Defining the Hallmarks of Metastasis

Cancer Research - Tập 79 Số 12 - Trang 3011-3027 - 2019
Danny R. Welch1, Douglas R. Hurst2
11Department of Cancer Biology and The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas.
22Department of Pathology and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.

Tóm tắt

AbstractMetastasis is the primary cause of cancer morbidity and mortality. The process involves a complex interplay between intrinsic tumor cell properties as well as interactions between cancer cells and multiple microenvironments. The outcome is the development of a nearby or distant discontiguous secondary mass. To successfully disseminate, metastatic cells acquire properties in addition to those necessary to become neoplastic. Heterogeneity in mechanisms involved, routes of dissemination, redundancy of molecular pathways that can be utilized, and the ability to piggyback on the actions of surrounding stromal cells makes defining the hallmarks of metastasis extraordinarily challenging. Nonetheless, this review identifies four distinguishing features that are required: motility and invasion, ability to modulate the secondary site or local microenvironments, plasticity, and ability to colonize secondary tissues. By defining these first principles of metastasis, we provide the means for focusing efforts on the aspects of metastasis that will improve patient outcomes.

Từ khóa


Tài liệu tham khảo

Eccles, 2007, Metastasis: recent discoveries and novel treatment strategies, Lancet, 369, 1742, 10.1016/S0140-6736(07)60781-8

Klein, 2011, Framework models of tumor dormancy from patient-derived observations, Curr Opin Genet Dev, 21, 42, 10.1016/j.gde.2010.10.011

Steeg, 2007, Metastasis: a therapeutic target for cancer, Nat Clin Pract Oncol, 5, 206, 10.1038/ncponc1066

Hanahan, 2000, The hallmarks of cancer, Cell, 100, 57, 10.1016/S0092-8674(00)81683-9

Hanahan, 2011, Hallmarks of cancer: the next generation, Cell, 144, 646, 10.1016/j.cell.2011.02.013

Lazebnik, 2010, What are the hallmarks of cancer?, Nat Rev Cancer, 10, 232, 10.1038/nrc2827

Welch, 1985, Implications of tumor progression on clinical oncology, Clin Exp Metastasis, 3, 151, 10.1007/BF01786761

Heppner, 1998, The cellular basis of tumor progression, Int Rev Cytol, 177, 1

Foulds, 1957, Tumor progression, Cancer Res, 17, 355

Vogelstein, 1988, Genetic alterations during colorectal-tumor development, N Engl J Med, 319, 525, 10.1056/NEJM198809013190901

Kunz-Schughart, 2002, Tumor-associated fibroblasts (part I): active stromal participants in tumor development and progression?, Histol Histopathol, 17, 599

Dvorak, 1983, Fibrin as a component of the tumor stroma - origins and biological significance, Cancer Metastasis Rev, 2, 41, 10.1007/BF00046905

Welch, 2006, Defining a cancer metastasis, AACR Education Book 2006, 111

Steeg, 2016, Targeting metastasis, Nat Rev Cancer, 16, 201, 10.1038/nrc.2016.25

Fidler, 1991, The biology of cancer metastasis or, you cannot fix it if you do not know how it works, Bioessays, 13, 551, 10.1002/bies.950131010

Fidler, 2003, Timeline - The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited, Nat Rev Cancer, 3, 453, 10.1038/nrc1098

Sleeman, 2011, Do all roads lead to Rome? Routes to metastasis development, Int J Cancer, 128, 2511, 10.1002/ijc.26027

Roh, 2015, Perineural growth in head and neck squamous cell carcinoma: a review, Oral Oncol, 51, 16, 10.1016/j.oraloncology.2014.10.004

Marchesi, 2010, Molecular mechanisms of perineural invasion, a forgotten pathway of dissemination and metastasis, Cytokine Growth Factor Rev, 21, 77, 10.1016/j.cytogfr.2009.11.001

Lugassy, 2004, Pericyte-like location of GFP-tagged melanoma cells: ex vivo and in vivo studies of extravascular migratory metastasis, Am J Pathol, 164, 1191, 10.1016/S0002-9440(10)63207-5

Blanpain, 2013, Tracing the cellular origin of cancer, Nat Cell Biol, 15, 126, 10.1038/ncb2657

Yamamoto, 2003, Determination of clonality of metastasis by cell-specific color-coded fluorescent-protein imaging, Cancer Res, 63, 7785

Talmadge, 1982, Evidence for the clonal origin of spontaneous metastases, Science, 217, 361, 10.1126/science.6953592

Navin, 2011, Tumour evolution inferred by single-cell sequencing, Nature, 472, 90, 10.1038/nature09807

McGranahan, 2017, Cancer evolution constrained by the immune microenvironment, Cell, 170, 825, 10.1016/j.cell.2017.08.012

McGranahan, 2017, Clonal heterogeneity and tumor evolution: past, present, and the future, Cell, 168, 613, 10.1016/j.cell.2017.01.018

Folkman, 2006, Angiogenesis, Annu Rev Med, 57, 1, 10.1146/annurev.med.57.121304.131306

Paulis, 2010, Signalling pathways in vasculogenic mimicry, Biochim Biophys Acta, 1806, 18

Sleeman, 2009, Tumor metastasis and the lymphatic vasculature, Int J Cancer, 125, 2747, 10.1002/ijc.24702

Folberg, 2004, Vasculogenic mimicry, APMIS, 112, 508, 10.1111/j.1600-0463.2004.apm11207-0810.x

