Next Article in Journal
Aphanizomenon flos-aquae (AFA) Extract Prevents Neurodegeneration in the HFD Mouse Model by Modulating Astrocytes and Microglia Activation
Next Article in Special Issue
MYCN Amplification, along with Wild-Type RB1 Expression, Enhances CDK4/6 Inhibitors’ Efficacy in Neuroblastoma Cells
Previous Article in Journal
Modified mRNA as a Treatment for Myocardial Infarction
Previous Article in Special Issue
Quiescent Cancer Cells—A Potential Therapeutic Target to Overcome Tumor Resistance and Relapse
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overcoming Acquired Drug Resistance to Cancer Therapies through Targeted STAT3 Inhibition

1
Singh Biotechnology, 1547 Fox Grape Loop, Lutz, FL 33558, USA
2
Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
3
Department of Surgery, Division of Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(5), 4722; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054722
Submission received: 1 February 2023 / Revised: 21 February 2023 / Accepted: 25 February 2023 / Published: 1 March 2023
(This article belongs to the Special Issue Novel Therapeutic Targets in Cancers)

Abstract

:
Anti-neoplastic agents for cancer treatment utilize many different mechanisms of action and, when combined, can result in potent inhibition of cancer growth. Combination therapies can result in long-term, durable remission or even cure; however, too many times, these anti-neoplastic agents lose their efficacy due to the development of acquired drug resistance (ADR). In this review, we evaluate the scientific and medical literature that elucidate STAT3-mediated mechanisms of resistance to cancer therapeutics. Herein, we have found that at least 24 different anti-neoplastic agents—standard toxic chemotherapeutic agents, targeted kinase inhibitors, anti-hormonal agents, and monoclonal antibodies—that utilize the STAT3 signaling pathway as one mechanism of developing therapeutic resistance. Targeting STAT3, in combination with existing anti-neoplastic agents, may prove to be a successful therapeutic strategy to either prevent or even overcome ADR to standard and novel cancer therapies.

1. Introduction

Cancer encompasses a diverse group of diseases with common features and behaviors [1]. Within each histologic type of malignancy, there is often tremendous heterogeneity, which develops from the highly unstable genome of cancer cells. This genomic instability leads to development of cancer cell variants within the bulk tumor. When selection pressure is applied by exposing cancers to anti-neoplastic treatments such as chemotherapeutics, targeted therapeutics, anti-hormone therapeutics, and monoclonal antibodies, clonal selection may occur in patients, and oftentimes acquired drug resistance (ADR) develops.
Drug resistance is a very complex and heterogenous problem that has developed through a variety of many mechanisms. Even within the same patient, several modes of ADR may be present across different tumors [2]. Drug resistance can be intrinsic (i.e., de novo) or conditional to an initial response to an anti-neoplastic agent followed by progression of the cancer—otherwise known as ADR. Signal transducer and activators of transcription 3 (STAT3) has been implicated in the development and maintenance of ADR in multiple cancers in response to various therapies. In this review, we will focus on the role of STAT3 in the development of ADR and clinically relevant drugs that are susceptible to STAT3-mediated ADR. Combination therapy of STAT3 inhibitors with therapies prone to ADR may prove to be synergistic and a compelling strategy to overcome therapeutic resistance in the clinical setting.
Human cells have evolved to develop complex regulatory mechanisms, including positive feedback loops and significant crosstalk among oncogenic signaling pathways. In its simplistic form, ADR can develop when the inhibition of one pathway induces the activation of another, which may impair any therapeutic effect. Twelve pathways, critical to ADR, have been identified, and the STAT3 signaling pathway, described as the “master regulator of antitumor immune response” is one of them [3,4].
In general, the anti-neoplastic agents that have limited efficacy as result of the development of ADR can be divided into four groups: (1) traditional chemotherapeutic drugs, (2) targeted therapeutics, (3) anti-hormone therapeutics, and (4) monoclonal antibodies (Table 1) [5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30]. The traditional chemotherapeutic drugs that develop ADR and are discussed here include doxorubicin [31,32,33,34], gemcitabine [35,36,37,38,39,40,41,42,43,44], cisplatin [34,45,46,47,48,49,50,51,52,53,54,55], temozolomide [56,57,58,59,60], and paclitaxel [61,62,63,64,65]. Targeted therapeutics for which the development of ADR has been documented included in this review are afatinib [37,60,66,67], crizotinib [68], dasatinib [69], and erlotinib [70,71,72,73,74]. The anti-hormone therapeutics that develop ADR are flutamide [75,76,77], enzalutamide [78,79], and tamoxifen [80,81,82,83]. ADR is also developed to monoclonal antibodies such as cetuximab [84,85,86], bevacizumab [87], trastuzumab [88,89,90,91], and to the immune checkpoint inhibitors (ICIs) [92,93,94,95,96,97,98,99,100,101,102,103,104,105,106], such as pembrolizumab [101], nivolumab [102,104], and ipilimumab [103]. In response to all these anti-neoplastic agents, cancer cells utilize STAT3 as one mechanism of escaping their therapeutic effects and promoting ADR. There is a very large body of scientific and medical literature to support the use of anti-STAT3 therapeutics to overcome ADR in these cases. While other mechanisms of ADR exist, here we focus on STAT3 as a key mechanism for the development of ADR.
It should be stated that STAT3 is present in all mammalian cells and plays an important role in physiological functions. Under normal conditions the duration of STAT3 activity is short and transient but in pathological situations, such as cancer, a stronger activation is maintained over long periods of time [117,118]. Activated STAT3 refers to the phosphorylated STAT3 (tyrosine or serine phosphorylated) and is measured and or quantified and described as p-STAT3. This pathological form is referred to in the literature by many different terms such as aberrant, constitutive, dysregulated, etc. STAT3 present in cancer cells is p-STAT3, the constitutively phosphorylated form responsible for the acquired resistance described in this publication [117,118]. STAT3 is located intracellularly, downstream many kinases at an exchanging point of the most important signaling pathways involved in cancer. Oftentimes, when an administered drug blocks a specific kinase pathway, the STAT3 pathway is triggered as result of the crosstalk amongst upstream pathways, resulting in the aberrantly persistent form of p-STAT3.

2. STATs (Signal Transducer and Activators of Transcription)

There are seven STATs (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6) that are intracellular proteins which function as signal messengers and transcription factors. They transmit signals from cytokines, growth factors, intracellular kinases, mutated oncoproteins, and other signaling pathways to the nucleus. Tyrosine phosphorylation cascade occurs after ligand binding by many extracellular molecules such as epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), interleukin-6 (IL-6), IL-5, oncostatin-M (OSM), granulocyte colony stimulating factor (GCSF), colony stimulating factor-1 receptor (CSF1R), leukemia inhibitory factor (LIF), c-kit, c-Met, insulin receptor, angiotensin-II receptor (AgtR2), interferons (IFNs), G-protein coupled receptors (GPCRs), and others. After such ligands bind the extracellular portion of their receptors, their intracellular portion attracts the Janus Kinase family (JAK1, JAK2, JAK3, and Tyk2) of proteins, which become phosphorylated. The JAK protein then phosphorylates STAT3 (pSTAT3) at tyrosine 705 and sometimes serine 727 to activate STAT3. Other intracellular kinases, which can directly activate STAT3 are Src and BCR–ABL, the mutant fusion protein in chronic myelogenous leukemia (CML) [26]. P-STAT3 then forms dimers, which translocate to the nucleus via chaperone proteins. There p-STAT3 dimers bind to specific nine base pair sequences in regulatory genomic regions to regulate transcription of specific genes. The signaling function of p-STAT3 is carefully regulated by inhibitory molecules such as protein inhibitors of activated STAT (PIAS), protein tyrosine phosphatases (PTPases), and suppressors of cytokine signaling (SOCS). Dysregulation of the normal physiologic balance of p-STAT3 and unphosphorylated STAT3 can occur due to upstream mutations or protein overexpression. This results in constitutive expression of p-STAT3 and continuous transcription of pro-oncogenic and anti-apoptotic genes, which promotes cancer growth, proliferation, cell cycle re-entry, angiogenesis, immunosuppression, and, metastasis when anticancer agents apply selective pressure might induce the development of ADR.

3. The Role of STAT3 in Cell Cycle Arrest and Regulation

STAT3 play critical roles within neoplastic cells, immune cells, and other stromal cells, such as cancer-associated fibroblasts (CAFs). Once activated within tumor cells, phosphorylated STAT3 (p-STAT3) regulates the transcription of various immunosuppressive cytokines such as VEGF, IL-10, and TGF-β. p-STAT3 can promote tumor progression by increasing transcription of genes associated with stemness and epithelial to mesenchymal transition (EMT) [119]. Additionally, p-STAT3 is involved in two apoptotic processes, autophagy and anoikis, both contributors to ADR development.
Autophagy, a cellular degradation process, is another regulatory mechanism that plays a major role in maintaining homeostasis, and its dysfunction has been implicated in cancer progression and ADR. The signaling pathways that control inducible autophagy and cell death are closely associated and incorporated into the tumor regulatory network of autophagy related proteins, ultimately affecting the fate of tumor cells [120]. The crosstalk between autophagy and other stress response pathways including STAT3, determines the survival or death of a cell. Nuclear STAT3 regulates autophagy in various forms. For instance, STAT3 inhibits autophagy by activating BCL2 or increases it by upregulating and stabilizing HIF1A under hypoxia; however, it has been determined that cytoplasmatic STAT3 regulates autophagy in a more direct way [121]. Autophagy initially prevents cancer progression but under stressful situations improves the survival of cancer cells [122] contributing to ADR and therapy failure. p-STAT3 has been found to be associated with aberrant autophagy activity in many oncological studies [123]. The anti-autophagy action is partly due to STAT3-mediated inhibition of the BEBN1/PIKC3 complex, resulting in reduced Beclin-1 activity. There is a link between ADR to chemotherapeutics, sometimes described as chemoresistance, and autophagy. The autophagic process vary depending on the tumor stage. In some cases, high dosage chemotherapy may induce protective autophagy that leads to ADR. Some proteins such as mTOR, Beclin-1, miRNA, and autophagy-related genes play a role during treatment of some cancers such as osteosarcoma. The use of autophagy inhibitors in combination with chemotherapeutics is being studied as a new treatment of cancer that might avoid chemoresistance [124]. STAT3 inhibition increases autophagy by increasing transcription of key activators of autophagy. [125]. The importance of autophagy in tumor immunity and ADR is now recognized and has been reported that optimal induction or inhibition of autophagy may induce effective treatments when combined with immunotherapy [126].
Anoikis, another type of apoptosis, is triggered by loss of cell adhesion [127]. It might be activated during tumorigenesis to clear off extracellular matrix (ECM) and detached cells. Cancer cells that develop the ability to survive are called anoikis-resistant cells. These cells become very aggressive and drug resistant, developing the capacity to invade and migrate to metastatic sites. Several features have been identified as responsible for modulating anoikis resistance, one of which is STAT3. STAT3-related anoikis-resistance has been reported in cancer cells of human pancreatic cancer, melanoma, cholangiocarinoma, esophageal squamous cell carcinoma, squamous cell carcinoma, nasopharyngeal carcinoma, and lung carcinoma [128,129,130,131,132]. These cancer cells were reported to have enhanced cell migration, invasion capability and high metastatic potential, and inhibition of STAT3 led to sensitization of all those anoikis-resistant cells [133].
Nicotinamide N-methyltransferase (NNMT) participates in the development of ADR. NNMT, a cytoplasmic enzyme that methylates nicotinamide, is regulated by STAT3 and has been shown to be overexpressed in solid tumors. Furthermore, STAT3 activation intensifies the expression of NNMT and stimulates its activity [134]. NNMT has been identified as an oncogene in intrahepatic cholangiocarcinoma [135]. NNMT is upregulated in cutaneous squamous cell carcinoma, induces cellular invasion via EMT-related gene expression [136] and plays critical roles in the incidence and development of various cancers [137].
Evidence that NNMT plays an important role in cancer can be seen by the fact that NNMT knockdown reduces tumorigenesis and chemoresistance and that Yuanhuadine, a natural inhibitor of NNM, reverses EGFR inhibitors ADR [138]. Chemoresistance or ADR to adriamycin and paclitaxel in breast cancer has been also reported by Wang et al., 2019. This group found that reversal of NNMT related ADR can be accomplished by using the SIRT1 inhibitor, EX527 or using siRNA therapy. SIRT1 also represses the activation of STAT3 and NF-κB proteins via deacetylation [139].
The major function of the tumor suppressor p53 is to induce transient cell cycle arrest, cellular senescence, and apoptosis, a significant barrier to the development of tumors; however, p-STAT3 can inhibit these repressive functions of p53 [140]. This crosstalk between STAT3 and p53 also contributes to the development of ADR and the loss of the pharmacologic effects of anticancer agents [141]. STAT3 inhibition upregulates the expression of p53 and increases cellular apoptotic activity, thereby reversing ADR. Another important signaling pathway for growth and proliferation is the RAS/mitogen activated pathway kinase (MAPK). The crosstalk between STAT3 and p53/RAS signaling regulates metastasis and cisplatin resistance in ovarian cancer through the Slug/MAPK/PI3K/AKT-mediated regulation of EMT and autophagy [142]. Therefore, RAS and STAT3 activation promote ovarian cancer growth, metastasis, and cisplatin resistance. Dual inhibition of STAT3 and KRAS, achieved by nano-antibody SBT-100, would be an ideal treatment for this type of cancer to overcome ADR in ovarian and many other types of cancer [143].
As previously mentioned, p-STAT3-mediated ADR occurs in response to anti-neoplastic agent therapy by utilizing multiple intracellular signaling pathways. As illustrated in Figure 1, treatment with a receptor tyrosine kinase inhibitor, which blocks MAPK pathways, results in the cancer cells secreting ligands, which bind to receptors on the cancer cells themselves in an autocrine fashion or to CAFs, intratumor macrophages, and other cells in the tumor microenvironment (TME) in a paracrine fashion. This ligand binding to its cognate receptor results in STAT3 activation, turning on numerous genes that promote growth, proliferation, cell cycle re-entry, anti-apoptosis, angiogenesis, immunosuppression, and metastasis, and ultimately circumventing the anti-neoplastic therapy being used resulting in ADR.

4. ADR Development to Chemotherapeutics

Doxorubicin is an anthracycline compound and currently one of the most effective classes of anti-cancer agents in clinical applications; however its use is limited by its chronic and acute toxicities [107]. It binds to topoisomerase I and II, resulting in intercalation of the base pairs of the DNA double helix and inhibition DNA replication. Because of this mechanism of action, doxorubicin has been highly effective in treating a wide variety of malignancies. Its efficacy is unfortunately limited in many cases by ADR due to STAT3 upregulation. A well-known STAT3 inhibitor, Stattic, was formulated in nanostructured lipid carriers to enhance the efficacy of doxorubicin against melanoma cancer cells [108,109]. Doxorubicin induces p-STAT3 in human breast cancer MCF cell line (ER+, non-metastatic) and human triple negative breast cancer MDA-MB-231 cell line (metastatic) [110]. The p-STAT3 was then suppressed by tyrphostin AG490 (an inhibitor of the upstream activating Janus kinases), transfection with a dominant-negative form of STAT3, and with satraplatin (a tetravalent platinum derivate that inhibits STAT3 phosphorylation) [110]. These treatments downregulated p-STAT3 and sensitized the cancer cells to doxorubicin.
Alantolactone (ALT), a sesquiterpene lactone component of Inula helenium, has anti-neoplastic effect against a variety of malignancies. Mechanistic research demonstrated that ALT abrogated STAT3 phosphorylation by promoting STAT3 glutathionylation. Reactive oxygen species scavenger NAC reverted ALT-mediated STAT3 glutathionylation and abrogation of STAT3 activation. With lung adenocarcinoma (A549 cell line), STAT3 inhibition by ALT enhanced chemosensitivity to doxorubicin via oxidative stress [111]. In the above three examples, genes transcribed by p-STAT3 dimers that are necessary for malignant cell behavior include BCL2L1 (Bcl-xL), BIRC5 (survivin), HIF1A, HIF1B (HIF-1), and MMP9 had their expression reduced [145].
Human osteosarcoma (SJSA-1) tumors when treated in vivo with doxorubicin undergoes significant growth suppression during a 14-day course of treatment; however, only 28% of the treated mice survived the 3-week xenograft study. The doxorubicin-associated toxicity was killing the mice. It is not clear if doxorubicin’s effect on normal cells caused this, or the induction of p-STAT3 in the SJSA-1 cells or a combination of both may have contributed to the death of the mice. When xenograft mice received doxorubicin with SBT-100, a STAT3 inhibitor, the osteosarcoma tumor growth was significantly suppressed, and survival of the mice increased to 71%. By some mechanism, SBT-100 was protecting the mice from doxorubicin toxicity. SBT-100 is a camelid derived single domain antibody that penetrates the cell membrane and blood brain barrier (BBB) rapidly in vivo and inhibits STAT3. SBT-100 has broad range of efficacy against many human malignancies such as ER + PR+ breast cancer, HER2-amplified breast cancer, triple negative breast cancer (TNBC), pancreatic cancer, prostate cancer, glioblastoma, osteosarcoma, fibrosarcoma, and leukemia [143].
Cisplatin is a platinum-based anti-neoplastic agent that binds DNA and inhibits its replication. It is used to treat ovarian, cervical, testicular, head and neck, colorectal, esophageal, bladder, lung, and breast cancers. Some mechanisms by which cisplatin resistance can develop include decreased drug import, increased drug export, increased DNA damage repair, increased drug inactivation by detoxification enzymes, and inactivation of cell death signaling, which occur within cancer cells [146]. Another mechanism of cisplatin resistance involves STAT3 overexpression. Sun et al have summarized utilization of STAT3 inhibition to reverse cisplatin induced resistance [52]. They summarized a large variety of STAT3 inhibitors which reverse cisplatin resistance in lung cancer, ovarian cancer, cervical cancer, breast cancer, laryngeal cancer, head and neck cancers, esophageal cancer, and hepatocellular carcinoma [52].
Morelli et al found, through network analysis and classification of proteome analysis of A549 cells (lung adenocarcinoma), that there were pathways altered in cisplatin resistant A549 cells. The resistance profile of these A549 cells included STAT3 overexpression. Furthermore, p-STAT3 is a marker of poor prognosis and cisplatin resistance in lung cancer. Generation of A549 STAT3 knockout cells resulted in impairment of clonogenic survival and mesenchymal phenotype in these A549 cells. These STAT3 knockouts do not develop cisplatin resistance nor over activation of mammalian target of rapamycin (mTOR) signaling with cis treatment. Moreover, the A549 knockout cells are more sensitive to mTOR inhibition by rapamycin [147].
Ovarian cancer can be effectively treated with paclitaxel; however, the development of resistance remains a major problem. Sheng et al have shown that STAT3 directly activates the pentose–phosphate pathway to cause pro-oncogenic behavior of paclitaxel resistant ovarian cancer [112]. Furthermore, they discovered that STAT3, p-STAT3, and glucose-6-phosphate dehydrogenase (G6PD) protein levels are increased in paclitaxel resistant cell lines versus paclitaxel sensitive cell lines. Blocking STAT3 decreased G6PD mRNA expression and increased the sensitivity of paclitaxel resistant ovarian cancer cells to paclitaxel. In addition, they demonstrated that STAT3 directly binds to the G6PD DNA promoter region and increases the expression of G6PD at the transcriptional level. In summary, their research reveals overexpression of STAT3 increases ovarian cancer colony formation, proliferation, and resistance to paclitaxel by increasing G6PD expression and pentose–phosphate metabolism flux [112].
With cervical cancer, paclitaxel is an important chemotherapeutic agent, but here too resistance to paclitaxel develops. Fan et al compared microRNA (miRNA) expression in cervical cancer cell lines to their paclitaxel resistant cervical cancer counterparts [113]. They found miR-125a to be a significantly decreased miRNA among paclitaxel-resistant cervical cancer cells and these cells also developed cisplatin resistance. Upregulating miR-125a sensitized resistant cervical cancers to paclitaxel in vitro and in vivo and to cisplatin in vitro. Importantly, they showed miR-125a increased sensitivity of cervical cancers to paclitaxel and cisplatin by decreasing STAT3. MiR-125a improved paclitaxel and cisplatin sensitivity by causing chemotherapy induced apoptosis. Clinically, miR-125a expression was linked to increased responsiveness to cisplatin combined with paclitaxel and this resulted in improved outcome. Their data suggests that miR-125a may provide a method which allows treatment of resistant cervical cancer. In addition, miR-125a may function as a biomarker for predicting resistance to cisplatin and paclitaxel in cervical cancer patients.
Temozolomide, a chemotherapeutic that penetrates the BBB is used for the treatment of the heterogenous glioblastoma and anaplastic astrocytoma. Despite treatment with surgery, chemotherapy, and radiation, survival is maximum 15 months. Hyperactivated STAT3 has been demonstrated to modulate the behavior of gliomas and promote ADR and the STAT3 inhibitor pacritinib in combination with temozolimide has been shown to be effective in glioblastoma overwhelming STAT3/miR-21/PDCD4 signaling [114,115,148]. Moreover, the antipsychotic pimozide, a repurposed STAT3 inhibitor, reduces STAT3, triggers an autophagy-dependent, lysosomal type of cell death and improves survival in GBM cells [149,150]. A rational therapy for the treatment of glioblastoma would be the combination of temozolomide with the STAT3 inhibitor SBT-100, two anticancer compounds that penetrate the BBB [143].

5. ADR Development to Targeted Therapies

Sun et al have described numerous studies that have shown that STAT3 activation can result in the failure of many different types of targeted therapies, especially EGFR targeted therapies [20,52]. For example, afatinib-induced STAT3 activation decreases the suppression of lung cancer cells to afatinib, and inhibiting IL-6R/JAK1/STAT3 signaling reverses the resistance. Blocking STAT3 can prevent ADR induced by EGFR inhibitors. Rhein, a lipophilic anthraquinone, sensitizes pancreatic cancer cells to erlotinib by inhibiting STAT3. Alantolactone, a natural sesquiterpene lactone, also sensitizes pancreatic cancer cells to erlotinib and also to afatinib by inhibiting STAT3 [116]. Silibinin, a polyphenolic flavonoid, is a direct inhibitor of STAT3, and it reverses ADR of crizotinib in ALK-rearranged lung cancer cells [68]. In addition, silibinin synergistically improves the response to sorafenib by hepatocellular carcinoma (HCC) cells by blocking STAT3 [151].
Pancreatic adenocarcinoma (PDAC) is a lethal malignancy that presents in late stages and responds poorly to current therapeutic regimens with an overall 5-year survival of 11%. It is characterized by an extensively dense fibrotic tumor stroma with poor vascularity, which hinder the intratumor delivery of anti-neoplastic agents [152]. Examining the response of PDAC to monotherapy with gemcitabine, dasatinib (Src inhibitor), and erlotinib (EGFR inhibitor) reveals that the upregulation of p-STAT3 is causing ADR. When combination therapy of dasatinib and erlotinib is used p-STAT3 is downregulated. This results in tumor collagen (types 1 and 4) and fibrosis to decrease within the tumor stroma in an orthotopic mouse model of PDAC. Here Dosch et al used PANC-1, which has a KRAS (G12D) mutation and constitutive expression of p-STAT3, and BxPC3, which has wild type KRAS and constitutive expression of p-STAT3. Interestingly, the addition of gemcitabine to combine with dasatinib and erlotinib therapy did not reverse the antifibrotic effects of this drug combination [69]. This two-drug combination inhibits the EGFR and Src signaling pathway and reduces p-STAT3. In turn, an increase in tumor vascularity occurs in vivo, and to determine this, treated PDAC were examined for CD31 (PECAM-1) by immunohistochemical (IHC) staining. CD31 is an endothelial marker that is associated with vascular normalization, maturity, and has been correlated with chemotherapeutic response in PDAC [11,28]. Monotherapy with dasatinib or erlotinib versus control showed no significant increase in CD31 positive staining; however, combined in vivo treatment with dasatinib plus erlotinib resulted in significant increase in CD31 positive endothelial cells. This finding was sustained with gemcitabine added to the two-drug combination. Dosch et al showed levels of gemcitabine is nearly undetectable in tumors treated with erlotinib or dasatinib monotherapy or in combination with gemcitabine [69]. When combination therapy with dasatinib plus erlotinib was administered, a marked increase in gemcitabine levels within PDAC tumors was detected. These findings demonstrated that combined Src and EGFR inhibition decreases p-STAT3 activity, which increases the microvascular density within PDAC tumors, which ultimately results in increased delivery of cytotoxic chemotherapy into the tumor mass.
The above orthotopic PDAC studies were conducted on athymic nude mice. Transgenic PKT mice (Ptf1aCre/+; LSL-KrasG12D/+; Tgfbr2flox/flox) were used for in vivo studies to examine tumor volume with the pancreas and overall survival of combined Src and EGFR inhibition. This immunocompetent, spontaneous mouse model of PDAC underwent treatment with dasatinib, erlotinib, and gemcitabine either alone or in combination. This therapy was continued for 4 weeks, after which the mice were sacrificed and the PDAC tumors harvested for histo-pathology evaluation. In PKT tumors, dasatinib plus erlotinib, and dasatinib plus erlotinib with gemcitabine treatments significantly reduced tumor weight at the end of the study. Furthermore, stromal remodeling of the PDAC tumors occurred as it did in the orthotopic tumors with decreased stromal fibrosis, decreased collagen type 1 and 4, increased microvascular density, and increased number of CD31 positive endothelial cells. Moreover, p-STAT3 levels were significantly decreased with combined treatment of dasatinib plus erlotinib, and dasatinib plus erlotinib with gemcitabine in the PKT tumor samples. These two combination regimens also prevented the progression of PDAC tumors in the PKT mice and increased their overall survival. Furthermore, they have previously shown tumor cell-derived IL1α induces stromal-derived IL-6, reciprocally activating tumor cell-autonomous STAT3 signaling, a well-known indicator of chemoresistance and oncogenic signaling in PDAC [126,133,153].
Lee et al performed extensive and elegant work on defining STAT3 as an escape mechanism for many cancers treated with targeted pharmaceutical therapeutics [154]. They discovered that many drug treated “oncogene addicted” malignancies use a positive feedback loop resulting in STAT3 hyperactivation. As a result, promoting cancer cell proliferation, survival, and decreasing response to targeted drug therapy. This was noted in malignant cells driven by different activated kinases such as HER2, EGFR, MET, ALT, and mutant KRAS [10,13]. MEK inhibition resulted in autocrine activation of STAT3 via FGFR and JAK kinases. Importantly, simultaneous drug suppression of MEK, JAK, and FGFR increased tumor regression. Their data implies that blocking the STAT3 feedback loop enhances the response to a wide range of pharmacologic therapeutics that inhibit pathways of oncogene addiction.

6. ADR Development to Monoclonal Antibody Treatment

Monoclonal antibodies (mAbs) to cell surface receptors, [e.g., human epidermal growth factor receptor 2 (HER2)], and to extracellular protein, e.g., vascular endothelial growth factor (VEGF), have greatly improved the treatment of patients with cancer. HER2 is a receptor tyrosine kinase (RTK) that controls differentiation and cell growth signaling pathways. In about 20–25% of breast cancers and in 30% of gastric cancers, HER2 is significantly overexpressed. This results in a very aggressive cancer phenotype and poor prognosis. Trastuzumab is a humanized mAb that binds to an extracellular domain of the HER2 molecule and inhibits its function. It provides significant benefit in patient outcome; however, treatment resistance does develop in some patients. Li et al found that p-STAT3 is hyper-expressed in de novo and acquired trastuzumab-resistant gastric cancer and breast cancer cells [96]. Here, increased STAT3 activation and signaling is caused by elevated levels of IL-6, fibronectin (FN), and EGF in an autocrine manner. This leads to ADR by upregulating the expression of MUC1 and MUC4. Both are downstream targets of p-STAT3. MUC1 and MUC4 can induce trastuzumab resistance by maintaining HER2 activation and masking of trastuzumab to prevent HER2 binding, respectively. Knocking down STAT3 expression and blocking STAT3 function with a small molecule inhibitor abrogated STAT3 activation, which allowed trastuzumab sensitivity of resistant cells in vitro and in vivo [89].
Trastuzumab–emtansine (T-DM1) is an antibody drug conjugate made with the trastuzumab mAb linked to a cytotoxic moiety DM1 (a derivative of maytansine) and it was developed to overcome ADR associated with trastuzumab use. T-DM1 has demonstrated great efficacy clinically; however, ADR to its use has emerged and is a significant problem for patients. Wang et al used BT-474/KR cells, a T-DM1 resistant cell line developed from HER2-positive BT-474 breast cancer cells, to show that STAT3 activation mediated by leukemia inhibitory factor receptor (LIFR) overexpression results in T-DM1 resistance. Furthermore, they demonstrated STAT3 inhibition sensitizes resistant cell to T-DM1 both in vitro and in vivo [90].
Anti-VEGF treatments help several types of cancer patients, but ADR can develop with therapy. There are several VEGF pathway inhibitors, which include bevacizumab (anti-VEGF mAb), aflibercept (decoy receptor that binds VEGF-A), and ramucirumab (anti-VEGF receptor 2 mAb), which inhibit tumor growth in preclinical cancer models and improve cancer patients’ survival. Eichten et al developed cell lines from anti-VEGF resistant tumor xenografts and one called A431-V epidermoid carcinoma developed partial resistance to aflibercept [87]. A431-V tumors secreted much more IL-6 and produced higher amounts of p-STAT3 compared to parental tumors. Combined inhibition of IL-6 receptor (IL-6R) and VEGF resulted in enhanced suppression of A431-V tumors. In addition, inhibition of IL-6R increased the suppression of DU145 prostate cancer cells using aflibercept. These DU145s have high endogenous IL-6R activity. These data indicate that ADR to anti-VEGF therapy is mediated in part by increased IL-6/STAT3 signaling in cancer cells. Inhibition of IL-6 signaling on cancer cells can overcome this ADR.
Immune checkpoint inhibitors (ICIs) provide significant benefit to some cancer patients and improve survival in a minority of patients. Moreover, some might even be cured [99]. Tumor-intrinsic resistance is the reason for the lack of response [97]; however, when ICIs are used for the first time, less than 45% respond and most responders eventually develop ADR [101]. ADR has been reported in several types of cancer patients and animal models treated with ICIs due to overactivation of STAT3. This undesirable effect has been observed in the case of anti-PD-1, anti PD-L1, and anti-CTLA-4 antibodies. STAT3 can directly or indirectly regulate these immune checkpoint molecules. There is a clear relation between STAT3 and PD-1, PD-L1, and PD-L2 [6]. STAT3 can increase their expression by direct binding to their promoters [105]. STAT3 binds to the CD274 (PD-L1) gene promoter and is required for CD274 gene expression. The NPM/ALK carrying T cell lymphoma (ALK + TCL) cells strongly express PD-L1 that is regulated by STAT3 [100]. These investigators at that time were already suggesting that the treatment of this lymphoma should combine inhibition of both NPM/ALK and STAT3. Under hypoxic conditions overactivated STAT3 interacts with PD-L1 and enables its nuclear translocation by the importin α and β pathways [95]. It was shown that in nasopharyngeal carcinoma, LMP1 upregulates PD-L1 through STAT3, AP-1, and NF-kB [94]. In gastric cancer, the suppressor gene miR-502-5p reduces PD-L1 expression through inhibition of the CD40/STAT3 pathway [106].
To obtain better results, combinations must be used. Nivolumab plus ipilimumab improved outcomes in 43% of patients with metastatic renal cell carcinoma but ADR was found in the rest of patients receiving this combination. Nonresponders exhibited significant increases in cytokines and higher levels of p-STAT3. The addition of a STAT3 inhibitor to the combination of the two ICIs showed significant tumor growth inhibition in a syngeneic model [92]. These investigators suggest that anti-PD-1 therapy administered along with a STAT3 inhibitor is a rational combination and should be further explored. In patients with melanoma, less than 20% respond to ICIs. Studies in a melanoma mouse model showed that the addition of STAT3 inhibitors to an ICI increases the response to the ICI-resistant tumor. These data suggest that the combination of ICIs with STAT3 inhibitors might be effective in patients with melanoma [96]. In drug-resistant BRAF-mutant melanoma, a combined blockade of STAT3 and PD-1 overcomes resistance [8,14,19,98]. In PDAC, the addition of MEK inhibitors plus STAT3 inhibitors to Nivolumab overcomes ICI resistance [93].
Ipilimumab is efficacious only in a subset of patients with prostate cancer. In a syngeneic prostate cancer mouse model, the combination of an anti-CTLA-4 with a STAT3 inhibitor significantly inhibited tumor growth and enhanced survival possibly by blocking STAT3 mediated resistance mechanisms such as Tregs in the immunosuppressive environment. These investigators raise the possibility that STAT3 inhibition in combination with anti-CTLA-4 could constitute a future novel treatment approach in advanced prostate cancer [103]. In summary, it appears that combining a STAT3 inhibitor with an ICI is an attractive way to prevent the development of ADR and increase their efficacy [92,93,96,106].

7. Discussion

Several mechanisms involved in the development of ADR have been studied in animal models of several types of cancer and in patients. As result of the unique cytoplasmatic location of the STAT3 signaling pathway downstream of major pathways involved in cancer and the significant crosstalk that occurs among them, it is activated by the inhibition of many other pathways and in most types of cancers becomes the constitutively activated p-STAT3. This explains why the administration of any anticancer therapeutic and the inhibition of its specific pathway through the crosstalk relations induces the production of p-STAT3 that appears to be responsible for the various changes that end in the development of ADR and the loss of their therapeutic effects. p-STAT3 alters autophagy and anoikis, two apoptotic processes that normally eliminates unwanted cells and the lack or reduction of their actions contribute to ADR and the progression of the tumor or hematological cancer. The physiological relation between STAT3, and p53, or NNMT is altered when STAT3 becomes constitutively activated. These alterations act as factors contributing to the development of ADR. Participation of STAT3 in ARD after administration of any type of anticancer therapy—including the newer targeted agents such as the eight new ICIs and the two KRAS inhibitors—indicates that the use of a STAT3 inhibitor should be part of any rational pharmacological treatment including radiotherapy. The inclusion of a STAT3 inhibitor in anticancer regimens increases their efficacy and most likely prevents the development of ADR. Some natural products, such as curcumin, are STAT3 inhibitors. A limitation of curcumin is the low oral bioavailability; however, new delivery technologies have improved it oral absorption [155]. When ADR is already present, the administration of a STAT3 inhibitor reverses it and restores the efficacy of the anticancer therapeutic.
Many investigators have shown that STAT3 inhibition can reverse ADR, restore the efficacy of anticancer agents [118,156], enhance anti-cancer immune responses, and rescue the suppressed immunologic system [157]. The treatment of cancer usually requires combination therapy, and two or more agents might be needed. As soon as direct STAT3 inhibitors reach the market, a rational combination for the pharmacologic treatment of cancer patients should include a STAT3 inhibitor to prevent and or reverse ADR and thereby increase the efficacy and duration of the therapeutic regimen.

Author Contributions

Conception and design: S.S., H.J.G., S.T., S.P.S. and A.S.P. Writing, review, and revision of manuscript: S.S., H.J.G., S.T., S.P.S. and A.S.P. Administrative, technical, or material support: S.S., S.P.S. Study supervision: S.S., H.J.G., A.S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Figure 1 was created with BioRender on 29 January 2023. Agreement number: MS24Y72CSM (www.biorender.com).

Conflicts of Interest

Sunanda Singh, Hector J. Gomez, and Ashutosh S. Parihar are employed by and shareholders of Singh Biotechnology.

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Floros, K.V.; Hata, A.N.; Faber, A.C. Investigating New Mechanisms of Acquired Resistance to Targeted Therapies: If You Hit Them Harder, Do They Get Up Differently? Cancer Res. 2020, 80, 25–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Rebe, C.; Ghiringhelli, F. STAT3, a Master Regulator of Anti-Tumor Immune Response. Cancers 2019, 11, 1280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef] [PubMed]
  5. Cao, W.; Liu, Y.; Zhang, R.; Zhang, B.; Wang, T.; Zhu, X.; Mei, L.; Chen, H.; Zhang, H.; Ming, P.; et al. Homoharringtonine induces apoptosis and inhibits STAT3 via IL-6/JAK1/STAT3 signal pathway in Gefitinib-resistant lung cancer cells. Sci. Rep. 2015, 5, 8477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Chen, Y.; Li, F.; Li, D.; Liu, W.; Zhang, L. Atezolizumab and blockade of LncRNA PVT1 attenuate cisplatin resistant ovarian cancer cells progression synergistically via JAK2/STAT3/PD-L1 pathway. Clin. Immunol. 2021, 227, 108728. [Google Scholar] [CrossRef]
  7. Chiu, H.C.; Chou, D.L.; Huang, C.T.; Lin, W.H.; Lien, T.W.; Yen, K.J.; Hsu, J.T. Suppression of Stat3 activity sensitizes gefitinib-resistant non small cell lung cancer cells. Biochem. Pharmacol. 2011, 81, 1263–1270. [Google Scholar] [CrossRef]
  8. Hartsough, E.J.; Aplin, A.E. A STATement on vemurafenib-resistant melanoma. J. Investig. Dermatol. 2013, 133, 1928–1929. [Google Scholar] [CrossRef] [Green Version]
  9. Hatiboglu, M.A.; Kong, L.Y.; Wei, J.; Wang, Y.; McEnery, K.A.; Fuller, G.N.; Qiao, W.; Davies, M.A.; Priebe, W.; Heimberger, A.B. The tumor microenvironment expression of p-STAT3 influences the efficacy of cyclophosphamide with WP1066 in murine melanoma models. Int. J. Cancer 2012, 131, 8–17. [Google Scholar] [CrossRef] [Green Version]
  10. Huang, W.C.; Hung, C.M.; Wei, C.T.; Chen, T.M.; Chien, P.H.; Pan, H.L.; Lin, Y.M.; Chen, Y.J. Interleukin-6 expression contributes to lapatinib resistance through maintenance of stemness property in HER2-positive breast cancer cells. Oncotarget 2016, 7, 62352–62363. [Google Scholar] [CrossRef] [Green Version]
  11. Huser, L.; Kokkaleniou, M.M.; Granados, K.; Dworacek, J.; Federico, A.; Vierthaler, M.; Novak, D.; Arkhypov, I.; Hielscher, T.; Umansky, V.; et al. HER3-Receptor-Mediated STAT3 Activation Plays a Central Role in Adaptive Resistance toward Vemurafenib in Melanoma. Cancers 2020, 12, 3761. [Google Scholar] [CrossRef]
  12. Li, C.; Zang, Y.; Sen, M.; Leeman-Neill, R.J.; Man, D.S.; Grandis, J.R.; Johnson, D.E. Bortezomib up-regulates activated signal transducer and activator of transcription-3 and synergizes with inhibitors of signal transducer and activator of transcription-3 to promote head and neck squamous cell carcinoma cell death. Mol. Cancer Ther. 2009, 8, 2211–2220. [Google Scholar] [CrossRef] [Green Version]
  13. Liao, Q.; Feng, X.; Li, X.; Chen, G.; Chen, J.; Yang, B.; Li, K.; Ai, J. Lapatinib-induced inhibition of ovarian function is counteracted by the STAT3 pathway both in vivo and in vitro. Oncol. Rep. 2020, 44, 1127–1135. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, F.; Cao, J.; Wu, J.; Sullivan, K.; Shen, J.; Ryu, B.; Xu, Z.; Wei, W.; Cui, R. Stat3-targeted therapies overcome the acquired resistance to vemurafenib in melanomas. J. Investig. Dermatol. 2013, 133, 2041–2049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Liu, Z.; Ma, L.; Sun, Y.; Yu, W.; Wang, X. Targeting STAT3 signaling overcomes gefitinib resistance in non-small cell lung cancer. Cell Death Dis. 2021, 12, 561. [Google Scholar] [CrossRef] [PubMed]
  16. Manni, S.; Brancalion, A.; Mandato, E.; Tubi, L.Q.; Colpo, A.; Pizzi, M.; Cappellesso, R.; Zaffino, F.; Di Maggio, S.A.; Cabrelle, A.; et al. Protein kinase CK2 inhibition down modulates the NF-kappaB and STAT3 survival pathways, enhances the cellular proteotoxic stress and synergistically boosts the cytotoxic effect of bortezomib on multiple myeloma and mantle cell lymphoma cells. PLoS ONE 2013, 8, e75280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Mencalha, A.L.; Correa, S.; Salles, D.; Du Rocher, B.; Santiago, M.F.; Abdelhay, E. Inhibition of STAT3-interacting protein 1 (STATIP1) promotes STAT3 transcriptional up-regulation and imatinib mesylate resistance in the chronic myeloid leukemia. BMC Cancer 2014, 14, 866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Sayed, D.; Badrawy, H.; Gaber, N.; Khalaf, M.R. p-Stat3 and bcr/abl gene expression in chronic myeloid leukemia and their relation to imatinib therapy. Leuk. Res. 2014, 38, 243–250. [Google Scholar] [CrossRef] [PubMed]
  19. Sinnberg, T.; Makino, E.; Krueger, M.A.; Velic, A.; Macek, B.; Rothbauer, U.; Groll, N.; Potz, O.; Czemmel, S.; Niessner, H.; et al. A Nexus Consisting of Beta-Catenin and Stat3 Attenuates BRAF Inhibitor Efficacy and Mediates Acquired Resistance to Vemurafenib. EBioMedicine 2016, 8, 132–149. [Google Scholar] [CrossRef] [Green Version]
  20. Song, X.; Tang, W.; Peng, H.; Qi, X.; Li, J. FGFR leads to sustained activation of STAT3 to mediate resistance to EGFR-TKIs treatment. Investig. New Drugs 2021, 39, 1201–1212. [Google Scholar] [CrossRef]
  21. Vangala, J.R.; Dudem, S.; Jain, N.; Kalivendi, S.V. Regulation of PSMB5 protein and beta subunits of mammalian proteasome by constitutively activated signal transducer and activator of transcription 3 (STAT3): Potential role in bortezomib-mediated anticancer therapy. J. Biol. Chem. 2014, 289, 12612–12622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, X.; Goldstein, D.; Crowe, P.J.; Yang, M.; Garrett, K.; Zeps, N.; Yang, J.L. Overcoming resistance of targeted EGFR monotherapy by inhibition of STAT3 escape pathway in soft tissue sarcoma. Oncotarget 2016, 7, 21496–21509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wu, K.; Chang, Q.; Lu, Y.; Qiu, P.; Chen, B.; Thakur, C.; Sun, J.; Li, L.; Kowluru, A.; Chen, F. Gefitinib resistance resulted from STAT3-mediated Akt activation in lung cancer cells. Oncotarget 2013, 4, 2430–2438. [Google Scholar] [CrossRef] [Green Version]
  24. Yang, M.H.; Jung, S.H.; Chinnathambi, A.; Alahmadi, T.A.; Alharbi, S.A.; Sethi, G.; Ahn, K.S. Attenuation of STAT3 Signaling Cascade by Daidzin Can Enhance the Apoptotic Potential of Bortezomib against Multiple Myeloma. Biomolecules 2019, 10, 23. [Google Scholar] [CrossRef] [Green Version]
  25. Yao, Y.; Sun, Y.; Shi, M.; Xia, D.; Zhao, K.; Zeng, L.; Yao, R.; Zhang, Y.; Li, Z.; Niu, M.; et al. Piperlongumine induces apoptosis and reduces bortezomib resistance by inhibiting STAT3 in multiple myeloma cells. Oncotarget 2016, 7, 73497–73508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Zhang, W.W.; Cortes, J.E.; Yao, H.; Zhang, L.; Reddy, N.G.; Jabbour, E.; Kantarjian, H.M.; Jones, D. Predictors of primary imatinib resistance in chronic myelogenous leukemia are distinct from those in secondary imatinib resistance. J. Clin. Oncol. 2009, 27, 3642–3649. [Google Scholar] [CrossRef] [Green Version]
  27. Zhang, Z.; Wang, W.; Ma, D.; Xiong, J.; Kuang, X.; Zhang, S.; Fang, Q.; Wang, J. Heme oxygenase-1 inhibition mediates Gas6 to enhance bortezomib-sensitivity in multiple myeloma via ERK/STAT3 axis. Aging 2020, 12, 6611–6629. [Google Scholar] [CrossRef]
  28. Zheng, Q.; Dong, H.; Mo, J.; Zhang, Y.; Huang, J.; Ouyang, S.; Shi, S.; Zhu, K.; Qu, X.; Hu, W.; et al. A novel STAT3 inhibitor W2014-S regresses human non-small cell lung cancer xenografts and sensitizes EGFR-TKI acquired resistance. Theranostics 2021, 11, 824–840. [Google Scholar] [CrossRef]
  29. Berman, H.; Henrick, K.; Nakamura, H. Announcing the worldwide Protein Data Bank. Nat. Struct. Biol. 2003, 10, 980. [Google Scholar] [CrossRef]
  30. Byun, W.S.; Bae, E.S.; Cui, J.; Park, H.J.; Oh, D.C.; Lee, S.K. Antitumor Activity of Pulvomycin via Targeting Activated-STAT3 Signaling in Docetaxel-Resistant Triple-Negative Breast Cancer Cells. Biomedicines 2021, 9, 436. [Google Scholar] [CrossRef]
  31. Chen, D.; Ma, Y.; Li, P.; Liu, M.; Fang, Y.; Zhang, J.; Zhang, B.; Hui, Y.; Yin, Y. Piperlongumine Induces Apoptosis and Synergizes with Doxorubicin by Inhibiting the JAK2-STAT3 Pathway in Triple-Negative Breast Cancer. Molecules 2019, 24, 2338. [Google Scholar] [CrossRef] [Green Version]
  32. Di Sotto, A.; Di Giacomo, S.; Rubini, E.; Macone, A.; Gulli, M.; Mammola, C.L.; Eufemi, M.; Mancinelli, R.; Mazzanti, G. Modulation of STAT3 Signaling, Cell Redox Defenses and Cell Cycle Checkpoints by beta-Caryophyllene in Cholangiocarcinoma Cells: Possible Mechanisms Accounting for Doxorubicin Chemosensitization and Chemoprevention. Cells 2020, 9, 858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Liu, C.; Xing, H.; Guo, C.; Yang, Z.; Wang, Y.; Wang, Y. MiR-124 reversed the doxorubicin resistance of breast cancer stem cells through STAT3/HIF-1 signaling pathways. Cell Cycle 2019, 18, 2215–2227. [Google Scholar] [CrossRef]
  34. Wu, X.; Xiao, H.; Wang, R.; Liu, L.; Li, C.; Lin, J. Persistent GP130/STAT3 Signaling Contributes to the Resistance of Doxorubicin, Cisplatin, and MEK Inhibitor in Human Rhabdomyosarcoma Cells. Curr. Cancer Drug Targets 2016, 16, 631–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Gong, J.; Munoz, A.R.; Pingali, S.; Payton-Stewart, F.; Chan, D.E.; Freeman, J.W.; Ghosh, R.; Kumar, A.P. Downregulation of STAT3/NF-kappaB potentiates gemcitabine activity in pancreatic cancer cells. Mol. Carcinog. 2017, 56, 402–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Guo, Y.; Xiao, Y.; Guo, H.; Zhu, H.; Chen, D.; Wang, J.; Deng, J.; Lan, J.; Liu, X.; Zhang, Q.; et al. The anti-dysenteric drug fraxetin enhances anti-tumor efficacy of gemcitabine and suppresses pancreatic cancer development by antagonizing STAT3 activation. Aging 2021, 13, 18545–18563. [Google Scholar] [CrossRef]
  37. Ioannou, N.; Seddon, A.M.; Dalgleish, A.; Mackintosh, D.; Solca, F.; Modjtahedi, H. Acquired resistance of pancreatic cancer cells to treatment with gemcitabine and HER-inhibitors is accompanied by increased sensitivity to STAT3 inhibition. Int. J. Oncol. 2016, 48, 908–918. [Google Scholar] [CrossRef] [Green Version]
  38. Lankadasari, M.B.; Aparna, J.S.; Mohammed, S.; James, S.; Aoki, K.; Binu, V.S.; Nair, S.; Harikumar, K.B. Targeting S1PR1/STAT3 loop abrogates desmoplasia and chemosensitizes pancreatic cancer to gemcitabine. Theranostics 2018, 8, 3824–3840. [Google Scholar] [CrossRef]
  39. Luo, F.; Lu, F.T.; Qiu, M.Z.; Zhou, T.; Ma, W.J.; Luo, M.; Zeng, K.M.; Luo, Q.Y.; Pan, W.T.; Zhang, L.; et al. Gemcitabine and APG-1252, a novel small molecule inhibitor of BCL-2/BCL-XL, display a synergistic antitumor effect in nasopharyngeal carcinoma through the JAK-2/STAT3/MCL-1 signaling pathway. Cell Death Dis. 2021, 12, 772. [Google Scholar] [CrossRef]
  40. Nagaraj, N.S.; Washington, M.K.; Merchant, N.B. Combined blockade of Src kinase and epidermal growth factor receptor with gemcitabine overcomes STAT3-mediated resistance of inhibition of pancreatic tumor growth. Clin. Cancer Res. 2011, 17, 483–493. [Google Scholar] [CrossRef] [Green Version]
  41. Nagathihalli, N.S.; Castellanos, J.A.; Shi, C.; Beesetty, Y.; Reyzer, M.L.; Caprioli, R.; Chen, X.; Walsh, A.J.; Skala, M.C.; Moses, H.L.; et al. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology 2015, 149, 1932–1943.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Venkatasubbarao, K.; Peterson, L.; Zhao, S.; Hill, P.; Cao, L.; Zhou, Q.; Nawrocki, S.T.; Freeman, J.W. Inhibiting signal transducer and activator of transcription-3 increases response to gemcitabine and delays progression of pancreatic cancer. Mol. Cancer 2013, 12, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wu, X.; Tang, W.; Marquez, R.T.; Li, K.; Highfill, C.A.; He, F.; Lian, J.; Lin, J.; Fuchs, J.R.; Ji, M.; et al. Overcoming chemo/radio-resistance of pancreatic cancer by inhibiting STAT3 signaling. Oncotarget 2016, 7, 11708–11723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhao, T.; Jin, F.; Xiao, D.; Wang, H.; Huang, C.; Wang, X.; Gao, S.; Liu, J.; Yang, S.; Hao, J. IL-37/ STAT3/ HIF-1alpha negative feedback signaling drives gemcitabine resistance in pancreatic cancer. Theranostics 2020, 10, 4088–4100. [Google Scholar] [CrossRef]
  45. Chang, W.M.; Chang, Y.C.; Yang, Y.C.; Lin, S.K.; Chang, P.M.; Hsiao, M. AKR1C1 controls cisplatin-resistance in head and neck squamous cell carcinoma through cross-talk with the STAT1/3 signaling pathway. J. Exp. Clin. Cancer Res. 2019, 38, 245. [Google Scholar] [CrossRef] [Green Version]
  46. Chen, K.B.; Yang, W.; Xuan, Y.; Lin, A.J. miR-526b-3p inhibits lung cancer cisplatin-resistance and metastasis by inhibiting STAT3-promoted PD-L1. Cell Death Dis. 2021, 12, 748. [Google Scholar] [CrossRef]
  47. Fan, H.; Ou, Q.; Su, Q.; Li, G.; Deng, Z.; Huang, X.; Bi, J. ZIPK activates the IL-6/STAT3 signaling pathway and promotes cisplatin resistance in gastric cancer cells. FEBS Open Bio 2021, 11, 2655–2667. [Google Scholar] [CrossRef]
  48. Jin, P.; Liu, Y.; Wang, R. STAT3 regulated miR-216a promotes ovarian cancer proliferation and cisplatin resistance. Biosci. Rep. 2018, 38, BSR20180547. [Google Scholar] [CrossRef]
  49. Kuo, W.Y.; Hwu, L.; Wu, C.Y.; Lee, J.S.; Chang, C.W.; Liu, R.S. STAT3/NF-kappaB-Regulated Lentiviral TK/GCV Suicide Gene Therapy for Cisplatin-Resistant Triple-Negative Breast Cancer. Theranostics 2017, 7, 647–663. [Google Scholar] [CrossRef] [Green Version]
  50. Lin, C.C.; Yeh, H.H.; Huang, W.L.; Yan, J.J.; Lai, W.W.; Su, W.P.; Chen, H.H.; Su, W.C. Metformin enhances cisplatin cytotoxicity by suppressing signal transducer and activator of transcription-3 activity independently of the liver kinase B1-AMP-activated protein kinase pathway. Am. J. Respir. Cell Mol. Biol. 2013, 49, 241–250. [Google Scholar] [CrossRef]
  51. Liu, W.H.; Chen, M.T.; Wang, M.L.; Lee, Y.Y.; Chiou, G.Y.; Chien, C.S.; Huang, P.I.; Chen, Y.W.; Huang, M.C.; Chiou, S.H.; et al. Cisplatin-selected resistance is associated with increased motility and stem-like properties via activation of STAT3/Snail axis in atypical teratoid/rhabdoid tumor cells. Oncotarget 2015, 6, 1750–1768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Sun, C.Y.; Nie, J.; Huang, J.P.; Zheng, G.J.; Feng, B. Targeting STAT3 inhibition to reverse cisplatin resistance. Biomed. Pharmacother. 2019, 117, 109135. [Google Scholar] [CrossRef]
  53. Tan, W.X.; Sun, G.; Shangguan, M.Y.; Gui, Z.; Bao, Y.; Li, Y.F.; Jia, Z.H. Novel role of lncRNA CHRF in cisplatin resistance of ovarian cancer is mediated by miR-10b induced EMT and STAT3 signaling. Sci. Rep. 2020, 10, 14768. [Google Scholar] [CrossRef] [PubMed]
  54. Tao, L.; Huang, G.; Wang, R.; Pan, Y.; He, Z.; Chu, X.; Song, H.; Chen, L. Cancer-associated fibroblasts treated with cisplatin facilitates chemoresistance of lung adenocarcinoma through IL-11/IL-11R/STAT3 signaling pathway. Sci. Rep. 2016, 6, 38408. [Google Scholar] [CrossRef]
  55. Zhu, X.; Shen, H.; Yin, X.; Long, L.; Chen, X.; Feng, F.; Liu, Y.; Zhao, P.; Xu, Y.; Li, M.; et al. IL-6R/STAT3/miR-204 feedback loop contributes to cisplatin resistance of epithelial ovarian cancer cells. Oncotarget 2017, 8, 39154–39166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Filppu, P.; Tanjore Ramanathan, J.; Granberg, K.J.; Gucciardo, E.; Haapasalo, H.; Lehti, K.; Nykter, M.; Le Joncour, V.; Laakkonen, P. CD109-GP130 interaction drives glioblastoma stem cell plasticity and chemoresistance through STAT3 activity. JCI Insight 2021, 6. [Google Scholar] [CrossRef] [PubMed]
  57. Kohsaka, S.; Wang, L.; Yachi, K.; Mahabir, R.; Narita, T.; Itoh, T.; Tanino, M.; Kimura, T.; Nishihara, H.; Tanaka, S. STAT3 inhibition overcomes temozolomide resistance in glioblastoma by downregulating MGMT expression. Mol. Cancer Ther. 2012, 11, 1289–1299. [Google Scholar] [CrossRef] [Green Version]
  58. Lee, E.S.; Ko, K.K.; Joe, Y.A.; Kang, S.G.; Hong, Y.K. Inhibition of STAT3 reverses drug resistance acquired in temozolomide-resistant human glioma cells. Oncol. Lett. 2011, 2, 115–121. [Google Scholar] [CrossRef] [Green Version]
  59. Xu, P.; Wang, H.; Pan, H.; Chen, J.; Deng, C. Anlotinib combined with temozolomide suppresses glioblastoma growth via mediation of JAK2/STAT3 signaling pathway. Cancer Chemother. Pharmacol. 2022, 89, 183–196. [Google Scholar] [CrossRef]
  60. Vengoji, R.; Macha, M.A.; Nimmakayala, R.K.; Rachagani, S.; Siddiqui, J.A.; Mallya, K.; Gorantla, S.; Jain, M.; Ponnusamy, M.P.; Batra, S.K.; et al. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells. J. Exp. Clin. Cancer Res. 2019, 38, 266. [Google Scholar] [CrossRef]
  61. Ding, Y.; Zhen, Z.; Nisar, M.A.; Ali, F.; Din, R.U.; Khan, M.; Mughal, T.A.; Alam, G.; Liu, L.; Saleem, M.Z. Sesquiterpene Lactones Attenuate Paclitaxel Resistance Via Inhibiting MALAT1/STAT3/ FUT4 Axis and P-Glycoprotein Transporters in Lung Cancer Cells. Front. Pharmacol. 2022, 13, 795613. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, L.; Zhang, F.; Cui, J.Y.; Chen, L.; Chen, Y.T.; Liu, B.W. CAFs enhance paclitaxel resistance by inducing EMT through the IL-6/JAK2/STAT3 pathway. Oncol. Rep. 2018, 39, 2081–2090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Wang, S.; Yao, Y.; Yao, M.; Fu, P.; Wang, W. Interleukin-22 promotes triple negative breast cancer cells migration and paclitaxel resistance through JAK-STAT3/MAPKs/AKT signaling pathways. Biochem. Biophys. Res. Commun. 2018, 503, 1605–1609. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, Y.; Zhang, J.; Wu, J.; Zhong, S.; Li, H. Inhibition of JAK2 Reverses Paclitaxel Resistance in Human Ovarian Cancer Cells. Int. J. Gynecol. Cancer 2015, 25, 1557–1564. [Google Scholar] [CrossRef] [PubMed]
  65. Zhao, C.; Chen, H.Y.; Zhao, F.; Feng, H.J.; Su, J.P. Acylglycerol kinase promotes paclitaxel resistance in nasopharyngeal carcinoma cells by regulating FOXM1 via the JAK2/STAT3 pathway. Cytokine 2021, 148, 155595. [Google Scholar] [CrossRef]
  66. Kim, S.M.; Kwon, O.J.; Hong, Y.K.; Kim, J.H.; Solca, F.; Ha, S.J.; Soo, R.A.; Christensen, J.G.; Lee, J.H.; Cho, B.C. Activation of IL-6R/JAK1/STAT3 signaling induces de novo resistance to irreversible EGFR inhibitors in non-small cell lung cancer with T790M resistance mutation. Mol. Cancer Ther. 2012, 11, 2254–2264. [Google Scholar] [CrossRef] [Green Version]
  67. Shu, D.; Xu, Y.; Chen, W. Knockdown of lncRNA BLACAT1 reverses the resistance of afatinib to non-small cell lung cancer via modulating STAT3 signalling. J. Drug Target 2020, 28, 300–306. [Google Scholar] [CrossRef]
  68. Cuyas, E.; Perez-Sanchez, A.; Micol, V.; Menendez, J.A.; Bosch-Barrera, J. STAT3-targeted treatment with silibinin overcomes the acquired resistance to crizotinib in ALK-rearranged lung cancer. Cell Cycle 2016, 15, 3413–3418. [Google Scholar] [CrossRef] [Green Version]
  69. Dosch, A.R.; Dai, X.; Reyzer, M.L.; Mehra, S.; Srinivasan, S.; Willobee, B.A.; Kwon, D.; Kashikar, N.; Caprioli, R.; Merchant, N.B.; et al. Combined Src/EGFR Inhibition Targets STAT3 Signaling and Induces Stromal Remodeling to Improve Survival in Pancreatic Cancer. Mol. Cancer Res. 2020, 18, 623–631. [Google Scholar] [CrossRef] [Green Version]
  70. Harada, D.; Takigawa, N.; Kiura, K. The Role of STAT3 in Non-Small Cell Lung Cancer. Cancers 2014, 6, 708–722. [Google Scholar] [CrossRef] [Green Version]
  71. Li, R.; Hu, Z.; Sun, S.Y.; Chen, Z.G.; Owonikoko, T.K.; Sica, G.L.; Ramalingam, S.S.; Curran, W.J.; Khuri, F.R.; Deng, X. Niclosamide overcomes acquired resistance to erlotinib through suppression of STAT3 in non-small cell lung cancer. Mol. Cancer Ther. 2013, 12, 2200–2212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Salzberg, M.P.; Merchant, N.B. Overcoming erlotinib resistance with STAT3 inhibition in pancreatic cancer. J. Clin. Oncol. 2017, 35, 304. [Google Scholar] [CrossRef]
  73. Stanam, A.; Love-Homan, L.; Joseph, T.S.; Espinosa-Cotton, M.; Simons, A.L. Upregulated interleukin-6 expression contributes to erlotinib resistance in head and neck squamous cell carcinoma. Mol. Oncol. 2015, 9, 1371–1383. [Google Scholar] [CrossRef] [Green Version]
  74. Tang, C.H.; Qin, L.; Gao, Y.C.; Chen, T.Y.; Xu, K.; Liu, T.; Ren, T. APE1 shRNA-loaded cancer stem cell-derived extracellular vesicles reverse Erlotinib resistance in non-small cell lung cancer via the IL-6/STAT3 signalling. Clin. Transl. Med. 2022, 12, e876. [Google Scholar] [CrossRef]
  75. Iguchi, T.; Tamada, S.; Kato, M.; Yasuda, S.; Yamasaki, T.; Nakatani, T. Enzalutamide versus flutamide for castration-resistant prostate cancer after combined androgen blockade therapy with bicalutamide: Study protocol for a multicenter randomized phase II trial (the OCUU-CRPC study). BMC Cancer 2019, 19, 339. [Google Scholar] [CrossRef] [PubMed]
  76. Giacinti, S.; Poti, G.; Roberto, M.; Macrini, S.; Bassanelli, M.; Di Pietro, F.; Aschelter, A.M.; Ceribelli, A.; Ruggeri, E.M.; Marchetti, P. Molecular Basis of Drug Resistance and Insights for New Treatment Approaches in mCRPC. Anticancer Res. 2018, 38, 6029–6039. [Google Scholar] [CrossRef]
  77. Schroeder, A.; Herrmann, A.; Cherryholmes, G.; Kowolik, C.; Buettner, R.; Pal, S.; Yu, H.; Muller-Newen, G.; Jove, R. Loss of androgen receptor expression promotes a stem-like cell phenotype in prostate cancer through STAT3 signaling. Cancer Res. 2014, 74, 1227–1237. [Google Scholar] [CrossRef] [Green Version]
  78. Liu, C.; Zhu, Y.; Lou, W.; Cui, Y.; Evans, C.P.; Gao, A.C. Inhibition of constitutively active Stat3 reverses enzalutamide resistance in LNCaP derivative prostate cancer cells. Prostate 2014, 74, 201–209. [Google Scholar] [CrossRef] [Green Version]
  79. Liu, Q.; Tong, D.; Liu, G.; Xu, J.; Do, K.; Geary, K.; Zhang, D.; Zhang, J.; Zhang, Y.; Li, Y.; et al. Metformin reverses prostate cancer resistance to enzalutamide by targeting TGF-beta1/STAT3 axis-regulated EMT. Cell Death Dis. 2017, 8, e3007. [Google Scholar] [CrossRef] [Green Version]
  80. Yao, J.; Deng, K.; Huang, J.; Zeng, R.; Zuo, J. Progress in the Understanding of the Mechanism of Tamoxifen Resistance in Breast Cancer. Front. Pharmacol. 2020, 11, 592912. [Google Scholar] [CrossRef]
  81. Lv, C.; Huang, Y.; Huang, R.; Wang, Q.; Zhang, H.; Jin, J.; Lu, D.; Zhou, Y.; Shen, Y.; Zhang, W.; et al. Narciclasine targets STAT3 via distinct mechanisms in tamoxifen-resistant breast cancer cells. Mol. Ther. Oncolytics 2022, 24, 340–354. [Google Scholar] [CrossRef]
  82. Moon, S.Y.; Lee, H.; Kim, S.; Hong, J.H.; Chun, S.H.; Lee, H.Y.; Kang, K.; Kim, H.S.; Won, H.S.; Ko, Y.H. Inhibition of STAT3 enhances sensitivity to tamoxifen in tamoxifen-resistant breast cancer cells. BMC Cancer 2021, 21, 931. [Google Scholar] [CrossRef] [PubMed]
  83. Zhu, N.; Zhang, J.; Du, Y.; Qin, X.; Miao, R.; Nan, J.; Chen, X.; Sun, J.; Zhao, R.; Zhang, X.; et al. Loss of ZIP facilitates JAK2-STAT3 activation in tamoxifen-resistant breast cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 15047–15054. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, W.; Shen, X.; Xia, X.; Xu, G.; Ma, T.; Bai, X.; Liang, T. NSC 74859-mediated inhibition of STAT3 enhances the anti-proliferative activity of cetuximab in hepatocellular carcinoma. Liver Int. 2012, 32, 70–77. [Google Scholar] [CrossRef] [PubMed]
  85. O’Keefe, R.A.; Bhola, N.E.; Lee, D.S.; Johnson, D.E.; Grandis, J.R. Interleukin 6 is increased in preclinical HNSCC models of acquired cetuximab resistance, but is not required for maintenance of resistance. PLoS ONE 2020, 15, e0227261. [Google Scholar] [CrossRef] [PubMed]
  86. Willey, C.D.; Anderson, J.C.; Trummell, H.Q.; Naji, F.; de Wijn, R.; Yang, E.S.; Bredel, M.; Thudi, N.K.; Bonner, J.A. Differential escape mechanisms in cetuximab-resistant head and neck cancer cells. Biochem. Biophys. Res. Commun. 2019, 517, 36–42. [Google Scholar] [CrossRef]
  87. Eichten, A.; Su, J.; Adler, A.P.; Zhang, L.; Ioffe, E.; Parveen, A.A.; Yancopoulos, G.D.; Rudge, J.; Lowy, I.; Lin, H.C.; et al. Resistance to Anti-VEGF Therapy Mediated by Autocrine IL6/STAT3 Signaling and Overcome by IL6 Blockade. Cancer Res. 2016, 76, 2327–2339. [Google Scholar] [CrossRef] [Green Version]
  88. Jiang, L.; Ren, L.; Chen, H.; Pan, J.; Zhang, Z.; Kuang, X.; Chen, X.; Bao, W.; Lin, C.; Zhou, Z.; et al. NCAPG confers trastuzumab resistance via activating SRC/STAT3 signaling pathway in HER2-positive breast cancer. Cell Death Dis. 2020, 11, 547. [Google Scholar] [CrossRef]
  89. Li, G.; Zhao, L.; Li, W.; Fan, K.; Qian, W.; Hou, S.; Wang, H.; Dai, J.; Wei, H.; Guo, Y. Feedback activation of STAT3 mediates trastuzumab resistance via upregulation of MUC1 and MUC4 expression. Oncotarget 2014, 5, 8317–8329. [Google Scholar] [CrossRef] [Green Version]
  90. Wang, L.; Wang, Q.; Gao, M.; Fu, L.; Li, Y.; Quan, H.; Lou, L. STAT3 activation confers trastuzumab-emtansine (T-DM1) resistance in HER2-positive breast cancer. Cancer Sci. 2018, 109, 3305–3315. [Google Scholar] [CrossRef] [Green Version]
  91. Yang, Z.; Guo, L.; Liu, D.; Sun, L.; Chen, H.; Deng, Q.; Liu, Y.; Yu, M.; Ma, Y.; Guo, N.; et al. Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of IL-6/STAT3/Jagged-1/Notch positive feedback loop. Oncotarget 2015, 6, 5072–5087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Chehrazi-Raffle, A.; Meza, L.; Alcantara, M.; Dizman, N.; Bergerot, P.; Salgia, N.; Hsu, J.; Ruel, N.; Salgia, S.; Malhotra, J.; et al. Circulating cytokines associated with clinical response to systemic therapy in metastatic renal cell carcinoma. J. Immunother. Cancer 2021, 9. [Google Scholar] [CrossRef] [PubMed]
  93. Datta, J.; Dai, X.; Bianchi, A.; De Castro Silva, I.; Mehra, S.; Garrido, V.T.; Lamichhane, P.; Singh, S.P.; Zhou, Z.; Dosch, A.R.; et al. Combined MEK and STAT3 Inhibition Uncovers Stromal Plasticity by Enriching for Cancer-Associated Fibroblasts With Mesenchymal Stem Cell-Like Features to Overcome Immunotherapy Resistance in Pancreatic Cancer. Gastroenterology 2022, 163, 1593–1612. [Google Scholar] [CrossRef]
  94. Fang, W.; Zhang, J.; Hong, S.; Zhan, J.; Chen, N.; Qin, T.; Tang, Y.; Zhang, Y.; Kang, S.; Zhou, T.; et al. EBV-driven LMP1 and IFN-gamma up-regulate PD-L1 in nasopharyngeal carcinoma: Implications for oncotargeted therapy. Oncotarget 2014, 5, 12189–12202. [Google Scholar] [CrossRef] [PubMed]
  95. Hou, J.; Zhao, R.; Xia, W.; Chang, C.W.; You, Y.; Hsu, J.M.; Nie, L.; Chen, Y.; Wang, Y.C.; Liu, C.; et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat.Cell Biol. 2020, 22, 1264–1275. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, L.; Xu, Y.; Fang, J.; Liu, W.; Chen, J.; Liu, Z.; Xu, Q. Targeting STAT3 Abrogates Tim-3 Upregulation of Adaptive Resistance to PD-1 Blockade on Regulatory T Cells of Melanoma. Front. Immunol. 2021, 12, 654749. [Google Scholar] [CrossRef] [PubMed]
  97. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef]
  98. Kim, T.W.; Kim, Y.; Keum, H.; Jung, W.; Kang, M.; Jon, S. Combination of a STAT3 inhibitor with anti-PD-1 immunotherapy is an effective treatment regimen for a vemurafenib-resistant melanoma. Mol. Ther. Oncolytics 2022, 26, 1–14. [Google Scholar] [CrossRef]
  99. MacManus, M.; Hegi-Johnson, F. Overcoming immunotherapy resistance in NSCLC. Lancet Oncol. 2022, 23, 191–193. [Google Scholar] [CrossRef]
  100. Marzec, M.; Zhang, Q.; Goradia, A.; Raghunath, P.N.; Liu, X.; Paessler, M.; Wang, H.Y.; Wysocka, M.; Cheng, M.; Ruggeri, B.A.; et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc. Natl. Acad. Sci. USA 2008, 105, 20852–20857. [Google Scholar] [CrossRef] [Green Version]
  101. Reck, M.; Rodriguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csoszi, T.; Fulop, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Venkatraman, S.; Meller, J.; Hongeng, S.; Tohtong, R.; Chutipongtanate, S. Transcriptional Regulation of Cancer Immune Checkpoints: Emerging Strategies for Immunotherapy. Vaccines 2020, 8, 735. [Google Scholar] [CrossRef] [PubMed]
  103. Witt, K.; Evans-Axelsson, S.; Lundqvist, A.; Johansson, M.; Bjartell, A.; Hellsten, R. Inhibition of STAT3 augments antitumor efficacy of anti-CTLA-4 treatment against prostate cancer. Cancer Immunol. Immunother. 2021, 70, 3155–3166. [Google Scholar] [CrossRef] [PubMed]
  104. Woods, D.M.; Ramakrishnan, R.; Laino, A.S.; Berglund, A.; Walton, K.; Betts, B.C.; Weber, J.S. Decreased Suppression and Increased Phosphorylated STAT3 in Regulatory T Cells are Associated with Benefit from Adjuvant PD-1 Blockade in Resected Metastatic Melanoma. Clin. Cancer Res. 2018, 24, 6236–6247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zou, S.; Tong, Q.; Liu, B.; Huang, W.; Tian, Y.; Fu, X. Targeting STAT3 in Cancer Immunotherapy. Mol. Cancer 2020, 19, 145. [Google Scholar] [CrossRef] [PubMed]
  106. You, W.; Liu, X.; Yu, Y.; Chen, C.; Xiong, Y.; Liu, Y.; Sun, Y.; Tan, C.; Zhang, H.; Wang, Y.; et al. miR-502-5p affects gastric cancer progression by targeting PD-L1. Cancer Cell Int. 2020, 20, 395. [Google Scholar] [CrossRef]
  107. McGowan, J.V.; Chung, R.; Maulik, A.; Piotrowska, I.; Walker, J.M.; Yellon, D.M. Anthracycline Chemotherapy and Cardiotoxicity. Cardiovasc. Drugs Ther. 2017, 31, 63–75. [Google Scholar] [CrossRef] [Green Version]
  108. Mohammadian, J.; Mahmoudi, S.; Pourmohammad, P.; Pirouzpanah, M.; Salehnia, F.; Maroufi, N.F.; Samadi, N.; Sabzichi, M. Formulation of Stattic as STAT3 inhibitor in nanostructured lipid carriers (NLCs) enhances efficacy of doxorubicin in melanoma cancer cells. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 2315–2323. [Google Scholar] [CrossRef]
  109. Schust, J.; Sperl, B.; Hollis, A.; Mayer, T.U.; Berg, T. Stattic: A small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006, 13, 1235–1242. [Google Scholar] [CrossRef] [Green Version]
  110. Gariboldi, M.B.; Ravizza, R.; Molteni, R.; Osella, D.; Gabano, E.; Monti, E. Inhibition of Stat3 increases doxorubicin sensitivity in a human metastatic breast cancer cell line. Cancer Lett. 2007, 258, 181–188. [Google Scholar] [CrossRef]
  111. Maryam, A.; Mehmood, T.; Zhang, H.; Li, Y.; Khan, M.; Ma, T. Alantolactone induces apoptosis, promotes STAT3 glutathionylation and enhances chemosensitivity of A549 lung adenocarcinoma cells to doxorubicin via oxidative stress. Sci. Rep. 2017, 7, 6242. [Google Scholar] [CrossRef] [Green Version]
  112. Sheng, H.; Feng, Q.; Quan, Q.; Sheng, X.; Zhang, P. Inhibition of STAT3 reverses Taxol-resistance in ovarian cancer by down-regulating G6PD expression in vitro. Biochem. Biophys. Res. Commun. 2022, 617, 62–68. [Google Scholar] [CrossRef] [PubMed]
  113. Fan, Z.; Cui, H.; Yu, H.; Ji, Q.; Kang, L.; Han, B.; Wang, J.; Dong, Q.; Li, Y.; Yan, Z.; et al. MiR-125a promotes paclitaxel sensitivity in cervical cancer through altering STAT3 expression. Oncogenesis 2016, 5, e223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Chuang, H.Y.; Su, Y.K.; Liu, H.W.; Chen, C.H.; Chiu, S.C.; Cho, D.Y.; Lin, S.Z.; Chen, Y.S.; Lin, C.M. Preclinical Evidence of STAT3 Inhibitor Pacritinib Overcoming Temozolomide Resistance via Downregulating miR-21-Enriched Exosomes from M2 Glioblastoma-Associated Macrophages. J. Clin. Med. 2019, 8, 959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ouedraogo, Z.G.; Biau, J.; Kemeny, J.L.; Morel, L.; Verrelle, P.; Chautard, E. Role of STAT3 in Genesis and Progression of Human Malignant Gliomas. Mol. Neurobiol. 2017, 54, 5780–5797. [Google Scholar] [CrossRef]
  116. Zheng, H.; Yang, L.; Kang, Y.; Chen, M.; Lin, S.; Xiang, Y.; Li, C.; Dai, X.; Huang, X.; Liang, G.; et al. Alantolactone sensitizes human pancreatic cancer cells to EGFR inhibitors through the inhibition of STAT3 signaling. Mol. Carcinog. 2019, 58, 565–576. [Google Scholar] [CrossRef]
  117. Groner, B. Determinants of the extent and duration of STAT3 signaling. JAKSTAT 2012, 1, 211–215. [Google Scholar] [CrossRef] [Green Version]
  118. Wang, H.Q.; Man, Q.W.; Huo, F.Y.; Gao, X.; Lin, H.; Li, S.R.; Wang, J.; Su, F.C.; Cai, L.; Shi, Y.; et al. STAT3 pathway in cancers: Past, present, and future. MedComm (2020) 2022, 3, e124. [Google Scholar] [CrossRef]
  119. Gide, T.N.; Wilmott, J.S.; Scolyer, R.A.; Long, G.V. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin. Cancer Res. 2018, 24, 1260–1270. [Google Scholar] [CrossRef] [Green Version]
  120. Morselli, E.; Galluzzi, L.; Kepp, O.; Vicencio, J.M.; Criollo, A.; Maiuri, M.C.; Kroemer, G. Anti- and pro-tumor functions of autophagy. Biochim. Biophys. Acta 2009, 1793, 1524–1532. [Google Scholar] [CrossRef] [Green Version]
  121. You, L.; Wang, Z.; Li, H.; Shou, J.; Jing, Z.; Xie, J.; Sui, X.; Pan, H.; Han, W. The role of STAT3 in autophagy. Autophagy 2015, 11, 729–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Li, X.; Zhou, Y.; Li, Y.; Yang, L.; Ma, Y.; Peng, X.; Yang, S.; Liu, J.; Li, H. Autophagy: A novel mechanism of chemoresistance in cancers. Biomed. Pharmacother. 2019, 119, 109415. [Google Scholar] [CrossRef] [PubMed]
  123. Xu, J.; Zhang, J.; Mao, Q.F.; Wu, J.; Wang, Y. The Interaction Between Autophagy and JAK/STAT3 Signaling Pathway in Tumors. Front. Genet. 2022, 13, 880359. [Google Scholar] [CrossRef] [PubMed]
  124. Pu, Y.; Wang, J.; Wang, S. Role of autophagy in drug resistance and regulation of osteosarcoma (Review). Mol. Clin. Oncol. 2022, 16, 72. [Google Scholar] [CrossRef]
  125. Wu, L.; Shen, B.; Li, J.; Zhang, H.; Zhang, K.; Yang, Y.; Zu, Z.; Shen, D.; Luo, M. STAT3 exerts pro-tumor and anti-autophagy roles in cervical cancer. Diagn. Pathol. 2022, 17, 13. [Google Scholar] [CrossRef]
  126. Jiang, T.; Chen, X.; Ren, X.; Yang, J.M.; Cheng, Y. Emerging role of autophagy in anti-tumor immunity: Implications for the modulation of immunotherapy resistance. Drug Resist. Updat. 2021, 56, 100752. [Google Scholar] [CrossRef]
  127. Adeshakin, F.O.; Adeshakin, A.O.; Liu, Z.; Lu, X.; Cheng, J.; Zhang, P.; Yan, D.; Zhang, G.; Wan, X. Upregulation of V-ATPase by STAT3 Activation Promotes Anoikis Resistance and Tumor Metastasis. J. Cancer 2021, 12, 4819–4829. [Google Scholar] [CrossRef]
  128. Du, X.L.; Yang, H.; Liu, S.G.; Luo, M.L.; Hao, J.J.; Zhang, Y.; Lin, D.C.; Xu, X.; Cai, Y.; Zhan, Q.M.; et al. Calreticulin promotes cell motility and enhances resistance to anoikis through STAT3-CTTN-Akt pathway in esophageal squamous cell carcinoma. Oncogene 2009, 28, 3714–3722. [Google Scholar] [CrossRef] [Green Version]
  129. Fofaria, N.M.; Srivastava, S.K. STAT3 induces anoikis resistance, promotes cell invasion and metastatic potential in pancreatic cancer cells. Carcinogenesis 2015, 36, 142–150. [Google Scholar] [CrossRef] [Green Version]
  130. Huyen, N.T.; Prachayasittikul, V.; Chan-On, W. Anoikis-resistant cholangiocarcinoma cells display aggressive characteristics and increase STAT3 activation. J. Hepatobiliary Pancreat Sci. 2016, 23, 397–405. [Google Scholar] [CrossRef]
  131. Lin, Y.T.; Wang, H.C.; Hsu, Y.C.; Cho, C.L.; Yang, M.Y.; Chien, C.Y. Capsaicin Induces Autophagy and Apoptosis in Human Nasopharyngeal Carcinoma Cells by Downregulating the PI3K/AKT/mTOR Pathway. Int. J. Mol. Sci. 2017, 18, 1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Neiva, K.G.; Zhang, Z.; Miyazawa, M.; Warner, K.A.; Karl, E.; Nor, J.E. Cross talk initiated by endothelial cells enhances migration and inhibits anoikis of squamous cell carcinoma cells through STAT3/Akt/ERK signaling. Neoplasia 2009, 11, 583–593. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, L.N.; Zhang, Z.T.; Wang, L.; Wei, H.X.; Zhang, T.; Zhang, L.M.; Lin, H.; Zhang, H.; Wang, S.Q. TGF-beta1/SH2B3 axis regulates anoikis resistance and EMT of lung cancer cells by modulating JAK2/STAT3 and SHP2/Grb2 signaling pathways. Cell Death Dis. 2022, 13, 472. [Google Scholar] [CrossRef] [PubMed]
  134. Tomida, M.; Ohtake, H.; Yokota, T.; Kobayashi, Y.; Kurosumi, M. Stat3 Up-Regulates Expression of Nicotinamide N-Methyltransferase in Human Cancer Cells. J. Cancer Res. Clin. Oncol. 2008, 134, 551–559. [Google Scholar] [CrossRef] [PubMed]
  135. Lu, S.; Ke, S.; Wang, C.; Xu, Y.; Li, Z.; Song, K.; Bai, M.; Zhou, M.; Yu, H.; Yin, B.; et al. NNMT promotes the progression of intrahepatic cholangiocarcinoma by regulating aerobic glycolysis via the EGFR-STAT3 axis. Oncogenesis 2022, 11, 39. [Google Scholar] [CrossRef]
  136. Hah, Y.S.; Cho, H.Y.; Jo, S.Y.; Park, Y.S.; Heo, E.P.; Yoon, T.J. Nicotinamide Nmethyltransferase Induces the Proliferation and Invasion of Squamous Cell Carcinoma Cells. Oncol. Rep. 2019, 2, 1805–1814. [Google Scholar]
  137. Wang, W.; Yang, C.; Wang, T.; Deng, H. Complex roles of nicotinamide N-methyltransferase in cancer progression. Cell Death Dis. 2022, 13, 267. [Google Scholar] [CrossRef]
  138. Li, X.-Y.; Pi, Y.-N.; Chen, Y.; Zhu, Q.; Xia, B.-R. Nicotinamide N-Methyltransferase: A Promising Biomarker and Target for Human Cancer Therapy. Front. Oncol. 2022, 12, 894744. [Google Scholar] [CrossRef]
  139. Lu, J.; Zhang, L.; Chen, X.; Lu, Q.; Yang, Y.; Liu, J.; Ma, X. SIRT1 counteracted the activation of STAT3 and NF-κB to repress the gastric cancer growth. Int. J. Clin. Exp. Med. 2014, 7, 5050–5058. [Google Scholar]
  140. Niu, G.; Wright, K.L.; Ma, Y.; Wright, G.M.; Huang, M.; Irby, R.; Briggs, J.; Karras, J.; Cress, W.D.; Pardoll, D.; et al. Role of Stat3 in regulating p53 expression and function. Mol. Cell Biol. 2005, 25, 7432–7440. [Google Scholar] [CrossRef] [Green Version]
  141. Pham, T.H.; Park, H.M.; Kim, J.; Hong, J.T.; Yoon, D.Y. STAT3 and p53: Dual Target for Cancer Therapy. Biomedicines 2020, 8, 637. [Google Scholar] [CrossRef] [PubMed]
  142. Liang, F.; Ren, C.; Wang, J.; Wang, S.; Yang, L.; Han, X.; Chen, Y.; Tong, G.; Yang, G. The crosstalk between STAT3 and p53/RAS signaling controls cancer cell metastasis and cisplatin resistance via the Slug/MAPK/PI3K/AKT-mediated regulation of EMT and autophagy. Oncogenesis 2019, 8, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Singh, S.; Murillo, G.; Richner, J.; Singh, S.P.; Berleth, E.; Kumar, V.; Mehta, R.; Ramiya, V.; Parihar, A.S. A Broad-Based Characterization of a Cell-Penetrating, Single Domain Camelid Bi-Specific Antibody Monomer That Targets STAT3 and KRAS Dependent Cancers. Int. J. Mol. Sci. 2022, 23, 7565. [Google Scholar] [CrossRef] [PubMed]
  144. Yang, L.; Lin, S.; Xu, L.; Lin, J.; Zhao, C.; Huang, X. Novel activators and small-molecule inhibitors of STAT3 in cancer. Cytokine Growth Factor Rev. 2019, 49, 10–22. [Google Scholar] [CrossRef]
  145. Joshi, N.; Hajizadeh, F.; Ansari Dezfouli, E.; Zekiy, A.O.; Nabi Afjadi, M.; Mousavi, S.M.; Hojjat-Farsangi, M.; Karpisheh, V.; Mahmoodpoor, A.; Hassannia, H.; et al. Silencing STAT3 enhances sensitivity of cancer cells to doxorubicin and inhibits tumor progression. Life Sci. 2021, 275, 119369. [Google Scholar] [CrossRef]
  146. Chen, S.H.; Chang, J.Y. New Insights into Mechanisms of Cisplatin Resistance: From Tumor Cell to Microenvironment. Int. J. Mol. Sci. 2019, 20, 4136. [Google Scholar] [CrossRef] [Green Version]
  147. Morelli, A.P.; Tortelli, T.C., Jr.; Mancini, M.C.S.; Pavan, I.C.B.; Silva, L.G.S.; Severino, M.B.; Granato, D.C.; Pestana, N.F.; Ponte, L.G.S.; Peruca, G.F.; et al. STAT3 contributes to cisplatin resistance, modulating EMT markers, and the mTOR signaling in lung adenocarcinoma. Neoplasia 2021, 23, 1048–1058. [Google Scholar] [CrossRef]
  148. Tan, M.S.Y.; Sandanaraj, E.; Chong, Y.K.; Lim, S.W.; Koh, L.W.H.; Ng, W.H.; Tan, N.S.; Tan, P.; Ang, B.T.; Tang, C. A STAT3-based gene signature stratifies glioma patients for targeted therapy. Nat. Commun. 2019, 10, 3601. [Google Scholar] [CrossRef] [Green Version]
  149. Nelson, E.A.; Sharma, S.V.; Settleman, J.; Frank, D.A. A chemical biology approach to developing STAT inhibitors: Molecular strategies for accelerating clinical translation. Oncotarget 2011, 2, 518–524. [Google Scholar] [CrossRef] [Green Version]
  150. Remy, J.; Linder, B.; Weirauch, U.; Day, B.W.; Stringer, B.W.; Herold-Mende, C.; Aigner, A.; Krohn, K.; Kogel, D. STAT3 Enhances Sensitivity of Glioblastoma to Drug-Induced Autophagy-Dependent Cell Death. Cancers 2022, 14, 339. [Google Scholar] [CrossRef]
  151. Gu, H.R.; Park, S.C.; Choi, S.J.; Lee, J.C.; Kim, Y.C.; Han, C.J.; Kim, J.; Yang, K.Y.; Kim, Y.J.; Noh, G.Y.; et al. Combined treatment with silibinin and either sorafenib or gefitinib enhances their growth-inhibiting effects in hepatocellular carcinoma cells. Clin. Mol. Hepatol. 2015, 21, 49–59. [Google Scholar] [CrossRef]
  152. Dosch, A.R.; Singh, S.; Dai, X.; Mehra, S.; Silva, I.C.; Bianchi, A.; Srinivasan, S.; Gao, Z.; Ban, Y.; Chen, X.; et al. Targeting Tumor-Stromal IL6/STAT3 Signaling through IL1 Receptor Inhibition in Pancreatic Cancer. Mol. Cancer Ther. 2021, 20, 2280–2290. [Google Scholar] [CrossRef] [PubMed]
  153. Nagathihalli, N.S.; Castellanos, J.A.; VanSaun, M.N.; Dai, X.; Ambrose, M.; Guo, Q.; Xiong, Y.; Merchant, N.B. Pancreatic stellate cell secreted IL-6 stimulates STAT3 dependent invasiveness of pancreatic intraepithelial neoplasia and cancer cells. Oncotarget 2016, 7, 65982–65992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Lee, H.J.; Zhuang, G.; Cao, Y.; Du, P.; Kim, H.J.; Settleman, J. Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell 2014, 26, 207–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Sabet, S.; Rashidinejad, A.; Melton, L.D.; Duncan, J. Recent advances to improve curcumin oral bioavailability. Trend Food Sci. Technol. 2021, 110, 253–266. [Google Scholar] [CrossRef]
  156. Zhao, C.; Li, H.; Lin, H.J.; Yang, S.; Lin, J.; Liang, G. Feedback Activation of STAT3 as a Cancer Drug-Resistance Mechanism. Trends Pharmacol. Sci. 2016, 37, 47–61. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, Y.; Shen, Y.; Wang, S.; Shen, Q.; Zhou, X. The role of STAT3 in leading the crosstalk between human cancers and the immune system. Cancer Lett. 2018, 415, 117–128. [Google Scholar] [CrossRef]
Figure 1. Graphical schematic depicting molecular mechanism of STAT3-mediated acquired drug resistance in response to receptor tyrosine kinases within cancer cells. Adapted from Yang et al. [144].
Figure 1. Graphical schematic depicting molecular mechanism of STAT3-mediated acquired drug resistance in response to receptor tyrosine kinases within cancer cells. Adapted from Yang et al. [144].
Ijms 24 04722 g001
Table 1. Chemical structures and functions for standard chemotherapies, receptor tyrosine kinases, hormonal therapies, and monoclonal antibodies utilized to treat various cancers.
Table 1. Chemical structures and functions for standard chemotherapies, receptor tyrosine kinases, hormonal therapies, and monoclonal antibodies utilized to treat various cancers.
Generic Name
(Brand Name)
Chemical Structure & Formula Drug TargetMechanism of ActionReference Numbers
Cisplatin
(Platinol)
Ijms 24 04722 i001
cis-[Pt(NH3)2Cl2]
The N7 reactive center on purine nucleotide residues of DNAInterferes with DNA replication, commonly by forming 1,2 intra- or interstrand crosslinks [34,45,46,47,48,49,50,51,52,53,54,55]
Docetaxel
(Taxotere)
Ijms 24 04722 i002C43H53NO14Intracellular microtubular network and Bcl-2Stabilizes microtubule structures to impair depolymerization and attenuation of Bcl-2 expression effects[30,47]
Doxorubicin
(Adriamycin)
Ijms 24 04722 i003C27H29NO11DNA and the topoisomerase II complex of DNAIntercalates DNA and inhibits progression of topoisomerase II to stop the replication process[30,31,32,33,107,108,109,110,111]
Gemcitabine
(Gemzar)
Ijms 24 04722 i004C9H11F2N3O4DNA nucleotidesActs as a cytidine analog to replace nucleotides during DNA replication, causing cell arrest and apoptosis [34,35,36,37,38,39,40,41,42,43]
Paclitaxel
(Taxol and Others)
Ijms 24 04722 i005C47H51NO14Tublin subunits of intracellular microtubular networkHyperstabilizes microtubules to impair disassembly and ultimately block mitosis progression[60,61,62,63,64,112]
Cyclophosphamide
(Cytoxan)
Ijms 24 04722 i006
C7H15Cl2N2O2P
DNA strands at guanine N-7 positionsForms intrastrand and interstrand DNA cross-linkages[9]
Temozolomide
(Temodar)
C6H6N6O2The N7 position of guanine, N3 of adenine and the O6 position of guanineBroken down to a methyl 80-diazonium cation that methylates adenosine and guanine residues, causing DNA lesions to trigger apoptosis[55,56,57,58,59,113,114,115]
Afatinib
(Gilotrif)
Ijms 24 04722 i007
C24H25ClFN5O3
ErbB family:
EGFR (ErB1), HER2 (ErbB2), and HER4 (ErbB4)
Covalently binds to kinase domains, irreversibly preventing autophosphorylation by homo- or heterodimers[36,59,65,66]
Bortezomib
(Velcade)
Ijms 24 04722 i008C19H25BN4O426S proteasomeInhibits a ubiquitin-proteasome, preventing the degradation of pro-apoptotic factors to induce cell cycle arrest [12,16,21,24,25,27]
Crizotinib
(Xalkori)
Ijms 24 04722 i009
C21H22Cl2FN5O
ALK, HGFR, cMET, cROS, and RONInhibits phosphorylation in and creates an inactive protein confirmation[67]
Dasatinib
(Sprycel)
Ijms 24 04722 i010
C22H26ClN7O2S
BCR-ABL, SRC family, c-KIT, EPHA2, and PDGFRβCompetitively binds to ATP binding site of the kinase domain[68]
Erlotinib
(Tarceva)
Ijms 24 04722 i011C22H23N3O4Epidermal Growth Factor Receptor
(EGFR)
Inhibits intracellular phosphorylation (not fully characterized)[69,70,71,72,73]
Gefitinib
(Iressa)
Ijms 24 04722 i012
C22H24ClFN4O3
EGFRCompetitively binds to ATP binding site of the kinase domain[5,7,15,23,116]
Lapatinib
(Tykerb)
Ijms 24 04722 i013C29H26ClFN4O4SEGFR and
Epidermal Growth Factor Receptor 2 (HER2)
Competitively binds to intracellular ATP binding site of the receptor[10,13]
Vemurafenib
(Zelboraf)
Ijms 24 04722 i014
C23H18ClF2N3O3S
BRAF (V600E)Competitively binds to kinase domain of mutant form of BRAF [8,11,14,19,97]
Imatinib
(Gleevec)
Ijms 24 04722 i015C29H31N7OBCR-ABL, ABL, CSF1R, FLT-3, c-KIT, PDGFRCompetitively binds to activate site of kinase to block phosphorylation[17,18,26]
Enzalutamide
(Xtandi)
Ijms 24 04722 i016C21H16F4N4O2SAndrogen Receptor (AR)Competitively binds to the androgen receptor, preventing gene expression of AR targets[66,67,68]
Flutamide
(Eulexin)
Ijms 24 04722 i017C11H11F3N2O3Androgen ReceptorCompetitively binds to the androgen receptor blocking binding and uptake of testosterones [74,75,76]
Tamoxifen
(Nolvadex)
Ijms 24 04722 i018
C26H29NO
Estrogen Receptor (ER)Competitively inhibits binding of estrogen to its receptor [79,80,81,82]
Cetuximab
(Erbitux)
Ijms 24 04722 i019C6484H10042N1732O2023S36EGFRCompetitively inhibits the binding of epidermal growth factor (EGF) and other ligands to block the extended conformation of EGFR [83,84,85]
Trastuzumab
(Herceptin)
Ijms 24 04722 i020
C6470H10012N1726O2013S42
HER2Binds to extracellular ligand-binding domain, blocking its cleavage and inducing downregulation of the receptor[87,88,89,90,96]
Bevacizumab
(Avastin)
Ijms 24 04722 i021C6538H10034N1716O2033S44Vascular endothelial growth factor (VEGF)Binds and inactivates circulating VEGF, preventing its binding to receptor[86]
Pembrolizumab
(Keytruda)
Ijms 24 04722 i022C6504H10004N1716O2036S46PD-1
(CD279)
Binds to PD-1 on T cells to antagonize interactions with ligands, PD-L1 and PD-L2 [100]
Nivolumab
(Opdivo)
Ijms 24 04722 i023C6362H9862N1712O1995S42PD-1Binds to PD-1 on T cells to antagonize interactions with ligands, PD-L1 and PD-L2 [91,92,101,103]
Ipilimumab
(Yervoy)
Ijms 24 04722 i024C6572H10126N1734O2080S40CTLA-4
(CD152)
Binds CTLA-4, preventing its binding to CD28 on antigen presenting cells or T cells [91,92,95,102,105]
Atezolizumab
(Tecentriq)
Ijms 24 04722 i025
C6446H9902N1706O1998S42
PD-L1
(CD274)
Selectively binds to PD-L1, blocking its interaction with PD-1 or B7-1[6]
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

Singh, S.; Gomez, H.J.; Thakkar, S.; Singh, S.P.; Parihar, A.S. Overcoming Acquired Drug Resistance to Cancer Therapies through Targeted STAT3 Inhibition. Int. J. Mol. Sci. 2023, 24, 4722. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054722

AMA Style

Singh S, Gomez HJ, Thakkar S, Singh SP, Parihar AS. Overcoming Acquired Drug Resistance to Cancer Therapies through Targeted STAT3 Inhibition. International Journal of Molecular Sciences. 2023; 24(5):4722. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054722

Chicago/Turabian Style

Singh, Sunanda, Hector J. Gomez, Shreya Thakkar, Samara P. Singh, and Ashutosh S. Parihar. 2023. "Overcoming Acquired Drug Resistance to Cancer Therapies through Targeted STAT3 Inhibition" International Journal of Molecular Sciences 24, no. 5: 4722. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054722

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