• 沒有找到結果。

結合帝盟多及CD8+為主的腫瘤?潤細胞針對惡性膠質母細胞瘤之CD133(+)腫瘤幹細胞的毒殺作用分析

N/A
N/A
Protected

Academic year: 2021

Share "結合帝盟多及CD8+為主的腫瘤?潤細胞針對惡性膠質母細胞瘤之CD133(+)腫瘤幹細胞的毒殺作用分析"

Copied!
6
0
0

加載中.... (立即查看全文)

全文

(1)

Printed in the USA. All rights reserved. DOI: 10.3727/096368910X532774

Copyright 2011 Cognizant Comm. Corp. E-ISSN 1555-3892

www.cognizantcommunication.com

The Role of Cancer Stem Cells (CD133

+

) in Malignant Gliomas

Der-Yang Cho,*† Shinn-Zong Lin,*† Wen-Kuang Yang,* Den-Mei Hsu,*

Hung-Lin Lin,* Han-Chung Lee,* Wen-Yeun Lee,* and Shao-Chih Chiu†

*Department of Neurosurgery, Neuropsychiatric Center, Cell/Gene Therapy Research Laboratory, China Medical University Hospital, Taichung, Taiwan

†Graduate Institute of Immunology, China Medical University, Taichung, Taiwan

Malignant gliomas, particularly glioblastoma multiforme (GBM) tumors, are very difficult to treat by con-ventional approaches. Although most of the tumor mass can be removed by surgical resection, radiotherapy, and chemotherapy, it eventually recurs. There is growing evidence that cancer stem cells (CSCs) play an important role in tumor recurrence. These stem cells are radioresistant and chemoresistant. The most com-monly used tumor marker for CSCs is CD133. The amount of CSC component is closely correlated with tumor malignancy grading. Isolating, identifying, and treating CSCs as the target is crucial for treating malignant gliomas. CSC-associated vascular endothelial growth factor (VEGF) promotes tumor angiogen-esis, tumor hemorrhage, and tumor infiltration. Micro-RNA (miRNA) plays a role in CSC gene expression, which may regulate oncogenesis or suppression to influence tumor development or progression. The antigen-esis of CSCs and normal stem cells may be different. The CSCs may escape the T-cell immune response. Identifying a new specific antigen from CSCs for vaccine treatment is a key point for immunotherapy. On the other hand, augmented treatment with radiosensitizer or chemosensitizer may lead to reduction of CSCs and lead to CSCs being vulnerable to radiotherapy and chemotherapy. The control of signaling pathway and cell differentiation to CSC growth is another new hope for treatment of malignant gliomas. Although the many physiological behavioral differences between CSCs and normal stem cells are unclear, the more we know about these differences the better we will be able to treat CSCs effectively.

Key words: Cancer stem cells (CSCs); CD133+; Glioblastoma multiforme (GBM); Malignant gliomas;

Micro-RNA (miRNA); Signaling pathway

INTRODUCTION neural progenitor proteins such as nestin (8), Sox2, Oct 4, and Musashi (4).

Various stem cell markers are found in different ma-Malignant gliomas are the most common malignant

primary intracranial tumors. Their incidence is 5.4/ lignant tumors. The CD133 tumor marker is specific and commonly used for CSCs of malignant gliomas (30). 100,000 (11). Glioblastoma multiforme (GBM) (WHO

IV) tumors are the most common highly invasive malig- Recent studies showed that biological behaviors of

CD133+ were like the CSC subpopulation that confers nant gliomas. Their prognosis is still dismal even though

aggressive surgery, radiotherapy, and chemotherapy are glioma radio- and chemoresistance (21,34). These stem cells may be the source of tumor recurrence after radia-used for treatment (40). The median survival is only

12–15 months for GBM (22,37). It seems to be an incur- tion (25). Oka et al. (29) reported that CD133+ CSCs were identified in the peripheral brain regions adjacent able disease due to its highly infiltrative phenotype.

There is growing evidence that GBM harbors small cell to the tumor. They were frequently localized around vas-cular structures (the vasvas-cular niche). Singh et al. (36) populations that may sustain tumor formation and

growth. These cells are called cancer stem cells (CSCs) reported that CD133+ CSCs in GBM might be

trans-plantable to xenograft tumors in SCID (severe combined (31,32). Cancer stem cells are not only found in

leuke-mia, multiple myeloma, and breast cancer but also in immunodeficiency disease) mice brain, which

recap-tured the features of the original tumor regarding mor-GBM. The CSCs share many properties with normal

stem cells including self-renewal and multipotency (14). phology lineage and marker expression, and generated both CD133+and CD133− cells. However, in the same They have also been shown to express various specific

Online prepub date: September 30, 2010.

Address correspondence to Wen-Kuang Yang, M.D., Ph.D., Professor, Cell/Gene Therapy Research Laboratory, China Medical University Hospital, No. 2 Yu-Der Road, Taichung, Taiwan, R.O.C. Tel: 886-42-2052121, ext. 2779; Fax: 886-42-2079649, ext. 5035; E-mail: wkyang@mail.cmu.edu.tw

(2)

patient from whom the CD133 cells were obtained, the of tumor CSCs varied from 1.02% to 2.32% based on flow cytometric analysis. Singh et al. (36) reported less cells could not form neurospheres in culture medium

and could not form tumor growth in SCID mice (18). than 20% of CD133+in total tumor mass, but wide rang-ing variations in the CD133+ cell ratio (0.1–50% in The CD133 CSC marker is analyzed by

immunohis-tochemistry staining and Western blotting. The compo- GBM) were reported. Low-grade gliomas also contain

glioma stem cells, but in relatively low numbers. Thon nent amount of CD133+cells closely correlates with

gli-oma malignancy. The quantities of CD133+CSCs within et al. (38) also reported that the number of CD133+cells quantitatively correlated with tumor grade.

the tumor mass might show a clear quantitative correla-tion with glioma grading (WHO II, III, and IV). These

NEOANGIOGENESIS IN CSCs

findings support the concept of CD133+CSC-dependent

gliomagenesis (38). The CD133+ cells from gliomas of different grades

might contribute to intratumoral neoangiogenesis. CD133+

ISOLATION AND IDENTIFICATION

cells frequently accumulate within highly vascularized

OF CSCs IN GLIOMAS

regions, particularly in the case of GBM. Bao et al. (2) The definition of CSCs must meet the following four

reported evidence that the mutual influence of stem cells criteria: 1) generate clonally derived cells that form

neu-and endothelium account for neoangiogenesis. Thon et rospheres; 2) possess properties of cell renewal and

pro-al. (38) also reported that CD133+cells exhibit expres-liferation; 3) differentiate and express typical CSC

sion of endothelial progenitor and display a typical en-markers of tumor cells; 4) generate tumors after in vivo

dothelial “cobblestone” growing pattern. Vascular endo-transplantation in animal models that resemble the

origi-thelial growth factor (VEGF) promotes tumorigenesis nal donor tumors (39).

via angiogenesis (28). VEGF-repressing CSCs lead to CSCs of brain tumors were first identified in primary

the massive expansion of vascular-rich GBM and tumor-brain tumors (e.g., GBM, medulloblastoma, pilocytic

associated hemorrhage, thereby causing high morbidity. astrocytoma, and ganglioglioma). They formed

clono-VEGF provides a vascular-rich tumor environment stem genic spheres (neurospheres) in serum-free culture

me-cell niche. dia containing epidermal growth factor (EGF) and basic

fibroblast growth factor (bFGF). The CD133 marker is MICRO-RNA REGULATION OF CSCs

a 120-kDa transmembrane cell-surface protein. In the

Micro-RNAs (miRNAs) are a novel group of short course of enzymatic digestion of tumor tissue, an

exces-RNAs, about 22 nucleotides in length, that regulate gene sively long incubation period easily damages CSCs and

expression in a posttranscriptional manner by paring breaks the structure of cell-surface antigens. The CD133 with complementary nucleotide sequences in the 3′-tumor marker can be also present in other 3′-tumors

includ-untranslatable region of target mRNA. The miRNAs ing prostate tumors and colon cancers. We may identify

regulate cell functions, either oncogenesis or tumor sup-CSCs in cell culture by neurosphere formation with

pos-pression, which influence cancer development and pro-itive CD133+ and nestin neural stem cell markers (42).

gression. Recently, 7, 21, and miRNA-When the CSCs of brain tumor cells are transferred into

451 were found to be involved in regulation of brain the nude or SCID mice brain they may induce tumor

differentiation (16). MiRNA-451 may disperse neu-formation (26). Singh et al. (36) demonstrated that as

rospheres and inhibit the growth of GBM cells. MiRNAs

few as 100 CD133+ cells derived from human GBM

may drive CD133+ cells to differentiate and lose their could form GBM in SCID mice; however, the injection

stem cell character. When the miRNA levels in CD133+ of 5× 104 to 1× 105 CD133cells did not. The CSCs

cells are raised by transfection, there may be inhibition from gliomas are concentrated by cell sorting or

mag-of cell growth and proliferation from loss mag-of cell renewal netic columns using specific immunoreactivities. The

potential. MiRNA-124 and miRNA-137 may also inhibit CSCs are thought to maintain their drug (Hoechst 33342

proliferation of GBM cells and induce differentiation of dye) efflux ability, which makes it possible to separate

brain tumor stem cells (35). CSCs in unstained cell fractions (17). Kang et al. (19)

reported radiation might increase CSCs from 2–3% ini- IMMUNE RESPONSE TO CSCs

tially to 5–7% in recurrent tumor. Low-dose radiation

The brain is not an immunologically exempt organ or hypoxic challenge might be used in the future for

without elicitation of inflammation and immune re-experimental study to increase the concentration of

sponse (1). According to tumor immunosurveillance the-CSCs in cell culture (20).

ory, tumor can be recognized and eliminated as a result

THE COMPONENT AMOUNT of the natural antitumor immune response (9). But in

OF CSCs IN GLIOMAS tumor development, tumor cells may influence immu-nity either from the tumor environment, rendering a tu-CD133+cell markers are positive in 60–70% of

(3)

the antitumor response. Most cancers are composed of a cells also involve tumor stem cell proliferation. Know-ing how to identify such tumor-specific signalKnow-ing path-mixture of normal stem cell and CSCs. The antigenesis

of normal stem cells and CSCs may be different. The ways and their inactivation mechanism will provide in-formation on key targets for glioma treatment. In CSCs frequently escape the T-cell immune response. It

is critical to fully characterize the immunological fea- comparison to CD133−tumor cells, CD133+CSCs have

higher DNA repair capacity, which results in a selective tures of CSCs and to develop immunotherapeutic

ap-proaches to eliminate CSCs without excessive toxicity postirradiation increase in the CD133+population (15). CSCs express multidrug resistance genes (e.g., ABCG2 to normal stem cells (41).

and BCRPI) that aide in the efflux of drugs and in the

CHEMOSENSITIZER AND selective promotion of CSC survival (15). The sonic RADIOSENSITIZER FOR CSCs hedgehog (shh) (10) and the Notch pathway are very

important for brain development and for maintaining Liu et al. (24) reported that CD133+CSCs play an

important role in tumor resistance to chemotherapy. This cell “stemness.” An shh pathway inhibitor, cyclopamine, depletes CSCs (3). In the Notch pathway, γ-secretase resistance is probably contributed to by CD133+ cells

with greater expression of breast cancer resistance pro- inhibitors also decrease the CD133+ fraction (13). Pro-moting the differentiation of stem cells to normal cells tein (BCRP1) and methyl-guanine methyltransferase

(MGMT) as well as the antiapoptosis proteins and inhib- is another possible approach for CSC treatment. Picciri-llo et al. (33) reported that bone morphogenetic protein itors of apoptosis protein families. Miqueli et al. (27)

reported that the monoclonal antibodies for anti-epider- (BMP4) might reduce the CSCs in GBM by differentiat-ing the CSCs to astrocytes.

mal growth factor receptors can increase radiosensitiza-tion to GMB CSCs. Chang et al. (6) reported that the

CONCLUSIONS

mean survival rate of GBM with CD133+ mice treated

with radiation was significantly improved by knock- Cancer stem cell investigation is a good starting point down of silencing information regulator (SirT1), a mem- for controlling tumor growth and recurrence (23). Al-ber of the sirtuin family, which is an NAD-dependent though many differences in physiological behavior be-histone deacetylase and essential mediator of longevity tween CSCs and normal stem cells are unclear, the more in normal cells. Ehtesham et al. (12) showed that cell we know about CSCs the more we can treat them effec-surface chemokine receptor (CCR4), a mediator of can- tively.

cer proliferation and invasion, was overexpressed in

ACKNOWLEDGMENTS: This study is supported in part by GBM CSCs. Administration of CXCL12, the only known Taiwan Department of Health Cancer Research Center of Ex-ligand for CXCR4, stimulates a specific and significant cellence (DOH-TD-C-111-005), and in part by Taiwan De-partment of Health Clinical Trial and Research Center of Ex-proliferative response in progenitors but not in

differen-cellence (DOH-TD-B-111-004). It also is partially granted tiated tumor cells.

from National Science Council (NSC-2314-B-039-012-MY2). Kang and Kang (20) reported pharmacological

block-age of chloride channel synergistically enhanced apopto- REFERENCES

sis in drug-resistant CSCs. Casper et al. (5) reported that

1. Akasaki, Y.; Black, K. L.; Yu, J. S. Dendritic cell-based acetaminophen, acetylsalicylate, and ibuprofen also

in-immunotherapy for malignant gliomas. Expert Rev. Neu-creased radiosensitivity in culture and reduced glioma rother. 5:497–508; 2005.

cell growth. Similarly, other nonsteroidal anti-inflamma- 2. Bao, S.; Wu, Q.; McLendon, R. E.; Hao, Y.; Shi, Q.; Hjel-meland, A. B. Glioma stem cell promote radioresistance tory drugs (NSAIDs) also directly act on tumor cell

by preferential activation of the DNA damage response. growth by the inhibition of prostaglandin synthesis.

Nature 444:756–760; 2006. NSAIDs increase the concentration of arachidonic acid,

3. Bar, E. E.; Chaudhry, A.; Lin, A.; Fan, X.; Schreck, K.; a precursor of prostaglandins, which induces ceramide Matsui, M.; Piccirillo, S.; Vescovi, A. L.; Dimeco, F.; formation from sphingomyelin. Subsequently, ceramide- Olivi, A.; Eberhart, C. G. Cyclopamine-mediated hedge-hog pathway inhibition depletes stem-like cancer cells in induced apoptosis ensues (7). The inhibition of

prosta-glioblastoma. Stem Cells 25:2524–2533; 2007. glandin synthesis also decreases tumor size directly by

4. Bleau, A. M.; Howard, B. M.; Taylor, L. A.; Gursel, D.; inhibiting angiogenesis (43). Whether these

radiosensi-Greenfield, J. P.; Lim Tung, H. Y.; Holland, E. C.; Boock-tizers and chemosensiBoock-tizers are effective for CSCs too var, J. A. New strategy for the analysis of phenotypic

needs to be further evaluated. marker antigens in brain tumor-derived neurospheres in

mice and humans. Neurosurg. Focus 24:E27; 2008.

IMPLICATION OF CSCs 5. Casper, D.; Lekhuaj, R.; Yaparpalvi, U. S.; Pidel, A.; Jaggernauth, W. A.; Werner, P.; Tribius, S.; Rowe, J. D.;

FOR GLIOMA TREATMENT

LaSala, P. A. Acetaminophen selectively reduces glioma As with normal tissue stem cells, CSCs are mostly cell growth and increases radiosensitivity in culture. J. quiescent and resistant to conventional treatment. Sig- Neurooncol. 46:215–229; 2000.

6. Chang, C. J.; Hsu, C. C.; Yung, M. C.; Chen, K. Y.; Tzao, naling pathways involving activation of normal stem

(4)

C.; Wu, W. F.; Chou, H. Y.; Lee, Y. Y.; Lu, K. H.; Chiou, 21. Kang, M. K.; Kang, S. K. Tumorigenesis of chemothera-peutic drug-resistant cancer stem-like cells in brain gli-S. H.; Ma, H. I. Enhanced radiosensitivity and

radiation-induced apoptosis in glioma CD133-positive cells by oma. Stem Cells Dev. 16:837–847; 2007.

22. Kelly, K. A.; Kirkwood, J. M.; Kapp, D. S. Glioblastoma knockdown of SirT1 expression. Biochem. Biophys. Res.

Commun. 380:236–242; 2009. multiforme: Pathology, natural history and treatment.

Can-cer Treat. Rev. 11:1–26; 1984. 7. Chang, T. A.; Morin, P. T.; Vogelstein, B.; Kinzler,

K. W. Mechanisms underlying nonsteroidal antiflamma- 23. Kobayashi, N.; Navarro-Alvarez, N.; Soto-Gutierrez, A.; Kawamoto, H.; Kondo, Y.; Yatsuji, T.; Shirakawa, Y.; tory drug-mediated apoptosis. Proc. Natl. Acad. Sci. USA

95:681–686; 1998. Naomoto, Y.; Tanaka, N. Cancer stem cell research:

Cur-rent situation and problems. Cell Transplant. 17:19–25; 8. Chinnayan, P.; Wang, M.; Rojiani, A. M.; Tofilon, P. J.;

Chakravarti, A.; Ang, K. K.; Zhang, H. Z.; Hammond, E.; 2008.

24. Liu, G.; Yuan, X.; Zeng, Z.; Tunici, P.; Ng, H.; Abdul-Curran, Jr., W. C.; Mehta, M. P. The prognostic value of

nestin expression in newly diagnosed glioblastoma: Re- kadir, I. R.; Lu, L.; Irvin, D.; Black, K. L.; Yu, J. S. Anal-ysis of gene expression and chemoresistance of CD133+ port from the radiation therapy oncology group. Radiat.

Oncol. 3:32; 2008. cancer stem cells in glioblastoma. Mol. Cancer 5:67;

2006. 9. Cho, D. Y.; Lin, S. Z.; Yang, W. K.; Hsu, S. M.; Lee,

H. C.; Lee, W. Y.; Liu, S. P. Dendritic cells (DCs)-based 25. Lubensky, I. A.; Vortmeyer, A. O.; Kim, S.; Lonser, R. R.; Park, D. M.; Ikejiri, B.; Li, J.; Okamoto, H.; Walbridge, immunotherapy for malignant gliomas. Cell Transplant.

18:977–983; 2009. S.; Ryschkewitsch, C.; Major, E.; Oldfield, E. H.; Zhuang,

Z. Identification of tumor precursor cells in the brains of 10. Clement, V.; Sanchez, P.; de Tribolet, N.; Radovanovic,

I.; Altaba, A. R. HEDGEHOG-GLI1 signaling regulated primates with radiation-induced de novo glioblastoma multiforme. Cell Cycle 16:452–456; 2006.

human glioma growth, cancer stem cell renewal and

tu-morigenicity. Curr. Biol. 23:165–172; 2007. 26. McLeod, M.; Hong, M.; Sen, A.; Sadi, D.; Ulalia, R.; Behie, L. A.; Mendez, I. Transplantation of bioreactor-11. Davis, F. G.; Frees, S.; Grutsch, J.; Barlas, S.; Brem, S.

Survival rates in patients with primary malignant brain tu- produced neural stem cells into the rodent brain. Cell Transplant. 15:689–697; 2006.

mors stratified by patient age and tumor histological type:

An analysis base on surveillance, epidemiology, end re- 27. Miqueli, A. D.; Rolff, J.; Lemm, M.; Fichtner, I.; Perez, R.; Montero, E. Radiosensitisation of U87MG brain tu-sults (SEER) data 1973–1991. J. Neurosurg. 88:1–10;

1998 mors by anti-epidermal growth factor receptor monoclonal

antibodies. Br. J. Cancer 100:950–958; 2009. 12. Ehtesham, M.; Mapara, K. Y.; Stevenson, C. B.;

Thomp-son, R. C. CXCR4 mediates the proliferation of glioblas- 28. Oka, N.; Soeda, A.; Inagaki, A.; Onodera, M.; Maruyama, H.; Hara, A.; Kunisada, T.; Mori, H.; Iwama, T. VEGF toma progenitor cells. Cancer Lett. 18:305–312; 2009.

13. Fan, X.; Mastsui, W.; Khaki, L. Notch pathway inhibition promotes tumorigenesis and angiogenesis of human gli-oblastoma stem cells. Biochem. Biophys. Res. Commun. depletes stlike cells and blocks engraftment in

em-bryonal brain tumors. Cancer Res. 66:7445–7452; 2006. 360:553–559; 2007.

29. Oka, N.; Soeda, A.; Noda, S.; Iwama, T. Brain tumor stem 14. Fan, X.; Salford, L. G.; Widegren, B. Glioma stem cell:

Evidence and limitation. Semin. Cancer Biol. 17:214–218; cell from an adenoid glioblastoma multiforme. Neurol. Med. Chir. 49:146–151; 2009.

2007.

15. Gal, H.; Makovitzki, A.; Amariglio, N.; Rechavi, G.; 30. Okamoto, O. K.; Oba-Shinjo, S. M.; Lopes, L.; Nagahashi Marie, S. K. Expression of HOXC9 and E2F2 are up-Ram, Z.; Givol, D. A rapid assay for drug sensitivity of

glioblastoma stem cells. Biochem. Biophys. Res. Com- regulated in CD 133+ cells isolated from human astrocy-tomas and associated with transformation of human astro-mun. 358:908–913; 2007.

16. Gal, H.; Pandi, G.; Kanner, A. A.; Ram, Z.; Lithwick- cytes. Biochim. Biophys. Acta 1769:437–442; 2007. 31. Perez Castillo, A.; Aguilar-Morante, D.; Morales-Garcia, Yanai, G.; Amariglio, N.; Rechavi, G.; Givol, D.

MIR-451 and Imatinib mesylate inhibit tumor growth of gli- J. A.; Dorado, J. Cancer stem cell and brain tumors. Clin. Transl. Oncol. 10:262–267; 2008.

oblastoma stem cells. Biochem. Biophys. Res. Commun.

376:86–90; 2008. 32. Piccirillo, S. G.; Combi, R.; Cajola, L.; Patrizi, A.;

Redaelli, S.; Bentivegna, A.; Baronchelli, S.; Maira, G.; 17. Hide, T.; Takezaki, T.; Nakamura, H.; Kuratsu, J.; Kondo,

T. Brain tumor stem cells as research and treatment tar- Pollo, B.; Mangiola, A.; Dimeco, F.; Dalpra, L.; Vescovi, A. L. Distinct pools of cancer stem-like cells coexist gets. Brain Tumor Pathol. 25:67–72; 2008.

18. Joo, K. M.; Kim, S. Y.; Jin, X.; Song, S. Y.; Kong, D. S.; within human glioblastoma and display different tumori-genicity and independent genomic evolution. Oncogene Lee, J. I.; Jeon, J. W.; Kim, M. H.; Kang, B. G.; Jung, Y.;

Jin, J.; Hong, S. C.; Park, W. Y.; Lee, D. S.; Kim, H.; 28:1807–1811; 2009.

33. Piccirillo, S. G.; Reynolds, B. A.; Zanetti, N.; Lamorte, Nam, D. H. Clinical and biological implications of CD

133-positive and CD 133-negative cells in glioblastomas. G.; Binda, E.; Broggi, G.; Brem, H.; Olivi, A.; Dimeco, F.; Vescovi, A. L. Bone morphogenetic proteins inhibit Lab. Invest. 88:808–815; 2008.

19. Kang, M. K.; Hur, B. I.; Kim, C. H.; Cha, S. H.; Kang, the tumorigenic potential of humane tumor initialing cells. Nature 444:761–765; 2006.

S. K. Potential identify of multi-potential cancer stem-like

subpopulation after radiation of cultured brain glioma. 34. Sakariassen, P. O.; Immervoll, H.; Chekenya, M. Cancer stem cells as mediators of treatment resistance in brain BMC Neurosci. 9:15; 2008.

20. Kang, M. K.; Kang, S. K. Pharmacologic blockade of tumors: Status and controversies. Neoplasm 9:882–892; 2007.

chloride channel synergistically enhances apoptosis of

chemotherapeutic drug-resistant cancer stem cells. Bio- 35. Silber, J.; Lim, D. A.; Petritsch, C.; Persson, A. I.; Mauna-kea, A. K.; Yu, M.; Vandenberg, S. R.; Ginzinger, D. G.; chem. Biophys. Res. Commun. 373:539–544; 2008.

(5)

James, C. D.; Costello, J. F.; Bergers, G.; Weiss, W. A.; 39. Tunici, P.; Irvin, D.; Liu, G.; Yuan, X.; Zhaohui, Z.; Ng, H.; Yu, J. S. Brain tumor stem cells: New targets for clini-Alvarez-Buylla, A.; Hodgson, J. G. miR-124 and miR-137

inhibit proliferation of glioblastoma multiforme cells and cal treatments? Neurosurg. Focus 26:E27; 2006.

40. Wen, P. Y.; Kesari, S. Malignant gliomas in adults. N. induce differentiation of brain tumor stem cells. BMC

Med. 6:14; 2007. Engl. J. Med. 359:492–507; 2008.

41. Yang, X. F. Immunology of stem cells and cancer stem 36. Singh, S. K.; Hawkins, C.; Clarke, I. D.; Squire, J. A.;

Bayani, J.; Hide, T.; Henkelman, R. M.; Cusimano, cells. Cell. Mol. Immunol. 4:161–171; 2007.

42. Yuan, X.; Curtin, J.; Xiong, Y.; Liu, G.; Waschsmann-M. D.; Dirks, P. B. Identification of human brain tumor

initiating cells. Nature 432:396–401; 2004. Hogiu, S.; Farkas, D. L.; Black, K. L.; Yu, J. S. Isolation of cancer stem cells from adult glioblastoma multiforme. 37. Surawicz, T. S.; Davis, F.; Frells, S.; Laws, Jr., E. R.;

Menck, H. R. Brain tumor survival: Results from the Na- Oncogene 23:9392–9400; 2004.

43. Ziche, M.; Jones, J.; Gullino, P. M. Role of prostaglandin tional Cancer Data Base. J. Neurooncol. 40:151–160;

1998. E1 and cooper in angiogenesis. J. Natl. Cancer Inst. 69:

475–482; 1982. 38. Thon, N.; Damianoff, K.; Hegermann, J.; Grau, S.; Krebs,

B.; Schnell, O.; Tonn, J. C.; Goldbrunner, R. Presence of pluripotent CD133+ cell correlates with malignancy of gliomas. Mol. Cell. Neurosci. 43:51–59; 2010.

(6)

參考文獻

相關文件

 Stain (-) because the main body of the tumor consisted of epithelioid or epithelial cells that contained few positive smooth muscle cells.  No clear ductal structures or

Although Central Giant Cell Granuloma (CGCG) is a benign tumor of the jaw and aneurysmal bone cyst seen in children, its aggressive behavior causes extensive loss of hard

Primordial odontogenic tumor (POT) is a benign mixed odontogenic tumor comprised of a loose connective tissue with a similar morphology with dental papilla and exhibiting in

Although DOI, based on its prognostic value on overall disease outcomes, has been incorporated into the current AJCC staging system such that it upstages an OTSCC from T1 to T2 stage

Histological sections showed tumor cells at a distance of 8 mm from the clinical and radiographic limit of the lesion, so the elimination of at least 1 cm of peripheral bone margin

2.滴加蒸餾水或亞甲藍液製成水埋標本 3.觀察 石細胞 (厚壁細胞)及

• 由於細胞代謝、紫外線的電離輻射、不良的生 活習慣 ( 煙、酒、肥胖 ) 、各種感染物所引起 的中性粒細胞、巨噬細胞的激活, 會催化分子 氧發生單價還原產生的 "

SF12144A 張基晟 比較 Nanoplatin 併用 Pemetrexed 和 Cisplatin 併用 Pemetrexed 作為第一線治 療用於非鱗狀細胞性之非小細胞肺癌