Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Antimicrob Agents Chemother ; 68(5): e0121123, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38563751

RESUMEN

Helminthiasis remains a public health issue in endemic areas. Various drugs have been proposed to improve efficacy against helminths. The study aimed to assess the safety and efficacy of three different anthelmintic combinations to treat Trichuris trichiura infections. We conducted a randomized assessors-blind clinical trial involving children aged 2-17 years with T. trichiura. Participants were randomly assigned to one of three treatment arms. On the first and third days, all participants got albendazole 400 mg, and on the second day, albendazole (arm A), mebendazole 500 mg (arm B), or pyrantel 125 mg/kg (arm C). We assessed treatment efficacy using the cure rate (CR) and egg reduction rate (ERR) at 3 and 6 weeks post-treatment. At 3 weeks post-treatment, ERR and CR were highest in study arm A [ERR = 94%, 95% confidence interval (CI): 92-95; CR = 71%; 95% CI: 58-81] compared to the B and C arms. Decrease in ERR was significant only for arm B versus arm A (P-value <0.001); decrease in ERR was significant for arms B and C (P-value <0.001). No statistical difference was observed in CR when comparing arms A and B (P-value =1.00) and C (P-value =0.27). At 6 weeks, a decrease in ERR was observed in three arms, significant only for arm C, 81% (95% CI: 78-83). A significant increase in egg counts was observed between 3 and 6 weeks post-treatment. All treatments were safe with mild adverse events. Albendazole 400 mg/day (arm A) showed the highest efficacy against trichuriasis. Nonetheless, this treatment regimen was able to cure half of the treated individuals highlighting concerns about controlling the transmission of T. trichiura.CLINICAL TRIALRegistered at ClinicalTrials.gov (NCT04326868).


Asunto(s)
Albendazol , Antihelmínticos , Mebendazol , Pirantel , Tricuriasis , Trichuris , Humanos , Albendazol/uso terapéutico , Albendazol/efectos adversos , Albendazol/administración & dosificación , Niño , Mebendazol/uso terapéutico , Tricuriasis/tratamiento farmacológico , Masculino , Femenino , Trichuris/efectos de los fármacos , Animales , Preescolar , Antihelmínticos/uso terapéutico , Antihelmínticos/efectos adversos , Antihelmínticos/administración & dosificación , Adolescente , Pirantel/uso terapéutico , Quimioterapia Combinada , Resultado del Tratamiento , Recuento de Huevos de Parásitos
2.
J Neurooncol ; 168(1): 125-138, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38563850

RESUMEN

PURPOSE: Triple-negative breast cancer (TNBC) often metastasizes to the central nervous system (CNS) and has the highest propensity among breast cancer subtypes to develop leptomeningeal disease (LMD). LMD is a spread of cancer into leptomeningeal space that speeds up the disease progression and severely aggravates the prognosis. LMD has limited treatment options. We sought to test whether the common anti-helminthic drug mebendazole (MBZ) may be effective against murine TNBC LMD. METHODS: A small-molecule screen involving TNBC cell lines identified benzimidazoles as potential therapeutic agents for further study. In vitro migration assays were used to evaluate cell migration capacity and the effect of MBZ. For in vivo testing, CNS metastasis was introduced into BALB/c athymic nude mice through internal carotid artery injections of brain-tropic MDA-MB-231-BR or MCF7-BR cells. Tumor growth and spread was monitored by bioluminescence imaging and immunohistochemistry. MBZ was given orally at 50 and 100 mg/kg doses. MBZ bioavailability was assayed by mass spectrometry. RESULTS: Bioinformatic analysis and migration assays revealed higher migratory capacity of TNBC compared to other breast cancer subtypes. MBZ effectively slowed down migration of TNBC cell line MDA-MB-231 and its brain tropic derivative MDA-MB-231-BR. In animal studies, MBZ reduced leptomeningeal spread, and extended survival in brain metastasis model produced by MDA-MB-231-BR cells. MBZ did not have an effect in the non-migratory MCF7-BR model. CONCLUSIONS: We demonstrated that MBZ is a safe and effective oral agent in an animal model of TNBC CNS metastasis. Our findings are concordant with previous efforts involving MBZ and CNS pathology and support the drug's potential utility to slow down leptomeningeal spread.


Asunto(s)
Movimiento Celular , Reposicionamiento de Medicamentos , Mebendazol , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Animales , Humanos , Femenino , Mebendazol/farmacología , Mebendazol/uso terapéutico , Ratones , Movimiento Celular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/secundario , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos
3.
Pediatr Blood Cancer ; 71(4): e30874, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38234020

RESUMEN

BACKGROUND: High-grade gliomas (HGG) have a dismal prognosis despite multimodal therapy. Mebendazole is an anti-helminthic benzimidazole that has demonstrated efficacy in numerous in vitro cancer models, and is able to cross the blood-brain barrier. We conducted a phase 1 trial (NCT01837862) to evaluate the safety of mebendazole in combination with bevacizumab and irinotecan in children and young adults with HGG. OBJECTIVE: To determine the maximally tolerated dose of mebendazole when given in combination with bevacizumab and irinotecan in children with HGG; to describe the progression-free survival (PFS) and overall survival (OS) for this group. DESIGN/METHOD: Patients between 1 and 21 years of age with HGG were enrolled in a 3 + 3 design to escalating doses of mebendazole in combination with bevacizumab (10 mg/kg/dose) and irinotecan (150 mg/m2 /dose). Subjects were eligible upfront after completion of radiation or at the time of progression. Mebendazole was taken orally twice per day continuously, and bevacizumab and irinotecan were given intravenously on Days 1 and 15 of 28-day cycles. RESULTS: Between 2015 and 2020, 10 subjects were enrolled at mebendazole doses of 50 mg/kg/day (n = 3), 100 mg/kg/day (n = 4), and 200 mg/kg/day (n = 3). One subject assigned to 100 mg/kg/day was not evaluable. Seven subjects had a diagnosis of diffuse midline glioma, one subject had anaplastic astrocytoma, and one subject had a spinal HGG. All subjects received radiation. There were no dose-limiting toxicities. The most frequent G3/4 adverse events were neutropenia (n = 3) and lymphopenia (n = 4). The overall response rate was 33%, with two subjects achieving a partial response and one subject achieving a complete response sustained for 10 months. The mean PFS and OS from the start of study treatment were 4.7 and 11.4 months, respectively. CONCLUSION: Mebendazole was safe and well tolerated when administered with bevacizumab and irinotecan at doses up to 200 mg/kg/day. Further studies are needed to determine the efficacy of this treatment.


Asunto(s)
Glioma , Mebendazol , Niño , Adulto Joven , Humanos , Bevacizumab , Irinotecán/efectos adversos , Mebendazol/efectos adversos , Camptotecina/efectos adversos , Glioma/tratamiento farmacológico
4.
Int J Mol Sci ; 24(2)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36674870

RESUMEN

Repurposing approved non-antitumor drugs is a promising and affordable strategy in drug discovery to identify new therapeutic uses different from the original medical indication that may help increase the number of possible, effective anticancer drugs. The use of drugs in ways other than their original FDA-approved indications could offer novel avenues such as bypassing the chemoresistance and recurrence seen with conventional therapy and treatment; moreover, it can offer a safe and economic strategy for combination therapy. Recent works have demonstrated the anticancer properties of the FDA-approved drug Mebendazole. This synthetic benzimidazole proved effective against a broad spectrum of intestinal Helminthiasis. Mebendazole can penetrate the blood-brain barrier and has been shown to inhibit the malignant progression of glioma by targeting signaling pathways related to cell proliferation, apoptosis, or invasion/migration, or by increasing the sensitivity of glioma cells to conventional chemotherapy or radiotherapy. Moreover, several preclinical models and ongoing clinical trials explore the efficacy of Mebendazole in multiple cancers, including acute myeloid leukemia, brain cancer, oropharyngeal squamous cell carcinoma, breast cancer, gastrointestinal cancer, lung carcinoma, adrenocortical carcinoma, prostate cancer, and head and neck cancer. The present review summarizes central literature regarding the anticancer effects of MBZ in cancer cell lines, animal tumor models, and clinical trials to suggest possible strategies for safe and economical combinations of anticancer therapies in brain cancer. Mebendazole might be an excellent candidate for the treatment of brain tumors because of its efficacy both when used as monotherapy and in combination as an enhancement to standard chemotherapeutics and radiotherapy, due to its effectiveness on tumor angiogenesis inhibition, cell cycle arrest, apoptosis induction, and targeting of critical pathways involved in cancer such as Hedgehog signaling. Therefore, attention to MBZ repurposing has recently increased because of its potential therapeutic versatility and significant clinical implications, such as reducing medical care costs and optimizing existing therapies. Using new treatments is essential, particularly when current therapeutics for patients with brain cancer fail.


Asunto(s)
Antiinfecciosos , Antineoplásicos , Neoplasias Encefálicas , Glioma , Neoplasias de Cabeza y Cuello , Masculino , Animales , Mebendazol/farmacología , Mebendazol/uso terapéutico , Antiparasitarios , Línea Celular Tumoral , Proteínas Hedgehog , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Antiinfecciosos/uso terapéutico , Glioma/tratamiento farmacológico
5.
Breast Cancer Res ; 24(1): 98, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36578038

RESUMEN

Breast cancer is the most diagnosed cancer among women. Approximately 15-20% of all breast cancers are highly invasive triple-negative breast cancer (TNBC) and lack estrogen, progesterone, and ERBB2 receptors. TNBC is challenging to treat due to its aggressive nature with far fewer targeted therapies than other breast cancer subtypes. Current treatments for patients with TNBC consist of cytotoxic chemotherapies, surgery, radiation, and in some instances PARP inhibitors and immunotherapy. To advance current therapeutics, we repurposed mebendazole (MBZ), an orally available FDA-approved anthelmintic that has shown preclinical efficacy for cancers. MBZ has low toxicity in humans and efficacy in multiple cancer models including breast cancer, glioblastoma multiforme, medulloblastoma, colon cancer, pancreatic and thyroid cancer. MBZ was well-tolerated in a phase I clinical trial of adults recently diagnosed with glioma. We determined that the half-maximal inhibitory concentration (IC50) of MBZ in four breast cancer cell lines is well within the range reported for other types of cancer. MBZ reduced TNBC cell proliferation, induced apoptosis, and caused G2/M cell cycle arrest. MBZ reduced the size of primary tumors and prevented lung and liver metastases. In addition, we uncovered a novel mechanism of action for MBZ. We found that MBZ reduces integrin ß4 (ITGß4) expression and cancer stem cell properties. ITGß4 has previously been implicated in promoting "cancer stemness," which may contribute to the efficacy of MBZ. Collectively, our results contribute to a growing body of evidence suggesting that MBZ should be considered as a therapeutic to slow tumor progression and prevent metastasis.


Asunto(s)
Mebendazol , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Mebendazol/farmacología , Mebendazol/uso terapéutico , Integrina beta4 , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Línea Celular Tumoral
6.
Chemistry ; 28(6): e202103589, 2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-34962330

RESUMEN

Among all possible NMR crystallography approaches for crystal-structure determination, crystal structure prediction - NMR crystallography (CSP-NMRX) has recently turned out to be a powerful method. In the latter, the original procedure exploited solid-state NMR (SSNMR) information during the final steps of the prediction. In particular, it used the comparison of computed and experimental chemical shifts for the selection of the correct crystal packing. Still, the prediction procedure, generally carried out with DFT methods, may require important computational resources and be quite time-consuming, especially if there are no available constraints to use at the initial stage. Herein, the successful application of this combined prediction method, which exploits NMR information also in the input step to reduce the search space of the predictive algorithm, is presented. Herein, this method was applied on mebendazole, which is characterized by desmotropism. The use of SSNMR data as constraints for the selection of the right tautomer and the determination of the number of independent molecules in the unit cell led to a considerably faster process, reducing the number of calculations to be performed. In this way, the crystal packing was successfully predicted for the three known phases of mebendazole. To evaluate the quality of the predicted structures, these were compared to the experimental ones. The crystal structure of phase B of mebendazole, in particular, was determined de novo by powder diffraction and is presented for the first time in this paper.


Asunto(s)
Imagen por Resonancia Magnética , Mebendazol , Cristalografía por Rayos X , Espectroscopía de Resonancia Magnética , Modelos Moleculares
7.
BMC Pregnancy Childbirth ; 22(1): 60, 2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35065622

RESUMEN

BACKGROUND: The use of deworming drugs is one of the important antenatal strategies in preventing anaemia in pregnancy. Little is known about the factors associated with the use of deworming drugs, which accounts for the aim of this study. METHOD: The study used data from the 2015-16 Tanzania HIV Demographic and Health Survey and Malaria Indicators Survey (2015-16 TDHS-MIS). A total of 6924 women of active reproductive age from 15 to 49 were included in the analysis. Both univariate and multiple logistic regression analyses were used. RESULTS: The majority of interviewed women 3864(60.1%) took deworming drugs. In a weighed multiple logistic regression, women residing in urban areas reported greater use of deworming drugs than women residing in rural areas [AOR = 1.73, p = 0.01, 95% CI (1.26-2.38)]. In the four areas of residence, compared to women residing in mainland rural areas, women residing in mainland urban areas and Pemba islands reported greater use of deworming drugs [mainland urban (AOR = 2.56 p < 0.001,95% CI(1.78-3.75), and Pemba Island (AOR = 1.18, p < 0.001, 95% CI(1.17-1.20)]. However, women residing in Zanzibar Island (Unguja) were less likely to use deworming drugs compared to women in mainland rural women (AOR = 0.5, p < 0.001, 95% CI (0.45-0.55). Similarly, compared to women under 20 years of age, women between 20 to 34 years reported significantly greater use of deworming drugs [20 to 34 years (AOR = 1.30, p = 0.03, 95% CI(1.02-1.65). Likewise, greater use of deworming drugs was reported in women with a higher level of education compared to no education [higher education level (AOR = 3.25, p = 0.01,95% CI(1.94-7.92)], rich women compared to poor [rich (AOR = 1.43, p = 0.003, 95% CI (1.13-1.80)] and in women who initiated antenatal care on their first trimester of pregnancy compared to those who initiated later [AOR = 1.37, p < 0.001, 95% CI (1.17-1.61)]. CONCLUSION: Women who were more likely to use the deworming drugs were those residing in urban compared to rural areas, aged between 20 and 34 years, those with a higher level of formal education, wealthier, and women who book the antenatal clinic (ANC) within their first trimester of pregnancy. Considering the outcomes of anaemia in pregnancy, a well-directed effort is needed to improve the use of deworming drugs.


Asunto(s)
Anemia Ferropénica/prevención & control , Antiparasitarios/uso terapéutico , Mebendazol/uso terapéutico , Cooperación del Paciente , Complicaciones Parasitarias del Embarazo/prevención & control , Atención Prenatal , Adolescente , Adulto , Femenino , Humanos , Persona de Mediana Edad , Embarazo , Determinantes Sociales de la Salud , Factores Sociodemográficos , Encuestas y Cuestionarios , Tanzanía/epidemiología , Adulto Joven
8.
J Nanobiotechnology ; 20(1): 169, 2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35361226

RESUMEN

BACKGROUND: Mebendazole (MBZ) is a well-known anti-parasite drug with significant anti-cancer properties. However, MBZ exhibits low solubility, limited absorption efficacy, extensive first-pass effect, and low bioavailability. Therefore, multiple oral administration of high dose MBZ is required daily for achieving the therapeutic serum level which can cause severe side effects and patients' non-compliance. METHOD: In the present study, MBZ-loaded/folic acid-targeted chitosan nanoparticles (CS-FA-MBZ) were synthesized, characterized, and used to form cylindrical subcutaneous implants for 4T1 triple-negative breast tumor (TNBC) treatment in BALB/c mice. The therapeutic efficacy of the CS-FA-MBZ implants was investigated after subcutaneous implantation in comparison with Control, MBZ (40 mg/kg, oral administration, twice a week for 2 weeks), and CS-FA implants, according to 4T1 tumors' growth progression, metastasis, and tumor-bearing mice survival time. Also, their biocompatibility was evaluated by blood biochemical analyzes and histopathological investigation of vital organs. RESULTS: The CS-FA-MBZ implants were completely degraded 15 days after implantation and caused about 73.3%, 49.2%, 57.4% decrease in the mean tumors' volume in comparison with the Control (1050.5 ± 120.7 mm3), MBZ (552.4 ± 76.1 mm3), and CS-FA (658.3 ± 88.1 mm3) groups, respectively. Average liver metastatic colonies' number per microscope field at the CS-FA-MBZ group (2.3 ± 0.7) was significantly (P < 0.05) lower than the Control (9.6 ± 1.7), MBZ (5.0 ± 1.5), and CS-FA (5.2 ± 1) groups. In addition, the CS-FA-MBZ treated mice exhibited about 52.1%, 27.3%, and 17% more survival days after the cancer cells injection in comparison with the Control, MBZ, and CS-FA groups, respectively. Moreover, the CS-FA-MBZ implants were completely biocompatible based on histopathology and blood biochemical analyzes. CONCLUSION: Taking together, CS-FA-MBZ implants were completely biodegradable and biocompatible with high therapeutic efficacy in a murine TNBC model.


Asunto(s)
Quitosano , Nanopartículas , Neoplasias de la Mama Triple Negativas , Animales , Quitosano/química , Ácido Fólico/química , Humanos , Concentración de Iones de Hidrógeno , Mebendazol/química , Mebendazol/farmacología , Ratones , Nanopartículas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
9.
Int J Mol Sci ; 23(24)2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36555137

RESUMEN

Breast cancer is the most commonly diagnosed cancer worldwide and ranks first in terms of both prevalence and cancer-related mortality in women. In this study, we aimed to evaluate the anticancer effect of mebendazole (MBZ) and radiotherapy (RT) concomitant use in triple-negative breast cancer (TNBC) cells and elucidate the underlying mechanisms of action. Breast cancer mouse models and several types of breast cancer cells, including TNBC-derived RT-resistant (RT-R) MDA-MB-231 cells, were treated with MBZ and/or RT. In mice, changes in body weight, renal and liver toxicity, tumor volume, and number of lung metastases were determined. In cells, cell viability, colony formation, scratch wound healing, Matrigel invasion, and protein expression using western blotting were determined. Our findings showed that MBZ and RT combined treatment increased the anticancer effect of RT without additional toxicity. In addition, we noted that cyclin B1, PH2AX, and natural killer (NK) cell-mediated cytotoxicity increased following MBZ + RT treatment compared to unaided RT. Our results suggest that MBZ + RT have an enhanced anticancer effect in TNBC which acquires radiation resistance through blocking cell cycle progression, initiating DNA double-strand breaks, and promoting NK cell-mediated cytotoxicity.


Asunto(s)
Mebendazol , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Ratones , Animales , Mebendazol/farmacología , Mebendazol/uso terapéutico , Línea Celular Tumoral , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/radioterapia , Neoplasias de la Mama Triple Negativas/patología , Apoptosis , Células Asesinas Naturales , Proliferación Celular
10.
Int J Mol Sci ; 23(8)2022 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-35457043

RESUMEN

Mebendazole (MBZ) is a synthetic benzimidazole known for its antiparasitic properties. In recent years, growing evidence showed that MBZ was also used as an anti-tumor agent. However, whether (and to what extent) this drug treatment affected the male reproductive system was not well-understood. In this study, male C57BL/6 mice were injected with 40 mg/kg/day of MBZ. The treatment was for 3 and 7 days. Our results showed that the injected mice exhibited an abnormal spermatogenic phase with a significant decrease in sperm. We further detected microtubule disruption and transient functional destruction of the blood-testes barrier (BTB) in the MBZ-injected mice testes (BTB). Our data confirmed that MBZ suppressed the expression of the BTB junction-associated proteins and disrupted the Sertoli cells' function in vivo. Moreover, MBZ-treated mice demonstrated an aberrant caspase-3 signalling pathway, which resulted in the apoptosis of the germ cells. Here, we present our data, indicating that MBZ impairs BTB by reducing the expression of the microtubules' and BTB junction-associated proteins. The last leads to activating the caspase-3 pathway, which triggers extensive germ cell apoptosis.


Asunto(s)
Barrera Hematotesticular , Mebendazol , Animales , Apoptosis , Barrera Hematotesticular/metabolismo , Caspasa 3/metabolismo , Masculino , Mebendazol/farmacología , Ratones , Ratones Endogámicos C57BL , Microtúbulos , Células de Sertoli/metabolismo , Testículo
11.
Int J Mol Sci ; 23(7)2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35408808

RESUMEN

Microtubule targeting agents (MTAs) have been exploited mainly as anti-cancer drugs because of their impact on cellular division and angiogenesis. Additionally, microtubules (MTs) are key structures for intracellular transport, which is frequently hijacked during viral infection. We have analyzed the antiviral activity of clinically used MTAs in the infection of DNA and RNA viruses, including SARS-CoV-2, to find that MT destabilizer agents show a higher impact than stabilizers in the viral infections tested, and FDA-approved anti-helminthic benzimidazoles were among the most active compounds. In order to understand the reasons for the observed antiviral activity, we studied the impact of these compounds in motor proteins-mediated intracellular transport. To do so, we used labeled peptide tools, finding that clinically available MTAs impaired the movement linked to MT motors in living cells. However, their effect on viral infection lacked a clear correlation to their effect in motor-mediated transport, denoting the complex use of the cytoskeleton by viruses. Finally, we further delved into the molecular mechanism of action of Mebendazole by combining biochemical and structural studies to obtain crystallographic high-resolution information of the Mebendazole-tubulin complex, which provided insights into the mechanisms of differential toxicity between helminths and mammalians.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Mebendazol , Animales , Antivirales/farmacología , Mamíferos , Mebendazol/farmacología , Microtúbulos , SARS-CoV-2 , Tubulina (Proteína)
12.
Molecules ; 27(23)2022 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-36500220

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) has one of the highest mortality rates and requires the development of highly efficacious medications that can improve the efficiency of existing treatment methods. In particular, in PDAC, resistance to conventional chemotherapy reduces the effectiveness of anticancer drugs, decreasing the therapeutic efficiency. Sphingosine 1-phosphate (S1P), produced by sphingosine kinase (SK), plays a vital role in cancer growth, metastasis, chemotherapy, and drug resistance. Focusing on the structural characteristics of mebendazole (MBZ), we studied whether MBZ would affect metastasis, invasion, and drug resistance in cancer by lowering S1P production through inhibition of SK activity. MBZ selectively inhibited SK1 more than SK2 and regulated the levels of sphingolipids. MBZ inhibited the proliferation and migration of cancer cells in other PDAC cell lines. To determine whether the effect of MBZ on cancer cell growth and migration is S1P-mediated, S1P was treated, and the growth and migration of cancer cells were observed. It was found that MBZ inhibited S1P-induced cancer cell growth, and MBZ showed a growth inhibitory effect by regulating the JAK2/STAT3/Bcl-2 pathway. The phosphorylation of focal adhesion kinase (FAK), a transcription factor that regulates migration, was inhibited by MBZ, so it was found that the effect of MBZ regulates the migration of cancer cells through the S1P/FAK/vimentin pathway. In conclusion, our study suggests that the anthelmintic MBZ can be used as a potential therapeutic agent for treating PDAC and for structural synthesis studies of its analogs.


Asunto(s)
Lisofosfolípidos , Neoplasias Pancreáticas , Humanos , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina , Mebendazol/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Proliferación Celular , Neoplasias Pancreáticas
13.
J Environ Sci Health B ; 57(11): 883-889, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36217592

RESUMEN

The heterologous strategy could improve the sensitivity of competitive enzyme-linked immunosorbent assay (ELISA) for detection of chemical contaminants in food samples. In this study, the heterologous coating antigen ELISA was developed to evaluate its sensitivity for mebendazole (MBZ). Results showed that the heterologous ELISA had a linear range of (IC20-IC80) 0.34-10.54 ng/mL, an IC50 value of 1.83 ng/mL, and a limit of detection (LOD) of 0.13 ng/mL, in which the sensitivity of ELISA improved 1.7- and 2-fold (IC50 value dropping from 7.41 and 3.65 ng/mL to 4.27 and 1.83 ng/mL) than that of rabbit IgG- and chicken IgY-based homologous ELISA for MBZ, respectively. The heterologous coating antigen ELISA showed negligible cross reactivity (<0.2%) with its structural analogues, including hydroxy-MBZ, albendazole, oxfendazole, fenbendazole, and flubendazole, except the value of 72.6% for amino-MBZ. The average recoveries of MBZ spiked in pork and chicken muscle samples by the assay ranged from 83.7% to 109.8% and agreed well with those of high-performance liquid chromatography. The results suggested that using heterologous coating antigen could distinctly improve the sensitivity of ELISA for routine screening of MBZ residues in food samples.


Asunto(s)
Antígenos Heterófilos , Mebendazol , Animales , Conejos , Antígenos Heterófilos/análisis , Ensayo de Inmunoadsorción Enzimática/métodos , Cromatografía Líquida de Alta Presión , Límite de Detección
14.
J Environ Sci Health B ; 57(1): 47-53, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34978273

RESUMEN

Chicken egg yolk IgY has proven to be qualified for analysis of targets in immunoassays. In order to explore the feasibility of chicken IgY-based ELISA for detection of mebendazole (MEB), the chicken IgY against MEB was generated in the laying hens. An enzyme-linked immunosorbent assay (ELISA) based on chicken IgY was developed for detection of MEB with a half-maximum signal inhibition concentration (IC50) of 3.65 ng mL-1 and a limit of detection of 0.25 ng mL-1. The assay showed a lower cross reactivity (less than 1%) with other structures analogues (except amino-MEB with the values of 70.7%). The average recoveries of MEB spiked in pork and mutton muscle samples ranged from 93.6% to 106.3% with relative standard deviation less than 8.78% and 10.85% for intra-assay and inter-assay, respectively, and agreed well with those of high-performance liquid chromatography. Our results indicate that generated IgY could be used as a robust reagent for routine screening analysis of small molecular compounds residues in food samples.


Asunto(s)
Carne de Cerdo , Carne Roja , Animales , Pollos , Yema de Huevo/química , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Inmunoglobulinas/análisis , Mebendazol/análisis , Porcinos
15.
Vet Med (Praha) ; 67(12): 620-627, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38845786

RESUMEN

The study aimed to assess the effects of a therapeutic bath of five different antiparasitic drugs, in different baths and durations: fenbendazole (25 mg l-1, 12 h and 2 × 12 h), formaldehyde (0.17 ml l-1, 15 min), ivermectin (0.031 mg l-1, 1 h), mebendazole (1 mg l-1, 12 h) and levamisole (50 mg l-1, 2 h and 3 × 1 h) on the reduction on the intensity and prevalence of a monogenean infection (Dactylogyrus anchoratus) in juvenile carp. The best effect on reducing the parasite number was achieved with the bath in formaldehyde (0.17 ml l-1, 15 min) and fenbendazole (25 mg l-1, 2 × 12 h with 24 h break), where the infection was reduced by more than 90%. Registered veterinary medicinal products (VMPs) with the active substance of fenbendazole can successfully replace the use of unregistered formaldehyde in the treatment of monogenean infections.

16.
Medicina (Kaunas) ; 58(9)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36143915

RESUMEN

Over the last years, repurposed agents have provided growing evidence of fast implementation in oncology treatment such as certain antimalarial, anthelmintic, antibiotics, anti-inflammatory, antihypertensive, antihyperlipidemic, antidiabetic agents. In this study, the four agents of choice were present in our patients' daily treatment for nonmalignant-associated pathology and have known, light toxicity profiles. It is quite common for a given patient's daily administration schedule to include two or three of these drugs for the duration of their treatment. We chose to review the latest literature concerning metformin, employed as a first-line treatment for type 2 diabetes; mebendazole, as an anthelmintic; atorvastatin, as a cholesterol-lowering drug; propranolol, used in cardiovascular diseases as a nonspecific inhibitor of beta-1 and beta-2 adrenergic receptors. At the same time, certain key action mechanisms make them feasible antitumor agents such as for mitochondrial ETC inhibition, activation of the enzyme adenosine monophosphate-activated protein kinase, amelioration of endogenous hyperinsulinemia, inhibition of selective tyrosine kinases (i.e., VEGFR2, TNIK, and BRAF), and mevalonate pathway inhibition. Despite the abundance of results from in vitro and in vivo studies, the only solid data from randomized clinical trials confirm metformin-related oncological benefits for only a small subset of nondiabetic patients with HER2-positive breast cancer and early-stage colorectal cancer. At the same time, clinical studies confirm metformin-related detrimental/lack of an effect for lung, breast, prostate cancer, and glioblastoma. For atorvastatin we see a clinical oncological benefit in patients and head and neck cancer, with a trend towards radioprotection of critical structures, thus supporting the role of atorvastatin as a promising agent for concomitant association with radiotherapy. Propranolol-related increased outcomes were seen in clinical studies in patients with melanoma, breast cancer, and sarcoma.


Asunto(s)
Antihelmínticos , Antimaláricos , Antineoplásicos , Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Metformina , Adenosina Monofosfato/uso terapéutico , Antagonistas Adrenérgicos beta/uso terapéutico , Antihelmínticos/uso terapéutico , Antibacterianos/uso terapéutico , Antihipertensivos/uso terapéutico , Antimaláricos/uso terapéutico , Antineoplásicos/uso terapéutico , Atorvastatina/uso terapéutico , Neoplasias de la Mama/patología , Colesterol , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipoglucemiantes/uso terapéutico , Masculino , Mebendazol/uso terapéutico , Metformina/uso terapéutico , Ácido Mevalónico/uso terapéutico , Propranolol/uso terapéutico , Proteínas Quinasas/metabolismo , Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf , Receptores Adrenérgicos beta 2/uso terapéutico , Tirosina
17.
Gynecol Oncol ; 160(1): 302-311, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33131904

RESUMEN

OBJECTIVE: Mebendazole and other anti-parasitic drugs are being used off-prescription based on social media and unofficial accounts of their anti-cancer activity. The purpose of this study was to conduct a controlled evaluation of mebendazole's therapeutic efficacy in cell culture and in vivo models of ovarian cancer. The majority of ovarian cancers harbor p53 null or missense mutations, therefore the effects of p53 mutations and a mutant p53 reactivator, PRIMA-1MET (APR246) on mebendazole activity were evaluated. METHODS: Mebendazole was evaluated in cisplatin-resistant high grade serous stage 3C ovarian cancer patient derived xenograft (PDX) models: PDX-0003 (p53 null) and PDX-0030 (p53 positive), and on ovarian cancer cell lines: MES-OV (p53 R282W), ES2 (p53 S241F), A2780 (p53 wild type), SKOV3 parental (p53 null) and isogenic sublines, SKOV3 R273H p53 and SKOV3 R248W p53. Drug synergy and mechanisms were evaluated in cell cultures using isobolograms, clonogenic assays and western blots. Prevention of tumor establishment was studied in a MES-OV orthotopic model. RESULTS: Mebendazole inhibited growth of ovarian cancer cell cultures at nanomolar concentrations and PDXs at doses up to 50 mg/kg, and reduced orthotopic tumor establishment at 50 mg/kg. The mechanism of mebendazole was associated with p53-independent induction of p21 and tubule depolymerization. PRIMA-1MET also inhibited tumor establishment and worked synergistically with mebendazole in cell culture to inhibit growth and induce intrinsic apoptosis through a p53- and tubule destabilization-independent mechanism. CONCLUSION: This work demonstrates the therapeutic potential of repurposing mebendazole and supports clinical development of mebendazole for ovarian cancer therapy and maintenance.


Asunto(s)
Mebendazol/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Reposicionamiento de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Fenbendazol/farmacología , Humanos , Mebendazol/administración & dosificación , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Quinuclidinas/administración & dosificación , Quinuclidinas/farmacología , Distribución Aleatoria , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Parasitology ; 148(5): 630-635, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33517933

RESUMEN

The present work aimed at studying the efficacy of mebendazole (MBZ) compared to artemisinin (ART) for the treatment of trichinellosis at various phases of infection. Seventy Swiss albino mice were orally infected by 300 Trichinella spiralis (T. spiralis) larvae. Mice were divided into infected untreated control group and infected groups treated with 50 mg kg-1 MBZ and 300 mg kg-1 ART for three and five consecutive days, respectively, at the enteral phase [2-4 days post infection (PI)], invasive phase (10-12 days PI) and encapsulated phase (28-30 days PI). All mice were sacrificed 35-42 days PI. MBZ and ART revealed a significant decrease in mean larval counts and increase of larval per cent reduction (LR %) when treatment was initiated during the enteral phase compared to the other phases. MBZ showed significantly higher LR % (99.7, 83.95 and 89.65%) than ART (80.58, 67.0 and 79.2%) when administered at the three infection phases. Histopathological study showed a decrease in the number of encysted larvae, their surrounding cellular infiltrates and increased regenerative muscles in all treated mice. In conclusion, ART possesses a substantial anthelmintic activity against T. spiralis infection in mice both at the enteral and encapsulated phases, yet, significantly lower than MBZ.


Asunto(s)
Antihelmínticos/farmacología , Artemisininas/farmacología , Mebendazol/farmacología , Trichinella spiralis/efectos de los fármacos , Triquinelosis/tratamiento farmacológico , Animales , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Ratones , Trichinella spiralis/crecimiento & desarrollo , Triquinelosis/parasitología
19.
Int J Mol Sci ; 22(19)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34639011

RESUMEN

The prognosis of elderly AML patients is still poor due to chemotherapy resistance. The Hedgehog (HH) pathway is important for leukemic transformation because of aberrant activation of GLI transcription factors. MBZ is a well-tolerated anthelmintic that exhibits strong antitumor effects. Herein, we show that MBZ induced strong, dose-dependent anti-leukemic effects on AML cells, including the sensitization of AML cells to chemotherapy with cytarabine. MBZ strongly reduced intracellular protein levels of GLI1/GLI2 transcription factors. Consequently, MBZ reduced the GLI promoter activity as observed in luciferase-based reporter assays in AML cell lines. Further analysis revealed that MBZ mediates its anti-leukemic effects by promoting the proteasomal degradation of GLI transcription factors via inhibition of HSP70/90 chaperone activity. Extensive molecular dynamics simulations were performed on the MBZ-HSP90 complex, showing a stable binding interaction at the ATP binding site. Importantly, two patients with refractory AML were treated with MBZ in an off-label setting and MBZ effectively reduced the GLI signaling activity in a modified plasma inhibitory assay, resulting in a decrease in peripheral blood blast counts in one patient. Our data prove that MBZ is an effective GLI inhibitor that should be evaluated in combination to conventional chemotherapy in the clinical setting.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Mebendazol/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Moduladores de Tubulina/farmacología , Proteína con Dedos de Zinc GLI1/metabolismo , Estudios de Casos y Controles , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Proteolisis , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Proteína con Dedos de Zinc GLI1/química
20.
Molecules ; 26(17)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34500557

RESUMEN

In this study, we aimed to evaluate the anticancer effect of benzimidazole derivatives on triple-negative breast cancer (TNBC) and investigate its underlying mechanism of action. Several types of cancer and normal breast cells including MDA-MB-231, radiotherapy-resistant (RT-R) MDA-MB-231, and allograft mice were treated with six benzimidazole derivatives including mebendazole (MBZ). Cells were analyzed for viability, colony formation, scratch wound healing, Matrigel invasion, cell cycle, tubulin polymerization, and protein expression by using Western blotting. In mice, liver and kidney toxicity, changes in body weight and tumor volume, and incidence of lung metastasis were analyzed. Our study showed that MBZ significantly induced DNA damage, cell cycle arrest, and downregulation of cancer stem cell markers CD44 and OCT3/4, and cancer progression-related ESM-1 protein expression in TNBC and RT-R-TNBC cells. In conclusion, MBZ has the potential to be an effective anticancer agent that can overcome treatment resistance in TNBC.


Asunto(s)
Antineoplásicos/farmacología , Bencimidazoles/farmacología , Mebendazol/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Biomarcadores de Tumor/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Pulmonares/metabolismo , Células MCF-7 , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda