Systemic NK4 gene therapy inhibits tumor growth and metastasis of melanoma and lung carcinoma in syngeneic mouse tumor models

2009 ◽  
Vol 100 (7) ◽  
pp. 1351-1358 ◽  
Author(s):  
Yuko Kishi ◽  
Keiji Kuba ◽  
Takahiro Nakamura ◽  
Jinhua Wen ◽  
Yoshinori Suzuki ◽  
...  
2016 ◽  
Author(s):  
Rafael Cubas ◽  
Marina Moskalenko ◽  
Jeanne Cheung ◽  
Shiuh-Ming Luoh ◽  
Erin McNamara ◽  
...  

2018 ◽  
Vol 46 (1) ◽  
pp. 46-56 ◽  
Author(s):  
Chao Cheng ◽  
Li Deng ◽  
Rongxiu Li

Background/Aims: The abnormally activated EGFR promotes tumor growth, invasion and metastasis. Current therapeutics targeting EGFR have markedly improved the clinical outcome, but they are limited in use due to transient efficacy, frequent administration, high cost and significant toxicity. Methods: We rationally designed a multiepitope immunogen against EGFR, named as DEGFRm. The immunogen is composed of an epitope peptide (EGFR265-283) and the extracellular domain III (EGFR334-505) of mouse EGFR. EGFR265-283 is grafted onto the translocation domain of diphtheria toxin (DTT), and EGFR334-505 is fused to C-terminal of DTT. Next, the immunogenicity and anti-tumor efficacies of DEGFRm vaccine were examined in mouse tumor models. Results: When formulated with Alum and CpG, DEGFRm vaccine elicits Th 1 immune responses and inhibits tumor growth in both prophylactic and therapeutic mouse tumor models. Moreover, the tumor microvasculature is markedly reduced and the tumor infiltration of CD8+ T lymphocytes is greatly enhanced. Conclusions: These data suggest that active immunization with DEGFRm vaccine is a promising strategy for therapy of various EGFR+ cancers.


2018 ◽  
Author(s):  
Matthew J. Robinson ◽  
Ines Osma-Garcia ◽  
Jane Coates-Ulrichisen ◽  
Amanda Watkins ◽  
Suzanne Mosely ◽  
...  

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A602-A602
Author(s):  
Matthew Lakins ◽  
Wenjia Liao ◽  
Emma McConnell ◽  
Quincy Kaka ◽  
Jennifer Ofoedu ◽  
...  

BackgroundImmune checkpoint inhibitors have demonstrated durable clinical responses and an increase in overall survival for some patients with cancer. Next generation cancer immunotherapies, such as tumor necrosis factor receptor superfamily (TNFRSF) agonists, have potential to further improve on this success. FS120 is a tetravalent bispecific antibody targeting OX40 and CD137 (4-1BB), currently being evaluated in a Phase I clinical trial (NCT04648202). FS120 activates CD4+ and CD8+ T cells by concurrent binding to both targets via an FcgR-independent mechanism [1]. In preclinical tumor models, FS120 induced T cell proliferation and cytokine production associated with significant tumor regression, better than that observed with a monoclonal antibody combination. Here, we demonstrate the ability of FS120 to improve anti-PD-1 induced T cell activity, increasing tumor growth inhibition and survival, in syngeneic mouse tumor models, compared to monotherapy.MethodsFS120 < i >in vitro</i > activity in combination with anti-PD-1 was assessed by utilizing staphylococcal enterotoxin A (SEA) superantigen assays and mixed leukocyte reaction (MLR) assays. An anti-mouse OX40/CD137 bispecific antibody (FS120 surrogate) was tested in CT26 syngeneic mouse tumor models in combination with an anti-mouse PD-1 antibody to assess efficacy and pharmacodynamic endpoints, including T cell proliferation by < i>ex vivo</i> flow cytometry and serum cytokine levels.ResultsFS120 in combination with anti-PD-1 enhanced primary human T cell activity, when compared to either monotherapy, in both SEA and MLR assays. FS120 surrogate significantly improved survival of CT26 tumor-bearing mice treated with anti-mPD-1 antibody. FS120 surrogate and anti-PD-1 combination significantly enhanced serum interferon-gamma levels and increased proliferating granzyme B+ CD8+ T cells in the blood of tumor-bearing mice, when compared to either monotherapy treatments.ConclusionsFS120 combination with anti-PD-1 enhances T cell activity in multiple human primary immune assays. In combination with anti-PD-1, FS120 surrogate increased the antitumor efficacy with pharmacodynamic changes related specifically to T cell activation, when compared to monotherapies. These data support the development of FS120 in combination with anti-PD-1 in patients with hard-to-treat cancers who may not benefit fully from either treatment as a monotherapy.ReferencesGaspar M, Pravin J, Rodrigues L, Uhlenbroich S, Everett K L, Wollerton F, Morrow M, Tuna M, Brewis N. CD137/OX40 Bispecific Antibody Induces Potent Antitumor Activity that Is Dependent on Target Coengagement. Cancer Immunol Res. 2020; (8) (6) 781–793Ethics ApprovalMurine studies were conducted under a U.K. Home Office License in accordance with the U.K. Animal (Scientific Procedures) Act 1986 and EU Directive EU 2010/63.


Sign in / Sign up

Export Citation Format

Share Document