Cargando…

Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo

Acute lymphoblastic leukemia (ALL) is an aggressive malignancy. Adults with ALL have more than 50% relapse rates. We have previously validated that overexpression of nucleophosmin (NPM) is involved in the multidrug resistance (MDR) development during ALL; and a synthetically engineered recombinant N...

Descripción completa

Detalles Bibliográficos
Autores principales: Gan, Donghui, Chen, Yuwen, Wu, Zhengjun, Luo, Liping, Yirga, Shimuye Kalayu, Zhang, Na, Ye, Fu, Chen, Haijun, Hu, Jianda, Chen, Yingyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8193937/
https://www.ncbi.nlm.nih.gov/pubmed/34122059
http://dx.doi.org/10.3389/fphar.2021.607755
_version_ 1783706322466766848
author Gan, Donghui
Chen, Yuwen
Wu, Zhengjun
Luo, Liping
Yirga, Shimuye Kalayu
Zhang, Na
Ye, Fu
Chen, Haijun
Hu, Jianda
Chen, Yingyu
author_facet Gan, Donghui
Chen, Yuwen
Wu, Zhengjun
Luo, Liping
Yirga, Shimuye Kalayu
Zhang, Na
Ye, Fu
Chen, Haijun
Hu, Jianda
Chen, Yingyu
author_sort Gan, Donghui
collection PubMed
description Acute lymphoblastic leukemia (ALL) is an aggressive malignancy. Adults with ALL have more than 50% relapse rates. We have previously validated that overexpression of nucleophosmin (NPM) is involved in the multidrug resistance (MDR) development during ALL; and a synthetically engineered recombinant NPM binding protein (NPMBP) has been developed in our group; NPMBP and doxorubicin (DOX) can be conjugated in a nanoparticle-based drug delivery system named DOX-PMs-NPMBP to counteract MDR during ALL. Here, we evaluated the antileukemia potential of DOX-PMs-NPMBP in resistant ALL cells. This study demonstrates that DOX-PMs-NPMBP significantly enhances chemosensitivity to DOX in ALL cells. Despite at variable concentrations, both resistant and primary ALL cells from relapsed patients were sensitive to DOX-PMs-NPMBP. In detail, the half maximal inhibitory concentration (IC50) values of DOX-PMs-NPMBP were between 1.6- and 7.0-fold lower than those of DOX in cell lines and primary ALL cells, respectively; and apoptotic cells ratio was over 2-fold higher in DOX-PMs-NPMBP than DOX. Mechanistically, p53-driven apoptosis induction and cell cycle arrest played essential role in DOX-PMs-NPMBP-induced anti-leukemia effects. Moreover, DOX-PMs-NPMBP significantly inhibited tumor growth and prolonged mouse survival of ALL xenograft models; and no systemic toxicity occurrence was observed after treatment during follow-up. In conclusion, these data indicate that DOX-PMs-NPMBP may significantly exert growth inhibition and apoptosis induction, and markedly improve DOX antileukemia activity in resistant ALL cells. This novel drug delivery system may be valuable to develop as a new therapeutic strategy against multidrug resistant ALL.
format Online
Article
Text
id pubmed-8193937
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-81939372021-06-12 Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo Gan, Donghui Chen, Yuwen Wu, Zhengjun Luo, Liping Yirga, Shimuye Kalayu Zhang, Na Ye, Fu Chen, Haijun Hu, Jianda Chen, Yingyu Front Pharmacol Pharmacology Acute lymphoblastic leukemia (ALL) is an aggressive malignancy. Adults with ALL have more than 50% relapse rates. We have previously validated that overexpression of nucleophosmin (NPM) is involved in the multidrug resistance (MDR) development during ALL; and a synthetically engineered recombinant NPM binding protein (NPMBP) has been developed in our group; NPMBP and doxorubicin (DOX) can be conjugated in a nanoparticle-based drug delivery system named DOX-PMs-NPMBP to counteract MDR during ALL. Here, we evaluated the antileukemia potential of DOX-PMs-NPMBP in resistant ALL cells. This study demonstrates that DOX-PMs-NPMBP significantly enhances chemosensitivity to DOX in ALL cells. Despite at variable concentrations, both resistant and primary ALL cells from relapsed patients were sensitive to DOX-PMs-NPMBP. In detail, the half maximal inhibitory concentration (IC50) values of DOX-PMs-NPMBP were between 1.6- and 7.0-fold lower than those of DOX in cell lines and primary ALL cells, respectively; and apoptotic cells ratio was over 2-fold higher in DOX-PMs-NPMBP than DOX. Mechanistically, p53-driven apoptosis induction and cell cycle arrest played essential role in DOX-PMs-NPMBP-induced anti-leukemia effects. Moreover, DOX-PMs-NPMBP significantly inhibited tumor growth and prolonged mouse survival of ALL xenograft models; and no systemic toxicity occurrence was observed after treatment during follow-up. In conclusion, these data indicate that DOX-PMs-NPMBP may significantly exert growth inhibition and apoptosis induction, and markedly improve DOX antileukemia activity in resistant ALL cells. This novel drug delivery system may be valuable to develop as a new therapeutic strategy against multidrug resistant ALL. Frontiers Media S.A. 2021-05-28 /pmc/articles/PMC8193937/ /pubmed/34122059 http://dx.doi.org/10.3389/fphar.2021.607755 Text en Copyright © 2021 Gan, Chen, Wu, Luo, Yirga, Zhang, Ye, Chen, Hu and Chen. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Gan, Donghui
Chen, Yuwen
Wu, Zhengjun
Luo, Liping
Yirga, Shimuye Kalayu
Zhang, Na
Ye, Fu
Chen, Haijun
Hu, Jianda
Chen, Yingyu
Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo
title Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo
title_full Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo
title_fullStr Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo
title_full_unstemmed Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo
title_short Doxorubicin/Nucleophosmin Binding Protein-Conjugated Nanoparticle Enhances Anti-leukemia Activity in Acute Lymphoblastic Leukemia Cells in vitro and in vivo
title_sort doxorubicin/nucleophosmin binding protein-conjugated nanoparticle enhances anti-leukemia activity in acute lymphoblastic leukemia cells in vitro and in vivo
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8193937/
https://www.ncbi.nlm.nih.gov/pubmed/34122059
http://dx.doi.org/10.3389/fphar.2021.607755
work_keys_str_mv AT gandonghui doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT chenyuwen doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT wuzhengjun doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT luoliping doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT yirgashimuyekalayu doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT zhangna doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT yefu doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT chenhaijun doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT hujianda doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo
AT chenyingyu doxorubicinnucleophosminbindingproteinconjugatednanoparticleenhancesantileukemiaactivityinacutelymphoblasticleukemiacellsinvitroandinvivo