1
|
Fatima I, Ahmad R, Barman S, Gowrikumar S, Pravoverov K, Primeaux M, Fisher KW, Singh AB, Dhawan P. Albendazole inhibits colon cancer progression and therapy resistance by targeting ubiquitin ligase RNF20. Br J Cancer 2024; 130:1046-1058. [PMID: 38278978 PMCID: PMC10951408 DOI: 10.1038/s41416-023-02570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND The repurposing of FDA-approved drugs for anti-cancer therapies is appealing due to their established safety profiles and pharmacokinetic properties and can be quickly moved into clinical trials. Cancer progression and resistance to conventional chemotherapy remain the key hurdles in improving the clinical management of colon cancer patients and associated mortality. METHODS High-throughput screening (HTS) was performed using an annotated library of 1,600 FDA-approved drugs to identify drugs with strong anti-CRC properties. The candidate drug exhibiting most promising inhibitory effects in in-vitro studies was tested for its efficacy using in-vivo models of CRC progression and chemoresistance and patient derived organoids (PTDOs). RESULTS Albendazole, an anti-helminth drug, demonstrated the strongest inhibitory effects on the tumorigenic potentials of CRC cells, xenograft tumor growth and organoids from mice. Also, albendazole sensitized the chemoresistant CRC cells to 5-fluorouracil (5-FU) and oxaliplatin suggesting potential to treat chemoresistant CRC. Mechanistically, Albendazole treatment modulated the expression of RNF20, to promote apoptosis in CRC cells by delaying the G2/M phase and suppressing anti-apoptotic-Bcl2 family transcription. CONCLUSIONS Albendazole, an FDA approved drug, carries strong therapeutic potential to treat colon cancers which are aggressive and potentially resistant to conventional chemotherapeutic agents. Our findings also lay the groundwork for further clinical testing.
Collapse
Affiliation(s)
- Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kristina Pravoverov
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kurt W Fisher
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
2
|
Yanke AB, Day KE, Taylor AR, Cruz-Espindola C, Boothe DM. Pharmacokinetics of mebendazole in plasma and cerebrospinal fluid following a single oral dose in healthy dogs. Front Vet Sci 2023; 10:1231769. [PMID: 37701529 PMCID: PMC10493385 DOI: 10.3389/fvets.2023.1231769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023] Open
Abstract
Novel therapies are needed for treatment of gliomas. Mebendazole previously demonstrated anti-neoplastic effects on canine glioma cell lines at in vitro mean inhibitory concentrations (IC50) of 10 ng/mL. Our study aimed to titrate the oral dose of mebendazole necessary to achieve concentrations ≥10 ng/mL in cerebrospinal fluid (CSF) of healthy dogs. We hypothesized that an oral dose up to 200 mg/kg would be necessary. Phase one was a dose titration study using a total of 6 mixed breed dogs that described dose vs. plasma concentrations for 72 h after single oral dosing of either 50 mg/kg (n = 2), 100 mg/kg (n = 2), or 200 mg/kg (n = 2). Based on phase one, phase two dogs (total of 9) received 100 mg/kg (n = 4) or 200 mg/kg (n = 5) orally and blood samples were collected intermittently for 60 h with CSF samples collected intermittently for 24 h. Mebendazole was quantitated in plasma and CSF using high performance liquid chromatography. Median peak plasma concentrations (Cmax) were reached at 7 ± 2 h (100 mg/kg) of 220 ng/mL (81, 283) and at 15 ± 4 h (200 mg/kg) of 147 ng/ml (112, 298). The respective area under the curve (AUC: ng/ml/h) reported as a median was 2,119 (1,876, 3,288) vs. 3,115 (1,559, 4,972). Median plasma concentrations (ng/ml) for 100 vs. 200 mg/kg were 47 (32, 52) vs. 65 (35, 104), respectively. For CSF, the median value for Cmax (at 100 mg/kg vs. 200 mg/kg) was 8 (2, 28) vs. 21 (12, 27) and AUC was 87 (22, 157) vs. 345 (92, 372), respectively. Relative bioavailability in CSF vs. plasma was 4 to 10%. Although several animals demonstrated clinical signs indicative of gastrointestinal upset [i.e., vomiting (n = 2), diarrhea (n = 2), or both (n = 1)], these events were not considered serious. The in vitro IC50 for gliomas can be reached in CSF at 100 mg/kg (n = 1), however a 200 mg/kg dose yielded more consistent concentrations.
Collapse
Affiliation(s)
- Amy B. Yanke
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Kendall E. Day
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- BluePearl Pet Hospital North Dallas, Lewisville, TX, United States
| | - Amanda R. Taylor
- Southeast Veterinary Neurology, Boynton Beach, FL, United States
| | - Crisanta Cruz-Espindola
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Dawn M. Boothe
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
3
|
Becerril-Vega J, Hernández-Campos A, González-Hernández I, Flores-Ramos M, Castillo R, Leyva-Gómez G, Mayet-Cruz L, Jung-Cook H. Development and Pharmacokinetic Evaluation of Two Parenteral Formulations of Albendazole Using Prodrug and Cosolvent Approaches. AAPS PharmSciTech 2023; 24:158. [PMID: 37498473 DOI: 10.1208/s12249-023-02613-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/28/2023] [Indexed: 07/28/2023] Open
Abstract
Albendazole is a broad-spectrum anthelmintic drug used for parasitic infections. In addition, due to its mechanism of action, it has been studied as an anticancer agent. However, poor and highly variable bioavailability are limiting factors for its use in systemic illnesses. The present study aimed to develop two parenteral formulations of albendazole and to compare its pharmacokinetic profile with the conventional oral administration. Parenteral formulations were developed using two different approaches: a phosphonooxymethylated prodrug and cosolvents. For the albendazole prodrug, once synthetized, its solubility and hydrolysis with alkaline phosphatase were evaluated. A factorial design of experiments was used for the cosolvent formulation. Stability and hemolytic activity were assessed. A pharmacokinetic study was performed on New Zealand rabbits. Both formulations were administered intravenously, and the prodrug was also administered intramuscularly. Results were compared with those obtained after the oral administration of albendazole. A 20,000-fold and 6000-fold increase in albendazole solubility was found with the prodrug and cosolvent formulations, respectively. Both parenteral formulations displayed higher albendazole plasma concentrations for the first 2 h compared with oral administration, even when the oral dose was doubled. The absolute bioavailability of oral albendazole was 15.5% while for the intramuscular administration of the prodrug was 102.6%. Both parenteral formulations showed a significant decrease in the formation of albendazole sulfoxide (ANOVA p<0.05) and allowed greater exposure to albendazole. Albendazole cosolvent parenteral formulation could be a promising option in systemic illnesses considering its ease of preparation and superb pharmacokinetic performance.
Collapse
Affiliation(s)
- José Becerril-Vega
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Alicia Hernández-Campos
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Iliana González-Hernández
- Laboratorio de Neuropsicofarmacología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Miguel Flores-Ramos
- Escuela Nacional de Estudios Superiores, Unidad Mérida, Universidad Nacional Autónoma de México, Yucatán, Mexico
| | - Rafael Castillo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lourdes Mayet-Cruz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Helgi Jung-Cook
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
4
|
Villarruel-Melquiades F, Mendoza-Garrido ME, García-Cuellar CM, Sánchez-Pérez Y, Pérez-Carreón JI, Camacho J. Current and novel approaches in the pharmacological treatment of hepatocellular carcinoma. World J Gastroenterol 2023; 29:2571-2599. [PMID: 37213397 PMCID: PMC10198058 DOI: 10.3748/wjg.v29.i17.2571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/19/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignant tumours worldwide. The mortality-to-incidence ratio is up to 91.6% in many countries, representing the third leading cause of cancer-related deaths. Systemic drugs, including the multikinase inhibitors sorafenib and lenvatinib, are first-line drugs used in HCC treatment. Unfortunately, these therapies are ineffective in most cases due to late diagnosis and the development of tumour resistance. Thus, novel pharmacological alternatives are urgently needed. For instance, immune checkpoint inhibitors have provided new approaches targeting cells of the immune system. Furthermore, monoclonal antibodies against programmed cell death-1 have shown benefits in HCC patients. In addition, drug combinations, including first-line treatment and immunotherapy, as well as drug repurposing, are promising novel therapeutic alternatives. Here, we review the current and novel pharmacological approaches to fight HCC. Preclinical studies, as well as approved and ongoing clinical trials for liver cancer treatment, are discussed. The pharmacological opportunities analysed here should lead to significant improvement in HCC therapy.
Collapse
Affiliation(s)
- Fernanda Villarruel-Melquiades
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - María Eugenia Mendoza-Garrido
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Claudia M García-Cuellar
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Julio Isael Pérez-Carreón
- Instituto Nacional de Medicina Genómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| |
Collapse
|
5
|
Qiao W, Zhang P, Jiang N, Zhang S, Bai H, Xie L, Sun L, Wang X. Albumin nanostructure assisted ABZ anti-parasite immune therapy for T. spiralis muscle infection. BIOMATERIALS ADVANCES 2023; 150:213434. [PMID: 37087912 DOI: 10.1016/j.bioadv.2023.213434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Currently, the treatment of Trichinella spiralis (T. spiralis) intracellular infection by oral administration of albendazole (ABZ) is hampered by its poor aqueous solubility and rapid metabolism. Herein, the nanoparticles with BSA and ABZ (ABZ-BSA Nps) were constructed by a desolvation technique in the study. The anti-parasite activity and pharmacokinetics of ABZ-BSA Nps were evaluated for T. spiralis muscle larvae during the encysted phase. The immune-responsive cytokines of ABZ-BSA Nps were quantitatively analyzed. The results showed that ABZ-BSA Nps could eliminate the muscle larvae by triggering the unbalance of Th1/Th2 immune-response in the infection mice. For ABZ-BSA Nps treatment group, the plasma concentration of ABZSO (ABZ active metabolite) was higher than ABZ and the muscle larvae were reduced by 70.2 %. In conclusion, the study had constructed a successful prospective protein nanoparticle delivery ABZ and evidenced the ABZ could be used for intracellular parasite therapy by triggering the anti-parasite immunity of hosts.
Collapse
Affiliation(s)
- Weidong Qiao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Peng Zhang
- College of Chemistry, Jilin University, Changchun 130012, PR China; Electron Microscope Center, Jilin University, Changchun 130012, PR China
| | - Ning Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Shuyan Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Huifang Bai
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Lingfeng Xie
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Lin Sun
- College of Chemistry, Jilin University, Changchun 130012, PR China.
| | - Xuelin Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, PR China.
| |
Collapse
|
6
|
Stolfi C, Pacifico T, Luiz-Ferreira A, Monteleone G, Laudisi F. Anthelmintic Drugs as Emerging Immune Modulators in Cancer. Int J Mol Sci 2023; 24:ijms24076446. [PMID: 37047419 PMCID: PMC10094506 DOI: 10.3390/ijms24076446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Despite recent advances in treatment approaches, cancer is still one of the leading causes of death worldwide. Restoration of tumor immune surveillance represents a valid strategy to overcome the acquired resistance and cytotoxicity of conventional therapies in oncology and immunotherapeutic drugs, such as immune checkpoint inhibitors and immunogenic cell death inducers, and has substantially progressed the treatment of several malignancies and improved the clinical management of advanced disease. Unfortunately, because of tumor-intrinsic and/or -extrinsic mechanisms for escaping immune surveillance, only a fraction of patients clinically respond to and benefit from cancer immunotherapy. Accumulating evidence derived from studies of drug repositioning, that is, the strategy to identify new uses for approved or investigational drugs that are outside the scope of the original medical indication, has suggested that some anthelmintic drugs, in addition to their antineoplastic effects, exert important immunomodulatory actions on specific subsets of immune cell and related pathways. In this review, we report and discuss current knowledge on the impact of anthelmintic drugs on host immunity and their potential implication in cancer immunotherapy.
Collapse
|
7
|
Lee YT, Tan YJ, Oon CE. Benzimidazole and its derivatives as cancer therapeutics: The potential role from traditional to precision medicine. Acta Pharm Sin B 2023; 13:478-497. [PMID: 36873180 PMCID: PMC9978992 DOI: 10.1016/j.apsb.2022.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer is the second leading cause of mortality globally which remains a continuing threat to human health today. Drug insensitivity and resistance are critical hurdles in cancer treatment; therefore, the development of new entities targeting malignant cells is considered a high priority. Targeted therapy is the cornerstone of precision medicine. The synthesis of benzimidazole has garnered the attention of medicinal chemists and biologists due to its remarkable medicinal and pharmacological properties. Benzimidazole has a heterocyclic pharmacophore, which is an essential scaffold in drug and pharmaceutical development. Multiple studies have demonstrated the bioactivities of benzimidazole and its derivatives as potential anticancer therapeutics, either through targeting specific molecules or non-gene-specific strategies. This review provides an update on the mechanism of actions of various benzimidazole derivatives and the structure‒activity relationship from conventional anticancer to precision healthcare and from bench to clinics.
Collapse
Affiliation(s)
- Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| |
Collapse
|
8
|
Zhang JY, Zhu XQ. Comparison of the Hydride-Donating Ability and Activity of Five- and Six-Membered Benzoheterocyclic Compounds in Acetonitrile. Molecules 2022; 27:7252. [PMID: 36364079 PMCID: PMC9658978 DOI: 10.3390/molecules27217252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 08/01/2023] Open
Abstract
In this work, we compared the hydride-donating ability of five-membered benzoheterocyclic compounds (FMB) and six-membered benzoheterocyclic compounds (SMB), isomers of DMBI and DMIZ and of DMPZ and DMPX, using detailed thermodynamic driving forces [ΔGo (XH)], kinetic intrinsic barriers (ΔG≠XH/X), and thermo-kinetic parameters [ΔG≠° (XH)]. For DMBI and DMIZ, the values of ΔGo (XH), ΔG≠XH/X, and ΔG≠° (XH) are 49.2 and 53.7 kcal/mol, 35.88 and 42.04 kcal/mol, and 42.54 and 47.87 kcal/mol, respectively. For DMPZ and DMPX, the values of ΔGo (XH), ΔG≠XH/X, and ΔG≠° (XH) are 73.2 and 79.5 kcal/mol, 35.34 and 25.02 kcal/mol, and 54.27 and 52.26 kcal/mol, respectively. It is easy to see that the FMB isomers are thermodynamically dominant and that the SMB isomers are kinetically dominant. Moreover, according to the analysis of ΔG≠° (XH), compared to the SMB isomers, the FMB isomers have a stronger hydride-donating ability in actual chemical reactions.
Collapse
|
9
|
Alzahrani SAS, Nazreen S, Elhenawy AA, Ahmad A, Alam MM. Benzimidazole‐1,3,4‐Oxadiazole Hybrids: Synthesis, Anticancer Evaluation, Docking and DFT Studies. ChemistrySelect 2022. [DOI: 10.1002/slct.202201559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Syed Nazreen
- Department of Chemistry Faculty of Science Al-Baha University Al-Baha Kingdom of Saudi Arabia
| | - Ahmed A. Elhenawy
- Department of Chemistry Faculty of Science Al-Baha University Al-Baha Kingdom of Saudi Arabia
- Chemistry Department Faculty of Science Al-Azhar University 11884 Nasr City Cairo Egypt
| | - Abrar Ahmad
- Department of Biochemistry Faculty of Science King Abdulaziz University Jeddah Kingdom of Saudi Arabia
| | - Mohammad Mahboob Alam
- Department of Chemistry Faculty of Science Al-Baha University Al-Baha Kingdom of Saudi Arabia
| |
Collapse
|
10
|
Song B, Park EY, Kim KJ, Ki SH. Repurposing of Benzimidazole Anthelmintic Drugs as Cancer Therapeutics. Cancers (Basel) 2022; 14:cancers14194601. [PMID: 36230527 PMCID: PMC9559625 DOI: 10.3390/cancers14194601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/06/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Although non-prescription anthelmintics are often used for cancer treatment, there is a lack of information regarding their anti-cancer effects in clinical settings. The aims of our review are to describe the possibilities and limitations of the anti-cancer effects of benzimidazole anthelmintics and to suggest ways to overcome these limitations. The results of the current review illustrate the potential development of anthelmintics as a useful strategy for cancer treatment based on much preclinical evidence. Furthermore, they suggest that more rigorous studies on whole anti-cancer pathways and development strategies, including formulations, could result in significantly enhanced anti-cancer effects of benzimidazoles as a repurposed cancer therapy in clinical settings. Abstract Benzimidazoles have shown significant promise for repurposing as a cancer therapy. The aims of this review are to investigate the possibilities and limitations of the anti-cancer effects of benzimidazole anthelmintics and to suggest ways to overcome these limitations. This review included studies on the anti-cancer effects of 11 benzimidazoles. Largely divided into three parts, i.e., preclinical anti-cancer effects, clinical anti-cancer effects, and pharmacokinetic properties, we examine the characteristics of each benzimidazole and attempt to elucidate its key properties. Although many studies have demonstrated the anti-cancer effects of benzimidazoles, there is limited evidence regarding their effects in clinical settings. This might be because the clinical trials conducted using benzimidazoles failed to restrict their participants with specific criteria including cancer entities, cancer stages, and genetic characteristics of the participants. In addition, these drugs have limitations including low bioavailability, which results in insufficient plasma concentration levels. Additional studies on whole anti-cancer pathways and development strategies, including formulations, could result significant enhancements of the anti-cancer effects of benzimidazoles in clinical situations.
Collapse
Affiliation(s)
- Bomi Song
- Graduate School of Clinical Pharmacy, Chosun University, Gwangju 61452, Korea
| | - Eun Young Park
- College of Pharmacy, Mokpo National University, Mokpo 58554, Korea
| | - Kwang Joon Kim
- College of Pharmacy, Mokpo National University, Mokpo 58554, Korea
- Correspondence: (K.J.K.); (S.H.K.); Tel.: +82-61-450-2334 (K.J.K.); +82-62-230-6639 (S.H.K.)
| | - Sung Hwan Ki
- Graduate School of Clinical Pharmacy, Chosun University, Gwangju 61452, Korea
- Correspondence: (K.J.K.); (S.H.K.); Tel.: +82-61-450-2334 (K.J.K.); +82-62-230-6639 (S.H.K.)
| |
Collapse
|
11
|
Albendazole-induced autophagy blockade contributes to elevated apoptosis in cholangiocarcinoma cells through AMPK/mTOR activation. Toxicol Appl Pharmacol 2022; 454:116214. [PMID: 36055539 DOI: 10.1016/j.taap.2022.116214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 12/09/2022]
Abstract
Albendazole (ABZ) is a broad-spectrum anti-parasitic drug that exhibits antitumor effects against several carcinomas. The effects of ABZ on cholangiocarcinoma (CCA) and its underlying mechanisms are still unclear. Our study aims to investigate the role of ABZ in inducing autophagy-mediated apoptosis of cholangiocarcinoma cells. The antitumor effects of ABZ were evaluated against CCA cells and HIBEC intrahepatic biliary epithelial cells. Furthermore, the apoptosis rates, and autophagy flux in RBE and FRH-0201 cells treated with ABZ were investigated. ABZ inhibited proliferation, induced cell death and apoptosis in CCA cells in vitro. In vivo, tumors from ABZ- treated BALB/c nude mice were significantly smaller than untreated mice. ABZ also induced the initiation of autophagy via AMPK/mTOR pathways, resulting in the formation of autophagosome. In addition, ABZ blocked autophagic flux by inhibiting the fusion of autophagosome-lysosome, which increased the apoptotic death of CCA cells. However, the apoptotic death of CCA cells induced by ABZ was reversed by 3-methyladenine (3-MA), an autophagosome formation inhibitor, but increased by chloroquine (CQ), an autophagosome-lysosome fusion inhibitor.Our work provides novel mechanisms for anti-tumor effects of ABZ on CCA, suggesting that ABZ may be used as a potent autophagy inhibitor in the treatment of CCA.
Collapse
|
12
|
Badripour A, Behzadi M, Hassanipour A, Azar PRS, Rahbar A, Abbaslou Z, Ehghaghi E, Piranviseh A, Khavandi MM, Ahmadi-Tafti SM, Ashouri M, Soltani ZE, Dehpour A. Albendazole ameliorates inflammatory response in a rat model of acute mesenteric ischemia reperfusion injury. Biomed Pharmacother 2022; 153:113320. [PMID: 35752010 DOI: 10.1016/j.biopha.2022.113320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acute mesenteric ischemia is known as a life threatening condition. Re-establishment of blood flow in this condition can lead to mesenteric ischemia reperfusion (MIR) injury which is accompanied by inflammatory response. Still, clear blueprint of inflammatory mechanism underlying MIR injury has not been provided. Interestingly, Albendazole has exhibited notable effects on inflammation and cytokine production. In this study, we aimed to evaluate outcomes of MIR injury following pretreatment with Albendazole with respect to assessment of mesenteric inflammation and ischemia threshold. METHODS Male rats were randomly divided into sham operated, vehicle treated, Albendazole 100 mg/kg and Albendazole 200 mg/kg groups. MIR injury was induced by occlusion of superior mesenteric artery for 30 min followed by 120 min of reperfusion. Samples were utilized for assessment of epithelial survival and villous height. Immunohistochemistry study revealed intestinal expression of TNF-α and HIF-1-α. Gene expression of NF-κB/TLR4/TNF-α/IL-6 was measured using RTPCR. Also protein levels of inflammatory cytokines in serum and intestine were assessed by ELISA method. RESULTS Histopathological study demonstrated that pretreatment with Albendazole could ameliorate decline in villous height and epithelial survival following MIR injury. Also, systemic inflammation was suppressed after administration of Albendazole. Analysis of possible participating inflammatory pathway could demonstrate that intestinal expression of NF-κB/TLR4/TNF-α/IL-6 is significantly attenuated in treated groups. Eventually, IHC study illustrated concordant decline in mesenteric expression of HIF-1-α/TNF-α. CONCLUSION Single dose pretreatment with Albendazole could ameliorate inflammatory response and enhance ischemia threshold following induction of MIR injury. More studies would clarify existing causality in this phenomenon.
Collapse
Affiliation(s)
- Abolfazl Badripour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Behzadi
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Hassanipour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Pasha Reza Shams Azar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Rahbar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zhaleh Abbaslou
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus Via Mersin 10, Turkey
| | - Elnaz Ehghaghi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus Via Mersin 10, Turkey
| | - Ashkan Piranviseh
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdi Khavandi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Ahmadi-Tafti
- Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran; Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ashouri
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zahra Ebrahim Soltani
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Sultana T, Jan U, Lee JI. Double Repositioning: Veterinary Antiparasitic to Human Anticancer. Int J Mol Sci 2022; 23:ijms23084315. [PMID: 35457127 PMCID: PMC9029030 DOI: 10.3390/ijms23084315] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 12/18/2022] Open
Abstract
Drug repositioning, the approach of discovering different uses for existing drugs, has gained enormous popularity in recent years in the anticancer drug discovery field due to the increasing demand for anticancer drugs. Additionally, the repurposing of veterinary antiparasitic drugs for the treatment of cancer is gaining traction, as supported by existing literature. A prominent example is the proposal to implement the use of veterinary antiparasitics such as benzimidazole carbamates and halogenated salicylanilides as novel anticancer drugs. These agents have revealed pronounced anti-tumor activities and gained special attention for “double repositioning”, as they are repurposed for different species and diseases simultaneously, acting via different mechanisms depending on their target. As anticancer agents, these compounds employ several mechanisms, including the inhibition of oncogenic signal transduction pathways of mitochondrial respiration and the inhibition of cellular stress responses. In this review, we summarize and provide valuable information about the experimental, preclinical, and clinical trials of veterinary antiparasitic drugs available for the treatment of various cancers in humans. This review suggests the possibility of new treatment options that could improve the quality of life and outcomes for cancer patients in comparison to the currently used treatments.
Collapse
Affiliation(s)
- Tania Sultana
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (T.S.); (U.J.)
| | - Umair Jan
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (T.S.); (U.J.)
| | - Jeong Ik Lee
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea; (T.S.); (U.J.)
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Correspondence: ; Tel.: +82-2-2049-6234
| |
Collapse
|
14
|
Novel therapeutic strategy for melanoma based on albendazole and the CDK4/6 inhibitor palbociclib. Sci Rep 2022; 12:5706. [PMID: 35383224 PMCID: PMC8983746 DOI: 10.1038/s41598-022-09592-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Although an increasing number of patients benefit from immunotherapy and targeted therapies, melanoma remains incurable with increasing incidence. Drug repositioning and repurposing is an alternative strategy to discover and develop novel anticancer drugs or combined therapeutic regimens. In this study, we demonstrated that albendazole (ABZ), an Food and Drug Administration (FDA)-approved broad-spectrum antiparasitic agent, significantly inhibits the proliferation of melanoma cells in vitro and in vivo. RNA sequencing and flow cytometry analysis revealed that ABZ arrests melanoma cells at the G2/M phase of the cell cycle and induces cell apoptosis. More importantly, the CDK4/6 inhibitor palbociclib, as a member of the first and only class of highly specific CDK inhibitors approved for cancer treatment to date, showed significant synergistic effects with ABZ treatment in melanoma cells and mouse models. Taken together, we revealed a previously unappreciated function of ABZ in antimelanoma proliferation by inducing cell cycle arrest and apoptosis and provided a novel combined therapeutic regimen of ABZ plus CDK4/6 inhibitor treatment in melanoma.
Collapse
|
15
|
Ghaferi M, Zahra W, Akbarzadeh A, Ebrahimi Shahmabadi H, Alavi SE. Enhancing the efficacy of albendazole for liver cancer treatment using mesoporous silica nanoparticles: an in vitro study. EXCLI JOURNAL 2022; 21:236-249. [PMID: 35221842 PMCID: PMC8859643 DOI: 10.17179/excli2021-4491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022]
Abstract
The present study aimed to synthesize albendazole (ABZ)-loaded Mobil Composition of Matter No. 41 (MCM-41 NPs) to increase the efficacy of the drug against liver cancer. ABZ was loaded into MCM-41 NPs, and after in vitro characterization, such as size, size distribution, zeta potential, morphology, chemical composition, thermal profile, drug release, surface and pore volume, and pore size, their biological effects were evaluated using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) cell migration assays. The results demonstrated that monodispersed and spherical NPs with a size of 220 ± 11.5 and 293 ± 8.7 nm, for MCM-41 NPs and ABZ-loaded MCM-41 NPs, respectively, and drug loading efficiency of 30 % were synthesized. ABZ was loaded physically into MCM-41 NPs, leading to a decrease in surface volume, pore size, and pore volume. Also, MCM-41 NPs could increase the cytotoxicity effects of ABZ by 2.9-fold (IC50 = 23 and 7.9 µM for ABZ and ABZ-loaded MCM-41 NPs, respectively). In addition, both ABZ and ABZ-loaded MCM-41 NPs could restrain the cell migration by 12 %. Overall, the results of the present study suggest evaluating the potency of MCM-41 NPs, as a potent nanoplatform, for ABZ delivery in vivo environment. See also the Graphical Abstract(Fig. 1).
Collapse
Affiliation(s)
- Mohsen Ghaferi
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Warda Zahra
- Nishtar Medical University and Hospital, Multan 60000, Pakistan
| | - Azim Akbarzadeh
- Department of Pilot Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Ebrahimi Shahmabadi
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Seyed Ebrahim Alavi
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
16
|
Saber S, Nasr M, Saad AS, Mourad AAE, Gobba NA, Shata A, Hafez AM, Elsergany RN, Elagamy HI, El-Ahwany E, Amin NA, Girgis S, Elewa YHA, Mahmoud MH, Batiha GES, El-Rous MA, Kamal I, Kaddah MMY, Khodir AE. Albendazole-loaded cubosomes interrupt the ERK1/2-HIF-1α-p300/CREB axis in mice intoxicated with diethylnitrosamine: A new paradigm in drug repurposing for the inhibition of hepatocellular carcinoma progression. Biomed Pharmacother 2021; 142:112029. [PMID: 34416629 DOI: 10.1016/j.biopha.2021.112029] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/28/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer related deaths worldwide. It was suggested that albendazole (ABZ) is a powerful inhibitor of several carcinoma types. However, the bioavailability of ABZ is very poor. Additionally, the mechanisms underlying the antitumor effects of ABZ may go beyond its tubulin-inhibiting activity. Therefore, we aimed to examine the effects of ABZ suspension (i.p. and p.o.) and ABZ-loaded cubosomes (LC) on the diethylnitrosamine-induced HCC in mice. ABZ-loaded nanoparticles exhibited a mean particle size of 48.17 ± 0.65 nm and entrapped 93.26 ± 2.48% of ABZ. The in vivo absorption study confirmed a two-fold improvement in the relative bioavailability compared with aqueous ABZ suspension. Furthermore, the oral administration of ABZ cubosomal dispersion demonstrated regression of tumor production rates that was comparable with ABZ (i.p.). ABZ relieved oxidative stress, improved liver function, and decreased necroinflammation score. The antiangiogenic activity was evident as ABZ effectively downregulated tissue expression of CD34, mRNA expression of CD309 and VEGF at the protein expression level. Besides, lower levels of MMP-9 and CXCR4 indicated antimetastatic activity. ABZ showed a considerable level of apoptotic activity as indicated by increased mRNA expression level of p53 and the increased Bax/BCL-2 ratio and active caspase-3. Additionally, Ki-67 expression levels were downregulated showing an antiproliferative potential. These protective effects contributed to increasing survival rate of diethylnitrosamine-treated mice. These effects found to be mediated via interrupting ERK1/2-HIF-1α-p300/CREB interactions. Therefore, our findings revealed that disrupting ERK1/2-HIF-1α-p300/CREB interplay might create a novel therapeutic target for the management of HCC.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| | - Mohamed Nasr
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Ahmed S Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port-Said University, Port-Said, Egypt
| | - Ahmed A E Mourad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port-Said University, Port-Said, Egypt
| | - Naglaa A Gobba
- Department of Pharmacology and Toxicology, College of Pharmacy, Misr University for Science and Technology, Egypt
| | - Ahmed Shata
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Clinical Pharmacy, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Abdel-Moneim Hafez
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Egypt; Department of Physiology, College of Medicine, Qassim University, Saudi Arabia
| | - Ramy N Elsergany
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Heba I Elagamy
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Noha A Amin
- Department of Haematology, Theodor Bilharz Research Institute, Egypt
| | - Samuel Girgis
- Department of Pharmaceutics, Faculty of Pharmacy, Alsalam University, Egypt
| | - Yaser H A Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Hokkaido, Japan
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Magdy Abou El-Rous
- Department of Biochemistry, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Islam Kamal
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Mohamed M Y Kaddah
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications, New Borg El-Arab 21934, Alexandria, Egypt
| | - Ahmed E Khodir
- Department of Pharmacology, Faculty of Pharmacy, Horus University, Egypt
| |
Collapse
|
17
|
Albendazole inhibits NF-κB signaling pathway to overcome tumor stemness and bortezomib resistance in multiple myeloma. Cancer Lett 2021; 520:307-320. [PMID: 34390764 DOI: 10.1016/j.canlet.2021.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
Multiple myeloma (MM) is incurable and the second most common hematologic malignancy in plasma cells. Multiple myeloma stem cell-like cells (MMSCs), a rare population of MM cells, are believed to be the major cause of drug resistance and high recurrence rates in patients with MM. Therefore, developing novel strategies to eradicate MMSCs may favor myeloma treatment. In this study, based on the drug repositioning strategy, we found that albendazole (ABZ), a broad-spectrum antiparasitic drug, selectively suppresses the proliferation of multiple myeloma cells in vitro and in vivo and decreases number of aldehyde dehydrogenase (ALDH)-positive MMSCs in MM. Furthermore, RNA-seq of MM cells after ABZ treatment revealed that inhibition of the nuclear factor kappa-B (NF-κB) pathway is a key mediator of ABZ against MM. Moreover, we demonstrated that ABZ can resensitize cells resistant to bortezomib and overcome MMSCs-induced bortezomib resistance by decreasing ALDH1+ MMSCs numbers. Our findings provide preclinical evidence for utilizing the previously known pharmacologically active drug albendazole for the treatment of multiple myeloma.
Collapse
|
18
|
Noura M, Morita K, Kiyose H, Okuno Y, Matsuo H, Koyama A, Nishinaka-Arai Y, Kamikubo Y, Adachi S. Albendazole induces the terminal differentiation of acute myeloid leukaemia cells to monocytes by stimulating the Krüppel-like factor 4-dihydropyrimidinase-like 2A (KLF4-DPYSL2A) axis. Br J Haematol 2021; 194:598-603. [PMID: 34227104 DOI: 10.1111/bjh.17557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 11/27/2022]
Abstract
Differentiation therapy is a less toxic but still a very effective treatment for a subset of acute myeloid leukaemia (AML) cases. With the goal to identify novel compounds that can effectively and safely induce the terminal differentiation of non-acute promyelocytic leukaemia (APL) AML cells, we performed a chemical screening and identified albendazole (ABZ), a widely used anti-helminthic drug, as a promising lead compound that can differentiate non-APL AML cells by stimulating the Krüppel-like factor 4-dihydropyrimidinase-like 2A (KLF4-DPYSL2A) differentiation axis to the monocytes. Our in vitro and in vivo findings demonstrate that ABZ is an attractive candidate drug as a novel differentiation chemotherapy for patients with non-APL AML.
Collapse
Affiliation(s)
- Mina Noura
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken Morita
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroki Kiyose
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukiko Okuno
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidemasa Matsuo
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Asami Koyama
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoko Nishinaka-Arai
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yasuhiko Kamikubo
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Dimov S, Mavrova AT, Yancheva D, Nikolova B, Tsoneva I. Thieno[2,3-d]pyrimidin-4(3H)-one Derivatives of Benzimidazole as Potential Anti- Breast Cancer (MDA-MB-231, MCF-7) Agents. Anticancer Agents Med Chem 2021; 21:1441-1450. [PMID: 32698751 DOI: 10.2174/1871520620666200721131431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 11/22/2022]
Abstract
AIMS The purpose of this study was the synthesis of some new thienopyrimidine derivatives of 1,3-disubstituted benzimidazoles and the evaluation of their cytotoxicity against MDA-MB-231, MCF-7, and 3T3 cells lines. BACKGROUND An overexpression or mutational activation of TK receptors EGFR and HER2/neu is characteristic of tumors. It has been found that some thieno[2,3-d]pyrimidines exhibited better inhibitory activity against Epidermal Growth Factor Receptor (EGFR/ErbB-2) tyrosine kinase in comparison to aminoquinazolines. Breast cancer activity towards MDA-MB-231 and MCF-7 cell lines by inhibiting EGFR was revealed by a novel 2-arylbenzimidazole. This motivated the synthesis of new thienopyrimidines possessing benzimidazole fragments in order to evaluate their cytotoxicity to the above-mentioned cell lines. OBJECTIVE The objectives of the study were to design and synthesize a novel series of thieno[2,3-d]pyrimidines bearing biologically active moieties, such as 1,3-disubstituted-benzimidazole heterocycle, structurally similar to diaryl ureas in order to evaluate their cytotoxicity against MDA-MB-231, and MCF-7 breast cancer cell lines. METHODS N,N-disubstituted benzimidazole-2-one carbonitriles were synthesized by Aza-Michael addition and used as precursors to generate some of the new thieno[2,3-d]pyrimidines in acidic medium The interaction of chloroethyl-2-thienopyrimidines, 2-amino-benzimidazole and benzimidazol-2-one nitriles under solid-liquid transfer catalysis conditions led to new thienopyrimidines. MTT assay for cell survival was performed in order to evaluate the cytotoxicity of the tested compounds. A fluorescence study was conducted to elucidate some aspects of the mechanism of action. RESULTS The effects of nine synthesized compounds were investigated towards MDA-MB-231, MCF-7 and 3T3 cell lines. Thieno[2,3-d]pyirimidine-4-one 16 (IC50 - 0.058μM) and 21 (IC50 - 0.029μM) possess high cytotoxicity against MDA-MB-231 cells after 24h. The most cytotoxic compounds against breast cancer MCF-7 cells was compound 21 (IC50 - 0.074μM), revealing lower cytotoxicity against mouse fibroblast 3T3 cells with IC50 - 0.20μM. SAR analysis was performed. Fluorescence study of the treatment of MDA-MB cells with compound 21 was carried out in order to clarify some aspects of the mechanism of action. CONCLUSION The relationship between cytotoxicity of compounds 14 and 20 against MCF-7 and 3T3 cells can suggest a similar mechanism of action. The antitumor potential of the tested compounds proves the necessity for further investigation to estimate the exact inhibition pathway in the cellular processes. The fluorescence study of the treatment of MDA-MB cells with compound 21 showed a rapid process of apoptosis.
Collapse
Affiliation(s)
- Stefan Dimov
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - Anelia Ts Mavrova
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Build. 9, 1113 Sofia, Bulgaria
| | - Biliana Nikolova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Build. 21, 1113 Sofia, Bulgaria
| | - Iana Tsoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Build. 21, 1113 Sofia, Bulgaria
| |
Collapse
|
20
|
Identification of Small Molecule Inhibitors of a Mir155 Transcriptional Reporter in Th17 Cells. Sci Rep 2021; 11:11498. [PMID: 34075120 PMCID: PMC8169650 DOI: 10.1038/s41598-021-90944-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 05/12/2021] [Indexed: 11/09/2022] Open
Abstract
MicroRNA miR-155 is an important regulatory molecule in the immune system and is highly expressed and functional in Th17 cells, a subset of CD4+ T helper cells which are key players in autoimmune diseases. Small molecules that can modulate miR-155 may potentially provide new therapeutic avenues to inhibit Th17 cell-mediated autoimmune diseases. Here, we present a novel high-throughput screening assay using primary T cells from genetically engineered Mir155 reporter mice, and its use to screen libraries of small molecules to identify novel modulators of Th17 cell function. We have discovered a chemical series of (E)-1-(phenylsulfonyl)-2-styryl-1H-benzo[d] imidazoles as novel down-regulators of Mir155 reporter and cytokine expression in Th17 cells. In addition, we found that FDA approved antiparasitic agents belonging to the 'azole' family also down-regulate Mir155 reporter and cytokine expression in Th17 cells, and thus could potentially be repurposed to treat Th17-driven immunopathologies.
Collapse
|
21
|
Chai JY, Jung BK, Hong SJ. Albendazole and Mebendazole as Anti-Parasitic and Anti-Cancer Agents: an Update. THE KOREAN JOURNAL OF PARASITOLOGY 2021; 59:189-225. [PMID: 34218593 PMCID: PMC8255490 DOI: 10.3347/kjp.2021.59.3.189] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022]
Abstract
The use of albendazole and mebendazole, i.e., benzimidazole broad-spectrum anthelmintics, in treatment of parasitic infections, as well as cancers, is briefly reviewed. These drugs are known to block the microtubule systems of parasites and mammalian cells leading to inhibition of glucose uptake and transport and finally cell death. Eventually they exhibit ovicidal, larvicidal, and vermicidal effects on parasites, and tumoricidal effects on hosts. Albendazole and mebendazole are most frequently prescribed for treatment of intestinal nematode infections (ascariasis, hookworm infections, trichuriasis, strongyloidiasis, and enterobiasis) and can also be used for intestinal tapeworm infections (taeniases and hymenolepiasis). However, these drugs also exhibit considerable therapeutic effects against tissue nematode/cestode infections (visceral, ocular, neural, and cutaneous larva migrans, anisakiasis, trichinosis, hepatic and intestinal capillariasis, angiostrongyliasis, gnathostomiasis, gongylonemiasis, thelaziasis, dracunculiasis, cerebral and subcutaneous cysticercosis, and echinococcosis). Albendazole is also used for treatment of filarial infections (lymphatic filariasis, onchocerciasis, loiasis, mansonellosis, and dirofilariasis) alone or in combination with other drugs, such as ivermectin or diethylcarbamazine. Albendazole was tried even for treatment of trematode (fascioliasis, clonorchiasis, opisthorchiasis, and intestinal fluke infections) and protozoan infections (giardiasis, vaginal trichomoniasis, cryptosporidiosis, and microsporidiosis). These drugs are generally safe with few side effects; however, when they are used for prolonged time (>14-28 days) or even only 1 time, liver toxicity and other side reactions may occur. In hookworms, Trichuris trichiura, possibly Ascaris lumbricoides, Wuchereria bancrofti, and Giardia sp., there are emerging issues of drug resistance. It is of particular note that albendazole and mebendazole have been repositioned as promising anti-cancer drugs. These drugs have been shown to be active in vitro and in vivo (animals) against liver, lung, ovary, prostate, colorectal, breast, head and neck cancers, and melanoma. Two clinical reports for albendazole and 2 case reports for mebendazole have revealed promising effects of these drugs in human patients having variable types of cancers. However, because of the toxicity of albendazole, for example, neutropenia due to myelosuppression, if high doses are used for a prolonged time, mebendazole is currently more popularly used than albendazole in anti-cancer clinical trials.
Collapse
Affiliation(s)
- Jong-Yil Chai
- Institute of Parasitic Diseases, Korea Association of Health Promotion, Seoul 07649,
Korea
- Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Bong-Kwang Jung
- Institute of Parasitic Diseases, Korea Association of Health Promotion, Seoul 07649,
Korea
| | - Sung-Jong Hong
- Department of Environmental Medical Biology, Chung-Ang University College of Medicine, Seoul 06974,
Korea
| |
Collapse
|
22
|
Esfahani MKM, Islam N, Cabot PJ, Izake EL. Development of Thiabendazole-Loaded Mesoporous Silica Nanoparticles for Cancer Therapy. ACS Biomater Sci Eng 2021; 8:4153-4162. [PMID: 34056895 DOI: 10.1021/acsbiomaterials.1c00066] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Thiabendazole (TBZ) is an anthelmintic drug currently studied for anticancer purposes. However, due to its low solubility, its biomedical application has been limited. Using mesoporous silica nanoparticles (MSNPs), such as Mobil Composition of Matter Number 41 (MCM-41), as a drug carrier, is a promising approach to improve the solubility of low water-soluble drugs. In the present work, we aim to develop TBZ-loaded MCM-41 (TBZ MCM-41) nanoparticles to improve the solubility and the therapeutic efficacy of TBZ against prostate cancer PC-3 cells. TBZ MCM-41 nanoparticles were synthesized with a size of 215.9 ± 0.07 nm, a spherical shape, a hexagonal array of channels, and a drug loading capacity of 19.1%. The biological effects of the nanoformulation on PC-3 cells were then evaluated using a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT), IncuCyte live-cell imaging system, cell migration, and reactive oxygen species (ROS) assays. The results demonstrated that TBZ was released from MCM-41 nanoparticles in a controlled manner at pH values of 1.2 and 6.8. The cell viability measurements revealed that the TBZ MCM-41 nanoparticles caused a considerable 2.8-fold increase in the cytotoxicity of TBZ (IC50 127.3 and 46 μM for TBZ and TBZ MCM-41 nanoparticles, respectively). The results of the proliferation assay were in agreement with those of the cell viability measurements, where the MCM-41 increased the cytotoxicity of TBZ in a concentration-dependent manner. Also, the TBZ MCM-41 nanoparticles were found to enhance the potency of the drug and inhibit PC-3 cell migration. In addition, the ROS assay confirmed that TBZ MCM-41 nanoparticles were approximately 15% more potent than TBZ to produce ROS. Overall, the results demonstrated that MCM-41 nanoparticles are a promising carrier to improve the therapeutic efficacy of TBZ against PC-3 cells and suggest evaluating the efficacy of the formulation in vivo.
Collapse
Affiliation(s)
- Maedeh Koohi Moftakhari Esfahani
- School of Chemistry and Physics, Science and Engineering Faculty, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia.,Centre for Materials Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
| | - Nazrul Islam
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane, 4000, Australia
| | - Peter J Cabot
- School of Pharmacy, The University of Queensland, Woolloongabba 4102, Australia
| | - Emad L Izake
- School of Chemistry and Physics, Science and Engineering Faculty, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia.,Centre for Materials Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
| |
Collapse
|
23
|
Weidong L, Sanna L, Bordoni V, Tiansheng Z, Chengxun L, Murineddu G, Pinna GA, Kelvin DJ, Bagella L. Target identification of a novel unsymmetrical 1,3,4-oxadiazole derivative with antiproliferative properties. J Cell Physiol 2021; 236:3789-3799. [PMID: 33089499 DOI: 10.1002/jcp.30120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/22/2020] [Accepted: 10/10/2020] [Indexed: 02/05/2023]
Abstract
1,3,4-Oxadiazole derivatives are widely used in research on antineoplastic drugs. Recently, we discovered a novel unsymmetrical 1,3,4-oxadiazole compound with antiproliferative properties called 2j. To further investigate its possible targets and molecular mechanisms, RNA-seq was performed and the differentially expressed genes (DEGs) were obtained after treatment. Data were analyzed using functional (Gene Ontology term) and pathway (Kyoto Encyclopedia of Genes and Genomes) enrichment of the DEGs. The hub genes were determined by the analysis of protein-protein interaction networks. The connectivity map (CMap) information provided insight into the model action of antitumor small molecule drugs. Hub genes have been identified through function gene networks using STRING analysis. The small molecular targets obtained by CMap comparison showed that 2j is a tubulin inhibitor and it acts mainly affecting tumor cells through the cell cycle, FoxO signaling pathway, apoptotic, and p53 signaling pathways. The possible targets of 2j could be TUBA1A and TUBA4A. Molecular docking results indicated that 2j interacts at the colchicine-binding site on tubulin.
Collapse
Affiliation(s)
- Lyu Weidong
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Laboratory of Immunity, Shantou University Medical College, Shantou, Guangdong, China
| | - Luca Sanna
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Valentina Bordoni
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Zeng Tiansheng
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Laboratory of Immunity, Shantou University Medical College, Shantou, Guangdong, China
| | - Li Chengxun
- Laboratory of Immunity, Shantou University Medical College, Shantou, Guangdong, China
| | - Gabriele Murineddu
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Gerard A Pinna
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - David J Kelvin
- Laboratory of Immunity, Shantou University Medical College, Shantou, Guangdong, China
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Kim U, Shin C, Kim CY, Ryu B, Kim J, Bang J, Park JH. Albendazole exerts antiproliferative effects on prostate cancer cells by inducing reactive oxygen species generation. Oncol Lett 2021; 21:395. [PMID: 33777218 PMCID: PMC7988661 DOI: 10.3892/ol.2021.12656] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Benzimidazole derivatives are used for their antihelmintic properties, but have also been reported to exert anticancer effects. In the present study, the anticancer effects of albendazole on prostate cancer cells were assessed using proliferation, clonogenic and migration assays. To investigate the anticancer mechanisms of albendazole, reactive oxygen species (ROS) levels were measured, and the expression of genes associated with oxidative stress and Wnt/β-catenin signaling was confirmed by reverse transcription-quantitative PCR and western blotting. Albendazole selectively inhibited the proliferation of the PC3, DU145, LNCaP and AT2 prostate cancer cell lines at concentrations that did not affect the proliferation of a normal prostate cell line (RWPE-1). Albendazole also inhibited the colony formation and migration of PC3 and DU145 cells, as well as inducing ROS production. Diphenyleneiodonium chloride, an inhibitor of NADPH oxidase (NOX), one of the sources of ROS, decreased basal ROS levels in the PC3 and DU145 cells, but did not reduce albendazole-associated ROS production, suggesting that ROS production following albendazole treatment was NOX-independent. The anticancer effect was decreased when albendazole-induced ROS was reduced by treatment with antioxidants (glutathione and N-acetylcysteine). Furthermore, albendazole decreased the mRNA expression of CDGSH iron sulfur domain 2, which regulates antioxidant activity against ROS, as well as the antioxidant enzymes catalase, and glutathione peroxidase 1 and 3. Albendazole also decreased the mRNA expression of catenin β1 and transcription factor 4, which regulate Wnt/β-catenin signaling and its associated targets, Twist family BHLH transcription factor 1 and BCL2. The albendazole-related decrease in the expression levels of oxidative stress-related genes and Wnt/β-catenin signaling proteins was thought to be associated with ROS production. These results suggest that the antihelmintic drug, albendazole, has inhibitory effects against prostate cancer cells in vitro. Therefore, albendazole may potentially be used as a novel anticancer agent for prostate cancer.
Collapse
Affiliation(s)
- Ukjin Kim
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Changsoo Shin
- Department of Energy Resources Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Bokyeong Ryu
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Kim
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Junpil Bang
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Hak Park
- Department of Laboratory Animal Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
25
|
Yang MH, Ha IJ, Um JY, Ahn KS. Albendazole Exhibits Anti-Neoplastic Actions against Gastric Cancer Cells by Affecting STAT3 and STAT5 Activation by Pleiotropic Mechanism(s). Biomedicines 2021; 9:biomedicines9040362. [PMID: 33807326 PMCID: PMC8065911 DOI: 10.3390/biomedicines9040362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
Albendazole (ABZ) has been reported to display anti-tumoral actions against various maliganncies, but possible impact of ABZ on gastric cancer has not been deciphered. As aberrant phosphorylation of STAT3 and STAT5 proteins can regulate the growth and progression of gastric cancer, we postulated that ABZ may interrupt the activation of these oncogenic transcription factors. We found that ABZ exposure abrogated STAT3/5 activation, inhibited phosphorylation of Janus-activated kinases 1/2 and Src and enhanced the levels of SHP-1 protein. Silencing of SHP-1 gene by small interfering RNA (siRNA) reversed the ABZ-promoted attenuation of STAT3 as well as STAT5 activation and cellular apoptosis. In addition, these effects were noted to be driven by an augmented levels of reactive oxygen species caused by drug-induced GSH/GSSG imbalance. Thus, the data indicates that ABZ can modulate the activation of STAT3 and STAT5 by pleiotropic mechanisms in gastric cancer cells.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea;
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Korea;
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea;
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-961-2316
| |
Collapse
|
26
|
Chung I, Zhou K, Barrows C, Banyard J, Wilson A, Rummel N, Mizokami A, Basu S, Sengupta P, Shaikh B, Sengupta S, Bielenberg DR, Zetter BR. Unbiased Phenotype-Based Screen Identifies Therapeutic Agents Selective for Metastatic Prostate Cancer. Front Oncol 2021; 10:594141. [PMID: 33738243 PMCID: PMC7962607 DOI: 10.3389/fonc.2020.594141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023] Open
Abstract
In American men, prostate cancer is the second leading cause of cancer-related death. Dissemination of prostate cancer cells to distant organs significantly worsens patients' prognosis, and currently there are no effective treatment options that can cure advanced-stage prostate cancer. In an effort to identify compounds selective for metastatic prostate cancer cells over benign prostate cancer cells or normal prostate epithelial cells, we applied a phenotype-based in vitro drug screening method utilizing multiple prostate cancer cell lines to test 1,120 different compounds from a commercial drug library. Top drug candidates were then examined in multiple mouse xenograft models including subcutaneous tumor growth, experimental lung metastasis, and experimental bone metastasis assays. A subset of compounds including fenbendazole, fluspirilene, clofazimine, niclosamide, and suloctidil showed preferential cytotoxicity and apoptosis towards metastatic prostate cancer cells in vitro and in vivo. The bioavailability of the most discerning agents, especially fenbendazole and albendazole, was improved by formulating as micelles or nanoparticles. The enhanced forms of fenbendazole and albendazole significantly prolonged survival in mice bearing metastases, and albendazole-treated mice displayed significantly longer median survival times than paclitaxel-treated mice. Importantly, these drugs effectively targeted taxane-resistant tumors and bone metastases - two common clinical conditions in patients with aggressive prostate cancer. In summary, we find that metastatic prostate tumor cells differ from benign prostate tumor cells in their sensitivity to certain drug classes. Taken together, our results strongly suggest that albendazole, an anthelmintic medication, may represent a potential adjuvant or neoadjuvant to standard therapy in the treatment of disseminated prostate cancer.
Collapse
Affiliation(s)
- Ivy Chung
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Kun Zhou
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Courtney Barrows
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
| | - Jacqueline Banyard
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Arianne Wilson
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
| | - Nathan Rummel
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Washington, DC, United States
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Sudipta Basu
- Laboratory for Nanomedicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Poulomi Sengupta
- Laboratory for Nanomedicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Badaruddin Shaikh
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Washington, DC, United States
| | - Shiladitya Sengupta
- Laboratory for Nanomedicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Diane R. Bielenberg
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Bruce R. Zetter
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
27
|
Racoviceanu R, Trandafirescu C, Voicu M, Ghiulai R, Borcan F, Dehelean C, Watz C, Aigner Z, Ambrus R, Coricovac DE, Cîrcioban D, Mioc A, Szuhanek CA, Şoica C. Solid Polymeric Nanoparticles of Albendazole: Synthesis, Physico-Chemical Characterization and Biological Activity. Molecules 2020; 25:E5130. [PMID: 33158183 PMCID: PMC7663605 DOI: 10.3390/molecules25215130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Albendazole is a benzimidazole derivative with documented antitumor activity and low toxicity to healthy cells. The major disadvantage in terms of clinical use is its low aqueous solubility which limits its bioavailability. Albendazole was incorporated into stable and homogeneous polyurethane structures with the aim of obtaining an improved drug delivery system model. Spectral and thermal analysis was used to investigate the encapsulation process and confirmed the presence of albendazole inside the nanoparticles. The in vitro anticancer properties of albendazole encapsulated in polyurethane structures versus the un-encapsulated compound were tested on two breast cancer cell lines, MCF-7 and MDA-MB-231, in terms of cellular viability and apoptosis induction. The study showed that the encapsulation process enhanced the antitumor activity of albendazole on the MCF-7 and MDA-MB-23 breast cancer lines. The cytotoxic activity manifested in a concentration-dependent manner and was accompanied by changes in cell morphology and nuclear fragmentation.
Collapse
Affiliation(s)
- Roxana Racoviceanu
- Department of Pharmaceutical Chemistry, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.R.); (C.T.); (C.Ş.)
| | - Cristina Trandafirescu
- Department of Pharmaceutical Chemistry, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.R.); (C.T.); (C.Ş.)
| | - Mirela Voicu
- Department of Pharmacology and Clinical Pharmacy, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Roxana Ghiulai
- Department of Pharmaceutical Chemistry, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.R.); (C.T.); (C.Ş.)
| | - Florin Borcan
- Department of Analytical Chemistry, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (F.B.); (D.C.)
| | - Cristina Dehelean
- Department of Toxicology, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.D.); (D.E.C.)
| | - Claudia Watz
- Department of Pharmaceutical Physics, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Zoltán Aigner
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, 6th Eotvos Str., 6720 Szeged, Hungary; (Z.A.); (R.A.)
| | - Rita Ambrus
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, 6th Eotvos Str., 6720 Szeged, Hungary; (Z.A.); (R.A.)
| | - Dorina Elena Coricovac
- Department of Toxicology, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.D.); (D.E.C.)
| | - Denisa Cîrcioban
- Department of Analytical Chemistry, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (F.B.); (D.C.)
| | - Alexandra Mioc
- Department of Anatomy, Physiology and Physiopathology, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Camelia Alexandrina Szuhanek
- Department of Orthodontics, Victor Babeș University of Medicine and Pharmacy, 9th Revolutiei din 1989 Bvd, 300041 Timisoara, Romania;
| | - Codruţa Şoica
- Department of Pharmaceutical Chemistry, Victor Babeș University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.R.); (C.T.); (C.Ş.)
| |
Collapse
|
28
|
Nath J, Paul R, Ghosh SK, Paul J, Singha B, Debnath N. Drug repurposing and relabeling for cancer therapy: Emerging benzimidazole antihelminthics with potent anticancer effects. Life Sci 2020; 258:118189. [DOI: 10.1016/j.lfs.2020.118189] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 02/08/2023]
|
29
|
Cortez-Maya S, Moreno-Herrera A, Palos I, Rivera G. Old Antiprotozoal Drugs: Are They Still Viable Options for Parasitic Infections or New Options for Other Diseases? Curr Med Chem 2020; 27:5403-5428. [DOI: 10.2174/0929867326666190628163633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 01/16/2023]
Abstract
Parasitic diseases, caused by helminths (ascariasis, hookworm, trichinosis, and schistosomiasis)
and protozoa (chagas, leishmaniasis, and amebiasis), are considered a serious public
health problem in developing countries. Additionally, there is a limited arsenal of anti-parasitic
drugs in the current pipeline and growing drug resistance. Therefore, there is a clear need for the
discovery and development of new compounds that can compete and replace these drugs that have
been controlling parasitic infections over the last decades. However, this approach is highly resource-
intensive, expensive and time-consuming. Accordingly, a drug repositioning strategy of the
existing drugs or drug-like molecules with known pharmacokinetics and safety profiles is alternatively
being used as a fast approach towards the identification of new treatments. The artemisinins,
mefloquine, tribendimidine, oxantel pamoate and doxycycline for the treatment of helminths, and
posaconazole and hydroxymethylnitrofurazone for the treatment of protozoa are promising candidates.
Therefore, traditional antiprotozoal drugs, which were developed in some cases decades ago,
are a valid solution. Herein, we review the current status of traditional anti-helminthic and antiprotozoal
drugs in terms of drug targets, mode of action, doses, adverse effects, and parasite resistance
to define their suitability for repurposing strategies. Current antiparasitic drugs are not only
still viable for the treatment of helminth and protozoan infections but are also important candidates
for new pharmacological treatments.
Collapse
Affiliation(s)
- Sandra Cortez-Maya
- Instituto de Quimica, Universidad Nacional Autonoma de Mexico, Cd. Universitaria, Circuito Exterior, Coyoacan, 04510 Ciudad de Mexico, Mexico
| | - Antonio Moreno-Herrera
- Laboratorio de Biotecnologia Farmaceutica, Centro de Biotecnologia Genomica, Instituto Politecnico Nacional, 88710 Reynosa, Mexico
| | - Isidro Palos
- Unidad AcadEmica Multidisciplinaria Reynosa-Rodhe, Universidad AutOnoma de Tamaulipas, 88710 Reynosa, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnologia Farmaceutica, Centro de Biotecnologia Genomica, Instituto Politecnico Nacional, 88710 Reynosa, Mexico
| |
Collapse
|
30
|
Jubie S, Durai U, Latha S, Ayyamperumal S, Wadhwani A, Prabha T. Repurposing of Benzimidazole Scaffolds for HER2 Positive Breast Cancer Therapy: An In-Silico Approach. Curr Drug Res Rev 2020; 13:73-83. [PMID: 32955008 DOI: 10.2174/2589977512999200821170221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/06/2020] [Accepted: 06/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND A newer trend has been seen recently to reuse the conventional drugs with distinct indications for the newer applications to speed up the drug discovery and development based on earlier records and safety data. Most of the non-cancerous agents could afford a little or tolerable side effects in individuals. However, the repositioning of these non-cancerous agents for successful anticancer therapy is an outstanding strategy for future anti-cancer drug development. Since more diverse and selective cancer drug targets are being discovered and developed, the approved drug collections are particularly useful to quickly identify clinically advanced anticancer drugs against those targets. OBJECTIVE Antihelminthic drugs such as Mebendazole and Albendazole (Benzimidazole class) have been reported to exhibit cytotoxicity (or anticancer activities) against several types of cancer. Therefore, this study aims to repurpose the benzimidazole scaffold for breast cancer treatment. METHODS In the present study, three hydrazone analogs having a benzimidazole motif in their structural frame were synthesized. Their in-silico binding studies against HER2 receptor (PDB ID: 4LQM) and ADMET studies were carried out using Accelrys drug discovery studio 4.1. Cytotoxicity of the synthesized compounds against HER2 overexpressed MCF-7 cell lines was determined by MTT assay. RESULTS One of the compounds 2-[2-(2,4-dinitrophenyl)hydrazinylidene]-2,3-dihydro-1H-benzimidazole (U1) has shown good cytotoxicity when compared to the standard Lapatinib, which is a well known HER2 inhibitor. CONCLUSIONS Thus, the designed benzimidazole scaffold might serve as the best leads for treating breast cancer, which is additionally confirmed by performing their docking study via Accelrys discovery studio.
Collapse
Affiliation(s)
- Selvaraj Jubie
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Researchooty, Nilgiris, Ooty-643001, Tamilnadu, India
| | - Uma Durai
- Department of Chemistry, PSG College of Arts & Science, Avinasi Road, Coimbatore- 641 014, Tamilnadu, India
| | - Subbiah Latha
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, Anna University, Bharathidasan Institute of Technology Campus, Tiruchirappalli, Tamilnadu, India
| | - Selvaraj Ayyamperumal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Researchooty, Nilgiris, Ooty-643001, Tamilnadu, India
| | - Ashish Wadhwani
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Researchooty, Nilgiris, Ooty-643001, Tamilnadu, India
| | - Thangavelu Prabha
- Department of Pharmaceutical Chemistry, Nandha College of Pharmacy, Affiliated to The Tamilnadu Dr. MGR. Medical University-Chennai, Erode-638052, Tamilnadu, India
| |
Collapse
|
31
|
Selvaraj J, Prabha T, Yadav N. Identification of Drug Candidates for Breast Cancer Therapy Through Scaffold Repurposing: A Brief Review. Curr Drug Res Rev 2020; 13:3-15. [PMID: 32838729 DOI: 10.2174/2589977512666200824103019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 06/10/2020] [Accepted: 07/02/2020] [Indexed: 11/22/2022]
Abstract
Conventional drug discovery is a time consuming and expensive expedition with less clinical preference achievement proportion intended for breast cancer therapy. Even if numerous novel approaches to the conformation of drugs have been introduced for breast cancer therapy, they are yet to be implemented in clinical practice. This tempting strategy facilitates a remarkable chance to take the entire benefit of existing drugs. Despite drug repurposing significantly decrease the investigational period and cost, it has got many objections and issues. Scaffold repurposing is an approach that procures a novel significance on the decrepit motto of "to commencement with a pristine drug" . Hence, we move into a probable and nearer approach, the exploitation of scaffolds, which was originally developed for other purposes, including anti-tumor activity. In this review, we summarize different drugs and scaffolds used in breast cancer therapy.
Collapse
Affiliation(s)
- Jubie Selvaraj
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research Ooty, Nilgiris, Tamilnadu, India
| | - Thangavelu Prabha
- Department of Pharmaceutical Chemistry, Nandha College of Pharmacy, Koorapalayam Pirivu, Pitchandam Palayam Post, Erode-638052, Tamilnadu, India
| | - Neetu Yadav
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research Ooty, Nilgiris, Tamilnadu, India
| |
Collapse
|
32
|
Son DS, Lee ES, Adunyah SE. The Antitumor Potentials of Benzimidazole Anthelmintics as Repurposing Drugs. Immune Netw 2020; 20:e29. [PMID: 32895616 PMCID: PMC7458798 DOI: 10.4110/in.2020.20.e29] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
The development of refractory tumor cells limits therapeutic efficacy in cancer by activating mechanisms that promote cellular proliferation, migration, invasion, metastasis, and survival. Benzimidazole anthelmintics have broad-spectrum action to remove parasites both in human and veterinary medicine. In addition to being antiparasitic agents, benzimidazole anthelmintics are known to exert anticancer activities, such as the disruption of microtubule polymerization, the induction of apoptosis, cell cycle (G2/M) arrest, anti-angiogenesis, and blockage of glucose transport. These antitumorigenic effects even extend to cancer cells resistant to approved therapies and when in combination with conventional therapeutics, enhance anticancer efficacy and hold promise as adjuvants. Above all, these anthelmintics may offer a broad, safe spectrum to treat cancer, as demonstrated by their long history of use as antiparasitic agents. The present review summarizes central literature regarding the anticancer effects of benzimidazole anthelmintics, including albendazole, parbendazole, fenbendazole, mebendazole, oxibendazole, oxfendazole, ricobendazole, and flubendazole in cancer cell lines, animal tumor models, and clinical trials. This review provides valuable information on how to improve the quality of life in patients with cancers by increasing the treatment options and decreasing side effects from conventional therapy.
Collapse
Affiliation(s)
- Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neurosciences and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA
| | - Samuel E Adunyah
- Department of Biochemistry, Cancer Biology, Neurosciences and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
33
|
Albendazole negatively regulates keratinocyte proliferation. Clin Sci (Lond) 2020; 134:907-920. [PMID: 32236445 DOI: 10.1042/cs20191215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increased keratinocyte proliferation occurs in the skin of psoriatic patients and is supposed to play a role in the pathogenesis of this disorder. Compounds interfering with keratinocyte proliferation could be useful in the management of psoriatic patients. AIM To investigate whether albendazole, an anti-helmintic drug that regulates epithelial cell function in various systems, inhibits keratinocyte proliferation in models of psoriasis. METHODS Aldara-treated mice received daily topical application of albendazole. Keratinocyte proliferation and keratin (K) 6 and K16 expression were evaluated by immunohistochemistry and Western blotting and inflammatory cells/mediators were analysed by immunohistochemistry and real-time PCR. In human keratinocytes (HEKa and HaCaT) treated with albendazole, cell cycle and proliferation, keratins and cell cycle-associated factors were evaluated by flow cytometry, colorimetric assay and Western blotting respectively. RESULTS Aldara-treated mice given albendazole exhibited reduced epidermal thickness, decreased number of proliferating keratinocytes and K6/K16 expression. Reduction of CD3- and Ly6G-positive cells in the skin of albendazole-treated mice associated with inhibition of IL-6, TNF-α, IL-1β, IL-17A, IL-36, CCL17, CXCL1, CXCL2 and CXCL5 expression. Treatment of keratinocytes with albendazole reduced K6/K16 expression and reversibly inhibited cell growth by promoting accumulation of cells in S-phase. This phenomenon was accompanied by down-regulation of CDC25A, a phosphatase regulating progression of cell cycle through S-phase, and PKR-dependent hyper-phosphorylation of eIF2α, an inhibitor of CDC25 translation. In Aldara-treated mice, albendazole activated PKR, enhanced eIF2α phosphorylation and reduced CDC25A expression. CONCLUSIONS Data show that albendazole inhibits keratinocyte proliferation and exerts therapeutic effect in a murine model of psoriasis.
Collapse
|
34
|
Laudisi F, Marônek M, Di Grazia A, Monteleone G, Stolfi C. Repositioning of Anthelmintic Drugs for the Treatment of Cancers of the Digestive System. Int J Mol Sci 2020; 21:ijms21144957. [PMID: 32668817 PMCID: PMC7404055 DOI: 10.3390/ijms21144957] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/20/2022] Open
Abstract
Tumors of the digestive system, when combined together, account for more new cases and deaths per year than tumors arising in any other system of the body and their incidence continues to increase. Despite major efforts aimed at discovering and validating novel and effective drugs against these malignancies, the process of developing such drugs remains lengthy and costly, with high attrition rates. Drug repositioning (also known as drug repurposing), that is, the process of finding new uses for approved drugs, has been gaining popularity in oncological drug development as it provides the opportunity to expedite promising anti-cancer agents into clinical trials. Among the drugs considered for repurposing in oncology, compounds belonging to some classes of anthelmintics—a group of agents acting against infections caused by parasitic worms (helminths) that colonize the mammalian intestine—have shown pronounced anti-tumor activities and attracted particular attention due to their ability to target key oncogenic signal transduction pathways. In this review, we summarize and discuss the available experimental and clinical evidence about the use of anthelmintic drugs for the treatment of cancers of the digestive system.
Collapse
Affiliation(s)
- Federica Laudisi
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy; (F.L.); (A.D.G.); (G.M.)
| | - Martin Marônek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia;
| | - Antonio Di Grazia
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy; (F.L.); (A.D.G.); (G.M.)
| | - Giovanni Monteleone
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy; (F.L.); (A.D.G.); (G.M.)
| | - Carmine Stolfi
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy; (F.L.); (A.D.G.); (G.M.)
- Division of Clinical Biochemistry and Clinical Molecular Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-06-72596163
| |
Collapse
|
35
|
Gandhi SR, Quintans JDSS, Gandhi GR, Araújo AADS, Quintans Júnior LJ. The use of cyclodextrin inclusion complexes to improve anticancer drug profiles: a systematic review. Expert Opin Drug Deliv 2020; 17:1069-1080. [DOI: 10.1080/17425247.2020.1776261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sathiyabama Rajiv Gandhi
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Sergipe, Brazil
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Campus Prof. João Cardoso Nascimento, Sergipe, Brazil
| | - Jullyana De Souza Siqueira Quintans
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Sergipe, Brazil
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Campus Prof. João Cardoso Nascimento, Sergipe, Brazil
| | - Gopalsamy Rajiv Gandhi
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Sergipe, Brazil
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Campus Prof. João Cardoso Nascimento, Sergipe, Brazil
- Department of Pharmacy, Federal University of Sergipe, Sergipe, Brazil
| | - Adriano Antunes De Souza Araújo
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Campus Prof. João Cardoso Nascimento, Sergipe, Brazil
- Department of Pharmacy, Federal University of Sergipe, Sergipe, Brazil
| | - Lucindo José Quintans Júnior
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Sergipe, Brazil
- Postgraduate Program of Health Sciences (PPGCS), University Hospital, Federal University of Sergipe (HU-UFS), Campus Prof. João Cardoso Nascimento, Sergipe, Brazil
| |
Collapse
|
36
|
Maqbool F, Falconer JR, Moyle PM. Supercritical fluid assembly of albendazole liposomes targeting gastrin-releasing peptide receptor overexpressing tumors. Nanomedicine (Lond) 2020; 15:1315-1330. [PMID: 32484025 DOI: 10.2217/nnm-2020-0048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To develop albendazole (ABZ)-loaded bombesin(6-14) (BBN(6-14)) functionalized liposomes for targeting GRPR to enhance delivery to cancer cells. Materials & methods: ABZ-loaded liposomes were formulated using supercritical CO2 technology; functionalized with a GRPR-targeted lipid-anchored BBN(6-14) peptide; and evaluated for effects on cell viability, particle size and targeted cell uptake. Results: BBN(6-14)-coated ABZ liposomes decreased cell viability compared with nonfunctionalized ABZ liposomes. The level of GRPR expression positively correlated with intracellular uptake and decreased cell viability. The reduced cell viability, higher cell uptake and GRPR expression were observed in the order PC-3 > Caco-2 > HepG2 cells. Conclusion: BBN(6-14)-functionalized ABZ liposomes showed enhanced reduction in cell viability compared with nonfunctionalized ABZ liposomes.
Collapse
Affiliation(s)
- Faheem Maqbool
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - James R Falconer
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Peter M Moyle
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
37
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
38
|
Gonzalez-Fierro A, Dueñas-González A. Drug repurposing for cancer therapy, easier said than done. Semin Cancer Biol 2019; 68:123-131. [PMID: 31877340 DOI: 10.1016/j.semcancer.2019.12.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/26/2019] [Accepted: 12/15/2019] [Indexed: 12/24/2022]
Abstract
Drug repurposing for cancer therapy is currently a hot topic of research. Theoretically, in contrast to the known hurdles of developing new molecular entities, the approach of repurposing has several advantages. Mostly, it is said that it is faster, safer, easier, and cheaper. In the real world, however, there are only three repurposed drugs so far, that are listed in widely recognized cancer guidelines, but a large number of them are being studied. Among the many barriers to repurposing cancer drugs, economical-driven are the most important that difficult the clinical development of them. In this review, we provide an overview of the current status of drug repurposing for cancer therapy and the barriers that need to be overcome to realize the benefit of this approach. It means to have repositioned drugs for cancer therapy accepted as standard therapy for cancer indications at low cost.
Collapse
Affiliation(s)
| | - Alfonso Dueñas-González
- Division of Basic Researach, Instituto Nacional de Cancerología, Mexico City, Mexico; Unit of Biomedical Research in Cancer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico NAM/ Instituto Nacional de Cancerología, Mexico City, Mexico.
| |
Collapse
|
39
|
Wang D, Chen G, Li X, Jia Q. Hypercrosslinked β-cyclodextrin porous polymer as adsorbent for effective uptake towards albendazole from aqueous media. Sep Purif Technol 2019. [DOI: 10.1016/j.seppur.2019.115720] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
40
|
Design and Synthesis of Benzimidazole-Chalcone Derivatives as Potential Anticancer Agents. Molecules 2019; 24:molecules24183259. [PMID: 31500191 PMCID: PMC6767017 DOI: 10.3390/molecules24183259] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 11/17/2022] Open
Abstract
Numerous reports have shown that conjugated benzimidazole derivatives possess various kinds of biological activities, including anticancer properties. In this report, we designed and synthesized 24 new molecules comprising a benzimidazole ring, arene, and alkyl chain-bearing cyclic moieties. The results showed that the N-substituted benzimidazole derivatives bearing an alkyl chain and a nitrogen-containing 5- or 6-membered ring enhanced the cytotoxic effects on human breast adenocarcinoma (MCF-7) and human ovarian carcinoma (OVCAR-3) cell lines. Among the 24 synthesized compounds, (2E)-1-(1-(3-morpholinopropyl)-1H-benzimidazol-2 -yl)-3-phenyl-2-propen-1-one) (23a) reduced the proliferation of MCF-7 and OVCAR-3 cell lines demonstrating superior outcomes to those of cisplatin.
Collapse
|
41
|
Philoppes JN, Lamie PF. Design and synthesis of new benzoxazole/benzothiazole-phthalimide hybrids as antitumor-apoptotic agents. Bioorg Chem 2019; 89:102978. [DOI: 10.1016/j.bioorg.2019.102978] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
|
42
|
Benzimidazoles Downregulate Mdm2 and MdmX and Activate p53 in MdmX Overexpressing Tumor Cells. Molecules 2019; 24:molecules24112152. [PMID: 31181622 PMCID: PMC6600429 DOI: 10.3390/molecules24112152] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor suppressor p53 is mutated in about 50% of cancers. Most malignant melanomas carry wild-type p53, but p53 activity is often inhibited due to overexpression of its negative regulators Mdm2 or MdmX. We performed high throughput screening of 2448 compounds on A375 cells carrying p53 activity luciferase reporter construct to reveal compounds that promote p53 activity in melanoma. Albendazole and fenbendazole, two approved and commonly used benzimidazole anthelmintics, stimulated p53 activity and were selected for further studies. The protein levels of p53 and p21 increased upon the treatment with albendazole and fenbendazole, indicating activation of the p53–p21 pathway, while the levels of Mdm2 and MdmX decreased in melanoma and breast cancer cells overexpressing these proteins. We also observed a reduction of cell viability and changes of cellular morphology corresponding to mitotic catastrophe, i.e., G2/M cell cycle arrest of large multinucleated cells with disrupted microtubules. In summary, we established a new tool for testing the impact of small molecule compounds on the activity of p53 and used it to identify the action of benzimidazoles in melanoma cells. The drugs promoted the stability and transcriptional activity of wild-type p53 via downregulation of its negative regulators Mdm2 and MdmX in cells overexpressing these proteins. The results indicate the potential for repurposing the benzimidazole anthelmintics for the treatment of cancers overexpressing p53 negative regulators.
Collapse
|
43
|
Cho YK, Shin EY, Uehara H, Ambati B. Antiangiogenesis Effect of Albendazole on the Cornea. J Ocul Pharmacol Ther 2019; 35:254-261. [PMID: 31033390 DOI: 10.1089/jop.2018.0103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose: To investigate the anti-(lymph)angiogenic and anti-inflammatory effects of albendazole and to study whether these effects are additive with bevacizumab therapy in a murine corneal suture model. Methods: Corneal neovascularization (NV) and lymphangiogenesis (LY) were compared in a corneal suture model after administration of a subconjunctival injection of albendazole, bevacizumab, dexamethasone, or phosphate-buffered saline (PBS). Immunohistochemical staining and analysis were performed in each group. Real-time polymerase chain reaction (RT-PCR) was performed to quantify the expression of inflammatory cytokines (tumor necrosis factor [TNF]-alpha and interleukin-6), vascular endothelial growth factor (VEGF)-A, VEGF-C, vascular endothelial growth factor receptor (VEGFR)-2, and VEGFR-3. To evaluate the additive effect of albendazole, corneal NV and LY were also analyzed in a combined group of albendazole and bevacizumab therapy and the additive effect was compared with that in the group of double dose of bevacizumab. Results: The albendazole group showed less NV and less LY compared with the PBS control group (P < 0.01). When albendazole was combined with bevacizumab therapy, a significant decrease in NV and LY was seen compared with bevacizumab treatment alone, and with albendazole alone (all P values <0.05). The combination group showed better antilymphangiogenesis effect than the group of double dose bevacizumab. The albendazole-treated group showed reduced expression of VEGF-A, VEGF-C, TNF-alpha, and VEGFR-2 compared with corneas from the PBS group (P value <0.05 in all respective comparisons). Conclusion: Albendazole significantly decreased NV and LY in the cornea. This beneficial effect is additively enhanced when combined with bevacizumab treatment.
Collapse
Affiliation(s)
- Yang Kyung Cho
- 1 Department of Ophthalmology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Eun Young Shin
- 2 Research Institute of Medical Science, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Hironori Uehara
- 3 Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Balamurali Ambati
- 3 Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
44
|
Nowak-Sliwinska P, Scapozza L, Ruiz i Altaba A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim Biophys Acta Rev Cancer 2019; 1871:434-454. [PMID: 31034926 PMCID: PMC6528778 DOI: 10.1016/j.bbcan.2019.04.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 02/08/2023]
Abstract
The strategy of using existing drugs originally developed for one disease to treat other indications has found success across medical fields. Such drug repurposing promises faster access of drugs to patients while reducing costs in the long and difficult process of drug development. However, the number of existing drugs and diseases, together with the heterogeneity of patients and diseases, notably including cancers, can make repurposing time consuming and inefficient. The key question we address is how to efficiently repurpose an existing drug to treat a given indication. As drug efficacy remains the main bottleneck for overall success, we discuss the need for machine-learning computational methods in combination with specific phenotypic studies along with mechanistic studies, chemical genetics and omics assays to successfully predict disease-drug pairs. Such a pipeline could be particularly important to cancer patients who face heterogeneous, recurrent and metastatic disease and need fast and personalized treatments. Here we focus on drug repurposing for colorectal cancer and describe selected therapeutics already repositioned for its prevention and/or treatment as well as potential candidates. We consider this review as a selective compilation of approaches and methodologies, and argue how, taken together, they could bring drug repurposing to the next level.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland; Translational Research Center in Oncohaematology, University of Geneva, Rue Michel Servet 1, 1211 Geneva 4, Switzerland.
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| |
Collapse
|
45
|
Non-mitotic effect of albendazole triggers apoptosis of human leukemia cells via SIRT3/ROS/p38 MAPK/TTP axis-mediated TNF-α upregulation. Biochem Pharmacol 2019; 162:154-168. [DOI: 10.1016/j.bcp.2018.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/05/2018] [Indexed: 12/27/2022]
|
46
|
Zhao A, Li Y, Orahoske CM, Schnur B, Sabbagh A, Zhang W, Li B, Su B. Lead optimization of selective tubulin inhibitors as anti-trypanosomal agents. Bioorg Med Chem 2019; 27:1517-1528. [PMID: 30833159 DOI: 10.1016/j.bmc.2019.02.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/15/2019] [Accepted: 02/22/2019] [Indexed: 12/16/2022]
Abstract
Previously synthesized tubulin inhibitors showed promising in vitro selectivity and activity against Human African Trypanosomiasis. Current aim is to improve the ligand efficiency and reduce overall hydrophobicity of the compounds, by lead optimization. Via combinatorial chemistry, 60 new analogs were synthesized. For biological assay Trypanosoma brucei brucei Lister 427 cell line were used as the parasite model and for the host model human embryonic kidney cell line HEK-293 and mouse macrophage cell line RAW 264.7 were used to test efficacy. Of the newly synthesized compounds 5, 39, 40, and 57 exhibited IC50s below 5 µM inhibiting the growth of trypanosome cells and not harming the mammalian cells at equipotent concentration. Comparably, the newly synthesized compounds have a reduced amount of aromatic moieties resulting in a decrease in molecular weight. Due to importance of tubulin polymerization during protozoan life cycle its activity was assessed by western blot analyses. Our results indicated that compound 5 had a profound effect on tubulin function. A detailed structure activity relationship (SAR) was summarized that will be used to guide future lead optimization.
Collapse
Affiliation(s)
- Anran Zhao
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Yaxin Li
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Cody M Orahoske
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Brittny Schnur
- Department of Biology, Geo. & Env. Sciences, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Abboud Sabbagh
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Wenjing Zhang
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Bibo Li
- Department of Biology, Geo. & Env. Sciences, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA.
| | - Bin Su
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA.
| |
Collapse
|
47
|
Vinarov Z, Gancheva G, Katev V, Tcholakova SS. Albendazole solution formulation via vesicle-to-micelle transition of phospholipid-surfactant aggregates. Drug Dev Ind Pharm 2018; 44:1130-1138. [DOI: 10.1080/03639045.2018.1438461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Zahari Vinarov
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Gabriela Gancheva
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Vladimir Katev
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Slavka S. Tcholakova
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| |
Collapse
|
48
|
|
49
|
Movahedi F, Li L, Gu W, Xu ZP. Nanoformulations of albendazole as effective anticancer and antiparasite agents. Nanomedicine (Lond) 2017; 12:2555-2574. [PMID: 28954575 DOI: 10.2217/nnm-2017-0102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Initially emerging as a widely used clinical antiparasitic drug, albendazole (ABZ) has been increasingly recognized as an effective anticancer agent due to its outstanding advantage, in other words, low toxicity to normal cells but high effectiveness against parasites and some tumors. The major challenge is its poor water solubility and subsequently low bioavailability. This article thus first reviews the brief achievements in using ABZ to treat parasites and cancers, and summarizes the basic mechanisms of action of ABZ. Then this article critically reviews recent nanotechnological strategies, in other words, formulating/conjugating it with carriers into nanoformulations, in practices of improving aqueous solubility and efficacy in treatment of tumors and parasites. Our expert opinions in this field are provided for more effective delivery of ABZ to treat tumors and parasites in vivo.
Collapse
Affiliation(s)
- Fatemeh Movahedi
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
50
|
Methyl 5-[(1H-indol-3-yl)selanyl]-1H-benzoimidazol-2-ylcarbamate (M-24), a novel tubulin inhibitor, causes G2/M arrest and cell apoptosis by disrupting tubulin polymerization in human cervical and breast cancer cells. Toxicol In Vitro 2017; 42:139-149. [DOI: 10.1016/j.tiv.2017.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/02/2016] [Accepted: 04/12/2017] [Indexed: 11/20/2022]
|