Stem Cell Rev DOI 10.1007/s12015-008-9009-1

Targeting Cancer Stem Cells to Modulate Alternative Vascularization Mechanisms Elena Monzani & Caterina AM La Porta

# Humana Press Inc. 2008

Abstract Recently, many papers have shown that tumor vascularization can be explained by angiogenesis, recruitment, cooption, vasculogenic mimicry and by mosaic vessels. In particular, vasculogenic mimicry seems to be different from mosaic blood vessels, where tumor cells form a part of the surface of the vessel while the remaining part is covered by endothelium. In this case, tumor cells in apparent contact with the lumen do not show an endothelial phenotype. More recently, vasculogenic mimicry was proposed to occur in patients with multiple myeloma due to bone marrow macrophages. Herein, all these data are, for the first time, discussed critically in comparison to cancer stem cells—which show high trans-differentiative capacity— and bone-marrow derived stem cells. In fact, the presence of alternative vasculogenic patterns might be due to the presence of stem cell population (cancer stem cells or bone-marrow stem cells). In this connection, the literature is discussed extensively and possible models are proposed. Pharmacological perspectives will also discuss. Keywords Cancer stem cell . Vasculogenic mimicry . Vascularization . Endothelial progenitor cells . Melanoma Recently the cancer stem cell (CSC) hypothesis suggests that neoplastic clones are maintained exclusively by a rare fraction of cells with stem cell proprieties. Like normal stem cells, these rare CSCs possess the extensive proliferative and self-renewal potential necessary to create a new tumor and generate a hierarchy of phenotypically diverse E. Monzani : C. A. La Porta (*) Molecular Oncology Laboratory, Department of Biomolecular Science and Biotechnology, University of Milan, 20133 Milan, Italy e-mail: [email protected]

downstream cells, which are successively more limited in these properties. On the other hand, like as normal tissue stem cells, CSCs should display the ability to undergo a broad range of differentiation events. Recently a stem cell population was found in melanoma biopsy [1] and it was demonstrated to increase during melanoma progression [2]. Considering that melanoma is one of the most aggressive forms of skin cancer and it is strongly resistant to conventional therapeutic agents [3], the molecular biology of CSCs opens interesting new perspectives from the pharmacological point of view [3].

Non Sprouting Angiogenesis and Melanoma Until recently, vascularization of malignant tumors was considered the exclusive result of directed capillary ingrowth. However, recent advances have been made in identifying the processes involved in vascular remodelling. The simplistic model of an invading capillary sprout has been deemed insufficient to describe the entire spectrum of morphogenic and molecular events required to form a neovascular network. The different forms of “non sprouting angiogenesis” are vascular co-option of pre-existing vessels, intussusceptive microvascular growth, postnatal vasculogenesis, glomeruloid angiogenesis and vasculogenic mimicry. In vessel co-option a subset of tumors, organised around few functional vessels, rapidly coopts existing host vessels to form an initially well-vascularized tumor mass that, regressing, leading to a secondarily avascular tumor and massive tumor cell loss [4–7]. This process has now been observed in different tumour types like murine Lewis lung carcinoma, murine ovarian cancer, human melanoma and human Kaposi sarcoma [4–7]. However, the remaining

