scholarly journals Immune Checkpoint Inhibitors Regulate K+ Channel Activity in Cytotoxic T Lymphocytes of Head and Neck Cancer Patients

2021 ◽  
Vol 12 ◽  
Author(s):  
Vaibhavkumar S. Gawali ◽  
Ameet A. Chimote ◽  
Hannah S. Newton ◽  
Manuel G. Feria-Garzón ◽  
Martina Chirra ◽  
...  

Programmed death receptor-1 (PD-1) and its ligand (PD-L1) interaction negatively regulates T cell function in head and neck squamous cell carcinoma (HNSCC). Overexpression of PD-1 reduces intracellular Ca2+ fluxes, and thereby T cell effector functions. In HNSCC patients, PD-1 blockade increases KCa3.1 and Kv1.3 activity along with Ca2+ signaling and mobility in CD8+ peripheral blood T cells (PBTs). The mechanism by which PD-L1/PD-1 interaction regulates ion channel function is not known. We investigated the effects of blocking PD-1 and PD-L1 on ion channel functions and intracellular Ca2+ signaling in CD8+ PBTs of HNSCC patients and healthy donors (HDs) using single-cell electrophysiology and live microscopy. Anti-PD-1 and anti-PD-L1 antibodies increase KCa3.1 and Kv1.3 function in CD8+ PBTs of HNSCC patients. Anti-PD-1 treatment increases Ca2+ fluxes in a subset of HSNCC patients. In CD8+ PBTs of HDs, exposure to PD-L1 reduces KCa3.1 activity and Ca2+ signaling, which were restored by anti-PD-1 treatment. The PD-L1-induced inhibition of KCa3.1 channels was rescued by the intracellular application of the PI3 kinase modulator phosphatidylinositol 3-phosphate (PI3P) in patch-clamp experiments. In HNSCC CD8+ PBTs, anti-PD-1 treatment did not affect the expression of KCa3.1, Kv1.3, Ca2+ release activated Ca2+ (CRAC) channels, and markers of cell activation (CD69) and exhaustion (LAG-3 and TIM-3). Our data show that immune checkpoint blockade improves T cell function by increasing KCa3.1 and Kv1.3 channel activity in HNSCC patients.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3138-3138
Author(s):  
Mark-Alexander Schwarzbich ◽  
Arantxa Romero-Toledo ◽  
John G. Gribben

Abstract Background: Chronic lymphocytic leukaemia (CLL) is associated with global immunodeficiency, hypogammagobulinemia and T-cell exhaustion. We hypothesise that repairing T cell functions would improve cancer immune-editing, improve outcome and decrease infectious complications which cause significant morbidity in CLL patients. Chronic B-cell receptor (BCR) activation as well as close interactions with the tumour microenvironment promote survival of malignant CLL B-cells, supporting their ability to induce immune suppression. To date, the most clinically successful approach to BCR-signalling inhibition is by the use of BTK inhibitors (BTKi). It has been suggested that the BTKi Ibrutinib has the ability to modulate T-helper cell polarity from Th2 to Th1 and thus would be a step towards repairing CLL associated T-cell defects (1). We examined the impact of Ibrutinib on T cell function and immune phenotype in vivo in Eµ-TCL1 mice with CLL. Materials and Methods: C57/Bl6 animals 2.5 months of age were injected with 40x10e6 purified CLL B-cells pooled from Eµ-TCL1 mice with CLL. When peripheral blood CLL load reach >10% animals were randomized (mean day 14) to either vehicle treatment (2% HPBCD) or Ibrutinib treatment (0.15.mg/kg in 2% HPBCD) for 21 days. 17 animals per group were analysed. Splenic cells were isolated, the cellular component characterized by CyTOF and T cell function assessed by multi-parameter flow cytometry and T-cell synapse formation assay. We demonstrated that Ibrutinib administration this way led to high levels of BTK occupancy. Results: Treatment with both Ibrutinib resulted in increased expression of IL2 (p=0.0004) in CD4+ T cells and decreased expression of IL4 among both CD4+ T cells (p=0.0015) and CD8+ T-cells (p<0.0001). Interferon gamma production was reduced in CD4+ (p=0.0056) and CD8+ T-cells (p=0.0020) with Ibrutinib treatment, which also resulted in an increase in CD107a+/CD107a- ratio among both CD44+ (p=0.0002) and CD44- CD8+ cytotoxic T-cells (p=0.0463). Ibrutinib treatment increases T-cell synapse area (p<0.0001) (Figure 1). We find a trend towards less antigen experienced CD44+ T-cells with Ibrutinib treatment with decreased expression of PD-1 in both CD44+ and CD44- negative T-cells but more pronounced in the antigen experienced T-cells. (Figure 2A). In addition, decreased expression of immune checkpoint receptor KLRG-1 on antigen experienced CD44+ T-cells was observed, most pronounced in the CD4+ subsets (Figure 2 B). Among NK 1.1+ NK-cells we find a strong trend towards decreased expression of immune checkpoint receptor KLRG-1 (Figure 2C). In white pulp myelomonocytic splenocytes (WPMC) we find a shift away from Ly6c low macrophage/patrolling monocyte-like cells towards more Ly6C high inflammatory monocyte-like cells. Moreover, we find a trend towards decreased expression of PD-L1, which is highly expressed among Ly6c low cells and shows little to no expression among Ly6C high cells (Figure 2D). Conclusion: In vivo Ibrutinib treatment in this mouse model resulted in alteration in T cell function with cytokine secretion changes in keeping with a switch away from Th2 towards Th1 polarity as well as increased in cytotoxic T-cell function. The typical exhaustion phenotype of T-cell subsets is significantly ameliorated by Ibrutinib including a decrease in PD-1 expression. Moreover, we demonstrate a decrease in numbers of KLRG-1 high NK1.1+ NK cells. WPMC cells are shifted away from a potentially more tumour promoting Ly6C low PD-L1+ phenotype towards a more inflammatory Ly6c high PD-L1 low phenotype. These findings may point to a potential synergism of the combination of BTK inhibitors with immune checkpoint blockade for the treatment of CLL. References Dubovsky JA, Beckwith KA, Natarajan G, Woyach JA, Jaglowski S, Zhong Y, et al. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood. 2013;122(15):2539-49. Disclosures Gribben: Cancer Research UK: Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Unum: Equity Ownership; Abbvie: Honoraria; Novartis: Honoraria; TG Therapeutics: Honoraria; Janssen: Honoraria, Research Funding; Acerta Pharma: Honoraria, Research Funding; NIH: Research Funding; Pharmacyclics: Honoraria; Wellcome Trust: Research Funding; Roche: Honoraria; Kite: Honoraria; Medical Research Council: Research Funding.


Blood ◽  
2018 ◽  
Vol 131 (1) ◽  
pp. 58-67 ◽  
Author(s):  
Behzad Rowshanravan ◽  
Neil Halliday ◽  
David M. Sansom

Abstract CD28 and CTLA-4 are members of a family of immunoglobulin-related receptors that are responsible for various aspects of T-cell immune regulation. The family includes CD28, CTLA-4, and ICOS as well as other proteins, including PD-1, BTLA, and TIGIT. These receptors have both stimulatory (CD28, ICOS) and inhibitory roles (CTLA-4, PD-1, BTLA, and TIGIT) in T-cell function. Increasingly, these pathways are targeted as part of immune modulatory strategies to treat cancers, referred to generically as immune checkpoint blockade, and conversely to treat autoimmunity and CTLA-4 deficiency. Here, we focus on the biology of the CD28/CTLA-4 pathway as a framework for understanding the impacts of therapeutic manipulation of this pathway.


2021 ◽  
Author(s):  
Emi Hifumi ◽  
Hiroaki Taguchi ◽  
Tamami Nonaka ◽  
Takunori Harada ◽  
Taizo Uda

Programmed cell death 1 (PD-1) is an immune checkpoint regulating T-cell function. A catalytic antibody light chain, H34, could enzymatically degrade the PD-1 molecule. In addition, it inhibited the binding of PD-1 with PD-L1.


2020 ◽  
Vol 8 (2) ◽  
pp. e000844
Author(s):  
Hannah S Newton ◽  
Vaibhavkumar S Gawali ◽  
Ameet A Chimote ◽  
Maria A Lehn ◽  
Sarah M Palackdharry ◽  
...  

BackgroundImmunotherapy has emerged as a promising treatment modality for head and neck squamous cell carcinoma (HNSCC). Pembrolizumab, an anti-programmed death 1 antibody, is an immunotherapy agent currently approved for metastatic HNSCC and curative intent clinical trials. Although clinical responses to pembrolizumab are promising, many patients fail to respond. However, it is well known that T cell cytotoxicity and chemotaxis are critically important in the elimination of HNSCC tumors. These functions depend on ion channel activity and downstream Ca2+ fluxing abilities, which are defective in patients with HNSCC. The purpose of this study was to elucidate the effects of pembrolizumab on potassium (K+) channel (KCa3.1 and Kv1.3) activity, Ca2+ fluxes, and chemotaxis in the cytotoxic T cells of patients with HNSCC and to determine their correlation with treatment response.MethodsFunctional studies were conducted in CD8+ peripheral blood T cells (PBTs) and tumor infiltrating lymphocytes (TILs) from patients with HNSCC treated with pembrolizumab. Untreated patients with HNSCC were used as controls. The ion channel activity of CD8+ T cells was measured by patch-clamp electrophysiology; single-cell Ca2+ fluxing abilities were measured by live microscopy. Chemotaxis experiments were conducted in a three-dimensional collagen matrix. Pembrolizumab patients were stratified as responders or non-responders based on pathological response (percent of viable tumor remaining at resection; responders: ≤80% viable tumor; non-responders: >80% viable tumor).ResultsPembrolizumab increased K+ channel activity and Ca2+ fluxes in TILs independently of treatment response. However, in PBTs from responder patients there was an increased KCa3.1 activity immediately after pembrolizumab treatment that was accompanied by a characteristic increase in Kv1.3 and Ca2+ fluxes as compared with PBTs from non-responder patients. The effects on Kv1.3 and Ca2+ were prolonged and persisted after tumor resection. Chemotaxis was also improved in responder patients’ PBTs. Unlike non-responders’ PBTs, pembrolizumab increased their ability to chemotax in a tumor-like, adenosine-rich microenvironment immediately after treatment, and additionally they maintained an efficient chemotaxis after tumor resection.ConclusionsPembrolizumab enhanced K+ channel activity, Ca2+ fluxes and chemotaxis of CD8+ T cells in patients with HNSCC, with a unique pattern of response in responder patients that is conducive to the heightened functionality of their cytotoxic T cells.


2001 ◽  
Vol 120 (5) ◽  
pp. A314-A315
Author(s):  
D FRANCHIMONT ◽  
J GALON ◽  
M VACCHIO ◽  
R VISCONTI ◽  
G CHROUSOS ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document