Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Pharmacother ; 175: 116782, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38776682

ABSTRACT

LAG3 is an inhibitory immune checkpoint expressed on activated T and NK cells. Blocking the interaction of LAG3 with its ligands MHC-II and FGL1 renders T cells improved cytotoxicity to cancer cells. Current study generated a panel of LAG3 monoclonal antibodies (mAbs) through immunization of mice followed by phage display. Some of them bound to the D1-D2 domain of LAG3, which is known for the engagement of its ligands FGL1 and MHC-II. Three outperformers, M208, M226, and M234, showed stronger blocking activity than Relatlimab in the FGL1 binding. Furthermore, M234 showed dual inhibition of FGL1 (IC50 of 20.6 nM) and MHC-II binding (IC50 of 6.2 nM) to LAG3. In vitro functional tests showed that M234 significantly stimulated IFN-γ secretion from activated PBMC cells. In vivo studies in a mouse model of hepatocellular carcinoma xenografts demonstrated that combining M234 IgG with GPC3-targeted bispecific antibodies significantly improved efficacy. In addition, GPC3-targeted CAR-T cells secreting IL-21-M234 scFv fusion protein exhibited enhanced activity in inhibiting tumor growth and greatly increased the survival rate of mice. Taken together, M234 has potential in cancer immunotherapy and warrants further clinical trial.


Subject(s)
Antibodies, Neutralizing , Antigens, CD , Immunotherapy , Lymphocyte Activation Gene 3 Protein , Animals , Humans , Mice , Antigens, CD/immunology , Antigens, CD/metabolism , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/immunology , Ligands , Immunotherapy/methods , Cell Line, Tumor , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Xenograft Model Antitumor Assays , Liver Neoplasms/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Mice, Inbred BALB C , Protein Binding , Female , Antibodies, Monoclonal/pharmacology
2.
Pharmacol Res ; 203: 107186, 2024 May.
Article in English | MEDLINE | ID: mdl-38641176

ABSTRACT

Chimeric antigen receptor (CAR)-modified T cell therapy has achieved remarkable efficacy in treating hematological malignancies, but it confronts many challenges in treating solid tumors, such as the immunosuppressive microenvironment of the solid tumors. These factors reduce the antitumor activity of CAR-T cells in clinical trials. Therefore, we used the immunocytokine interleukin-12 (IL-12) to enhance the efficacy of CAR-T cell therapy. In this study, we engineered CAR-IL12R54 T cells that targeted mesothelin (MSLN) and secreted a single-chain IL-12 fused to a scFv fragment R54 that recognized a different epitope on mesothelin. The evaluation of the anti-tumor activity of the CAR-IL12R54 T cells alone or in combination with anti-PD-1 antibody in vitro and in vivo was followed by the exploration of the functional mechanism by which the immunocytokine IL-12 enhanced the antitumor activity. CAR-IL12R54 T cells had potency to lyse mesothelin positive tumor cells in vitro. In vivo studies demonstrated that CAR-IL12R54 T cells were effective in controlling the growth of established tumors in a xenograft mouse model with fewer side effects than CAR-T cells that secreted naked IL-12. Furthermore, combination of PD-1 blockade antibody with CAR-IL12R54 T cells elicited durable anti-tumor responses. Mechanistic studies showed that IL12R54 enhanced Interferon-γ (IFN-γ) production and dampened the activity of regulatory T cells (Tregs). IL12R54 also upregulated CXCR6 expression in the T cells through the NF-κB pathway, which facilitated T cell infiltration and persistence in the tumor tissues. In summary, the studies provide a good therapeutic option for the clinical treatment of solid tumors.


Subject(s)
Immunotherapy, Adoptive , Interleukin-12 , Mesothelin , Receptors, Chimeric Antigen , Animals , Interleukin-12/immunology , Interleukin-12/genetics , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Cell Line, Tumor , Mice , Xenograft Model Antitumor Assays , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/antagonists & inhibitors , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/therapy , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , T-Lymphocytes/immunology
3.
J Cancer Res Clin Oncol ; 149(16): 15027-15038, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37612388

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T cell therapy has shown promising results in treating blood cancers, but it has limited efficacy against solid tumors that express mesothelin (MSLN). One of the reasons is the immunosuppressive tumor microenvironment, which consists of physical barriers, multiple mechanisms of immune evasion, and various biochemical factors that favor tumor growth and survival. These factors reduce the antitumor activity of MSLN-targeted CAR T cells in clinical trials. Therefore, new therapeutic strategies are needed to enhance the effectiveness of MSLN-targeted CAR T cell therapy. METHODS: To investigate whether the antitumor efficacy of anti-MSLN CAR-T cells depends on the epitopes they recognize, we generated MSLN-targeted CAR T cells that bind to different regions of MSLN (Region I, II, III and Full length). We then evaluated the antitumor activity of MSLN-targeted CAR T cells alone or in combination with the chemotherapeutic drug irinotecan or an anti-PD-1 antibody in vitro and in vivo. RESULTS: We found that MSLN-targeted CAR T cells effectively killed MSLN-positive cancer cells (H9, H226 and Panc-1), but not MSLN-negative cells (A431) in vitro. In a mouse model of H9 tumor xenografts, all CAR T cells showed similar tumor suppression, but an MSLN-targeted scFv with Region I epitope, R47, performed slightly better. Combining irinotecan with CAR_R47 T cells enhanced tumor control synergistically in both H9 xenograft mice and patient-derived xenograft mice. CONCLUSIONS: Our results suggest that irinotecan can enhance the antitumor activity of MSLN-targeted CAR T cells, and offer a promising combination therapy strategy for MSLN-positive solid tumors.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Animals , Mice , Mesothelin , Immunotherapy, Adoptive/methods , Irinotecan , GPI-Linked Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Neoplasms/drug therapy
4.
Curr Med Sci ; 43(5): 855-868, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37558865

ABSTRACT

Smad ubiquitylation regulatory factor 1 (Smurf1) is an important homologous member of E6-AP C-terminus type E3 ubiquitin ligase. Initially, Smurf1 was reportedly involved in the negative regulation of the bone morphogenesis protein (BMP) pathway. After further research, several studies have confirmed that Smurf1 is widely involved in various biological processes, such as bone homeostasis regulation, cell migration, apoptosis, and planar cell polarity. At the same time, recent studies have provided a deeper understanding of the regulatory mechanisms of Smurf1's expression, activity, and substrate selectivity. In our review, a brief summary of recent important biological functions and regulatory mechanisms of E3 ubiquitin ligase Smurf1 is proposed.

SELECTION OF CITATIONS
SEARCH DETAIL
...