» Articles » PMID: 35605191

NOTCH1 Signaling During CD4+ T-cell Activation Alters Transcription Factor Networks and Enhances Antigen Responsiveness

Overview
Journal Blood
Publisher Elsevier
Specialty Hematology
Date 2022 May 23
PMID 35605191
Authors
Affiliations
Soon will be listed here.
Abstract

Adoptive transfer of T cells expressing chimeric antigen receptors (CAR-T) effectively treats refractory hematologic malignancies in a subset of patients but can be limited by poor T-cell expansion and persistence in vivo. Less differentiated T-cell states correlate with the capacity of CAR-T to proliferate and mediate antitumor responses, and interventions that limit tumor-specific T-cell differentiation during ex vivo manufacturing enhance efficacy. NOTCH signaling is involved in fate decisions across diverse cell lineages and in memory CD8+ T cells was reported to upregulate the transcription factor FOXM1, attenuate differentiation, and enhance proliferation and antitumor efficacy in vivo. Here, we used a cell-free culture system to provide an agonistic NOTCH1 signal during naïve CD4+ T-cell activation and CAR-T production and studied the effects on differentiation, transcription factor expression, cytokine production, and responses to tumor. NOTCH1 agonism efficiently induced a stem cell memory phenotype in CAR-T derived from naïve but not memory CD4+ T cells and upregulated expression of AhR and c-MAF, driving heightened production of interleukin-22, interleukin-10, and granzyme B. NOTCH1-agonized CD4+ CAR-T demonstrated enhanced antigen responsiveness and proliferated to strikingly higher frequencies in mice bearing human lymphoma xenografts. NOTCH1-agonized CD4+ CAR-T also provided superior help to cotransferred CD8+ CAR-T, driving improved expansion and curative antitumor responses in vivo at low CAR-T doses. Our data expand the mechanisms by which NOTCH can shape CD4+ T-cell behavior and demonstrate that activating NOTCH1 signaling during genetic modification ex vivo is a potential strategy for enhancing the function of T cells engineered with tumor-targeting receptors.

Citing Articles

A multiomic atlas identifies a treatment-resistant, bone marrow progenitor-like cell population in T cell acute lymphoblastic leukemia.

Xu J, Chen C, Sussman J, Yoshimura S, Vincent T, Polonen P Nat Cancer. 2024; 6(1):102-122.

PMID: 39587259 PMC: 11779640. DOI: 10.1038/s43018-024-00863-5.


Optimizing CAR-T cell therapy for solid tumors: current challenges and potential strategies.

Ai K, Liu B, Chen X, Huang C, Yang L, Zhang W J Hematol Oncol. 2024; 17(1):105.

PMID: 39501358 PMC: 11539560. DOI: 10.1186/s13045-024-01625-7.


Engineering synthetic agonists for targeted activation of Notch signaling.

Perez D, Antfolk D, Bustos X, Medina E, Chang S, Ramadan A bioRxiv. 2024; .

PMID: 39149362 PMC: 11326249. DOI: 10.1101/2024.08.06.606897.


SARS-CoV-2 inflammation durably imprints memory CD4 T cells.

Gray-Gaillard S, Solis S, Chen H, Monteiro C, Ciabattoni G, Samanovic M Sci Immunol. 2024; 9(96):eadj8526.

PMID: 38905326 PMC: 11824880. DOI: 10.1126/sciimmunol.adj8526.


Harnessing the cytotoxic granule exocytosis to augment the efficacy of T-cell-engaging bispecific antibody therapy.

Casey M, Lee C, Hoyte S, Johnston R, Kwok W, Law S Haematologica. 2024; 109(7):2131-2143.

PMID: 38268493 PMC: 11215359. DOI: 10.3324/haematol.2023.284435.


References
1.
Amsen D, Helbig C, Backer R . Notch in T Cell Differentiation: All Things Considered. Trends Immunol. 2015; 36(12):802-814. DOI: 10.1016/j.it.2015.10.007. View

2.
Kelliher M, Roderick J . NOTCH Signaling in T-Cell-Mediated Anti-Tumor Immunity and T-Cell-Based Immunotherapies. Front Immunol. 2019; 9:1718. PMC: 6109642. DOI: 10.3389/fimmu.2018.01718. View

3.
Bailis W, Yashiro-Ohtani Y, Fang T, Hatton R, Weaver C, Artis D . Notch simultaneously orchestrates multiple helper T cell programs independently of cytokine signals. Immunity. 2013; 39(1):148-59. PMC: 3762693. DOI: 10.1016/j.immuni.2013.07.006. View

4.
Radens C, Blake D, Jewell P, Barash Y, Lynch K . Meta-analysis of transcriptomic variation in T-cell populations reveals both variable and consistent signatures of gene expression and splicing. RNA. 2020; 26(10):1320-1333. PMC: 7491319. DOI: 10.1261/rna.075929.120. View

5.
Takeshita M, Suzuki K, Kassai Y, Takiguchi M, Nakayama Y, Otomo Y . Polarization diversity of human CD4+ stem cell memory T cells. Clin Immunol. 2015; 159(1):107-17. DOI: 10.1016/j.clim.2015.04.010. View