Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2017

Open Access 01-12-2017 | Research

Exosomes isolated from cancer patients’ sera transfer malignant traits and confer the same phenotype of primary tumors to oncosuppressor-mutated cells

Authors: Mohamed Abdouh, Dana Hamam, Zu-Hua Gao, Vincenzo Arena, Manuel Arena, Goffredo Orazio Arena

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2017

Login to get access

Abstract

Background

Horizontal transfer of malignant traits from the primary tumor to distant organs, through blood circulating factors, has recently become a thoroughly studied metastatic pathway to explain cancer dissemination. Recently, we reported that oncosuppressor gene-mutated human cells undergo malignant transformation when exposed to cancer patients’ sera. We also observed that oncosuppressor mutated cells would show an increased uptake of cancer-derived exosomes and we suggested that oncosuppressor genes might protect the integrity of the cell genome by blocking integration of cancer-derived exosomes. In the present study, we tested the hypothesis that cancer patients’ sera-derived exosomes might be responsible for the malignant transformation of target cells and that oncosuppressor mutation would promote their increased uptake. We also sought to unveil the mechanisms behind the hypothesized phenomena.

Methods

We used human BRCA1 knockout (BRCA1-KO) fibroblasts as target cells. Cells were treated in vitro with cancer patients’ sera or cancer patients’ sera-derived exosomes. Treated cells were injected into NOD-SCID mice. Immunohistochemical analyses were performed to determine the differentiation state of the xenotransplants. Mass spectrometry analyses of proteins from cancer exosomes and the BRCA1-KO fibroblasts’ membrane were performed to investigate possible de novo expression of molecules involved in vesicles uptake. Blocking of the identified molecules in vitro was performed and in vivo experiments were conducted to confirm the role of these molecules in the malignant transformation carried out by cancer-derived exosomes.

Results

Cells treated with exosomes isolated from cancer patients’ sera underwent malignant transformation and formed tumors when transplanted into immunodeficient mice. Histological analyses showed that the tumors were carcinomas that differentiated into the same lineage of the primary tumors of blood donors. Oncosuppressor mutation promoted the de novo expression, on the plasma membrane of target cells, of receptors, responsible for the increased uptake of cancer-derived exosomes. The selective blocking of these receptors inhibited the horizontal transfer of malignant traits.

