Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Nov 25.
Published in final edited form as: Cell Cycle. 2006 Aug 15;5(16):1740–1743. doi: 10.4161/cc.5.16.3165

The Problem of Cancer Dormancy

Understanding the Basic Mechanisms and Identifying Therapeutic Opportunities

Julio A Aguirre-Ghiso 1
PMCID: PMC2587296  NIHMSID: NIHMS53909  PMID: 16929164

Abstract

The hiatus observed in the progression of cancer after diagnosis and treatment in a large proportion of patients has led to the notion that a state of cancer dormancy must exist during tumor progression. However, research on this stage of cancer has been limited due to the lack of appropriate models and clinical correlates. Fortunately, the last decade has seen the development of new cancer dormancy models, whole animal and intravital imaging techniques and the molecular characterization of minimal residual disease. These studies enabled researchers to reveal intriguing mechanisms and molecular determinants that define tumor dormancy. It is imperative to understand the basic mechanisms of dormancy, as this will accelerate the development of new markers of progression and novel therapeutic opportunities to induce dormancy and/or eradicate dormant disease. This issue of Cell Cycle includes a “Spotlight on Cancer Dormancy” highlighting major contributions to the field of cancer dormancy from basic and clinical studies. We anticipate that this will initiate a forum of discussion on the problem of cancer dormancy and stimulate investigators to study this rather unexplored but undeniably relevant clinical stage of cancer progression.

Keywords: quiescence, angiogenesis, metastasis, stem cells, signal transduction

A STAGE OF DORMANCY IN CANCER PROGRESSION

It is an indisputable clinical fact that cancer cells, which disseminate from the primary tumor before its removal, can persist in a dormant state for a very long time, before initiating overt metastases.1,2 Because in the past the existence of such cells could only be deduced, and not actually proven, it was impossible to attribute a specific mechanism to the phenomenon of dormancy.2-5 Currently, genetic analyses of the primary tumors can predict with relative certainty whether spread has occurred.6,7 Moreover, with the advent of new techniques of detection, isolation and characterization of very small number of cells from patients,8 as well as generation of experimental models of dormancy9-11 (reviewd in this issue, refs. 12-20), new insights are beginning to emerge. It appears that dormant cancer cells can persist by completely withdrawing from the cell cycle, or by continuing to slowly cycle and die at an equivalent rate. It is believed that the former is the result of loss of pro-proliferative signals while the latter is due to an inability to mount an angiogenic response.11,21-23 Also, it appears that the disseminated cells that don’t resume proliferation but persist, even in face of chemotherapy,24 might have developed adaptive mechanisms to survive under stress.25 Some of these properties resemble those of stem cells.26

It is my opinion that identifying mechanisms that keep disseminated cells in a persistent dormant state will allow interventions that might prevent their reemergence as overt metastases, while overcoming their survival response to stress might lead to their elimination. Both scenarios would be of benefit to cancer patients.

In this introductory article I provide a focused synopsis of the salient findings described in the reviews in the “Spotlight on Cancer Dormancy” in this issue of Cell Cycle, and put them in context of the important, and mostly unanswered questions in the field.

CLUES ABOUT DORMANCY FROM THE CLINIC AND MOUSE MODELS: IS THERE A ROLE FOR TUMOR STEM CELLS?

Three reviews in this issue (Marches et al.,13 Laufs et al.16 and Klein and Hötzel18) provide tantalizing information from minimal residual disease (MRD) studies regarding how dormancy may manifest itself in the clinic.1-3,8,27 Klein and Hötzel18 suggest that almost all tumors undergo what appears to be an obligatory dormancy stage that can last in some cases ~5 years. They show that very few genetic alterations are found in a large proportion of early disseminated tumor cells suggesting that either genetic alterations not detected by CGH are taking place or that the lack of growth of these initial departing tumor cells may be driven initially more by epigenetic than genetic alterations. Laufs et al.,16 describe the use of staining for the urokinase receptor (uPAR) along with cytokeratin-18 in bone marrow disseminated tumor cells as a way to stage cancer patients. Their findings showing that uPAR staining is an accurate predictor of poor prognosis and recurrence in gastric cancer28,29 correlated well with mechanistic studies from the Ossowski lab (see Ranganathan et al. and Laufs et al. refs. 16 and 19) where it was found that uPAR overexpression and signaling21,30 was important to prevent dormancy of malignant carcinoma cells.31 Thus, these studies1,28 support further exploration of strategies such as humanized monoclonal antibodies or small molecules that may restrict disease recurrence by blocking uPAR function.

