Tumor Growth Inhibition by a Purine Analogue

2009 ◽  
Vol 7 (9) ◽  
pp. 271-273
PLoS ONE ◽  
2012 ◽  
Vol 7 (10) ◽  
pp. e48654 ◽  
Author(s):  
Giovanna Bianchi ◽  
Fabio Morandi ◽  
Michele Cilli ◽  
Antonio Daga ◽  
Chiara Bocelli-Tyndall ◽  
...  

1999 ◽  
Vol 82 (08) ◽  
pp. 846-849 ◽  
Author(s):  
Steven Shapiro

SummaryIn the mouse, macrophage elastase is critical to macrophage proteolysis. The use of gene-targeting has uncovered both pathological roles, including destructive effects in aneurysm formation and emphysema, and physiological roles, such as tumor growth inhibition and regulation of inflammation. Translation of findings from mouse to human biology depends upon how well the disease models replicate the human conditions and the similarity of enzyme profile between species. We know that human MMP-12 is associated with these diseases, but as opposed to the mouse, other MMPs may also be of importance (MMP-9, and perhaps MMP-7, in particular). Our interpretation is that findings in mice reflect the critical role of macrophage proteolysis in these disease processes.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A804-A804
Author(s):  
Luis Zuniga ◽  
Karan Uppal ◽  
Kathy Bang ◽  
Enping Hong ◽  
Simran Sabharwal ◽  
...  

BackgroundThe use of pattern recognition receptor agonists (PRRAs) such as Toll-like receptor (TLR) agonists is an attractive approach for cancer immunotherapy. TLR agonism elicits anti-tumor activity by activating antigen presenting cells (APCs) to promote a proinflammatory microenvironment and anti-tumor immunity. Local delivery of TLR agonists has shown encouraging preclinical and clinical anti-tumor benefit. However, intratumoral (IT) delivery of naked PRRAs may lead to rapid effusion from the tumor microenvironment, potentially impacting their effectiveness in inducing local inflammation and may promote systemic cytokine release, increasing the risk of adverse effects.MethodsTransConTM TLR7/8 Agonist was designed to address the current limitations of PRRA therapies and IT delivery through sustained and controlled release of resiquimod, a potent TLR7/8 agonist, following IT administration of a hydrogel depot.ResultsA single IT injection of TransCon TLR7/8 Agonist induced potent tumor growth inhibition in a dose-dependent manner in syngeneic mouse CT26 tumors. Following IT TransCon TLR7/8 Agonist treatment, acute and sustained upregulation of cell surface markers indicative of activation of APCs, such as CD54, CD69, and CD86, in the tumor was observed by fluorescence activated flow cytometry (FACs). Additionally, TransCon TLR7/8 Agonist treatment was associated with an increase in the frequency of APCs with an activated phenotype in tumor draining lymph nodes (LNs). Further, a concomitant potentiation in the frequency of activated CD4 and CD8 T cells in tumor draining LNs following IT TransCon TLR7/8 Agonist treatment was observed, as demonstrated by increased expression of Ki67, ICOS, or granzyme B.ConclusionsThese data support that a single IT dose of TransCon TLR7/8 Agonist can mediate robust anti-tumor activity as a monotherapy in the CT26 syngeneic mouse tumor model while promoting local activation of intratumoral APCs. Such activation may promote tumor antigen uptake and migration to tumor-associated lymphoid tissue, as evidenced by an increase in APCs with an activated phenotype in tumor draining LNs following TransCon TLR7/8 Agonist treatment. Activated tumor antigen-bearing APCs can promote the priming and activation of tumor-specific T cells in the tumor-draining LNs. Consistently, a dose-dependent increase in the frequency of T cells with an activated effector phenotype in tumor draining LNs following administration of TransCon TLR7/8 Agonist was observed. These preclinical data further support TransCon TLR7/8 Agonist as a novel and potentially efficacious PRRA therapy. A clinical trial to evaluate safety and efficacy of TransCon TLR7/8 Agonist as monotherapy, and in combination with pembrolizumab, in cancer patients has been initiated (transcendIT-101; NCT04799054).Ethics ApprovalThe animal studies performed described were performed in accordance with the “Guide for the Care and Use of Laboratory Animals: Eighth Edition” and approved by the institutional animal care and use committee (IACUC).


Author(s):  
Kae Tsutsumi ◽  
Tatsuhiko Kasaoka ◽  
Hyi-Man Park ◽  
Hiroko Nishiyama ◽  
Motowo Nakajima ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1844-1844
Author(s):  
John Richards ◽  
Myriam N Bouchlaka ◽  
Robyn J Puro ◽  
Ben J Capoccia ◽  
Ronald R Hiebsch ◽  
...  

AO-176 is a highly differentiated, humanized anti-CD47 IgG2 antibody that is unique among agents in this class of checkpoint inhibitors. AO-176 works by blocking the "don't eat me" signal, the standard mechanism of anti-CD47 antibodies, but also by directly killing tumor cells. Importantly, AO-176 binds preferentially to tumor cells, compared to normal cells, and binds even more potently to tumors in their acidic microenvironment (low pH). Hematological neoplasms are the fourth most frequently diagnosed cancers in both men and women and account for approximately 10% of all cancers. Here we describe AO-176, a highly differentiated anti-CD47 antibody that potently targets hematologic cancers in vitro and in vivo. As a single agent, AO-176 not only promotes phagocytosis (15-45%, EC50 = 0.33-4.1 µg/ml) of hematologic tumor cell lines (acute myeloid leukemia, non-Hodgkin's lymphoma, multiple myeloma, and T cell leukemia) but also directly targets and kills tumor cells (18-46% Annexin V positivity, EC50 = 0.63-10 µg/ml) in a non-ADCC manner. In combination with agents targeting CD20 (rituximab) or CD38 (daratumumab), AO-176 mediates enhanced phagocytosis of lymphoma and multiple myeloma cell lines, respectively. In vivo, AO-176 mediates potent monotherapy tumor growth inhibition of hematologic tumors including Raji B cell lymphoma and RPMI-8226 multiple myeloma xenograft models in a dose-dependent manner. Concomitant with tumor growth inhibition, immune cell infiltrates were observed with elevated numbers of macrophage and dendritic cells, along with increased pro-inflammatory cytokine levels in AO-176 treated animals. When combined with bortezomib, AO-176 was able to elicit complete tumor regression (100% CR in 10/10 animals treated with either 10 or 25 mg/kg AO-176 + 1 mg/kg bortezomib) with no detectable tumor out to 100 days at study termination. Overall survival was also greatly improved following combination therapy compared to animals treated with bortezomib or AO-176 alone. These data show that AO-176 exhibits promising monotherapy and combination therapy activity, both in vitro and in vivo, against hematologic cancers. These findings also add to the previously reported anti-tumor efficacy exhibited by AO-176 in solid tumor xenografts representing ovarian, gastric and breast cancer. With AO-176's highly differentiated MOA and binding characteristics, it may have the potential to improve upon the safety and efficacy profiles relative to other agents in this class. AO-176 is currently being evaluated in a Phase 1 clinical trial (NCT03834948) for the treatment of patients with select solid tumors. Disclosures Richards: Arch Oncology Inc.: Employment, Equity Ownership, Other: Salary. Bouchlaka:Arch Oncology Inc.: Consultancy, Equity Ownership. Puro:Arch Oncology Inc.: Employment, Equity Ownership. Capoccia:Arch Oncology Inc.: Employment, Equity Ownership. Hiebsch:Arch Oncology Inc.: Employment, Equity Ownership. Donio:Arch Oncology Inc.: Employment, Equity Ownership. Wilson:Arch Oncology Inc.: Employment, Equity Ownership. Chakraborty:Arch Oncology Inc.: Employment, Equity Ownership. Sung:Arch Oncology Inc.: Employment, Equity Ownership. Pereira:Arch Oncology Inc.: Employment, Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document