Bentolila, 2016, Imaging of angiotropism/vascular co-option in a murine model of brain melanoma: implications for melanoma progression along extravascular pathways, Sci Rep, 6, 23834, 10.1038/srep23834

Maniotis, 1999, Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry, Am J Pathol, 155, 739, 10.1016/S0002-9440(10)65173-5

Welch, 2016, Tumor heterogeneity–a ‘contemporary concept’ founded on historical insights and predictions, Cancer Res, 76, 4, 10.1158/0008-5472.CAN-15-3024

Heppner, 1993, Cancer cell societies and tumor progression, Stem Cells, 11, 199, 10.1002/stem.5530110306

Hunter, 2018, Genetic insights into the morass of metastatic heterogeneity, Nat Rev Cancer, 18, 211, 10.1038/nrc.2017.126

El-Kebir, 2018, Inferring parsimonious migration histories for metastatic cancers, Nat Genet, 50, 718, 10.1038/s41588-018-0106-z

Baslan, 2017, Unravelling biology and shifting paradigms in cancer with single-cell sequencing, Nat Rev Cancer, 17, 557, 10.1038/nrc.2017.58

Abbosh, 2017, Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution, Nature, 545, 446, 10.1038/nature22364

Gundem, 2015, The evolutionary history of lethal metastatic prostate cancer, Nature, 520, 353, 10.1038/nature14347

Fidler, 1986, Evidence that intraveously derived murine pulmonary melanoma metastases can originate from the expansion of a single tumor cell, Cancer Res, 46, 5167

Wolman, 1985, Development and progression of karyotypic variability in melanoma K1735 following X-irradiation, Cancer Res, 45, 1839

Poste, 1981, Interactions among clonal subpopulations affect stability of the metastatic phenotype in polyclonal populations of B16 melanoma cells, Proc Natl Acad Sci U S A, 78, 6226, 10.1073/pnas.78.10.6226

Poste, 1982, Evolution of tumor cell heterogeneity during progressive growth of individual lung metastases, Proc Natl Acad Sci U S A, 79, 6574, 10.1073/pnas.79.21.6574

Fidler, 1977, Metastasis results from pre-existing variant cells within a malignant tumor, Science, 197, 893, 10.1126/science.887927

Liu, 2007, The prognostic role of a gene signature from tumorigenic breast-cancer cells, N Engl J Med, 356, 217, 10.1056/NEJMoa063994

Kang, 2003, A multigenic program mediating breast cancer metastasis to bone, Cancer Cell, 3, 537, 10.1016/S1535-6108(03)00132-6

Nowell, 1976, The clonal evolution of tumor cell populations, Science, 194, 23, 10.1126/science.959840

Fidler, 1973, Selection of successive tumor lines for metastasis, Nat New Biol, 242, 148, 10.1038/newbio242148a0

Nicolson, 1988, Organ specificity of tumor-metastasis - Role of preferential adhesion, invasion and growth of malignant-cells at specific secondary sites, Cancer Metastasis Rev, 7, 143, 10.1007/BF00046483

Bohl, 2014, Metastasis suppressors in breast cancers: mechanistic insights and clinical potential, J Mol Med, 92, 13, 10.1007/s00109-013-1109-y

Welch, 2012, Preface. Metastasis genes, Cancer Metastasis Rev, 31, 417, 10.1007/s10555-012-9386-4

Welch, 2009, Metastsis suppressors: Discovery, mechanisms and translation, AACR Education Book 2009, 191

Thiery, 2002, Epithelial-mesenchymal transitions in tumour progression, Nat Rev Cancer, 2, 442, 10.1038/nrc822

Illmensee, 1976, Totipotency and normal differentiation of single teratocarcinoma cells cloned by injection into blastocysts, Proc Natl Acad Sci U S A, 73, 549, 10.1073/pnas.73.2.549

Kulesa, 2006, Reprogramming metastatic melanoma cells to assume a neural crest cell-like phenotype in an embryonic microenvironment, Proc Natl Acad Sci U S A, 103, 3752, 10.1073/pnas.0506977103

Kasemeier-Kulesa, 2018, NGF reprograms metastatic melanoma to a bipotent glial-melanocyte neural crest-like precursor, Biol Open, 7

van der Horst, 2012, Epithelial plasticity, cancer stem cells, and the tumor-supportive stroma in bladder carcinoma, Mol Cancer Res, 10, 995, 10.1158/1541-7786.MCR-12-0274

Psaila, 2009, The metastatic niche: adapting the foreign soil, Nat Rev Cancer, 9, 285, 10.1038/nrc2621

Kaplan, 2006, Preparing the "soil": the premetastatic niche, Cancer Res, 66, 11089, 10.1158/0008-5472.CAN-06-2407

Peinado, 2017, Pre-metastatic niches: organ-specific homes for metastases, Nat Rev Cancer, 17, 302, 10.1038/nrc.2017.6

Zhang, 2018, Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation, Nat Cell Biol, 20, 332, 10.1038/s41556-018-0040-4

Costa-Silva, 2015, Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver, Nat Cell Biol, 17, 816, 10.1038/ncb3169

Sansone, 2017, Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer, Proc Natl Acad Sci U S A, 114, E9066, 10.1073/pnas.1704862114

Matei, 2017, Unshielding exosomal RNA unleashes tumor growth and metastasis, Cell, 170, 223, 10.1016/j.cell.2017.06.047