Marches et al.,13 describe the detection of circulating tumor cells (CTCs) in blood >20 years after patients have been deemed cured.27 They hypothesize that the existence of tumor stem cells may explain this sub-clinical condition of cancer. They propose that rather than the existence of a tumor stem cell population remaining alive in circulation it is possible that tumor stem cells that have disseminated and formed micrometastases might shed cells into circulation, which may explain their findings. This hypothesis is in agreement with the studies reviewed by Felsher20 on Myc inactivation in a transgenic liver tumor mouse model.26,32 It is proposed that two possibilities might explain the transition between tumor cell growth and quiescence due to Myc inactivation: either Myc transformation occurred in a liver stem cell that upon oncogene inactivation differentiated into a normal appearing and functioning cell, but that still retains stem cell properties that allow them to resume growth after Myc-reactivation or, alternatively Myc may activate a stem cell program in cells that had already committed to a specific lineage.26,32 It is possible that many of the questions regarding tumor cell dormancy will be answered if it can be demonstrated that disseminated tumor stem cells can regain quiescence. Given that the primary tumor may generate communal signals that “recruit” the tumor stem cells, it is possible that upon dissemination and lodging in distant organs tumor stem cells that are unable to reproduce their expansion niche may enter a program of dormancy, a natural state for normal stem cells.26,33,34 Alternatively, the bone marrow, where disseminated tumor cells can be found, may serve as a survival reservoir for tumor stem cells. Thus, it will be important to determine the functional relationship between dormant tumor cells and their surrounding bone marrow or target organ stroma (e.g., extracellular matrix, fibroblasts, inflammatory cells).2,35 Further, the possibility that a tumor stem cell that carries mutations that can give origin to a primary tumor would reprogram into a low proliferation or growth arrest mode, questions the dominance of oncogenic signaling over cellular programs that can induce quiescence.

An interesting possibility is that tumor cell dormancy may represent an adaptive response to a non-propitious microenvironment.9 Tumor cells that are not fit to survive the dissemination may be rapidly cleared by apoptosis.36 Tumor cells that have all the tools to resume growth will do so.37 A third scenario is a phenotype where the genetic and/or epigenetic alterations in tumor cells do not favor immediate growth but give a selective advantage through the adaptation to stress (e.g., hypoxia, hyperoxia, low nutrient availability, chemotherapy).9,25 This phenotype could be dependent on programs described by Ranganathan et al.,19 where pathways that allow cells adapt to stress (e.g., p38SAPK, endoplasmic reticulum stress) would halt tumor cell growth until the conditions are once more favorable.25 This behavior could similarly apply to a model of tumor stem cell dormancy, as stem cells are known to withdraw from cell cycle and to resist drug treatments due to the expression of MDR pumps.26,33,34 Progress made in the tumor stem cell research field will very likely provide insight into these possibilities.

Most of the mouse models of cancer attempt recapitulating carcinogenesis and primary tumor development. Comparatively, much less effort has been dedicated to model the progression of MRD and metastasis development and these models rarely include chemotherapy. Only recently some mouse models have taken advantage of inducible expression systems for oncogenes such as Myc and Her2/neu or malignancy promoting factors such as TGFβ.38-40 While some of these studies have addressed the problem of metastasis they remain mostly focused on primary tumor development. Nevertheless, such transgenic mouse systems may be useful to model the progression of the disease including surgery of the primary tumor, chemotherapeutic treatment, MRD, dormancy and recurrence of disseminated disease.

TWO POTENTIAL MECHANISMS FOR TUMOR DORMANCY: WHAT CAN WE LEARN FROM THEM?

The general consensus that emerges from these reviews is that two stages might explain tumor dormancy: one that involves the induction of growth arrest and prolonged survival of disseminated single or small groups of tumor cells21,41 and a second that occurs in a small tumor mass where a constant balance between apoptosis and proliferation keeps the lesion small and undiagnosed.42,43 As these are general outputs of tumor cell behavior it is anticipated, and reflected in the reviews presented here, that different mechanisms might regulate these outcomes.

The existence of cancer dormancy due to a growth arrest of tumor cells is supported by evidence that within tissues where primary tumors are developing (see White et al. and Ranganathan et al., refs. 15 and 19, this issue) or that harbor disseminated cells (Townson and Chambers12 and Ranganathan et al.,19 this issue) and have a functional vasculature, tumor cells are found to be in a nonproliferative mode. Dormant tumor cells have been found to be in a G0/G1 arrest which was linked to reduced ERK mitogenic signaling,21,31 and to be associated with negative staining for proliferation markers (e.g., PCNA or Ki67).1,22 In models of breast and head and neck cancer it appears that a failure of tumor cells to coordinate integrin (e.g., α5β1.β1) and growth factor signaling (e.g., EGFR, Her2/neu) may result in the deactivation of mitogenic signaling21,39,44,45 and activation of p38SAPK stress pathways,46 that can lead to a state of tumor dormancy.

Naumov et al.17 and Indraccolo et al.14 describe how a failure to activate the angiogenic “switch” can maintain the tumor cell population in a constant balance between apoptosis and proliferation23,47 and cause dormancy. Indraccolo et al.14 provide novel insight as to the requirement for high levels of angiogenic factors to interrupt dormancy and subsequently low or moderate levels to maintain a functional vasculature in the now growing tumors. Marches et al.13 describe how in a model of B-cell lymphoma, immunizing toward the BCL1 Ig idiotype can induce tumor dormancy.11 Although in this model there is a balance between proliferation and cell death, it does not seem to depend on the angiogenic capacity of the tumor cells but on B-cell receptor signaling and upregulation of p21CIP/WAF1.11,48 It will be interesting to learn how a tumor cell population that achieves a balance of apoptosis and proliferation for years and accumulates genetic modifications can avoid undergoing an angiogenic switch and escape dormancy and whether VEGFR and PDGFR inhibitors targeting the endothelium and pericytes, respectively,49 can induce and maintain dormancy. Similarly it will be interesting to learn how this balance and the angiogenic switch is regulated in tumor stem cells, given that they could potentially enter a growth arrest and remain quiescent.26,32-34 As highlighted by Marches et al., the precision of the events controlling the balance between proliferation and death seems extraordinary. Therefore, it is important to understand its underlying operation to be able to activate cell death pathways, which would allow eradicating the residual disease.27

The notion of reprogramming malignant cells is not new.20,26 Still, the existence of tumor dormancy, which could be defined as a reprogramming towards normalcy, challenges some well-established concepts in cancer progression. For example how is it possible that tumor cells that have accumulated genetic and epigenetic alterations that led to the formation of a primary tumor37 somehow revert to a growth arrest mode after dissemination? This implies that common genetic alterations thought to be dominant (e.g., Ras mutations, ErbB2 amplification) may be overridden by signals that force the cells into a quiescent state. If this is true it suggests that the ability of tumor cells to fulfill the hallmarks of cancer37 may be context dependent. This is an exciting possibility as it suggests that there is still plasticity in these malignant cells to convert them into a dormant phenotype.

POTENTIAL THERAPEUTIC APPROACHES

Understanding the mechanisms that induce dormancy and drug resistance of dormant cells would allow generating at least two strategies to improve treatment. Therapies aimed at inducing dormancy may help overcome conventional drug resistance that is based on the ability of drugs to induce cell killing. Thus, reprogramming malignant cells into a growth arrest would allow converting the disease that would be otherwise untreatable, into a chronic asymptomatic condition. The combination of mitogenic signaling inhibitors (e.g., Mek, EGFR, Met, IGFR inhibitors)50-52 with activators of stress pathways (e.g., p38, JNK) may induce reprogramming of tumor cells into dormancy (see Ranganathan et al., ref. 19). A second option, which is highlighted in the reviews presented here, envisions inducing killing of dormant tumor cells during the dormancy stage24,25 (see Townson and Chambers, Marches et al., and Ranganathan et al., refs. 12, 13 and19). However, in order to conclusively determine whether a patient has dormant disseminated disease, new markers might have to be developed (see also Marches et al., ref. 13). Studies in the MRD field have identified several markers to achieve this goal.1 However, most of these markers inform about the recurrence of the disease (e.g., uPAR, ErbB2)1,28,29 rather than the state of tumor cell dormancy. It is possible that ongoing research in experimental models combined with the molecular characterization of CTCs or tumor cells in bone marrow will very likely reveal markers of dormancy. To achieve this goal it will be important to enhance collaborations between basic research labs and clinical labs to test the markers of dormancy identified in experimental models in samples from bone marrow, lymph nodes and CTCs. These will be important for staging of cancer and for developing novel targets for therapies.

The data covered by these reviews describe new exciting and provocative hypotheses and in depth data to support them. Collectively they represent major advances in our understanding of the problems associated with cancer dormancy.

Acknowledgments

This work is supported by a grant from the Samuel Waxman Cancer Research Foundation Tumor Dormancy Program and the NIH/National Cancer Institute grant CA109182 (to J.A. Aguirre-Ghiso). I would like to thank the members of my laboratory and Liliana Ossowski (Mount Sinai School of Medicine, NY) for helpful comments and stimulating discussions.

References

  • 1.Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer. 2004;4:448–56. doi: 10.1038/nrc1370. [DOI] [PubMed] [Google Scholar]
  • 2.Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M, Ahr A, Blankenstein TJ, Kaufmann M, Diebold J, Arnholdt H, Muller P, Bischoff J, Harich D, Schlimok G, Riethmuller G, Eils R, Klein CA. From latent disseminated cells to overt metastasis: Genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci USA. 2003;100:7737–42. doi: 10.1073/pnas.1331931100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Riethmuller G, Klein CA. Early cancer cell dissemination and late metastatic relapse: Clinical reflections and biological approaches to the dormancy problem in patients. Semin Cancer Biol. 2001;11:307–11. doi: 10.1006/scbi.2001.0386. [DOI] [PubMed] [Google Scholar]
  • 4.Demicheli R. Tumour dormancy: Findings and hypotheses from clinical research on breast cancer. Semin Cancer Biol. 2001;11:297–306. doi: 10.1006/scbi.2001.0385. [DOI] [PubMed] [Google Scholar]
  • 5.Demicheli R, Miceli R, Moliterni A, Zambetti M, Hrushesky WJ, Retsky MW, Valagussa P, Bonadonna G. Breast cancer recurrence dynamics following adjuvant CMF is consistent with tumor dormancy and mastectomy-driven acceleration of the metastatic process. Ann Oncol. 2005;16:1449–57. doi: 10.1093/annonc/mdi280. [DOI] [PubMed] [Google Scholar]
  • 6.Roepman P, Wessels LF, Kettelarij N, Kemmeren P, Miles AJ, Lijnzaad P, Tilanus MG, Koole R, Hordijk GJ, van der Vliet PC, Reinders MJ, Slootweg PJ, Holstege FC. An expression profile for diagnosis of lymph node metastases from primary head and neck squamous cell carcinomas. Nat Genet. 2005;37:182–6. doi: 10.1038/ng1502. [DOI] [PubMed] [Google Scholar]
  • 7.Akervall J. Gene profiling in squamous cell carcinoma of the head and neck. Cancer Metastasis Rev. 2005;24:87–94. doi: 10.1007/s10555-005-5049-z. [DOI] [PubMed] [Google Scholar]
  • 8.Schardt JA, Meyer M, Hartmann CH, Schubert F, Schmidt-Kittler O, Fuhrmann C, Polzer B, Petronio M, Eils R, Klein CA. Genomic analysis of single cytokeratin-positive cells from bone marrow reveals early mutational events in breast cancer. Cancer Cell. 2005;8:227–39. doi: 10.1016/j.ccr.2005.08.003. [DOI] [PubMed] [Google Scholar]
  • 9.Aguirre-Ghiso JA, Ossowski L, Rosenbaum SK. Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res. 2004;64:7336–45. doi: 10.1158/0008-5472.CAN-04-0113. [DOI] [PubMed] [Google Scholar]
  • 10.Chambers AF, Naumov GN, Varghese HJ, Nadkarni KV, MacDonald IC, Groom AC. Critical steps in hematogenous metastasis: An overview. Surg Oncol Clin N Am. 2001;10:243–55. vii. [PubMed] [Google Scholar]
  • 11.Uhr JW, Marches R. Dormancy in a model of murine B cell lymphoma. Semin Cancer Biol. 2001;11:277–83. doi: 10.1006/scbi.2001.0383. [DOI] [PubMed] [Google Scholar]
