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.
Mol Cancer Ther ; 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38670552

ABSTRACT

Delta-like ligand 3 (DLL3) is expressed in more than 70% of small cell lung cancers (SCLCs) and other neuroendocrine-derived tumor types. SCLC is highly aggressive and limited therapeutic options lead to poor prognosis for patients. HPN328 is a tri-specific T cell activating construct (TriTAC) consisting of three binding domains: a CD3 binder for T cell engagement, an albumin binder for half-life extension, and a DLL3 binder for tumor cell engagement. In vitro assays, rodent models and non-human primates were used to assess the activity of HPN328. HPN328 induces potent dose-dependent killing of DLL3-expressing SCLC cell lines in vitro concomitant with T cell activation and cytokine release. In an NCI-H82 xenograft model with established tumors, HPN328 treatment led to T cell recruitment and anti-tumor activity. In an immunocompetent mouse model expressing a human CD3ε epitope, mice previously treated with HPN328 withstood tumor rechallenge, demonstrating long-term anti-tumor immunity. When repeat doses were administered to cynomolgus monkeys, HPN328 was well tolerated up to 10 mg/kg. Pharmacodynamic changes, such as transient cytokine elevation, were observed, consistent with the expected mechanism of action of T cell engagers. HPN328 exhibited linear pharmacokinetic in the given dose range with a serum half-life of 78 to 187 hours, supporting weekly or less frequent administration of HPN328 in humans. Preclinical and nonclinical characterization suggests that HPN328 is a highly efficacious, safe, and novel therapeutic candidate. A phase 1/2 clinical trial is currently underway testing safety and efficacy in patients with DLL3 expressing malignancies.

2.
Mol Cancer Ther ; 20(1): 109-120, 2021 01.
Article in English | MEDLINE | ID: mdl-33203731

ABSTRACT

T cells have a unique capability to eliminate cancer cells and fight malignancies. Cancer cells have adopted multiple immune evasion mechanisms aimed at inhibiting T cells. Dramatically improved patient outcomes have been achieved with therapies genetically reprogramming T cells, blocking T-cell inhibition by cancer cells, or transiently connecting T cells with cancer cells for redirected lysis. This last modality is based on antibody constructs that bind a surface antigen on cancer cells and an invariant component of the T-cell receptor. Although high response rates were observed with T-cell engagers specific for CD19, CD20, or BCMA in patients with hematologic cancers, the treatment of solid tumors has been less successful. Here, we developed and characterized a novel T-cell engager format, called TriTAC (for Trispecific T-cell Activating Construct). TriTACs are engineered with features to improve patient safety and solid tumor activity, including high stability, small size, flexible linkers, long serum half-life, and highly specific and potent redirected lysis. The present study establishes the structure/activity relationship of TriTACs and describes the development of HPN424, a PSMA- (FOLH1-) targeting TriTAC in clinical development for patients with metastatic castration-resistant prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , T-Lymphocytes/metabolism , Albumins/pharmacology , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , CD3 Complex/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Half-Life , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macaca fascicularis , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Prostate-Specific Antigen/metabolism , T-Lymphocytes/drug effects
3.
Clin Cancer Res ; 27(5): 1452-1462, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33262134

ABSTRACT

PURPOSE: Mesothelin (MSLN) is a glycophosphatidylinositol-linked tumor antigen overexpressed in a variety of malignancies, including ovarian, pancreatic, lung, and triple-negative breast cancer. Early signs of clinical efficacy with MSLN-targeting agents have validated MSLN as a promising target for therapeutic intervention, but therapies with improved efficacy are still needed to address the significant unmet medical need posed by MSLN-expressing cancers. EXPERIMENTAL DESIGN: We designed HPN536, a 53-kDa, trispecific, T-cell-activating protein-based construct, which binds to MSLN-expressing tumor cells, CD3ε on T cells, and to serum albumin. Experiments were conducted to assess the potency, activity, and half-life of HPN536 in in vitro assays, rodent models, and in nonhuman primates (NHP). RESULTS: HPN536 binds to MSLN-expressing tumor cells and to CD3ε on T cells, leading to T-cell activation and potent redirected target cell lysis. A third domain of HPN536 binds to serum albumin for extension of plasma half-life. In cynomolgus monkeys, HPN536 at doses ranging from 0.1 to 10 mg/kg demonstrated MSLN-dependent pharmacologic activity, was well tolerated, and showed pharmacokinetics in support of weekly dosing in humans. CONCLUSIONS: HPN536 is potent, is well tolerated, and exhibits extended half-life in NHPs. It is currently in phase I clinical testing in patients with MSLN-expressing malignancies (NCT03872206).


Subject(s)
Immunotherapy/methods , Lymphocyte Activation/immunology , Mesothelin/antagonists & inhibitors , Neoplasms/drug therapy , Single-Domain Antibodies/pharmacology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis , Cell Proliferation , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Peptide Fragments/immunology , Single-Domain Antibodies/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Biotechnol Prog ; 31(2): 431-7, 2015.
Article in English | MEDLINE | ID: mdl-25683459

ABSTRACT

Virus clearance by depth filtration has not been well-understood mechanistically due to lack of quantitative data on filter charge characteristics and absence of systematic studies. It is generally believed that both electrostatic interactions and sized based mechanical entrapment contribute to virus clearance by depth filtration. In order to establish whether the effectiveness of virus clearance correlates with the charge characteristics of a given depth filter, a counter-ion displacement technique was employed to determine the ionic capacity for several depth filters. Two depth filters (Millipore B1HC and X0HC) with significant differences in ionic capacities were selected and evaluated for their ability to eliminate viruses. The high ionic capacity X0HC filter showed complete porcine parvovirus (PPV) clearance (eliminating the spiked viruses to below the limit of detection) under low conductivity conditions (≤2.5 mS/cm), achieving a log10 reduction factor (LRF) of > 4.8. On the other hand, the low ionic capacity B1HC filter achieved only ∼2.1-3.0 LRF of PPV clearance under the same conditions. These results indicate that parvovirus clearance by these two depth filters are mainly achieved via electrostatic interactions between the filters and PPV. When much larger xenotropic murine leukemia virus (XMuLV) was used as the model virus, complete retrovirus clearance was obtained under all conditions evaluated for both depth filters, suggesting the involvement of mechanisms other than just electrostatic interactions in XMuLV clearance.


Subject(s)
Filtration/instrumentation , Filtration/methods , Viruses/isolation & purification , Animals , Cats , Cell Line , Osmolar Concentration , Parvovirus, Porcine , Static Electricity , Swine , Xenotropic murine leukemia virus-related virus
5.
Bioinformation ; 5(3): 132-5, 2010 Sep 20.
Article in English | MEDLINE | ID: mdl-21364793

ABSTRACT

MicroRNAs have been implicated for the regulation of gene expression. These miRNA are a class of single stranded non coding RNAs, formed from endogenous transcripts and measure typically about 19-25 nucleotides in length. They are important regulators of the various biological and metabolic functions taking place in humans. Many miRNAs show tissue specific expression. Human heart is a complex organ which during various diseased and developed conditions shows differential expression of miRNA. Here, we overview the recent findings on miRNA in cardiac diseases and report the presence of high AU content in differentially expressed miRNA in developed and diseased condition of heart as compared to all the miRNA present in the human. A total of 905 human miRNA sequences taken from miRBase were computationally analyzed. Trend analysis was performed to study the influence of positional frequency of the nucleotides. This study will help us in understanding the significance of AU rich elements in miRNA during the development of cardiac diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...