@article {Semmriche003488, author = {Monika Semmrich and Jean-Baptiste Marchand and Laetitia Fend and Matilda Rehn and Christelle Remy and Petra Holmkvist and Nathalie Silvestre and Carolin Svensson and Patricia Kleinpeter and Jules Deforges and Fred Junghus and Kirstie L Cleary and Mimoza Bod{\'e}n and Linda M{\r a}rtensson and Johann Foloppe and Ingrid Teige and Eric Qu{\'e}m{\'e}neur and Bj{\"o}rn Frend{\'e}us}, title = {Vectorized Treg-depleting αCTLA-4 elicits antigen cross-presentation and CD8+ T cell immunity to reject {\textquoteleft}cold{\textquoteright} tumors}, volume = {10}, number = {1}, elocation-id = {e003488}, year = {2022}, doi = {10.1136/jitc-2021-003488}, publisher = {BMJ Specialist Journals}, abstract = {Background Immune checkpoint blockade (ICB) is a clinically proven concept to treat cancer. Still, a majority of patients with cancer including those with poorly immune infiltrated {\textquoteleft}cold{\textquoteright} tumors are resistant to currently available ICB therapies. Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is one of few clinically validated targets for ICB, but toxicities linked to efficacy in approved αCTLA-4 regimens have restricted their use and precluded full therapeutic dosing. At a mechanistic level, accumulating preclinical and clinical data indicate dual mechanisms for αCTLA-4; ICB and regulatory T cell (Treg) depletion are both thought to contribute efficacy and toxicity in available, systemic, αCTLA-4 regimens. Accordingly, strategies to deliver highly effective, yet safe αCTLA-4 therapies have been lacking. Here we assess and identify spatially restricted exposure to a novel strongly Treg-depleting, checkpoint-blocking, vectorized αCTLA-4, as a highly efficacious and potentially safe strategy to target CTLA-4.Methods A novel human IgG1 CTLA-4 antibody (4-E03) was identified using function-first screening for monoclonal antibodies (mAbs) and targets associated with superior Treg-depleting activity. A tumor-selective oncolytic vaccinia vector was then engineered to encode this novel, strongly Treg-depleting, checkpoint-blocking, αCTLA-4 antibody or a matching surrogate antibody, and Granulocyte-macrophage colony-stimulating factor (GM-CSF) (VVGM-αCTLA-4).Results The identified 4-E03 antibody showed significantly stronger Treg depletion, but equipotent checkpoint blockade, compared with clinically validated αCTLA-4 ipilimumab against CTLA-4-expressing Treg cells in a humanized mouse model in vivo. Intratumoral administration of VVGM-αCTLA-4 achieved tumor-restricted CTLA-4 receptor saturation and Treg depletion, which elicited antigen cross-presentation and stronger systemic expansion of tumor-specific CD8+ T cells and antitumor immunity compared with systemic αCTLA-4 antibody therapy. Efficacy correlated with FcγR-mediated intratumoral Treg depletion. Remarkably, in a clinically relevant mouse model resistant to systemic ICB, intratumoral VVGM-αCTLA-4 synergized with αPD-1 to reject cold tumors.Conclusion Our findings demonstrate in vivo proof of concept for spatial restriction of Treg depletion-optimized immune checkpoint blocking, vectorized αCTLA-4 as a highly effective and safe strategy to target CTLA-4. A clinical trial evaluating intratumoral VVGM-αhCTLA-4 (BT-001) alone and in combination with αPD-1 in metastatic or advanced solid tumors has commenced.The datasets generated during and/or analyzed during this current study have been deposited or are available from the corresponding author on reasonable request. The assigned accession number for RNAseq data reported in this paper is GEO: GSE176052.}, URL = {https://jitc.bmj.com/content/10/1/e003488}, eprint = {https://jitc.bmj.com/content/10/1/e003488.full.pdf}, journal = {Journal for ImmunoTherapy of Cancer} }