Skip to main content
Top
Published in: Molecular Cancer 1/2018

Open Access 01-12-2018 | Research

Exosomes derived from mesenchymal stem cells enhance radiotherapy-induced cell death in tumor and metastatic tumor foci

Authors: Virgínea de Araujo Farias, Francisco O’Valle, Santiago Serrano-Saenz, Per Anderson, Eduardo Andrés, Jesús López-Peñalver, Isabel Tovar, Ana Nieto, Ana Santos, Francisco Martín, José Expósito, F. Javier Oliver, José Mariano Ruiz de Almodóvar

Published in: Molecular Cancer | Issue 1/2018

Login to get access

Abstract

Background

We have recently shown that radiotherapy may not only be a successful local and regional treatment but, when combined with MSCs, may also be a novel systemic cancer therapy. This study aimed to investigate the role of exosomes derived from irradiated MSCs in the delay of tumor growth and metastasis after treatment with MSC + radiotherapy (RT).

Methods

We have measured tumor growth and metastasis formation, of subcutaneous human melanoma A375 xenografts on NOD/SCID-gamma mice, and the response of tumors to treatment with radiotherapy (2 Gy), mesenchymal cells (MSC), mesenchymal cells plus radiotherapy, and without any treatment. Using proteomic analysis, we studied the cargo of the exosomes released by the MSC treated with 2 Gy, compared with the cargo of exosomes released by MSC without treatment.

Results

The tumor cell loss rates found after treatment with the combination of MSC and RT and for exclusive RT, were: 44.4% % and 12,1%, respectively. Concomitant and adjuvant use of RT and MSC, increased the mice surviving time 22,5% in this group, with regard to the group of mice treated with exclusive RT and in a 45,3% respect control group. Moreover, the number of metastatic foci found in the internal organs of the mice treated with MSC + RT was 60% less than the mice group treated with RT alone. We reasoned that the exosome secreted by the MSC, could be implicated in tumor growth delay and metastasis control after treatment.

