MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice

Abstract Background and aims Several oncolytic viruses applications have been approved in the clinic or in different phases of clinical trials. However, these methods have some rudimentary problems. Therefore, to enhance the delivery and quality of treatment, considering the advantage of cell carrie...

Full description

Bibliographic Details
Main Authors: Seyed-Mahmood Seyed-Khorrami, Hoorieh Soleimanjahi, Sara Soudi, Ala Habibian
Format: Article
Language:English
Published: BMC 2021-05-01
Series:Cancer Cell International
Subjects:
MOI
Online Access:https://doi.org/10.1186/s12935-021-01848-5
id doaj-4c9963efeda041a380480ba600ae6e20
record_format Article
spelling doaj-4c9963efeda041a380480ba600ae6e202021-05-02T11:21:23ZengBMCCancer Cell International1475-28672021-05-0121111910.1186/s12935-021-01848-5MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 miceSeyed-Mahmood Seyed-Khorrami0Hoorieh Soleimanjahi1Sara Soudi2Ala Habibian3Department of Virology, Faculty of Medical Sciences, Tarbiat Modares UniversityDepartment of Virology, Faculty of Medical Sciences, Tarbiat Modares UniversityDepartment of Immunology, Faculty of Medical Sciences, Tarbiat Modares UniversityDepartment of Virology, Faculty of Medical Sciences, Tarbiat Modares UniversityAbstract Background and aims Several oncolytic viruses applications have been approved in the clinic or in different phases of clinical trials. However, these methods have some rudimentary problems. Therefore, to enhance the delivery and quality of treatment, considering the advantage of cell carrier-based methods such as Mesenchymal Stem Cells (MSC) have been proposed. This study was designed to evaluate the performance and quality of cancer treatment based on MSCs loaded by oncolytic reovirus in the cancerous C57BL/6 mouse model. Also, we evaluated MSCs migration potency in vitro and in vivo following the oncolytic reovirus infection. Methods C57BL/6 mice were inoculated with TC-1 cell lines and tumors were established in the right flank. Mice were systemically treated with reovirus, MSCs-loaded with reovirus, MSCs, and PBS as a control in separated groups. Effects of infected AD-MSCs with reovirus on tumor growth and penetration in the tumor site were monitored. All groups of mice were monitored for two months in order to therapeutic and anticancer potential. After treatments, tumor size alteration and apoptosis rate, as well as cytokine release pattern was assessed. Results The results of the current study indicated that the effect of reovirus infection on AD-MSCs is not devastating the migration capacity especially in MOI 1 and 5 while intact cells remain. On the other hand, MSCs play an efficient role as a carrier to deliver oncolytic virus into the tumor site in comparison with systemic administration of reovirus alone. Apoptosis intensity relies on viral titration and passing time. Followed by systemic administration, treatment with oncolytic reovirus-infected AD-MSCs and MSCs alone had shown significant inhibition in tumor growth. Also, treatment by reovirus causes an increase in IFN-γ secretion. Conclusion The results of in vitro and in vivo study confirmed the tumor-homing properties of infected AD-MSCs and the significant antitumor activity of this platform. Hence, our results showed that the cell carrier strategy using oncolytic reovirus-loaded AD-MSCs enhanced virus delivery, infiltration, and antitumor activity can be effectively applied in most cancers.https://doi.org/10.1186/s12935-021-01848-5Drug Delivery SystemsTargeted therapyIFN-γAnti-tumor effectMOIVirus titer
collection DOAJ
language English
format Article
sources DOAJ
author Seyed-Mahmood Seyed-Khorrami
Hoorieh Soleimanjahi
Sara Soudi
Ala Habibian
spellingShingle Seyed-Mahmood Seyed-Khorrami
Hoorieh Soleimanjahi
Sara Soudi
Ala Habibian
MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice
Cancer Cell International
Drug Delivery Systems
Targeted therapy
IFN-γ
Anti-tumor effect
MOI
Virus titer
author_facet Seyed-Mahmood Seyed-Khorrami
Hoorieh Soleimanjahi
Sara Soudi
Ala Habibian
author_sort Seyed-Mahmood Seyed-Khorrami
title MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice
title_short MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice
title_full MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice
title_fullStr MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice
title_full_unstemmed MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice
title_sort mscs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing c57bl/6 mice
publisher BMC
series Cancer Cell International
issn 1475-2867
publishDate 2021-05-01
description Abstract Background and aims Several oncolytic viruses applications have been approved in the clinic or in different phases of clinical trials. However, these methods have some rudimentary problems. Therefore, to enhance the delivery and quality of treatment, considering the advantage of cell carrier-based methods such as Mesenchymal Stem Cells (MSC) have been proposed. This study was designed to evaluate the performance and quality of cancer treatment based on MSCs loaded by oncolytic reovirus in the cancerous C57BL/6 mouse model. Also, we evaluated MSCs migration potency in vitro and in vivo following the oncolytic reovirus infection. Methods C57BL/6 mice were inoculated with TC-1 cell lines and tumors were established in the right flank. Mice were systemically treated with reovirus, MSCs-loaded with reovirus, MSCs, and PBS as a control in separated groups. Effects of infected AD-MSCs with reovirus on tumor growth and penetration in the tumor site were monitored. All groups of mice were monitored for two months in order to therapeutic and anticancer potential. After treatments, tumor size alteration and apoptosis rate, as well as cytokine release pattern was assessed. Results The results of the current study indicated that the effect of reovirus infection on AD-MSCs is not devastating the migration capacity especially in MOI 1 and 5 while intact cells remain. On the other hand, MSCs play an efficient role as a carrier to deliver oncolytic virus into the tumor site in comparison with systemic administration of reovirus alone. Apoptosis intensity relies on viral titration and passing time. Followed by systemic administration, treatment with oncolytic reovirus-infected AD-MSCs and MSCs alone had shown significant inhibition in tumor growth. Also, treatment by reovirus causes an increase in IFN-γ secretion. Conclusion The results of in vitro and in vivo study confirmed the tumor-homing properties of infected AD-MSCs and the significant antitumor activity of this platform. Hence, our results showed that the cell carrier strategy using oncolytic reovirus-loaded AD-MSCs enhanced virus delivery, infiltration, and antitumor activity can be effectively applied in most cancers.
topic Drug Delivery Systems
Targeted therapy
IFN-γ
Anti-tumor effect
MOI
Virus titer
url https://doi.org/10.1186/s12935-021-01848-5
work_keys_str_mv AT seyedmahmoodseyedkhorrami mscsloadedwithoncolyticreovirusmigrationandinvivovirusdeliverypotentialforevaluatinganticancereffectintumorbearingc57bl6mice
AT hooriehsoleimanjahi mscsloadedwithoncolyticreovirusmigrationandinvivovirusdeliverypotentialforevaluatinganticancereffectintumorbearingc57bl6mice
AT sarasoudi mscsloadedwithoncolyticreovirusmigrationandinvivovirusdeliverypotentialforevaluatinganticancereffectintumorbearingc57bl6mice
AT alahabibian mscsloadedwithoncolyticreovirusmigrationandinvivovirusdeliverypotentialforevaluatinganticancereffectintumorbearingc57bl6mice
_version_ 1721492282239090688