Your browser doesn't support javascript.
loading
Nitric oxide-releasing nanoparticles improve doxorubicin anticancer activity.
Alimoradi, Houman; Greish, Khaled; Barzegar-Fallah, Anita; Alshaibani, Lama; Pittalà, Valeria.
Afiliación
  • Alimoradi H; Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.
  • Greish K; College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit, Princess Al-Jawhara Centre for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Kingdom of Bahrain, khaledfg@agu.edu.bh.
  • Barzegar-Fallah A; Department of Oncology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt, khaledfg@agu.edu.bh.
  • Alshaibani L; Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.
  • Pittalà V; College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit, Princess Al-Jawhara Centre for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Kingdom of Bahrain, khaledfg@agu.edu.bh.
Int J Nanomedicine ; 13: 7771-7787, 2018.
Article en En | MEDLINE | ID: mdl-30538458
ABSTRACT

PURPOSE:

Anticancer drug delivery systems are often limited by hurdles, such as off-target distribution, slow cellular internalization, limited lysosomal escape, and drug resistance. To overcome these limitations, we have developed a stable nitric oxide (NO)-releasing nanoparticle (polystyrene-maleic acid [SMA]-tert-dodecane S-nitrosothiol [tDodSNO]) with the aim of enhancing the anticancer properties of doxorubicin (Dox) and a Dox-loaded nanoparticle (SMA-Dox) carrier. MATERIALS AND

METHODS:

Effects of SMA-tDodSNO and/or in combination with Dox or SMA-Dox on cell viability, apoptosis, mitochondrial membrane potential, lysosomal membrane permeability, tumor tissue, and tumor growth were studied using in vitro and in vivo model of triple-negative breast cancer (TNBC). In addition, the concentrations of SMA-Dox and Dox in combination with SMA-tDodSNO were measured in cells and tumor tissues.

RESULTS:

Combination of SMA-tDodSNO and Dox synergistically decreased cell viability and induced apoptosis in 4T1 (TNBC cells). Incubation of 4T1 cells with SMA-tDodSNO (40 µM) significantly enhanced the cellular uptake of SMA-Dox and increased Dox concentration in the cells resulting in a twofold increase (P<0.001). Lysosomal membrane integrity, evaluated by acridine orange (AO) staining, was impaired by 40 µM SMA-tDodSNO (P<0.05 vs control) and when combined with SMA-Dox, this effect was significantly potentiated (P<0.001 vs SMA-Dox). Subcutaneous administration of SMA-tDodSNO (1 mg/kg) to xenografted mice bearing 4T1 cells showed that SMA-tDodSNO alone caused a twofold decrease in the tumor size compared to the control group. SMA-tDodSNO in combination with SMA-Dox resulted in a statistically significant 4.7-fold reduction in the tumor volume (P<0.001 vs control), without causing significant toxicity as monitored through body weight loss.

CONCLUSION:

Taken together, these results suggest that SMA-tDodSNO can be used as a successful strategy to increase the efficacy of Dox and SMA-Dox in a model of TNBC.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Doxorrubicina / Nanopartículas / Liberación de Fármacos / Antineoplásicos / Óxido Nítrico Límite: Animals / Female / Humans Idioma: En Revista: Int J Nanomedicine Año: 2018 Tipo del documento: Article País de afiliación: Nueva Zelanda

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Doxorrubicina / Nanopartículas / Liberación de Fármacos / Antineoplásicos / Óxido Nítrico Límite: Animals / Female / Humans Idioma: En Revista: Int J Nanomedicine Año: 2018 Tipo del documento: Article País de afiliación: Nueva Zelanda