Next Article in Journal
Predicting the Compression Index of Clayey Soils Using a Hybrid Genetic Programming and XGBoost Model
Next Article in Special Issue
Immunotherapy and MASLD-Related HCC: Should We Reconsider the Role of Etiology in the Therapeutic Approach to HCC?
Previous Article in Journal
Thermal Behavior of Alveoli with Phase Change Materials in Fruit Packaging
Previous Article in Special Issue
Glioma Stem Cells: GPRC5A as a Novel Predictive Biomarker and Therapeutic Target Associated with Mesenchymal and Stemness Features
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential and Challenges of Pioglitazone in Cancer Treatment

by
Maria Vasileiou
1,†,
Sotirios Charalampos Diamantoudis
2,†,
Christina Tsianava
3 and
Nam P. Nguyen
4,*
1
Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece
2
School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
3
Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
4
Department of Radiation Oncology, Howard University, Washington, DC 20060, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2025, 15(4), 1925; https://doi.org/10.3390/app15041925
Submission received: 22 November 2024 / Revised: 8 February 2025 / Accepted: 10 February 2025 / Published: 13 February 2025

Abstract

:
Pioglitazone (ACTOS) is a thiazolidinedione for peroxisome proliferator-activated receptor γ (PPAR-γ) that has been well established for the second or third line treatment of type 2 diabetes mellitus. Beyond the effects on glucose metabolism, pioglitazone displays positive effects on lipid metabolism, blood pressure, endothelial function, bone density, and apoptosis of cancer cells. In fact, according to in vitro experiments and preclinical studies, PPAR-γ ligand is currently considered a potential target for both chemoprevention and cancer therapy. PPAR-γ ligands are known to inhibit cancer cell proliferation and metastasis through terminal differentiation and underexpression of inflammatory mediators. Despite its anticancer properties, pioglitazone was withdrawn by the national medicine agencies of France and Germany, due to reports of increased incidence of bladder cancer. These reports were associated with European populations undergoing higher doses and longer durations of treatment. In this review, we discuss the pharmacokinetics, therapeutic potential, and limitations regarding the clinical use of pioglitazone, with a focus on cancer treatment.

1. Introduction

Thiazolidinediones (TZDs) are a class of oral hypoglycemic agents for the management of diabetes. TZDs are ligands for PPAR α and γ, a member of the nuclear receptor family which regulates glucose and lipid metabolism, inflammation, and cell proliferation [1,2,3]. PPARs are classified into three subtypes: PPAR-α, PPAR-β/δ, and PPAR-γ. Specifically, PPAR-α receptors are expressed in the heart, liver, and skeletal muscle, while PPAR-γ receptors are expressed in adipose tissue. The former regulate fatty acid oxidation followed by decreased low-density lipoprotein cholesterol (LDL-C) and increased high-density lipoprotein cholesterol (HDL-C) levels, while the latter regulate cell differentiation, leptin expression and insulin sensitivity [4,5]. As a result, TZDs find therapeutic application in the treatment of type 2 diabetes mellitus (T2DM), as well as non-alcoholic fatty liver disease (NASH) and multiple types of cancer [6,7].
Initially approved by the FDA in 1999, pioglitazone is marketed under the brand name ACTOS by Takeda Pharmaceuticals. It has been established as a second- or third-line treatment for the management of T2DM when first-line treatment with metformin is inadequate [8,9]. In this scenario, metformin is combined with sulfonylureas, glinides, alpha-glucosidase inhibitors, insulin, glucagon-like peptide-1 receptor agonist, dipeptidyl peptidase 4 inhibitors, sodium-glucose co-transporter 2 inhibitors, and thiazolidinediones, such as pioglitazone [10]. Pioglitazone is typically administered orally, with the optimal dose customized based on clinical status. According to the European Medicines Agency (EMA) recommendations, the initial dose ranges from 15 to 45 mg once daily. The dose may be increased if inadequate glycemic control is achieved. Alternatively, it may need to be decreased if the patient is receiving pioglitazone along with other diabetes medications (insulin, chlorpropamide, glibenclamide, gliclazide, and tolbutamide). The tablets should be administered once a day with water, either with or without food. After three to six months, the patients should be evaluated and treatment discontinued if considered non-beneficial [11].
However, despite its effectiveness in enhancing glycemic control, concerns have been raised about its safety profile, one of which regards an increased bladder cancer risk. Notably, indications of a higher incidence of bladder cancer among pioglitazone users prompted the French and German pharmaceutical regulators to withdraw the medication from the market in May 2011. Nevertheless, the medicine remained available throughout the rest of Europe [12].
The aim of this literature review is to thoroughly evaluate pioglitazone’s safety profile and efficacy, with an emphasis on its association with bladder cancer. This review summarizes clinical data, epidemiological studies, and regulatory recommendations in an effort to evaluate the risk–benefit ratio of pioglitazone. Its evaluation will shed light into current clinical practice and direct future studies to explore the multiple therapeutic applications of pioglitazone.

2. Overview of Pioglitazone

Pioglitazone, also known as 5-{4-[2-(5-ethyl-2-pyridinyl)ethoxy]benzyl}-1,3-thiazolidine-2,4-dione based on the International Union of Pure and Applied Chemistry (IUPAC), is a TZD compound, as shown in Figure 1, which is derived from a pyridine derivative through a nucleophilic aromatic substitution reaction with a 72% yield [13]. Pioglitazone appears as needles from dimethylformamide and water, while pioglitazone hydrochloride (HCl) appears as colorless prisms from ethanol, with a melting point of 183–184 °C and 193–194 °C, respectively [14]. The solubility of pioglitazone hydrochloride (HCl) increases with increasing temperature [15]. Low aqueous solubility and oral bioavailability can be overcome through temperature increase, cosolvents, and formulation strategies such as cyclodextrins inclusion complexes or nanostructured lipid carriers (NLCs). More specifically, polyethylene glycols (PEGs) are safe cosolvents for oral or parenteral pioglitazone HCl formulations. The additions of PEGs 200, 400, or 600, and a temperature increase significantly enhanced the solubility profile of pioglitazone [15]. Positive results were achieved through the addition of cyclodextrins in ethanol solution or encapsulation of pioglitazone HCl. Cyclodextrins caused a 6-fold increase in dissolution rate of pioglitazone HCl-β-cyclodextrin when compared to the pure drug, while lipid carriers achieved controlled release up to 24 h [16,17]. The latter was also observed during the administration of a newly discovered racemic compound that serves as a more thermodynamically stable and less soluble form of pioglitazone HCl [18].
The molecular structure and properties of pioglitazone have been well-characterized, providing a solid foundation for its pharmacological profile. The pharmacological activity of pioglitazone has been attributed to its hydroxy and ketone derivatives during in vivo studies on rats and dogs [13]. Similarly to other thiazolidinediones, pioglitazone activates the PPAR-γ receptor, increasing insulin sensitivity, without inducing insulin release from pancreatic cells. It is typically administered once a day with or without food, metabolized by the cytochrome P450 hepatic enzymes with a half-life of approximately 9 h, and excreted in the urine. The pharmacokinetics of pioglitazone are dose and time-dependent, leading to non-linear changes in plasma concentration and potential toxicity. According to clinical evidence, pioglitazone follows the same pharmacokinetic profile in healthy participants, diabetic patients, and patients with renal failure, irrespective of age, gender, and race. However, dose adjustment is required for patients with hepatic failure due to decreased metabolic function and by extension decreased plasma concentration [19].

3. Therapeutic Use of Pioglitazone

3.1. Diabetes Mellitus Type 2 (T2DM)

Thiazolidinediones (TZDs), also known as glitazones, have been well-established for the treatment of type 2 diabetes mellitus (T2DM) [20]. Given the pathophysiology of T2DM, targeting mechanisms regulating lipid metabolism is a viable option [21]. TZDs are peroxisome-proliferator activated receptor γ (PPAR-γ) agonists which regulate gene expression related to the uptake, distribution, and metabolism of lipids, resulting in reduced lipid levels and sensitization to insulin. However, the use of TZDs has been limited during the past years due to major safety and toxicity concerns, such as edema, cardiovascular disease, weight gain, and carcinogenesis reports, as analyzed below [22].

3.2. Nonalcoholic Fatty Liver Disease (NAFLD) and Nonalcoholic Steatohepatitis (NASH)

NASH is characterized as one of the most extreme forms of NAFLD, having the potential to lead to severe and often terminal hepatic disorders such as fibrosis, cirrhosis, and hepatocellular cancer [23]. Considering the association of NASH with T2DM and dyslipidemia, pioglitazone is a reasonable option against NASH in both diabetic and non-diabetic patients [24]. According to the study conducted by Della Pepa et al. (2021), pioglitazone has demonstrated positive effects against NASH, even in relatively small doses. This is achieved through the increase of the peripheral and adipose tissue sensitivity to insulin, leading to the reduction of hepatic inflammation and steatosis in a glucose-independent manner [25]. It is important to note several investigations of combination therapies against NASH, with the most significant one being pioglitazone combined with vitamin E [26,27].

3.3. Cardiovascular Protection

By lowering insulin resistance, TZDs can have cardioprotective and anti-atherosclerotic activity, and prevent ischemic events to an extent in patients with prior history of insulin resistance [28,29,30]. Additionally, it has been noted that, in murine models and in vitro studies, pioglitazone has demonstrated the capability of limiting cardiac hypertrophy in addition to arresting cardiac fibrosis and providing protection against oxidative stress [31,32,33].

3.4. Neurodegenerative Diseases (NDs)

There have been promising indications that PPAR-γ agonists, such as pioglitazone, possess the capability of reducing neuroinflammation, as well as the concentration phosphorylated tau protein and synaptophysin. This is complemented by the elevated permeability of the blood–brain barrier [34]. Focusing on Alzheimer’s Disease (AD), given that PPAR-γ regulates the expression of key enzymes related to the production and metabolism of amyloid plates, the inhibition of its phosphorylation, and therefore deactivation by pioglitazone, suggests favorable results, as illustrated in Figure 2 [35]. The neuroprotective properties of pioglitazone that mainly stem from the anti-inflammatory effects and the mitochondrial enhancement can also be applied for Parkinson’s Disease (PD), as well as amyotrophic lateral sclerosis (ALS), in which case the effects on the muscular tissue are also of significance [36,37]. As such, based on the results of studies conducted in rodents and humans, it is evident that pioglitazone can have a positive impact in the cognition and other neurological functions of patients with NDSs [38,39,40].

3.5. Anticancer Properties

3.5.1. Overview

As established, pioglitazone has been associated with elevated risk of bladder cancer, especially in the Caucasian population, to such extend that it has catalyzed its withdrawal from the French, German, and Indian markets, and led the Food and Drug Administration (FDA) to introduce a mandate for the providing of information regarding potential risk [41,42]. However, there have been several reports and studies that point out the antineoplastic properties of pioglitazone and the related biochemical pathways that PPAR-γ is implicated in. Given the pleiotropic effects of PPAR-γ and its agonists, we consider that investigating the exact molecular interactions will contribute to shedding some light on the mechanism of action and toxicity, as well as the interactions and effects of pioglitazone, and potentially suggest new applications and optimalization/personalization of interventions involving TZDs.

3.5.2. Molecular Pharmacology

PPAR-γ is classified as a nuclear receptor which, after binding with its ligand, translocates to the nucleus, heterodimerizes the retinoid X receptor (RXR), and acts as gene regulator via its interaction with the peroxisome proliferator response element (PPRE) of selected genes. The latter is related to metabolism in physiological as well as cancer cells [43]. As such, the receptor possesses the capability of affecting key cancer cell processes related to proliferation, progression, differentiation, metastasis, apoptosis, etc. Activation of PPAR-γ receptors suppresses tumorgenicity through their interaction with insulin growth factor, as illustrated in Figure 3. Additional molecular mechanisms of therapeutic significance are summarized in Table 1.

3.6. Therapeutic Effect on Different Types of Cancer

3.6.1. Breast Cancer

According to retrospective cohort studies, there is insignificant to almost no elevated breast cancer (BC) risk related to the use of pioglitazone [64]. It is important to note that PPAR-γ levels are reduced and methylation of PPAR-γ promoter is increased in BC cells, leading to the indication of its unfavorable role in cancer proliferation. In the example of triple negative breast cancer (TNBC), pioglitazone has proven its value as an adjuvant in the combined use with doxorubicin, as it decreases the invasiveness and tendency for migration, an issue commonly associated with the use of the latter. Similar findings were also reported for the apoptosis-inducing capabilities of cisplatin during in vitro testing [50,65]. PPAR-γ facilitates major regulatory processes such as the promotion of CXCR4 and CXCR7 genes, inhibition of eIF2α, and intracellular accumulation of glucose-affecting related migration pathways [66,67]. Despite the aforementioned evidence, there is no clinical application yet [68,69].

3.6.2. Lung Cancer

According to clinical evidence, PPAR-γ agonists play a major role lung cancer progression. In fact, PPAR-γ agonists have been associated with differentiation, tumor size, BCL2/BAX ratio and c-MYC expression levels, in cases of non-small cell lung cancer (NSCLC), lung adenocarcinoma, and squamous cell lung cancer (SCLC) [70]. Pioglitazone has demonstrated the ability to reduce NSCLC proliferation and invasiveness through downregulation and altering pathways of major significance in proliferation, apoptosis, angiogenesis, and metastatic potential. Indeed, rodent studies have shown that pioglitazone has the potential to reduce tumor formation and volume [71]. Additionally, inflammation is also affected, as pioglitazone inhibits prostaglandin 2 (PGE2) production while bypassing cyclooxygenase 2 (COX-2) [59]. As a chemopreventive medication, pioglitazone has proven to have little value considering the double-blind clinical trial conducted by Keith et al. (2019), which reported that no significant alterations in the endpoints were noted other than some minor effect on lesions [72]. Chemopreventive studies were also carried out by Seabloom et al. (2017), who noted significant reduction in adenoma formation but no noteworthy alteration to the effect of metformin when combined with it, and by a team of similar composure, who evaluated the effectiveness of pioglitazone in an aerosol form [73]. They concluded from animal tests that a dose of 150–450 μg/kg was tolerable, with limited to no adverse effects, and effective, given the reduction in adenoma formation after biochemical and histological assessments [74]. As with BC, pioglitazone has also been considered for combination therapy along with more established anticarcinogenic agents, as studied in the following cases:
  • The use with EGFR Tyrosine Kinase Inhibitors such as gefitinib for NSCLC, as it was found that PPAR-γ mediated upregulation of phosphate, and tensin homolog (PTEN) downregulated the PI3K/Akt pathway that is correlated to resistance to said TKIs [70].
  • Combination of pioglitazone with clarithromycin and a relatively small dose of chemotherapeutic agent was compared against nivolumab in the ModuLung trial by Heudobler et al. (2021) [75]. This trial was terminated early due to the approval of checkpoint inhibitors as first line treatment, with the conclusion that nivolumab was superior to this combination therapy; however, with a difference in the overall survival rate and quality of life between the two regimens being similar, the latter seems to be a viable alternative to be assessed in future trials and be considered in cases with few other options [75].
Granted that inactivation of PPAR-γ combined with increased presence of prostaglandins is a dangerous sign, as it leads to loss of control over Ras/Raf/Mek activation and NF-Κβ mediated proliferation, Kiran et al. (2022) tested pioglitazone along with COX-2 specific antagonist celecoxib on rodents. The results were coherent with the hypothesis as decreases in tumor size, alterations to tumor architecture, and increase in lifespan, as well as improvement of its quality, were noted [76].
However, PPAR-γ also possesses tumor promoting effects regarding lung cancer [77], such as the case of increased tumor progression in orthotropic mice after systemic PPAR-γ activation in the cancer cells as well as the tumor microenvironment [59], and, as such, additional studies regarding the exact effects of pioglitazone in a clinical setting and risk–benefit assessment in the given context are needed.

3.6.3. Renal Cancer

It has been noted that the antidiabetic use of pioglitazone is not associated with elevation in the risk of renal cancer [78]. Pioglitazone itself induced apoptosis in Caki cells, a model used for the study of clear cell renal cell carcinoma, as shown in Table 1 [54]. Such effects are expected, considering the aforementioned, as well as the elevated presence of PPAR-γ receptors in renal carcinoma cells. Another cell line, 769-P cells, was heavily affected in a dose-dependent manner with imminent apoptosis as the application of the molecule led to significant morphological alterations, including cell density. The suggested mechanism of apoptosis, with limited proof, in this case, was mitochondrial with membrane irregularities and release of cytochrome c. This cell line also exhibited increased sensitivity to methotrexate after treatment with pioglitazone, something that was not evident in the Vero cell line [79]. Finally, there is a lot of interest in the combination of pioglitazone with cisplatin, as it showed nephroprotective effects in rodent models HK-2 cells by preventing tissue damage and oxidative stress and it potentiated the latter’s effects on renal adenocarcinoma cells inhibiting NF-κB-mediated inflammatory response and activation of Nrf2-mediated antioxidative stress signaling [80].

3.6.4. Hepatocellular Carcinoma

Assessing the preventative potential of pioglitazone, metanalytical results indicate that the use of TZDs in patients with T2DM is associated with protection against liver cancer [81]. Complementary rodent studies indicated prevention of liver fibrosis and carcinogenesis following cirrhosis [82]. Results originating from preclinical studies showcase pioglitazone’s anticancer properties. The increased presence of the receptor for advanced glycation end products (RAGE) is deemed as a negative biomarker for the progression of hepatocellular carcinoma which possibly indicates invasion. Pioglitazone decreased the expression of RAGE along with HMGB1, NF-κβ, and p38MAPK, resulting in reduced proliferation, invasion, and metastatic potential [83]. Additionally, it was reported that metabolomic alterations triggered by pioglitazone, mostly regarding the metabolism of lipids, leading to the death of chemically induced hypoxic HepG2 cells from oxidative stress [84].

3.6.5. Colorectal Cancer

Similarly to hepatocellular carcinoma, it has been indicated that TZDs reduce the risk of colorectal cancer occurrence in patients with T2DM after metanalytical assessment of about 2.5 million T2DM patients [85]. Direct effects were reported during in vitro experiments with pioglitazone and its analog, Δ2-pioglitazone treated HCT116 and HT29 cells. There were similarities in the endpoints, such as cell growth arrest and differentiation, in addition to key differences in the ways those were achieved, namely arrest in the S phase when treated with the former and in the G0/G1 phase when treated with the latter [86]. Additionally, xenograft studies on rodents using HT29 and SW480 cell lines indicated antiproliferative and antimetastatic activity towards the liver activity [85]. As an adjuvant, pioglitazone can be used in immunotherapy due to the autophagic degradation of PD-L1 mediated by PPAR-γ whose binding with the immunomodulatory ligand is similarly enhanced by said xenobiotic agent in the case of lung cancer [62]. Investigations in the field of pharmaceutics addressing pioglitazone’s poor solubility have shown that its combination with capecitabine, an established chemotherapeutic molecule against colorectal cancer, in the form of nanoparticles, enhances its bioavailability and overall proapoptotic effect on HT29 and HCT119 cells [87].

3.6.6. Thyroid Cancer

The use of pioglitazone has been correlated with neither increased nor decreased risk of thyroid cancer according to a study comprising mostly Eastern Asian individuals [88]. A widely studied finding is the PAX8-PPAR-γ Fusion Protein (PPFP) and how pioglitazone alters its function. PPFP is a result of a fused gene containing the significant majority of the Paired Box Gene 8 (PAX8) transcription factor, related to thyroid development and function, gene, and the entirety of the PPAR-γ gene and, as such, a binding site for TZDs [89]. As evident by the imminent intracellular lipid accumulation and the expression of adipocyte-related biomarkers upon treatment, pioglitazone induces the differentiation of thyroid cancer cells into adipocytes. Moreover, it is accepted that pioglitazone blocks the interaction between PPFP and Thyroid Transcription Factor 1 (TTF-1), which otherwise leads to the severe limiting of the differentiation induction by the former via the suspension of recruitment of synergistic molecules such as coactivators and corepressors [90]. Considering immunity, pioglitazone also advanced the infiltration of T cells and macrophages in the tumor microenvironment when tested on mice, and exhibited favorable properties in a clinical setting [91]. Unfortunately, Giordano et al. (2018), in their examination of 40 thyroid cancer patients, found that only one tested positive for PPFP, potentially indicating that it is a rare phenomenon in these cases [92]. Finally, studies have suggested that pioglitazone and metformin have a synergistic effect against thyroid cancer, especially in the induction of apoptosis [93].

3.6.7. Glioma

As mentioned before, pioglitazone can cross the blood–brain barrier, a capability that can be utilized in the case of glioma, as it is one of the most aggressive brain malignancies. Cytotoxic activity has been confirmed, as it has been reported to reduce glioma cell proliferation in vitro in the U87MG, T98G, and U251MG cell lines, as well as LN-229 cells in a xenograft model [94,95]. However, in another xenograft study using Gl261 cells, pioglitazone seemed to significantly alter the fate of the test subjects only when injected into the cerebrum [96]. Antineoplastic action can be by conventional, PPARγ-dependent and -independent pathways previously mentioned, namely alterations in the levels of cyclin D1, MMP9, N-Cadhenin and caspase 3, in addition to β-catenin inhibition, as revealed by knockout studies [97,98]. Invasiveness is also affected, as it was limited to C6 rat glioma cells [99]. Complimentary to this is the evident increase in the expression of the Excitatory Amino Acid Transporter 2 (EAAT2) in the same cell lines, except T98G. Lack of this transporter is commonly found in glioma cells as the resulting elevation of extracellular glutamate concentration leads to excitotoxicity, assisting in the thriving of malignant cells, as an excitatory neurotransmitter, and sets the field for tumor associated epilepsy (TAE). As such, by increasing EAAT2 levels in glioma cells, pioglitazone worsens the conditions, not favoring the development of glioma and, potentially, the associated symptoms such as TAE [100]. The effect of pioglitazone has been further investigated in glioma stem cells (GSCs), which do not seem to be immune from the effects of pioglitazone either. In vitro tests by Cilibrasi et al. (2016) involving treatment of GSCs revealed that G166, GliNS2, GBM2, and G144 (with latent effects) cells reduced their metabolic rate and alteration in the proteomic profile regarding the expression of markers related to differentiation and stemness. These changes in the behavior of the cells were not accompanied by any alterations in their morphology [101]. Data originating from the clinical data reveal a preventative relationship between pioglitazone and glioma [94]. A synergy between TZDs (with pioglitazone being the most potent of them) and statins has been suggested by Tapia-Perez et al. (2010), as they observed a significant reduction in the population in in vitro tests using the U87 and RG II cell lines, even in hypoxic conditions [102]. Follow-up rodent model tests, also by Tapia Perez et al. (2016), revealed no significant differences in overall survival rate compared to the controls, and that the only combination displaying reduction in tumor size was lovastatin/atorvastatin + plus pioglitazone [103]. Related to cancer care, pioglitazone has also been found to protect against radiation induced cognitive decline in patients with brain malignancy and is relatively safe for use in radiation treatment [104].

