Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters











Publication year range
1.
Expert Rev Anticancer Ther ; 24(7): 493-512, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38690706

ABSTRACT

INTRODUCTION: As a vital mechanism of survival, lymphopoiesis requires the collaboration of different signaling molecules to orchestrate each step of cell development and maturation. The PI3K pathway is considerably involved in the maturation of lymphatic cells and therefore, its dysregulation can immensely affect human well-being and cause some of the most prevalent malignancies. As a result, studies that investigate this pathway could pave the way for a better understanding of the lymphopoiesis mechanisms, the undesired changes that lead to cancer progression, and how to design drugs to solve this issue. AREAS COVERED: The present review addresses the aforementioned aspects of the PI3K pathway and helps pave the way for future therapeutic approaches. In order to access the articles, databases such as Medicine Medline/PubMed, Scopus, Google Scholar, and Science Direct were utilized. The search formula was established by identifying main keywords including PI3K/Akt/mTOR pathway, Lymphopoiesis, Lymphoid malignancies, and inhibitors. EXPERT OPINION: The PI3K pathway is crucial for lymphocyte development and differentiation, making it a potential target for therapeutic intervention in lymphoid cancers. Studies are focused on developing PI3K inhibitors to impede the progression of hematologic malignancies, highlighting the pathway's significance in lymphoma and lymphoid leukemia.


Subject(s)
Drug Development , Lymphoma , Lymphopoiesis , Phosphatidylinositol 3-Kinases , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Humans , Phosphatidylinositol 3-Kinases/metabolism , Animals , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Lymphoma/pathology , Lymphoma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Disease Progression , Molecular Targeted Therapy , Drug Design , Cell Differentiation
2.
Acta Cardiol ; : 1-6, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38032242

ABSTRACT

BACKGROUND: The clinical safety and consequences of upgrade procedures compared with de novo cardiac resynchronisation therapy (CRT) implantation in heart failure remain unclear. The present study aimed to assess clinical and procedural consequences of patients undergoing CRT upgrade as compared to de novo CRT implantations. METHODS: In this prospective cohort study, two subgroups were considered as the study population as (1) de novo group that CRT was considered on optimised medical treatment with heart failure of NYHA functional class from II to IV, left ventricular ejection fraction (LVEF) of ≤35%, and QRS width of >130 ms and (2) upgrade group including the patients with previously implantable cardioverter defibrillator (ICD) with the indications for upgrading to CRT. The two groups were compared regarding the changes in clinical outcome and echocardiography parameters. RESULTS: The procedure was successful in 95.9% of patients who underwent CRT upgrade and 100% of those who underwent de novo CRT implantation. It showed a significant improvement in LVEF, severity of mitral regurgitation and NYHA functional classification, without any difference between the two study groups. Overall procedural related complications were reported in 10.8% and 3.8% (p = .093) and cardiac death in 5.4% and 2.5% (p = .360), respectively, with no overall difference in postoperative outcome between the two groups. CONCLUSIONS: Upgrading to CRT is a safe and effective procedure regarding improvement of functional class, left ventricular function status and post-procedural outcome.

3.
Nutr Metab Cardiovasc Dis ; 33(11): 2055-2066, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37567791

ABSTRACT

AIMS: Studies have indicated inconsistent results regarding the association between plasma levels of Lipoprotein(a) [Lp(a)] and coronary artery calcification (CAC). We performed a systematic review and meta-analysis to investigate the association between elevated levels of Lp(a) and risk of CAC in populations free of cardiovascular disease (CVD) symptoms. DATA SYNTHESIS: PubMed, Web of Science, Embase, and Scopus were searched up to July 2022 and the methodological quality was assessed using Newcastle-Ottawa Scale (NOS) scale. Random-effects meta-analysis was used to estimate pooled odds ratio (OR) and 95% confidence interval. Out of 298 studies, data from 8 cross-sectional (n = 18,668) and 4 cohort (n = 15,355) studies were used in meta-analysis. Cohort studies demonstrated a positive significant association between Lp(a) and CAC, so that individuals with Lp(a)≥30-50 exposed to about 60% risk of CAC incidence compared to those with lower Lp(a) concentrations in asymptomatic CVD subjects (OR, 1.58; 95% CI, 1.38-1.80; l2, 0.0%; P, 0.483); Subgroup analysis showed that a cut-off level for Lp(a) measurement could not statistically affect the association, but race significantly affected the relationship between Lp(a) and CAC (OR,1.60; 95% CI, 1.41-1.81). Analyses also revealed that both men and women with higher Lp(a) concentrations are at the same risk for increased CAC. CONCLUSIONS: Blood Lp(a) level was significantly associated with CAC incidence in asymptomatic populations with CVD, indicating that measuring Lp(a) may be a useful biomarker for diagnosing subclinical atherosclerosis in individuals at higher risk of CAC score. PROSPERO REGISTRATION NUMBER: CRD42022350297.

4.
Geriatr Gerontol Int ; 22(8): 616-622, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35734811

ABSTRACT

AIM: This study aimed to assess the effects of telenursing on patients' activities of daily living and instrumental activities of daily living (ADLs and IADLs) following a myocardial infarction (MI). METHODS: This randomized, parallel-group, controlled trial was conducted on 95 patients post-MI from 2018 to 2019. Patients were randomly assigned to the intervention (telenursing) and control groups using permuted block randomization. Through telephone calls, telenursing was performed twice a week during the first six consecutive weeks, then once a week until week 12. ADL and IADL questionnaires were completed by both groups before intervention and 12 weeks later. The CONSORT 2010 checklist was used to report the study protocol. RESULTS: The mean age of patients was 56.8 ± 11.07 and 54.2 ± 9.8 years in the telenursing and control group, respectively. The mean ADL and IADL scores in the telenursing group were substantially greater than in the control group [4.57 (3.18, 5.97); P < 0.001 and 4.40 (3.06, 5.75); P < 0.001, respectively]. The odds of a higher degree of independence (no disabilities vs. mild disabilities and disability as well as no disabilities and mild disabilities vs. disability) regarding ADLs and IADLs were significantly greater in the telenursing group as compared with the control group (P < 0.001 and P < 0.001, respectively). CONCLUSIONS: Our findings suggest that the use of telenursing intervention may increase patients' ADLs and IADLs after an MI and may enhance their independence. Geriatr Gerontol Int 2022; 22: 616-622.


Subject(s)
Disabled Persons , Myocardial Infarction , Telenursing , Activities of Daily Living , Aged , Humans , Surveys and Questionnaires
5.
J Cancer Res Clin Oncol ; 148(1): 57-70, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34981193

ABSTRACT

PURPOSE: Therapy resistance is the principal obstacle to achieving cures in cancer patients and its successful tackling requires a deep understanding of the resistance mediators. Increasing evidence indicates that tumor phosphatases are novel and druggable targets in translational oncology and their modulation may hinder tumor growth and motility and potentiate therapeutic sensitivity in various neoplasms via regulation of various signal transduction pathways. Dual-specificity phosphatases (DUSPs) are key players of cell growth, survival and death and have essential roles in tumor initiation, malignant progression and therapy resistance through regulation of the MAPK signaling pathway. In this review, different aspects of DUSPs are discussed. METHODS: A comprehensive literature review was performed using various websites including PubMed. RESULTS: We provide mechanistic insights into the roles of well-known DUSPs in resistance to a wide range of cancer therapeutic approaches including chemotherapy, radiation and molecular targeted therapy in human malignancies. Moreover, we discuss the development of DUSP modulators, with a focus on DUSP1 and 6 inhibitors. Ultimately, the preclinical investigations of small molecule inhibitors of DUSP1 and 6 are outlined. CONCLUSION: Emerging evidence indicates that the DUSP family is aberrantly expressed in human malignancies and plays critical roles in determining sensitivity to a wide range of cancer therapeutic strategies through regulation of the MAPK signaling pathways. Consequently, targeting DUSPs and their downstream molecules can pave the way for more effective cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 6/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Benzofurans/pharmacology , Carcinogenesis/pathology , Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 1/biosynthesis , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 6/biosynthesis , Dual Specificity Phosphatase 6/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Targeted Therapy/methods , Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Cell Physiol ; 237(1): 346-372, 2022 01.
Article in English | MEDLINE | ID: mdl-34498289

ABSTRACT

Despite remarkable advances in different types of cancer therapies, an effective therapeutic strategy is still a major and significant challenge. One of the most promising approaches in this regard is immunotherapy, which takes advantage of the patients' immune system; however, the many mechanisms that cancerous cells harbor to extend their survival make it impossible to gain perfect eradication of tumors. The response rate to cancer immunotherapies, especially checkpoint inhibitors and adoptive T cell therapy, substantially differs in various cancer types with the highest rates in advanced melanoma and non-small cell lung cancer. Indeed, the lack of response in many tumors indicates primary resistance that can originate from either tumor cells (intrinsic) or tumor microenvironment (extrinsic). On the other hand, some tumors show an initial response to immunotherapy followed by relapse in few months (acquired resistance). Understanding the underlying molecular mechanisms of immunotherapy resistance makes it possible to develop effective strategies to overcome this hurdle and boost therapy outcomes. In this review, we take a look at immunotherapy strategies and go through a number of primary and acquired resistance mechanisms. Also, we present various ongoing methods to overcoming resistance and introduce some promising fields to improve the outcome of immunotherapy in patients affected with cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neoplasms , Humans , Immunotherapy , Neoplasm Recurrence, Local , Neoplasms/pathology , Tumor Microenvironment
7.
Mol Biol Rep ; 48(12): 8045-8059, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34689281

ABSTRACT

Along with evolution, a considerable number of signaling cascades have evolved within cells to meet their multifaceted needs. Among transmitting molecules, phosphoinositide 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR) have teamed up to build a signaling axis that effectively regulates various cellular processes including cell proliferation and migration. Given the extensive output of the PI3K/Akt/mTOR signaling axis, its aberrancy could subsequently lead to the formation of a wide range of human cancers spanning from hematologic malignancies to different types of solid tumors. Despite the high frequency of the PI3K pathway over-activation in most malignancies, mutations in the DNA sequence are not equally common. Such incompatibility sheds light on the possible effects of post-translational modification mechanisms that may take control of this pathway, some of the most important ones of which are through microRNAs (miRNAs or miRs). The present review is designed to take off the veil from the regulatory role of these small non-coding RNAs on the PI3K/Akt/mTOR signaling axis in carcinogenesis.


Subject(s)
MicroRNAs/genetics , Neoplasms/genetics , Signal Transduction/physiology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Humans , MicroRNAs/metabolism , Neoplasms/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism
8.
Urol Case Rep ; 39: 101818, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34485086

ABSTRACT

Renal arteriovenous fistula (AVF), an abnormal connection between artery and vein, results from development failure or following vascular injury. AVFs may cause various complications, i.e., secondary hypertension and hematuria. To manage AVF, it is recommended to bypass it from blood circulation surgically or by percutaneous embolization. The present study describes a woman with AVF, who primarily was managed percutaneously and then surgically.

9.
Urol Case Rep ; 38: 101651, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33868939

ABSTRACT

Blunt perineum trauma rarely leads to massive urethrorrhagia due to the formation of a bulbar aneurysm. A 29-year-old man with unstable hemodynamic underwent a digital subtraction angiography (DSA), which revealed a pseudoaneurysm in the penile bulb supplied from both internal pudendal arteries fistulized to the urethral duct bulb. A catheter was inserted into the distal part of pudendal arteries at the pseudoaneurysm's proximity, and an intermittent embolizing agent (Gel-foam) was injected. The pseudoaneurysm was filled with Gel-foam. Despite the superiority of conservative management, urethrorrhagia's life-threatening nature calls for angiographic intervention, successfully embolized using gel-foam with negligible complications.

10.
Int Immunopharmacol ; 90: 107202, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33278749

ABSTRACT

PURPOSE: Acute myeloid leukemia (AML) is one of the most severe blood cancers. Many studies have revealed that inflammation has an essential role in the progression of hematopoietic malignancies. Since the toll-like receptor 4 (TLR4) pathway, an important pathway involved in inflammation induction, has previously been associated with solid tumors, we hypothesized that it would be correlated with the pathophysiological characteristics of AML patients and could be considered as an anticancer target. METHOD: We evaluated the mRNA expression of TLR4, MyD88, RelB, and NF-кB using qRT-PCR in bone-marrow samples of 40 AML patients categorized into four groups according to prognosis, cell type, age, and drug response. Next, we explored the expression of these genes in three AML cell lines (NB4, U937, and KG-1) and used TAK-242, a specific inhibitor of TLR4, to investigate whether this inhibition could suppress AML cell proliferation using cell-cycle analysis. The effect of TAK-242 on arsenic trioxide (ATO) cytotoxicity was also assessed. RESULT: The results of qRT-PCR showed that most genes had higher expression in patients with poor prognosis or drug-resistant statues. They were also overexpressed in patients with less-differentiated cells. Moreover, TAK-242 inhibited cell proliferation of all the cell lines and altered their cell cycle distribution. It could also intensify the cytotoxicity of ATO in combination therapy. CONCLUSION: In sum, the TLR4 pathway was related to pathophysiological characteristics of AML and its inhibition using TAK-242 could be considered as a promising treatment strategy in the TLR4 expressing AML cells, individually or in combination with ATO.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arsenic Trioxide/pharmacology , Cell Cycle/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , U937 Cells , Young Adult
11.
J Cell Physiol ; 236(6): 4121-4137, 2021 06.
Article in English | MEDLINE | ID: mdl-33230811

ABSTRACT

The toll-like receptor (TLR) family consists of vital receptors responsible for pattern recognition in innate immunity, making them the core proteins involved in pathogen detection and eliciting immune responses. The most studied member of this family, TLR4, has been the center of attention regarding its contributory role in many inflammatory diseases including sepsis shock and asthma. Notably, mounting pieces of evidence have proved that this receptor is aberrantly expressed on the tumor cells and the tumor microenvironment in a wide range of cancer types and it is highly associated with the initiation of tumorigenesis as well as tumor progression and drug resistance. Cancer therapy using TLR4 inhibitors has recently drawn scientists' attention, and the promising results of such studies may pave the way for more investigation in the foreseeable future. This review will introduce the key proteins of the TLR4 pathway and how they interact with major growth factors in the tumor microenvironment. Moreover, we will discuss the many aspects of tumor progression affected by the activation of this receptor and provide an overview of the recent therapeutic approaches using various TLR4 antagonists.


Subject(s)
Neoplasms/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antineoplastic Agents/therapeutic use , Disease Progression , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Receptor Cross-Talk , Receptors, Growth Factor/metabolism , Signal Transduction , Toll-Like Receptor 4/antagonists & inhibitors , Tumor Microenvironment
12.
Clin Case Rep ; 8(12): 3208-3212, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33363908

ABSTRACT

Leishmaniasis is a rare complication in adult cases even in endemic areas. Here, the first report of visceral leishmaniasis in a young woman in northeast of Iran has been described.

13.
Life Sci ; 257: 118060, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32645343

ABSTRACT

AIMS: Despite the remarkable anti-proliferative effects of Arsenic trioxide (ATO) in breast cancer cells, the requirement of high, toxic concentrations to induce apoptosis may cause serious side effects in patients. In the present study, we aimed to use BIBR1532, an hTERT inhibitor, in combination with ATO to sensitize MCF7 and MDA-231 cells to lower concentrations of ATO. MAIN METHODS: Breast cancer cell lines MCF7 and MDA-231 were cultured and treated with different doses of ATO and BIBR1532 for 48 h and its effects on cell survival and proliferation were analyzed by MTT, crystal violet staining, colony formation assay, cell cycle, AnnexinV/PI and Real-time PCR tests. KEY FINDINGS: ATO and BIBR1532 synergistically inhibited proliferation and colony-forming ability of breast cancer cells. Besides, BIBR1532 augmented ATO-induced cytotoxic effects via triggering G1 cell cycle arrest and induction of apoptosis coupled with the down-regulation of NF-κB target genes that were involved in cell cycle progression (e.g. CCND1 and CDK6) and prevention of apoptosis such as Bcl-2, Bcl-xl, c-IAP2, and Survivin Respectively. Moreover, ATO-BIBR1532 significantly reduced the mRNA expression level of RELA, NFKB1, and several validated target genes of the NF-κB signaling pathway including NFKBIA, VEGFC, c-Myc, and hTERT. SIGNIFICANCE: The combination of ATO and BIBR1532 synergistically induced its anti-proliferative effect in breast cancer cells by targeting the two key cancer-related pathways, hTERT and NF-κB, and disrupting their feed-forward loop at the same time which result in the reduction of NF-κB transcriptional activity and subsequent down-regulation of its target genes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Aminobenzoates/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Arsenic Trioxide/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , MCF-7 Cells , NF-kappa B/metabolism , Naphthalenes/administration & dosage , Signal Transduction/drug effects
14.
Eur J Pharmacol ; 882: 173298, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32593665

ABSTRACT

Prostate Cancer is the second cause of cancer-related death in men and development of metastatic castration-resistant prostate cancer (mCRPC) is the major reason for its high mortality rate. Despite various treatments, all patients succumb to resistant disease, suggesting that there is a pressing need for novel and more efficacious treatments. Members of the vascular endothelial growth factor (VEGF) family play key roles in the tumorigenesis of mCRPC, indicating that VEGF-targeted therapies may have potential anti-tumor efficacy in this malignancy. However, due to compensatory activation of other family members, clinical trials with single-targeted VEGF inhibitors were discouraging. Here, we determined the anti-neoplastic activity of Cediranib, a pan-VEGF receptor inhibitor, in the mCRPC cell lines. Anti-growth effects of Cediranib were studied by MTT and BrdU cell proliferation assays and crystal violet staining. Annexin V/PI, radiation therapy and cell motility assays were carried out to examine the effects of Cediranib on apoptosis, radio-sensitivity and cell motility. Quantitative reverse transcription-PCR (qRT-PCR) and Western blot analyses were conducted to determine the molecular mechanisms underlying the anti-tumor activity of Cediranib. Cediranib decreased cell viability and induced apoptosis via inhibition of the anti-apoptotic proteins. Combination with Cediranib synergistically increased Docetaxel sensitivity and potentiated the effects of radiation therapy. Furthermore, Cediranib impaired cell motility via decrease in the expression of the epithelial-to-mesenchymal transition markers. These findings suggest that Cediranib may have anti-tumor activity in mCRPC cells and warrant further investigation on the therapeutic activity of this pan-VEGF receptor inhibitor in mCRPC.


Subject(s)
Adenocarcinoma , Antineoplastic Agents/pharmacology , Prostatic Neoplasms , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Docetaxel/pharmacology , Gamma Rays , Gene Expression Regulation/drug effects , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Receptors, Vascular Endothelial Growth Factor/genetics
15.
Cancer Chemother Pharmacol ; 85(1): 47-59, 2020 01.
Article in English | MEDLINE | ID: mdl-31786654

ABSTRACT

BACKGROUND: Despite all advances in the treatment of ovarian cancer (OC), it remains the most lethal gynecological malignancy worldwide. There are growing amounts of evidence indicating the role of inflammation in initiating chemoresistance. Therefore, Toll-like receptor 4 (TLR4), a mediator of inflammation in cancer cells, may be a proper anticancer target. METHODS: The effects of TLR4 activation by LPS was studied using MTT, colony formation, staining, scratch, and qRT-PCR assays as the first step. Then the same assays, in addition to anoikis resistance, cell cycle and annexin V/PI apoptosis tests, were used to investigate whether the inhibition of TLR4 using a small molecule inhibitor, TAK-242, could suppress the proliferation of various OC cell lines: A2780CP, 2008C13, SKOV3, and A2780S. RESULTS: The activation of TLR4 using LPS showed enhanced proliferation and invasion in the TLR4-expressing cell line (SKOV3). Next, treatment with the inhibitor revealed that TAK-242 suppressed the inflammatory condition of ovarian cancer cells, as evident by the down-regulation of IL-6 gene expression. We also found that TAK-242 halted cancer cell proliferation by inducing cell cycle arrest and apoptosis through the modulation of genes involved in these processes. Given the fact that the overexpression of TLR4 contributes to drug resistance, it was tempting to investigate the effect of TAK-242 in a combined-modality strategy. Interestingly, we found enhanced cytotoxicity when TAK-242 was used in combination with doxorubicin. CONCLUSION: TAK-242 serves as an appealing therapeutic strategy in the TLR4-expressing OC cells, either in the context of monotherapy or in combination with a chemotherapeutic drug.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Anoikis/drug effects , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Doxorubicin/pharmacology , Drug Synergism , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Tumor Cells, Cultured
16.
J Cell Biochem ; 121(2): 1623-1634, 2020 02.
Article in English | MEDLINE | ID: mdl-31535397

ABSTRACT

Increasing pieces of evidence indicate that inflammatory processes facilitate tumorigenesis; tumor cells simulate the mechanisms by which innate immune cells produce pro-inflammatory cytokines to exploit them for their own survival and proliferation. Toll-like receptor 4 (TLR4), which serves as one of the most well-known receptors on the surface of the immune cells, is often expressed ectopically in the tumor cells resulting in tumor progression, invasion, and chemoresistance. In this study, we examined the anticancer effects of TAK-242, a small molecule inhibitor of TLR4, on different breast cancer cell lines: MCF7, SKBR3, MDA-MB-231, and BT-474. Our results showed that the TLR4 inhibition, as revealed by the downregulation of TLR4 downstream genes, exerted desirable cytotoxicity on the TLR4-expressing cells, at least partly, through the downregulation of EGFR and c-Myc genes. TAK-242 also inhibited the proliferation of anoikis-resistant cells and suppressed the clonal growth of the indicated cells. The results of this study propose a mechanistic pathway by which the inhibition of TLR4 using TAK-242 could augment apoptotic cell death through the alteration of both nuclear factor-кB- and p53-related apoptosis genes in breast cancer cells, especially cells with overexpression of TLR4. Taken together, this study supports the idea that the activation of inflammatory pathways may have a crucial role in breast cancer progression and the inhibition of TLR4 using TAK-242, either as a single agent or in combination, seems to be a novel promising strategy that could be clinically available in foreseeable future.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle , Cell Movement , Cell Proliferation , Drug Therapy, Combination , Female , Humans , Tumor Cells, Cultured
17.
Eur J Pharmacol ; 863: 172705, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31574259

ABSTRACT

Gastric adenocarcinoma (GAC), the most common malignancy of the stomach, is the fourth most common and the second cause of cancer-related death worldwide. Although HER family plays a cardinal role in tumorigenesis of GAC, trastuzumab is the only approved anti-HER drug for this malignancy and development of resistance to trastuzumab is inevitable. Additionally, single-targeted HER inhibitors have demonstrated limited activity in GAC. Hence, there is a pressing need to devise more efficacious anti-HER therapeutic strategies. Here, we examined the anti-tumor activity of neratinb, a pan-HER inhibitor, on GAC cells. Anti-proliferative effects of neratinib were determined using a cell proliferation assay and crystal violet staining. Annexin V/PI staining, radiation therapy and anoikis resistance and wound healing assays were carried out to examine the effects of neratinib on apoptosis, radio-sensitivity and cell motility, respectively. Quantitative reverse transcription-PCR (qRT-PCR) analyses were applied to further investigate the anti-tumor activity of neratinib. We found that neratinib sensitized GAC cells to 5FU, carboplatin and oxaliplatin. Moreover, we found that neratinib was synergistic with trametinib (an approved MEK inhibitor) and foretinib (a c-MET inhibitor) and potentiated radio-sensitivity of GAC cells. Furthermore, we found that neratinib diminished GAC cell proliferation along with downregulation of FOXM1 and its targets. Additionally, neratinib induced apoptosis along with upregulation of pro-apoptotic and downregulation of anti-apoptotic genes. Treatment with neratinib attenuated invasive ability of GAC cells as shown by reduced anoikis resistance, downregulation of EMT markers, and reduced width in scratch assay. Our findings indicate that neratinib provides the therapeutic potential in the treatment of GAC.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Stomach Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Neoplasm Invasiveness
18.
Int J Biochem Cell Biol ; 116: 105615, 2019 11.
Article in English | MEDLINE | ID: mdl-31539632

ABSTRACT

Although conventional therapeutic approaches have brought remarkable advantages for the treatment of breast cancer (BC), drug-resistance still remains as a leading cause of tumor recurrence in this malignancy. In the present study, we designed experiments to evaluate the therapeutic value of PI3K inhibition in combination with Arsenic trioxide (ATO) in MCF-7 cells. The results of our study manifested that BKM120 sensitized MCF-7 cells to the lower concentrations of ATO. The significant anti-cancer effect of PI3K inhibition became even more evident when we found that BKM120, either as a single agent or in combination with ATO, reduced clonogenic ability of anoikis-resistant stem-like MCF-7 cells. Our findings also showed that BKM120 augmented ATO-induced anti-proliferative effects through inducing G1 arrest and reducing the incorporation of BrdU into the synthesized DNA of drugs-treated cells, which was coupled with c-Myc-mediated suppression of hTERT expression. Moreover, we found that in the presence of PI3K inhibitor, ATO is able to more profoundly induce apoptosis in MCF-7 cells, as revealed by the increment in the percentage of haplodiploid sub-G1 cells and the externalization of phosphatidylserine. Real-time PCR analysis also revealed that probably down-regulation of survivin coupled with up-regulation of forkhead family transcription factors is responsible for the enhancive effect of drugs in this cell line. Conclusively, this study shed lights on the effect of PI3K inhibition in chemo-sensitivity of MCF-7 cells, disclosing that combination of BKM120 and ATO could be a promising therapeutic approach in BC.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Anoikis/drug effects , Anoikis/genetics , Cell Line , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Combinations , Drug Synergism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Humans , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylserines/metabolism , Signal Transduction , Survivin/antagonists & inhibitors , Survivin/genetics , Survivin/metabolism , Telomerase/antagonists & inhibitors , Telomerase/genetics , Telomerase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Eur J Pharmacol ; 853: 256-263, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30930249

ABSTRACT

Numerous links exist between inflammation and tumor development. Toll-like receptor 4 (TLR4) expression by tumor cells can be a contributing factor that promotes tumor cell proliferation, survival, migration, and metastasis. In this study, we explored the impact of TLR4 inhibition using TAK-242, a specific inhibitor of TLR4, on the invasion properties of ovarian (A2780CP, 2008C13, SKOV3, and A2780S) and breast (MCF7, SKBR3, MDA-MB-231, and BT-474) cancer cell lines. Six out of eight cell lines expressed TLR4 and its downstream mediators (MyD88, NF-ĸB1, and RELB), indicating that these cell lines could be proper candidates for the TLR4 inhibition. TAK-242 induced a cytotoxic effect on all tested cell lines; however, a different cell sensitivity pattern was noticeable. Interestingly, in the TLR4-expressing cell lines, there was a significant correlation between the TLR4/MyD88 expressions and the cancer cell response to TAK-242: the higher the expression, the higher the IC50. To the best of our knowledge, no study has addressed the effects of TAK-242 on invasive abilities of cancer cells and our study suggests for the first time that TAK-242 could considerably decrease invasion properties of ovarian and breast cancer cell lines. We found that not only did TAK-242 reduce the enzymatic activity of MMP2 and MMP9, but also down-regulated gene expressions of epithelial-mesenchymal transition (EMT)-related genes. In sum, it seems that targeting TLR4 using TAK-242 possesses novel promising potential in cancer treatment strategies and may prevent invasion in patients suffering from ovarian and breast cancers, especially in those with over-expression of TLR4.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix/drug effects , Ovarian Neoplasms/pathology , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation/drug effects , Extracellular Matrix/metabolism , Female , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Toll-Like Receptor 4/metabolism
20.
Immunol Res ; 67(6): 505-516, 2019 12.
Article in English | MEDLINE | ID: mdl-32026322

ABSTRACT

It is believed that pathways of the immune system are responsible for eradicating cancer cells; however, their over-activation and also their ectopic expression in tumor cells and microenvironment are major contributors to tumor growth and chemoresistance. Toll-like receptor 4 (TLR4) pathway is an innate immune-related pathway which is usually overexpressed in tumor cells that leads to excessive pro-inflammatory cytokines and eventually results in tumor survival, drug resistance, and metastasis. In this study, we investigated whether TLR4 expression is affected upon the treatment of breast and ovarian cancer cells with common chemotherapeutics (paclitaxel, cisplatin, doxorubicin, and arsenic trioxide) and if TLR4 inhibition using its specific inhibitor TAK-242 could enhance cancer cells' response to the drugs. Both breast (MCF7) and ovarian (2008C13) cancer cells experienced an elevated expression of TLR4 after treatment with the drugs. The expression of this receptor was also upregulated in cisplatin-resistant 2008C13 cells; however, it was significantly higher upon short-term treatment with cisplatin. More importantly, the combination treatment of the drugs with TAK-242 intensified the chemosensitivity of six different breast and ovarian cancer cells to chemotherapeutic drugs. It was also identified that co-treatment of paclitaxel and TAK-242 not only led to enhanced G2/M arrest and apoptosis but also satisfactorily decreased the expression of TLR4 and different interleukins in these cells. Taken together, the results of the present study emphasize that chemotherapy may lead to chemoresistance through inducing TLR4 expression, and therefore inhibiting this receptor using TAK-242 could be a promising approach to improve the outcome of chemotherapy in foreseeable future.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Drug Synergism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , MCF-7 Cells , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL