Insulin-producing cells derived from induced pluripotent stem cells (iPSCs) are promising candidates for beta cell replacement in type 1 diabetes. However, the risk of teratoma formation due to residual undifferentiated iPSCs contaminating the differentiated cells is still a critical concern for clinical application. Here, we hypothesized that pretreatment of iPSC-derived insulin-producing cells with an anti-CD30 antibody-drug conjugate could prevent in vivo teratoma formation by selectively killing residual undifferentiated cells. CD30 is expressed in all human iPSCs clones tested by flow cytometry (n = 7) but not in iPSC-derived beta cells (i beta s). Concordantly, anti-CD30 treatment in vitro for 24 h induced a dose-dependent cell death (up to 90%) in human iPSCs while it did not kill i beta s nor had an impact on i beta identity and function, including capacity to secrete insulin in response to stimuli. In a model of teratoma assay associated with i beta transplantation, the pretreatment of cells with anti-CD30 for 24 h before the implantation into NOD-SCID mice completely eliminated teratoma development (0/10 vs. 8/8, p < 0.01). These findings suggest that short-term in vitro treatment with clinical-grade anti-CD30, targeting residual undifferentiated cells, eliminates the tumorigenicity of iPSC-derived beta cells, potentially providing enhanced safety for iPSC-based beta cell replacement therapy in clinical scenarios.

Treating iPSC-Derived β Cells with an Anti-CD30 Antibody-Drug Conjugate Eliminates the Risk of Teratoma Development upon Transplantation / Pellegrini, Silvia; Zamarian, Valentina; Landi, Elisa; Cospito, Alessandro; Lombardo, Marta Tiffany; Manenti, Fabio; Citro, Antonio; Schiavo Lena, Marco; Piemonti, Lorenzo; Sordi, Valeria. - In: INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES. - ISSN 1422-0067. - 23:17(2022), p. 9699. [10.3390/ijms23179699]

Treating iPSC-Derived β Cells with an Anti-CD30 Antibody-Drug Conjugate Eliminates the Risk of Teratoma Development upon Transplantation

Piemonti, Lorenzo;
2022-01-01

Abstract

Insulin-producing cells derived from induced pluripotent stem cells (iPSCs) are promising candidates for beta cell replacement in type 1 diabetes. However, the risk of teratoma formation due to residual undifferentiated iPSCs contaminating the differentiated cells is still a critical concern for clinical application. Here, we hypothesized that pretreatment of iPSC-derived insulin-producing cells with an anti-CD30 antibody-drug conjugate could prevent in vivo teratoma formation by selectively killing residual undifferentiated cells. CD30 is expressed in all human iPSCs clones tested by flow cytometry (n = 7) but not in iPSC-derived beta cells (i beta s). Concordantly, anti-CD30 treatment in vitro for 24 h induced a dose-dependent cell death (up to 90%) in human iPSCs while it did not kill i beta s nor had an impact on i beta identity and function, including capacity to secrete insulin in response to stimuli. In a model of teratoma assay associated with i beta transplantation, the pretreatment of cells with anti-CD30 for 24 h before the implantation into NOD-SCID mice completely eliminated teratoma development (0/10 vs. 8/8, p < 0.01). These findings suggest that short-term in vitro treatment with clinical-grade anti-CD30, targeting residual undifferentiated cells, eliminates the tumorigenicity of iPSC-derived beta cells, potentially providing enhanced safety for iPSC-based beta cell replacement therapy in clinical scenarios.
2022
CD30
beta cells
cell therapy
induced pluripotent stem cells
teratoma
type 1 diabetes
Animals
Cell Differentiation
Humans
Insulin
Ki-1 Antigen
Mice
Mice, Inbred NOD
Mice, SCID
Antineoplastic Agents
Immunoconjugates
Induced Pluripotent Stem Cells
Teratoma
File in questo prodotto:
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/20.500.11768/131954
Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus 2
  • ???jsp.display-item.citation.isi??? 2
social impact