Skip to main content
Gastrointestinal Cancer Research : GCR logoLink to Gastrointestinal Cancer Research : GCR
. 2008 Jul-Aug;2(4):169–174.

Cancer Stem Cells, Endothelial Progenitors, and Mesenchymal Stem Cells: “Seed and Soil” Theory Revisited

Edward H Lin 1,2,3, Yixing Jiang 1,2,3, Yanhong Deng 1,2,3, Ritu Lapsiwala 1,2,3, Tongyu Lin 1,2,3, C Anthony Blau 1,2,3
PMCID: PMC2632830  PMID: 19259284

Abstract

Isolation of putative cancer stem cells (CSCs) in various tumors has generated much excitement among researchers who consider these cells the potential “culprits” behind resistance to conventional therapy. Both cancer and cardiovascular disease are believed to be stem cell disorders involving circulating endothelial progenitors (CEPs) and mesenchymal stem cells (MSCs). CD133 and CD44, markers of CSCs in many tumors, also enrich CEPs and MSCs, respectively. We propose an integrated tumorigenesis model that involves all three interdependent stem cell (CSC, CEP, MSC) compartments by revisiting the “seed and soil” model. Developing therapeutics that can effectively target CSCs and spare normal cardiovascular tissue will remain a challenge. Preliminary laboratory and clinical data on monitoring and targeting colon CSCs, using such a modeling system, are discussed.


More than a century ago, Virchow and Paget indirectly implied the existence of cancer stem cells (CSCs)1 and a CSC niche,2 respectively. The term “cancer stem cells” was coined in the early 1980s,3 but research in the field was hampered by the lack of specific markers, cell-sorting technology, and the extremely low frequency of CSCs. The first isolation of putative CSCs in acute myelogenous leukemia (AML) by Dick and colleagues in 19944 spurred a wave of discoveries of CSCs in acute lymphocytic leukemia (ALL),5,6 chronic myelogenous leukemia (CML),7,8 multiple myeloma,9 and in cancers of the colon,1012 breast,13 prostate,1416 brain,17,18 head and neck,19 retina,20 lung,21,22 pancreas,23 melanoma,24,25 kidney,26 and liver.27,28

Researchers postulated that CSCs might be the “culprits” capable of evading conventional therapy; thus, eradication of CSCs could lead to complete cure.29,30 However, CSCs are quiescent or slow cycling,31,32 they overexpress antiapoptotic proteins,24,25,33 possess multidrug resistance proteins,24,25,34,35 and are characteristically similar to normal stem cells, except that their ability to differentiate is impaired.429

In 1997, Asahara et al first identified bone-marrow derived circulating endothelial progenitors (CEPs) vital in postnatal physiologic and pathologic angiogenesis.36 In the same year, Yin et al discovered CD133, a member of the prominin family that identifies and enriches CEPs.37 Two large prospective studies showed that decreased CEP levels independently predict increased cardiovascular death and correlate with all known cardiovascular risk factors.38,39 Conversely, elevated CEP levels were found in patients with leukemia,40 myeloma,41,42 myelodysplastic syndrome,43 cancers of the lung,44 breast,45,46 liver,47 colon and rectum,4850 prostate,49 kidney,26 and in infantile hemangioma.51 Furthermore, the presence of elevated levels of CEPs or CD133 mRNA is predictive of poor outcomes and death in certain tumors.44,4749,52

Circulating endothelial progenitors are known to form the premetastatic niche essential in the earliest step to tumor angiogenesis,53 and it has been shown that targeting CEPs inhibits the development of macrometastases from micrometastases in mouse lung metastasis models.54 Moreover, it has been demonstrated that bone marrow-derived stem cells can transdifferentiate into endothelial precursor cells with the potential to acquire mutations and contribute “cancerous” microvascular endothelial cells to tumor vessels.55 In 1997, Zohar and others identified mesenchymal stem cells (MSCs) using CD44, a glycoprotein ligand that binds with hyaluronate and E-selectin.56,57 Variant forms of CD44 are expressed in many human cancers and correlate with cancer outcomes in certain tumors.5867 Interestingly, CD133 and CD44 have both been used to isolate CSCs in many tumors.425 The purpose of this review is to integrate these complex, confusing data on cancer stem cells, circulating endothelial progenitors, and mesenchymal stem cells and to discuss strategies directed at these novel cellular targets by revisiting Paget’s “seed and soil” theory2 in the context of today’s understanding of CSCs, CEPs, and MSCs.

CANCER STEM CELLS: “THE SEED”

Normal stem cells are defined by two properties; (1) self renewal, and (2) the capacity to differentiate into a wide array of specialized cells in the body. To date, stem cells have been identified in hematopoietic tissue,68 lung,69 skin,70 and colon,71 and in less regenerative tissues, such as those of the heart,72 and central nervous system.73 Epithelial-mesenchymal transition (EMT) dictates the fate of morphologic and functional diversities among these tissues and are regulated by common signaling pathways, such as Wnt/β-catenin, Notch, and bone morphogenetic protein (BMP) for stem cell renewal, maintenance, activation, and differentiation.74

CSC Research

Cancer stem cell research has advanced considerably since Virchow first described the similarities between neoplasia and embryonic tissues in 1885.1 An elegant summary of CSC research authored by Huntly and Gilliland was published in 2005.75 Cancer stem cells are rare, with plating efficiency ranging from 1/1,000–5,000 in solid tumors,3 to 1/1,000,000 in leukemia.4 Identification of stem cell markers CD133 and CD44 and the use of non-obese diabetic mice with severe combined immunodeficiency disease (NOD/SCID mice) xenografts led to the successful isolation of CSCs in many tumors.824

Leukemia stem cells, characterized by CD34+ and CD38 low/-, comprised 0.1% of the total leukemia population and were capable of inducing all subtypes of AML observed in patients, except for the M3 subtype.4 BCR-ABL fusion gene can only be found in the leukemia initiating fraction of ALL and CML in blast crisis with similar phenotypes of CD34+/CD38-.68 Singh et al first isolated CD133+ CSCs from patients with glioblastoma.17,76 Later, two groups independently identified colon CSCs estimated at a frequency of one per 5.7x104 colon cancer cells.10,11 One out of 262 CD133-enriched tumor cells contains a colon cancer stem cell, and as few as 100–500 of these enriched cells with either CD133+ or CD44+ tumor cells could recapitulate human tumors in NOD-SCID mice.4,913,16,17 Tumor spheres generated from CD133+ cells contain self-renewal units that could be passaged in vitro over 1 year.11,13,14,76 G-protein coupled receptor GPR) 49, a new colon stem cell marker, may also identify colon CSCs.71 It is important to point out that Ricci-Vitiani reported 2% of CEP cells in the enriched CD133+ colon CSC fraction only using CD45 and CD31 staining. CD133 enriched hematopoietic stem cells express many “stemness” genes that are also mutated or abnormally expressed in leukemia.77

The CSC model is being challenged in a report by Kelly and Strauss, who showed that low efficiency tumor grafting may be due to poor grafting between humanmouse interface, as 1/10 mouse leukemia and lymphoma cells can establish xenograft. 78 Table 1 summarizes estimated CSC frequency, their surface markers, and major signaling pathways.

Table 1.

Stem cells and their respective markers and pathways

Sites Frequency in cancer Surface markers Target pathways
Colon 10–12 0.7–24.5% CD133, CD44, GRP49 Wnt, Sonic Hedgehog

Liver27,119,120 <2% CD133, G Akt/PKB, BCL-2

Pancreas23 0.2–0.8% CD44/CD24/ESA/CD133 Wnt?

Kidney26 0.9±0.17% CD133

Prostate14,15,121 1% CD133/CD44/α2β1,Sca-1,CXCR2 PTEN/AKT, SDF1/CXCR4

Melanoma24,25 <1% CD133/ABCG2 Notch4

Lung & Head/neck19,21 10% CD133 ?

Brain17,18,74,122 1–27.5% CD133 Notch

Myeloid (AML)46 0.2% CD34+/CD38− Fas, Wnt

Breast13 2–4% CD44+/CD24−/ESA+ Wnt, Oct4

Niche And Tumor Angiogenesis: Roles of MSCs and CEPs

The differences between normal stem cells and CSCs are also determined by their dependence on the stem cell niche, a specialized microenvironment where stem cells reside, proliferate, and differentiate.7981 The niche maintains stem cell homeostasis, serves as a shield from tumorigenesis, and provides regulatory signals for tissue regeneration.7981 Niche normally generates dominant signals that inhibit stem cell differentiation and restrict normal stem cells from overproliferation for self renewal and multilineage commitment.70 Disruption of the niche signal can lead to either loss of stem cells or cancer (Figure 1).81 One of the best studied models is the intestinal stem cell niche characterized by high turnover rate and homeostasis through intestinal crypts.79 However, stem cell and niche is a potent combination, and a single stem cell is able to regenerate an entire crypt or generate additional crypts through crypt fission following radiation damage8284 and can also convert into cancer initiation cells.84,85

Figure 1.

Figure 1

Normal stem cell homeostasis and revised “Seed and Soil” model involving CSCs, CEPs, and MSCs.

In contrast to the belief that endothelial cells of tumor vessels are considered normal diploid cells that do not acquire mutations, a recent study showed that 15% to 85% of microvascular endothelial cells in B-cell lymphomas harbored lymphoma-specific chromosomal translocations with numerically similar chromosomal aberrations, supporting the idea that change in the niche signal also contributes to tumorigenesis by providing stimulatory signals.55

Tumor angiogenesis, one of six essential hallmarks of cancer, is a dysregulated modeling process that involves branching of pre-existing vessels, as well as mobilization of CEPs as a result of tumor hypoxia and excess proangiogenic factors.8690 CEPs are recruited to form the premetastatic niche conducive to the earliest steps of tumor angiogenesis.53,54,90 Angiogenesis from CEPs differs from the branching out of pre-existing blood vessels in that CEPs possess higher proliferative capacity than activated endothelial cells capable of expanding a thousandfold in vitro.91 Compared to as high as 90%CEPs found in xenograft models, CEPs range from 1% to 12% in human cancers, depending on the surface markers.90 Given the proliferative and differentiation potential of CEPs, which lose CD133 expression, enumerating CEPs based on CD133 expression in established tumors may underestimate the true CEP contribution to tumor angiogenesis. Bone marrow derived CEPs are mobilized by cytokines secreted by tumors and directed by chemokines to sites of hypoxia and neovascularization.90 Stimulation of CEPs promotes tumor angiogenesis and lymphogenesis, while inhibition of CEPs retards tumor angiogenesis and tumor growth.54,90,92,93

As discussed previously, CEP levels measured by flow cytometry (or its marker CD133 mRNA) were elevated in a variety of cancers and were associated with poor clinical outcomes in cancers of the lung,44 breast,46 liver,47 and colon,52 and in solid tumor patients with bone metastases.49 It is estimated that 1,400–700,000 circulating tumor cells exist based on the observation that there are 2–1,000 circulating tumor cells per 7 mL of blood.94 If 1% of circulating colon cancer cells were CSCs, only 14–7,000 circulating CSCs would be present in a 70 kg patient (or ~ 5 liters of blood), supporting the clinical observation that most patients had only one metastasis to few metastases. Furthermore, any condition in which CSCs could commandeer and mobilize CEPs will also limit the number of metastases. Given its sensitivity, CD133 mRNA assay could have identified CEPs as well as CSCs if the latter are present in peripheral blood. We observed that elevated CD133 mRNA levels ≥ 4.79 predict colon cancer recurrence with an odds ratio of 22.2 (P = .02) compared to 17.2 for patients with stage III/IV vs. stage I/II disease (P = .01). Likewise, CEP levels were also found to correlate with stage in patients with invasive breast cancers46 and are emerging as a useful surrogate marker of response in a variety of solid tumors.44,4749,95

Standard CD44 isoform (CD44s) identifies and enriches CD34-, CD133- MSCs that can differentiate into all stroma components, including fibroblast, bone, and cartilage. 56,57,96 A variant form of CD44v is found in many epithelial cancers and is the ligand for E- and P-selection and hyaluronate. The prognosis values of CD44 and its soluble forms are controversial.5261,97 Recently, Liu and colleagues studied 186 gene expression profiles of CD44+/CD24-/low putative breast cancer stem cells to that of normal breast epithelium and found a significant association between the gene signature in both overall and metastasisfree survival (P < .001, for both) independent of established clinical and pathologic variables. The unique gene signatures could further stratify women with high-risk early breast cancer into good prognosis (10-year metastasis-free survival of 81%) and poor prognosis (10-year metastasis-free survival of 57%, P = .01) groups when combined with National Institutes of Health prognostic criteria. The gene signatures are also associated with prognosis in medulloblastoma (P = .004), lung cancer (P = .03), and prostate cancer (P = .01).98

Given the dynamic plasticity and interdependency among all three stem cell compartments (CSC, MSC, and CEP), we hypothesize that “cancer stemness” functions in a complex “seed and soil” organizational hierarchy (Figure 1). Karnoub et al showed that mixing low potency tumor with MSCs dramatically improved the xenograft potential.99 One alternative explanation to Kelly and Strauss’s findings would be that mouse leukemia and lymphoma progenitors are proficient in establishing their niche and angiogenesis given mouse origin and tumor types selected. Not all tumor types would need to fit in a CSC model, and a marked increase in tumor stem cell marker in the majority of tumor cells suggests that these are not necessarily stem cells but cells that had acquired stem cells or stem-cell-like characteristics.

TARGETING STEM CELLS IN CANCER THERAPY

Cancer stem cells possess many inherent in vitro resistance mechanisms;24,25,3335 however, most clinically relevant is the observation that demonstrated host-tumor interaction conributes to the evolution of in vivo resistance to chemotherapy.100 In vivo drug resistance occurs through EMT via MSC activation and angiogenesis through CEP mobilization by cytotoxic chemotherapy. 92,101,102 Gemcitabine stimulates CD44 expression in pancreatic cancer cell lines,103 whereas 5-fluorouracil (5-FU) or oxaliplatin leads to a sixteen- to thirtyfold increase in CD133 expression and a twofold increase in CD44 expression in colon cancer cell lines.104

In addition to normalization of tumor vasculature,87 bevacizumab, a VEGF inhibitor may also affect stem cell vascular niches and disrupt cancer stem cell maintenance, as mice bearing glioblastoma treated with bevacizumab showed depleted vasculature and a dramatic reduction in the number of glioblastoma stem cells without affecting their proliferation.105

Given that CD44 and CD133 are coexpressed in three stem cell compartments, targeting CD44,106 or CD133, though logical,107,108 may lead to potential toxicities in heart, renal tubule,109 and retina.110 Inhibitors of Wnt signaling,111,112 Sonic Hedgehog,113 and AKT28 appears promising in certain tumor models. Use of genetically modified MSCs as a vehicle for targeting CSCs remains experimental at this stage.114 Novel immunotherapy and nanotechnology targeting of CSCs is also being explored. Successful strategies targeting CSCs, however, would require an in-depth understanding of the biology of all three stem cell compartments: CSC, CEP, and MSC. Targeting cancer stem cells using conventional phase I dose escalation may not be relevant or applicable, and well-designed prospective clinical trials are needed to validate surrogate markers, such as CTCs and CEPs, for drug development.

Therapy directed at these stem cell compartments may lead to significant survival benefits in patients with metastatic colorectal cancer. The median overall survival (OS) for patients with metastatic colorectal cancer reached 20 months with chemotherapy.115 Though complete response is rare (4%) and not durable, median OS reached 44 months, comparable to those who underwent surgical resection of metastasis.116,117

Integrating multimodality therapy with long-term maintenance (XCEL) with capecitabine and celecoxib, a selective COX-2 inhibitor, we reported a 28% complete response rate in patients with unresectable metastatic colorectal cancer.118 Among 19 patients who achieved complete radiographic responses, we observed a paradoxic 3-year relapse-free survival of 70% for those who did not undergo resection or had R1-2 resections vs. 20% for those who had R0 resection.119 Median OS reached 73.3 months in this group of patients whose median survival was projected to be around 20–30 months, based on their clinical risk scores.

Our unpublished preclinical data confirmed findings that 5-FU enriches CD133 expression,104 and maintenance XCEL may effectively target colon CSCs. Furthermore, celecoxib can also inhibit CEP proliferation by inhibiting AKT and caspase 3 activation. 120 Celecoxib can reduce high-risk polyps by 50%, probably through Wnt signaling. 121,122 The cardiovascular toxicities of celecoxib and bevacizumab may be due to their disturbance in CEP function and levels.115,121 Interestingly, celecoxib failed to demonstrate any survival advantage when combined with irinotecan-based chemotherapy in patients with metastatic colorectal cancer.123

DISCUSSION

Cancer and cardiovascular disease are believed to be stem cell-related disorders, with opposing CEP levels. Tumorigenesis involves CSCs, CEPs, and MSCs, three distinct, interdependent stem cell compartments essential to the delopment of in vivo resistance to chemotherapy and radiation. Targeting CSC/CEP/MSC compartments may lead to tumor dormancy, a new paradigm in cancer therapy (Figure 1); however, it is important to avoid serious injury to cardiovascular and other normal tissue, given the striking similarities within all three stem cell compartments.38,39,124 The revised “Seed and Soil” theory invokes new levels of molecular and cellular complexity, which may help researchers to narrow the search for “ideal” targets or pathways within these cellular compartments.

Footnotes

Disclosures of Potential Conflicts of Interest

The authors indicated no potential conflicts of interest.

REFERENCES

  • 1.Virchow R.Editorial. Virchows Arch Pathol Anat Physiol Klin Med 3.1855
  • 2.Paget S. The distribution of secondary growths in cancer of the breast. Lancet. 1889;1:571–573. [PubMed] [Google Scholar]
  • 3.Carney DN, Gazdar AF, Bunn PA, Jr, et al. Demonstration of the stem cell nature of clonogenic tumor cells from lung cancer patients. Stem Cells. 1982;1:149–164. [PubMed] [Google Scholar]
  • 4.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
  • 5.Anastasi J, Feng J, Dickstein JI, et al. Lineage involvement by BCR/ABL in Ph+ lymphoblastic leukemias: chronic myelogenous leukemia presenting in lymphoid blast vs Ph+ acute lymphoblastic leukemia. Leukemia. 1996;10:795–802. [PubMed] [Google Scholar]
  • 6.Cobaleda C, Gutierrez-Cianca N, Perez-Losada J, et al. A primitive hematopoietic cell is the target for the leukemic transformation in human philadelphia-positive acute lymphoblastic leukemia. Blood. 2000;95:1007–1013. [PubMed] [Google Scholar]
  • 7.Eaves C, Udomsakdi C, Cashman J, et al. The biology of normal and neoplastic stem cells in CML. Leuk Lymphoma. 1993;11(suppl 1):245–253. doi: 10.3109/10428199309047894. [DOI] [PubMed] [Google Scholar]
  • 8.Jamieson CH, Ailles LE, Dylla SJ, et al. Granulocyte- macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004;351:657–667. doi: 10.1056/NEJMoa040258. [DOI] [PubMed] [Google Scholar]
  • 9.Matsui W, Huff CA, Wang Q, et al. Characterization of clonogenic multiple myeloma cells. Blood. 2004;103:2332–2336. doi: 10.1182/blood-2003-09-3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.O'Brien CA, Pollett A, Gallinger S, et al. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445:106–110. doi: 10.1038/nature05372. [DOI] [PubMed] [Google Scholar]
  • 11.Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification and expansion of human coloncancer-initiating cells. Nature. 2007;445:111–115. doi: 10.1038/nature05384. [DOI] [PubMed] [Google Scholar]
  • 12.Dalerba P, Dylla SJ, Park IK, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A. 2007;104:10158–10163. doi: 10.1073/pnas.0703478104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–3988. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gu G, Yuan J, Wills M, et al. Prostate cancer cells with stem cell characteristics reconstitute the original human tumor in vivo. Cancer Res. 2007;67:4807–4815. doi: 10.1158/0008-5472.CAN-06-4608. [DOI] [PubMed] [Google Scholar]
  • 15.Lawson DA, Xin L, Lukacs R, et al. Prostate stem cells and prostate cancer. Cold Spring Harb Symp Quant Biol. 2005;70:187–196. doi: 10.1101/sqb.2005.70.003. [DOI] [PubMed] [Google Scholar]
  • 16.Patrawala L, Calhoun T, Schneider-Broussard R, et al. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene. 2006;25:1696–1708. doi: 10.1038/sj.onc.1209327. [DOI] [PubMed] [Google Scholar]
  • 17.Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401. doi: 10.1038/nature03128. [DOI] [PubMed] [Google Scholar]
  • 18.Dirks PB. Cancer: stem cells and brain tumours. Nature. 2006;444:687–688. doi: 10.1038/444687a. [DOI] [PubMed] [Google Scholar]
  • 19.Prince ME, Sivanandan R, Kaczorowski A, et al. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci U S A. 2007;104:973–978. doi: 10.1073/pnas.0610117104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Seigel GM, Hackam AS, Ganguly A, et al. Human embryonic and neuronal stem cell markers in retinoblastoma. Mol Vis. 2007;13:823–832. [PMC free article] [PubMed] [Google Scholar]
  • 21.Hilbe W, Dirnhofer S, Oberwasserlechner F, et al. CD133 positive endothelial progenitor cells contribute to the tumour vasculature in nonsmall cell lung cancer. J Clin Pathol. 2004;57:965–969. doi: 10.1136/jcp.2004.016444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Donnenberg VS, Landreneau RJ, Donnenberg AD. Tumorigenic stem and progenitor cells: implications for the therapeutic index of anticancer agents. J Control Release. 2007;122:385–391. doi: 10.1016/j.jconrel.2007.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030–1037. doi: 10.1158/0008-5472.CAN-06-2030. [DOI] [PubMed] [Google Scholar]
  • 24.Monzani E, Facchetti F, Galmozzi E, et al. Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer. 2007;43:935–946. doi: 10.1016/j.ejca.2007.01.017. [DOI] [PubMed] [Google Scholar]
  • 25.Frank NY, Margaryan A, Huang Y, et al. ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res. 2005;65:4320–4333. doi: 10.1158/0008-5472.CAN-04-3327. [DOI] [PubMed] [Google Scholar]
  • 26.Bruno S, Bussolati B, Grange C, et al. CD133+ renal progenitor cells contribute to tumor angiogenesis. Am J Pathol. 2006;169:2223–2235. doi: 10.2353/ajpath.2006.060498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Suetsugu A, Nagaki M, Aoki H, et al. Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun. 2006;351:820–824. doi: 10.1016/j.bbrc.2006.10.128. [DOI] [PubMed] [Google Scholar]
  • 28.Ma S, Lee TK, Zheng BJ, et al. CD133(+) HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene. 2008;27:1749–1758. doi: 10.1038/sj.onc.1210811. [DOI] [PubMed] [Google Scholar]
  • 29.Reya T, Morrison SJ, Clarke MF, et al. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–111. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • 30.Clarke MF, Dick JE, Dirks PB, et al. Cancer stem cells—perspectives on current status and future directions: AACR Workshop on Cancer Stem Cells. Cancer Res. 2006;66:9339–9344. doi: 10.1158/0008-5472.CAN-06-3126. [DOI] [PubMed] [Google Scholar]
  • 31.Passegue E, Wagers AJ. Regulating quiescence: new insights into hematopoietic stem cell biology. Dev Cell. 2006;10:415–417. doi: 10.1016/j.devcel.2006.03.002. [DOI] [PubMed] [Google Scholar]
  • 32.Sharpless NE, DePinho RA. Telomeres, stem cells, senescence, and cancer. J Clin Invest. 2004;113:160–168. doi: 10.1172/JCI20761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ghods AJ, Irvin D, Liu G, et al. Spheres isolat- ed from 9L gliosarcoma rat cell line possess chemoresistant and aggressive cancer stemlike cells. Stem Cells. 2007;25:1645–1653. doi: 10.1634/stemcells.2006-0624. [DOI] [PubMed] [Google Scholar]
  • 34.Liu G, Yuan X, Zeng Z, et al. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 2006;5:67. doi: 10.1186/1476-4598-5-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lou H, Dean M. Targeted therapy for cancer stem cells: the patched pathway and ABC transporters. Oncogene. 2007;26:1357–1360. doi: 10.1038/sj.onc.1210200. [DOI] [PubMed] [Google Scholar]
  • 36.Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–967. doi: 10.1126/science.275.5302.964. [DOI] [PubMed] [Google Scholar]
  • 37.Yin AH, Miraglia S, Zanjani ED, et al. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 1997;90:5002–5012. [PubMed] [Google Scholar]
  • 38.Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348:593–600. doi: 10.1056/NEJMoa022287. [DOI] [PubMed] [Google Scholar]
  • 39.Werner N, Kosiol S, Schiegl T, et al. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005;353:999–1007. doi: 10.1056/NEJMoa043814. [DOI] [PubMed] [Google Scholar]
  • 40.Rigolin GM, Mauro E, Ciccone M, et al. Neoplastic circulating endothelial-like cells in patients with acute myeloid leukaemia. Eur J Haematol. 2007;78:365–373. doi: 10.1111/j.1600-0609.2007.00839.x. [DOI] [PubMed] [Google Scholar]
  • 41.Braunstein M, Ozcelik T, Bagislar S, et al. Endothelial progenitor cells display clonal restriction in multiple myeloma. BMC Cancer. 2006;6:161. doi: 10.1186/1471-2407-6-161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zhang H, Vakil V, Braunstein M, et al. Circulating endothelial progenitor cells in multiple myeloma: implications and significance. Blood. 2005;105:3286–3294. doi: 10.1182/blood-2004-06-2101. [DOI] [PubMed] [Google Scholar]
  • 43.Auberger J, Dlaska M, Auberger T, et al. Increased CD133 expression in bone marrow of myelodysplastic syndromes. Leuk Res. 2005;29:995–1001. doi: 10.1016/j.leukres.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • 44.Dome B, Timar J, Dobos J, et al. Identification and clinical significance of circulating endothelial progenitor cells in human non-small cell lung cancer. Cancer Res. 2006;66:7341–7347. doi: 10.1158/0008-5472.CAN-05-4654. [DOI] [PubMed] [Google Scholar]
  • 45.Furstenberger G, von Moos R, Senn HJ, et al. Real-time PCR of CD146 mRNA in peripheral blood enables the relative quantification of circulating endothelial cells and is an indicator of angiogenesis. Br J Cancer. 2005;93:793–798. doi: 10.1038/sj.bjc.6602782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Naik RP, Jin D, Chuang E, et al. Circulating endothelial progenitor cells correlate to stage in patients with invasive breast cancer. Breast Cancer Res Treat. 2008;107:133–138. doi: 10.1007/s10549-007-9519-6. [DOI] [PubMed] [Google Scholar]
  • 47.Ho JW, Pang RW, Lau C, et al. Significance of circulating endothelial progenitor cells in hepatocellular carcinoma. Hepatology. 2006;44:836–843. doi: 10.1002/hep.21353. [DOI] [PubMed] [Google Scholar]
  • 48.Lin EH, Hassan M, Li Y, et al. Elevated circulating endothelial progenitor marker CD133 messenger RNA levels predict colon cancer recurrence. Cancer. 2007;110:534–542. doi: 10.1002/cncr.22774. [DOI] [PubMed] [Google Scholar]
  • 49.Mehra N, Penning M, Maas J, et al. Progenitor marker CD133 mRNA is elevated in peripheral blood of cancer patients with bone metastases. Clin Cancer Res. 2006;12:4859–4866. doi: 10.1158/1078-0432.CCR-06-0422. [DOI] [PubMed] [Google Scholar]
  • 50.Willett CG, Boucher Y, Duda DG, et al. Surrogate markers for antiangiogenic therapy and dose-limiting toxicities for bevacizumab with radiation and chemotherapy: continued experience of a phase I trial in rectal cancer patients. J Clin Oncol. 2005;23:8136–8139. doi: 10.1200/JCO.2005.02.5635. [DOI] [PubMed] [Google Scholar]
  • 51.Yu Y, Flint AF, Mulliken JB, et al. Endothelial progenitor cells in infantile hemangioma. Blood. 2004;103:1373–1375. doi: 10.1182/blood-2003-08-2859. [DOI] [PubMed] [Google Scholar]