Hoshino, 2015, Tumour exosome integrins determine organotropic metastasis, Nature, 527, 329, 10.1038/nature15756

Cox, 2015, The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase, Nature, 522, 106, 10.1038/nature14492

Hou, 2009, Deformability study of breast cancer cells using microfluidics, Biomed Microdevices, 11, 557, 10.1007/s10544-008-9262-8

Ochalek, 1988, Correlation between cell deformability and metastatic potential in B16-F1 melanoma cell variants, Cancer Res, 48, 5124

Skau, 2016, FMN2 makes perinuclear actin to protect nuclei during confined migration and promote metastasis, Cell, 167, 1571, 10.1016/j.cell.2016.10.023

Hou, 2008, Permissive and repulsive cues and signalling pathways of axonal outgrowth and regeneration, Int Rev Cell Mol Biol, 267, 125, 10.1016/S1937-6448(08)00603-5

Kruger, 2005, Semaphorins command cells to move, Nat Rev Mol Cell Biol, 6, 789, 10.1038/nrm1740

Seiki, 1991, Comparison of autocrine mechanisms promoting motility in two metastatic cell lines: human melanoma and ras-transfected NIH3T3 cells, Int J Cancer, 49, 717, 10.1002/ijc.2910490515

Stuelten, 2018, Cell motility in cancer invasion and metastasis: insights from simple model organisms, Nat Rev Cancer, 18, 296, 10.1038/nrc.2018.15

McCarthy, 1984, Laminin and fibronectin promote the haptotactic migration of B16 mouse melanoma cells in vitro, J Cell Biol, 98, 1474, 10.1083/jcb.98.4.1474

Te Boekhorst, 2016, Plasticity of cell migration in vivo and in silico, Annu Rev Cell Dev Biol, 32, 491, 10.1146/annurev-cellbio-111315-125201

van Helvert, 2018, Mechanoreciprocity in cell migration, Nat Cell Biol, 20, 8, 10.1038/s41556-017-0012-0

Chaffer, 2016, EMT, cell plasticity and metastasis, Cancer Metastasis Rev, 35, 645, 10.1007/s10555-016-9648-7

Lambert, 2017, Emerging biological principles of metastasis, Cell, 168, 670, 10.1016/j.cell.2016.11.037

Mani, 2008, The epithelial-mesenchymal transition generates cells with properties of stem cells, Cell, 133, 704, 10.1016/j.cell.2008.03.027

Chaffer, 2006, Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: role of fibroblast growth factor receptor-2, Cancer Res, 66, 11271, 10.1158/0008-5472.CAN-06-2044

Tsai, 2012, Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis, Cancer Cell, 22, 725, 10.1016/j.ccr.2012.09.022

Yu, 2013, Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition, Science, 339, 580, 10.1126/science.1228522

Nieto, 2013, Epithelial plasticity: a common theme in embryonic and cancer cells, Science, 342, 1234850, 10.1126/science.1234850

Saitoh, 2018, Involvement of partial EMT in cancer progression, J Biochem, 164, 257, 10.1093/jb/mvy047

Tsuji, 2009, Epithelial-mesenchymal transition and cell cooperativity in metastasis, Cancer Res, 69, 7135, 10.1158/0008-5472.CAN-09-1618

Tsuji, 2008, Epithelial-mesenchymal transition induced by growth suppressor p12CDK2-AP1 promotes tumor cell local invasion but suppresses distant colony growth, Cancer Res, 68, 10377, 10.1158/0008-5472.CAN-08-1444

Zheng, 2015, Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer, Nature, 527, 525, 10.1038/nature16064

Fischer, 2015, Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance, Nature, 527, 472, 10.1038/nature15748

Esserman, 2019, Should we rename low risk cancers?, BMJ, 364, k4699, 10.1136/bmj.k4699

Liotta, 1986, Tumor Invasion and Metastases Role of the Extracellular-Matrix - Rhoads Memorial Award Lecture, Cancer Res, 46, 1

Friedl, 2012, Classifying collective cancer cell invasion, Nat Cell Biol, 14, 777, 10.1038/ncb2548

Chambers, 1997, Changing views of the role of matrix metalloproteinases in metastasis, J Natl Cancer Inst, 89, 1260, 10.1093/jnci/89.17.1260

Naba, 2014, Extracellular matrix signatures of human mammary carcinoma identify novel metastasis promoters, Elife, 3, e01308, 10.7554/eLife.01308

Salvador, 2017, Lysyl oxidase-like protein LOXL2 promotes lung metastasis of breast cancer, Cancer Res, 77, 5846, 10.1158/0008-5472.CAN-16-3152

Moon, 2013, MCF-7 cells expressing nuclear associated lysyl oxidase-like 2 (LOXL2) exhibit an epithelial-to-mesenchymal transition (EMT) phenotype and are highly invasive in vitro, J Biol Chem, 288, 30000, 10.1074/jbc.C113.502310

Patel, 2018, An extracellular matrix fragment drives epithelial remodeling and airway hyperresponsiveness, Sci Transl Med, 10, 10.1126/scitranslmed.aaq0693

Arroyo, 2010, Extracellular matrix, inflammation, and the angiogenic response, Cardiovasc Res, 86, 226, 10.1093/cvr/cvq049

Duca, 2004, Elastin as a matrikine, Crit Rev Oncol Hematol, 49, 235, 10.1016/j.critrevonc.2003.09.007

