Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 137: 112424, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38878486

ABSTRACT

Colorectal cancer is a major global health burden, with limited efficacy of traditional treatment modalities in improving survival rates. However, recently advances in immunotherapy has improved treatment outcomes for patients with this cancer. To address the continuing need for improved treatment efficacy, this study introduced a novel tri-specific antibody, IMT030122, that targets EpCAM, 4-1BB, and CD3. We evaluated the pharmacological efficacy and mechanism of action of IMT030122 in vitro and in vivo. In in vitro studies, IMT030122 exhibited differential binding to antigens and cells expressing EpCAM, 4-1BB, and CD3. Moreover, IMT030122 relied on EpCAM-targeted activation of intracellular CD3 and 4-1BB signaling and mediated T cell cytotoxicity specific to HCT116 colorectal cancer cells. In vivo, IMT030122 demonstrated potent anti-tumor activity, significantly inhibiting the growth of colon cancer HCT116 and MC38-hEpCAM subcutaneous grafts. Further pharmacological analysis revealed that IMT030122 recruited lymphocytes from peripheral blood into colorectal cancer tissue and exerted durable anti-tumor activity, predominantly by promoting the activation, proliferation, and differentiation of CD8T cells. Notably, IMT030122 still exhibited anti-tumor efficacy even in the presence of significantly depleted lymphocytes in colorectal cancer tissue. The potent pharmacological activity and anti-tumor effects of IMT030122 suggest it may enhance treatment efficacy and substantially extend the survival of patients with colorectal cancer in the future.


Subject(s)
CD3 Complex , Colorectal Neoplasms , Epithelial Cell Adhesion Molecule , Animals , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Epithelial Cell Adhesion Molecule/metabolism , CD3 Complex/immunology , Mice , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , HCT116 Cells , Xenograft Model Antitumor Assays , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Female , Cell Line, Tumor , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Mice, Inbred BALB C , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Immunotherapy/methods
2.
Cancer Immunol Immunother ; 72(12): 4015-4030, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37863852

ABSTRACT

BACKGROUND: Additional immunotherapies are still warranted for non-responders to checkpoint inhibitors with refractory or relapsing cancers, especially for patients with "cold" tumours lacking significant immune infiltration at treatment onset. We developed XFab-α4-1BB/CD40L, a bispecific antibody targeting 4-1BB and CD40 for dendritic cell activation and priming of tumour-reactive T cells to inhibit tumours. METHODS: XFab-α4-1BB/CD40L was developed by engineering an anti-4-1BB Fab arm into a CD40L trimer based on XFab® platform. Characterisation of the bispecific antibody was performed by cell-based reporter assays, maturation of dendritic cell assays, and mixed lymphocyte reactions. The abilities of antigen-specific T-cell expansion and antitumour efficacy were assessed in syngeneic mouse tumour models. Toxicological and pharmacodynamic profiles were investigated in non-human primates. RESULTS: XFab-α4-1BB/CD40L demonstrated independent CD40 agonistic activity and conditional 4-1BB activity mediated by CD40 crosslinking, leading to dendritic cell maturation and T-cell proliferation in vitro. We confirmed the expansion of antigen-specific T cells in the vaccination model and potent tumour regression induced by the bispecific antibody alone or in combination with gemcitabine in vivo, concomitant with improved tumour-reactive T-cell infiltration. XFab-α4-1BB/CD40L showed no signs of liver toxicity at doses up to 51 mg/kg in a repeated-dose regimen in non-human primates. CONCLUSIONS: XFab-α4-1BB/CD40L is capable of enhancing antitumour immunity by modulating dendritic cell and T-cell functions via targeting 4-1BB agonism to areas of CD40 expression. The focused, potent, and safe immune response induced by the bispecific antibody supports further clinical investigations for the treatment of solid tumours.


Subject(s)
CD40 Ligand , Neoplasms , Humans , Mice , Animals , T-Lymphocytes/metabolism , Neoplasms/therapy , Neoplasms/metabolism , CD40 Antigens , Primates/metabolism , Dendritic Cells
3.
Comp Med ; 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37072182

ABSTRACT

Current follicle-stimulating hormone (FSH) drugs meet safety criteria but have suboptimal efficacy, poor patient compliance, and high cost. Alternative FSH-like drugs would help to meet the high market demand. Here, we evaluated X002, an FSH-Fc fusion protein, for bioactivity and half-life in vitro and in vivo. In all cases, the effects of X002 were compared with those of a commercially available short-acting FSH recombinant hormone. First, female Kunming mice (age, 21 to 24 d) were stimulated with pregnant mare serum gonadotropin (PMSG) for 46 h, after which naked oocytes were harvested, treated with X002 or the comparison agent at 37 °C for 4 h, and then evaluated for germinal vesicle breakdown. Second, cumulus-oocyte complexes (COC) were collected from PMSG-stimulated mice and cocultured with X002 or the comparison agent for 14 h; the COC diameters were then measured, and the expression of genes involved in COC expansion were evaluated using quantitative RT-PCR analysis. Third, to assess the pharmacokinetics of X002, female Sprague-Dawley rats (age, 6 to 8 wk) were injected subcutaneously with X002 or the comparison agent; serum samples then were collected at various times and assessed via ELISA. Fourth, to evaluate X002 pharmacodynamics, 26-d-old female Sprague-Dawley rats were treated with X002 or the comparison agent; 84 h later, the rats were stimulated with human chorionic gonadotropin (hCG). At 12 h after hCG injection, euthanasia was performed. Ovaries were removed and weighed, and serum levels of estradiol and progesterone were measured. Finally, to assess superovulation, the oocytes in the fallopian tubes were counted at 108 h after in vivo treatment of rats with X002 or the comparison agent. The data show that X002, a long-acting agent, promoted germinal vesicle breakdown and COC expansion in vitro and in vivo ovarian weight gain and superovulation to a degree similar to the short-acting comparison agent.

4.
Int J Mol Sci ; 23(19)2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36233320

ABSTRACT

Bio-macromolecules have potential applications in cancer treatment due to their high selectivity and efficiency in hitting therapeutic targets. However, poor cell membrane permeability has limited their broad-spectrum application in cancer treatment. The current study developed highly internalizable anti-c-MET antibody Fab fusion proteins with intracellular epitope peptide chimera to achieve the dual intervention from the extracellular to intracellular targets in tumor therapy. In vitro experiments demonstrated that the fusion proteins could interfere with the disease-associated intracellular signaling pathways and inhibit the uncontrolled proliferation of tumor cells. Importantly, investigation of the underlying mechanism revealed that these protein chimeras could induce vacuolation in treated cells, thus interfering with the normal extension and arrangement of microtubules as well as the mitosis, leading to the induction of methuosis-mediated cell death. Furthermore, in vivo tumor models indicated that certain doses of fusion proteins could inhibit the A549 xenograft tumors in NOD SCID mice. This study thus provides new ideas for the intracellular delivery of bio-macromolecules and the dual intervention against tumor cell signaling pathways.


Subject(s)
Proto-Oncogene Proteins c-met , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibodies/metabolism , Epitopes , GRB2 Adaptor Protein/metabolism , Humans , Mice , Mice, SCID , Peptides/chemistry , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism
5.
Immunobiology ; 227(6): 152283, 2022 11.
Article in English | MEDLINE | ID: mdl-36198215

ABSTRACT

The claudin 18.2(CLDN18.2) antigen is highly expressed in gastric mucosa epithelial cells and frequently expressed in malignant tumors. Positive clinical outcomes have popularized claudin 18.2 as a novel cellular and antibody therapeutic. Here, we designed a bispecific antibody-ZWB67 using the XFab® platform, aimed at redirecting CD3+ effector T cells to CLDN18.2+ target cells or tissues. Physicochemical characterization, binding properties, T cell stimulatory activity, and T cell-dependent cellular cytotoxicity of ZWB67 were evaluated in dosage intervals using antigens of CD3 and target cells expressing CLDN18.2 or CD3. Then, the anti-tumor activity was assessed in humanized CD3EDG mice bearing MC-38-hCLDN18.2 tumors. Our data demonstrate that ZWB67 specifically binds to the human CD3e antigen (KD = 1.04E-08 M) and binds more strongly to CLDN18.2+ cells than to CD3+ cells (4.3- to 9.2-fold difference). ZWB67 showed good activity in the luciferase reporter system and exhibited dose-dependent activation, cytotoxicity of T cells, and cytokine release when co-cultured with CLDN18.2+ cells and CD3+ T cells. ZWB67 also exhibited high in vivo efficacy in the MC-38-hCLDN18.2 xenograft mouse model. In conclusion, the novel anti-CLDN18.2 × anti-CD3 bispecific antibody exhibited low affinity for anti-CD3, highly specific binding, potent cytotoxicity, and anti-tumor activity. These data provide a basis for future preclinical and clinical development of this therapeutic strategy.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Mice , Animals , CD3 Complex , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , T-Lymphocytes , Neoplasms/drug therapy , Claudins
SELECTION OF CITATIONS
SEARCH DETAIL
...