Conclusions

Our results show that exosomes derived form MSCs, combined with radiotherapy, are determinant in the enhancement of radiation effects observed in the control of metastatic spread of melanoma cells and suggest that exosome-derived factors could be involved in the bystander, and abscopal effects found after treatment of the tumors with RT plus MSC. Radiotherapy itself may not be systemic, although it might contribute to a systemic effect when used in combination with mesenchymal stem cells owing the ability of irradiated MSCs-derived exosomes to increase the control of tumor growth and metastasis.
Appendix
Available only for authorised users
Literature
1.
go back to reference Atun R, Jaffray DA, Barton MB, Bray F, Baumann M, Vikram B, Hanna TP, Knaul FM, Lievens Y, Lui TY, et al. Expanding global access to radiotherapy. The Lancet Oncology. 2015;16:1153–86.CrossRefPubMed Atun R, Jaffray DA, Barton MB, Bray F, Baumann M, Vikram B, Hanna TP, Knaul FM, Lievens Y, Lui TY, et al. Expanding global access to radiotherapy. The Lancet Oncology. 2015;16:1153–86.CrossRefPubMed
2.
go back to reference Barton MB, Jacob S, Shafiq J, Wong K, Thompson SR, Hanna TP, Delaney GP. Estimating the demand for radiotherapy from the evidence: a review of changes from 2003 to 2012. Radiother Oncol. 2014;112:140–4.CrossRefPubMed Barton MB, Jacob S, Shafiq J, Wong K, Thompson SR, Hanna TP, Delaney GP. Estimating the demand for radiotherapy from the evidence: a review of changes from 2003 to 2012. Radiother Oncol. 2014;112:140–4.CrossRefPubMed
3.
go back to reference Scaife JE, Barnett GC, Noble DJ, Jena R, Thomas SJ, West CM, Burnet NG. Exploiting biological and physical determinants of radiotherapy toxicity to individualize treatment. Br J Radiol. 2015;88:20150172.CrossRefPubMedPubMedCentral Scaife JE, Barnett GC, Noble DJ, Jena R, Thomas SJ, West CM, Burnet NG. Exploiting biological and physical determinants of radiotherapy toxicity to individualize treatment. Br J Radiol. 2015;88:20150172.CrossRefPubMedPubMedCentral
4.
go back to reference Burnet NG, Wurm R, Nyman J, Peacock JH: Normal tissue radiosensitivity--how important is it? Clinical oncology (Royal College of Radiologists (Great Britain)) 1996, 8:25–34. Burnet NG, Wurm R, Nyman J, Peacock JH: Normal tissue radiosensitivity--how important is it? Clinical oncology (Royal College of Radiologists (Great Britain)) 1996, 8:25–34.
5.
go back to reference Lopez E, Guerrero R, Nunez MI, del Moral R, Villalobos M, Martinez-Galan J, Valenzuela MT, Munoz-Gamez JA, Oliver FJ, Martin-Oliva D, Ruiz de Almodovar JM. early and late skin reactions to radiotherapy for breast cancer and their correlation with radiation-induced DNA damage in lymphocytes. Breast Cancer Res. 2005;7:R690–8.CrossRefPubMedPubMedCentral Lopez E, Guerrero R, Nunez MI, del Moral R, Villalobos M, Martinez-Galan J, Valenzuela MT, Munoz-Gamez JA, Oliver FJ, Martin-Oliva D, Ruiz de Almodovar JM. early and late skin reactions to radiotherapy for breast cancer and their correlation with radiation-induced DNA damage in lymphocytes. Breast Cancer Res. 2005;7:R690–8.CrossRefPubMedPubMedCentral
6.
go back to reference Dietrich A, Koi L, Zophel K, Sihver W, Kotzerke J, Baumann M, Krause M. Improving external beam radiotherapy by combination with internal irradiation. Br J Radiol. 2015;88:20150042.CrossRefPubMedPubMedCentral Dietrich A, Koi L, Zophel K, Sihver W, Kotzerke J, Baumann M, Krause M. Improving external beam radiotherapy by combination with internal irradiation. Br J Radiol. 2015;88:20150042.CrossRefPubMedPubMedCentral
7.
go back to reference Lopez E, Nunez MI, Guerrero MR, del Moral R, de Dios LJ, del Mar RM, Valenzuela MT, Villalobos M, Ruiz de Almodovar JM. breast cancer acute radiotherapy morbidity evaluated by different scoring systems. Breast Cancer Res Treat. 2002;73:127–34.CrossRefPubMed Lopez E, Nunez MI, Guerrero MR, del Moral R, de Dios LJ, del Mar RM, Valenzuela MT, Villalobos M, Ruiz de Almodovar JM. breast cancer acute radiotherapy morbidity evaluated by different scoring systems. Breast Cancer Res Treat. 2002;73:127–34.CrossRefPubMed
8.
go back to reference Nicolson GL. Cell membrane fluid-mosaic structure and cancer metastasis. Cancer Res. 2015;75:1169–76.CrossRefPubMed Nicolson GL. Cell membrane fluid-mosaic structure and cancer metastasis. Cancer Res. 2015;75:1169–76.CrossRefPubMed
9.
go back to reference Kumar D, Gupta D, Shankar S, Srivastava RK. Biomolecular characterization of exosomes released from cancer stem cells: possible implications for biomarker and treatment of cancer. Oncotarget. 2015;6:3280–91.PubMed Kumar D, Gupta D, Shankar S, Srivastava RK. Biomolecular characterization of exosomes released from cancer stem cells: possible implications for biomarker and treatment of cancer. Oncotarget. 2015;6:3280–91.PubMed
10.
go back to reference Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A, O'Connor ST, Chin AR, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25:501–15.CrossRefPubMedPubMedCentral Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A, O'Connor ST, Chin AR, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25:501–15.CrossRefPubMedPubMedCentral
11.
12.
go back to reference Prockop DJ, Prockop SE, Bertoncello I. Are clinical trials with mesenchymal stem/progenitor cells too far ahead of the science? Lessons from experimental hematology. Stem Cells. 2014;32:3055–61.CrossRefPubMedPubMedCentral Prockop DJ, Prockop SE, Bertoncello I. Are clinical trials with mesenchymal stem/progenitor cells too far ahead of the science? Lessons from experimental hematology. Stem Cells. 2014;32:3055–61.CrossRefPubMedPubMedCentral
13.
go back to reference Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev. 2012;64:739–48.CrossRefPubMed Shah K. Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev. 2012;64:739–48.CrossRefPubMed
14.
go back to reference Zhao Q, Gregory CA, Lee RH, Reger RL, Qin L, Hai B, Park MS, Yoon N, Clough B, McNeill E, et al. MSCs derived from iPSCs with a modified protocol are tumor-tropic but have much less potential to promote tumors than bone marrow MSCs. Proc Natl Acad Sci U S A. 2015;112:530–5.CrossRefPubMed Zhao Q, Gregory CA, Lee RH, Reger RL, Qin L, Hai B, Park MS, Yoon N, Clough B, McNeill E, et al. MSCs derived from iPSCs with a modified protocol are tumor-tropic but have much less potential to promote tumors than bone marrow MSCs. Proc Natl Acad Sci U S A. 2015;112:530–5.CrossRefPubMed
15.
go back to reference Kim SM, Oh JH, Park SA, Ryu CH, Lim JY, Kim DS, Chang JW, Oh W, Jeun SS. Irradiation enhances the tumor tropism and therapeutic potential of tumor necrosis factor-related apoptosis-inducing ligand-secreting human umbilical cord blood-derived mesenchymal stem cells in glioma therapy. Stem Cells. 2010;28:2217–28.CrossRefPubMed Kim SM, Oh JH, Park SA, Ryu CH, Lim JY, Kim DS, Chang JW, Oh W, Jeun SS. Irradiation enhances the tumor tropism and therapeutic potential of tumor necrosis factor-related apoptosis-inducing ligand-secreting human umbilical cord blood-derived mesenchymal stem cells in glioma therapy. Stem Cells. 2010;28:2217–28.CrossRefPubMed
16.
go back to reference Cojocneanu Petric R, Braicu C, Raduly L, Zanoaga O, Dragos N, Monroig P, Dumitrascu D, Berindan-Neagoe I. Phytochemicals modulate carcinogenic signaling pathways in breast and hormone-related cancers. Onco Targets Ther. 2015;8:2053–66.CrossRefPubMedPubMedCentral Cojocneanu Petric R, Braicu C, Raduly L, Zanoaga O, Dragos N, Monroig P, Dumitrascu D, Berindan-Neagoe I. Phytochemicals modulate carcinogenic signaling pathways in breast and hormone-related cancers. Onco Targets Ther. 2015;8:2053–66.CrossRefPubMedPubMedCentral
17.
go back to reference Tran TH, Mattheolabakis G, Aldawsari H, Amiji M. Exosomes as nanocarriers for immunotherapy of cancer and inflammatory diseases. Clin Immunol. 2015;160:46–58.CrossRefPubMed Tran TH, Mattheolabakis G, Aldawsari H, Amiji M. Exosomes as nanocarriers for immunotherapy of cancer and inflammatory diseases. Clin Immunol. 2015;160:46–58.CrossRefPubMed
18.
go back to reference Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015; Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;
19.
go back to reference Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, Garcia-Santos G, Ghajar C, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.CrossRefPubMedPubMedCentral Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, Garcia-Santos G, Ghajar C, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.CrossRefPubMedPubMedCentral
20.
go back to reference Shin JW, Son JY, Raghavendran HR, Chung WK, Kim HG, Park HJ, Jang SS, Son CG. High-dose ionizing radiation-induced hematotoxicity and metastasis in mice model. Clinical & Experimental Metastasis. 2011;28:803–10.CrossRef Shin JW, Son JY, Raghavendran HR, Chung WK, Kim HG, Park HJ, Jang SS, Son CG. High-dose ionizing radiation-induced hematotoxicity and metastasis in mice model. Clinical & Experimental Metastasis. 2011;28:803–10.CrossRef
21.
go back to reference Klein D, Schmetter A, Imsak R, Wirsdorfer F, Unger K, Jastrow H, Stuschke M, Jendrossek V. Therapy with multipotent mesenchymal stromal cells protects lungs from radiation-induced injury and reduces the risk of lung metastasis. Antioxid Redox Signal. 2016;24:53–69.CrossRefPubMed Klein D, Schmetter A, Imsak R, Wirsdorfer F, Unger K, Jastrow H, Stuschke M, Jendrossek V. Therapy with multipotent mesenchymal stromal cells protects lungs from radiation-induced injury and reduces the risk of lung metastasis. Antioxid Redox Signal. 2016;24:53–69.CrossRefPubMed
22.
go back to reference de Araujo FV, O'Valle F, Lerma BA, Ruiz de Almodovar C, Lopez-Penalver JJ, Nieto a, Santos a, Fernandez BI, Guerra-Librero a, Ruiz-Ruiz MC, et al. human mesenchymal stem cells enhance the systemic effects of radiotherapy. Oncotarget. 2015;6:31164–80. de Araujo FV, O'Valle F, Lerma BA, Ruiz de Almodovar C, Lopez-Penalver JJ, Nieto a, Santos a, Fernandez BI, Guerra-Librero a, Ruiz-Ruiz MC, et al. human mesenchymal stem cells enhance the systemic effects of radiotherapy. Oncotarget. 2015;6:31164–80.
23.
go back to reference Gomez-Millan J, Katz IS, Farias Vde A, Linares-Fernandez JL, Lopez-Penalver J, Ortiz-Ferron G, Ruiz-Ruiz C, Oliver FJ, Ruiz de Almodovar JM. the importance of bystander effects in radiation therapy in melanoma skin-cancer cells and umbilical-cord stromal stem cells. Radiother Oncol. 2012;102:450–8.CrossRefPubMed Gomez-Millan J, Katz IS, Farias Vde A, Linares-Fernandez JL, Lopez-Penalver J, Ortiz-Ferron G, Ruiz-Ruiz C, Oliver FJ, Ruiz de Almodovar JM. the importance of bystander effects in radiation therapy in melanoma skin-cancer cells and umbilical-cord stromal stem cells. Radiother Oncol. 2012;102:450–8.CrossRefPubMed
24.
go back to reference Farias VA, Lopez-Penalver JJ, Sires-Campos J, Lopez-Ramon MV, Moreno-Castilla C, Oliver FJ, Ruiz de Almodovar JM. growth and spontaneous differentiation of umbilical-cord stromal stem cells on activated carbon cloth. J Mater Chem B. 2013;1:3359–68.CrossRef Farias VA, Lopez-Penalver JJ, Sires-Campos J, Lopez-Ramon MV, Moreno-Castilla C, Oliver FJ, Ruiz de Almodovar JM. growth and spontaneous differentiation of umbilical-cord stromal stem cells on activated carbon cloth. J Mater Chem B. 2013;1:3359–68.CrossRef
25.
go back to reference Farias VA, Linares-Fernandez JL, Penalver JL, Paya Colmenero JA, Ferron GO, Duran EL, Fernandez RM, Olivares EG, O'Valle F, Puertas A, et al. Human umbilical cord stromal stem cell express CD10 and exert contractile properties. Placenta. 2011;32:86–95.CrossRefPubMed Farias VA, Linares-Fernandez JL, Penalver JL, Paya Colmenero JA, Ferron GO, Duran EL, Fernandez RM, Olivares EG, O'Valle F, Puertas A, et al. Human umbilical cord stromal stem cell express CD10 and exert contractile properties. Placenta. 2011;32:86–95.CrossRefPubMed
26.
go back to reference Siles E, Villalobos M, Valenzuela MT, Nunez MI, Gordon A, McMillan TJ, Pedraza V, Ruiz de Almodovar JM. relationship between p53 status and radiosensitivity in human tumour cell lines. Br J Cancer. 1996;73:581–8.CrossRefPubMedPubMedCentral Siles E, Villalobos M, Valenzuela MT, Nunez MI, Gordon A, McMillan TJ, Pedraza V, Ruiz de Almodovar JM. relationship between p53 status and radiosensitivity in human tumour cell lines. Br J Cancer. 1996;73:581–8.CrossRefPubMedPubMedCentral
27.
go back to reference van der Vlist EJ, Nolte-'t Hoen EN, Stoorvogel W, Arkesteijn GJ, Wauben MH. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nat Protoc. 2012;7:1311–26.CrossRefPubMed van der Vlist EJ, Nolte-'t Hoen EN, Stoorvogel W, Arkesteijn GJ, Wauben MH. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nat Protoc. 2012;7:1311–26.CrossRefPubMed
28.
go back to reference Maitland ML, Schwartz LH, Ratain MJ. Time to tumor growth: a model end point and new metric system for oncology clinical trials. J Clin Oncol. 2013;31:2070–2.CrossRefPubMed Maitland ML, Schwartz LH, Ratain MJ. Time to tumor growth: a model end point and new metric system for oncology clinical trials. J Clin Oncol. 2013;31:2070–2.CrossRefPubMed