  • 12.Townson JL, Chambers AF. Dormancy of solitary metastatic cells. Cell Cycle. 2006 doi: 10.4161/cc.5.16.2864. In Press. [DOI] [PubMed] [Google Scholar]
  • 13.Marches R, Scheuermann RH, Uhr J. Cancer dormancy from mice to man: A review. Cell Cycle. 2006 doi: 10.4161/cc.5.16.2995. In Press. [DOI] [PubMed] [Google Scholar]
  • 14.Indraccolo S, Favaro E, Amadori A. Dormant tumors awaken by a short-term angiogenic burst: The spike hypothesis. Cell Cycle. 2006 doi: 10.4161/cc.5.16.2985. In Press. [DOI] [PubMed] [Google Scholar]
  • 15.White DE, Rayment JH, Muller W. Addressing the role of cell adhesion in tumor cell dormancy. Cell Cycle. 2006 doi: 10.4161/cc.5.16.2993. In Press. [DOI] [PubMed] [Google Scholar]
  • 16.Laufs S, Schumacher J, Allgayer H. Urokinase-receptor (u-PAR)-an essential player in multiple games of cancer: A review on its role in tumor progression, invasion, metastasis, proliferation/dormancy, clinical outcome and minimal residual disease. Cell Cycle. 2006 doi: 10.4161/cc.5.16.2994. In Press. [DOI] [PubMed] [Google Scholar]
  • 17.Naumov GN, Akslen LA, Folkman J. Role of angiogenesis in human tumor dormancy: Animal models of the angiogenic switch. Cell Cycle. 2006 doi: 10.4161/cc.5.16.3018. In Press. [DOI] [PubMed] [Google Scholar]
  • 18.Klein CA, Hölzel D. Systemic cancer progression and tumor dormancy: Mathematical models meet single cell genomics. Cell Cycle. 2006 doi: 10.4161/cc.5.16.3097. In Press. [DOI] [PubMed] [Google Scholar]
  • 19.Ranganathan AC, Adam AP, Aguirre-Ghiso JA. Opposing roles of mitogenic and stress signaling pathways in the induction of cancer dormancy. Cell Cycle. 2006 doi: 10.4161/cc.5.16.3109. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Felsher DW. Tumor dormancy: Death and resurrection of cancer as seen through transgenic mouse models. Cell Cycle. 2006 doi: 10.4161/cc.5.16.3111. In Press. [DOI] [PubMed] [Google Scholar]
  • 21.Aguirre-Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by downregulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol. 1999;147:89–104. doi: 10.1083/jcb.147.1.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Naumov GN, MacDonald IC, Weinmeister PM, Kerkvliet N, Nadkarni KV, Wilson SM, Morris VL, Groom AC, Chambers AF. Persistence of solitary mammary carcinoma cells in a secondary site: A possible contributor to dormancy. Cancer Res. 2002;62:2162–8. [PubMed] [Google Scholar]
  • 23.Naumov GN, Bender E, Zurakowski D, Kang SY, Sampson D, Flynn E, Watnick RS, Straume O, Akslen LA, Folkman J, Almog N. A model of human tumor dormancy: An angiogenic switch from the nonangiogenic phenotype. J Natl Cancer Inst. 2006;98:316–25. doi: 10.1093/jnci/djj068. [DOI] [PubMed] [Google Scholar]
  • 24.Naumov GN, Townson JL, MacDonald IC, Wilson SM, Bramwell VH, Groom AC, Chambers AF. Ineffectiveness of doxorubicin treatment on solitary dormant mammary carcinoma cells or late-developing metastases. Breast Cancer Res Treat. 2003;82:199–206. doi: 10.1023/B:BREA.0000004377.12288.3c. [DOI] [PubMed] [Google Scholar]
  • 25.Ranganathan AC, Zhang L, Adam AP, Aguirre-Ghiso JA. Functional coupling of p38-induced upregulation of BiP and activation of RNA-dependent protein kinase-like endoplasmic reticulum kinase to drug resistance of dormant carcinoma cells. Cancer Res. 2006;66:1702–11. doi: 10.1158/0008-5472.CAN-05-3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Shachaf CM, Felsher DW. Tumor dormancy and MYC inactivation: Pushing cancer to the brink of normalcy. Cancer Res. 2005;65:4471–4. doi: 10.1158/0008-5472.CAN-05-1172. [DOI] [PubMed] [Google Scholar]
  • 27.Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, Beitsch PD, Leitch M, Hoover S, Euhus D, Haley B, Morrison L, Fleming TP, Herlyn D, Terstappen LW, Fehm T, Tucker TF, Lane N, Wang J, Uhr JW. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10:8152–62. doi: 10.1158/1078-0432.CCR-04-1110. [DOI] [PubMed] [Google Scholar]
  • 28.Allgayer H, Heiss MM, Riesenberg R, Grutzner KU, Tarabichi A, Babic R, Schildberg FW. Urokinase plasminogen activator receptor (uPA-R): One potential characteristic of metastatic phenotypes in minimal residual tumor disease. Cancer Res. 1997;57:1394–9. [PubMed] [Google Scholar]
  • 29.Heiss MM, Allgayer H, Gruetzner KU, Funke I, Babic R, Jauch KW, Schildberg FW. Individual development and uPA-receptor expression of disseminated tumour cells in bone marrow: A reference to early systemic disease in solid cancer. Nat Med. 1995;1:1035–9. doi: 10.1038/nm1095-1035. [DOI] [PubMed] [Google Scholar]
  • 30.Yu W, Kim J, Ossowski L. Reduction in surface urokinase receptor forces malignant cells into a protracted state of dormancy. J Cell Biol. 1997;137:767–77. doi: 10.1083/jcb.137.3.767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Aguirre-Ghiso JA, Estrada Y, Liu D, Ossowski L. ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK) Cancer Res. 2003;63:1684–95. [PubMed] [Google Scholar]
  • 32.Shachaf CM, Kopelman AM, Arvanitis C, Karlsson A, Beer S, Mandl S, Bachmann MH, Borowsky AD, Ruebner B, Cardiff RD, Yang Q, Bishop JM, Contag CH, Felsher DW. MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature. 2004;431:1112–7. doi: 10.1038/nature03043. [DOI] [PubMed] [Google Scholar]
  • 33.Salm SN, Burger PE, Coetzee S, Goto K, Moscatelli D, Wilson EL. TGF-{beta} maintains dormancy of prostatic stem cells in the proximal region of ducts. J Cell Biol. 2005;170:81–90. doi: 10.1083/jcb.200412015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Talpaz M, Estrov Z, Kantarjian H, Ku S, Foteh A, Kurzrock R. Persistence of dormant leukemic progenitors during interferon-induced remission in chronic myelogenous leukemia. Analysis by polymerase chain reaction of individual colonies. J Clin Invest. 1994;94:1383–9. doi: 10.1172/JCI117473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pantel K, Otte M. Occult micrometastasis: Enrichment, identification and characterization of single disseminated tumour cells. Semin Cancer Biol. 2001;11:327–37. doi: 10.1006/scbi.2001.0388. [DOI] [PubMed] [Google Scholar]
  • 36.Wong CW, Lee A, Shientag L, Yu J, Dong Y, Kao G, Al-Mehdi AB, Bernhard EJ, Muschel RJ. Apoptosis: An early event in metastatic inefficiency. Cancer Res. 2001;61:333–8. [PubMed] [Google Scholar]
  • 37.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 38.Muraoka-Cook RS, Kurokawa H, Koh Y, Forbes JT, Roebuck LR, Barcellos-Hoff MH, Moody SE, Chodosh LA, Arteaga CL. Conditional overexpression of active transforming growth factor beta1 in vivo accelerates metastases of transgenic mammary tumors. Cancer Res. 2004;64:9002–11. doi: 10.1158/0008-5472.CAN-04-2111. [DOI] [PubMed] [Google Scholar]
  • 39.White DE, Kurpios NA, Zuo D, Hassell JA, Blaess S, Mueller U, Muller WJ. Targeted disruption of beta1-integrin in a transgenic mouse model of human breast cancer reveals an essential role in mammary tumor induction. Cancer Cell. 2004;6:159–70. doi: 10.1016/j.ccr.2004.06.025. [DOI] [PubMed] [Google Scholar]
  • 40.Tilli MT, Furth PA. Conditional mouse models demonstrate oncogene-dependent differences in tumor maintenance and recurrence. Breast Cancer Res. 2003;5:202–5. doi: 10.1186/bcr614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Naumov GN, MacDonald IC, Chambers AF, Groom AC. Solitary cancer cells as a possible source of tumour dormancy? Semin Cancer Biol. 2001;11:271–6. doi: 10.1006/scbi.2001.0382. [DOI] [PubMed] [Google Scholar]
  • 42.Folkman J. Role of angiogenesis in tumor growth and metastasis. Semin Oncol. 2002;29:15–8. doi: 10.1053/sonc.2002.37263. [DOI] [PubMed] [Google Scholar]
  • 43.Folkman J, Hanahan D. Switch to the angiogenic phenotype during tumorigenesis. Princess Takamatsu Symp. 1991;22:339–47. [PubMed] [Google Scholar]
  • 44.Weaver VM, Petersen OW, Wang F, Larabell CA, Briand P, Damsky C, Bissell MJ. Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies. J Cell Biol. 1997;137:231–45. doi: 10.1083/jcb.137.1.231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Liu D, Aguirre-Ghiso J, Estrada Y, Ossowski L. EGFR is a transducer of the urokinase receptor initiated signal that is required for in vivo growth of a human carcinoma. Cancer Cell. 2002;1:445–57. doi: 10.1016/s1535-6108(02)00072-7. [DOI] [PubMed] [Google Scholar]
  • 46.Aguirre-Ghiso JA, Liu D, Mignatti A, Kovalski K, Ossowski L. Urokinase receptor and fibronectin regulate the ERK(MAPK) to p38(MAPK) activity ratios that determine carcinoma cell proliferation or dormancy in vivo. Mol Biol Cell. 2001;12:863–79. doi: 10.1091/mbc.12.4.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Watnick RS, Cheng YN, Rangarajan A, Ince TA, Weinberg RA. Ras modulates Myc activity to repress thrombospondin-1 expression and increase tumor angiogenesis. Cancer Cell. 2003;3:219–31. doi: 10.1016/s1535-6108(03)00030-8. [DOI] [PubMed] [Google Scholar]
  • 48.Marches R, Hsueh R, Uhr JW. Cancer dormancy and cell signaling: Induction of p21(waf1) initiated by membrane IgM engagement increases survival of B lymphoma cells. Proc Natl Acad Sci USA. 1999;96:8711–5. doi: 10.1073/pnas.96.15.8711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Erber R, Thurnher A, Katsen AD, Groth G, Kerger H, Hammes HP, Menger MD, Ullrich A, Vajkoczy P. Combined inhibition of VEGF and PDGF signaling enforces tumor vessel regression by interfering with pericyte-mediated endothelial cell survival mechanisms. Faseb J. 2004;18:338–40. doi: 10.1096/fj.03-0271fje. [DOI] [PubMed] [Google Scholar]
  • 50.Sebolt-Leopold JS, Dudley DT, Herrera R, Van Becelaere K, Wiland A, Gowan RC, Tecle H, Barrett SD, Bridges A, Przybranowski S, Leopold WR, Saltiel AR. Blockade of the MAP kinase pathway suppresses growth of colon tumors in vivo. Nat Med. 1999;5:810–6. doi: 10.1038/10533. [DOI] [PubMed] [Google Scholar]
  • 51.Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, Harris PL, Driscoll DR, Fidias P, Lynch TJ, Rabindran SK, McGinnis JP, Wissner A, Sharma SV, Isselbacher KJ, Settleman J, Haber DA. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci USA. 2005;102:7665–70. doi: 10.1073/pnas.0502860102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mendelsohn J. Targeting the epidermal growth factor receptor for cancer therapy. J Clin Oncol. 2002;20:1S–13S. [PubMed] [Google Scholar]

RESOURCES