Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Res ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38683145

ABSTRACT

The prognosis of patients with acute myeloid leukemia (AML) is limited, especially for elderly or unfit patients not eligible for hematopoietic stem cell (HSC) transplantation. The disease is driven by leukemic stem cells (LSCs), which are characterized by clonal heterogeneity and resistance to conventional therapy. These cells are therefore believed to be a major cause of progression and relapse. We designed MP0533, a multispecific CD3-engaging DARPin (designed ankyrin repeat protein) that can simultaneously bind to three antigens on AML cells (CD33, CD123, and CD70), aiming to enable avidity-driven T cell-mediated killing of AML cells co-expressing at least two of the antigens. In vitro, MP0533 induced selective T cell-mediated killing of AML cell lines, as well as patient-derived AML blasts and LSCs, expressing two or more target antigens, while sparing healthy HSCs, blood, and endothelial cells. The higher selectivity also resulted in markedly lower levels of cytokine release in normal human blood compared to single antigen-targeting T-cell engagers. In xenograft AML mouse models, MP0533 induced tumor-localized T-cell activation and cytokine release, leading to complete eradication of the tumors while having no systemic adverse effects. These studies show that the multispecific-targeting strategy used with MP0533 holds promise for improved selectivity towards LSCs and efficacy against clonal heterogeneity, potentially bringing a new therapeutic option to this group of patients with high unmet need. MP0533 is currently being evaluated in a dose-escalation phase 1 study in patients with relapsed or refractory AML (NCT05673057).

2.
Nat Biotechnol ; 40(12): 1845-1854, 2022 12.
Article in English | MEDLINE | ID: mdl-35864170

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with potential resistance to existing drugs emphasizes the need for new therapeutic modalities with broad variant activity. Here we show that ensovibep, a trispecific DARPin (designed ankyrin repeat protein) clinical candidate, can engage the three units of the spike protein trimer of SARS-CoV-2 and inhibit ACE2 binding with high potency, as revealed by cryo-electron microscopy analysis. The cooperative binding together with the complementarity of the three DARPin modules enable ensovibep to inhibit frequent SARS-CoV-2 variants, including Omicron sublineages BA.1 and BA.2. In Roborovski dwarf hamsters infected with SARS-CoV-2, ensovibep reduced fatality similarly to a standard-of-care monoclonal antibody (mAb) cocktail. When used as a single agent in viral passaging experiments in vitro, ensovibep reduced the emergence of escape mutations in a similar fashion to the same mAb cocktail. These results support further clinical evaluation of ensovibep as a broad variant alternative to existing targeted therapies for Coronavirus Disease 2019 (COVID-19).


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Cricetinae , Humans , SARS-CoV-2/genetics , Designed Ankyrin Repeat Proteins , Cryoelectron Microscopy , Antibodies, Monoclonal/therapeutic use , Combined Antibody Therapeutics , Antibodies, Neutralizing
3.
Methods Mol Biol ; 988: 19-29, 2013.
Article in English | MEDLINE | ID: mdl-23475711

ABSTRACT

When recombinant glycoproteins for therapeutic use are to be produced on an industrial scale, there is a crucial need for technologies that can engineer fast-growing stable cells secreting the protein drug at a high rate and with a defined and safe glycosylation profile. Current cell lines approved for drug production are essentially from rodent origin. Their glycosylation machinery often adds undesired carbohydrate determinants which may alter protein folding, induce immunogenicity, and reduce circulatory life span of the drug. Notably, sialic acid as N-acetylneuraminic acid is not efficiently added in most mammalian cells and the 6-linkage is missing in rodent cells. Engineering cells with the various enzymatic activities required for sialic acid transfer has not yet succeeded in providing a human-like pattern of glycoforms to protein drugs. To date, there is a need for engineering animal cells and get highly sialylated products that resemble as closely as possible to human proteins. We have designed ST6Gal minigenes to optimize the ST6GalI sialyltransferase activity and used them to engineer ST6(+)CHO cells. When stably transfected in cells expressing a protein of interest or not, these constructs have proven to equip cell clones with efficient transfer activity of 6-linked sialic acid. In this chapter, we describe a methodology for generating healthy stable adherent clones with hypersialylation activity and high secretion rate.


Subject(s)
Glycoproteins/biosynthesis , Protein Engineering/methods , Protein Processing, Post-Translational , Sialyltransferases/biosynthesis , Animals , CHO Cells , Cricetinae , Gene Expression , Glycoproteins/genetics , Glycosylation , Humans , Sialyltransferases/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...