36 results on '"Eyquem J"'
Search Results
2. Targeted Integration of Transgenes in a Safe Harbor Locus using Engineered Meganuclease and TALEN (TM)-based Homologous Recombination Strategies
- Author
-
Rodriguez-Fornes, F., Quintana-Bustamante, O., Lozano, M. L., Cerrato, L., Eyquem, J., Gouble, A., Galetto, R., Poirot, L., Smith, J., Segovia, J. C., Guenechea, G., and Juan Bueren
3. Author Correction: Generation of T-cell-receptor-negative CD8αβ-positive CAR T cells from T-cell-derived induced pluripotent stem cells.
- Author
-
van der Stegen SJC, Lindenbergh PL, Petrovic RM, Xie H, Diop MP, Alexeeva V, Shi Y, Mansilla-Soto J, Hamieh M, Eyquem J, Cabriolu A, Wang X, Abujarour R, Lee T, Clarke R, Valamehr B, Themeli M, Riviere I, and Sadelain M
- Published
- 2024
- Full Text
- View/download PDF
4. Scalable intracellular delivery via microfluidic vortex shedding enhances the function of chimeric antigen receptor T-cells.
- Author
-
Sytsma BJ, Allain V, Bourke S, Faizee F, Fathi M, Berdeaux R, Ferreira LMR, Brewer WJ, Li L, Pan FL, Rothrock AG, Nyberg WA, Li Z, Wilson LH, Eyquem J, and Pawell RS
- Abstract
Adoptive chimeric antigen receptor T-cell (CAR-T) therapy is transformative and approved for hematologic malignancies. It is also being developed for the treatment of solid tumors, autoimmune disorders, heart disease, and aging. Despite unprecedented clinical outcomes, CAR-T and other engineered cell therapies face a variety of manufacturing and safety challenges. Traditional methods, such as lentivirus transduction and electroporation, result in random integration or cause significant cellular damage, which can limit the safety and efficacy of engineered cell therapies. We present hydroporation as a gentle and effective alternative for intracellular delivery. Hydroporation resulted in 1.7- to 2-fold higher CAR-T yields compared to electroporation with superior cell viability and recovery. Hydroporated cells exhibited rapid proliferation, robust target cell lysis, and increased pro-inflammatory and regulatory cytokine secretion in addition to improved CAR-T yield by day 5 post-transfection. We demonstrate that scaled-up hydroporation can process 5 × 10
8 cells in less than 10 s, showcasing the platform as a viable solution for high-yield CAR-T manufacturing with the potential for improved therapeutic outcomes., Competing Interests: B.J.S., S.B., F.F., L.H.W., and R.S.P. are or were employed by and have an equity interest in Indee Labs. L.M.R.F., J.B., L.L., F.L.P. and J.E. are or were consultants to Indee Labs. R.S.P. is an investor in and a venture partner at both Pioneer Fund and Axial, which have a financial interest in Indee Labs. The Ferreira Lab received support from Indee Labs as a subaward from the National Institute of Diabetes and Digestive and Kidney Diseases (Grant No. 1R43DK133029-01). Indee Labs has a commercial interest in developing patents related to Hydropore™ (WO2016109864A1 & WO2019084624A1). M.F. and R.B. are or were employed by and have an equity interest in CellChorus. CellChorus received support from the National Center for Translational Sciences (R44TR005137) and the National Institute of General Medical Sciences (R44GM149106) of the National Institutes of Health and the National Science Foundation (NSF2229323). CellChorus has a commercial interest in developing the TIMING assay. J.E. is a compensated co-founder at Mnemo Therapeutics; owns stocks in Mnemo Therapeutics and Cytovia Therapeutics; is a compensated scientific advisor for Enterome, Treefrog Therapeutics and Resolution Therapeutics; and is a holder of patents pertaining to but not resulting from this work. The Eyquem Lab received research support from Cytovia Therapeutic, Mnemo Therapeutics, Takeda and Indee Labs as a subaward from the National Cancer Institute (Contract No. 7591022C00053).- Published
- 2024
- Full Text
- View/download PDF
5. An improved approach to generate IL-15 +/+ /TGFβR2 -/- iPSC-derived natural killer cells using TALEN.
- Author
-
Chen AP, Gao P, Lin L, Ashok P, He H, Ma C, Zou DL, Allain V, Boyne A, Juillerat A, Duchateau P, Rath A, Teper D, Arulanandam A, Chang HM, Eyquem J, and Li W
- Subjects
- Humans, Cell Differentiation, Transcription Activator-Like Effector Nucleases metabolism, Transcription Activator-Like Effector Nucleases genetics, Gene Editing methods, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells cytology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Interleukin-15 genetics, Interleukin-15 metabolism, Receptor, Transforming Growth Factor-beta Type II genetics, Receptor, Transforming Growth Factor-beta Type II metabolism
- Abstract
We present a TALEN-based workflow to generate and maintain dual-edited (IL-15
+/+ /TGFβR2-/- ) iPSCs that produce enhanced iPSC-derived natural killer (iNK) cells for cancer immunotherapy. It involves using a cell lineage promoter for knocking in (KI) gene(s) to minimize the potential effects of expression of any exogenous genes on iPSCs. As a proof-of-principle, we KI IL-15 under the endogenous B2M promoter and show that it results in high expression of the sIL-15 in iNK cells but minimal expression in iPSCs. Furthermore, given that it is known that knockout (KO) of TGFβR2 in immune cells can enhance resistance to the suppressive TGF-β signaling in the tumor microenvironment, we develop a customized medium containing Nodal that can maintain the pluripotency of iPSCs with TGFβR2 KO, enabling banking of these iPSC clones. Ultimately, we show that the dual-edited IL-15+/+ /TGFβR2-/- iPSCs can be efficiently differentiated into NK cells that show enhanced autonomous growth and are resistant to the suppressive TGF-β signaling., Competing Interests: Declaration of interests A.-P.C., P.G., L.L., P.A., H.H., H.-M.C., D.Z., A.R., D.T., A.A., and W.L. are employees of and hold stock or stock options in Cytovia Therapeutics. J.E. is a consultant to Cytovia Therapeutics and received funding support from Cytovia Therapeutics., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) in primary human immune cells and hematopoietic stem cells.
- Author
-
Sahu S, Castro M, Muldoon JJ, Asija K, Wyman SK, Krishnappa N, de Onate L, Eyquem J, Nguyen DN, and Wilson RC
- Abstract
Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) is a new approach for ex vivo genome editing of primary human cells. PERC uses a single amphiphilic peptide reagent to mediate intracellular delivery of the same pre-formed CRISPR ribonucleoprotein enzymes that are broadly used in research and therapeutics, resulting in high-efficiency editing of stimulated immune cells and cultured hematopoietic stem and progenitor cells (HSPCs). PERC facilitates nuclease-mediated gene knockout, precise transgene knock-in, and base editing. PERC involves mixing the CRISPR ribonucleoprotein enzyme with peptide and then incubating the formulation with cultured cells. For efficient transgene knock-in, adeno-associated virus (AAV) bearing homology-directed repair template DNA may be included. In contrast to electroporation, PERC is appealing as it requires no dedicated hardware and has less impact on cell phenotype and viability. Due to the gentle nature of PERC, delivery can be performed multiple times without substantial impact to cell health or phenotype. Here we report methods for improved PERC-mediated editing of T cells as well as novel methods for PERC-mediated editing of HSPCs, including knockout and precise knock-in. Editing efficiencies can surpass 90% using either Cas9 or Cas12a in primary T cells or HSPCs. Because PERC calls for only three readily available reagents - protein, RNA, and peptide - and does not require dedicated hardware for any step, PERC demands no special expertise and is exceptionally straightforward to adopt. The inherent compatibility of PERC with established cell engineering pipelines makes this approach appealing for rapid deployment in research and clinical settings.
- Published
- 2024
- Full Text
- View/download PDF
7. Localized in vivo gene editing of murine cancer-associated fibroblasts.
- Author
-
Kuhn NF, Zaleta-Linares I, Nyberg WA, Eyquem J, and Krummel MF
- Abstract
Discovering the role of fibroblasts residing in the tumor microenvironment (TME) requires controlled, localized perturbations because fibroblasts play critical roles in regulating immunity and tumor biology at multiple sites. Systemic perturbations can lead to unintended, confounding secondary effects, and methods to locally genetically engineer fibroblasts are lacking. To specifically investigate murine stromal cell perturbations restricted to the TME, we developed an adeno-associated virus (AAV)-based method to target any gene-of-interest in fibroblasts at high efficiency (>80%). As proof of concept, we generated single (sKO) and double gene KOs (dKO) of Osmr , Tgfbr2 , and Il1r1 in cancer-associated fibroblasts (CAFs) and investigated how their cell states and those of other cells of the TME subsequently change in mouse models of melanoma and pancreatic ductal adenocarcinoma (PDAC). Furthermore, we developed an in vivo knockin-knockout (KIKO) strategy to achieve long-term tracking of CAFs with target gene KO via knocked-in reporter gene expression. This validated in vivo gene editing toolbox is fast, affordable, and modular, and thus holds great potential for further exploration of gene function in stromal cells residing in tumors and beyond., Competing Interests: M.F.K. is a founder and shareholder of FOUNDERY innovations. J.E. is a compensated co-founder at Mnemo Therapeutics and a compensated scientific advisor to Cytovia Therapeutics. J.E. owns stocks in Mnemo Therapeutics and Cytovia Therapeutics. J.E. has received a consulting fee from Casdin Capital, Resolution Therapeutics and Treefrog Therapeutics. The J.E. lab has received research support from Cytovia Therapeutics, Mnemo Therapeutics, and Takeda Pharmaceutical Company.
- Published
- 2024
- Full Text
- View/download PDF
8. Spleen Tyrosine Kinase (SYK) negatively regulates ITAM-mediated human NK cell signaling and CD19-CAR NK cell efficacy.
- Author
-
Millan AJ, Allain V, Nayak I, Aguilar OA, Arakawa-Hoyt JS, Ureno G, Rothrock AG, Shemesh A, Eyquem J, Das J, and Lanier LL
- Abstract
NK cells express activating receptors that signal through ITAM-bearing adapter proteins. The phosphorylation of each ITAM creates binding sites for SYK and ZAP70 protein tyrosine kinases to propagate downstream signaling including the induction of Ca 2 + influx. While all immature and mature human NK cells co-express SYK and ZAP70, clonally driven memory or adaptive NK cells can methylate SYK genes and signaling is mediated exclusively using ZAP70. Here, we examined the role of SYK and ZAP70 in a clonal human NK cell line KHYG1 by CRISPR-based deletion using a combination of experiments and mechanistic computational modeling. Elimination of SYK resulted in more robust Ca + + influx after cross-linking of the CD16 and NKp30 receptors and enhanced phosphorylation of downstream proteins, whereas ZAP70 deletion diminished these responses. By contrast, ZAP70 depletion increased proliferation of the NK cells. As immature T cells express both SYK and ZAP70 but mature T cells often express only ZAP70, we transduced the human Jurkat cell line with SYK and found that expression of SYK increased proliferation but diminished TCR-induced Ca 2 + flux and activation. We performed transcriptional analysis of the matched sets of variant Jurkat and KHYG1 cells and observed profound alterations caused by SYK expression. As depletion of SYK in NK cells increased their activation, primary human NK cells were transduced with a CD19-targeting CAR and were CRISPR edited to ablate SYK or ZAP70 . Deletion of SYK resulted in more robust cytotoxic activity and cytokine production, providing a new therapeutic strategy of NK cell engineering for cancer immunotherapy.
- Published
- 2024
- Full Text
- View/download PDF
9. Ultra-high efficiency T cell reprogramming at multiple loci with SEED-Selection.
- Author
-
Chang CR, Vykunta VS, Goodman DB, Muldoon JJ, Nyberg WA, Liu C, Allain V, Rothrock A, Wang CH, Marson A, Shy BR, and Eyquem J
- Abstract
Multiplexed reprogramming of T cell specificity and function can generate powerful next-generation cellular therapies. However, current manufacturing methods produce heterogenous mixtures of partially engineered cells. Here, we develop a one-step process to enrich for unlabeled cells with knock-ins at multiple target loci using a family of repair templates named Synthetic Exon/Expression Disruptors (SEEDs). SEED engineering associates transgene integration with the disruption of a paired endogenous surface protein, allowing non-modified and partially edited cells to be immunomagnetically depleted (SEED-Selection). We design SEEDs to fully reprogram three critical loci encoding T cell specificity, co-receptor expression, and MHC expression, with up to 98% purity after selection for individual modifications and up to 90% purity for six simultaneous edits (three knock-ins and three knockouts). These methods are simple, compatible with existing clinical manufacturing workflows, and can be readily adapted to other loci to facilitate production of complex gene-edited cell therapies., Competing Interests: Competing Interests C.R.C., V.S.V., A.M., B.S., and J.E. are inventors on patent filings related to this work. J.E. is a compensated co-founder at Mnemo Therapeutics and a compensated scientific advisor to Cytovia Therapeutics. J.E. owns stocks in Mnemo Therapeutics and Cytovia Therapeutics. J.E. has received a consulting fee from Casdin Capital, Resolution Therapeutics, and Treefrog Therapeutics. The Eyquem lab has received research support from Cytovia Therapeutics, Mnemo Therapeutics, and Takeda Pharmaceutical Company. A.M. is a co-founder of Arsenal Biosciences, Function Bio, Spotlight Therapeutics, and Survey Genomics, serves on the boards of directors at Function Bio, Spotlight Therapeutics and Survey Genomics, is a member of the scientific advisory boards of Arsenal Biosciences, Function Bio, Spotlight Therapeutics, Survey Genomics, NewLimit, Amgen, Tenaya, and Lightcast, owns stock in Arsenal Biosciences, Function Bio, Spotlight Therapeutics, NewLimit, Survey Genomics, Tenaya, and Lightcast, and has received fees from Arsenal Biosciences, Spotlight Therapeutics, NewLimit, 23andMe, PACT Pharma, Juno Therapeutics, Tenaya, Lightcast, Trizell, Vertex, Merck, Amgen, Genentech, AlphaSights, Rupert Case Management, Bernstein, GLG, ClearView Healthcare Partners, and ALDA. A.M. is an investor in and informal advisor to Offline Ventures and a client of EPIǪ. The Marson laboratory has received research support from Juno Therapeutics, Epinomics, Sanofi, GlaxoSmithKline, Gilead, and Anthem.
- Published
- 2024
- Full Text
- View/download PDF
10. Base-editing mutagenesis maps alleles to tune human T cell functions.
- Author
-
Schmidt R, Ward CC, Dajani R, Armour-Garb Z, Ota M, Allain V, Hernandez R, Layeghi M, Xing G, Goudy L, Dorovskyi D, Wang C, Chen YY, Ye CJ, Shy BR, Gilbert LA, Eyquem J, Pritchard JK, Dodgson SE, and Marson A
- Subjects
- Humans, Amino Acids genetics, CRISPR-Cas Systems genetics, RNA, Guide, CRISPR-Cas Systems genetics, Lymphocyte Activation, Cytokines biosynthesis, Cytokines metabolism, Gain of Function Mutation, Loss of Function Mutation, Alleles, Gene Editing, Mutagenesis genetics, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
CRISPR-enabled screening is a powerful tool for the discovery of genes that control T cell function and has nominated candidate targets for immunotherapies
1-6 . However, new approaches are required to probe specific nucleotide sequences within key genes. Systematic mutagenesis in primary human T cells could reveal alleles that tune specific phenotypes. DNA base editors are powerful tools for introducing targeted mutations with high efficiency7,8 . Here we develop a large-scale base-editing mutagenesis platform with the goal of pinpointing nucleotides that encode amino acid residues that tune primary human T cell activation responses. We generated a library of around 117,000 single guide RNA molecules targeting base editors to protein-coding sites across 385 genes implicated in T cell function and systematically identified protein domains and specific amino acid residues that regulate T cell activation and cytokine production. We found a broad spectrum of alleles with variants encoding critical residues in proteins including PIK3CD, VAV1, LCP2, PLCG1 and DGKZ, including both gain-of-function and loss-of-function mutations. We validated the functional effects of many alleles and further demonstrated that base-editing hits could positively and negatively tune T cell cytotoxic function. Finally, higher-resolution screening using a base editor with relaxed protospacer-adjacent motif requirements9 (NG versus NGG) revealed specific structural domains and protein-protein interaction sites that can be targeted to tune T cell functions. Base-editing screens in primary immune cells thus provide biochemical insights with the potential to accelerate immunotherapy design., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
11. Structural surfaceomics reveals an AML-specific conformation of integrin β 2 as a CAR T cellular therapy target.
- Author
-
Mandal K, Wicaksono G, Yu C, Adams JJ, Hoopmann MR, Temple WC, Izgutdina A, Escobar BP, Gorelik M, Ihling CH, Nix MA, Naik A, Xie WH, Hübner J, Rollins LA, Reid SM, Ramos E, Kasap C, Steri V, Serrano JAC, Salangsang F, Phojanakong P, McMillan M, Gavallos V, Leavitt AD, Logan AC, Rooney CM, Eyquem J, Sinz A, Huang BJ, Stieglitz E, Smith CC, Moritz RL, Sidhu SS, Huang L, and Wiita AP
- Subjects
- Humans, T-Lymphocytes, Integrins metabolism, Immunotherapy, Adoptive methods, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Safely expanding indications for cellular therapies has been challenging given a lack of highly cancer-specific surface markers. Here we explore the hypothesis that tumor cells express cancer-specific surface protein conformations that are invisible to standard target discovery pipelines evaluating gene or protein expression, and these conformations can be identified and immunotherapeutically targeted. We term this strategy integrating cross-linking mass spectrometry with glycoprotein surface capture 'structural surfaceomics'. As a proof of principle, we apply this technology to acute myeloid leukemia (AML), a hematologic malignancy with dismal outcomes and no known optimal immunotherapy target. We identify the activated conformation of integrin β
2 as a structurally defined, widely expressed AML-specific target. We develop and characterize recombinant antibodies to this protein conformation and show that chimeric antigen receptor T cells eliminate AML cells and patient-derived xenografts without notable toxicity toward normal hematopoietic cells. Our findings validate an AML conformation-specific target antigen and demonstrate a tool kit for applying these strategies more broadly., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
12. Mitigation of chromosome loss in clinical CRISPR-Cas9-engineered T cells.
- Author
-
Tsuchida CA, Brandes N, Bueno R, Trinidad M, Mazumder T, Yu B, Hwang B, Chang C, Liu J, Sun Y, Hopkins CR, Parker KR, Qi Y, Hofman L, Satpathy AT, Stadtmauer EA, Cate JHD, Eyquem J, Fraietta JA, June CH, Chang HY, Ye CJ, and Doudna JA
- Subjects
- Humans, Chromosomes, DNA Damage, Clinical Trials as Topic, CRISPR-Cas Systems genetics, Gene Editing methods, T-Lymphocytes, Chromosome Aberrations
- Abstract
CRISPR-Cas9 genome editing has enabled advanced T cell therapies, but occasional loss of the targeted chromosome remains a safety concern. To investigate whether Cas9-induced chromosome loss is a universal phenomenon and evaluate its clinical significance, we conducted a systematic analysis in primary human T cells. Arrayed and pooled CRISPR screens revealed that chromosome loss was generalizable across the genome and resulted in partial and entire loss of the targeted chromosome, including in preclinical chimeric antigen receptor T cells. T cells with chromosome loss persisted for weeks in culture, implying the potential to interfere with clinical use. A modified cell manufacturing process, employed in our first-in-human clinical trial of Cas9-engineered T cells (NCT03399448), reduced chromosome loss while largely preserving genome editing efficacy. Expression of p53 correlated with protection from chromosome loss observed in this protocol, suggesting both a mechanism and strategy for T cell engineering that mitigates this genotoxicity in the clinic., Competing Interests: Declaration of interests C.A.T., J.A.D., and the Regents of the University of California have patents pending or issued related to the use of CRISPR genome editing technologies. R.B. is an employee of BioMarin Pharmaceutical Inc., J.L. is an employee of Altos Labs, and K.R.P. is a co-founder and employee of Cartography Biosciences. A.T.S. is a co-founder of Immunai and Cartography Biosciences. A.T.S. has received research support from Arsenal Biosciences, Allogene Therapeutics, and 10x Genomics. J.H.D.C. is a co-founder of Initial Therapeutics. J.E. is a co-founder of Mnemo Therapeutics, a scientific advisory board member of Cytovia Therapeutics, and a consultant for Casdin Capital, Resolution Therapeutics, IndeeLabs, and Treefrog Therapeutics. J.E. has received research support from Cytovia Therapeutics, Mnemo Therapeutics, and Takeda Pharmaceutical Company. J.A.F. has received research support from Tmunity. C.H.J. and the University of Pennsylvania have patents pending or issued related to the use of gene modification in T cells for adoptive T cell therapy. C.H.J. is a co-founder of Tmunity. H.Y.C. is a co-founder of Accent Therapeutics, Boundless Bio, Cartography Biosciences, and Orbital Therapeutics, and an advisor to 10x Genomics, Arsenal Biosciences, Chroma Medicine, Spring Discovery, and Vida Ventures. C.J.Y. is a co-founder of Survey Genomics, and a scientific advisory board member of Related Sciences and Immunai. C.J.Y. is a consultant for Maze Therapeutics, TReX Bio, ImYoo, and Santa Ana Bio. C.J.Y. has received research support from the Chan Zuckerberg Initiative, Chan Zuckerberg Biohub, Genentech, BioLegend, ScaleBio, and Illumina. J.A.D. is a co-founder of Editas Medicine, Intellia Therapeutics, Caribou Biosciences, Mammoth Biosciences, and Scribe Therapeutics, and a scientific advisory board member of Intellia Therapeutics, Caribou Biosciences, Mammoth Biosciences, Scribe Therapeutics, Vertex Pharmaceuticals, Felix Biosciences, The Column Group, Inari, and Isomorphic Labs. J.A.D. is the Chief Science Advisor at Sixth Street and a Director at Johnson & Johnson, Tempus, and Altos Labs. J.A.D. has sponsored research projects through Apple Tree Partners, Genentech, and Roche., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Modular pooled discovery of synthetic knockin sequences to program durable cell therapies.
- Author
-
Blaeschke F, Chen YY, Apathy R, Daniel B, Chen AY, Chen PA, Sandor K, Zhang W, Li Z, Mowery CT, Yamamoto TN, Nyberg WA, To A, Yu R, Bueno R, Kim MC, Schmidt R, Goodman DB, Feuchtinger T, Eyquem J, Jimmie Ye C, Carnevale J, Satpathy AT, Shifrut E, Roth TL, and Marson A
- Subjects
- Humans, Gene Library, Immunotherapy, Research, Cell- and Tissue-Based Therapy, Exercise
- Abstract
Chronic stimulation can cause T cell dysfunction and limit the efficacy of cellular immunotherapies. Improved methods are required to compare large numbers of synthetic knockin (KI) sequences to reprogram cell functions. Here, we developed modular pooled KI screening (ModPoKI), an adaptable platform for modular construction of DNA KI libraries using barcoded multicistronic adaptors. We built two ModPoKI libraries of 100 transcription factors (TFs) and 129 natural and synthetic surface receptors (SRs). Over 30 ModPoKI screens across human TCR- and CAR-T cells in diverse conditions identified a transcription factor AP4 (TFAP4) construct that enhanced fitness of chronically stimulated CAR-T cells and anti-cancer function in vitro and in vivo. ModPoKI's modularity allowed us to generate an ∼10,000-member library of TF combinations. Non-viral KI of a combined BATF-TFAP4 polycistronic construct enhanced fitness. Overexpressed BATF and TFAP4 co-occupy and regulate key gene targets to reprogram T cell function. ModPoKI facilitates the discovery of complex gene constructs to program cellular functions., Competing Interests: Declaration of interests F.B. received research awards (Gilead and Kite and Bristol Myers Squibb Foundation Immunonkologie). E.S. was an advisor for Arsenal Biosciences. J.E. is a compensated co-founder at Mnemo Therapeutics and compensated scientific advisor to Cytovia Therapeutics. J.E. owns stocks in Mnemo Therapeutics and Cytovia Therapeutics. J.E. has received a consulting fee from Casdin Capital. The Eyquem lab has received research support from Cytovia Therapeutics and Takeda. T.L.R. is a compensated co-founder, member of the scientific advisory board, and previously worked as the CSO of Arsenal Biosciences. A.T.S. is a founder of Immunai and Cartography Biosciences and receives research funding from Allogene Therapeutics and Merck Research Laboratories. C.T.M. is a compensated Bio+Health Venture Fellow at Andreessen Horowitz. C.J.Y. is founder for and holds equity in DropPrint Genomics (now ImmunAI) and Survey Genomics, a scientific advisory board member for and holds equity in Related Sciences and ImmunAI, a consultant for and holds equity in Maze Therapeutics, and a consultant for TReX Bio, HiBio, ImYoo, and Santa Ana. C.J.Y. has received research support from Chan Zuckerberg Initiative, Chan Zuckerberg Biohub, Genentech, BioLegend, ScaleBio, and Illumina. A.M. is a co-founder of Arsenal Biosciences, Spotlight Therapeutics, and Survey Genomics, serves on the boards of directors at Spotlight Therapeutics and Survey Genomics, is a board observer (and former member of the board of directors) at Arsenal Biosciences, is a member of the scientific advisory boards of Arsenal Biosciences, Spotlight Therapeutics, Survey Genomics, NewLimit, Amgen, Tenaya, and Lightcast, owns stock in Arsenal Biosciences, Spotlight Therapeutics, NewLimit, Survey Genomics, PACT Pharma, Tenaya, and Lightcast and has received fees from Arsenal Biosciences, Spotlight Therapeutics, Survey Genomics, NewLimit, 23andMe, PACT Pharma, Juno Therapeutics, Tenaya, Lightcast, GLG, Gilead, Trizell, Vertex, Merck, Amgen, Genentech, AlphaSights, Rupert Case Management, Bernstein, and ALDA. A.M. is an investor in and informal advisor to Offline Ventures and a client of EPIQ. The Marson laboratory received research support from Juno Therapeutics, Epinomics, Sanofi, GlaxoSmithKline, Gilead, and Anthem. T.L.R., F.B., A.M., R.A., Y.Y.C., C.T.M., and E.S. are listed on patent applications related to this work., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Novel extragenic genomic safe harbors for precise therapeutic T-cell engineering.
- Author
-
Odak A, Yuan H, Feucht J, Cantu VA, Mansilla-Soto J, Kogel F, Eyquem J, Everett J, Bushman FD, Leslie CS, and Sadelain M
- Subjects
- Animals, Mice, Humans, Genetic Vectors, Immunotherapy, Adoptive, Cell Engineering, Genomics, Antigens, CD19, T-Lymphocytes, Receptors, Antigen, T-Cell
- Abstract
Cell therapies that rely on engineered immune cells can be enhanced by achieving uniform and controlled transgene expression in order to maximize T-cell function and achieve predictable patient responses. Although they are effective, current genetic engineering strategies that use γ-retroviral, lentiviral, and transposon-based vectors to integrate transgenes, unavoidably produce variegated transgene expression in addition to posing a risk of insertional mutagenesis. In the setting of chimeric antigen receptor (CAR) therapy, inconsistent and random CAR expression may result in tonic signaling, T-cell exhaustion, and variable T-cell persistence. Here, we report and validate an algorithm for the identification of extragenic genomic safe harbors (GSH) that can be efficiently targeted for DNA integration and can support sustained and predictable CAR expression in human peripheral blood T cells. The algorithm is based on 7 criteria established to minimize genotoxicity by directing transgene integration away from functionally important genomic elements, maximize efficient CRISPR/Cas9-mediated targeting, and avert transgene silencing over time. T cells engineered to express a CD19 CAR at GSH6, which meets all 7 criteria, are curative at low cell dose in a mouse model of acute lymphoblastic leukemia, matching the potency of CAR T cells engineered at the TRAC locus and effectively resisting tumor rechallenge 100 days after their infusion. The identification of functional extragenic GSHs thus expands the human genome available for therapeutic precision engineering., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
15. Peptide-mediated delivery of CRISPR enzymes for the efficient editing of primary human lymphocytes.
- Author
-
Foss DV, Muldoon JJ, Nguyen DN, Carr D, Sahu SU, Hunsinger JM, Wyman SK, Krishnappa N, Mendonsa R, Schanzer EV, Shy BR, Vykunta VS, Allain V, Li Z, Marson A, Eyquem J, and Wilson RC
- Subjects
- Humans, Mice, Animals, T-Lymphocytes metabolism, Peptides genetics, Ribonucleoproteins, CRISPR-Cas Systems, Gene Editing methods
- Abstract
CRISPR-mediated genome editing of primary human lymphocytes is typically carried out via electroporation, which can be cytotoxic, cumbersome and costly. Here we show that the yields of edited primary human lymphocytes can be increased substantially by delivering a CRISPR ribonucleoprotein mixed with an amphiphilic peptide identified through screening. We evaluated the performance of this simple delivery method by knocking out genes in T cells, B cells and natural killer cells via the delivery of Cas9 or Cas12a ribonucleoproteins or an adenine base editor. We also show that peptide-mediated ribonucleoprotein delivery paired with an adeno-associated-virus-mediated homology-directed repair template can introduce a chimaeric antigen receptor gene at the T-cell receptor α constant locus, and that the engineered cells display antitumour potency in mice. The method is minimally perturbative, does not require dedicated hardware, and is compatible with multiplexed editing via sequential delivery, which minimizes the risk of genotoxicity. The peptide-mediated intracellular delivery of ribonucleoproteins may facilitate the manufacturing of engineered T cells., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
16. High-yield genome engineering in primary cells using a hybrid ssDNA repair template and small-molecule cocktails.
- Author
-
Shy BR, Vykunta VS, Ha A, Talbot A, Roth TL, Nguyen DN, Pfeifer WG, Chen YY, Blaeschke F, Shifrut E, Vedova S, Mamedov MR, Chung JJ, Li H, Yu R, Wu D, Wolf J, Martin TG, Castro CE, Ye L, Esensten JH, Eyquem J, and Marson A
- Subjects
- Humans, Genome, Recombinational DNA Repair, Mutation, DNA, Gene Editing, DNA End-Joining Repair, CRISPR-Cas Systems genetics, DNA, Single-Stranded genetics
- Abstract
Enhancing CRISPR-mediated site-specific transgene insertion efficiency by homology-directed repair (HDR) using high concentrations of double-stranded DNA (dsDNA) with Cas9 target sequences (CTSs) can be toxic to primary cells. Here, we develop single-stranded DNA (ssDNA) HDR templates (HDRTs) incorporating CTSs with reduced toxicity that boost knock-in efficiency and yield by an average of around two- to threefold relative to dsDNA CTSs. Using small-molecule combinations that enhance HDR, we could further increase knock-in efficiencies by an additional roughly two- to threefold on average. Our method works across a variety of target loci, knock-in constructs and primary human cell types, reaching HDR efficiencies of >80-90%. We demonstrate application of this approach for both pathogenic gene variant modeling and gene-replacement strategies for IL2RA and CTLA4 mutations associated with Mendelian disorders. Finally, we develop a good manufacturing practice (GMP)-compatible process for nonviral chimeric antigen receptor-T cell manufacturing, with knock-in efficiencies (46-62%) and yields (>1.5 × 10
9 modified cells) exceeding those of conventional approaches., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2023
- Full Text
- View/download PDF
17. An evolved AAV variant enables efficient genetic engineering of murine T cells.
- Author
-
Nyberg WA, Ark J, To A, Clouden S, Reeder G, Muldoon JJ, Chung JY, Xie WH, Allain V, Steinhart Z, Chang C, Talbot A, Kim S, Rosales A, Havlik LP, Pimentel H, Asokan A, and Eyquem J
- Subjects
- Animals, Mice, CRISPR-Cas Systems genetics, Gene Targeting, Dependovirus genetics, Genetic Engineering methods, T-Lymphocytes
- Abstract
Precise targeting of large transgenes to T cells using homology-directed repair has been transformative for adoptive cell therapies and T cell biology. Delivery of DNA templates via adeno-associated virus (AAV) has greatly improved knockin efficiencies, but the tropism of current AAV serotypes restricts their use to human T cells employed in immunodeficient mouse models. To enable targeted knockins in murine T cells, we evolved Ark313, a synthetic AAV that exhibits high transduction efficiency in murine T cells. We performed a genome-wide knockout screen and identified QA2 as an essential factor for Ark313 infection. We demonstrate that Ark313 can be used for nucleofection-free DNA delivery, CRISPR-Cas9-mediated knockouts, and targeted integration of large transgenes. Ark313 enables preclinical modeling of Trac-targeted CAR-T and transgenic TCR-T cells in immunocompetent models. Efficient gene targeting in murine T cells holds great potential for improved cell therapies and opens avenues in experimental T cell immunology., Competing Interests: Declaration of interests J.A., W.A.N., J.E., and A.A. are co-inventors on a patent application filed on the subject matter of this study. A.A. is a cofounder and board member at StrideBio and TorqueBio. J.E. is a compensated co-founder at Mnemo Therapeutics and a compensated scientific advisor to Cytovia Therapeutics. J.E. owns stocks in Mnemo Therapeutics and Cytovia Therapeutics. J.E. has received a consulting fee from Casdin Capital, Resolution Therapeutics and Treefrog Therapeutics. The J.E. lab has received research support from Cytovia Therapeutics, Mnemo Therapeutics, and Takeda Pharmaceutical Company., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Pooled screening of CAR T cells identifies diverse immune signaling domains for next-generation immunotherapies.
- Author
-
Goodman DB, Azimi CS, Kearns K, Talbot A, Garakani K, Garcia J, Patel N, Hwang B, Lee D, Park E, Vykunta VS, Shy BR, Ye CJ, Eyquem J, Marson A, Bluestone JA, and Roybal KT
- Subjects
- Humans, B-Cell Maturation Antigen, Immunotherapy, Adoptive methods, T-Lymphocytes, Immunotherapy, Signal Transduction, Neoplasm Recurrence, Local metabolism, Receptors, Chimeric Antigen metabolism
- Abstract
Chimeric antigen receptors (CARs) repurpose natural signaling components to retarget T cells to refractory cancers but have shown limited efficacy in persistent, recurrent malignancies. Here, we introduce "CAR Pooling," a multiplexed approach to rapidly identify CAR designs with clinical potential. Forty CARs with signaling domains derived from a range of immune cell lineages were evaluated in pooled assays for their ability to stimulate critical T cell effector functions during repetitive stimulation that mimics long-term tumor antigen exposure. Several domains were identified from the tumor necrosis factor (TNF) receptor family that have been primarily associated with B cells. CD40 enhanced proliferation, whereas B cell-activating factor receptor (BAFF-R) and transmembrane activator and CAML interactor (TACI) promoted cytotoxicity. These functions were enhanced relative to clinical benchmarks after prolonged antigen stimulation, and CAR T cell signaling through these domains fell into distinct states of memory, cytotoxicity, and metabolism. BAFF-R CAR T cells were enriched for a highly cytotoxic transcriptional signature previously associated with positive clinical outcomes. We also observed that replacing the 4-1BB intracellular signaling domain with the BAFF-R signaling domain in a clinically validated B cell maturation antigen (BCMA)-specific CAR resulted in enhanced activity in a xenotransplant model of multiple myeloma. Together, these results show that CAR Pooling is a general approach for rapid exploration of CAR architecture and activity to improve the efficacy of CAR T cell therapies.
- Published
- 2022
- Full Text
- View/download PDF
19. Generation of T-cell-receptor-negative CD8αβ-positive CAR T cells from T-cell-derived induced pluripotent stem cells.
- Author
-
van der Stegen SJC, Lindenbergh PL, Petrovic RM, Xie H, Diop MP, Alexeeva V, Shi Y, Mansilla-Soto J, Hamieh M, Eyquem J, Cabriolu A, Wang X, Abujarour R, Lee T, Clarke R, Valamehr B, Themeli M, Riviere I, and Sadelain M
- Subjects
- Mice, Animals, Humans, T-Lymphocytes, Receptors, Antigen, T-Cell, CD8 Antigens metabolism, Induced Pluripotent Stem Cells metabolism, Receptors, Chimeric Antigen metabolism
- Abstract
The production of autologous T cells expressing a chimaeric antigen receptor (CAR) is time-consuming, costly and occasionally unsuccessful. T-cell-derived induced pluripotent stem cells (TiPS) are a promising source for the generation of 'off-the-shelf' CAR T cells, but the in vitro differentiation of TiPS often yields T cells with suboptimal features. Here we show that the premature expression of the T-cell receptor (TCR) or a constitutively expressed CAR in TiPS promotes the acquisition of an innate phenotype, which can be averted by disabling the TCR and relying on the CAR to drive differentiation. Delaying CAR expression and calibrating its signalling strength in TiPS enabled the generation of human TCR
- CD8αβ+ CAR T cells that perform similarly to CD8αβ+ CAR T cells from peripheral blood, achieving effective tumour control on systemic administration in a mouse model of leukaemia and without causing graft-versus-host disease. Driving T-cell maturation in TiPS in the absence of a TCR by taking advantage of a CAR may facilitate the large-scale development of potent allogeneic CD8αβ+ T cells for a broad range of immunotherapies., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
20. NUDT21 limits CD19 levels through alternative mRNA polyadenylation in B cell acute lymphoblastic leukemia.
- Author
-
Witkowski MT, Lee S, Wang E, Lee AK, Talbot A, Ma C, Tsopoulidis N, Brumbaugh J, Zhao Y, Roberts KG, Hogg SJ, Nomikou S, Ghebrechristos YE, Thandapani P, Mullighan CG, Hochedlinger K, Chen W, Abdel-Wahab O, Eyquem J, and Aifantis I
- Subjects
- Antigens, CD19 genetics, Antigens, CD19 metabolism, Cleavage And Polyadenylation Specificity Factor metabolism, Humans, Immunotherapy, Adoptive adverse effects, Membrane Glycoproteins metabolism, Polyadenylation, RNA, Messenger genetics, RNA, Messenger metabolism, Trans-Activators metabolism, Burkitt Lymphoma, Lymphoma, B-Cell, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Receptors, Chimeric Antigen metabolism
- Abstract
B cell progenitor acute lymphoblastic leukemia (B-ALL) treatment has been revolutionized by T cell-based immunotherapies-including chimeric antigen receptor T cell therapy (CAR-T) and the bispecific T cell engager therapeutic, blinatumomab-targeting surface glycoprotein CD19. Unfortunately, many patients with B-ALL will fail immunotherapy due to 'antigen escape'-the loss or absence of leukemic CD19 targeted by anti-leukemic T cells. In the present study, we utilized a genome-wide CRISPR-Cas9 screening approach to identify modulators of CD19 abundance on human B-ALL blasts. These studies identified a critical role for the transcriptional activator ZNF143 in CD19 promoter activation. Conversely, the RNA-binding protein, NUDT21, limited expression of CD19 by regulating CD19 messenger RNA polyadenylation and stability. NUDT21 deletion in B-ALL cells increased the expression of CD19 and the sensitivity to CD19-specific CAR-T and blinatumomab. In human B-ALL patients treated with CAR-T and blinatumomab, upregulation of NUDT21 mRNA coincided with CD19 loss at disease relapse. Together, these studies identify new CD19 modulators in human B-ALL., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
21. RASA2 ablation in T cells boosts antigen sensitivity and long-term function.
- Author
-
Carnevale J, Shifrut E, Kale N, Nyberg WA, Blaeschke F, Chen YY, Li Z, Bapat SP, Diolaiti ME, O'Leary P, Vedova S, Belk J, Daniel B, Roth TL, Bachl S, Anido AA, Prinzing B, Ibañez-Vega J, Lange S, Haydar D, Luetke-Eversloh M, Born-Bony M, Hegde B, Kogan S, Feuchtinger T, Okada H, Satpathy AT, Shannon K, Gottschalk S, Eyquem J, Krenciute G, Ashworth A, and Marson A
- Subjects
- Animals, Bone Marrow, CRISPR-Cas Systems, Disease Models, Animal, Gene Knockdown Techniques, Humans, Immunotherapy, Adoptive, Leukemia immunology, Leukemia pathology, Leukemia therapy, Mice, Receptors, Antigen, T-Cell immunology, Receptors, Chimeric Antigen immunology, Time Factors, Xenograft Model Antitumor Assays, Antigens, Neoplasm immunology, Neoplasms immunology, Neoplasms pathology, Neoplasms therapy, T-Lymphocytes immunology, T-Lymphocytes metabolism, ras GTPase-Activating Proteins deficiency, ras GTPase-Activating Proteins genetics
- Abstract
The efficacy of adoptive T cell therapies for cancer treatment can be limited by suppressive signals from both extrinsic factors and intrinsic inhibitory checkpoints
1,2 . Targeted gene editing has the potential to overcome these limitations and enhance T cell therapeutic function3-10 . Here we performed multiple genome-wide CRISPR knock-out screens under different immunosuppressive conditions to identify genes that can be targeted to prevent T cell dysfunction. These screens converged on RASA2, a RAS GTPase-activating protein (RasGAP) that we identify as a signalling checkpoint in human T cells, which is downregulated upon acute T cell receptor stimulation and can increase gradually with chronic antigen exposure. RASA2 ablation enhanced MAPK signalling and chimeric antigen receptor (CAR) T cell cytolytic activity in response to target antigen. Repeated tumour antigen stimulations in vitro revealed that RASA2-deficient T cells show increased activation, cytokine production and metabolic activity compared with control cells, and show a marked advantage in persistent cancer cell killing. RASA2-knockout CAR T cells had a competitive fitness advantage over control cells in the bone marrow in a mouse model of leukaemia. Ablation of RASA2 in multiple preclinical models of T cell receptor and CAR T cell therapies prolonged survival in mice xenografted with either liquid or solid tumours. Together, our findings highlight RASA2 as a promising target to enhance both persistence and effector function in T cell therapies for cancer treatment., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
22. The surfaceome of multiple myeloma cells suggests potential immunotherapeutic strategies and protein markers of drug resistance.
- Author
-
Ferguson ID, Patiño-Escobar B, Tuomivaara ST, Lin YT, Nix MA, Leung KK, Kasap C, Ramos E, Nieves Vasquez W, Talbot A, Hale M, Naik A, Kishishita A, Choudhry P, Lopez-Girona A, Miao W, Wong SW, Wolf JL, Martin TG 3rd, Shah N, Vandenberg S, Prakash S, Besse L, Driessen C, Posey AD Jr, Mullins RD, Eyquem J, Wells JA, and Wiita AP
- Subjects
- Drug Resistance, Humans, Immunotherapy methods, Lenalidomide therapeutic use, Proteomics, Tumor Microenvironment, Multiple Myeloma drug therapy, Multiple Myeloma pathology
- Abstract
The myeloma surface proteome (surfaceome) determines tumor interaction with the microenvironment and serves as an emerging arena for therapeutic development. Here, we use glycoprotein capture proteomics to define the myeloma surfaceome at baseline, in drug resistance, and in response to acute drug treatment. We provide a scoring system for surface antigens and identify CCR10 as a promising target in this disease expressed widely on malignant plasma cells. We engineer proof-of-principle chimeric antigen receptor (CAR) T-cells targeting CCR10 using its natural ligand CCL27. In myeloma models we identify proteins that could serve as markers of resistance to bortezomib and lenalidomide, including CD53, CD10, EVI2B, and CD33. We find that acute lenalidomide treatment increases activity of MUC1-targeting CAR-T cells through antigen upregulation. Finally, we develop a miniaturized surface proteomic protocol for profiling primary plasma cell samples with low inputs. These approaches and datasets may contribute to the biological, therapeutic, and diagnostic understanding of myeloma., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
23. HLA-independent T cell receptors for targeting tumors with low antigen density.
- Author
-
Mansilla-Soto J, Eyquem J, Haubner S, Hamieh M, Feucht J, Paillon N, Zucchetti AE, Li Z, Sjöstrand M, Lindenbergh PL, Saetersmoen M, Dobrin A, Maurin M, Iyer A, Garcia Angus A, Miele MM, Zhao Z, Giavridis T, van der Stegen SJC, Tamzalit F, Rivière I, Huse M, Hendrickson RC, Hivroz C, and Sadelain M
- Subjects
- Animals, Antigens, CD19, Histocompatibility Antigens, Humans, Immunotherapy, Adoptive, Mice, Receptors, Antigen, T-Cell, Xenograft Model Antitumor Assays, Leukemia, Myeloid, Acute, Receptors, Chimeric Antigen metabolism
- Abstract
Chimeric antigen receptors (CARs) are receptors for antigen that direct potent immune responses. Tumor escape associated with low target antigen expression is emerging as one potential limitation of their efficacy. Here we edit the TRAC locus in human peripheral blood T cells to engage cell-surface targets through their T cell receptor-CD3 complex reconfigured to utilize the same immunoglobulin heavy and light chains as a matched CAR. We demonstrate that these HLA-independent T cell receptors (HIT receptors) consistently afford high antigen sensitivity and mediate tumor recognition beyond what CD28-based CARs, the most sensitive design to date, can provide. We demonstrate that the functional persistence of HIT T cells can be augmented by constitutive coexpression of CD80 and 4-1BBL. Finally, we validate the increased antigen sensitivity afforded by HIT receptors in xenograft mouse models of B cell leukemia and acute myeloid leukemia, targeting CD19 and CD70, respectively. Overall, HIT receptors are well suited for targeting cell surface antigens of low abundance., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
24. The CD28-Transmembrane Domain Mediates Chimeric Antigen Receptor Heterodimerization With CD28.
- Author
-
Muller YD, Nguyen DP, Ferreira LMR, Ho P, Raffin C, Valencia RVB, Congrave-Wilson Z, Roth TL, Eyquem J, Van Gool F, Marson A, Perez L, Wells JA, Bluestone JA, and Tang Q
- Subjects
- Antigens, CD19 immunology, Dimerization, Humans, Lymphocyte Activation immunology, Signal Transduction immunology, T-Lymphocytes immunology, CD28 Antigens immunology, Protein Domains immunology, Receptors, Chimeric Antigen immunology
- Abstract
Anti-CD19 chimeric antigen receptor (CD19-CAR)-engineered T cells are approved therapeutics for malignancies. The impact of the hinge domain (HD) and the transmembrane domain (TMD) between the extracellular antigen-targeting CARs and the intracellular signaling modalities of CARs has not been systemically studied. In this study, a series of 19-CARs differing only by their HD (CD8, CD28, or IgG
4 ) and TMD (CD8 or CD28) was generated. CARs containing a CD28-TMD, but not a CD8-TMD, formed heterodimers with the endogenous CD28 in human T cells, as shown by co-immunoprecipitation and CAR-dependent proliferation of anti-CD28 stimulation. This dimerization was dependent on polar amino acids in the CD28-TMD and was more efficient with CARs containing CD28 or CD8 HD than IgG4 -HD. The CD28-CAR heterodimers did not respond to CD80 and CD86 stimulation but had a significantly reduced CD28 cell-surface expression. These data unveiled a fundamental difference between CD28-TMD and CD8-TMD and indicated that CD28-TMD can modulate CAR T-cell activities by engaging endogenous partners., Competing Interests: A provisional patent on CAR-CD28 heterodimerization has been submitted. JAB and QT are co-founders of Sonoma Biotherapeutics. AM and TR are co-founders of Arsenal Biosciences. AM is also a co-founder of Spotlight Therapeutics. JAB and AM have served as advisors to Juno Therapeutics. AM was a member of the scientific advisory board at PACT Pharma and was an advisor to Trizell. QT, JAB, and AM have received sponsored research support from Juno Therapeutics. AM is cofounder, member of the Boards of Directors and member of Scientific Advisory Boards of Spotlight Therapeutics and Arsenal Biosciences. AM has served as an advisor to Juno Therapeutics, was a member of the scientific advisory board at PACT Pharma and was an advisor to Trizell. AM has received honoraria from Merck, a consulting fee from AlphaSights, and is an investor in and informal advisor to Offline Ventures. AM owns stock in Arsenal Biosciences, Spotlight Therapeutics and PACT Pharma. AM has received research support from Epinomics, Sanofi, GlaxoSmithKline, and gifts from Gilead and Anthem. JAW is co-Founder of Soteria Biotherapeutics developing small molecule switchable biologics, on the SAB of Spotlight, and recipient of sponsored research from Bristol Myers Squibb. JE is an advisor for Mnemo Thérapeutics and Cytovia and received research support from Cytovia. TLR is a co-founder, member of Scientific Advisory Board, and founding Chief Scientific Officer of Arsenal Biosciences. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Muller, Nguyen, Ferreira, Ho, Raffin, Valencia, Congrave-Wilson, Roth, Eyquem, Van Gool, Marson, Perez, Wells, Bluestone and Tang.)- Published
- 2021
- Full Text
- View/download PDF
25. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape.
- Author
-
Hamieh M, Dobrin A, Cabriolu A, van der Stegen SJC, Giavridis T, Mansilla-Soto J, Eyquem J, Zhao Z, Whitlock BM, Miele MM, Li Z, Cunanan KM, Huse M, Hendrickson RC, Wang X, Rivière I, and Sadelain M
- Subjects
- 4-1BB Ligand immunology, Animals, CD28 Antigens immunology, Cytotoxicity, Immunologic, Female, Immunotherapy, Adoptive, Leukemia pathology, Male, Mice, Mice, Inbred NOD, Neoplasm Recurrence, Local immunology, T-Lymphocytes cytology, Antigens, Neoplasm immunology, Antigens, Neoplasm metabolism, Leukemia immunology, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Tumor Escape immunology
- Abstract
Chimeric antigen receptors (CARs) are synthetic antigen receptors that reprogram T cell specificity, function and persistence
1 . Patient-derived CAR T cells have demonstrated remarkable efficacy against a range of B-cell malignancies1-3 , and the results of early clinical trials suggest activity in multiple myeloma4 . Despite high complete response rates, relapses occur in a large fraction of patients; some of these are antigen-negative and others are antigen-low1,2,4-9 . Unlike the mechanisms that result in complete and permanent antigen loss6,8,9 , those that lead to escape of antigen-low tumours remain unclear. Here, using mouse models of leukaemia, we show that CARs provoke reversible antigen loss through trogocytosis, an active process in which the target antigen is transferred to T cells, thereby decreasing target density on tumour cells and abating T cell activity by promoting fratricide T cell killing and T cell exhaustion. These mechanisms affect both CD28- and 4-1BB-based CARs, albeit differentially, depending on antigen density. These dynamic features can be offset by cooperative killing and combinatorial targeting to augment tumour responses to immunotherapy.- Published
- 2019
- Full Text
- View/download PDF
26. Publisher Correction: Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency.
- Author
-
Feucht J, Sun J, Eyquem J, Ho YJ, Zhao Z, Leibold J, Dobrin A, Cabriolu A, Hamieh M, and Sadelain M
- Abstract
In the version of this article originally published, there was an error in the legend for Extended Data Fig. 7. The legend for panel f was originally: "f, FACS analysis of IL7R
- , CD62L- and CD45RA- expression on TRAC-1928ζ and TRAC-1XX CAR T cells at day 63 post CAR infusion (representative for at least n = 3 mice per group in one independent experiment)." The legend should have been: "f, FACS analysis of IL7R+ , CD62L+ and CD45RA+ expression on TRAC-1928ζ and TRAC-1XX CAR T cells at day 63 post CAR infusion (representative for at least n = 3 mice per group in one independent experiment)." The error has been corrected in the HTML and PDF versions of this article.- Published
- 2019
- Full Text
- View/download PDF
27. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency.
- Author
-
Feucht J, Sun J, Eyquem J, Ho YJ, Zhao Z, Leibold J, Dobrin A, Cabriolu A, Hamieh M, and Sadelain M
- Subjects
- Animals, Calibration, Cell Line, Male, Mice, Protein Domains, Receptors, Antigen, T-Cell chemistry, Cell Lineage, Immunotherapy, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes immunology
- Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that target and reprogram T cells to acquire augmented antitumor properties
1 . CD19-specific CARs that comprise CD28 and CD3ζ signaling motifs2 have induced remarkable responses in patients with refractory leukemia3-5 and lymphoma6 and were recently approved by the US Food and Drug Administration7 . These CARs program highly performing effector functions that mediate potent tumor elimination4,8 despite the limited persistence they confer on T cells3-6,8 . Extending their functional persistence without compromising their potency should improve current CAR therapies. Strong T cell activation drives exhaustion9,10 , which may be accentuated by the redundancy of CD28 and CD3ζ signaling11,12 as well as the spatiotemporal constraints imparted by the structure of second-generation CARs2 . Thus, we hypothesized that calibrating the activation potential of CD28-based CARs would differentially reprogram T cell function and differentiation. Here, we show that CARs encoding a single immunoreceptor tyrosine-based activation motif direct T cells to different fates by balancing effector and memory programs, thereby yielding CAR designs with enhanced therapeutic profiles.- Published
- 2019
- Full Text
- View/download PDF
28. Antibody with Infinite Affinity for In Vivo Tracking of Genetically Engineered Lymphocytes.
- Author
-
Krebs S, Ahad A, Carter LM, Eyquem J, Brand C, Bell M, Ponomarev V, Reiner T, Meares CF, Gottschalk S, Sadelain M, Larson SM, and Weber WA
- Subjects
- Animals, Cell Line, Tumor, Genes, Reporter, Heterocyclic Compounds, 1-Ring, Humans, Immunoglobulin Fragments genetics, Immunoglobulin Fragments metabolism, Immunotherapy, Adoptive, Lutetium, Male, Mice, Mice, Inbred ICR, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Positron Emission Tomography Computed Tomography methods, Radiation Dosage, Radioisotopes, Radiopharmaceuticals chemistry, Radiopharmaceuticals pharmacokinetics, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Single Photon Emission Computed Tomography Computed Tomography methods, T-Lymphocytes metabolism, Tissue Distribution, Xenograft Model Antitumor Assays, Yttrium Radioisotopes, Cell Tracking methods, T-Lymphocytes cytology, T-Lymphocytes immunology
- Abstract
There remains an urgent need for the noninvasive tracking of transfused chimeric antigen receptor (CAR) T cells to determine their biodistribution, viability, expansion, and antitumor functionality. DOTA antibody reporter 1 (DAbR1) comprises a single-chain fragment of the antilanthanoid-DOTA antibody 2D12.5/G54C fused to the human CD4-transmembrane domain and binds irreversibly to lanthanoid ( S )-2-(4-acrylamidobenzyl)-DOTA (AABD). The aim of this study was to investigate whether DAbR1 can be expressed on lymphocytes and used as a reporter gene as well as a suicide gene for therapy of immune-related adverse effects. Methods: DAbR1 was subcloned together with green fluorescent protein into an SFG-retroviral vector and used to transduce CD3/CD28-activated primary human T cells and second-generation 1928z (CAR) T cells. Cell surface expression of DAbR1 was confirmed by cell uptake studies with radiolabeled AABD. In addition, the feasibility of imaging of DAbR1-positive T cells in vivo after intravenous injection of
86 Y/177 Lu-AABD was studied and radiation doses determined. Results: A panel of DAbR1-expressing T cells and CAR T cells exhibited greater than 8-fold increased uptake of86 Y-AABD in vitro when compared with nontransduced cells. Imaging studies showed86 Y-AABD was retained by DAbR1-positive T cells while it continuously cleared from normal tissues, allowing for in vivo tracking of intravenously administered CAR T cells. Normal-organ dose estimates were favorable for repeated PET/CT studies. Selective T cell ablation in vivo with177 Lu-AABD seems feasible for clustered T-cell populations. Conclusion: We have demonstrated for the first time that T cells can be modified with DAbR1, enabling their in vivo tracking via PET and SPECT. The favorable biodistribution and high image contrast observed warrant further studies of this new reporter gene., (© 2018 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2018
- Full Text
- View/download PDF
29. Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape.
- Author
-
DeSelm C, Palomba ML, Yahalom J, Hamieh M, Eyquem J, Rajasekhar VK, and Sadelain M
- Subjects
- Animals, Antigens, CD19 immunology, Antigens, Neoplasm chemistry, Antigens, Neoplasm immunology, Antigens, Neoplasm radiation effects, CA-19-9 Antigen, Combined Modality Therapy, Disease Models, Animal, Humans, Insulin-Secreting Cells immunology, Insulin-Secreting Cells radiation effects, Mice, Oligosaccharides chemistry, Oligosaccharides immunology, Oligosaccharides therapeutic use, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Radiation, Radiation Dosage, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen therapeutic use, Sequence Analysis, RNA, TNF-Related Apoptosis-Inducing Ligand immunology, Antigens, CD19 therapeutic use, Immunotherapy, Adoptive, Pancreatic Neoplasms immunology, Pancreatic Neoplasms radiotherapy, TNF-Related Apoptosis-Inducing Ligand genetics
- Abstract
CD19 chimeric antigen receptors (CARs) have demonstrated great efficacy against a range of B cell malignancies. However, antigen escape and, more generally, heterogeneous antigen expression pose a challenge to applying CAR therapy to a wide range of cancers. We find that low-dose radiation sensitizes tumor cells to immune rejection by locally activated CAR T cells. In a model of pancreatic adenocarcinoma heterogeneously expressing sialyl Lewis-A (sLeA), we show that not only sLeA
+ but also sLeA- tumor cells exposed to low-dose radiation become susceptible to CAR therapy, reducing antigen-negative tumor relapse. RNA sequencing analysis of low-dose radiation-exposed tumors reveals the transcriptional signature of cells highly sensitive to TRAIL-mediated death. We find that sLeA-targeted CAR T cells produce TRAIL upon engaging sLeA+ tumor cells, and eliminate sLeA- tumor cells previously exposed to systemic or local low-dose radiation in a TRAIL-dependent manner. These findings enhance the prospects for successfully applying CAR therapy to heterogeneous solid tumors. Local radiation is integral to many tumors' standard of care and can be easily implemented as a CAR conditioning regimen., (Copyright © 2018 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
30. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade.
- Author
-
Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, and Sadelain M
- Subjects
- Animals, Humans, Interleukin 1 Receptor Antagonist Protein metabolism, Interleukin-1 metabolism, Mice, Myeloid Cells metabolism, Neoplasms immunology, Neoplasms pathology, Syndrome, Cytokines metabolism, Immunotherapy, Adoptive, Interleukin-1 antagonists & inhibitors, Macrophages metabolism
- Abstract
Chimeric antigen receptor (CAR) therapy targeting CD19 is an effective treatment for refractory B cell malignancies, especially acute lymphoblastic leukemia (ALL)
1 . Although a majority of patients will achieve a complete response following a single infusion of CD19-targeted CAR-modified T cells (CD19 CAR T cells)2-4 , the broad applicability of this treatment is hampered by severe cytokine release syndrome (CRS), which is characterized by fever, hypotension and respiratory insufficiency associated with elevated serum cytokines, including interleukin-6 (IL-6)2,5 . CRS usually occurs within days of T cell infusion at the peak of CAR T cell expansion. In ALL, it is most frequent and more severe in patients with high tumor burden2-4 . CRS may respond to IL-6 receptor blockade but can require further treatment with high dose corticosteroids to curb potentially lethal severity2-9 . Improved therapeutic and preventive treatments require a better understanding of CRS physiopathology, which has so far remained elusive. Here we report a murine model of CRS that develops within 2-3 d of CAR T cell infusion and that is potentially lethal and responsive to IL-6 receptor blockade. We show that its severity is mediated not by CAR T cell-derived cytokines, but by IL-6, IL-1 and nitric oxide (NO) produced by recipient macrophages, which enables new therapeutic interventions.- Published
- 2018
- Full Text
- View/download PDF
31. Integrating Proteomics and Transcriptomics for Systematic Combinatorial Chimeric Antigen Receptor Therapy of AML.
- Author
-
Perna F, Berman SH, Soni RK, Mansilla-Soto J, Eyquem J, Hamieh M, Hendrickson RC, Brennan CW, and Sadelain M
- Subjects
- Cell Line, Tumor, Humans, Leukemia, Myeloid, Acute immunology, Recombinant Fusion Proteins metabolism, T-Lymphocytes metabolism, Antigens, CD19, Gene Expression Profiling, Immunotherapy methods, Leukemia, Myeloid, Acute therapy, Proteomics, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes immunology
- Abstract
Chimeric antigen receptor (CAR) therapy targeting CD19 has yielded remarkable outcomes in patients with acute lymphoblastic leukemia. To identify potential CAR targets in acute myeloid leukemia (AML), we probed the AML surfaceome for overexpressed molecules with tolerable systemic expression. We integrated large transcriptomics and proteomics datasets from malignant and normal tissues, and developed an algorithm to identify potential targets expressed in leukemia stem cells, but not in normal CD34
+ CD38- hematopoietic cells, T cells, or vital tissues. As these investigations did not uncover candidate targets with a profile as favorable as CD19, we developed a generalizable combinatorial targeting strategy fulfilling stringent efficacy and safety criteria. Our findings indicate that several target pairings hold great promise for CAR therapy of AML., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
32. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection.
- Author
-
Eyquem J, Mansilla-Soto J, Giavridis T, van der Stegen SJ, Hamieh M, Cunanan KM, Odak A, Gönen M, and Sadelain M
- Subjects
- Animals, Antigens, CD19 immunology, Cell Differentiation genetics, Cell Differentiation immunology, Disease Models, Animal, Gene Expression Regulation, Genetic Loci genetics, Humans, Lymphocyte Activation, Male, Mice, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Promoter Regions, Genetic genetics, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta immunology, T-Lymphocytes cytology, T-Lymphocytes metabolism, Translational Research, Biomedical, CRISPR-Cas Systems, Gene Editing, Immunotherapy methods, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology
- Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that redirect and reprogram T cells to mediate tumour rejection. The most successful CARs used to date are those targeting CD19 (ref. 2), which offer the prospect of complete remission in patients with chemorefractory or relapsed B-cell malignancies. CARs are typically transduced into the T cells of a patient using γ-retroviral vectors or other randomly integrating vectors, which may result in clonal expansion, oncogenic transformation, variegated transgene expression and transcriptional silencing. Recent advances in genome editing enable efficient sequence-specific interventions in human cells, including targeted gene delivery to the CCR5 and AAVS1 loci. Here we show that directing a CD19-specific CAR to the T-cell receptor α constant (TRAC) locus not only results in uniform CAR expression in human peripheral blood T cells, but also enhances T-cell potency, with edited cells vastly outperforming conventionally generated CAR T cells in a mouse model of acute lymphoblastic leukaemia. We further demonstrate that targeting the CAR to the TRAC locus averts tonic CAR signalling and establishes effective internalization and re-expression of the CAR following single or repeated exposure to antigen, delaying effector T-cell differentiation and exhaustion. These findings uncover facets of CAR immunobiology and underscore the potential of CRISPR/Cas9 genome editing to advance immunotherapies.
- Published
- 2017
- Full Text
- View/download PDF
33. Multiplex Genome-Edited T-cell Manufacturing Platform for "Off-the-Shelf" Adoptive T-cell Immunotherapies.
- Author
-
Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, Potrel P, Bas C, Lemaire L, Galetto R, Lebuhotel C, Eyquem J, Cheung GW, Duclert A, Gouble A, Arnould S, Peggs K, Pule M, Scharenberg AM, and Smith J
- Subjects
- Alemtuzumab, Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Humanized pharmacology, Antigens, CD genetics, Antigens, CD19 immunology, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Base Sequence, CD52 Antigen, Cytotoxicity, Immunologic, Drug Resistance, Glycoproteins deficiency, Glycoproteins genetics, Graft vs Host Disease prevention & control, Humans, Lymphocyte Activation, Lymphoma therapy, Mice, Mice, Mutant Strains, Molecular Sequence Data, RNA, Messenger, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell, alpha-beta deficiency, Receptors, Antigen, T-Cell, alpha-beta genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, T-Lymphocytes drug effects, T-Lymphocytes immunology, Transfection, Xenograft Model Antitumor Assays, Gene Knockout Techniques, Immunotherapy, Adoptive, T-Lymphocytes transplantation
- Abstract
Adoptive immunotherapy using autologous T cells endowed with chimeric antigen receptors (CAR) has emerged as a powerful means of treating cancer. However, a limitation of this approach is that autologous CAR T cells must be generated on a custom-made basis. Here we show that electroporation of transcription activator-like effector nuclease (TALEN) mRNA allows highly efficient multiplex gene editing in primary human T cells. We use this TALEN-mediated editing approach to develop a process for the large-scale manufacturing of T cells deficient in expression of both their αβ T-cell receptor (TCR) and CD52, a protein targeted by alemtuzumab, a chemotherapeutic agent. Functionally, T cells manufactured with this process do not mediate graft-versus-host reactions and are rendered resistant to destruction by alemtuzumab. These characteristics enable the administration of alemtuzumab concurrently or prior to engineered T cells, supporting their engraftment. Furthermore, endowing the TALEN-engineered cells with a CD19 CAR led to efficient destruction of CD19(+) tumor targets even in the presence of the chemotherapeutic agent. These results demonstrate the applicability of TALEN-mediated genome editing to a scalable process, which enables the manufacturing of third-party CAR T-cell immunotherapies against arbitrary targets. As such, CAR T-cell immunotherapies can therefore be used in an "off-the-shelf" manner akin to other biologic immunopharmaceuticals, (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
34. A platform for rapid prototyping of synthetic gene networks in mammalian cells.
- Author
-
Duportet X, Wroblewska L, Guye P, Li Y, Eyquem J, Rieders J, Rimchala T, Batt G, and Weiss R
- Subjects
- Animals, Cell Line, Cloning, Molecular, Gene Library, Humans, Cell Engineering methods, Gene Regulatory Networks
- Abstract
Mammalian synthetic biology may provide novel therapeutic strategies, help decipher new paths for drug discovery and facilitate synthesis of valuable molecules. Yet, our capacity to genetically program cells is currently hampered by the lack of efficient approaches to streamline the design, construction and screening of synthetic gene networks. To address this problem, here we present a framework for modular and combinatorial assembly of functional (multi)gene expression vectors and their efficient and specific targeted integration into a well-defined chromosomal context in mammalian cells. We demonstrate the potential of this framework by assembling and integrating different functional mammalian regulatory networks including the largest gene circuit built and chromosomally integrated to date (6 transcription units, 27kb) encoding an inducible memory device. Using a library of 18 different circuits as a proof of concept, we also demonstrate that our method enables one-pot/single-flask chromosomal integration and screening of circuit libraries. This rapid and powerful prototyping platform is well suited for comparative studies of genetic regulatory elements, genes and multi-gene circuits as well as facile development of libraries of isogenic engineered cell lines., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2014
- Full Text
- View/download PDF
35. Freedom space for rivers: a sustainable management approach to enhance river resilience.
- Author
-
Biron PM, Buffin-Bélanger T, Larocque M, Choné G, Cloutier CA, Ouellet MA, Demers S, Olsen T, Desjarlais C, and Eyquem J
- Subjects
- Conservation of Natural Resources trends, Geography, Quebec, Climate, Conservation of Natural Resources methods, Floods classification, Models, Theoretical, Rivers, Wetlands
- Abstract
River systems are increasingly under stress and pressure from agriculture and urbanization in riparian zones, resulting in frequent engineering interventions such as bank stabilization or flood protection. This study provides guidelines for a more sustainable approach to river management based on hydrogeomorphology concepts applied to three contrasted rivers in Quebec (Canada). Mobility and flooding spaces are determined for the three rivers, and three levels of "freedom space" are subsequently defined based on the combination of the two spaces. The first level of freedom space includes very frequently flooded and highly mobile zones over the next 50 years, as well as riparian wetlands. It provides the minimum space for both fluvial and ecological functionality of the river system. On average for the three studied sites, this minimum space was approximately 1.7 times the channel width, but this minimum space corresponds to a highly variable width which must be determined from a thorough hydrogeomorphic assessment and cannot be predicted using a representative average. The second level includes space for floods of larger magnitude and provides for meanders to migrate freely over a longer time period. The last level of freedom space represents exceptional flood zones. We propose the freedom space concept to be implemented in current river management legislation because it promotes a sustainable way to manage river systems, and it increases their resilience to climate and land use changes in comparison with traditional river management approaches which are based on frequent and spatially restricted interventions.
- Published
- 2014
- Full Text
- View/download PDF
36. Characterization of three loci for homologous gene targeting and transgene expression.
- Author
-
Eyquem J, Poirot L, Galetto R, Scharenberg AM, and Smith J
- Subjects
- Biotechnology methods, Cell Line, Genes, Reporter, Genomic Instability, Humans, Promoter Regions, Genetic, Gene Expression, Gene Targeting, Mutagenesis, Insertional methods, Transgenes
- Abstract
Integrative gene transfer is widely used for bioproduction, drug screening, and therapeutic applications but usual viral methods lead to random and multicopy insertions, contribute to unstable transgene expression and can disturb endogenous gene expression. Homologous targeting of an expression cassette using rare-cutting endonucleases is a potential solution; however the number of studied loci remains limited. Furthermore, the behavior and performance of various types of gene cassettes following gene targeting is poorly defined. Here we have evaluated three loci for gene targeting, including one locus compatible with the proposed Safe Harbor criteria for human translational applications. Using optimized conditions for homologous gene targeting, reporter genes under the control of different promoters were efficiently inserted at each locus in both sense and antisense orientations. Sustainable expression was achieved at all three loci without detectable disturbance of flanking gene expression. However, the promoter, the integration locus and the cassette orientation have a strong impact on transgene expression. Finally, single targeted integrations exhibited greatly improved transgene expression stability versus multicopy or random integration. Taken together, our data suggest a potential set of loci for site-specific transgene integration, suitable for a variety of biotechnological applications., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.