Stem Cell Rev

tumor is ultimately rescued by robust angiogenesis at the tumor margin [4, 8]. In vascular co-option, therefore, fused vessels together with the newly synthesized connective tissue, is incorporated into the tumor [9]. The expression patterns of Vascular endothelial growth factor (VEGF) and the natural Tie-2 receptor antagonist Ang-2 seem to play complementary and coordinated roles in this process [4, 10]. An additional mode of tumor angiogenesis different from sprouting, is intussusceptive (growth within itself) angiogenenesis (IMG), described approximately 18 years ago. It is a fast process, occurs within hours or even minutes, that does not need proliferation of endothelial cells but the remodelling of these cells by increasing in volume, in complexity (intussusceptive microvascular growth), and becoming thinner; for this region it is more economical from the energetic and metabolic point of view [11]. It is characterized by physiological levels of vascular transpermeability, a condition essential for uncompromized tissue and organ function and it may represent the unique means in the mechanisms of vascular tree formation and vascular remodelling [11]. Recently, IMG growth has been reported during angiogenesis in the mesenteric and peritoneal tissue induced by tumor ascites fluid [12] and in a human colon adenocarcinoma [13]. The fact that IMG angiogenesis does not involve intense cell proliferation, implies that neo-vascularisation by this mechanism would be resistant to cytotoxic treatment. As a consequence, cancerous cells could still cover their nutritional needs and continue to grow [14]. In general, the mechanisms that are involved in the regulation of intussusception are poorly understood but there is good evidence that flow alterations have a major influence in initiating the process of pillar formation [11]. The rapid vascular remodelling by IMG could possibly contribute to intermittent blood flow in tumors. Factors, that are known to be involved in these interactions in sprouting angiogenesis, such as the angiopoietins and their Tie receptors [15], platelet derived growth factor-B [16], monocyte chemotactic protein-1 [17], ephrins and EphB-receptors [18], are candidates for the mediation of IMG [11]. Postnatal angiogenesis is due to circulating bone marrow-derived endothelial progenitor cells (EPCs) that home to sites of physiological and pathological neovascularization and differentiate into endothelial cells. Until 1997, the growth of new blood vessels in adults was considered to exclusively occur through the mechanism of sprouting and intussusceptive angiogenesis. This paradigm of vascular development changed after the discovery, by Asahara et al. of CD34-enriched subpopulation of mononuclear blood cells named EPCs or angioblasts [19], located in a distinct zone of the vascular wall identified to be localized between smooth muscle and adventitial layer of human adult vascular wall [20]. Vascular wall areas

containing EPCs are found in large and middle sized arteries and veins of different organs [20]. These data suggest the existence of a ‘vasculogenic zone’ in the wall of adult human blood vessels not directly exposed to shear stress, which may serve as niche for progenitor cells for postnatal vasculogenesis, contributing to tumor vascularization and local immune response [20]. These cells express several endothelial specific markers like CD31, VEGF receptor (R)-2, Tie-2 [21] and CD14 [22] and have a role in maintenance of vascular integrity (postnatal vasculogenesis) [19]. On the other hand, bone marrow-derived circulating EPCs or angioblasts have the capacity to proliferate, migrate and differentiate into endothelial lineage cells and can contribute to tumor angiogenesis and growth of certain tumours [23]. Their discovery led to the new concept that vasculogenesis and angiogenesis may occur simultaneously in the postnatal life because these cells are able to differentiate when needed into vascular endothelium, through a mechanism recapitulating embryonic vasculogenesis [21, 24]. In particular, recruitment of EPCs from the bone marrow into the blood system, triggered by the increased availability of angiogenic growth factors or chemokines produced by tumours, such as VEGF and angiopoietin [25, 26], induce their incorporation into the sites of active neovascularization during tissue ischemia, vascular trauma or tumor growth and then promote differentiation in endothelial cells (ECs) [24, 27, 28]. EC progenitors have been identified in the blood vessels of mouse colon tumors and in xenografts of lymphoma, Lewis lung carcinoma, Ewing’s sarcoma, and breast cancer [24, 28–30]. Hypoxia, that can results from the tumour growth, can also mobilize EPCs from the bone marrow in the same way [23]. Some studies have demonstrated an impaired role of EPCs in angiogenesis after specific interventions. In a study by Capillo et al. [31], endostatin was described as a potent inhibitor of mobilization and clonogenic potential of EPCs. Similarly, simultaneous inhibition of VEGFR-2 and VEGFR-1 demonstrated an effective inhibition of mobilization and incorporation of EPCs in tumour vasculature [28]. Glomeruloid microvascular proliferations (GMPs) are focal proliferative buddings of endothelial cells resembling a renal glomerulus [32, 33], that was recently found in 13– 23% of various human tumours (melanoma, breast-, endometrial- and prostate cancer), and this vascular signature was significantly associated with an impaired prognosis [34]. The function of these structural units is not well understood. Formation of GMPs has been considered a dysregulated angiogenic response, due to imbalanced expression of angiogenic stimulators and inhibitors, but little is known about its detailed pathogenesis [35]. Presence of GMPs was associated with increased expression in tumour endothelium of the VEGF-A, receptors KDR, FLT-1 and neuropilin-1, as well as VEGF-D protein [36].

