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.
Hum Gene Ther ; 31(11-12): 679-691, 2020 06.
Article in English | MEDLINE | ID: mdl-32160795

ABSTRACT

Adoptive T cell immunotherapy in combination with gene therapy is a promising treatment concept for chronic infections and cancer. Recently, receptor-targeted lentiviral vectors (LVs) were shown to enable selective gene transfer into particular types of lymphocytes both in vitro and in vivo. This approach might facilitate the genetic engineering of a patient's own T lymphocytes, possibly even shifting this concept from personalized medicine to an off-the shelf therapy in future. Here, we describe novel high-affinity binders for CD8 consisting of designed ankyrin repeat proteins (DARPins), which were selected to bind to the CD8 receptor of human and nonhuman primate (NHP) cells. These binders were identified by ribosome display screening of DARPin libraries using recombinant human CD8 followed by receptor binding analysis on primary lymphocytes. CD8-targeted LVs (CD8-LVs) were then generated that delivered genes exclusively and specifically to human and NHP T lymphocytes by using the same targeting domain. These CD8-LVs were as specific for human T lymphocytes as their single-chain variable fragment-based counterpart, but they could be produced to higher titers. Moreover, they were superior in transducing cytotoxic T cells both in vitro and in vivo when equal particle numbers were applied. Since the here described CD8-LVs transduced primary T lymphocytes from NHP and human donors equally well, they offer the opportunity for preclinical studies in different animal models including large animals such as NHPs without the need for modifications in vector design.


Subject(s)
Ankyrin Repeat , CD8-Positive T-Lymphocytes/metabolism , Genetic Vectors , Receptors, Antigen, T-Cell/metabolism , Single-Chain Antibodies/genetics , Animals , Cell Line , Chronic Disease/therapy , Gene Transfer Techniques , Genetic Therapy/methods , HEK293 Cells , Humans , Lentivirus , Leukocytes, Mononuclear , Macaca mulatta , Macaca nemestrina , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes, Cytotoxic/metabolism , Transduction, Genetic
2.
PLoS One ; 7(10): e46667, 2012.
Article in English | MEDLINE | ID: mdl-23071609

ABSTRACT

Lentiviral vectors (LVs) are potent gene transfer vehicles frequently applied in research and recently also in clinical trials. Retargeting LV entry to cell types of interest is a key issue to improve gene transfer safety and efficacy. Recently, we have developed a targeting method for LVs by incorporating engineered measles virus (MV) glycoproteins, the hemagglutinin (H), responsible for receptor recognition, and the fusion protein into their envelope. The H protein displays a single-chain antibody (scFv) specific for the target receptor and is ablated for recognition of the MV receptors CD46 and SLAM by point mutations in its ectodomain. A potential hindrance to systemic administration in humans is pre-existing MV-specific immunity due to vaccination or natural infection. We compared transduction of targeting vectors and non-targeting vectors pseudotyped with MV glycoproteins unmodified in their ectodomains (MV-LV) in presence of α-MV antibody-positive human plasma. At plasma dilution 1:160 MV-LV was almost completely neutralized, whereas targeting vectors showed relative transduction efficiencies from 60% to 90%. Furthermore, at plasma dilution 1:80 an at least 4-times higher multiplicity of infection (MOI) of MV-LV had to be applied to obtain similar transduction efficiencies as with targeting vectors. Also when the vectors were normalized to their p24 values, targeting vectors showed partial protection against α-MV antibodies in human plasma. Furthermore, the monoclonal neutralizing antibody K71 with a putative epitope close to the receptor binding sites of H, did not neutralize the targeting vectors, but did neutralize MV-LV. The observed escape from neutralization may be due to the point mutations in the H ectodomain that might have destroyed antibody binding sites. Furthermore, scFv mediated cell entry via the target receptor may proceed in presence of α-MV antibodies interfering with entry via the natural MV receptors. These results are promising for in vivo applications of targeting vectors in humans.


Subject(s)
Lentivirus/genetics , Measles virus/genetics , Transduction, Genetic , Viral Fusion Proteins/genetics , Virus Internalization , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Genetic Therapy/methods , Genetic Vectors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , HEK293 Cells , Hemagglutinins, Viral/genetics , Humans , Lentivirus/immunology , Lentivirus/physiology , Measles virus/immunology , Recombinant Fusion Proteins/genetics
3.
Virology ; 418(2): 85-92, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21835422

ABSTRACT

The R peptide in the cytoplasmic tail (C-tail) of γ-retroviral envelope proteins (Env) prevents membrane fusion before budding. To analyse its role in the formation of replication competent, infectious particles, we developed chimeric murine leukaemia viruses (MLV) with unmodified or R-peptide deleted Env proteins of the gibbon ape leukaemia virus (GaLV). While titres of these viruses were unaffected, R-peptide deficiency led to strongly impaired spreading. Most remarkably, we isolated an escape mutant which had restored an open reading frame for a C-terminal extension of the truncated C-tail. A reconstituted virus encoding this escape C-tail replicated in cell culture. In contrast to R-peptide deficient Env, particle incorporation of the escape Env was effective due to an enhanced protein expression and restored intracellular co-localisation with Gag proteins. Our data demonstrate that the R peptide not only regulates membrane fusion but also mediates efficient Env protein particle incorporation in γ-retrovirus infected cells.


Subject(s)
Gene Deletion , Gene Expression Regulation, Viral/physiology , Oligopeptides/metabolism , Retroviridae/classification , Retroviridae/genetics , Animals , Cell Line , Humans , Oligopeptides/genetics , Reassortant Viruses/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly , Virus Replication
4.
Nat Methods ; 7(11): 929-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20935652

ABSTRACT

We present a flexible and highly specific targeting method for lentiviral vectors based on single-chain antibodies recognizing cell-surface antigens. We generated lentiviral vectors specific for human CD105(+) endothelial cells, human CD133(+) hematopoietic progenitors and mouse GluA-expressing neurons. Lentiviral vectors specific for CD105 or for CD20 transduced their target cells as efficiently as VSV-G pseudotyped vectors but discriminated between endothelial cells and lymphocytes in mixed cultures. CD133-targeted vectors transduced CD133(+) cultured hematopoietic progenitor cells more efficiently than VSV-G pseudotyped vectors, resulting in stable long-term transduction. Lentiviral vectors targeted to the glutamate receptor subunits GluA2 and GluA4 exhibited more than 94% specificity for neurons in cerebellar cultures and when injected into the adult mouse brain. We observed neuron-specific gene modification upon transfer of the Cre recombinase gene into the hippocampus of reporter mice. This approach allowed targeted gene transfer to many cell types of interest with an unprecedented degree of specificity.


Subject(s)
Endothelial Cells/metabolism , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Neurons/metabolism , AC133 Antigen , Animals , Antigens, CD/genetics , Antigens, CD20/genetics , Cells, Cultured , Glycoproteins/genetics , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Peptides/genetics , Receptors, AMPA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...