scholarly journals Naive and in vitro-activated primary mouse CD8+ T cells retain in vivo immune responsiveness after electroporation-based CRISPR/Cas9 genetic engineering

2021 ◽  
Author(s):  
Petra Pfenninger ◽  
Laura Yerly ◽  
Jun Abe

CRISPR/Cas9 technology has revolutionized genetic engineering of primary cells. Although its use is gaining momentum in studies on CD8+ T cell biology, it remains elusive to what extent CRISPR/Cas9 affects in vivo function of CD8+ T cells. Here, we optimized nucleofection-based CRISPR/Cas9 genetic engineering of naive and in vitro-activated primary mouse CD8+ T cells and tested their in vivo immune responses. Nucleofection of naive CD8+ T cells preserved their in vivo antiviral immune responsiveness to an extent that is indistinguishable from non-nucleofected cells, whereas in vitro activation of CD8+ T cells prior to nucleofection led to slightly impaired expansion/survival. Of note, different target proteins displayed distinct decay rates after gene editing. This is in stark contrast to a comparable period of time required to complete gene inactivation. Thus, for optimal experimental design, it is crucial to determine the kinetics of the loss of target gene product to adapt incubation period after gene editing. In sum, nucleofection-based CRISPR/Cas9 genome editing achieves efficient and rapid generation of mutant CD8+ T cells without imposing detrimental constraints on their in vivo functions.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 27-28
Author(s):  
A. Samer Al-Homsi ◽  
Sebastien Anguille ◽  
Jason Brayer ◽  
Dries Deeren ◽  
Nathalie Meuleman ◽  
...  

Background Autologous CAR T-cell therapy targeting the B-cell maturation antigen (BCMA) has shown impressive objective response rates in patients with advanced multiple myeloma (MM). Clinical grade manufacturing of autologous CAR T-cells has limitations including vein-to-vein delivery time delay and potentially sub-optimal immunological capability of T-cells isolated from patients with advanced disease. Allogeneic CAR T-cell products, whereby cells from healthy third-party donors are used to generate an "off-the-shelf" CAR T-cell product, have the potential to overcome some of these issues. To circumvent the primary potential risk of graft-versus-host disease (GvHD) associated with the use of allogeneic T-cells, abrogation of the T-cell receptor (TCR) expression in the CAR T-cells, via gene editing, is being actively pursued. To avoid the potential safety risks and manufacturing challenges associated with gene editing, the allogeneic CYAD-211 CAR T-cell product exploits short hairpin RNA (shRNA) interference technology to down-regulate TCR expression thus avoiding the risk of life-threatening GvHD. Aim The aim is to generate a BCMA-specific allogeneic CAR T-cell product using a non-gene editing approach and study its activity both in vitro and in vivo. CYAD-211 combines a BCMA-specific CAR with a single optimized shRNA targeting the TCR CD3ζ subunit. Downregulation of CD3ζ impairs the TCR expression on the surface of the donor T-cells, preventing their reactivity with the normal host tissue cells and potential GvHD induction. Maintaining all the elements required for the therapy within a single vector (all-in-one vector) provides some significant manufacturing advantages, as a solitary selection step will isolate cells expressing all the desired traits. Results CYAD-211 cells produce high amounts of interferon-gamma (IFN-γ) during in vitro co-cultures with various BCMA-expressing MM cell lines (i.e., RPMI-8226, OPM-2, U266, and KMS-11). Cytotoxicity experiments confirmed that CYAD-211 efficiently kills MM cell lines in a BCMA-specific manner. The anti-tumor efficacy of CYAD-211 was further confirmed in vivo, in xenograft MM models using the RPMI-8226 and KMS-11 cell lines. Preclinical data also showed no demonstrable evidence of GvHD when CYAD-211 was infused in NSG mice confirming efficient inhibition of TCR-induced activation. Following FDA acceptance of the IND application, IMMUNICY-1, a first-in-human, open-label dose-escalation phase I clinical study evaluating the safety and clinical activity of CYAD-211 for the treatment of relapsed or refractory MM patients to at least two prior MM treatment regimens, is scheduled to begin recruitment. IMMUNICY-1 will evaluate three dose-levels of CYAD-211 (3x107, 1x108 and 3x108 cells/infusion) administered as a single infusion after a non-myeloablative conditioning (cyclophosphamide 300 mg/m²/day and fludarabine 30 mg/m²/day, daily for 3 days) according to a classical Fibonacci 3+3 design. Description of the study design and preliminary safety and clinical data from the first cohort will be presented at ASH 2020. Conclusion CYAD-211 is the first generation of non-gene edited allogeneic CAR T-cell product based on shRNA technology. The IMMUNICY-1 clinical study seeks to provide proof of principle that single shRNA-mediated knockdown can generate fully functional allogeneic CAR T-cells in humans without GvHD-inducing potential. We anticipate that subsequent generations of this technology will incorporate multiple shRNA hairpins within a single vector system. This will enable the production of allogeneic CAR T-cells in which multiple genes of interest are modulated simultaneously thereby providing a platform approach that can underpin the future of this therapeutic modality. Figure 1 Disclosures Al-Homsi: Celyad: Membership on an entity's Board of Directors or advisory committees. Brayer:Janssen: Consultancy; Bristol-Myers Squibb, WindMIL Therapeutics: Research Funding; Bristol-Myers Squibb, Janssen, Amgen: Speakers Bureau. Nishihori:Novartis: Other: Research support to institution; Karyopharm: Other: Research support to institution. Sotiropoulou:Celyad Oncology: Current Employment. Twyffels:Celyad Oncology: Current Employment. Bolsee:Celyad Oncology: Current Employment. Braun:Celyad Oncology: Current Employment. Lonez:Celyad Oncology: Current Employment. Gilham:Celyad Oncology: Current Employment. Flament:Celyad Oncology: Current Employment. Lehmann:Celyad Oncology: Current Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1469-1469
Author(s):  
Xiuli Wang ◽  
Wen-Chung Chang ◽  
ChingLam W Wong ◽  
David Colcher ◽  
Mark Sherman ◽  
...  

Abstract Abstract 1469 Hematopoietic cell-based therapies, including genetically manipulated cell products derived from either hematopoietic stem cells or T cells, is an emerging area in applied biotechnology. In both of these venues, a variety of genetic engineering approaches are being studied to endow cells with novel attributes, to increase their therapeutic potency and/or safety. Common to the field of ex vivo cellular genetic engineering is the need to purify cells that express desired quantities of therapeutic transgene(s) and cull out non-expressing cells that either lack transgene endowed therapeutic activity or safety features. However, current drug selection strategies are associated with prolonged ex vivo culture that drives terminal differentiation of the T cells, which has in turn been found to be associated with impaired antitumor efficacy of adoptively transferred CD8+ T cells in vivo. Thus, we were interested in developing a single transgene encoded polypeptide that can serve both as an ex vivo selection epitope and in vivo tracking marker/target for mAb-mediated cell ablation, while fulfilling the criteria of being functionally inert, non immunogenic, and amenable to commercially available cGMP-grade selection systems appropriate for clinical use. Here we describe a truncated human EGFR polypeptide (huEGFRt) devoid of extracellular N-terminal ligand binding domains and intracellular receptor tyrosine kinase domains. Retained features of huEGFRt include type I transmembrane cell surface localization and a conformationally intact binding epitope for pharmaceutical-grade anti-EGFR mAb, cetuximab/Erbitux™. Applying this system to cellular immunotherapy, we designed lentiviral vector prototypes housing multifunctional constructs combining huEGFRt with CD19-specific chimeric antigen receptors (CARs), and demonstrate that biotinylated-cetuximab immunomagnetic selection of transduced human T cells results in coordinate enrichment of CAR+ cells from 2% to over 90%. The huEGFRt-mediated selection did not affect the phenotype (i.e., TCR, CD3, CD4, CD8, CD28, and granzyme A expression), the in vitro expansion potential, nor the in vivo engraftment fitness (upon transfer into immunodeficient mice) of the T cells. Direct examination of EGF-binding and phospho-tyrosine analysis confirmed that this selection marker is functionally inert and has no negative effect on the T cell product. In addition, cytotoxicity against B cell malignancies and IFN-g/TNF-a production through the CD19-specific CAR was dramatically enhanced in the huEGFRt-selected population. The utility of huEGFRt in tracking the gene modified, transferred cells in vivo within easily obtained human tissues such as blood, bone marrow and tissue biopsies was then also proven via detection of huEGFRt using multiparameter flow cytometric analysis or FDA approved immunohistochemical techniques/reagents. In addition, we were able to demonstrate that Erbitux™ could mediate ADCC of huEGFRt+ T cells in vitro and inhibit the growth of huEGFRt+ CTLL2 cells in NOD/Scid mice, supporting the use of huEGFRt as a suicide gene via cetuximab-mediated ADCC after adoptive transfer. Together these data suggest that huEGFRt is a superior selection marker for any transduction system that can be applied to the generation of cell products for hematopoietic cell-based medical therapies. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 12 (546) ◽  
pp. eaay6422 ◽  
Author(s):  
Yuchi Honaker ◽  
Nicholas Hubbard ◽  
Yufei Xiang ◽  
Logan Fisher ◽  
David Hagin ◽  
...  

Thymic regulatory T cells (tTregs) are potent inhibitors of autoreactive immune responses, and loss of tTreg function results in fatal autoimmune disease. Defects in tTreg number or function are also implicated in multiple autoimmune diseases, leading to growing interest in use of Treg as cell therapies to establish immune tolerance. Because tTregs are present at low numbers in circulating blood and may be challenging to purify and expand and also inherently defective in some subjects, we designed an alternative strategy to create autologous Treg-like cells from bulk CD4+ T cells. We used homology-directed repair (HDR)–based gene editing to enforce expression of FOXP3, the master transcription factor for tTreg. Targeted insertion of a robust enhancer/promoter proximal to the first coding exon bypassed epigenetic silencing, permitting stable and robust expression of endogenous FOXP3. HDR-edited T cells, edTregs, manifested a transcriptional program leading to sustained expression of canonical markers and suppressive activity of tTreg. Both human and murine edTregs mediated immunosuppression in vivo in models of inflammatory disease. Further, this engineering strategy permitted generation of antigen-specific edTreg with robust in vitro and in vivo functional activity. Last, edTreg could be enriched and expanded at scale using clinically relevant methods. Together, these findings suggest that edTreg production may permit broad future clinical application.


2004 ◽  
Vol 78 (10) ◽  
pp. 5244-5257 ◽  
Author(s):  
Xiaoduan Weng ◽  
Elena Priceputu ◽  
Pavel Chrobak ◽  
Johanne Poudrier ◽  
Denis G. Kay ◽  
...  

ABSTRACT The cellular and molecular mechanisms of dysfunction and depletion of CD4+ T lymphocytes over the course of human immunodeficiency virus type 1 (HIV-1) infection are still incompletely understood, but chronic immune activation is thought to play an important role in disease progression. We studied CD4+ T-cell biology in CD4C/HIV transgenic (Tg) mice, in which Nef expression is sufficient to induce a severe AIDS-like disease including a preferential decrease of CD4+ T cells. We show here that Nef-expressing Tg CD4+ T cells exhibit an activated/memory-like phenotype which appears to be independent of antigenic stimulation, as documented in experiments involving breeding with AD10 TcR Tg mice. In addition, in vivo bromodeoxyuridine incorporation showed that a larger proportion of Tg than non-Tg CD4+ T cells entered the S phase. However, in vitro, Tg CD4+ T cells were found to have a very limited capacity to divide in response to stimulation with anti-CD3 and anti-CD28 or in allogeneic mixed leukocyte reactions. Interestingly, despite these observations, the deletion of Tg CD4+ T cells had little impact on the development of other AIDS-like organ phenotypes. Thus, the Nef-induced chronic activation of CD4+ T cells may exhaust the T-cell pool and may contribute to the thymic atrophy and the low number of CD4+ T cells observed in these Tg mice.


2016 ◽  
Vol 213 (9) ◽  
pp. 1881-1900 ◽  
Author(s):  
Martin Chopra ◽  
Marlene Biehl ◽  
Tim Steinfatt ◽  
Andreas Brandl ◽  
Juliane Kums ◽  
...  

Donor CD4+Foxp3+ regulatory T cells (T reg cells) suppress graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HCT [allo-HCT]). Current clinical study protocols rely on the ex vivo expansion of donor T reg cells and their infusion in high numbers. In this study, we present a novel strategy for inhibiting GvHD that is based on the in vivo expansion of recipient T reg cells before allo-HCT, exploiting the crucial role of tumor necrosis factor receptor 2 (TNFR2) in T reg cell biology. Expanding radiation-resistant host T reg cells in recipient mice using a mouse TNFR2-selective agonist before allo-HCT significantly prolonged survival and reduced GvHD severity in a TNFR2- and T reg cell–dependent manner. The beneficial effects of transplanted T cells against leukemia cells and infectious pathogens remained unaffected. A corresponding human TNFR2-specific agonist expanded human T reg cells in vitro. These observations indicate the potential of our strategy to protect allo-HCT patients from acute GvHD by expanding T reg cells via selective TNFR2 activation in vivo.


2015 ◽  
Vol 1 (2) ◽  
pp. 122-128
Author(s):  
Syuichi Koarada ◽  
Yuri Sadanaga ◽  
Natsumi Nagao ◽  
Satoko Tashiro ◽  
Rie Suematsu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document