  • 52.Lindvall C, Bu W, Williams BO, et al. Wnt signaling, stem cells, and the cellular origin of breast cancer. Stem Cell Rev. 2007;3:157–168. doi: 10.1007/s12015-007-0025-3. [DOI] [PubMed] [Google Scholar]
  • 53.Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–827. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gao D, Nolan DJ, Mellick AS, et al. Endothelial progenitor cells control the angiogenic switch in mouse lung metastasis. Science. 2008;319:195–198. doi: 10.1126/science.1150224. [DOI] [PubMed] [Google Scholar]
  • 55.Streubel B, Chott A, Huber D, et al. Lymphoma- specific genetic aberrations in microvascular endothelial cells in B-cell lymphomas. N Engl J Med. 2004;351:250–259. doi: 10.1056/NEJMoa033153. [DOI] [PubMed] [Google Scholar]
  • 56.Reyes M, Lund T, Lenvik T, et al. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood. 2001;98:2615–2625. doi: 10.1182/blood.v98.9.2615. [DOI] [PubMed] [Google Scholar]
  • 57.Zohar R, Sodek J, McCulloch CA. Characterization of stromal progenitor cells enriched by flow cytometry. Blood. 1997;90:3471–3481. [PubMed] [Google Scholar]
  • 58.Yamaguchi A, Urano T, Goi T, et al. Expression of a CD44 variant containing exons 8 to 10 is a useful independent factor for the prediction of prognosis in colorectal cancer patients. J Clin Oncol. 1996;14:1122–1127. doi: 10.1200/JCO.1996.14.4.1122. [DOI] [PubMed] [Google Scholar]
  • 59.Gansauge F, Gansauge S, Rau B, et al. Low serum levels of soluble CD44 variant 6 are significantly associated with poor prognosis in patients with pancreatic carcinoma. Cancer. 1997;80:1733–1739. doi: 10.1002/(sici)1097-0142(19971101)80:9<1733::aid-cncr6>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 60.Jung K, Lein M, Weiss S, et al. Soluble CD44 molecules in serum of patients with prostate cancer and benign prostatic hyperplasia. Eur J Cancer. 1996;32A:627–630. doi: 10.1016/0959-8049(95)00634-6. [DOI] [PubMed] [Google Scholar]
  • 61.Franzmann EJ, Reategui EP, Carraway KL, et al. Salivary soluble CD44: a potential molecular marker for head and neck cancer. Cancer Epidemiol Biomarkers Prev. 2005;14:735–739. doi: 10.1158/1055-9965.EPI-04-0546. [DOI] [PubMed] [Google Scholar]
  • 62.Mulder JW, Kruyt PM, Sewnath M, et al. Colorectal cancer prognosis and expression of exon-v6-containing CD44 proteins. Lancet. 1994;344:1470–1472. doi: 10.1016/s0140-6736(94)90290-9. [DOI] [PubMed] [Google Scholar]
  • 63.Saito H, Tsujitani S, Katano K, et al. Serum concentration of CD44 variant 6 and its relation to prognosis in patients with gastric carcinoma. Cancer. 1998;83:1094–1101. [PubMed] [Google Scholar]
  • 64.Niitsu N, Iijima K. High serum soluble CD44 is correlated with a poor outcome of aggressive non-Hodgkin's lymphoma. Leuk Res. 2002;26:241–248. doi: 10.1016/s0145-2126(01)00122-9. [DOI] [PubMed] [Google Scholar]
  • 65.De Rossi G, Marroni P, Paganuzzi M, et al. Increased serum levels of soluble CD44 standard, but not of variant isoforms v5 and v6, in B cell chronic lymphocytic leukemia. Leukemia. 1997;11:134–141. doi: 10.1038/sj.leu.2400525. [DOI] [PubMed] [Google Scholar]
  • 66.Sasaki K, Niitsu N. Elevated serum levels of soluble CD44 variant 6 are correlated with shorter survival in aggressive non-Hodgkin's lymphoma. Eur J Haematol. 2000;65:195–202. doi: 10.1034/j.1600-0609.2000.065003195.x. [DOI] [PubMed] [Google Scholar]
  • 67.Zeimet AG, Widschwendter M, Uhl-Steidl M, et al. High serum levels of soluble CD44 variant isoform v5 are associated with favourable clinical outcome in ovarian cancer. Br J Cancer. 1997;76:1646–1651. doi: 10.1038/bjc.1997.611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Spangrude GJ, Heimfeld S, Weissman IL. Purification Purification and characterization of mouse hematopoietic stem cells. Science. 1988;241:58–62. doi: 10.1126/science.2898810. [DOI] [PubMed] [Google Scholar]
  • 69.Kim CF, Jackson EL, Woolfenden AE, et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005;121:823–835. doi: 10.1016/j.cell.2005.03.032. [DOI] [PubMed] [Google Scholar]
  • 70.Tumbar T, Guasch G, Greco V, et al. Defining the epithelial stem cell niche in skin. Science. 2004;303:359–363. doi: 10.1126/science.1092436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Barker N, van Es JH, Kuipers J, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–1007. doi: 10.1038/nature06196. [DOI] [PubMed] [Google Scholar]
  • 72.Yamada Y, Yokoyama S, Wang XD, et al. Cardiac stem cells in brown adipose tissue express CD133 and induce bone marrow nonhematopoietic cells to differentiate into cardiomyocytes. Stem Cells. 2007;25:1326–1333. doi: 10.1634/stemcells.2006-0588. [DOI] [PubMed] [Google Scholar]
  • 73.Song H, Stevens CF, Gage FH. Astroglia induce neurogenesis from adult neural stem cells. Nature. 2002;417:39–44. doi: 10.1038/417039a. [DOI] [PubMed] [Google Scholar]
  • 74.Blanpain C, Horsley V, Fuchs E. Epithelial stem cells: turning over new leaves. Cell. 2007;128:445–458. doi: 10.1016/j.cell.2007.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Huntly BJ, Gilliland DG. Leukaemia stem cells and the evolution of cancer-stem-cell research. Nat Rev Cancer. 2005;5:311–321. doi: 10.1038/nrc1592. [DOI] [PubMed] [Google Scholar]
  • 76.Singh SK, Clarke ID, Terasaki M, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–5828. [PubMed] [Google Scholar]
  • 77.Toren A, Bielorai B, Jacob-Hirsch J, et al. CD133-positive hematopoietic stem cell “stemness” genes contain many genes mutated or abnormally expressed in leukemia. Stem Cells. 2005;23:1142–1153. doi: 10.1634/stemcells.2004-0317. [DOI] [PubMed] [Google Scholar]
  • 78.Kelly PN, Dakic A, Adams JM, et al. Tumor growth need not be driven by rare cancer stem cells. Science. 2007;317:337. doi: 10.1126/science.1142596. [DOI] [PubMed] [Google Scholar]
  • 79.Yen TH, Wright NA. The gastrointestinal tract stem cell niche. Stem Cell Rev. 2006;2:203–212. doi: 10.1007/s12015-006-0048-1. [DOI] [PubMed] [Google Scholar]
  • 80.Li HC, Stoicov C, Rogers AB, et al. Stem cells and cancer: evidence for bone marrow stem cells in epithelial cancers. World J Gastroenterol. 2006;12:363–371. doi: 10.3748/wjg.v12.i3.363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Li L, Neaves WB. Normal stem cells and cancer stem cells: the niche matters. Cancer Res. 2006;66:4553–4557. doi: 10.1158/0008-5472.CAN-05-3986. [DOI] [PubMed] [Google Scholar]
  • 82.Ponder BA, Schmidt GH, Wilkinson MM, et al. Derivation of mouse intestinal crypts from single progenitor cells. Nature. 1985;313:689–691. doi: 10.1038/313689a0. [DOI] [PubMed] [Google Scholar]
  • 83.Rizvi AZ, Swain JR, Davies PS, et al. Bone marrow-derived cells fuse with normal and transformed intestinal stem cells. Proc Natl Acad Sci U S A. 2006;103:6321–6325. doi: 10.1073/pnas.0508593103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bi CL, Fang JS, Chen FH, et al. [Chemoresistance of CD133(+) tumor stem cells from human brain glioma] Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2007;32:568–573. [PubMed] [Google Scholar]
  • 85.Houghton J, Stoicov C, Nomura S, et al. Gastric cancer originating from bone marrow-derived cells. Science. 2004;306:1568–1571. doi: 10.1126/science.1099513. [DOI] [PubMed] [Google Scholar]
  • 86.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]
  • 87.Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257. doi: 10.1038/35025220. [DOI] [PubMed] [Google Scholar]
  • 88.Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1:27–31. doi: 10.1038/nm0195-27. [DOI] [PubMed] [Google Scholar]
  • 89.Folkman J. Seminars in Medicine of the Beth Israel Hospital, Boston. Clinical applications of research on angiogenesis. N Engl J Med. 1995;333:1757–1763. doi: 10.1056/NEJM199512283332608. [DOI] [PubMed] [Google Scholar]
  • 90.Rafii S, Lyden D, Benezra R, et al. Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy? Nat Rev Cancer. 2002;2:826–835. doi: 10.1038/nrc925. [DOI] [PubMed] [Google Scholar]
  • 91.Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood. 2000;95:952–958. [PubMed] [Google Scholar]
  • 92.Shaked Y, Ciarrocchi A, Franco M, et al. Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Science. 2006;313:1785–1787. doi: 10.1126/science.1127592. [DOI] [PubMed] [Google Scholar]
  • 93.Bertolini F, Paul S, Mancuso P, et al. Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells. Cancer Res. 2003;63:4342–4346. [PubMed] [Google Scholar]
  • 94.Meropol NJ CS, Iannotti N, et al. Circulating tumor cells (CTC) predicts progression free (PFS) and overall survival (OS) in patients with metastatic colorectal cancer. 2007 ASCO Annual Meeting Proceedings. J Clin Oncol. 2007;25:18S. (abstr 4010) [Google Scholar]
  • 95.Lin E LY, Trent J, Patel S, Burgess MA, Steinert DM, Benjamin RS, Li D, Chen L. Quantitative real time-PCR of CD133 mRNA: A potential surrogate angiogenic marker of response for patients with metastatic sarcoma. 2006 ASCO Annual Meeting Proceedings. J Clin Oncol. 2006;24:18S. (abstr 20034) [Google Scholar]
  • 96.Herrera MB, Bruno S, Buttiglieri S, et al. Isolation and characterization of a stem cell population from adult human liver. Stem Cells. 2006;24:2840–2850. doi: 10.1634/stemcells.2006-0114. [DOI] [PubMed] [Google Scholar]
  • 97.Naor D, Nedvetzki S, Golan I, et al. CD44 in cancer. Crit Rev Clin Lab Sci. 2002;39:527–579. doi: 10.1080/10408360290795574. [DOI] [PubMed] [Google Scholar]
  • 98.Liu R, Wang X, Chen GY, et al. The prognostic role of a gene signature from tumorigenic breast-cancer cells. N Engl J Med. 2007;356:217–226. doi: 10.1056/NEJMoa063994. [DOI] [PubMed] [Google Scholar]
  • 99.Karnoub AE, Dash AB, Vo AP, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449:557–563. doi: 10.1038/nature06188. [DOI] [PubMed] [Google Scholar]
  • 100.Teicher BA, Herman TS, Holden SA, et al. Tumor resistance to alkylating agents conferred by mechanisms operative only in vivo. Science. 1990;247:1457–1461. doi: 10.1126/science.247.4949.1457. [DOI] [PubMed] [Google Scholar]
  • 101.Ortiz LA, Gambelli F, McBride C, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A. 2003;100:8407–8411. doi: 10.1073/pnas.1432929100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mueller LP, Luetzkendorf J, Mueller T, et al. Presence of mesenchymal stem cells in human bone marrow after exposure to chemotherapy: evidence of resistance to apoptosis induction. Stem Cells. 2006;24:2753–2765. doi: 10.1634/stemcells.2006-0108. [DOI] [PubMed] [Google Scholar]
  • 103.Shah AN, Summy JM, Zhang J, et al. Development and characterization of gemcitabine- resistant pancreatic tumor cells. Ann Surg Oncol. 2007;14:3629–3637. doi: 10.1245/s10434-007-9583-5. [DOI] [PubMed] [Google Scholar]
  • 104.Dallas NA XL, Fan F, et al. Resistance of colorectal cancer cells to 5-fluorouracil or oxaliplatin enriches for tumor stem cells. Proceedings of 2008 ASCO Gastrointestinal Cancers Symposium; (abstr 253) [Google Scholar]
  • 105.Calabrese C, Poppleton H, Kocak M, et al. A perivascular niche for brain tumor stem cells. Cancer Cell. 2007;11:69–82. doi: 10.1016/j.ccr.2006.11.020. [DOI] [PubMed] [Google Scholar]
  • 106.Jin L, Hope KJ, Zhai Q, et al. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. 2006;12:1167–1174. doi: 10.1038/nm1483. [DOI] [PubMed] [Google Scholar]
  • 107.Van Orden KL, Birse CE, He T.al: Proteomic analysis of colorectal tumor cells identifies CD133/Prominin-1, a cancer stem cell marker, as a monoclonal antibody therapeutic candidate Proc Am Assoc Cancer Res 2007(abstr LB 1287) [Google Scholar]
  • 108.Smith LM, Nesterova A, Ryan MC, et al. CD133/prominin-1 is a novel antibody therapeutic target in pancreatic, gastric and hepatocellular cancers. Proc Am Assoc Cancer Res. 2007 (abstr 1332) [Google Scholar]
  • 109.Florek M, Haase M, Marzesco AM, et al. Prominin-1/CD133, a neural and hematopoietic stem cell marker, is expressed in adult human differentiated cells and certain types of kidney cancer. Cell Tissue Res. 2005;319:15–26. doi: 10.1007/s00441-004-1018-z. [DOI] [PubMed] [Google Scholar]
  • 110.Maw MA, Corbeil D, Koch J, et al. A frameshift mutation in prominin (mouse)-like 1 causes human retinal degeneration. Hum Mol Genet. 2000;9:27–34. doi: 10.1093/hmg/9.1.27. [DOI] [PubMed] [Google Scholar]
  • 111.Hanai J, Gloy J, Karumanchi SA, et al. Endostatin is a potential inhibitor of Wnt signaling. J Cell Biol. 2002;158:529–539. doi: 10.1083/jcb.200203064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Gregory CA, Singh H, Perry AS, et al. The Wnt signaling inhibitor dickkopf-1 is required for reentry into the cell cycle of human adult stem cells from bone marrow. J Biol Chem. 2003;278:28067–28078. doi: 10.1074/jbc.M300373200. [DOI] [PubMed] [Google Scholar]
  • 113.Feldmann G, Dhara S, Fendrich V, et al. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67:2187–2196. doi: 10.1158/0008-5472.CAN-06-3281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ramasamy R, Lam EW, Soeiro I, et al. Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth. Leukemia. 2007;21:304–310. doi: 10.1038/sj.leu.2404489. [DOI] [PubMed] [Google Scholar]
  • 115.Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 116.Dy GK, Krook JE, Green EM, et al. Impact of complete response to chemotherapy on overall survival in advanced colorectal cancer: results from Intergroup N9741. J Clin Oncol. 2007;25:3469–3474. doi: 10.1200/JCO.2007.10.7128. [DOI] [PubMed] [Google Scholar]
  • 117.Benoist S, Brouquet A, Penna C, et al. Complete response of colorectal liver metastases after chemotherapy: does it mean cure? J Clin Oncol. 2006;24:3939–3945. doi: 10.1200/JCO.2006.05.8727. [DOI] [PubMed] [Google Scholar]
  • 118.Lin EH, Curley SA, Crane CC, et al. Retrospective study of capecitabine and celecoxib in metastatic colorectal cancer: potential benefits and COX-2 as the common mediator in pain, toxicities and survival? Am J Clin Oncol. 2006;29:232–239. doi: 10.1097/01.coc.0000217818.07962.67. [DOI] [PubMed] [Google Scholar]
  • 119.Zhang M CS, Ng C. Long-term maintenance capecitabine and celecoxib improved clinical outcomes by targeting colorectal cancer micrometastasis. 2007 ASCO Annual Meeting Proceedings. J Clin Oncol. 2007;25:18S. (abstr 14513) [Google Scholar]
  • 120.Colleselli D, Bijuklic K, Mosheimer BA, et al. Inhibition of cyclooxygenase (COX)-2 affects endothelial progenitor cell proliferation. Exp Cell Res. 2006;312:2933–2941. doi: 10.1016/j.yexcr.2006.05.021. [DOI] [PubMed] [Google Scholar]
  • 121.Arber N, Eagle CJ, Spicak J, et al. Celecoxib for the prevention of colorectal adenomatous polyps. N Engl J Med. 2006;355:885–895. doi: 10.1056/NEJMoa061652. [DOI] [PubMed] [Google Scholar]
  • 122.Buchanan FG, DuBois RN. Connecting COX-2 and Wnt in cancer. Cancer Cell. 2006;9:6–8. doi: 10.1016/j.ccr.2005.12.029. [DOI] [PubMed] [Google Scholar]
  • 123.Fuchs CS, Marshall J, Mitchell E, et al. Randomized, controlled trial of irinotecan plus infusional, bolus, or oral fluoropyrimidines in first-line treatment of metastatic colorectal cancer: results from the BICC-C Study. J Clin Oncol. 2007;25:4779–4786. doi: 10.1200/JCO.2007.11.3357. [DOI] [PubMed] [Google Scholar]
  • 124.Mangi AA, Noiseux N, Kong D, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9:1195–1201. doi: 10.1038/nm912. [DOI] [PubMed] [Google Scholar]

Articles from Gastrointestinal Cancer Research : GCR are provided here courtesy of International Society of Gastrointestinal Oncology

RESOURCES