207 Small molecule inhibitors of Sec61 cotranslational translocation regulate the phagocytosis checkpoint molecule CD47

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A223-A223
Author(s):  
Jennifer Whang ◽  
Andrea Fan ◽  
Christopher Kirk ◽  
Eric Lowe ◽  
Dustin McMinn ◽  
...  

BackgroundMany tumor cells escape immune cell clearance by overexpressing CD47, a multi-pass transmembrane protein, which binds signal regulatory protein α (SIRPα) on macrophages leading to decreased phagocytic activity. Blockade of CD47/SIRPα interactions enhances macrophage phagocytosis and is being targeted with antibody-based drugs, some of which are used in combination therapies in clinical trials. A novel method to target CD47 is through the inhibition of cotranslational translocation of transmembrane proteins. Immediately after exiting the ribosome, signal sequences that are unique to each protein are directed through the Sec61 channel into the ER for extracellular expression.1 Several Sec61-targeting compounds have been identified to suppress translocation in a signal sequence-specific manner.2 We previously described Sec61 inhibitors capable of selectively targeting immune checkpoint proteins and enhancing T cell function.3 Here, we demonstrate the blockade of CD47 expression on tumor cells and enhancement of macrophage phagocytosis with small molecule inhibitors of Sec61.MethodsSec61-dependent expression of target proteins was assayed using HEK293 cells overexpressing constructs comprised of signal sequences fused to a luciferase reporter. Stimulated PBMCs or tumor cells were incubated with Sec61 inhibitors, and surface expression of checkpoint molecules were examined by flow cytometry. Necrotic and apoptotic cells were assessed by Annexin V and 7AAD labeling. Human CD14+ monocytes were differentiated to M1- or M2-type macrophages. Jurkat or SKBR3 cells were incubated with Sec61 inhibitors, labeled with a pH sensitive dye and co-cultured with macrophages to assess phagocytosis.ResultsWe identified Sec61 inhibitors that block select immune checkpoint proteins. Compounds demonstrated either selective or multi-target profiles in transient transfection screens, which was supported by decreased protein expression on activated T cells. KZR-9275 targeted multiple checkpoint molecules, including PD-1, LAG-3 and CD73, along with a potent inhibition of the CD47 signal sequence reporter. CD47 surface expression was decreased on Jurkat and SKBR3 cells following 72 hours of compound treatment. KZR-9275 treatment of SKBR3 cells induced a minor increase in apoptotic cells, which was not detected in Jurkat cells. Increased macrophage phagocytosis, especially with M2-type macrophages, was observed when Jurkat or SKBR3 cells were pre-treated with KZR-9275.ConclusionsOur findings demonstrate that Sec61 inhibitors can block the expression of CD47, a phagocytosis checkpoint protein, on tumor cells and subsequently modulate macrophage phagocytic activity. Small molecule inhibitors of Sec61 provide an opportunity to target multiple checkpoint proteins on various cell populations. Future in vivo tumor models will assess the efficacy of Sec61 inhibitors to provide combination-like therapy.ReferencesPark E, Rapoport TA. Mechanisms of Sec61/SecY-mediated protein translocation across membranes. Annu Rev Biophys 2012; 41:1–20.Van Puyenbroeck V, Vermeire K. Inhibitors of protein translocation across membranes of the secretory pathway: novel antimicrobial and anticancer agents. Cell Mol Life Sci 2018; 75:1541–1558.Whang J, Anderl J, Fan A, Kirk C, Lowe E, McMinn D, et al. Targeting multiple immune checkpoint proteins with novel small molecule inhibitors of Sec61-dependent cotranslational translocation. 34th Annual Meeting & Pre-Conference Programs of the Society for Immunotherapy of Cancer (SITC 2019): part 2. J Immunother Cancer 2019; 7: 283. Abstract 815.

2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Pottayil G. Sasikumar ◽  
Naremaddepalli S. Sudarshan ◽  
Srinivas Adurthi ◽  
Raghuveer K. Ramachandra ◽  
Dodderi S. Samiulla ◽  
...  

AbstractSmall molecule immune checkpoint inhibitors targeting PD-1 and other pathways may offer advantages including ease of dosing, ability to manage immune-related adverse events (irAEs) due to their shorter pharmacokinetic exposure and opportunity to target more than one pathway for improving efficacy. Here we describe the identification and characterization of CA-170, an amino acid inspired small molecule inhibitor of PD-L1 and VISTA derived from the interface of PD-1 and PD-L1. CA-170 exhibited potent rescue of proliferation and effector functions of T cells inhibited by PD-L1/L2 and VISTA with selectivity over other immune checkpoint proteins as well as a broad panel of receptors and enzymes. Observed blocking of PD-L1 signaling and binding to PD-L1 in the cellular context without preventing the assembly of PD-1:PD-L1 complex support the formation of a defective ternary complex as the mechanism of action of CA-170. Oral administration of CA-170 resulted in increased proliferation and activation of T cells in the tumor, and significant anti-tumor efficacy in a number of immunocompetent mouse tumor models either as a single agent or in combination with approved therapeutics. These results prompted the advancement of CA-170 to human clinical trials.


2020 ◽  
Vol 63 (19) ◽  
pp. 11271-11285
Author(s):  
Magdalena Konieczny ◽  
Bogdan Musielak ◽  
Justyna Kocik ◽  
Lukasz Skalniak ◽  
Dominik Sala ◽  
...  

2016 ◽  
Vol 18 (suppl 3) ◽  
pp. iii143.4-iii143
Author(s):  
Eric Ring ◽  
Blake Moore ◽  
Li Nan ◽  
Tina Etminan ◽  
James Markert ◽  
...  

2017 ◽  
Vol 77 (6) ◽  
pp. 1245-1249 ◽  
Author(s):  
Sangbin Lim ◽  
Joshua B. Phillips ◽  
Luciana Madeira da Silva ◽  
Ming Zhou ◽  
Oystein Fodstad ◽  
...  

2020 ◽  
Author(s):  
Hend Mohamed Abdel Hamid ◽  
Zeinab El Sayed Darwish ◽  
Sahar Mohamed Elsheikh ◽  
Ghada Mourad ◽  
Hanaa Donia ◽  
...  

Abstract Background: The concept of personalized therapy has been proven to be a promising approach. A popular approach is to utilize gold nanoparticles (AuNPs) as drug delivery vectors for cytotoxic drugs and small molecule inhibitors to target and eradicate oral cancer cells in vitro and in vivo. While it is currently accepted that the cytotoxic drug’s mode of action remains the key regulator of the therapeutic outcome and toxicity beside nanocarrier design. None of the leading studies have compared multiple chemotherapeutics to their baseline free drugs nor used multiple nanocarriers to calculate drugs impact versus nanocarriers effect. We hypothesized that similarly constructed nanocarriers play a greater role than only acting as cargo-carriers. If proven, AuNPs may have a therapeutic role beyond bypassing cancer cell membrane and delivering their loaded drugs. We propose that similarly constructed AuNPs can flexibly leverage different conjugated drugs irrelevant to their mode of action enhancing the therapeutic outcome.Methods: We conjugated 5- fluorouracil (5Fu), camptothecin (CPT), and a fibroblast growth factor receptor1-inhibitor (FGFR1i) to gold nanospheres (AuNSs). We followed their trajectories in Syrian hamsters with chemically induced buccal carcinomas.Results: Flow cytometry and cell cycle data shows that 5Fu- and CPT- induced a similar ratio of S-phase cell cycle arrest as nanoconjugates and in their free forms. On the other hand, FGFR1i-AuNSs induced significant sub-G1 cell population compared with its free form. Despite cell cycle dynamics variability, there was no significant difference in tumor cells’ proliferation rate between CPT-, 5Fu- and FGFR1i- AuNSs treated groups. Clinically, FGFR1i-AuNSs induced the highest tumor reduction rates followed by 5Fu- AuNSs. CPT-AuNSs induced significantly lower tumor reduction rates compared with the 5Fu- and FGFR1i- AuNSs despite showing similar proliferative rates in tumor cells.Conclusions: Our data indicates that the cellular biological events do not predict the clinical outcome. Furthermore, our results suggest that AuNSs selectively enhances the therapeutic effect of small molecule inhibitors such as FGFR1i than potent anticancer drugs. Future studies are required to better understand the underlying mechanism.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Mohammed Akhtar ◽  
Sameera Rashid ◽  
Issam A. Al-Bozom

Abstract Background Immune checkpoint proteins, especially PD-L1 and PD-1, play a crucial role in controlling the intensity and duration of the immune response, thus preventing the development of autoimmunity. These proteins play a vital role in enabling cancer cells to escape immunity, proliferate and progress. Methods This brief review highlights essential points related to testing for immune checkpoint therapy that histopathologists need to know. Results In recent years, several inhibitors of these proteins have been used to reactivate the immune system to fight cancer. Selection of patients for such therapy requires demonstration of PD-L1 activation on the tumor cells, best done by immunohistochemical staining of the tumor and immune cells using various antibodies with predetermined thresholds. Conclusions Immune checkpoint therapy appears to be promising and is rapidly expanding to include a large variety of cancers.


Sign in / Sign up

Export Citation Format

Share Document