Skip to main content

Advertisement

Log in

Development of minimally invasive cancer immunotherapy using anti-disialoganglioside GD2 antibody-producing mesenchymal stem cells for a neuroblastoma mouse model

  • Original Article
  • Published:
Pediatric Surgery International Aims and scope Submit manuscript

Abstract

Purpose

Mouse IgG anti-disialoganglioside GD2 antibody-secreting mouse mesenchymal stem cells (anti-GD2-MSCs) were developed, and their anti-tumor effects were validated in an in vivo neuroblastoma mouse model.

Methods

Anti-GD2 antibody constructs were generated, incorporating FLAG-tagged single-chain fragment variables against GD2 fused to a linker sequence, and a fragment of a stationary portion was changed from human IgG to mouse IgG and GFP protein. The construct was lentivirally introduced into mouse MSCs. A syngeneic mouse model was established through the subcutaneous transplantation of a tumor tissue fragment from a TH-MYCN transgenic mouse, and the homing effects of anti-GD2-MSCs were validated by In vivo imaging system imaging. The syngeneic model was divided into three groups according to topical injection materials: anti-GD2-MSCs with IL-2, IL-2, and PBS. The tumors were removed, and natural killer (NK) cells were counted.

Results

Anti-GD2-MSCs showed homing effects in syngeneic models. The growth rate of subcutaneous tumors was significantly suppressed by anti-GD2-MSCs with IL-2 (p < 0.05). Subcutaneous tumor immunostaining showed an increased NK cell infiltration in the same group (p < 0.01).

Conclusion

Anti-GD2-MSCs using mouse IgG showed a homing effect and significant tumor growth suppression in syngeneic models. Anti-GD2-MSC-based cellular immunotherapy could be a novel therapeutic strategy for intractable neuroblastoma.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability statement

The datasets generated and analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Tolbert VP, Matthay KK (2018) Neuroblastoma: clinical and biological approach to risk stratification and treatment. Cell Tissue Res 372:195–209. https://doi.org/10.1007/s00441-018-2821-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mujoo K, Cheresh DA, Yang HM, Reisfeld RA (1987) Disialoganglioside GD2 on human neuroblastoma cells: target antigen for monoclonal antibody-mediated cytolysis and suppression of tumor growth. Cancer Res 47:1098–1104

    CAS  PubMed  Google Scholar 

  3. Schulz G, Cheresh DA, Varki NM, Yu A, Staffileno LK, Reisfeld RA (1984) Detection of ganglioside GD2 in tumor tissues and sera of neuroblastoma patients. Cancer Res 44:5914–5920

    CAS  PubMed  Google Scholar 

  4. Kramer K, Gerald WL, Kushner BH, Larson SM, Hameed M, Cheung NK (1998) Disialoganglioside G(D2) loss following monoclonal antibody therapy is rare in neuroblastoma. Clin Cancer Res 4:2135–2139

    CAS  PubMed  Google Scholar 

  5. Stephenson J (1995) Reengineered monoclonal antibodies step up to the plate in cancer studies. Jama 274:1821–1822. https://doi.org/10.1001/jama.1995.03530230007002

    Article  CAS  PubMed  Google Scholar 

  6. Colucci F, Caligiuri MA, Di Santo JP (2003) What does it take to make a natural killer? Nat Rev Immunol 3:413–425. https://doi.org/10.1038/nri1088

    Article  CAS  PubMed  Google Scholar 

  7. Lammie G, Cheung N, Gerald W, Rosenblum M, Cordoncardo C (1993) Ganglioside gd2 expression in the human nervous-system and in neuroblastomas - an immunohistochemical study. Int J Oncol 3:909–915. https://doi.org/10.3892/ijo.3.5.909

    Article  CAS  PubMed  Google Scholar 

  8. Xiao WH, Yu AL, Sorkin LS (1997) Electrophysiological characteristics of primary afferent fibers after systemic administration of anti-GD2 ganglioside antibody. Pain 69:145–151. https://doi.org/10.1016/s0304-3959(96)03280-0

    Article  CAS  PubMed  Google Scholar 

  9. Fleurence J, Fougeray S, Bahri M, Cochonneau D, Clémenceau B, Paris F, Heczey A, Birklé S (2017) Targeting O-Acetyl-GD2 ganglioside for cancer immunotherapy. J Immunol Res. https://doi.org/10.1155/2017/5604891

    Article  PubMed  PubMed Central  Google Scholar 

  10. Alvarez-Rueda N, Desselle A, Cochonneau D, Chaumette T, Clemenceau B, Leprieur S, Bougras G, Supiot S, Mussini JM, Barbet J, Saba J, Paris F, Aubry J, Birklé S (2011) A monoclonal antibody to O-acetyl-GD2 ganglioside and not to GD2 shows potent anti-tumor activity without peripheral nervous system cross-reactivity. PLoS One 6:e25220

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini F (2008) Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene therapy 15:730–738

    Article  CAS  PubMed  Google Scholar 

  12. Chamberlain G, Fox J, Ashton B, Middleton J (2007) Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 25:2739–2749

    Article  CAS  PubMed  Google Scholar 

  13. Devine SM, Cobbs C, Jennings M, Bartholomew A, Hoffman R (2003) Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 101:2999–3001

    Article  CAS  PubMed  Google Scholar 

  14. Yong RL, Shinojima N, Fueyo J, Gumin J, Vecil GG, Marini FC, Bogler O, Andreeff M, Lang FF (2009) Human bone marrow-derived mesenchymal stem cells for intravascular delivery of oncolytic adenovirus Delta24-RGD to human gliomas. Cancer Res 69:8932–8940

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, Shi Y, Abbruzzese J, Konopleva M, Andreeff M, Marini FC (2010) Mesenchymal stromal cells alone or expressing interferon-beta suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy 12:615–625. https://doi.org/10.3109/14653241003631815

    Article  CAS  PubMed  Google Scholar 

  16. Deng L, Gao X, Fan G, Yang C (2019) Effects of GDNF-Transfected Marrow Stromal Cells on Rats with Intracerebral Hemorrhage. J Stroke Cerebrovasc 28:2555–2562. https://doi.org/10.1016/j.jstrokecerebrovasdis.2019.06.002

    Article  Google Scholar 

  17. Wang J, Zhu L, Chen X, Huang R, Wang S, Dong P (2019) Human bone marrow mesenchymal stem cells functionalized by hybrid baculovirus-adeno-associated viral vectors for targeting hypopharyngeal carcinoma. Stem Cells Dev 28:543–553. https://doi.org/10.1089/scd.2018.0252

    Article  CAS  PubMed  Google Scholar 

  18. Relation T, Yi T, Guess AJ, La Perle K, Otsuru S, Hasgur S, Dominici M, Breuer C, Horwitz EM (2018) Intratumoral delivery of interferonγ-secreting mesenchymal stromal cells repolarizes tumor-associated macrophages and suppresses neuroblastoma proliferation in vivo. Stem Cells 36:915–924

    Article  CAS  PubMed  Google Scholar 

  19. Kimura K, Kishida T, Wakao J, Tanaka T, Higashi M, Fumino S, Aoi S, Furukawa T, Mazda O, Tajiri T (2016) Tumor-homing effect of human mesenchymal stem cells in a TH-MYCN mouse model of neuroblastoma. J Pediatr Surg 51:2068–2073. https://doi.org/10.1016/j.jpedsurg.2016.09.041

    Article  PubMed  Google Scholar 

  20. Maniwa J, Fumino S, Kimura K, Tanaka T, Higashi M, Kishida T, Mazda O, Tajiri T (2019) Novel mesenchymal stem cell delivery system as targeted therapy against neuroblastoma using the TH-MYCN mouse model. J Pediatr Surg 54:2600–2605. https://doi.org/10.1016/j.jpedsurg.2019.08.023

    Article  PubMed  Google Scholar 

  21. Rossig C, Bollard CM, Nuchtern JG, Merchant DA, Brenner MK (2001) Targeting of G(D2)-positive tumor cells by human T lymphocytes engineered to express chimeric T-cell receptor genes. Int J Cancer 94:228–236

    Article  CAS  PubMed  Google Scholar 

  22. Heczey A, Louis CU, Savoldo B, Dakhova O, Durett A, Grilley B, Liu H, Wu MF, Mei Z, Gee A, Mehta B, Zhang H, Mahmood N, Tashiro H, Heslop HE, Dotti G, Rooney CM, Brenner MK (2017) CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther 25:2214–22124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E, Liu H, Wu MF, Gee AP, Mei Z, Rooney CM, Heslop HE, Brenner MK (2011) Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118:6050–6056

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. William A, Weiss KA, Mohapatra Gayatry, Feuerstein Burt G, Michael Bishop J (1997) Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J 16:2985–2995. https://doi.org/10.1093/emboj/16.11.2985

    Article  Google Scholar 

  25. Ari P, Kars M, Meany H, Pestieau S (2018) Treatment of transient peripheral neuropathy during chimeric 14.18 antibody therapy in children with neuroblastoma: a case Series. J Pediatr Hematology/oncology 40:e113–e116. https://doi.org/10.1097/MPH.0000000000000889

    Article  Google Scholar 

  26. Kushner BH, Cheung NK (1989) GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood 73:1936–1941

    Article  CAS  PubMed  Google Scholar 

  27. Ladenstein R, Pötschger U, Valteau-Couanet D, Luksch R, Castel V, Yaniv I, Laureys G, Brock P, Michon JM, Owens C, Trahair T, Chan GCF, Ruud E, Schroeder H, Beck Popovic M, Schreier G, Loibner H, Ambros P, Holmes K, Castellani MR, Gaze MN, Garaventa A, Pearson ADJ, Lode HN (2018) Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): a multicentre, randomised, phase 3 trial. Lancet Oncol 19:1617–1629. https://doi.org/10.1016/S1470-2045(18)30578-3

    Article  CAS  PubMed  Google Scholar 

  28. Munn DH, Cheung NK (1989) Antibody-dependent antitumor cytotoxicity by human monocytes cultured with recombinant macrophage colony-stimulating factor. Induction of efficient antibody-mediated antitumor cytotoxicity not detected by isotope release assays. J Exp Med 170:511–526

    Article  CAS  PubMed  Google Scholar 

  29. Kroesen M, Brok IC, Reijnen D, van Hout-Kuijer MA, Zeelenberg IS, Den Brok MH, Hoogerbrugge PM, Adema GJ (2015) Intra-adrenal murine TH-MYCN neuroblastoma tumors grow more aggressive and exhibit a distinct tumor microenvironment relative to their subcutaneous equivalents. Cancer Immunol Immunother CII 64:563–572

    Article  CAS  PubMed  Google Scholar 

  30. Li J, Qian W, Qin T, Xiao Y, Cheng L, Cao J, Chen X, Ma Q, Wu Z (2019) Mouse-derived allografts: a complementary model to the kpc mice on researching pancreatic cancer in vivo. Comput Struct Biotechnol J 17:498–506

    Article  PubMed  PubMed Central  Google Scholar 

  31. Michiel Kroesen SN, Ansems Marleen, Wassink Melissa, Orentas Rimas J, Boon Louis, den Brok Martijn H, Hoogerbrugge Peter M, Adema Gosse J (2014) A transplantable TH-MYCN transgenic tumor model in C57Bl/6 mice for preclinical immunological studies in neuroblastoma. Int J Cancer 134:1335–1345. https://doi.org/10.1002/ijc.28463

    Article  CAS  PubMed  Google Scholar 

  32. Gonzalez ME, Martin EE, Anwar T, Arellano-Garcia C, Medhora N, Lama A, Chen YC, Tanager KS, Yoon E, Kidwell KM, Ge C, Franceschi RT, Kleer CG (2017) Mesenchymal stem cell-induced DDR2 mediates stromal-breast cancer interactions and metastasis growth. Cell Reports 18:1215–1228

    Article  CAS  PubMed  Google Scholar 

  33. Błogowski W, Bodnarczuk T, Starzyńska T (2016) Concise review: pancreatic cancer and bone marrow-derived stem cells. Stem Cells Transl Med 57:938–945

    Article  Google Scholar 

Download references

Acknowledgements

We are grateful to the member of the Department of Immunology, Kyoto Prefectural University of Medicine for useful comments. The English used in this manuscript was reviewed by enago.

Funding

This work was supported by a Grant-in-Aid for Exploratory Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan (MEXT KAKENHI grant number 19H03719), and by the Practical Research for Innovative Cancer Control from the Japan Agency for Medical Research and Development, AMED (grant number 20ck0106609h0001), and Funding for Research Projects, Children’s Cancer Association of Japan.

Author information

Authors and Affiliations

Authors

Contributions

We are grateful to Dr. S. Yagyu for collaboration on many phases of this work. We thank M. Higashi and M. Iguchi for technical assistance with the experiments. Finally, we are grateful to the member of the Department of Immunology, Kyoto Prefectural University of Medicine for useful comments. All authors reviewed the manuscript.

Corresponding author

Correspondence to Kosuke Kambe.

Ethics declarations

Competing interests

The authors declare no competing interests.

Conflict of interest

The authors declare no conflicts of interest in association with the present study.

Ethical approval

All experimental animal procedures and protocols conformed to the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Committee for Animal Research of Kyoto Prefectural University of Medicine.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kambe, K., Iguchi, M., Higashi, M. et al. Development of minimally invasive cancer immunotherapy using anti-disialoganglioside GD2 antibody-producing mesenchymal stem cells for a neuroblastoma mouse model. Pediatr Surg Int 39, 43 (2023). https://doi.org/10.1007/s00383-022-05310-z

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00383-022-05310-z

Keywords

Navigation