Stem Cell Rev

Finally vasculogenic mimicry was first described by the unique ability of aggressive melanoma cells to express an endothelial phenotype and to form vessel-like networks in three dimensional cultures, “mimicking” the pattern of embryonic vascular networks and recapitulating the patterned networks seen in patients with aggressive tumors correlated with poor prognosis (Fig. 1) [37]. In fact, the word ‘‘vasculogenic’’ was selected to indicate the generation of the pathway de novo and ‘‘mimicry’’ was used because the tumour uses cell pathways for transporting

a tumor

fluid in tissues that were clearly not blood vessels. Additional studies have reported vasculogenic mimicry in several other tumor types, including breast, prostate, ovarian, chorio-, lung carcinomas, synovial-, rhabdomyosarcoma, Ewing sarcomas and paeochromocytoma [38]. Interestingly, global gene analysis of aggressive and poorly aggressive human cutaneous and uveal melanoma cell lines unexpectedly revealed the ability of aggressive tumor cells to express genes associated with multiple cellular phenotypes and stem cells including endothelial, epithelial, fibroblast, and several other cell types [38–40]. These findings were interpreted considering that aggressive melanoma cells might revert to an undifferentiated, embryonic-like phenotype [37]. In this connection, aggressive melanoma cells express endothelium-associated genes and form extracellular matrix rich vasculogenic like networks in three dimensional cultures [41–49]. Microarray analysis indicated a genetic reversion of aggressive melanoma cells to an undifferentiated embryonic-like phenotype [38]. Endothelium associated genes such as VE-cadherin, Ephrin A2 and tissue factor pathway inhibitors, CD34, tyrosine kinase receptor 1, neuropilin 1, E-selectin and endoglin (CD105) had a more than twofold increased expression in

Blood

Other types of cells

b

EPC VEGF

tumor angiogenesis

Fig. 1 Blood vessels form through angiogenesis when sprouts grow out from existing vessels. However, alternative vascularization mechanisms occur in tumors such as vasculogenic mimicry. Recent evidences demonstrate that endothelial precursor cells (EPC) or macrophage cells can contribute to build neovessels (a). Factors such as VEGF produced by tumors may result in mobilization of these cells in the peripheral circulation and enhance their recruitment into the tumor vasculature (b). In red endothelial cells, in blue other types of cells (i.e., tumor cell, macrophages, EPC)

Table 1 Summary of angiogenic/vasculogeic factors as well as adhesion and stem cell markers expressed by human melanoma cancer stem cells [1] Gene

Expressed (+)

VEGF Flt-1 Flk-1 Nrp-1 Nrp-2 Ang-1 Ang-2 Tie-1 Tie-2 Ephrine A2 CD31 Endogline Notch 4 JAG1 JAG2 DLL1 DLL3 DLL4 Sox18 Semaphorine 3A Semaphorine 3F E-cadherin N-cadherin P-cadherin

+ − + + + + + − + + + + + − − − − − − + − − + −

Stem Cell Rev

vasculogenic mimicry positive cells, while genes related to a melanocytic phenotype, like Melan-A, microphthalmia associated transcription factor and tyrosinase, were more than 20-fold downregulated [8]. Another possibility is that the endothelial cell lining is replaced by tumour cells, resulting in the so-called ‘‘mosaic vessels’’, where both endothelial and tumour cells contribute to the formation of the vascular tube [50]. Mosaic blood vessels constitute 4% of the total surface area of tumor microcirculation. Some researchers believe that they are not related to vasculogenic mimicry (VM) channels and are a mosaicism of endothelial and tumor cells [50]. In contrast, Zhang and co-workers proposed a three-stage phenomenon among VM channels, mosaic blood vessels, and endothelium-dependent blood vessels, according to which all three patterns participate in tumor blood supply [51].

Vasculogenic Mimicry and Cancer Stem Cells: New Therapeutic Perspectives The transdifferentiative capacity of normal stem cells is a common properties of CSCs. Regarding to melanoma CSCs

like as all the other CSCs, they can transdifferentiate into different phenotype [1], they express neurogenic, angiogenic, vasculogenic and also lymphatic markers [1] (Table 1) and they are able to organize pseudovascular network in a physiological assay such as aorta ring [52] (Fig. 2). Thereby, we suggest that CSC subpopulation inside the tumor is able to organize vasculogenic mimicry or a mosaic network in dependence of the environmental condition. In this connection, a transdifferentiative capacity was demonstrated for bone marrow macrophages [53] and for dendritic cells [54], recently. Thereby we believe that, at least for melanoma, VM or mosaic vessel is due to the transdifferentiative capacity of CSCs subpopulation. The evidence of such a subpopulation opens of course new perspectives for the treatment of melanoma. In particular since the expression of proangiogenic/lymphatic factors might confer additional transdifferentiative capacity and a more aggressive phenotype, the use of anti-angiogenic drugs in combination with drugs acting on specific target of CSCs subpopulation, open interesting new therapeutic perspectives. To this aim, many efforts need to be done to better understand the biology of CSCs and to identify specific targets expressed by this subpopulation.

M M Aorta

Aorta

M

Aorta Fig. 2 CSC melanoma cells (WM115) was able to organize a pseudovascular network in rat aorta ring model (vasculogenic mimicry). Rat aorta ring and WM115 spheroids (M) were cocultivated in the presence of VEGF (20 ng/ml) in eight multi-wells collagen-coated. After 8 days, the cells were fixed, treated with 0.5% TRITON X-100 and stained with anti-VE cadherin (1:10) overnight at

M Aorta 4°C to detect human melanoma cells (red) or with fluorescein Griffonia (Bandeiraea) simplicifonia lactin 1, isolectin B4 (Vector) for 30 min at room temperature to detect rat endothelium (green). Nuclei were counterstained with DAPI (1:100) for 30 min. Arrowheads, rat aorta vessels; arrows, human melanoma vessels

Stem Cell Rev Acknowledgment We thank Riccardo Bazzotti for the relevant contribution to the vasculogenic mimicry picture. 18.

References 1. Monzani, E., Facchetti, F., Galmozzi, E., Corsini, E., Benetti, A., Cavazzin, C., et al. (2007). Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. European Journal of Cancer, 43, 935–946. 2. Klein, W. M., Wu, B. P., Zhao, S., Wu, H., Klein-Szanto, A. J., & Tahan, S. R. (2007). Increased expression of stem cell markers in malignant melanoma. Modern Pathology, 20, 102–107. 3. La Porta, C. A. (2007). Drug resistance in melanoma: new perspectives. Current Medicinal Chemistry, 14, 387–391. 4. Holash, J., Maisonpierre, P. C., Compton, D., Boland, P., Alexander, C. R., Zagzag, D., et al. (1999). Vessel cooption, regression and growth in tumors mediated by angiopoietins and VEGF. Science, 284, 1994–1998. 5. Zhang, L., Yang, N., Park, J. W., Katsaros, D., Fracchioli, S., Cao, G., et al. (2003). Tumor-derived vascular endothelial growth factor up-regulates angiopoietin-2 in host endothelium and destabilizes host vasculature, supporting angiogenesis in ovarian cancer. Cancer Research, 63, 3403–3412. 6. Dome, B., Paku, S., Somlai, B., & Timar, J. (2002). Vascularization of cutaneous melanoma involves vessel co-option and has clinical significance. The Journal of Pathology, 197, 355–362. 7. Kim, E. S., Serur, A., Huang, J., Manley, C. A., McCrudden, K. W., Frischer, J. S., et al. (2002). Potent VEGF blockade causes regression of coopted vessels in a model of neuroblastoma. Proceedings of the National Academy of Sciences of the United States of America, 99, 11399–11404. 8. Hillen, F., & Griffioen, A. W. (2007). Tumour vascularization: sprouting angiogenesis and beyond. Cancer Metastasis Reviews, 26, 489–502. 9. Paku, S., Kopper, L., & Nagy, P. (2005). Development of the vasculature in “pushing-type” liver metastases of an experimental colorectal cancer. International Journal of Cancer, 115, 893–902. 10. Maisonpierre, P. C., Suri, C., Jones, P. F., Bartunkova, S., Wiegand, S. J., Radziejewski, C., et al. (1997). Angiopoietin-2, a Natural Antagonist for Tie-2 That Disrupts in vivo Angiogenesis. Science, 277, 55–60. 11. Burri, P. H., Hlushchuk, R., & Djonov, V. (2004). Intussusceptive angiogenesis: Its emergence, its characteristics, and its significance. Developmental Dynamics, 231, 474–488. 12. Nagy, J. A., Morgan, E. S., Herzberg, K. T., Manseau, E. J., Dvorak, A. M., & Dvorak, H. F. (1995). Pathogenesis of ascites tumor growth: angiogenesis, vascular remodeling and stroma formation in the peritoneal lining. Cancer Research, 55, 376–385. 13. Patan, S., Munn, L. L., & Jain, R. K. (1996). Intussusceptive microvascular growth in a human colon adenocarcinoma xenograft: a novel mechanism of tumor angiogenesis. Microvascular Research, 51, 260–272. 14. Burri, P. H., & Djonov, V. (2002). Intussusceptive angiogenesis–– the alternative to capillary sprouting. Molecular Aspects of Medicine, 23, S1–S27. 15. Folkman, J., & D’Amore, P. A. (1996). Blood vessel formation: what is its molecular basis? Cell, 87, 1153–1155. 16. Hellstrom, M., Kalen, M., Lindahl, P., Abramsson, A., & Betsholtz, C. (1999). Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development, 126, 3047–3055. 17. Shyy, Y. J., Hsieh, H. J., Usami, S., & Chien, S. (1994). Fluid shear stress induces a biphasic response of human monocyte

19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

chemotactic protein 1 gene expression in vascular endothelium. Proceedings of the National Academy of Sciences of the United States of America, 91, 4678–4682. Gale, N. W., Baluk, P., Pan, L., Kwan, M., Holash, J., DeChiara, T. M., et al. (2001). Ephrin-B2 selectively marks arterial vessels and neovascularization sites in the adult, with expression in both endothelial and smooth-muscle cells. Developmental Biology, 230, 151–160. Döme, B., Hendrix, M. J., Paku, S., Tóvári, J., & Tímár, J. (2007). Alternative vascularization mechanisms in cancer: pathology and therapeutic implications. The American Journal of Pathology, 170, 1–15. Zengin, E., Chalajour, F., Gehling, U. M., Ito, W. D., Treede, H., Lauke, H., et al. (2006). Vascular wall resident progenitor cells: a source for postnatal vasculogenesis. Development, 133, 1543– 1551. Asahara, T., Murohara, T., Sullivan, A., Kalka, C., Pastore, C., Silver, M., et al. (1997). Isolation of putative progenitor endothelial cells for angiogenesis. Science, 275, 964–967. Kalka, C., Masuda, H., Takahashi, T., Kalka-Moll, W. M., Silver, M., Kearney, M., et al. (2000). Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proceedings of the National Academy of Sciences of the United States of America, 97, 3422–3427. Ribatti, D. (2004). The involvement of endothelial progenitor cells in tumour angiogenesis. Journal of Cellular and Molecular Medicine, 8, 294–300. Asahara, T., Masuda, H., Takahashi, T., Kalka, C., Pastore, C., Silver, M., et al. (1999). Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circulation Research, 85, 221–228. Hattori, K., Dias, S., Heissig, B., Hackett, N. R., Lyden, D., Tateno, M., et al. (2001). Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells. The Journal of Experimental Medicine, 193, 1005–1014. Iwaguro, H., Yamaguchi, J., Kalka, C., Murasawa, S., Masuda, H., Hayashi, S., et al. (2002). Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation, 105, 732–738. Gill, M., Dias, K., Hattori, M. L., Rivera, D., Hicklin, L., Witte, L., et al. (2001). Vascular trauma induces rapid but transient mobilization of VEGFR2(+) AC133(+) endothelial precursor cells. Circulation Research, 88, 167–174. Lyden, D., Hattori, K., Dias, S., Costa, C., Blaikie, P., Butros, L., et al. (2001). Impaired recruitment of bone marrow derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nature Medicine, 7, 1194–1201. Bolontrade, M. F., Zhou, R. R., & Kleinerman, E. S. (2002). Vasculogenesis plays a role in the growth of Ewing’s sarcoma in vivo. Clinical Cancer Research, 8, 3622–3627. Shirakawa, K., Furuhata, S., Watanabe, I., Hayase, H., Shimizu, A., Ikarashi, Y., et al. (2002). Induction of vasculogenesis in breast cancer models. British Journal of Cancer, 87, 1454–1461. Capillo, M., Mancuso, P., Gobbi, A., Monestiroli, S., Pruneri, G., Dell’Agnola, C., et al. (2003). Continuous infusion of endostatin inhibits differentiation, mobilization, and clonogenic potential of endothelial cell progenitors. Clinical Cancer Research, 9, 377–382. Pettersson, A., Nagy, J. A., Brown, L. F., Sundberg, C., Morgan, E., Jungles, S., et al. (2000). Heterogeneity of the angiogenic response induced in different normal adult tissues by vascular permeability factor/vascular endothelial growth factor. Laboratory Investigation, 80, 99–115. Wesseling, P., Vandersteenhoven, J. J., Downey, B. T., Ruiter, D. J., & Burger, P. C. (1993). Cellular components of microvas-

Stem Cell Rev

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

cular proliferation in human glial and metastatic brain neoplasms. A light microscopic and immunohistochemical study of formalinfixed, routinely processed material. Acta Neuropathologica, 85, 508–514. Straume, O., Chappuis, P. O., Salvesen, H. B., Halvorsen, O. J., Haukaas, S. A., Goffin, J. R., et al. (2002). Prognostic importance of glomeruloid microvascular proliferation indicates an aggressive angiogenic phenotype in human cancers. Cancer Research, 62, 6808–6811. Brat, D. J., Castellano-Sanchez, A., Kaur, B., & Van Meir, E. G. (2002). Genetic and biologic progression in astrocytomas and their relation angiogenic dysregulation. Advances in Anatomic Pathology, 9, 24–36. Straume, O., & Akslen, L. A. (2003). Increased expression of VEGF-receptors (FLT-1, KDR, NRP-1) and thrombospondin-1 is associated with glomeruloid microvascular proliferation, an aggressive angiogenic phenotype, in malignant melanoma. Angiogenesis, 6, 295–301. Maniotis, A. J., Folberg, R., Hess, A., Seftor, E. A., Gardner, L. M., Pe'er, J., et al. (1999). Vascular channel formation by human melanoma cells in vivo and in vitro, vasculogenic mimicry. The American Journal of Pathology, 155, 739–752. Hendrix, M. J., Seftor, E. A., Hess, A. R., & Seftor, R. E. (2003). Vasculogenic mimicry and tumor cell plasticity: lessons from melanoma. Nature Reviews Cancer, 3, 411–421. Seftor, E. A., Meltzer, P. S., Schatteman, G. C., Gruman, L. M., Hess, A. R., Kirschmann, D. A., et al. (2002). Expression of multiple molecular phenotypes by aggressive melanoma tumor cells: role in vasculogenic mimicry. Critical Reviews in Oncology/ hematology, 44, 17–27. Hendrix, M. J., Seftor, E. A., Hess, A. R., & Seftor, R. E. (2003). Molecular plasticity of human melanoma cells. Oncogene, 22, 3070–3075. Folberg, R., Rummelt, V., Parys-Van Ginderdeuren, R., Hwang, T., Woolson, R. F., & Pe’er, J., et al. (1993). The prognostic value of tumor blood vessel morphology in primary uveal melanoma. Ophthalmology, 100, 1389–1398. Makitie, T., Summanen, P., Tarkkanen, A., & Kivela, T. (1999). Microvascular loops and networks as prognostic indicators in choroidal and ciliary body melanomas. Journal of National Cancer Institute, 91, 359–367. Sakamoto, T., Sakamoto, M., Yoshikawa, H., Hata, Y., Ishibashi, T., Ohnishi, Y., et al. (1996). Histologic findings and prognosis of

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

uveal malignant melanoma in Japanese patients. American Journal of Ophthalmology, 121, 276–283. Seregard, S., Spangberg, B., Juul, C., & Oskarsson, M. (1998). Prognostic accuracy of the mean of the largest nucleoli, vascular patterns, and PC-10 in posterior uveal melanoma. Ophthalmology, 105, 485–491. Thies, A., Mangold, U., Moll, I., & Schumacher, U. (2001). PASpositive loops and networks as a prognostic indicator in cutaneous malignant melanoma. The Journal of Pathology, 195, 537–542. Warso, M. A., Maniotis, A. J., Chen, X., Majumdar, D., Patel, M. K., & Shilkaitis, A., et al. (2001). Prognostic significance of periodic acid-Schiff-positive patterns in primary cutaneous melanoma. Clinical Cancer Research, 7, 473–477. Rummelt, V., Mehaffey, M. G., Campbell, R. J., Pe’er, J., Bentler, S. E., & Woolson, R. F., et al. (1998). Microcirculation architecture of metastases from primary ciliary body and choroidal melanomas. American Journal of Ophthalmology, 126, 303–305. Shirakawa, K., Kobayashi, H., Heike, Y., Kawamoto, S., Brechbiel, M. W., & Kasumi, F., et al. (2002). Hemodynamics in vasculogenic mimicry and angiogenesis of inflammatory breast cancer xenograft. Cancer Research, 62, 560–566. Zhou, Y., Fisher, S. J., Janatpour, M., Genbacev, O., Dejana, E., Wheelock, M., et al. (1997). Human cytotrophoblasts adopt a vascular phenotype as they differentiate. A strategy for successful endovascular invasion. The Journal of Clinical Investigation, 99, 2139–2151. Chang, Y. S., di Tomaso, E., Mc Donald, D. M., Jones, R., Jain, R. K., & Munn, L. L. (2000). Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proceedings of the National Academy of Sciences of the United States of America, 97, 14608–14613. Zhang, S., Guo, H., Zhang, D., Zhang, W., Zhao, X., Ren, Z., et al. (2006). Microcirculation patterns in different stages of melanoma growth. Oncology Reports, 15, 15–20. Nicosia, R. F., & Ottinetti, A. (1990). Growth of microvessels in serum-free matrix culture of rat aorta. A quantitative assay of angiogenesis in vitro. Laboratory Investigation, 63, 115–122. Scavelli, C., Nico, B., Cirulli, T., Ria, R., Di Pietro, G., Mangieri, D., et al. (2007). A vasculogenic mimicry by bone marrow macrophages in patients with multiple myeloma. Oncogene, 30, 1–12. Sozzani, S., Rusnati, M., Riboldi, E., Mitola, S., & Presta, M. (2007). Dendritic cell-endothelial cell cross-talk in angiogenesis. Trends in Immunology, 28, 385–392.

Targeting Cancer Stem Cells to Modulate Alternative ...

frequency of cancer cells in contact with flowing blood. Proceedings of the National Academy of Sciences of the United. States of America, 97, 14608–14613. 51.

