Anat Cell Biol.  2015 Mar;48(1):44-53. 10.5115/acb.2015.48.1.44.

Glioblastoma specific antigens, GD2 and CD90, are not involved in cancer stemness

Affiliations
  • 1Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, Korea. kmjoo@skku.edu
  • 2Center for Molecular Medicine, Samsung Biomedical Research Institute, Seoul, Korea.
  • 3Department of Anatomy and Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
  • 4Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.

Abstract

Glioblastoma multiforme (GBM) is the most malignant World Health Organization grade IV brain tumor. GBM patients have a poor prognosis because of its resistance to standard therapies, such as chemotherapy and radiation. Since stem-like cells have been associated with the treatment resistance of GBM, novel therapies targeting the cancer stem cell (CSC) population is critically required. However, GBM CSCs share molecular and functional characteristics with normal neural stem cells (NSCs). To elucidate differential therapeutic targets of GBM CSCs, we compared surface markers of GBM CSCs with adult human NSCs and found that GD2 and CD90 were specifically overexpressed in GBM CSCs. We further tested whether the GBM CSC specific markers are associated with the cancer stemness using primarily cultured patient-derived GBM cells. However, results consistently indicated that GBM cells with or without GD2 and CD90 had similar in vitro sphere formation capacity, a functional characteristics of CSCs. Therefore, GD2 and CD90, GBM specific surface markers, might not be used as specific therapeutic targets for GBM CSCs, although they could have other clinical utilities.

Keyword

Glioblastoma multiforme; Cancer stem cell; Therapeutic marker; GD2; CD90

MeSH Terms

Adult
Brain Neoplasms
Drug Therapy
Glioblastoma*
Humans
Neoplastic Stem Cells
Neural Stem Cells
Prognosis
World Health Organization

Figure

  • Fig. 1 Sox2 and nestin were highly regulated in both normal neural stem cells (NSCs) and glioblastoma multiforme (GBM) cells. 464T and 532T GBM cells, as well as 682 and 779 normal NSCs were stained by anti-Sox2 (upper, green) and anti-nestin (lower, green) antibodies for immunocytochemistry. Nuclei of all cells were stained by DAPI (blue).

  • Fig. 2 Glioblastoma multiforme cells unlike neural stem cells generated tumor mass in orthotopic brain xenograft models. Hematoxylin and eosin (H&E) and immunohistochemistry staining for proliferating cell nuclear antigen (PCNA) expression were performed in representative whole brains of the orthotopic xenograft. Bar chart summary of average staining intensity of PCNA in 3 randomly selected hot spot regions (*P<0.05).

  • Fig. 3 Cell surface markers of 242 kinds were analyzed in normal neural stem cells (NSCs) and glioblastoma multiforme (GBM) cells. 464T (B) and 532T (D) GBM cells, as well as 682 (A) and 779 (C) normal NSCs were stained by BD Lyoplate Human Cell Surface Marker Screening Panel containg 242 antibodies. All cells were counted by flow cytometer BD Caliber & LSR II. Data were analyzed by BD FACSDive software.

  • Fig. 4 Positive expression of GD2 and CD90 were significantly increased in glioblastoma multiforme (GBM) cells than neural stem cells (NSCs). BD Lyoplate Human Cell Surface Marker Screening showing high regulated markers of >50% were selected in GBM cells, and analyzed with the expressed changes in NSCs. Data was expressed by the fold changes of each value in GBM cells as compared with NSCs for the selected 11 markers.

  • Fig. 5 GBM cells with high levels of GD2 or CD90 protein do not have characteristics of cancer stemness. 464T cells were stained by anti-GD2 (A) and anti-CD90 (B) antibodies. The high-regulated and low-regulated cells of GD2 or CD90, were sorted by fluorescence activated cell sorting analysis. The sorted cells and parent cells were tested for the ability of neurosphere formation according to the expressed levels of GD2 (C) or CD90 (D) by the limiting dilution assay.


Reference

1. Kohler BA, Ward E, McCarthy BJ, Schymura MJ, Ries LA, Eheman C, Jemal A, Anderson RN, Ajani UA, Edwards BK. Annual report to the nation on the status of cancer, 1975-2007, featuring tumors of the brain and other nervous system. J Natl Cancer Inst. 2011; 103:714–736.
2. Mangum R, Nakano I. Glioma stem cells and their therapy resistance. J Carcinog Mutagen. 2011; S1:002.
3. Kang MK, Kang SK. Tumorigenesis of chemotherapeutic drug-resistant cancer stem-like cells in brain glioma. Stem Cells Dev. 2007; 16:837–847.
4. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006; 444:756–760.
5. Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN, Mikkelsen T, Brodie C. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int J Cancer. 2009; 125:717–722.
6. McKay R. Stem cells in the central nervous system. Science. 1997; 276:66–71.
7. Temple S. The development of neural stem cells. Nature. 2001; 414:112–117.
8. Goffart N, Kroonen J, Rogister B. Glioblastoma-initiating cells: relationship with neural stem cells and the micro-environment. Cancers (Basel). 2013; 5:1049–1071.
9. Stiles CD, Rowitch DH. Glioma stem cells: a midterm exam. Neuron. 2008; 58:832–846.
10. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001; 414:105–111.
11. Singh SK, Clarke ID, Hide T, Dirks PB. Cancer stem cells in nervous system tumors. Oncogene. 2004; 23:7267–7273.
12. Nicolis SK. Cancer stem cells and "stemness" genes in neuro-oncology. Neurobiol Dis. 2007; 25:217–229.
13. Han ME, Oh SO. Gastric stem cells and gastric cancer stem cells. Anat Cell Biol. 2013; 46:8–18.
14. Ignatova TN, Kukekov VG, Laywell ED, Suslov ON, Vrionis FD, Steindler DA. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro. Glia. 2002; 39:193–206.
15. Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI. Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A. 2003; 100:15178–15183.
16. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003; 63:5821–5828.
17. Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004; 64:7011–7021.
18. Tunici P, Bissola L, Lualdi E, Pollo B, Cajola L, Broggi G, Sozzi G, Finocchiaro G. Genetic alterations and in vivo tumorigenicity of neurospheres derived from an adult glioblastoma. Mol Cancer. 2004; 3:25.
19. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004; 432:396–401.
20. Joo KM, Kim SY, Jin X, Song SY, Kong DS, Lee JI, Jeon JW, Kim MH, Kang BG, Jung Y, Jin J, Hong SC, Park WY, Lee DS, Kim H, Nam DH. Clinical and biological implications of CD133-positive and CD133-negative cells in glioblastomas. Lab Invest. 2008; 88:808–815.
21. Joo KM, Kang BG, Yeon JY, Cho YJ, An JY, Song HS, Won JH, Kim SJ, Hong SC, Nam DH. Experimental and clinical factors influencing long-term stable in vitro expansion of multipotent neural cells from human adult temporal lobes. Exp Neurol. 2013; 240:168–177.
22. Kim KH, Seol HJ, Kim EH, Rheey J, Jin HJ, Lee Y, Joo KM, Lee J, Nam DH. Wnt/beta-catenin signaling is a key downstream mediator of MET signaling in glioblastoma stem cells. Neuro Oncol. 2013; 15:161–171.
23. Bez A, Corsini E, Curti D, Biggiogera M, Colombo A, Nicosia RF, Pagano SF, Parati EA. Neurosphere and neurosphere-forming cells: morphological and ultrastructural characterization. Brain Res. 2003; 993:18–29.
24. Shi X, Gipp J, Bushman W. Anchorage-independent culture maintains prostate stem cells. Dev Biol. 2007; 312:396–406.
25. Legler JM, Ries LA, Smith MA, Warren JL, Heineman EF, Kaplan RS, Linet MS. Cancer surveillance series [corrected]: brain and other central nervous system cancers: recent trends in incidence and mortality. J Natl Cancer Inst. 1999; 91:1382–1390.
26. Ritter G, Livingston PO. Ganglioside antigens expressed by human cancer cells. Semin Cancer Biol. 1991; 2:401–409.
27. Chang HR, Cordon-Cardo C, Houghton AN, Cheung NK, Brennan MF. Expression of disialogangliosides GD2 and GD3 on human soft tissue sarcomas. Cancer. 1992; 70:633–638.
28. Heiner JP, Miraldi F, Kallick S, Makley J, Neely J, Smith-Mensah WH, Cheung NK. Localization of GD2-specific monoclonal antibody 3F8 in human osteosarcoma. Cancer Res. 1987; 47:5377–5381.
29. Cheung NK, Saarinen UM, Neely JE, Landmeier B, Donovan D, Coccia PF. Monoclonal antibodies to a glycolipid antigen on human neuroblastoma cells. Cancer Res. 1985; 45:2642–2649.
30. Mujoo K, Cheresh DA, Yang HM, Reisfeld RA. Disialoganglioside GD2 on human neuroblastoma cells: target antigen for monoclonal antibody-mediated cytolysis and suppression of tumor growth. Cancer Res. 1987; 47:1098–1104.
31. Wu ZL, Schwartz E, Seeger R, Ladisch S. Expression of GD2 ganglioside by untreated primary human neuroblastomas. Cancer Res. 1986; 46:440–443.
32. Tsuchida T, Saxton RE, Morton DL, Irie RF. Gangliosides of human melanoma. J Natl Cancer Inst. 1987; 78:45–54.
33. Battula VL, Shi Y, Evans KW, Wang RY, Spaeth EL, Jacamo RO, Guerra R, Sahin AA, Marini FC, Hortobagyi G, Mani SA, Andreeff M. Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis. J Clin Invest. 2012; 122:2066–2078.
34. Yu AL, Uttenreuther-Fischer MM, Huang CS, Tsui CC, Gillies SD, Reisfeld RA, Kung FH. Phase I trial of a human-mouse chimeric anti-disialoganglioside monoclonal antibody ch14.18 in patients with refractory neuroblastoma and osteosarcoma. J Clin Oncol. 1998; 16:2169–2180.
35. Zeytin HE, Tripathi PK, Bhattacharya-Chatterjee M, Foon KA, Chatterjee SK. Construction and characterization of DNA vaccines encoding the single-chain variable fragment of the anti-idiotype antibody 1A7 mimicking the tumor-associated antigen disialoganglioside GD2. Cancer Gene Ther. 2000; 7:1426–1436.
36. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, Shimada H, Grupp SA, Seeger R, Reynolds CP, Buxton A, Reisfeld RA, Gillies SD, Cohn SL, Maris JM, Sondel PM. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010; 363:1324–1334.
37. Simon T, Hero B, Faldum A, Handgretinger R, Schrappe M, Niethammer D, Berthold F. Infants with stage 4 neuroblastoma: the impact of the chimeric anti-GD2-antibody ch14.18 consolidation therapy. Klin Padiatr. 2005; 217:147–152.
38. Dennis JE, Esterly K, Awadallah A, Parrish CR, Poynter GM, Goltry KL. Clinical-scale expansion of a mixed population of bone-marrow-derived stem and progenitor cells for potential use in bone-tissue regeneration. Stem Cells. 2007; 25:2575–2582.
39. Baum CM, Weissman IL, Tsukamoto AS, Buckle AM, Peault B. Isolation of a candidate human hematopoietic stem-cell population. Proc Natl Acad Sci U S A. 1992; 89:2804–2808.
40. Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PW, Lam CT, Poon RT, Fan ST. Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell. 2008; 13:153–166.
41. Bayati V, Hashemitabar M, Gazor R, Nejatbakhsh R, Bijannejad D. Expression of surface markers and myogenic potential of rat bone marrow- and adipose-derived stem cells: a comparative study. Anat Cell Biol. 2013; 46:113–121.
42. He J, Liu Y, Zhu T, Zhu J, Dimeco F, Vescovi AL, Heth JA, Muraszko KM, Fan X, Lubman DM. CD90 is identified as a candidate marker for cancer stem cells in primary high-grade gliomas using tissue microarrays. Mol Cell Proteomics. 2012; 11:M111.010744.
43. Parry PV, Engh JA. CD90 is identified as a marker for cancer stem cells in high-grade gliomas using tissue microarrays. Neurosurgery. 2012; 70:N23–N24.
Full Text Links
  • ACB
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr