Skip to main content
Top
Published in: Cellular Oncology 2/2020

01-04-2020 | Metastasis | Review

Tumor dormancy as an alternative step in the development of chemoresistance and metastasis - clinical implications

Authors: Federico Rossari, Cristina Zucchinetti, Gabriele Buda, Enrico Orciuolo

Published in: Cellular Oncology | Issue 2/2020

Login to get access

Abstract

Background

The ability of a tumor to become dormant in response to suboptimal conditions has recently been recognized as a key step in tumor progression. Tumor dormancy has been found to be implicated in several tumor types as the culprit of therapy resistance and metastasis development, the deadliest features of a cancer. Several lines of evidence indicate that the development of these traits may rely on the de-differentiation of committed tumor cells that regain stem-like properties during a dormant state. Presently, dormancy is classified into cell- and population-level, according to the preponderance of cellular mechanisms that keep tumor cells quiescent or to a balance between overall cell division and death, respectively. Cellular dormancy is characterized by autophagy, stress-tolerance signaling, microenvironmental cues and, of prime relevance, epigenetic modifications. It has been found that the epigenome alters during cellular quiescence, thus representing the driving force for short-term cancer progression. Population-level dormancy is characterized by processes that counteract proliferation, such as inappropriate blood supply and intense immune responses. The latter two mechanisms are not mutually exclusive and may affect tumor masses both simultaneously and subsequently.

Conclusions

Overall, tumor dormancy may represent an additional step in the acquisition of cancer characteristics, and its comprehension may clarify both theoretical and practical aspects of cancer development. Clinically, only a deep understanding of dormancy may explain the course of tumor development in different patients, thus representing a process that may be targeted to prevent and/or treat advanced-stage cancers. That is especially the case for breast cancer, against which the mTOR inhibitor everolimus displays potent antitumor activity in patients with metastatic disease by impeding autophagy and tumor dormancy onset. Here we will also discuss other targeted therapies directed towards tumor dormancy onset, e.g. specific inhibitors of SFK and MEK, or aimed at keeping tumor cells dormant, e.g. prosaposin derivatives, that may shortly enter clinical assessment in breast, and possibly other cancer types.
Literature
2.
3.
4.
go back to reference L. Tong, L. Yi, P. Liu, I.R. Abeysekera, L. Hai, T. Li, Z. Tao, H. Ma, Y. Xie, Y. Huang, S. Yu, J. Li, F. Yuan, X. Yang, Tumour cell dormancy as a contributor to the reduced survival of GBM patients who received standard therapy. Oncol. Rep. 40, 463–471 (2018)PubMed L. Tong, L. Yi, P. Liu, I.R. Abeysekera, L. Hai, T. Li, Z. Tao, H. Ma, Y. Xie, Y. Huang, S. Yu, J. Li, F. Yuan, X. Yang, Tumour cell dormancy as a contributor to the reduced survival of GBM patients who received standard therapy. Oncol. Rep. 40, 463–471 (2018)PubMed
5.
go back to reference D.M. Schewe, J.A. Aguirre-Ghiso, ATF6α-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo. Proc. Natl. Acad. Sci. U. S. A. 105, 10519–10524 (2008) D.M. Schewe, J.A. Aguirre-Ghiso, ATF6α-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo. Proc. Natl. Acad. Sci. U. S. A. 105, 10519–10524 (2008)
6.
go back to reference X. Deng, D.Z. Ewton, E. Friedman, Mirk dyrk1B maintains the viability of quiescent pancreatic cancer cells by reducing levels of reactive oxygen species. Cancer Res. 69, 3317–3324 (2009)CrossRefPubMedPubMedCentral X. Deng, D.Z. Ewton, E. Friedman, Mirk dyrk1B maintains the viability of quiescent pancreatic cancer cells by reducing levels of reactive oxygen species. Cancer Res. 69, 3317–3324 (2009)CrossRefPubMedPubMedCentral
7.
go back to reference P. Walter, D. Ron, The unfolded protein response: From stress pathway to homeostatic regulation. Science 334, 1081–1086 (2011)CrossRefPubMed P. Walter, D. Ron, The unfolded protein response: From stress pathway to homeostatic regulation. Science 334, 1081–1086 (2011)CrossRefPubMed
8.
go back to reference F. Crea, N.R. Nur Saidy, C.C. Collins, Y. Wang, The epigenetic/noncoding origin of tumor dormancy. Trends Mol. Med. 21, 206–211 (2015)CrossRefPubMed F. Crea, N.R. Nur Saidy, C.C. Collins, Y. Wang, The epigenetic/noncoding origin of tumor dormancy. Trends Mol. Med. 21, 206–211 (2015)CrossRefPubMed
9.
go back to reference P. Hahnfeldt, The host support niche as a control point for tumor dormancy: Implications for tumor development and beyond. Adv. Exp. Med. Biol. 734, 19–35 (2013)CrossRefPubMed P. Hahnfeldt, The host support niche as a control point for tumor dormancy: Implications for tumor development and beyond. Adv. Exp. Med. Biol. 734, 19–35 (2013)CrossRefPubMed
10.
go back to reference J.W. Uhr, K. Pantel, Controversies in clinical cancer dormancy. Proc. Natl. Acad. Sci. U. S. A. 108, 12396–12400 (2011) J.W. Uhr, K. Pantel, Controversies in clinical cancer dormancy. Proc. Natl. Acad. Sci. U. S. A. 108, 12396–12400 (2011)
12.
13.
go back to reference J.A. Krall, F. Reinhardt, O.A. Mercury, D.R. Pattabiraman, M.W. Brooks, M. Dougan, A.W. Lambert, B. Bierie, H.L. Ploegh, S.K. Dougan, R.A. Weinberg, The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci. Transl. Med. 10, eean3464 (2018) J.A. Krall, F. Reinhardt, O.A. Mercury, D.R. Pattabiraman, M.W. Brooks, M. Dougan, A.W. Lambert, B. Bierie, H.L. Ploegh, S.K. Dougan, R.A. Weinberg, The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci. Transl. Med. 10, eean3464 (2018)
14.
go back to reference J. Pfitzenmaier, W.J. Ellis, E.W. Arfman, S. Hawley, P.O. Mclaughlin, P.H. Lange, R.L. Vessella, Telomerase activity in disseminated prostate cancer cells. BJU Int. 97, 1309–1313 (2006)CrossRefPubMed J. Pfitzenmaier, W.J. Ellis, E.W. Arfman, S. Hawley, P.O. Mclaughlin, P.H. Lange, R.L. Vessella, Telomerase activity in disseminated prostate cancer cells. BJU Int. 97, 1309–1313 (2006)CrossRefPubMed
15.
go back to reference X.H.-F. Zhang, Q. Wang, W. Gerald, C.A. Hudis, L. Norton, M. Smid, J.A. Foekens, J. Massagué, Latent bone metastasis in breast Cancer tied to Src-dependent survival signals. Cancer Cell 16, 67–78 (2009)CrossRefPubMedPubMedCentral X.H.-F. Zhang, Q. Wang, W. Gerald, C.A. Hudis, L. Norton, M. Smid, J.A. Foekens, J. Massagué, Latent bone metastasis in breast Cancer tied to Src-dependent survival signals. Cancer Cell 16, 67–78 (2009)CrossRefPubMedPubMedCentral
17.
go back to reference T. Lyu, N. Jia, J. Wang, X. Yan, Y. Yu, Z. Lu, R.C. Bast, K. Hua, W. Feng, Expression and epigenetic regulation of angiogenesis-related factors during dormancy and recurrent growth of ovarian carcinoma. Epigenetics 8, 1330–1346 (2013)CrossRefPubMedPubMedCentral T. Lyu, N. Jia, J. Wang, X. Yan, Y. Yu, Z. Lu, R.C. Bast, K. Hua, W. Feng, Expression and epigenetic regulation of angiogenesis-related factors during dormancy and recurrent growth of ovarian carcinoma. Epigenetics 8, 1330–1346 (2013)CrossRefPubMedPubMedCentral
18.
go back to reference H. Easwaran, H.-C. Tsai, S.B. Baylin, Cancer epigenetics: Tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol. Cell 54, 716–727 (2014)CrossRefPubMedPubMedCentral H. Easwaran, H.-C. Tsai, S.B. Baylin, Cancer epigenetics: Tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol. Cell 54, 716–727 (2014)CrossRefPubMedPubMedCentral
19.
go back to reference D. Gao, D.J. Nolan, A.S. Mellick, K. Bambino, K. McDonnell, V. Mittal, Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science 319, 195–198 (2008)CrossRefPubMed D. Gao, D.J. Nolan, A.S. Mellick, K. Bambino, K. McDonnell, V. Mittal, Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science 319, 195–198 (2008)CrossRefPubMed
20.
go back to reference M. Elkabets, A.M. Gifford, C. Scheel, B. Nilsson, F. Reinhardt, M.A. Bray, A.E. Carpenter, K. Jirström, K. Magnusson, B.L. Ebert, F. Pontén, R.A. Weinberg, S.S. McAllister, Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J. Clin. Invest. 121, 784–799 (2011)CrossRefPubMedPubMedCentral M. Elkabets, A.M. Gifford, C. Scheel, B. Nilsson, F. Reinhardt, M.A. Bray, A.E. Carpenter, K. Jirström, K. Magnusson, B.L. Ebert, F. Pontén, R.A. Weinberg, S.S. McAllister, Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J. Clin. Invest. 121, 784–799 (2011)CrossRefPubMedPubMedCentral
21.
go back to reference D. Barkan, L.H. El Touny, A.M. Michalowski, J.A. Smith, I. Chu, A.S. Davis, J.D. Webster, S. Hoover, R.M. Simpson, J. Gauldie, J.E. Green, Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 70, 5706–5716 (2010)CrossRefPubMedPubMedCentral D. Barkan, L.H. El Touny, A.M. Michalowski, J.A. Smith, I. Chu, A.S. Davis, J.D. Webster, S. Hoover, R.M. Simpson, J. Gauldie, J.E. Green, Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 70, 5706–5716 (2010)CrossRefPubMedPubMedCentral
24.
go back to reference D.J. Weisenberger, K.D. Siegmund, M. Campan, J. Young, T.I. Long, M.A. Faasse, G.H. Kang, M. Widschwendter, D. Weener, D. Buchanan, H. Koh, L. Simms, M. Barker, B. Leggett, J. Levine, M. Kim, A.J. French, S.N. Thibodeau, J. Jass, R. Haile, P.W. Laird, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat. Genet. 38, 787–793 (2006)CrossRefPubMed D.J. Weisenberger, K.D. Siegmund, M. Campan, J. Young, T.I. Long, M.A. Faasse, G.H. Kang, M. Widschwendter, D. Weener, D. Buchanan, H. Koh, L. Simms, M. Barker, B. Leggett, J. Levine, M. Kim, A.J. French, S.N. Thibodeau, J. Jass, R. Haile, P.W. Laird, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat. Genet. 38, 787–793 (2006)CrossRefPubMed
26.
go back to reference B.T. Tan, C.Y. Park, L.E. Ailles, I.L. Weissman, The cancer stem cell hypothesis: A work in progress. Lab. Investig. 86, 1203–1207 (2006)CrossRefPubMed B.T. Tan, C.Y. Park, L.E. Ailles, I.L. Weissman, The cancer stem cell hypothesis: A work in progress. Lab. Investig. 86, 1203–1207 (2006)CrossRefPubMed
27.
go back to reference N.D. Marjanovic, R.A. Weinberg, C.L. Chaffer, Cell plasticity and heterogeneity in cancer. Clin. Chem. 59, 168–179 (2013)CrossRefPubMed N.D. Marjanovic, R.A. Weinberg, C.L. Chaffer, Cell plasticity and heterogeneity in cancer. Clin. Chem. 59, 168–179 (2013)CrossRefPubMed
28.
go back to reference S.A. Mani, W. Guo, M.J. Liao, E.N. Eaton, A. Ayyanan, A.Y. Zhou, M. Brooks, F. Reinhard, C.C. Zhang, M. Shipitsin, L.L. Campbell, K. Polyak, C. Brisken, J. Yang, R.A. Weinberg, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008)CrossRefPubMedPubMedCentral S.A. Mani, W. Guo, M.J. Liao, E.N. Eaton, A. Ayyanan, A.Y. Zhou, M. Brooks, F. Reinhard, C.C. Zhang, M. Shipitsin, L.L. Campbell, K. Polyak, C. Brisken, J. Yang, R.A. Weinberg, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008)CrossRefPubMedPubMedCentral
29.
go back to reference A.-P. Morel, M. Lièvre, C. Thomas, G. Hinkal, S. Ansieau, A. Puisieux, Generation of breast Cancer stem cells through epithelial-mesenchymal transition. PLoS One 3, e2888 (2008)CrossRefPubMedPubMedCentral A.-P. Morel, M. Lièvre, C. Thomas, G. Hinkal, S. Ansieau, A. Puisieux, Generation of breast Cancer stem cells through epithelial-mesenchymal transition. PLoS One 3, e2888 (2008)CrossRefPubMedPubMedCentral
32.
go back to reference D.D. Tran, C.A.S. Corsa, H. Biswas, R.L. Aft, G.D. Longmore, Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast Cancer recurrence. Mol. Cancer Res. 9, 1644–1657 (2011)CrossRefPubMedPubMedCentral D.D. Tran, C.A.S. Corsa, H. Biswas, R.L. Aft, G.D. Longmore, Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast Cancer recurrence. Mol. Cancer Res. 9, 1644–1657 (2011)CrossRefPubMedPubMedCentral
33.
go back to reference A.D. Rhim, E.T. Mirek, N.M. Aiello, A. Maitra, J.M. Bailey, F. McAllister, M. Reichert, G.L. Beatty, A.K. Rustgi, R.H. Vonderheide, S.D. Leach, B.Z. Stanger, EMT and dissemination precede pancreatic tumor formation. Cell 148, 349–361 (2012)CrossRefPubMedPubMedCentral A.D. Rhim, E.T. Mirek, N.M. Aiello, A. Maitra, J.M. Bailey, F. McAllister, M. Reichert, G.L. Beatty, A.K. Rustgi, R.H. Vonderheide, S.D. Leach, B.Z. Stanger, EMT and dissemination precede pancreatic tumor formation. Cell 148, 349–361 (2012)CrossRefPubMedPubMedCentral
34.
go back to reference O. Karaosmanoğlu, S. Banerjee, H. Sivas, Identification of biomarkers associated with partial epithelial to mesenchymal transition in the secretome of slug over-expressing hepatocellular carcinoma cells. Cell. Oncol. 41, 439–453 (2018)CrossRef O. Karaosmanoğlu, S. Banerjee, H. Sivas, Identification of biomarkers associated with partial epithelial to mesenchymal transition in the secretome of slug over-expressing hepatocellular carcinoma cells. Cell. Oncol. 41, 439–453 (2018)CrossRef
35.
37.
go back to reference O.H. Ocaña, R. Córcoles, Á. Fabra, G. Moreno-Bueno, H. Acloque, S. Vega, A. Barrallo-Gimeno, A. Cano, M.A. Nieto, Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012)CrossRefPubMed O.H. Ocaña, R. Córcoles, Á. Fabra, G. Moreno-Bueno, H. Acloque, S. Vega, A. Barrallo-Gimeno, A. Cano, M.A. Nieto, Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012)CrossRefPubMed
38.
go back to reference X. Li, M.T. Lewis, J. Huang, C. Gutierrez, C.K. Osborne, M.F. Wu, S.G. Hilsenbeck, A. Pavlick, X. Zhang, G.C. Chamness, H. Wong, J. Rosen, J.C. Chang, Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl. Cancer Inst. 100, 672–679 (2008)CrossRefPubMed X. Li, M.T. Lewis, J. Huang, C. Gutierrez, C.K. Osborne, M.F. Wu, S.G. Hilsenbeck, A. Pavlick, X. Zhang, G.C. Chamness, H. Wong, J. Rosen, J.C. Chang, Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl. Cancer Inst. 100, 672–679 (2008)CrossRefPubMed
39.
go back to reference R.J. Bowater, P.E. Lilford, R.J. Lilford, Estimating changes in overall survival using progression-free survival in metastatic breast and colorectal cancer. Int. J. Technol. Assess. Health Care 27, 207–214 (2011)CrossRefPubMed R.J. Bowater, P.E. Lilford, R.J. Lilford, Estimating changes in overall survival using progression-free survival in metastatic breast and colorectal cancer. Int. J. Technol. Assess. Health Care 27, 207–214 (2011)CrossRefPubMed
40.
go back to reference F. Sclafani, D. Gonzalez, D. Cunningham, S. Hulkki Wilson, C. Peckitt, J. Tabernero, B. Glimelius, A. Cervantes, A. Dewdney, A. Wotherspoon, G. Brown, D. Tait, J. Oates, I. Chau, TP53 Mutational Status and Cetuximab Benefit in Rectal Cancer: 5-Year Results of the EXPERT-C. Trial. J. Natl. Cancer Inst. 106, dju121 (2014)PubMed F. Sclafani, D. Gonzalez, D. Cunningham, S. Hulkki Wilson, C. Peckitt, J. Tabernero, B. Glimelius, A. Cervantes, A. Dewdney, A. Wotherspoon, G. Brown, D. Tait, J. Oates, I. Chau, TP53 Mutational Status and Cetuximab Benefit in Rectal Cancer: 5-Year Results of the EXPERT-C. Trial. J. Natl. Cancer Inst. 106, dju121 (2014)PubMed
41.
go back to reference N.S. Ruppender, C. Morrissey, P.H. Lange, R.L. Vessella, Dormancy in solid tumors: Implications for prostate cancer. Cancer Metastasis Rev. 32, 501–509 (2013)CrossRefPubMed N.S. Ruppender, C. Morrissey, P.H. Lange, R.L. Vessella, Dormancy in solid tumors: Implications for prostate cancer. Cancer Metastasis Rev. 32, 501–509 (2013)CrossRefPubMed
42.
go back to reference P. Ost, D. Reynders, K. Decaestecker, V. Fonteyne, N. Lumen, A. DeBruycker, B. Lambert, L. Delrue, R. Bultijnck, T. Claeys, E. Goetghebeur, G. Villeirs, K. De Man, F. Ameye, I. Billiet, S. Joniau, F. Vanhaverbeke, G. De Meerleer, Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: A prospective, randomized, multicenter phase II trial. J. Clin. Oncol. 36, 446–453 (2018)CrossRefPubMed P. Ost, D. Reynders, K. Decaestecker, V. Fonteyne, N. Lumen, A. DeBruycker, B. Lambert, L. Delrue, R. Bultijnck, T. Claeys, E. Goetghebeur, G. Villeirs, K. De Man, F. Ameye, I. Billiet, S. Joniau, F. Vanhaverbeke, G. De Meerleer, Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: A prospective, randomized, multicenter phase II trial. J. Clin. Oncol. 36, 446–453 (2018)CrossRefPubMed
43.
go back to reference J.A. Hensel, T.W. Flaig, D. Theodorescu, Clinical opportunities and challenges in targeting tumour dormancy. Nat. Rev. Clin. Oncol. 10, 41–51 (2013)CrossRefPubMed J.A. Hensel, T.W. Flaig, D. Theodorescu, Clinical opportunities and challenges in targeting tumour dormancy. Nat. Rev. Clin. Oncol. 10, 41–51 (2013)CrossRefPubMed
44.
go back to reference T.H. Cheung, T.A. Rando, Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013)CrossRefPubMed T.H. Cheung, T.A. Rando, Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013)CrossRefPubMed
45.
go back to reference A. Kobayashi, H. Okuda, F. Xing, P.R. Pandey, M. Watabe, S. Hirota, S.K. Pai, W. Liu, K. Fukuda, C. Chambers, A. Wilber, K. Watabe, Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. J. Exp. Med. 208, 2641–2655 (2011)CrossRefPubMedPubMedCentral A. Kobayashi, H. Okuda, F. Xing, P.R. Pandey, M. Watabe, S. Hirota, S.K. Pai, W. Liu, K. Fukuda, C. Chambers, A. Wilber, K. Watabe, Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. J. Exp. Med. 208, 2641–2655 (2011)CrossRefPubMedPubMedCentral
46.
go back to reference H. Jo, Y. Jia, K.K. Subramanian, H. Hattori, H.R. Luo, Cancer cell-derived Clusterin modulates the phosphatidylinositol 3’-kinase-Akt pathway through attenuation of insulin-like growth factor 1 during serum deprivation. Mol. Cell. Biol. 28, 4285–4299 (2008)CrossRefPubMedPubMedCentral H. Jo, Y. Jia, K.K. Subramanian, H. Hattori, H.R. Luo, Cancer cell-derived Clusterin modulates the phosphatidylinositol 3’-kinase-Akt pathway through attenuation of insulin-like growth factor 1 during serum deprivation. Mol. Cell. Biol. 28, 4285–4299 (2008)CrossRefPubMedPubMedCentral
47.
go back to reference A.P. Adam, A. George, D. Schewe, P. Bragado, B.V. Iglesias, A.C. Ranganathan, A. Kourtidis, D.S. Conklin, J.A. Aguirre-Ghiso, Computational identification of a p38SAPK-regulated transcription factor network required for tumor cell quiescence. Cancer Res. 69, 5664–5672 (2009) A.P. Adam, A. George, D. Schewe, P. Bragado, B.V. Iglesias, A.C. Ranganathan, A. Kourtidis, D.S. Conklin, J.A. Aguirre-Ghiso, Computational identification of a p38SAPK-regulated transcription factor network required for tumor cell quiescence. Cancer Res. 69, 5664–5672 (2009)
48.
go back to reference Z. Lu, R.Z. Luo, Y. Lu, X. Zhang, Q. Yu, S. Khare, S. Kondo, Y. Kondo, Y. Yu, G.B. Mills, W.S.L. Liao, R.C. Bast, The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J. Clin. Invest. 118, 3917–3929 (2008)PubMedPubMedCentral Z. Lu, R.Z. Luo, Y. Lu, X. Zhang, Q. Yu, S. Khare, S. Kondo, Y. Kondo, Y. Yu, G.B. Mills, W.S.L. Liao, R.C. Bast, The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J. Clin. Invest. 118, 3917–3929 (2008)PubMedPubMedCentral
49.
go back to reference L. Vera-Ramirez, S.K. Vodnala, R. Nini, K.W. Hunter, J.E. Green, Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat. Commun. 9, 1944 (2018)CrossRefPubMedPubMedCentral L. Vera-Ramirez, S.K. Vodnala, R. Nini, K.W. Hunter, J.E. Green, Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat. Commun. 9, 1944 (2018)CrossRefPubMedPubMedCentral
50.
go back to reference E. Friedman, The kinase mirk/dyrk1B: A possible therapeutic target in pancreatic cancer. Cancers 2, 1492–1512 (2010) E. Friedman, The kinase mirk/dyrk1B: A possible therapeutic target in pancreatic cancer. Cancers 2, 1492–1512 (2010)
51.
go back to reference G. Wu, Z. Ma, W. Hu, D. Wang, B. Gong, C. Fan, S. Jiang, T. Li, J. Gao, Y. Yang, Molecular insights of Gas6/TAM in cancer development and therapy. Cell Death Dis. 8, e2700 (2017)CrossRefPubMedPubMedCentral G. Wu, Z. Ma, W. Hu, D. Wang, B. Gong, C. Fan, S. Jiang, T. Li, J. Gao, Y. Yang, Molecular insights of Gas6/TAM in cancer development and therapy. Cell Death Dis. 8, e2700 (2017)CrossRefPubMedPubMedCentral
52.
go back to reference Y. Shiozawa, A.M. Havens, K.J. Pienta, R.S. Taichman, The bone marrow niche: Habitat to hematopoietic and mesenchymal stem cells, and unwitting host to molecular parasites. Leukemia 22, 941–950 (2008)CrossRefPubMedPubMedCentral Y. Shiozawa, A.M. Havens, K.J. Pienta, R.S. Taichman, The bone marrow niche: Habitat to hematopoietic and mesenchymal stem cells, and unwitting host to molecular parasites. Leukemia 22, 941–950 (2008)CrossRefPubMedPubMedCentral
53.
go back to reference A. Del Gobbo, N. Fusco, M. Barella, G. Ercoli, A. Sciarra, A. Palleschi, F. Pagni, C. Marchiò, M. Papotti, S. Ferrero, CXCL12 expression is a bona fide predictor of recurrence in lung neuroendocrine tumours; a multicentric study with emphasis on atypical carcinoids - a short report. Cell. Oncol. 41, 687–691 (2018)CrossRef A. Del Gobbo, N. Fusco, M. Barella, G. Ercoli, A. Sciarra, A. Palleschi, F. Pagni, C. Marchiò, M. Papotti, S. Ferrero, CXCL12 expression is a bona fide predictor of recurrence in lung neuroendocrine tumours; a multicentric study with emphasis on atypical carcinoids - a short report. Cell. Oncol. 41, 687–691 (2018)CrossRef
54.
go back to reference H. Gao, G. Chakraborty, A.P. Lee-Lim, Q. Mo, M. Decker, A. Vonica, R. Shen, E. Brogi, A.H. Brivanlou, F.G. Giancotti, The BMP inhibitor Coco reactivates breast Cancer cells at lung metastatic sites. Cell 150, 764–779 (2012)CrossRefPubMedPubMedCentral H. Gao, G. Chakraborty, A.P. Lee-Lim, Q. Mo, M. Decker, A. Vonica, R. Shen, E. Brogi, A.H. Brivanlou, F.G. Giancotti, The BMP inhibitor Coco reactivates breast Cancer cells at lung metastatic sites. Cell 150, 764–779 (2012)CrossRefPubMedPubMedCentral
55.
go back to reference C.M.C. Li, V. Gocheva, M.J. Oudin, A. Bhutkar, S.Y. Wang, S.R. Date, S.R. Ng, C.A. Whittaker, R.T. Bronson, E.L. Snyder, F.B. Gertler, T. Jacks, Foxa2 and Cdx2 cooperate with Nkx2-1 to inhibit lung adenocarcinoma metastasis. Genes Dev. 29, 1850–1862 (2015)CrossRefPubMedPubMedCentral C.M.C. Li, V. Gocheva, M.J. Oudin, A. Bhutkar, S.Y. Wang, S.R. Date, S.R. Ng, C.A. Whittaker, R.T. Bronson, E.L. Snyder, F.B. Gertler, T. Jacks, Foxa2 and Cdx2 cooperate with Nkx2-1 to inhibit lung adenocarcinoma metastasis. Genes Dev. 29, 1850–1862 (2015)CrossRefPubMedPubMedCentral
56.
go back to reference S. Malladi, D.G. MacAlinao, X. Jin, L. He, H. Basnet, Y. Zou, E. De Stanchina, J. Massagué, Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 165, 45–60 (2016)CrossRefPubMedPubMedCentral S. Malladi, D.G. MacAlinao, X. Jin, L. He, H. Basnet, Y. Zou, E. De Stanchina, J. Massagué, Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 165, 45–60 (2016)CrossRefPubMedPubMedCentral
58.
go back to reference P. Bragado, Y. Estrada, F. Parikh, S. Krause, C. Capobianco, H.G. Farina, D.M. Schewe, J.A. Aguirre-Ghiso, TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38α/β signalling. Nat. Cell Biol. 15, 1351–1361 (2013)CrossRefPubMedPubMedCentral P. Bragado, Y. Estrada, F. Parikh, S. Krause, C. Capobianco, H.G. Farina, D.M. Schewe, J.A. Aguirre-Ghiso, TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38α/β signalling. Nat. Cell Biol. 15, 1351–1361 (2013)CrossRefPubMedPubMedCentral
59.
go back to reference R. Sharma, R. Sharma, T.P. Khaket, C. Dutta, B. Chakraborty, T.K. Mukherjee, Breast cancer metastasis: Putative therapeutic role of vascular cell adhesion molecule-1. Cell. Oncol. 40, 199–208 (2017)CrossRef R. Sharma, R. Sharma, T.P. Khaket, C. Dutta, B. Chakraborty, T.K. Mukherjee, Breast cancer metastasis: Putative therapeutic role of vascular cell adhesion molecule-1. Cell. Oncol. 40, 199–208 (2017)CrossRef
60.
go back to reference D.M. Sosnoski, R.J. Norgard, C.D. Grove, S.J. Foster, A.M. Mastro, Dormancy and growth of metastatic breast cancer cells in a bone-like microenvironment. Clin. Exp. Metastasis 32, 335–344 (2015)CrossRefPubMed D.M. Sosnoski, R.J. Norgard, C.D. Grove, S.J. Foster, A.M. Mastro, Dormancy and growth of metastatic breast cancer cells in a bone-like microenvironment. Clin. Exp. Metastasis 32, 335–344 (2015)CrossRefPubMed
61.
go back to reference S. Hapke, H. Kessler, N. Arroyo de Prada, A. Benge, M. Schmitt, E. Lengyel, U. Reuning, Integrin alpha(v)beta(3)/vitronectin interaction affects expression of the urokinase system in human ovarian cancer cells. J. Biol. Chem. 276, 26340–26348 (2001)CrossRefPubMed S. Hapke, H. Kessler, N. Arroyo de Prada, A. Benge, M. Schmitt, E. Lengyel, U. Reuning, Integrin alpha(v)beta(3)/vitronectin interaction affects expression of the urokinase system in human ovarian cancer cells. J. Biol. Chem. 276, 26340–26348 (2001)CrossRefPubMed
63.
go back to reference L.H. El Touny, M.J. Hoenerhoff, J.E. Green, L.H. El Touny, A. Vieira, A. Mendoza, C. Khanna, M.J. Hoenerhoff, J.E. Green, Combined SFK / MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J. Clin. Invest. 124, 156–168 (2014)CrossRefPubMed L.H. El Touny, M.J. Hoenerhoff, J.E. Green, L.H. El Touny, A. Vieira, A. Mendoza, C. Khanna, M.J. Hoenerhoff, J.E. Green, Combined SFK / MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J. Clin. Invest. 124, 156–168 (2014)CrossRefPubMed
64.
go back to reference R. Sriram, V. Lo, B. Pryce, L. Antonova, A.J. Mears, M. Daneshmand, B. McKay, S.J. Conway, W.J. Muller, L.A. Sabourin, Loss of periostin/OSF-2 in ErbB2/Neu-driven tumors results in androgen receptor-positive molecular apocrine-like tumors with reduced Notch1 activity. Breast Cancer Res. 17, 7 (2015)CrossRefPubMedPubMedCentral R. Sriram, V. Lo, B. Pryce, L. Antonova, A.J. Mears, M. Daneshmand, B. McKay, S.J. Conway, W.J. Muller, L.A. Sabourin, Loss of periostin/OSF-2 in ErbB2/Neu-driven tumors results in androgen receptor-positive molecular apocrine-like tumors with reduced Notch1 activity. Breast Cancer Res. 17, 7 (2015)CrossRefPubMedPubMedCentral
65.
go back to reference S.E. Wheeler, A.M. Clark, D.P. Taylor, C.L. Young, V.C. Pillai, D.B. Stolz, R. Venkataramanan, D. Lauffenburger, L. Griffith, A. Wells, Spontaneous dormancy of metastatic breast cancer cells in an all human liver microphysiologic system. Br. J. Cancer 111, 2342–2350 (2014)CrossRefPubMedPubMedCentral S.E. Wheeler, A.M. Clark, D.P. Taylor, C.L. Young, V.C. Pillai, D.B. Stolz, R. Venkataramanan, D. Lauffenburger, L. Griffith, A. Wells, Spontaneous dormancy of metastatic breast cancer cells in an all human liver microphysiologic system. Br. J. Cancer 111, 2342–2350 (2014)CrossRefPubMedPubMedCentral
66.
go back to reference C.M. Ghajar, H. Peinado, H. Mori, I.R. Matei, K.J. Evason, H. Brazier, D. Almeida, A. Koller, K.A. Hajjar, D.Y.R. Stainier, E.I. Chen, D. Lyden, M.J. Bissell, The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 15, 807–817 (2013)CrossRefPubMedPubMedCentral C.M. Ghajar, H. Peinado, H. Mori, I.R. Matei, K.J. Evason, H. Brazier, D. Almeida, A. Koller, K.A. Hajjar, D.Y.R. Stainier, E.I. Chen, D. Lyden, M.J. Bissell, The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 15, 807–817 (2013)CrossRefPubMedPubMedCentral
67.
go back to reference D.G. DeNardo, J.B. Barreto, P. Andreu, L. Vasquez, D. Tawfik, N. Kolhatkar, L.M. Coussens, CD4+ T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16, 91–102 (2009) D.G. DeNardo, J.B. Barreto, P. Andreu, L. Vasquez, D. Tawfik, N. Kolhatkar, L.M. Coussens, CD4+ T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16, 91–102 (2009)
69.
go back to reference L.M. Nusblat, M.J. Carroll, C.M. Roth, Crosstalk between M2 macrophages and glioma stem cells. Cell. Oncol. 40, 471–482 (2017)CrossRef L.M. Nusblat, M.J. Carroll, C.M. Roth, Crosstalk between M2 macrophages and glioma stem cells. Cell. Oncol. 40, 471–482 (2017)CrossRef
70.
go back to reference Y. Jing, N. Ma, T. Fan, C. Wang, X. Bu, G. Jiang, R. Li, L. Gao, D. Li, M. Wu, L. Wei, Tumor necrosis factor-alpha promotes tumor growth by inducing vascular endothelial growth factor. Cancer Investig. 29, 485–493 (2011) Y. Jing, N. Ma, T. Fan, C. Wang, X. Bu, G. Jiang, R. Li, L. Gao, D. Li, M. Wu, L. Wei, Tumor necrosis factor-alpha promotes tumor growth by inducing vascular endothelial growth factor. Cancer Investig. 29, 485–493 (2011)
71.
go back to reference Y. Kienast, L. Von Baumgarten, M. Fuhrmann, W.E.F. Klinkert, R. Goldbrunner, J. Herms, F. Winkler, Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 16, 116–122 (2010)CrossRefPubMed Y. Kienast, L. Von Baumgarten, M. Fuhrmann, W.E.F. Klinkert, R. Goldbrunner, J. Herms, F. Winkler, Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 16, 116–122 (2010)CrossRefPubMed
72.
go back to reference S. Singh, U.P. Singh, W.E. Grizzle, J.W. Lillard, CXCL12–CXCR4 interactions modulate prostate cancer cell migration, metalloproteinase expression and invasion. Lab. Invest. 84, 1666–1676 (2004) S. Singh, U.P. Singh, W.E. Grizzle, J.W. Lillard, CXCL12–CXCR4 interactions modulate prostate cancer cell migration, metalloproteinase expression and invasion. Lab. Invest. 84, 1666–1676 (2004)
73.
go back to reference Y. Shiozawa, E.a. Pedersen, A.M. Havens, Y. Jung, A. Mishra, J. Joseph, J.K. Kim, L.R. Patel, C. Ying, A.M. Ziegler, Others, Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011)CrossRefPubMedPubMedCentral Y. Shiozawa, E.a. Pedersen, A.M. Havens, Y. Jung, A. Mishra, J. Joseph, J.K. Kim, L.R. Patel, C. Ying, A.M. Ziegler, Others, Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011)CrossRefPubMedPubMedCentral
74.
go back to reference I. Bruns, D. Lucas, S. Pinho, J. Ahmed, M.P. Lambert, Y. Kunisaki, C. Scheiermann, L. Schiff, M. Poncz, A. Bergman, P.S. Frenette, Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion. Nat. Med. 20, 1315–1320 (2014)CrossRefPubMedPubMedCentral I. Bruns, D. Lucas, S. Pinho, J. Ahmed, M.P. Lambert, Y. Kunisaki, C. Scheiermann, L. Schiff, M. Poncz, A. Bergman, P.S. Frenette, Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion. Nat. Med. 20, 1315–1320 (2014)CrossRefPubMedPubMedCentral
75.
