Theranostics 2020; 10(6):2621-2630. doi:10.7150/thno.41763 This issue Cite

Research Paper

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

Jin Zhang1, Qianqian Du2, Xiaowei Song1, Shanshan Gao1, Xuechao Pang1, Yan Li2, Ruiping Zhang1, Zeper Abliz1,3, Jiuming He1✉

1. State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
2. Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
3. Center for Imaging and Systems Biology, Minzu University of China, Beijing, 100081, China.

Citation:
Zhang J, Du Q, Song X, Gao S, Pang X, Li Y, Zhang R, Abliz Z, He J. Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging. Theranostics 2020; 10(6):2621-2630. doi:10.7150/thno.41763. https://www.thno.org/v10p2621.htm
Other styles

File import instruction

Abstract

Graphic abstract

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.

Keywords: quantitative mass spectrometry imaging, whole-body animal, paclitaxel, tumor-targeting efficiency, intratumor heterogeneity


Citation styles

APA
Zhang, J., Du, Q., Song, X., Gao, S., Pang, X., Li, Y., Zhang, R., Abliz, Z., He, J. (2020). Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging. Theranostics, 10(6), 2621-2630. https://doi.org/10.7150/thno.41763.

ACS
Zhang, J.; Du, Q.; Song, X.; Gao, S.; Pang, X.; Li, Y.; Zhang, R.; Abliz, Z.; He, J. Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging. Theranostics 2020, 10 (6), 2621-2630. DOI: 10.7150/thno.41763.

NLM
Zhang J, Du Q, Song X, Gao S, Pang X, Li Y, Zhang R, Abliz Z, He J. Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging. Theranostics 2020; 10(6):2621-2630. doi:10.7150/thno.41763. https://www.thno.org/v10p2621.htm

CSE
Zhang J, Du Q, Song X, Gao S, Pang X, Li Y, Zhang R, Abliz Z, He J. 2020. Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging. Theranostics. 10(6):2621-2630.

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.
Popup Image