Next Article in Journal
Heat-Inactivated Pediococcus acidilactici pA1c®HI Maintains Glycemic Control and Prevents Body Weight Gain in High-Fat-Diet-Fed Mice
Previous Article in Journal
Short-Term Probiotic Colonization Alters Molecular Dynamics of 3D Oral Biofilms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulation of Endoplasmic Reticulum Stress in Experimental Anti-Cancer Therapy

by
Natalia Ivanovna Agalakova
Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Avenue, Saint-Petersburg 194223, Russia
Int. J. Mol. Sci. 2025, 26(13), 6407; https://doi.org/10.3390/ijms26136407
Submission received: 23 May 2025 / Revised: 28 June 2025 / Accepted: 1 July 2025 / Published: 3 July 2025

Abstract

The growth of tumor cells is accompanied by an increased rate of endoplasmic reticulum stress (ERS), the accumulation of misfolded proteins, and the activation of a network of adaptive signaling pathways known as the unfolded protein response (UPR). Although the UPR is an adaptive reaction aiming to restore ER proteostasis, prolonged and severe ERS leads to cell death. Taking into account that the components of the ERS/UPR machinery in cancers of different types can be overexpressed or downregulated, both the induction of excessive ERS and suppression of UPR have been proposed as therapeutic strategies to sensitize cells to conventional chemotherapy. This narrative review presents a several examples of using natural and synthetic compounds that can either induce persistent ERS by selectively blocking ER Ca2+ pumps (SERCA) to disrupt ER Ca2+ homeostasis, or altering the activity of UPR chaperones and sensors (GRP78, PERK, IRE1α, and ATF6) to impair protein degradation signaling. The molecular alterations induced by miscellaneous inhibitors of ERS/UPR effectors are described as well. These agents showed promising therapeutic effects as a part of combination therapy in preclinical experimental settings; however, the number of clinical trials is still limited, while their results are inconsistent. Multiple side effects, high toxicity to normal cells, or poor bioavailability also hampers their clinical application. Since the pharmacological modulation of ERS/UPR is a valuable approach to sensitize cancer cells to standard chemotherapy, the search for more selective agents with better stability and low toxicity, as well as the development of more efficient delivery systems that can increase their therapeutic specificity, are highly required goals for future studies.

1. Introduction

The progression of malignancies of different origin shares a common pathogenesis, including an increased rate of endoplasmic reticulum stress (ERS) caused by genetic mutations and genomic rearrangements, the accumulation of misfolded proteins, and the subsequent activation of multiple signaling pathways that are together known as the unfolded protein response (UPR) [1,2]. A sustained UPR plays an important role in supporting cancer growth, survival, invasion, and metastasis. The ability of tumor cells to rapidly adapt to stress conditions such as limited oxygen and nutrient supplies by activating/deactivating the UPR determines either the resistance to chemotherapy/radiotherapy or a positive therapeutic response [1,2,3,4]. Additionally, the defense mechanisms induced by ERS are implicated in the processes occurring in the tumor microenvironment, such as angiogenesis [5]. On the other hand, persistent ERS promotes cell death, and its induction via different mechanisms is believed to be a promising approach in anti-tumor therapy [3,6]. Moreover, since different components of ERS/UPR pathways in cancers are upregulated or repressed, their expression profiles might be used for diagnostics, determining the prognosis of disease progression, choosing therapeutic schemes, and assessing the response to chemotherapeutics.
The development of novel anti-cancer drugs is extremely time-consuming and cost-ineffective; therefore, the modulation of ERS/UPR for cancer treatment is an attractive alternative in clinical oncology to sensitize the tumor cells to conventional cytotoxins. In this rapidly growing field of research, not only the development of new, selective agents targeting ERS/UPR effectors but also the repurposing of long known herbal or synthetic drugs in combination therapy have emerged [3,6,7,8]. Pro-apoptotic ERS-dependent anti-tumor activity could be achieved either by suppressing the adaptive UPR or by exaggerating ERS [4]. This narrative review highlights recent experimental pharmacological strategies involving a few natural and synthetic blockers used to induce ERS or disrupt UPR signaling. It focuses on miscellaneous compounds either suppressing ER Ca2+ pumps (SERCA) to compromise ER Ca2+ homeostasis or directly targeting UPR sensors to impair the protein degradation machinery (Figure 1).

2. Endoplasmic Reticulum Stress and the Unfolded Protein Response

The endoplasmic reticulum (ER) is a continuous interconnected membrane network consisting of rough ER serving for the synthesis, folding, maturation, and transport of proteins, and smooth ER responsible for the biosynthesis of lipids. Moreover, the ER is a cellular Ca2+ store. Such multiple functions make the ER an important contributor to the maintenance and dynamic regulation of cellular proteostasis, lipid metabolism, redox balance, and Ca2+ homeostasis and signaling [9,10]. The disruption of the ER folding capacity accompanied by the accumulation of unfolded/aggregated proteins leads to a condition known as ER stress (ERS) [1,2,10]. ERS can be induced by various physiological and pathological stimuli such as metabolic alterations, oxidative stress, hypoxia, chronic inflammation, senescence, and a Ca2+ imbalance. To clear the misfolded proteins and enhance the ER folding capacity, cells evolved a protective mechanism—unfolded protein response (UPR)—that includes several transcriptional and translational modifications serving to attenuate protein synthesis and restore ER homeostasis [10,11,12].
The UPR is directed by three transmembrane ER sensors: PERK (PKR-like endoplasmic reticulum kinase), IRE1 (inositol-requiring enzyme 1α) and ATF6 (activating transcription factor 6) [1,2,12,13]. Under healthy conditions, these proteins are inactive due to binding of their luminal domains to the chaperone GRP78 (glucose-regulated protein 78, also known as binding immunoglobulin protein, BiP, or human heat shock protein 5, HSPA5). In response to an increased ER load with misfolded/aggregated proteins, ER sensors dissociate from GRP78 and activate downstream signaling pathways, while GRP78 interacts with unfolded proteins. PERK undergoes dimerization and autophosphorylation allowing it to phosphorylate eIF2α (eukaryotic initiation factor 2α), which inhibits translation and stops the global synthesis of new proteins, a key mechanism of the UPR [14,15]. Another PERK target is ATF4 (activating transcription factor 4), which controls the transcription of adaptive UPR genes and the apoptotic/autophagy genes CHOP (C/EBP protein homolog) and GADD34 (growth arrest and DNA damage-inducible protein 34) [16]. Similar dimerization and autophosphorylation of IRE1 facilitates its RNase activity and triggers the splicing of XBP1 (X-Box binding protein 1) [17,18]. sXBP1 functions as a transcription factor that is able to restore ER homeostasis by enhancing the expression of genes encoding ER chaperones and effectors of the ERAD (ER-associated degradation) system. Another function of IRE1 is the selective destruction of mRNAs and miRNAs encoding compromised ER proteins through a mechanism known as RIDD (regulated IRE1-dependent decay) [19]. The third branch of the UPR includes conformational changes in ATF6, which drives its trafficking to the Golgi apparatus for proteolytic cleavage by serine protease site-1 (S1P) and metalloprotease site-2 (S2P) into the active transcription fragment pATF6α [2,13]. This factor further translocates to the nucleus and stimulates the transcription of XBP1, UPR genes including GRP78, and genes encoding ERAD proteins. In concert, UPR pathways can reduce ERS or restore the ER protein balance by repairing the moderately misfolded proteins but degrading terminally damaged ones. However, if ERS is excessive or persistent, the cells are unable to restore ER homeostasis, and the UPR switches from pro-survival to pro-death signaling [13,20,21]. The effectors connecting the UPR with apoptosis and autophagy include CHOP; JNK (c-Jun N-terminal kinase); the pro-apoptotic Bcl-2 family protein NOXA; caspase-4, which is selectively stimulated by disturbed ER homeostasis; and caspase-12, while the cell destiny depends on the ratio between the activities of these molecules.

3. ERS in Cancer Cells

3.1. Ca2+ Homeostasis, SERCA, and Cancer

The compartmentalization and precise control of the Ca2+ distribution in the cells, with a high Ca2+ concentration in the ER (100–800 µM) and low cytoplasmic Ca2+ levels (50–100 nM), are the phenomena enabling the regulation of various physiological processes, such as gene transcription, cell differentiation, growth, metabolism, migration, survival, or death [22,23]. Strict regulation of the ER Ca2+ content is necessary for proper protein synthesis, folding, and maturation. Even a partial ER Ca2+ loss enhances the accumulation of unfolded proteins and the UPR with the stimulation of PERK and IREα, while ER Ca2+ replenishing leads to complete IRE1α and PERK dephosphorylation [24]. However, a cellular Ca2+ imbalance due to altered activities of plasma membrane and ER membrane Ca2+ channels and pumps are involved in cancer spread and chemoresistance [23,25,26,27].
SERCA (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase) is a P-type transporter located on ER membranes and existing as a few isoforms (1a, 1b, 2a, 2b, and 3). It is responsible for replenishing the ER Ca2+ stores by transporting Ca2+ from the cytoplasm against its concentration gradient [22,28,29]. In concert with plasma membrane and mitochondrial Ca2+ pumps and channels, it plays a crucial role in maintaining cellular Ca2+ homeostasis. However, aberrant SERCA activity results in the depletion or overload of ER Ca2+ stores and triggers ERS. Since luminal Ca2+ binds to ER chaperones like GRP78, calnexin, or calreticulin, an ER Ca2+ loss changes their functioning [22,28].
The overexpression, mutations, or loss of different SERCA isoforms are implicated in various malignancies (Table 1). The upregulation of SERCA1-encoding genes is linked to the reduced survival of patients with breast [30] and colorectal [31] cancers. Increased SERCA2 expression in the tissues of patients with colorectal adenoma and carcinoma positively correlates with the tumor grade and metastasis [32,33,34]. The downregulation of SERCA3 determines the invasion and metastasis of gastric carcinomas [35,36], gliomas, and glioblastomas [37], although it is not linked to the survival of patients with colorectal carcinoma [38]. Decreased SERCA3 expression is observed in choroid plexus tumors [39], while strong SERCA3 immunopositivity is found in gastrointestinal tumors [40]. Both SERCA3 loss [41] and overexpression [37] are found in breast carcinomas of various types.
In spite of such variable expression in tumors of different types, SERCAs attracted great attention as targets for anti-cancer therapy [25,26,27,28,29]. Since SERCA overexpression is a predominant phenomenon, the therapeutic strategies are mainly focused on blocking its activity. SERCA suppression induces aberrant ERS signaling and the depletion of luminal Ca2+, while concomitant excessive cytoplasmic Ca2+ levels cause its entrance into the mitochondrial matrix, mitochondrial Ca2+ overload, and depolarization of inner mitochondrial membranes. The following events include the activation of pro-apoptotic Bcl-2 family proteins (Bax and Bak), Cytochrome C release, and the formation of the apoptosome Apaf-1. High cytosolic Ca2+ levels also stimulate the protease calpain, which cleaves ER-associated procaspase-12, leading to the activation of caspases-9 and -3, and eventually cell death [2,22].

3.2. UPR Components and Cancer

Abnormal activities of all UPR effectors (PERK, IRE1α, and ATF6) have been shown to be implicated in the survival, metastasis, angiogenesis, and chemoresistance of glioblastoma [42], pancreatic [43,44], lung [45], colon/colorectal [46,47], and urological [48] cancers at different stages of the disease. The overexpression, downregulation, phosphorylation, and mutation of several eIF subunits (eIFs) in malignancies of various origins have been reported as well [49,50]. The IRE1α/XBP1 branch is implicated in the development of a variety of hematologic [51,52] and pancreatic [53] cancers. GRP78 also emerges as a central player in the pathogenesis of and chemoresistance in many cancers [54,55].
PERK/eIFα. Increased PERK levels in the tumor tissues are associated with a poor prognosis for patients with prostate cancer [56], pancreatic duct adenocarcinoma (PDAC) [57], kidney renal papillary cell carcinoma, gliomas, breast carcinoma, and thyroid carcinoma [58], but determined a favorable prognosis for head and neck squamous cell carcinoma (HNSCC) [58] (Table 2). In contrast, it was downregulated in Hodgkin’s lymphoma, T-cell lymphomas, and gastrointestinal stromal tumors [58].
eIF2α overexpression was found in tumors originating from melanocytes and the colonic epithelium [59], in aggressive thyroid cancers [60], Hodgkin’s lymphoma [61], and the tumor tissues and blood plasma of breast cancer patients [62,63]. Sustained eIF2α phosphorylation was linked with a poor prognosis, higher risk of metastasis, and lower disease-free survival of patients with prostate cancer [56,64], PDAC [57], pancreatic adenocarcinoma (PAAD) [65], hepatocellular carcinoma (HCC) [66], and brain tumors [67]. On the other hand, eIF2α hyperphosphorylation correlated with better disease-free survival and favorable clinical outcomes for patients with stomach, colon and sigma-rectum carcinomas [68], non-small cell lung cancer (NSCLC) [69], and triple-negative breast cancer (TNBC) [70].
IRE1α/XBP1. The upregulation of IRE1α was found to be an independent factor predicting a higher recurrence of prostate cancer [56,71]. In contrast, in lung adenocarcinoma, IRE1α overexpression was associated with a favorable prognosis [72].
XBP1 overexpression and splicing (sXBP1) was reported in the tumor tissues of patients with HCC [73], breast adenocarcinoma [74], melanoma [75], and glioblastoma [76]. XBP1 upregulation predicted a poor response to therapy and shorter survival of patients with aggressive diffuse large B-cell and plasmablastic lymphomas [77], acute lymphoblastic leukemia (ALL) [78], aggressive luminal B and ER+ breast cancers [79,80], and prostate cancer [81]. On the other hand, increased levels of mature and spliced XBP1 forms were associated with better overall survival of patients with acute myeloid leukemia (AML) [82,83]. Moderate sXBP1 expression sensitized the cells to cytotoxic treatments, while sustained XBP1 activation induced apoptosis [84]. However, in pancreatic cancer, XBP1 upregulation was not associated with recurrence and overall survival [85].
ATF6. Increased levels of the ATF6 mRNA and its transcriptionally active nuclear product pATF6α were observed in the tissues of patients with moderately to poorly differentiated HCC [73] and osteosarcoma [86]. ATF6 overexpression was associated with chemoresistance and poor clinical outcomes of ovarian cancer [87], gastric cancer [88], pancreatic cancer [85], oral squamous cell carcinoma [89], and HNSCC [90]. However, high ATF6 level did not correlate with the overall survival of patients with biliopancreatic carcinoma [91], prostate [56], and colon [92] cancers, although it was a predictor of a shorter recurrence time.
GRP78. High expression of GRP78 was found in the tissue lesions and bone marrow of patients with breast cancer [74,93,94]. Abnormally elevated levels of GRP78 correlated with stronger aggressiveness, lower overall survival, increased invasion and metastasis, and low sensitivity to anti-cancer agents in patients with HCC [73,95,96], gastric cancer [97], gliomas [98], prostate cancer [99], PDAC [100], lung cancer [101,102], and HNSCC [103]. In contrast, high GRP78 expression in the tissues of patients with colorectal cancer was associated with improved survival after surgery [104].
Table 2. Examples of the aberrant expression of UPR effectors in human cancer tissues.
Table 2. Examples of the aberrant expression of UPR effectors in human cancer tissues.
EffectorExpression
Profile
Cancer TypeClinical OutcomeReferences
PERKOverexpressionProstate cancerPoor prognosis[56]
Pancreatic duct adenocarcinoma[57]
Kidney renal papillary cell carcinoma[58]
Brain glioma[58]
Breast carcinoma[58]
Thyroid carcinoma[58]
Head and neck squamous cell carcinomaFavorable prognosis[58]
eIF2αOverexpression, PhosphorylationMelanoma, colonic adenoma and adenocarcinoma-[59]
Aggressive thyroid carcinoma and papillary carcinoma[60]
Hodgkin’s lymphoma[61]
Prostate cancerPoor prognosis
Lower overall and disease-free survival
Metastasis
Chemoresistance
[56]
Pancreatic duct adenocarcinoma[57]
Pancreatic adenocarcinoma[65]
Hepatocellular carcinoma[66]
Brain meningioma, astrocytoma and oligodendroglial tumors[67]
Overexpression, PhosphorylationStomach, colon, sigma-rectum carcinomaLonger survival
Better disease-free survival
[68]
Non-small cell lung cancer[69]
Triple-negative breast cancer[70]
IRE1αOverexpressionProstate cancerHigher recurrence[56,71]
Lung adenocarcinomaFavorable prognosis[72]
XBP1/sXBP1Overexpression
Splicing
Hepatocellular carcinoma-[73]
Breast adenocarcinoma[74]
Melanoma[75]
Glioblastoma[76]
Diffuse large B-cell lymphomaShorter overall and disease-free survival
Poor response to therapy
[77]
Acute lymphoblastic leukemia[78]
Aggressive luminal B breast cancer[79]
ER+ breast cancer[80]
Prostate cancer[81]
Acute myeloid leukemiaBetter disease-free and overall survival
Lower relapse rate
[82,83]
Pancreatic cancerNo correlation with survival[85]
ATF6OverexpressionHepatocellular carcinomaPoor prognosis
Chemoresistance, lower overall survival
[73]
Osteosarcoma[86]
Ovarian cancer[87]
Gastric cancer[88]
Pancreatic cancer[85]
Oral squamous cell carcinoma[89]
Head and neck squamous carcinoma[90]
Biliopancreatic carcinomaNo correlation with survival[91]
Prostate cancer[56]
Colon cancer[92]
GRP78OverexpressionBreast adenocarcinoma-[74,93,94]
Hepatocellular carcinomaPoor prognosis
Lower overall survival
Invasion
Metastasis
Chemoresistance
[73,95,96]
Gastric cancer[97]
Gliomas[98]
Prostate cancer[99]
Pancreatic duct adenocarcinoma[100]
Lung cancer[101,102]
Head and neck squamous cell carcinomas[103]
Colorectal cancerImproved survival[104]
Comments: - not analyzed.
Such diverse findings encouraged using both the induction of ERS and inhibition of UPR effectors as therapeutic approaches. Strong anti-tumor activity caused by chronic ERS followed by apoptosis was achieved in primary and stable leukemic cell lines [51] and ovarian cancer [105]. A variety of natural, synthetic, and semisynthetic compounds either activating or suppressing IRE1α, PERK, ATF6, or GRP78 has been tested for the treatment of breast cancer [7,106], thyroid cancer [8], and glioma/glioblastoma [107,108]. A few agents that stimulate the UPR entered clinical trials for pancreatic cancers [44]. Suppression of the adaptive UPR makes tumor cells more susceptible to chemotherapy as well [109,110].

4. SERCA as a Target for Cancer Treatment

To date, a few chemically and functionally diverse SERCA inhibitors of natural (terpenoids, polyphenols, and flavonoids) or synthetic origin have been developed and examined [29,111,112].

4.1. Terpenoids

Thapsigargin (TG), a terpenoid with a long history of use in traditional medicine, is a highly selective irreversible SERCA blocker that prevents Ca2+ transport from the cytosol to ER, thus depleting the ER Ca2+ stores but increasing the cytosolic Ca2+ content [113,114]. TG is a SERCA inhibitor that has been probably most extensively tested for anti-cancer therapy [115,116]. It effectively suppressed the growth of cultured prostate, renal, bladder, colon, thyroid, and breast cancer cells [117]. In conventional (MCF7) and metastatic (MDA-MB-231) breast cancer cells, TG increased the generation of ROS, activated PARP and caspases-8, -9 and -3 [118,119], reduced the mRNA and protein levels of Ca2+-binding protein S100A4 [120], and induced the contraction and rearrangement of actin cytoskeleton with the phosphorylation of myosin light chain 2 kinase and myosin phosphatase 1 [121] (Table 3). The effects of TG on prostate cancer LNCaP and PC3 cells include the sustained activation of the death receptor DR5, chaperones GRP94 and GRP78, PERK/ATF4/CHOP and IRE1/XBP1/JNK cascades, and cleavage of PARP and caspase-8 downstream of MAP1LC3B [118,122]. In patient-derived stem cell-enriched glioblastoma cultures, TG impaired the ability of cells to form neurospheres by decreasing the expression of the stem cell transcription factor SOX2 but stimulating the UPR and apoptosis [123]. The disturbance in ER Ca2+ homeostasis in neuroblastoma SH-SY5Y cells was accompanied by the activation of caspase-4 [124]. Mitochondrial apoptosis and ERS through JNK signaling with the upregulation of the UPR and autophagy are implicated in TG’s effects on adrenocortical carcinoma SW-13 and NCI-H295R cells and mouse ACC xenograft models [125]. Increased expression of CHOP, DR5, and Bid and the cleavage of PARP and caspases-3 and 8 were observed in TG-exposed liposarcoma SW872 cells [126].
In combination therapy, TG exerted additive or synergistic effects with a variety of cytotoxic or anti-proliferative agents. It enhanced the responses of metastatic and cisplatin-resistant breast cancer cells to cisplatin [127,128]. Although not effective as a single agent, TG increased the sensitivity of patient-derived papillary thyroid cancer (PTC) YUMC cells and mouse YUMC xenograft models to paclitaxel, sorafenib and lenvatinib [129,130]. In oral cancer CAL 27 and Ca 9–22 cells, TG potentiated the effect of the Ca2+ channel blocker manoalide, a sesterterpenoid isolated from the marine sponge Luffariella variabilis [131]. In breast cancer MCF7 and MDA-MB-231 cells, it synergized with the anti-proliferative effect of nodakenin, a bioactive coumarin glycoside from Angelica gigas [132], and physapruin A, a withanolide from the plant Physalis peruviana [119].
Thapsigargin analogues. In spite of strong ERS induction and the promising results obtained for cultured cancer cells and animal xenograft models, clinical TG application is not possible due to the unacceptably high toxicity to normal cells [113,117]. As a result, a series of TG water-soluble cell-impermeable prodrugs conjugated to peptides that are specifically cleaved by PSA (prostate-specific antigen) or PSMA (prostate-specific membrane antigen) have been designed [133,134]. Some of them suppressed the viability of prostate cancer cells and the growth of LNCaP prostate xenografts in mice without substantial systemic toxicity [117,118]. One of the PSMA-hydrolyzed prodrugs, mipsagargin (G-202), was accepted for the therapy of multiple advanced solid tumors (prostate cancer, glioblastoma, and hepatocellular carcinoma) in clinical trials [133,134]. However, although G-202 was relatively well tolerated and some patients previously heavily treated with chemotherapy or radiotherapy demonstrated longer disease stabilization, objective clinical responses were not found [135,136]. A recently completed trial that examined the efficacy and safety of G-202 for the treatment of glioblastoma did not show any clear clinical outcomes as well [137]. Due to reported side-effects and other reasons, G-202 was not registered [134], and its role as an anti-cancer agent remains uncertain.
Other terpenoids. Lathyrol is a natural terpenoid extracted from the seeds of the Asian plant Euphorbiae lathyrism. In lung tumor A549 and H460 cells, it triggered apoptosis and G2/M cell cycle arrest accompanied by a cytoplasmic Ca2+ elevation, ROS production, and the upregulation of apoptotic and ERS-related proteins [138]. It also greatly suppressed the growth of subcutaneous H460 tumors in mice without significant effects on the liver, heart, lungs, and kidneys [138]. The downregulation of Ki67, Bcl-2, metalloproteinases, and p-Akt, but overexpression of Bax and cleaved caspases underlie the impaired proliferation and increased apoptosis of renal cell carcinoma exposed to lathyrol [139]. Dihydroartemisinin (DHA), a derivative of natural artemisinin that was used to treat malaria, exerted synergistic anti-proliferative, anti-metastatic, and pro-apoptotic effects in combination with a series of chemotherapy drugs. However, although it effectively inhibits SERCA activity and increases ERS, its clinical application is limited due to low solubility and bioavailability [140]. Another group of natural compounds showing cytotoxic and antimetastatic activity in cancer cells of different origins, including those resistant to chemotherapy, are triterpenes isolated from Alisma species plants, including alisol B [141].

