4.7 Review

Targeting Cbl-b in cancer immunotherapy

Journal

JOURNAL FOR IMMUNOTHERAPY OF CANCER
Volume 11, Issue 2, Pages -

Publisher

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2022-006007

Keywords

CTLA-4 antigen; drug therapy; combination; immunotherapy; review; therapies; investigational

Ask authors/readers for more resources

Cancer immunotherapy using immune-checkpoint blockade has improved outcomes for many cancer patients, but there are still many who do not benefit or develop resistance. Efforts are being made to target additional inhibitory receptors, costimulatory proteins, and intracellular mediators to overcome anti-PD1 resistance. CD28 and Cbl-b have been identified as important pathways in immune activation and tumor microenvironment modulation. Novel platforms including DNA encoded library screening, allosteric drug targeting, and gene editing have enabled the development of Cbl-b inhibitors. These inhibitors have shown promising results in reversing immunosuppression, stimulating cytotoxic T cell activity, and promoting tumor regression when combined with PD1 blockade.
Cancer immunotherapy with immune-checkpoint blockade has improved the outcomes of patients with various malignancies, yet a majority do not benefit or develop resistance. To address this unmet need, efforts across the field are targeting additional coinhibitory receptors, costimulatory proteins, and intracellular mediators that could prevent or bypass anti-PD1 resistance mechanisms. The CD28 costimulatory pathway is necessary for antigen-specific T cell activation, though prior CD28 agonists did not translate successfully to clinic due to toxicity. Casitas B lymphoma-b (Cbl-b) is a downstream, master regulator of both CD28 and CTLA-4 signaling. This E3 ubiquitin ligase regulates both innate and adaptive immune cells, ultimately promoting an immunosuppressive tumor microenvironment (TME) in the absence of CD28 costimulation. Recent advances in pharmaceutical screening and computational biology have enabled the development of novel platforms to target this once 'undruggable' protein. These platforms include DNA encoded library screening, allosteric drug targeting, small-interfering RNA inhibition, CRISPR genome editing, and adoptive cell therapy. Both genetic knock-out models and Cbl-b inhibitors have been shown to reverse immunosuppression in the TME, stimulate cytotoxic T cell activity, and promote tumor regression, findings augmented with PD1 blockade in experimental models. In translating Cbl-b inhibitors to clinic, we propose specific gene expression profiles that may identify patient populations most likely to benefit. Overall, novel Cbl-b inhibitors provide antigen-specific immune stimulation and are a promising therapeutic tool in the field of immuno-oncology.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available