861 Development of FPA157, an anti-CCR8 depleting antibody engineered to preferentially eliminate tumor-infiltrating T regulatory cells

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A914-A914
Author(s):  
Andrew Rankin ◽  
Edwina Naik

BackgroundThe clinical success of PD-1- and CTLA-4- immune checkpoint inhibitors highlights the key contribution of immunosuppression to limiting effective anti-tumor responses. However, as many patients do not respond to anti-PD1 or CTLA4 therapy1-3 novel therapeutics that target additional immune-suppressive mechanisms are needed. Regulatory T cells (Tregs) inhibit immune responses in the tumor microenvironment via multiple suppressive mechanisms.4 5 Existing Treg-targeting agents lack specificity for intratumoral Tregs and can also deplete effector cells, a property that has likely contributed to the lack of clinical activity observed to date. CCR8 (C-C chemokine receptor 8) is selectively expressed on highly activated intratumoral Tregs, its high expression correlates with poor prognosis in multiple human tumor types6 7 and depletion of CCR8+ Tregs in preclinical models elicited potent anti-tumor activity. These observations provided rationale for the development of a CCR8-specific human depleting antibody.MethodsHuman FOXP3 and CCR8 expression was correlated across multiple tumor types using TCGA datasets and expression of CCR8 evaluated in primary tumor explants and PBMCs by flow cytometry. The efficacy of anti-CCR8 antibody treatment was evaluated in the MC38 and CT26 murine tumor models. The depletion of Tregs following anti-CCR8 treatment was assessed by flow cytometry. Flow cytometric-based binding assays were performed using cell lines expressing human or cynomolgus CCR8. Purified human NK cells were co-cultured with CCR8+ target cells and flow cytometry used to evaluate antibody-dependent killing activity.ResultsCCR8 expression was highly correlated with FoxP3 across multiple cancer subtypes and was low to absent on effector T cells. Importantly, CCR8 was not detected on any peripheral human leukocyte subset. In murine tumor models, anti-CCR8 antibody treatment reduced tumor growth in a dose- and Fc-gamma-receptor-dependent manner and resulted in complete regressions and the development of memory. Tumor shrinkage was associated with a reduction in intratumoral Tregs and increased representation of intratumoral CD8 T cells. FPA157 is a highly specific human and cynomolgus crossreactive CCR8 antibody that does not bind closely related chemokine receptors. FPA157 was engineered to enhance antibody-dependent cell-mediated cytotoxicity (eADCC) and elicited potent NK-mediated killing of target cells expressing CCR8 at levels observed on human intratumoralTregs.ConclusionsFPA157 is a CCR8-specific monoclonal antibody with eADCC activity that is being developed for the treatment of cancer. Depletion of CCR8+ Tregs induced substantial anti-tumor activity in pre-clinical models, thus supporting the clinical evaluation of FPA157 as a novel approach to alleviate immune suppression in the microenvironment of human solid tumors.ReferencesHellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, Minenza E, Linardou H, Burgers S, Salman P, Borghaei H, Ramalingam SS, Brahmer J, Reck M, O’Byrne KJ, Geese WJ, Green G, Chang H, Szustakowski J, Bhagavatheeswaran P, Healey D, Fu Y, Nathan F, Paz-Ares L. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med 2018;378(22):2093-2104.Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, Lao CD, Wagstaff J, Schadendorf D, Ferrucci PF, Smylie M, Dummer R, Hill A, Hogg D, Haanen J, Carlino MS, Bechter O, Maio M, Marquez-Rodas I, Guidoboni M, McArthur G, Lebbé C, Ascierto PA, Long GV, Cebon J, Sosman J, Postow MA, Callahan MK, Walker D, Rollin L, Bhore R, Hodi FS, Larkin J. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017;377(14):1345-1356.Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, Powles T, Donskov F, Neiman V, Kollmannsberger CK, Salman P, Gurney H, Hawkins R, Ravaud A, Grimm MO, Bracarda S, Barrios CH, Tomita Y, Castellano D, Rini BI, Chen AC, Mekan S, McHenry MB, Wind-Rotolo M, Doan J, Sharma P, Hammers HJ, Escudier B; CheckMate 214 Investigators. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med 2018 Apr 5;378(14):1277-1290.Teng MW, Ngiow SF, von Scheidt B, McLaughlin N, Sparwasser T, Smyth MJ. Conditional regulatory T-cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth [published correction appears in Cancer Res. 2010; 70(23):10014]. Cancer Res 2010;70(20):7800-7809.Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, Roddie C, Henry JY, Yagita H, Wolchok JD, Peggs KS, Ravetch JV, Allison JP, Quezada SA. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med 2013;210(9):1695-710.Plitas G, Konopacki C, Wu K, Bos PD, Morrow M, Putintseva EV, Chudakov DM, Rudensky AY. Regulatory T cells exhibit distinct features in human breast cancer. Immunity 2016;45(5):1122-1134.De Simone M, Arrigoni A, Rossetti G, Gruarin P, Ranzani V, Politano C, Bonnal RJP, Provasi E, Sarnicola ML, Panzeri I, Moro M, Crosti M, Mazzara S, Vaira V, Bosari S, Palleschi A, Santambrogio L, Bovo G, Zucchini N, Totis M, Gianotti L, Cesana G, Perego RA, Maroni N, Pisani Ceretti A, Opocher E, De Francesco R, Geginat J, Stunnenberg HG, Abrignani S, Pagani M. Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells. Immunity 2016;45(5):1135-1147.

Author(s):  
Anne M. Pesenacker ◽  
Lucy R. Wedderburn

In recent years, there have been many new developments in the field of regulatory T cells (Treg), challenging the consensus on their behaviour, classification and role(s) in disease. The role Treg might play in autoimmune disease appears to be more complex than previously thought. Here, we discuss the current knowledge of regulatory T cells through animal and human research and illustrate the recent developments in childhood autoimmune arthritis (juvenile idiopathic arthritis (JIA)). Furthermore, this review summarises our understanding of the fields and assesses current and future implications for Treg in the treatment of JIA.


2019 ◽  
Vol 37 (8_suppl) ◽  
pp. 70-70 ◽  
Author(s):  
Ayman Oweida ◽  
Laurel Darragh ◽  
Shilpa Bhatia ◽  
David Raben ◽  
Lynn Heasley ◽  
...  

70 Background: Head and neck tumors are highly enriched in regulatory T cells which dampen the response to radiotherapy by creating an immune-inhibitory microenvironment. We explored mechanisms of Treg infiltration and assessed their modulation by RT in murine models of HNSCC. Methods: Mechanisms of Treg infiltration were investigated in murine HNSCC tumors using whole genome sequencing and flow cytometry. Mice were treated with anti-CTLA-4, anti-CD-25 and/or anti-PD-L1 alone and in combination with RT. Tumor growth and survival were assessed. Flow cytometry was used to assess phenotypic and functional changes in intratumoral T cell populations. Multiplex ELISA was performed for assessment of cytokines. RNA Sequencing was performed to interrogate mechanisms of response and resistance to treatment. Results: Treatment with anti-CD-25 concurrently with RT led to significant tumor growth delay, enhanced T cell cytotoxicity, decreased Tregs and improved survival. In contrast CTLA-4 blockade did not affect tumor growth or survival. Treg depletion induced an influx of CD8 and CD4 T cells when combined with RT. In addition, Treg depletion in combination with RT transformed myeloid populations decreasing M2 macrophages and MDSCs and increasing M1 macrophages. Mechanistically, tumors secrete CCL20, a potent Treg chemoattractant responsible for creating a highly immunuosuppressive tumor microenvironment and potentially responsible for treatment resistance. Conclusions: These data reveal a critical role for regulatory T cells in mediating resistance to RT. Targeted depletion of Tregs represents an important mechanism of sensitizing tumors to RT. Our data support the design of clinical trials integrating targeted Treg inhibitors in the standard of care for cancer patients receiving RT.


Blood ◽  
2007 ◽  
Vol 110 (7) ◽  
pp. 2217-2218
Author(s):  
Jerzy W. Kupiec-Weglinski

The OX40 T-cell costimulatory molecule, critical for both survival and proliferation of activated T cells, has now been identified as a key negative regulator of Foxp3+ T regulatory cells (Tregs).


2017 ◽  
Vol 63 (1) ◽  
pp. 104-109
Author(s):  
Andrey Savchenko ◽  
Aleksandr Borisov ◽  
Igor Kudryavtsev ◽  
Anton Moshev

The aim of the study was to investigate the features of the relation to the number and the phenotype of the cytotoxic T-lymphocytes and NKT-cell from regulatory T-cells content in the blood by patients with renal cell carcinoma. The study included patients with renal cell carcinoma (T3N0M0, clear cell type) at the age of 40-55 years before surgery. Lymphocyte immunophenotyping was performed by flow cytometry. It is found that in the peripheral blood of the patients with renal cell carcinoma accompanied by increased number of T-regulatory cells observed decrease content of the cytotoxic T-lymphocytes and increased levels of the NKT-cells. It is assumed that no change in the number of activated T-regulatory cells and cytotoxic T-lymphocyte determined migration from the blood. Increasing the amount of the NKT-cells in renal cancer is determined by the increase of activated and effector cells but at lower levels of the regulatory subpopulation. The content of the T-regulatory cells in healthy people weakly correlated with the effector subpopulations of T-lymphocytes. In patients with renal cancer the number of the activated T-regulatory cells is closely correlated with the various NKT-lymphocytes fractions. Moreover, if the mature and regulatory NKT-cells subset detected negative relations, so with the NKT-cells expressing CD28 and CD57 markers found positive correlations that characterizes the codirectional dynamics the activated of the regulatory and effector T-lymphocyte subpopulations levels in the background of tumor growth. A canonical analysis demonstrated that the highest significance kidney cancer patients have activated regulatory T-cells, cytotoxic T cells and NKT-cells. A canonical analysis demonstrated that the highest significance by renal cancer patients have activated regulatory T-cells, cytotoxic T-cells and NKT-cells.


Stroke ◽  
2017 ◽  
Vol 48 (suppl_1) ◽  
Author(s):  
Hilda Ahnstedt ◽  
Anthony Patrizz ◽  
Javiera Bravo-Alegria ◽  
Monica Sphychala ◽  
Meaghan Roy-O’Reilly ◽  
...  

Introduction: Sex differences are increasingly recognized in stroke. Stroke incidence is higher in men than in women until an advanced age and women have higher mortality and morbidity. Inflammatory responses are key determinants of outcome but have not been well studies in aged animals. Hypothesis: We hypothesize that there are important sex differences in inflammation in aged male and female mice. Results: We quantified T cells and T regulatory cells, as well as functional outcome after stroke in aged mice. Male and female C57BL/6 mice (20-21 months) were subjected to 60 min middle cerebral artery occlusion (n=16) by the filament model, or sham surgery (n=7). Immune responses in brain, blood and spleen were investigated 15 days post-stroke by flow cytometry. Functional outcomes were assessed at day 3, 7 and 14. Mortality was 25% in females and occurred at day 1, while in males 50% mortality was seen between day 3 and 9. Hemorrhagic transformation was seen in 53% of the males that died (5/8) and in none of the females (0/4). Flow cytometry analysis at day 15 showed significantly higher levels of CD8 + T cells in the blood after stroke in males compared to females (73±5% of CD3 + T cells vs 52±3%, P<0.001). CD8 + T cells were found in brains from males and females after stroke but levels were not different from sham (Male Stroke: 64±7%, Female Stroke: 58±5%). Independent of the type of surgery (stroke or sham), we observed higher levels of splenic CD8 + T cells in males (P<0.01). T regulatory cells were significantly increased in the brain after stroke in males compared to sham (P<0.01), but not in females. Open-field testing showed a decrease in center visits after stroke in both sexes at 3 days, however males spent more time immobile (P<0.01) indicating decreased locomotor activity and/or anxiety. This difference equalized at 7 and 14 days post-stroke. Conclusion: Higher mortality and hemorrhagic transformation rates were seen in aged males than in females after stroke. This may be associated with the elevated levels of CD8 + T cells in blood and possible unfavorable effects on blood brain barrier permeability.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A740-A740
Author(s):  
Douglas Hodges ◽  
Christina Kochel ◽  
Michael Totagrande ◽  
Jeffrey Jones ◽  
Megan Welch ◽  
...  

BackgroundHuman leukocyte antigen-G (HLA-G) is an immune checkpoint molecule that belongs to the non-classical HLA-class I family of receptors. HLA-G restrains immune cell activation and effector function by engaging with inhibitory receptors ILT2 and ILT4. While expression of HLA-G is highly restricted under normal healthy conditions, we have demonstrated that its expression in cancer is aberrantly upregulated and broadly detected across a variety of tumor types. Tizona Therapeutics has generated a novel, fully human antibody that specifically targets HLA-G and reverses HLA-G-mediated immunosuppression. Here we present in vitro and in vivo data demonstrating the functional impact of HLA-G blockade on immune cells and evidence to support the use of TTX-080 in the clinic to treat patients with advanced solid tumors.MethodsEvaluation of HLA-G expression in cancer was performed using immunohistochemistry, flow cytometry, and gene profiling. Expression of ILT2 and ILT4 was assessed on tumor infiltrating leukocytes by flow cytometry. To demonstrate the suppressive function of HLA-G, primary human NK cells, T cells, and monocyte-derived macrophages were cultured with target cells expressing HLA-G. TTX-080 was then evaluated for its ability to reverse this suppression. In addition, TTX-080 was investigated in vivo using a disseminated xenograft tumor model.ResultsExpression of HLA-G was detected on tumor cells and tumor infiltrating leukocytes across a variety of solid tumor types. TTX-080 blocked interaction of HLA-G with both ILT2 and ILT4 and restored cytotoxicity in multiple assays using either primary NK cells or NKL cell lines. Monocyte-derived macrophages expressing ILT2 and ILT4 exhibited decreased phagocytosis of HLA-G+ target cells; this inhibition was reversed with an antigen-binding fragment of TTX-080. TTX-080 was also able to reverse HLA-G-mediated suppression of ILT2+ CD8+ T cells as assessed by degranulation and proinflammatory cytokine secretion. Notably, mice with disseminated tumors had extended median survival when treated with a single dose of TTX-080.ConclusionsTTX-080 reverses HLA-G-mediated suppression of ILT2+ and ILT4+ immune cells that are found within the tumor microenvironment. Blockade of HLA-G using TTX-080 therefore has the potential to reverse broad immune suppression in patients with advanced solid tumors by reinvigorating CD8+ T cells, enhancing NK cytolytic activity, and increasing macrophage phagocytosis.


2016 ◽  
Vol 2016 ◽  
pp. 1-12 ◽  
Author(s):  
Asiel Arce-Sillas ◽  
Diana Denisse Álvarez-Luquín ◽  
Beatriz Tamaya-Domínguez ◽  
Sandra Gomez-Fuentes ◽  
Abel Trejo-García ◽  
...  

T regulatory cells play a key role in the control of the immune response, both in health and during illness. While the mechanisms through which T regulatory cells exert their function have been extensively described, their molecular effects on effector cells have received little attention. Thus, this revision is aimed at summarizing our current knowledge on those regulation mechanisms on the target cells from a molecular perspective.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A298-A298
Author(s):  
David Draper ◽  
Philip Lapinski ◽  
Scott Wise

BackgroundBladder cancer (BC) is the thirteenth leading cause of cancer-related deaths.1 Five checkpoint immunotherapies that target the PD-1/PD-L1 axis are FDA approved, and gene- and protein-based approaches are helping to identify new combination treatment strategies for therapeutic intervention.2 Using the murine MB49 model for BC, we demonstrate how non-targeted immune gene expression profiling can combine with flow cytometry to provide a gene and cell-specific signature for the tumor microenvironment and help identify potential targets for novel treatment approaches.MethodsAnimals with established MB49 tumors were treated with anti-mPD-1 or isotype control antibodies. Tumors were collected 7 days after the last treatment. Flow cytometry examined anti-mPD-1 treatment-induced immunophenotypic modulation for eleven tumor-infiltrating immune subsets. The mouse PanCancer IO 360™ panel (NanoString) provided transcriptomic analysis of 770 immuno-oncology-related genes. The ROSALIND™ platform (OnRamp BioInformatics) was used to identify differentially regulated genes between treatment groups (±1.5 fold-change; p <0.05).ResultsAnti-mPD-1 had moderate anti-tumor activity, with a 58% tumor growth inhibition at day 18 post-implant in treated compared to control animals. Flow cytometry revealed anti-mPD-1 triggered an increase in tumor-infiltrating CD8+ T cells (45%) compared to control animals. Furthermore, the CD8+ T cell phenotype was altered by anti-mPD-1 treatment. The percentage of CD8+ T cells that expressed ICOS and LAG-3 was increased in tumors from anti-mPD-1 treated animals (22% and 35% respectively). A reduction in PD-1 expression was also observed (33%). In myeloid cells, iNOS expression increased in tumor-associated macrophages from treated animals compared to controls. NanoString analysis revealed 62 genes were differentially regulated in tumors from anti-mPD-1 treated animals compared to controls. ROSALIND analysis classified 30 of the genes as regulators of interferon, cytotoxicity, antigen presentation, and cytokine/chemokine signaling. Also, among the genes upregulated by anti-mPD-1 were IDO, HAVCR2 (TIM-3), and CSFR1, which can promote tumor growth and are clinical targets actively being investigated for new immunotherapies.ConclusionsNanoString analysis complemented flow cytometry to provide a comprehensive profile of the MB49 tumor. Together, these data demonstrate that anti-mPD1 increases T cell recruitment into the tumor and upregulates the expression of genes known to enhance T cell recruitment and anti-tumor activity. iNOS protein upregulation indicates that anti-mPD-1 treatment may also exert effects by reprogramming M2 macrophages towards an M1 phenotype. Upregulation of IDO, HAVCR2, and CSFR1 genes may effectively counteract anti-mPD-1 treatment. Further investigation may elucidate clinical implications for inhibitors of these gene products as combination treatment partners with anti-mPD-1.ReferencesSaginala K, Barsouk A, Aluru JS, Rawla P, Padala SA, Barsouk A. Epidemiology of bladder cancer. Med Sci 2020;1:15–26. Lopez-Beltran A, Cimadamore A, Blanca A, Massari F, Vau N, Scarpelli M, Cheng L, Montironi R. Immune checkpoint inhibitors for the treatment of bladder cancer. Cancers 2021;1:131–146.Ethics ApprovalN/A


Sign in / Sign up

Export Citation Format

Share Document