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










Publication year range
1.
Cell Rep Med ; 2(4): 100247, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33948577

ABSTRACT

Sickle cell disease (SCD) is caused by a 20A > T mutation in the ß-globin gene. Genome-editing technologies have the potential to correct the SCD mutation in hematopoietic stem cells (HSCs), producing adult hemoglobin while simultaneously eliminating sickle hemoglobin. Here, we developed high-efficiency viral vector-free non-footprint gene correction in SCD CD34+ cells with electroporation to deliver SCD mutation-targeting guide RNA, Cas9 endonuclease, and 100-mer single-strand donor DNA encoding intact ß-globin sequence, achieving therapeutic-level gene correction at DNA (∼30%) and protein (∼80%) levels. Gene-edited SCD CD34+ cells contributed corrected cells 6 months post-xenograft mouse transplant without off-target δ-globin editing. We then developed a rhesus ß-to-ßs-globin gene conversion strategy to model HSC-targeted genome editing for SCD and demonstrate the engraftment of gene-edited CD34+ cells 10-12 months post-transplant in rhesus macaques. In summary, gene-corrected CD34+ HSCs are engraftable in xenograft mice and non-human primates. These findings are helpful in designing HSC-targeted gene correction trials.


Subject(s)
Anemia, Sickle Cell/genetics , Antigens, CD34/metabolism , Hematopoietic Stem Cells/metabolism , Heterografts/immunology , Macaca mulatta/genetics , Animals , Gene Editing/methods , Gene Targeting/methods , Hematopoietic Stem Cell Transplantation/methods , Hemoglobin, Sickle/genetics , Humans , Mice , RNA, Guide, Kinetoplastida/metabolism , beta-Globins/genetics
2.
Hum Gene Ther ; 29(5): 614-625, 2018 05.
Article in English | MEDLINE | ID: mdl-29334771

ABSTRACT

CD19-targeted chimeric antigen receptor (CAR) engineered T/natural killer (NK)-cell therapies can result in durable clinical responses in B-cell malignancies. However, CAR-based immunotherapies have been much less successful in solid cancers, in part due to "on-target off-tumor" toxicity related to expression of target tumor antigens on normal tissue. Based on preliminary observations of safety and clinical activity in proof-of-concept clinical trials, tumor antigen-specific messenger RNA (mRNA) CAR transfection into selected, activated, and expanded T/NK cells may permit prospective control of "on-target off-tumor" toxicity. To develop a commercial product for solid tumors, mesothelin was selected as an antigen target based on its association with poor prognosis and overexpression in multiple solid cancers. It was hypothesized that selecting, activating, and expanding cells ex vivo prior to mRNA CAR transfection would not be necessary, thus simplifying the complexity and cost of manufacturing. Now, the development of anti-human mesothelin mRNA CAR transfected peripheral blood lymphocytes (CARMA-hMeso) is reported, demonstrating the manufacture and cryopreservation of multiple cell aliquots for repeat administrations from a single human leukapheresis. A rapid, automated, closed system for cGMP-compliant transfection of mRNA CAR in up to 20 × 109 peripheral blood lymphocytes was developed. Here we show that CARMA-hMeso cells recognize and lyse tumor cells in a mesothelin-specific manner. Expression of CAR was detectable over approximately 7 days in vitro, with a progressive decline of CAR expression that appears to correlate with in vitro cell expansion. In a murine ovarian cancer model, a single intraperitoneal injection of CARMA-hMeso resulted in the dose-dependent inhibition of tumor growth and improved survival of mice. Furthermore, repeat weekly intraperitoneal administrations of the optimal CARMA-hMeso dose further prolonged disease control and survival. No significant off-target toxicities were observed. These data support further investigation of CARMA-hMeso as a potential treatment for ovarian cancer and other mesothelin-expressing cancers.


Subject(s)
GPI-Linked Proteins/immunology , Natural Killer T-Cells/transplantation , Ovarian Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , Animals , Cell Line, Tumor , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/therapeutic use , Humans , Immunotherapy, Adoptive , Lymphocytes/immunology , Mesothelin , Mice , Natural Killer T-Cells/immunology , Ovarian Neoplasms/immunology , RNA, Messenger/genetics , RNA, Messenger/therapeutic use , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Xenograft Model Antitumor Assays
3.
Sci Transl Med ; 9(372)2017 01 11.
Article in English | MEDLINE | ID: mdl-28077679

ABSTRACT

Gene repair of CD34+ hematopoietic stem and progenitor cells (HSPCs) may avoid problems associated with gene therapy, such as vector-related mutagenesis and dysregulated transgene expression. We used CRISPR (clustered regularly interspaced short palindromic repeat)/Cas9 (CRISPR-associated 9) to repair a mutation in the CYBB gene of CD34+ HSPCs from patients with the immunodeficiency disorder X-linked chronic granulomatous disease (X-CGD). Sequence-confirmed repair of >20% of HSPCs from X-CGD patients restored the function of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase and superoxide radical production in myeloid cells differentiated from these progenitor cells in vitro. Transplant of gene-repaired X-CGD HSPCs into NOD (nonobese diabetic) SCID (severe combined immunodeficient) γc-/- mice resulted in efficient engraftment and production of functional mature human myeloid and lymphoid cells for up to 5 months. Whole-exome sequencing detected no indels outside of the CYBB gene after gene correction. CRISPR-mediated gene editing of HSPCs may be applicable to other CGD mutations and other monogenic disorders of the hematopoietic system.


Subject(s)
CRISPR-Cas Systems , Genetic Therapy , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Cell Differentiation , DNA Repair , Female , Granulomatous Disease, Chronic/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mutagenesis , Mutation , NADPH Oxidase 2/genetics , Oligonucleotides/genetics
4.
Front Immunol ; 7: 105, 2016.
Article in English | MEDLINE | ID: mdl-27047492

ABSTRACT

For more than a decade, investigators have pursued methods to genetically engineer natural killer (NK) cells for use in clinical therapy against cancer. Despite considerable advances in viral transduction of hematopoietic stem cells and T cells, transduction efficiencies for NK cells have remained disappointingly low. Here, we show that NK cells can be genetically reprogramed efficiently using a cGMP-compliant mRNA electroporation method that induces rapid and reproducible transgene expression in nearly all transfected cells, without negatively influencing their viability, phenotype, and cytotoxic function. To study its potential therapeutic application, we used this approach to improve key aspects involved in efficient lymphoma targeting by adoptively infused ex vivo-expanded NK cells. Electroporation of NK cells with mRNA coding for the chemokine receptor CCR7 significantly promoted migration toward the lymph node-associated chemokine CCL19. Further, introduction of mRNA coding for the high-affinity antibody-binding receptor CD16 (CD16-158V) substantially augmented NK cell cytotoxicity against rituximab-coated lymphoma cells. Based on these data, we conclude that this approach can be utilized to genetically modify multiple modalities of NK cells in a highly efficient manner with the potential to improve multiple facets of their in vivo tumor targeting, thus, opening a new arena for the development of more efficacious adoptive NK cell-based cancer immunotherapies.

5.
Nat Biotechnol ; 34(4): 424-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950749

ABSTRACT

Gene therapy with genetically modified human CD34(+) hematopoietic stem and progenitor cells (HSPCs) may be safer using targeted integration (TI) of transgenes into a genomic 'safe harbor' site rather than random viral integration. We demonstrate that temporally optimized delivery of zinc finger nuclease mRNA via electroporation and adeno-associated virus (AAV) 6 delivery of donor constructs in human HSPCs approaches clinically relevant levels of TI into the AAVS1 safe harbor locus. Up to 58% Venus(+) HSPCs with 6-16% human cell marking were observed following engraftment into mice. In HSPCs from patients with X-linked chronic granulomatous disease (X-CGD), caused by mutations in the gp91phox subunit of the NADPH oxidase, TI of a gp91phox transgene into AAVS1 resulted in ∼15% gp91phox expression and increased NADPH oxidase activity in ex vivo-derived neutrophils. In mice transplanted with corrected HSPCs, 4-11% of human cells in the bone marrow expressed gp91phox. This method for TI into AAVS1 may be broadly applicable to correction of other monogenic diseases.


Subject(s)
Antigens, CD34/chemistry , Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Animals , Cells, Cultured , Humans , Mice , Mice, Transgenic
6.
J Biomol Screen ; 20(4): 545-51, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25520372

ABSTRACT

In recent years, researchers have turned to transient gene expression (TGE) as an alternative to CHO stable cell line generation for early-stage antibody development. Despite advances in transfection methods and culture optimization, the majority of CHO-based TGE systems produce insufficient antibody titers for extensive use within biotherapeutic development pipelines. Flow electroporation using the MaxCyte STX Scalable Transfection System is a highly efficient, scalable means of CHO-based TGE for gram-level production of antibodies without the need for specialized expression vectors or genetically engineered CHO cell lines. CHO cell flow electroporation is easily scaled from milligram to multigram quantities without protocol reoptimization while maintaining transfection performance and antibody productivity. In this article, data are presented that demonstrate the reproducibility, scalability, and antibody production capabilities of CHO-based TGE using the MaxCyte STX. Data show optimization of posttransfection parameters such as cell density, media composition, and feed strategy that result in secreted antibody titers >1 g/L and production of multiple grams of antibody within 2 weeks of a single CHO-S cell transfection. In addition, data are presented to demonstrate the application of scalable electroporation for the rapid generation of high-yield stable CHO cell lines to bridge the gap between early- and late-stage antibody development activities.


Subject(s)
Electroporation , Gene Expression , Animals , CHO Cells , Cricetinae , Cricetulus
7.
Int Immunopharmacol ; 15(3): 488-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23474736

ABSTRACT

Recent commercial approval of cancer vaccine, demonstrating statistically significant improvement in overall survival of prostate cancer patients has spurred renewed interest in active immunotherapies; specifically, strategies that lead to enhanced biological activity and robust efficacy for dendritic cell vaccines. A simple, widely used approach to generating multivalent cancer vaccines is to load tumor whole cell lysates into dendritic cells (DCs). Current DC vaccine manufacturing processes require co-incubation of tumor lysate antigens with immature DCs and their subsequent maturation. We compared electroloading of tumor cell lysates directly into mature DCs with the traditional method of lysate co-incubation with immature DCs. Electroloaded mature DCs were more potent in vitro, as judged by their ability to elicit significantly (p < 0.05) greater expansion of peptide antigen-specific CD8(+) T cells, than either lysate-electroloaded immature DCs or lysate-co-incubated immature DCs, both of which must be subsequently matured. Expanded CD8(+) T cells were functional as judged by their ability to produce IFN-γ upon antigen-specific re-stimulation. The electroloading technology used herein is an automated, scalable, functionally closed cGMP-compliant manufacturing technology supported by a Master File at CBER, FDA and represents an opportunity for translation of enhanced potency DC vaccines at clinical/commercial scale.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/metabolism , Electroporation/methods , Immunotherapy, Adoptive/methods , Melanoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , CD8 Antigens/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Feasibility Studies , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Melanoma/therapy
8.
Methods Mol Biol ; 969: 127-38, 2013.
Article in English | MEDLINE | ID: mdl-23296932

ABSTRACT

Genetic modification for enhancing cellular function has been continuously pursued for fighting diseases. Messenger RNA (mRNA) transfection is found to be a promising solution in modifying hematopoietic and immune cells for therapeutic purpose. We have developed a flow electroporation-based system for large volume electroporation of cells with various molecules, including mRNA. This allows robust and scalable mRNA transfection of primary cells of different origin. Here we describe transfection of chimeric antigen receptor (CAR) mRNA into NK cells to modulate the ability of NK cells to target tumor cells. High levels of CAR expression in NK cells can be maintained for 3-7 days post transfection. CD19-specific CAR mRNA transfected NK cells demonstrate targeted lysis of CD19-expressing tumor cells OP-1, primary B-CLL tumor cells, and autologous CD19+ B cells in in vitro assays with enhanced potency: >80% lysis at effector-target ratio of 1:1. This allows current good manufacturing practices (cGMP) and regulatory compliant manufacture of CAR mRNA transfected NK cells for clinical delivery.


Subject(s)
Electroporation/methods , Killer Cells, Natural , RNA, Messenger/chemistry , Transfection/methods , Animals , Antigens, CD19/biosynthesis , Antigens, CD19/genetics , Antigens, CD19/immunology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , Receptors, Antigen/biosynthesis , Receptors, Antigen/genetics , Receptors, Antigen/immunology
9.
Hum Gene Ther ; 23(2): 243-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21933028

ABSTRACT

Lentiviral vectors are beginning to emerge as a viable choice for human gene therapy. Here, we describe a method that combines the convenience of a suspension cell line with a scalable, nonchemically based, and GMP-compliant transfection technique known as flow electroporation (EP). Flow EP parameters for serum-free adapted HEK293FT cells were optimized to limit toxicity and maximize titers. Using a third generation, HIV-based, lentiviral vector system pseudotyped with the vesicular stomatitis glycoprotein envelope, both small- and large-volume transfections produced titers over 1×10(8) infectious units/mL. Therefore, an excellent option for implementing large-scale, clinical lentiviral productions is flow EP of suspension cell lines.


Subject(s)
Genetic Vectors/biosynthesis , Lentivirus/genetics , Vesiculovirus/genetics , Viral Proteins/genetics , Bioreactors , Cell Survival , Culture Media, Serum-Free , Deoxyribonuclease I/metabolism , Electroporation , Genetic Vectors/genetics , HEK293 Cells , Humans , Plasmids , Recombinant Proteins/metabolism , Rheology , Transfection , Vesiculovirus/chemistry , Viral Proteins/chemistry
10.
Cytotherapy ; 13(9): 1128-39, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745159

ABSTRACT

BACKGROUND AIMS: Several studies have demonstrated that the immunogenicity of chronic lymphocytic leukemia (CLL) cells can be increased by manipulation of the CD40/CD40-ligand (CD40L) pathway. Although immunologic, and perhaps clinical, benefits have been obtained with an autologous CLL tumor vaccine obtained by transgenic expression of CD40L and interleukin (IL)-2, there is little information about the optimal gene transfer strategies. METHODS: We compared two different CLL vaccines prepared by adenoviral gene transfer and plasmid electroporation, analyzing their phenotype and immunostimulatory activity. RESULTS: We found that higher expression of transgenic CD40L was mediated by adenoviral gene transfer than by plasmid transduction, and that adenoviral transfer of CD40L was associated with up-regulation of the co-stimulatory molecules CD80 and CD86 and adhesion molecule CD54. In contrast, transgenic IL-2 secretion was greater following plasmid transduction. These phenotypic differences in the vaccines were associated with different functionality, both ex vivo and following administration to patients. Thus adenoviral vaccines induced greater activation of leukemia-reactive T cells ex vivo than plasmid vaccines. In treated patients, specific T-cell (T helper 1 (Th1) and T helper 2 (Th2)) and humoral anti-leukemia responses were detected following administration of the adenoviral vaccine (n = 15), while recipients of the plasmid vaccine (n = 9) manifested only a low-level Th2 response. Progression-free survival at 2 years was 46.7% in the adenoviral vaccine recipients, versus 11.1 % in those receiving plasmid vaccine. CONCLUSIONS: CLL vaccines expressing the same transgenes but produced by distinct methods of gene transfer may differ in the polarity of the immune response they induce in patients.


Subject(s)
CD40 Ligand/metabolism , Cancer Vaccines , Gene Transfer Techniques , Interleukin-2/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Adenoviridae/genetics , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Antigens, Neoplasm/immunology , CD40 Ligand/genetics , Cell Adhesion , Cell Differentiation , Female , Humans , Interleukin-2/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphocyte Activation , Male , Middle Aged , Plasmids/genetics , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...