4.2. Curcumin and Its Analogues

Curcumin, a polyphenol isolated from the plant Curcuma longa, is a potent allosteric SERCA blocker that induces conformational changes in its molecule, thus preventing ATP binding [142,143]. This compound is well known from traditional medicine and was tested as a part of combination therapy for breast, oral, lung, prostate, hematological, and brain cancers [144]. The cellular effects of curcumin include the inhibition of proteins of the Wnt/β-catenin, PI3K/Akt, EGFR and NF-kB signaling pathways; cell cycle arrest; ROS production; and changes in miRNA expression [145,146]. It interferes with miscellaneous transcription factors, growth factors, inflammatory cytokines, apoptotic proteins, protein kinases, receptors, and cell survival proteins [144].
A causative link between the anti-neoplastic effects of curcumin and the disruption of Ca2+ homeostasis was shown in ovarian cancer SKOV3, MDAH2774, and PA1 cells [147]. In human liposarcoma SW872 cells and SW872 xenografts, curcumin enhanced eIF2α phosphorylation and increased the expression of ATF4, CHOP, DR5 receptor, and its effectors (caspases-8 and -3, and Bid) [126] (Table 3). The effect of curcumin on glioma LN229 and U87 cells and LN229 xenografts was linked to the suppression of ERK1/2 phosphorylation, metalloproteinases, and cell cycle proteins [148]. By activating both apoptosis and ERS, it increased the sensitivity of cisplatin-resistant NSCLC A549 and H1299 cells to cisplatin [149]. In a combination therapy, curcumin derivatives synergistically enhanced the effect of temozolomide on glioblastoma U87 and LN18 cells [150]. A recent meta-analysis has confirmed its potency to reduce the volume of glioma xenografts in animals [151].
However, in spite of the promising in vitro effects, the effectiveness of curcumin in clinical trials is still not clear due to the high heterogeneity of the obtained results [152]. Thus, the therapy of breast cancer with docetaxel, paclitaxel, gemcitabine, or radiotherapy in combination with curcumin improved the objective response rate and reduced the severity of side effects such as dermatitis [146]. On the other hand, the addition of curcumin to first- or second-line docetaxel treatment of advanced metastatic breast cancer was not effective, with only an insignificant tendency to longer 12-month progression-free survival [153]. The causes of such inconsistency are probably the low chemical stability of curcumin, its limited water solubility, poor bioavailability after oral intake, and rapid elimination from an organism. The development of more effective ways of delivering curcumin to cancer cells would increase its therapeutic specificity in clinical applications.
Curcumin analogues. To enhance the therapeutic potential of curcumin, its derivatives with better bioavailability and stability that might improve anti-cancer effectiveness have been developed [154]. The curcumin analogue F36 exhibited more potency in suppressing SERCA2 and inducing ERS-associated apoptosis in human colon adenocarcinoma epithelial SW480 cells [33]. Similar effects were observed on SW480 cells and mice with xenografts that were treated with the curcumin analog RL71 [155].

4.3. Flavonoids

Quercetin. Among the miscellaneous natural SERCA blockers, flavonoids emerge as valuable agents for preventive or therapeutic anti-cancer purposes [29,111]. Quercetin and its derivatives are able to induce conformational changes in the SERCA molecule [156]. They exert anti-proliferative and anti-apoptotic activities in cancers of different origins by modulating the activities of components of the PI3K/Akt/mTOR, MAPK/ERK, JAK/STAT, and NF-κB signaling pathways [157,158]. Thus, in hepatocellular carcinoma, quercetin promoted apoptosis by inhibiting PI3K/Akt/mTOR cascade proteins and P4HA2 (proline 4-hydroxylase II), an enzyme involved in the biosynthesis of collagen [159]. In acute myeloid leukemia HL-60 cells, it induced both apoptosis and autophagy through the CaMKKβ/-dependent pathway, which included AMPK phosphorylation and the suppression of p-mTOR [160]. The pro-apoptotic effect of quercetin on cutaneous melanoma A375 cells was associated with the activation of GPER (G-protein coupled estrogen receptor) and enhanced expression of p-ERK and c-Myc [161]. A derivative of quercetin, dihydroquercetin, has been tested as anti-cancer agent as well [162].
Luteolin. Luteolin, another natural flavonoid with anti-inflammatory, antioxidant, and anti-cancer properties, induced the same intracellular changes as quercetin in melanoma A375 cells [161]. The anti-proliferative and pro-apoptotic effects of luteolin on non-small cell lung cancer H1299 and A549 cells were associated with the downregulation of the components of the WDR72 (WD repeat domain 72), Akt and EMT (epithelial–mesenchymal transition) axis necessary for protein interactions, cytoskeletal regulation, and cell migration [163]. It also reduced the formation of subcutaneous H1299 tumors in mice without evident damage to major organs [164]. In bladder cancer EJ138 cells, luteolin increased the expression levels of genes encoding apoptosis and autophagy-related proteins [163]. The anti-tumor activity of luteolin in cultured diffuse large B-cell lymphoma U2932 and OCI-LY10 cells and mouse subcutaneous U2932 xenograft models included the suppression of JAK2/STAT3 phosphorylation alongside with changes in the expression of apoptotic proteins [165].
However, as other natural compounds, flavonoids have limited bioavailability. To enhance their potency and chemosensitivity, a variety of more efficient delivery systems, including liposomes and nanoparticles, is currently under development [166,167,168,169].

4.4. Other SERCA Inhibitors

The attempts to design agents with SERCA-targeted, ERS-inducible properties but low toxicity continues. A few compounds of different chemical natures (named by the authors as compounds 7, 13, 40, 42, etc.) [129,130], as well as the small inhibitors CKP1 and CKP2 with high SERCA1 binding affinity but lower cardiac toxicity than TG [170], augmented apoptosis in patient-derived PTC YUMC cells, including chemotherapy-resistant lines, in the presence of paclitaxel, sorafenib or lenvatinib, and reduced the growth of mouse xenograft originated from YUMC stem-like cells. Four small molecules were lately proposed as potential SERCA1 inhibitors for colorectal carcinoma cells [31]. The natural arylnaphthalide lignan diphyllin, a blocker of vacuolar H+-ATPase, was also recently tested as a SERCA2 inhibitor. In NSCLC cells, diphyllin promoted ERS and apoptosis by directly suppressing SERCA2 activity, leading to ER Ca2+ depletion, cytoplasmic and mitochondrial Ca2+ accumulation, increased ROS production, a decreased mitochondrial membrane potential, and cytochrome C release [171]. Moreover, it exerted synergistic anti-tumor effects in combination with cisplatin in vitro and in vivo. A few diphyllin derivatives with longer half-lives, better bioavailability and metabolic stability more effectively blocked autophagy and induced apoptosis in pancreatic cancer [172] and HNSCC cells and xenografts [173].
Table 3. Examples of SERCA inhibitors used for the induction of unresolved ERS in cancer cells.
Table 3. Examples of SERCA inhibitors used for the induction of unresolved ERS in cancer cells.
Cell TypeTreatmentMolecular ChangesCellular EffectsReferences
Terpenoids
Breast cancer MCF7 and MDA-MB-231 cellsTG 6–100 nM
6–48 h
↓ ER Ca2+, ↑ROS
↑ Cleaved PARP
↑ Caspases-8, 9, 3
↑ Cells in subG1 phase
↓ Proliferation
↓ Viability, ↑ Apoptosis
[118,119]
Breast cancer cells MDA-MB-231 and MDA-MB-436TG 2–10 µM
6 or 24 h
↑ Cytoplasmic Ca2+
↑ p-Myosin light chain 2 kinase
↑ p-Myosin phosphatase 1
Actin contraction and rearrangement
Changed morphology
[121]
Prostate cancer LNCaP, PC3 cellsTG 30–100 nM
6–48 h
↓ ER Ca2+, ↑ GRP94, ↑ GRP78,
↑ ATF4, ↑ cleaved PARP, ↑ CHOP
↓ Proliferation, ↑Death Changed morphology[118]
Transfected prostate cancer LNCaP cellsTG 100 nM
30–48 h
↑ DR5, ↑ PERK, ↑ ATF4, ↑ CHOP
↑ IRE1, ↑ XBP1, ↑ JNK
↑ Cleaved PARP, ↑ Caspases-3, 8
↑ Cell death[122]
Patient-derived stem cell-enriched glioblastoma
culture
TG 1–10 µM
24–48 h
↑ p-PERK, ↑ ATF4, ↑ CHOP, ↑GRP78, ↑ sXBP1, ↑ ATF6,
↓ SOX2,
↑ Cleaved PARP, ↑ Caspases-3/7
↓ Viability
↓ Neurosphere-forming ability
[123]
Neuroblastoma SH-SY5Y cellsTG 300 nM
30 min or 4 h
↓ ER Ca2+, ↑ ROS
↑ Hypodiploid nuclei, ↑ GRP78, ↑ ATF4, ↑ p-PERK, ↑ Caspase-4
↓ Viability[124]
ACC SW-13 and NCI-H295R cellsTG 0.5–32 µM
48 h
↑ p-JNK/JNK, ↑ PERK, ↑ ATF6,
↑ LC3B, ↑ HSAP, ↑ Bcl-2
↓ Viability, ↑ Apoptosis, ↓ Migration, invasion[125]
Mice with SW-13 cell xenograftsTG 1 mg/kg
14 days
↑ p-JNK/JNK, ↑ p-ERK/ERK,
↑ p-PERK/PERK, ↑ GRP78, ↑ IRE1
↓ Tumor growth[125]
Patient-derived PTC YUMC cells resistant to PTX, SOR, and LENTG 10–200 µM + PTX, or SOR, or LEN
10–200 µM 40 h
↑ CHOP, ↑p-PERK
↑ Cytochrome c
↑ Cleaved caspase-3
↑ Sensitivity to PTX, SOR or LEN
↓ Viability
[129,130]
Mice with PTC YUMC xenografts resistant
to PTX, SOR, and LEN
TG 25 mg/kg PO + PTX 25 mg/kg IP, or SOR 80 mg/kg PO, or LEN 10 mg/kg PO↑ CHOP↓ Tumor weight
↑ Sensitivity to PTX, SOR or LEN
[129,130]
Oral cancer CAL 27 and Ca9–22 cellsTG 10 nM + Manoalide 5–10 µM
24 h
↑ Caspase 3/7↓ Viability, ↑ Autophagy
↑ Sensitivity to manoalide
[131]
Breast cancer MCF7 and MDA-MB-231s cellsTG 3 µM + nodakenin 40 µM 24 h↑ p-PERK
↑ p-eIF2α
↑ CHOP, ↑ ATF4
↑ Sensitivity to nodakenin
↑ Cell death
[132]
Hepatocellular carcinomaG-202 40 mg on 1–3 d, or 40 mg on 1 d and 66.8 mg on 2–3 d of 28-d cycle No complete response
No progressive disease
Stable disease
Partial response
[135,136]
Glioblastoma multiformeG-202 IV for 3 days of 28-d cycle No clear conclusions[137]
Lung cancer A549
and H460 cells
Lathyrol 30–120 µg/mL 24 h or 14 d↑ Cytosolic Ca2+, ↑ GRP78, ↑ PERK
↑ p-eIF2α, ↑ CHOP, ↑ ATF4, ↑ Bax,
↓ Bcl-2, ↑ Caspase-3, ↑ Cyt C
↓ Viability, ↑ Apoptosis[138]
Mice with H460
cell xenografts
Lathyrol 10–40 mg/kg IP 16 d ↓ Tumor volume and weight[138]
Renal cell carcinoma
786-O cells
Lathyrol 10–375
µg/mL 24 h
↓ Bcl-2, ↑ Bax, ↓ p-Akt, ↓ MMP2,
↓ MMP9, ↓ Ki67, ↑ Caspase-3, 9
↓ Viability, ↓ Invasion,
↓ Migration, ↑ Apoptosis
[139]
Curcumin and its analogues
Human liposarcoma
SW872 cells
Curcumin 5–20 µM
24 or 48 h
↓ Ca2+-ATPase activity, ↑ DR5,
↑ Caspase-8, ↑ Caspase-3, ↑ Bid,
↑ PARP, ↑ CHOP, ↑ p-eIF2a, ↑ ATF4
↓ Cell growth
↑ Apoptosis
[126]
SCID mice injected with SW872 cellsCurcumin 100 mg/kg IP 40 d↑ Caspase-8, ↑ Caspase-3,
↑ Cleaved PARP, ↑ CHOP,
↓ Tumor growth[126]
Human glioma
LN229 and U87 cells
Curcumin 8–32 μM
24–72 h
↓ Cyclin D1, ↓ CDK46/6
↓ Bcl-2, ↓ Bcl-XL, ↓ MMP-2,
↓ MMP-9, ↓ p-ERK1/2
↓ Proliferation
↓ Migration and invasion
[148]
Mice with subcutaneous LN229 xenograftsCurcumin 60 mg/kg/d 4 w↓ MMP-2 and MMP-9
↓ CD147
↓ Tumor growth
[148]
CIS-resistant
NSCLC A549
and H1299 cells
Curcumin 2.5 μg/mL
+ CIS 2 μg/mL
48 h
↑ Cleaved PARP, ↑ Caspase-3
↑ GRP78, ↑ ATF6, ↑ XBP1
↑ Caspase-4, ↑ CHOP
↓ Viability
↑ Apoptosis
↑ Sensitivity to CIS
[149]
Glioblastoma U87
and LN18 cells
Curcumin 10 µg/mL
+ TMZ 200 µM
Alterations in
actin network
Cell cycle arrest,
↓ Viability, ↑ Apoptosis,
↑ Sensitivity to TMZ
[150]
Colon carcinoma SW480 cellsF36 1–10 µM
24–72 h
↑ Cleaved PARP, ↑ Caspase-3,
↑ CHOP, ↑ ATF4, ↑ p-eIF2a
↓ Proliferation
↑ Apoptosis
[33]
Advanced
metastatic
breast cancer
Curcumin 6 g (7 d) + DOC 100 mg/m2 every 3 w
6 cycles
No significant difference in the objective response rate and 12-month overall survival[153]
Colon carcinoma SW480 cellsRL71 0.5–10 µM
24–72 h
↑ GRP78, ↑ ATF4, ↑ CHOP,
↑ cleaved PARP
↓ Viability
G2/M cell cycle arrest
[155]
Mice with SW480
xenografts
RL71 1–4 mg/kg
14 d
↑ Cleaved PARP, ↑ CHOP,
↑ cleaved Caspase-3
↓ Tumor growth[155]
Flavonoids
HCC SNU-449
and Hep-3B cells
Quercetin 6.5–75 µM 24–48 h↓ p-PI3K, ↓ p-Akt, ↓ p-mTOR,
↓ Bcl-2, ↑ Bax, ↑ cleaved PARP, ↑ cleaved Caspase-3, ↓ P4HA2
↓ Viability, ↑ Apoptosis, ↓ Proliferation, ↓ Colony formation[159]
AML HL-60 cellsQuercetin 25–100 µM 24–72 h↑ LC3II/I ↓ Bcl-2 ↑ Bax
↑ p-AMPK ↓ p-mTOR ↑ Caspase-3
↓ Viability, ↑ Apoptosis, ↑ Autophagy,
↓ Colony formation
[160]
Melanoma
A375 cells
Quercetin 1–100 µM 24–72 h↑ p-ERK, ↑ p-Akt,
↑ GPER, ↑ c-Myc
↓ Viability,
Cell cycle arrest, Changed morphology,
↑ Apoptosis/necrosis
[161]
NSCLC A549
and H1299 cells
Luteolin 0.1–1000 µM 12–72 h
or 50 µM 24 h
↓ WDR72, ↓ Bcl-2, ↑ Caspase-3,
↓ p-Akt, ↓ E-cadherin, ↓ β-catenin,
↓ N-cadherin, ↓ ZEB1
↓ Viability, ↓ Migration and invasion,
↓ Proliferation
[163]
Mice with NSCLC
H1299 xenografts
Luteolin 50 mg/kg IP once/2 d 21 d↓ WDR72 mRNA↓ Tumor growth[163]
Bladder cancer
EJ138 cells
Luteolin 20–50 µM
24–48 h
↑ P53, ↑ ULK1,
↑ ATG12, ↓ BCL2
↓ Viability, ↑ Apoptosis G2/M phase arrest[164]
DLBCL U2932
and OCI-LY10 cells
Luteolin 5–20 µM
24 h
↑ Bax, ↓ Bcl-2, ↑ cleaved PARP, ↑ Caspase-3, ↓ p-JAK2, ↓ p-STAT3↑ Apoptosis[165]
Mice with U2932
tumors
Luteolin 12.5–50 mg/kg IP 14 d↑ Bax, ↓ Bcl-2, ↑ cleaved PARP, ↑ cleaved Caspase-3,
↓ p-JAK2, ↓ p-STAT3
↓ Tumor volume
and weight
[165]
Other SERCA inhibitors
Patient-derived
PTX-, SOR-, or LEN-
resistant PTC
YUMC cells
Compounds 7, 13, 40, 42 (10–200 µM) + PTX, SOR, LEN 10–200 µM 40 h↑ p-PERK, ↑ CHOP,
↑ Cytochrome c,
↑ Cleaved caspase-3
↑ Sensitivity to PTX, SOR, or LEN
↓ Viability
[129,130]
Mice with xenografts of PTX-, SOR-, or LEN-sensitive and
-resistant YUMC PTC cells
Compounds 7, 13, 40, 42 (25 mg/kg) + PTX 25 mg/kg, or SOR 80 mg/kg, or LEN 10 mg/kg ↓ Tumor weight
↑ Sensitivity to PTX, SOR, or LEN
[129,130]
NSCLCDiphyllin↓ Ca2+ levels in the ER,
↑ ROS, ↓ MMP,
Cytochrome C release
↓ Proliferation,
↓ Migration, ↑ Apoptois,
Synergy with CIS
[171]
Abbreviations: ACC—adrenocortical carcinoma, PTC—papillary thyroid cancer, NSCLC—non-small cell lung cancer, HCC—hepatocellular carcinoma, AML—acute myeloid leukemia, DLBCL—diffuse large B-cell lymphoma. PTX—paclitaxel, LEN—lenvatinib, SOR—sorafenib, TMZ—temozolomide, DOC—docetaxel, CIS—cisplatin, PO—per oral, PI—intravenous, ↑—increase, ↓—suppression.

