Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 46(1): 8-14, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37962127

RESUMEN

Osteosarcoma is the most common primary bone tumor. Using the multiple ligands simultaneous docking method, we found that bazedoxifene could bind to the GP130 D1 domain. We then demonstrated that bazedoxifene can decrease cell viability and cell migration of osteosarcoma cells by inhibiting interleukin 6 (IL-6) and IL-11/GP130 signaling. Consistently, treatment with IL-6 or IL-11 antibody or knockdown of GP130 by siRNA silenced the activation of STAT3, ERK, and AKT. Similarly, recombinant IL-6 and IL-11 proteins antagonized the inhibitory effect of bazedoxifene on osteosarcoma cells. Finally, the combinational treatment of temsirolimus and bazedoxifene synergistically suppressed osteosarcoma development in vitro and in vivo. Our findings suggest that bazedoxifene directly prompts the deactivation of GP130 and inhibits the osteosarcoma progression in vitro and in vivo. Therefore, bazedoxifene could be effectively applied as a therapeutic drug for human osteosarcoma in the future.


Asunto(s)
Interleucina-6 , Osteosarcoma , Humanos , Receptor gp130 de Citocinas/metabolismo , Interleucina-11/farmacología , Línea Celular Tumoral , Transducción de Señal , Osteosarcoma/tratamiento farmacológico
2.
Molecules ; 28(2)2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36677735

RESUMEN

In the past 25 years, a number of efforts have been made toward the development of small molecule interleukin-6 (IL-6) signaling inhibitors, but none have been approved to date. Monosaccharides are a diverse class of bioactive compounds, but thus far have been unexplored as a scaffold for small molecule IL-6-signaling inhibitor design. Therefore, in this present communication, we combined a structure-based drug design approach with carbohydrate building blocks to design and synthesize novel IL-6-signaling inhibitors targeting glycoprotein 130 (gp130). Of this series of compounds, LS-TG-2P and LS-TF-3P were the top lead compounds, displaying IC50 values of 6.9 and 16 µM against SUM159 cell lines, respectively, while still retaining preferential activity against the IL-6-signaling pathway. The carbohydrate moiety was found to improve activity, as N-unsubstituted triazole analogues of these compounds were found to be less active in vitro compared to the leads themselves. Thus, LS-TG-2P and LS-TF-3P are promising scaffolds for further development and study as IL-6-signaling inhibitors.


Asunto(s)
Antineoplásicos , Interleucina-6 , Antineoplásicos/farmacología , Carbohidratos/farmacología , Línea Celular Tumoral , Proliferación Celular , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Interleucina-6/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Transducción de Señal , Relación Estructura-Actividad , Humanos
3.
J Cell Mol Med ; 24(8): 4748-4761, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32164044

RESUMEN

The role of IL-6 signalling in hypertensive heart disease and its sequelae is controversial. Our group demonstrated that Bazedoxifene suppressed IL-6/gp130 signalling in cancer cells but its effect on myocardial pathology induced by pressure overload is still unknown. We explored whether Bazedoxifene could confer benefits in wild-type C57BL/6J mice suffering from transverse aortic constriction (TAC) and the potential mechanisms in H9c2 myoblasts. Mice were randomized into three groups (Sham, TAC, TAC+Bazedoxifene, n = 10). Morphological and histological observations suggested TAC aggravated myocardial remodelling while long-term intake of Bazedoxifene (5 mg/kg, intragastric) attenuated pressure overload-induced pathology. Echocardiographic results indicated Bazedoxifene rescued cardiac function in part. We found Bazedoxifene decreased the mRNA expression of IL-6, MMP2, Col1A1, Col3A1 and periostin in murine hearts after 8-week surgery. By Western blot detection, we found Bazedoxifene exhibited an inhibition of STAT3 activation in mice three hours and 8 weeks after TAC. Acute TAC stress (3 hours) led to down-regulated ratio of LC3-Ⅱ/LC3-Ⅰ, while in mice after long-term (8 weeks) TAC this ratio becomes higher than that in Sham mice. Bazedoxifene inverted the autophagic alteration induced by TAC at both two time-points. In H9c2 myoblasts, Bazedoxifene suppressed the IL-6-induced STAT3 activation. Moreover, IL-6 reduced the ratio of LC3-Ⅱ/LC3-Ⅰ, promoted P62 expression but Bazedoxifene reversed both changes in H9c2 cells. Our data suggested Bazedoxifene inhibited IL-6/gp130 signalling and protected against cardiac remodelling together with function deterioration in TAC mice.


Asunto(s)
Receptor gp130 de Citocinas/genética , Hipertensión/tratamiento farmacológico , Indoles/farmacología , Interleucina-6/genética , Factor de Transcripción STAT3/genética , Animales , Autofagia/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Hipertensión/genética , Hipertensión/patología , Ratones , Ratas , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética
4.
Breast Cancer Res Treat ; 181(1): 31-41, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32240456

RESUMEN

PURPOSE: Persistent STAT3 signaling is frequently detected in many cancer types including triple-negative breast cancer, and thus could potentially serve as a viable therapeutic target. We have designed a novel non-peptide compound LLY17 targeting STAT3 using Advanced Multiple Ligand Simultaneous Docking (AMLSD) methods. However, the efficacy of LLY17 has not been evaluated extensively in human and murine triple-negative breast cancer cells. In this study, we tested LLY17 in multiple human and murine triple-negative breast cancer cell lines. METHODS: Human triple-negative breast cancer MDA-MB-468, MDA-MB-231, SUM159, and BT-549 cells, and murine triple-negative breast cancer 4T1 cells were used to study the inhibition effects of LLY17. The inhibition of STAT3 activation of LLY17 was investigated using western blot analysis. Cell viability, apoptosis and migration assays were carried out by MTT assay, Caspase-3/7 assay and wound healing assay, respectively. A mammary fat pad syngeneic mouse model was used to evaluate the antitumor effect of LLY17 in vivo. RESULTS: LLY17 inhibited IL-6-mediated induction of STAT3 phosphorylation but had no effect on IFN-γ-induced STAT1 phosphorylation or EGF-induced ERK phosphorylation. LLY17 inhibited STAT3 phosphorylation and induced apoptosis in human and murine triple-negative breast cancer cells but exhibited minimal toxicity toward Luminal A subtype breast cancer MCF-7 cells. RNAi attenuation experiments supported the requirement of STAT3 for LLY17-mediated inhibition of cell viability in triple-negative breast cancer cells. In addition, LLY17 inhibited cell migration of human and murine triple-negative breast cancer cells. Furthermore, LLY17 suppressed tumor growth and STAT3 phosphorylation of triple-negative breast cancer cells in a mammary fat pad syngeneic mouse model in vivo. CONCLUSIONS: Together, our findings suggest that targeting persistent STAT3 signaling by novel small molecule LLY17 may be a potential approach for the therapy of triple-negative breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Sci ; 110(3): 950-961, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30648776

RESUMEN

The interleukin (IL)-6/glycoprotein (GP)130/signal transducer and activator of transcription (STAT)3 pathway is emerging as a target for the treatment of hepatocellular carcinoma. IL-6 binds to IL-6R, forming a binary complex, which further combines with GP130 to transduce extracellular signaling by activating STAT3. Therefore, blocking the interaction between IL-6 and GP130 may inhibit the IL-6/GP130/STAT3 signaling pathway and its biological effects. It has been reported that bazedoxifene acetate (BAZ), a selective estrogen receptor modulator approved by the US Food and Drug Administration, could inhibit IL-6/GP130 protein-protein interactions. Western blot, immunofluorescence staining, wound healing and colony formation assays were used to detect the effect of BAZ on liver cancer cells. Cell viability was evaluated by MTT assay. Apoptosis of cells was determined using the Annexin V-FITC detection kit. Mouse xenograft tumor models were utilized to evaluate the effect of BAZ in vivo. Our data showed that BAZ inhibited STAT3 phosphorylation (P-STAT3) and expression of STAT3 downstream genes, inducing apoptosis in liver cancer cells. BAZ inhibited P-STAT3 induced by IL-6, but not by leukemia inhibitory factor. BAZ inhibited P-STAT1 and P-STAT6 less significantly as elicited by interferon-α, interferon-γ and IL-4. In addition, pretreatment of BAZ impeded the translocation of STAT3 to nuclei induced by IL-6. BAZ inhibited cell viability, wound healing and colony formation in vitro. Furthermore, tumor growth in HEPG2 mouse xenografts were significantly inhibited by daily intragastric gavage of BAZ. Our results suggest that BAZ inhibited the growth of hepatocellular carcinoma in vitro and in vivo, indicating another potential strategy for HCC prevention and therapy.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Glicoproteínas/metabolismo , Indoles/farmacología , Interleucina-6/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Células Hep G2 , Humanos , Interleucina-4/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Receptores de Interleucina-6/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Breast Cancer Res Treat ; 175(3): 553-566, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30852762

RESUMEN

PURPOSE: Triple-negative breast cancer (TNBC) has been ranked as one of the devastating malignancy worldwide. Its disease progression and treatment obstacle is associated with the negligible expression of estrogen receptors (ER-), progesterone receptors (PR-), and HER2 (HER2-). Due to a lack of growth hormone receptors, TNBC is desperately demanding effective therapeutic regimens. A growing body of evidence indicated that glycoprotein 130 kDa (GP130), the pivotal mediator involved in interleukin 6 (IL-6) and signal transducer and activator of transcription 3 (STAT3) signaling pathways, is strongly correlated with tumor progression. Therefore, GP130 could become a novel target for treating TNBC. In our earlier studies, we demonstrated bazedoxifene as being a novel GP130 inhibitor. METHODS: In the current report, anti-tumor effect of bazedoxifene on TNBC was further evaluated in TNBC cell lines SUM159, MDA-MB-231, and MDA-MB-468. We assessed anti-TNBC potency of bazedoxifene by carrying out various analysis encompassing western blot, cell proliferation, cell migration, colony formation, and growth of tumors in the xenograft mice. RESULTS: Our findings demonstrated that bazedoxifene not only decreased the expression of P-STAT3, IL-6/GP130-mediated downstream target genes P-AKT and P-ERK, but also blocked mitogen effects stimulated by IL-6, including cell viability, and overall cell survive, proliferation as well as cell migration. Likewise in laboratory animal model, tumor growth in mice was remarkably suppressed by bazedoxifene via an oral administration route. Combinational treatment of bazedoxifene plus the conventional chemotherapeutic agent, paclitaxel, synergistically impeded cell viability, colony formation, and cell migration far more significantly than the one from single-drug alone. CONCLUSIONS: Taken together, our data suggest that bazedoxifene may be developed as a promising small molecular therapeutic agent for eradicating TNBC intrinsically associated with constitutively active IL-6/GP130/STAT3 signaling cascade.


Asunto(s)
Antineoplásicos/administración & dosificación , Receptor gp130 de Citocinas/antagonistas & inhibidores , Indoles/administración & dosificación , Interleucina-6/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Administración Oral , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Receptor gp130 de Citocinas/metabolismo , Regulación hacia Abajo , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Ratones , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biochem Biophys Res Commun ; 491(1): 159-165, 2017 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-28711499

RESUMEN

Persistent activation of IL-6/STAT3 signaling pathway has been frequently detected in human cancer including breast cancer, colon cancer and multiple myeloma. IL-6/STAT3 can be a promising target for cancer prevent and treatment. However, few STAT3 inhibitors with high efficiency, specificity and safety is available for present clinical cancer therapy. Evista (Raloxifene·HCl) is known as selective estrogen receptor modulator which has been used for the prevention and treatment of osteoporosis and was approved for reducing the risk of invasive breast cancer. Our previous study found that Raloxifene inhibited IL-6/GP130 interaction, resulting in blockade of STAT3 phosphorylation. In our present study, we examined the effect on IL-6/GP130/STAT3 signaling pathway and cancer cell viability with Evista. We first demonstrated Evista inhibited constitutive activation of STAT3 in breast cancer cell line MDB-MB-231, colon cancer cell line HCT116 and multiple myeloma cancer cell line U266. Evista also inhibited phosphorylation of STAT3 induced by IL-6 in MCF-7, HT29 and MM.1S cancer cell lines. Induction of apoptosis was exerted in MDA-MB-231, HCT116 and U266 as evidenced by increased caspase-3 cleavage. However, Evista did not inhibit STAT1, STAT2, STAT4 or STAT6 phosphorylation elicited by IFN-α, IFN-γ and IL-4, nor phosphorylation of STAT3 induced by LIF in MCF-7 cell lines. Evista attenuated STAT3 phosphorylation, decreased STAT3 transcriptional activity but much less in pGL3 and AP1 transcriptional luciferase activity, and decreased cell viability in vitro. These results suggest that it may be possible for Evista to emerge as a chemoprevention agent for breast cancer and other cancers such as colon cancer or multiple myeoloma by targeting IL-6/STAT3 signaling.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Interleucina-6/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Clorhidrato de Raloxifeno/administración & dosificación , Clorhidrato de Raloxifeno/farmacología , Factor de Transcripción STAT3/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Relación Dosis-Respuesta a Droga , Células HCT116 , Células HT29 , Humanos , Células MCF-7 , Terapia Molecular Dirigida , Neoplasias Experimentales/patología , Transducción de Señal/efectos de los fármacos
8.
J Biol Chem ; 290(6): 3418-29, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25313399

RESUMEN

Signal transducers and activators of transcription 3 (STAT3) signaling is persistently activated and could contribute to tumorigenesis of medulloblastoma. Numerous studies have demonstrated that inhibition of the persistent STAT3 signaling pathway results in decreased proliferation and increased apoptosis in human cancer cells, indicating that STAT3 is a viable molecular target for cancer therapy. In this study, we investigated a novel non-peptide, cell-permeable small molecule, named LY5, to target STAT3 in medulloblastoma cells. LY5 inhibited persistent STAT3 phosphorylation and induced apoptosis in human medulloblastoma cell lines expressing constitutive STAT3 phosphorylation. The inhibition of STAT3 signaling by LY5 was confirmed by down-regulating the expression of the downstream targets of STAT3, including cyclin D1, bcl-XL, survivin, and micro-RNA-21. LY5 also inhibited the induction of STAT3 phosphorylation by interleukin-6 (IL-6), insulin-like growth factor (IGF)-1, IGF-2, and leukemia inhibitory factor in medulloblastoma cells, but did not inhibit STAT1 and STAT5 phosphorylation stimulated by interferon-γ (IFN-γ) and EGF, respectively. In addition, LY5 blocked the STAT3 nuclear localization induced by IL-6, but did not block STAT1 and STAT5 nuclear translocation mediated by IFN-γ and EGF, respectively. A combination of LY5 with cisplatin or x-ray radiation also showed more potent effects than single treatment alone in the inhibition of cell viability in human medulloblastoma cells. Furthermore, LY5 demonstrated a potent inhibitory activity on cell migration and angiogenesis. Taken together, these findings indicate LY5 inhibits persistent and inducible STAT3 phosphorylation and suggest that LY5 is a promising therapeutic drug candidate for medulloblastoma by inhibiting persistent STAT3 signaling.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/farmacología , Transporte Activo de Núcleo Celular , Aminopiridinas/síntesis química , Aminopiridinas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Cisplatino/farmacología , Ciclina D/genética , Ciclina D/metabolismo , Citocinas/farmacología , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Meduloblastoma/metabolismo , MicroARNs/genética , Fosforilación , Transducción de Señal , Sulfonamidas/síntesis química , Sulfonamidas/química , Survivin , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
9.
Bioorg Med Chem ; 23(6): 1348-55, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25698618

RESUMEN

Inhibition of the signaling pathways of signal transducer and activator of transcription 3 (STAT 3) has shown to be a promising strategy to combat cancer. In this paper we report the design, synthesis and evaluation of a novel class of small molecule inhibitors, that is, XZH-5 and its analogues, as promising leads for further development of STAT3 inhibitors. Preliminary SARs was established for XZH-5 and its derivatives; and the binding modes were predicted by molecular docking. Lead compounds with IC50 as low as 6.5µM in breast cancer cell lines and 7.6µM in pancreatic cancer cell lines were identified.


