Abstract 4885: Intratumoral injection of G100 (TLR4 agonist glycopyranosyl lipid A) modulates tumor microenvironment and induces CD8 T cell-dependent, systemic anti-tumor immunity

Author(s):  
Hailing Lu ◽  
Jessica Hewitt ◽  
Jan ter Meulen.
2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A894-A894
Author(s):  
Haixing Kehoe ◽  
Alexandre Iannello ◽  
Keith Cheung ◽  
Bret Peterson ◽  
Marie Marotel ◽  
...  

BackgroundIn a metastatic setting, systemically-administered therapies that overcome the immunosuppressive tumor microenvironment to promote T-cell recruitment and T-cell cytolytic function will be required to elicit durable anti-tumor immunity. To accomplish this, the STACT (S. Typhimurium-Attenuated Cancer Therapy) platform was developed. STACT is a live bacterial product that has been highly modified using precision genome editing for the following properties: (1) enhanced tolerability after IV dosing, (2) tumor-specific enrichment, (3) phagocytosis by tumor-resident antigen-presenting cells (APCs) with a lack of epithelial cell infectivity, (4) multiplexed genetic cargo delivery, and (5) attenuation of bacterial pathways that impair CD8+ T-cell function. An extensive screening campaign was performed to identify ideal encoded immunomodulatory payload combinations delivered by STACT for efficacy against T-cell excluded tumors.MethodsChromosomal edits to the STACT platform strain were made using PCR. A panel of immunomodulatory proteins, including cytokines, type I interferon (IFN)-inducing factors, co-stimulatory receptors, checkpoint antibodies and TGFβR-Fc decoys were tested for combinatorial potency using STACT. An engineered STING (eSTING) was designed through an extensive protein engineering campaign to identify optimal variants. Combinations were evaluated in primary human APCs using in vitro functional assays, where STACT IL-15Rα-IL-15 (IL-15) + eSTING (ACTM-838) emerged as a lead candidate. ACTM-838 was then evaluated in multiple murine tumor models for therapeutic efficacy and mechanism, as well as tolerability in rodents and primates after systemic administration.ResultsCombinatorial target profiling led to the discovery of ACTM-838, a STACT encoding IL-15 + eSTING. In vitro, ACTM-838 payloads synergistically produced high levels of type I IFN and T-cell recruitment and activation factors from primary human APCs. In vivo, ACTM-838 demonstrated a high degree of complete tumor responses that were entirely CD8+ T-cell dependent. In an autochthonous breast cancer model that lacks any significant lymphocyte infiltrate, ACTM-838 was able to uniformly enrich in each spontaneous lesion to high levels after IV dosing and resulted in significant CD8+ T-cell infiltration. In primates, ACTM-838 was well-tolerated, rapidly cleared, and elicited minimal cytokine response after IV dosing.ConclusionsACTM-838 is a highly attenuated, precision genome-engineered bacterial immunotherapy that delivers IL-15 + eSTING to phagocytic APCs of the solid tumor microenvironment after systemic administration. In preclinical studies, ACTM-838 promotes CD8+ T-cell mediated tumor clearance in T-cell excluded tumors and elicits durable anti-tumor immunity, and is well tolerated in primates. Based on these data, ACTM-838 was nominated for clinical development and has entered cGMP manufacturing and IND-enabling studies.Ethics ApprovalAll animals were used according to protocols approved by an Institutional Animal Care and Use Committee and maintained in specific pathogen-free conditions in a barrier facility.


Cancers ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 515
Author(s):  
Sungmin Jung ◽  
Jea-Hyun Baek

T cell factor 1 (TCF1) is a transcription factor that has been highlighted to play a critical role in the promotion of T cell proliferation and maintenance of cell stemness in the embryonic and CD8+ T cell populations. The regulatory nature of TCF1 in CD8+ T cells is of great significance, especially within the context of T cell exhaustion, which is linked to the tumor and viral escape in pathological contexts. Indeed, inhibitory signals, such as programmed cell death 1 (PD-1) and cytotoxic-T-lymphocyte-associated protein 4 (CTLA-4), expressed on exhausted T lymphocytes (TEX), have become major therapeutic targets in immune checkpoint blockade (ICB) therapy. The significance of TCF1 in the sustenance of CTL-mediated immunity against pathogens and tumors, as well as its recently observed necessity for an effective anti-tumor immune response in ICB therapy, presents TCF1 as a potentially significant biomarker and/or therapeutic target for overcoming CD8+ T cell exhaustion and resistance to ICB therapy. In this review, we aim to outline the recent findings on the role of TCF1 in T cell development and discuss its implications in anti-tumor immunity.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A275-A275
Author(s):  
Rebecca Ward ◽  
Elena Paltrinieri ◽  
Marilyn Marques ◽  
Priyadarshini Iyer ◽  
Sylvia Dietrich ◽  
...  

BackgroundT-cell immunoreceptor with Ig and ITIM domains (TIGIT) is an important negative regulator of the immune response to cancer that contributes to resistance/relapse to anti-PD-1 therapy.1 In clinical trials, anti-human (h) TIGIT antibodies have shown promising activity in combination with anti-PD-1/PD-L1 antibodies for the treatment of various solid tumors.2 However, the optimal format for anti-TIGIT antibodies remains controversial. Here we describe a novel Fcγ receptor (FcγR)-dependent mechanism of action that is critical for enhancing T and NK cell anti-tumor immunity, and, further informs on the optimal design of anti-TIGIT antibodies.MethodsWe investigated a panel of Fc-silent, Fc-competent, and Fc-engineered anti-mouse (m) TIGIT antibody variants in syngeneic murine CT26 tumor-bearing or B16F10 pseudo-metastases models. To further elucidate the relative contribution of T and NK cells in controlling tumor growth, we assessed the activity of Fc-engineered anti-TIGIT antibodies in NK cell-depleted or T cell-deficient (Nu-Foxn1nu) CT26 tumor-bearing mice. Immune-related pharmacodynamic changes in the tumor microenvironment were assessed by flow cytometry. We further validated these findings in primary human T and NK cell activation assays using Fc-engineered anti-human TIGIT antibodies.ResultsThe Fc-engineered anti-mTIGIT antibody, which demonstrates enhanced binding to mouse FcγRIV, was the only variant to deliver single agent anti-tumor activity. The Fc-enhanced variant outperformed the Fc-competent variant while the Fc-inert variant had no anti-tumor activity. Tumor control by anti-mTIGIT antibodies was not dependent on Treg depletion, but rather on increased frequency of CD8+ T cells and activated NK cells (Ki67, IFNγ, CD107a and TRAIL) in the tumor microenvironment. Concordant with observations in the mouse, Fc-engineered anti-hTIGIT antibodies with improved binding to FcγRIIIA demonstrate superior T and NK cell activation in PBMC-based assays compared to a standard hIgG1 variant. Notably, superior activity of the Fc-engineered anti-hTIGIT antibody was observed from PBMC donors that express either high or low affinity FcγRIIIA. Blockade of FcγRIIIA or depletion of CD14+ and CD56+ cells reduced the functional activity of the Fc-enhanced anti-TIGIT antibody, confirming the requirement for FcγR co-engagement to maximize T cell responses.ConclusionsOur data demonstrate the importance of FcγR co-engagement by anti-TIGIT antibodies to promote immune activation and tumor control. First generation anti-TIGIT antibodies are not optimally designed to co-engage all FcγRIIIA variants. However, Fc-enhanced anti-TIGIT antibodies unlock a novel FcγR-dependent mechanism of action to enhance T and NK cell-dependent anti-tumor immunity and further improve therapeutic outcomes.ReferencesJohnston RJ, et al., The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell 2014; 26:923–37.Rodriguez-Abreu D, et al., Primary analysis of a randomized, double-blind, phase II study of the anti-TIGIT antibody tiragolumab (tira) plus atezolizumab (atezo) versus placebo plus atezo as first-line (1L) treatment in patients with PD-L1-selected NSCLC (CITYSCAPE). Journal of Clinical Oncology 2020; 38:15_suppl, 9503–9503.


2019 ◽  
Vol 7 (5) ◽  
pp. 707-718 ◽  
Author(s):  
Quentin Haas ◽  
Kayluz Frias Boligan ◽  
Camilla Jandus ◽  
Christoph Schneider ◽  
Cedric Simillion ◽  
...  

2015 ◽  
Vol 23 (10) ◽  
pp. 1653-1662 ◽  
Author(s):  
Daniel O Villarreal ◽  
Megan C Wise ◽  
Rebekah J Siefert ◽  
Jian Yan ◽  
Laurence M Wood ◽  
...  

Nanoscale ◽  
2021 ◽  
Author(s):  
Simeng Liu ◽  
Huimin Liu ◽  
Xiaoshuang Song ◽  
Ailing Jiang ◽  
Yuchuan Deng ◽  
...  

Efficient tumor-targeting delivery of CpG or BMS-202 by adoptive T-cells coupled with drug loaded liposomes reversed the immunosuppressive tumor microenvironment, restoring T cell viability and effectively inhibiting the growth of melanoma.


Sign in / Sign up

Export Citation Format

Share Document