Conclusion

These findings strengthen the hypothesis that oncogenic factors transferred via circulating cancer exosomes, induce malignant transformation of target cells even at distance. Oncosuppressor genes might protect the integrity of the cell genome by inhibiting the uptake of cancer-derived exosomes.
Appendix
Available only for authorised users
Literature
2.
go back to reference Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.CrossRefPubMed Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.CrossRefPubMed
3.
go back to reference Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ specific colonization. Nat Rev Cancer. 2009;9:274–84.CrossRefPubMed Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ specific colonization. Nat Rev Cancer. 2009;9:274–84.CrossRefPubMed
4.
go back to reference Skog J, Wurdinger T, van Rijn S. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–81.CrossRefPubMedPubMedCentral Skog J, Wurdinger T, van Rijn S. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–81.CrossRefPubMedPubMedCentral
5.
go back to reference Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–801.CrossRefPubMed Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–801.CrossRefPubMed
6.
go back to reference Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71(15):5346–56.CrossRefPubMed Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011;71(15):5346–56.CrossRefPubMed
7.
go back to reference Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a prometastatic phenotype through MET. Nat Med. 2012;18:883–91.CrossRefPubMedPubMedCentral Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a prometastatic phenotype through MET. Nat Med. 2012;18:883–91.CrossRefPubMedPubMedCentral
8.
go back to reference Abdel-Mageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moraz K, et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells. 2014;32:983–97.CrossRefPubMedPubMedCentral Abdel-Mageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moraz K, et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells. 2014;32:983–97.CrossRefPubMedPubMedCentral
9.
go back to reference Fujita Y, Yoshioka Y, Ochiya T. Extracellular vesicle transfer of cancer pathogenic components. Cancer Sci. 2016. [DOI: 10.1111Epub ahead of print]. Fujita Y, Yoshioka Y, Ochiya T. Extracellular vesicle transfer of cancer pathogenic components. Cancer Sci. 2016. [DOI: 10.1111Epub ahead of print].
10.
go back to reference Garcıa-Olmo D, Garcıa-Olmo DC, Ontanon J, Martinez E, Vallejo M. Tumor DNA circulating in the plasma might play a role in metastasis. The hypothesis of the genometastasis. Histol Histopathol. 1999;14:1159–64.PubMed Garcıa-Olmo D, Garcıa-Olmo DC, Ontanon J, Martinez E, Vallejo M. Tumor DNA circulating in the plasma might play a role in metastasis. The hypothesis of the genometastasis. Histol Histopathol. 1999;14:1159–64.PubMed
11.
go back to reference García-Olmo DC, Domínguez C, García-Arranz M, Anker P, Stroun M, García-Verdugo JM, et al. Cell-free nucleic acids circulating in the plasma of colorectal cancer patients induce the oncogenic transformation of susceptible cultured cells. Cancer Res. 2010;70:560–7.CrossRefPubMed García-Olmo DC, Domínguez C, García-Arranz M, Anker P, Stroun M, García-Verdugo JM, et al. Cell-free nucleic acids circulating in the plasma of colorectal cancer patients induce the oncogenic transformation of susceptible cultured cells. Cancer Res. 2010;70:560–7.CrossRefPubMed
12.
go back to reference Trejo-Becerril C, Perez-Cardenas E, Taja-Chayeb L, Anker P, Herrera-Goepfert R, Medina-Velazquez LA, et al. Cancer Progression Mediated by Horizontal Gene Transfer in an In Vivo Model. PLOS ONE. 2012;7(12):e52754.CrossRefPubMedPubMedCentral Trejo-Becerril C, Perez-Cardenas E, Taja-Chayeb L, Anker P, Herrera-Goepfert R, Medina-Velazquez LA, et al. Cancer Progression Mediated by Horizontal Gene Transfer in an In Vivo Model. PLOS ONE. 2012;7(12):e52754.CrossRefPubMedPubMedCentral
13.
go back to reference Abdouh M, Zhou S, Arena V, Arena M, Lazaris A, Onerheim R, et al. Transfer of malignant trait to immortalized human cells following exposure to human cancer serum. J Exp Clin Cancer Res. 2014;33(1):86.CrossRefPubMedPubMedCentral Abdouh M, Zhou S, Arena V, Arena M, Lazaris A, Onerheim R, et al. Transfer of malignant trait to immortalized human cells following exposure to human cancer serum. J Exp Clin Cancer Res. 2014;33(1):86.CrossRefPubMedPubMedCentral
14.
go back to reference Hamam D, Abdouh M, Gao ZH, Arena V, Arena M, Arena GO. Transfer of malignant trait to BRCA1 deficient human fibroblasts following exposure to serum of cancer patients. J Exp Clin Cancer Res. 2016;35:80.CrossRefPubMedPubMedCentral Hamam D, Abdouh M, Gao ZH, Arena V, Arena M, Arena GO. Transfer of malignant trait to BRCA1 deficient human fibroblasts following exposure to serum of cancer patients. J Exp Clin Cancer Res. 2016;35:80.CrossRefPubMedPubMedCentral
15.
go back to reference Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9.CrossRefPubMed Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9.CrossRefPubMed
16.
go back to reference Runz S, Keller S, Rupp C, Stoeck A, Issa Y, Koensgen D. Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM. Gynecol Oncol. 2007;107:563–71.CrossRefPubMed Runz S, Keller S, Rupp C, Stoeck A, Issa Y, Koensgen D. Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM. Gynecol Oncol. 2007;107:563–71.CrossRefPubMed
17.
go back to reference Pisetsky DS, Gauley J, Ullal AJ. Microparticles as a source of extracellular DNA. Immunol Res. 2010;49:227–34.CrossRef Pisetsky DS, Gauley J, Ullal AJ. Microparticles as a source of extracellular DNA. Immunol Res. 2010;49:227–34.CrossRef
18.
go back to reference Subra C, Grand D, Laulagnier K, Stella A, Lambeau G, Paillasse M, et al. Exosomes account for vesicle mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 2010;51:2105–20.CrossRefPubMedPubMedCentral Subra C, Grand D, Laulagnier K, Stella A, Lambeau G, Paillasse M, et al. Exosomes account for vesicle mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 2010;51:2105–20.CrossRefPubMedPubMedCentral
19.
go back to reference Gaiffe E, Pretet JL, Launay S, Jacquin L, Saunier M, Hetzel G. Apoptotic HPV positive cancer cells exhibit transforming properties. PLoS One. 2012;7:e36766.CrossRefPubMedPubMedCentral Gaiffe E, Pretet JL, Launay S, Jacquin L, Saunier M, Hetzel G. Apoptotic HPV positive cancer cells exhibit transforming properties. PLoS One. 2012;7:e36766.CrossRefPubMedPubMedCentral
20.
go back to reference Balaj L, Lessard R, Dai L, Cho YJ, Pomeroy SL, Breakefield XO. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180.CrossRefPubMedPubMedCentral Balaj L, Lessard R, Dai L, Cho YJ, Pomeroy SL, Breakefield XO. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011;2:180.CrossRefPubMedPubMedCentral
21.
go back to reference Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007;1775(1):181–232.PubMed Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007;1775(1):181–232.PubMed
22.
go back to reference Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79.PubMed Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79.PubMed
23.
24.
go back to reference Candelario KM, Steindler DA. The role of extracellular vesicles in the progression of neurodegenerative disease and cancer. Trends Mol Med. 2014;20(7):368–74.CrossRefPubMedPubMedCentral Candelario KM, Steindler DA. The role of extracellular vesicles in the progression of neurodegenerative disease and cancer. Trends Mol Med. 2014;20(7):368–74.CrossRefPubMedPubMedCentral
25.
go back to reference Pant S, Hilton H, Burczynski ME. The multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker oppertunities. Biochem Pharmacol. 2012;83(11):1484–94.CrossRefPubMed Pant S, Hilton H, Burczynski ME. The multifaceted exosome: biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker oppertunities. Biochem Pharmacol. 2012;83(11):1484–94.CrossRefPubMed
26.
go back to reference Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.CrossRefPubMed Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.CrossRefPubMed
28.
go back to reference D’Asti E, Chennakrishnaiah S, Lee TH, Rak J. Extracellular vesicles in brain tumor progression. Cell Mol Neurobiol. 2016;36(3):383–407.CrossRefPubMed D’Asti E, Chennakrishnaiah S, Lee TH, Rak J. Extracellular vesicles in brain tumor progression. Cell Mol Neurobiol. 2016;36(3):383–407.CrossRefPubMed
29.
go back to reference Soldevilla B, Rodriguez M, San Millan C, Garcia V, Fernandez-Perianez R, Gil-Calderon B, et al. Tumor-derived exosomes are enriched in DNp73, which promotes oncogenic potential in acceptor cells and correlates with patient survival. Hum Mol Genet. 2014;23(2):467–78.CrossRefPubMed Soldevilla B, Rodriguez M, San Millan C, Garcia V, Fernandez-Perianez R, Gil-Calderon B, et al. Tumor-derived exosomes are enriched in DNp73, which promotes oncogenic potential in acceptor cells and correlates with patient survival. Hum Mol Genet. 2014;23(2):467–78.CrossRefPubMed
30.
go back to reference Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Kosh M, et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNS in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014;289(7):3869–75.CrossRefPubMedPubMedCentral Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Kosh M, et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNS in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 2014;289(7):3869–75.CrossRefPubMedPubMedCentral
31.
go back to reference Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24:766–9.CrossRefPubMedPubMedCentral Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24:766–9.CrossRefPubMedPubMedCentral
32.
go back to reference Antonyak MA, Li B, Boroughs LK, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci U S A. 2011;108:4852–7.CrossRefPubMedPubMedCentral Antonyak MA, Li B, Boroughs LK, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci U S A. 2011;108:4852–7.CrossRefPubMedPubMedCentral
33.
go back to reference He M, Qin H, Poon TC, Sze SC, Ding X, Co NN, et al. Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis. 2015;36(9):1008–18.CrossRefPubMed He M, Qin H, Poon TC, Sze SC, Ding X, Co NN, et al. Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis. 2015;36(9):1008–18.CrossRefPubMed
34.
go back to reference Fanz Z, van de Rijn M, Montgomery K, Rouse RV. Hep par 1 antibody stain for the differential diagnosis of hepatocellular carcinoma: 676 tumors tested using tissue microarrays and conventional tissue sections. Mod Pathol. 2003;16(2):137–44.CrossRef Fanz Z, van de Rijn M, Montgomery K, Rouse RV. Hep par 1 antibody stain for the differential diagnosis of hepatocellular carcinoma: 676 tumors tested using tissue microarrays and conventional tissue sections. Mod Pathol. 2003;16(2):137–44.CrossRef
35.
go back to reference Kakar S, Muir T, Murphy LM, Lloyd RV, Burgart LJ. Immunoreactivity of Hep par 1 in hepatic and extrahepatic tumors and its correlation with albumin in situ hybridization in hepatocellular carcinoma. Am J Clin Pathol. 2003;119:361–6.CrossRefPubMed Kakar S, Muir T, Murphy LM, Lloyd RV, Burgart LJ. Immunoreactivity of Hep par 1 in hepatic and extrahepatic tumors and its correlation with albumin in situ hybridization in hepatocellular carcinoma. Am J Clin Pathol. 2003;119:361–6.CrossRefPubMed
36.
go back to reference Lykke-Andersen S, Brodersen DE, Jensen TH. Origins and activities of the eukaryotic exosome. J Cell Sci. 2009;122(Pt 10):1487–94.CrossRefPubMed Lykke-Andersen S, Brodersen DE, Jensen TH. Origins and activities of the eukaryotic exosome. J Cell Sci. 2009;122(Pt 10):1487–94.CrossRefPubMed
37.
go back to reference Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20(9):1487–95.CrossRefPubMed Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20(9):1487–95.CrossRefPubMed
38.
go back to reference Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic M. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.CrossRefPubMedPubMedCentral Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic M. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35.CrossRefPubMedPubMedCentral
40.
go back to reference Kastelowitz N, Yin H. Exosomes and microvesicles: identification and targeting by particle size and lipid chemical probes. Chembiochem. 2014;15(7):923–8.CrossRefPubMedPubMedCentral Kastelowitz N, Yin H. Exosomes and microvesicles: identification and targeting by particle size and lipid chemical probes. Chembiochem. 2014;15(7):923–8.CrossRefPubMedPubMedCentral
41.
go back to reference Hosseini-Beheshti E, Pham S, Adomat H, Li N, Tomlinson Guns ES. Exosomes as biomarker enriched microvesicles: characterization of exosomal proteins derived from a panel of prostate cell lines with distinct AR phenotypes. Mol Cell Proteomics. 2012;11(10):863–85.CrossRefPubMedPubMedCentral Hosseini-Beheshti E, Pham S, Adomat H, Li N, Tomlinson Guns ES. Exosomes as biomarker enriched microvesicles: characterization of exosomal proteins derived from a panel of prostate cell lines with distinct AR phenotypes. Mol Cell Proteomics. 2012;11(10):863–85.CrossRefPubMedPubMedCentral
42.
go back to reference McKelvey KJ, Powell KL, Ashton AW, Morris JM, McCracken SA. Exosomes: mechanisms of uptake. J Circ Biomark. 2015;4:1–9.CrossRef McKelvey KJ, Powell KL, Ashton AW, Morris JM, McCracken SA. Exosomes: mechanisms of uptake. J Circ Biomark. 2015;4:1–9.CrossRef
43.
go back to reference Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:2464.CrossRef Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:2464.CrossRef
44.
go back to reference Arena G, Arena V, Arena M, Abdouh M. Transfer of malignant traits as opposed to migration of cells: a novel concept to explain metastatic disease. Med Hypotheses. 2017;100:82–6.CrossRefPubMed Arena G, Arena V, Arena M, Abdouh M. Transfer of malignant traits as opposed to migration of cells: a novel concept to explain metastatic disease. Med Hypotheses. 2017;100:82–6.CrossRefPubMed
45.
go back to reference Kawada M, Amemiya M, Ishizuka M, Takeuchi T. Cytostatin, an inhibitor of cell adhesion to extracellular matrix, selectively inhibits protein phosphatase 2A. Biochim Biophys Acta. 1999;1452(2):209–17.CrossRefPubMed Kawada M, Amemiya M, Ishizuka M, Takeuchi T. Cytostatin, an inhibitor of cell adhesion to extracellular matrix, selectively inhibits protein phosphatase 2A. Biochim Biophys Acta. 1999;1452(2):209–17.CrossRefPubMed
46.
go back to reference Christianson HC, Svensson KJ, van Kuppevelt TH, Li JP, Belting M. Cancer cell exosomes depend on cell-surface heparin sulfate proteoglycans for their internalization and functional activity. PNAS. 2013;110(43):17380–5.CrossRefPubMedPubMedCentral Christianson HC, Svensson KJ, van Kuppevelt TH, Li JP, Belting M. Cancer cell exosomes depend on cell-surface heparin sulfate proteoglycans for their internalization and functional activity. PNAS. 2013;110(43):17380–5.CrossRefPubMedPubMedCentral
47.
go back to reference Abdouh M, Hamam D, Arena V, Arena M, Alamri H, Arena G. Novel blood test to predict neoplastic activity in healthy patients and metastatic recurrence after primary tumor resection. Journal of Circulating Biomarkers. 2016;5:1–10. Abdouh M, Hamam D, Arena V, Arena M, Alamri H, Arena G. Novel blood test to predict neoplastic activity in healthy patients and metastatic recurrence after primary tumor resection. Journal of Circulating Biomarkers. 2016;5:1–10.
48.
go back to reference Barreto S, Pandanaboyana S, Ironside N, Windsor J. Does revision of resection margins based on frozen section improve overall survival following pancreaticoduodenectomy for pancreatic ductal adenocarcinoma? A meta-analysis. HPB (Oxford). 2017; (17): 30534–30548. Barreto S, Pandanaboyana S, Ironside N, Windsor J. Does revision of resection margins based on frozen section improve overall survival following pancreaticoduodenectomy for pancreatic ductal adenocarcinoma? A meta-analysis. HPB (Oxford). 2017; (17): 30534–30548.
49.
go back to reference Ribelles N, Santonja A, Pajares B, Llacer C, Alba E. The seed and soil hypothesis revisited: current state of knowledge of inherited genes on prognosis in breast cancer. Cancer Treat Rev. 2014;40:293–9.CrossRefPubMed Ribelles N, Santonja A, Pajares B, Llacer C, Alba E. The seed and soil hypothesis revisited: current state of knowledge of inherited genes on prognosis in breast cancer. Cancer Treat Rev. 2014;40:293–9.CrossRefPubMed
50.
go back to reference Luzzi K, MacDonald I, Schmidt E, Kerkvliet N, Morris V, et al. Multistep nature of metastatic inefficiency. Am J Pathology. 1998;153(3):865–73.CrossRef Luzzi K, MacDonald I, Schmidt E, Kerkvliet N, Morris V, et al. Multistep nature of metastatic inefficiency. Am J Pathology. 1998;153(3):865–73.CrossRef
51.
go back to reference Suzuki M, Tanin D. Gene expression profiling of human lynph node metastases and matched primary breast carcinomas: clinical implications. Mol Oncol. 2007;1(2):172–80.CrossRefPubMedPubMedCentral Suzuki M, Tanin D. Gene expression profiling of human lynph node metastases and matched primary breast carcinomas: clinical implications. Mol Oncol. 2007;1(2):172–80.CrossRefPubMedPubMedCentral
52.
go back to reference Huang S, Chen Y, Podsypanina K, Li Y. Comparison of expression profiles of metastatic versus primary mammary tumors in MMTV-Wnt-1 and MMTV-Neu transgenic mice. Neoplasia. 2008;10(2):118–24.CrossRefPubMedPubMedCentral Huang S, Chen Y, Podsypanina K, Li Y. Comparison of expression profiles of metastatic versus primary mammary tumors in MMTV-Wnt-1 and MMTV-Neu transgenic mice. Neoplasia. 2008;10(2):118–24.CrossRefPubMedPubMedCentral
54.
go back to reference Yoshida A, Okamoto N, Tozawa-Ono A, KoizUmi H, Kiguchi K, Ishizuka B, et al. Proteomic analysis of differential protein expression by brain metastases of gynecological malignancies. Hum Cell. 2013;26(2):56–66.CrossRefPubMedPubMedCentral Yoshida A, Okamoto N, Tozawa-Ono A, KoizUmi H, Kiguchi K, Ishizuka B, et al. Proteomic analysis of differential protein expression by brain metastases of gynecological malignancies. Hum Cell. 2013;26(2):56–66.CrossRefPubMedPubMedCentral
55.
go back to reference Qian W, Wang J, Van Houten B. The role of dynamin-related protein 1 in cancer growth: a promising therapeutic target? Expert Opin Ther Targets. 2013;17(9):997–1001.CrossRefPubMedPubMedCentral Qian W, Wang J, Van Houten B. The role of dynamin-related protein 1 in cancer growth: a promising therapeutic target? Expert Opin Ther Targets. 2013;17(9):997–1001.CrossRefPubMedPubMedCentral
56.
go back to reference Sindrewicz P, Lian LY, Yu LG. Interaction of the Oncofetal Thomsen-Friedenreich antigen with galectins in cancer progression and metastasis. Front Oncol. 2016;6:79.CrossRefPubMedPubMedCentral Sindrewicz P, Lian LY, Yu LG. Interaction of the Oncofetal Thomsen-Friedenreich antigen with galectins in cancer progression and metastasis. Front Oncol. 2016;6:79.CrossRefPubMedPubMedCentral
58.
go back to reference Dai M, Yuan F, Fu C, Shen G, Hu S, Shen G. Relationship between epithelial cell adhesion molecule (EpCAM) overexpression and gastric cancer patients: a systematic review and meta-analysis. PLoS One. 2017;12(4):e0175357.CrossRefPubMedPubMedCentral Dai M, Yuan F, Fu C, Shen G, Hu S, Shen G. Relationship between epithelial cell adhesion molecule (EpCAM) overexpression and gastric cancer patients: a systematic review and meta-analysis. PLoS One. 2017;12(4):e0175357.CrossRefPubMedPubMedCentral
Metadata
Title
Exosomes isolated from cancer patients’ sera transfer malignant traits and confer the same phenotype of primary tumors to oncosuppressor-mutated cells
Authors
Mohamed Abdouh
Dana Hamam
Zu-Hua Gao
Vincenzo Arena
Manuel Arena
Goffredo Orazio Arena
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2017
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-017-0587-0

Other articles of this Issue 1/2017

Journal of Experimental & Clinical Cancer Research 1/2017 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