Asunto(s)
Diseño de Fármacos , Histidina/análogos & derivados , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Histidina/síntesis química , Histidina/química , Histidina/farmacología , Humanos , Estructura Molecular , Compuestos de Fenilurea/química , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
10.
Eur Respir Rev ; 33(171)2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38355149

RESUMEN

Lung cancer is the leading cause of cancer-related deaths worldwide, and ∼85% of lung cancers are classified as nonsmall cell lung cancer (NSCLC). These malignancies can proliferate indefinitely, in part due to dysregulation of the cell cycle and the resulting abnormal cell growth. The specific activation of cyclin-dependent kinases 4 and 6 (CDK4/6) is closely linked to tumour proliferation. Approximately 80% of human tumours exhibit abnormalities in the cyclin D-CDK4/6-INK4-RB pathway. Specifically, CDK4/6 inhibitors either as monotherapy or combination therapy have been investigated in pre-clinical and clinical studies for the treatment of NSCLC, and promising results have been achieved. This review article focuses on research regarding the use of CDK4/6 inhibitors in NSCLC, including the characteristics and mechanisms of action of approved drugs and progress of pre-clinical and clinical research.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quinasa 4 Dependiente de la Ciclina/metabolismo , Proliferación Celular , Inhibidores de Proteínas Quinasas/efectos adversos
11.
Cancers (Basel) ; 15(13)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37444617

RESUMEN

Mounting evidence links the phenomenon of enhanced recruitment of tumor-associated macrophages towards cancer bulks to neoplastic growth, invasion, metastasis, immune escape, matrix remodeling, and therapeutic resistance. In the context of cancer progression, naïve macrophages are polarized into M1 or M2 subtypes according to their differentiation status, gene signatures, and functional roles. While the former render proinflammatory and anticancer effects, the latter subpopulation elicits an opposite impact on pancreatic ductal adenocarcinoma. M2 macrophages have gained increasing attention as they are largely responsible for molding an immune-suppressive landscape. Through positive feedback circuits involving a paracrine manner, M2 macrophages can be amplified by and synergized with neighboring neoplastic cells, fibroblasts, endothelial cells, and non-cell autonomous constituents in the microenvironmental niche to promote an advanced disease state. This review delineates the molecular cues expanding M2 populations that subsequently convey notorious clinical outcomes. Future therapeutic regimens shall comprise protocols attempting to abolish environmental niches favoring M2 polarization; weaken cancer growth typically assisted by M2; promote the recruitment of tumoricidal CD8+ T lymphocytes and dendritic cells; and boost susceptibility towards gemcitabine as well as other chemotherapeutic agents.

12.
Int J Cancer ; 130(6): 1459-69, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21520044

RESUMEN

We characterized the effects of a newly developed signal transducers and activators of transcription 3 (STAT3) inhibitor, LLL12 in multiple myeloma (MM) cells. LLL12 specifically inhibited STAT3 phosphorylation, nuclear localization, DNA binding activity, down-regulated STAT3 downstream genes, and induced apoptosis in MM cells. Importantly, LLL12 significantly inhibited STAT3 phosphorylation, induced apoptosis in primary MM cells which came from patients that were clinically resistant to lenalidomide and bortezomib. LLL12 is a potent inhibitor of cell proliferation with IC50 values ranging between 0.26 and 1.96 µM in MM and primary MM cells. LLL12 also inhibited STAT3 phosphorylation induced by interleukin-6 (IL-6) and interferon-α but not STAT1, STAT2, STAT4 and STAT6 phosphorylation induced by interferon-α, interferon-γ and IL-4 indicating the selectivity of LLL12 for STAT3. The selectively of LLL12 on STAT3 was further demonstrated on 21 protein kinases, which LLL12 had IC50 values ≥ 73.92 µM. In addition, the pretreatment of LLL12 blocked the promotion of the cell proliferation and resistance to lenalidomide by IL-6. Furthermore, LLL12 significantly blocked tumor growth of MM cells in mouse model. Our results indicate that LLL12 blocks constitutive STAT3 and IL-6 induced STAT3 signaling and may be a potential therapeutic agent for MM.


Asunto(s)
Antraquinonas/farmacología , Interleucina-6/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Sulfonamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Interferón-alfa/metabolismo , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
13.
Invest New Drugs ; 30(3): 916-26, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21340507

RESUMEN

Constitutive activation of Signal Transducers and Activators of Transcription 3 (STAT3) is frequently detected in osteosarcoma, and hence, may serve as a therapeutic target. In order to target STAT3, we tested two new STAT3 inhibitors, LLL12 and FLLL32. LLL12 and FLLL32 inhibit STAT3 phosphorylation and STAT3 downstream targets. LLL12 and FLLL32 also inhibit IL-6 induced STAT3 phosphorylation. The inhibition of STAT3 by LLL12 and FLLL32 resulted in the induction of apoptosis, reduction of plating efficiency, and migration in osteosarcoma cells. Furthermore, LLL12 and FLLL32 inhibited SJSA osteosarcoma cells and OS-33 tumor growth in murine xenografts. These results provide evidence that constitutive STAT3 signaling is required for osteosarcoma survival and migration in vitro and tumor growth in vivo. Blocking persistent STAT3 signaling by LLL12 and FLLL32 may be a novel therapeutic approach for osteosarcoma.


Asunto(s)
Antraquinonas/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Curcumina/análogos & derivados , Osteosarcoma/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Animales , Antraquinonas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Curcumina/uso terapéutico , Femenino , Humanos , Interleucina-6/farmacología , Ratones , Ratones Desnudos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Sulfonamidas/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nutr Cancer ; 64(8): 1228-35, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145775

RESUMEN

Reactivation of tumor suppressor genes (TSGs) involved in carcinogenesis by nontoxic bioactive food component represents a promising strategy for cancer chemoprevention. Recently, curcumin has been demonstrated to inhibit a bacterial DNA methyltransferase (M. Sss I) activity, induce global DNA hypomethylation in leukemia cells, and reactivate several hypermethylation silenced genes in lung and prostate cancer cells. Herein, we demonstrated that curcumin can enhance the mRNA and protein levels of ras-association domain family protein 1A (RASSF1A), 1 hypermethylation-silenced TSG, and decrease its promoter methylation in breast cancer cells. Mechanistic study demonstrated that curcumin can decrease DNA methylation activity of nuclear extract and downregulate the mRNA and protein levels of DNMT1 in MCF-7 cells, which may be associated with curcumin-induced disruption of NF-κB/Sp1 complex bound to the promoter region of DNMT1. Altogether, this study reveals a novel molecular mechanism of curcumin as a chemo-preventive agent for breast cancer through hypomethylation reactivation of RASSF1A.


Asunto(s)
Neoplasias de la Mama/prevención & control , Curcumina/farmacología , Activación Transcripcional/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Animales , Neoplasias de la Mama/química , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , Proteínas Represoras/análisis , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/análisis , Ensayos Antitumor por Modelo de Xenoinjerto
15.
BMC Vet Res ; 8: 244, 2012 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-23244668

RESUMEN

BACKGROUND: STAT3 [1] has been shown to be dysregulated in nearly every major cancer, including osteosarcoma (OS). Constitutive activation of STAT3, via aberrant phosphorylation, leads to proliferation, cell survival and resistance to apoptosis. The present study sought to characterize the biologic activity of a novel allosteric STAT3 inhibitor, LLL12, in canine OS cell lines. RESULTS: We evaluated the effects of LLL12 treatment on 4 canine OS cell lines and found that LLL12 inhibited proliferation, induced apoptosis, reduced STAT3 phosphorylation, and decreased the expression of several transcriptional targets of STAT3 in these cells. Lastly, LLL12 exhibited synergistic anti-proliferative activity with the chemotherapeutic doxorubicin in the OS lines. CONCLUSION: LLL12 exhibits biologic activity against canine OS cell lines through inhibition of STAT3 related cellular functions supporting its potential use as a novel therapy for OS.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Neoplasias Óseas/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Osteosarcoma/veterinaria , Factor de Transcripción STAT3/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting/veterinaria , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Enfermedades de los Perros/metabolismo , Enfermedades de los Perros/patología , Perros , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Osteosarcoma/patología , Fosforilación/efectos de los fármacos , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/metabolismo
16.
Cancers (Basel) ; 14(2)2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35053449

RESUMEN

Breast cancer is the most common malignancy among females in western countries, where women have an overall lifetime risk of >10% for developing invasive breast carcinomas. It is not a single disease but is composed of distinct subtypes associated with different clinical outcomes and is highly heterogeneous in both the molecular and clinical aspects. Although tumor initiation is largely driven by acquired genetic alterations, recent data suggest microenvironment-mediated immune evasion may play an important role in neoplastic progression. Beyond surgical resection, radiation, and chemotherapy, additional therapeutic options include hormonal deactivation, targeted-signaling pathway treatment, DNA repair inhibition, and aberrant epigenetic reversion. Yet, the fatality rate of metastatic breast cancer remains unacceptably high, largely due to treatment resistance and metastases to brain, lung, or bone marrow where tumor bed penetration of therapeutic agents is limited. Recent studies indicate the development of immune-oncological therapy could potentially eradicate this devastating malignancy. Evidence suggests tumors express immunogenic neoantigens but the immunity towards these antigens is frequently muted. Established tumors exhibit immunological tolerance. This tolerance reflects a process of immune suppression elicited by the tumor, and it represents a critical obstacle towards successful antitumor immunotherapy. In general, immune evasive mechanisms adapted by breast cancer encompasses down-regulation of antigen presentations or recognition, lack of immune effector cells, obstruction of anti-tumor immune cell maturation, accumulation of immunosuppressive cells, production of inhibitory cytokines, chemokines or ligands/receptors, and up-regulation of immune checkpoint modulators. Together with altered metabolism and hypoxic conditions, they constitute a permissive tumor microenvironment. This article intends to discern representative incidents and to provide potential innovative therapeutic regimens to reinstate tumoricidal immunity.

17.
Int J Oncol ; 60(5)2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35315502

RESUMEN

Ovarian cancer is the most lethal gynecological cancer type in the United States. The success of current chemotherapies is limited by chemoresistance and side effects. Targeted therapy is a promising future direction for cancer therapy. In the present study, the efficacy of co­targeting IL­6 and IL­8 in human ovarian cancer cells by bazedoxifene (Baze) + SCH527123 (SCH) treatment was examined. ELISA, cell viability, cell proliferation, cell migration, cell invasion, western blotting and peritoneal ovarian tumor mouse model analyses were performed to analyze the expression levels of IL­6 and IL­8, tumor growth, tumor migration and invasion, and the possible pathways of human ovarian cancer cell lines (SKOV3, CAOV3 and OVCAR3) and patient­derived OV75 ovarian cancer cells. Each cell line was treated by monotherapy or combination therapy. The results demonstrated that IL­6 and IL­8 were secreted by human ovarian cancer cell lines. Compared with the DMSO control, the combination of IL­6/glycoprotein 130 inhibitor Baze and IL­8 inhibitor SCH synergistically inhibited cell viability in ovarian cancer cells. Baze + SCH also inhibited cell migration and invasion, suppressed ovarian tumor growth and inhibited STAT3 and AKT phosphorylation, as well as survivin expression. Therefore, co­targeting the IL­6 and IL­8 signaling pathways may be an effective approach for ovarian cancer treatment.


Asunto(s)
Benzamidas/farmacología , Ciclobutanos/farmacología , Indoles/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Benzamidas/administración & dosificación , Benzamidas/uso terapéutico , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclobutanos/administración & dosificación , Ciclobutanos/uso terapéutico , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Indoles/administración & dosificación , Indoles/uso terapéutico , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Ratones , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico
18.
Biomedicines ; 10(8)2022 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-36009550

RESUMEN

Persistent STAT3 signaling plays a pivotal role in human tumor malignancy, including triple-negative breast cancer (TNBC). There are few treatment options currently available for TNBC; thus, given its importance to cancer, STAT3 is a potential cancer therapeutic target and is the focus of drug discovery efforts. In this study, we tested a novel orally bioavailable small-molecule STAT3 inhibitor, LLL12B, in human MDA-MB-231, SUM159, and murine 4T1 TNBC cell lines. TNBC cells frequently expressed persistent STAT3 phosphorylation and their cell viability was sensitive to STAT3 knockdown by siRNA. LLL12B selectively inhibited the IL-6-mediated induction of STAT3 phosphorylation, but had little effect on the IFN-γ-mediated induction of STAT1 phosphorylation nor the EGF-mediated induction of ERK phosphorylation. In addition, targeting STAT3 with LLL12B induced apoptosis, reduced colony formation ability, and inhibited cell migration in TNBC cells. Furthermore, LLL12B suppressed the tumor growth of the MDA-MB-231 TNBC cells in a mammary fat pad mouse tumor model in vivo. Together, our findings support the concept that targeting persistent STAT3 signaling using the novel small-molecule LLL12B is a potential approach for TNBC therapy.

19.
J Biol Chem ; 285(35): 27429-27439, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20562100

RESUMEN

Interleukin-6 (IL-6) is a multifunctional cytokine, which may block apoptosis during inflammation to protect cells under very toxic conditions. However, IL-6 also activates STAT3 in many types of human cancer. Recent studies demonstrate that high levels of IL-6 are associated with hepatocellular carcinoma, the most common type of liver cancer. Here we reported that IL-6 promoted survival of human liver cancer cells through activating STAT3 in response to doxorubicin treatment. Endogenous IL-6 levels in SNU-449 cells were higher than in Hep3B cells. Meanwhile, SNU-449 cells were more resistant to doxorubicin than Hep3B cells. Addition of IL-6 induced STAT3 activation in Hep3B cells and led to protection against doxorubicin. In contrast, neutralizing IL-6 with anti-IL-6 antibody decreased survival of SNU-449 cells in response to doxorubicin. To elucidate the mechanism of the anti-apoptotic function of IL-6, we investigated if STAT3 mediated this drug resistance. Targeting STAT3 with STAT3 siRNA reduced the protection of IL-6 against doxorubicin-induced apoptosis, indicating that STAT3 signaling contributed to the anti-apoptotic effect of IL-6. Moreover, we further explored if a STAT3 small molecule inhibitor could abolish this anti-apoptotic effect. LLL12, a STAT3 small molecule inhibitor, blocked IL-6-induced STAT3 phosphorylation, resulting in attenuation of the anti-apoptotic activity of IL-6. Finally, neutralization of endogenous IL-6 with anti-IL-6 antibody or blockade of STAT3 with LLL12 lowered the recovery in SNU-449 cells after doxorubicin treatment. Therefore, our results demonstrated that targeting STAT3 signaling could interrupt the anti-apoptotic function of IL-6 in human liver cancer cells.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/metabolismo , Resistencia a Antineoplásicos , Interleucina-6/metabolismo , Neoplasias Hepáticas/metabolismo , Factor de Transcripción STAT3/metabolismo , Antraquinonas/farmacología , Antibióticos Antineoplásicos/farmacología , Anticuerpos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Humanos , Interleucina-6/antagonistas & inhibidores , Neoplasias Hepáticas/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
20.
Cancer Sci ; 102(7): 1381-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21435102

RESUMEN

Signal transducers and activators of transcription 3 (STAT3) signaling is constitutively activated in many types of human cancers and cancer cell lines and represents a promising target for cancer therapy. We previously reported that the STAT3 signaling pathway is constitutively activated in human rhabodomyosarcoma cell lines (RH28, RH30 and RD2). We also demonstrated that inhibition of the STAT3 pathway led to apoptosis in human rhabdomyosarcoma cells. In the present study, we investigated the inhibitory effects of a novel small molecule, XZH-5, on the STAT3 signaling pathway in human rhabdomyosarcoma cells. XZH-5 was designed based on STAT3 structure, and our idea was to design peptide mimics to bind to the phosphorylated Tyr705 site and the side pocket. We found that XZH-5 downregulated STAT3 phosphorylation. The inhibition of STAT3 by XZH-5 was confirmed by the inhibition of STAT3 DNA binding ability and the downregulation of STAT3 downstream genes, such as Bcl-2, Bcl-xL, Cyclin D1 and Survivin; we also demonstrated that blockade of STAT3 phosphorylation in human rhabdomyosarcoma cells with XZH-5 caused apoptosis and suppressed colony-forming ability and cell migration. In addition to reducing constitutive STAT3 phosphorylation, XZH-5 also exhibited the potency to block interleukin-6 (IL-6)-induced STAT3 phosphorylation and nuclear translocation but did not inhibit the stimulation of STAT1 phosphorylation by interferon (IFN)-γ. Our findings indicate that XZH-5 has the potential for targeting human rhabdomyosarcoma cells expressing constitutive STAT3.


Asunto(s)
Histidina/análogos & derivados , Interleucina-6/farmacología , Compuestos de Fenilurea/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Transporte Activo de Núcleo Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Descubrimiento de Drogas , Histidina/farmacología , Humanos , Fosforilación , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Factor de Transcripción STAT3/metabolismo , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA