important functions of mTORC1 in cisplatin resistance takes place at the level of translational regulation. mTORC1 phosphorylation results in the activation of the p70S6K kinase activity, which, in turn, has many substrates, including ribosomal protein S6 [54]. In its unphosphorylated state, 4E-BP1 binds and inhibits eIF4E to prevent translation initiation. The phosphorylation of 4E-BP1 by mTORC1 releases eIF4E, which can now bind to eIF4G and initiate translation [55,56]. The phosphorylation of p70S6K and 4E-BP1 was elevated in cells that acquired resistance to cisplatin compared to parental sensitive cells in small-cell lung cancer [57] and ovarian cancer cells [58]. The S6K1 inhibitor RAME, which blocks the interaction between S6K1 and mTOR, promoted autophagy and apoptosis and greatly enhanced the antitumor effect of cisplatin in cisplatin-resistant cervical cancer cells [59].
Programmed cell death 4 (PDCD4) protein, which is not a direct target of mTORC1, can suppress protein translation by interacting with EIF4A and EIF4G to inhibit the formation of the translation initiation complex. An overexpression of PDCD4 enhances platinum sensitivity, while a knockdown of PDCD4 reduces platinum sensitivity in ovarian cells and in a xenograft model [60].
EIF3A, the largest subunit of the eIF3 translational initiation complex, downregulates the translation of a number of nucleotide excision repair proteins. EIF3A knockdown or ectopic overexpression, respectively, increases or decreases cellular resistance to cisplatin in a number of cancer cell lines, including nasopharyngeal and ovarian carcinoma and lung cancer cell lines, likely due to EIF3A’s role in the regulation of NER proteins [61,62]. Another recently proposed mechanism is that EIF3A regulates S6K1 activity by inhibiting mTORC1 kinase via regulating the synthesis of its subunit RAPTOR. Thus, not only does mTORC1 regulate translational control, but it can also be regulated by translational control in a feedback loop, which involves S6K1, contributing to cellular response to cisplatin [63].
Translationally controlled tumor protein (TCTP) stimulates mTORC1 by positively regulating RHEB activity [64]. TCTP is overexpressed in many human tumor tissues [65]. The inhibition of mTORC1 by rapamycin in human lung cancer cells and an A549 lung cancer xenograft model induces ubiquitin–proteasome degradation of TCTP. Moreover, the minimal dose of rapamycin required to induce TCTP proteolysis enhances the efficacy of cisplatin through the induction of apoptotic cell death in vitro and in vivo. This synergistic cytotoxicity was induced irrespective of the functional status of p53 [66].
Chemotherapy can induce oxidative and genotoxic stress, triggering a senescence-like state, which in many cancer cells causes treatment resistance, supporting tumor proliferation and cancer recurrence. mTORC1 activity is upregulated in senescent cells, which are insensitive to serum and amino acid starvation. How this can be related to the senescence-induced drug resistance was addressed in a recent study by Jiang et al., who investigated the role of five small GTPases that can activate mTORC1 in response to amino acid stimulation [67]. In the senescence-like hepatoma cell line HepG2, RAGC and RHEB, but not RAB1A, RAB5 or ARF1, were required for persistent mTORC1 activity. One of the reasons could be that senescence-like cells rely on amino acid supply resulting from the lysosomal degradation of cellular components via autophagy. Out of the five studied small GTPases, only RAGC and RHEB localized to the lysosome, while RAB1A and ARF1 preferentially localized to ER/Golgi, and RAB5 to endosomes. The knocking down of RAGC or RHEB, but not the other three GTPases, increased cisplatin sensitivity in senescence-like HEPG2, which were significantly more resistant than their proliferating counterparts. Accordingly, the expression of RAGC and RHEB, but not the other GTPases, is associated with a poor prognosis in liver cancer patients. It will be important to find out if this effect can be observed in different tumors and if other RAGs can also have a role in the cisplatin resistance of senescence-like cancer cells. That would be of a particular interest, given the recent finding that RAGs in heterodimers are not functionally redundant. This is especially relevant to the function of transcription factors TFE3 and TFEB, which regulate the transcription of many lysosomal genes. mTORC1 phosphorylates TFE3 and TFEB at the lysosomal membrane, leading to their inactivation and sequestration in the cytoplasm [68]. RAGD preferentially promotes mTORC1 phosphorylation of TFEB, which is linked to a stronger association of RAGD with the lysosome, compared to RAGC, while TEF3 is preferentially recruited by RAGC [69]. Recent findings demonstrate that TFEB regulates the expression of ATP7B, a copper transporter involved in cisplatin efflux. The suppression of TFEB inhibits ATP7B expression and sensitizes initially resistant ovarian cancer cells to cisplatin [70]. In addition, in ovarian cancer cells, TFEB is involved in cisplatin resistance by regulating the tumor microenvironment. Cisplatin treatment induced TFEB nuclear translocation, increasing the expression of programmed cell death-ligands 1 and 2 (PD-L1 and PD-L2) to foster an immunosuppressive tumor microenvironment that mediates immune evasion and drug resistance [71].
An increased expression of branched-chain amino acid transaminase 1 (BCAT1) is correlated with a poor prognosis in many solid tumors and is also associated with decreased cisplatin sensitivity. BCAT1 is responsible for facilitating the synthesis of α-ketoglutarates from branched-chain amino acids (i.e., leucine), which are important activators of the mTORC1 pathway. A recent study has demonstrated that BCAT1 decreases cisplatin sensitivity by activating autophagy via the mTORC1 pathway [72]. Following treatment with cisplatin, BCAT1 expression is initially upregulated, resulting in a decrease in branched-chain amino acid levels due to the enzymatic activity of BCAT1. The deficiency of amino acids, especially leucine, suppresses mTORC1 activity and subsequently enhances autophagy, leading to a decrease in cisplatin sensitivity. Treatment with leucine or chloroquine, as well as the knockdown of BCAT1, inhibits autophagy and increases cisplatin sensitivity in cervical cancer and hepatocellular carcinoma cell lines and in mouse models. Thus, BCAT1 can be considered as a potential pharmacological target to overcome cisplatin resistance.
One of the major upstream regulators of the mTORC1 pathway, the GATOR1 complex, is involved in the regulation of nutrient sensing and responding. Various mutations of the genes encoding GATOR1 proteins have been detected in many solid tumors [73]. Notably, a low expression of NPRL2, one of the GATOR1 components, in different types of lung cancers is correlated with cisplatin resistance [74,75]. The overexpression of NPRL2 in NPRL2-deficient and cisplatin-resistant NSCLC cells reactivates cellular response to cisplatin and promotes tumor suppression activity in vitro and in mouse models [74]. The reason for this resistance is still not clear, but it could be related to the role of NPRL2 in DNA damage response [75,76].
Recently, a number of different mTOR inhibitors in combination with various platinum drugs have been applied to sensitize resistant cells. mTORC1 inhibitors can be generally divided into four groups: (1) rapamycin and its analogs (rapalogues), which preferentially target mTORC1, yet the 4E-BP1 branch is only partially sensitive to these inhibitors, activating a negative feedback loop of PI3K–AKT signaling; (2) pan-mTORC1 inhibitors (TORKIs), which target the catalytic core of mTOR kinase and therefore suppress both mTORC1 and mTORC2 (pp242, torin1, AZD8055); (3) bivalent inhibitors that target two domains of mTOR and are effective in inhibiting the 4E-BP1 phosphorylation (RapaLink1); and (4) dual inhibitors that can act on mTOR kinase and another protein, most often targeting PI3K/AKT signaling (NVP-BEZ235) [45].
The suppression of the mTOR activity by various inhibitors has been applied to enhance cisplatin-induced apoptosis in ovarian cancer [77], head and neck cancer [26], hepatocellular carcinoma [78], lung cancer [79] and nasopharyngeal carcinoma [80]. The treatment of platinum-resistant ovarian cancer cells with carboplatin and a dual mTORC1/2 inhibitor resulted in a strong inhibition of cancer cell proliferation, improved tumor control, reduced metastasis and significantly increased survival in mice [77]. Due to these promising results, several clinical trials (phase I/II) have been initiated recently in patients with triple-negative breast cancer to see the effects of a combined therapy involving platinum drugs and the mTORC1 inhibitor everolimus [81] or the dual PI3K/mTOR inhibitor gedatolisib [82]. The results of these clinical trials did not demonstrate a significantly improved response rate compared to cisplatin monotherapy. However, these results may have been compromised by the small sample size (14 patients in the first trial, ~100 patients in the second) and nonrandomized study design. Earlier clinical trials on different solid tumors (phase I only) had a very small patient cohort and did not provide conclusive results [83,84,85]. Further clinical trials encompassing a broader range of cancer types and larger patient cohorts are necessary to determine the efficacy of combining platinum drugs with PI3K/mTOR inhibitors and their potential to improve the outcomes of cancer patients.

4. Autophagy in Cisplatin Resistance

The activation of the mTORC1 pathway results in autophagy inhibition, while cisplatin treatment generally promotes autophagy (Figure 6). One of the first reports about the association between autophagy and cisplatin resistance dates back to 2010, when it was demonstrated that the acquired cisplatin resistance in lung adenocarcinoma cells A549 was associated with elevated autophagy [86]. The inhibition of autophagy is often observed in cisplatin-sensitive cells, whereas the basal level of autophagy is elevated in cisplatin-resistant cells. Accordingly, the suppression of autophagy, for example by chloroquine, increases drug toxicity and can improve sensitivity in cisplatin-resistant cancer cells [87]. A recently published resource database of genes associated with platinum resistance in cancer demonstrates that genes involved in the production of autophagosomes, including ATG5, ATG7, ATG12, ATG14 and BECN1, promote platinum resistance [88]. In the same line, an elevated expression of LC3A was shown to be associated with platinum resistance and a worse prognosis in ovarian clear cell carcinomas [89]. Thus, the inhibition of autophagy can be considered as a strategy for improving cisplatin sensitivity.
Autophagy can also have a cytoprotective function, which is particularly important in the context of adverse effects during cisplatin treatment. Notably, up to 30% of patients receiving cisplatin develop acute kidney injury (AKI), leading to a rapid loss of renal function or renal failure. Autophagy is activated in renal tubules to protect against neurotoxicity during the acute phase. On the other hand, the sustained activation of autophagy will limit kidney repair. Thus, it is important to manipulate autophagy differently at the beginning and after the cisplatin treatment, in order to protect kidneys and allow for their effective recovery [90]. A recent review on cisplatin and autophagy extensively considered many aspects of this catabolic process in drug treatment [91]. Therefore, here, we only outline the most important features of the role of autophagy in cisplatin resistance.
Following cisplatin treatment, autophagy induction is detected in both cisplatin-sensitive and resistant cancer cells. Therefore, drug resistance is not necessarily a consequence of autophagy induction. It is important, though, to evaluate what type of autophagy is induced upon drug treatment because autophagy can participate in both cell survival (cytoprotective autophagy) and cell death (cytotoxic autophagy) [92]. Moreover, Gewirtz proposed to consider two additional types of autophagy: nonprotective and cytostatic [93]. These four forms of autophagy can be identified by the functional outcomes of their inhibition by drugs, but so far, there are no reliable morphological or biochemical methods that would allow us to distinguish between autophagy types. Cisplatin generally induces autophagy, which has a prosurvival role for cancer cells and, therefore, can participate in drug resistance. The inhibition of this cytoprotective autophagy results in chemosensitization. In contrast, the inhibition of nonprotective autophagy does not lead to changes in drug sensitivity [94,95]. Moreover, the functions of autophagy can be changed in response to stimuli, stress or genetic alterations through a so-called “autophagic switch”.
One example of this switch relevant to cisplatin treatment was recently demonstrated by Patel et al. when comparing wild-type and functionally null p53 NSCLC H460 cells [94]. Cisplatin induced cytoprotective autophagy in p53-deficient cells, while p53 wild-type cells had nonprotective autophagy. p53 wild-type H460 cells, where autophagy was nonprotective, were more sensitive to cisplatin than the H460crp53 cells. However, both types of cells had the same degree of apoptosis. Thus, the type of autophagy, and not p53 function, largely determined cisplatin sensitivity. The inhibition of cytoprotective autophagy was sufficient to restore cisplatin sensitivity in p53-deficient cells [94]. Therefore, the combination of autophagy inhibitors and cisplatin seems to have a strong therapeutic potential for resistant tumors, where autophagy plays a cytoprotective role, but this may not be applicable to cisplatin-sensitive cancers. Nevertheless, it is currently unclear how these findings can be applied in the clinic, especially because there are no valid methods to identify the nature of induced autophagy.
Many reports describe the regulation of cisplatin-induced autophagy in tumor cells by various noncoding RNAs (ncRNAs), including long noncoding RNAs, microRNAs and circular RNAs [91,96]. In the majority of cases, these ncRNAs inhibit autophagy via different mechanisms and increase sensitivity to the drug in both sensitive and resistant cancer cells. For example, the inhibition of autophagy via miR-205 sensitized castration-resistant prostate cancer cells to cisplatin [97]. Another study demonstrated that the overexpression of miRNA-1, -181, -223, -425-3p and lncRNA BLACAT1 in cisplatin-resistant lung cancer cells upregulated cisplatin-induced autophagy [91]. LncRNA LUCAT1, highly expressed in cisplatin-resistant NSCLC tissues and cells, contributes to cisplatin resistance by negatively regulating miR-514a-3p, another noncoding RNA. miR-514a-3p, in turn, negatively modulates ULK1 expression. LUCAT1 silencing enhances cisplatin sensitivity by inducing cell apoptosis, suppressing autophagy and inhibiting cell metastasis in NSCLC. Moreover, LUCAT1 regulates the sensitivity of NSCLC cells to cisplatin by upregulating ULK1 via sponging miR-514a-3p [98].
The inhibition of autophagy via different micro-RNAs also sensitized gastric cancer cells to cisplatin [99,100]. Thus, miR-148a-3p modulates cisplatin sensitivity by simultaneously regulating RAB12-mediated autophagy and AKAP1-mediated mitochondrial fission [100]. RAB12 GTPase, a member of the RAS oncogene family, induces autophagy by inhibiting mTORC1 activity [101] and accelerating autolysosome maturation [102]. RAB12 also interacts with optineurin (OPTN), an important mitophagy receptor. The suppression of mTORC1 by RAB12 facilitates early autophagosome formation to protect gastric cancer cells from cisplatin-induced cell death. miR-148a-3p can significantly reduce autophagic flux and autophagosome formation by regulating RAB12. A-kinase anchoring protein 1 (AKAP1) is upregulated in cisplatin-resistant gastric cancer tissues and antagonizes cisplatin-induced mitochondrial fission by the phosphorylation of dynamin-related proteins 1 (DRP1), an important mitochondrial fission factor [100]. Because AKAP1 restrains mitochondrial fission and reinforces cisplatin resistance in gastric cancer cells, targeting this protein by miR-148a-3 sensitizes cells to cisplatin. The role of DRP1 in cisplatin resistance is somewhat controversial and can be the opposite in different cancers. For example, in cisplatin-resistant ovarian cancer cell lines SKOV3, DRP1 expression was downregulated, and the knockdown of DRP1 in parental sensitive cell lines provoked cisplatin resistance [11]. Similarly, the ROS-promoted downregulation of DRP1 phosphorylation triggered mitochondrial fission and cisplatin resistance in a number of ovarian cancer cells. The inhibition of DRP1 by the specific inhibitor Mdivi-1 increased the cisplatin sensitivity of ovarian cancer cells under hypoxia [103]. On the other hand, the inhibition of DRP1 by Mdivi-1 sensitized lung, breast, colon and renal carcinoma cell lines to cisplatin [104].
Despite promising in vivo and in vitro results in the application of noncoding RNAs to reverse drug resistance, these strategies are far from being applied in the clinic. The main difficulties concern the methods of expressing ncRNAs in a cancerous tissue or in organs, the dosage in combination with cisplatin and other anticancer drugs used in the patient undergoing therapy.
Finally, cisplatin resistance can also be associated with the changes of mitochondrial autophagy (mitophagy). As mentioned above, cisplatin activates DRP1, leading to mitochondrial fragmentation. This results in the segregation of dysfunctional organelles and their enhanced removal by mitophagy. The suppression of DRP1-mediated mitophagy by Mdivi-1 increases the sensitivity of hepatocellular carcinoma cells to cisplatin [105]. Similarly, in cisplatin-resistant ovarian carcinoma and osteosarcoma cell lines, mitochondria are more fragmented. These cells exhibit an increased expression of mitophagy receptor BNIP3. A silencing of BNIP3 resensitizes resistant cells to the drug [106]. A study in cisplatin-resistant lung cancer cells also revealed that under hypoxic conditions, BNIP3-dependent autophagy induction was enhanced, allowing cell survival, thus also pointing to the importance of the microenvironment in cisplatin resistance [107].
In contrast to mTORC1, which can now be targeted by the fourth generation of drugs, the only autophagy inhibitors on the market remain chloroquine and hydroxychloroquine. Recently, many efforts have been applied to search for autophagy-selective drugs, but none of them are currently found in clinical development [108]. However, given the progress in understanding the autophagy pathway at the molecular level that have been achieved during recent decades and a growing number of biotech companies investing in the developing of autophagic modulators, it is reasonable to expect the appearance of new drugs that can specifically inhibit (or induce) autophagy. Most probably, some of these drugs will be effective in combination with platinum drugs to sensitize cancers that have been resistant so far to chemotherapies.

5. Conclusions

Cisplatin resistance is a significant challenge in cancer treatment, leading to decreased efficacy and poorer patient outcomes. Recent studies on the involvement of the mTORC1 pathway and autophagy in this resistance mechanism offer potential avenues for therapeutic intervention. Many mTORC1 signaling components and effectors are frequently deregulated or altered in different cancers. Cisplatin-resistant tumors often have a constitutive activation of mTORC1 signaling. Targeting the mTORC1 pathway with inhibitors has shown promising results in restoring cisplatin sensitivity, sensitizing resistant cells to the drug. Autophagy has emerged as another important player in cisplatin resistance because it can have a prosurvival role by protecting cancer cells from cisplatin-induced stress. Combination therapies that inhibit mTORC1 while modulating autophagy may provide a more effective strategy to sensitize resistant cancer cells to cisplatin. More clinical trials covering different cancer types and larger patient cohorts are necessary to determine the efficacy of such therapies.

Author Contributions

Conceptualization, Z.P. and S.D.; writing—original draft preparation, Z.P., H.Z. and S.D.; writing—review and editing, S.D.; visualization, Z.P. and H.Z.; supervision, S.D.; funding acquisition, S.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by La Ligue contre le Cancer 94 (Comité du Val-de-Marne). Z.P. and H.Z. are recipients of Chinese Scholarship Council fellowships.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to Catherine Brenner for helpful discussions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Howell, S.B.; Safaei, R.; Larson, C.A.; Sailor, M.J. Copper Transporters and the Cellular Pharmacology of the Platinum-Containing Cancer Drugs. Mol. Pharmacol. 2010, 77, 887–894. [Google Scholar] [CrossRef] [Green Version]
  2. Planells-Cases, R.; Lutter, D.; Guyader, C.; Gerhards, N.M.; Ullrich, F.; Elger, D.A.; Kucukosmanoglu, A.; Xu, G.; Voss, F.K.; Reincke, S.M.; et al. Subunit Composition of VRAC Channels Determines Substrate Specificity and Cellular Resistance to P T-based Anti-cancer Drugs. EMBO J. 2015, 34, 2993–3008. [Google Scholar] [CrossRef]
  3. Lukanović, D.; Herzog, M.; Kobal, B.; Černe, K. The Contribution of Copper Efflux Transporters ATP7A and ATP7B to Chemoresistance and Personalized Medicine in Ovarian Cancer. Biomed. Pharmacother. 2020, 129, 110401. [Google Scholar] [CrossRef]
  4. Petruzzelli, R.; Polishchuk, R.S. Activity and Trafficking of Copper-Transporting ATPases in Tumor Development and Defense against Platinum-Based Drugs. Cells 2019, 8, 1080. [Google Scholar] [CrossRef] [Green Version]
  5. Jamieson, E.R.; Lippard, S.J. Structure, Recognition, and Processing of Cisplatin−DNA Adducts. Chem. Rev. 1999, 99, 2467–2498. [Google Scholar] [CrossRef]
  6. Roberts, J.J.; Friedlos, F. Quantitative Aspects of the Formation and Loss of DNA Interstrand Crosslinks in Chinese Hamster Cells Following Treatment with Cis-Diamminedichloroplatinum(II) (Cisplatin) I. Proportion of DNA-Platinum Reactions Involved in DNA Crosslinking. Biochim. Biophys. Acta-Nucleic Acids Protein Synth. 1981, 655, 146–151. [Google Scholar] [CrossRef]
  7. Martinho, N.; Santos, T.C.B.; Florindo, H.F.; Silva, L.C. Cisplatin-Membrane Interactions and Their Influence on Platinum Complexes Activity and Toxicity. Front. Physiol. 2019, 9, 1898. [Google Scholar] [CrossRef] [Green Version]
  8. Cocetta, V.; Ragazzi, E.; Montopoli, M. Mitochondrial Involvement in Cisplatin Resistance. Int. J. Mol. Sci. 2019, 20, 3384. [Google Scholar] [CrossRef] [Green Version]
  9. Wang, K.; Lu, J.; Li, R. The Events That Occur When Cisplatin Encounters Cells. Coord. Chem. Rev. 1996, 151, 53–88. [Google Scholar] [CrossRef]
  10. Rong, Z.; Tu, P.; Xu, P.; Sun, Y.; Yu, F.; Tu, N.; Guo, L.; Yang, Y. The Mitochondrial Response to DNA Damage. Front. Cell Dev. Biol. 2021, 9, 669379. [Google Scholar] [CrossRef]
  11. Zou, G.-P.; Yu, C.-X.; Shi, S.-L.; Li, Q.-G.; Wang, X.-H.; Qu, X.-H.; Yang, Z.-J.; Yao, W.-R.; Yan, D.-D.; Jiang, L.-P.; et al. Mitochondrial Dynamics Mediated by DRP1 and MFN2 Contributes to Cisplatin Chemoresistance in Human Ovarian Cancer SKOV3 Cells. J. Cancer 2021, 12, 7358–7373. [Google Scholar] [CrossRef]
  12. Oun, R.; Moussa, Y.E.; Wheate, N.J. The Side Effects of Platinum-Based Chemotherapy Drugs: A Review for Chemists. Dalton Trans. 2018, 47, 6645–6653. [Google Scholar] [CrossRef]
  13. Ghosh, S. Cisplatin: The First Metal Based Anticancer Drug. Bioorgan. Chem. 2019, 88, 102925. [Google Scholar] [CrossRef]
  14. Go, R.S.; Adjei, A.A. Review of the Comparative Pharmacology and Clinical Activity of Cisplatin and Carboplatin. J. Clin. Oncol. 1999, 17, 409. [Google Scholar] [CrossRef]
  15. Knox, R.J.; Friedlos, F.; Lydall, D.A.; Roberts, J.J. Mechanism of Cytotoxicity of Anticancer Platinum Drugs: Evidence That Cis-Diamminedichloroplatinum(II) and Cis-Diammine-(1,1-Cyclobutanedicarboxylato)Platinum(II) Differ Only in the Kinetics of Their Interaction with DNA. Cancer Res. 1986, 46, 1972–1979. [Google Scholar]
  16. Bonetti, A.; Giuliani, J.; Muggia, F. Targeted Agents and Oxaliplatin-Containing Regimens for the Treatment of Colon Cancer. Anticancer Res. 2014, 34, 423–434. [Google Scholar]
  17. Ranasinghe, R.; Mathai, M.L.; Zulli, A. Cisplatin for Cancer Therapy and Overcoming Chemoresistance. Heliyon 2022, 8, e10608. [Google Scholar] [CrossRef]
  18. Chen, Z.; Han, F.; Du, Y.; Shi, H.; Zhou, W. Hypoxic Microenvironment in Cancer: Molecular Mechanisms and Therapeutic Interventions. Signal Transduct. Target. Ther. 2023, 8, 70. [Google Scholar] [CrossRef]
  19. Chen, S.-H.; Chang, J.-Y. New Insights into Mechanisms of Cisplatin Resistance: From Tumor Cell to Microenvironment. Int. J. Mol. Sci. 2019, 20, 4136. [Google Scholar] [CrossRef] [Green Version]
  20. Galluzzi, L.; Vitale, I.; Michels, J.; Brenner, C.; Szabadkai, G.; Harel-Bellan, A.; Castedo, M.; Kroemer, G. Systems Biology of Cisplatin Resistance: Past, Present and Future. Cell Death Dis. 2014, 5, e1257. [Google Scholar] [CrossRef] [Green Version]
  21. Siddik, Z.H. Cisplatin: Mode of Cytotoxic Action and Molecular Basis of Resistance. Oncogene 2003, 22, 7265–7279. [Google Scholar] [CrossRef] [Green Version]
  22. Romani, A.M.P. Cisplatin in Cancer Treatment. Biochem. Pharmacol. 2022, 206, 115323. [Google Scholar] [CrossRef]
  23. Kryczka, J.; Kryczka, J.; Czarnecka-Chrebelska, K.H.; Brzeziańska-Lasota, E. Molecular Mechanisms of Chemoresistance Induced by Cisplatin in NSCLC Cancer Therapy. Int. J. Mol. Sci. 2021, 22, 8885. [Google Scholar] [CrossRef]
  24. Rottenberg, S.; Disler, C.; Perego, P. The Rediscovery of Platinum-Based Cancer Therapy. Nat. Rev. Cancer 2021, 21, 37–50. [Google Scholar] [CrossRef]
  25. Wang, Y.; Liu, Z.; Shu, S.; Cai, J.; Tang, C.; Dong, Z. AMPK/MTOR Signaling in Autophagy Regulation During Cisplatin-Induced Acute Kidney Injury. Front. Physiol. 2020, 11, 619730. [Google Scholar] [CrossRef]
  26. Marquard, F.E.; Jücker, M. PI3K/AKT/MTOR Signaling as a Molecular Target in Head and Neck Cancer. Biochem. Pharmacol. 2020, 172, 113729. [Google Scholar] [CrossRef]
  27. Wangpaichitr, M.; Theodoropoulos, G.; Nguyen, D.J.M.; Wu, C.; Spector, S.A.; Feun, L.G.; Savaraj, N. Cisplatin Resistance and Redox-Metabolic Vulnerability: A Second Alteration. Int. J. Mol. Sci. 2021, 22, 7379. [Google Scholar] [CrossRef]
  28. Wang, L.; Zhao, X.; Fu, J.; Xu, W.; Yuan, J. The Role of Tumour Metabolism in Cisplatin Resistance. Front. Mol. Biosci. 2021, 8, 691795. [Google Scholar] [CrossRef]
  29. Battaglioni, S.; Benjamin, D.; Wälchli, M.; Maier, T.; Hall, M.N. MTOR Substrate Phosphorylation in Growth Control. Cell 2022, 185, 1814–1836. [Google Scholar] [CrossRef]
  30. Ben-Sahra, I.; Manning, B.D. MTORC1 Signaling and the Metabolic Control of Cell Growth. Curr. Opin. Cell Biol. 2017, 45, 72–82. [Google Scholar] [CrossRef] [Green Version]
  31. Liu, G.Y.; Sabatini, D.M. MTOR at the Nexus of Nutrition, Growth, Ageing and Disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 183–203. [Google Scholar] [CrossRef]
  32. Sancak, Y.; Peterson, T.R.; Shaul, Y.D.; Lindquist, R.A.; Thoreen, C.C.; Bar-Peled, L.; Sabatini, D.M. The Rag GTPases Bind Raptor and Mediate Amino Acid Signaling to MTORC1. Science 2008, 320, 1496–1501. [Google Scholar] [CrossRef] [Green Version]
  33. Kim, E.; Goraksha-Hicks, P.; Li, L.; Neufeld, T.P.; Guan, K.-L. Regulation of TORC1 by Rag GTPases in Nutrient Response. Nat. Cell Biol. 2008, 10, 935–945. [Google Scholar] [CrossRef] [Green Version]
  34. Bar-Peled, L.; Schweitzer, L.D.; Zoncu, R.; Sabatini, D.M. Ragulator Is a GEF for the Rag GTPases That Signal Amino Acid Levels to MTORC1. Cell 2012, 150, 1196–1208. [Google Scholar] [CrossRef] [Green Version]
  35. Zoncu, R.; Bar-Peled, L.; Efeyan, A.; Wang, S.; Sancak, Y.; Sabatini, D.M. MTORC1 Senses Lysosomal Amino Acids through an Inside-out Mechanism That Requires the Vacuolar H+-ATPase. Science 2011, 334, 678–683. [Google Scholar] [CrossRef] [Green Version]
  36. Long, X.; Ortiz-Vega, S.; Lin, Y.; Avruch, J. Rheb Binding to Mammalian Target of Rapamycin (MTOR) Is Regulated by Amino Acid Sufficiency. J. Biol. Chem. 2005, 280, 23433–23436. [Google Scholar] [CrossRef] [Green Version]
  37. Hoxhaj, G.; Manning, B.D. The PI3K-AKT Network at the Interface of Oncogenic Signalling and Cancer Metabolism. Nat. Rev. Cancer 2020, 20, 74–88. [Google Scholar] [CrossRef]
  38. Demetriades, C.; Plescher, M.; Teleman, A.A. Lysosomal Recruitment of TSC2 Is a Universal Response to Cellular Stress. Nat. Commun. 2016, 7, 10662. [Google Scholar] [CrossRef] [Green Version]
  39. Yang, S.; Zhang, Y.; Ting, C.-Y.; Bettedi, L.; Kim, K.; Ghaniam, E.; Lilly, M.A. The Rag GTPase Regulates the Dynamic Behavior of TSC Downstream of Both Amino Acid and Growth Factor Restriction. Dev. Cell 2020, 55, 272–288.e5. [Google Scholar] [CrossRef]
  40. Demetriades, C.; Doumpas, N.; Teleman, A.A. Regulation of TORC1 in Response to Amino Acid Starvation via Lysosomal Recruitment of TSC2. Cell 2014, 156, 786–799. [Google Scholar] [CrossRef] [Green Version]
  41. Bar-Peled, L.; Chantranupong, L.; Cherniack, A.D.; Chen, W.W.; Ottina, K.A.; Grabiner, B.C.; Spear, E.D.; Carter, S.L.; Meyerson, M.; Sabatini, D.M. A Tumor Suppressor Complex with GAP Activity for the Rag GTPases That Signal Amino Acid Sufficiency to MTORC1. Science 2013, 340, 1100–1106. [Google Scholar] [CrossRef] [Green Version]
  42. Hu, Y.; Reggiori, F. Molecular Regulation of Autophagosome Formation. Biochem. Soc. Trans. 2022, 50, 55–69. [Google Scholar] [CrossRef]
  43. Licheva, M.; Raman, B.; Kraft, C.; Reggiori, F. Phosphoregulation of the Autophagy Machinery by Kinases and Phosphatases. Autophagy 2022, 18, 104–123. [Google Scholar] [CrossRef]
  44. Settembre, C.; Zoncu, R.; Medina, D.L.; Vetrini, F.; Erdin, S.; Erdin, S.; Huynh, T.; Ferron, M.; Karsenty, G.; Vellard, M.C.; et al. A Lysosome-to-Nucleus Signalling Mechanism Senses and Regulates the Lysosome via MTOR and TFEB: Self-Regulation of the Lysosome via MTOR and TFEB. EMBO J. 2012, 31, 1095–1108. [Google Scholar] [CrossRef] [Green Version]
  45. Ma, Y.; Vassetzky, Y.; Dokudovskaya, S. MTORC1 Pathway in DNA Damage Response. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 1293–1311. [Google Scholar] [CrossRef]
  46. Du Rusquec, P.; Blonz, C.; Frenel, J.S.; Campone, M. Targeting the PI3K/Akt/MTOR Pathway in Estrogen-Receptor Positive HER2 Negative Advanced Breast Cancer. Ther. Adv. Med. Oncol. 2020, 12, 175883592094093. [Google Scholar] [CrossRef]
  47. Li, H.; Zeng, J.; Shen, K. PI3K/AKT/MTOR Signaling Pathway as a Therapeutic Target for Ovarian Cancer. Arch. Gynecol. Obstet. 2014, 290, 1067–1078. [Google Scholar] [CrossRef]
  48. Tan, A.C. Targeting the PI3K/Akt/MTOR Pathway in Non-small Cell Lung Cancer (NSCLC). Thorac. Cancer 2020, 11, 511–518. [Google Scholar] [CrossRef] [Green Version]
  49. Peng, Y.; Wang, Y.; Zhou, C.; Mei, W.; Zeng, C. PI3K/Akt/MTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? Front. Oncol. 2022, 12, 819128. [Google Scholar] [CrossRef]
  50. Popova, N.V.; Jücker, M. The Role of MTOR Signaling as a Therapeutic Target in Cancer. Int. J. Mol. Sci. 2021, 22, 1743. [Google Scholar] [CrossRef]
  51. Grabiner, B.C.; Nardi, V.; Birsoy, K.K.; Possemato, R.; Shen, K.; Sinha, S.; Jordan, A.; Beck, A.H.; Sabatini, D.M. A Diverse Array of Cancer-Associated MTOR Mutations Are Hyperactivating and Can Predict Rapamycin Sensitivity. Cancer Discov. 2014, 4, 554–563. [Google Scholar] [CrossRef] [Green Version]
  52. Seeboeck, R.; Sarne, V.; Haybaeck, J. Current Coverage of the MTOR Pathway by Next-Generation Sequencing Oncology Panels. Int. J. Mol. Sci. 2019, 20, 690. [Google Scholar] [CrossRef] [Green Version]
  53. Gremke, N.; Polo, P.; Dort, A.; Schneikert, J.; Elmshäuser, S.; Brehm, C.; Klingmüller, U.; Schmitt, A.; Reinhardt, H.C.; Timofeev, O.; et al. MTOR-Mediated Cancer Drug Resistance Suppresses Autophagy and Generates a Druggable Metabolic Vulnerability. Nat. Commun. 2020, 11, 4684. [Google Scholar] [CrossRef]
  54. Artemenko, M.; Zhong, S.S.W.; To, S.K.Y.; Wong, A.S.T. P70 S6 Kinase as a Therapeutic Target in Cancers: More than Just an MTOR Effector. Cancer Lett. 2022, 535, 215593. [Google Scholar] [CrossRef]
  55. Gingras, A.C.; Gygi, S.P.; Raught, B.; Polakiewicz, R.D.; Abraham, R.T.; Hoekstra, M.F.; Aebersold, R.; Sonenberg, N. Regulation of 4E-BP1 Phosphorylation: A Novel Two-Step Mechanism. Genes Dev. 1999, 13, 1422–1437. [Google Scholar] [CrossRef] [Green Version]
  56. Maracci, C.; Motta, S.; Romagnoli, A.; Costantino, M.; Perego, P.; Di Marino, D. The MTOR/4E-BP1/EIF4E Signalling Pathway as a Source of CancerDrug Targets. Curr. Med. Chem. 2022, 29, 3501–3529. [Google Scholar] [CrossRef]
  57. Dhar, R.; Basu, A. Constitutive Activation of P70 S6 Kinase Is Associated with Intrinsic Resistance to Cisplatin. Int. J. Oncol. 2008, 32, 1133–1137. [Google Scholar]
  58. Liu, J.; Zhang, L.; Zhang, X.; Xing, X. Rapamycin Enhanced the Antitumor Efficacy of Oxaliplatin in Cisplatin-Resistant Ovarian Cancer Cells A2780cis Both in Vitro and in Vivo. J. Chemother. 2015, 27, 358–364. [Google Scholar] [CrossRef]
  59. Nam, K.H.; Yi, S.A.; Nam, G.; Noh, J.S.; Park, J.W.; Lee, M.G.; Park, J.H.; Oh, H.; Lee, J.; Lee, K.R.; et al. Identification of a Novel S6K1 Inhibitor, Rosmarinic Acid Methyl Ester, for Treating Cisplatin-Resistant Cervical Cancer. BMC Cancer 2019, 19, 773. [Google Scholar] [CrossRef]
  60. Zhang, X.; Wang, X.; Song, X.; Liu, C.; Shi, Y.; Wang, Y.; Afonja, O.; Ma, C.; Chen, Y.H.; Zhang, L. Programmed Cell Death 4 Enhances Chemosensitivity of Ovarian Cancer Cells by Activating Death Receptor Pathway in Vitro and in Vivo. Cancer Sci. 2010, 101, 2163–2170. [Google Scholar] [CrossRef]
  61. Liu, R.-Y.; Dong, Z.; Liu, J.; Yin, J.-Y.; Zhou, L.; Wu, X.; Yang, Y.; Mo, W.; Huang, W.; Khoo, S.K.; et al. Role of EIF3a in Regulating Cisplatin Sensitivity and in Translational Control of Nucleotide Excision Repair of Nasopharyngeal Carcinoma. Oncogene 2011, 30, 4814–4823. [Google Scholar] [CrossRef] [Green Version]
  62. Yin, J.-Y.; Shen, J.; Dong, Z.-Z.; Huang, Q.; Zhong, M.-Z.; Feng, D.-Y.; Zhou, H.-H.; Zhang, J.-T.; Liu, Z.-Q. Effect of EIF3a on Response of Lung Cancer Patients to Platinum-Based Chemotherapy by Regulating DNA Repair. Clin. Cancer Res. 2011, 17, 4600–4609. [Google Scholar] [CrossRef] [Green Version]
  63. Ma, S.; Dong, Z.; Huang, Y.; Liu, J.-Y.; Zhang, J.-T. EIF3a Regulation of MTOR Signaling and Translational Control via HuR in Cellular Response to DNA Damage. Oncogene 2022, 41, 2431–2443. [Google Scholar] [CrossRef]
  64. Hsu, Y.-C.; Chern, J.J.; Cai, Y.; Liu, M.; Choi, K.-W. Drosophila TCTP Is Essential for Growth and Proliferation through Regulation of DRheb GTPase. Nature 2007, 445, 785–788. [Google Scholar] [CrossRef]
  65. Bommer, U.-A.; Telerman, A. Dysregulation of TCTP in Biological Processes and Diseases. Cells 2020, 9, 1632. [Google Scholar] [CrossRef]
  66. Jeong, M.; Jeong, M.H.; Kim, J.E.; Cho, S.; Lee, K.J.; Park, S.; Sohn, J.; Park, Y.G. TCTP Protein Degradation by Targeting MTORC1 and Signaling through S6K, Akt, and Plk1 Sensitizes Lung Cancer Cells to DNA-Damaging Drugs. Sci. Rep. 2021, 11, 20812. [Google Scholar] [CrossRef]
  67. Jiang, W.; Ou, Z.; Zhu, Q.; Zai, H. RagC GTPase Regulates MTOR to Promote Chemoresistance in Senescence-like HepG2 Cells. Front. Physiol. 2022, 13, 949737. [Google Scholar] [CrossRef]
  68. Napolitano, G.; Esposito, A.; Choi, H.; Matarese, M.; Benedetti, V.; Di Malta, C.; Monfregola, J.; Medina, D.L.; Lippincott-Schwartz, J.; Ballabio, A. MTOR-Dependent Phosphorylation Controls TFEB Nuclear Export. Nat. Commun. 2018, 9, 3312. [Google Scholar] [CrossRef]
  69. Li, K.; Wada, S.; Gosis, B.S.; Thorsheim, C.; Loose, P.; Arany, Z. Folliculin Promotes Substrate-Selective MTORC1 Activity by Activating RagC to Recruit TFE3. PLoS Biol. 2022, 20, e3001594. [Google Scholar] [CrossRef]
  70. Petruzzelli, R.; Mariniello, M.; De Cegli, R.; Catalano, F.; Guida, F.; Di Schiavi, E.; Polishchuk, R.S. TFEB Regulates ATP7B Expression to Promote Platinum Chemoresistance in Human Ovarian Cancer Cells. Cells 2022, 11, 219. [Google Scholar] [CrossRef]
  71. Liu, W.; Wang, Y.; Xie, Y.; Dai, T.; Fan, M.; Li, C.; Zou, Y. Cisplatin Remodels the Tumor Immune Microenvironment via the Transcription Factor EB in Ovarian Cancer. Cell Death Discov. 2021, 7, 136. [Google Scholar] [CrossRef]
  72. Luo, L.; Sun, W.; Zhu, W.; Li, S.; Zhang, W.; Xu, X.; Fang, D.; Grahn, T.H.M.; Jiang, L.; Zheng, Y. BCAT1 Decreases the Sensitivity of Cancer Cells to Cisplatin by Regulating MTOR-Mediated Autophagy via Branched-Chain Amino Acid Metabolism. Cell Death Dis. 2021, 12, 169. [Google Scholar] [CrossRef] [PubMed]
  73. Loissell-Baltazar, Y.A.; Dokudovskaya, S. SEA and GATOR 10 Years Later. Cells 2021, 10, 2689. [Google Scholar] [CrossRef]
  74. Ueda, K.; Kawashima, H.; Ohtani, S.; Deng, W.-G.G.; Ravoori, M.; Bankson, J.; Gao, B.; Girard, L.; Minna, J.D.; Roth, J.A.; et al. The 3p21.3 Tumor Suppressor NPRL2 Plays an Important Role in Cisplatin-Induced Resistance in Human Non-Small-Cell Lung Cancer Cells. Cancer Res. 2006, 66, 9682–9690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Jayachandran, G.; Ueda, K.; Wang, B.; Roth, J.A.; Ji, L. NPRL2 Sensitizes Human Non-Small Cell Lung Cancer (NSCLC) Cells to Cisplatin Treatment by Regulating Key Components in the DNA Repair Pathway. PLoS ONE 2010, 5, e11994. [Google Scholar] [CrossRef]
  76. Ma, Y.; Silveri, L.; LaCava, J.; Dokudovskaya, S. Tumor Suppressor NPRL2 Induces ROS Production and DNA Damage Response. Sci. Rep. 2017, 7, 15311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Musa, F.; Alard, A.; David-West, G.; Curtin, J.P.; Blank, S.V.; Schneider, R.J. Dual MTORC1/2 Inhibition as a Novel Strategy for the Resensitization and Treatment of Platinum-Resistant Ovarian Cancer. Mol. Cancer Ther. 2016, 15, 1557–1567. [Google Scholar] [CrossRef] [Green Version]
  78. Sheng, J.; Shen, L.; Sun, L.; Zhang, X.; Cui, R.; Wang, L. Inhibition of PI3K/MTOR Increased the Sensitivity of Hepatocellular Carcinoma Cells to Cisplatin via Interference with Mitochondrial-lysosomal Crosstalk. Cell Prolif. 2019, 52, e12609. [Google Scholar] [CrossRef] [Green Version]
  79. Wangpaichitr, M.; Wu, C.; You, M.; Kuo, M.T.; Feun, L.; Lampidis, T.; Savaraj, N. Inhibition of MTOR Restores Cisplatin Sensitivity through Down-Regulation of Growth and Anti-Apoptotic Proteins. Eur. J. Pharmacol. 2008, 591, 124–127. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, J.; Jiang, Y.; Yu, Y.; Li, J. Preclinical Evaluation of the Dual MTORC1/2 Inhibitor Sapanisertib in Combination with Cisplatin in Nasopharyngeal Carcinoma. Eur. J. Pharmacol. 2022, 915, 174688. [Google Scholar] [CrossRef]
  81. Park, I.H.; Kong, S.-Y.; Kwon, Y.; Kim, M.K.; Sim, S.H.; Joo, J.; Lee, K.S. Phase I/II Clinical Trial of Everolimus Combined with Gemcitabine/Cisplatin for Metastatic Triple-Negative Breast Cancer. J. Cancer 2018, 9, 1145–1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Curigliano, G.; Shapiro, G.I.; Kristeleit, R.S.; Abdul Razak, A.R.; Leong, S.; Alsina, M.; Giordano, A.; Gelmon, K.A.; Stringer-Reasor, E.; Vaishampayan, U.N.; et al. A Phase 1B Open-Label Study of Gedatolisib (PF-05212384) in Combination with Other Anti-Tumour Agents for Patients with Advanced Solid Tumours and Triple-Negative Breast Cancer. Br. J. Cancer 2023, 128, 30–41. [Google Scholar] [CrossRef]
  83. Fury, M.G.; Sherman, E.; Ho, A.L.; Xiao, H.; Tsai, F.; Nwankwo, O.; Sima, C.; Heguy, A.; Katabi, N.; Haque, S.; et al. A Phase 1 Study of Everolimus plus Docetaxel plus Cisplatin as Induction Chemotherapy for Patients with Locally and/or Regionally Advanced Head and Neck Cancer: Phase 1 Everolimus Plus TP in H&N CA. Cancer 2013, 119, 1823–1831. [Google Scholar] [CrossRef] [Green Version]
  84. Costello, B.A.; Borad, M.J.; Qi, Y.; Kim, G.P.; Northfelt, D.W.; Erlichman, C.; Alberts, S.R. Phase I Trial of Everolimus, Gemcitabine and Cisplatin in Patients with Solid Tumors. Investig. New Drugs 2014, 32, 710–716. [Google Scholar] [CrossRef]
  85. Fury, M.G.; Sherman, E.; Haque, S.; Korte, S.; Lisa, D.; Shen, R.; Wu, N.; Pfister, D. A Phase I Study of Daily Everolimus plus Low-Dose Weekly Cisplatin for Patients with Advanced Solid Tumors. Cancer Chemother. Pharmacol. 2012, 69, 591–598. [Google Scholar] [CrossRef]
  86. Ren, J.-H.; He, W.-S.; Nong, L.; Zhu, Q.-Y.; Hu, K.; Zhang, R.-G.; Huang, L.-L.; Zhu, F.; Wu, G. Acquired Cisplatin Resistance in Human Lung Adenocarcinoma Cells Is Associated with Enhanced Autophagy. Cancer Biother. Radiopharm. 2010, 25, 75–80. [Google Scholar] [CrossRef]
  87. Hwang, J.R.; Kim, W.Y.; Cho, Y.-J.; Ryu, J.-Y.; Choi, J.-J.; Jeong, S.Y.; Kim, M.-S.; Kim, J.H.; Paik, E.S.; Lee, Y.-Y.; et al. Chloroquine Reverses Chemoresistance via Upregulation of P21WAF1/CIP1 and Autophagy Inhibition in Ovarian Cancer. Cell Death Dis. 2020, 11, 1034. [Google Scholar] [CrossRef]
  88. Huang, D.; Savage, S.R.; Calinawan, A.P.; Lin, C.; Zhang, B.; Wang, P.; Starr, T.K.; Birrer, M.J.; Paulovich, A.G. A Highly Annotated Database of Genes Associated with Platinum Resistance in Cancer. Oncogene 2021, 40, 6395–6405. [Google Scholar] [CrossRef]
  89. Miyamoto, M.; Takano, M.; Aoyama, T.; Soyama, H.; Yoshikawa, T.; Tsuda, H.; Furuya, K. Inhibition of Autophagy Protein LC3A as a Therapeutic Target in Ovarian Clear Cell Carcinomas. J. Gynecol. Oncol. 2017, 28, e33. [Google Scholar] [CrossRef] [Green Version]
  90. Hu, X.; Ma, Z.; Wen, L.; Li, S.; Dong, Z. Autophagy in Cisplatin Nephrotoxicity during Cancer Therapy. Cancers 2021, 13, 5618. [Google Scholar] [CrossRef]
  91. Xu, J.; Gewirtz, D.A. Is Autophagy Always a Barrier to Cisplatin Therapy? Biomolecules 2022, 12, 463. [Google Scholar] [CrossRef]
  92. Yu, G.; Klionsky, D.J. Life and Death Decisions—The Many Faces of Autophagy in Cell Survival and Cell Death. Biomolecules 2022, 12, 866. [Google Scholar] [CrossRef]
  93. Gewirtz, D.A. The Four Faces of Autophagy: Implications for Cancer Therapy. Cancer Res. 2014, 74, 647–651. [Google Scholar] [CrossRef] [Green Version]
  94. Patel, N.H.; Xu, J.; Saleh, T.; Wu, Y.; Lima, S.; Gewirtz, D.A. Influence of Nonprotective Autophagy and the Autophagic Switch on Sensitivity to Cisplatin in Non-Small Cell Lung Cancer Cells. Biochem. Pharmacol. 2020, 175, 113896. [Google Scholar] [CrossRef]
  95. Michaud, M.; Martins, I.; Sukkurwala, A.Q.; Adjemian, S.; Ma, Y.; Pellegatti, P.; Shen, S.; Kepp, O.; Scoazec, M.; Mignot, G.; et al. Autophagy-Dependent Anticancer Immune Responses Induced by Chemotherapeutic Agents in Mice. Science 2011, 334, 1573–1577. [Google Scholar] [CrossRef]
  96. Wang, S.; Li, M.-Y.; Liu, Y.; Vlantis, A.C.; Chan, J.Y.; Xue, L.; Hu, B.-G.; Yang, S.; Chen, M.-X.; Zhou, S.; et al. The Role of MicroRNA in Cisplatin Resistance or Sensitivity. Expert Opin. Ther. Targets 2020, 24, 885–897. [Google Scholar] [CrossRef]
  97. Pennati, M.; Lopergolo, A.; Profumo, V.; De Cesare, M.; Sbarra, S.; Valdagni, R.; Zaffaroni, N.; Gandellini, P.; Folini, M. MiR-205 Impairs the Autophagic Flux and Enhances Cisplatin Cytotoxicity in Castration-Resistant Prostate Cancer Cells. Biochem. Pharmacol. 2014, 87, 579–597. [Google Scholar] [CrossRef]
  98. Shen, Q.; Xu, Z.; Xu, S. Long Non-coding RNA LUCAT1 Contributes to Cisplatin Resistance by Regulating the MiR-514a-3p/ULK1 Axis in Human Non-small Cell Lung Cancer. Int. J. Oncol. 2020, 57, 967–979. [Google Scholar] [CrossRef]
  99. Zhao, J.; Nie, Y.; Wang, H.; Lin, Y. MiR-181a Suppresses Autophagy and Sensitizes Gastric Cancer Cells to Cisplatin. Gene 2016, 576, 828–833. [Google Scholar] [CrossRef]
  100. Li, B.; Wang, W.; Li, Z.; Chen, Z.; Zhi, X.; Xu, J.; Li, Q.; Wang, L.; Huang, X.; Wang, L.; et al. MicroRNA-148a-3p Enhances Cisplatin Cytotoxicity in Gastric Cancer through Mitochondrial Fission Induction and Cyto-Protective Autophagy Suppression. Cancer Lett. 2017, 410, 212–227. [Google Scholar] [CrossRef]
  101. Matsui, T.; Fukuda, M. Rab12 Regulates MTORC1 Activity and Autophagy through Controlling the Degradation of Amino-acid Transporter PAT4. EMBO Rep. 2013, 14, 450–457. [Google Scholar] [CrossRef] [Green Version]
  102. Sirohi, K.; Chalasani, M.L.S.; Sudhakar, C.; Kumari, A.; Radha, V.; Swarup, G. M98K-OPTN Induces Transferrin Receptor Degradation and RAB12—Mediated Autophagic Death in Retinal Ganglion Cells. Autophagy 2013, 9, 510–527. [Google Scholar] [CrossRef] [Green Version]
  103. Han, Y.; Kim, B.; Cho, U.; Park, I.S.; Kim, S.I.; Dhanasekaran, D.N.; Tsang, B.K.; Song, Y.S. Mitochondrial Fission Causes Cisplatin Resistance under Hypoxic Conditions via ROS in Ovarian Cancer Cells. Oncogene 2019, 38, 7089–7105. [Google Scholar] [CrossRef]
  104. Woo, S.M.; Min, K.; Kwon, T.K. Inhibition of Drp1 Sensitizes Cancer Cells to Cisplatin-Induced Apoptosis through Transcriptional Inhibition of c-FLIP Expression. Molecules 2020, 25, 5793. [Google Scholar] [CrossRef]
  105. Ma, M.; Lin, X.; Liu, H.; Zhang, R.; Chen, R. Suppression of DRP1-mediated Mitophagy Increases the Apoptosis of Hepatocellular Carcinoma Cells in the Setting of Chemotherapy. Oncol. Rep. 2020, 43, 1010–1018. [Google Scholar] [CrossRef]
  106. Vianello, C.; Cocetta, V.; Catanzaro, D.; Dorn, G.W.; De Milito, A.; Rizzolio, F.; Canzonieri, V.; Cecchin, E.; Roncato, R.; Toffoli, G.; et al. Cisplatin Resistance Can Be Curtailed by Blunting Bnip3-Mediated Mitochondrial Autophagy. Cell Death Dis. 2022, 13, 398. [Google Scholar] [CrossRef]
  107. Wu, H.-M.; Jiang, Z.-F.; Ding, P.-S.; Shao, L.-J.; Liu, R.-Y. Hypoxia-Induced Autophagy Mediates Cisplatin Resistance in Lung Cancer Cells. Sci. Rep. 2015, 5, 12291. [Google Scholar] [CrossRef] [Green Version]
  108. Kocak, M.; Ezazi Erdi, S.; Jorba, G.; Maestro, I.; Farrés, J.; Kirkin, V.; Martinez, A.; Pless, O. Targeting Autophagy in Disease: Established and New Strategies. Autophagy 2022, 18, 473–495. [Google Scholar] [CrossRef]
Figure 1. Cancers where cisplatin is used as a mainstream drug.
Figure 1. Cancers where cisplatin is used as a mainstream drug.
Ijms 24 10651 g001
Figure 2. Main factors of cisplatin resistance. Cancer cells can develop resistance to cisplatin via decreased drug influx or increased efflux, because of the drug inactivation through the interaction with glutathione (GSH) and metallothioneins (MT), because of enhanced DNA damage response and alternations in signaling pathways.
Figure 2. Main factors of cisplatin resistance. Cancer cells can develop resistance to cisplatin via decreased drug influx or increased efflux, because of the drug inactivation through the interaction with glutathione (GSH) and metallothioneins (MT), because of enhanced DNA damage response and alternations in signaling pathways.
Ijms 24 10651 g002
Figure 3. Cisplatin and its derivatives used in the treatment of different types of cancers. Side effects for each drug are listed.
Figure 3. Cisplatin and its derivatives used in the treatment of different types of cancers. Side effects for each drug are listed.
Ijms 24 10651 g003
Figure 4. Overview of mTORC1 signaling.
Figure 4. Overview of mTORC1 signaling.
Ijms 24 10651 g004
Figure 5. mTORC1 pathway factors and effectors involved in cisplatin resistance. The factors and effectors of the same signaling path are indicated with the same color. EIF3A translational factor negatively regulates mTORC1 activity. Accordingly, EIF3A downregulation (black arrow) increases mTORC1 activity. The activation of p70S6K by mTORC1 can be suppressed by RAME. Many oncogenic mutations have been detected in eukaryotic translation initiation factors, a group of mTORC1 downstream effectors. In addition, PDCD4, which is not a direct target of mTORC1, can suppress protein translation by interacting with EIF4A and EIF4G to inhibit the formation of the translation initiation complex. A knockdown of PDCD4 (black arrow) reduces sensitivity to platinum drugs. BCAT1 overexpression results in a decrease of leucine and other branched-chain amino acid levels. As a consequence, mTORC1 cannot be effectively activated (dashed arrow), resulting in enhanced autophagy and cisplatin resistance. NPRL2 downregulation (white arrow) results in mTORC1 activation, compromised DNA damage response and cisplatin resistance. See text for more details. RAGD promotes TFEB inhibition through its phosphorylation by mTORC1 and sequestration in the cytoplasm, where TFEB cannot exert its function as a transcription factor. Cisplatin treatment induces TFEB nuclear translocation and activation. Active TFEB increases the expression of programmed cell death-ligands 1 and 2 (PD-L1 and PD-L2) to foster an immunosuppressive tumor microenvironment that mediates drug resistance. The suppression of TFEB inhibits the expression of the copper transporter ATP7B involved in cisplatin efflux and sensitizes initially resistant ovarian cancer cells to cisplatin.
Figure 5. mTORC1 pathway factors and effectors involved in cisplatin resistance. The factors and effectors of the same signaling path are indicated with the same color. EIF3A translational factor negatively regulates mTORC1 activity. Accordingly, EIF3A downregulation (black arrow) increases mTORC1 activity. The activation of p70S6K by mTORC1 can be suppressed by RAME. Many oncogenic mutations have been detected in eukaryotic translation initiation factors, a group of mTORC1 downstream effectors. In addition, PDCD4, which is not a direct target of mTORC1, can suppress protein translation by interacting with EIF4A and EIF4G to inhibit the formation of the translation initiation complex. A knockdown of PDCD4 (black arrow) reduces sensitivity to platinum drugs. BCAT1 overexpression results in a decrease of leucine and other branched-chain amino acid levels. As a consequence, mTORC1 cannot be effectively activated (dashed arrow), resulting in enhanced autophagy and cisplatin resistance. NPRL2 downregulation (white arrow) results in mTORC1 activation, compromised DNA damage response and cisplatin resistance. See text for more details. RAGD promotes TFEB inhibition through its phosphorylation by mTORC1 and sequestration in the cytoplasm, where TFEB cannot exert its function as a transcription factor. Cisplatin treatment induces TFEB nuclear translocation and activation. Active TFEB increases the expression of programmed cell death-ligands 1 and 2 (PD-L1 and PD-L2) to foster an immunosuppressive tumor microenvironment that mediates drug resistance. The suppression of TFEB inhibits the expression of the copper transporter ATP7B involved in cisplatin efflux and sensitizes initially resistant ovarian cancer cells to cisplatin.
Ijms 24 10651 g005
Figure 6. Main factors of autophagy involved in cisplatin resistance. HQ/CQ: chloroquine and hydroxychloroquine.
Figure 6. Main factors of autophagy involved in cisplatin resistance. HQ/CQ: chloroquine and hydroxychloroquine.
Ijms 24 10651 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pan, Z.; Zhang, H.; Dokudovskaya, S. The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics. Int. J. Mol. Sci. 2023, 24, 10651. https://doi.org/10.3390/ijms241310651

AMA Style

Pan Z, Zhang H, Dokudovskaya S. The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics. International Journal of Molecular Sciences. 2023; 24(13):10651. https://doi.org/10.3390/ijms241310651

Chicago/Turabian Style

Pan, Zhenrui, Hanxiao Zhang, and Svetlana Dokudovskaya. 2023. "The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics" International Journal of Molecular Sciences 24, no. 13: 10651. https://doi.org/10.3390/ijms241310651

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

Article Metrics

Back to TopTop