go back to reference S. Bekaert, M. Fillet, B. Detry, M. Pichavant, R. Marée, A. Noel, N. Rocks, D. Cataldo, Inflammation-generated extracellular matrix fragments drive lung metastasis. Cancer Growth Metastasis 10, 1–11 (2017)CrossRef S. Bekaert, M. Fillet, B. Detry, M. Pichavant, R. Marée, A. Noel, N. Rocks, D. Cataldo, Inflammation-generated extracellular matrix fragments drive lung metastasis. Cancer Growth Metastasis 10, 1–11 (2017)CrossRef
76.
go back to reference J.M. De Cock, T. Shibue, A. Dongre, Z. Keckesova, F. Reinhardt, R.A. Weinberg, Inflammation triggers Zeb1-dependent escape from tumor latency. Cancer Res. 76, 6778–6784 (2016)CrossRefPubMedPubMedCentral J.M. De Cock, T. Shibue, A. Dongre, Z. Keckesova, F. Reinhardt, R.A. Weinberg, Inflammation triggers Zeb1-dependent escape from tumor latency. Cancer Res. 76, 6778–6784 (2016)CrossRefPubMedPubMedCentral
77.
go back to reference J. Albrengues, M.A. Shields, D. Ng, C.G. Park, A. Ambrico, M.E. Poindexter, P. Upadhyay, D.L. Uyeminami, A. Pommier, V. Küttner, E. Bružas, L. Maiorino, C. Bautista, E.M. Carmona, P.A. Gimotty, D.T. Fearon, K. Chang, S.K. Lyons, K.E. Pinkerton, L.C. Trotman, M.S. Goldberg, J.T.-H. Yeh, M. Egeblad, Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 361, 4227 (2018) J. Albrengues, M.A. Shields, D. Ng, C.G. Park, A. Ambrico, M.E. Poindexter, P. Upadhyay, D.L. Uyeminami, A. Pommier, V. Küttner, E. Bružas, L. Maiorino, C. Bautista, E.M. Carmona, P.A. Gimotty, D.T. Fearon, K. Chang, S.K. Lyons, K.E. Pinkerton, L.C. Trotman, M.S. Goldberg, J.T.-H. Yeh, M. Egeblad, Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 361, 4227 (2018)
78.
go back to reference M. Retsky, R. Demicheli, W.J.M. Hrushesky, P. Forget, M. De Kock, I. Gukas, R.A. Rogers, M. Baum, V. Sukhatme, J.S. Vaidya, Reduction of breast cancer relapses with perioperative non-steroidal anti-inflammatory drugs: New findings and a review. Curr. Med. Chem. 20, 4163–4176 (2013)CrossRefPubMedPubMedCentral M. Retsky, R. Demicheli, W.J.M. Hrushesky, P. Forget, M. De Kock, I. Gukas, R.A. Rogers, M. Baum, V. Sukhatme, J.S. Vaidya, Reduction of breast cancer relapses with perioperative non-steroidal anti-inflammatory drugs: New findings and a review. Curr. Med. Chem. 20, 4163–4176 (2013)CrossRefPubMedPubMedCentral
79.
go back to reference I. Ben-Porath, M.W. Thomson, V.J. Carey, R. Ge, G.W. Bell, A. Regev, R.A. Weinberg, An embryonic stem cell–like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 40, 499–507 (2008)CrossRefPubMedPubMedCentral I. Ben-Porath, M.W. Thomson, V.J. Carey, R. Ge, G.W. Bell, A. Regev, R.A. Weinberg, An embryonic stem cell–like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 40, 499–507 (2008)CrossRefPubMedPubMedCentral
80.
go back to reference K. Eppert, K. Takenaka, E.R. Lechman, L. Waldron, B. Nilsson, P. van Galen, K.H. Metzeler, A. Poeppl, V. Ling, J. Beyene, A.J. Canty, J.S. Danska, S.K. Bohlander, C. Buske, M.D. Minden, T.R. Golub, I. Jurisica, B.L. Ebert, J.E. Dick, Stem cell gene expression programs influence clinical outcome in human leukemia. Nat. Med. 17, 1086–1093 (2011)CrossRefPubMed K. Eppert, K. Takenaka, E.R. Lechman, L. Waldron, B. Nilsson, P. van Galen, K.H. Metzeler, A. Poeppl, V. Ling, J. Beyene, A.J. Canty, J.S. Danska, S.K. Bohlander, C. Buske, M.D. Minden, T.R. Golub, I. Jurisica, B.L. Ebert, J.E. Dick, Stem cell gene expression programs influence clinical outcome in human leukemia. Nat. Med. 17, 1086–1093 (2011)CrossRefPubMed
81.
go back to reference S. Gawrzak, L. Rinaldi, S. Gregorio, E.J. Arenas, F. Salvador, J. Urosevic, C. Figueras-Puig, F. Rojo, I.D.B. Barrantes, J.M. Cejalvo, M. Palafox, M. Guiu, A. Berenguer-Llergo, A. Symeonidi, A. Bellmunt, D. Kalafatovic, A. Arnal-Estapé, E. Fernández, B. Müllauer, R. Groeneveld, K. Slobodnyuk, C.S.-O. Attolini, C. Saura, J. Arribas, J. Cortes, A. Rovira, M. Muñoz, A. Lluch, V. Serra, J. Albanell, A. Prat, A.R. Nebreda, S.A. Benitah, R.R. Gomis, MSK1 regulates luminal cell differentiation and metastatic dormancy in ER+breast cancer. Nat. Cell Biol. 20, 211–221 (2018)CrossRefPubMed S. Gawrzak, L. Rinaldi, S. Gregorio, E.J. Arenas, F. Salvador, J. Urosevic, C. Figueras-Puig, F. Rojo, I.D.B. Barrantes, J.M. Cejalvo, M. Palafox, M. Guiu, A. Berenguer-Llergo, A. Symeonidi, A. Bellmunt, D. Kalafatovic, A. Arnal-Estapé, E. Fernández, B. Müllauer, R. Groeneveld, K. Slobodnyuk, C.S.-O. Attolini, C. Saura, J. Arribas, J. Cortes, A. Rovira, M. Muñoz, A. Lluch, V. Serra, J. Albanell, A. Prat, A.R. Nebreda, S.A. Benitah, R.R. Gomis, MSK1 regulates luminal cell differentiation and metastatic dormancy in ER+breast cancer. Nat. Cell Biol. 20, 211–221 (2018)CrossRefPubMed
82.
go back to reference M.L. Asselin-Labat, K.D. Sutherland, H. Barker, R. Thomas, M. Shackleton, N.C. Forrest, L. Hartley, L. Robb, F.G. Grosveld, J. van der Wees, G.J. Lindeman, J.E. Visvader, Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat. Cell Biol. 9, 201–207 (2007)CrossRefPubMed M.L. Asselin-Labat, K.D. Sutherland, H. Barker, R. Thomas, M. Shackleton, N.C. Forrest, L. Hartley, L. Robb, F.G. Grosveld, J. van der Wees, G.J. Lindeman, J.E. Visvader, Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat. Cell Biol. 9, 201–207 (2007)CrossRefPubMed
84.
go back to reference R. Bhattacharya, S. Banerjee Mustafi, M. Street, A. Dey, S.K.D. Dwivedi, Bmi-1: At the crossroads of physiological and pathological biology. Genes Dis 2, 225–239 (2015)CrossRefPubMedPubMedCentral R. Bhattacharya, S. Banerjee Mustafi, M. Street, A. Dey, S.K.D. Dwivedi, Bmi-1: At the crossroads of physiological and pathological biology. Genes Dis 2, 225–239 (2015)CrossRefPubMedPubMedCentral
85.
go back to reference B.H. Lee, S. Yegnasubramanian, X. Lin, W.G. Nelson, Procainamide is a specific inhibitor of DNA methyltransferase 1. J. Biol. Chem. 280, 40749–40756 (2005)CrossRefPubMed B.H. Lee, S. Yegnasubramanian, X. Lin, W.G. Nelson, Procainamide is a specific inhibitor of DNA methyltransferase 1. J. Biol. Chem. 280, 40749–40756 (2005)CrossRefPubMed
86.
go back to reference L. Wang, J. Wang, MicroRNA-mediated breast cancer metastasis: From primary site to distant organs. Oncogene 31, 2499–2511 (2012)CrossRefPubMed L. Wang, J. Wang, MicroRNA-mediated breast cancer metastasis: From primary site to distant organs. Oncogene 31, 2499–2511 (2012)CrossRefPubMed
87.
go back to reference P.K. Lim, S.A. Bliss, S.A. Patel, M. Taborga, M.A. Dave, L.A. Gregory, S.J. Greco, M. Bryan, P.S. Patel, P. Rameshwar, Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 71, 1550–1560 (2011)CrossRefPubMed P.K. Lim, S.A. Bliss, S.A. Patel, M. Taborga, M.A. Dave, L.A. Gregory, S.J. Greco, M. Bryan, P.S. Patel, P. Rameshwar, Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 71, 1550–1560 (2011)CrossRefPubMed
88.
go back to reference V. Sundararajan, F.H. Sarkar, T.S. Ramasamy, The versatile role of exosomes in cancer progression: Diagnostic and therapeutic implications. Cell. Oncol. 41, 223–252 (2018)CrossRef V. Sundararajan, F.H. Sarkar, T.S. Ramasamy, The versatile role of exosomes in cancer progression: Diagnostic and therapeutic implications. Cell. Oncol. 41, 223–252 (2018)CrossRef
89.
go back to reference M. Makita, T. Sakai, A. Ogiya, D. Kitagawa, H. Morizono, Y. Miyagi, K. Iijima, T. Iwase, Optimal surveillance for postoperative metastasis in breast cancer patients. Breast Cancer 23, 286–294 (2016)CrossRefPubMed M. Makita, T. Sakai, A. Ogiya, D. Kitagawa, H. Morizono, Y. Miyagi, K. Iijima, T. Iwase, Optimal surveillance for postoperative metastasis in breast cancer patients. Breast Cancer 23, 286–294 (2016)CrossRefPubMed
91.
go back to reference R.M. MacKie, R. Reid, B. Junor, Fatal melanoma transferred in a donated kidney 16 years after melanoma surgery. N. Engl. J. Med. 348, 567–568 (2003)CrossRefPubMed R.M. MacKie, R. Reid, B. Junor, Fatal melanoma transferred in a donated kidney 16 years after melanoma surgery. N. Engl. J. Med. 348, 567–568 (2003)CrossRefPubMed
92.
go back to reference Y.A.H. Matser, M.L. Terpstra, S. Nadalin, G.D. Nossent, J. de Boer, B.C. van Bemmel, S. van Eeden, K. Budde, S. Brakemeier, F.J. Bemelman, Transmission of breast cancer by a single multiorgan donor to 4 transplant recipients. Am. J. Transplant. 18, 1810–1814 (2018)CrossRefPubMed Y.A.H. Matser, M.L. Terpstra, S. Nadalin, G.D. Nossent, J. de Boer, B.C. van Bemmel, S. van Eeden, K. Budde, S. Brakemeier, F.J. Bemelman, Transmission of breast cancer by a single multiorgan donor to 4 transplant recipients. Am. J. Transplant. 18, 1810–1814 (2018)CrossRefPubMed
93.
go back to reference L. Biavati, F. Rossari, E. Orciuolo, M.I. Ferreri, C. Zucchinetti, U. Boggi, E.M. Ciancia, F. Caracciolo, E. Benedetti, G. Buda, E. Ciabatti, F. Guerrini, S. Grassi, S. Galimberti, M. Petrini, Role of circulating DNA in the precocious diagnosis of lymphoma: An example in a transplant-related aggressive lymphoma. Clin. Oncol. Res. 2, 1–5 (2019) L. Biavati, F. Rossari, E. Orciuolo, M.I. Ferreri, C. Zucchinetti, U. Boggi, E.M. Ciancia, F. Caracciolo, E. Benedetti, G. Buda, E. Ciabatti, F. Guerrini, S. Grassi, S. Galimberti, M. Petrini, Role of circulating DNA in the precocious diagnosis of lymphoma: An example in a transplant-related aggressive lymphoma. Clin. Oncol. Res. 2, 1–5 (2019)
94.
go back to reference M.H. Manjili, The inherent premise of immunotherapy for cancer dormancy. Cancer Res. 74, 6745–6749 (2014)CrossRefPubMed M.H. Manjili, The inherent premise of immunotherapy for cancer dormancy. Cancer Res. 74, 6745–6749 (2014)CrossRefPubMed
95.
go back to reference C.M. Koebel, W. Vermi, J.B. Swann, N. Zerafa, S.J. Rodig, L.J. Old, M.J. Smyth, R.D. Schreiber, Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450, 903–907 (2007)CrossRefPubMed C.M. Koebel, W. Vermi, J.B. Swann, N. Zerafa, S.J. Rodig, L.J. Old, M.J. Smyth, R.D. Schreiber, Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450, 903–907 (2007)CrossRefPubMed
96.
go back to reference N. Müller-Hermelink, H. Braumüller, B. Pichler, T. Wieder, R. Mailhammer, K. Schaak, K. Ghoreschi, A. Yazdi, R. Haubner, C.A. Sander, R. Mocikat, M. Schwaiger, I. Förster, R. Huss, W.A. Weber, M. Kneilling, M. Röcken, TNFR1 signaling and IFN-γ signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 13, 507–518 (2008)CrossRefPubMed N. Müller-Hermelink, H. Braumüller, B. Pichler, T. Wieder, R. Mailhammer, K. Schaak, K. Ghoreschi, A. Yazdi, R. Haubner, C.A. Sander, R. Mocikat, M. Schwaiger, I. Förster, R. Huss, W.A. Weber, M. Kneilling, M. Röcken, TNFR1 signaling and IFN-γ signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 13, 507–518 (2008)CrossRefPubMed
98.
go back to reference D. Panigrahy, M.L. Edin, C.R. Lee, S. Huang, D.R. Bielenberg, C.E. Butterfield, C.M. Barnés, A. Mammoto, T. Mammoto, A. Luria, O. Benny, D.M. Chaponis, A.C. Dudley, E.R. Greene, J.-A. Vergilio, G. Pietramaggiori, S.S. Scherer-Pietramaggiori, S.M. Short, M. Seth, F.B. Lih, K.B. Tomer, J. Yang, R.A. Schwendener, B.D. Hammock, J.R. Falck, V.L. Manthati, D.E. Ingber, A. Kaipainen, P.A. D’Amore, M.W. Kieran, D.C. Zeldin, Epoxyeicosanoids stimulate multiorgan metastasis and tumor dormancy escape in mice. J. Clin. Invest. 122, 178–191 (2012) D. Panigrahy, M.L. Edin, C.R. Lee, S. Huang, D.R. Bielenberg, C.E. Butterfield, C.M. Barnés, A. Mammoto, T. Mammoto, A. Luria, O. Benny, D.M. Chaponis, A.C. Dudley, E.R. Greene, J.-A. Vergilio, G. Pietramaggiori, S.S. Scherer-Pietramaggiori, S.M. Short, M. Seth, F.B. Lih, K.B. Tomer, J. Yang, R.A. Schwendener, B.D. Hammock, J.R. Falck, V.L. Manthati, D.E. Ingber, A. Kaipainen, P.A. D’Amore, M.W. Kieran, D.C. Zeldin, Epoxyeicosanoids stimulate multiorgan metastasis and tumor dormancy escape in mice. J. Clin. Invest. 122, 178–191 (2012)
99.
go back to reference J. Eyles, A.-L. Puaux, X. Wang, B. Toh, C. Prakash, M. Hong, T.G. Tan, L. Zheng, L.C. Ong, Y. Jin, M. Kato, A. Prévost-Blondel, P. Chow, H. Yang, J.-P. Abastado, Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J. Clin. Invest. 120, 2030–2039 (2010)CrossRefPubMedPubMedCentral J. Eyles, A.-L. Puaux, X. Wang, B. Toh, C. Prakash, M. Hong, T.G. Tan, L. Zheng, L.C. Ong, Y. Jin, M. Kato, A. Prévost-Blondel, P. Chow, H. Yang, J.-P. Abastado, Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J. Clin. Invest. 120, 2030–2039 (2010)CrossRefPubMedPubMedCentral
100.
go back to reference W. Zou, Regulatory T cells, tumour immunity and immunotherapy. Nat. Rev. Immunol. 6, 295–307 (2006)CrossRefPubMed W. Zou, Regulatory T cells, tumour immunity and immunotherapy. Nat. Rev. Immunol. 6, 295–307 (2006)CrossRefPubMed
101.
go back to reference N. Said, S. Smith, M. Sanchez-Carbayo, D. Theodorescu, Tumor endothelin-1 enhances metastatic colonization of the lung in mouse xenograft models of bladder cancer. J. Clin. Invest. 121, 132–147 (2011)CrossRefPubMed N. Said, S. Smith, M. Sanchez-Carbayo, D. Theodorescu, Tumor endothelin-1 enhances metastatic colonization of the lung in mouse xenograft models of bladder cancer. J. Clin. Invest. 121, 132–147 (2011)CrossRefPubMed
102.
go back to reference A. Borodovsky, V. Salmasi, S. Turcan, A.W.M. Fabius, G.S. Baia, C.G. Eberhart, J.D. Weingart, G.L. Gallia, S.B. Baylin, T.A. Chan, G.J. Riggins, 5-azacytidine reduces methylation, promotes differentiation and induces tumor regression in a patient-derived IDH1 mutant glioma xenograft. Oncotarget 4, 1737–1747 (2013)CrossRefPubMedPubMedCentral A. Borodovsky, V. Salmasi, S. Turcan, A.W.M. Fabius, G.S. Baia, C.G. Eberhart, J.D. Weingart, G.L. Gallia, S.B. Baylin, T.A. Chan, G.J. Riggins, 5-azacytidine reduces methylation, promotes differentiation and induces tumor regression in a patient-derived IDH1 mutant glioma xenograft. Oncotarget 4, 1737–1747 (2013)CrossRefPubMedPubMedCentral
103.
go back to reference S.-Y. Kang, O.J. Halvorsen, K. Gravdal, N. Bhattacharya, J.M. Lee, N.W. Liu, B.T. Johnston, A.B. Johnston, S.A. Haukaas, K. Aamodt, S. Yoo, L.A. Akslen, R.S. Watnick, Prosaposin inhibits tumor metastasis via paracrine and endocrine stimulation of stromal p53 and Tsp-1. Proc. Natl. Acad. Sci. U. S. A. 106, 12115–12120 (2009) S.-Y. Kang, O.J. Halvorsen, K. Gravdal, N. Bhattacharya, J.M. Lee, N.W. Liu, B.T. Johnston, A.B. Johnston, S.A. Haukaas, K. Aamodt, S. Yoo, L.A. Akslen, R.S. Watnick, Prosaposin inhibits tumor metastasis via paracrine and endocrine stimulation of stromal p53 and Tsp-1. Proc. Natl. Acad. Sci. U. S. A. 106, 12115–12120 (2009)
104.
go back to reference S. Demaria, N. Kawashima, A.M. Yang, M.L. Devitt, J.S. Babb, J.P. Allison, S.C. Formenti, Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 11, 728–734 (2005)PubMed S. Demaria, N. Kawashima, A.M. Yang, M.L. Devitt, J.S. Babb, J.P. Allison, S.C. Formenti, Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 11, 728–734 (2005)PubMed
105.
go back to reference M. Kmieciak, K.K. Payne, X.-Y. Wang, M.H. Manjili, IFN-γ Rα is a key determinant of CD8+ T cell-mediated tumor elimination or tumor escape and relapse in FVB mouse. PLoS One 8, e82544 (2013)CrossRefPubMedPubMedCentral M. Kmieciak, K.K. Payne, X.-Y. Wang, M.H. Manjili, IFN-γ Rα is a key determinant of CD8+ T cell-mediated tumor elimination or tumor escape and relapse in FVB mouse. PLoS One 8, e82544 (2013)CrossRefPubMedPubMedCentral
106.
go back to reference F. Rossari, F. Minutolo, E. Orciuolo, Past, present, and future of Bcr-Abl inhibitors: From chemical development to clinical efficacy. J. Hematol. Oncol. 11, 1–14 (2018)CrossRef F. Rossari, F. Minutolo, E. Orciuolo, Past, present, and future of Bcr-Abl inhibitors: From chemical development to clinical efficacy. J. Hematol. Oncol. 11, 1–14 (2018)CrossRef
107.
go back to reference A. Sadarangani, G. Pineda, K.M. Lennon, H.J. Chun, A. Shih, A.E. Schairer, A.C. Court, D.J. Goff, S.L. Prashad, I. Geron, R. Wall, J.D. McPherson, R.A. Moore, M. Pu, L. Bao, A. Jackson-Fisher, M. Munchhof, T. VanArsdale, T. Reya, S.R. Morris, M.D. Minden, K. Messer, H.K.A. Mikkola, M.A. Marra, T.J. Hudson, C.H.M. Jamieson, GLI2 inhibition abrogates human leukemia stem cell dormancy. J. Transl. Med. 13, 98 (2015)CrossRefPubMedPubMedCentral A. Sadarangani, G. Pineda, K.M. Lennon, H.J. Chun, A. Shih, A.E. Schairer, A.C. Court, D.J. Goff, S.L. Prashad, I. Geron, R. Wall, J.D. McPherson, R.A. Moore, M. Pu, L. Bao, A. Jackson-Fisher, M. Munchhof, T. VanArsdale, T. Reya, S.R. Morris, M.D. Minden, K. Messer, H.K.A. Mikkola, M.A. Marra, T.J. Hudson, C.H.M. Jamieson, GLI2 inhibition abrogates human leukemia stem cell dormancy. J. Transl. Med. 13, 98 (2015)CrossRefPubMedPubMedCentral
108.
go back to reference P.S. Becker, Dependence of acute myeloid leukemia on adhesion within the bone marrow microenvironment. Sci. World J. 2012, 856467 (2012)CrossRef P.S. Becker, Dependence of acute myeloid leukemia on adhesion within the bone marrow microenvironment. Sci. World J. 2012, 856467 (2012)CrossRef
109.
go back to reference B. Boyerinas, M. Zafrir, A.E. Yesilkanal, T.T. Price, E.M. Hyjek, D.A. Sipkins, Adhesion to osteopontin in the bone marrow niche regulates lymphoblastic leukemia cell dormancy. Blood 121, 4821–4831 (2013)CrossRefPubMedPubMedCentral B. Boyerinas, M. Zafrir, A.E. Yesilkanal, T.T. Price, E.M. Hyjek, D.A. Sipkins, Adhesion to osteopontin in the bone marrow niche regulates lymphoblastic leukemia cell dormancy. Blood 121, 4821–4831 (2013)CrossRefPubMedPubMedCentral
110.
go back to reference C. Kollmannsberger, D. Soulieres, R. Wong, A. Scalera, R. Gaspo, G. Bjarnason, Sunitinib therapy for metastatic renal cell carcinoma: Recommendations for management of side effects. Can. Urol. Assoc. J. 1, S41–S54 (2007) C. Kollmannsberger, D. Soulieres, R. Wong, A. Scalera, R. Gaspo, G. Bjarnason, Sunitinib therapy for metastatic renal cell carcinoma: Recommendations for management of side effects. Can. Urol. Assoc. J. 1, S41–S54 (2007)
111.
go back to reference J. Baselga, M. Campone, M. Piccart, H.A. Burris, H.S. Rugo, T. Sahmoud, S. Noguchi, M. Gnant, K.I. Pritchard, F. Lebrun, J.T. Beck, Y. Ito, D. Yardley, I. Deleu, A. Perez, T. Bachelot, L. Vittori, Z. Xu, P. Mukhopadhyay, D. Lebwohl, G.N. Hortobagyi, Everolimus in postmenopausal hormone-receptor–positive advanced breast Cancer. N. Engl. J. Med. 366, 520–529 (2012)CrossRefPubMed J. Baselga, M. Campone, M. Piccart, H.A. Burris, H.S. Rugo, T. Sahmoud, S. Noguchi, M. Gnant, K.I. Pritchard, F. Lebrun, J.T. Beck, Y. Ito, D. Yardley, I. Deleu, A. Perez, T. Bachelot, L. Vittori, Z. Xu, P. Mukhopadhyay, D. Lebwohl, G.N. Hortobagyi, Everolimus in postmenopausal hormone-receptor–positive advanced breast Cancer. N. Engl. J. Med. 366, 520–529 (2012)CrossRefPubMed
112.
go back to reference P. Hadji, R. Coleman, M. Gnant, Bone effects of mammalian target of rapamycin (mTOR) inhibition with everolimus. Crit. Rev. Oncol. Hematol. 87, 101–111 (2013)CrossRefPubMed P. Hadji, R. Coleman, M. Gnant, Bone effects of mammalian target of rapamycin (mTOR) inhibition with everolimus. Crit. Rev. Oncol. Hematol. 87, 101–111 (2013)CrossRefPubMed
113.
go back to reference L.M. Glode, A. Barqawi, F. Crighton, E.D. Crawford, R. Kerbel, Metronomic therapy with cyclophosphamide and dexamethasone for prostate carcinoma. Cancer 98, 1643–1648 (2003)CrossRefPubMed L.M. Glode, A. Barqawi, F. Crighton, E.D. Crawford, R. Kerbel, Metronomic therapy with cyclophosphamide and dexamethasone for prostate carcinoma. Cancer 98, 1643–1648 (2003)CrossRefPubMed
114.
go back to reference Y. Takahashi, G. Sawada, J. Kurashige, R. Uchi, T. Matsumura, H. Ueo, Y. Takano, S. Akiyoshi, H. Eguchi, T. Sudo, K. Sugimachi, Y. Doki, M. Mori, K. Mimori, Paired related homoeobox 1, a new EMT inducer, is involved in metastasis and poor prognosis in colorectal cancer. Br. J. Cancer 109, 307–311 (2013)CrossRefPubMedPubMedCentral Y. Takahashi, G. Sawada, J. Kurashige, R. Uchi, T. Matsumura, H. Ueo, Y. Takano, S. Akiyoshi, H. Eguchi, T. Sudo, K. Sugimachi, Y. Doki, M. Mori, K. Mimori, Paired related homoeobox 1, a new EMT inducer, is involved in metastasis and poor prognosis in colorectal cancer. Br. J. Cancer 109, 307–311 (2013)CrossRefPubMedPubMedCentral
115.
go back to reference S.V. Shmelkov, J.M. Butler, A.T. Hooper, A. Hormigo, J. Kushner, T. Milde, R.S. Clair, M. Baljevic, I. White, D.K. Jin, A. Chadburn, A.J. Murphy, D.M. Valenzuela, N.W. Gale, G. Thurston, G.D. Yancopoulos, M. D’Angelica, N. Kemeny, D. Lyden, S. Rafii, CD133 expression is not restricted to stem cells, and both CD133 + and CD133 - metastatic colon cancer cells initiate tumors. J. Clin. Invest. 118, 2111–2120 (2008)PubMedPubMedCentral S.V. Shmelkov, J.M. Butler, A.T. Hooper, A. Hormigo, J. Kushner, T. Milde, R.S. Clair, M. Baljevic, I. White, D.K. Jin, A. Chadburn, A.J. Murphy, D.M. Valenzuela, N.W. Gale, G. Thurston, G.D. Yancopoulos, M. D’Angelica, N. Kemeny, D. Lyden, S. Rafii, CD133 expression is not restricted to stem cells, and both CD133 + and CD133 - metastatic colon cancer cells initiate tumors. J. Clin. Invest. 118, 2111–2120 (2008)PubMedPubMedCentral
116.
go back to reference T. Lotan, J. Hickson, J. Souris, D. Huo, J. Taylor, T. Li, K. Otto, S.D. Yamada, K. Macleod, C.W. Rinker-Schaeffer, c-Jun NH2-Terminal Kinase Activating Kinase 1/Mitogen-Activated Protein Kinase Kinase 4-Mediated Inhibition of SKOV3ip.1 Ovarian Cancer Metastasis Involves Growth Arrest and p21 Up-regulation. Cancer Res. 68, 2166–2175 (2008)CrossRefPubMedPubMedCentral T. Lotan, J. Hickson, J. Souris, D. Huo, J. Taylor, T. Li, K. Otto, S.D. Yamada, K. Macleod, C.W. Rinker-Schaeffer, c-Jun NH2-Terminal Kinase Activating Kinase 1/Mitogen-Activated Protein Kinase Kinase 4-Mediated Inhibition of SKOV3ip.1 Ovarian Cancer Metastasis Involves Growth Arrest and p21 Up-regulation. Cancer Res. 68, 2166–2175 (2008)CrossRefPubMedPubMedCentral
117.
go back to reference A.C. Ranganathan, A.P. Adam, L. Zhang, J.A. Aguirre-Ghiso, Tumor cell dormancy induced by p38SAPK and ER-stress signaling: An adaptive advantage for metastatic cells? Cancer Biol. Ther. 5, 729–735 (2006) A.C. Ranganathan, A.P. Adam, L. Zhang, J.A. Aguirre-Ghiso, Tumor cell dormancy induced by p38SAPK and ER-stress signaling: An adaptive advantage for metastatic cells? Cancer Biol. Ther. 5, 729–735 (2006)
118.
go back to reference A.W. Lambert, D.R. Pattabiraman, R.A. Weinberg, Emerging Biological Principles of Metastasis Cell 168, 670–691 (2017) A.W. Lambert, D.R. Pattabiraman, R.A. Weinberg, Emerging Biological Principles of Metastasis Cell 168, 670–691 (2017)
119.
go back to reference Y. Kang, P.M. Siegel, W. Shu, M. Drobnjak, S.M. Kakonen, C. Cordón-Cardo, T.A. Guise, J. Massagué, A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003)CrossRefPubMed Y. Kang, P.M. Siegel, W. Shu, M. Drobnjak, S.M. Kakonen, C. Cordón-Cardo, T.A. Guise, J. Massagué, A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003)CrossRefPubMed
120.
go back to reference P.D. Bos, X.H.F. Zhang, C. Nadal, W. Shu, R.R. Gomis, D.X. Nguyen, A.J. Minn, M.J. Van De Vijver, W.L. Gerald, J.A. Foekens, J. Massagué, Genes that mediate breast cancer metastasis to the brain. Nature 459, 1005–1009 (2009)CrossRefPubMedPubMedCentral P.D. Bos, X.H.F. Zhang, C. Nadal, W. Shu, R.R. Gomis, D.X. Nguyen, A.J. Minn, M.J. Van De Vijver, W.L. Gerald, J.A. Foekens, J. Massagué, Genes that mediate breast cancer metastasis to the brain. Nature 459, 1005–1009 (2009)CrossRefPubMedPubMedCentral
121.
go back to reference A.M. Clark, S.E. Wheeler, C.L. Young, L. Stockdale, J. Shepard Neiman, W. Zhao, D.B. Stolz, R. Venkataramanan, D. Lauffenburger, L. Griffith, A. Wells, A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties. Lab Chip 17, 156–168 (2017)CrossRef A.M. Clark, S.E. Wheeler, C.L. Young, L. Stockdale, J. Shepard Neiman, W. Zhao, D.B. Stolz, R. Venkataramanan, D. Lauffenburger, L. Griffith, A. Wells, A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties. Lab Chip 17, 156–168 (2017)CrossRef
122.
go back to reference H. Gao, G. Chakraborty, Z. Zhang, I. Akalay, M. Gadiya, Y. Gao, S. Sinha, J. Hu, C. Jiang, M. Akram, E. Brogi, B. Leitinger, F.G. Giancotti, Multi-organ site metastatic reactivation mediated by non-canonical Discoidin domain receptor 1 signaling. Cell 166, 47–62 (2016)CrossRefPubMedPubMedCentral H. Gao, G. Chakraborty, Z. Zhang, I. Akalay, M. Gadiya, Y. Gao, S. Sinha, J. Hu, C. Jiang, M. Akram, E. Brogi, B. Leitinger, F.G. Giancotti, Multi-organ site metastatic reactivation mediated by non-canonical Discoidin domain receptor 1 signaling. Cell 166, 47–62 (2016)CrossRefPubMedPubMedCentral
124.
go back to reference J.M. Butler, H. Kobayashi, S. Rafii, Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer 10, 138–146 (2010)CrossRefPubMedPubMedCentral J.M. Butler, H. Kobayashi, S. Rafii, Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer 10, 138–146 (2010)CrossRefPubMedPubMedCentral
125.
go back to reference N.N. Rahbari, M. Aigner, K. Thorlund, N. Mollberg, E. Motschall, K. Jensen, M.K. Diener, M.W. Büchler, M. Koch, J. Weitz, Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer. Gastroenterol. 138, 1714–1726 (2010) N.N. Rahbari, M. Aigner, K. Thorlund, N. Mollberg, E. Motschall, K. Jensen, M.K. Diener, M.W. Büchler, M. Koch, J. Weitz, Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal  cancer. Gastroenterol. 138, 1714–1726 (2010)
126.
go back to reference X.-L. Ma, Z.-L. Xiao, L. Liu, X.-X. Liu, W. Nie, P. Li, N.-Y. Chen, Y.-Q. Wei, Meta-analysis of circulating tumor cells as a prognostic marker in lung cancer. Asian Pac. J. Cancer Prev. 13, 1137–1144 (2012)CrossRefPubMed X.-L. Ma, Z.-L. Xiao, L. Liu, X.-X. Liu, W. Nie, P. Li, N.-Y. Chen, Y.-Q. Wei, Meta-analysis of circulating tumor cells as a prognostic marker in lung cancer. Asian Pac. J. Cancer Prev. 13, 1137–1144 (2012)CrossRefPubMed
127.
go back to reference M. Berdasco, M. Esteller, Aberrant epigenetic landscape in cancer: How cellular identity goes awry. Dev. Cell 19, 698–711 (2010) M. Berdasco, M. Esteller, Aberrant epigenetic landscape in cancer: How cellular identity goes awry. Dev. Cell 19, 698–711 (2010)
128.
go back to reference I.P. Torres Filho, M. Leunig, F. Yuan, M. Intaglietta, R.K. Jain, Noninvasive measurement of microvascular and interstitial oxygen profiles in a human tumor in SCID mice. Proc. Natl. Acad. Sci. U. S. A. 91, 2081–2095 (1994)CrossRefPubMedPubMedCentral I.P. Torres Filho, M. Leunig, F. Yuan, M. Intaglietta, R.K. Jain, Noninvasive measurement of microvascular and interstitial oxygen profiles in a human tumor in SCID mice. Proc. Natl. Acad. Sci. U. S. A. 91, 2081–2095 (1994)CrossRefPubMedPubMedCentral
129.
go back to reference P. Bhat, G. Leggatt, N. Waterhouse, I.H. Frazer, Interferon-γ derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 8, e2836 (2017)CrossRefPubMedPubMedCentral P. Bhat, G. Leggatt, N. Waterhouse, I.H. Frazer, Interferon-γ derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis. 8, e2836 (2017)CrossRefPubMedPubMedCentral
130.
go back to reference C. Cekic, D. Sag, Y. Li, D. Theodorescu, R.M. Strieter, J. Linden, Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J. Immunol. 188, 198–205 (2012)CrossRefPubMed C. Cekic, D. Sag, Y. Li, D. Theodorescu, R.M. Strieter, J. Linden, Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J. Immunol. 188, 198–205 (2012)CrossRefPubMed
131.
132.
go back to reference A. Chatterjee, E.J. Rodger, M.R. Eccles, Epigenetic drivers of tumourigenesis and cancer metastasis. Semin. Cancer Biol. 51, 149–159 (2018)CrossRefPubMed A. Chatterjee, E.J. Rodger, M.R. Eccles, Epigenetic drivers of tumourigenesis and cancer metastasis. Semin. Cancer Biol. 51, 149–159 (2018)CrossRefPubMed
Metadata
Title
Tumor dormancy as an alternative step in the development of chemoresistance and metastasis - clinical implications
Authors
Federico Rossari
Cristina Zucchinetti
Gabriele Buda
Enrico Orciuolo
Publication date
01-04-2020
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 2/2020
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-019-00467-7

Other articles of this Issue 2/2020

Cellular Oncology 2/2020 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