29.
go back to reference Claret L, Gupta M, Han K, Joshi A, Sarapa N, He J, Powell B, Bruno R. Evaluation of tumor-size response metrics to predict overall survival in western and Chinese patients with first-line metastatic colorectal cancer. J Clin Oncol. 2013;31:2110–4.CrossRefPubMed Claret L, Gupta M, Han K, Joshi A, Sarapa N, He J, Powell B, Bruno R. Evaluation of tumor-size response metrics to predict overall survival in western and Chinese patients with first-line metastatic colorectal cancer. J Clin Oncol. 2013;31:2110–4.CrossRefPubMed
30.
31.
32.
go back to reference Valeriote F, Lin H. Synergistic interaction of anticancer agents: a cellular perspective. Cancer Chemother Rep. 1975;59:895–900.PubMed Valeriote F, Lin H. Synergistic interaction of anticancer agents: a cellular perspective. Cancer Chemother Rep. 1975;59:895–900.PubMed
33.
go back to reference Thery C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J Cell Biol. 1999;147:599–610.CrossRefPubMedPubMedCentral Thery C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J Cell Biol. 1999;147:599–610.CrossRefPubMedPubMedCentral
34.
go back to reference Villalobos M, Olea N, Brotons JA, Olea-Serrano MF, Ruiz de Almodovar JM, Pedraza V. the E-screen assay: a comparison of different MCF7 cell stocks. Environ Health Perspect. 1995;103:844–50.CrossRefPubMedPubMedCentral Villalobos M, Olea N, Brotons JA, Olea-Serrano MF, Ruiz de Almodovar JM, Pedraza V. the E-screen assay: a comparison of different MCF7 cell stocks. Environ Health Perspect. 1995;103:844–50.CrossRefPubMedPubMedCentral
35.
go back to reference Haynesworth SE, Baber MA, Caplan AI. Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: effects of dexamethasone and IL-1 alpha. J Cell Physiol. 1996;166:585–92.CrossRefPubMed Haynesworth SE, Baber MA, Caplan AI. Cytokine expression by human marrow-derived mesenchymal progenitor cells in vitro: effects of dexamethasone and IL-1 alpha. J Cell Physiol. 1996;166:585–92.CrossRefPubMed
36.
go back to reference Nicolay NH, Liang YY, Perez RL, Bostel T, Trinh T, Sisombath S, Weber KJ, Ho AD, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells are resistant to carbon ion radiotherapy. Oncotarget. 2015;6:2076–87.PubMed Nicolay NH, Liang YY, Perez RL, Bostel T, Trinh T, Sisombath S, Weber KJ, Ho AD, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells are resistant to carbon ion radiotherapy. Oncotarget. 2015;6:2076–87.PubMed
37.
38.
go back to reference Loebinger MR, Sage EK, Davies D, Janes SM. TRAIL-expressing mesenchymal stem cells kill the putative cancer stem cell population. Br J Cancer. 2010;103:1692–7.CrossRefPubMedPubMedCentral Loebinger MR, Sage EK, Davies D, Janes SM. TRAIL-expressing mesenchymal stem cells kill the putative cancer stem cell population. Br J Cancer. 2010;103:1692–7.CrossRefPubMedPubMedCentral
39.
go back to reference Rager TM, Olson JK, Zhou Y, Wang Y, Besner GE. Exosomes secreted from bone marrow-derived mesenchymal stem cells protect the intestines from experimental necrotizing enterocolitis. J Pediatr Surg. 2016;51:942–7.CrossRefPubMedPubMedCentral Rager TM, Olson JK, Zhou Y, Wang Y, Besner GE. Exosomes secreted from bone marrow-derived mesenchymal stem cells protect the intestines from experimental necrotizing enterocolitis. J Pediatr Surg. 2016;51:942–7.CrossRefPubMedPubMedCentral
40.
go back to reference Weyd H. More than just innate affairs - on the role of annexins in adaptive immunity. Biol Chem. 2016;397:1017–29.CrossRefPubMed Weyd H. More than just innate affairs - on the role of annexins in adaptive immunity. Biol Chem. 2016;397:1017–29.CrossRefPubMed
Metadata
Title
Exosomes derived from mesenchymal stem cells enhance radiotherapy-induced cell death in tumor and metastatic tumor foci
Authors
Virgínea de Araujo Farias
Francisco O’Valle
Santiago Serrano-Saenz
Per Anderson
Eduardo Andrés
Jesús López-Peñalver
Isabel Tovar
Ana Nieto
Ana Santos
Francisco Martín
José Expósito
F. Javier Oliver
José Mariano Ruiz de Almodóvar
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2018
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0867-0

Other articles of this Issue 1/2018

Molecular Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine