Antitumor effects of the GM3(Neu5Gc) ganglioside-specific humanized antibody 14F7hT against Cmah-transfected cancer cells.

Dorvignit, Denise; Frias Boligan, Kayluz; Relova-Hernández, Ernesto; Clavell, Marilyn; López, Armando; Labrada, Mayrel; Simon, Hans-Uwe; López-Requena, Alejandro; Mesa, Circe; von Gunten, Stephan (2019). Antitumor effects of the GM3(Neu5Gc) ganglioside-specific humanized antibody 14F7hT against Cmah-transfected cancer cells. Scientific Reports, 9(1), p. 9921. Nature Publishing Group 10.1038/s41598-019-46148-1

[img]
Preview
Text
Simon_Von Gunten_Antitumor effects of the GM3(Neu5Gc) ganglioside-specific humanized antibody 14F7hT.pdf - Published Version
Available under License Creative Commons: Attribution (CC-BY).

Download (2MB) | Preview

The GM3(Neu5Gc) ganglioside represents a tumor-specific antigen that is considered a promising target for cancer immunotherapy. We previously demonstrated that the humanized antibody 14F7hT, specific for this ganglioside, exhibited significant antitumor effects in preclinical hematological tumor models. As this antibody recognizes human tumor tissues from several origins, we addressed its potential effect on different tumor types. The use of cell lines for testing GM3(Neu5Gc)-targeting strategies, in particular for human malignancies, is complicated by the absence in humans of functional cytidine monophospho-N-acetyl-neuraminic acid hydroxylase (CMAH), the enzyme required for Neu5Gc sialic acid biosynthesis. Quantitative flow cytometry revealed the absence of surface GM3(Neu5Gc) in several human but also mouse cell lines, in the last case due to low expression of the enzyme. Hypoxia-induced expression of this ganglioside on human SKOV3 cells was observed upon culture in Neu5Gc-containing medium without evidence for CMAH-independent biosynthesis. However, only transfection of the mouse Cmah gene into human SKOV3 and mouse 3LL cells induced a stable expression of GM3(Neu5Gc) on the cancer cell surface, resulting in effective models to evaluate the antitumor responses by 14F7hT in vitro and in vivo. This antibody exerted antibody-dependent cell-mediated cytotoxicity (ADCC) and in vivo antitumor effects on these Cmah-transfected non-hematological tumors from both mouse and human origin. These results contribute to validate GM3(Neu5Gc) as a relevant target for cancer immunotherapy and reinforces the value of 14F7hT as a novel anti-cancer drug.

Item Type:

Journal Article (Original Article)

Division/Institute:

04 Faculty of Medicine > Pre-clinic Human Medicine > Institute of Pharmacology

UniBE Contributor:

Frias Boligan, Kayluz, Simon, Hans-Uwe, von Gunten, Stephan

Subjects:

600 Technology > 610 Medicine & health

ISSN:

2045-2322

Publisher:

Nature Publishing Group

Language:

English

Submitter:

Celine Joray

Date Deposited:

26 Aug 2019 15:55

Last Modified:

05 Dec 2022 15:30

Publisher DOI:

10.1038/s41598-019-46148-1

PubMed ID:

31289278

BORIS DOI:

10.7892/boris.132369

URI:

https://boris.unibe.ch/id/eprint/132369

Actions (login required)

Edit item Edit item
Provide Feedback