5. UPR Modulators

5.1. PERK Inhibitors

GSK2606414 is a highly potent, synthetic first-generation PERK inhibitor that competitively targets its ATP-binding site [174]. As a single agent, GSK2606414 suppressed the proliferation of PDAC SW1990 cells with a high level of the tumorigenic BZW1 protein (basic leucine zipper and W2 domain-containing protein 1) and the growth of SW1990 xenografts [175] (Table 4). The application of GSK2606414 with the autophagy inhibitor simvastatin sensitizes glioblastoma U87 and U251 cells to temozolomide, although it differentially affected autophagosome formation (LC3β-II/LC3β-I ratio) [176]. The combination of GSK2606414 and irradiation led to sustained ERS followed by apoptosis in glioblastoma cells in vitro, and enhanced the overall survival of mice with orthotopic glioblastoma by preventing tumor recurrence [177]. PERK suppression sensitized multidrug-resistant colorectal cancer S1-M1–80 cells overexpressing the cancer resistance protein ABCG2 to doxorubicin and mitoxantrone [178]. As a single agent or in combination with bortezomib, GSK2606414 triggered apoptosis in multiple human myeloma cells [179]. Among the other described effects of GSK2606414 is synergistic efficacy with tumor-selective oncolytic reovirus type 3 Dearing in HNSCC HN5 and FaDu cells [180], an ability to reduce the volume of subcutaneous HN5 xenografts in mice treated with doxycycline and the reovirus [180], the suppression of the phenotypic transition of cancer-associated fibroblasts in pancreatic cancer [181], synergistic anti-tumor effects with digoxin on human leukemia cells K562 and THP-1 [182], the inhibition of PERK/eIF2α/CHOP pathway leading to accelerated apoptosis in small-cell lung cancer (SCLC) H1688 and H446 cells [183], and enhanced therapeutic responses of mice SCLC xenografts to the natural compound oridonin [183].
GSK2656157 is another highly selective ATP-competitive PERK blocker [184]. In pancreatic BxPC3 cells stressed by tunicamycin or TG, GSK2656157 suppressed PERK/eIF2 phosphorylation, decreased ATF4 and CHOP levels, and changed the expression of UPR-associated genes, while its oral administration inhibited the growth of pancreatic and multiple myeloma xenografts in mice accompanied by impaired tumor angiogenesis [185]. The growth of esophageal squamous cell carcinoma cells was also sensitive to GSK2656157 application [186]. However, an addition of GSK2656157 did not sensitize mice with subcutaneous leukemic K562 cell xenografts to imatinib [187]. Additionally, despite its anti-tumor activity, GSK2656157 exerts a serious off-tumor side effects associated with the inhibition of PERK activity in the pancreas, which leads to impaired pancreatic function [185].
Other PERK inhibitors. In the search for less toxic PERK inhibitors, a few other agents have been examined. The compound NCI 159,456 decreased the viability and increased the death of normal and ER-stressed non-small cell lung cancer A549 cells, which was accompanied by excessive ROS production, DNA damage, and increased mRNA levels of pro-apoptotic genes [188]. Cancer-associated PERK activity in rhabdomyosarcoma cell lines was diminished by the new PERK inhibitor AMGEN44 [189].

5.2. eIF2α Inhibitors

Since eIF2α phosphorylation can have pro-survival or apoptotic outcomes, both the suppression and stimulation of its activity have been proposed as potential strategies in anti-cancer therapy.
ISRIB. Integrated stress response inhibitor (ISRIB) is a small synthetic drug-like molecule that stabilizes eIF2B by binding its subunits, thus reversing eIF2α phosphorylation [190]. In spite of its insolubility, it showed a good safety profile and promising results in in vitro experiments and preclinical animal tumor models. ISRIB effectively decreased the growth and metastases of xenografts originating from patient-derived aggressive metastatic prostate cancer [64], lung tumor [191], and pancreatic duct adenocarcinoma SW 1990 cells with high level of BZW1 in mice [175], thus increasing the overall animal survival. It suppressed the migration and invasion of triple-negative breast cancer Hs576T and MDA-MB-231 cells overexpressing ETHE1 (ethylmalonic encephalopathy protein 1), which upregulates eIF2α phosphorylation [192], and diminished the growth and metastatic potential of subcutaneous MDA-MB-231 xenografts in mice either as a single agent or in combination with doxorubicin [193]. ISRIB and imatinib suppressed the proliferation of TG-stressed leukemic K562 and LAMA84 cells, whereas the administration of ISRIB to mice bearing K562 xenografts enhanced the sensitivity of the tumors to imatinib [187]. Unfortunately, in spite of its scientific value, the therapeutic potential of ISRIB in clinical trials, as well as its effectiveness, safety, and side effects, were not evaluated consistently [194].
Salubrinal. Salubrinal is an example of synthetic phosphatase inhibitor that specifically blocks eIF2α dephosphorylation by the complexes GADD34/PP1 and CREP/PP1, thus inducing sustained eIF2α hyperphosphorylation and eventually cell death [195,196]. It suppressed the proliferation of inflammatory breast cancer SUM149PT and SUM190PT cells by enhancing ROS production and changing the expression of ERS genes/proteins [197]. In adrenocortical carcinoma SW-13 and NCI-H295R cells, salubrinal increased the cytosolic Ca2+ content and upregulated components of the PERK/eIF2α/ATF4 and apoptosis signaling pathways [198]. A combination of salubrinal and ionizing radiation was proposed as a novel approach for the treatment of pediatric glioblastoma [199]. By disrupting the cell cycle and dephosphorylating cyclin A and cyclin-dependent kinases, salubrinal impaired the viability and clonogenic capacity of patient-derived and cultured (SCC4 and FaDu) head and neck squamous cells carcinomas, while in FaDu cells, it exerted synergistic or additive effects with chemotherapeutic drugs [200]. However, it did not enhance the sensitivity of glioblastoma U87 and U251 cells to the combination of temozolomide and simvastatin, although it promoted apoptosis as a single agent [176]. Excessive ROS production and enhanced death via the upregulation of xCT (SLC7A11), an antiporter of cystine and glutamate, was observed in breast, gastric, and oral squamous cell carcinoma cells treated with salubrinal under conditions of glucose deprivation [201]. Although ineffective as a single treatment, salubrinal in combination with 4E1RCat, an inhibitor of cap-dependent translation, synergistically suppressed the viability of melanoma UACC 903 cells and the development of subcutaneous UACC 903 xenograft tumors [202]. In triple-negative breast cancer BT549, SUM159 and MCF-10A cells, salubrinal potentiated the cytotoxicity and apoptosis induced by silver nanoparticles, AgNPs [203].

5.3. IRE1α/XBP1 Inhibitors

IRE1α inhibitors are classified as kinase (ATP-binding) domain and RNase domain blockers. The kinase inhibitors in turn include ATP-competitive agents that are able to stimulate the RNase domain, leading to XBP1 mRNA splicing (type I), and the compounds that inactivate the RNase (type II) [8,204]. The examples of type I IRE1α kinase inhibitors are APY29 and sunitinib, and the type II IRE1α kinase blockers are KIRAs (kinase inhibiting RNase attenuators) [204,205,206]. IRE1α RNase inhibitors include MKC-3946, MKC8866, 4µ8c, STF-83010, toyocamycin, salicylaldehydes, and hydroxyl-aryl-aldehydes [207,208,209,210]. They suppress XBP1 splicing, resulting in unresolved ER stress.
Sunitinib. Sunitinib, a synthetic indolinone derivative, has been approved for the standard first-line therapy of metastatic renal cell carcinoma (RCC) [211], and was examined as a part of combination therapy for ovarian [212] and breast [213] cancers. However, sunitinib is a multi-kinase inhibitor that also targets, for example, VEGF and PDGF receptors. Thus, although sunitinib alone caused a similar level of apoptosis to gemcitabine in PDAC cells and a synergized chemotherapeutic effect on mice with orthotopic PDAC xenografts, it did not suppress XBP1 splicing, which indicates an absence of its direct effect on IRE1α activity [100]. Additionally, in some cases, it showed poor tolerance alone [213]. Other significant obstacles are the development of resistance and lower efficacy than that of other drugs. For instance, the combined application of lenvatinib + pembrolizumab as the first-line treatment for patients with advanced RCC had higher therapeutic effect versus sunitinib as a standard [214].
KIRAs. The blockage of XBP1 splicing with KIRA6 led to apoptosis or suppressed the proliferation of cultured mast cell leukemia [215]. A more selective compound, 18/KIRA8, reduced the viability of multiple myeloma and non-myeloma cancer cell lines, diminished the growth of subcutaneous or orthometastatic myeloma xenograft tumors in mice, and potentiated the efficacy of the cytotoxic agents bortezomib and lenalidomide [216]. It also reversed the sensitivity of pancreatic tumor to radiotherapy [217].
MKC-3946. MKC-3946 exerted modest cytotoxicity toward multiple myeloma cells under in vitro conditions, but significantly suppressed the growth of subcutaneous myeloma in mice by inhibiting XBP1 splicing and enhancing the apoptotic effects of the anti-cancer drugs bortezomib and 17-AAG [207]. The cytotoxicity of MKC-3946 toward acute myeloid leukemia cells was also associated with the blockade of XBP1 mRNA splicing [83]. In stable (U87MG and A172) and patient-derived glioblastoma cells, MKC-3946 reduced the viability and colony formation, and potentiated the effect of temozolomide, particularly in cells with a methylated O6-methylguanine-DNA methyl transferase gene promoter [76].
MKC8866. MKC8866 (ORIN1001) is the only IRE1α inhibitor that entered clinical trials as an agent enhancing the effects of chemical/targeted therapy of advanced solid tumors [218]. In experiments, MKC8866 suppressed the proliferation and colony formation of mast leukemia HMC-1.2 [215] and rhabdomyosarcoma [189] cells. In prostate cancer cells and mouse xenograft models, MKC8866 was effective alone and exhibited additive or synergistic effects with antiandrogens (abiraterone acetate and enzalutamide) and taxanes (cabazitaxel and paclitaxel) [81]. It also increased the responsiveness of a mouse model of prostate cancer to anti-PD-1 antibody therapy [71]. In a series of breast cancer cells, MKC8866 treatment reduced the production of the pro-inflammatory factors IL-6, IL-8, CXCL1, GM-CSF, and TGFβ2, and enhanced the effectiveness of paclitaxel both in vitro and in vivo [219]. It increased the sensitivity of mice with intracerebral glioblastoma GL261-Luc to combined treatment with irradiation and temozolomide [220], and exerted synergistic effect with the tyrosine kinase inhibitor nilotinib on acute lymphoblastic leukemia [221] and with AZD1775 (an inhibitor of the WEE1 G2 checkpoint kinase) in ovarian cancer cells [222]. However, MKC8866 did not alter the rate of temozolomide + simvastatin-induced death of glioblastoma U87 and U251 cells, although it reduced the autophagic flux triggered by these drugs and decreased cell viability as a single agent [176].
4μ8C. This compound significantly inhibited the colony-forming ability of aggressive luminal B breast cancer SUM52 cells [79], suppressed the proliferation and migration of hepatocellular carcinoma HepG2 or Huh7 cells in vitro, and reduced fibrotic diethylnitrosamine-induced HCC tumor and collagen deposition in mice [223]. In both HCC xenograft and in vitro HCC models (HepG2, SNU449, Huh7 cells, and patient-derived organoids), 4μ8C synergized with doxorubicin’s effects by altering lipid metabolism and the oxygen consumption rate, and downregulated ERS markers [224]. It was effective at suppressing the proliferation of colon cancer stem cells in vitro and enhanced the efficacy of 5-FU chemotherapy in mice bearing 5-FU-resistant colon cancer [225,226].
STF-083010. The IRE1α RNase-specific inhibitor, STF-083010, induced apoptosis and blocked the growth of mast cell leukemia [215], and increased miR-34a expression in a few AML cell lines [83]. It suppressed the proliferation of a series of pancreatic cancer cell lines by increasing the expression of apoptotic proteins and arresting the cell cycle at G1 or G2/M phases, and potentiated the effect of bortezomib [227]. In tunicamycin-stressed PDAC cells, STF-083010 increased the number of mature autophagy-associated lysosomes and had additive effect with gemcitabine [100]. It restored the sensitivity of breast cancer MCF7 cells to tamoxifen in vitro, while both drugs synergistically suppressed the growth of mammary tumors in mice [80]. However, although STF-083010 showed anti-tumor effects in experimental settings, it is unstable in organisms and its application in clinical trials is under question.
Other IRE1α RNase inhibitors. HNA (2-hydroxy-1-naphthaldehyde), toyocamycin (an adenosine analog produced by Actinomycetes), and 3ETH (3-ethoxy-5,6-dibromosalicylaldehyde) suppressed XBP1 mRNA splicing in acute myeloid leukemia cells, followed by caspase-dependent apoptosis, G1 cell cycle arrest, and altered expression of chaperones, Bcl-2 family, G1 phase-controlling proteins, and miR-34a, whereas its synergic effect with bortezomib was linked with p-JNK and p-PI3K [83]. The blockade of tunicamycin-induced XBP1 splicing in pancreatic cancer cells by these compounds reduced proliferation and the colony-forming ability, arrested the cell cycle at G1 or G2/M phases, enhanced the expression of apoptotic proteins, decreased the mitochondrial membrane potential, and potentiated the anti-tumor activity of bortezomib in mice with pancreatic cancer xenograft [227]. The urea-based compound Z4P was shown to diminish the migratory capacity of glioblastoma cells (U251, RADH85, and RADH87), arrested IRE1 phosphorylation in U87 cells, reduced the burden of xenograft tumors in mice, prolonged relapse-free survival, and sensitized glioblastoma to temozolomide [228].

5.4. ATF6 Inhibitors

The inhibitors of ATF6 signaling are rare. Two decades ago, a serine protease inhibitor AEBSF (4-(2-aminoethyl) benzenesulfonyl fluoride) was shown to prevent the cleavage of ATF6α/ATF6β, thus suppressing the transcription of ATF6 target genes [229]. However, the only highly specific inhibitors of ATF6α developed to date are the pyrazole amides ceapins [230]. Ceapin-A7 impaired the survival of castration-resistance prostate cancer (CRPC) cells [231] and sensitized the radioresistant pancreatic cancer cells to radiotherapy by increasing apoptosis and G1 cell cycle arrest [217].

5.5. GRP78 Inhibitors

YUM70. YUM70 is a newly discovered derivative of 8-hydroxyquinoline that inhibits GRP78 enzymatic activity by direct binding. Such ability impairs the potency of GRP78 to correct misfolded proteins and adapt to ERS. As a monotherapy, YUM70 activated ERS-associated apoptosis, suppressed the growth of pancreatic cancer cells in vitro, and reduced the subcutaneous pancreatic PaCa-2 tumors in mice without toxicity to other organs, while when administered in combination with the cytotoxic drugs topotecan, vorinostat, MG132, and 5-FU, it had synergic or additive effects on the clonogenic capacity of cells [232]. It efficiently inhibited oncogenic KRAS expression in a panel of human lung, colon, and pancreatic cancer cell lines both in vitro and in vivo [233]. The addition of YUM70 reversed the sensitivity of cisplatin-resistant HNSCC to chemotherapy by reducing their viability and clonogenic capacity [234]. The treatment of HNSCC (SCC25 and SCC15) cells, their cisplatin-resistant clones, TNBC (MDA-MB-231), and PDAC (MIA PaCa-2) cells with YUM70 diminished the expression of oncogenic protein c-Myc, which led to apoptosis, and suppressed the growth of pancreatic xenografts [235].
HA15. Another potent and specific GRP78 inhibitor is thiazole benzenesulfonamide HA15, which suppresses its ATPase activity by promoting conformational changes. It triggered early ERS followed by apoptosis and autophagy in a series of melanoma cell lines, including patient-derived melanocytes resistant to BRAF inhibitors, and suppressed the growth of BRAF-resistant melanoma xenografts in mice [236]. In adrenocortical carcinoma H295R cells, HA15 suppressed proliferation and steroidogenesis [237]. Its ability to reduce the stemness led to the apoptosis of colorectal cancer HCT116 and HT29 cells and impaired formation of tumor spheres [238], while in a few lung, colon, and pancreatic cancer cell lines, it diminished oncogenic KRAS expression [233]. HA15-induced apoptosis in lung cancer cells was accompanied by the formation of autophagosomes with increased expression of apoptosis and autophagy genes [101]. The pro-apoptotic effects of HA15 in NSCLC HCC827-GR, H1993-GR and H1993-ER cells with acquired resistance to therapeutic EGFR tyrosine kinase inhibitors included enhanced ROS production [239]. Similar to YUM70, HA15 reduced c-Myc expression and upregulated the eukaryotic translation inhibitor 4E-BP1 in multiple c-Myc-dependent 2D cell culture, 3D spheroid, and xenograft cancer models [235].
Epigallocatechin gallate. The compound (−)-Epigallocatechin-3-gallate (EGCG) is one of the polyphenol flavonoids found in green tea. It is believed to directly interact with GRP78 at its ATP-binding site, thus inducing conformational changes and preventing the formation of the anti-apoptotic GRP78/caspase-7 complex [240]. EGCG has been intensively studied as a part of preventive therapy for various types of malignancies, including gliomas [98,241], breast [240,242], adrenal [243], and colorectal [244] cancers. Although poorly effective as a single agent, it sensitized breast cancer cells to etoposide [240] and paclitaxel in vitro and in vivo by enhancing JNK phosphorylation [242]. The increased sensitivity of glioma cells to temozolomide in the presence of EGCG included the activation of CHOP and caspase-7 [98]. Although EGCG did not improve the survival of mice with intracranially implanted U87 (p53 wild type) or U251 (p53 mutant) glioblastoma cells, it significantly enhanced the effect of temozolomide in combination therapy [245]. However, the cellular effects of EGCG are variable and cannot be attributed solely to GRP78 suppression.
Table 4. Examples of using inhibitors of UPR effectors in experimental cancer treatment.
Table 4. Examples of using inhibitors of UPR effectors in experimental cancer treatment.
Cell TypeTreatmentMolecular ChangesCellular EffectsReferences
PERK inhibitors
PDAC SW1990 cells with ↑ BZW1GSK2606414
10 µM 12 h
↓ p-eIF2α, ↓ HIF1α, ↓ c-Myc, ↓ HIF1A, ↓ MYC IRES↓ Cell survival
↓ Proliferation
[175]
Mice with PDAC SW1990 BZW1 xenograftsGSK2606414 100 mg/kg
IP twice/w
↓ Ki67 staining
↓ TUNEL staining
↓ Tumor growth
↓ Cell proliferation
↑ Apoptosis
[175]
Glioblastoma U87
and U251 cells
GSK2606414
1–20 µM + simvastatin
+ TMZ 72 h
↑ p62, ↓ p-eIF2α,
↓ LC3B-II/I in U87 cells,
↑ LC3B-II/I in U251 cells
↓ Viability
↑ Sensitivity to simvastatin+TMZ
[176]
Multidrug-resistant colorectal cancer
S1-M1–80 cells
GSK2606414 1–3 µM + mitoxantrone or DOX
10 µM 24–72 h
↑ Sensitivity to mitoxantrone
and DOX
[178]
Human myeloma L363, H929, U266,
and KMS11 cells
GSK2606414 1–100 µM
or 10 µM +
BTZ 4 nM 24–48 h
↓ PERK, ↓ ATF4, ↓ eIF2α,
Changes in the expression
of UPR genes
↓ Cell survival
↑ Apoptosis
↑ Sensitivity to BTZ
[179]
SCLC H1688
and H446 cells
GSK2606414 10 µM
+ Oridonin 20 µM 24 h
↓ p62
↓ LC3B-II/LC3B-I
↑ Apoptosis, ↑ Oridonin effect, ↑ Autophagy[183]
Mice with SCLC H1688
cell xenografts
GSK2606414
50 mg/kg +
oridonin 10 mg/kg
↓ GRP78, ↓ p-PERK,
↓ p-eIF2α, ↓ ATF4, ↓ CHOP
↓ Tumor size[183]
Himan pancreatic adenocarcinoma BxPC3 cellsGSK2656157
1 µM + tunicamycin
or TG 6 h
↓ p-PERK, ↓ ATF4,
↓ p-eIF2α, ↓ CHOP,
↓ UPR gene expression
[185]
Mice with pancreatic
cancer xenografts
GSK2656157 50 or 150 mg/kg twice/d OR
14 d
↓ p-PERK, changes in
genes expression
↓ Tumor growth
↓ Blood vessel density
[185]
Myeloid leukemia K562 and LAMA 84 cellsGSK2656157 0.1–10 µM + TG 100 nM + IMA 1 μM
16 h
↓ CHOP mRNA
↓ GADD34 mRNA
[187]
Mice subcutaneous K562 xenograftGSK2656157
20 mg/kg/d + IMA 50 mg/kg twice/d 2 w
No significant decrease in the tumor mass[187]
Intact and ER-stressed NSCLC A549 linesNCI 159456
3–100 or 50 µM +
TG 500 nM 24 h
DNA damage, ↑ ATF4,
DDTI3, ↑ BAX, ↓ BCL2, ↑ Caspase-3, ↑ ROS
↓ Viability
↑ Apoptosis
[188]
eIF2 inhibitors
Mice with prostate
cancer xenografts
ISRIB 10 mg/kg, 6 w ↓ Tumor growth,
↓ Metastases, ↑ Survival
[64]
Mice with PDAC SW1990 BZW1 cell xenograftsISRIB 2.5 mg/kg
IP twice/w
↓ Tumor volume
↑ Animal survival
[175]
TNBC Hs576T and MDA-MB-231/ETHE1 cellsISRIB 200 nM
24 h
↓ p-eIF2
↓ ATF4
↓ Migration
↓ Invasion
[192]
Mice with subcutaneous TNBC MDA-MB-231 cell xenograftsISRIB for 15 d +
DOX 6 injections
↑ Cleaved caspase-3↓ Tumor volume
↓ Tumor weight
↑ Sensitivity to DOX
[193]
Human ML K562
and LAMA84 cells
ISRIB 250 nM +
IMA 0.5–1 µM 16 h
↓ p-STAT,
↓ m-TOR, ↓ p-GSK3
↓ Proliferation[187]
Mice with subcutaneous K562 xenograftsISRIB 2 mg/kg/d +
IMA 100 mg/kg/d 2 w
↑ Sensitivity to IMA
↓ Tumor mass
[187]
Inflammatory breast cancer SUM149PT
and SUM190PT cells
Salubrinal 10 µM
24–48 h
↑ p-eIF2a, ↓ PERK,
↑ CHOP, ↓ GRP78, ↑ ATF4,
↑ ROS, ↑ Bax, ↑ cleaved PARP, ↑ Caspase-3,
↓ p-Akt, ↓ p-NFkB
↓ Proliferation[197]
ACC SW-13 and
NCI–H295R cells
Salubrinal 100 µM
24 h
↑ p-eIF2α, ↑ p-PERK,
↑ ATF4, ↑ Ca2+, ↑Bcl-2
↓ Viability, migration
↑ Apoptosis
[198]
Primary pediatric GB SU-DIPG
and KNS-42 lines
Salubrinal 2.5–8 µM + irradiation↑ p-eIF2a↑ Sensitivity to irradiation
↓ Cell survival
[199]
HNSCC SCC4 and FaDu cells, patient-derived 3D spheresSalubrinal 10–50 µM
24–72 h
↑ p-eIF2a, ↓ p-RB1,
↓ E2F1, ↓ Cyclin A,
↑ p21
↓ Viability
↓ Clonogenic ability
Cell cycle arrest
[200]
Glioblastoma U87
and U251 cells
Salubrinal 1–20 µM or 15 µM + simvastatin + temozolomide 72 h↑ p-eIF2α↓ Viability
No synergistic effect with cytotoxic drugs
[176]
Melanoma
UACC 903 cells
Salubrinal 40 µM +
4E1RCat 10 µM 48 h
↓ Protein synthesis, ↓ Cyclins, ↓ CDK2, ↓ Polysomes↓ Cell cycle progression
↓ Viability
[202]
Mice with subcutaneous UACC 903
melanoma
Salubrinal 1 mg/kg + 4E1RCat 2.5–15 mg/kg I
P one/2 d 3–4 w
↓Tumor volume[202]
TNBS BT549, SUM159, and MCF-10A cellsSalubrinal 5–10 µM
+ AgNPs 24 h
↑ p-eIF2a, ↑ CHOP,
↑ cleaved Caspases-3/7/9
↓ Viability
↑ AgN-induced death
[203]
IRE1 inhibitors
PDAC cells Panc3.27, Pan02, Miapaca-2Sunitinib 10 µM +
GEM 100–250 nM
48–72 h
↓ Lysosomal degradation,
DNA fragmentation
↓ Viability, ↑ Apoptosis,
↓ Autophagy,
↑ GEM effect
[100]
Mice with orthotopic
PDAC Pan02 or KPCP1 xenografts
Sunitinib 25 mg/kg/d OR + GEM 25 mg/kg/w IP + PTX 10 mg/kg/w IP until mortality or for 4 w↓ Ki67-positive cells
↓ TUNEL-positive cells
↓ GRP78 immunosignal
↓ Tumor growth
↑ Overall survival
↑ Chemotherapy effect
[100]
Myeloma patient-derived INA6 and
RPMI 8226 cells
MKC-3946 10 µM + BTZ 2.5–10 nM or 17-AAG
125–1000 nM 2–24 h
↓ XBP1s, ↑ CHOP, ↑ ATF4,
↑ p-eIF2α, ↑ Caspase-3, ↑ cleaved PARP
↑ Growth inhibition
↑ Apoptosis
↑ BTZ, 17-AAG effect
[207]
Mice with subcutaneous RPMI 8226 myelomaMKC-3946 100 mg/kg/d IP + BTZ 0.15 mg/kg IV 2/w 21 d↓ XBP1s
↑ CHOP mRNA
↓ Tumor growth
↑ Overall survival
[207]
GBM patient-derived U87MG, A172, BAH1 TMZ-resistant cellsMKC-3496 10 μM +
TMZ 50 μM 24–72 h
↓ sXBP1 mRNA↓ Colony formation
↓ Viability
↑ Efficacy of TMZ
[76]
Mouse prostate
cancer LNCaP,
VCap, 22Rv1, and C4–2B
cells xenograft models
MKC8866 300 mg/kg/d
or once/2 d OR + enzalutamide, abiraterone acetate, cabazitaxel, PTX
↓ sXBP1
↑ Cleaved Caspase-3
↓ PCNA
↓ Tumor growth
↓ Proliferation,
Additive/synergic effects with anti-cancer drugs, ↑ apoptosis
[81]
Mice with subcutaneous Myc-CaP prostate cancer xenograftsMKC8866 150–300 mg/kg/2 d + anti-PD-1 10 mg/kg/w IP 18–38 d ↓ Tumor volume/weight
↑ Anti-PD-1 immunotherapy
[71]
Breast cancer cells MCF7, SKBR3,
MDA-MB-231
MKC8866 5–20 μM +
PTX 10 nM
72 h
↓ XBP1s, ↓ IL-6, ↓ IL-8,
↓ CXCL1, ↓ TGFβ
↓ Proliferation
Cell cycle arrest
↓ Mammospheres
[219]
Mice with MDA-MB-231 xenograftsMKC8866 300 mg/kg/d OR + PTX 10 mg/kg/w IV
up to 60 d
↓ XBP1s
↓ Tumor growth, ↑ sensitivity to PTX
↑ Survival
[219]
Glioblastoma U87
and U251 cells
MKC8866 10–80 µM
or 30 µM + SIM and
TMZ 72 h
↓ p62, ↓ Beclin-1, ↓ XBP1s,
↑ LC3B-II/LC3B-I in
U251 cells
↓ Viability
No synergistic effect with TMZ
and SIM on death
[176]
Mice with intracerebral GL261-Luc cell glioblastomaMKC8866 + IR 2 Gy +
TMZ 25 mg/kg 5 d → TMZ 30–50 mg/kg 4 w
↑ Active caspase-3↑ Survival
↑ Apoptosis
[220]
HCC HepG2, Huh7 + stellate LX2 cells4µ8C 50–100 µM
48 h
↓ PCNA mRNA
↓ ROS
↓ Proliferation
↓ Migration
[223]
Mice with DEN-induced HCC4µ8C 10 mg/g 2/w
until 25th week
↓ Oncogenic proteins
↓ PCNA, ↓ HCC promoters PRDX5 and DDAH1, ↓ sXBP1/XBP1
↓ Tumor growth
↓ Collagen deposition
↓ Smooth muscle actin
[223]
HCC cells
(HepG2, SNU449, Huh7) and patient-derived organoids
4µ8C 10–1000 µM +
DOX 1 µM
24 h
↓ sXBP1 ↓ ATF4
↓ Lipid metabolism genes expression
↓ Viability, ↑ Death,
↓ lipid metabolism,
↑ DOX effect, ↓ oxygen consumption
[224]
Mice HCC tumor induced by DEN4µ8C 10 mg/g IP +
DOX 4 mg/g IV bi-weekly
3 w
↑ Caspase-3, ↓ ATF4,
↓ Ki67-positive cells,
↓ aSMA mRNA, ↓ CHOP,
↓ CXCL4, ↓ IL-1, ↓ ALT,
↓ Tumors, ↑ DOX effect, ↓ CD68, ↓ Triglycerides, ↓ Inflammation, ↓ Fibrosis, ↓ Collagen[224]
Patient-derived
and blast AML cells
STF-083010 50 μM
24 h
↓ sXBP1
↑ miR-34a expression
↑ Cytotoxicity[83]
Mice with breast cancer MCF-7-TAM-resistant xenograftsSTF-083010 30 mg/kg/w
+ TAM 100 µg/kg/d IP
↓ sXBP1↑ Effect of TAM
↓ Tumor growth
↑ Caspase-3
[80]
Patient-derived
AML blast cells
HNA 2–25 μM
+ BTZ 2.5–10 μM
48–72 h
↓ sXBP1, ↑ CHOP, ↓ Bcl-2,
↑ Bim, ↓ Cyclin D, ↑ p21cip1, ↑ p27kip1, ↑ Cleaved PARP,
↑ Caspase-3, ↑ miR-34a
↑ Cytotoxicity, ↑ BTZ effect, ↓ Colony formation,
↓ Viability, ↑ Apoptosis
[83]
Pancreatic cancer cells MiaPaCa-2, SU8686, Panc0403, and Panc0327HNA 10–50 μM
6–24 h
↓ sXBP1, ↑ CHOP, ↑ p-JNK, ↑ cells in sub-G1 phase,
↑ cleaved PARP, ↓ Bcl-2, ↑ Bim
↑ Apoptosis,
↓ Colony formation,
↓ Proliferation, ↓ MMP
[227]
Pancreatic cancer cells MiaPaCa-2, SU8686, Panc0403, and Panc0327Toyocamycin 0.5–5 μM
24 h
↑ Cleaved PARP
↓ Bcl-2
↑ CHOP
↓ Proliferation
↓ Colony formation
↓ Mitochondrial membrane potential
[227]
Pancreatic cancer cells MiaPaCa-2, SU8686, Panc0403, and Panc03273ETH 1–10 μM
24 h
↓ Proliferation
↓ Colony formation
[227]
Mice with pancreatic BxPc3 xenografts3ETH 20 mg/kg 3 times/w 4 w ↓ Tumor growth[227]
Glioblastoma U87 cellsZ4 0.5–25 μM 4–24 h↓ sXBP1, ↓ p-IRE1,
↓ SPARC
↓ Viability, ↓ Migration[228]
Mice with orthotopic glioblastomaZ4 300 mg/kg/d 182 d +TMZ 10 mg/kg/d 21d ↑ Effect of TMZ
↑ Relapse-free survival
[228]
GRP78 inhibitors
Pancreatic cancer PaCa-2, PANC, and BxPC-3 cellsYUM70 1–5–15 µM
24–48 h
↑ GRP78, ↑ CHOP, ↑ FAM 129A, ↑ p-eIF2α, ↑ ATF4, ↓ c-MYC, ↓ eIF4A, eIF4E, ↓ eIF5A, ↑ 4E-BP1, ↓ p-4E-BP1, ↑ cleaved PARP,
↑ Caspase-3/7
↓ Viability, ↑ Apoptosis,
↓ Cell proliferation,
↓ Colony formation,
Synergistic/additive effects with topotecan, vorinostat, or 5-FU
[232]
Mice bearing
PaCa-2 cell xenografts
YUM70 30 mg/kg IP
5 d/w 7 w
↓ Ki67 staining, ↑ CHOP,
↑ FAM 129A, ↑ Caspase-3
↓ Tumor growth
↑ Apoptosis
[233,234]
HNSCC cells SCC15, SCC25, and SCC351YUM70 1.25–30 µM +
CIS 12–24 µM
48 h
↑ GRP78, ↑ CHOP,
↑ cleaved PARP,
↑ Caspase-7
↓ Viability, ↑ Apoptosis,
↓ Clonogenicity,
↑ Sensitivity to CIS
[235,236]
TNBC MDA-MB-231 cells and HNSCC SCC15 and
SCC25 cells
YUM70 10 μM
24 h
↓ c-MYC, ↓ eIF4A, ↓ eIF4E, ↓ eIF5A, ↑ 4E-BP1, ↓ p-4E-BP1, ↑ cleaved PARP↓ Viability
↑ Apoptosis
[235]
Melanoma A375, Mel501, SKMel28, and patient-derived cellsHA15 10 μM,
48 h or 1–24 h
↑ p-PERK/EIF2AK3,
↑ p-elF2a, ↑ ATF4, ↑ ERN1, ↑ ATF6, ↑ DDIT3, ↑ LC3, ↑ sXBP1, ↑ JUN, ↑ BCL2
↓ Viability
↑ ER stress
↑ Apoptosis
↑ Autophagy
[236]
Mice with melanoma A375 xenografts sensitive/resistant to BRAF inhibitorsHA15 0.7 mg/day
2 w
↑ CHOP
↑ LC3B
↑ Autophagosomes
↓ Tumor growth
↑ Apoptosis
↑ Autophagy
[236]
Lung cancer A549, H460,
and H1975 cells
HA15 2–10 μM 48 h
or 10 μM 24 h
ATF4, ↑ ATF6, ↑ XBP1, ↑ IRE1, ↑ CHOP, ↑ Atg5, ↑ Atg7, ↑ Atg12, ↑ LC3, ↑ ULK1, ↑ Bax, ↑ CHOP↓ Proliferation,
Cell cycle arrest,
↑ Autophagosomes,
↓ Viability, ↑ Apoptosis
[239]
Pancreatic, lung, and colon cancer cells,
KRAS mutant
HA15 10 μM 2
4–48 h
↑ Cleaved PARP,
↑ CHOP, ↑ Caspase-7
↓ Viability
↑ Apoptosis
[233]
HNSSCC SCC25 and SCC15 cells, TNBC
MDA-MB-231 cells
HA15 10 μM
24–48 h
↓ c-MYC,
↑ 4E-BP1, ↓ p-4E-BP1,
↓ eIF4A, ↓ eIF4E, ↓ eIF5A
↓ Viability
↑ Apoptosis
[101]
Breast cancer MDA-MB-231
and T-47D cells
EGCG 10 μM + ETO
20–40 μM
24-48 h
↑ Caspase-7,
↓ GRP78/caspase-7 complex
↑ Apoptosis
↓ Colony formation
↑ Sensitivity to ETO
[241]
Breast cancer cells 4T1, MCF-7, and MDA-MB-231EGCG 20 μM +
PTX 1 μM 48 h
↑ p-JNK
↓ GRP78
↑ Apoptosis
↑ Sensitivity to PTX
[242]
Mice with breast 4T1 xenograftsEGCG 30 mg/kg +
PTX 10 mg/kg IP 24 d
↑ p-JNK
↓ GRP78
↓ Tumor growth[242]
Abbreviations. PDAC—pancreatic duct adenocarcinoma, HCC—hepatocellular carcinoma, MM—multiple myeloma, GBM—glioblastoma, HHC—hepatocellular carcinoma, AML—acute myeloid leukemia, TNBC—triple-negative breast cancer, HNSCC—head and neck squamous cell carcinoma, NSCLC—non-small cell lung carcinoma, OSCC—oral squamous cell carcinoma, TMZ—temozolomide, BTZ—bortezomib, PTX—paclitaxel, GEM—gemcitabine, CIS—cisplatin, IMA—imatinib, SIM—simvastatin, TM—tamoxifen, ETO—etoposide, DEN—diethylnitrosamine, HNA—2-hydroxy-1-naphthaldehyde, 3ETH—3-ethoxy-5,6-dibromosalicylaldehyde, IR—irradiation, PCNA—proliferating cell nuclear antigen, MMP—mitochondrial membrane potential, ↑—increase, ↓—suppression.

6. Conclusions

Numerous experimental in vitro and in vivo studies have shown that enhanced ERS and abnormal activities of components of the protein degradation machinery are common phenomena that are implicated in tumor growth, metastasis, and the emergence of resistance to chemotherapy. Modulation of ERS and the UPR is believed to be a promising and valuable approach to sensitize the malignancies of different origins to conventional cytotoxic drugs. The attempts to overcome the adaptation of cancer cells to apoptosis resulted in development of a variety of natural and synthetic compounds that are able to selectively suppress or activate ERS sensors and the components of UPR downstream signaling pathways. However, the majority of agents targeting ERS/UPR effectors have been examined only in experimental in vitro and in vivo settings. Although many of these compounds have entered clinical trials (clinicaltrials.gov database), only few of them, like sunitinib, have been approved for routine anti-cancer therapy. Moreover, the number of completed clinical trials for which the results have been published in scientific journals and detailed descriptions of the tumor types, number of patients, side effects and proper statistical analysis have been provided is limited. As a result, the available data are heterogeneous, inconsistent and often contradictory. The possible cause of such a situation is the quite variable expression of ERS/UPR genes and proteins in different cancers, which greatly depends on the cell type and presence of mutations. It means that the application of the agents targeting ERS/UPR components cannot be universal. The existence of other cellular targets, possible toxicities to normal cells, drug–drug interactions, poor bioavailability, insolubility, and other factors are still obstacles. The design of more specific compounds with low toxicity, the absence of side effects, and better bioavailability, either as the derivatives of existing natural drugs or new synthetic agents, is urgently required. Another way to increase the therapeutic specificity of ERS/UPR-modulating compounds is the development of more efficient delivery systems like nanoparticles and liposomes.

Funding

This work was performed within the state assignment of Federal Agency of Scientific Organizations of Russia (theme No. 075-00263-25-00).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Cherubini, A.; Zito, E. ER stress as a trigger of UPR and ER-phagy in cancer growth and spread. Front. Oncol. 2022, 12, 997235. [Google Scholar] [CrossRef] [PubMed]
  2. Ghemrawi, R.; Kremesh, S.; Mousa, W.K.; Khair, M. The role of ER stress and the unfolded protein response in cancer. Cancer Genom. Proteom. 2025, 22, 363–381. [Google Scholar] [CrossRef] [PubMed]
  3. Bonsignore, G.; Martinotti, S.; Ranzato, E. Endoplasmic reticulum stress and cancer: Could unfolded protein response be a druggable target for cancer therapy? Int. J. Mol. Sci. 2023, 24, 1566. [Google Scholar] [CrossRef] [PubMed]
  4. Lines, C.L.; McGrath, M.J.; Dorwart, T.; Conn, C.S. The integrated stress response in cancer progression: A force for plasticity and resistance. Front. Oncol. 2023, 13, 1206561. [Google Scholar] [CrossRef]
  5. Urra, H.; Aravena, R.; González-Johnson, L.; Hetz, C. The UPRising connection between endoplasmic reticulum stress and the tumor microenvironment. Trends Cancer 2024, 10, 1161–1173. [Google Scholar] [CrossRef]
  6. Mishra, T.; Dubey, N.; Basu, S. Small molecules for impairing endoplasmic reticulum in cancer. Org. Biomol. Chem. 2024, 22, 8689–8699. [Google Scholar] [CrossRef]
  7. Choudhary, M.K.; Pancholi, B.; Kumar, M.; Babu, R.; Garabadu, D. A review on endoplasmic reticulum-dependent anti-breast cancer activity of herbal drugs: Possible challenges and opportunities. J. Drug Target. 2025, 33, 206–231. [Google Scholar] [CrossRef]
  8. Kaczmarzyk, I.; Nowak-Perlak, M.; Woźniak, M. Promising approaches in plant-based therapies for thyroid cancer: An overview of In Vitro, In Vivo, and clinical trial studies. Int. J. Mol. Sci. 2024, 25, 4463. [Google Scholar] [CrossRef]
  9. Hetz, C.; Dillin, A. Central role of the ER proteostasis network in healthy aging. Trends Cell Biol. 2024, S0962-8924(24)00209-5. [Google Scholar] [CrossRef]
  10. Almanza, A.; Carlesso, A.; Chintha, C.; Creedican, S.; Doultsinos, D.; Leuzzi, B.; Luís, A.; McCarthy, N.; Montibeller, L.; More, S.; et al. Endoplasmic reticulum stress signalling—From basic mechanisms to clinical applications. FEBS J. 2019, 286, 241–278. [Google Scholar] [CrossRef]
  11. Akman, M.; Belisario, D.C.; Salaroglio, I.C.; Kopecka, J.; Donadelli, M.; De Smaele, E.; Riganti, C. Hypoxia, endoplasmic reticulum stress and chemoresistance: Dangerous liaisons. J. Exp. Clin. Cancer Res. 2021, 40, 28. [Google Scholar] [CrossRef] [PubMed]
  12. Acosta-Alvear, D.; Harnoss, J.M.; Walter, P.; Ashkenazi, A. Homeostasis control in health and disease by the unfolded protein response. Nat. Rev. Mol. Cell Biol. 2025, 26, 193–212. [Google Scholar] [CrossRef] [PubMed]
  13. Kapuy, O. Mechanism of decision making between autophagy and apoptosis induction upon endoplasmic reticulum stress. Int. J. Mol. Sci. 2024, 25, 4368. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, Z.; Lv, Y.; Zhao, N.; Guan, G.; Wang, J. Protein kinase R-like ER kinase and its role in endoplasmic reticulum stress-decided cell fate. Cell Death Dis. 2015, 6, e1822. [Google Scholar] [CrossRef]
  15. Ong, G.; Ragetli, R.; Mnich, K.; Doble, B.W.; Kammouni, W.; Logue, S.E. IRE1 signaling increases PERK expression during chronic ER stress. Cell Death Dis. 2024, 15, 276. [Google Scholar] [CrossRef]
  16. Wortel, I.M.; van der Meer, L.T.; Kilberg, M.S.; van Leeuwen, F.N. Surviving stress: Modulation of ATF4-mediated stress responses in normal and malignant cells. Trends Endocrinol. Metab. 2017, 28, 794–806. [Google Scholar] [CrossRef]
  17. Siwecka, N.; Rozpędek-Kamińska, W.; Wawrzynkiewicz, A.; Pytel, D.; Diehl, J.A.; Majsterek, I. The structure, activation and signaling of IRE1 and its role in determining cell fate. Biomedicines 2021, 9, 156. [Google Scholar] [CrossRef]
  18. Park, S.M.; Kang, T.I.; So, J.S. Roles of XBP1s in transcriptional regulation of target genes. Biomedicines 2021, 9, 791. [Google Scholar] [CrossRef]
  19. Maurel, M.; Chevet, E.; Tavernier, J.; Gerlo, S. Getting RIDD of RNA: IRE1 in cell fate regulation. Trends Biochem. Sci. 2014, 39, 245–254. [Google Scholar] [CrossRef]
  20. Lin, Y.; Jiang, M.; Chen, W.; Zhao, T.; Wei, Y. Cancer and ER stress: Mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed. Pharmacother. 2019, 118, 109249. [Google Scholar] [CrossRef]
  21. Fu, X.; Cui, J.; Meng, X.; Jiang, P.; Zheng, Q.; Zhao, W.; Chen, X. Endoplasmic reticulum stress, cell death and tumor: Association between endoplasmic reticulum stress and the apoptosis pathway in tumors (Review). Oncol. Rep. 2021, 45, 801–808. [Google Scholar] [CrossRef] [PubMed]
  22. Varghese, E.; Samuel, S.M.; Sadiq, Z.; Kubatka, P.; Liskova, A.; Benacka, J.; Pazinka, P.; Kruzliak, P.; Büsselberg, D. Anti-cancer agents in proliferation and cell death: The calcium connection. Int. J. Mol. Sci. 2019, 20, 3017. [Google Scholar] [CrossRef] [PubMed]
  23. Papp, B.; Launay, S.; Gélébart, P.; Arbabian, A.; Enyedi, A.; Brouland, J.P.; Carosella, E.D.; Adle-Biassette, H. Endoplasmic reticulum calcium pumps and tumor cell differentiation. Int. J. Mol. Sci. 2020, 21, 3351. [Google Scholar] [CrossRef] [PubMed]
  24. Pontisso, I.; Ornelas-Guevara, R.; Chevet, E.; Combettes, L.; Dupont, G. Gradual ER calcium depletion induces a progressive and reversible UPR signaling. PNAS Nexus 2024, 3, 229. [Google Scholar] [CrossRef]
  25. Cui, C.; Merritt, R.; Fu, L.; Pan, Z. Targeting calcium signaling in cancer therapy. Acta Pharm. Sin. B 2017, 7, 3–17. [Google Scholar] [CrossRef]
  26. Capitani, C.; Chioccioli Altadonna, G.; Santillo, M.; Lastraioli, E. Ion channels in lung cancer: Biological and clinical relevance. Front. Pharmacol. 2023, 14, 1283623. [Google Scholar] [CrossRef]
  27. Su, M.; Zheng, S.; Liu, H.; Tang, T.S.; Hu, Y. Ca2+ homeostasis: A potential target for cancer therapies. Biophys. Rep. 2024, 10, 283–292. [Google Scholar] [CrossRef]
  28. Makio, T.; Chen, J.; Simmen, T. ER stress as a sentinel mechanism for ER Ca2+ homeostasis. Cell Calcium 2024, 124, 102961. [Google Scholar] [CrossRef]
  29. Song, W.; Zhang, Q.; Cao, Z.; Jing, G.; Zhan, T.; Yuan, Y.; Kang, N.; Zhang, Q. Targeting SERCA2 in anti-tumor drug discovery. Curr. Drug Targets 2025, 26, 1–16. [Google Scholar] [CrossRef]
  30. Christodoulou, P.; Yiallouris, A.; Michail, A.; Christodoulou, M.I.; Politis, P.K.; Patrikios, I. Altered SERCA expression in breast cancer. Medicina 2021, 57, 1074. [Google Scholar] [CrossRef]
  31. Zhang, G.; Shang, H.; Liu, B.; Wu, G.; Wu, D.; Wang, L.; Li, S.; Wang, Z.; Wang, S.; Yuan, J. Increased ATP2A1 predicts poor prognosis in patients with colorectal carcinoma. Front. Genet. 2022, 13, 661348. [Google Scholar] [CrossRef] [PubMed]
  32. Chung, F.Y.; Lin, S.R.; Lu, C.Y.; Yeh, C.S.; Chen, F.M.; Hsieh, J.S.; Huang, T.J.; Wang, J.Y. Sarco/endoplasmic reticulum calcium-ATPase 2 expression as a tumor marker in colorectal cancer. Am. J. Surg. Pathol. 2006, 30, 969–974. [Google Scholar] [CrossRef]
  33. Fan, L.; Li, A.; Li, W.; Cai, P.; Yang, B.; Zhang, M.; Gu, Y.; Shu, Y.; Sun, Y.; Shen, Y.; et al. Novel role of Sarco/endoplasmic reticulum calcium ATPase 2 in development of colorectal cancer and its regulation by F36, a curcumin analog. Biomed. Pharmacother. 2014, 68, 1141–1148. [Google Scholar] [CrossRef] [PubMed]
  34. Youssef, H.M.; Radi, D.A.; Abd El-Azeem, M.A. Expression of TSP50, SERCA2 and IL-8 in colorectal adenoma and carcinoma: Correlation to clinicopathological factors. Pathol. Oncol. Res. 2021, 27, 1609990. [Google Scholar] [CrossRef]
  35. Xu, X.Y.; Gou, W.F.; Yang, X.; Wang, G.L.; Takahashi, H.; Yu, M.; Mao, X.Y.; Takano, Y.; Zheng, H.C. Aberrant SERCA3 expression is closely linked to pathogenesis, invasion, metastasis, and prognosis of gastric carcinomas. Tumour Biol. 2012, 33, 1845–1854. [Google Scholar] [CrossRef]
  36. Meneses-Morales, I.; Izquierdo-Torres, E.; Flores-Peredo, L.; Rodríguez, G.; Hernández-Oliveras, A.; Zarain-Herzberg, A. Epigenetic regulation of the human ATP2A3 gene promoter in gastric and colon cancer cell lines. Mol. Carcinog. 2019, 58, 887–897. [Google Scholar] [CrossRef]
  37. Li, J.; Li, X.; Huang, H.; Tao, L.; Zhang, C.; Xie, Y.; Jiang, Y. Role of SERCA3 in the prognosis and immune function in pan-cancer. J. Oncol. 2022, 2022, 9359879. [Google Scholar] [CrossRef]
  38. Gou, W.F.; Niu, Z.F.; Zhao, S.; Takano, Y.; Zheng, H.C. Aberrant SERCA3 expression during the colorectal adenoma-adenocarcinoma sequence. Oncol. Rep. 2014, 31, 232–240. [Google Scholar] [CrossRef]
  39. Aït-Ghezali, L.; Arbabian, A.; Jeibmann, A.; Hasselblatt, M.; Hallaert, G.G.; Broecke, C.V.D.; Gray, F.; Brouland, J.-P.; Varin-Blank, N.; Papp, B. Loss of endoplasmic reticulum calcium pump expression in choroid plexus tumours. Neuropathol. Appl. Neurobiol. 2014, 40, 726–735. [Google Scholar] [CrossRef]
  40. Adle-Biassette, H.; Ricci, R.; Martin, A.; Martini, M.; Ravegnini, G.; Kaci, R.; Gélébart, P.; Poirot, B.; Sándor, Z.; Lehman-Che, J.; et al. Sarco/endoplasmic reticulum calcium ATPase 3 (SERCA3) expression in gastrointestinal stromal tumours. Pathology 2024, 56, 343–356. [Google Scholar] [CrossRef]
  41. Papp, B.; Brouland, J.-P. Altered endoplasmic reticulum calcium pump expression during breast tumorigenesis. Breast Cancer Basic Clin. Res. 2011, 5, 163–174. [Google Scholar] [CrossRef] [PubMed]
  42. Simbilyabo, L.Z.; Yang, L.; Wen, J.; Liu, Z. The unfolded protein response machinery in glioblastoma genesis, chemoresistance and as a druggable target. CNS Neurosci. Ther. 2024, 30, e14839. [Google Scholar] [CrossRef] [PubMed]
  43. Borrello, M.T.; Martin, M.B.; Pin, C.L. The unfolded protein response: An emerging therapeutic target for pancreatitis and pancreatic ductal adenocarcinoma. Pancreatology 2022, 22, 148–159. [Google Scholar] [CrossRef] [PubMed]
  44. Botrus, G.; Miller, R.M.; Uson Junior, P.L.S.; Kannan, G.; Han, H.; Von Hoff, D.D. Increasing stress to induce apoptosis in pancreatic cancer via the unfolded protein response (UPR). Int. J. Mol. Sci. 2023, 24, 577. [Google Scholar] [CrossRef]
  45. Jadhav, A.; Menon, A.; Gupta, K.; Singh, N. Molecular and therapeutic insight into ER Stress signaling in NSCLC. J. Drug Target. 2025, 33, 877–886. [Google Scholar] [CrossRef]
  46. Salaroglio, I.C.; Panada, E.; Moiso, E.; Buondonno, I.; Provero, P.; Rubinstein, M.; Kopecka, J.; Riganti, C. PERK induces resistance to cell death elicited by endoplasmic reticulum stress and chemotherapy. Mol. Cancer 2017, 16, 91. [Google Scholar] [CrossRef]
  47. Liu, Z.; Liu, Q.; Zeng, A.; Song, L. Regulatory function of endoplasmic reticulum stress in colorectal cancer: Mechanism, facts, and perspectives. Int. Immunopharmacol. 2025, 147, 114024. [Google Scholar] [CrossRef]
  48. Farahani, N.; Alimohammadi, M.; Raei, M.; Nabavi, N.; Aref, A.R.; Hushmandi, K.; Daneshi, S.; Razzaghi, A.; Taheriazam, A.; Hashemi, M. Exploring the dual role of endoplasmic reticulum stress in urological cancers: Implications for tumor progression and cell death interactions. J. Cell Commun. Signal. 2024, 18, e12054. [Google Scholar] [CrossRef]
  49. Ali, M.U.; Ur Rahman, M.S.; Jia, Z.; Jiang, C. Eukaryotic translation initiation factors and cancer. Tumor Biol. 2017, 39, 1010428317709805. [Google Scholar] [CrossRef]
  50. Hao, P.; Yu, J.; Ward, R.; Liu, Y.; Hao, Q.; An, S.; Xu, T. Eukaryotic translation initiation factors as promising targets in cancer therapy. Cell Commun. Signal. 2020, 18, 175. [Google Scholar] [CrossRef]
  51. Féral, K.; Jaud, M.; Philippe, C.; Di Bella, D.; Pyronnet, S.; Rouault-Pierre, K.; Mazzolini, L.; Touriol, C. ER stress and unfolded protein response in leukemia: Friend, foe, or both? Biomolecules 2021, 11, 199. [Google Scholar] [CrossRef] [PubMed]
  52. Wiese, W.; Siwecka, N.; Wawrzynkiewicz, A.; Rozpędek-Kamińska, W.; Kucharska, E.; Majsterek, I. IRE1α inhibitors as a promising therapeutic strategy in blood malignancies. Cancers 2022, 14, 2526. [Google Scholar] [CrossRef] [PubMed]
  53. Lucas, D.; Sarkar, T.; Niemeyer, C.Y.; Harnoss, J.C.; Schneider, M.; Strowitzki, M.J.; Harnoss, J.M. IRE1 is a promising therapeutic target in pancreatic cancer. Am. J. Physiol. Cell Physiol. 2025, 328, C806–C824. [Google Scholar] [CrossRef] [PubMed]
  54. Lin, M.; Mo, Y.; Li, C.M.; Liu, Y.Z.; Feng, X.P. GRP78 as a potential therapeutic target in cancer treatment: An updated review of its role in chemoradiotherapy resistance of cancer cells. Med. Oncol. 2025, 42, 49. [Google Scholar] [CrossRef]
  55. Song, W.; Rahimian, N.; Bashkandi, A.H. GRP78: A new promising candidate in colorectal cancer pathogenesis and therapy. Eur. J. Pharmacol. 2025, 995, 177308. [Google Scholar] [CrossRef]
  56. Liu, J.; Xiao, M.; Li, J.; Wang, D.; He, Y.; He, J.; Gao, F.; Mai, L.; Li, Y.; Liang, Y.; et al. Activation of UPR signaling pathway is associated with the malignant progression and poor prognosis in prostate cancer. Prostate 2017, 77, 274–281. [Google Scholar] [CrossRef]
  57. Wang, E.M.; Akasaka, H.; Zhao, J.; Varadhachary, G.R.; Lee, J.E.; Maitra, A.; Fleming, J.B.; Hung, M.C.; Wang, H.; Katz, M.H. Expression and clinical significance of Protein Kinase RNA-like endoplasmic reticulum kinase and phosphorylated Eukaryotic Initiation Factor 2α in pancreatic ductal adenocarcinoma. Pancreas 2019, 48, 323–328. [Google Scholar] [CrossRef]
  58. Wang, P.; Han, L.; Yu, M.; Cao, Z.; Li, X.; Shao, Y.; Zhu, G. The prognostic value of PERK in cancer and its relationship with immune cell infiltration. Front. Mol. Biosci. 2021, 8, 648752. [Google Scholar] [CrossRef]
  59. Rosenwald, I.B.; Wang, S.; Savas, L.; Woda, B.; Pullman, J. Expression of translation initiation factor eIF-2alpha is increased in benign and malignant melanocytic and colonic epithelial neoplasms. Cancer 2003, 98, 1080–1088. [Google Scholar] [CrossRef]
  60. Wang, S.; Lloyd, R.V.; Hutzler, M.J.; Rosenwald, I.B.; Safran, M.S.; Patwardhan, N.A.; Khan, A. Expression of eukaryotic translation initiation factors 4E and 2alpha correlates with the progression of thyroid carcinoma. Thyroid 2001, 11, 1101–1107. [Google Scholar] [CrossRef]
  61. Rosenwald, I.B.; Koifman, L.; Savas, L.; Chen, J.J.; Woda, B.A.; Kadin, M.E. Expression of the translation initiation factors eIF-4E and eIF-2* is frequently increased in neoplastic cells of Hodgkin lymphoma. Hum. Pathol. 2008, 39, 910–916. [Google Scholar] [CrossRef] [PubMed]
  62. Li, F.; Sun, H.; Li, Y.; Bai, X.; Dong, X.; Zhao, N.; Meng, J.; Sun, B.; Zhang, D. High expression of eIF4E is associated with tumor macrophage infiltration and leads to poor prognosis in breast cancer. BMC Cancer 2021, 21, 1305. [Google Scholar] [CrossRef] [PubMed]
  63. Yoon, K.H.; Chu, H.; Kim, H.; Huh, S.; Kim, E.K.; Kang, U.B.; Shin, H.C. Comparative profiling by data-independent acquisition mass spectrometry reveals featured plasma proteins in breast cancer: A pilot study. Ann. Surg. Treat. Res. 2024, 106, 195–202. [Google Scholar] [CrossRef]
  64. Nguyen, H.G.; Conn, C.S.; Kye, Y.; Xue, L.; Forester, C.M.; Cowan, J.E.; Hsieh, A.C.; Cunningham, J.T.; Truillet, C.; Tameire, F.; et al. Development of a stress response therapy targeting aggressive prostate cancer. Sci. Transl. Med. 2018, 10, eaar2036. [Google Scholar] [CrossRef]
  65. Cao, Z.; Jing, Y.; Cheng, C.; Wang, F.; Guan, M.; Zhang, K.; Jiao, J.; Ruan, L.; Chen, Z. EIF2Ss, a novel c-Myc-correlated gene family, is associated with poor prognosis and immune infiltration in pancreatic adenocarcinoma. Front. Biosci. 2024, 29, 119. [Google Scholar] [CrossRef]
  66. Ji, P.; Wang, H.; Cheng, Y.; Liang, S. Prognostic prediction and gene regulation network of EIF2S2 in hepatocellular carcinoma based on data mining. J. Gastrointest. Oncol. 2021, 12, 3061–3078. [Google Scholar] [CrossRef]
  67. Tejada, S.; Lobo, M.V.; García-Villanueva, M.; Sacristán, S.; Pérez-Morgado, M.I.; Salinas, M.; Martín, M.E. Eukaryotic initiation factors (eIF) 2alpha and 4E expression, localization, and phosphorylation in brain tumors. J. Histochem. Cytochem. 2009, 57, 503–512. [Google Scholar] [CrossRef]
  68. Lobo, M.V.; Martín, M.E.; Pérez, M.I.; Alonso, F.J.; Redondo, C.; Alvarez, M.I.; Salinas, M. Levels, phosphorylation status and cellular localization of translational factor eIF2 in gastrointestinal carcinomas. Histochem. J. 2000, 32, 139–150. [Google Scholar] [CrossRef]
  69. He, Y.; Correa, A.M.; Raso, M.G.; Hofstetter, W.L.; Fang, B.; Behrens, C.; Roth, J.A.; Zhou, Y.; Yu, L.; Wistuba, I.I.; et al. The role of PKR/eIF2alpha signaling pathway in prognosis of non-small cell lung cancer. PLoS ONE 2011, 6, e24855. [Google Scholar] [CrossRef]
  70. Guo, L.; Chi, Y.; Xue, J.; Ma, L.; Shao, Z.; Wu, J. Phosphorylated EIF2 predicts disease-free survival in triple-negative breast cancer patients. Sci. Rep. 2017, 7, 44674. [Google Scholar] [CrossRef]
  71. Unal, B.; Kuzu, O.F.; Jin, Y.; Osorio, D.; Kildal, W.; Pradhan, M.; Kung, S.H.; Oo, H.Z.; Daugaard, M.; Vendelbo, M.; et al. Targeting IRE1α reprograms the tumor microenvironment and enhances anti-tumor immunity in prostate cancer. Nat. Commun. 2024, 15, 8895. [Google Scholar] [CrossRef] [PubMed]
  72. Sakatani, T.; Maemura, K.; Hiyama, N.; Amano, Y.; Watanabe, K.; Kage, H.; Fukayama, M.; Nakajima, J.; Yatomi, Y.; Nagase, T.; et al. High expression of IRE1 in lung adenocarcinoma is associated with a lower rate of recurrence. Jpn. J. Clin. Oncol. 2017, 47, 543–550. [Google Scholar] [CrossRef] [PubMed]
  73. Shuda, M.; Kondoh, N.; Imazeki, N.; Tanaka, K.; Okada, T.; Mori, K.; Hada, A.; Arai, M.; Wakatsuki, T.; Matsubara, O.; et al. Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: A possible involvement of the ER stress pathway in hepatocarcinogenesis. J. Hepatol. 2003, 38, 605–614. [Google Scholar] [CrossRef] [PubMed]
  74. Scriven, P.; Coulson, S.; Haines, R.; Balasubramanian, S.; Cross, S.; Wyld, L. Activation and clinical significance of the unfolded protein response in breast cancer. Br. J. Cancer 2009, 101, 1692–1698. [Google Scholar] [CrossRef]
  75. Chen, C.; Zhang, X. IRE1α-XBP1 pathway promotes melanoma progression by regulating IL-6/STAT3 signaling. J. Transl. Med. 2017, 15, 42. [Google Scholar] [CrossRef]
  76. Dowdell, A.; Marsland, M.; Faulkner, S.; Gedye, C.; Lynam, J.; Griffin, C.P.; Marsland, J.; Jiang, C.C.; Hondermarck, H. Targeting XBP1 mRNA splicing sensitizes glioblastoma to chemotherapy. FASEB Bioadv. 2023, 5, 211–220. [Google Scholar] [CrossRef]
  77. Balague, O.; Mozos, A.; Martinez, D.; Hernandez, L.; Colomo, L.; Mate, J.L.; Teruya-Feldstein, J.; Lin, O.; Campo, E.; Lopez-Guillermo, A.; et al. Activation of the Endoplasmic Reticulum Stress-Associated Transcription Factor X Box-Binding Protein-1 Occurs in a Subset of Normal Germinal-Center B Cells and in Aggressive B-Cell Lymphomas with Prognostic Implications. Am. J. Pathol. 2009, 174, 2337–2346. [Google Scholar] [CrossRef]
  78. Masouleh, B.K.; Geng, H.; Hurtz, C.; Chan, L.N.; Logan, A.C.; Chang, M.S.; Huang, C.; Swaminathan, S.; Sun, H.; Paietta, E.; et al. Mechanistic rationale for targeting the unfolded protein response in pre-B acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. USA 2014, 111, E2219–E2228. [Google Scholar] [CrossRef]
  79. Zhang, K.; Liu, H.; Song, Z.; Jiang, Y.; Kim, H.; Samavati, L.; Nguyen, H.M.; Yang, Z.Q. The UPR transducer IRE1 promotes breast cancer malignancy by degrading tumor suppressor microRNAs. iScience 2020, 23, 101503. [Google Scholar] [CrossRef]
  80. Ming, J.; Ruan, S.; Wang, M.; Ye, D.; Fan, N.; Meng, Q.; Tian, B.; Huang, T. A novel chemical, STF-083010, reverses Tamoxifen-related drug resistance in breast cancer by inhibiting IRE1/XBP1. Oncotarget 2015, 6, 40692–40703. [Google Scholar] [CrossRef]
  81. Sheng, X.; Nenseth, H.Z.; Qu, S.; Kuzu, O.F.; Frahnow, T.; Simon, L.; Greene, S.; Zeng, Q.; Fazli, L.; Rennie, P.S.; et al. IRE1α-XBP1s pathway promotes prostate cancer by activating c-MYC signaling. Nat. Commun. 2019, 10, 323. [Google Scholar] [CrossRef] [PubMed]
  82. Schardt, J.A.; Weber, D.; Eyholzer, M.; Mueller, B.U.; Pabst, T. Activation of the Unfolded protein response is associated with favorable prognosis in acute myeloid leukemia. Clin. Cancer Res. 2009, 15, 3834–3841. [Google Scholar] [CrossRef] [PubMed]
  83. Sun, H.; Lin, D.C.; Guo, X.; Kharabi Masouleh, B.; Gery, S.; Cao, Q.; Alkan, S.; Ikezoe, T.; Akiba, C.; Paquette, R.; et al. Inhibition of IRE1α-driven pro-survival pathways is a promising therapeutic application in acute myeloid leukemia. Oncotarget 2016, 7, 18736–18749. [Google Scholar] [CrossRef] [PubMed]
  84. Philippe, C.; Jaud, M.; Féral, K.; Gay, A.; Van Den Berghe, L.; Farce, M.; Bousquet, M.; Pyronnet, S.; Mazzolini, L.; Rouault-Pierre, K.; et al. Pivotal role of the endoplasmic reticulum stress-related XBP1s/miR-22/SIRT1 axis in acute myeloid leukemia apoptosis and response to chemotherapy. Leukemia 2024, 38, 1764–1776. [Google Scholar] [CrossRef]
  85. Xiao, W.; Cao, R.C.; Yang, W.J.; Tan, J.H.; Liu, R.Q.; Kan, H.P.; Zhou, L.; Zhang, N.; Chen, Z.Y.; Chen, X.M.; et al. Roles and clinical significances of ATF6, EMC6, and APAF1 in prognosis of pancreatic cancer. Front. Genet. 2022, 12, 730847. [Google Scholar] [CrossRef]
  86. Yarapureddy, S.; Abril, J.; Foote, J.; Kumar, S.; Asad, O.; Sharath, V.; Faraj, J.; Daniel, D.; Dickman, P.; White-Collins, A.; et al. ATF6α activation enhances survival against chemotherapy and serves as a prognostic indicator in osteosarcoma. Neoplasia 2019, 21, 516–532. [Google Scholar] [CrossRef]
  87. Meng, J.; Liu, K.; Shao, Y.; Feng, X.; Ji, Z.; Chang, B.; Wang, Y.; Xu, L.; Yang, G. ID1 confers cancer cell chemoresistance through STAT3/ATF6-mediated induction of autophagy. Cell Death Dis. 2020, 11, 137. [Google Scholar] [CrossRef]
  88. Zhang, Q.; Zhu, X.D.; Sun, Q.N.; Wang, D. The UPR signaling molecule ATF6 is a poor prognostic marker in gastric cancer. Asian J. Surg. 2022, 45, 2836–2837. [Google Scholar] [CrossRef]
  89. Wu, Y.; Xie, Q.; Wu, L.; Li, Z.; Li, X.; Zhang, L.; Zhang, B. Identification of activating transcription factor 6 (ATF6) as a novel prognostic biomarker and potential target in oral squamous cell carcinoma. Gene 2024, 915, 148436. [Google Scholar] [CrossRef]
  90. Miao, Y.; Chen, Q.; Liu, X.; Bu, J.; Zhang, Z.; Liu, T.; Yue, Z.; Huang, L.; Sun, S.; Li, H.; et al. Comprehensive analysis of endoplasmic reticulum stress related signature in head and neck squamous carcinoma. Sci. Rep. 2024, 14, 16972. [Google Scholar] [CrossRef]
  91. Martinez-Useros, J.; Georgiev-Hristov, T.; Borrero-Palacios, A.; Fernandez-Aceñero, M.J.; Rodríguez-Remírez, M.; Del Puerto-Nevado, L.; Cebrian, A.; Gomez Del Pulgar, M.T.; Cazorla, A.; Vega-Bravo, R.; et al. Identification of poor-outcome biliopancreatic carcinoma patients with two-marker signature based on ATF6α and p-p38 “STARD compliant”. Medicine 2015, 94, e1972. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, C.Y.; Hsu, C.C.; Huang, T.T.; Lee, C.H.; Chen, J.L.; Yang, S.H.; Jiang, J.K.; Chen, W.S.; Lee, K.D.; Teng, H.W. ER stress-related ATF6 upregulates CIP2A and contributes to poor prognosis of colon cancer. Mol. Oncol. 2018, 12, 1706–1717. [Google Scholar] [CrossRef] [PubMed]
  93. Fernandez, P.; Tabbara, S.; Jacobs, L.; Manning, F.; Tsangaris, T.; Schwartz, A.; Kennedy, K.; Patierno, S. Overexpression of the glucoseregulated stress gene GRP78 in malignant but not benign human breast lesions. Breast Cancer Res. Treat. 2000, 59, 15–26. [Google Scholar] [CrossRef] [PubMed]
  94. Bartkowiak, K.; Effenberger, K.E.; Harder, S.; Andreas, A.; Buck, F.; Peter-Katalinic, J.; Pantel, K.; Brandt, B.H. Discovery of a novel unfolded protein response phenotype of cancer stem/progenitor cells from the bone marrow of breast cancer patients. J. Proteome Res. 2010, 9, 3158–3168. [Google Scholar] [CrossRef]
  95. Al-Rawashdeh, F.Y.; Scriven, P.; Cameron, I.C.; Vergani, P.V.; Wyld, L. Unfolded protein response activation contributes to chemoresistance in hepatocellular carcinoma. Eur. J. Gastroenterol. Hepatol. 2010, 22, 1099–1105. [Google Scholar] [CrossRef]
  96. Su, R.; Li, Z.; Li, H.; Song, H.; Bao, C.; Wei, J.; Cheng, L. Grp78 promotes the invasion of hepatocellular carcinoma. BMC Cancer 2010, 10, 20. [Google Scholar] [CrossRef]
  97. Zhang, J.; Jiang, Y.; Jia, Z.; Li, Q.; Gong, W.; Wang, L.; Wei, D.; Yao, J.; Fang, S.; Xie, K. Association of elevated GRP78 expression with increased lymph node metastasis and poor prognosis in patients with gastric cancer. Clin. Exp. Metastasis 2006, 23, 401–410. [Google Scholar] [CrossRef]
  98. Pyrko, P.; Schönthal, A.H.; Hofman, F.M.; Chen, T.C.; Lee, A.S. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007, 67, 9809–9816. [Google Scholar] [CrossRef]
  99. Daneshmand, S.; Quek, M.L.; Lin, E.; Lee, C.; Cote, R.J.; Hawes, D.; Cai, J.; Groshen, S.; Lieskovsky, G.; Skinner, D.G.; et al. Glucose-regulated protein GRP78 is up-regulated in prostate cancer and correlates with recurrence and survival. Hum. Pathol. 2007, 38, 1547–1552. [Google Scholar] [CrossRef]
  100. Thakur, P.C.; Miller-Ocuin, J.L.; Nguyen, K.; Matsuda, R.; Singhi, A.D.; Zeh, H.J.; Bahary, N. Inhibition of endoplasmic-reticulum-stress-mediated autophagy enhances the effectiveness of chemotherapeutics on pancreatic cancer. J. Transl. Med. 2018, 16, 190. [Google Scholar] [CrossRef]
  101. Wu, J.; Wu, Y.; Lian, X. Targeted inhibition of GRP78 by HA15 promotes apoptosis of lung cancer cells accompanied by ER stress and autophagy. Biol. Open 2020, 9, bio053298. [Google Scholar] [CrossRef] [PubMed]
  102. Xia, S.; Duan, W.; Liu, W.; Zhang, X.; Wang, Q. GRP78 in lung cancer. J. Transl. Med. 2021, 19, 118. [Google Scholar] [CrossRef] [PubMed]
  103. Schneider, M.; Winkler, K.; Kell, R.; Pfaffl, M.W.; Atkinson, M.J.; Moertl, S. The Chaperone protein GRP78 promotes survival and migration of head and neck cancer after direct radiation exposure and extracellular vesicle-transfer. Front. Oncol. 2022, 12, 842418. [Google Scholar] [CrossRef] [PubMed]
  104. Thornton, M.; Aslam, M.A.; Tweedle, E.M.; Ang, C.; Campbell, F.; Jackson, R.; Costello, E.; Rooney, P.S.; Vlatković, N.; Boyd, M.T. The unfolded protein response regulator GRP78 is a novel predictive biomarker in colorectal cancer. Int. J. Cancer 2013, 133, 1408–1418. [Google Scholar] [CrossRef]
  105. Chen, Q.; Li, C.; Wei, W.; Li, J.; Liu, F.; Fu, Y.; Tang, L.; Han, F. Endoplasmic reticulum stress response pathway-mediated cell death in ovarian cancer. Front. Oncol. 2024, 14, 1446552. [Google Scholar] [CrossRef]
  106. Xu, D.; Liu, Z.; Liang, M.X.; Fei, Y.J.; Zhang, W.; Wu, Y.; Tang, J.H. Endoplasmic reticulum stress targeted therapy for breast cancer. Cell Commun. Signal. 2022, 20, 174. [Google Scholar] [CrossRef]
  107. Markouli, M.; Strepkos, D.; Papavassiliou, A.G.; Piperi, C. Targeting of endoplasmic reticulum (ER) stress in gliomas. Pharmacol. Res. 2020, 157, 104823. [Google Scholar] [CrossRef]
  108. Zhai, K.; Mazurakova, A.; Koklesova, L.; Kubatka, P.; Büsselberg, D. Flavonoids synergistically enhance the anti-glioblastoma effects of chemotherapeutic drugs. Biomolecules 2021, 11, 1841. [Google Scholar] [CrossRef]
  109. Yu, M.; Lun, J.; Zhang, H.; Wang, L.; Zhang, G.; Zhang, H.; Fang, J. Targeting UPR branches, a potential strategy for enhancing efficacy of cancer chemotherapy. Acta Biochim. Biophys. Sin. 2021, 53, 1417–1427. [Google Scholar] [CrossRef]
  110. Mu, W.; Zhi, Y.; Zhou, J.; Wang, C.; Chai, K.; Fan, Z.; Lv, G. Endoplasmic reticulum stress and quality control in relation to Cisplatin resistance in tumor cells. Front. Pharmacol. 2024, 15, 1419468. [Google Scholar] [CrossRef]
  111. Michelangeli, F.; East, J.M. A diversity of SERCA Ca2+ pump inhibitors. Biochem. Soc. Trans. 2011, 39, 789–797. [Google Scholar] [CrossRef] [PubMed]
  112. Peterková, L.; Kmoníčková, E.; Ruml, T.; Rimpelová, S. Sarco/endoplasmic reticulum calcium ATPase inhibitors: Beyond anticancer perspective. J. Med. Chem. 2020, 63, 1937–1963. [Google Scholar] [CrossRef] [PubMed]
  113. Xu, C.; Ma, H.; Inesi, G.; Al-Shawi, M.K.; Toyoshima, C. Specific structural requirements for the inhibitory effect of Thapsigargin on the Ca2+ ATPase SERCA. J. Biol. Chem. 2004, 279, 17973–17979. [Google Scholar] [CrossRef]
  114. Suresh, A.; Bagchi, D.; Kaliappan, K.P. Thapsigargin: A promising natural product with diverse medicinal potential—A review of synthetic approaches and total syntheses. Org. Biomol. Chem. 2024, 22, 8551–8569. [Google Scholar] [CrossRef]
  115. Jaskulska, A.; Janecka, A.E.; Gach-Janczak, K. Thapsigargin-from traditional medicine to anticancer drug. Int. J. Mol. Sci. 2020, 22, 4. [Google Scholar] [CrossRef]
  116. Khurram, I.; Khan, M.U.; Ibrahim, S.; Ghani, M.U.; Amin, I.; Falzone, L.; Herrera-Bravo, J.; Setzer, W.N.; Sharifi-Rad, J.; Calina, D. Thapsigargin and its prodrug derivatives: Exploring novel approaches for targeted cancer therapy through calcium signaling disruption. Med. Oncol. 2024, 42, 7. [Google Scholar] [CrossRef]
  117. Denmeade, S.R.; Jakobsen, C.M.; Janssen, S.; Khan, S.R.; Garrett, E.S.; Lilja, H.; Christensen, S.B.; Isaacs, J.T. Prostate-specific antigen-activated Thapsigargin prodrug as targeted therapy for prostate cancer. J. Natl. Cancer Inst. 2003, 95, 990–1000. [Google Scholar] [CrossRef]
  118. Sehgal, P.; Szalai, P.; Olesen, C.; Praetorius, H.A.; Nissen, P.; Christensen, S.B.; Engedal, N.; Møller, J.V. Inhibition of the sarco/endoplasmic reticulum (ER) Ca2+-ATPase by Thapsigargin analogs induces cell death via ER Ca2+ depletion and the unfolded protein response. J. Biol. Chem. 2017, 292, 19656–19673. [Google Scholar] [CrossRef]
  119. Yu, T.J.; Shiau, J.P.; Tang, J.Y.; Farooqi, A.A.; Cheng, Y.B.; Hou, M.F.; Yen, C.H.; Chang, H.W. Physapruin A exerts endoplasmic reticulum stress to trigger breast cancer cell apoptosis via oxidative stress. Int. J. Mol. Sci. 2023, 24, 8853. [Google Scholar] [CrossRef]
  120. Kotnova, A.P.; Lyanova, B.M.; Dukhanina, E.A.; Portseva, T.N.; Ilyin, Y.V.; Georgieva, S.G.; Stepchenko, A.G.; Pankratova, E.V. Thapsigargin, inhibitor of sarco-endoplasmic Ca2+-ATPase, effectively suppresses the expression of S100A4 protein in human breast cancer cell line. Dokl. Biochem. Biophys. 2019, 486, 181–183. [Google Scholar] [CrossRef]
  121. Chang, K.T.; Thompson, K.N.; Pratt, S.J.P.; Ju, J.A.; Lee, R.M.; Mathias, T.J.; Mull, M.L.; Annis, D.A.; Ory, E.C.; Stemberger, M.B.; et al. Elevation of cytoplasmic calcium suppresses microtentacle formation and function in breast tumor cells. Cancers 2023, 15, 884. [Google Scholar] [CrossRef] [PubMed]
  122. Lindner, P.; Christensen, S.B.; Nissen, P.; Møller, J.V.; Engedal, N. Cell death induced by the ER stressor Thapsigargin involves death receptor 5, a non-autophagic function of MAP1LC3B, and distinct contributions from unfolded protein response components. Cell Commun. Signal. 2020, 18, 12. [Google Scholar] [CrossRef]
  123. Peñaranda-Fajardo, N.M.; Meijer, C.; Liang, Y.; Dijkstra, B.M.; Aguirre-Gamboa, R.; den Dunnen, W.F.; Kruyt, F.A. ER stress and UPR activation in glioblastoma: Identification of a noncanonical PERK mechanism regulating GBM stem cells through SOX2 modulation. Cell Death Dis. 2019, 10, 690. [Google Scholar] [CrossRef] [PubMed]
  124. Pecoraro, M.; Serra, A.; Pascale, M.; Franceschelli, S. The ER stress induced in human neuroblastoma cells can be reverted by lumacaftor, a CFTR corrector. Curr. Issues Mol. Biol. 2024, 46, 9342–9358. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, L.; Huang, X.; Kuang, Y.; Xing, Z.; Deng, X.; Luo, Z. Thapsigargin induces apoptosis in adrenocortical carcinoma by activating endoplasmic reticulum stress and the JNK signaling pathway: An in vitro and in vivo study. Drug Des. Devel. Ther. 2019, 13, 2787–2798. [Google Scholar] [CrossRef]
  126. Wang, L.; Wang, L.; Song, R.; Shen, Y.; Sun, Y.; Gu, Y.; Shu, Y.; Xu, Q. Targeting sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2 by curcumin induces ER stress-associated apoptosis for treating human liposarcoma. Mol. Cancer Ther. 2011, 10, 461–471. [Google Scholar] [CrossRef]
  127. Al-Taweel, N.; Varghese, E.; Florea, A.M.; Büsselberg, D. Cisplatin (CDDP) triggers cell death of MCF-7 cells following disruption of intracellular calcium ([Ca2+]i) homeostasis. J. Toxicol. Sci. 2014, 39, 765–774. [Google Scholar] [CrossRef]
  128. Balakrishnan, P.; Arasu, A.; Velusamy, T. Targeting altered calcium homeostasis and uncoupling protein-2 promotes sensitivity in drug-resistant breast cancer cells. J. Biochem. Mol. Toxicol. 2024, 38, e23575. [Google Scholar] [CrossRef]
  129. Kim, S.M.; Park, K.; Lim, J.H.; Yun, H.J.; Kim, S.Y.; Choi, K.H.; Kim, C.W.; Lee, J.H.; Weicker, R.; Pan, C.H.; et al. Potential therapeutic agents against Paclitaxel- and Sorafenib-resistant papillary thyroid carcinoma. Int. J. Mol. Sci. 2022, 23, 10378. [Google Scholar] [CrossRef]
  130. Kim, J.; Chang, H.S.; Yun, H.J.; Chang, H.J.; Park, K.C. New small-molecule SERCA inhibitors enhance treatment efficacy in Lenvatinib-resistant papillary thyroid cancer. Int. J. Mol. Sci. 2024, 25, 10646. [Google Scholar] [CrossRef]
  131. Peng, S.Y.; Tang, J.Y.; Lan, T.H.; Shiau, J.P.; Chen, K.L.; Jeng, J.H.; Yen, C.Y.; Chang, H.W. Oxidative-stress-mediated ER stress is involved in regulating Manoalide-induced antiproliferation in oral cancer cells. Int. J. Mol. Sci. 2023, 24, 3987. [Google Scholar] [CrossRef] [PubMed]
  132. Kim, T.W. Nodakenin induces ROS-dependent apoptotic cell death and ER stress in radioresistant breast cancer. Antioxidants 2023, 12, 492. [Google Scholar] [CrossRef] [PubMed]
  133. Doan, N.T.; Christensen, S.B. Thapsigargin, origin, chemistry, structure-activity relationships and prodrug development. Curr. Pharm. Des. 2015, 21, 5501–5517. [Google Scholar] [CrossRef]
  134. Isaacs, J.T.; Brennen, W.N.; Christensen, S.B.; Denmeade, S.R. Mipsagargin: The beginning—Not the end—Of Thapsigargin prodrug-based cancer therapeutics. Molecules 2021, 26, 7469. [Google Scholar] [CrossRef] [PubMed]
  135. Mahalingam, D.; Wilding, G.; Denmeade, S.; Sarantopoulas, J.; Cosgrove, D.; Cetnar, J.; Azad, N.; Bruce, J.; Kurman, M.; Allgood, V.E.; et al. Mipsagargin, a novel Thapsigargin-based PSMA-activated prodrug: Results of a first-in-man phase I clinical trial in patients with refractory, advanced or metastatic solid tumours. Br. J. Cancer 2016, 114, 986–994. [Google Scholar] [CrossRef]
  136. Mahalingam, D.; Mahalingam, D.; Arora, S.P.; Sarantopoulos, J.; Peguero, J.; Campos, L.; Cen, P.; Rowe, J.; Allgood, V.; Tubb, B.; et al. A phase II, multicenter, single-arm study of Mipsagargin (G-202) as a second-line therapy following sorafenib for adult patients with progressive advanced hepatocellular carcinoma. Cancers 2019, 11, 833. [Google Scholar] [CrossRef]
  137. NCT02067156; Efficacy, Safety and CNS Exposure of G-202 (Mipsagargin) in Patients with Recurrent or Progressive Glioblastoma. GenSpera, Inc.: San Antonio, TX, USA, 2024.
  138. Chen, P.; Li, Y.; Zhou, Z.; Pan, C.; Zeng, L. Lathyrol promotes ER stress-induced apoptosis and proliferation inhibition in lung cancer cells by targeting SERCA2. Biomed. Pharmacother. 2023, 158, 114123. [Google Scholar] [CrossRef]
  139. Song, S.; Tai, L.; Zhou, L.; Jiang, J.; Zhao, J. Lathyrol affects the expression of AR and PSA and inhibits the malignant behavior of RCC cells. Open Med. 2025, 20, 20241136. [Google Scholar] [CrossRef]
  140. Rani, K.; Chand Sahu, R.; Chaudhuri, A.; Kumar, D.N.; Arora, S.; Kumar, D.; Agrawal, A.K. Exploring combinations of dihydroartemisinin for cancer therapy: A comprehensive review. Biochem. Biophys. Res. Commun. 2025, 765, 151854. [Google Scholar] [CrossRef]
  141. Jang, E.; Lee, J.H. Promising anticancer activities of Alismatis rhizome and its triterpenes via p38 and PI3K/Akt/mTOR signaling pathways. Nutrients 2021, 13, 2455. [Google Scholar] [CrossRef]
  142. Bilmen, J.G.; Khan, S.Z.; Javed, M.H.; Michelangeli, F. Inhibition of the SERCA Ca2+ pumps by curcumin. Curcumin putatively stabilizes the interaction between the nucleotide-binding and phosphorylation domains in the absence of ATP. Eur. J. Biochem. 2001, 268, 6318–6327. [Google Scholar] [CrossRef] [PubMed]
  143. Paula, S.; Floruta, S.; Pajazetovic, K.; Sobota, S.; Almahmodi, D. The molecular determinants of calcium ATPase inhibition by curcuminoids. Biochim. Biophys. Acta Biomembr. 2024, 1866, 184367. [Google Scholar] [CrossRef] [PubMed]
  144. Giordano, A.; Tommonaro, G. Curcumin and cancer. Nutrients 2019, 11, 2376. [Google Scholar] [CrossRef]
  145. Mayo, B.; Penroz, S.; Torres, K.; Simón, L. Curcumin administration routes in breast cancer treatment. Int. J. Mol. Sci. 2024, 25, 11492. [Google Scholar] [CrossRef]
  146. Shi, W.; Wang, Q.; Jiang, S.; Wu, Y.; Li, C.; Xie, Y.; Chen, Q.; Luo, X. Evaluating the efficacy of Curcumin in the management of oral potentially malignant disorders: A systematic review and meta-analysis. PeerJ 2024, 12, e18492. [Google Scholar] [CrossRef]
  147. Seo, J.A.; Kim, B.; Dhanasekaran, D.N.; Tsang, B.K.; Song, Y.S. Curcumin induces apoptosis by inhibiting sarco/endoplasmic reticulum Ca2+ ATPase activity in ovarian cancer cells. Cancer Lett. 2016, 371, 30–37. [Google Scholar] [CrossRef]
  148. Wang, P.; Hao, X.; Li, X.; Yan, Y.; Tian, W.; Xiao, L.; Wang, Z.; Dong, J. Curcumin inhibits adverse psychological stress-induced proliferation and invasion of glioma cells via down-regulating the ERK/MAPK pathway. J. Cell Mol. Med. 2021, 25, 7190–7203. [Google Scholar] [CrossRef]
  149. Wang, L.; Hu, R.; Dai, A. Curcumin increased the sensitivity of non-small-cell lung cancer to Cisplatin through the endoplasmic reticulum stress pathway. Evid. Based Complement. Alternat. Med. 2022, 2022, 6886366. [Google Scholar] [CrossRef]
  150. Serafino, A.; Krasnowska, E.K.; Romanò, S.; De Gregorio, A.; Colone, M.; Dupuis, M.L.; Bonucci, M.; Ravagnan, G.; Stringaro, A.; Fuggetta, M.P. The synergistic combination of curcumin and polydatin improves temozolomide efficacy on glioblastoma cells. Int. J. Mol. Sci. 2024, 25, 10572. [Google Scholar] [CrossRef]
  151. Luís, Â.; Amaral, L.; Domingues, F.; Pereira, L.; Cascalheira, J.F. Action of Curcumin on glioblastoma growth: A systematic review with meta-analysis of animal model studies. Biomedicines 2024, 12, 268. [Google Scholar] [CrossRef]
  152. Gutsche, L.C.; Dörfler, J.; Hübner, J. Curcumin as a complementary treatment in oncological therapy: A systematic review. Eur. J. Clin. Pharmacol. 2024, 81, 1–33. [Google Scholar] [CrossRef]
  153. Judith, P.J.; Maureen, B.; Mélanie, P.; Fabrice, K.; Isabelle, V.D.; Pascale, D.L.; Catherine, A.; Jean-Marc, N.; Hervé, C.; Valérie, D.; et al. Curcumin’s effect in advanced and metastatic breast cancer patients treated with first or second-line Docetaxel: A randomized trial. Health Sci. Rep. 2024, 7, e70052. [Google Scholar] [CrossRef] [PubMed]
  154. Kuzminska, J.; Szyk, P.; Mlynarczyk, D.T.; Bakun, P.; Muszalska-Kolos, I.; Dettlaff, K.; Sobczak, A.; Goslinski, T.; Jelinska, A. Curcumin derivatives in medicinal chemistry: Potential applications in cancer treatment. Molecules 2024, 29, 5321. [Google Scholar] [CrossRef] [PubMed]
  155. Yang, B.; Zhang, M.; Gao, J.; Li, J.; Fan, L.; Xiang, G.; Wang, X.; Wang, X.; Wu, X.; Sun, Y.; et al. Small molecule RL71 targets SERCA2 at a novel site in the treatment of human colorectal cancer. Oncotarget 2015, 6, 37613–37625. [Google Scholar] [CrossRef] [PubMed]
  156. Rezbarikova, P.; Viskupicova, J.; Majekova, M.; Horakova, L. Interaction of quercetin and its derivatives with Ca2+-ATPase from sarcoplasmic reticulum: Kinetic and molecular modeling studies. Gen. Physiol. Biophys. 2023, 42, 457–468. [Google Scholar] [CrossRef]
  157. Silva-Pinto, P.A.; de Pontes, J.T.C.; Aguilar-Morón, B.; Canales, C.S.C.; Pavan, F.R.; Roque-Borda, C.A. Phytochemical insights into flavonoids in cancer: Mechanisms, therapeutic potential, and the case of quercetin. Heliyon 2025, 11, e42682. [Google Scholar] [CrossRef]
  158. Hoinoiu, T.; Dumitrascu, V.; Pit, D.; Schipor, D.A.; Jabri-Tabrizi, M.; Hoinoiu, B.; Petreuș, D.E.; Seiman, C. Quercetin as a potential therapeutic agent for malignant melanoma-a review of current evidence and future directions. Medicina 2025, 61, 656. [Google Scholar] [CrossRef]
  159. Zhang, J.; Guo, J.; Qian, Y.; Yu, L.; Ma, J.; Gu, B.; Tang, W.; Li, Y.; Li, H.; Wu, W. Quercetin induces apoptosis through downregulating P4HA2 and inhibiting the PI3K/Akt/mTOR axis in hepatocellular carcinoma cells: An In Vitro study. Cancer Rep. 2025, 8, e70220. [Google Scholar] [CrossRef]
  160. Xiao, J.; Zhang, B.; Yin, S.; Xie, S.; Huang, K.; Wang, J.; Yang, W.; Liu, H.; Zhang, G.; Liu, X.; et al. Quercetin induces autophagy-associated death in HL-60 cells through CaMKKbeta/AMPK/mTOR signal pathway. Acta Biochim. Biophys. Sin. 2022, 54, 1244–1256. [Google Scholar] [CrossRef]
  161. Hussein, S.A.; Ababneh, N.A.; Tarawneh, N.; Ismail, M.A.; Awidi, A.; Abdalla, S. Antitumor effects of quercetin and luteolin in A375 cutaneous melanoma cell line are mediated by upregulation of P-ERK, c-Myc, and the upstream GPER. Life 2025, 15, 417. [Google Scholar] [CrossRef]
  162. Zhang, C.; Zeng, Y.; Wu, B.; Wang, L.; Wu, P.; Shen, A.; Wang, L. Anti-cancer properties and mechanistic insights of Dihydroquercetin. Curr. Pharm. Biotechnol, 2025; Online ahead of print. [Google Scholar] [CrossRef]
  163. Shi, G.; Wei, J.; Rahemu, S.; Zhou, J.; Li, X. Study on the regulatory mechanism of luteolin inhibiting WDR72 on the proliferation and metastasis of non small cell lung cancer. Sci. Rep. 2025, 15, 12398. [Google Scholar] [CrossRef] [PubMed]
  164. Vatankhah, M.A.; Ziyabakhsh, A.; Vakili Ojarood, M. Regulation of apoptosis, autophagy, and metastasis by luteolin in human bladder cancer EJ138 cells: An experimental study. Iran. J. Pharm. Res. 2024, 23, e153408. [Google Scholar] [CrossRef] [PubMed]
  165. Zhan, X.Z.; Bo, Y.W.; Zhang, Y.; Zhang, H.D.; Shang, Z.H.; Yu, H.; Chen, X.L.; Kong, X.T.; Zhao, W.Z.; Teimonen, T.; et al. Luteolin inhibits diffuse large B-cell lymphoma cell growth through the JAK2/STAT3 signaling pathway. Front. Pharmacol. 2025, 16, 1545779. [Google Scholar] [CrossRef]
  166. Pawar, C.S.; Balamurugan, K.; Baskar, S.; Prasad, N.R.; Khan, H.A. Enhancing chemosensitivity in drug-resistant breast cancer cells using beta-cyclodextrin-loaded Quercetin and Doxorubicin inclusion complex via modulating SRC/PI3K/Akt Pathway. Appl. Biochem. Biotechnol. 2025, 197, 4068–4095. [Google Scholar] [CrossRef]
  167. Tiburzi, S.; Lezcano, V.; Principe, G.; Montiel Schneider, M.G.; Miravalles, A.B.; Lassalle, V.; Bruzzone, A.; González-Pardo, V. Quercetin-loaded magnetic nanoparticles: A promising tool for antitumor treatment in human breast cancer cells. J. Drug Target. 2025, 13, 1–16. [Google Scholar] [CrossRef]
  168. Sun, C.; Xie, F.; Zhang, H.; Feng, L.; Wang, Y.; Huang, C.; Cui, Z.; Luo, C.; Zhang, L.; Wang, Q. Paclitaxel/Luteolin coloaded dual-functional liposomes for esophageal cancer therapy. Adv. Sci. 2025, 29, e2411930. [Google Scholar] [CrossRef]
  169. Mukherjee, A.; Ghosh, S.; Ganguli, S.; Basu, J.; Basu, B. Antiproliferative and apoptotic efficacy of nano-PLGA encapsulated Quercetin molecules by downregulation of Akt in K-ras mutated NSCLC cell lines, A549 and H460. J. Biochem. Mol. Toxicol. 2025, 39, e70240. [Google Scholar] [CrossRef]
  170. Kim, S.M.; Park, K.; Yun, H.J.; Kim, J.M.; Choi, K.H.; Park, K.C. Identification of new small molecules for selective inhibition of SERCA1 in patient-derived metastatic papillary thyroid cancer. Br. J. Pharmacol. 2025, 182, 2392–2408. [Google Scholar] [CrossRef]
  171. Xu, Z.; Shi, Y.; Zhu, L.; Luo, J.; Hu, Q.; Jiang, S.; Xiao, M.; Jiang, X.; Wang, H.; Xu, Y.; et al. Novel SERCA2 inhibitor Diphyllin displays anti-tumor effect in non-small cell lung cancer by promoting endoplasmic reticulum stress and mitochondrial dysfunction. Cancer Lett. 2024, 598, 217075. [Google Scholar] [CrossRef]
  172. Li, Y.; Lu, Q.; Xiao, R.; Ma, J.; Tang, Y.; Chen, W.; Zhang, R.; Jiang, L.; Chen, H.; Shen, B.; et al. Synthesis and anti-tumor activity of nitrogen-containing derivatives of the natural product Diphyllin. Eur. J. Med. Chem. 2022, 243, 114708. [Google Scholar] [CrossRef]
  173. Tang, Y.; Pan, X.; Shang, F.F.; Li, Y.; Zhang, C.; Ma, H.; Zhang, A.; Wang, X.; Ding, C.; Chen, W. Synthesis and pharmacological evaluation of natural product Diphyllin derivatives against head and neck squamous cell carcinoma. Eur. J. Med. Chem. 2025, 285, 117215. [Google Scholar] [CrossRef] [PubMed]
  174. Axten, J.M.; Medina, J.R.; Feng, Y.; Shu, A.; Romeril, S.P.; Grant, S.W.; Li, W.H.; Heerding, D.A.; Minthorn, E.; Mencken, T.; et al. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). J. Med. Chem. 2012, 55, 7193–7207. [Google Scholar] [CrossRef] [PubMed]
  175. Li, Z.; Ge, Y.; Dong, J.; Wang, H.; Zhao, T.; Wang, X.; Liu, J.; Gao, S.; Shi, L.; Yang, S.; et al. BZW1 Facilitates glycolysis and promotes tumor growth in pancreatic ductal adenocarcinoma through potentiating eIF2alpha phosphorylation. Gastroenterology 2022, 162, 1256–1271. [Google Scholar] [CrossRef]
  176. Dastghaib, S.; Shojaei, S.; Mostafavi-Pour, Z.; Sharma, P.; Patterson, J.B.; Samali, A.; Mokarram, P.; Ghavami, S. Simvastatin induces unfolded protein response and enhances Temozolomide-induced cell death in glioblastoma cells. Cells 2020, 9, 2339. [Google Scholar] [CrossRef]
  177. Ketkar, M.; Desai, S.; Rana, P.; Thorat, R.; Epari, S.; Dutt, A.; Dutt, S. Inhibition of PERK-mediated unfolded protein response acts as a switch for reversal of residual senescence and as senolytic therapy in glioblastoma. Neuro Oncol. 2024, 26, 2027–2043. [Google Scholar] [CrossRef]
  178. Yu, Z.Z.; Xu, B.Q.; Wang, Y.Y.; Zhang, P.W.; Shu, Y.B.; Shi, Z. GSK2606414 Sensitizes ABCG2-overexpressing multidrug-resistant colorectal cancer cells to chemotherapeutic drugs. Biomedicines 2023, 11, 3103. [Google Scholar] [CrossRef]
  179. Bagratuni, T.; Patseas, D.; Mavrianou-Koutsoukou, N.; Liacos, C.I.; Sklirou, A.D.; Rousakis, P.; Gavriatopoulou, M.; Terpos, E.; Tsitsilonis, O.E.; Trougakos, I.P.; et al. Characterization of a PERK kinase inhibitor with anti-myeloma activity. Cancers 2020, 12, 2864. [Google Scholar] [CrossRef]
  180. McLaughlin, M.; Pedersen, M.; Roulstone, V.; Bergerhoff, K.F.; Smith, H.G.; Whittock, H.; Kyula, J.N.; Dillon, M.T.; Pandha, H.S.; Vile, R.; et al. The PERK inhibitor GSK2606414 enhances reovirus infection in head and neck squamous cell carcinoma via an ATF4-dependent mechanism. Mol. Ther. Oncolytics 2020, 16, 238–249. [Google Scholar] [CrossRef]
  181. Cai, W.; Sun, X.; Jin, F.; Xiao, D.; Li, H.; Sun, H.; Wang, Y.; Lu, Y.; Liu, J.; Huang, C.; et al. PERK-eIF2α-ERK1/2 axis drives mesenchymal-endothelial transition of cancer-associated fibroblasts in pancreatic cancer. Cancer Lett. 2021, 515, 86–95. [Google Scholar] [CrossRef]
  182. Zhang, X.H.; Wang, X.Y.; Zhou, Z.W.; Bai, H.; Shi, L.; Yang, Y.X.; Zhou, S.F.; Zhang, X.C. The combination of Digoxin and GSK2606414 exerts synergistic anticancer activity against leukemia In Vitro and In Vivo. Biofactors 2017, 43, 812–820. [Google Scholar] [CrossRef]
  183. Xu, L.; Jiang, Y.; Bi, Y.; Zheng, S.; Wu, Y.; Wu, Y.; Xu, Y.; Chen, J. Suppression of PERK/eIF2α/CHOP pathway enhances Oridonin-induced apoptosis by inhibiting autophagy in small-cell lung cancer cells. Biomed. Pharmacother. 2024, 175, 116684. [Google Scholar] [CrossRef] [PubMed]
  184. Axten, J.M.; Romeril, S.P.; Shu, A.; Ralph, J.; Medina, J.R.; Feng, Y.; Li, W.H.; Grant, S.W.; Heerding, D.A.; Minthorn, E.; et al. Discovery of GSK2656157: An optimized PERK inhibitor selected for preclinical development. ACS Med. Chem. Lett. 2013, 4, 964–968. [Google Scholar] [CrossRef] [PubMed]
  185. Atkins, C.; Liu, Q.; Minthorn, E.; Zhang, S.Y.; Figueroa, D.J.; Moss, K.; Stanley, T.B.; Sanders, B.; Goetz, A.; Gaul, N.; et al. Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res. 2013, 73, 1993–2002. [Google Scholar] [CrossRef] [PubMed]
  186. Wang, S.Q.; Wang, X.; Zheng, K.; Liu, K.S.; Wang, S.X.; Xie, C.H. Simultaneous targeting PI3K and PERK pathways promotes cell death and improves the clinical prognosis in esophageal squamous carcinoma. Biochem. Biophys. Res. Commun. 2017, 493, 534–541. [Google Scholar] [CrossRef]
  187. Dudka, W.; Hoser, G.; Mondal, S.S.; Turos-Korgul, L.; Swatler, J.; Kusio-Kobialka, M.; Wołczyk, M.; Klejman, A.; Brewinska-Olchowik, M.; Kominek, A.; et al. Targeting integrated stress response with ISRIB combined with Imatinib treatment attenuates RAS/RAF/MAPK and STAT5 signaling and eradicates chronic myeloid leukemia cells. BMC Cancer 2022, 22, 1254. [Google Scholar] [CrossRef]
  188. Rozpędek-Kamińska, W.; Galita, G.; Siwecka, N.; Granek, Z.; Barczuk, J.; Saramowicz, K.; Majsterek, I. NCI 159456 PERK inhibitor as a targeted therapy for lung cancer: An In Vitro study. Biomedicines 2024, 12, 889. [Google Scholar] [CrossRef]
  189. McCarthy, N.; Dolgikh, N.; Logue, S.; Patterson, J.B.; Zeng, Q.; Gorman, A.M.; Samali, A.; Fulda, S. The IRE1 and PERK arms of the unfolded protein response promote survival of rhabdomyosarcoma cells. Cancer Lett. 2020, 490, 76–88. [Google Scholar] [CrossRef]
  190. Anand, A.A.; Walter, P. Structural insights into ISRIB, a memory-enhancing inhibitor of the integrated stress response. FEBS J. 2020, 287, 239–245. [Google Scholar] [CrossRef]
  191. Ghaddar, N.; Wang, S.; Woodvine, B.; Krishnamoorthy, J.; van Hoef, V.; Darini, C.; Kazimierczak, U.; Ah-Son, N.; Popper, H.; Johnson, M.; et al. The integrated stress response is tumorigenic and constitutes a therapeutic liability in KRAS-riven lung cancer. Nat. Commun. 2021, 12, 4651. [Google Scholar] [CrossRef]
  192. Yang, S.Y.; Liao, L.; Hu, S.Y.; Deng, L.; Andriani, L.; Zhang, T.M.; Zhang, Y.L.; Ma, X.Y.; Zhang, F.L.; Liu, Y.Y.; et al. ETHE1 accelerates triple-negative breast cancer metastasis by activating GCN2/eIF2alpha/ATF4 signaling. Int. J. Mol. Sci. 2023, 24, 14566. [Google Scholar] [CrossRef]
  193. Wang, X.; Chen, T.; Li, C.; Li, W.; Zhou, X.; Li, Y.; Luo, D.; Zhang, N.; Chen, B.; Wang, L.; et al. CircRNA-CREIT inhibits stress granule assembly and overcomes doxorubicin resistance in TNBC by destabilizing PKR. J. Hematol. Oncol. 2022, 15, 122. [Google Scholar] [CrossRef] [PubMed]
  194. Boretti, A.; Banik, B. Maximizing ISRIB Potential requires addressing specificity, long-term safety, and disease-specific considerations. Curr. Med. Chem. 2024. [Google Scholar] [CrossRef] [PubMed]
  195. Matsuoka, M.; Komoike, Y. Experimental evidence shows Salubrinal, an eIF2α dephosphorylation inhibitor, reduces xenotoxicant-induced cellular damage. Int. J. Mol. Sci. 2015, 16, 16275–16287. [Google Scholar] [CrossRef] [PubMed]
  196. da Silva, D.C.; Valentao, P.; Andrade, P.B.; Pereira, D.M. Endoplasmic reticulum stress signaling in cancer and neurodegenerative disorders: Tools and strategies to understand its complexity. Pharmacol. Res. 2020, 155, 104702. [Google Scholar] [CrossRef]
  197. Alsterda, A.; Asha, K.; Powrozek, O.; Repak, M.; Goswami, S.; Dunn, A.M.; Memmel, H.C.; Sharma-Walia, N. Salubrinal exposes anticancer properties in inflammatory breast cancer cells by manipulating the endoplasmic reticulum stress pathway. Front. Oncol. 2021, 11, 654940. [Google Scholar] [CrossRef]
  198. Wu, L.; Liang, C.; Huang, X.; Deng, X.; Jiang, J.; Luo, Z. Salubrinal regulates the apoptosis of adrenocortical carcinoma cells via the PERK/eIF2α/ATF4 signaling pathway. Int. J. Endocrinol. 2021, 2021, 5038130. [Google Scholar] [CrossRef]
  199. Eytan, K.; Versano, Z.; Oren, R.; Jacob-Hirsch, J.; Leitner, M.; Harmelin, A.; Rechavi, G.; Toren, A.; Paglin, S.; Yalon, M. Pediatric glioblastoma cells are sensitive to drugs that inhibit eIF2a dephosphorylation and its phosphomimetic S51D variant. Front. Oncol. 2022, 12, 959133. [Google Scholar] [CrossRef]
  200. Cyran, A.M.; Kleinegger, F.; Nass, N.; Naumann, M.; Haybaeck, J.; Arens, C. Inhibition of EIF2α dephosphorylation decreases cell viability and synergizes with standard-of-care chemotherapeutics in head and neck squamous cell carcinoma. Cancers 2023, 15, 5350. [Google Scholar] [CrossRef]
  201. Chen, M.C.; Hsu, L.L.; Wang, S.F.; Pan, Y.L.; Lo, J.F.; Yeh, T.S.; Tseng, L.M.; Lee, H.C. Salubrinal enhances cancer cell death during glucose deprivation through the upregulation of xCT and mitochondrial oxidative stress. Biomedicines 2021, 9, 1101. [Google Scholar] [CrossRef]
  202. Kardos, G.R.; Gowda, R.; Dinavahi, S.S.; Kimball, S.; Robertson, G.P. Salubrinal in combination with 4E1RCat synergistically impairs melanoma development by disrupting the protein synthetic machinery. Front. Oncol. 2020, 10, 834. [Google Scholar] [CrossRef]
  203. Snyder, C.M.; Mateo, B.; Patel, K.; Fahrenholtz, C.D.; Rohde, M.M.; Carpenter, R.; Singh, R.N. Enhancement of triple-negative breast cancer-specific induction of cell death by silver nanoparticles by combined treatment with proteotoxic stress response inhibitors. Nanomaterials 2024, 14, 1564. [Google Scholar] [CrossRef] [PubMed]
  204. Shao, A.; Xu, Q.; Spalek, W.T.; Cain, C.F.; Kang, C.W.; Tang, C.A.; Del Valle, J.R.; Hu, C.A. Development of tumor-targeting IRE-1 inhibitors for B-cell cancer therapy. Mol. Cancer Ther. 2020, 19, 2432–2444. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, L.; Perera, B.G.; Hari, S.B.; Bhhatarai, B.; Backes, B.J.; Seeliger, M.A.; Schurer, S.C.; Oakes, S.A.; Papa, F.R.; Maly, D.J. Divergent allosteric control of the IRE1a endoribonuclease using kinase inhibitors. Nat. Chem. Biol. 2012, 8, 982–989. [Google Scholar] [CrossRef]
  206. Carlesso, A.; Chintha, C.; Gorman, A.M.; Samali, A.; Eriksson, L.A. Effect of kinase inhibiting RNase attenuator (KIRA) compounds on the formation of face-to-face dimers of inositol-requiring enzyme 1: Insights from computational modeling. Int. J. Mol. Sci. 2019, 20, 5538. [Google Scholar] [CrossRef]
  207. Mimura, N.; Fulciniti, M.; Gorgun, G.; Tai, Y.T.; Cirstea, D.; Santo, L.; Hu, Y.; Fabre, C.; Minami, J.; Ohguchi, H.; et al. Blockade of XBP1 splicing by inhibition of IRE1α is a promising therapeutic option in multiple myeloma. Blood 2012, 119, 5772–5781. [Google Scholar] [CrossRef]
  208. Cross, B.C.; Bond, P.J.; Sadowski, P.G.; Jha, B.K.; Zak, J.; Goodman, J.M.; Silverman, R.H.; Neubert, T.A.; Baxendale, I.R.; Ron, D.; et al. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proc. Natl. Acad. Sci. USA 2012, 109, E869–E878. [Google Scholar] [CrossRef]
  209. Volkmann, K.; Lucas, J.L.; Vuga, D.; Wang, X.; Brumm, D.; Stiles, C.; Kriebel, D.; Der-Sarkissian, A.; Krishnan, K.; Schweitzer, C.; et al. Potent and selective inhibitors of the inositol-requiring enzyme 1 endoribonuclease. J. Biol. Chem. 2011, 286, 12743–12755. [Google Scholar] [CrossRef]
  210. Shao, A.; Xu, Q.; Kang, C.W.; Cain, C.F.; Lee, A.C.; Tang, C.A.; Del Valle, J.R.; Hu, C.A. IRE-1-targeting caged prodrug with endoplasmic reticulum stress-inducing and XBP-1S-inhibiting activities for cancer therapy. Mol. Pharm. 2022, 19, 1059–1067. [Google Scholar] [CrossRef]
  211. Aldin, A.; Besiroglu, B.; Adams, A.; Monsef, I.; Piechotta, V.; Tomlinson, E.; Hornbach, C.; Dressen, N.; Goldkuhle, M.; Maisch, P.; et al. First-line therapy for adults with advanced renal cell carcinoma: A systematic review and network meta-analysis. Cochrane Database Syst. Rev. 2023, 5, CD013798. [Google Scholar] [CrossRef]
  212. Wen, W.; Han, E.S.; Dellinger, T.H.; Lu, L.X.; Wu, J.; Jove, R.; Yim, J.H. Synergistic anti-tumor activity by targeting multiple signaling pathways in ovarian cancer. Cancers 2020, 12, 2586. [Google Scholar] [CrossRef]
  213. Fullana, B.; Morales, S.; Petit, A.; Alay, A.; Verdaguer, H.; Climent, F.; Navarro-Perez, V.; Cejuela, M.; Galvan, P.; Gumà, A.; et al. Neoadjuvant Sunitinib plus Exemestane in post-menopausal women with hormone receptor-positive/HER2-negative early-stage breast cancer (SUT_EXE-08): A phase I/II trial. Sci. Rep. 2024, 14, 23626. [Google Scholar] [CrossRef] [PubMed]
  214. Motzer, R.J.; Porta, C.; Eto, M.; Hutson, T.E.; Rha, S.Y.; Merchan, J.R.; Winquist, E.; Gurney, H.; Grünwald, V.; George, S.; et al. Biomarker analyses from the phase III randomized CLEAR trial: Lenvatinib plus Pembrolizumab versus Sunitinib in advanced renal cell carcinoma. Ann. Oncol. 2025, 36, 375–386. [Google Scholar] [CrossRef] [PubMed]
  215. Wilhelm, T.; Bick, F.; Peters, K.; Mohta, V.; Tirosh, B.; Patterson, J.B.; Kharabi-Masouleh, B.; Huber, M. Infliction of proteotoxic stresses by impairment of the unfolded protein response or proteasomal inhibition as a therapeutic strategy for mast cell leukemia. Oncotarget 2017, 9, 2984–3000. [Google Scholar] [CrossRef] [PubMed]
  216. Harnoss, J.M.; Le Thomas, A.; Shemorry, A.; Marsters, S.A.; Lawrence, D.A.; Lu, M.; Chen, Y.A.; Qing, J.; Totpal, K.; Kan, D.; et al. Disruption of IRE1α through its kinase domain attenuates multiple myeloma. Proc. Natl. Acad. Sci. USA 2019, 116, 16420–16429. [Google Scholar] [CrossRef]
  217. Kern, J.; Schilling, D.; Schneeweis, C.; Schmid, R.M.; Schneiderm, G.; Combs, S.E.; Dobiasch, S. Identification of the unfolded protein response pathway as target for radiosensitization in pancreatic cancer. Radiother. Oncol. 2024, 191, 110059. [Google Scholar] [CrossRef]
  218. Li, X.; Bo, Y.; Zeng, Q.; Diao, L.; Greene, S.; Patterson, J.; Liu, L.; Yang, F. Population pharmacokinetic model for oral ORIN1001 in Chinese patients with advanced solid tumors. Front. Pharmacol. 2024, 15, 1322557. [Google Scholar] [CrossRef]
  219. Logue, S.E.; McGrath, E.P.; Cleary, P.; Greene, S.; Mnich, K.; Almanza, A.; Chevet, E.; Dwyer, R.M.; Oommen, A.; Legembre, P.; et al. Inhibition of IRE1 RNase activity modulates the tumor cell secretome and enhances response to chemotherapy. Nat. Commun. 2018, 9, 3267. [Google Scholar] [CrossRef]
  220. Le Reste, P.J.; Pineau, R.; Voutetakis, K.; Samal, J.; Jégou, G.; Lhomond, S.; Gorman, A.M.; Samali, A.; Patterson, J.B.; Zeng, Q.; et al. Local intracerebral inhibition of IRE1 by MKC8866 sensitizes glioblastoma to irradiation/chemotherapy In Vivo. Cancer Lett. 2020, 494, 73–83. [Google Scholar] [CrossRef]
  221. Vieri, M.; Preisinger, C.; Schemionek, M.; Salimi, A.; Patterson, J.B.; Samali, A.; Brümmendorf, T.H.; Appelmann, I.; Kharabi Masouleh, B. Targeting of BCR-ABL1 and IRE1α induces synthetic lethality in Philadelphia-positive acute lymphoblastic leukemia. Carcinogenesis 2021, 42, 272–284. [Google Scholar] [CrossRef]
  222. Xiao, R.; You, L.; Zhang, L.; Guo, X.; Guo, E.; Zhao, F.; Yang, B.; Li, X.; Fu, Y.; Lu, F.; et al. Inhibiting the IRE1α axis of the unfolded protein response enhances the antitumor effect of AZD1775 in TP53 mutant ovarian cancer. Adv. Sci. 2022, 9, e2105469. [Google Scholar] [CrossRef]
  223. Pavlović, N.; Calitz, C.; Thanapirom, K.; Mazza, G.; Rombouts, K.; Gerwins, P.; Heindryckx, F. Inhibiting IRE1α-endonuclease activity decreases tumor burden in a mouse model for hepatocellular carcinoma. Elife 2020, 9, e55865. [Google Scholar] [CrossRef] [PubMed]
  224. Kopsida, M.; Clavero, A.L.; Khaled, J.; Balgoma, D.; Luna-Marco, C.; Chowdhury, A.; Nyman, S.S.; Rorsman, F.; Ebeling Barbier, C.; Bergsten, P.; et al. Inhibiting the endoplasmic reticulum stress response enhances the effect of D221oxorubicin by altering the lipid metabolism of liver cancer cells. Mol. Metab. 2024, 79, 101846. [Google Scholar] [CrossRef] [PubMed]
  225. Li, X.X.; Zhang, H.S.; Xu, Y.M.; Zhang, R.J.; Chen, Y.; Fan, L.; Qin, Y.Q.; Liu, Y.; Li, M.; Fang, J. Knockdown of IRE1alpha inhibits colonic tumorigenesis through decreasing beta-catenin and IRE1alpha targeting suppresses colon cancer cells. Oncogene 2017, 36, 6738–6746. [Google Scholar] [CrossRef] [PubMed]
  226. Gao, Q.; Li, X.X.; Xu, Y.M.; Zhang, J.Z.; Rong, S.D.; Qin, Y.Q.; Fang, J. IRE1alpha-targeting downregulates ABC transporters and overcomes drug resistance of colon cancer cells. Cancer Lett. 2020, 476, 67–74. [Google Scholar] [CrossRef]
  227. Chien, W.; Ding, L.W.; Sun, Q.Y.; Torres-Fernandez, L.A.; Tan, S.Z.; Xiao, J.; Lim, S.L.; Garg, M.; Lee, K.L.; Kitajima, S.; et al. Selective inhibition of unfolded protein response induces apoptosis in pancreatic cancer cells. Oncotarget 2014, 5, 4881–4894. [Google Scholar] [CrossRef]
  228. Pelizzari-Raymundo, D.; Doultsinos, D.; Pineau, R.; Sauzay, C.; Koutsandreas, T.; Langlais, T.; Carlesso, A.; Gkotsi, E.; Negroni, L.; Avril, T.; et al. A novel IRE1 kinase inhibitor for adjuvant glioblastoma treatment. iScience 2023, 26, 106687. [Google Scholar] [CrossRef]
  229. Okada, T.; Haze, K.; Nadanaka, S.; Yoshida, H.; Seidah, N.G.; Hirano, Y.; Sato, R.; Negishi, M.; Mori, K. A serine protease inhibitor prevents endoplasmic reticulum stress-induced cleavage but not transport of the membrane bound transcription factor ATF6. J. Biol. Chem. 2003, 278, 31024–31032. [Google Scholar] [CrossRef]
  230. Gallagher, C.M.; Garri, C.; Cain, E.L.; Ang, K.K.; Wilson, C.G.; Chen, S.; Hearn, B.R.; Jaishankar, P.; Aranda-Diaz, A.; Arkin, M.R.; et al. Ceapins are a new class of unfolded protein response inhibitors, selectively targeting the ATF6α branch. Elife 2016, 5, e11878. [Google Scholar] [CrossRef]
  231. Zhou, H.; Zhang, T.; Chen, L.; Cui, F.; Xu, C.; Peng, J.; Ma, W.; Huang, J.; Sheng, X.; Liu, M.; et al. The functional implication of ATF6α in castration-resistant prostate cancer cells. FASEB J. 2023, 37, e22758. [Google Scholar] [CrossRef]
  232. Samanta, S.; Yang, S.; Debnath, B.; Xue, D.; Kuang, Y.; Ramkumar, K.; Lee, A.S.; Ljungman, M.; Neamati, N. The hydroxyquinoline analogue YUM70 inhibits GRP78 to induce ER stress-mediated apoptosis in pancreatic cancer. Cancer Res. 2021, 81, 1883–1895. [Google Scholar] [CrossRef]
  233. Ha, D.P.; Huang, B.; Wang, H.; Rangel, D.F.; Van Krieken, R.; Liu, Z.; Samanta, S.; Neamati, N.; Lee, A.S. Targeting GRP78 suppresses oncogenic KRAS protein expression and reduces viability of cancer cells bearing various KRAS mutations. Neoplasia 2022, 33, 100837. [Google Scholar] [CrossRef] [PubMed]
  234. Yamamoto, V.; Wang, B.; Lee, A.S. Suppression of head and neck cancer cell survival and Cisplatin resistance by GRP78 small molecule inhibitor YUM70. Front. Oncol. 2023, 12, 104469. [Google Scholar] [CrossRef] [PubMed]
  235. Yamamoto, V.; Ha, D.P.; Liu, Z.; Huang, M.; Samanta, S.; Neamati, N.; Lee, A.S. GRP78 inhibitor YUM70 upregulates 4E-BP1 and suppresses c-MYC expression and viability of oncogenic c-MYC tumors. Neoplasia 2024, 55, 101020. [Google Scholar] [CrossRef]
  236. Cerezo, M.; Lehraiki, A.; Millet, A.; Rouaud, F.; Plaisant, M.; Jaune, E.; Botton, T.; Ronco, C.; Abbe, P.; Amdouni, H.; et al. Compounds triggering ER stress exert anti-melanoma effects and overcome BRAF inhibitor resistance. Cancer Cell 2016, 29, 805–819. [Google Scholar] [CrossRef]
  237. Ruggiero, C.; Doghman-Bouguerra, M.; Ronco, C.; Benhida, R.; Rocchi, S.; Lalli, E. The GRP78/BiP inhibitor HA15 synergizes with mitotane action against adrenocortical carcinoma cells through convergent activation of ER stress pathways. Mol. Cell. Endocrinol. 2018, 474, 57–64. [Google Scholar] [CrossRef]
  238. Cheng, C.C.; Yang, B.L.; Chen, W.C.; Ho, A.S.; Sie, Z.L.; Lin, H.C.; Chang, C.C. STAT3 mediated miR-30a-5p Inhibition enhances proliferation and inhibits apoptosis in colorectal cancer cells. Int. J. Mol. Sci. 2020, 21, 7315. [Google Scholar] [CrossRef]
  239. Park, J.; Purushothaman, B.; Hong, S.; Choi, M.; Jegal, K.H.; Park, M.; Song, J.M.; Kang, K.W. GRP78 blockade overcomes acquired resistance to EGFR-tyrosine kinase inhibitors in non-small cell lung cancer. Life Sci. 2024, 348, 122681. [Google Scholar] [CrossRef]
  240. Ermakova, S.P.; Kang, B.S.; Choi, B.Y.; Choi, H.S.; Schuster, T.F.; Ma, W.Y.; Bode, A.M.; Dong, Z. (−)-Epigallocatechin gallate overcomes resistance to Etoposide-induced cell death by targeting the molecular chaperone glucose-regulated protein 78. Cancer Res. 2006, 66, 9260–9269. [Google Scholar] [CrossRef]
  241. Le, C.T.; Leenders, W.P.; Molenaar, R.J.; van Noorden, C.J. Effects of the green tea polyphenol Epigallocatechin-3-Gallate on glioma: A critical evaluation of the literature. Nutr. Cancer 2018, 70, 317–333. [Google Scholar] [CrossRef]
  242. Luo, T.; Wang, J.; Yin, Y.; Hua, H.; Jing, J.; Sun, X.; Li, M.; Zhang, Y.; Jiang, Y. (−)-Epigallocatechin gallate sensitizes breast cancer cells to Paclitaxel in a murine model of breast carcinoma. Breast Cancer Res. 2010, 12, R8. [Google Scholar] [CrossRef]
  243. Wu, P.P.; Kuo, S.C.; Huang, W.W.; Yang, J.S.; Lai, K.C.; Chen, H.J.; Lin, K.L.; Chiu, Y.J.; Huang, L.J.; Chung, J.G. (−)-Epigallocatechin gallate induced apoptosis in human adrenal cancer NCI-H295 cells through caspase-dependent and caspase-independent pathway. Anticancer Res. 2009, 29, 1435–1442. [Google Scholar] [PubMed]
  244. La, X.; Zhang, L.; Li, Z.; Li, H.; Yang, Y. (−)-Epigallocatechin gallate (EGCG) enhances the sensitivity of colorectal cancer cells to 5-FU by inhibiting GRP78/NF-κB/miR-155-5p/MDR1 pathway. J. Agric. Food Chem. 2019, 67, 2510–2518. [Google Scholar] [CrossRef] [PubMed]
  245. Chen, T.C.; Wang, W.; Golden, E.B.; Thomas, S.; Sivakumar, W.; Hofman, F.M.; Louie, S.G.; Schönthal, A.H. Green tea Epigallocatechin Gallate enhances therapeutic efficacy of temozolomide in orthotopic mouse glioblastoma models. Cancer Lett. 2011, 302, 100–108. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic diagram illustrating the strategies to modulate endoplasmic reticulum (ER) stress in cancer cells. Shown are the unfolded protein response (UPR) molecular network and the compounds used to induce ER stress or disrupt UPR signaling. SERCA—sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (pump). SERCA transports Ca2+ into the ER lumen, thus maintaining a high ER Ca2+ content but a low cytoplasmic Ca2+ content. Thapsigargin, lathyrol, DHA, alisol B, curcumin, F36, RL71, quercetin, and luteolin—SERCA inhibitors used to induce ER stress. Targeting SERCA causes ER Ca2+ depletion and ER stress, but increases the cytosolic Ca2+ level followed by mitochondrial Ca2+ overload and dysfunction leading to apoptosis. PERK, IRE1α, and ATF6α are ER stress sensors. Upon the accumulation of unfolded/aggregated proteins in the ER lumen (ER stress) they are released from the chaperone GRP78 and trigger multiple signaling cascades involving downstream transcription factors (ATF4, XBP1s, and pATF6α) and associated proteins, while GRP78 interacts with unfolded proteins. In concert, these mechanisms are able to resolve the misfolded ER protein load and restore protein homeostasis (UPR). GSK2606414 and GSK2656157—PERK inhibitors; salubrinal and ISRIB—eIF2α inhibitors; MKC8866, MKC-3946, STF-083010, toyocamycin, 4µ8c, HNA, 3ETH, and Z4P—IRE1α/XBP1 inhibitors; ceapins and AEBSF—ATF6α blockers; YUM70, HA15, and EGCG—GRP78 blockers. The inhibition of ER sensors or the chaperone GRP78 decreases the ability of cells to cope with ER stress and suppresses the transcription of UPR genes, thus inducing persistent ER stress and promoting pro-apoptotic signaling leading to cell death. X—inhibition, ↑—increase, ↓—downregulation, P—phosphorylation.
Figure 1. Schematic diagram illustrating the strategies to modulate endoplasmic reticulum (ER) stress in cancer cells. Shown are the unfolded protein response (UPR) molecular network and the compounds used to induce ER stress or disrupt UPR signaling. SERCA—sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (pump). SERCA transports Ca2+ into the ER lumen, thus maintaining a high ER Ca2+ content but a low cytoplasmic Ca2+ content. Thapsigargin, lathyrol, DHA, alisol B, curcumin, F36, RL71, quercetin, and luteolin—SERCA inhibitors used to induce ER stress. Targeting SERCA causes ER Ca2+ depletion and ER stress, but increases the cytosolic Ca2+ level followed by mitochondrial Ca2+ overload and dysfunction leading to apoptosis. PERK, IRE1α, and ATF6α are ER stress sensors. Upon the accumulation of unfolded/aggregated proteins in the ER lumen (ER stress) they are released from the chaperone GRP78 and trigger multiple signaling cascades involving downstream transcription factors (ATF4, XBP1s, and pATF6α) and associated proteins, while GRP78 interacts with unfolded proteins. In concert, these mechanisms are able to resolve the misfolded ER protein load and restore protein homeostasis (UPR). GSK2606414 and GSK2656157—PERK inhibitors; salubrinal and ISRIB—eIF2α inhibitors; MKC8866, MKC-3946, STF-083010, toyocamycin, 4µ8c, HNA, 3ETH, and Z4P—IRE1α/XBP1 inhibitors; ceapins and AEBSF—ATF6α blockers; YUM70, HA15, and EGCG—GRP78 blockers. The inhibition of ER sensors or the chaperone GRP78 decreases the ability of cells to cope with ER stress and suppresses the transcription of UPR genes, thus inducing persistent ER stress and promoting pro-apoptotic signaling leading to cell death. X—inhibition, ↑—increase, ↓—downregulation, P—phosphorylation.
Ijms 26 06407 g001
Table 1. Examples of aberrant SERCA expression in human cancer tissues.
Table 1. Examples of aberrant SERCA expression in human cancer tissues.
SchemeExpression ProfileCancer TypeClinical OutcomeReferences
SERCA1OverexpressionBreast cancerPoor prognosis
Reduced survival
[30]
Colorectal carcinoma[31]
SERCA2OverexpressionColon and rectal
adenomas and
carcinomas
Lower survival
Invasion
Metastasis
[32,33,34]
SERCA3DownregulationGastric carcinomaMetastasis
Poor prognosis
[35,36]
Glioma/glioblastoma[37]
Colorectal carcinomaNo correlation with survival[38]
Choroid plexus papillomas and carcinomas-[39]
Breast carcinomas[41]
Comments: - not analyzed.
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

Agalakova, N.I. Modulation of Endoplasmic Reticulum Stress in Experimental Anti-Cancer Therapy. Int. J. Mol. Sci. 2025, 26, 6407. https://doi.org/10.3390/ijms26136407

AMA Style

Agalakova NI. Modulation of Endoplasmic Reticulum Stress in Experimental Anti-Cancer Therapy. International Journal of Molecular Sciences. 2025; 26(13):6407. https://doi.org/10.3390/ijms26136407

Chicago/Turabian Style

Agalakova, Natalia Ivanovna. 2025. "Modulation of Endoplasmic Reticulum Stress in Experimental Anti-Cancer Therapy" International Journal of Molecular Sciences 26, no. 13: 6407. https://doi.org/10.3390/ijms26136407

APA Style

Agalakova, N. I. (2025). Modulation of Endoplasmic Reticulum Stress in Experimental Anti-Cancer Therapy. International Journal of Molecular Sciences, 26(13), 6407. https://doi.org/10.3390/ijms26136407

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