388KB Sizes 1 Downloads 191 Views

Recommend Documents

Most C6 Cells Are Cancer Stem Cells: Evidence ... - Cancer Research
Apr 15, 2007 - cell line using clonal and population analyses, rather than isolating .... Phone: 86-571-87784606; Fax: 86-571-87783757; E-mail: pheiphei@. 163.com. ..... multipotentiality or a cell's ability to give rise to multiple cell types.

Eradication of Cancer Stem Cells
tion of cells: these are the stem cells (SC). Tissue specific SC are ... specific SC, including its rate of replication, ..... Dingli, D., Traulsen, A., and Michor, F. 2007a.

Identifying Cancer Stem Cells in Solid Tumors - Cancer Research
Feb 15, 2006 - Top, theoretical concerns; bottom, technical concerns. (discussed in ... of initiating tumor growth when injected back into animals. Technical ..... http://cancerres.aacrjournals.org/content/66/4/1891.full.html#ref-list-1. This article

Normal Stem Cells and Cancer Stem Cells: The Niche ...
Sep 14, 1982 - E-mail: [email protected]. I2006 American ... progenitor cells causes a blast crisis in some patients with chronic myeloid leukemia (14).

Normal Stem Cells and Cancer Stem Cells: The Niche ...
Sep 14, 1982 - Likewise, integrin is required for stem cell migration as evidenced in both hematopoietic and ... cancer cell migration. (69). In addition, recent data support the role of the vascular niche .... template DNA strands. J Cell Sci 2002 .

Stem Cells PH v9.pdf
There is evidence to suggest that regenerative cell therapies using cell types such as endothelial progenitor cells. (EPCs) and mesenchymal stem cells (MSCs) ...

MR studies of stem cells
MR-TRACKING OF MAGNETICALLY LABELED NEURAL STEM CELLS ON TISSUE ...... A potential application of these interesting properties of NSCs is to treat.

Re: "stem cells and cloning"
Aug 11, 2001 - Concerning organs received from a dead person, in a letter dated 26 June 1956, also ..... We will also visit briefly the evolution of the Universe. ...... upright animal receives 60% less heat compared to four-legged companion.

Re: "stem cells and cloning" - Semantic Scholar
Aug 11, 2001 - 3. Summary of chapter on Citric Acid—Conversion of fuel into Energy. 4. ...... Planets orbit around stars, which in turn are part of a galaxy.

Re: "stem cells and cloning" - Semantic Scholar
Aug 11, 2001 - incubator: this is essentially a matter that concerns science, and as such should be ...... had already suffered through two courses in Organic Chemistry in undergraduate school. .... The fact that you can design randomness through a c

Stochastic Dynamics of Hematopoietic Tumor Stem Cells
http://www.landesbioscience.com/journals/cc/abstract.php?id=3853. Once the issue is complete and page numbers have been assigned, the citation will change ...

Stem cells - European Medicines Agency - Europa EU
Jun 27, 2016 - An agency of the European Union ... European Medicines Agency, 2016. ... agents poses a real challenge taken into account that the ideal. 37.

[PDF] Pancreatic Stem Cells
pancreatic development and regeneration. The many strategies to differentiate adult and embryonic stem cells into pancreatic beta cells are also discussed in the context of potential therapeutic interventions for type I diabetes. Book details. Author

NCIM Cancer Program - Nova Cells Institute
Back during 1999 Wake Forest researcher Zheng Cui, MD, PhD injected mice with an ultra aggressive form of cancer that should have killed all the treated ...

A Novel Response of Cancer Cells to Radiation ...
Jan 15, 2001 - Increased red:green (R/G) fluorescence ratio in irradiated cells is radiation ... AVO formation or function may serve as a tool to increase cell .... Anderson, R. G. W., Falck, J. R., Goldstein, J. L., and Brown, M. S. Visualization of

Ingrowth of human mesenchymal stem cells into porous ...
c Centre for Material and Fibre Innovation, Deakin University, Geelong, Victoria, Australia. 10 d Department of .... and at reasonable costs due to the commodity textile business. 139 ..... (GraphPad Software Inc., San Diego, CA). Significance ...