Tran, 2004, Extracellular matrix signaling through growth factor receptors during wound healing, Wound Repair Regen, 12, 262, 10.1111/j.1067-1927.2004.012302.x

Liotta, 1992, Cancer cell invasion and metastasis, Sci Am, 266, 54, 10.1038/scientificamerican0292-54

Hehlgans, 2007, Signalling via integrins: implications for cell survival and anticancer strategies, Biochim Biophys Acta, 1775, 163

Schwartz, 2002, Networks and crosstalk: integrin signalling spreads, Nat Cell Biol, 4, E65, 10.1038/ncb0402-e65

Hamidi, 2018, Every step of the way: integrins in cancer progression and metastasis, Nat Rev Cancer, 18, 533, 10.1038/s41568-018-0038-z

Fingleton, 2006, Matrix metalloproteinases: roles in cancer and metastasis, Front Biosci, 11, 479, 10.2741/1811

Coussens, 2002, Cancer therapy - Matrix metalloproteinase inhibitors and cancer: trials and tribulations, Science, 295, 2387, 10.1126/science.1067100

Egeblad, 2002, New functions for the matrix metalloproteinases in cancer progression, Nat Rev Cancer, 2, 161, 10.1038/nrc745

Mason, 2011, Proteolytic networks in cancer, Trends Cell Biol, 21, 228, 10.1016/j.tcb.2010.12.002

Allgayer, 2010, Translational research on u-PAR, Eur J Cancer, 46, 1241, 10.1016/j.ejca.2010.02.029

Denais, 2016, Nuclear envelope rupture and repair during cancer cell migration, Science, 352, 353, 10.1126/science.aad7297

Weaver, 2006, Invadopodia: specialized cell structures for cancer invasion, Clin Exp Metastasis, 23, 97, 10.1007/s10585-006-9014-1

Revach, 2013, The interplay between the proteolytic, invasive, and adhesive domains of invadopodia and their roles in cancer invasion, Cell Adh Migr, 8

Murphy, 2011, The ‘ins’ and ‘outs’ of podosomes and invadopodia: characteristics, formation and function, Nat Rev Mol Cell Biol, 12, 413, 10.1038/nrm3141

Charras, 2014, Physical influences of the extracellular environment on cell migration, Nat Rev Mol Cell Biol, 15, 813, 10.1038/nrm3897

Bonnans, 2014, Remodelling the extracellular matrix in development and disease, Nat Rev Mol Cell Biol, 15, 786, 10.1038/nrm3904

Huang, 2005, Cell tension, matrix mechanics, and cancer development, Cancer Cell, 8, 175, 10.1016/j.ccr.2005.08.009

Mouw, 2014, Extracellular matrix assembly: a multiscale deconstruction, Nat Rev Mol Cell Biol, 15, 771, 10.1038/nrm3902

Kumar, 2009, Mechanics, malignancy, and metastasis: the force journey of a tumor cell, Cancer Metastasis Rev, 28, 113, 10.1007/s10555-008-9173-4

Erdogan, 2017, Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin, J Cell Biol, 216, 3799, 10.1083/jcb.201704053

Friedl, 2009, Proteolytic interstitial cell migration: a five-step process, Cancer Metastasis Rev, 28, 129, 10.1007/s10555-008-9174-3

Friedl, 2008, Tube travel: the role of proteases in individual and collective cancer cell invasion, Cancer Res, 68, 7247, 10.1158/0008-5472.CAN-08-0784

Allavena, 2008, The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance, Immunol Rev, 222, 155, 10.1111/j.1600-065X.2008.00607.x

Carron, 2017, Macrophages promote the progression of premalignant mammary lesions to invasive cancer, Oncotarget, 8, 50731, 10.18632/oncotarget.14913

Piccard, 2012, On the dual roles and polarized phenotypes of neutrophils in tumor development and progression, Crit Rev Oncol Hematol, 82, 296, 10.1016/j.critrevonc.2011.06.004

Attieh, 2016, The hallmarks of CAFs in cancer invasion, Eur J Cell Biol, 95, 493, 10.1016/j.ejcb.2016.07.004

Aeed, 1988, The role of polymorphonuclear leukocytes (PMN) on the growth and metastatic potential of 13762NF mammary adenocarcinoma cells, Int J Cancer, 42, 748, 10.1002/ijc.2910420521

McGary, 1995, Highly metastatic 13762NF rat mammary adenocarcinoma cell clones stimulate bone marrow by secretion of granulocyte-macrophage colony-stimulating factor/interleukin-3 activity, Am J Pathol, 147, 1668

Safari, 2019, Myeloid-derived suppressor cells and tumor: current knowledge and future perspectives, J Cell Physiol, 234, 9966, 10.1002/jcp.27923

Talmadge, 2007, Inflammatory cell infiltration of tumors: Jekyll or Hyde, Cancer Metastasis Rev, 26, 373, 10.1007/s10555-007-9072-0

Wyckoff, 2007, Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors, Cancer Res, 67, 2649, 10.1158/0008-5472.CAN-06-1823

Yamaguchi, 2005, Cell migration in tumors, Curr Opin Cell Biol, 17, 559, 10.1016/j.ceb.2005.08.002

Condeelis, 2006, Macrophages: obligate partners for tumor cell migration, invasion, and metastasis, Cell, 124, 263, 10.1016/j.cell.2006.01.007

Huber, 1991, Regulation of transendothelial neutrophil migration by endogenous interleukin-8, Science, 254, 99, 10.1126/science.1718038

Nicolson, 1982, Metastatic tumor cell attachment and invasion assay utilizing vascular endothelial cell monolayers, J Histochem Cytochem, 30, 214, 10.1177/30.3.7061823

De Palma, 2017, Microenvironmental regulation of tumour angiogenesis, Nat Rev Cancer, 17, 457, 10.1038/nrc.2017.51

Stacker, 2014, Lymphangiogenesis and lymphatic vessel remodelling in cancer, Nat Rev Cancer, 14, 159, 10.1038/nrc3677

Keck, 1989, Vasular permeability factor, an endothelial cell mitogen related to PDGF, Science, 246, 1309, 10.1126/science.2479987

Kramer, 1979, Interactions of tumor cells with vascular endothelial cell monolayers: a model for metastatic invasion, Proc Natl Acad Sci U S A, 76, 5704, 10.1073/pnas.76.11.5704

Overholtzer, 2007, A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion, Cell, 131, 966, 10.1016/j.cell.2007.10.040

Coman, 1944, Decreased mutual adhesiveness, a property of cells from squamous cell carcinomas, Cancer Res, 4, 625

Zeidman, 1950, Factors affecting the number of tumor metastases; experiments with a transplantable mouse tumor, Cancer Res, 10, 357

Wong, 2017, Mitosis-mediated intravasation in a tissue-engineered tumor-microvessel platform, Cancer Res, 77, 6453, 10.1158/0008-5472.CAN-16-3279

Butler, 1975, Quantitation of cell shedding into efferent blood of mammary adenocarcinoma, Cancer Res, 35, 512

Jain, 1987, Transport of molecules accross tumor vasculature, Cancer Metastasis Rev, 6, 559, 10.1007/BF00047468

Fidler, 1970, Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125I-5-iodo-2′-deoxyuridine, J Natl Cancer Inst, 45, 773

Tarin, 1984, Mechanisms of human tumor metastasis studied in patients with peritoneovenous shunts, Cancer Res, 44, 3584

Ao, 2015, Identification of cancer-associated fibroblasts in circulating blood from patients with metastatic breast cancer, Cancer Res, 75, 4681, 10.1158/0008-5472.CAN-15-1633

Fidler, 1973, The relationship of embolic heterogeneity, number size and viability to the incidence of experimental metastasis, Eur J Cancer, 9, 223, 10.1016/S0014-2964(73)80022-2

Updyke, 1986, Malignant melanoma cell lines selected in vitro for increased homotypic adhesion properties have increased experimental metastasis potential, CEM, 4, 273

Weiss, 1992, Comments on hematogenous metastatic patterns in humans as revealed by autopsy, Clin Exp Metastasis, 10, 191, 10.1007/BF00132751

Ewing, 1919, Neoplastic disease, 10.1097/00000658-191903000-00014

Gkountela, 2019, Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding, Cell, 176, 98, 10.1016/j.cell.2018.11.046

Krog, 2018, Biomechanics of the circulating tumor cell microenvironment, Adv Exp Med Biol, 1092, 209, 10.1007/978-3-319-95294-9_11

Paoletti, 2015, Significance of circulating tumor cells in metastatic triple-negative breast cancer patients within a randomized, phase II trial: TBCRC 019, Clin Cancer Res, 21, 2771, 10.1158/1078-0432.CCR-14-2781

Aceto, 2015, En route to metastasis: circulating tumor cell clusters and epithelial-to-mesenchymal transition, Trends Cancer, 1, 44, 10.1016/j.trecan.2015.07.006

Aceto, 2014, Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis, Cell, 158, 1110, 10.1016/j.cell.2014.07.013

Crissman, 1985, Arrest and extravasation of B16 amelanotic melanoma in murine lungs: a light and EM study, Lab Invest, 53, 470

Ito, 2001, Real-time observation of micrometastasis formation in the living mouse liver using a green fluorescent protein gene-tagged rat tongue carcinoma cell line, Int J Cancer, 93, 212, 10.1002/ijc.1318

Springer, 1994, Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm, Cell, 76, 301, 10.1016/0092-8674(94)90337-9

Stupack, 2001, Apoptosis of adherent cells by recruitment of caspase-8 to unligated integrins, J Cell Biol, 155, 459, 10.1083/jcb.200106070

Alby, 1984, Differential adhesion of tumor cells to capillary endothelial cells in vitro, Proc Natl Acad Sci U S A, 81, 5739, 10.1073/pnas.81.18.5739

Auerbach, 1987, Specificity of adhesion between murine tumor cells and capillary endothelium: an in vitro correlate of preferential metastasis in vivo, Cancer Res, 47, 1492

Belloni, 1989, Microvascular endothelial cell heterogeneity: interactions with leukocytes and tumor cells, Cancer Metastasis Rev, 8, 353, 10.1007/BF00052608

Belloni, 1988, Differential expression of cell-surface glycoproteins on various organ-derived microvascular endothelia and endothelial-cell cultures, J Cell Physiol, 136, 398, 10.1002/jcp.1041360303

Pasqualini, 2002, Probing the structural and molecular diversity of tumor vasculature, Trends Mol Med, 8, 563, 10.1016/S1471-4914(02)02429-2

Pasqualini, 1996, Organ targeting in vivo using phage display peptide libraries, Nature, 380, 364, 10.1038/380364a0

Lala, 1998, Role of nitric oxide in tumor progression: lessons from experimental tumors, Cancer Metastasis Rev, 17, 91, 10.1023/A:1005960822365

Orr, 1987, Effects of neutrophil-mediated pulmonary endothelial injury on the localization and metastasis of circulating Walker carcinosarcoma cells, Invasion Metastasis, 7, 183

Orr, 1986, Promotion of pulmonary metastasis in mice by bleomycin-induced endothelial injury, Cancer Res, 46, 891

Sugarbaker, 1977, Mechanisms and prevention of cancer dissemination: an overview, Semin Oncol, 4, 19

Cho, 2010, Breast cancer cutaneous metastasis at core needle biopsy site, Ann Dermatol, 22, 238, 10.5021/ad.2010.22.2.238

Retsky, 2008, Dormancy and surgery-driven escape from dormancy help explain some clinical features of breast cancer, APMIS, 116, 730, 10.1111/j.1600-0463.2008.00990.x

Osborne, 1990, Leukocyte adhesion to endothelium in inflammation, Cell, 62, 3, 10.1016/0092-8674(90)90230-C

Chambers, 2002, Dissemination and growth of cancer cells in metastatic sites, Nat Rev Cancer, 2, 563, 10.1038/nrc865

Al-Mehdi, 2000, Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis, Nat Med, 6, 100, 10.1038/71429

Naumov, 2008, Tumor-vascular interactions and tumor dormancy, APMIS, 116, 569, 10.1111/j.1600-0463.2008.01213.x

Hendrix, 2003, Vasculogenic mimicry and tumour-cell plasticity: lessons from melanoma, Nat Rev Cancer, 3, 411, 10.1038/nrc1092

Kaplan, 2005, VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche, Nature, 438, 820, 10.1038/nature04186

Wculek, 2015, Neutrophils support lung colonization of metastasis-initiating breast cancer cells, Nature, 528, 413, 10.1038/nature16140

Albrengues, 2018, Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice, Science, 361, 10.1126/science.aao4227

Cedervall, 2016, Tumor-induced NETosis as a risk factor for metastasis and organ failure, Cancer Res, 76, 4311, 10.1158/0008-5472.CAN-15-3051

Demers, 2012, Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis, Proc Natl Acad Sci U S A, 109, 13076, 10.1073/pnas.1200419109

Langley, 2011, The seed and soil hypothesis revisited–the role of tumor-stroma interactions in metastasis to different organs, Int J Cancer, 128, 2527, 10.1002/ijc.26031

Paget, 1889, The distribution of secondary growths in cancer of the breast, Lancet, 1, 571, 10.1016/S0140-6736(00)49915-0

Beadnell, 2018, Roles of the mitochondrial genetics in cancer metastasis: not to be ignored any longer, Cancer Metastasis Rev, 37, 615, 10.1007/s10555-018-9772-7

Brinker, 2017, Mitochondrial haplotype alters mammary cancer tumorigenicity and metastasis in an oncogenic driver-dependent manner, Cancer Res, 77, 6941, 10.1158/0008-5472.CAN-17-2194

Feeley, 2015, Mitochondrial genetics regulate breast cancer tumorigenicity and metastatic potential, Cancer Res, 75, 4429, 10.1158/0008-5472.CAN-15-0074

Telonis, 2018, Race disparities in the contribution of miRNA isoforms and tRNA-derived fragments to triple-negative breast cancer, Cancer Res, 78, 1140, 10.1158/0008-5472.CAN-17-1947

Warburg, 1956, On the origin of cancer cells, Science, 123, 309, 10.1126/science.123.3191.309

Weber, 2016, Metabolism in cancer metastasis, Int J Cancer, 138, 2061, 10.1002/ijc.29839

Valcarcel-Jimenez, 2017, Mitochondrial metabolism: yin and yang for tumor progression, Trends Endocrinol Metab, 28, 748, 10.1016/j.tem.2017.06.004

Tan, 2015, Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA, Cell Metab, 21, 81, 10.1016/j.cmet.2014.12.003

Porporato, 2015, Paving the way for therapeutic prevention of tumor metastasis with agents targeting mitochondrial superoxide, Mol Cell Oncol, 2, e968043, 10.4161/23723548.2014.968043

Liu, 2013, Metastasis suppressor KISS1 appears to reverse the Warburg effect by enhancing mitochondrial biogenesis, Cancer Res, 74, 954, 10.1158/0008-5472.CAN-13-1183

LeBleu, 2014, PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis, Nat Cell Biol, 16, 992, 10.1038/ncb3039

Santidrian, 2013, Mitochondrial complex I activity and NAD+/NADH balance regulate breast cancer progression, J Clin Invest, 123, 1068, 10.1172/JCI64264

Seo, 2018, Syntaphilin ubiquitination regulates mitochondrial dynamics and tumor cell movements, Cancer Res, 78, 4215, 10.1158/0008-5472.CAN-18-0595

Altieri, 2019, Mitochondrial dynamics and metastasis, Cell Mol Life Sci, 76, 827, 10.1007/s00018-018-2961-2

Lisanti, 2016, Transgenic expression of the mitochondrial chaperone TNFR-associated protein 1 (TRAP1) accelerates prostate cancer development, J Biol Chem, 291, 25247, 10.1074/jbc.M116.745950

Caino, 2016, A neuronal network of mitochondrial dynamics regulates metastasis, Nat Commun, 7, 13730, 10.1038/ncomms13730

Mowers, 2017, Autophagy in cancer metastasis, Oncogene, 36, 1619, 10.1038/onc.2016.333

Vlahakis, 2017, The interconnections between autophagy and integrin-mediated cell adhesion, J Mol Biol, 429, 515, 10.1016/j.jmb.2016.11.027

Pauli, 1990, Organ-preference of metastasis. The role of endothelial cell adhesion molecules, Cancer Metastasis Rev, 9, 175, 10.1007/BF00046359

Nicolson, 1989, Adhesive, invasive, and growth properties of selected metastatic variants of a murine large-cell lymphoma, Invasion Metastasis, 9, 102

Nicolson, 1985, Preferential organ attachment and invasion in vitro by B16 melanoma cells selected for differing metastatic colonization and invasive properties, Invasion Metastasis, 5, 144

Aguirre-Ghiso, 2018, Emerging topics on disseminated cancer cell dormancy and the paradigm of metastasis, Annu Rev Cancer Biol, 2, 377, 10.1146/annurev-cancerbio-030617-050446

Hosseini, 2016, Early dissemination seeds metastasis in breast cancer, Nature, 540, 552, 10.1038/nature20785

Hoffman, 2015, Patient-derived orthotopic xenografts: better mimic of metastasis than subcutaneous xenografts, Nat Rev Cancer, 15, 451, 10.1038/nrc3972

Ghinassi, 2010, Evidence for organ-specific stem cell microenvironments, J Cell Physiol, 223, 460, 10.1002/jcp.22055

Minn, 2005, Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors, J Clin Invest, 115, 44, 10.1172/JCI22320

Boire, 2017, Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis, Cell, 168, 1101, 10.1016/j.cell.2017.02.025

Bos, 2009, Genes that mediate breast cancer metastasis to the brain, Nature, 459, 1005, 10.1038/nature08021

van der Weyden, 2017, Genome-wide in vivo screen identifies novel host regulators of metastatic colonization, Nature, 541, 233, 10.1038/nature20792

Nicolson, 1986, Organ specificity of metastatic tumor colonization is related to organ-selective growth-properties of malignant-cells, Int J Cancer, 38, 289, 10.1002/ijc.2910380221

Altorki, 2019, The lung microenvironment: an important regulator of tumour growth and metastasis, Nat Rev Cancer, 19, 9, 10.1038/s41568-018-0081-9

Zhuang, 2017, Differential effects on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1, Nat Cell Biol, 19, 1274, 10.1038/ncb3613

Bowman, 2016, Macrophage ontogeny underlies differences in tumor-specific education in brain malignancies, Cell Rep, 17, 2445, 10.1016/j.celrep.2016.10.052

Kim, 2011, Astrocytes upregulate survival genes in tumor cells and induce protection from chemotherapy, Neoplasia, 13, 286, 10.1593/neo.11112

Jackson, 2017, Role of megakaryocytes in breast cancer metastasis to bone, Cancer Res, 77, 1942, 10.1158/0008-5472.CAN-16-1084

Ku, 2016, Tumor-induced MDSC act via remote control to inhibit L-selectin-dependent adaptive immunity in lymph nodes, Elife, 5, 10.7554/eLife.17375

Muller, 2001, Involvement of chemokine receptors in breast cancer metastasis, Nature, 410, 50, 10.1038/35065016

Gay, 2017, The sleeping ugly: Tumour microenvironment's act to make or break the spell of dormancy, Biochim Biophys Acta Rev Cancer, 1868, 231, 10.1016/j.bbcan.2017.05.002

Yeh, 2015, Mechanisms of cancer cell dormancy-another hallmark of cancer?, Cancer Res, 75, 5014, 10.1158/0008-5472.CAN-15-1370

Sosa, 2014, Mechanisms of disseminated cancer cell dormancy: an awakening field, Nat Rev Cancer, 14, 611, 10.1038/nrc3793

Hadfield, 1954, The dormant cancer cell, Br Med J, 2, 607, 10.1136/bmj.2.4888.607

Vishnoi, 2015, The isolation and characterization of CTC subsets related to breast cancer dormancy, Sci Rep, 5, 17533, 10.1038/srep17533

Naumov, 2006, A model of human tumor dormancy: an angiogenic switch from the nonangiogenic phenotype, J Natl Cancer Inst, 98, 316, 10.1093/jnci/djj068

Manjili, 2018, A theoretical basis for the efficacy of cancer immunotherapy and immunogenic tumor dormancy: the adaptation model of immunity, Adv Cancer Res, 137, 17, 10.1016/bs.acr.2017.11.005

Yumoto, 2014, Molecular pathways: niches in metastatic dormancy, Clin Cancer Res, 20, 3384, 10.1158/1078-0432.CCR-13-0897

Kaur, 2019, Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility, Cancer Discov, 9, 64, 10.1158/2159-8290.CD-18-0193

Fidler, 1977, Fate of recirculating B16 melanoma metastatic variant cells in parabiotic syngeneic recipients, J Natl Cancer Inst, 58, 1867, 10.1093/jnci/58.6.1867

Bartkowiak, 2015, Disseminated tumor cells persist in the bone marrow of breast cancer patients through sustained activation of the unfolded protein response, Cancer Res, 75, 5367, 10.1158/0008-5472.CAN-14-3728

Pantel, 1999, Detection and clinical importance of micrometastatic disease, J Natl Cancer Inst, 91, 1113, 10.1093/jnci/91.13.1113

Friberg, 2015, Cancer metastases: early dissemination and late recurrences, Cancer Growth Metastasis, 8, 43, 10.4137/CGM.S31244

Harper, 2016, Mechanism of early dissemination and metastasis in Her2(+) mammary cancer, Nature, 540, 588, 10.1038/nature20609

Leonard, 2004, Ductal carcinoma in situ, complexities and challenges, J Natl Cancer Inst, 96, 906, 10.1093/jnci/djh164

Silver, 1998, Mammary ductal carcinoma in situ with microinvasion, Cancer, 82, 2382, 10.1002/(SICI)1097-0142(19980615)82:12<2382::AID-CNCR12>3.0.CO;2-L

Narod, 2015, Breast cancer mortality after a diagnosis of ductal carcinoma in situ, JAMA Oncol, 1, 888, 10.1001/jamaoncol.2015.2510

Turajlic, 2016, Metastasis as an evolutionary process, Science, 352, 169, 10.1126/science.aaf2784

Subarsky, 2003, The hypoxic tumour microenvironment and metastatic progression, Clin Exp Metastasis, 20, 237, 10.1023/A:1022939318102

De Jaeger, 2001, Relationship of hypoxia to metastatic ability in rodent tumours, Br J Cancer, 84, 1280, 10.1054/bjoc.2001.1743

Gilkes, 2014, Hypoxia and the extracellular matrix: drivers of tumour metastasis, Nat Rev Cancer, 14, 430, 10.1038/nrc3726

Gilkes, 2014, Hypoxia-inducible factors mediate coordinated RhoA-ROCK1 expression and signaling in breast cancer cells, Proc Natl Acad Sci U S A, 111, E384, 10.1073/pnas.1321510111

Castro-Vega, 2015, The senescent microenvironment promotes the emergence of heterogeneous cancer stem-like cells, Carcinogenesis, 36, 1180, 10.1093/carcin/bgv101

Pan, 2017, 20-year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years, N Engl J Med, 377, 1836, 10.1056/NEJMoa1701830

Alix-Panabieres, 2016, Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy, Cancer Discov, 6, 479, 10.1158/2159-8290.CD-15-1483

Christiano, 2000, Development of markers of prostate cancer metastasis. Review and perspective, Urol Oncol, 5, 217, 10.1016/S1078-1439(00)00070-3

Floor, 2012, Hallmarks of cancer: of all cancer cells, all the time?, Trends Mol Med, 18, 509, 10.1016/j.molmed.2012.06.005

Lawson, 2018, Tumour heterogeneity and metastasis at single-cell resolution, Nat Cell Biol, 20, 1349, 10.1038/s41556-018-0236-7

Welch, 1987, Biologic considerations for drug targeting in cancer patients, Cancer Treat Rev, 14, 351, 10.1016/0305-7372(87)90029-6

Heppner, 1984, Tumor heterogeneity, Cancer Res, 44, 2259

Rak, 1993, Growth advantage ("clonal dominance") of metastatically competent tumor cell variants expressed under selective two- or three-dimensional tissue culture conditions, In Vitro Cell Dev Biol Anim, 29A, 742, 10.1007/BF02631431

Miller, 1986, Metabolic cooperation between mouse mammary tumor subpopulations in three-dimensional collagen gel cultures, Cancer Res, 46, 89

Miller, 1986, Subpopulation interactions in drug resistance, metastasis and tumor progression, Neo-adjuvant chemotherapy, 69

Miller, 1985, Factors affecting growth and drug sensitivity of mouse mammary tumor lines in collagen gel cultures, Cancer Res, 45, 4200

Miller, 1988, Dominance of a tumor subpopulation line in mixed heterogeneous mouse mammary tumors, Cancer Res, 48, 5747

Miller, 1990, Cellular interactions in metastasis, Cancer Metastasis Rev, 9, 21, 10.1007/BF00047586

Somarelli, 2016, Distinct routes to metastasis: plasticity-dependent and plasticity-independent pathways, Oncogene, 35, 4302, 10.1038/onc.2015.497

Ishay-Ronen, 2019, Gain fat-lose metastasis: converting invasive breast cancer cells into adipocytes inhibits cancer metastasis, Cancer Cell, 35, 17, 10.1016/j.ccell.2018.12.002

Acharyya, 2015, Invasion and Metastasis, The Molecular Basis of Cancer, 269, 10.1016/B978-1-4557-4066-6.00018-4

Boral, 2017, Molecular characterization of breast cancer CTCs associated with brain metastasis, Nat Commun, 8, 196, 10.1038/s41467-017-00196-1

Robinson, 2017, Integrative clinical genomics of metastatic cancer, Nature, 548, 297, 10.1038/nature23306

Naxerova, 2017, Origins of lymphatic and distant metastases in human colorectal cancer, Science, 357, 55, 10.1126/science.aai8515

Naxerova, 2015, Using tumour phylogenetics to identify the roots of metastasis in humans, Nat Rev Clin Oncol, 12, 258, 10.1038/nrclinonc.2014.238

Lytle, 2018, Stem cell fate in cancer growth, progression and therapy resistance, Nat Rev Cancer, 18, 669, 10.1038/s41568-018-0056-x

Welch, 1983, Heterogeneous response and clonal drift of sensitivities of metastatic 13762NF mammary adenocarcinoma clones to gamma-radiation in vitro, Cancer Res, 43, 6

Welch, 2017, Beyond the primary tumor: progression, invasion, and metastasis, The molecular basis of human cancer, 203, 10.1007/978-1-59745-458-2_12