3.6.8. Hematological Malignancies

There is a great amount of interest in the potential of pioglitazone in blood cancers. Studies conducted by Esmaeili et al. (2021) on U937 cells, modeling acute myeloid leukemia (AML) cancer cells, have shown in their results that pioglitazone dampened their survival and development capabilities, led to accumulation of cells in the G1 and pre-G1 cell cycle phases, and induction of apoptosis, as indicated by the increase in the population of cells that test positive for Annexin-V and PI [105]. Similar targeted antiproliferative effects were also observed on HL60 (modeling Acute Pro-myelocytic Leukemia-APL), K562 (modeling Chronic Myeloid Leukemia), and Jurkat (modeling T-cell lymphoma) cells, and the cell cycle of the HL60 line also demonstrated accumulation in the G1 phase and decrease in the G2 and metaphasis populations [106]. Jurkat cells were also assessed with the combination of pioglitazone and valproic acid, with optimistic results as cell cycle deregulation was noted [107,108,109]. According to in vitro studies in different types of leukemia—AML in U937 cells, APL in Jurkat cells, ALL in SD-1, IM-9, Sup-B15, and NALM-6 cells—the primary mode of action for pioglitazone is cell cycle arrest [110]. Adding to the above are the tests on Philadelphia chromosome positive leukemic cells by Okabe et al. (2017), which reported the antineoplastic effects of pioglitazone on cells bearing the T315I mutation while sparing normal CD34 expressing cells and inducing the phosphorylation of the AMP-activated protein kinase (AMPK) [111]. The combination of pioglitazone with PI3K inhibitors can potentiate its cytotoxic effects, as was observed on NB4 cells (APL) when they were treated with pioglitazone, and PI3K inhibitors CAL-101 and BKM120. The suggested mechanism of synergy is the cell cycle arrest, as analyzed before, mediated by p21 [112]. However, in NALM-6 cell line experiments, where similar synergy was noted, it was found that said effects are interfered with by NF-κβ signaling and autophagy [113]. Potential synergy was also noticed in more conventional chemotherapeutics with a statistically insignificant remission rate being reported for cytarabine and daunorubicin in the clinical study conducted by Ghadiany et al. (2019) [114]. Additionally, as noted in Table 1, pioglitazone is correlated with elevated effectiveness of arsenic trioxide [115]. Adjuvant use of pioglitazone has been proven to positively influence interventions utilizing BCR-ABL1 tyrosine kinase inhibitors (TKIs) in Chronic Myeloid Leukemia (CML). In vitro experiments carried out on K562 cells by Glodkowska-Mrowka et al. (2016) resulted in the eradication of leukemia stem cells (LSCs), progenitors, and differentiated CML cells [116,117]. A small phase II clinical trial by Rousselot et al. (2016) involving 24 subjects treated with imatinib, who were administered pioglitazone at a dosage ranging from 30mg/d to 45mg/d, exhibited that about half of the patients showed response within one year, with no significant adverse effects other than edema, higher than the rate of 23% in monotherapy with imatinib, thus suggesting correlation between the synergy of pioglitazone and favorable outcomes [118]. Additional discontinuation trials such as EDI-PIO in Brazil have raised the issue of alterations in the lipid metabolism and metabolomic profile of the patients, given the effects of pioglitazone on the adipose tissue, and the aspect of mitochondrial dysfunction that influences the proliferation of CML cells [106]. However, results originating from the same study, suggest that pioglitazone does not affect STAT5 expression and, therefore, the levels of downstream molecules to it, such as HIF-2α, contrary to the statements of previous reports [119].

3.6.9. Pancreatic Cancer

There has been contradictory evidence regarding the association between the antidiabetic use of pioglitazone and the elevation of the risk of pancreatic cancer. As such, the effect the molecule has on pancreatic cancer occurrence, and the extent of it, is yet to be determined [120,121,122,123]. Nevertheless, antitumor activity has been observed in cell lines modeling pancreatic cancer, including Capan-1, Apsc-1 BxPC-3, PANC-1, and MIApaCa-2. Xenograft studies involving BxPC-3 revealed antiproliferative and antimetastatic properties, as tumor size was limited along with lymph metastasis [124]. Finally, pioglitazone has been reported to enhance the anticancer effects of gemcitabine through inhibition of the NF-κβ, induction of apoptosis, and setting of the platform to include HDAC inhibitors that have proven to be helpful in the activity of PPAR-γ, as illustrated in Figure 4 [125].

3.7. Immunotherapy

PPAR-γ immunomodulatory effects have long been known, and there have been multiple studies seeking to assess the significance of said effects in cancer. A study conducted on mice hosting solid Ehrlich carcinoma cells by El-Sisi et al. (2014) concluded that pioglitazone administration led to a statistically significant reduction in tumor size and TNF-α amount, increase in peripheral neutrophil and splenic T- lymphocyte populations, and increase in splenic T-lymphocyte activity, CD4+/CD8+ ratio and immunoglobin G (IgG) levels, indicating elevated immune response. Similar results, although to a varying extent and except peripheral neutrophil counts, were also obtained from co-administration with doxorubicin [126]. Immunomodulation is a property that can be exploited in the context of immunotherapy. An example of it is the use of PPAR-γ agonists to reprogram T cells; however, there were no specific reports on pioglitazone [127,128].

3.8. Clinical Evidence

To evaluate the antineoplastic potential of pioglitazone, it is important to review the latest clinical trials (Supplementary Table S1—from clinicaltrials.gov platform) [129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155]. Moghareabed et al. investigated the combined use of pioglitazone with chemotherapy for metastatic breast cancer treatment. Despite the study’s statistical power being significantly restricted due to the small sample size of just 60 female participants, it was concluded that combining pioglitazone with chemotherapy was more effective than using taxol alone. This combination resulted in better radiologic responses, disease stability, and halted progression, ultimately providing a greater overall clinical benefit [156]. In contrast, the study by Agulló-Ortuño et al. showed no clear superiority of one combination over the other, when participants received carboplatin (AUC = 2) and paclitaxel (80 mg/m2) weekly in combination with either hydroxyurea (1000 mg and 1500 mg) or pioglitazone (30, 45 and 60 mg) daily. However, treatment with hydroxyurea demonstrated slightly more favorable indications based on the measurement of uncoupling protein-2 gene expression and rate of cells with double-strand breaks [157].
Besides therapeutic use, pioglitazone can also play a role in the diagnosis of cancer. Han et al. have studied the effect of pioglitazone on 18F-2-fluoro-2-deoxy-D-glucose (18F-FDG) uptake by neoplastic lesions. In the study group of 43 patients (13F/30M, Median Age 64.0), the participants were scanned 1h after intravenous administration of 18F-FDG (5.5 MBq/kg). The same procedure was repeated the next day, 2h after the oral administration of pioglitazone (15 or 30 mg). Results indicated a dose-independent betterment in all assessed aspects of positron emission tomography (PET) imaging in the group that responded to the administration of pioglitazone, with discrimination between the effect on malignant and inflammatory lesions. However, it must be highlighted that, like the rest of the trials discussed, this one also suffers from a small sample size, dampening the statistical significance of the results [158].
Despite the substantial number of clinical trials, evidence regarding the use of pioglitazone for cancer prevention and treatment remains insufficient. This can be attributed to the low number of participants and lack of reports, which limit the statistical value of the reported results. Thus, a large number of clinical trials is required to properly assess the antineoplastic potential of pioglitazone. More specifically, there is a need to further investigate combination therapies and adverse events. For this purpose, multiple parameters should be taken into consideration, such as population, duration of treatment, and follow-up appointments. Regarding population, its size and diversity need to be increased, especially in phase II and III trials, for future studies to possess enough power to be considered for applications in standardized cancer care.

4. Discussion

Concerns have been raised regarding the elevated risk of bladder cancer, which has been reported since 2012. This can be attributed to overexpression of PPARγ, which is present in the urinary bladder and several other tissues. This adverse event is prevalent, especially in cases of prolonged pioglitazone administration, indicating a time-dependent and dose-dependent pharmacological profile. However, analysis does not suggest a strong dose-dependence relationship, since bladder cancer risk can be attributed to comorbidities and administration of antidiabetic drugs, such as metformin. Existing studies suggest a slightly, but significantly, elevated risk of bladder cancer associated with pioglitazone administration in a time- and dose-dependent manner [41]. This suggestion is confirmed by the cohort study conducted by Mamtani et al. (2012), who initiated TZD or SU treatment in 18,459 and 41,396 T2DM patients, respectively. Out of the 59,855 patients, 33,730 were males and 26,125 were females. Regarding bladder cancer incidence, 60 cases were reported in the TZD group and 137 cases were reported in the SU group. No difference was found between the two groups. Interestingly, in the TZD group long-term treatment (≥5 years) was associated with an increased risk of bladder cancer, but not among those with shorter duration of exposure to TZDs. This observation was not limited only to pioglitazone, but may apply to other TZDs [159].
Another cohort study which gathered data from the French national health insurance information system, indicates the time- and dose-dependent pharmacological profile of pioglitazone. The study included 155,535 T2DM patients, out of whom 83,755 were males and 71,780 were females. The risk was increased by 75% for cumulative doses ≥28,000 mg over the span of ≥ 24 months. Analysis by sex revealed a significant association between pioglitazone and bladder cancer for males, but not for females [160]. This was contradicted by the meta-analysis of Tang et al. (2018), who found no gender difference, though they did indicate a higher bladder cancer risk for Europeans versus US or Asian populations, which could be attributed to dissimilar genetic backgrounds or socioeconomic factors [161]. Lastly, it is important to acknowledge that T2DM patients have an increased risk of bladder cancer at baseline. Other confounding factors, such as cigarette smoking, exposure to arylamines, and schistosomal infection, should be taken into consideration. Thus, it is plausible that the TZDs may exacerbate a pre-existing high-risk state of T2DM patients [162].
Due to the structure of these studies, there is no conclusive data regarding the risk–benefit ratio of pioglitazone. To date, there is no study that proves the direct cause–effect relationship between pioglitazone and bladder cancer [41].
Similar concerns have been raised regarding the association of metformin with risk of bladder cancer. While metformin does not decrease bladder cancer risk, it improves clinical parameters such as recurrence-free survival (RFS), progression-free survival (PFS), and cancer-specific survival (CSS) [121]. Although no effect was reported between overall survival (OS) and bladder cancer, studies show a positive outlook [122]. Another meta-analysis conducted in bladder cancer patients showed neither a protective nor a harmful effect of metformin exposure [123]. Few reports indicate decreased bladder cancer risk in metformin users versus never-users. However, the link between metformin and bladder cancer risk is not clear due to co-administration of other drugs and small proportion of metformin users versus never-users [124]. Overall, meta-analyses show no association of metformin with bladder cancer risk. Considering that these studies are limited to T2DM or bladder cancer patients, a consensus cannot be drawn regarding the risk of bladder cancer in the general population.

5. Conclusions

Based on the aforementioned, it is safe to assume that pioglitazone, as well as other TZDs, possess antineoplastic agents properties. PPARγ receptors, being implicated in multiple pathways, lead to diverse results in tumors. Although in vitro studies have been carried out with positive results, the adverse events presented indicate that additional studies are required in order to fully assess the risk to benefit ratio in an evidence-based manner. While in vitro studies have been carried out with positive results, cohort studies indicate a time- and dose-dependent relationship between pioglitazone administration and bladder cancer risk. Higher risk is observed in T2DM patients undergoing treatment for more than 2 years and at higher cumulative doses. Considering that overall risk remains relatively low, pioglitazone administration is recommended with caution for individuals at higher risk.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/app15041925/s1, Table S1: Molecular mechanisms of pioglitazone.

Author Contributions

Conceptualization, M.V., S.C.D., C.T. and N.P.N.; investigation, M.V., S.C.D., C.T. and N.P.N.; writing—original draft preparation, M.V., S.C.D., C.T. and N.P.N.; writing—review and editing, M.V., S.C.D., C.T. and N.P.N.; supervision, N.P.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Figure 1 was created using ACD/ChemSketch, version 2021.2.0 (Advanced Chemistry Development, Inc., Canada; www.acdlabs.com, 2022 (accessed on 25 January 2025)). Figure 2, Figure 3 and Figure 4 were created using https://biorender.com. URL accessed on 25 January 2024.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mal, S.; Dwivedi, A.R.; Kumar, V.; Kumar, N.; Kumar, B.; Kumar, V. Role of Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) in Different Disease States: Recent Updates. Curr. Med. Chem. 2021, 28, 3193–3215. [Google Scholar] [CrossRef]
  2. Souza-Tavares, H.; Miranda, C.S.; Macedo, I.; Sandoval, C.; Santana-Oliveira, D.A.; Silva-Veiga, F.M.; Fernandes-da-Silva, A.; Souza-Mello, V. Peroxisome Proliferator-Activated Receptors as Targets to Treat Metabolic Diseases: Focus on the Adipose Tissue, Liver, and Pancreas. World J. Gastroenterol. 2023, 29, 4136–4155. [Google Scholar] [CrossRef] [PubMed]
  3. Dixit, G.; Prabhu, A. The Pleiotropic Peroxisome Proliferator Activated Receptors: Regulation and Therapeutics. Exp. Mol. Pathol. 2022, 124, 104723. [Google Scholar] [CrossRef] [PubMed]
  4. Qaoud, M.T.; Al-masri, I.; Önkol, T. Peroxisome Proliferator-Activated Receptors as Superior Targets for Treating Diabetic Disease, Design Strategies—Review Article. Turk. J. Pharm. Sci. 2022, 19, 353–370. [Google Scholar] [CrossRef] [PubMed]
  5. Dana, N.; Ferns, G.A.; Nedaeinia, R.; Javanmard, S.H. Leptin Signaling in Breast Cancer and Its Crosstalk with Peroxisome Proliferator-Activated Receptors α and γ. Clin. Transl. Oncol. 2022, 25, 601–610. [Google Scholar] [CrossRef] [PubMed]
  6. Chandra, A.; Kaur, P.; Sahu, S.K.; Mittal, A. A New Insight into the Treatment of Diabetes by Means of Pan PPAR Agonists. Chem. Biol. Drug Des. 2022, 100, 947–967. [Google Scholar] [CrossRef] [PubMed]
  7. Staels, B.; Butruille, L.; Francque, S. Treating NASH by Targeting Peroxisome Proliferator-Activated Receptors. J. Hepatol. 2023, 79, 1302–1316. [Google Scholar] [CrossRef]
  8. Takada, N.; Genda, K. Pioglitazone (Actos, Glustin). In Drug Discovery in Japan; Springer: Singapore, 2019; pp. 169–181. [Google Scholar] [CrossRef]
  9. Dahlén, A.D.; Dashi, G.; Maslov, I.; Attwood, M.M.; Jonsson, J.; Trukhan, V.; Schiöth, H.B. Trends in Antidiabetic Drug Discovery: FDA Approved Drugs, New Drugs in Clinical Trials and Global Sales. Front. Pharmacol. 2022, 12, 807548. [Google Scholar] [CrossRef] [PubMed]
  10. Singh, A.K.; Singh, R.; Chakraborty, P.P. Diabetes Monotherapies versus Metformin-Based Combination Therapy for the Treatment of Type 2 Diabetes. Int. J. Gen. Med. 2021, 14, 3833–3848. [Google Scholar] [CrossRef] [PubMed]
  11. European Medicines Agency. Actos: EPAR—Product Information. 2024. Available online: https://www.ema.europa.eu/en/documents/product-information/actos-epar-product-information_en.pdf (accessed on 25 January 2025).
  12. Papaetis, G.S. Pioglitazone, Bladder Cancer, and the Presumption of Innocence. Curr. Drug Saf. 2022, 17, 294–318. [Google Scholar] [CrossRef] [PubMed]
  13. Tanis, S.P.; Parker, T.T.; Colca, J.R.; Fisher, R.M.; Kletzein, R.F. Synthesis and Biological Activity of Metabolites of the Antidiabetic, Antihyperglycemic Agent Pioglitazone. J. Med. Chem. 1996, 39, 5053–5063. [Google Scholar] [CrossRef] [PubMed]
  14. Patel, T.; Patel, V. Thiazolidinediones: Recent Development in Analytical Methodologies. J. Chromatogr. Sci. 2023, 62, 789–802. [Google Scholar] [CrossRef] [PubMed]
  15. Soltanpour, S.; Zohrabi, F.; Bastami, Z. Thermodynamic Solubility of Pioglitazone HCl in Polyethylene Glycols 200, 400 or 600+Water Mixtures at 303.2 and 308.2K—Data Report and Modeling. Fluid Phase Equilibria 2014, 379, 180–184. [Google Scholar] [CrossRef]
  16. Beloshe, S.P.; Chougule, D.D.; Shah, R.R.; Ghodke, D.S.; Pawar, N.D.; Ghaste, R.P. Effect of Method of Preparation on Pioglitazone HCl-β-Cyclodextrin Inclusion Complexes. Asian J. Pharm. 2010, 4, 168–172. [Google Scholar] [CrossRef]
  17. Faiz, S.; Arshad, S.; Kamal, Y.; Imran, S.; Asim, M.H.; Mahmood, A.; Inam, S.; Irfan, H.M.; Riaz, H. Pioglitazone-Loaded Nanostructured Lipid Carriers: In-Vitro and In-Vivo Evaluation for Improved Bioavailability. J. Drug Deliv. Sci. Technol. 2023, 79, 104041. [Google Scholar] [CrossRef]
  18. Zhang, C.; Matzger, A.J. A Newly Discovered Racemic Compound of Pioglitazone Hydrochloride Is More Stable than the Commercial Conglomerate. Cryst. Growth Des. 2017, 17, 414–417. [Google Scholar] [CrossRef]
  19. Eckland, D.; Danhof, M. Clinical Pharmacokinetics of Pioglitazone. Exp. Clin. Endocrinol. Diabetes 2000, 108, 234–242. [Google Scholar] [CrossRef]
  20. Taylor, S.I.; Yazdi, Z.S.; Beitelshees, A.L. Pharmacological Treatment of Hyperglycemia in Type 2 Diabetes. J. Clin. Investig. 2021, 131, e142243. [Google Scholar] [CrossRef] [PubMed]
  21. Singh, G.; Kumar, R.; Desna, D.S.; Chaudhary, M.; Kaur, C.; Khurrana, N. Thiazolidinedione as a Promising Medicinal Scaffold for the Treatment of Type 2 Diabetes. Curr. Diabetes Rev. 2023, 20, e201023222411. [Google Scholar] [CrossRef] [PubMed]
  22. Maheshwari, A.; Dhoat, P. USE, Safety and Adverse Effects of Thiazolidinediones—A Review. World J. Pharm. Res. 2023, 12, 111–119. [Google Scholar]
  23. Zhao, Y.; Zhao, W.; Wang, H.; Zhao, Y.; Bu, H.; Takahashi, H. Pioglitazone on Nonalcoholic Steatohepatitis: A Systematic Review and Meta-Analysis of 15 RCTs. Medicine 2022, 101, e31508. [Google Scholar] [CrossRef]
  24. Wang, Z.; Du, H.; Zhao, Y.; Ren, Y.; Ma, C.; Chen, H.; Li, M.; Tian, J.; Xue, C.; Long, G.; et al. Response to Pioglitazone in Non-Alcoholic Fatty Liver Disease Patients with vs. without Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Front. Endocrinol. 2023, 14, 1111430. [Google Scholar] [CrossRef] [PubMed]
  25. Della Pepa, G.; Russo, M.; Vitale, M.; Carli, F.; Vetrani, C.; Masulli, M.; Riccardi, G.; Vaccaro, O.; Gastaldelli, A.; Rivellese, A.A.; et al. Pioglitazone Even at Low Dosage Improves NAFLD in Type 2 Diabetes: Clinical and Pathophysiological Insights from a Subgroup of the TOSCA.IT Randomised Trial. Diabetes Res. Clin. Pract. 2021, 178, 108984. [Google Scholar] [CrossRef]
  26. Mazhar, I.; Yasir, M.; Sarfraz, S.; Shlaghya, G.; Narayana, S.; Mushtaq, U.; Ameen, B.S.; Nie, C.; Nechi, D.; Penumetcha, S. Vitamin E and Pioglitazone: A Comprehensive Systematic Review of Their Efficacy in Non-Alcoholic Fatty Liver Disease. Cureus 2023, 15, e43635. [Google Scholar] [CrossRef] [PubMed]
  27. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  28. Nesti, L.; Tricò, D.; Mengozzi, A.; Natali, A. Rethinking Pioglitazone as a Cardioprotective Agent: A New Perspective on an Overlooked Drug. Cardiovasc. Diabetol. 2021, 20, 109. [Google Scholar] [CrossRef] [PubMed]
  29. Spence, J.D.; Viscoli, C.; Kernan, W.N.; Young, L.H.; Furie, K.; DeFronzo, R.; Abdul-Ghani, M.; Dandona, P.; Inzucchi, S.E. Efficacy of Lower Doses of Pioglitazone after Stroke or Transient Ischaemic Attack in Patients with Insulin Resistance. Diabetes Obes. Metab. 2022, 24, 1150–1158. [Google Scholar] [CrossRef]
  30. Camafort, M.; Yang, E.; Ponte, C.I.; Coca, A. Therapeutic Strategies to Prevent Recurrent Stroke. In Hypertension and Brain Damage. Updates in Hypertension and Cardiovascular Protection; Springer: Cham, Swizterland, 2024; pp. 229–243. [Google Scholar] [CrossRef]
  31. Teng, Y.M.; Liu, W.Z.; Yang, L. Pioglitazone Ameliorates Hypertension Induced Cardiac Hypertrophy and down Regulates Cardiac Hypoxia Inducible Factor-l α in Rats. Pak. J. Zool. 2023, 55, 1657–1663. [Google Scholar] [CrossRef]
  32. Zhang, X.D.; Sun, G.X.; Guo, J.J.; Hu, C.C.; Sun, R.C.; Yu, H.C. Effects of PPARγ agonist pioglitazone on cardiac fibrosis in diabetic mice by regulating PTEN/AKT/FAK pathway. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 812–819. [Google Scholar]
  33. Mannucci, E.; Giaccari, A.; Gallo, M.; Targher, G.; Pintaudi, B.; Candido, R.; Monami, M. Effects of Pioglitazone on Cardiovascular Events and All-Cause Mortality in Patients with Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Nutr. Metab. Cardiovasc. Dis. 2021, 32, 529–536. [Google Scholar] [CrossRef] [PubMed]
  34. Alhowail, A.; Alsikhan, R.; Alsaud, M.; Aldubayan, M.; Rabbani, S.I. Protective Effects of Pioglitazone on Cognitive Impairment and the Underlying Mechanisms: A Review of Literature. Drug Des. Dev. Ther. 2022, 16, 2919–2931. [Google Scholar] [CrossRef] [PubMed]
  35. Saunders, A.M.; Burns, D.K.; Gottschalk, W.K. Reassessment of Pioglitazone for Alzheimer’s Disease. Front. Neurosci. 2021, 15, 666958. [Google Scholar] [CrossRef]
  36. Zamanian, M.Y.; Terefe, E.M.; Taheri, N.; Kujawska, M.; Tork, Y.J.; Abdelbasset, W.K.; Shoukat, S.; Jade, M.; Heidari, M.; Alesaeidi, S. Neuroprotective and Anti-Inflammatory Effects of Pioglitazone on Parkinson’s Disease: A Comprehensive Narrative Review of Clinical and Experimental Findings. CNS Neurol. Disord. Drug Targets 2023, 22, 1453–1461. [Google Scholar] [CrossRef]
  37. Wan, M.; Zhang, L.; Huo, J.; Fu, Y.; Huang, T.; Fan, D. Genetic Variation in Targets of Antidiabetic Drugs and Amyotrophic Lateral Sclerosis Risk. Biomedicines 2024, 12, 2733. [Google Scholar] [CrossRef]
  38. Adamu, A.; Li, S.; Gao, F.; Xue, G. The Role of Neuroinflammation in Neurodegenerative Diseases: Current Understanding and Future Therapeutic Targets. Front. Aging Neurosci. 2024, 16, 1347987. [Google Scholar] [CrossRef]
  39. Ha, J.; Choi, D.W.; Kim, K.J.; Kim, K.Y.; Nam, C.M.; Kim, E. Pioglitazone Use and Reduced Risk of Dementia in Patients with Diabetes Mellitus with a History of Ischemic Stroke. Neurology 2023, 100, e1799–e1811. [Google Scholar] [CrossRef]
  40. Chen, L.; Tao, Y.; Li, J.; Kang, M. Pioglitazone Use Is Associated with Reduced Risk of Parkinson’s Disease in Patients with Diabetes: A Systematic Review and Meta-Analysis. J. Clin. Neurosci. 2022, 106, 154–158. [Google Scholar] [CrossRef] [PubMed]
  41. Malhotra, B.; Hiteshi, P.; Khalkho, P.; Malik, R.; Bhadada, S.K.; Bhansali, A.; Shafiq, N.; Malhotra, S.; Kumar, N.; Rajput, R.; et al. Bladder Cancer with Pioglitazone: A Case-Control Study. Diabetes Metab. Syndr. 2022, 16, 102637. [Google Scholar] [CrossRef] [PubMed]
  42. Hashmi, A. Pioglitazone Suspension and Its Aftermath: A Wake up Call for the Indian Drug Regulatory Authorities. J. Pharmacol. Pharmacother. 2013, 4, 227. [Google Scholar] [CrossRef]
  43. Morones, J.; Pérez, M.; Muñoz, M.; Sánchez, E.; Ávila, M.; Topete, J.; Ventura, J.; Martínez, S. Evaluation of the Effect of an α-Adrenergic Blocker, a PPAR-γ Receptor Agonist, and a Glycemic Regulator on Chronic Kidney Disease in Diabetic Rats. Int. J. Mol. Sci. 2024, 25, 11372. [Google Scholar] [CrossRef] [PubMed]
  44. Yousefnia, S.; Momenzadeh, S.; Forootan, F.S.; Ghaedi, K.; Nasr-Esfahani, M.H. The Influence of Peroxisome Proliferator-Activated Receptor γ (PPARγ) Ligands on Cancer Cell Tumorigenicity. Gene 2018, 649, 14–22. [Google Scholar] [CrossRef]
  45. Wang, Y.; Lei, F.; Lin, Y.; Han, Y.; Yang, L.; Tan, H. Peroxisome Proliferator-Activated Receptors as Therapeutic Target for Cancer. J. Cell. Mol. Med. 2023, 28, 17931. [Google Scholar] [CrossRef] [PubMed]
  46. Chi, T.; Wang, M.; Wang, X.; Yang, K.; Xie, F.; Liao, Z.; Wei, P. PPAR-γ Modulators as Current and Potential Cancer Treatments. Front. Oncol. 2021, 11, 737776. [Google Scholar] [CrossRef]
  47. Jiao, X.X.; Lin, S.Y.; Lian, S.X.; Qiu, Y.R.; Li, Z.H.; Chen, Z.H.; Lu, W.Q.; Zhang, Y.; Deng, L.; Jiang, Y.; et al. Inhibition of the Breast Cancer by PPARγ Agonist Pioglitazone through JAK2/STAT3 Pathway. Neoplasma 2020, 67, 834–842. [Google Scholar] [CrossRef] [PubMed]
  48. Olatunde, A.; Nigam, M.; Singh, R.K.; Panwar, A.S.; Lasisi, A.; Alhumaydhi, F.A.; Jyoti kumar, V.; Mishra, A.P.; Sharifi-Rad, J. Cancer and Diabetes: The Interlinking Metabolic Pathways and Repurposing Actions of Antidiabetic Drugs. Cancer Cell Int. 2021, 21, 499. [Google Scholar] [CrossRef]
  49. Yun, S.H.; Han, S.H.; Park, J.I. Peroxisome Proliferator-Activated Receptor γ and PGC-1α in Cancer: Dual Actions as Tumor Promoter and Suppressor. PPAR Res. 2018, 2018, 6727421. [Google Scholar] [CrossRef]
  50. Chen, M.; Wang, H.; Cui, Q.; Shi, J.; Hou, Y. Dual Function of Activated PPARγ by Ligands on Tumor Growth and Immunotherapy. Med. Oncol. 2024, 41, 114. [Google Scholar] [CrossRef]
  51. Goleij, P.; Amin, M.; Rabie, A.; Mohamed, L.; Hamed, A.; Abdu, M.; Moahmmednoor, M.; Khan, H. Molecular Secrets Revealed: How Diabetes May Be Paving the Way for Leukemia. Curr. Treat. Options Oncol. 2024, 25, 1563–1579. [Google Scholar] [CrossRef]
  52. Tsubaki, M.; Takeda, T.; Tomonari, Y.; Kawashima, K.; Itoh, T.; Imano, M.; Satou, K.; Nishida, S. Pioglitazone Inhibits Cancer Cell Growth through STAT3 Inhibition and Enhanced AIF Expression via a PPARγ-Independent Pathway. J. Cell. Physiol. 2017, 233, 3638–3647. [Google Scholar] [CrossRef] [PubMed]
  53. Elrod, H.A.; Sun, S.-Y. PPARγ and Apoptosis in Cancer. PPAR Res. 2008, 2008, 704165. [Google Scholar] [CrossRef] [PubMed]
  54. Jang, J.H.; Lee, T.J.; Sung, E.G.; Song, I.H.; Kim, J.Y. Pioglitazone Mediates Apoptosis in Caki Cells via Downregulating C-FLIP(L) Expression and Reducing Bcl-2 Protein Stability. Oncol. Lett. 2021, 22, 743. [Google Scholar] [CrossRef] [PubMed]
  55. Rahman, H.S. Preclinical Drug Discovery in Colorectal Cancer: A Focus on Natural Compounds. Curr. Drug Targets 2021, 22, 977–997. [Google Scholar] [CrossRef] [PubMed]
  56. Ballav, S.; Biswas, B.; Sahu, V.K.; Ranjan, A.; Basu, S. PPAR-γ Partial Agonists in Disease-Fate Decision with Special Reference to Cancer. Cells 2022, 11, 3215. [Google Scholar] [CrossRef] [PubMed]
  57. Sun, J.; Yu, L.; Qu, X.; Huang, T. The Role of Peroxisome Proliferator-Activated Receptors in the Tumor Microenvironment, Tumor Cell Metabolism, and Anticancer Therapy. Front. Pharmacol. 2023, 14, 1184794. [Google Scholar] [CrossRef] [PubMed]
  58. Dicitore, A.; Caraglia, M.; Gaudenzi, G.; Manfredi, G.; Amato, B.; Mari, D.; Persani, L.; Arra, C.; Vitale, G. Type I Interferon-Mediated Pathway Interacts with Peroxisome Proliferator Activated Receptor-γ (PPAR-γ): At the Cross-Road of Pancreatic Cancer Cell Proliferation. Biochim. Et Biophys. Acta (BBA) Rev. Cancer 2014, 1845, 42–52. [Google Scholar] [CrossRef] [PubMed]
  59. Joharapurkar, A.; Patel, V.; Kshirsagar, S.; Patel, M.S.; Savsani, H.; Jain, M. Effect of Dual PPAR-α/γ Agonist Saroglitazar on Diabetic Retinopathy and Oxygen-Induced Retinopathy. Eur. J. Pharmacol. 2021, 899, 174032. [Google Scholar] [CrossRef] [PubMed]
  60. Hernandez-Quiles, M.; Broekema, M.F.; Kalkhoven, E. PPARgamma in Metabolism, Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front. Endocrinol. 2021, 12, 624112. [Google Scholar] [CrossRef]
  61. Esmaeili, S.; Safaroghli-Azar, A.; Pourbagheri-Sigaroodi, A.; Salari, S.; Gharehbaghian, A.; Hamidpour, M.; Bashash, D. Stimulation of Peroxisome Proliferator-Activated Receptor-Gamma (PPARγ) Using Pioglitazone Decreases the Survival of Acute Promyelocytic Leukemia Cells through Up-Regulation of PTEN Expression. Anti-Cancer Agents Med. Chem. 2020, 21, 108–119. [Google Scholar] [CrossRef] [PubMed]
  62. Jia, X.; Qian, J.; Chen, H.; Liu, Q.; Hussain, S.; Jin, J.; Shi, J.; Hou, Y. PPARγ Agonist Pioglitazone Enhances Colorectal Cancer Immunotherapy by Inducing PD-L1 Autophagic Degradation. Eur. J. Pharmacol. 2023, 950, 175749. [Google Scholar] [CrossRef]
  63. Ciaramella, V.; Sasso, F.C.; Liello, R.D.; Della, M.; Barra, G.; Viscardi, G.; Esposito, G.; Sparano, F.; Troiani, T.; Martinelli, E.; et al. Activity and Molecular Targets of Pioglitazone via Blockade of Proliferation, Invasiveness and Bioenergetics in Human NSCLC. J. Exp. Clin. Cancer Res. 2019, 38, 178. [Google Scholar] [CrossRef] [PubMed]
  64. Tseng, C.H. Pioglitazone and Breast Cancer Risk in Female Patients with Type 2 Diabetes Mellitus: A Retrospective Cohort Analysis. BMC Cancer 2022, 22, 559. [Google Scholar] [CrossRef]
  65. Alqahtani, Q.H.; Alkharashi, L.A.; Alajami, H.; Alkharashi, I.; Alkharashi, L.; Alhinti, S.N. Pioglitazone Enhances Cisplatin’s Impact on Triple-Negative Breast Cancer: Role of PPARγ in Cell Apoptosis. Saudi Pharm. J. 2024, 32, 102059. [Google Scholar] [CrossRef]
  66. Malakouti, P.; Mohammadi, M.; Boshagh, M.A.; Amini, A.; Rezaee, M.A.; Rahmani, M.R. Combined Effects of Pioglitazone and Doxorubicin on Migration and Invasion of MDA-MB-231 Breast Cancer Cells. J. Egypt. Natl. Cancer Inst. 2022, 34, 13. [Google Scholar] [CrossRef]
  67. Loo, S.Y.; Syn, N.L.; Koh, A.P.-F.; Teng, J.C.-F.; Deivasigamani, A.; Tan, T.Z.; Thike, A.A.; Vali, S.; Kapoor, S.; Wang, X.; et al. Epigenetic Derepression Converts PPARγ into a Druggable Target in Triple-Negative and Endocrine-Resistant Breast Cancers. Cell Death Discov. 2021, 7, 265. [Google Scholar] [CrossRef]
  68. Augimeri, G.; Bonofiglio, D. PPARgamma: A Potential Intrinsic and Extrinsic Molecular Target for Breast Cancer Therapy. Biomedicines 2021, 9, 543. [Google Scholar] [CrossRef] [PubMed]
  69. Zhao, B.; Xin, Z.; Ren, P.; Wu, H. The Role of PPARs in Breast Cancer. Cells 2022, 12, 130. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, J.; Tang, M.; Shang, J. PPARγ Modulators in Lung Cancer: Molecular Mechanisms, Clinical Prospects, and Challenges. Biomolecules 2024, 14, 190. [Google Scholar] [CrossRef]
  71. de Carvalho, M.V.; Gonçalves-de-Albuquerque, C.F.; Silva, A.R. PPAR Gamma: From Definition to Molecular Targets and Therapy of Lung Diseases. Int. J. Mol. Sci. 2021, 22, 805. [Google Scholar] [CrossRef] [PubMed]
  72. Keith, R.L.; Blatchford, P.J.; Merrick, D.T.; Bunn, P.A.; Bagwell, B.; Dwyer-Nield, L.D.; Jackson, M.K.; Geraci, M.W.; Miller, Y.E. A Randomized Phase II Trial of Pioglitazone for Lung Cancer Chemoprevention in High-Risk Current and Former Smokers. Cancer Prev. Res. 2019, 12, 721–730. [Google Scholar] [CrossRef]
  73. Seabloom, D.E.; Galbraith, A.R.; Haynes, A.M.; Antonides, J.D.; Wuertz, B.R.; Miller, W.A.; Miller, K.A.; Steele, V.E.; Miller, M.S.; Clapper, M.L.; et al. Fixed-Dose Combinations of Pioglitazone and Metformin for Lung Cancer Prevention. Cancer Prev. Res. 2017, 10, 116–123. [Google Scholar] [CrossRef] [PubMed]
  74. Seabloom, D.E.; Galbraith, A.R.; Haynes, A.M.; Antonides, J.D.; Wuertz, B.R.; Miller, W.A.; Miller, K.A.; Steele, V.E.; Suen, C.S.; O’Sullivan, M.G.; et al. Safety and Preclinical Efficacy of Aerosol Pioglitazone on Lung Adenoma Prevention in A/J Mice. Cancer Prev. Res. 2017, 10, 124–132. [Google Scholar] [CrossRef] [PubMed]
  75. Heudobler, D.; Schulz, C.; Fischer, J.R.; Staib, P.; Wehler, T.; Südhoff, T.; Schichtl, T.; Wilke, J.; Hahn, J.; Lüke, F.; et al. A Randomized Phase II Trial Comparing the Efficacy and Safety of Pioglitazone, Clarithromycin and Metronomic Low-Dose Chemotherapy with Single-Agent Nivolumab Therapy in Patients with Advanced Non-Small Cell Lung Cancer Treated in Second or Further Line (ModuLung). Front. Pharmacol. 2021, 12, 599598. [Google Scholar] [CrossRef]
  76. Kiran, A.V.V.V.R.; Kumari, G.K.; Krishnamurthy, P.T. Preliminary Evaluation of Anticancer Efficacy of Pioglitazone Combined with Celecoxib for the Treatment of Non-Small Cell Lung Cancer. Investig. New Drugs 2022, 40, 1–9. [Google Scholar] [CrossRef]
  77. Xu, R.; Luo, X.; Ye, X.; Li, H.; Liu, H.; Du, Q.; Zhai, Q. SIRT1/PGC-1α/PPAR-γ Correlate with Hypoxia-Induced Chemoresistance in Non-Small Cell Lung Cancer. Front. Oncol. 2021, 11, 682762. [Google Scholar] [CrossRef]
  78. Tseng, C.H. Pioglitazone Does Not Affect the Risk of Kidney Cancer in Patients with Type 2 Diabetes. Metabolism 2014, 63, 1049–1055. [Google Scholar] [CrossRef]
  79. Piątkowska-Chmiel, I.; Gawrońska-Grzywacz, M.; Natorska-Chomicka, D.; Herbet, M.; Sysa, M.; Iwan, M.; Korga, A.; Dudka, J. Pioglitazone as a Modulator of the Chemoresistance of Renal Cell Adenocarcinoma to Methotrexate. Oncol. Rep. 2020, 43, 1019–1030. [Google Scholar] [CrossRef]
  80. Alharbi, M.; Mahmoud, M.A.; Alshammari, A.; Almutairi, M.M.; Al-Ghamdi, J.M.; Alsabhan, J.F.; Shabanah, O.A.; Alshalawi, N.A.; Alzarea, S.I.; Alasmari, A.F. The Ameliorative Effect of Pioglitazone against Colistin-Induced Nephrotoxicity Is Mediated by Inhibition of NF-ΚB and Restoration of Nrf2 Signaling: An Integrative Bioinformatics Prediction-Guided in Vitro Study. PLoS ONE 2024, 19, e0314092. [Google Scholar] [CrossRef]
  81. Bell, D.S.; Jerkins, T. In Praise of Pioglitazone: An Economically Efficacious Therapy for Type 2 Diabetes and Other Manifestations of the Metabolic Syndrome. Diabetes Obes. Metab. 2023, 25, 3093–3102. [Google Scholar] [CrossRef]
  82. Li, S.; Ghoshal, S.; Sojoodi, M.; Arora, G.; Masia, R.; Erstad, D.J.; Lanuti, M.; Hoshida, Y.; Baumert, T.; Tanabe, K.K.; et al. Pioglitazone Reduces Hepatocellular Carcinoma Development in Two Rodent Models of Cirrhosis. J. Gastrointest. Surg. 2019, 23, 101–111. [Google Scholar] [CrossRef] [PubMed]
  83. Yang, Y.; Zhao, L.H.; Huang, B.; Wang, R.Y.; Yuan, S.X.; Tao, Q.F.; Xu, Y.; Sun, H.Y.; Lin, C.; Zhou, W.P. Pioglitazone, a PPARγ Agonist, Inhibits Growth and Invasion of Human Hepatocellular Carcinoma via Blockade of the Rage Signaling. Mol. Carcinog. 2014, 54, 1584–1595. [Google Scholar] [CrossRef] [PubMed]
  84. Huang, G.; Zhang, M.; Wang, M.; Xu, W.; Duan, X.; Han, X.; Ren, J. Pioglitazone, a Peroxisome Proliferator-Activated Receptor γ Agonist, Induces Cell Death and Inhibits the Proliferation of Hypoxic HepG2 Cells by Promoting Excessive Production of Reactive Oxygen Species. Oncol. Lett. 2024, 27, 1–11. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, G.H.; Li, S.-F.; Wei, R.; Jiang, Z. Diabetes and Colorectal Cancer Risk: Clinical and Therapeutic Implications. J. Diabetes Res. 2022, 2022, 1747326. [Google Scholar] [CrossRef]
  86. Huber, S.; Valente, S.; Chaimbault, P.; Schohn, H. Evaluation of Δ2-Pioglitazone, an Analogue of Pioglitazone, on Colon Cancer Cell Survival: Evidence of Drug Treatment Association with Autophagy and Activation of the Nrf2/Keap1 Pathway. International J. Oncol. 2014, 45, 426–438. [Google Scholar] [CrossRef] [PubMed]
  87. Pouya, F.D.; Salehi, R.; Rasmi, Y.; Kheradmand, F.; Fathi-Azarbayjani, A. Combination Chemotherapy against Colorectal Cancer Cells: Co-Delivery of Capecitabine and Pioglitazone Hydrochloride by Polycaprolactone-Polyethylene Glycol Carriers. Life Sci. 2023, 332, 122083. [Google Scholar] [CrossRef]
  88. Kushchayeva, Y.; Kushchayev, S.; Jensen, K.; Brown, R.J. Impaired Glucose Metabolism, Anti-Diabetes Medications, and Risk of Thyroid Cancer. Cancers 2022, 14, 555. [Google Scholar] [CrossRef]
  89. Sakaguchi, K.; Okiyama, Y.; Tanaka, S. In Silico Search for Drug Candidates Targeting the PAX8–PPARγ Fusion Protein in Thyroid Cancer. Int. J. Mol. Sci. 2024, 25, 5347. [Google Scholar] [CrossRef]
  90. Xu, B.; O’Donnell, M.; O’Donnell, J.; Yu, J.; Zhang, Y.; Sartor, M.A.; Koenig, R.J. Adipogenic Differentiation of Thyroid Cancer Cells through the Pax8-PPARγ Fusion Protein Is Regulated by Thyroid Transcription Factor 1 (TTF-1). J. Biol. Chem. 2016, 291, 19274–19286. [Google Scholar] [CrossRef]
  91. Greco, A.; Coperchini, F.; Croce, L.; Magri, F.; Teliti, M.; Rotondi, M. Drug Repositioning in Thyroid Cancer Treatment: The Intriguing Case of Anti-Diabetic Drugs. Front. Pharmacol. 2023, 14, 1303844. [Google Scholar] [CrossRef] [PubMed]
  92. Giordano, T.J.; Haugen, B.R.; Sherman, S.I.; Shah, M.H.; Caoili, E.M.; Koenig, R.J. Pioglitazone Therapy of PAX8-PPARγ Fusion Protein Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2018, 103, 1277–1281. [Google Scholar] [CrossRef]
  93. Ozdemir Kutbay, N.; Biray Avci, C.; Sarer Yurekli, B.; Caliskan Kurt, C.; Shademan, B.; Gunduz, C.; Erdogan, M. Effects of Metformin and Pioglitazone Combination on Apoptosis and AMPK/MTOR Signaling Pathway in Human Anaplastic Thyroid Cancer Cells. J. Biochem. Mol. Toxicol. 2020, 34, e22547. [Google Scholar] [CrossRef]
  94. Basilotta, R.; Lanza, M.; Casili, G.; Chisari, G.; Munao, S.; Colarossi, L.; Cucinotta, L.; Campolo, M.; Esposito, E.; Paterniti, I. Potential Therapeutic Effects of PPAR Ligands in Glioblastoma. Cells 2022, 11, 621. [Google Scholar] [CrossRef] [PubMed]
  95. Grommes, C.; Karlo, J.C.; Caprariello, A.; Blankenship, D.; DeChant, A.; Landreth, G.E. The PPARγ Agonist Pioglitazone Crosses the Blood–Brain Barrier and Reduces Tumor Growth in a Human Xenograft Model. Cancer Chemother. Pharmacol. 2013, 71, 929–936. [Google Scholar] [CrossRef]
  96. Lichtor, T.; Spagnolo, A.; Glick, R.P.; Feinstein, D.L. PPAR—Thiazolidinedione Agonists and Immunotherapy in the Treatment of Brain Tumors. PPAR Res. 2008, 2008, 547470. [Google Scholar] [CrossRef]
  97. Ching, J.; Amiridis, S.; Stylli, S.S.; Morokoff, A.P.; O’Brien, T.J.; Kaye, A.H. A Novel Treatment Strategy for Glioblastoma Multiforme and Glioma Associated Seizures: Increasing Glutamate Uptake with PPARγ Agonists. J. Clin. Neurosci. 2015, 22, 21–28. [Google Scholar] [CrossRef]
  98. Ranjan, G.; Ranjan, S.; Sunita, P.; Pattanayak, S.P. Thiazolidinedione Derivatives in Cancer Therapy: Exploring Novel Mechanisms, Therapeutic Potentials, and Future Horizons in Oncology. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 1–21. [Google Scholar] [CrossRef]
  99. de Vasconcelos, A.; Boeira, A.J.Z.; Drawanz, B.B.; Pedra, N.S.; Bona, N.P.; Stefanello, F.M.; Cunico, W. 2,4-Thiazolidinedione as Precursor to the Synthesis of Compounds with Anti-Glioma Activities in C6 and GL261 Cells. Med. Chem. 2021, 17, 601–610. [Google Scholar] [CrossRef] [PubMed]
  100. Biegański, M.; Szeliga, M. Disrupted Glutamate Homeostasis as a Target for Glioma Therapy. Pharmacol. Rep. 2024, 76, 1305–1317. [Google Scholar] [CrossRef]
  101. Cilibrasi, C.; Butta, V.; Riva, G.; Bentivegna, A. Pioglitazone Effect on Glioma Stem Cell Lines: Really a Promising Drug Therapy for Glioblastoma? PPAR Res. 2016, 2016, 7175067. [Google Scholar] [CrossRef] [PubMed]
  102. Tapia-Pérez, J.H.; Kirches, E.; Mawrin, C.; Firsching, R.; Schneider, T. Cytotoxic Effect of Different Statins and Thiazolidinediones on Malignant Glioma Cells. Cancer Chemother. Pharmacol. 2010, 67, 1193–1201. [Google Scholar] [CrossRef]
  103. Tapia-Pérez, J.H.; Preininger, R.; Kirches, E.; Reinhold, A.; Butzmann, J.; Prilloff, S.; Mawrin, C.; Schneider, T. Simultaneous Administration of Statins and Pioglitazone Limits Tumor Growth in a Rat Model of Malignant Glioma. Anticancer Res. 2016, 36, 6357–6365. [Google Scholar] [CrossRef] [PubMed]
  104. Cramer, C.K.; Alphonse-Sullivan, N.K.; Isom, S.; Metheny-Barlow, L.J.; Cummings, T.L.; Page, B.R.; Brown, D.; Blackstock, A.W.; Peiffer, A.M.; Strowd, R.E.; et al. Safety of Pioglitazone during and after Radiation Therapy in Patients with Brain Tumors: A Phase I Clinical Trial. J. Cancer Res. Clin. Oncol. 2019, 145, 337–344. [Google Scholar] [CrossRef] [PubMed]
  105. Esmaeili, S.; Salari, S.; Kaveh, V.; Ghaffari, S.H.; Bashash, D. Alteration of PPAR-GAMMA (PPARG.; PPARγ) and PTEN Gene Expression in Acute Myeloid Leukemia Patients and the Promising Anticancer Effects of PPARγ Stimulation Using Pioglitazone on AML Cells. Mol. Genet. Genom. Med. 2021, 9, 1818. [Google Scholar] [CrossRef]
  106. Saiki, M.; Hatta, Y.; Yamazaki, T.; Itoh, T.; Enomoto, Y.; Takeuchi, J.; Sawada, U.; Aizawa, S.; Horie, T. Pioglitazone Inhibits the Growth of Human Leukemia Cell Lines and Primary Leukemia Cells While Sparing Normal Hematopoietic Stem Cells. Int. J. Oncol. 2006, 29, 437–443. [Google Scholar] [CrossRef] [PubMed]
  107. Póvoa, V.M.O.; Delafiori, J.; Dias-Audibert, F.L.; de Oliveira, A.N.; Lopes, A.B.P.; de Paula, E.V.; Pagnano, K.B.B.; Catharino, R.R. Metabolic Shift of Chronic Myeloid Leukemia Patients under Imatinib–Pioglitazone Regimen and Discontinuation. Med. Oncol. 2021, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
  108. Hijazi, Μ.A.; Gessner, A.; El-Najjar, Ν. Repurposing of Chronically Used Drugs in Cancer Therapy: A Chance to Grasp. Cancers 2023, 15, 3199. [Google Scholar] [CrossRef]
  109. Ghasemi, H.; Jamshidi, A.; Ghatee, M.A.; Mazhab-Jafari, K.; Khorasani, M.; Rahmati, M.; Mohammadi, S. PPARγ Activation by Pioglitazone Enhances the Anti-Proliferative Effects of Doxorubicin on Pro-Monocytic THP-1 Leukemia Cells via Inducing Apoptosis and G2/M Cell Cycle Arrest. J. Recept. Signal Transduct. Res. 2021, 42, 429–438. [Google Scholar] [CrossRef]
  110. Ryu, S.; Kim, D.S.; Lee, M.W.; Lee, J.W.; Sung, K.W.; Koo, H.H.; Yoo, K.H. Anti-Leukemic Effects of PPARγ Ligands. Cancer Lett. 2018, 418, 10–19. [Google Scholar] [CrossRef] [PubMed]
  111. Okabe, S.; Tauchi, T.; Tanaka, Y.; Ohyashiki, K. Peroxisome Proliferator-Activated Receptors: Targets for the Treatment of Philadelphia Chromosome-Positive Leukemia Cells. Blood 2017, 130, 5241. [Google Scholar]
  112. Esmaeili, S.; Yousefi, A.-M.; Delshad, M.; Bashash, D. Synergistic Effects of PI3K Inhibition and Pioglitazone against Acute Promyelocytic Leukemia Cells. Mol. Genet. Genom. Med. 2022, 11, 2106. [Google Scholar] [CrossRef]
  113. Mokhtari, Y.; Yousefi, A.-M.; Bashash, D. Inhibition of PI3K Signaling Intensified the Antileukemic Effects of Pioglitazone: New Insight into the Application of PPARγ Stimulators in Acute Lymphoblastic Leukemia. Indian J. Hematol. Blood Transfus. 2023, 39, 546–556. [Google Scholar] [CrossRef] [PubMed]
  114. Ghadiany, M.; Tabarraee, M.; Salari, S.; Haghighi, S.; Rezvani, H.; Ghasemi, S.N.; Karimi-Sari, H. Adding Oral Pioglitazone to Standard Induction Chemotherapy of Acute Myeloid Leukemia: A Randomized Clinical Trial. Clin. Lymphoma Myeloma Leuk. 2019, 19, 206–212. [Google Scholar] [CrossRef]
  115. Esmaeili, S.; Safaroghli-azar, A.; Pourbagheri-Sigaroodi, A.; Salari, S.; Gharehbaghian, A.; Hamidpour, M.; Bashash, D. Activation of PPARγ Intensified the Effects of Arsenic Trioxide in Acute Promyelocytic Leukemia through the Suppression of PI3K/Akt Pathway: Proposing a Novel Anticancer Effect for Pioglitazone. Int. J. Biochem. Cell Biol. 2020, 122, 105739. [Google Scholar] [CrossRef] [PubMed]
  116. Glodkowska-Mrowka, E.; Manda-Handzlik, A.; Stelmaszczyk-Emmel, A.; Seferynska, I.; Stoklosa, T.; Przybylski, J.; Mrowka, P. PPARγ Ligands Increase Antileukemic Activity of Second- and Third-Generation Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia Cells. Blood Cancer J. 2016, 6, 377. [Google Scholar] [CrossRef]
  117. Jazi, M.S.; Mohammadi, S.; Yazdani, Y.; Sedighi, S.; Memarian, A.; Aghaei, M. Effects of Valproic Acid and Pioglitazone on Cell Cycle Progression and Proliferation of T-Cell Acute Lymphoblastic Leukemia Jurkat Cells. Iran. J. Basic Med. Sci. 2016, 19, 779. [Google Scholar]
  118. Rousselot, P.; Prost, S.; Guilhot, J.; Roy, L.; Etienne, G.; Legros, L.; Charbonnier, A.; Coiteux, V.; Cony-Makhoul, P.; Huguet, F.; et al. Pioglitazone Together with Imatinib in Chronic Myeloid Leukemia: A Proof of Concept Study. Cancer 2016, 123, 1791–1799. [Google Scholar] [CrossRef] [PubMed]
  119. Beatriz, A.; Miranda, E.C.; Oliveira, M.; Vergílio, B.R.; Pavan, C.; Delamain, M.T.; Duarte, G.B.; Souza, C.A.; De, E.V.; Pagnano, K.B. Pioglitazone Did Not Affect PPAR-Γ, STAT5, HIF2α and CITED2 Gene Expression in Chronic Myeloid Leukemia Patients with Deep Molecular Response. Blood 2019, 134, 1637. [Google Scholar] [CrossRef]
  120. Zhao, Z.; He, X.; Sun, Y. Hypoglycemic Agents and Incidence of Pancreatic Cancer in Diabetic Patients: A Meta-Analysis. Front. Pharmacol. 2023, 14, 1193610. [Google Scholar] [CrossRef]
  121. Chen, Y. Diabetes Medication Use and Risk of Breast, Colorectal, and Pancreatic Cancer: A Population-Based Cohort Study. Ph.D. Thesis, The University of British Columbia, Vancouver, BC, Canada, 2025. [Google Scholar] [CrossRef]
  122. Chen, Y.; Mushashi, F.; Son, S.; Bhatti, P.; Dummer, T.; Murphy, R.A. Diabetes Medications and Cancer Risk Associations: A Systematic Review and Meta-Analysis of Evidence over the Past 10 Years. Sci. Rep. 2023, 13, 11844. [Google Scholar] [CrossRef]
  123. See, L.-C.; Wu, C.-Y.; Tsai, C.-Y.; Lee, C.-C.; Chen, J.-J.; Jenq, C.-C.; Chen, C.-Y.; Chen, Y.-C.; Yen, C.-L.; Yang, H.-Y. PPAR-γ Agonist Pioglitazone and the Risks of Malignancy among Type2 Diabetes Mellitus Patients. Acta Diabetol. 2024. Online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  124. Ninomiya, I.; Yamazaki, K.; Oyama, K.; Hayashi, H.; Tajima, H.; Kitagawa, H.; Fushida, S.; Fujimura, T.; Ohta, T. Pioglitazone Inhibits the Proliferation and Metastasis of Human Pancreatic Cancer Cells. Oncol. Lett. 2014, 8, 2709–2714. [Google Scholar] [CrossRef]
  125. Koga, H. PPARγ Potentiates Anticancer Effects of Gemcitabine on Human Pancreatic Cancer Cells. Int. J. Oncol. 2011, 40, 679–685. [Google Scholar] [CrossRef]
  126. El-Sisi, A.E.; Sokar, S.S.; Salem, T.A.; Abu, S.E. PPARγ-Dependent Anti-Tumor and Immunomodulatory Actions of Pioglitazone. J. Immunotoxicol. 2014, 12, 308–316. [Google Scholar] [CrossRef] [PubMed]
  127. Bahrambeigi, S.; Molaparast, M.; Sohrabi, F.; Seifi, L.; Faraji, A.; Fani, S.; Shafiei-Irannejad, V. Targeting PPAR Ligands as Possible Approaches for Metabolic Reprogramming of T Cells in Cancer Immunotherapy. Immunol. Lett. 2020, 220, 32–37. [Google Scholar] [CrossRef]
  128. Xiong, Z.; Chan, S.L.; Zhou, J.; Joaquim; Kwong, T.T.; Zeng, X.; Wu, H.; Cao, J.; Tu, Y.; Feng, Y.; et al. Targeting PPAR-Gamma Counteracts Tumour Adaptation to Immune-Checkpoint Blockade in Hepatocellular Carcinoma. Cancers 2023, 15, 3199. [Google Scholar] [CrossRef] [PubMed]
  129. A Study of Pioglitazone and Carboplatin in Patients with Advanced Solid Tumors—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02133625?cond=cancer%20&intr=pioglitazone&rank=3 (accessed on 25 January 2025).
  130. Pioglitazone Hydrochloride in Preventing Head and Neck Cancer in Patients with Oral Leukoplakia—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT00099021?cond=cancer%20&intr=pioglitazone&rank=4 (accessed on 25 January 2025).
  131. A Phase II Study of Pioglitazone for Patients with Cancer of the Pancreas—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT01838317?cond=cancer%20&intr=pioglitazone&rank=9 (accessed on 25 January 2025).
  132. Preventing Squamous Cell Skin Cancer—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02347813?cond=cancer%20&intr=pioglitazone&rank=10 (accessed on 25 January 2025).
  133. Pioglitazone to Treat Adults Undergoing Surgery for Non-Small Cell Lung Cancer—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT00923949?cond=cancer%20&intr=pioglitazone&page=2&rank=11 (accessed on 25 January 2025).
  134. Study of Imatinib Discontinuation in Chronic Myeloid Leukemia with Deep Molecular Response (EDI-PIO)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02852486?cond=cancer%20&intr=pioglitazone&page=2&rank=13 (accessed on 25 January 2025).
  135. Pioglitazone Hydrochloride in Treating Patients with Stage IA-IIIA Non-small Cell Lung Cancer—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT01342770?cond=cancer%20&intr=pioglitazone&page=2&rank=15 (accessed on 25 January 2025).
  136. Study on Trabectedin in Combination with Pioglitazone in Patients Myxoid Liposarcomas with Stable Disease After T Alone. (TRABEPIO)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT04794127?cond=cancer%20&intr=pioglitazone&page=2&rank=1 (accessed on 25 January 2025).
  137. Pioglitazone-Metformin Combination Treatment for High Risk Oral Preneoplasia—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT05727761?cond=cancer%20&intr=pioglitazone&page=2&rank=20 (accessed on 25 January 2025).
  138. Pioglitazone for Oral Premalignant Lesions—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT00951379?cond=cancer%20&intr=pioglitazone&page=3&rank=21 (accessed on 25 January 2025).
  139. A Trial With Metronomic Low-Dose Treosulfan, Pioglitazone and Clarithromycin Versus Standard Treatment in NSCLC (ModuLung)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02852083?cond=cancer%20&intr=pioglitazone&page=3&rank=22 (accessed on 25 January 2025).
  140. Value of Using Pioglitazone in Chronic Myeloid Leukemia Treatment—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT04883125?cond=cancer%20&intr=pioglitazone&page=3&rank=27 (accessed on 25 January 2025).
  141. Pioglitazone for Lung Cancer Chemoprevention—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT00780234?cond=cancer%20&intr=pioglitazone&page=3&rank=28 (accessed on 25 January 2025).
  142. Safety and Efficiency Study of Pioglitazone in Combination with Imatinib Mesylate to Treat Chronic Myelogenous Leukemia (SESPI)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02687425?cond=cancer%20&intr=pioglitazone&page=3&rank=30 (accessed on 25 January 2025).
  143. Therapies in Combination or Sequentially with Tyrosine Kinase Inhibitors (TKIs) in Chronic Phase Chronic Myelogenous Leukemia Patients in CCR (ACTIW) (ACTIW)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02767063?cond=cancer%20&intr=pioglitazone&page=4&rank=31 (accessed on 25 January 2025).
  144. Second STOP After Pioglitazone Priming in CML Patients (PIO2STOP)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02889003?cond=cancer%20&intr=pioglitazone&page=4&rank=32 (accessed on 25 January 2025).
  145. Combined Antiinflammatory and Angiostatic Therapy in Patients with Hormone-refractory Prostate Cancer (INV342)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT00427999?cond=cancer%20&intr=pioglitazone&page=4&rank=33 (accessed on 25 January 2025).
  146. Pioglitazone as Second-Line in Patients with Metastatic Pancreatic Cancer After Treatment With Gemcitabine—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT00867126?cond=cancer%20&intr=pioglitazone&page=4&rank=37 (accessed on 25 January 2025).
  147. Pioglitazone and Tyrosine Kinase Inhibitor in Treating Patients with Relapsed Chronic Myeloid Leukemia—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02730195?cond=cancer%20&intr=pioglitazone&page=4&rank=38 (accessed on 25 January 2025).
  148. Phase I and Consecutive Phase II, Two-arm, Randomized Multi-center Trial in Patients with Advanced Melanoma—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT01614301?cond=cancer%20&intr=pioglitazone&page=5&rank=43 (accessed on 25 January 2025).
  149. ACTOplus Met XR in Treating Patients with Stage I-IV Oral Cavity or Oropharynx Cancer Undergoing Definitive Treatment—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02917629?cond=cancer%20&intr=pioglitazone&page=5&rank=44 (accessed on 25 January 2025).
  150. Third-line Therapy of Multiple Myeloma a Prospective Phase I/II Trial (MM03). Available online: https://clinicaltrials.gov/study/NCT01010243?cond=cancer%20&intr=pioglitazone&page=5&rank=46 (accessed on 25 January 2025).
  151. Modulatory Effect of Prodigiosin or Pioglitazone on TIME and the Crosstalk to Immune-Checkpoint Protein(s)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT06502249?cond=cancer%20&intr=pioglitazone&page=5&rank=48 (accessed on 25 January 2025).
  152. Pioglitazone and Imatinib for CML Patients (ACTIM)—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02888964?cond=cancer%20&intr=pioglitazone&page=5&rank=50 (accessed on 25 January 2025).
  153. A Study of Perpetrator Drug Interactions of Enasidenib in AML Patients—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT03720366?cond=cancer%20&intr=pioglitazone&page=6&rank=53&tab=table (accessed on 25 January 2025).
  154. Pioglitazone in Thyroid Cancers—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT01655719?cond=cancer%20&intr=pioglitazone&rank=1&tab=results (accessed on 25 January 2025).
  155. Incretin-based Drugs and Pancreatic Cancer—Full Text View. 2025. Available online: https://clinicaltrials.gov/study/NCT02475499?cond=cancer%20&intr=pioglitazone&page=6&rank=57 (accessed on 25 January 2025).
  156. Moghareabed, R.; Hemati, S.; Akhavan, A.; Emami, H.; Farghadani, M.; Roayaei, M.; Tavajoh, S.; Feizi, A. Randomized Phase II Clinical Trial of Pioglitazone plus Chemotherapy versus Chemotherapy Alone in Patients with Metastatic Breast Cancer. J. Glob. Oncol. 2019, 5, 83. [Google Scholar] [CrossRef]
  157. Agulló-Ortuño, M.T.; Pérez, C.; Díaz-García, C.V.; Homet, B.; Agudo-López, A.; Rodríguez Garzotto, A.; Prieto-García, E.; Adeva, J.; Riesco, M.C.; Rodríguez, R.; et al. Abstract 2575: Pharmacodynamic Evaluation of the Combination of Carboplatin and Paclitaxel Associated with Either Pioglitazone or Hydroxyurea, within a Randomized Phase 1 Dose Escalation Clinical Trial in Patients with Advanced Solid Tumors. Cancer Res. 2015, 75, 2575. [Google Scholar] [CrossRef]
  158. Han, Y.H.; Kwon, S.Y.; Kim, J.; Na, C.J.; Choi, S.; Min, J.J.; Bom, H.S.; Kim, Y.C.; Oh, I.J.; Chae, H.J.; et al. A Phase II Clinical Trial to Investigate the Effect of Pioglitazone on 18F-FDG Uptake in Malignant Lesions. Eur. J. Nucl. Med. Mol. Med. 2015, 5, 50. [Google Scholar] [CrossRef] [PubMed]
  159. Mamtani, R.; Haynes, K.; Bilker, W.B.; Vaughn, D.J.; Strom, B.L.; Glanz, K.; Lewis, J.D. Association between Longer Therapy with Thiazolidinediones and Risk of Bladder Cancer: A Cohort Study. JNCI J. Natl. Cancer Inst. 2012, 104, 1411–1421. [Google Scholar] [CrossRef]
  160. Neumann, A.; Weill, A.; Ricordeau, P.; Fagot, J.P.; Alla, F.; Allemand, H. Pioglitazone and Risk of Bladder Cancer among Diabetic Patients in France: A Population-Based Cohort Study. Diabetologia 2012, 55, 1953–1962. [Google Scholar] [CrossRef] [PubMed]
  161. Tang, H.; Shi, W.; Fu, S.; Wang, T.; Zhai, S.; Song, Y.; Han, J. Pioglitazone and Bladder Cancer Risk: A Systematic Review and Meta-Analysis. Cancer Med. 2018, 7, 1070–1080. [Google Scholar] [CrossRef]
  162. Xu, X.; Wu, J.; Mao, Y.; Zhu, Y.; Hu, Z.; Xu, X.; Lin, Y.; Chen, H.; Zheng, X.; Qin, J.; et al. Diabetes Mellitus and Risk of Bladder Cancer: A Meta-Analysis of Cohort Studies. PLoS ONE 2013, 8, e58079. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of pioglitazone (created using ACD/ChemSketch, version 2021.2.0 (Advanced Chemistry Development, Inc., (ACD/Labs), Toronto, ON, Canada; Available online: www.acdlabs.com (accessed on 25 January 2025)).
Figure 1. Chemical structure of pioglitazone (created using ACD/ChemSketch, version 2021.2.0 (Advanced Chemistry Development, Inc., (ACD/Labs), Toronto, ON, Canada; Available online: www.acdlabs.com (accessed on 25 January 2025)).
Applsci 15 01925 g001
Figure 2. Pharmacological effect of pioglitazone on the cardiovascular and nervous system, as well as hepatic and metabolic function. Effect of pioglitazone on different systems. Pioglitazone enhances insulin sensitivity, which improves endothelial, neuronal, pancreatic and liver cell function (created using https://biorender.com, URL accessed on 25 January 2025).
Figure 2. Pharmacological effect of pioglitazone on the cardiovascular and nervous system, as well as hepatic and metabolic function. Effect of pioglitazone on different systems. Pioglitazone enhances insulin sensitivity, which improves endothelial, neuronal, pancreatic and liver cell function (created using https://biorender.com, URL accessed on 25 January 2025).
Applsci 15 01925 g002
Figure 3. Pharmacological effect of pioglitazone facilitated through the inhibition of thromboxane A2 (TX-A2) and growth factor receptor pathways. The former regulates the expression of phospholipase A2 (PLA2), arachidonic acid (AA), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2) and prostacyclin I2 (PGI2). The latter regulates the expression of vascular endothelial growth factor (VEGF) through the phosphoinositide 3-Kinase pathway (PI3K) (created using https://biorender.com, URL accessed on 25 January 2025).
Figure 3. Pharmacological effect of pioglitazone facilitated through the inhibition of thromboxane A2 (TX-A2) and growth factor receptor pathways. The former regulates the expression of phospholipase A2 (PLA2), arachidonic acid (AA), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2) and prostacyclin I2 (PGI2). The latter regulates the expression of vascular endothelial growth factor (VEGF) through the phosphoinositide 3-Kinase pathway (PI3K) (created using https://biorender.com, URL accessed on 25 January 2025).
Applsci 15 01925 g003
Figure 4. Pioglitazone combination therapies for the treatment of breast cancer, lung cancer, renal cancer, hepatocellular carcinoma, colorectal cancer, thyroid cancer, glioma, pancreatic cancer, and hematological malignancies (created using https://biorender.com, URL accessed on 25 January 2025).
Figure 4. Pioglitazone combination therapies for the treatment of breast cancer, lung cancer, renal cancer, hepatocellular carcinoma, colorectal cancer, thyroid cancer, glioma, pancreatic cancer, and hematological malignancies (created using https://biorender.com, URL accessed on 25 January 2025).
Applsci 15 01925 g004
Table 1. Molecular mechanisms of pioglitazone.
Table 1. Molecular mechanisms of pioglitazone.
ProcessMechanismCancer Type
Reduced cell proliferationIncreased expression of excitatory amino acid transporter 2 [44].Neuroblastoma
Increased activity of p-Akt and p-GSK-3β [44].
Redifferentiation of tumor associated adipocytes [44].
Inhibited cell growth via mTOR and STAT5 pathway tampering by retinoid X receptor agonists [45].Glioma
Inhibited expression of estrogen receptor and aromatase via PGE2 and BRCA1 pathways [46].Breast cancer
Inhibition of JAK2/STAT3 pathway [47].
Increased expression of p21 and MAPK activity [48].
Inhibited CSC proliferation due to decreased STAT5 and HIF-2α levels [49].Chronic myeloid leukemia
Downregulated MAPK, RAS, MYC gene expression and phosphorylation of MAPK pathway proteins [50].Non-small cell lung cancer
Increased apoptosisDownregulation of BCL2 and SCD1 [44].Leukemia
Reduced expression phosphorylation of MEK1 and ERK phosphorylation [51].
Downregulation of STAT3 with ERK1/2, NF-κβ and p38MAPK molecules unaffected [49,52].
Reduced expression of Survivin [49,52].
Increased expression of TRAIL death ligand and apoptosis inducing factor [49,53].
Downregulation of BCLXL/BCL2 in a PPAR-γ and caspase-independent manner [51].Prostate cancer, squamous cell cancer (SCC)
Induction of apoptosis in a caspase-dependent manner assisted by downregulation of c-FLIP, leading to BCL2 downregulation and instability [54].Caki cells
Downregulation of X-linked inhibitor of apoptosis (XIAP) and cyclooxygenase-2 (COX-2) [55].Colorectal cancer
Upregulation of cyclinB1, CDC2, p21 and alteration of BAX/BCL2 ratio [55].
Reduced angiogenesis
[44,45,46,56,57,58]
Reduced expression of matrix metalloproteinase 2 (MMP2), vascular endothelial growth factor (VEGF), COX-2.Hepatocellular carcinoma
Inhibition of fibroblast growth factor 2 (FGF-2) and urokinase plasminogen activator.
Apoptosis of endothelial cells.
Combined downregulation of COX-2 and VEGF when coupled with clofibric acid [45].Ovarian cancer
Reduction of bFGF- and VEGF-initiated angiogenesis [59].Chick chorioallantoic membrane model
Drug SensitizationReduced expression of metallothionein and endorphin connected to S273 phosphorylation [60].Pancreatic cancer
Enhanced type I Interferon activity due to inhibition of the STAT-3 pathway [58].
Increased arsenic trioxide induced tumor toxicity through inhibition of the PI3K/AKT pathway [45].Leukemia
Doxorubicin sensitization via modulation of P-glycoprotein [56].Osteosarcoma
Reduced resistance to cisplatin [46].
Cell Cycle
Modification
Increased cisplatin and oxiplatin efficacy.Thyroid, lung, prostate, breast, kidney, esophageal and urothelial cancer
Inhibited EGFR/MDM2 mediated chemoresistance and PPAR-γ degradation [50].
Downregulation of cyclin dependent kinase 4 (CDK4).
Upregulation of CDK inhibitors including p19, p21, p27 and rho-related GTP binding protein.
Activation of Rb protein [51,60].
Downregulation of cyclins D, cyclin E, CDK2, CDK4, proliferating nuclear antigen and retinoblastoma protein [58].Breast and colorectal cancer
Induction of G1-arrest through the activation of p21 along with the upregulation of FOXO3a [61].Acute promyelotic leukemia (NB4 cells)
Accelerated differentiationInduced adipogenesis [57].Melanoma
ImmunomodulationIncreased β3 and α5 integrin expression [57].Colorectal cancer
Reduced PD-L1 levels due to autophagy [62].Lung, colorectal cancer
Bioenergetic disruptionReduced pyruvate oxidation and glutathione levels.Hepatocellular carcinoma
ROS-induced stress mediated by HIF-1 and NF-κβ signaling.
Reduced metastasis and invasivenessDownregulation of smad family member 3 (SMAD3), PDK1 and MCT-1.Breast cancer
Upregulation of plasminogen activator inhibitor-1 (PAI-1).
Downregulation of NF-κβ, eIF2α, MMP9 and fibronectin [46].Lung cancer
Upregulation of CXCR4, CXCR7, E-cadherin [47].
Downregulation of TGF-β.Glioma
Reduced expression and invasiveness of β-cantenin [46].Breast cancer
Downregulation of TGFβR1 and SMAD3 associated with epithelial to mesenchymal transition (EMT) [63].Non-small cell lung cancer
Increased autophagyUpregulation of HIF-1 and BNIP3 [51].Breast and prostate cancer
Activation of PI3K [61].Acute promyelotic leukemia (NB4 cells)
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

Vasileiou, M.; Diamantoudis, S.C.; Tsianava, C.; Nguyen, N.P. Therapeutic Potential and Challenges of Pioglitazone in Cancer Treatment. Appl. Sci. 2025, 15, 1925. https://doi.org/10.3390/app15041925

AMA Style

Vasileiou M, Diamantoudis SC, Tsianava C, Nguyen NP. Therapeutic Potential and Challenges of Pioglitazone in Cancer Treatment. Applied Sciences. 2025; 15(4):1925. https://doi.org/10.3390/app15041925

Chicago/Turabian Style

Vasileiou, Maria, Sotirios Charalampos Diamantoudis, Christina Tsianava, and Nam P. Nguyen. 2025. "Therapeutic Potential and Challenges of Pioglitazone in Cancer Treatment" Applied Sciences 15, no. 4: 1925. https://doi.org/10.3390/app15041925

APA Style

Vasileiou, M., Diamantoudis, S. C., Tsianava, C., & Nguyen, N. P. (2025). Therapeutic Potential and Challenges of Pioglitazone in Cancer Treatment. Applied Sciences, 15(4), 1925. https://doi.org/10.3390/app15041925

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