Cargando…

Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging

The development of improved or targeted drugs that discriminate between normal and tumor tissues is the key therapeutic issue in cancer research. However, the development of an analytical method with a high accuracy and sensitivity to achieve quantitative assessment of the tumor targeting of antican...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Jin, Du, Qianqian, Song, Xiaowei, Gao, Shanshan, Pang, Xuechao, Li, Yan, Zhang, Ruiping, Abliz, Zeper, He, Jiuming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7052894/
https://www.ncbi.nlm.nih.gov/pubmed/32194824
http://dx.doi.org/10.7150/thno.41763
_version_ 1783502938926219264
author Zhang, Jin
Du, Qianqian
Song, Xiaowei
Gao, Shanshan
Pang, Xuechao
Li, Yan
Zhang, Ruiping
Abliz, Zeper
He, Jiuming
author_facet Zhang, Jin
Du, Qianqian
Song, Xiaowei
Gao, Shanshan
Pang, Xuechao
Li, Yan
Zhang, Ruiping
Abliz, Zeper
He, Jiuming
author_sort Zhang, Jin
collection PubMed
description The development of improved or targeted drugs that discriminate between normal and tumor tissues is the key therapeutic issue in cancer research. However, the development of an analytical method with a high accuracy and sensitivity to achieve quantitative assessment of the tumor targeting of anticancer drugs and even intratumor heterogeneous distribution of these drugs at the early stages of drug research and development is a major challenge. Mass spectrometry imaging is a label-free molecular imaging technique that provides spatial-temporal information on the distribution of drugs and metabolites in organisms, and its application in the field of pharmaceutical development is rapidly increasing. Methods: The study presented here accurately quantified the distribution of paclitaxel (PTX) and its prodrug (PTX-R) in whole-body animal sections based on the virtual calibration quantitative mass spectrometry imaging (VC-QMSI) method, which is label-free and does not require internal standards, and then applied this technique to evaluate the tumor targeting efficiency in three treatment groups—the PTX-injection treatment group, PTX-liposome treatment group and PTX-R treatment group—in nude mice bearing subcutaneous A549 xenograft tumors. Results: These results indicated that PTX was widely distributed in multiple organs throughout the dosed body in the PTX-injection group and the PTX-liposome group. Notably, in the PTX-R group, both the prodrug and metabolized PTX were mainly distributed in the tumor tissue, and this group showed a significant difference compared with the PTX-liposome group, the relative targeting efficiency of PTX-R group was increased approximately 50-fold, leading to substantially decreased systemic toxicities. In addition, PTX-R showed a significant and specific accumulation in the poorly differentiated intratumor area and necrotic area. Conclusion: This method was demonstrated to be a reliable, feasible and easy-to-implement strategy to quantitatively map the absorption, distribution, metabolism and excretion (ADME) of a drug in the whole-body and tissue microregions and could therefore evaluate the tumor-targeting efficiency of anticancer drugs to predict drug efficacy and safety and provide key insights into drug disposition and mechanisms of action and resistance. Thus, this strategy could significantly facilitate the design and optimization of drugs at the early stage of drug research and development.
format Online
Article
Text
id pubmed-7052894
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-70528942020-03-19 Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging Zhang, Jin Du, Qianqian Song, Xiaowei Gao, Shanshan Pang, Xuechao Li, Yan Zhang, Ruiping Abliz, Zeper He, Jiuming Theranostics Research Paper The development of improved or targeted drugs that discriminate between normal and tumor tissues is the key therapeutic issue in cancer research. However, the development of an analytical method with a high accuracy and sensitivity to achieve quantitative assessment of the tumor targeting of anticancer drugs and even intratumor heterogeneous distribution of these drugs at the early stages of drug research and development is a major challenge. Mass spectrometry imaging is a label-free molecular imaging technique that provides spatial-temporal information on the distribution of drugs and metabolites in organisms, and its application in the field of pharmaceutical development is rapidly increasing. Methods: The study presented here accurately quantified the distribution of paclitaxel (PTX) and its prodrug (PTX-R) in whole-body animal sections based on the virtual calibration quantitative mass spectrometry imaging (VC-QMSI) method, which is label-free and does not require internal standards, and then applied this technique to evaluate the tumor targeting efficiency in three treatment groups—the PTX-injection treatment group, PTX-liposome treatment group and PTX-R treatment group—in nude mice bearing subcutaneous A549 xenograft tumors. Results: These results indicated that PTX was widely distributed in multiple organs throughout the dosed body in the PTX-injection group and the PTX-liposome group. Notably, in the PTX-R group, both the prodrug and metabolized PTX were mainly distributed in the tumor tissue, and this group showed a significant difference compared with the PTX-liposome group, the relative targeting efficiency of PTX-R group was increased approximately 50-fold, leading to substantially decreased systemic toxicities. In addition, PTX-R showed a significant and specific accumulation in the poorly differentiated intratumor area and necrotic area. Conclusion: This method was demonstrated to be a reliable, feasible and easy-to-implement strategy to quantitatively map the absorption, distribution, metabolism and excretion (ADME) of a drug in the whole-body and tissue microregions and could therefore evaluate the tumor-targeting efficiency of anticancer drugs to predict drug efficacy and safety and provide key insights into drug disposition and mechanisms of action and resistance. Thus, this strategy could significantly facilitate the design and optimization of drugs at the early stage of drug research and development. Ivyspring International Publisher 2020-02-03 /pmc/articles/PMC7052894/ /pubmed/32194824 http://dx.doi.org/10.7150/thno.41763 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Zhang, Jin
Du, Qianqian
Song, Xiaowei
Gao, Shanshan
Pang, Xuechao
Li, Yan
Zhang, Ruiping
Abliz, Zeper
He, Jiuming
Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
title Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
title_full Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
title_fullStr Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
title_full_unstemmed Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
title_short Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
title_sort evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7052894/
https://www.ncbi.nlm.nih.gov/pubmed/32194824
http://dx.doi.org/10.7150/thno.41763
work_keys_str_mv AT zhangjin evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT duqianqian evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT songxiaowei evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT gaoshanshan evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT pangxuechao evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT liyan evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT zhangruiping evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT ablizzeper evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging
AT hejiuming evaluationofthetumortargetingefficiencyandintratumorheterogeneityofanticancerdrugsusingquantitativemassspectrometryimaging