Your browser doesn't support javascript.
loading
Next-Generation Antisense Oligonucleotide of TGF-ß2 Enhances T Cell-Mediated Anticancer Efficacy of Anti-PD-1 Therapy in a Humanized Mouse Model of Immune-Excluded Melanoma.
Lee, Hong Kyu; Kim, Cho-Won; Ahn, Dohee; Go, Ryeo-Eun; Choi, Youngdong; Choi, Kyung-Chul.
Affiliation
  • Lee HK; Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea.
  • Kim CW; Division of Endocrinology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA.
  • Ahn D; Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea.
  • Go RE; Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea.
  • Choi Y; Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea.
  • Choi KC; Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea.
Cancers (Basel) ; 14(21)2022 Oct 25.
Article in En | MEDLINE | ID: mdl-36358638
ABSTRACT
Anti-programmed death-1 (PD-1) immunotherapy is one of the most promising therapeutic interventions for treating various tumors, including lung cancer, bladder cancer, and melanoma. However, only a subset of patients responds to anti-PD-1 therapy due to complicated immune regulation in tumors and the evolution of resistance. In the current study, we investigate the potential of a novel transforming growth factor-beta2 (TGF-ß2) antisense oligonucleotide (ngTASO), as a combination therapy with an anti-PD-1 antibody in melanoma. This study was conducted in a melanoma-bearing human immune system mouse model that recapitulates immune-excluded phenotypes. We observed that the TGF-ß2 blockade by ngTASO in combination with PD-1 inhibition downregulated the tumor intrinsic ß-catenin, facilitated the infiltration of CD8+ cytotoxic lymphocytes (CTLs) in the tumor, and finally, enhanced the antitumor immune potentials and tumor growth delays. Blockade of TGF-ß2 combined with PD-1 inhibition also resulted in downregulating the ratio of regulatory T cells to CTLs in the peripheral blood and tumor, resulting in increased granzyme B expression. In addition, co-treatment of ngTASO and anti-PD-1 augmented the PD-L1 expression in tumors, which is associated with an improved response to anti-PD-1 immunotherapy. These results indicate that the combination of ngTASO and anti-PD-1 exerts an enhanced T cell-mediated antitumor immune potential. Hence, co-inhibition of TGF-ß2 and PD-1 is a potentially promising immunotherapeutic strategy for immune-excluded melanoma.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: Cancers (Basel) Year: 2022 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: Cancers (Basel) Year: 2022 Document type: Article