Your browser doesn't support javascript.
loading
Humanized PD-1 Knock-in Mice Reveal Nivolumab's Inhibitory Effects on Glioblastoma Tumor Progression In Vivo.
Chiang, Chun-Yu; Huang, Meng-Chu; Tsai, Shih-Chong; Hsu, Fei-Ting; Liao, Tsai-Lan; Yu, Jei-Hwa; Lin, Tzu-Hsiang; Huang, Hua-Hsih; Liao, Pen-An.
Affiliation
  • Chiang CY; Ph.D. Program of Electrical and Communications Engineering, Feng Chia University, Taichung, Taiwan, R.O.C.
  • Huang MC; Department of Medical Imaging, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.
  • Tsai SC; Institute of Biologics, Development Center for Biotechnology, Taipei, Taiwan, R.O.C.
  • Hsu FT; Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
  • Liao TL; Department of Medical Imaging and Radiologic Sciences, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C.
  • Yu JH; Institute of Biologics, Development Center for Biotechnology, Taipei, Taiwan, R.O.C.
  • Lin TH; Department of Radiology, Cathay General Hospital, Taipei, Taiwan, R.O.C.
  • Huang HH; Department of Medical Imaging, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.; wyu@show.org.tw.
  • Liao PA; Department of Radiology, Cathay General Hospital, Taipei, Taiwan, R.O.C.; cgh17508@cgh.org.tw.
In Vivo ; 37(5): 1991-2000, 2023.
Article in En | MEDLINE | ID: mdl-37652472
ABSTRACT
BACKGROUND/

AIM:

Immunotherapy has been considered a promising approach for brain tumor treatment since the discovery of the brain lymphatic system. Glioblastoma (GBM), the most aggressive type of brain tumor, is associated with poor prognosis and a lack of effective treatment options. MATERIALS AND

METHODS:

To test the efficacy of human anti-PD-1, we used a humanized PD-1 knock-in mouse to establish an orthotopic GBM-bearing model.

RESULTS:

Nivolumab, a human anti-PD-1, effectively inhibited tumor growth, increased the survival rate of mice, enhanced the accumulation and function of cytotoxic T cells, reduced the accumulation and function of immunosuppressive cells and their related factors, and did not induce tissue damage or biochemical changes. The treatment also induced the accumulation and activation of CD8+ cytotoxic T cells, while reducing the accumulation and activation of myeloid-derived suppressor cells, regulatory T cells, and tumor-associated macrophages in the immune microenvironment.

CONCLUSION:

Nivolumab has the potential to be a treatment for GBM.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Neoplasms / Glioblastoma Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: In Vivo Journal subject: NEOPLASIAS Year: 2023 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain Neoplasms / Glioblastoma Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: In Vivo Journal subject: NEOPLASIAS Year: 2023 Document type: Article