46 results on '"Justin Eyquem"'
Search Results
2. 132 HLA-independent T cell receptors effectively target low abundance antigens
- Author
-
Zeguo Zhao, Mohamad Hamieh, Justin Eyquem, Michel Sadelain, Maria Sjöstrand, Jorge Mansilla-Soto, Sascha Haubner, Judith Feucht, Noémie Paillon, Andres Zucchetti, Zhuoning Li, Pieter Lindenbergh, Michelle Saetersmoen, Mathieu Maurin, Archana Iyer, Anton Dobrin, Andreina Garcia Angus, Matthew Miele, Theodoros Giavridis, Sjoukje van der Stegen, Fella Tamzalit, Morgan Huse, Ronald Hendrickson, and Claire Hivroz
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. The CD28-Transmembrane Domain Mediates Chimeric Antigen Receptor Heterodimerization With CD28
- Author
-
Yannick D. Muller, Duy P. Nguyen, Leonardo M. R. Ferreira, Patrick Ho, Caroline Raffin, Roxxana Valeria Beltran Valencia, Zion Congrave-Wilson, Theodore L. Roth, Justin Eyquem, Frederic Van Gool, Alexander Marson, Laurent Perez, James A. Wells, Jeffrey A. Bluestone, and Qizhi Tang
- Subjects
chimeric antigen receptor ,CAR T cell ,CD28 ,transmembrane domain ,hinge domain ,heterodimerization ,Immunologic diseases. Allergy ,RC581-607 - 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 IgG4) 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.
- Published
- 2021
- Full Text
- View/download PDF
4. High-yield genome engineering in primary cells using a hybrid ssDNA repair template and small-molecule cocktails
- Author
-
Brian R. Shy, Vivasvan S. Vykunta, Alvin Ha, Alexis Talbot, Theodore L. Roth, David N. Nguyen, Wolfgang G. Pfeifer, Yan Yi Chen, Franziska Blaeschke, Eric Shifrut, Shane Vedova, Murad R. Mamedov, Jing-Yi Jing Chung, Hong Li, Ruby Yu, David Wu, Jeffrey Wolf, Thomas G. Martin, Carlos E. Castro, Lumeng Ye, Jonathan H. Esensten, Justin Eyquem, and Alexander Marson
- Subjects
Gene Editing ,Genome ,DNA End-Joining Repair ,5.2 Cellular and gene therapies ,Biomedical Engineering ,Recombinational DNA Repair ,Bioengineering ,DNA ,Applied Microbiology and Biotechnology ,Article ,Single-Stranded ,Mutation ,Genetics ,Humans ,Molecular Medicine ,CRISPR-Cas Systems ,Development of treatments and therapeutic interventions ,Biotechnology - 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 × 109 modified cells) exceeding those of conventional approaches.
- Published
- 2022
- Full Text
- View/download PDF
5. Generation of T-cell-receptor-negative CD8αβ-positive CAR T cells from T-cell-derived induced pluripotent stem cells
- Author
-
Sjoukje J. C. van der Stegen, Pieter L. Lindenbergh, Roseanna M. Petrovic, Hongyao Xie, Mame P. Diop, Vera Alexeeva, Yuzhe Shi, Jorge Mansilla-Soto, Mohamad Hamieh, Justin Eyquem, Annalisa Cabriolu, Xiuyan Wang, Ramzey Abujarour, Tom Lee, Raedun Clarke, Bahram Valamehr, Maria Themeli, Isabelle Riviere, Michel Sadelain, CCA - Cancer biology and immunology, and VU University medical center
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,CD8 Antigens ,Induced Pluripotent Stem Cells ,Receptors, Antigen, T-Cell ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Article ,Computer Science Applications ,Mice ,Animals ,Humans ,Biotechnology - 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.
- Published
- 2022
- Full Text
- View/download PDF
6. Peptide-mediated delivery of CRISPR enzymes for the efficient editing of primary human lymphocytes
- Author
-
Dana V. Foss, Joseph J. Muldoon, David N. Nguyen, Daniel Carr, Srishti U. Sahu, John M. Hunsinger, Stacia K. Wyman, Netravathi Krishnappa, Rima Mendonsa, Elaine V. Schanzer, Brian R. Shy, Vivasvan S. Vykunta, Vincent Allain, Zhongmei Li, Alexander Marson, Justin Eyquem, and Ross C. Wilson
- Subjects
Gene Editing ,5.2 Cellular and gene therapies ,T-Lymphocytes ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Gene Therapy ,Computer Science Applications ,Mice ,Ribonucleoproteins ,Genetics ,Animals ,Humans ,CRISPR-Cas Systems ,Development of treatments and therapeutic interventions ,Peptides ,Biotechnology - 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.
- Published
- 2023
7. Supplementary Figures 1 - 5, Tables 1 - 3 from Multiplex Genome-Edited T-cell Manufacturing Platform for 'Off-the-Shelf' Adoptive T-cell Immunotherapies
- Author
-
Julianne Smith, Andrew M. Scharenberg, Martin Pule, Karl Peggs, Sylvain Arnould, Agnès Gouble, Aymeric Duclert, Gordon Weng-Kit Cheung, Justin Eyquem, Céline Lebuhotel, Roman Galetto, Laetitia Lemaire, Cécile Bas, Pierrick Potrel, Sophie Derniame, Isabelle Chion-Sotinel, Diane Le Clerre, Cécile Schiffer-Mannioui, Brian Philip, and Laurent Poirot
- Abstract
Supplementary figure S1 : Gene disruption analysis in HEK293 cells. Supplementary figure S2: Optimization of the generation of double KO cells Supplementary table S1: Deep sequencing analysis of TCR-negative cells Supplementary figure S3: Representative deletions found at disrupted TRAC gene Supplementary figure S4: Size distribution of the mutations found in TALEN-treated T cells Supplementary table S2: Size of insertions/deletions in mutated CD52/TRAC alleles Supplementary Table S3: NGS sequencing analysis of off target cleavage Supplementary figure S5: Gating strategy for definition of T-cellsubsets in UCART19
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Multiplex Genome-Edited T-cell Manufacturing Platform for 'Off-the-Shelf' Adoptive T-cell Immunotherapies
- Author
-
Julianne Smith, Andrew M. Scharenberg, Martin Pule, Karl Peggs, Sylvain Arnould, Agnès Gouble, Aymeric Duclert, Gordon Weng-Kit Cheung, Justin Eyquem, Céline Lebuhotel, Roman Galetto, Laetitia Lemaire, Cécile Bas, Pierrick Potrel, Sophie Derniame, Isabelle Chion-Sotinel, Diane Le Clerre, Cécile Schiffer-Mannioui, Brian Philip, and Laurent Poirot
- 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. Cancer Res; 75(18); 3853–64. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Methods from Multiplex Genome-Edited T-cell Manufacturing Platform for 'Off-the-Shelf' Adoptive T-cell Immunotherapies
- Author
-
Julianne Smith, Andrew M. Scharenberg, Martin Pule, Karl Peggs, Sylvain Arnould, Agnès Gouble, Aymeric Duclert, Gordon Weng-Kit Cheung, Justin Eyquem, Céline Lebuhotel, Roman Galetto, Laetitia Lemaire, Cécile Bas, Pierrick Potrel, Sophie Derniame, Isabelle Chion-Sotinel, Diane Le Clerre, Cécile Schiffer-Mannioui, Brian Philip, and Laurent Poirot
- Abstract
Supplementary Methods from Multiplex Genome-Edited T-cell Manufacturing Platform for “Off-the-Shelf” Adoptive T-cell Immunotherapies
- Published
- 2023
- Full Text
- View/download PDF
10. Mitigation of chromosome loss in clinical CRISPR-Cas9-engineered T cells
- Author
-
Connor A. Tsuchida, Nadav Brandes, Raymund Bueno, Marena Trinidad, Thomas Mazumder, Bingfei Yu, Byungjin Hwang, Christopher Chang, Jamin Liu, Yang Sun, Caitlin R. Hopkins, Kevin R. Parker, Yanyan Qi, Ansuman T. Satpathy, Edward A. Stadtmauer, Jamie H.D. Cate, Justin Eyquem, Joseph A. Fraietta, Carl H. June, Howard Y. Chang, Chun Jimmie Ye, and Jennifer A. Doudna
- Subjects
Article - Abstract
SummaryCRISPR-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 chromosome, including in pre-clinical 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,1dramatically 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.
- Published
- 2023
- Full Text
- View/download PDF
11. Highly Parallel Discovery of Synthetic Knockin Sequences for Enhanced Cancer Immunotherapies
- Author
-
Franziska Blaeschke, Yan Yi Chen, Ryan Apathy, Zhongmei Li, Cody T. Mowery, William A. Nyberg, Angela To, Ruby Yu, Raymund Bueno, Min Cheol Kim, Ralf Schmidt, Daniel B. Goodman, Tobias Feuchtinger, Justin Eyquem, Chun Jimmie Ye, Eric Shifrut, Theodore L. Roth, and Alexander Marson
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
12. Genome Editing Applications in Cancer T Cell Therapy
- Author
-
William A. Nyberg and Justin Eyquem
- Published
- 2022
- Full Text
- View/download PDF
13. Novel extragenic genomic safe harbors for precise therapeutic T cell engineering
- Author
-
Ashlesha Odak, Han Yuan, Judith Feucht, Vito Adrian Cantu, Jorge Mansilla-Soto, Friederike Kogel, Justin Eyquem, John K Everett, Frederic D. Bushman, Christina Leslie, and Michel Sadelain
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - 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 effective, current genetic engineering strategies that utilize g-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. We report and validate herein an algorithm for the identification of extragenic genomic safe harbors (GSH) that can be efficiently targeted for DNA integration and 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 resist tumor rechallenge 100 days after their infusion. The identification of functional extragenic GSHs thus expands the human genome available for therapeutic precision engineering.
- Published
- 2023
- Full Text
- View/download PDF
14. Pooled screening of CAR T cells identifies diverse immune signaling domains for next-generation immunotherapies
- Author
-
Daniel B. Goodman, Camillia S. Azimi, Kendall Kearns, Alexis Talbot, Kiavash Garakani, Julie Garcia, Nisarg Patel, Byungjin Hwang, David Lee, Emily Park, Vivasvan S. Vykunta, Brian R. Shy, Chun Jimmie Ye, Justin Eyquem, Alexander Marson, Jeffrey A. Bluestone, and Kole T. Roybal
- Subjects
Receptors, Chimeric Antigen ,5.2 Cellular and gene therapies ,T-Lymphocytes ,Adoptive ,Chimeric Antigen ,General Medicine ,Biological Sciences ,Immunotherapy, Adoptive ,Medical and Health Sciences ,Vaccine Related ,Neoplasm Recurrence ,Local ,Receptors ,Humans ,Immunization ,Immunotherapy ,Neoplasm Recurrence, Local ,B-Cell Maturation Antigen ,Development of treatments and therapeutic interventions ,Signal Transduction ,Cancer ,Biotechnology - 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
15. 323 TALEN®-based gene edited iPSC-derived NK (iNK) cells demonstrate enhanced antitumor activity
- Author
-
An-Ping Chen, Peng Gao, Preeti Ashok, Marshall Chao Ma, David Zou, Andrea Chambers, Liang Lin, Hao-Ming Chang, Antonio Arulanandam, Justin Eyquem, Elisabetta Burchi, Armin Rath, Stanley Frankel, Alex Boyne, Alexandre Juillerat, Philippe Duchateau, Daniel Teper, Nejmi Dilmac, and Wei Li
- Published
- 2022
- Full Text
- View/download PDF
16. NUDT21 limits CD19 levels through alternative mRNA polyadenylation in B cell acute lymphoblastic leukemia
- Author
-
Matthew T. Witkowski, Soobeom Lee, Eric Wang, Anna K. Lee, Alexis Talbot, Chao Ma, Nikolaos Tsopoulidis, Justin Brumbaugh, Yaqi Zhao, Kathryn G. Roberts, Simon J. Hogg, Sofia Nomikou, Yohana E. Ghebrechristos, Palaniraja Thandapani, Charles G. Mullighan, Konrad Hochedlinger, Weiqiang Chen, Omar Abdel-Wahab, Justin Eyquem, and Iannis Aifantis
- Subjects
Pediatric Research Initiative ,Lymphoma, B-Cell ,Lymphoma ,Childhood Leukemia ,Pediatric Cancer ,Antigens, CD19 ,Messenger ,Adoptive ,Immunology ,Polyadenylation ,Immunotherapy, Adoptive ,Article ,Vaccine Related ,Rare Diseases ,Stem Cell Research - Nonembryonic - Human ,Receptors ,Genetics ,Immunology and Allergy ,Humans ,2.1 Biological and endogenous factors ,RNA, Messenger ,Antigens ,Aetiology ,Cancer ,Pediatric ,Receptors, Chimeric Antigen ,Membrane Glycoproteins ,CD19 ,Cleavage And Polyadenylation Specificity Factor ,Human Genome ,B-Cell ,Chimeric Antigen ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Stem Cell Research ,Burkitt Lymphoma ,Orphan Drug ,Trans-Activators ,RNA ,Immunization ,Immunotherapy ,Biotechnology - 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.
- Published
- 2022
17. Modular Pooled Discovery of Synthetic Knockin Sequences to Program Durable Cell Therapies
- Author
-
Franziska Blaeschke, Yan Yi Chen, Ryan Apathy, Zhongmei Li, Cody T. Mowery, William A. Nyberg, Angela To, Ruby Yu, Raymund Bueno, Min Cheol Kim, Ralf Schmidt, Daniel B. Goodman, Tobias Feuchtinger, Justin Eyquem, Chun Jimmie Ye, Eric Shifrut, Theodore L. Roth, and Alexander Marson
- Abstract
SUMMARYChronic stimulation can cause T cell dysfunction and limit efficacy of cellular immunotherapies. CRISPR screens have nominated gene targets for engineered T cells, but improved methods are required to compare large numbers of synthetic knockin sequences to reprogram cell functions. Here, we developed Modular Pooled Knockin Screening (ModPoKI), an adaptable platform for modular construction of DNA knockin libraries using barcoded multicistronic adaptors. We built two ModPoKI libraries of 100 transcription factors (TFs) and 129 natural and synthetic surface receptors. Over 20 ModPoKI screens across human TCR and CAR T cells in diverse conditions identified a transcription factor AP4 (TFAP4) construct to enhance long-term T cell fitness and anti-cancer functionin vitroandin vivo. ModPoKI’s modularity allowed us to generate a ∼10,000-member library of TF combinations. Non-viral knockin of a combined BATF-TFAP4 polycistronic construct further enhanced functionin vivo. ModPoKI facilitates discovery of complex gene constructs to program cellular functions.HighlightsModular pooled knockins of hundreds of TF and surface receptor constructs combined with different antigen receptorsChronic stimulation screens discover programs to improve T cell persistenceCombinatorial knockin screens with ∼10,000 transcription factor combinationsBATF-TFAP4 dual knockin construct improves CAR T cell functionin vitroandin vivo
- Published
- 2022
- Full Text
- View/download PDF
18. Abstract 3187: Improved anti-tumor immune functions of iPSC-derived NK cells with TGFβR2 knock-out and/or IL-15 knock-in by TALEN® editing for use alone or in combination with GPC3 Flex-NKTM bispecific antibody
- Author
-
Liang Lin, Peng Gao, An-Ping Chen, Hao-Ming Chang, Preeti Ashok, David Zou, Marshall Chao Ma, Justin Eyquem, Alex Boyne, Alexandre Juillerat, Laurent Poirot, Philippe Duchateau, Armin Rath, Elisabetta Burchi, Daniel Teper, Antonio Arulanandam, and Wei Li
- Subjects
Cancer Research ,Oncology - Abstract
Background: Induced Pluripotent Stem Cell (iPSC)-derived NK cells (iNK) offer an opportunity to generate unlimited homogenous NK cells as allogeneic off-the-shelf therapies. Interleukin-15 (IL-15) signaling enhances proliferation, persistence, cytotoxicity, and metabolic fitness of NK cells. Activation of TGF-β signaling suppresses anti-tumor functions of immune cells, including NK cells, in the tumor microenvironment (TME). In addition, previously, we have reported that our Flex-NKTM bispecific antibody that engages NK cells through NKp46 can enhance the cytotoxicity of the non-edited iNK cells. Therefore, we hypothesized that iNK cells with IL-15 knock-in (KI) and/or TGFβR2 knock-out (KO) could exhibit improved immune function and overcome the immunosuppressive TME. Furthermore, the activity of these edited universal iNK cells could be enhanced when combined with CYT-303, a Flex-NKTM bispecific antibody NK engager targeting GPC3 expressed on many solid tumors including hepatocellular carcinoma (HCC). Methods: IL15 was knocked-in and/or TGFβR2 was knocked-out in iPSC using Cellectis TALEN® and the verified edited iPSC clones were differentiated and expanded into NK cells. The functional significance of these edits in iNKs were assessed in IL-15 and TGF-β dependent NK cell assays evaluating survival and proliferation, expression of activating receptors, as well as cytolysis of these iNK cells against HCC tumor cells. Cytotoxic activity of these edited cells was also tested in a serial killing assay with or without CYT-303 in the absence or presence of TGF-β. Results: Compared to the non-edited iNK cells, iNKs with IL-15 KI can extend persistence in vitro in the absence of exogeneous cytokines. In the presence of TGF-β, the expression level of a number of NK cell activating receptors, such as NKG2D, DNAM-1, and NKp30, was decreased and lower cytotoxicity against HCC tumor cells was observed. However, this TGF-β-mediated immune suppression was reversed in TGFβR2 KO iNKs cells which also presented enhanced cytotoxicity against HCC cells. Furthermore, the anti-HCC cytotoxic activity of either single (IL-15 KI) or double edited (TGFβR2 KO and Il-15 KI) iNK cells were further enhanced by CYT-303, even in the presence of TGF-β. Serial killing assays against HCC tumor cells showed that the iNK cell dysfunction observed in later rounds of killing could still be reversed by the combination of these gene edits and CYT-303. Conclusions: This work demonstrates that KI of IL-15 and KO of TGFβR2 is a promising strategy for TALEN®-engineered iNK cell therapies to overcome the immunosuppressive TME and mount a potent and persistent anti-tumor immune response. The data also provide a solid foundation for combining these edited iNK cells with CYT-303 to address the immunosuppressive TME towards a cure for HCC. Citation Format: Liang Lin, Peng Gao, An-Ping Chen, Hao-Ming Chang, Preeti Ashok, David Zou, Marshall Chao Ma, Justin Eyquem, Alex Boyne, Alexandre Juillerat, Laurent Poirot, Philippe Duchateau, Armin Rath, Elisabetta Burchi, Daniel Teper, Antonio Arulanandam, Wei Li. Improved anti-tumor immune functions of iPSC-derived NK cells with TGFβR2 knock-out and/or IL-15 knock-in by TALEN® editing for use alone or in combination with GPC3 Flex-NKTM bispecific antibody [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3187.
- Published
- 2023
- Full Text
- View/download PDF
19. Integrating Transcriptomics and Proteomics for the Discovery of Novel Antigen Targets on Surface of Malignant Plasma Cells Amenable for Chimeric Antigen Receptor-T (CAR-T) Cell Approach in the Treatment of Patients with Relapsed/Refractory Multiple Myeloma
- Author
-
Alexis Talbot, Solène Weill, Eléonore Fox, Léa Meunier, Guillaume Appé, Camille Marijon, Abdelkader Behdenna, Arun P Wiita, Akpéli Nordor, and Justin Eyquem
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
20. An evolved AAV variant enables efficient genetic engineering of murine T cells
- Author
-
William A. Nyberg, Jonathan Ark, Angela To, Sylvanie Clouden, Gabriella Reeder, Joseph J. Muldoon, Jing-Yi Chung, William H. Xie, Vincent Allain, Zachary Steinhart, Christopher Chang, Alexis Talbot, Sandy Kim, Alan Rosales, L. Patrick Havlik, Harold Pimentel, Aravind Asokan, and Justin Eyquem
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
21. 132 HLA-independent T cell receptors effectively target low abundance antigens
- Author
-
Sascha Haubner, Zeguo Zhao, Ronald C. Hendrickson, Andreina Garcia Angus, Claire Hivroz, Noémie Paillon, Judith Feucht, Mathieu Maurin, Michelle Saetersmoen, Archana Iyer, Michel Sadelain, Andrés Ernesto Zucchetti, Jorge Mansilla-Soto, Mohamad Hamieh, Sjoukje J. C. van der Stegen, Justin Eyquem, Anton Dobrin, Maria L. Sjostrand, Fella Tamzalit, Morgan Huse, Zhuoning Li, Pieter Lindenbergh, Matthew M. Miele, and Theodoros Giavridis
- Subjects
Pharmacology ,Cancer Research ,Immunology ,T-cell receptor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Human leukocyte antigen ,Biology ,Molecular biology ,Oncology ,Antigen ,Abundance (ecology) ,Molecular Medicine ,Immunology and Allergy ,RC254-282 - Abstract
BackgroundChimeric antigen receptors (CARs) engage antigen independently of HLA and enable sustained T cell proliferation when they are endowed with both activating and costimulatory functions. While remission rates have been noticeably elevated in numerous clinical trials targeting CD19, CD22 or BCMA, relapses are common. One of the several underlying relapse mechanisms is antigen escape, which refers to a relapsing tumor that is either negative for the targeted antigen or expresses the latter at a low level. Failure to eliminate antigen-low tumors raises questions about the sensitivity of CARs and the minimum antigen density that is required for effective tumor eradication. Unlike CARs, TCRs engage antigen in an HLA-dependent manner, and they do so with high sensitivity. We hypothesized that a TCR/CD3 complex containing the same heavy and light immunoglobulin chains as a CAR will display increased sensitivity to the target antigen.MethodsWe edited the TRAC locus in human primary T cells to establish a novel antigen receptor structure, termed HLA-independent TCR or HIT receptor, by incorporating into the TCR/CD3 complex the same heavy and light chains as those of a corresponding CAR. We assessed their antigen sensitivity against a panel of cell lines expressing different antigen levels, analyzing their cytotoxicity, cytokine secretion, signaling response and degranulation activity. HIT and CAR T cells were further evaluated for their anti-tumor response using established ALL and AML mouse models.ResultsCD19-TRAC-HIT and CD19-TRAC-CAR T cells lysed wild-type NALM6 (~27,000 CD19 molecules) and NALM6 variants with 100-fold less CD19. As CD19 levels decreased further, CAR T cells no longer killed their target, in contrast to HIT T cells. HIT T cells showed increased expression of IFN-gamma, IL-2 and TNF-alpha upon exposure to NALM6 cells expressing ~20 CD19 molecules per cell, compared to CAR T cells. This increased sensitivity of HIT receptors correlated to their greater signaling response, upon exposure to the low-antigen-density NALM6. Phospho-proteomic analyses further confirmed this increased response of HIT T cells to low antigen levels. Altogether, these results confirm that HIT receptors endow T cells with greater antigen sensitivity than canonical CARs. We further showed that HIT T cells have higher in vivo anti-tumor activity compared to CAR T cells in mice bearing low-antigen-density ALL or AML.ConclusionsHIT receptors consistently afford high antigen sensitivity and mediate tumor recognition beyond what current CARs can provide. HIT receptors open new prospects for targeting cell surface antigens of low abundance.Ethics ApprovalEight- to 12-week-old NOD/SCID/IL-2Rgamma-null (NSG) male mice (Jackson Laboratory) were used under a protocol approved by the MSKCC Institutional Animal Care and Use Committee.
- Published
- 2021
22. Hybrid ssDNA repair templates enable high yield genome engineering in primary cells for disease modeling and cell therapy manufacturing
- Author
-
Hong Li, Alvin Ha, Lumeng Ye, Franziska Blaeschke, Jonathan H. Esensten, Jeffrey L. Wolf, Alexis Talbot, Vivasvan Vykunta, Brian R. Shy, Alexander Marson, Yan Yi Chen, Justin Eyquem, Murad R. Mamedov, Thomas G. Martin, David N. Nguyen, Jing-Yi Chung, Shane Vedova, and Theodore L. Roth
- Subjects
Homology directed repair ,Cell therapy ,chemistry.chemical_compound ,chemistry ,Cas9 ,Transgene ,CRISPR ,Computational biology ,Biology ,Genome ,DNA ,Genome engineering - Abstract
CRISPR-Cas9 offers unprecedented opportunities to modify genome sequences in primary human cells to study disease variants and reprogram cell functions for next-generation cellular therapies. CRISPR has several potential advantages over widely used retroviral vectors including: 1) site-specific transgene insertion via homology directed repair (HDR), and 2) reductions in the cost and complexity of genome modification. Despite rapid progress with ex vivo CRISPR genome engineering, many novel research and clinical applications would be enabled by methods to further improve knock-in efficiency and the absolute yield of live knock-in cells, especially with large HDR templates (HDRT). We recently reported that Cas9 target sequences (CTS) could be introduced into double-stranded DNA (dsDNA) HDRTs to improve knock-in, but yields and efficiencies were limited by toxicity at high HDRT concentrations. Here we developed a novel system that takes advantage of lower toxicity with single-stranded DNA (ssDNA). We designed hybrid ssDNA HDRTs that incorporate CTS sites and were able to boost knock-in percentages by >5-fold and live cell yields by >7-fold relative to dsDNA HDRTs with CTS. Knock-in efficiency and yield with ssCTS HDRTs were increased further with small molecule inhibitor combinations to improve HDR. We demonstrate application of these methods across a variety of target loci, knock-in constructs, and primary human cell types to reach ultra-high HDR efficiencies (>80-90%) which we use for pathogenic gene variant modeling and universal gene replacement strategies for IL2RA and CTLA4 mutations associated with mendelian immune disorders. Finally, we develop a GMP-compatible method for fully non-viral CAR-T cell manufacturing, demonstrating knock-in efficiencies of 46-62% and generating yields of >1.5 x 109 CAR+ T cells, well above current doses for adoptive cellular therapies. Taken together, we present a comprehensive non-viral approach to model disease associated mutations and re-write targeted genome sequences to program immune cell therapies at a scale compatible with future clinical application.
- Published
- 2021
- Full Text
- View/download PDF
23. RASA2 ablation in T cells boosts antigen sensitivity and long-term function
- Author
-
Julia Carnevale, Eric Shifrut, Nupura Kale, William A. Nyberg, Franziska Blaeschke, Yan Yi Chen, Zhongmei Li, Sagar P. Bapat, Morgan E. Diolaiti, Patrick O’Leary, Shane Vedova, Julia Belk, Bence Daniel, Theodore L. Roth, Stefanie Bachl, Alejandro Allo Anido, Brooke Prinzing, Jorge Ibañez-Vega, Shannon Lange, Dalia Haydar, Marie Luetke-Eversloh, Maelys Born-Bony, Bindu Hegde, Scott Kogan, Tobias Feuchtinger, Hideho Okada, Ansuman T. Satpathy, Kevin Shannon, Stephen Gottschalk, Justin Eyquem, Giedre Krenciute, Alan Ashworth, and Alexander Marson
- Subjects
Time Factors ,General Science & Technology ,T-Lymphocytes ,Adoptive ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Mice ,Antigens, Neoplasm ,Bone Marrow ,Neoplasms ,Receptors ,Genetics ,2.1 Biological and endogenous factors ,Animals ,Humans ,Antigens ,Aetiology ,Cancer ,Multidisciplinary ,Leukemia ,Receptors, Chimeric Antigen ,5.2 Cellular and gene therapies ,Animal ,Prevention ,Chimeric Antigen ,T-Cell ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,5.1 Pharmaceuticals ,ras GTPase-Activating Proteins ,Antigen ,Gene Knockdown Techniques ,Disease Models ,Neoplasm ,Immunotherapy ,Development of treatments and therapeutic interventions ,CRISPR-Cas Systems ,Biotechnology - Abstract
The efficacy of adoptive T cell therapies for cancer treatment can be limited by suppressive signals from both extrinsic factors and intrinsic inhibitory checkpoints1,2. 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.
- Published
- 2021
24. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency
- Author
-
Michel Sadelain, Jie Sun, Judith Feucht, Annalisa Cabriolu, Josef Leibold, Anton Dobrin, Zeguo Zhao, Yu-Jui Ho, Justin Eyquem, and Mohamad Hamieh
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Antigen ,medicine ,Animals ,Cell Lineage ,Receptor ,Effector ,CD28 ,General Medicine ,Immunotherapy ,Chimeric antigen receptor ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Calibration - Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that target and reprogram T cells to acquire augmented antitumor properties1. 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. A novel chimeric antigen receptor (CAR) T cell design improves therapeutic efficacy by balancing effector and memory profiles.
- Published
- 2018
- Full Text
- View/download PDF
25. The CD28-transmembrane domain mediates chimeric antigen receptor heterodimerization with CD28
- Author
-
Roxxana Valeria Beltran Valencia, Alexander Marson, Frédéric Van Gool, Patrick Ho, Justin Eyquem, Theodore L. Roth, Jeffrey A. Bluestone, James A. Wells, Caroline Raffin, Qizhi Tang, Duy P. Nguyen, Leonardo M. R. Ferreira, Zion Congrave-Wilson, and Yannick D. Muller
- Subjects
CD86 ,Transmembrane domain ,Chemistry ,CD28 ,chemical and pharmacologic phenomena ,hemic and immune systems ,human activities ,Chimeric antigen receptor ,CD8 ,Intracellular ,CD80 ,Transmembrane protein ,Cell biology - Abstract
Anti-CD19 chimeric antigen receptor (CD19-CAR)-engineered T cells are approved therapeutics for malignancies. The impact of the hinge (HD) and transmembrane (TMD) domains between the extracellular antigen-targeting and the intracellular signaling modalities of CARs has not been systemically studied. Here, a series of CD19-CARs differing only by their HD (CD8/CD28/IgG4) and TMD (CD8/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 to anti-CD28 stimulation. This dimerization depended on polar amino-acids in the CD28-TMD. CD28-CAR heterodimerization was more efficient in CARs containing a CD8-HD or CD28-HD as compared to an IgG4-HD. CD28-CAR heterodimers did not respond to CD80 and CD86 stimulation but led to a significant reduction of CD28 cell-surface expression. These data unveil a new property of the CD28-TMD and suggest that TMDs can modulate CAR T-cell activities by engaging endogenous partners.Abstract Figure
- Published
- 2020
- Full Text
- View/download PDF
26. HLA-independent T cell receptors for targeting tumors with low antigen density
- Author
-
Jorge Mansilla-Soto, Justin Eyquem, Sascha Haubner, Mohamad Hamieh, Judith Feucht, Noémie Paillon, Andrés Ernesto Zucchetti, Zhuoning Li, Maria Sjöstrand, Pieter L. Lindenbergh, Michelle Saetersmoen, Anton Dobrin, Mathieu Maurin, Archana Iyer, Andreina Garcia Angus, Matthew M. Miele, Zeguo Zhao, Theodoros Giavridis, Sjoukje J. C. van der Stegen, Fella Tamzalit, Isabelle Rivière, Morgan Huse, Ronald C. Hendrickson, Claire Hivroz, and Michel Sadelain
- Subjects
Leukemia, Myeloid, Acute ,Mice ,Receptors, Chimeric Antigen ,Histocompatibility Antigens ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Animals ,Humans ,General Medicine ,Immunotherapy, Adoptive ,Xenograft Model Antitumor Assays ,General Biochemistry, Genetics and Molecular Biology ,Article - 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.
- Published
- 2020
27. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection
- Author
-
Michel Sadelain, Sjoukje J. C. van der Stegen, Mithat Gonen, Mohamad Hamieh, Theodoros Giavridis, Ashlesha Odak, Jorge Mansilla-Soto, Justin Eyquem, and Kristen M. Cunanan
- Subjects
Male ,0301 basic medicine ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Lymphocyte Activation ,CD19 ,Translational Research, Biomedical ,Mice ,03 medical and health sciences ,Cancer immunotherapy ,Genome editing ,medicine ,Animals ,Humans ,CRISPR ,Gene silencing ,Promoter Regions, Genetic ,Gene Editing ,Multidisciplinary ,biology ,Cas9 ,Cell Differentiation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Chimeric antigen receptor ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Genetic Loci ,Immunology ,biology.protein ,Cancer research ,Immunotherapy ,CRISPR-Cas Systems ,human activities - 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
28. Establishing cGMP manufacturing of CRISPR/Cas9-edited human CAR T cells
- Author
-
Xiuyan Wang, M. Wu, J. Mansilla-Soto, S. Zabierowski, Michael Sadelain, Justin Eyquem, C. Del Casale, and Isabelle Riviere
- Subjects
Cancer Research ,Transplantation ,biology ,CD3 ,T cell ,Transgene ,Immunology ,T-cell receptor ,Cell Biology ,CD19 ,Chimeric antigen receptor ,Cell biology ,Transduction (genetics) ,medicine.anatomical_structure ,Oncology ,biology.protein ,medicine ,Immunology and Allergy ,human activities ,Genetics (clinical) ,B cell - Abstract
Background & Aim Chimeric antigen receptors (CARs) are synthetic receptors that redirect and reprogram T cells to mediate tumor rejection. CD19 targeted CAR T cells have showed remarkable effacy for chemorefractory/relapsed B cell malignancies. Recombinant retroviral vectors mediated- CAR gene transfer is currently the standard method to generate cGMP grade CAR T cells. However, CAR gene expression is highly variable, owing to position effects and vector incorporation variations. CAR T cells engineered in this manner are capable of tumor eradication, but are prone to tonic signaling and accelerated exhaustion. We have developed a novel genetic engineering strategy to insert CAR genes into a precise genomic location in human peripheral blood T cells. Methods, Results & Conclusion T cells are electroporated with Cas9 mRNA or Cas9 protein and a guide RNA, followed by transduction with a recombinant Adeno-Associated Virus encoding the CAR sequence to facilitate the insertion of the CAR gene upstream of the constant region of TCR alpha chain. This results in the endogenous TCR promoter (TRAC) controlled CAR expression and abrogation of TCR surface expression. This strategy not only allows uniform CAR expression, but also delays T-cell differentiation and exhaustion, leading to enhanced T cell function and anti-tumor efficacy. The edited cells vastly outperformes retrovirally modified CAR T cells in a pre-B ALL NALM6 mouse model. The targeting of CARs to the TCR locus also provides a safer therapeutic T cell by minimizing the risks of insertional oncogenesis, and TCR-induced autoimmunity and alloreactivity (thus spanning both autologous and allogeneic T cell applications). In addition, we have incorporated in the CAR a mutant CD3z chain encoding a single immunoreceptor tyrosine-based activation motif that improves the balance between effector and memory T cells composition. We are in the process of translating this novel approach into the clinical setting by establishing cGMP conditions and protocols for the manufacturing of TRAC-CAR T cells. Using the 4D-LV electroporator, we have demonstrated that we can knockout the TCR at large scale (100 E06 CD3+ T cells) with high efficiency (70-80%) as that obtained at small scale. We have evaluated AAV6 to deliver the CAR transgene in the TCR locus. Data will be presented on optimizing the manufacturing of the TRAC-CAR T cells and on evaluating their anti-tumor efficacy in vivo.
- Published
- 2020
- Full Text
- View/download PDF
29. Targeted Integration of a CAR at a Novel Genomic Safe Harbor Directs Potent Therapeutic Outcomes
- Author
-
Ashlesha Odak, Han Yuan, Judith Feucht, Jorge Mansilla Soto, Michel Sadelain, Christina S. Leslie, and Justin Eyquem
- Subjects
Safe harbor ,business.industry ,Immunology ,Medicine ,Cell Biology ,Hematology ,business ,Bioinformatics ,Biochemistry - Abstract
Chimeric receptor antigen (CAR)-T cell therapy using CARs specific for CD19 have been remarkably successful for treating chemo-refractory/relapsed B cell malignancies. These successes not withstanding, therapeutic outcomes between patients are variable and occasional cases of clonal expansion of the transduced T cells have been observed, albeit without leukemic transformation. These issues are cause for concern and need to be addressed to achieve better and safer therapeutic outcomes. Clonal expansion due to insertional mutagenesis and variegated transgene expression due to position effects are well established to be the consequence of the semi-random integration pattern afforded by gamma-retroviral and lentiviral vectors. We previously established that integration of a CAR cDNA in the TCR alpha locus (TRAC) provides consistent, regulated expression of CD19 CARs and superior CAR T cell efficacy in a mouse model of B-cell acute lymphoblastic leukemia (B-ALL). Here, we identify a novel extragenic site devoid of any known function and remote from endogenous genes, i.e. a 'genomic safe harbor' (GSH), that can be efficiently targeted in human T cells and drives potent CAR T cell therapy in the B-ALL mouse model. To identify GSHs that could be efficiently targeted in T cells by CRISPR-Cas9 and that could also support durable transgene expression, we screened for genomic regions meeting both, GSH criteria and high chromatin accessibility in T cells as measured by ATAC-seq. In human primary T cells, we identified 379 such sites. Ten of the highest accessible sites were investigated. All showed high (>90%) cleavage efficiency and allowed for CAR cDNA targeted integration and expression which also translated into effective cytolytic activity of the CARs within a few days after transduction. However, thereafter CAR expression diminished over the course of a week at most but not all of these sites. In order to prevent possible heterochromatinization, we incorporated chromatin insulator elements with barrier activity flanking the CAR transcription unit. Incorporation of the chromatin insulator element dramatically improved CAR expression and functionality at one site, whereas 3 other GSHs tested were not affected. One of the 10 GSHs maintained long-term CAR expression without requiring an insulator and directed potent anti-leukemic CAR T cell efficacy in a B-ALL 'CAR stress test' mouse model, matching the T cell potency afforded by integrating the CAR cDNA at the TRAC locus. This finding highlights the major effect of the integration site on transgene expression and ensuing therapeutic efficacy. We identified an extragenic GSH site that can be used for effective T cell engineering and sustained expression of a CAR. Through this study, we provide a platform for identifying GSHs that could be reliably targeted for safe and predictable expression of CARs or other immunomodulatory transgenes to potentiate adoptive immunotherapy. Disclosures Sadelain: Fate Therapeutics: Patents & Royalties, Research Funding; Mnemo: Patents & Royalties; Atara: Patents & Royalties, Research Funding; Takeda: Patents & Royalties, Research Funding; Minerva: Other: Biotechnologies, Patents & Royalties.
- Published
- 2020
- Full Text
- View/download PDF
30. MM-378: Inhibitory and Stimulatory Interplay Between Myeloma Tumor and T Cells
- Author
-
Kole T. Roybal, Justin Eyquem, Thomas Martin, August Reich Dietrich, Cassandra E. Burnett, and Arun P. Wiita
- Subjects
CD86 ,Cancer Research ,Tumor microenvironment ,biology ,business.industry ,SLAMF7 ,Hematology ,CD48 ,CD19 ,Oncology ,Antigen ,biology.protein ,Cancer research ,Medicine ,B-cell activating factor ,business ,CD80 - Abstract
Context: T cells engineered to express chimeric antigen receptors (CAR-Ts) offer hope for improved outcomes in multiple myeloma. Patients relapse despite 80% or better response rates from CAR-Ts in clinical trials. Improvements in CAR design may come from a better understanding of the interplay between T cells and the tumor microenvironment. Objective: To describe this interplay, we deeply characterized the surfaceome of the ANBL-6 cell line shown by us to best represent the transcriptome of patients newly diagnosed with multiple myeloma. Design: ANBL-6 cells were cultured in RPMI Complete with 2 ng/mL IL-6 and were stained with antibodies directed toward 28 unique cell-surface antigens known to mark myeloma cells, as well as those known to have effects on T cells that are stimulatory, inhibitory, or both. Flow cytometry was employed to assess expression of cell-surface antigens on ANBL-6 cells incubated for 24 hours in and out of the presence of IFNγ, which is expressed by activated T cells, at 100 U/mL. Results: Canonical myeloma markers CD38 and CD138 were expressed. BCMA, BAFF, and the receptor to BAFF (BAFF-R) were not. CD19, CD44, CD56, CD72, CD307, CS1/SLAMF7, GPRC5D, and TACI were expressed. CD19, CD72, CD56, and CS1/SLAMF7 increased with IFNγ. TACI decreased. Stimulatory 4-1BB-L, CD48, CD86, ICOS-L, and OX40-L were present. 4-1BB-L increased with IFNγ. The inhibitory cell-surface antigens CD112, CEACAM, FasL, PD-L1, and PD-L2 were present. CEACAM, FasL, and PD-L1 all increased with IFNγ. Stimulatory and inhibitory CD155 and CD270 were both present and did not change with IFNγ. Inhibitory CD200 and stimulatory CD70 and CD80 were not expressed. Conclusions: ANBL-6 expressed five known T-cell stimulatory antigens and five known inhibitory antigens. One of five stimulatory antigens increased with IFNγ. Three of five inhibitory proteins increased with IFNγ. Thus, stimulated CAR-Ts may cause their own inhibition. These in vitro data in a single cell line offer insight into myeloma tumor dynamics but may not fully reflect in vivo tumor behavior. Characterization of multiple cell lines and primary tumor samples in vitro and in vivo may improve CAR design ranging from having better suited costimulatory domains to switching receptors that transform inhibitory ligands into costimulatory signals.
- Published
- 2020
- Full Text
- View/download PDF
31. Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape
- Author
-
Justin Eyquem, M. Lia Palomba, Michel Sadelain, Vinagolu K. Rajasekhar, Mohamad Hamieh, Carl J. DeSelm, and Joachim Yahalom
- Subjects
0301 basic medicine ,CA-19-9 Antigen ,Antigens, CD19 ,Oligosaccharides ,Radiation Dosage ,Immunotherapy, Adoptive ,CD19 ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,Pancreatic cancer ,Insulin-Secreting Cells ,Drug Discovery ,Genetics ,Medicine ,Animals ,Humans ,Molecular Biology ,B cell ,Pharmacology ,Radiation ,Receptors, Chimeric Antigen ,biology ,business.industry ,Sequence Analysis, RNA ,Sialyl-Lewis A ,medicine.disease ,Combined Modality Therapy ,Chimeric antigen receptor ,Pancreatic Neoplasms ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,Adenocarcinoma ,Original Article ,Car t cells ,business - 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.
- Published
- 2018
32. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade
- Author
-
Sjoukje J. C. van der Stegen, Alessandra Piersigilli, Theodoros Giavridis, Justin Eyquem, Mohamad Hamieh, and Michel Sadelain
- Subjects
0301 basic medicine ,T cell ,Immunotherapy, Adoptive ,General Biochemistry, Genetics and Molecular Biology ,CD19 ,Article ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Neoplasms ,medicine ,Animals ,Humans ,Myeloid Cells ,B cell ,biology ,business.industry ,Macrophages ,General Medicine ,Syndrome ,medicine.disease ,Pathophysiology ,Chimeric antigen receptor ,Blockade ,Cytokine release syndrome ,Interleukin 1 Receptor Antagonist Protein ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Immunology ,biology.protein ,Cytokines ,business ,Interleukin-1 - 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. Blocking IL-1 and iNOS prevents CAR T cell–induced cytokine release syndrome.
- Published
- 2018
33. Antibody with Infinite Affinity for In Vivo Tracking of Genetically Engineered Lymphocytes
- Author
-
Justin Eyquem, Steven M. Larson, Simone Krebs, Thomas Reiner, Meghan Bell, Christian Brand, Stephen Gottschalk, Afruja Ahad, Vladimir Ponomarev, Michel Sadelain, Wolfgang A. Weber, Lukas M. Carter, and Claude F. Meares
- Subjects
0301 basic medicine ,Male ,Biodistribution ,Single Photon Emission Computed Tomography Computed Tomography ,CD3 ,T cell ,T-Lymphocytes ,Mice, SCID ,Lutetium ,Radiation Dosage ,Immunotherapy, Adoptive ,03 medical and health sciences ,Heterocyclic Compounds, 1-Ring ,Mice ,In vivo ,Genes, Reporter ,Mice, Inbred NOD ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Tissue Distribution ,Yttrium Radioisotopes ,Lymphocytes ,Immunoglobulin Fragments ,Mice, Knockout ,Radioisotopes ,Reporter gene ,Mice, Inbred ICR ,Receptors, Chimeric Antigen ,biology ,Chemistry ,Suicide gene ,Molecular biology ,Adoptive Transfer ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Molecular Imaging ,030104 developmental biology ,medicine.anatomical_structure ,Cell Tracking ,Positron-Emission Tomography ,biology.protein ,Antibody ,Radiopharmaceuticals - 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 of (86)Y-AABD in vitro when compared with nontransduced cells. Imaging studies showed (86)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 with (177)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.
- Published
- 2018
34. Integrating Proteomics and Transcriptomics for Systematic Combinatorial Chimeric Antigen Receptor Therapy of AML
- Author
-
Fabiana Perna, Cameron Brennan, Mohamad Hamieh, Rajesh K. Soni, Ronald C. Hendrickson, Samuel H. Berman, Michel Sadelain, Justin Eyquem, and Jorge Mansilla-Soto
- Subjects
0301 basic medicine ,Proteomics ,Cancer Research ,medicine.medical_treatment ,Recombinant Fusion Proteins ,T-Lymphocytes ,Antigens, CD19 ,CD34 ,Receptors, Antigen, T-Cell ,Biology ,Bioinformatics ,Article ,03 medical and health sciences ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Humans ,Gene Expression Profiling ,Myeloid leukemia ,Cell Biology ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Haematopoiesis ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,Cancer research ,Stem cell - Abstract
Summary 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.
- Published
- 2017
35. A platform for rapid prototyping of synthetic gene networks in mammalian cells
- Author
-
Xavier Duportet, Patrick Guye, Yingqing Li, Tharathorn Rimchala, Justin Eyquem, Liliana Wroblewska, Gregory Batt, Julianne Rieders, Ron Weiss, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology. Synthetic Biology Center, Wroblewska, Liliana, Guye, Patrick, Li, Yinqing, Weiss, Ron, Rimchala, Tharathorn, Computational systems biology and optimization (Lifeware), Inria Paris-Rocquencourt, Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria), Department of Biological Engineering [Cambridge] (BE), Massachusetts Institute of Technology (MIT), Matière et Systèmes Complexes (MSC (UMR_7057)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Cellectis, Cellectis SA, Department of Electrical Engineering and Computer Science, MIT, DARPA CCM [HR0011–12-C-0067], DARPA Synbio BBN [DARPA-BAA-11–23], NIGMS of the NIH [P50GM098792], ANR-10-BINF-06-11,Iceberg,Des modèles de population aux populations de modèles: observation, modélisation et contrôle de l’expression génique au niveau de la cellule unique(2011), ANR-10-COSI-0007,Syne2Arti,Des réseaux de régulation génique aux tissus artificiels(2010), Centre National de la Recherche Scientifique (CNRS)-Université Paris Diderot - Paris 7 (UPD7), ANR-10-BINF-0006,Iceberg,Des modèles de population aux populations de modèles: observation, modélisation et contrôle de l'expression génique au niveau de la cellule unique(2010), and Matière et Systèmes Complexes (MSC)
- Subjects
Rapid prototyping ,Gene regulatory network ,Computational biology ,Biology ,Cell Line ,03 medical and health sciences ,Synthetic biology ,0302 clinical medicine ,Genetics ,Animals ,Humans ,Gene Regulatory Networks ,Cloning, Molecular ,Cell Engineering ,Gene ,Gene Library ,030304 developmental biology ,0303 health sciences ,Drug discovery ,business.industry ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Modular design ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Proof of concept ,DECIPHER ,ComputingMethodologies_GENERAL ,Synthetic Biology and Bioengineering ,business ,030217 neurology & neurosurgery - 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., National Institute of General Medical Sciences (U.S.) (P50GM098792), France. Agence nationale de la recherche (Syne2arti. ANR-10-COSINUS-007), France. Agence nationale de la recherche (Iceberg. ANR-IABI-3096), United States. Defense Advanced Research Projects Agency (Controlling Cellular Machinery. R0011–12-C-0067), United States. Defense Advanced Research Projects Agency (Synbio BBN. DARPA-BAA-11–23)
- Published
- 2014
- Full Text
- View/download PDF
36. Publisher Correction: Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency
- Author
-
Michel Sadelain, Jie Sun, Judith Feucht, Yu-Jui Ho, Mohamad Hamieh, Anton Dobrin, Justin Eyquem, Annalisa Cabriolu, Zeguo Zhao, and Josef Leibold
- Subjects
0301 basic medicine ,Calibration (statistics) ,T cell ,General Medicine ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,Potency ,Car t cells ,Mathematics - 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
37. Characterization of three loci for homologous gene targeting and transgene expression
- Author
-
Roman Galetto, Andrew M. Scharenberg, Laurent Poirot, Julianne Smith, and Justin Eyquem
- Subjects
Genetics ,Reporter gene ,Transgene ,Gene Expression ,Gene targeting ,Bioengineering ,Locus (genetics) ,Biology ,Applied Microbiology and Biotechnology ,Gene dosage ,Genomic Instability ,Cell Line ,Mutagenesis, Insertional ,Genes, Reporter ,Gene Targeting ,Gene expression ,Humans ,Transgenes ,Expression cassette ,Promoter Regions, Genetic ,Gene ,Biotechnology - 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.
- Published
- 2013
- Full Text
- View/download PDF
38. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part one
- Author
-
Andreas Lundqvist, Vincent van Hoef, Xiaonan Zhang, Erik Wennerberg, Julie Lorent, Kristina Witt, Laia Masvidal Sanz, Shuo Liang, Shannon Murray, Ola Larsson, Rolf Kiessling, Yumeng Mao, John-William Sidhom, Catherine A. Bessell, Jonathan Havel, Jonathan Schneck, Timothy A. Chan, Eliot Sachsenmeier, David Woods, Anders Berglund, Rupal Ramakrishnan, Andressa Sodre, Jeffrey Weber, Roberta Zappasodi, Yanyun Li, Jingjing Qi, Philip Wong, Cynthia Sirard, Michael Postow, Walter Newman, Henry Koon, Vamsidhar Velcheti, Margaret K. Callahan, Jedd D. Wolchok, Taha Merghoub, Lawrence G. Lum, Minsig Choi, Archana Thakur, Abhinav Deol, Gregory Dyson, Anthony Shields, Cara Haymaker, Marc Uemura, Ravi Murthy, Marihella James, Daqing Wang, Julie Brevard, Catherine Monaghan, Suzanne Swann, James Geib, Mark Cornfeld, Srinivas Chunduru, Sudhir Agrawal, Cassian Yee, Jennifer Wargo, Sapna P. Patel, Rodabe Amaria, Hussein Tawbi, Isabella Glitza, Scott Woodman, Wen-Jen Hwu, Michael A. Davies, Patrick Hwu, Willem W. Overwijk, Chantale Bernatchez, Adi Diab, Erminia Massarelli, Neil H. Segal, Vincent Ribrag, Ignacio Melero, Tara C. Gangadhar, Walter Urba, Dirk Schadendorf, Robert L. Ferris, Roch Houot, Franck Morschhauser, Theodore Logan, Jason J. Luke, William Sharfman, Fabrice Barlesi, Patrick A. Ott, Laura Mansi, Shivaani Kummar, Gilles Salles, Cecilia Carpio, Roland Meier, Suba Krishnan, Dan McDonald, Matthew Maurer, Xuemin Gu, Jaclyn Neely, Satyendra Suryawanshi, Ronald Levy, Nikhil Khushalani, Jennifer Wu, Jinyu Zhang, Fahmin Basher, Mark Rubinstein, Mark Bucsek, Guanxi Qiao, Cameron MacDonald, Bonnie Hylander, Elizabeth Repasky, Shilpak Chatterjee, Anusara Daenthanasanmak, Paramita Chakraborty, Kyle Toth, Megan Meek, Elizabeth Garrett-Mayer, Michael Nishimura, Chrystal Paulos, Craig Beeson, Xuezhong Yu, Shikhar Mehrotra, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nicole Scharping, Ashley V. Menk, Rebecca Moreci, Ryan Whetstone, Rebekah Dadey, Simon Watkins, Robert Ferris, Greg M. Delgoffe, Jonathan Peled, Sean Devlin, Anna Staffas, Melissa Lumish, Kori Porosnicu Rodriguez, Katya Ahr, Miguel Perales, Sergio Giralt, Ying Taur, Eric Pamer, Marcel R. M. van den Brink, Robert Jenq, Nicola Annels, Hardev Pandha, Guy Simpson, Hugh Mostafid, Kevin Harrington, Alan Melcher, Mark Grose, Bronwyn Davies, Gough Au, Roberta Karpathy, Darren Shafren, Jacob Ricca, Dmitriy Zamarin, Luciana Batista, Florence Marliot, Angela Vasaturo, Sabrina Carpentier, Cécile Poggionovo, Véronique Frayssinet, Jacques Fieschi, Marc Van den Eynde, Franck Pagès, Jérôme Galon, Fabienne Hermitte, Sean G. Smith, Khue Nguyen, Sruthi Ravindranathan, Bhanu Koppolu, David Zaharoff, Gustavo Schvartsman, Roland Bassett, Jennifer L. McQuade, Lauren E. Haydu, Douglas Kline, Xiufen Chen, Dominick Fosco, Justin Kline, Abigail Overacre, Maria Chikina, Erin Brunazzi, Gulidanna Shayan, William Horne, Jay Kolls, Tullia C. Bruno, Creg Workman, Dario Vignali, Prasad S. Adusumilli, Ephraim A Ansa-Addo, Zihai Li, Andrew Gerry, Joseph P. Sanderson, Karen Howe, Roslin Docta, Qian Gao, Eleanor A. L. Bagg, Nicholas Tribble, Miguel Maroto, Gareth Betts, Natalie Bath, Luca Melchiori, Daniel E. Lowther, Indu Ramachandran, Gabor Kari, Samik Basu, Gwendolyn Binder-Scholl, Karen Chagin, Lini Pandite, Tom Holdich, Rafael Amado, Hua Zhang, John Glod, Donna Bernstein, Bent Jakobsen, Crystal Mackall, Ryan Wong, Jonathan D. Silk, Katherine Adams, Garth Hamilton, Alan D. Bennett, Sara Brett, Junping Jing, Adriano Quattrini, Manoj Saini, Guy Wiedermann, Joanna Brewer, MyLinh Duong, An Lu, Peter Chang, Aruna Mahendravada, Nicholas Shinners, Kevin Slawin, David M. Spencer, Aaron E. Foster, J. Henri Bayle, Cristina Bergamaschi, Sinnie Sin Man Ng, Bethany Nagy, Shawn Jensen, Xintao Hu, Candido Alicea, Bernard Fox, Barbara Felber, George Pavlakis, Jessica Chacon, Tori Yamamoto, Thomas Garrabrant, Luis Cortina, Daniel J. Powell, Marco Donia, Julie Westerlin Kjeldsen, Rikke Andersen, Marie Christine Wulff Westergaard, Valentina Bianchi, Mateusz Legut, Meriem Attaf, Garry Dolton, Barbara Szomolay, Sascha Ott, Rikke Lyngaa, Sine Reker Hadrup, Andrew Kelvin Sewell, Inge Marie Svane, Aaron Fan, Takumi Kumai, Esteban Celis, Ian Frank, Amanda Stramer, Michelle A. Blaskovich, Seth Wardell, Maria Fardis, James Bender, Michael T. Lotze, Stephanie L. Goff, Nikolaos Zacharakis, Yasmine Assadipour, Todd D. Prickett, Jared J. Gartner, Robert Somerville, Mary Black, Hui Xu, Harshini Chinnasamy, Isaac Kriley, Lily Lu, John Wunderlich, Paul F. Robbins, Steven Rosenberg, Steven A. Feldman, Kasia Trebska-McGowan, Parisa Malekzadeh, Eden Payabyab, Richard Sherry, Aishwarya Gokuldass, Charlene Kopits, Brian Rabinovich, Daniel S. Green, Olena Kamenyeva, Kathryn C. Zoon, Christina M. Annunziata, Joanne Hammill, Christopher Helsen, Craig Aarts, Jonathan Bramson, Yui Harada, Yoshikazu Yonemitsu, Kenneth Mwawasi, Galina Denisova, Rajanish Giri, Benjamin Jin, Tracy Campbell, Lindsey M. Draper, Sanja Stevanovic, Zhiya Yu, Bianca Weissbrich, Nicholas P. Restifo, Cornelia L. Trimble, Christian S. Hinrichs, Kwong Tsang, Massimo Fantini, James W. Hodge, Rika Fujii, Ingrid Fernando, Caroline Jochems, Christopher Heery, James Gulley, Patrick Soon-Shiong, Jeffrey Schlom, Weiqing Jing, Jill Gershan, Grace Blitzer, James Weber, Laura McOlash, Bryon D. Johnson, Simin Kiany, Huang Gangxiong, Eugenie S. Kleinerman, Michael Klichinsky, Marco Ruella, Olga Shestova, Saad Kenderian, Miriam Kim, John Scholler, Carl H. June, Saar Gill, Duane Moogk, Shi Zhong, Ivan Liadi, William Rittase, Victoria Fang, Janna Dougherty, Arianne Perez-Garcia, Iman Osman, Cheng Zhu, Navin Varadarajan, Alan Frey, Michelle Krogsgaard, Daniel Landi, Kristen Fousek, Malini Mukherjee, Ankita Shree, Sujith Joseph, Kevin Bielamowicz, Tiara Byrd, Nabil Ahmed, Meenakshi Hegde, Sylvia Lee, David Byrd, John Thompson, Shailender Bhatia, Scott Tykodi, Judy Delismon, Liz Chu, Siddiq Abdul-Alim, Arpy Ohanian, Anna Marie DeVito, Stanley Riddell, Kim Margolin, Isabelle Magalhaes, Jonas Mattsson, Michael Uhlin, Satoshi Nemoto, Patricio Pérez Villarroel, Ryosuke Nakagawa, James J. Mule, Adam W. Mailloux, Melinda Mata, Phuong Nguyen, Claudia Gerken, Christopher DeRenzo, Stephen Gottschalk, Mélissa Mathieu, Sandy Pelletier, John Stagg, Simon Turcotte, Nicholas Minutolo, Prannda Sharma, Andrew Tsourkas, Nadine Mockel-Tenbrinck, Daniela Mauer, Katharina Drechsel, Carola Barth, Katharina Freese, Ulrike Kolrep, Silke Schult, Mario Assenmacher, Andrew Kaiser, John Mullinax, MacLean Hall, Julie Le, Krithika Kodumudi, Erica Royster, Allison Richards, Ricardo Gonzalez, Amod Sarnaik, Shari Pilon-Thomas, Morten Nielsen, Anders Krarup-Hansen, Dorrit Hovgaard, Michael Mørk Petersen, Anand Chainsukh Loya, Niels Junker, Charlotte Rivas, Robin Parihar, Cliona M. Rooney, Haiying Qin, Sang Nguyen, Paul Su, Chad Burk, Brynn Duncan, Bong-Hyun Kim, M. Eric Kohler, Terry Fry, Arjun A. Rao, Noam Teyssier, Jacob Pfeil, Nikolaos Sgourakis, Sofie Salama, David Haussler, Sarah A. Richman, Selene Nunez-Cruz, Zack Gershenson, Zissimos Mourelatos, David Barrett, Stephan Grupp, Michael Milone, Alba Rodriguez-Garcia, Matthew K. Robinson, Gregory P. Adams, João Santos, Riikka Havunen, Mikko Siurala, Víctor Cervera-Carrascón, Suvi Parviainen, Marjukka Antilla, Akseli Hemminki, Jyothi Sethuraman, Laurelis Santiago, Jie Qing Chen, Zhimin Dai, Huizi Sha, Shu Su, Naiqing Ding, Baorui Liu, Anna Pasetto, Sarah R. Helman, Steven A. Rosenberg, Melissa Burgess, Hui Zhang, Tien Lee, Hans Klingemann, Paul Nghiem, John M. Kirkwood, John M. Rossi, Marika Sherman, Allen Xue, Yueh-wei Shen, Lynn Navale, James N. Kochenderfer, Adrian Bot, Anandaraman Veerapathran, Doris Wiener, Edmund K. Waller, Jian-Ming Li, Christopher Petersen, Bruce R. Blazar, Jingxia Li, Cynthia R. Giver, Ziming Wang, Steven K. Grossenbacher, Ian Sturgill, Robert J. Canter, William J. Murphy, Congcong Zhang, Michael C. Burger, Lukas Jennewein, Anja Waldmann, Michel Mittelbronn, Torsten Tonn, Joachim P. Steinbach, Winfried S. Wels, Jason B. Williams, Yuanyuan Zha, Thomas F. Gajewski, LaTerrica C. Williams, Giedre Krenciute, Mamta Kalra, Chrystal Louis, Gang Xin, David Schauder, Aimin Jiang, Nikhil Joshi, Weiguo Cui, Xue Zeng, Zeguo Zhao, Mohamad Hamieh, Justin Eyquem, Gertrude Gunset, Neil Bander, Michel Sadelain, David Askmyr, Milad Abolhalaj, Kristina Lundberg, Lennart Greiff, Malin Lindstedt, Helen K. Angell, Kyoung-Mee Kim, Seung-Tae Kim, Sung Kim, Alan D. Sharpe, Julia Ogden, Anna Davenport, Darren R. Hodgson, Carl Barrett, Jeeyun Lee, Elaine Kilgour, Jodi Hanson, Richard Caspell, Alexey Karulin, Paul Lehmann, Tameem Ansari, Annemarie Schiller, Srividya Sundararaman, Diana Roen, Mark Ayers, Diane Levitan, Gladys Arreaza, Fang Liu, Robin Mogg, Yung-Jue Bang, Bert O’Neil, Razvan Cristescu, Philip Friedlander, Karl Wassman, Chrisann Kyi, William Oh, Nina Bhardwaj, Svetlana Bornschlegl, Michael P. Gustafson, Dennis A. Gastineau, Ian F. Parney, Allan B. Dietz, Daniel Carvajal-Hausdorf, Nikita Mani, Kurt Schalper, David Rimm, Serena Chang, John Kurland, Christoph Matthias Ahlers, Maria Jure-Kunkel, Lewis Cohen, Holden Maecker, Holbrook Kohrt, Shuming Chen, George Crabill, Theresa Pritchard, Tracee McMiller, Drew Pardoll, Fan Pan, Suzanne Topalian, Patrick Danaher, Sarah Warren, Lucas Dennis, Andrew M. White, Leonard D’Amico, Melissa Geller, Mary L. Disis, Joseph Beechem, Kunle Odunsi, Steven Fling, Roshanak Derakhshandeh, Tonya J. Webb, Sigrid Dubois, Kevin Conlon, Bonita Bryant, Jennifer Hsu, Nancy Beltran, Jürgen Müller, Thomas Waldmann, Rebekka Duhen, Thomas Duhen, Lucas Thompson, Ryan Montler, Andrew Weinberg, Max Kates, Brandon Early, Erik Yusko, Taylor H. Schreiber, Trinity J. Bivalacqua, Jared Lunceford, Michael Nebozhyn, Erin Murphy, Andrey Loboda, David R. Kaufman, Andrew Albright, Jonathan Cheng, S. Peter Kang, Veena Shankaran, Sarina A. Piha-Paul, Jennifer Yearley, Tanguy Seiwert, Antoni Ribas, Terrill K. McClanahan, Xinwei Sher, Xiao Qiao Liu, Andrew Joe, Elizabeth Plimack, Alex Forrest-Hay, Cheryl A. Guyre, Kohei Narumiya, Marc Delcommenne, Heather A. Hirsch, Amit Deshpande, Jason Reeves, Jenny Shu, Tong Zi, Jennifer Michaelson, Debbie Law, Elizabeth Trehu, Sriram Sathyanaryanan, Brendan P. Hodkinson, Natalie A. Hutnick, Michael E. Schaffer, Michael Gormley, Tyler Hulett, Carmen Ballesteros-Merino, Christopher Dubay, Michael Afentoulis, Ashok Reddy, Larry David, Kumar Jayant, Swati Agrawal, Rajendra Agrawal, Ghayathri Jeyakumar, Seongho Kim, Heejin Kim, Cynthia Silski, Stacey Suisham, Elisabeth Heath, Ulka Vaishampayan, Natalie Vandeven, Natasja Nielsen Viller, Alison O’Connor, Hui Chen, Bolette Bossen, Eric Sievers, Robert Uger, Lisa Johnson, Hsiang-Fong Kao, Chin-Fu Hsiao, Shu-Chuan Lai, Chun-Wei Wang, Jenq-Yuh Ko, Pei-Jen Lou, Tsai-Jan Lee, Tsang-Wu Liu, Ruey-Long Hong, Staci J. Kearney, Joshua C. Black, Benjamin J. Landis, Sally Koegler, Brooke Hirsch, Roberto Gianani, Jeffrey Kim, Ming-Xiao He, Bingqing Zhang, Nan Su, Yuling Luo, Xiao-Jun Ma, Emily Park, Dae Won Kim, Domenico Copploa, Nishi Kothari, Young doo Chang, Richard Kim, Namyong Kim, Melvin Lye, Ee Wan, Hanna A. Knaus, Sofia Berglund, Hubert Hackl, Judith E. Karp, Ivana Gojo, Leo Luznik, Henoch S. Hong, Sven D. Koch, Birgit Scheel, Ulrike Gnad-Vogt, Karl-Josef Kallen, Volker Wiegand, Linus Backert, Oliver Kohlbacher, Ingmar Hoerr, Mariola Fotin-Mleczek, James M. Billingsley, Yoshinobu Koguchi, Valerie Conrad, William Miller, Iliana Gonzalez, Tomasz Poplonski, Tanisha Meeuwsen, Ana Howells-Ferreira, Rogan Rattray, Mary Campbell, Carlo Bifulco, Keith Bahjat, Brendan Curti, E-K Vetsika, G. Kallergi, Despoina Aggouraki, Z. Lyristi, P. Katsarlinos, Filippos Koinis, V. Georgoulias, Athanasios Kotsakis, Nathan T. Martin, Famke Aeffner, Logan Cerkovnik, Luke Pratte, Rebecca Kim, Joseph Krueger, Amaia Martínez-Usatorre, Camilla Jandus, Alena Donda, Laura Carretero-Iglesia, Daniel E. Speiser, Dietmar Zehn, Nathalie Rufer, Pedro Romero, Anshuman Panda, Janice Mehnert, Kim M. Hirshfield, Greg Riedlinger, Sherri Damare, Tracie Saunders, Levi Sokol, Mark Stein, Elizabeth Poplin, Lorna Rodriguez-Rodriguez, Ann Silk, Nancy Chan, Melissa Frankel, Michael Kane, Jyoti Malhotra, Joseph Aisner, Howard L. Kaufman, Siraj Ali, Jeffrey Ross, Eileen White, Gyan Bhanot, Shridar Ganesan, Anne Monette, Derek Bergeron, Amira Ben Amor, Liliane Meunier, Christine Caron, Antigoni Morou, Daniel Kaufmann, Moishe Liberman, Igor Jurisica, Anne-Marie Mes-Masson, Kamel Hamzaoui, Rejean Lapointe, Ann Mongan, Yuan-Chieh Ku, Warren Tom, Yongming Sun, Alex Pankov, Tim Looney, Janice Au-Young, Fiona Hyland, Jeff Conroy, Carl Morrison, Sean Glenn, Blake Burgher, He Ji, Mark Gardner, Angela R. Omilian, Wiam Bshara, Omilian Angela, Joseph M. Obeid, Gulsun Erdag, Mark E. Smolkin, Donna H. Deacon, James W. Patterson, Lieping Chen, Timothy N. Bullock, Craig L. Slingluff, John T. Loffredo, Raja Vuyyuru, Sophie Beyer, Vanessa M. Spires, Maxine Fox, Jon M. Ehrmann, Katrina A. Taylor, Alan J. Korman, Robert F. Graziano, David Page, Katherine Sanchez, Maritza Martel, Mariana Petaccia De Macedo, Yong Qin, Alex Reuben, Christine Spencer, Michele Guindani, Adriana Racolta, Brian Kelly, Tobin Jones, Nathan Polaske, Noah Theiss, Mark Robida, Jeffrey Meridew, Iva Habensus, Liping Zhang, Lidija Pestic-Dragovich, Lei Tang, Ryan J. Sullivan, Thomas Olencki, Thomas Hutson, Joanna Roder, Shauna Blackmon, Heinrich Roder, John Stewart, Asim Amin, Marc S. Ernstoff, Joseph I. Clark, Michael B. Atkins, Jeffrey Sosman, David F. McDermott, Harriet Kluger, Ruth Halaban, Mario Snzol, Senait Asmellash, Arni Steingrimsson, Chichung Wang, Kristin Roman, Amanda Clement, Sean Downing, Clifford Hoyt, Nathalie Harder, Guenter Schmidt, Ralf Schoenmeyer, Nicolas Brieu, Mehmet Yigitsoy, Gabriele Madonna, Gerardo Botti, Antonio Grimaldi, Paolo A. Ascierto, Ralf Huss, Maria Athelogou, Harald Hessel, Alexander Buchner, Christian Stief, Gerd Binnig, Thomas Kirchner, Shankar Sellappan, Sheeno Thyparambil, Sarit Schwartz, Fabiola Cecchi, Andrew Nguyen, Charles Vaske, Todd Hembrough, Jan Spacek, Michal Vocka, Eva Zavadova, Helena Skalova, Pavel Dundr, Lubos Petruzelka, Nicole Francis, Rau T. Tilman, Arndt Hartmann, Irena Netikova, Julia Stump, Amanda Tufman, Frank Berger, Michael Neuberger, Rudolf Hatz, Michael Lindner, Rachel E. Sanborn, John Handy, Rudolf M. Huber, Hauke Winter, Simone Reu, Cheng Sun, Weihua Xiao, Zhigang Tian, Kshitij Arora, Niyati Desai, Anupriya Kulkarni, Mihir Rajurkar, Miguel Rivera, Vikram Deshpande, David Ting, Katy Tsai, Adi Nosrati, Simone Goldinger, Omid Hamid, Alain Algazi, Paul Tumeh, Jimmy Hwang, Jacqueline Liu, Lawrence Chen, Reinhard Dummer, Michael Rosenblum, Adil Daud, Tsu-Shuen Tsao, Julia Ashworth-Sharpe, Donald Johnson, Srabani Bhaumik, Christopher Bieniarz, Joseph Couto, Michael Farrell, Mahsa Ghaffari, Antony Hubbard, Jerome Kosmeder, Cleo Lee, Erin Marner, Diana Uribe, Hongjun Zhang, Jian Zhang, Wenjun Zhang, Yifei Zhu, Larry Morrison, Takahiro Tsujikawa, Rohan N. Borkar, Vahid Azimi, Sushil Kumar, Guillaume Thibault, Motomi Mori, Edward El Rassi, Daniel R. Clayburgh, Molly F. Kulesz-Martin, Paul W. Flint, Lisa M. Coussens, Lisa Villabona, Giuseppe V. Masucci, Gary Geiss, Brian Birditt, Qian Mei, Alan Huang, Maribeth A. Eagan, Eduardo Ignacio, Nathan Elliott, Dwayne Dunaway, Jaemyeong Jung, Chris Merritt, Isaac Sprague, Philippa Webster, Yan Liang, Jessica Wenthe, Gunilla Enblad, Hannah Karlsson, Magnus Essand, Barbara Savoldo, Gianpietro Dotti, Martin Höglund, Malcolm K. Brenner, Hans Hagberg, Angelica Loskog, Matthew J. Bernett, Gregory L. Moore, Michael Hedvat, Christine Bonzon, Seung Chu, Rumana Rashid, Kendra N. Avery, Umesh Muchhal, John Desjarlais, Matthew Kraman, Katarzyna Kmiecik, Natalie Allen, Mustapha Faroudi, Carlo Zimarino, Mateusz Wydro, Jacqueline Doody, Sreesha P. Srinivasa, Nagaraja Govindappa, Praveen Reddy, Aparajita Dubey, Sankar Periyasamy, Madhukara Adekandi, Chaitali Dey, Mary Joy, Pieter Fokko van Loo, Henrike Veninga, Setareh Shamsili, Mark Throsby, Harry Dolstra, Lex Bakker, Ajjai Alva, Juergen Gschwendt, Yohann Loriot, Joaquim Bellmunt, Dai Feng, Christian Poehlein, Thomas Powles, Emmanuel S. Antonarakis, Charles G. Drake, Haiyan Wu, Johann De Bono, Rajat Bannerji, John Byrd, Gareth Gregory, Stephen Opat, Jake Shortt, Andrew J. Yee, Noopur Raje, Seth Thompson, Arun Balakumaran, Shaji Kumar, Brian I. Rini, Toni K. Choueiri, Mariangela Mariani, Laurence Albiges, John B. Haanen, James Larkin, Manuela Schmidinger, Domenico Magazzù, Alessandra di Pietro, Robert J. Motzer, Troels Holz Borch, Per Kongsted, Magnus Pedersen, Özcan Met, Karim Boudadi, Hao Wang, James Vasselli, Jan E. Baughman, Jon Wigginton, Rehab Abdallah, Ashley Ross, Jiwon Park, Steven Grossenbacher, Jesus I. Luna, Sita Withers, William Culp, Mingyi Chen, Arta Monjazeb, Michael S. Kent, Smita Chandran, David Danforth, James Yang, Christopher Klebanoff, Stephanie Goff, Biman Paria, Arvind Sabesan, Abhishek Srivastava, Udai Kammula, Jon Richards, Mark Faries, Robert H. I. Andtbacka, Luis A. Diaz, Dung T. Le, Takayuki Yoshino, Thierry André, Johanna Bendell, Minori Koshiji, Yayan Zhang, S Peter Kang, Bao Lam, Dirk Jäger, Todd M. Bauer, Judy S. Wang, Jean K. Lee, Gulam A. Manji, Ragini Kudchadkar, John S. Kauh, Shande Tang, Naomi Laing, Gerald Falchook, Edward B. Garon, Balazs Halmos, Hui Rina, Natasha Leighl, Sung Sook Lee, William Walsh, Konstanin Dragnev, Bilal Piperdi, Luis Paz-Ares Rodriguez, Nabeegha Shinwari, Ziewn Wei, Mary L Maas, Michael Deeds, Adam Armstrong, Tim Peterson, Sue Steinmetz, Thomas Herzog, Floor J. Backes, Larry Copeland, Maria Del Pilar Estevez Diz, Thomas W. Hare, Warner Huh, Byoung-Gie Kim, Kathleen M. Moore, Ana Oaknin, William Small, Krishnansu S. Tewari, Bradley J. Monk, Ashish M. Kamat, Kijoeng Nam, Maria De Santis, Robert Dreicer, Noah M. Hahn, Rodolfo Perini, Arlene Siefker-Radtke, Guru Sonpavde, Ronald de Wit, J. Alfred Witjes, Stephen Keefe, Dean Bajorin, Philippe Armand, John Kuruvilla, Craig Moskowitz, Mehdi Hamadani, Pier Luigi Zinzani, Sabine Chlosta, Nancy Bartlett, Rachel Sabado, Yvonne Saenger, Loging William, Michael Joseph Donovan, Erlinda Sacris, John Mandeli, Andres M. Salazar, John Powderly, Joshua Brody, John Nemunaitis, Leisha Emens, Amita Patnaik, Ian McCaffery, Richard Miller, Ginna Laport, Andrew L. Coveler, David C. Smith, Juneko E. Grilley-Olson, Sanjay Goel, Shyra J. Gardai, Che-Leung Law, Gary Means, Thomas Manley, Kristen A. Marrone, Gary Rosner, Valsamo Anagnostou, Joanne Riemer, Jessica Wakefield, Cynthia Zanhow, Stephen Baylin, Barbara Gitlitz, Julie Brahmer, Sabina Signoretti, Wenting Li, Charles Schloss, Jean-Marie Michot, Wei Ding, Beth Christian, Patricia Marinello, Margaret Shipp, Yana G. Najjar, null Lin, Lisa H. Butterfield, Ahmad A. Tarhini, Diwakar Davar, Hassane Zarour, Elizabeth Rush, Cindy Sander, Siqing Fu, Todd Bauer, Chris Molineaux, Mark K. Bennett, Keith W. Orford, Kyriakos P. Papadopoulos, Sukhmani K. Padda, Sumit A. Shah, A Dimitrios Colevas, Sujata Narayanan, George A. Fisher, Dana Supan, Heather A. Wakelee, Rhonda Aoki, Mark D. Pegram, Victor M. Villalobos, Jie Liu, Chris H. Takimoto, Mark Chao, Jens-Peter Volkmer, Ravindra Majeti, Irving L. Weissman, Branimir I. Sikic, Wendy Yu, Alison Conlin, Janet Ruzich, Stacy Lewis, Anupama Acheson, Kathleen Kemmer, Kelly Perlewitz, Nicole M. Moxon, Staci Mellinger, Heather McArthur, Trine Juhler-Nøttrup, Jayesh Desai, Ben Markman, Shahneen Sandhu, Hui Gan, Michael L. Friedlander, Ben Tran, Tarek Meniawy, Joanne Lundy, Duncan Colyer, Malaka Ameratunga, Christie Norris, Jason Yang, Kang Li, Lai Wang, Lusong Luo, Zhen Qin, Song Mu, Xuemei Tan, James Song, Michael Millward, Matthew H. G. Katz, Todd W. Bauer, Gauri R. Varadhachary, Nicolas Acquavella, Nipun Merchant, Gina Petroni, Osama E. Rahma, Mei Chen, Yang Song, Markus Puhlmann, Arun Khattri, Ryan Brisson, Christopher Harvey, Jatin Shah, Maria Victoria Mateos, Morio Matsumoto, Hilary Blacklock, Albert Oriol Rocafiguera, Hartmut Goldschmidt, Shinsuke Iida, Dina Ben Yehuda, Enrique Ocio, Paula Rodríguez-Otero, Sundar Jagannath, Sagar Lonial, Uma Kher, Jesus San-Miguel, Moacyr Ribeiro de Oliveira, Habte Yimer, Robert Rifkin, Fredrik Schjesvold, Razi Ghori, Anna Spreafico, Victor Lee, Roger K. C. Ngan, Ka Fai To, Myung Ju Ahn, Quan Sing Ng, Jin-Ching Lin, Ramona F. Swaby, Christine Gause, Sanatan Saraf, Anthony T. C. Chan, Elaine Lam, Nizar M. Tannir, Funda Meric-Bernstam, Matt Gross, Andy MacKinnon, Sam Whiting, Martin Voss, Evan Y. Yu, Mark R. Albertini, Erik A. Ranheim, Jacquelyn A. Hank, Cindy Zuleger, Thomas McFarland, Jennifer Collins, Erin Clements, Sharon Weber, Tracey Weigel, Heather Neuman, Greg Hartig, David Mahvi, MaryBeth Henry, Jacek Gan, Richard Yang, Lakeesha Carmichael, KyungMann Kim, Stephen D. Gillies, Paul M. Sondel, Vivek Subbiah, Lori Noffsinger, Kyle Hendricks, Marnix Bosch, Jay M. Lee, Mi-Heon Lee, Jonathan W. Goldman, Felicita E. Baratelli, Dorthe Schaue, Gerald Wang, Frances Rosen, Jane Yanagawa, Tonya C. Walser, Ying Q. Lin, Sharon Adams, Franco M. Marincola, Paul C. Tumeh, Fereidoun Abtin, Robert Suh, Karen Reckamp, William D. Wallace, Gang Zeng, David A. Elashoff, Sherven Sharma, Steven M. Dubinett, Anna C. Pavlick, Brian Gastman, Brent Hanks, Tibor Keler, Tom Davis, Laura A. Vitale, Elad Sharon, Chihiro Morishima, Martin Cheever, Christopher R. Heery, Joseph W. Kim, Elizabeth Lamping, Jennifer Marte, Sheri McMahon, Lisa Cordes, Farhad Fakhrejahani, Ravi Madan, Rachel Salazar, Maggie Zhang, Christoph Helwig, James L Gulley, Roger Li, John Amrhein, Zvi Cohen, Monique Champagne, Ashish Kamat, M. Angela Aznar, Sara Labiano, Angel Diaz-Lagares, Manel Esteller, Juan Sandoval, Susannah D. Barbee, David I. Bellovin, John C. Timmer, Nebiyu Wondyfraw, Susan Johnson, Johanna Park, Amanda Chen, Mikayel Mkrtichyan, Amir S. Razai, Kyle S. Jones, Chelsie Y. Hata, Denise Gonzalez, Quinn Deveraux, Brendan P. Eckelman, Luis Borges, Rukmini Bhardwaj, Raj K. Puri, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Todd Bartkowiak, Ashvin Jaiswal, Casey Ager, Midan Ai, Pratha Budhani, Renee Chin, David Hong, Michael Curran, William D. Hastings, Maria Pinzon-Ortiz, Masato Murakami, Jason R. Dobson, David Quinn, Joel P. Wagner, Xianhui Rong, Pamela Shaw, Ernesta Dammassa, Wei Guan, Glenn Dranoff, Alexander Cao, Ross B. Fulton, Steven Leonardo, Kathryn Fraser, Takashi O. Kangas, Nadine Ottoson, Nandita Bose, Richard D. Huhn, Jeremy Graff, Jamie Lowe, Keith Gorden, Mark Uhlik, Thomas O’Neill, Jenifer Widger, Andrea Crocker, Li-Zhen He, Jeffrey Weidlick, Karuna Sundarapandiyan, Venky Ramakrishna, James Storey, Lawrence J. Thomas, Joel Goldstein, Henry C. Marsh, Jamison Grailer, Julia Gilden, Pete Stecha, Denise Garvin, Jim Hartnett, Frank Fan, Mei Cong, Zhi-jie Jey Cheng, Marlon J. Hinner, Rachida-Siham Bel Aiba, Corinna Schlosser, Thomas Jaquin, Andrea Allersdorfer, Sven Berger, Alexander Wiedenmann, Gabriele Matschiner, Julia Schüler, Ulrich Moebius, Christine Rothe, Olwill A. Shane, Brendan Horton, Stefani Spranger, Dayson Moreira, Tomasz Adamus, Xingli Zhao, Piotr Swiderski, Sumanta Pal, Marcin Kortylewski, Alyssa Kosmides, Kevin Necochea, Kathleen M. Mahoney, Sachet A. Shukla, Nikolaos Patsoukis, Apoorvi Chaudhri, Hung Pham, Ping Hua, Xia Bu, Baogong Zhu, Nir Hacohen, Catherine J. Wu, Edward Fritsch, Vassiliki A. Boussiotis, Gordon J. Freeman, Amy E. Moran, Fanny Polesso, Lisa Lukaesko, Emelie Rådestad, Lars Egevad, Berit Sundberg, Lars Henningsohn, Victor Levitsky, William Rafelson, John L. Reagan, Loren Fast, Pottayil Sasikumar, Naremaddepalli Sudarshan, Raghuveer Ramachandra, Nagesh Gowda, Dodheri Samiulla, Talapaneni Chandrasekhar, Sreenivas Adurthi, Jiju Mani, Rashmi Nair, Amit Dhudashia, Nagaraj Gowda, Murali Ramachandra, Alexander Sankin, Benjamin Gartrell, Kerwin Cumberbatch, Hongying Huang, Joshua Stern, Mark Schoenberg, Xingxing Zang, Ryan Swanson, Michael Kornacker, Lawrence Evans, Erika Rickel, Martin Wolfson, Sandrine Valsesia-Wittmann, Tala Shekarian, François Simard, Rodrigo Nailo, Aurélie Dutour, Anne-Catherine Jallas, Christophe Caux, and Aurélien Marabelle
- Subjects
Pharmacology ,0303 health sciences ,Cancer Research ,Side effect ,business.industry ,medicine.drug_class ,Immunology ,Phases of clinical research ,Monoclonal antibody ,Phase i study ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Molecular Medicine ,Immunology and Allergy ,Medicine ,In patient ,Programmed death 1 ,business ,030304 developmental biology - Published
- 2016
- Full Text
- View/download PDF
39. Pluripotent Cell-Derived Off-the-Shelf TCR-Less CAR-Targeted Cytotoxic T Cell Therapeutic for the Allogeneic Treatment of B Cell Malignancies
- Author
-
Yi-Shin Lai, Eigen Peralta, Bahram Valamehr, Mochtar Pribadi, Justin Eyquem, Judith Feucht, Ramzey Abujarour, Jason Dinella, Helen Chu, Mushtaq Husain, Chris Truong, Michel Sadelain, Laurel Stokely, Jorge Mansilla-Soto, Mohsen Sabouri-Ghomi, Chia-Wei Chang, Sjoukje J. C. van der Stegen, Thomas H. Lee, Raedun Clarke, Isabelle Riviere, and Miguel Meza
- Subjects
0301 basic medicine ,T cell ,Immunology ,T-cell receptor ,Juno Therapeutics ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CD19 ,Chimeric antigen receptor ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,medicine ,biology.protein ,Cytotoxic T cell ,IL-2 receptor ,B cell - Abstract
The advent of off-the-shelf chimeric antigen receptor (CAR) T cell therapeutics is widely recognized to be a major potential advancement for the treatment of cancer. Several obstacles currently hamper the broad use of CAR T cells, including the inherent variability and cost of manufacturing of autologous cellular populations, the absolute requirement for precise genetic editing in the allogeneic setting, and the challenge to keep pace with clonal heterogeneity. Here we present pre-clinical data for FT819, a first-of-kind off-the-shelf human induced pluripotent stem cell (hiPSC)-derived CAR T cell product. FT819 is defined by the precise genetic engineering of multiple targeting events at the single cell level to create a clonal master iPSC line. The engineered features include the targeted integration of a novel, modified CD19 CAR into the T cell receptor α (TRAC) locus to provide antigen specificity and enhanced efficacy while eliminating the possibility of graft versus host disease (GvHD), and the expression of a high-affinity, non-cleavable form of CD16 (hnCD16) to deliver an adjustable system to address tumor antigen escape. Through a proprietary cellular reprogramming platform, peripheral blood derived T cells are converted to hiPSCs, engineered to contain the modified CD19 CAR targeted into the TRAC locus and hnCD16, and clonally selected to create a master hiPSC line (TRAC-TiPSC, FT819). Molecular characterization of the TRAC-TiPSC master cell line by 5' junction, 3' junction and internal sequence PCR confirmed homology directed repair and bi-allelic targeting of the CD19 CAR into the TRAC locus. The origin of the clonal master cell bank was confirmed to be a TCRαβ T cell by PCR-mediated detection of TCRδ locus deletion and methyl-seq analysis of the TCRα locus. Flow cytometric analysis demonstrated the maintenance of a uniform population of hiPSCs (>95% SSEA4/TRA-1-81/OCT4/NANOG) and expression of hnCD16 transgene (>95% CD16). Utilizing our stage-specific T cell differentiation protocol, we demonstrate that the TRAC-TiPSCs yield TRAC-iT cells with uniform expression of the CAR (>95%), complete elimination of TCR surface expression and clinically enabling expansion through the manufacturing process (>50,000 fold). To confirm the lack of alloreactivity conferred by the deletion of endogenous TCR expression, mixed lymphocyte reactions were performed using TRAC-iT, primary TCR+ T cells and primary TCR+CAR+ T cells as responders and HLA-mismatched peripheral blood mononuclear cells (PBMCs) as targets. In comparison to primary T cells and primary CAR-T cells, TRAC-iT did not respond and proliferate in response to TCR stimulation or HLA-mismatched PBMCs indicating that the risk of GvHD was alleviated. In vitro functional studies established that TRAC-iT possess a potent cytotoxic T lymphocyte response to CD19 antigen challenge in a similar manner to peripheral blood CAR T cells as demonstrated by expression of markers indicative of degranulation (CD107a/b, Granzyme B), T cell activation (CD69, CD25), and production of INFγ, TNFα and IL2. Importantly, TRAC-iT targeted tumor in an antigen specific manner as verified by lysis of CD19+, but not CD19-, tumor cell lines as seen by in vitro cytolytic assays (50% killing E:T; TRAC-iT = 1:8, primary CAR-T = 1:4). In vivo studies demonstrated that TRAC-iT cells effectively control tumor progression in a mouse model of acute lymphoblastic leukemia Nalm6 (TRAC-iT versus no treatment, p Disclosures Clarke: Fate Therapeutics Inc.: Employment. Chang:Fate Therapeutics Inc.: Employment. Husain:Fate Therapeutics Inc.: Employment. Lai:Fate Therapeutics Inc.: Employment. Peralta:Fate Therapeutics Inc.: Employment. Stokely:Fate Therapeutics Inc.: Employment. Abujarour:Fate Therapeutics Inc.: Employment. Dinella:Fate Therapeutics Inc.: Employment. Lee:Fate Therapeutics Inc.: Employment. Pribadi:Fate Therapeutics Inc.: Employment. Chu:Fate Therapeutics Inc.: Employment. Truong:Fate Therapeutics Inc.: Employment. Sabouri-Ghomi:Fate Therapeutics Inc.: Employment. Meza:Fate Therapeutics Inc.: Employment. Riviere:Juno Therapeutics, a Celgene Company: Membership on an entity's Board of Directors or advisory committees, Research Funding; Fate Therapeutics Inc.: Research Funding. Sadelain:Juno Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; Fate Therapeutics Inc.: Research Funding. Valamehr:Fate Therapeutics Inc.: Employment.
- Published
- 2018
- Full Text
- View/download PDF
40. Calibrated CAR Activation Potential Directs Alternative T Cell Fates and Therapeutic Potency
- Author
-
Annalisa Cabriolu, Zeguo Zhao, Justin Eyquem, Judith Feucht, Jie Sun, Michel Sadelain, Anton Dobrin, Mohamad Hamieh, Yu-Jui Ho, and Josef Leibold
- Subjects
Chemistry ,T cell ,Immunology ,T-cell receptor ,Cell Biology ,Hematology ,Biochemistry ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,T cell differentiation ,Immunoreceptor tyrosine-based activation motif ,Disease remission ,medicine ,Potency ,Signal transduction - Abstract
Immunotherapy with second-generation chimeric antigen receptor (CAR) T cells has achieved great clinical success against hematological malignancies. CD19-specific CARs incorporating CD28 and CD3z signaling domains have demonstrated remarkable potency resulting in the frequent induction of complete remissions and were recently approved by the US FDA for use in some refractory B cell malignancies. We previously demonstrated that CD28-based CARs program strong effector functions, but impart a relatively limited T cell lifespan. Increasing functional T cell persistence is therefore likely to further enhance the therapeutic success of 1928z CAR T cells. We hypothesized that excessive signal strength arising from redundancy of combined CD3z and CD28 signals might foster terminal T cell differentiation and exhaustion. We therefore proceeded to titrate the activation potential of CD28-based CARs and assess the impact thereof on the function, longevity and therapeutic potency of CAR T cells. We analyzed the contribution of individual immunoreceptor tyrosine-based activation motifs (ITAMs) to the phenotype and function of 1928z CAR T cells. ITAM-mutated CAR T cells demonstrated similar expression levels in retrovirally transduced primary T cells and directed comparable short-term cytotoxicity and proliferation capacity in vitro. However, remarkable differences in their therapeutic potency emerged when T cells expressing different mutant CARs were tested in the pre-B acute lymphoblastic leukemia NALM6 mouse model. The CAR "stress test" model revealed that a CAR containing a single functional ITAM - depending on its position - outperformed wild-type 1928z CARs, achieving rapid and durable tumor eradication even at low T cell doses, by delaying T cell differentiation and exhaustion. CAR T cells retrieved from the bone marrow of treated mice demonstrated that inactivation of two ITAM domains augmented CAR persistence at the tumor sites with a higher percentage of central memory cells and a decreased proportion of terminally differentiated effector cells. Deletion mutants further revealed the importance of ITAM location within second-generation CAR T cells. These findings were rigorously tested by directing the mutant CAR cDNAs to the T-cell receptor α constant (TRAC) locus using CRISPR/Cas9 technology, thereby ruling out potentially confounding effects arising from different CAR expression levels. TRAC-1928z mutants demonstrated superior antitumor efficacy compared to conventional TRAC-1928z CARs and prevented terminal T cell differentiation and exhaustion. Genome-wide transcriptional profiles of TRAC-edited naïve peripheral blood T cells further demonstrated that CARs encoding different ITAMs direct T cells to different fates. While TRAC-1928z CARs demonstrated similarity to transcriptional profiles of effector cells, reducing the number of ITAMs to one ITAM preserved a less-differentiated T cell state and promoted greater T cell persistence. We identified one 1928z mutant CAR, which improved therapeutic potency and induced a transcriptional profile similar to that of stem cell memory T cells (TSCM). Another 1928z mutant CAR with further reduction of the activation potential resulted in a naïve-like phenotype with great proliferation potential and persistence, but greatly diminished anti-tumor efficacy. In conclusion, we demonstrate that the number and position of ITAMs in 1928z CAR T cells influence functional, phenotypic and transcriptional programs resulting in profound effects on therapeutic potency. Balancing T cell differentiation and acquisition of effector functions is essential to optimize therapeutic potency of CAR T cells and can be intrinsically regulated by defined mutations in the CD3z chain of 1928z CAR T cells. Improved therapeutic potency of CAR T cells can thus be achieved by calibrating activation strength, thus retaining memory functions and preventing exhaustion, without compromising effector functions. Importantly, we were able to identify a novel CAR design which programs a favorable balance of effector and memory signatures, inducing increased persistence of highly functional CARs with the replicative capacity of long-lived memory cells and potent effector functions. Clinical studies evaluating the new CAR design are in preparation. Disclosures Sadelain: Juno Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; Fate Therapeutics Inc.: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
41. Abstract 2567: Novel therapeutic interventions to alleviate CAR T cell-induced cytokine release syndrome
- Author
-
Sjoukje J. C. van der Stegen, Mohamad Hamieh, Justin Eyquem, Theodoros Giavridis, and Michel Sadelain
- Subjects
Cancer Research ,Myeloid ,biology ,business.industry ,medicine.medical_treatment ,medicine.disease ,Chimeric antigen receptor ,CD19 ,Cytokine release syndrome ,medicine.anatomical_structure ,Cytokine ,Oncology ,Intensive care ,Immunology ,medicine ,biology.protein ,Macrophage ,business ,B cell - Abstract
CD19 chimeric antigen receptor (CAR) therapy is highly effective for some refractory B cell malignancies. Its broad implementation is limited in part by the occurrence of cytokine release syndrome (CRS), which is characterized by fever, hypotension and respiratory insufficiency that may require intensive care to overcome. CRS is associated with elevated serum cytokines, including interleukin-6 (IL-6), the blockade of which abates CRS in some but not all patients. The biological mechanisms involved in the pathogenesis of CRS are currently unknown, including the specific role of CAR T cells, tumor cells and other host cells. The development of relevant animal models is imperative in order to study the biology of CRS, analyze the role of different cell populations and ultimately identify interventions that not only treat but prevent CRS without reducing the therapeutic efficacy of CAR T cells. Here we present a novel mouse model in which CD19 CAR-induced CRS is predictably and reproducibly elicited and mirrors the rapid onset and acuteness of clinical CRS. Symptoms present within 24 hours post CAR T cell administration, including rapid weight loss and eventual mortality. Survival correlates negatively with pro-inflammatory human (T cell-derived) and murine (host-derived) cytokine levels. A comparison between the cytokine fingerprint in patients with CRS and our mouse model revealed remarkable similarity between the two profiles. Importantly, IL-6 is elevated, as seen in patients receiving CAR therapy, but of mouse origin. In accordance with the demonstrated clinical benefit of tocilizumab, we demonstrate that CRS-associated mortality can be abrogated by murine IL-6 receptor blockade. We further show that CAR T cell - tumor interaction is required for the robust recruitment of myeloid cells to the tumor site, including neutrophils, eosinophils, macrophages, monocytes and dendritic cells. Cytokine measurements and the transcriptomic analysis of myeloid populations from multiple sites reveal that IL-6 is of myeloid cell origin, dependent on CAR T cell - myeloid proximity and interactions at the tumor site. Furthermore, we probe the impact of myeloid involvement by activating or suppressing macrophage functions to show that CRS outcomes are largely determined by macrophage engagement. Probing into the functions of macrophages led us to identify multiple therapeutic interventions that can ameliorate the toxicities of CRS. We have also designed a novel CAR construct that can prevent CRS-associated mortality while maintaining intact antitumor efficacy in a mouse model. Overall, our results suggest an indispensible role for the myeloid system in severe CRS, especially macrophages, representing a cellular compartment that has been largely overlooked in pre-clinical models to date. Moreover, our model should be useful to guide the rational design of novel CAR T cells that can autonomously ameliorate or prevent CRS. Citation Format: Theodoros Giavridis, Sjoukje J. van der Stegen, Justin Eyquem, Mohamad Hamieh, Michel Sadelain. Novel therapeutic interventions to alleviate CAR T cell-induced cytokine release syndrome [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2567.
- Published
- 2018
- Full Text
- View/download PDF
42. Abstract IA22: Targeting CARs to the TRAC locus enhances T-cell potency
- Author
-
Michel Sadelain and Justin Eyquem
- Subjects
Cancer Research ,biology ,T cell ,T-cell receptor ,CD19 ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,Genome editing ,medicine ,biology.protein ,Cancer research ,CRISPR ,Gene silencing ,B cell - Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that redirect and reprogram T cells to mediate tumor rejection. The most successful CARs used to date are those targeting CD19, which offer the prospect of complete remissions in patients with chemorefractory/relapsed B cell malignancies. CARs are typically transduced into patient T cells using γ-retroviral or other randomly-integrating retroviral vectors (RVs), which may result in variegated CAR expression and transcriptional silencing. Recent advances in genome editing enable efficient sequence-specific modifications in human primary cells, including site-specific transgene integration into the CCR5 and AAVS1 loci in T lymphocytes. Using CRISPR/Cas9 to edit human T cells, we have established conditions yielding efficient target gene disruption and CAR insertion in a single step. We have found that directing a CD19 CAR to the human T-cell receptor (TCR) alpha chain (TRAC) locus not only results in efficient and uniform CAR expression in human peripheral blood T cells, but, remarkably, also enhances T-cell potency, vastly outperforming that of conventionally generated CAR T cells. We further show that CAR gene expression under the control of the TCR alpha promoter minimizes tonic signaling and allows effective regulation of CAR expression. These findings provide important insights into CAR immunobiology and highlight the potential of CRISPR/Cas9 genome editing to advance immunotherapies. Citation Format: Michel Sadelain, Justin Eyquem. Targeting CARs to the TRAC locus enhances T-cell potency [abstract]. In: Proceedings of the Second AACR Conference on Hematologic Malignancies: Translating Discoveries to Novel Therapies; May 6-9, 2017; Boston, MA. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(24_Suppl):Abstract nr IA22.
- Published
- 2017
- Full Text
- View/download PDF
43. Multiplex Genome-Edited T-cell Manufacturing Platform for 'Off-the-Shelf' Adoptive T-cell Immunotherapies
- Author
-
Gordon Weng-Kit Cheung, Agnès Gouble, Sophie Derniame, Roman Galetto, Justin Eyquem, Brian Philip, Cécile Schiffer-Mannioui, Pierrick Potrel, Julianne Smith, Karl S. Peggs, Aymeric Duclert, Cécile Bas, Andrew M. Scharenberg, Laetitia Lemaire, Martin Pule, Diane Le Clerre, Céline Lebuhotel, Sylvain Arnould, Laurent Poirot, and Isabelle Chion-Sotinel
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,CD52 ,Lymphoma ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell, alpha-beta ,Recombinant Fusion Proteins ,T-Lymphocytes ,Antigens, CD19 ,Molecular Sequence Data ,Drug Resistance ,Receptors, Antigen, T-Cell ,Graft vs Host Disease ,Biology ,Antibodies, Monoclonal, Humanized ,Lymphocyte Activation ,Transfection ,Immunotherapy, Adoptive ,Gene Knockout Techniques ,Mice ,Cancer immunotherapy ,Genome editing ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,medicine ,Animals ,Humans ,RNA, Messenger ,Alemtuzumab ,Glycoproteins ,Transcription activator-like effector nuclease ,Base Sequence ,Antibodies, Monoclonal ,Immunotherapy ,Virology ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Mice, Mutant Strains ,Cell biology ,medicine.anatomical_structure ,Oncology ,CD52 Antigen - 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. Cancer Res; 75(18); 3853–64. ©2015 AACR.
- Published
- 2014
44. Probing the AML Surfaceome for Chimeric Antigen Receptor (CAR) Targets
- Author
-
Mohamad Hamieh, Rupa Juthani, Ronald C. Hendrickson, Fabiana Perna, Samuel H. Berman, Jorge Mansilla-Soto, Cameron Brennan, Rajesh K. Soni, Michel Sadelain, and Justin Eyquem
- Subjects
0301 basic medicine ,biology ,T cell ,Immunology ,CD34 ,Cell Biology ,Hematology ,Biochemistry ,CD19 ,Chimeric antigen receptor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Antigen ,Cancer stem cell ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Stem cell ,B cell - Abstract
Adoptive T cell therapy using chimeric antigen receptors (CARs) to redirect the specificity and function of T lymphocytes has demonstrated efficacy in patients with lymphoid malignancies, in particular acute lymphoblastic leukemia (ALL). CD19 CAR therapy can induce durable complete remissions in subjects with CD19+ malignancies for whom chemotherapies have led to drug resistance and tumor progression. We previously obtained "breakthrough designation" from the US FDA for CD19 CAR therapy for adult ALL. The success of CD19 CAR therapy bodes well for tackling other hematological malignancies, including Acute Myeloid Leukemia (AML). The challenge for developing an effective CAR therapy for AML lies in the lack of suitable CAR targets. In ALL, CD19 is expressed on most if not all tumor cells, including tumor-initiating cells, and is only expressed in the normal B cell lineage. In contrast, the AML CAR targets proposed to date (Le-Y, CD123, CD33 and folate receptor-β) do not share this profile. They have not yielded effective and safe tumor eradication in early clinical studies. The clonal heterogeneity of AML and the similarity of AML cancer stem cells to normal hematopoietic stem cells pose additional challenges. Searching for better targets is thus essential. However, the identification of CAR targets is limited by the lack of reliable tools to assess antigen expression across all human tissues. In order to identify suitable CAR targets for AML, we integrated multiple transcriptomic and proteomic data sources and created an algorithm to identify proteins that can be targeted by single and combinatorial CAR strategies. To comprehensively probe the AML surfaceome, we compiled 474 molecules identified in previous studies and 3,675 molecules we identified by membrane protein biotinylation followed by Mass-Spectometry analysis. To assess whole body antigen expression, we integrated large sets of protein and mRNA expression data and annotated the expression of each AML surface molecule in normal cell types, tissues and organs, including hematopoietic stem cells (HSCs). We then established a "quality control" filter to assign priority antigens, based on their presence in at least two protein expression datasets (Human Protein Atlas, Human Protein Map and/or Proteomics Database) and their membrane-association. This step yielded 1,694 AML molecules. We further selected molecules with low expression in 64 normal tissues/organs and excluded antigens with high expression in bone marrow HSCs. These steps reduced candidate targets to 215 proteins. From these, we identified 32 targets overexpressed in diverse AML cells and showing very low overall expression in normal tissues. We performed systematic validation analyses by flow cytometry and identified 11 top candidates with low expression in normal CD34+ and CD34+CD38- HSCs and high expression in a panel of AML cells. Further elimination of molecules expressed in T cells reduced the candidate target number to 4, including 2 G-protein coupled receptors not previously reported as CAR targets in AML. These molecules were however still minimally expressed in some normal tissues, which prompted us to search for pairs of antigens with non-overlapping expression in normal tissues as a strategy to reduce on-target/off-tumor cytotoxicity. We identified 55 such pairs. Our validation studies have so far identified 3 unique pairs of targets showing absent co-expression in normal tissues and ~100% co-expression in a small panel of AML cells. These promising pairs are targeted by T cells co-expressing a CAR specific for one antigen and a chimeric costimulatory receptor (CCR) specific for the other. In a proof-of-principle study, we demonstrate that dual-targeted CAR T cells specific for CD33 and CD70 effectively lyse AML cells with diminished reactivity relative to single-targeted CAR T cells. Further validation studies in larger AML panels are in progress. This novel discovery approach to CAR target identification should prove very useful to expand CAR therapy applications to AML and other malignancies. Disclosures Sadelain: Juno Therapeutics: Consultancy.
- Published
- 2016
- Full Text
- View/download PDF
45. Abstract 2309: T-cell development from T cell-derived induced pluripotent stem cell
- Author
-
Maria Themeli, Sjoukje J. C. van der Stegen, Michel Sadelain, Justin Eyquem, and Jorge Mansilla-Soto
- Subjects
Cancer Research ,Induced stem cells ,T cell ,T-cell receptor ,Notch signaling pathway ,Biology ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,Oncology ,T cell differentiation ,medicine ,Induced pluripotent stem cell ,CD8 - Abstract
The ability to differentiate T cells of defined specificity and function from T-cell derived induced pluripotent stem cells (TiPSC) may be useful for the treatment of a range of pathologies, including cancer, infection and immune deficits. We recently reported that genetic engineering of TiPSC to express a Chimeric Antigen Receptor (CAR) is an effective strategy to combine the unlimited availability of TiPSC and facilitate reprogramming of the T cell specificity and functional potential. In vivo, CD19-retargeted human TiPS-derived CAR T cells (CARTiPSC-T) display therapeutic potency in a lymphoma model. Surprisingly, despite expression of the endogenous αβ TCR, the CARTiPSC-T cells possessed innate-like phenotype and function, most similar to γδ T cells. Although innate T cells have anti-tumor activity, they lack some vital features for therapeutic efficacy including long-term memory and in vivo persistence, which characterize mature CD8+ and CD4+ αβ TCR T cells. Additional research into the mechanisms underlying in vitro T cell differentiation of TiPSC is required to improve the development of mature TCRαβ T cells and facilitate the employment of their therapeutic potential. T cell lineage determination depends in part on the balance between Notch and TCR signaling, we are investigating their respective roles, as well as that of the CAR, in determining lineage commitment. We hypothesize that the combined CAR and early CD3/TCRαβ expression disrupts the TCR/Notch signaling balance, prohibiting mature TCRαβ T cell development. To study the effects of the different Notch ligands, TiPSC were differentiated on OP9 cells expressing one of four Notch ligands (DLL1, DLL4, Jagged-1 or Jagged-2). Preliminary data suggests that DLL4 is able to facilitate T cell development to CD4/CD8 double positive (DP) stage, however, this is hindered by CAR expression. To determine the role of early TCRαβ expression, elimination of TCRαβ expression was facilitated by targeted disruption of the TCRα constant region using the CRISPR/Cas9 system. Elimination of early TCRαβ expression showed improved development towards DP stage of TiPSC on OP9-DLL1. TiPSC are a valuable system for the study of human T cell differentiation. In addition, and most importantly they are further amenable to genetic engineering with TCRs or CARs, which may be useful for the generation of therapeutic “off-the-shelf”, antigen-specific T cells. Citation Format: Sjoukje J.C. van der Stegen, Maria Themeli, Justin Eyquem, Jorge Mansilla-Soto, Michel Sadelain. T-cell development from T cell-derived induced pluripotent stem cell. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2309.
- Published
- 2016
- Full Text
- View/download PDF
46. 274. One-Step Generation of Universal CAR T Cells
- Author
-
Justin Eyquem, Ashlesha Odak, Michel Sadelain, and Jorge Mansilla-Soto
- Subjects
Pharmacology ,Genetics ,Transcription activator-like effector nuclease ,T cell ,Biology ,Sleeping Beauty transposon system ,Zinc finger nuclease ,Chimeric antigen receptor ,Viral vector ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Genome editing ,TAL effector ,030220 oncology & carcinogenesis ,Drug Discovery ,medicine ,Molecular Medicine ,Molecular Biology ,030215 immunology - Abstract
Adoptive immunotherapy using chimeric antigen receptors (CARs) has shown remarkable clinical results in the treatment of leukemia and is one of the most promising new strategies to treat cancer. Current clinical protocols utilize autologous T cells that are collected by apheresis and engineered with retroviral vectors to stably express the CAR. This approach therefore requires patient-specific cell manufacturing, which unavoidably results in patient-to-patient variability in the final cell product. Widespread implementation of this approach will further require progress in automation and miniaturization of cell manufacturing to meet the demand for CAR T cells. Furthermore, current approaches utilize randomly integrating vectors, including gamma-retroviral, lentiviral and transposons, which all result in semi-random integration and variable expression of the CAR owing to transgene variegation. Position effects may result in heterogeneous T cell function, transgene silencing and, potentially, insertional oncogenesis. Thus, the conjunction of autologous cell sourcing and random vector integration is prone to generating cell products with variable potency. Here we utilize gene editing to generate histocompatible T cell products with consistent and homogeneous CAR expression. Different tailored nucleases, including CRISPR/Cas9 system, Zinc Finger Nucleases or TAL effector nucleases (TALENs), have been previously used for gene disruption in a wide range of human cells including primary T cells. In some instances, these nucleases have been used to generate so-called “universal T cells” for allogeneic administration, by disrupting T cell receptor (TCR) or HLA class I expression, but viral vectors or the sleeping beauty transposon were used to deliver the CAR cDNA, all of which result in semi-random transgene integration and its downstream consequences. We present here a novel strategy for one-step generation of universal CAR T cells. We first compared the efficiency of TALEN and CRISPR/Cas9 to promote homologous recombination using AAV6 donor template in T cells and established conditions yielding more than 50% of universal CAR T cells combining target gene disruption and CAR insertion in a single single step. We molecularly confirmed the targeted integration of the CAR transgene, which results in highly homogeneous and stable CAR expression in human peripheral blood T cells. These T cells exhibited the same in vitro tumor lysis activity and proliferation than retrovirally transduced CAR T cells, which augur favorably for their in vivo anti-tumor activity. Deep sequencing analyses to evaluate off-target effects of the nucleases and random AAV integration are in progress, as are in vivo experiments comparing the anti-tumor activity and graft-versus-host disease potential of edited T cells vs conventional CAR T cells. The process we describe here, which combines the scalability of universal T cell manufacturing with the uniformity and safety of targeted CAR gene integration, should be useful for the development of off-the-shelf CAR therapy.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.