Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Med Chem ; 112: 117897, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39216383

RESUMEN

Due to their pivotal roles in regulating energy metabolism and apoptosis, mitochondria in cancer cells have been considered a vulnerable and feasible target. Many anticancer agents, e.g., metal-based compounds, are found to target and disturb mitochondria primarily, which may lead to the disturbance of energy metabolism and, more importantly, the initiation of apoptosis. In this work, a gold-based complex 7 (GC7) was synthesized and evaluated in a series of different cancer cell lines. The anticancer efficacies of GC7 on cell viability, apoptosis, and colony formation were determined. Cellular thioredoxin reductase (TrxR) activity, oxygen consumption rate (OCR), glucose uptake, and lactate production following GC7 treatment were evaluated and analyzed. The Jeko-1 and A549 xenograft models were used to assess GC7's tumor-suppressing effects. The results showed that GC7 possessed a broad-spectrum anticancer effect, with IC50 values ranging from 0.43 to 1.2 µM in multiple cancer cell lines, which was more potent than gold-based auranofin (∼2-6 folds). GC7 (0.3 and 1 µM) efficiently induced apoptosis of Jeko-1, A549, and HCT116 cells, and it suppressed the sphere formation of cancer stem cells GSC11 and GSC23 cells at 0.1 µM, and it completely eliminated colony at 0.3 µM. The preliminary mechanistic study showed that GC7 inhibited cellular TrxR activity, suppressed mitochondrial OCR, reduced mitochondrial membrane potential (MMP), decreased glucose uptake, and possibly suppressed glycolysis to reduce lactate production. GC7 was predicted to have a similar yet slightly different pharmacokinetic profile as auranofin. Finally, GC7 (20 mg/kg, oral, 5/week, or 3 mg/kg, IP, 3/week) significantly inhibited tumor growth. In conclusion, GC7 showed great potential in suppressing cancer cell proliferation, probably via inhibiting TrxR and impacting mitochondria-mediated energy metabolism.


Asunto(s)
Antineoplásicos , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Metabolismo Energético , Mitocondrias , Humanos , Proliferación Celular/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Metabolismo Energético/efectos de los fármacos , Animales , Ratones , Apoptosis/efectos de los fármacos , Relación Estructura-Actividad , Estructura Molecular , Oro/química , Oro/farmacología , Relación Dosis-Respuesta a Droga , Línea Celular Tumoral , Ratones Desnudos , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Ratones Endogámicos BALB C , Compuestos Orgánicos de Oro/farmacología , Compuestos Orgánicos de Oro/química , Compuestos Orgánicos de Oro/síntesis química
2.
Bioorg Med Chem ; 65: 116761, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35504208

RESUMEN

Survivin, a member of the inhibitor of apoptosis protein family, exists as a homodimer and is aberrantly upregulated in a wide spectrum of cancers. It was thought to be an ideal target due to its lack of expression in most adult normal tissues and importance in cancer cell survival. However, it has been challenging to target survivin due to its "undruggable" nature. We previously attempted to target its dimerization domain with a hypothesis that inhibiting survivin dimerization would promote its degradation in proteasome, which led to identification of a lead small-molecule inhibitor, LQZ-7F. LQZ-7F consists of a flat tetracyclic aromatic core with labile hydrazone linking a 1,2,5-oxadiazole moiety. In this study, we tested the hypothesis that LQZ-7F could be developed as a prodrug because the labile hydrazone linker could be hydrolyzed, releasing the tetracyclic aromatic core. To this end, we synthesized the tetracyclic aromatic core (LQZ-7F1) using reported procedure and tested LQZ-7F1 for its biological activities. Here we show that LQZ-7F1 has a significantly improved potency with submicromolar IC50's and induces spontaneous apoptosis in prostate cancer cells. It also more effectively inhibits survivin dimerization and induces survivin degradation in a proteasome-dependent manner than LQZ-7F. We also show that the combination of LQZ-7F1 and docetaxel have strong synergism in inhibiting prostate cancer cell survival. Together, we conclude that the hydrazone linker with the oxadiazole tail is dispensable for survivin inhibition and the survivin dimerization inhibitor, LQZ-7F, may be developed as a prodrug for prostate cancer treatment and to overcome docetaxel resistance.


Asunto(s)
Profármacos , Neoplasias de la Próstata , Apoptosis , Línea Celular Tumoral , Dimerización , Docetaxel/farmacología , Docetaxel/uso terapéutico , Humanos , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Profármacos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Survivin/metabolismo
3.
Drug Resist Updat ; 54: 100743, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33513557

RESUMEN

ATP-binding cassette (ABC) transporters mediate the ATP-driven translocation of structurally and mechanistically distinct substrates against steep concentration gradients. Among the seven human ABC subfamilies namely ABCA-ABCG, ABCC is the largest subfamily with 13 members. In this respect, 9 of the ABCC members are termed "multidrug resistance proteins" (MRPs1-9) due to their ability to mediate cancer multidrug resistance (MDR) by extruding various chemotherapeutic agents or their metabolites from tumor cells. Furthermore, MRPs are also responsible for the ATP-driven efflux of physiologically important organic anions such as leukotriene C4, folic acid, bile acids and cAMP. Thus, MRPs are involved in important regulatory pathways. Blocking the anticancer drug efflux function of MRPs has shown promising results in overcoming cancer MDR. As a result, many novel MRP modulators have been developed in the past decade. In the current review, we summarize the structure, tissue distribution, biological and pharmacological functions as well as clinical insights of MRPs. Furthermore, recent updates in MRP modulators and their therapeutic applications in clinical trials are also discussed.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/farmacología , Antineoplásicos/farmacología , Resistencia a Antineoplásicos/fisiología , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Transporte Biológico , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos
4.
Int J Mol Sci ; 23(20)2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36293028

RESUMEN

Reactive oxygen species (ROS) homeostasis and mitochondrial metabolism are critical for the survival of cancer cells, including cancer stem cells (CSCs), which often cause drug resistance and cancer relapse. Auranofin is a mono-gold anti-rheumatic drug, and it has been repurposed as an anticancer agent working by the induction of both ROS increase and mitochondrial dysfunction. Hypothetically, increasing auranofin's positive charges via incorporating more gold atoms to enhance its mitochondria-targeting capacity could enhance its anti-cancer efficacy. Hence, in this work, both mono-gold and bi-gold compounds were designed and evaluated to test our hypothesis. The results showed that bi-gold compounds generally suppressed cancer cells proliferation better than their mono-gold counterparts. The most potent compound, BGC2a, substantially inhibited the antioxidant enzyme TrxR and increased the cellular ROS. BGC2a induced cell apoptosis, which could not be reversed by the antioxidant agent vitamin C, implying that the ROS induced by TrxR inhibition might not be the decisive cause of cell death. As expected, a significant proportion of BGC2a accumulated within mitochondria, likely contributing to mitochondrial dysfunction, which was further confirmed by measuring oxygen consumption rate, mitochondrial membrane potential, and ATP production. Moreover, BGC2a inhibited colony formation and reduced stem-like side population (SP) cells of A549. Finally, the compound effectively suppressed the tumor growth of both A549 and PANC-1 xenografts. Our study showed that mitochondrial disturbance may be gold-based compounds' major lethal factor in eradicating cancer cells, providing a new approach to developing potent gold-based anti-cancer drugs by increasing mitochondria-targeting capacity.


Asunto(s)
Antirreumáticos , Neoplasias , Humanos , Especies Reactivas de Oxígeno/metabolismo , Auranofina/farmacología , Antioxidantes/farmacología , Mitocondrias/metabolismo , Apoptosis , Compuestos de Oro , Ácido Ascórbico/farmacología , Antirreumáticos/farmacología , Adenosina Trifosfato/metabolismo , Proliferación Celular , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
5.
Drug Resist Updat ; 48: 100663, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785545

RESUMEN

Drug resistance is a major obstacle in the field of pre-clinical and clinical therapeutics. The development of novel technologies and targeted therapies have yielded new modalities to overcome drug resistance, but multidrug resistance (MDR) remains one of the major challenges in the treatment of cancer. The ubiquitin-proteasome system (UPS) has a central role in regulating the levels and activities of a multitude of proteins as well as regulation of cell cycle, gene expression, response to oxidative stress, cell survival, cell proliferation and apoptosis. Therefore, inhibition of the UPS could represent a novel strategy for the treatment and overcoming of drug resistance in chemoresistant malignancies. In 2003, bortezomib was approved by the FDA for the treatment of multiple myeloma (MM). However, due to its limitations, second generation proteasome inhibitors (PIs) like carfilzomib, ixazomib, oprozomib, delanzomib and marizomib were introduced which displayed clinical activity in bortezomib-resistant tumors. Past studies have demonstrated that proteasome inhibition potentiates the anti-cancer efficacy of other chemotherapeutic drugs by: i) decreasing the expression of anti-apoptotic proteins such as TNF-α and NF-kB, ii) increasing the levels of Noxa, a pro-apoptotic protein, iii) activating caspases and inducing apoptosis, iv) degrading the pro-survival protein, induced myeloid leukemia cell differentiation protein (MCL1), and v) inhibiting drug efflux transporters. In addition, the mechanism of action of the immunoproteasome inhibitors, ONX-0914 and LU-102, suggested their therapeutic role in the combination treatment with PIs. In the current review, we discuss various PIs and their underlying mechanisms in surmounting anti-tumor drug resistance when used in combination with conventional chemotherapeutic agents.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Humanos , Neoplasias/metabolismo , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Transducción de Señal/efectos de los fármacos
6.
Int J Mol Sci ; 22(5)2021 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-33671108

RESUMEN

The overexpression of ATP-binding cassette transporter, ABCG2, plays an important role in mediating multidrug resistance (MDR) in certain types of cancer cells. ABCG2-mediated MDR can significantly attenuate or abrogate the efficacy of anticancer drugs by increasing their efflux from cancer cells. In this study, we determined the efficacy of the novel benzamide derivative, VKNG-2, to overcome MDR due to the overexpression of the ABCG2 transporter in the colon cancer cell line, S1-M1-80. In vitro, 5 µM of VKNG-2 reversed the resistance of S1-M1-80 cell line to mitoxantrone (70-fold increase in efficacy) or SN-38 (112-fold increase in efficacy). In contrast, in vitro, 5 µM of VKNG-2 did not significantly alter either the expression of ABCG2, AKT, and PI3K p110ß protein or the subcellular localization of the ABCG2 protein compared to colon cancer cells incubated with the vehicle. Molecular docking data indicated that VKNG-2 had a high docking score (-10.2 kcal/mol) for the ABCG2 transporter substrate-drug binding site whereas it had a low affinity on ABCB1 and ABCC1 transporters. Finally, VKNG-2 produced a significant concentration-dependent increase in ATPase activity (EC50 = 2.3 µM). In conclusion, our study suggests that in vitro, VKNG-2 reverses the resistance of S1-M1-80, a cancer cell line resistant to mitoxantrone and SN-38, by inhibiting the efflux function of the ABCG2 transporter.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Antineoplásicos/farmacología , Benzamidas/química , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Irinotecán/farmacología , Mitoxantrona/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Humanos , Inhibidores de Topoisomerasa I/farmacología , Células Tumorales Cultivadas
7.
Drug Resist Updat ; 41: 1-25, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30471641

RESUMEN

The successful treatment of cancer has significantly improved as a result of targeted therapy and immunotherapy. However, during chemotherapy, cancer cells evolve and can acquire "multidrug resistance" (MDR), which significantly limits the efficacy of cancer treatment and impacts patient survival and quality of life. Among the approaches to reverse MDR, modulating reactive oxidative species (ROS) may represent a strategy to kill MDR cancer cells that are mechanistically diverse. ROS in cancer cells play a central role in regulating and inducing apoptosis, thereby modulating cancer cells proliferation, survival and drug resistance. The levels of ROS and the activity of scavenging/anti-oxidant enzymes in drug resistant cancer cells are typically increased compared to non-MDR cancer and normal cells. Consequently, MDR cancer cells may be more susceptible to alterations in ROS levels. Numerous studies suggest that compounds modulating cellular ROS levels can enhance MDR cancer cell death and sensitize MDR cancer cells to certain chemotherapeutic drugs. In the current review, we discuss the critical and targetable redox-regulating enzymes, including mitochondrial electron transport chain (ETC) complexes, NADPH oxidases (NOXs), enzymes related to glutathione metabolism, glutamate/cystine antiporter xCT, thioredoxin reductases (TrxRs), nuclear factor erythroid 2-related factor 2 (Nrf2), and their roles in regulating cellular ROS levels, drug resistance as well as their clinical significance. We also discuss and summarize the findings in the past decade regarding the efficacy of ROS modulators for the treatment of MDR cancer alone or as sensitizing compounds. Compounds that are efficacious in modulating ROS generation represent a prominent class of drug candidates that warrants evaluation in clinical trials for patients harboring MDR cancers.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/efectos de los fármacos , Desarrollo de Medicamentos/métodos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Neoplasias/patología , Oxidación-Reducción/efectos de los fármacos , Calidad de Vida
8.
Int J Mol Sci ; 20(17)2019 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-31443367

RESUMEN

P-glycoprotein (P-gp), which is encoded by the ATP-binding cassette (ABC) transporter subfamily B member 1 (ABCB1) gene, is one of the most pivotal ABC transporters that transport its substrates across the cell membrane. Its overexpression is one of the confirmed causes of multidrug resistance (MDR), which results in the failure of cancer treatment. Here, we report that checkpoint kinase (Chk) 1 inhibitor MK-8776, a drug candidate in clinical trial, can restore the sensitivity of chemotherapeutics that are substrates of P-gp in KB-C2, SW620/Ad300 cells and human embryonic kidney (HEK)293/ABCB1 cells that overexpress P-gp. MK-8776 remarkably enhanced the cellular [3H]-paclitaxel accumulation and suppressed the efflux function of P-gp without reducing its expression and affecting its cellular localization in cancer cells. Furthermore, MK-8776 (0-40 µM) stimulated the activity of ATPase in P-gp, which was 4.1-fold greater than the control. In addition, MK-8776 formed a cation-π bond and π-π interaction with key residues of the substrate-binding site in P-gp, as indicated by computer-aided molecular docking study. Our study indicated that MK-8776 may significantly enhance the sensitivity of chemotherapeutics that are substrates of P-gp, providing important information for its application in the reversal of MDR.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Pirazoles/farmacología , Pirimidinas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/química , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/metabolismo , Antineoplásicos/química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Expresión Génica , Humanos , Concentración 50 Inhibidora , Transporte de Proteínas , Pirazoles/química , Pirimidinas/química , Relación Estructura-Actividad
10.
J Cell Biochem ; 117(1): 94-105, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26058377

RESUMEN

Doxorubicin (Dox) is an anthracycline antibiotic widely used in clinics as an anticancer agent. However, the use of Dox is limited by its cardiotoxicity. We have previously shown that a Danshensu (DSS) derivative, ADTM, displayed strong cardioprotective effects. With improved chemical stability and activity, a novel DSS derivative, D006, based on the structure of ADTM, was synthesized. In the present study, the protective effects of D006, indexed by attenuation of the cardiotoxicity induced by Dox as well as chemosensitizing effects that increase the antitumor activity of Dox, were investigated. Our results showed that D006 was more potent than either parental compound, or their use in combination, in ameliorating Dox-induced toxicity in H9c2 cells. In our zebrafish model, D006, but not DSS, alone significantly preserved the ventricular function of zebrafish after Dox treatment. Moreover, D006 upregulated mitochondrial biogenesis and increased mtDNA copy number after Dox treatment of H9c2 cells. D006 promoted the expression of HO-1 protein in a time-dependent manner while the HO-1 inhibitor, Znpp, reversed the protective effects of D006. In human breast tumor MCF-7 cells, D006 enhanced Dox-induced cytotoxicity by increasing apoptosis. In conclusion, our results indicate that a new DSS derivative exhibits promising protective effects against Dox-induced cardiotoxicity both in vivo and in vitro, an effect at least partially mediated by induction of HO-1 expression and the activation of mitochondrial biogenesis. Meanwhile, D006 also potentiated the anti-cancer effects of Dox in breast tumor cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Animales , Antibióticos Antineoplásicos/química , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , ADN Mitocondrial , Femenino , Humanos , Células MCF-7 , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pez Cebra
11.
J Environ Manage ; 132: 313-22, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24333742

RESUMEN

Pavement rehabilitation is carbon intensive and the choice of pavement type is a critical factor in controlling greenhouse gas (GHG) emissions. The existing body of knowledge is not able to support decision-making on pavement choice due to a lack of consensus on the system boundaries, the functional units and the estimation periods. Excessive data requirements further inhibit the generalization of the existing methodologies for design evaluation at the early planning stage. This study proposes a practical life-cycle GHG estimation approach, which is arguably effective to benchmark pavement emissions given project bid tabulation. A set of case studies conducted for this study suggest that recycled asphalt pavement (e.g., foam stabilized base (FSB), and warm mix asphalt (WMA)) would prevent up to 50% of GHGs from the initial construction phase. However, from a life-cycle perspective, pavement emissions are dictated largely by the traffic characteristics and the analysis period for the use phase. The benefits from using recycled materials (e.g., FSB) are likely to diminish if the recycled products do not perform as well as those properly proportioned with less recycled materials, or if the recycled materials are locally unavailable. When the AADT reaches 10,000, use phase releases more than 97% of the life cycle emissions and the emissions difference among alternative designs will be within 1%.


Asunto(s)
Contaminantes Atmosféricos/análisis , Materiales de Construcción/análisis , Monitoreo del Ambiente/métodos , Gases/análisis , Efecto Invernadero , Reciclaje
13.
J Med Chem ; 66(24): 16515-16545, 2023 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-38092421

RESUMEN

Survivin, a homodimeric protein and a member of the IAP family, plays a vital function in cell survival and cycle progression by interacting with various proteins and complexes. Its expression is upregulated in cancers but not detectable in normal tissues. Thus, it has been regarded and validated as an ideal cancer target. However, survivin is "undruggable" due to its lack of enzymatic activities or active sites for small molecules to bind/inhibit. Academic and industrial laboratories have explored different strategies to overcome this hurdle over the past two decades, with some compounds advanced into clinical testing. These strategies include inhibiting survivin expression, its interaction with binding partners and homodimerization. Here, we provide comprehensive analyses of these strategies and perspective on different small molecule survivin inhibitors to help drug discovery targeting "undruggable" proteins in general and survivin specifically with a true survivin inhibitor that will prevail in the foreseeable future.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis , Neoplasias , Humanos , Survivin/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias/metabolismo , Descubrimiento de Drogas , Dimerización , Apoptosis
14.
J Occup Environ Med ; 65(6): 458-466, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36804870

RESUMEN

OBJECTIVE: This study aimed to examine blood benzene, toluene, ethylbenzene, o-xylene, and m-/p-xylene (BTEX) concentrations and their trends contrasting construction workers with workers in other occupations from 1999 to 2014 in the United States. Methods: Using data from the National Health and Nutrition Examination Survey, quantile regressions were performed to investigate associations between occupation and blood BTEX concentrations. Results: We found that high-risk and construction occupations were associated with increased blood concentrations of toluene, o-xylene, and m-/p-xylene at the 50-90th percentiles (P 50-90 ), and ethylbenzene at P 70-90 . Moreover, although blood concentrations of ethylbenzene, o-xylene, and m-/p-xylene trended down among all US workers, no decreasing trend was observed for benzene and toluene among construction workers. Conclusions: Future studies are warranted to address questions about specific tasks to better assess VOC exposure from various trades.


Asunto(s)
Compuestos Orgánicos Volátiles , Humanos , Benceno/análisis , Encuestas Nutricionales , Xilenos , Derivados del Benceno , Tolueno , Monitoreo del Ambiente
15.
Front Chem ; 11: 1292115, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38148758

RESUMEN

Mitochondria are promising drug target for cancer treatment. We previously demonstrated that a bi-gold compound BGC2a was more potent than the mono-gold drug auranofin in suppressing cancer cells due to increased gold atom number that led to higher drug accumulation in and thereby inhibition of mitochondria. To exploit the potential of this new strategy, we further designed and synthesized a series of bi-gold mitocans, the compounds targeting mitochondria. The results showed that most of the newly synthesized mitocans exhibited obviously lower IC50 than auranofin, an old drug that is repurposed in clinical trials for cancer treatment. The best mitocan C3P4 was nearly 2-fold more potent than BGC2a in human non-small cell lung cancer A549 cells and mantle cell lymphoma Jeko-1 cells, exhibiting substantial colony formation-suppressing and tumor-suppressing effects in A549 cells xenograft model. C3P4 induced apoptosis in a dose-dependent manner and arrested cell cycle at G0/G1 phase. The mechanistic study showed that C3P4 significantly increased the global reactive oxygen species and mitochondrial superoxide level, and reduced the mitochondrial membrane potential. C3P4 preferentially accumulated in mitochondria as measured by the gold content and substantially inhibited oxygen consumption rate and ATP production. These results further validated our hypothesis that targeting mitochondria would be promising to develop more potent anticancer agents. C3P4 may be further evaluated as a drug candidate for lung cancer treatment.

16.
Front Pharmacol ; 14: 1235285, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37521473

RESUMEN

Introduction: The overexpression of ATP-binding cassette (ABC) transporters, ABCB1 and ABCG2, are two of the major mediators of multidrug resistance (MDR) in cancers. Although multiple ABCB1 and ABCG2 inhibitors have been developed and some have undergone evaluation in clinical trials, none have been clinically approved. The compound, MK-2206, an inhibitor of the protein kinases AKT1/2/3, is undergoing evaluation in multiple clinical trials for the treatment of certain types of cancers, including those resistant to erlotinib. In this in vitro study, we conducted in vitro experiments to determine if MK-2206 attenuates multidrug resistance in cancer cells overexpressing the ABCB1 or ABCG2 transporter. Methodology: The efficacy of MK-2206 (0.03-1 µM), in combination with the ABCB1 transporter sub-strates doxorubicin and paclitaxel, and ABCG2 transporter substrates mitoxantrone, SN-38 and topotecan, were determined in the cancer cell lines, KB-C2 and SW620/Ad300, which overexpress the ABCB1 transporter or H460/MX20 and S1-M1-80, which overexpress the ABCG2 transporter, respectively. The expression level and the localization of ABCG2 transporter on the cancer cells membranes were determined using western blot and immunofluorescence assays, respectively, following the incubation of cells with MK-2206. Finally, the interaction between MK-2206 and human ABCG2 transporter was predicted using computer-aided molecular modeling. Results: MK-2206 significantly increased the efficacy of anticancer compounds that were substrates for the ABCG2 but not the ABCB1 transporter. MK-2206 alone (0.03-1 µM) did not significantly alter the viability of H460/MX20 and S1-M1-80 cancer cells, which overexpress the ABCG2 transporter, compared to cells incubated with vehicle. However, MK-2206 (0.3 and 1 µM) significantly increased the anticancer efficacy of mitoxantrone, SN-38 and topotecan, in H460/MX20 and S1-M1-80 cancer cells, as indicated by a significant decrease in their IC50 values, compared to cells incubated with vehicle. MK-2206 significantly increased the basal activity of the ABCG2 ATPase (EC50 = 0.46 µM) but did not significantly alter its expression level and sub-localization in the membrane. The molecular modeling results suggested that MK-2206 binds to the active pocket of the ABCG2 transporter, by a hydrogen bond, hydrophobic interactions and π-π stacking. Conclusion: These in vitro data indicated that MK-2206 surmounts resistance to mitoxantrone, SN-38 and topotecan in cancer cells overexpressing the ABCG2 transporter. If these results can be translated to humans, it is possible that MK-2206 could be used to surmount MDR in cancer cells overexpressing the ABCG2 transporter.

17.
Am J Cancer Res ; 13(12): 6026-6037, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38187048

RESUMEN

FN-1501 is a potent FLT3 inhibitor with antitumor activity. A phase 1 trial of FN-1501 monotherapy in patients with advanced solid tumors and R/R AML is in progress. Since one of the primary causes of multidrug resistance (MDR) is the overexpression of ATP-binding cassette superfamily B member 1 (ABCB1), the objective of this study was to investigate the potential relationship between FN-1501 and the ABCB1 transporter. We found ABCB1 overexpressing-cancer cells conferred FN-1501 resistance, which could be reversed by an ABCB1 inhibitor. Molecular docking study revealed that FN-1501 docked the ligand binding site with an affinity score of -9.77 kcal/mol, denoting a strong interaction between FN-1501 and ABCB1. Additionally, the ABCB1 ATPase assay indicated that FN-1501 could significantly stimulate ABCB1 ATPase activity. Furthermore, we observed a similar trend of ABCB1-facilated FN-1501 resistance in tumor-bearing mice model. In sum, we demonstrate that FN-1501 is a substrate of ABCB1 transporter from both in vivo and in vitro studies. Therefore, our findings provide new insight on the mechanism of chemoresistance due to ABCB1 overexpression.

18.
Biochem Pharmacol ; 201: 115061, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35489394

RESUMEN

Drug resistance is the major obstacle that undermines effective cancer treatment. Recently, the application of gas signaling molecules, e.g., carbon monoxide (CO), in overcoming drug resistance has gained significant attention. Growing evidence showed that CO could inhibit mitochondria respiratory effect and glycolysis, two major ATP production pathways in cancer cells, and suppress angiogenesis and inhibit the activity of cystathionine ß-synthase that is important in regulating cancer cells homeostasis, leading to synergistic effects when combined with cisplatin, doxorubicin, or phototherapy, etc. in certain resistant cancer cells. In the current review, we attempted to have a summary of these research conducted in the past decade using CO in treating drug resistant cancers, and have a detailed interpretation of the underlying mechanisms. The critical challenges will be discussed and potential solutions will also be provided. The information collected in this work will hopefully evoke more effects in using CO for the treatment of drug resistant cancers.


Asunto(s)
Monóxido de Carbono , Neoplasias , Monóxido de Carbono/metabolismo , Cisplatino/farmacología , Doxorrubicina/farmacología , Humanos , Mitocondrias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
19.
Front Pharmacol ; 13: 843829, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281897

RESUMEN

The KRAS-G12C inhibitor ARS-1620, is a novel specific covalent inhibitor of KRAS-G12C, possessing a strong targeting inhibitory effect on KRAS-G12C mutant tumors. Overexpression of ATP-binding cassette super-family B member 1 (ABCB1/P-gp) is one of the pivotal factors contributing to multidrug resistance (MDR), and its association with KRAS mutations has been extensively studied. However, the investigations about the connection between the inhibitors of mutant KRAS and the level of ABC transporters are still missing. In this study, we investigated the potential drug resistance mechanism of ARS-1620 associated with ABCB1. The desensitization effect of ARS-1620 was remarkably intensified in both drug-induced ABCB1-overexpressing cancer cells and ABCB1-transfected cells as confirmed by cell viability assay results. This desensitization of ARS-1620 could be completely reversed when co-treated with an ABCB1 reversal agent. In mechanism-based studies, [3H] -paclitaxel accumulation assay revealed that ARS-1620 could be competitively pumped out by ABCB1. Additionally, it was found that ARS-1620 remarkably stimulated ATPase activity of ABCB1, and the HPLC drug accumulation assay displayed that ARS-1620 was actively transported out of ABCB1-overexpressing cancer cells. ARS-1620 acquired a high docking score in computer molecular docking analysis, implying ARS-1620 could intensely interact with ABCB1 transporters. Taken all together, these data indicated that ARS-1620 is a substrate for ABCB1, and the potential influence of ARS-1620-related cancer therapy on ABCB1-overexpressing cancer cells should be considered in future clinical applications.

20.
J Med Chem ; 65(20): 13681-13691, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36257066

RESUMEN

Fatty acid synthase (FASN), a sole cytosolic enzyme responsible for de-novo lipid synthesis, is overexpressed in cancer but not in normal non-lipogenic tissues. FASN has been targeted, albeit no such inhibitor has been approved. Proton pump inhibitors (PPIs), approved for digestive disorders, were found to inhibit FASN with anticancer activities in attempting to repurpose Food and Drug Administration-approved drugs. Indeed, PPI usage benefited breast cancer patients and increased their response rate. Due to structural similarity, we thought that their metabolites might extend anticancer effects of PPIs by inhibiting FASN. Here, we tested this hypothesis and found that 5-hydroxy lansoprazole sulfide (5HLS), the end lansoprazole metabolite, was more active than lansoprazole in inhibiting FASN function and regulation of NHEJ repair of oxidative DNA damage via PARP1. Surprisingly, 5HLS inhibits the enoyl reductase, whereas lansoprazole inhibits the thioesterase of FASN. Thus, PPI metabolites may contribute to the lasting anticancer effects of PPIs by inhibiting FASN.


Asunto(s)
Inhibidores de la Bomba de Protones , Neoplasias de la Mama Triple Negativas , Humanos , Lansoprazol/farmacología , Lansoprazol/uso terapéutico , Inhibidores de la Bomba de Protones/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Oxidorreductasas , Ácido Graso Sintasas/metabolismo , Sulfuros/farmacología , Lípidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA