Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 26: 144-156, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-35795778

ABSTRACT

Pre-clinical humanized mouse models are a powerful tool to evaluate immunotherapies. NSG-SGM3 mice reconstituted with human stem cells (huSGM3) develop pronounced human myeloid cells due to transgenic expression of stem cell factor, granulocyte-macrophage colony-stimulating factor, and interleukin-3 (IL-3) compared with the widely used humanized NSG (huNSG) model. We assessed in vivo generation of CD19-CAR T cells in huSGM3 mice upon single intravenous injection of the T cell-specific lentiviral vectors (LVs) CD4-LV and CD8-LV. While in vivo CAR T cell generation was clearly detectable in individual mice, generation appeared less efficient than previously observed for huNSG mice. Especially for the CD4-LV group, this correlated with increased IL-15 and decreased GM-CSF levels, indicating activation of monocytes and macrophages. Co-culture assays identified macrophages as a potential barrier for gene transfer. Refining CD4-LV and CD8-LV with a less immunogenic surface by using modified packaging cells substantially improved the transduction of lymphocytes in vitro in the presence of macrophages, as well in vivo in huSGM3 mice. Notably, two mice that developed less CAR T cells showed high interferon-α or -ß levels before vector injection. Our data emphasize the relevance of innate immune responses for in vivo generation of CAR T cells, which can be overcome by vector surface engineering.

2.
Nat Protoc ; 16(7): 3210-3240, 2021 07.
Article in English | MEDLINE | ID: mdl-33846629

ABSTRACT

Receptor targeting of vector particles is a key technology to enable cell type-specific in vivo gene delivery. For example, T cells in humanized mouse models can be modified by lentiviral vectors (LVs) targeted to human T-cell markers to enable them to express chimeric antigen receptors (CARs). Here, we provide detailed protocols for the generation of CD4- and CD8-targeted LVs (which takes ~9 d in total). We also describe how to humanize immunodeficient mice with hematopoietic stem cells (which takes 12-16 weeks) and precondition (over 5 d) and administer the vector stocks. Conversion of the targeted cell type is monitored by PCR and flow cytometry of blood samples. A few weeks after administration, ~10% of the targeted T-cell subtype can be expected to have converted to CAR T cells. By closely following the protocol, sufficient vector stock for the genetic manipulation of 10-15 humanized mice is obtained. We also discuss how the protocol can be easily adapted to use LVs targeted to other types of receptors and/or for delivery of other genes of interest.


Subject(s)
Genetic Engineering/methods , T-Lymphocytes/metabolism , Animals , Antigens, CD/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Mice , Models, Animal , Receptors, Chimeric Antigen/genetics
3.
Blood Adv ; 4(22): 5702-5715, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33216892

ABSTRACT

Genetic modification of T lymphocytes is a key issue in research and therapy. Conventional lentiviral vectors (LVs) are neither selective for T cells nor do they modify resting or minimally stimulated cells, which is crucial for applications, such as efficient in vivo modification of T lymphocytes. Here, we introduce novel CD3-targeted LVs (CD3-LVs) capable of genetically modifying human T lymphocytes without prior activation. For CD3 attachment, agonistic CD3-specific single-chain variable fragments were chosen. Activation, proliferation, and expansion mediated by CD3-LVs were less rapid compared with conventional antibody-mediated activation owing to lack of T-cell receptor costimulation. CD3-LVs delivered genes not only selectively into T cells but also under nonactivating conditions, clearly outperforming the benchmark vector vesicular stomatitis-LV glycoproteins under these conditions. Remarkably, CD3-LVs were properly active in gene delivery even when added to whole human blood in absence of any further stimuli. Upon administration of CD3-LV into NSG mice transplanted with human peripheral blood mononuclear cells, efficient and exclusive transduction of CD3+ T cells in all analyzed organs was achieved. Finally, the most promising CD3-LV successfully delivered a CD19-specific chimeric antigen receptor (CAR) into T lymphocytes in vivo in humanized NSG mice. Generation of CAR T cells was accompanied by elimination of human CD19+ cells from blood. Taken together, the data strongly support implementation of T-cell-activating properties within T-cell-targeted vector particles. These particles may be ideally suited for T-cell-specific in vivo gene delivery.


Subject(s)
Genetic Vectors , Lentivirus , Animals , Lentivirus/genetics , Leukocytes, Mononuclear , Mice , T-Lymphocytes , Transduction, Genetic
4.
Front Immunol ; 11: 2028, 2020.
Article in English | MEDLINE | ID: mdl-32983147

ABSTRACT

Natural killer (NK) cells are a noteworthy lymphocyte subset in cancer adoptive cell therapy. NK cells initiate innate immune responses against infections and malignancies with natural cytotoxicity, which is independent of foreign antigen recognition. Based on these substantive features, genetically modifying NK cells is among the prime goals in immunotherapy but is currently difficult to achieve. Recently, we reported a fully human CAR19 construct (huCAR19) with remarkable function in gene-modified T-cells. Here, we show efficient and stable gene delivery of huCAR19 to primary human NK cells using lentiviral vectors with transduction efficiencies comparable to those achieved with NK cell lines. These huCAR19 NK cells display specific and potent cytotoxic activity against target cells. To improve homing of NK cells to the bone marrow, we augmented huCAR19 NK cells with the human CXCR4 gene, resulting in transgenically augmented CAR NK cells (TRACKs). Compared to conventional CAR NK cells, TRACKs exhibit enhanced migration capacity in response to recombinant SDF-1 or bone marrow stromal cells while retaining functional and cytolytic activity against target cells. Based on these promising findings, TRACKs may become a novel candidate for immunotherapeutic strategies in clinical applications.


Subject(s)
Gene Expression , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, CXCR4/genetics , Receptors, Chimeric Antigen/metabolism , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cell Movement , Chemotaxis/genetics , Chemotaxis/immunology , Cytotoxicity, Immunologic , Humans , Immunomodulation , Immunophenotyping , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Transduction, Genetic
5.
Mol Ther ; 28(8): 1783-1794, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32485137

ABSTRACT

T cells modified with CD19-specific chimeric antigen receptors (CARs) result in significant clinical benefit for leukemia patients but constitute a challenge for manufacturing. We have recently demonstrated the in vivo generation of CD19-CAR T cells using the CD8-targeted lentiviral vector (CD8-LV). In this study, we investigated the in vivo generation of CD4+ CAR T cells using CD4-targeted LV (CD4-LV). Administration of CD4-LV into NSG mice transplanted with human peripheral blood mononuclear cells (PBMCs) led to 40%-60% of human CD4+ lymphocytes being CAR positive while CD8+ cells remained CAR negative. CAR+ T cells displayed a T helper 1 (Th1)/Th2 phenotype, which was accompanied by CD19+ B cell elimination. Intravenous administration of CD4-LV into NSG mice reconstituted with human CD34+ cells induced CAR expression and B cell elimination within 2-3 weeks post-injection. Preclinical analysis in a tumor mouse model revealed that mice administered CD4-LV exhibited faster and superior tumor cell killing compared to mice injected with CD8-LV alone or as a mixture with CD4-LV. Further analysis suggests that CD4+CAR+ cells may outperform CD8+CAR+ cells, especially at a high burden of target antigen, mainly since CD8 cells are more prone to exhaustion. This first description of in vivo-generated CD4+ CAR T cells supports their importance for cellular therapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Animals , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
Oncoimmunology ; 8(12): e1671761, 2019.
Article in English | MEDLINE | ID: mdl-31741773

ABSTRACT

Chimeric antigen receptor (CAR) T cells are in prime focus of current research in cancer immunotherapy. Facilitating CAR T cell generation is among the top goals. We have recently demonstrated direct in vivo generation of human CD19-CAR T cells by targeting CD8+ cells using lentiviral vectors (LVs). The anti-tumor potency of in vivo generated CAR T cells was assessed in human PBMC-transplanted NSG mice carrying i.v. injected CD19+ Nalm-6 tumor cells. A single injection of CD8-targeted LV delivering CD19-CAR was sufficient to completely eliminate the tumor cells from bone marrow and spleen, whereas control animals contained high levels of CD19+ cells. Tumor elimination was due to in vivo generated CAR+ cells. Notably, these were not only composed of T lymphocytes but also included CAR+ natural killer cells (NK and NKT). This is the first demonstration of tumor elimination by in vivo generated human CAR T cells.

7.
PLoS One ; 8(12): e82121, 2013.
Article in English | MEDLINE | ID: mdl-24324757

ABSTRACT

AIMS: In the present study we have investigated the comparative switching propensity of murine peritoneal and splenic B cell subpopulations to IgA in presence of retinoic acid (RA) and TGF-ß. METHODS AND RESULTS: To study the influence of RA and TGF-ß on switching of B cell subpopulations to IgA, peritoneal (B1a, B1b and B2 cells) and splenic (B1a, marginal zone, and B2) B cells from normal BALB/c mice were FACS purified, cultured for 4 days in presence of RA and TGF-ß and the number of IgA producing cells was determined by ELISPOT assay or FACS analysis. In presence of TGF-ß, peritoneal B1b cells switched to IgA more potently than other peritoneal B cell subpopulations. When TGF-ß was combined with retinoic acid (RA), switching to IgA was even more pronounced. Under these conditions, "innate" B cells like peritoneal and splenic B1 cells and MZ B cells produced IgA more readily than B2 cells. Additionally, high frequency of nucleotide exchanges indicating somatic hypermutation in VH regions was observed. Besides IgA induction, RA treatment of sorted PEC and splenic B cells led to expression of gut homing molecules - α4ß7 and CCR9. Intraperitoneal transfer of RA-treated B1 cells into Rag1(-/-) recipients resulted in IgA in serum and gut lavage, most efficiently amongst B1b cell recipients. CONCLUSION: Present study demonstrates the differential and synergistic effect of RA and TGF-ß on switching of different B cell subpopulations to IgA and establishes the prominence of peritoneal B1b cells in switching to IgA under the influence of these two factors. Our study extends our knowledge about the existing differences among B cell subpopulations with regards to IgA production and indicates towards their differential contribution to gut associated humoral immunity.


Subject(s)
Immunoglobulin A/metabolism , Peritoneum/cytology , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Animals , Cell Count , Cell Line , Cell Movement/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Humans , Immunoglobulin A/blood , Immunoglobulin Class Switching/drug effects , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Somatic Hypermutation, Immunoglobulin/drug effects , Spleen/cytology , Spleen/drug effects
8.
Indian J Crit Care Med ; 17(2): 111-2, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23983418

ABSTRACT

Children in rural India are a vulnerable group for snake bites. Improper elicitation of history and atypical presentations could lead to misdiagnosis and delay in treatment. We are reporting the case of an 8-year-old male child who presented with convulsions, unconsciousness and hypertension who was initially managed as a case of hypertensive encephalopathy showing no sign of improvement even after 20 hs. The history when reviewed suggested neurotoxic snake bite although the patient did not have any classical local findings. Anti-snake venom administration was followed by prompt recovery. We therefore suggest that snake bite should be considered in patients from rural background presenting with hypertension, convulsion and unconsciousness, even in the absence of classical features of snake bite.

9.
Eur J Immunol ; 43(8): 2023-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23677546

ABSTRACT

In mice, B-1 (B1a/B1b) cells are mainly located in the peritoneal cavity. B-1 cells are well known for their role in the early stages of Ab-mediated immune responses against pathogenic invasion as well as for the production of natural IgM antibodies. Although such B cells have been claimed to give rise to intestinal plasma cells producing IgA, a clear role of B-1 cells in IgA production in the gut-associated tissues is still not defined. Here, we employed the transgenic L2 mouse model characterized by the lack of B-2 cells and presence of B-1 cells as major B-cell subpopulation. The oligoclonality of the Ab repertoire in this mouse allowed us to take typical B1a cell VH sequences as indicators of the presence of IgM-producing B-1a cells in Peyer's patches as well as in lamina propria. However, amongst the IgAVH sequences recovered from the same tissues, none of the sequences showed B1a-cell specificity. Interestingly, all IgAVH sequences derived from the lamina propria of L2 mice displayed extensive numbers of nucleotide exchanges, indicating somatic hypermutation, and affinity maturation. This suggests that the contribution of natural unmutated IgA by B-1a cells to intestinal immunity is negligible.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunoglobulin A , Immunoglobulin Heavy Chains/immunology , Immunoglobulin M , Immunoglobulin Variable Region/immunology , Animals , Immunoglobulin A/biosynthesis , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin Class Switching , Immunoglobulin Heavy Chains/genetics , Immunoglobulin M/biosynthesis , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Immunoglobulin Variable Region/genetics , Intestinal Mucosa/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Mucous Membrane/immunology , Peritoneal Cavity , Peyer's Patches/immunology , Somatic Hypermutation, Immunoglobulin
10.
J Anaesthesiol Clin Pharmacol ; 28(1): 45-50, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22345945

ABSTRACT

CONTEXT: Snake bites are the common cause of morbidity and mortality in tropical countries. AIMS: To analyze the outcome of snake bite victims SETTINGS AND DESIGN: Retrospective analysis of data from Intensive care unit, Department of Anesthesiology. MATERIALS AND METHODS: All the patients admitted in the intensive care unit for snake bite management during the year May 2004 - April 2009 were retrospectively reviewed. The data included age, sex, month and time of incident, site of bite, dose of anti--snake venom, time of anti--snake venom, administration, duration of mechanical ventilation, complications and death of a victim. STATISTICAL ANALYSIS USED: Pearson's correlation test, paired samples t-test. RESULTS AND CONCLUSIONS: 113 patients reported to the Accident and Emergency with history of snake bite. 26 patients were referred to other hospital, 17 patients were brought dead, and 70 patients were admitted to the intensive care unit. In 59 snake-bite victims, maximum data could be recovered. Krait was the most common type of snake bite reported. There was a male preponderance (69.4%) with age ranging between 20 and 40 years (52.5%). The mean lag time (time elapsed between bite and first dose of anti--snake venom) was 5.3 ± 1.4 h and the mean anti-snake venom dose was 12.3 ± 2.4 vials. There was a positive and significant correlation between lag time and total dose of anti--snake venom (correlation coefficient =0.956, P<0.0001). Overall 72.9% patients required mechanical ventilation with a mean duration of 56.2 ± 16.1 h. 10.2% patients sustained cardiac arrest, 8.7% patients developed ventilator associated pneumonia, 6.7% suffered mild anti-snake venom reaction, 6.7% had hypotension and 5.1% patients developed renal failure. The overall mortality was 5.1%.

SELECTION OF CITATIONS
SEARCH DETAIL
...