Tumor Tropism of Mesenchymal Stem Cells

2013 ◽  
pp. 21-38 ◽  
Author(s):  
Paula Y. P. Lam ◽  
Ivy A. W. Ho
2007 ◽  
Vol 18 (7) ◽  
pp. 627-641 ◽  
Author(s):  
Tanja Hakkarainen ◽  
Merja Särkioja ◽  
Petri Lehenkari ◽  
Susanna Miettinen ◽  
Timo Ylikomi ◽  
...  

2018 ◽  
Vol 2018 ◽  
pp. 1-14 ◽  
Author(s):  
Jun Zhou ◽  
Tian Liang ◽  
Dejun Wang ◽  
Liru Li ◽  
Yan Cheng ◽  
...  

Background. Immunotherapy for cervical cancer with type I interferon (IFN) is limited because of the cytotoxicity that accompanies the high doses that are administered. In this study, we investigated the utilization of amniotic fluid-derived mesenchymal stem cells (AF-MSCs) as a means for delivering IFNα to local tumor sites for the suppression of cervical cancer in a mouse model using HeLa cell xenografts. Methods. The tumor tropism ability of AF-MSCs and AF-MSCs genetically modified to overexpress IFNα (IFNα-AF-MSCs) was examined through Transwell in vitro and through fluorescent images and immunohistochemistry in a mouse model. Tumor size and tumor apoptosis were observed to evaluate the efficacy of the targeting therapy. Mechanistically, tumor cell apoptosis was detected by cytometry and TUNEL, and oncogenic proteins c-Myc, p53, and Bcl-2 as well as microvessel density were detected by immunohistochemistry. Results. In this model, intravenously injected AF-MSCs selectively migrated to the tumor sites, participated in tumor construction, and promoted tumor growth. After being genetically modified to overexpress IFNα, the IFNα-AF-MSCs maintained their tumor tropism but could significantly suppress tumor growth. The restrictive efficacy of IFNα-AF-MSCs was associated with the suppression of angiogenesis, inhibition of tumor cell proliferation, and induction of apoptosis in tumor cells. Neither AF-MSCs nor IFNα-AF-MSCs trigger tumor formation. Conclusions. IFNα-AF-MSC-based therapy is feasible and shows potential for treating cervical cancer, suggesting that AF-MSCs may be promising vehicles for delivering targeted anticancer therapy.


RSC Advances ◽  
2016 ◽  
Vol 6 (51) ◽  
pp. 45553-45561 ◽  
Author(s):  
Tsai-Hua Chung ◽  
Chia-Chu Hsieh ◽  
Jong-Kai Hsiao ◽  
Szu-Chun Hsu ◽  
Ming Yao ◽  
...  

dex-IO NPs can activate the antitumor mechanism (tumor tropism) but inactivate protumor mechanisms to transform protumor MSCs (pT-MSCs) into antitumor MSCs (aT-MSCs).


Biomedicines ◽  
2021 ◽  
Vol 9 (11) ◽  
pp. 1592
Author(s):  
Natalia Yudintceva ◽  
Ekaterina Lomert ◽  
Natalia Mikhailova ◽  
Elena Tolkunova ◽  
Nikol Agadzhanian ◽  
...  

Despite multimodal approaches for the treatment of multiforme glioblastoma (GBM) advances in outcome have been very modest indicating the necessity of novel diagnostic and therapeutic strategies. Currently, mesenchymal stem cells (MSCs) represent a promising platform for cell-based cancer therapies because of their tumor-tropism, low immunogenicity, easy accessibility, isolation procedure, and culturing. In the present study, we assessed the tumor-tropism and biodistribution of the superparamagnetic iron oxide nanoparticle (SPION)-labeled MSCs in the orthotopic model of C6 glioblastoma in Wistar rats. As shown in in vitro studies employing confocal microscopy, high-content quantitative image cytometer, and xCelligence system MSCs exhibit a high migratory capacity towards C6 glioblastoma cells. Intravenous administration of SPION-labeled MSCs in vivo resulted in intratumoral accumulation of the tagged cells in the tumor tissues that in turn significantly enhanced the contrast of the tumor when high-field magnetic resonance imaging was performed. Subsequent biodistribution studies employing highly sensitive nonlinear magnetic response measurements (NLR-M2) supported by histological analysis confirm the retention of MSCs in the glioblastoma. In conclusion, MSCs due to their tumor-tropism could be employed as a drug-delivery platform for future theranostic approaches.


2020 ◽  
Vol 17 ◽  
Author(s):  
Mengying Xie ◽  
Lei Tao ◽  
Ziqi Zhang ◽  
Wei Wang

: Mesenchymal stem cells (MSCs) possess unique properties that make them potential carriers for cancer therapy. MSCs have been documented to have low immunogenicity, positive safety in clinical trials, and the ability to selectively homing to inflammation and tumor sites. Thisreview aims to introduce tumor tropism mechanism and effects of MSCs on tumor cells, and give an overview of MSCs in delivering gene therapeutic agents, oncolytic viruses and chemotherapeutics, as well as the application of MSCs-derived exosomes in tumor-targeted therapy.


Oncotarget ◽  
2019 ◽  
Vol 10 (58) ◽  
pp. 6049-6061 ◽  
Author(s):  
Lasse Dührsen ◽  
Sophie Hartfuß ◽  
Daniela Hirsch ◽  
Sabine Geiger ◽  
Cecile L. Maire ◽  
...  

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Zhijie Weng ◽  
Bowen Zhang ◽  
Chenzhou Wu ◽  
Fanyuan Yu ◽  
Bo Han ◽  
...  

AbstractExtracellular vesicles (EVs) are cell-derived membrane structures enclosing proteins, lipids, RNAs, metabolites, growth factors, and cytokines. EVs have emerged as essential intercellular communication regulators in multiple physiological and pathological processes. Previous studies revealed that mesenchymal stem cells (MSCs) could either support or suppress tumor progression in different cancers by paracrine signaling via MSC-derived EVs. Evidence suggested that MSC-derived EVs could mimic their parental cells, possessing pro-tumor and anti-tumor effects, and inherent tumor tropism. Therefore, MSC-derived EVs can be a cell-free cancer treatment alternative. This review discusses different insights regarding MSC-derived EVs' roles in cancer treatment and summarizes bioengineered MSC-derived EVs’ applications as safe and versatile anti-tumor agent delivery platforms. Meanwhile, current hurdles of moving MSC-derived EVs from bench to bedside are also discussed.


Sign in / Sign up

Export Citation Format

Share Document