52 results on '"Ramzey Abujarour"'
Search Results
2. Quadruple gene-engineered natural killer cells enable multi-antigen targeting for durable antitumor activity against multiple myeloma
- Author
-
Frank Cichocki, Ryan Bjordahl, Jodie P. Goodridge, Sajid Mahmood, Svetlana Gaidarova, Ramzey Abujarour, Zachary B. Davis, Aimee Merino, Katie Tuininga, Hongbo Wang, Akhilesh Kumar, Brian Groff, Alec Witty, Greg Bonello, Janel Huffman, Thomas Dailey, Tom T. Lee, Karl-Johan Malmberg, Bruce Walcheck, Uta Höpken, Armin Rehm, Bahram Valamehr, and Jeffrey S. Miller
- Subjects
Science - Abstract
The use of chimeric antigen receptor modified immune cell therapeutics has improved the treatment of a range of tumours. Here the authors explore a dual-target iPSC-derived NK cell product as a potential therapeutic for the treatment of multiple myeloma.
- Published
- 2022
- Full Text
- View/download PDF
3. Platform for Induction and Maintenance of Transgene-free hiPSCs Resembling Ground State Pluripotent Stem Cells
- Author
-
Bahram Valamehr, Megan Robinson, Ramzey Abujarour, Betsy Rezner, Florin Vranceanu, Thuy Le, Amanda Medcalf, Tom Tong Lee, Michael Fitch, David Robbins, and Peter Flynn
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Cell banking, disease modeling, and cell therapy applications have placed increasing demands on hiPSC technology. Specifically, the high-throughput derivation of footprint-free hiPSCs and their expansion in systems that allow scaled production remains technically challenging. Here, we describe a platform for the rapid, parallel generation, selection, and expansion of hiPSCs using small molecule pathway inhibitors in stage-specific media compositions. The platform supported efficient and expedited episomal reprogramming using just OCT4/SOX2/SV40LT combination (0.5%–4.0%, between days 12 and 16) in a completely feeder-free environment. The resulting hiPSCs are transgene-free, readily cultured, and expanded as single cells while maintaining a homogeneous and genomically stable pluripotent population. hiPSCs generated or maintained in the media compositions described exhibit properties associated with the ground state of pluripotency. The simplicity and robustness of the system allow for the high-throughput generation and rapid expansion of a uniform hiPSC product that is applicable to industrial and clinical-grade use.
- Published
- 2014
- Full Text
- View/download PDF
4. Dual antigen–targeted off-the-shelf NK cells show durable response and prevent antigen escape in lymphoma and leukemia
- Author
-
Frank Cichocki, Jodie P. Goodridge, Ryan Bjordahl, Sajid Mahmood, Zachary B. Davis, Svetlana Gaidarova, Ramzey Abujarour, Brian Groff, Alec Witty, Hongbo Wang, Katie Tuininga, Behiye Kodal, Martin Felices, Greg Bonello, Janel Huffman, Thomas Dailey, Tom T. Lee, Bruce Walcheck, Bahram Valamehr, and Jeffrey S. Miller
- Subjects
Killer Cells, Natural ,Leukemia ,Neoplasms ,Immunology ,Humans ,Cell Biology ,Hematology ,Antigenic Drift and Shift ,Biochemistry - Abstract
Substantial numbers of B cell leukemia and lymphoma patients relapse due to antigen loss or heterogeneity after anti-CD19 chimeric antigen receptor (CAR) T cell therapy. To overcome antigen escape and address antigen heterogeneity, we engineered induced pluripotent stem cell-derived NK cells to express both an NK cell-optimized anti-CD19 CAR for direct targeting and a high affinity, non-cleavable CD16 to augment antibody-dependent cellular cytotoxicity. In addition, we introduced a membrane-bound IL-15/IL-15R fusion protein to promote in vivo persistence. These engineered cells, termed iDuo NK cells, displayed robust CAR-mediated cytotoxic activity that could be further enhanced with therapeutic antibodies targeting B cell malignancies. In multiple in vitro and xenogeneic adoptive transfer models, iDuo NK cells exhibited robust anti-lymphoma activity. Furthermore, iDuo NK cells effectively eliminated both CD19+ and CD19− lymphoma cells and displayed a unique propensity for targeting malignant cells over healthy cells that expressed CD19, features not achievable with anti-CAR19 T cells. iDuo NK cells combined with therapeutic antibodies represent a promising approach to prevent relapse due to antigen loss and tumor heterogeneity in patients with B cell malignancies.
- Published
- 2022
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
6. 304 Off-the-shelf iPSC-derived CAR-T cells containing seven functional edits overcome antigen heterogeneity, improve trafficking, and withstand immunosuppression associated with failed tumor treatment
- Author
-
Martin Hosking, Soheila Shirinbak, Bishwas Shrestha, Joy Grant, Kyla Omilusik, Hannah Keegan, Demetrio Cardenas, Angela Gentile, Shilpi Chandra, Lorraine Loter, Lexe Linderhof, Nicholas Brookhouser, Stephanie Kennedy, Francisco Martinez, Loraine Campanati, Chris Ecker, Xu Yuan, Karina Palomares, Yi-Shin Lai, Lauren Fong, Yijia Pan, Shohreh Sikaroodi, Mark Jelcic, Philip Chu, Amit Mehta, Layton Smith, Eigen Peralta, Tom Lee, Ramzey Abujarour, Raedun Clarke, and Bob Valamehr
- Published
- 2022
7. Combined Genetic Ablation of CD54 and CD58 in CAR Engineered Cytotoxic Lymphocytes Effectively Averts Allogeneic Immune Cell Rejection
- Author
-
Quirin Hammer, Karlo Perica, Rina M Mbofung, Hanna van Ooijen, Erika Varady, Mark Jelcic, Yijia Pan, Pouria Momayyezi, Brian Groff, Ramzey Abujarour, Silje Zandstra Krokeide, Tom Lee, Alan M Williams, Jode P Goodridge, Bahram Valamehr, Bjorn Onfelt, Michel Sadelain, and Karl-Johan Malmberg
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
8. Alloimmune Defense Receptor Harnesses Host Immune Cell Activation to Potentiate Functional Persistence and Anti-Tumor Activity of Off-the-Shelf, Cell-Based Cancer Therapy
- Author
-
Alan M Williams, Ken L Hayama, Yijia Pan, Brian Groff, Rina M Mbofung, Amber Chang, Christine Chen, Lauren Fong, Nicholas Brookhouser, Berhan Mandefro, Ramzey Abujarour, Tom Lee, Maksim Mamonkin, Raedun Clarke, Ryan Bjordahl, Jode P Goodridge, and Bahram Valamehr
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
9. Abstract 2828: A novel synthetic stealth receptor that redirects host immune cell alloreactivity and potentiates functional persistence of adoptively transferred off-the-shelf cell-based cancer therapy
- Author
-
Alan M. Williams, Ken Hayama, Yijia Pan, Brian Groff, Rina Mbofung, Lauren Fong, Nicholas Brookhouser, Berhan Mandefro, Ramzey Abujarour, Tom Lee, Quirin Hammer, Karl-Johan Malmberg, Maksim Mamonkin, Ryan Bjordahl, Jode Goodridge, and Bahram Valamehr
- Subjects
Cancer Research ,Oncology - Abstract
Chimeric antigen receptor (CAR) T-cell therapies have revolutionized the treatment of hematologic malignancies and have shown significant potential in solid tumor indications. However, logistical complexities associated with patient-specific CAR T-cell therapies often limit broad accessibility. Many of these challenges can be overcome with an allogeneic cellular product, but immune cell-mediated rejection of allogeneic cellular therapies remains a significant concern. Both allogeneic and autologous cell therapies currently rely on lymphodepleting conditioning to modulate the immune system and create greater access to homeostatic cytokines. However, protracted lympho-conditioning has been associated with poor immune reconstitution and increased susceptibility to opportunistic infections. Therefore, an ideal allogeneic cell therapy would be able to avoid immune rejection while reducing or eliminating the need for chemotherapeutic conditioning to deplete host lymphocytes. To address many of these challenges, we engineered our novel alloimmune defense receptor (ADR) that targets 41BB+ alloreactive immune cells while providing a CD3z signaling boost into our off-the-shelf iPSC derived NK cells expressing anti-CD19 CAR (CAR iNK). The ability of ADR+ CAR iNK cells to resist alloimmune rejection was tested by co-culturing ADR+ CAR iNK cells with allogeneic PBMCs from ten donors in a mixed lymphocyte reaction assay. Notably, ADR+ CAR iNK cells co-cultured with allogeneic PBMCs persisted to similar levels as the PBMC-free culture while ADR negative CAR iNK cells were eliminated when co-cultured with allogeneic PBMCs. Furthermore, all PBMC donors screened in ADR+ CAR iNK cell co-cultures showed ablation of reactive 41BB+ NK and T cells, with non-activated T cells remaining intact. CAR iNK cells +/- ADR were then compared in a Nalm6 disseminated in vivo model for anti-tumor efficacy. ADR+ CAR iNK cells and their ADR negative counterparts were found to equivalently control tumor. Building on this tumor model, we co-infused allogeneic T cells to mimic an immuno-competent setting. The data demonstrated that ADR negative CAR iNK cells were depleted and were unable to control tumor growth while significant levels of allogeneic T cells persisted. In contrast, ADR+ CAR iNK cells were able to resist allogeneic T cell attack, control tumor, and persist when compared to the ADR negative control. Collectively, our preliminary data suggest that ADR-armed CAR iNK cells withstand immune cell-mediated rejection with uncompromised effector function. We are actively developing models to confirm our initial finding that ADR+ effector cells also benefit from their engagement with alloreactive cells in immuno-competent settings to promote enhanced anti-tumor responses, proliferation, and persistence. Citation Format: Alan M. Williams, Ken Hayama, Yijia Pan, Brian Groff, Rina Mbofung, Lauren Fong, Nicholas Brookhouser, Berhan Mandefro, Ramzey Abujarour, Tom Lee, Quirin Hammer, Karl-Johan Malmberg, Maksim Mamonkin, Ryan Bjordahl, Jode Goodridge, Bahram Valamehr. A novel synthetic stealth receptor that redirects host immune cell alloreactivity and potentiates functional persistence of adoptively transferred off-the-shelf cell-based cancer therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2828.
- Published
- 2022
10. Harnessing features of adaptive NK cells to generate iPSC-derived NK cells for enhanced immunotherapy
- Author
-
Svetlana Gaidarova, Hongbo Wang, Frank Cichocki, Jeffrey S. Miller, Bruce Walcheck, Brian Hancock, Miguel Meza, Ryan Bjordahl, Karrune Woan, Bruce R. Blazar, Bahram Valamehr, Janel Huffman, Melissa Khaw, Karl J. Malmberg, Ramzey Abujarour, Hansol Kim, Moyar Q. Ge, Bin Zhang, Thomas Dailey, John Goulding, Martin Felices, Cheng-Ying Wu, Tom Tong Lee, Yenan T. Bryceson, Greg Bonello, Laura Bendzick, Sajid Mahmood, Behiye Kodal, Zachary Davis, Paul Rogers, and Katie Tuininga
- Subjects
medicine.medical_treatment ,Induced Pluripotent Stem Cells ,Fc receptor ,CD38 ,Immunotherapy, Adoptive ,Article ,Natural killer cell ,Immune system ,Cell Line, Tumor ,Neoplasms ,Genetics ,medicine ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Gene Editing ,biology ,Interleukin ,Cell Biology ,Immunotherapy ,Cell biology ,Killer Cells, Natural ,medicine.anatomical_structure ,Cytokine ,biology.protein ,Molecular Medicine ,Multiple Myeloma - Abstract
Select subsets of immune effector cells have the greatest propensity to mediate antitumor responses. However, procuring these subsets is challenging, and cell-based immunotherapy is hampered by limited effector-cell persistence and lack of on-demand availability. To address these limitations, we generated a triple-gene-edited induced pluripotent stem cell (iPSC). The clonal iPSC line was engineered to express a high affinity, non-cleavable version of the Fc receptor CD16a and a membrane-bound interleukin (IL)-15/IL-15R fusion protein. The third edit was a knockout of the ecto-enzyme CD38, which hydrolyzes NAD+. Natural killer (NK) cells derived from these uniformly engineered iPSCs, termed iADAPT, displayed metabolic features and gene expression profiles mirroring those of cytomegalovirus-induced adaptive NK cells. iADAPT NK cells persisted in vivo in the absence of exogenous cytokine and elicited superior antitumor activity. Our findings suggest that unique subsets of the immune system can be modeled through iPSC technology for effective treatment of patients with advanced cancer. acceptedVersion
- Published
- 2021
11. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti-PD-1 therapy
- Author
-
Frank Cichocki, Martin Felices, Svetlana Gaidarova, Ryan Bjordahl, Bahram Valamehr, David Robbins, Laurel Stokely, Jeffrey S. Miller, Betsy Rezner, Hongbo Wang, Bruce R. Blazar, Moyar Q. Ge, Raedun Clarke, Ramzey Abujarour, Laura Bendzick, Megan Robinson, Tom Tong Lee, Zachary Davis, Paul Rogers, Dan S. Kaufman, Katie Tuininga, and Sajid Mahmood
- Subjects
medicine.medical_treatment ,T cell ,T-Lymphocytes ,Cell ,Induced Pluripotent Stem Cells ,Programmed Cell Death 1 Receptor ,Medical and Health Sciences ,Article ,Cell therapy ,Vaccine Related ,Immune system ,Neoplasms ,medicine ,Killer Cells ,Humans ,Induced pluripotent stem cell ,Cancer ,biology ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Chemistry ,General Medicine ,Immunotherapy ,Biological Sciences ,Stem Cell Research ,Killer Cells, Natural ,medicine.anatomical_structure ,Cytokine ,biology.protein ,Cancer research ,Natural ,Immunization ,Antibody ,Biotechnology - Abstract
The development of immunotherapeutic monoclonal antibodies targeting checkpoint inhibitory receptors, such as programmed cell death 1 (PD-1), or their ligands, such as PD-L1, has transformed the oncology landscape. However, durable tumor regression is limited to a minority of patients. Therefore, combining immunotherapies with those targeting checkpoint inhibitory receptors is a promising strategy to bolster antitumor responses and improve response rates. Natural killer (NK) cells have the potential to augment checkpoint inhibition therapies, such as PD-L1/PD-1 blockade, because NK cells mediate both direct tumor lysis and T cell activation and recruitment. However, sourcing donor-derived NK cells for adoptive cell therapy has been limited by both cell number and quality. Thus, we developed a robust and efficient manufacturing system for the differentiation and expansion of high-quality NK cells derived from induced pluripotent stem cells (iPSCs). iPSC-derived NK (iNK) cells produced inflammatory cytokines and exerted strong cytotoxicity against an array of hematologic and solid tumors. Furthermore, we showed that iNK cells recruit T cells and cooperate with T cells and anti-PD-1 antibody, further enhancing inflammatory cytokine production and tumor lysis. Because the iNK cell derivation process uses a renewable starting material and enables the manufacturing of large numbers of doses from a single manufacture, iNK cells represent an "off-the-shelf" source of cells for immunotherapy with the capacity to target tumors and engage the adaptive arm of the immune system to make a "cold" tumor "hot" by promoting the influx of activated T cells to augment checkpoint inhibitor therapies.
- Published
- 2020
12. Pluripotent stem cell–derived NK cells with high-affinity noncleavable CD16a mediate improved antitumor activity
- Author
-
Jianming Wu, Ramzey Abujarour, Pei Fang Tsai, Dan S. Kaufman, Svetlana Gaidarova, Robert Blum, Gregory B. Bonello, Bruce Walcheck, Bahram Valamehr, Paul Rogers, Ryan Bjordahl, Huang Zhu, Jeffrey S. Miller, and Tom Tong Lee
- Subjects
Lymphoma ,Hematopoiesis and Stem Cells ,Fc receptor ,Mice, SCID ,Cardiorespiratory Medicine and Haematology ,Biochemistry ,Cell therapy ,Mice ,Antineoplastic Agents, Immunological ,Mice, Inbred NOD ,Receptors ,Monoclonal ,Killer Cells ,Induced pluripotent stem cell ,Cancer ,Antibody-dependent cell-mediated cytotoxicity ,Ovarian Neoplasms ,Tumor ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,biology ,Chemistry ,Antibodies, Monoclonal ,Hematology ,Killer Cells, Natural ,Immunological ,Natural ,Female ,Development of treatments and therapeutic interventions ,Antibody ,Biotechnology ,Lymphoma, B-Cell ,IgG ,medicine.drug_class ,Clinical Sciences ,Immunology ,Induced Pluripotent Stem Cells ,Antineoplastic Agents ,SCID ,Monoclonal antibody ,Antibodies ,Cell Line ,Paediatrics and Reproductive Medicine ,Rare Diseases ,Antigen ,Cell Line, Tumor ,medicine ,Animals ,Humans ,CD20 ,Antigens ,Stem Cell Research - Induced Pluripotent Stem Cell ,5.2 Cellular and gene therapies ,Receptors, IgG ,B-Cell ,Antibody-Dependent Cell Cytotoxicity ,Cell Biology ,Stem Cell Research ,Antigens, CD20 ,Cell culture ,biology.protein ,Cancer research ,Inbred NOD - Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is a key effector mechanism of natural killer (NK) cells that is mediated by therapeutic monoclonal antibodies (mAbs). This process is facilitated by the Fc receptor CD16a on human NK cells. CD16a appears to be the only activating receptor on NK cells that is cleaved by the metalloprotease a disintegrin and metalloproteinase-17 upon stimulation. We previously demonstrated that a point mutation of CD16a prevents this activation-induced surface cleavage. This noncleavable CD16a variant is now further modified to include the high-affinity noncleavable variant of CD16a (hnCD16) and was engineered into human induced pluripotent stem cells (iPSCs) to create a renewable source for human induced pluripotent stem cell–derived NK (hnCD16-iNK) cells. Compared with unmodified iNK cells and peripheral blood–derived NK (PB-NK) cells, hnCD16-iNK cells proved to be highly resistant to activation-induced cleavage of CD16a. We found that hnCD16-iNK cells were functionally mature and exhibited enhanced ADCC against multiple tumor targets. In vivo xenograft studies using a human B-cell lymphoma demonstrated that treatment with hnCD16-iNK cells and anti-CD20 mAb led to significantly improved regression of B-cell lymphoma compared with treatment utilizing anti-CD20 mAb with PB-NK cells or unmodified iNK cells. hnCD16-iNK cells, combined with anti-HER2 mAb, also mediated improved survival in an ovarian cancer xenograft model. Together, these findings show that hnCD16-iNK cells combined with mAbs are highly effective against hematologic malignancies and solid tumors that are typically resistant to NK cell–mediated killing, demonstrating the feasibility of producing a standardized off-the-shelf engineered NK cell therapy with improved ADCC properties to treat malignancies that are otherwise refractory.
- Published
- 2020
13. Off-the-Shelf, Multiplexed-Engineered iPSC-Derived NK Cells Mediate Potent Multi-Antigen Targeting of B-Cell Malignancies with Reduced Cytotoxicity Against Healthy B Cells
- Author
-
Frank Cichocki, Peter Szabo, Janel Huffman, Thomas Dailey, Jode P Goodridge, Sarah Cooley, Behiye Kodal, Ramzey Abujarour, Thomas H. Lee, Hongbo Wang, Bahram Valamehr, Ryan Bjordahl, Wong Lilly L, Svetlana Gaidarova, Zachary Davis, Paul Rogers, Sajid Mahmood, Katie Tuininga, Martin Felices, Greg Bonello, and Jeffrey S. Miller
- Subjects
medicine.anatomical_structure ,Antigen Targeting ,Chemistry ,Immunology ,medicine ,Cancer research ,Off the shelf ,Cell Biology ,Hematology ,Cytotoxicity ,Biochemistry ,B cell - Abstract
Treatments for B-cell malignancies have improved over the past several decades with clinical application of the CD20-specific antibody rituximab and chimeric antigen receptor (CAR) T cells targeting CD19. Despite the success of these therapies, loss of CD20 after rituximab treatment has been reported in leukemia and lymphoma patients. Additionally, up to 50% of all patients receiving anti-CD19 CAR T-cell therapy relapse within the first year with many of those patients exhibiting CD19 loss. Thus, new therapeutic approaches are needed to address tumor antigen escape. Accordingly, we generated triple gene-modified iPSC-derived NK (iNK) cells, termed "iDuo" NK cells, tailored to facilitate multi-antigen targeting. The iPSC line was clonally engineered to express high-affinity, non-cleavable CD16a (hnCD16), an anti-CD19 CAR optimized for NK cell signaling, and a membrane-bound IL-15/IL-15R fusion (IL-15RF) molecule to enhance NK cell persistence (Fig. 1A). To model antigen escape, we generated CD19 knockout AHR77 lymphoma cells alongside wild type AHR77 cells (both CD20 +) as targets in cytotoxicity assays. Activated peripheral blood NK (PBNK) cells, non-transduced iNK cells, and iDuo NK cells were tested as effectors. Unlike PBNK cells or non-transduced iNK cells, iDuo NK cells efficiently eliminated wild type AHR77 cells with or without the addition of rituximab at all tested E:T ratios. Similarly, iDuo NK cells in combination with rituximab were uniquely able to efficiently eliminate CD19 KO AHR77 cells due to enhanced antibody-dependent cellular cytotoxicity (ADCC) driven by hnCD16 (Fig. 1B-E). Cytotoxicity mediated by iDuo NK cells was also evaluated using primary chronic lymphocytic leukemia (CLL) cells. Compared to expanded PBNK cells and non-transduced iNK cells, only iDuo NK cells (in the absence of rituximab) were able to kill primary CLL cells (Fig. 1F). Expression of IL-15RF by iDuo NK cells uniquely supports in vitro expansion without the need for cytokine supplementation. To determine whether IL-15RF supports in vivo persistence of iDuo NK cells, CD19 CAR iNK cells (lacking IL-15RF) and iDuo NK cells were injected into NSG mice without the addition of cytokines or CD19 antigen availability. iDuo NK cell numbers peaked within a week after injection and persisted at measurable levels for ~5 weeks, in marked contrast to CD19 CAR iNK cell numbers that were undetectable throughout (Fig. 1G). To evaluate the in vivo function of iDuo NK cells, NALM6 leukemia cells were engrafted into NSG mice. Groups of mice received tumor alone or were treated with 3 doses of thawed iDuo NK cells. iDuo NK cells alone were highly effective in this model as evidenced by complete survival of mice in the treatment group (Fig. 1H). To assess iDuo NK cells in a more aggressive model, Raji lymphoma cells were engrafted, and groups of mice received rituximab alone, iDuo NK cells alone, or iDuo NK cells plus rituximab. Mice given the combination of iDuo NK cells and rituximab provided extended survival compared to all other arms in the aggressive disseminated Raji lymphoma xenograft model (Fig. 1I). One disadvantage of anti-CD19 CAR T cells is their inability to discriminate between healthy and malignant B cells. Because NK cells express inhibitory receptors that enable "self" versus "non-self" discrimination, we reasoned that iDuo NK cells could have higher cytotoxicity against tumor cells relative to healthy B cells. To address this, we labeled Raji cells, CD19 + B cells from healthy donor peripheral blood mononuclear cells (PBMCs) and CD19 - PBMCs. Labeled populations of cells were co-cultured with iDuo NK cells, and specific killing was analyzed. As expected, iDuo NK cells did not target CD19 - PBMCs. Intriguingly, iDuo NK cells had much higher cytotoxic activity against Raji cells compared to primary CD19 + B cells, suggesting a preferential targeting of malignant B cells compared to healthy B cells. Together, these results demonstrate the potent multi-antigen targeting capability and in vivo antitumor function of iDuo NK cells. Further, these data suggest that iDuo NK cells may have an additional advantage over anti-CD19 CAR T cells by discriminating between healthy and malignant B cells. The first iDuo NK cell, FT596, is currently being tested in a Phase I clinical trial (NCT04245722) for the treatment of B-cell lymphoma. Figure 1 Figure 1. Disclosures Cichocki: Gamida Cell: Research Funding; Fate Therapeutics, Inc: Patents & Royalties, Research Funding. Bjordahl: Fate Therapeutics: Current Employment. Gaidarova: Fate Therapeutics, Inc: Current Employment. Abujarour: Fate Therapeutics, Inc.: Current Employment. Rogers: Fate Therapeutics, Inc: Current Employment. Huffman: Fate Therapeutics, Inc: Current Employment. Lee: Fate Therapeutics, Inc: Current Employment. Szabo: Fate Therapeutics, Inc: Current Employment. Wong: BMS: Current equity holder in publicly-traded company; Fate Therapeutics, Inc: Current Employment. Cooley: Fate Therapeutics, Inc: Current Employment. Valamehr: Fate Therapeutics, Inc.: Current Employment. Miller: Magenta: Membership on an entity's Board of Directors or advisory committees; ONK Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Vycellix: Consultancy; GT Biopharma: Consultancy, Patents & Royalties, Research Funding; Fate Therapeutics, Inc: Consultancy, Patents & Royalties, Research Funding; Sanofi: Membership on an entity's Board of Directors or advisory committees; Wugen: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
14. Clinical Manufacture of FT819: Use of a Clonal Multiplexed-Engineered Master Induced Pluripotent Stem Cell Line to Mass Produce Off-the-Shelf CAR T-Cell Therapy
- Author
-
Eric Sung, Jason ORourke, Raedun Clarke, Thomas H. Lee, Isabelle Riviere, Bi-Huei Yang, Rebecca Magdaleno, Gloria Hsia, Dell Farnan, Sjoukje J. C. van der Stegen, Stephanie Moreno, Chia-Wei Chang, Brigitte Senechal, Xu Yuan, Alma Gutierrez, Mark Plavsic, Meghan Eberhart, Bahram Valamehr, Abubakar Jalloh, Xiuyan Wang, Helena Shaked, Jerome Bressi, Yi-Shin Lai, Betsy Rezner, Devanjan S. Sikder, and Ramzey Abujarour
- Subjects
Immunology ,Off the shelf ,CAR T-cell therapy ,Cell Biology ,Hematology ,Line (text file) ,Biology ,Induced pluripotent stem cell ,Biochemistry ,Cell biology - Abstract
FT819 is a first-of-kind, allogeneic, off-the-shelf CAR T-cell therapy derived from a clonal master induced pluripotent stem cell (iPSC) line precisely engineered to insert a novel 1XX anti-CD19 chimeric antigen receptor (CAR) under the regulation of the T-cell receptor alpha constant (TRAC) locus for optimized control of anti-tumor activity and to completely delete T-cell receptor (TCR) expression to eliminate the potential of graft-versus-host disease (GvHD). Unlike conventional allogeneic CAR T-cell therapies which require repeatedly sourcing of T cells from various donors as the starting material, the use of a clonal master engineered iPSC line serves as a renewable starting cell source and ensures routine mass production of a uniformly engineered, homogenous CAR T-cell product for broad patient access. T cell-derived iPSCs were generated using a proprietary non-integrating cellular reprogramming system and genetically modified to integrate a novel anti-CD19 1XX CAR into both alleles of the TRAC gene. After single cell subcloning, each engineered iPSC clone was screened for multiple critical quality attributes including pluripotency, identity, genomic stability, cassette integration, on/off-target integration, T-cell differentiation propensity, and CAR T-cell function. Accordingly, the ideal single cell-derived engineered iPSC clone was selected as the clonal master iPSC line for FT819 and was converted into a master cell bank (MCB). The iPSC MCB serves as a renewable source for the routine GMP manufacture of FT819 drug product. The FT819 production process consists of three stages: 1) generation of CD34-expressing hematopoietic progenitor cells from iPSCs (>90% CD34+ cells post enrichment); 2) lineage-specification to T cells followed by T-cell expansion (>5e5 fold expansion); and 3) fill/finish and cryopreservation of the drug product. As an example, in an initial small-scale manufacturing campaign, a total of 2.5 × 10 10 FT819 CAR T-cells were generated and filled and finished starting from one vial of the MCB. The FT819 drug product was tested on safety, identity, purity, and potency. The final product was comprised of CD45+CD7+ lymphocytes (>99%), with homogeneous CAR expression (>99% CAR+) and lacking expression of TCRαβ (not detected) on the cell surface. Importantly, there were no residual iPSCs detected in the FT819 drug product. The FT819 drug product exhibited potent and consistent effector function against NALM6 leukemia cells. The FT819 drug product is currently being used in a landmark Phase I study (NCT04629729), the first-ever iPSC-derived T-cell therapy to undergo clinical investigation, for the treatment of patients with relapsed/refractory B-cell lymphoma, chronic lymphocytic leukemia and precursor B-cell acute lymphoblastic leukemia. In summary, FT819 is a first-of-kind, off-the-shelf, CAR T-cell therapy uniquely derived from a clonal multiplexed-engineered master iPSC line. The novel manufacturing paradigm enables mass production of a uniformly engineered, homogenous cell therapy product that is available on-demand for broad patient access. A multi-center Phase 1 study of FT819 is currently ongoing for the treatment of B-cell malignancies. Key Words: cancer immunotherapy, cell therapy, CAR-T, CD19, allogeneic, induced pluripotent stem cell, iPSC, clonal master iPSC line, engineered, off-the-shelf, cGMP, production, manufacturing, FT819 Disclosures Yuan: Fate Therapeutics, Inc.: Current Employment. Clarke: Fate Therapeutics, Inc.: Current Employment. Lai: Fate Therapeutics, Inc.: Current Employment. Chang: Fate Therapeutics, Inc.: Current Employment. Yang: Fate Therapeutics, Inc.: Current Employment. Hsia: Fate Therapeutics, Inc.: Current Employment. Abujarour: Fate Therapeutics, Inc.: Current Employment. Lee: Fate Therapeutics, Inc.: Current Employment. van der Stegen: Fate Therapeutics, Inc.: Current Employment. Shaked: Fate Therapeutics, Inc.: Current Employment. Jalloh: Fate Therapeutics, Inc.: Current Employment. Moreno: Fate Therapeutics, Inc.: Current Employment. ORourke: Fate Therapeutics, Inc.: Current Employment. Sung: Fate Therapeutics, Inc.: Current Employment. Gutierrez: Fate Therapeutics, Inc.: Current Employment. Rezner: Fate Therapeutics, Inc.: Current Employment. Eberhart: Fate Therapeutics, Inc.: Current Employment. Magdaleno: Fate Therapeutics, Inc.: Current Employment. Farnan: Fate Therapeutics, Inc.: Current Employment. Plavsic: Fate Therapeutics, Inc.: Current Employment. Bressi: Fate Therapeutics, Inc.: Current Employment. Rivière: Centre for Commercialization of Cancer Immunotherapy: Other: Provision of Services; Fate Therapeutics: Other: Provision of Services, Patents & Royalties; The Georgia Tech Research Corporation (GTRC): Other: Provision of Services (uncompensated); FloDesign Sonics: Other: Provision of Services; Juno Therapeutics: Patents & Royalties. Valamehr: Fate Therapeutics, Inc.: Current Employment.
- Published
- 2021
15. A Novel Stealth Strategy That Activates Adoptively Transferred Allogeneic Immune Cells and Avoids Rejection for Off-the-Shelf Cell-Based Cancer Therapy
- Author
-
Ken Hayama, Ryan Bjordahl, Nicholas Brookhouser, Lauren Fong, Jode P Goodridge, Berhan Mandefro, Brian Groff, Bahram Valamehr, Karl-Johan Malmberg, Ramzey Abujarour, Alan M Williams, Thomas H. Lee, Maksim Mamonkin, Quirin Hammer, Rina M Mbofung, and Yijia Pan
- Subjects
Immune system ,business.industry ,Immunology ,Cancer research ,Cancer therapy ,Off the shelf ,Medicine ,Cell Biology ,Hematology ,business ,Biochemistry ,Cell based - Abstract
Chimeric antigen receptor (CAR) T-cell therapies have revolutionized the treatment of hematologic malignancies, however, logistical complexities associated with patient-specific CAR T-cell therapies often limit broad accessibility to patients. Many of these challenges can be overcome with an allogeneic cellular product that is available off-the-shelf, and overcoming immune cell-mediated rejection of allogeneic cell therapy is an area of significant research. Conditioning chemotherapies, which are commonly administered with CAR T-cell therapy, suppress a patient's immune system and may create a suitable window of activity for allogeneic cell therapies to elicit clinical response. However, protracted lympho-conditioning has been associated with poor immune reconstitution and increased susceptibility to opportunistic infections. Deletion of human leukocyte antigen (HLA) surface expression is known to abrogate T-cell alloreactivity, but deletion of class I HLA must be combined with other immune-modulating strategies to avoid NK cell-mediated recognition. To this end, allogeneic models combining class I HLA deletion with NK cell inhibitory molecules, such as HLA-E and CD47, have been shown to abrogate NK cell reactivity in mouse models. However, since HLA-E is the canonical activator of NKG2C, a dominant activating receptor found on human NK cells, and since the ligand for CD47, SIRPα, is known to be expressed on macrophages and dendritic cells and not on human NK cells, the observed effects of these immune-modulating strategies may not translate into the patient-treatment setting. In assessment of their protective effects in a defined human system, we found that HLA-E or CD47 overexpression on class I HLA null human cells offer only partial protection in evading various human NK cell compartments. We found that class I HLA-null K562 cells engineered to over-express CD47 were ineffective in inhibiting NK cells (0 to 7% inhibition). Separately, K562 cells engineered to over-express HLA-E, while effective in inhibiting NKG2A+ NK cells (90.2% +/- 3.7% inhibition), were unable to completely inhibit CD56 dim NK cells (33.2% +/- 29.6% inhibition) and not only failed, but instead activated, NKG2C+ NK cells (167% +/- 69% activation). Our data highlight the limitations of engineered CD47 and HLA-E modalities in suppressing broad populations of NK cells in clinically relevant settings. We therefore evaluated expression of the alloimmune defense receptor (ADR) that uniquely targets alloreactive immune cells (Mo et al. Nat Biotechnol 2021). We have shown that the expression of ADR has the potential to evade host immune cells without the need for further genetic editing such as class I HLA deletion. To determine its applicability for off-the-shelf cell therapy, ADR expression was engineered into induced pluripotent stem cells (iPSCs) and iPSC-derived CAR-NK (CAR-iNK) cells were generated. CAR-iNK cells carrying the ADR modality (ADR+ CAR-iNK cells) showed normal patterns of differentiation (>99% CD56+ with co-expression of NK cell receptors such as NKG2D, NKp30 and NKp46), suggesting that ADR expression did not disrupt hematopoiesis or the expansion of iNK cells. Additionally, ADR+ and ADR-negative CAR-iNK cells produced similar cytotoxicity against tumor cells. We next tested the ability of ADR to provide resistance to alloimmune rejection by coculturing ADR+ CAR-iNK cells with allogeneic pBMCs in a mixed lymphocyte reaction (MLR) assay. Notably, ADR+ CAR-iNK cells maintained durable persistence throughout the entire duration of the MLR assay and suppressed the expansion of alloreactive T- and NK-cells in comparison to the control arm (Figure 1). Collectively, initial preclinical studies suggest that ADR-modified CAR-iNK cells resist host immune cell rejection, while eliciting a durable anti-tumor response. Our preliminary data show evidence toward a promising off-the-shelf solution for elimination of broad pools of alloreactive T- and NK- effector cells in the clinical setting without the need for lympho-depleting conditioning or genetic editing strategies. Figure 1 Figure 1. Disclosures Williams: Fate Therapeutics: Current Employment. Abujarour: Fate Therapeutics, Inc.: Current Employment. Lee: Fate Therapeutics, Inc.: Current Employment. Malmberg: Merck: Research Funding; Vycellix: Consultancy; Fate Therapeutics: Consultancy, Research Funding. Mamonkin: Beam Therapeutics: Other: Licensing payments; Fate Therapeutics: Other: Licensing payments; Allogene Therapeutics: Consultancy, Other: Licensing payments; Xenetic Biosciences: Consultancy, Membership on an entity's Board of Directors or advisory committees. Bjordahl: Fate Therapeutics: Current Employment. Valamehr: Fate Therapeutics, Inc.: Current Employment.
- Published
- 2021
16. 117 FT536 Path to IND: Ubiquitous targeting of solid tumors with an off-the-shelf, first-of-kind MICA/B-specific CAR-iNK cellular immunotherapy
- Author
-
Chia-Wei Chang, Svetlana Gaidarova, Natalie Marquez-Solorzano, Sajid Mahmood, Robert Blum, Yijia Pan, Raedun Clarke, Rina M Mbofung, Soheila Shirinbak, Christine Chen, Fernanda Rodrigues Cugola, Bi-Huei Yang, Kai W. Wucherpfennig, Janel Huffman, Andrew Burns, Lucas Ferrari de Andrade, Antonio Fernandez-Perez, John Goulding, Martin Hosking, Karina Palomares, Mochtar Pribadi, Lauren Fong, Brian Groff, Thomas Dailey, Tom Lee, Ramzey Abujarour, Wen-I Yeh, Joy Grant, Moyar Ge, Samvel Nazaretyan, Miguel Meza, Jason O’Rouke, Paul Rogers, Nicholas Brookhouser, Bryan Hancock, Pei-Fang Tsai, Riya Kanherkar, Hui-yi Chu, Bahram Valamehr, Ryan Bjordahl, Jerome Bressi, Shohreh Sikaroodi, and Aidan Keefe
- Subjects
Pharmacology ,Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,Chemistry ,medicine.drug_class ,Immunology ,Cell ,Monoclonal antibody ,Chimeric antigen receptor ,Cell therapy ,medicine.anatomical_structure ,Oncology ,Antigen ,Cancer cell ,Cancer research ,medicine ,Molecular Medicine ,Immunology and Allergy ,Induced pluripotent stem cell - Abstract
BackgroundChimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer treatment, but it is associated with significant dose-limiting toxicities, restricted tumor targeting (limited by specific antigen expression), and, notably, a lack of multi-antigen targeting capability to mitigate tumor associated immune evasion and heterogeneity. Furthermore, dysfunctional starting material, product inconsistency, and small manufacturing lot size limits the application and on-demand availability of CAR-T cell therapy.MethodsTo overcome these considerable limitations, we have developed FT536, a first-of-kind, induced pluripotent stem cell (iPSC)-derived NK (iNK) cell with a novel CAR that ubiquitously targets cancer cells through canonical stress ligand recognition. We have previously reported FT536 recognizes the conserved α3 domain of the pan-tumor associated antigens MICA and MICB (MICA/B), and is derived from a renewable master iPSC line that contains multiplexed genetic edits to enhance effector cell functionality, persistence, and multi-antigen targeting capabilities via high affinity non cleavable CD16 (hnCD16) mediated antibody dependent cellular cytotoxicity (ADCC). Here we preview the nonclinical study for the investigational new drug (IND) application for FT536.ResultsUtilizing a manufacturing process analogous to pharmaceutical drug product development, we demonstrate FT536 can be consistently and uniformly produced with a greater than 4x10E7 fold cellular expansion per manufacturing campaign. Furthermore, FT536 can be cryopreserved at clinical scale to support off-the-shelf clinical application, with rapid product thaw and immediate patient infusion in an out-patient setting. Functional evaluation demonstrated that FT536 uniquely possesses potent and persistent antigen specific cytolytic activity against an array of solid and hematological tumor lines. Through its hnCD16 modality, FT536 can be utilized in combination with monoclonal antibodies to provide multi-antigen targeting capabilities and in conjunction with chemotherapeutics and/or radiation that augment surface MICA/B expression. In addition, directly thawed and infused FT536 demonstrated significant tumor growth inhibition in multiple solid and liquid in vivo xenograft models, in which tumor control was further enhanced in combination with a therapeutic antibody (figure 1). Finally, ongoing studies utilizing a lung adenocarcinoma model have highlighted the sustained persistence of FT536 in lung tissue up to 33 days following a single dose infusion without the need for exogenous cytokine support.Abstract 117 Figure 1FT536 provides statistically significant in vivo anti-tumor activity which is enhanced in combination with ADCC active monoclonal antibody therapy. (A-B) FT536 significantly reduced the number of lung and liver (not shown) metastases compared to CAR negative iNK control cells in a murine metastatic melanoma model using B16-F10 cells engineered to overexpress human MICA. (C-D) FT536 alone, and in combination with Herceptin, demonstrate significant tumor growth inhibition (TGI) compared to Herceptin alone in an orthotopic xenograft model of human lung adenocarcinoma.ConclusionsCollectively, these studies demonstrate that FT536 is a highly potent, multi-tumor targeting CAR-iNK cell product that is uniform in composition and can be effectively and safely used off-the-shelf for on-demand treatment of multiple solid and hematological malignancies. An IND submission is planned for 2021, with an initial Phase 1 clinical trial to follow.
- Published
- 2021
17. Engineered iPSC-Derived NK Cells Expressing Recombinant CD64 for Enhanced ADCC
- Author
-
Robert Hullsiek, Jason Dinella, Ramzey Abujarour, Kate Dixon, Hui-yi Chu, Bahram Valamehr, Ryan Bjordahl, Melissa Khaw, Jeffrey S. Miller, Zachary Davis, Paul Rogers, Jianming Wu, Bruce Walcheck, Thomas H. Lee, and Kristin M. Snyder
- Subjects
CD64 ,Antibody-dependent cell-mediated cytotoxicity ,law ,Chemistry ,Immunology ,Recombinant DNA ,Cell Biology ,Hematology ,Biochemistry ,law.invention ,Cell biology - Abstract
Natural killer (NK) cells are innate cytotoxic lymphocytes. They target malignant cells via non-clonotypic receptors to induce natural cytotoxicity and also recognize tumor-bound antibodies to induce antibody-dependent cell-mediated cytotoxicity (ADCC). While ADCC by NK cells is a key mechanism of several clinically successful therapeutic monoclonal antibodies (mAbs), most patients exhibit or acquire resistance to mAb therapies. ADCC by human NK cells is exclusively mediated by the IgG Fc receptor, CD16A (FcγRIIIA). Studies have demonstrated that increasing the binding affinity between CD16A and therapeutic mAbs can augment their clinical efficacy. Given the exquisite specificity and diverse antigen detection of anti-tumor mAbs, we are interested in enhancing the ADCC potency of NK cell-based therapies for various malignancies. CD64 is the only high affinity FcγR family member and binds to the same IgG isotypes as CD16A (IgG1 and IgG3) but with > 30-fold higher affinity. CD64 (FcγRI) is normally expressed by certain myeloid cells but not by NK cells. We generated a recombinant version of this receptor consisting of the extracellular region of CD64 and the transmembrane and intracellular regions of human CD16A, referred to as CD64/16A (figure 1A). An important feature of CD64/16A is that due to its high affinity state, soluble monomeric anti-tumor mAbs can be pre-adsorbed to engineered NK cells expressing the recombinant FcγR, and these pre-absorbed mAbs can be switched or mixed for universal tumor antigen targeting (figure 1B). The engineered NK cells used in our study were derived from genetically edited and clonally derived induced pluripotent stem cells (iPSCs) through a series of stepwise differentiation stages (figure 2). Engineered iPSC-derived NK (iNK) cells can be produced in a uniform and clinically scalable manner (figure 2). In Figure 3, using an in vitro Delfia® ADCC assay, we show that iNK-CD64/16A cells mediated ADCC against SKOV3 cells, an ovarian adenocarcinoma cell line, in the presence of the anti-HER2 therapeutic mAb trastuzumab (Herceptin) or anti-EGFR1 therapeutic mAb cetuximab (Erbitux), when either added to the assay or pre-adsorbed to the iNK cells (figure 3). Considering the high affinity state of CD64, we examined the effects of free IgG in human serum on ADCC by iNK-CD64/16A cells. Using an IncuCyte® Live Cell Analysis System, ADCC was evaluated in the presence or absence of 5% human AB serum, in which free IgG was approximately 50-fold higher than the IgG saturation level of the CD64/16A receptors on iNK cells (data not shown). Despite the high levels of excess free IgG, iNK-CD64/16A cells mediated efficient ADCC when Herceptin was either added to the assay or pre-adsorbed to the cells (figure 4). ADCC assays were also performed with Raji cells, a Burkitt lymphoma cell line, as target cells and the therapeutic mAb rituximab (Rituxan). iNK-CD64/16A cells were added with or without pre-adsorbed Rituxan and the assay was performed in 10% AB serum. Again, iNK-CD64/16A cells mediated effective target cell killing in the presence of serum IgG (figure 5), demonstrating that saturating levels of free IgG did not prevent ADCC. To determine if we can further optimize the function of recombinant CD64, we engineered CD64 with the transmembrane regions of CD16A or NKG2D and signaling/co-signaling domain from CD28, 2B4 (CD244), 4-1BB (CD137), and CD3ζ (figure 6). CD64/16A signals by non-covalent association with the immunoreceptor tyrosine-based activation motif (ITAM)-containing signaling adapters CD3ζ and FcRγ found in the cell membrane, whereas the other recombinant CD64 constructs use ITAM and non-ITAM regions to mediate their signaling. The various recombinant CD64 constructs were initially expressed in NK92 cells (lacks expression of endogenous FcγRs) (figure 7). Using the Delfia® ADCC assay system, we examined the function of each recombinant CD64 construct and found all combinations are able to effectively induce ADCC (figure 8). We are in the process of generating iNK cells with these constructs and testing their ability to kill hematologic and solid tumors in vitro and in vivo. Our goal is to utilize this docking approach to pre-absorb mAbs to iNK cells for adoptive cell therapy. The mAbs would thus provide tumor-targeting elements that could be exchanged as a means of preventing tumor cell escape by selectively and easily altering NK cell specificity for tumor antigens. Figure Disclosures Lee: Fate Therapeutics, Inc.: Current Employment. Chu:Fate Therapeutics: Current Employment. Abujarour:Fate Therapeutics, Inc: Current Employment. Dinella:Fate Therapeutics: Current Employment. Rogers:Fate Therapeutics, Inc: Current Employment. Bjordahl:Fate Therapeutics: Current Employment. Miller:Fate Therapeutics, Inc: Consultancy, Patents & Royalties, Research Funding; Nektar: Honoraria, Membership on an entity's Board of Directors or advisory committees; Vycellix: Consultancy; GT Biopharma: Consultancy, Patents & Royalties, Research Funding; Onkimmune: Honoraria, Membership on an entity's Board of Directors or advisory committees. Valamehr:Fate Therapeutics, Inc: Current Employment, Current equity holder in publicly-traded company. Walcheck:Fate Therapeutics: Consultancy, Research Funding.
- Published
- 2020
18. cGMP Mass Production of FT538, a First-of-Kind, Off-the-Shelf, Multiplexed Engineered Natural Killer Cell Cancer Immunotherapy Derived from a Clonal Master Induced Pluripotent Stem Cell Line
- Author
-
Luane Reyes, Alice Lin, Ramzey Abujarour, Thomas H. Lee, Betsy Rezner, Bahram Valamehr, Luis A. Solchaga, and Wen Bo Wang
- Subjects
education.field_of_study ,medicine.medical_treatment ,Immunology ,Population ,Daratumumab ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,Biochemistry ,Natural killer cell ,medicine.anatomical_structure ,Cancer immunotherapy ,medicine ,Cancer research ,Clone (B-cell biology) ,Induced pluripotent stem cell ,education ,Reprogramming - Abstract
FT538 is an investigational, off-the-shelf, multiplexed engineered natural killer (NK) cell cancer immunotherapy that is derived from a clonal master induced pluripotent stem cell (iPSC) line engineered with three functional components to enhance innate immunity: a novel high-affinity, non-cleavable CD16 (hnCD16) Fc receptor; an IL-15/IL-15 receptor fusion (IL-15RF); and the elimination of CD38 expression. The use of a clonal master engineered iPSC line as a starting cell source enables routine mass production of FT538 and supports off-the-shelf product availability to reach many patients. The clonal master iPSC line for the manufacture of FT538 was made by reprogramming and engineering donor-consented human fibroblasts to induce pluripotency using a proprietary non-integrating system and to integrate a bicistronic cassette containing hnCD16 and IL-15RF into the CD38 locus, which resulted in complete disruption of the CD38 gene. The engineered iPSC population was then sorted to isolate single clones, and each engineered iPSC clone was screened for multiple critical quality attributes including pluripotency, identity, genomic stability, cassette integration, and off-target effects of engineering. Accordingly, a single engineered iPSC clone was selected as the FT538 clonal master iPSC line. The clonal master engineered iPSC line serves as a renewable source for the routine cGMP mass production of FT538 drug product. Routine cGMP manufacture of FT538 drug product consists of three stages: 1) differentiation of the clonal master engineered iPSC line to CD34-expressing hematopoietic progenitor cells; 2) further differentiation to and expansion of NK cells; and 3) fill/finish and cryopreservation of the FT538 drug product. A cGMP manufacturing campaign yielded sufficient CD34-expressing hematopoietic progenitor cells to support at least 15 batches of NK cell differentiation, the first batch of which was used to produce a total of 3 × 1011 FT538 NK cells filled into over 300 units of cryopreserved drug product. The cGMP campaign had a theoretical yield of 4.5 x 1012 FT538 NK cells. The FT538 drug product was characterized and found to be comprised of uniformly engineered CD56+ NK cells with homogeneous expression of hnCD16 and lacking expression of CD38. Importantly, there were no residual iPSCs detected in the FT538 drug product. Additionally, the FT538 drug product exhibited potent effector function in a candidate potency assay measuring IFN-γ release in response to RPMI 8226 target cells in the presence of daratumumab. An Investigational New Drug application for FT538 has been cleared by the U.S. Food and Drug Administration (FDA) for the conduct of a multicenter, multi-dose Phase I clinical trial for the treatment of patients with relapsed/refractory (r/r) acute myelogenous leukemia (AML) and multiple myeloma (MM). The dose-escalation utilizes a 3+3 design to identify the maximum tolerated dose of three doses of FT538 on Days 1, 8, and 15 as a monotherapy in r/r AML (Regimen A) and in combination with daratumumab (Regimen B) or elotuzumab (Regimen C) in r/r MM. The trial will test up to five FT538 dose levels ranging from 50 million to 1.5 billion cells, and up to 105 patients will be enrolled. The trial is expected to begin patient enrollment in 2020. Disclosures Rezner: Fate Therapeutics, Inc: Current Employment, Current equity holder in publicly-traded company. Solchaga:Fate Therapeutics, Inc: Current Employment. Reyes:Fate Therapeutics, Inc: Current Employment. Lin:Fate Therapeutics, Inc: Current Employment. Abujarour:Fate Therapeutics, Inc: Current Employment. Lee:Fate Therapeutics, Inc.: Current Employment. Valamehr:Fate Therapeutics, Inc: Current Employment, Current equity holder in publicly-traded company. Wang:Fate Therapeutics, Inc: Current Employment.
- Published
- 2020
19. Generation of Multiplexed Engineered, Off-the-Shelf CAR T Cells Uniformly Carrying Multiple Anti-Tumor Modalities to Prevent Tumor Relapse
- Author
-
Bahram Valamehr, Samuel LaBarge, Yi-Shin Lai, Mochtar Pribadi, Suzanna Gasparian, Mandal Mili, Alma Gutierrez, Matthew Denholtz, Gloria Hsia, Jason ORourke, May Sumi, Bi-Huei Yang, Cokey Nguyen, Sandeep Kothapally Hanok, Alec Witty, Eric Sung, Amit R. Mehta, Raedun Clarke, Ramzey Abujarour, Eigen Peralta, Emily Carron, Thomas H. Lee, Emily Driver, Angela Gentile, Natalie Navarrete, David J. Robbins, Wen-I Yeh, Philip Chu, Chia-Wei Chang, and Amanda D. Yzaguirre
- Subjects
medicine.medical_treatment ,T cell ,Immunology ,T-cell receptor ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Chimeric antigen receptor ,Cytokine ,medicine.anatomical_structure ,Antigen ,Cancer research ,medicine ,Induced pluripotent stem cell ,Cell bank ,Reprogramming - Abstract
The development of chimeric antigen receptor (CAR) T cell therapeutics is widely recognized as a significant advancement for the treatment of cancer. However, several obstacles currently impede the broad use of CAR T cells, including the inherent process variability, cost of manufacturing, the absolute requirement for precise and uniform genetic editing in the allogeneic setting, and the challenge to keep pace with clonal heterogeneity and tumor growth. Utilizing our previously described induced pluripotent stem cell (iPSC)-derived T (iT) cell platform, we illustrate here the unique ability to address these challenges by creating a consistent CAR iT cell product that can be repeatedly manufactured in large quantities from a renewable iPSC master cell bank that has been engineered to mitigate the occurrence of graft versus host disease (GvHD), antigen escape and tumor relapse. Utilizing our proprietary cellular reprogramming and engineering platform and stage-specific T cell differentiation protocol, we demonstrate that iPSC can be engineered at the single cell level to generate a fully characterized clonal iPSC line, which can then be accessed routinely to yield CAR iT cells in a highly scalable manufacturing process (>100,000 fold expansion). Through bi-allelic targeting of a CAR into the T cell receptor alpha constant (TRAC) region, we generated CAR iT cells with uniform CAR expression (99.0 ± 0.5% CAR+) and complete elimination of T cell receptor (TCR) expression to avoid GvHD in the allogeneic setting. We elected to utilize the 1XX-CAR configuration, which has demonstrated superior anti-tumor performance relative to other CAR designs and when introduced into iT cells displayed enhanced antigen specificity (% specific cytotoxicity at E:T=10:1, antigen positive group: 86.4 ± 7.8; antigen null group: 8.9 ± 3.5). To enhance persistence without reliance on exogenous cytokine support, we engineered signaling-fusion complexes, including IL-7 receptor fusion (RF), into iPSC and studied its impact on iT phenotype, persistence, and efficacy. In vitro, IL-7RF clones demonstrated improved anti-tumor activity in a serial antigen dependent tumor challenge assay (Day 10, relative tumor counts, IL-7RF group: 1.95 ± 0.01; control group: 57.56 ± 4.55, P Collectively, the described studies demonstrate that iPSCs are an ideal cellular source to generate large-quantities of uniformly multi-edited off-the-shelf CAR T cell products that include a best-in-class CAR design, enhanced product modalities, and complete elimination of TCR expression to avoid the potential of GvHD while maintaining high anti-tumor efficacy in allogeneic setting. Disclosures Hsia: Fate Therapeutics Inc.: Current Employment. Clarke:Fate Therapeutics Inc.: Current Employment, Current equity holder in publicly-traded company. Lee:Fate Therapeutics, Inc.: Current Employment. Robbins:Fate Therapeutics, Inc.: Current Employment. Denholtz:Fate Therapeutics, Inc: Current Employment. Hanok:Fate Therapeutics, Inc.: Current Employment. Carron:Fate Therapeutics, Inc.: Current Employment. Navarrete:Fate Therapeutics, Inc.: Current Employment. ORourke:Fate Therapeutics, Inc.: Current Employment. Sung:Fate Therapeutics, Inc.: Current Employment. Gentile:Fate Therapeutics, Inc.: Current Employment. Nguyen:Fate Therapeutics, Inc.: Current Employment. Valamehr:Fate Therapeutics, Inc: Current Employment, Current equity holder in publicly-traded company.
- Published
- 2020
20. Abstract 3245: FT819 path to IND: First-of-kind off-the-shelf CAR19 T-cell for B cell malignancies
- Author
-
Ramzey Abujarour, Cheng-Jang Wu, Jason ORourke, Mandal Mili, Bahram Valamehr, Jolanta Stefanski, Gloria Hsia, Alec Witty, Bi-Huei Yang, Sjoukje J. C. van der Stegen, Gilberto Hernandez, Michel Sadelain, Mochtar Pribadi, Chia-Wei Chang, Helen Chu, Yi-Shin Lai, Raedun Clarke, Thomas H. Lee, Meilan Wu, Juan Zhen, Isabelle Riviere, Mushtaq Husain, Laurel Stokely, Chad Dufaud, Helena Shaked, and Alma Gutierrez
- Subjects
Cancer Research ,T cell ,T-cell receptor ,Biology ,Molecular biology ,CD19 ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,Antigen ,medicine ,biology.protein ,Cytotoxic T cell ,Cell bank ,B cell - Abstract
Genetic engineering of T cells using a chimeric antigen receptor targeting CD19 antigen (CAR19) is now a well-established treatment of B cell malignancies. While cellular immunotherapies are entering front line treatment, substantial limitations currently hamper the broad application of adoptive T cell therapies in diverse patient population including dysfunctional starting material, lack of product consistency and purity post genetic engineering and inefficient quantity produced for true on-demand availability. FT819 is a first-of-kind off-the-shelf CAR19-T cell product generated from a renewable pluripotent stem cells for large-scale clinical manufacturing. We previously reported the engineering and characterization of the FT819 clonal master cell bank (MCB) derived from a single cell comprising targeted integration of a novel CD19 1XX CAR into the T-cell receptor (TCR) α constant locus to provide optimally regulated CAR expression and elimination of graft versus host (GvH) response. Here we preview the nonclinical study for the original investigational new drug application of FT819. Derived in a manufacturing process analogous to pharmaceutical drug product development, pilot runs from the MCB demonstrated FT819 can be consistently and uniformly manufactured in cGMP compliance, cryopreserved at clinical scale to support off-the-shelf clinical application with greater than 1e5 fold increase in cellular yield from the starting MCB and can be thawed and directly used for facilitated treatment. Repeatedly, FT819 displayed a uniform product profile of ≥95% CAR+, TCR-, CD45+, CD7+ and CD3+ [intracellular] with majority of CD8 T cells expressing CD8β. FT819 global gene expression profile displayed high similarity to primary CAR19-T cells confirming its identity as a T lymphocyte. Functional assessment demonstrated that FT819 possesses potent antigen specific cytolytic activity against leukemia and lymphoma cell lines (p=0.0004). Additional specificity studies demonstrated on-target, off-tumor cytolysis of CD19+ B cells in mixed lymphocyte reaction assay (85% lysis of CD19+ B cells versus < 2% lysis of T cells). Inability of FT819 to produce a GvH response was confirmed in a co-culture assay with anti-TCR crosslinking antibodies. Disseminated leukemia xenograft mouse studies demonstrated the ability of directly thawed and infused FT819 to control tumor growth (p=0.0003 at day 21). In a systemic administered leukemia model FT819 also showed sustained localization in the bone marrow up to 45 days post injection. Ongoing in vivo studies will assess long-term survival and avoidance of GvH disease. Collectively, these studies demonstrate that FT819 is a potent, consistent and uniform CAR19 T cell product and can be effectively and safely used off-the-shelf in the treatment of B cell malignancies with an original Phase 1 clinical trial planned in 2020. Citation Format: Mili Mandal, Raedun Clarke, Sjoukje van der Stegen, Chia-Wei Chang, Yi-Shin Lai, Alec Witty, Mushtaq Husain, Cheng-Jang Wu, Bi-Huei Yang, Chad Dufaud, Gloria Hsia, Helena Shaked, Laurel Stokely, Helen Chu, Mochtar Pribadi, Gilberto Hernandez, Jason ORourke, Alma Gutierrez, Ramzey Abujarour, Tom Lee, Jolanta Stefanski, Juan Zhen, Meilan Wu, Isabelle Riviere, Michel Sadelain, Bahram Valamehr. FT819 path to IND: First-of-kind off-the-shelf CAR19 T-cell for B cell malignancies [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3245.
- Published
- 2020
21. Myogenic Differentiation of Muscular Dystrophy-Specific Induced Pluripotent Stem Cells for Use in Drug Discovery
- Author
-
Kevin Lai, Thuy Ai Huyen Le, Monica Bennett, Bahram Valamehr, Peter Flynn, David J. Robbins, Tom Tong Lee, Ramzey Abujarour, and Megan Robinson
- Subjects
Pluripotent Stem Cells ,Cell type ,Cellular differentiation ,Muscle Fibers, Skeletal ,Gene Expression ,Biology ,MyoD ,MyoD Protein ,Drug Discovery ,medicine ,Humans ,Myocyte ,Cell Lineage ,Muscular dystrophy ,Induced pluripotent stem cell ,Cells, Cultured ,Embryonic Stem Cells/Induced Pluripotent Stem (iPS) Cells ,Myosin Heavy Chains ,Myogenesis ,Cell Differentiation ,Cell Biology ,General Medicine ,musculoskeletal system ,medicine.disease ,Research Highlight ,Molecular biology ,Cell biology ,Muscular Dystrophy, Duchenne ,tissues ,Developmental Biology - Abstract
Human induced pluripotent stem cells (iPSCs) represent a scalable source of potentially any cell type for disease modeling and therapeutic screening. We have a particular interest in modeling skeletal muscle from various genetic backgrounds; however, efficient and reproducible methods for the myogenic differentiation of iPSCs have not previously been demonstrated. Ectopic myogenic differentiation 1 (MyoD) expression has been shown to induce myogenesis in primary cell types, but the same effect has been unexpectedly challenging to reproduce in human iPSCs. In this study, we report that optimization of culture conditions enabled direct MyoD-mediated differentiation of iPSCs into myoblasts without the need for an intermediate step or cell sorting. MyoD induction mediated efficient cell fusion of mature myocytes yielding multinucleated myosin heavy chain-positive myotubes. We applied the same approach to dystrophic iPSCs, generating 16 iPSC lines from fibroblasts of four patients with Duchenne and Becker muscular dystrophies. As seen with iPSCs from healthy donors, within 36 hours from MyoD induction there was a clear commitment toward the myogenic identity by the majority of iPSCs in culture (50%–70%). The patient iPSC-derived myotubes successfully adopted the skeletal muscle program, as determined by global gene expression profiling, and were functionally responsive to treatment with hypertrophic proteins insulin-like growth factor 1 (IGF-1) and wingless-type MMTV integration site family, member 7A (Wnt7a), which are being investigated as potential treatments for muscular dystrophy in clinical and preclinical studies, respectively. Our results demonstrate that iPSCs have no intrinsic barriers preventing MyoD from inducing efficient and rapid myogenesis and thus providing a scalable source of normal and dystrophic myoblasts for use in disease modeling and drug discovery.
- Published
- 2014
22. FT819: Translation of Off-the-Shelf TCR-Less Trac-1XX CAR-T Cells in Support of First-of-Kind Phase I Clinical Trial
- Author
-
Mochtar Pribadi, Bahram Valamehr, Michel Sadelain, Alec Witty, Ramzey Abujarour, Hui-yi Chu, Helena Shaked, Mandal Mili, Jason ORourke, Brian Groff, Laurel Stokely, Mushtaq Husain, Alma Gutierrez, Yi-Shin Lai, Chia-Wei Chang, Raedun Clarke, Isabelle Riviere, Thomas H. Lee, Bi-Huei Yang, Sjoukje J. C. van der Stegen, and Pieter Lindenbergh
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Immunology ,T-cell receptor ,Juno Therapeutics ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Chimeric antigen receptor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Cancer research ,Cytotoxic T cell ,Clone (B-cell biology) ,CD8 ,B cell ,030215 immunology - Abstract
Long-term follow-up of adoptive transfer of autologous T cells expressing a chimeric antigen receptor (CAR) directed to CD19 antigen has demonstrated encouraging, durable clinical outcome in various B cell malignancies. However, to make such CAR-T cells available to a broader base and to reach a more diverse patient population, challenges associated with product consistency, cost of manufacture, precision genetic engineering and on-demand availability still need to be addressed. FT819 is a first-of-kind off-the-shelf CAR-T cell product candidate derived from a renewable master pluripotent cell line. FT819 comprises precise genetic engineering of multiple targeting events at the single cell level and is produced using a clonally-derived master cell bank (MCB) that serves as the starting material to support consistent and reproducible clinical manufacturing. The engineered features of FT819 include the targeted integration of a novel CD19 1XX-CAR into the T-cell receptor α constant (TRAC) locus to provide antigen specificity, enhanced efficacy and temporally-regulated CAR expression driven by an endogenous (TCR) promoter. Such features are designed to also eliminate the possibility of graft versus host disease (GvHD) by nullifying the TCR. To develop the MCB for FT819, αβ T cells were reprogrammed into induced pluripotent stem cells (iPSCs) and subsequently engineered to direct CD19 1XX-CAR into the TRAC locus with knockout of the TCR. To generate clonal lines, engineered iPSCs were sorted by flow cytometry for various markers and single cells were seeded into individual wells of feeder-free 96-well plates. Engineered iPSC clones were screened for integration of CAR into the TRAC locus by amplifying the genomic DNA flanking the homologous recombination site and confirmed by a SNP phasing assay. Clones were further screened for random integration of donor template by quantitative PCR and the CAR copy number was confirmed by droplet digital PCR. Genome stability of each clone was also confirmed by karyotype analysis. Overall, the described screening initiative surveyed 774 clones to select the ideal MCB for FT819. Utilizing our stage-specific T cell differentiation and expansion protocol, we demonstrated that T cells derived from the FT819 (FT819-iTs) expanded greater than 100,000-fold during the clinical manufacturing process and the cells expressed greater than 95% T lymphocyte markers such as CD45, CD7, intracellular CD3, and TRAC-regulated CAR. Further modifications to the T cell differentiation protocol resulted in enhanced expression of CD8 αβ from less than 25% to greater than 70% of the total population. In addition, expression of CD2, CD5, and CD27 was increased by approximately 5- to 20-fold. In vitro functional studies showed that FT819-iTs possess antigen specificity as confirmed by cytokine release and cytotoxic T lymphocytes (CTL) assays. Upon stimulation with a wild type acute lymphoblastic leukemia line, Nalm-6, FT819-iTs expressed 30% CD107a/b compared to 2% when stimulated by Nalm-6 CD19KO. In an in vitro CTL assay, greater than 80% of Nalm-6 WT cells were lysed with effector to target (E:T) ratio at 10:1 as compared to Nalm-6-CD19KO, which showed less than 10% lysis at the same E:T ratio. Finally, in an in vivo tumor model, FT819-iTs generated from our original and modified T cell differentiation protocols showed similar tumor burden control and prolonged survival rate when compared to primary CAR-T cells (days of survival >80days, p>0.1). In a more stringent in vivo model, FT819-iTs generated from the modified differentiation protocol demonstrated higher anti-tumor response and better animal survival rate compared to iTs from the original T cell differentiation protocol (Day 30 p Disclosures Chang: Fate Therapeutics: Employment. Van Der Stegen:Memorial Sloan Kettering Cancer Center: Employment. Mili:Fate Therapeutics: Employment. Clarke:Fate Therapeutics: Employment. Lai:Fate Therapeutics: Employment. Witty:Fate Therapeutics: Employment. Lindenbergh:Memorial Sloan Kettering Cancer Center: Employment. Yang:Fate Therapeutics: Employment. Husain:Fate Therapeutics: Employment. Shaked:Fate Therapeutics: Employment. Groff:FATE THERAPEUTICS: Employment. Stokely:Fate Therapeutics: Employment. Abujarour:Fate Therapeutics, Inc.: Employment. Lee:Fate Therapeutics, Inc.: Employment. Chu:Fate Therapeutics: Employment. Pribadi:Fate Therapeutics, Inc.: Employment. ORourke:Fate Therapeutics: Employment. Gutierrez:Fate Therapeutics: Employment. Riviere:Juno Therapeutics: Consultancy, Equity Ownership, Research Funding; Fate Therapeutics: Consultancy; Memorial Sloan Kettering Cancer Center: Employment. Sadelain:Memorial Sloan Kettering Cancer Center: Employment; Fate Therapeutics: Consultancy, Patents & Royalties; Juno Therapeutics: Consultancy, Patents & Royalties, Research Funding. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2019
23. FT538: Preclinical Development of an Off-the-Shelf Adoptive NK Cell Immunotherapy with Targeted Disruption of CD38 to Prevent Anti-CD38 Antibody-Mediated Fratricide and Enhance ADCC in Multiple Myeloma When Combined with Daratumumab
- Author
-
Thomas H. Lee, Megan Robinson, Bahram Valamehr, Dan S. Kaufman, Ryan Bjordahl, Janel Huffman, Karl-Johan Malmberg, Lauren Fong, Mochtar Pribadi, Frank Cichocki, Greg Bonello, Karrune Woan, Chelsea Ruller, Hui-yi Chu, Ramzey Abujarour, Jason Dinella, Svetlana Gaidarova, and Jeffrey S. Miller
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,medicine.medical_treatment ,Immunology ,Daratumumab ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,NKG2D ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Interleukin 15 ,Cancer research ,medicine ,Induced pluripotent stem cell ,Clone (B-cell biology) ,Reprogramming ,030215 immunology - Abstract
Monoclonal antibody (mAb) treatment is an effective therapeutic strategy for many cancer types, though there remains meaningful opportunity to improve mAb efficacy by optimizing the interaction with natural killer (NK) cells to enhance antibody-dependent cellular cytotoxicity (ADCC). NK cells are an ideal effector cell for combined use with tumor-targeting mAbs, as NK cells effect both innate tumoricidal capacity and ADCC. CD38-targeting mAbs, such as daratumumab, are effective in treating multiple myeloma (MM) and achieve their efficacy through multiple mechanisms, including ADCC. However, because activated NK cells express high levels of CD38, daratumumab induces NK cell depletion through fratricide, potentially reducing treatment effectiveness. Adoptive NK cell immunotherapy therefore has the potential to augment daratumumab's ADCC activity if fratricide can be reduced or prevented. FT538 is an off-the-shelf adoptive NK cell immunotherapy product candidate designed for enhanced cellular persistence and ADCC while avoiding anti-CD38 mAb induced fratricide. It is derived from induced pluripotent stem cells (iPSC) engineered to lack CD38 expression, which we have previously shown to eliminate daratumumab-induced fratricide among iPSC-derived NK cells, resulting in enhanced long-term daratumumab-mediated ADCC. FT538 is engineered to express an IL-15 receptor alpha fusion protein (IL-15RF; IL-15 tethered to IL-15 receptor α) to enhance persistence and a high-affinity non-cleavable CD16 (hnCD16, FcRγIII) to increase ADCC. To support the clinical translation of FT538, and to enable the repeatable and scalable cell production to support off-the-shelf availability of a uniform NK cell product, a clinical-grade master pluripotent stem cell line was developed. The FT538 master pluripotent stem cell line was created by reprogramming donor fibroblasts into iPSCs using our non-integrating cellular reprogramming platform, and cells were further genetically edited by targeting IL-15RF and hnCD16 to the CD38 locus. Clonal iPSC lines were generated and screened for precise knock-in and knock-out edits at the CD38 locus and a lack of off-target genome integration (15% total success rate for CD38-/-IL-15RF+CD16+). Selected engineered iPSC clones were confirmed to be free of reprogramming transgenes and to maintain genomic stability. Engineered iPSC clones were additionally tested for their NK cell differentiation potential and function, and a single clone was selected to serve as the renewable starting material for cGMP manufacturing and clinical development. Upon differentiation and expansion FT538 demonstrated a mature NK cell phenotype with expression of NK cell receptors including NKp30, NKp46, NKG2D, KIR, NKG2A, and DNAM-1. The functional impact of CD38 knockout on FT538 NK cells was confirmed in an in vitro fratricide assay, where peripheral blood (PB)-NK cells exhibited fratricide at a frequency of 33% after 3 hr culture with increasing daratumumab concentrations. In contrast, FT538 cells were entirely resistant ( Disclosures Bjordahl: Fate Therapeutics, Inc.: Employment. Gaidarova:Fate Therapeutics, Inc: Employment. Cichocki:Fate Therapeutics, Inc: Research Funding. Bonello:Fate Therapeutics, Inc.: Employment. Robinson:Fate Therapeutics, Inc.: Employment. Ruller:Fate Therapeutics, Inc.: Employment. Pribadi:Fate Therapeutics, Inc.: Employment. Dinella:Fate Therapeutics, Inc.: Employment. Fong:Fate Therapeutics, Inc.: Employment. Huffman:Fate Therapeutics, Inc.: Employment. Chu:FATE THERAPEUTICS: Employment. Lee:Fate Therapeutics, Inc.: Employment. Abujarour:Fate Therapeutics, Inc.: Employment. Kaufman:FATE Therapeutics: Consultancy, Research Funding. Malmberg:Fate Therapeutics, Inc.: Consultancy, Research Funding; Vycellix: Consultancy, Membership on an entity's Board of Directors or advisory committees. Miller:CytoSen: Membership on an entity's Board of Directors or advisory committees; Moderna: Membership on an entity's Board of Directors or advisory committees; OnKImmune: Membership on an entity's Board of Directors or advisory committees; GT BioPharma: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Dr. Reddys Laboratory: Membership on an entity's Board of Directors or advisory committees; Fate Therapeutics, Inc: Consultancy, Research Funding. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2019
24. FT576: A Novel Multiplexed Engineered Off-the-Shelf Natural Killer Cell Immunotherapy for the Dual-Targeting of CD38 and Bcma for the Treatment of Multiple Myeloma
- Author
-
Ryan Bjordahl, Janel Huffman, Mochtar Pribadi, Chelsea Ruller, Uta E. Höpken, Jason Dinella, Megan Robinson, Ramzey Abujarour, Bahram Valamehr, Sajid Mahmood, Svetlana Gaidarova, Jode P Goodridge, Thomas H. Lee, Armin Rehm, Greg Bonello, and Hui-yi Chu
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,medicine.drug_class ,medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,Monoclonal antibody ,Biochemistry ,Fusion protein ,Chimeric antigen receptor ,Natural killer cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Cancer research ,Induced pluripotent stem cell ,030215 immunology - Abstract
Multiple myeloma (MM) is a B cell neoplasm that originates from the malignant transformation of plasma cells, with treatment strategies that include chemotherapeutic agents and immunomodulatory drugs. Recently, significant effort has been applied to the development of monoclonal antibody (mAb) and chimeric antigen receptor (CAR) T cell therapies for the treatment of advanced MM. Anti-CD38 mAb therapy is at the forefront of these efforts, with clearly demonstrated clinical benefit and availability of a FDA-approved mAb in daratumumab. Antibody-dependent cellular cytotoxicity (ADCC) is a key mechanism of action of CD38-targeted mAbs; however, high CD38 expression on natural killer (NK) cells results in fratricide, which depletes the NK cells necessary for ADCC. In addition to CD38, targeting of other MM-associated cell-surface proteins has been explored. Of these antigens, the TNF-superfamily member BCMA is among the most researched and is under development by multiple groups as a CAR target. Several clinical trials in MM have shown promising initial results targeting BCMA with CAR T cells, however there remains significant opportunity to improve both relapse rates and treatment of relapsed patients. Collectively, clinical data would suggest that combinatorial targeting of both CD38 and BCMA may improve clinical efficacy compared with targeting either antigen alone. We have developed a multiple-target, adoptive NK cell immunotherapy approach for the treatment of MM. The strategy utilizes our off-the-shelf NK cell platform with four engineered attributes: 1) an anti-BCMA CAR for direct MM targeting, 2) high affinity non-cleavable CD16 (hnCD16) for enhanced ADCC in combination with anti-CD38 mAbs, 3) CD38 deletion for resistance to anti-CD38 mAb induced NK cell depletion, and 4) IL-15/IL-15 receptor α fusion protein (IL-15RF; IL-15 fused to IL-15Rα) for enhanced NK cell persistence. The anti-BCMA CAR consists of a unique single chain variable fragment (scFv) targeting domain with a BCMA binding affinity in the low nanomolar range, providing high functional avidity and efficacy in disease settings where BCMA antigen density is low. Our approach utilizes NK cells derived from a genetically engineered, clonally-derived master pluripotent stem cell line with uniform expression of anti-BCMA CAR, IL-15RF, hnCD16, and CD38 bi-allelic knockout. The engineered master pluripotent stem cell line serves as the starting material for consistent and repeatable manufacture of off-the-shelf NK cells that contain all described attributes in a homogenous manner (termed FT576) and that can be produced at a scale to support multi-dose treatment strategies and on-demand dose availability. In preclinical studies, FT576 NK cells exhibited uniform expression of CD16, CAR, and IL15-RF and did not express CD38 (99% cytotoxicity in a 3D-spheroid culture model. Preclinical studies are ongoing to support the advancement of FT576 as the first-of-kind cellular therapeutic for the combination of anti-BCMA CAR and mAb-directed targeting of MM. Disclosures Bjordahl: Fate Therapeutics, Inc.: Employment. Gaidarova:Fate Therapeutics, Inc: Employment. Goodridge:FATE THERAPEUTICS: Employment. Mahmood:Fate Therapeutics, Inc: Employment. Bonello:Fate Therapeutics, Inc.: Employment. Robinson:Fate Therapeutics, Inc.: Employment. Ruller:Fate Therapeutics, Inc.: Employment. Pribadi:Fate Therapeutics, Inc.: Employment. Lee:Fate Therapeutics, Inc.: Employment. Abujarour:Fate Therapeutics, Inc.: Employment. Dinella:Fate Therapeutics, Inc.: Employment. Huffman:Fate Therapeutics, Inc.: Employment. Chu:FATE THERAPEUTICS: Employment. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2019
25. Abstract LB-073: Generation of novel single cell-derived engineered master pluripotent cell line as a renewable source for off-the-shelf TCR-less CAR T cells in support of first-of-kind clinical trial
- Author
-
Jolanta Stefanski, Raedun Clarke, Isabelle Riviere, Yi-Shin Lai, Chelsea Ruller, Megan Robinson, Pieter Lindenbergh, Ramzey Abujarour, Helen Chu, Alec Witty, Jaeger Davis, Amanda Medcalf, Jason Dinella, Thomas H. Lee, Bahram Valamehr, Juan Zhen, Michel Sadelain, Janel Huffman, Mochtar Pribadi, Greg Bonello, Stacey K. Moreno, Xiuyan Wang, and Sjoukje J. C. van der Stegen
- Subjects
Cancer Research ,Adoptive cell transfer ,medicine.diagnostic_test ,T-cell receptor ,Biology ,Molecular biology ,CD19 ,Chimeric antigen receptor ,Flow cytometry ,genomic DNA ,Oncology ,biology.protein ,medicine ,Induced pluripotent stem cell ,Reprogramming - Abstract
Adoptive transfer of autologous T cells expressing chimeric antigen receptor (CAR) has shown great promise in the treatment of blood malignancies. Challenges for the application of current CAR T cell therapies to broader and more diverse patient populations include inherent variability, cost of manufacture, and the requirement for precise genetic engineering to generate a highly homogenous and consistent CAR T cell product. We have previously reported pre-clinical data supporting the development of FT819, a first-of-kind off-the-shelf CAR T cell product candidate. FT819 is generated from a renewable clonal master human induced pluripotent stem cell (hiPSC) line derived from a single cell engineered to contain bi-allelic disruption of the T cell receptor (TCR) and a novel CD19 CAR targeted into the T cell receptor α constant (TRAC) locus to provide antigen specificity and enhanced efficacy while eliminating the possibility of graft versus host disease. For the manufacture of a clinical-grade FT819 clonal master hiPSC line, we sourced peripheral blood mononuclear cells from a fully consented and eligible donor with protocol overseen by an independent Institutional Review Board. Sourced T cells were enriched (>98%) through positive selection for TCRαβ, and cryopreserved cells were confirmed to have stable genome by karyotyping. Using our proprietary non-integrating cellular reprogramming platform, αβ T cells were reprogrammed into hiPSCs. Concurrently with the reprogramming process, reprogrammed cells received nuclease and donor template to mediate targeting of CD19 CAR into the TRAC locus with bi-allelic knockout of the TCR. To generate clonal lines, engineered cells were sorted by flow cytometry for various markers and single cells were seeded into individual wells of feeder-free 96-well plates. hiPSC clones were screened for bi-allelic integration of CAR into the TRAC locus by amplifying the genomic DNA flanking the homologous recombination site and confirmed by a SNP phasing assay. Clones were further screened for random integration of donor template by quantitative PCR (qPCR), and the CAR copy number was confirmed by droplet digital PCR. Out of 545 hiPSC clones screened, 27 clones (5%) had bi-allelic TRAC targeting with no detectable random integration. Maintenance of pluripotency was confirmed in 19 out of the 27 engineered hiPSC clones (70%). Seventeen clones were further tested and were confirmed to be footprint-free of transgenic reprogramming factors. Of the 18 clones tested for genomic stability, 12 clones had normal karyotypes (67%). Validated, TRAC-targeted hiPSC clones were cryopreserved (~150 vials per clone) and are currently being assessed for off-target editing, differentiation propensity into highly-functional T cells, genomic stability, clone identity, sterility and lack of mycoplasma detection. In summary, using our novel iPSC technology platform for reprogramming, single cell engineering and multiplex high-throughput screening of hiPSCs, we have generated clinical-grade clonal master hiPSC lines in support of our first-of-kind clinical trials evaluating FT819 allogenic off-the-shelf hiPSC-derived TCR-less TRAC-CAR19 T cells for the treatment of blood malignancies. Citation Format: Ramzey Abujarour, Yi-Shin Lai, Mochtar Pribadi, Tom Lee, Megan Robinson, Chelsea Ruller, Sjoukje Van der Stegen, Xiuyan Wang, Jolanta Stefanski, Juan Zhen, Jason Dinella, Greg Bonello, Janel Huffman, Helen Chu, Raedun Clarke, Alec Witty, Amanda Medcalf, Jaeger Davis, Stacey Moreno, Pieter Lindenbergh, Isabelle Riviere, Michel Sadelain, Bahram Valamehr. Generation of novel single cell-derived engineered master pluripotent cell line as a renewable source for off-the-shelf TCR-less CAR T cells in support of first-of-kind clinical trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-073.
- Published
- 2019
26. Abstract 3191: FT516, an off-the-shelf engineered NK cell therapeutic product for universal anti-tumor targeting strategy in combination with monoclonal antibodies
- Author
-
Frank Cichocki, Paul Rogers, Moyar Q. Ge, Bahram Valamehr, Greg Bonello, Ryan Bjordahl, Huang Zhu, Ramzey Abujarour, Brian Groff, Svetlana Gaidarova, Jeffrey S. Miller, Bruce Walcheck, Thomas H. Lee, Dan S. Kaufman, Jode P Goodridge, Helen Chu, and Robert Blum
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,biology ,business.industry ,medicine.drug_class ,ZAP70 ,Fc receptor ,CD38 ,CD16 ,Monoclonal antibody ,Oncology ,Calcium flux ,biology.protein ,Cancer research ,Medicine ,Antibody ,business - Abstract
Monoclonal antibody (mAb) treatment is an effective therapeutic strategy for many cancer types, with significant opportunity to optimize natural killer (NK) cell and mAb interaction to improve antibody-dependent cellular cytotoxicity (ADCC). NK cells are critical mediators of ADCC, where they recognize and kill malignant cells coated with antibody through the Fc receptor CD16. However, NK cell function is often impaired in cancer patients, which limits the induction of ADCC in mAb therapy. To enhance ADCC in combination with commercialized mAb therapies, we have developed FT516; a novel, off-the-shelf NK cell immunotherapeutic engineered to uniformly express a high-affinity, non-cleavable version of CD16 (hnCD16). FT516 is manufactured from a renewable master induced pluripotent stem cell (iPSC) line with the potential to generate hundreds to thousands of doses of allogeneic NK cells uniformly expressing hnCD16 (hnCD16 iNK cells) per manufacturing run. In an in vivo xenograft model of disseminated lymphoma, FT516 reduced tumor burden below the limit of detection at day 28 after transplant when delivered in combination with rituximab, which was significantly more potent than peripheral blood NK cells (p = 0.03). This was attributed to enhanced CD16-mediated activation of FT516, as observed through improved calcium flux and enhanced activation of signaling pathways such as ERK (p = 0.016), LAT (p = 0.0007), and ZAP70 (p = 0.0003), leading to enhanced ADCC and cytokine production. FT516 also maintained tumor cell specificity, with preferential targeting of K562 leukemia cells when presented with a mixture of K562 and normal PBMC targets (p < 0.0001). We also explored strategies to further engineer therapeutic function to enhance FT516 efficacy. Combined expression of hnCD16 with an IL-15/IL-15ra fusion construct enhanced the persistence of iNK cells and allowed survival of up to 8 weeks in vivo without exogenous cytokine (p < 0.0001), with correlation to improved efficacy. In vitro modeling of FT516 with daratumumab demonstrated ADCC against multiple myeloma (MM) targets. However, as reported, daratumumab induced NK cell fratricide through binding of CD38 on NK cells. To rescue daratumumab-mediated fratricide, we specifically deleted CD38 at the iPSC level and demonstrated that fratricide was undetectable in hnCD16 CD38-/- iNK cells ( Citation Format: Ryan Bjordahl, Huang Zhu, Paul Rogers, Svetlana Gaidarova, Moyar Q. Ge, Robert Blum, Frank Cichocki, Jode Goodridge, Helen Chu, Greg Bonello, Tom Lee, Brian Groff, Ramzey Abujarour, Bruce Walcheck, Jeffrey Miller, Dan Kaufman, Bahram Valamehr. FT516, an off-the-shelf engineered NK cell therapeutic product for universal anti-tumor targeting strategy in combination with monoclonal antibodies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3191.
- Published
- 2019
27. 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
28. Abstract 3576: FT500, an off-the-shelf NK cell cancer immunotherapy derived from a master pluripotent cell line, enhances T-cell activation and recruitment to overcome checkpoint blockade resistance
- Author
-
Moyar Q. Ge, Svetlana Gaidarova, Sajid Mahmood, Laurel Stokely, Ryan Bjordahl, Raedun Clarke, Paul Rogers, Megan Robinson, Bahram Valamehr, Ramzey Abujarour, Tom Tong Lee, and Betsy Rezner
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,Adoptive cell transfer ,business.industry ,medicine.medical_treatment ,T cell ,Cell ,Immunotherapy ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,CXCL10 ,Cytotoxic T cell ,Induced pluripotent stem cell ,business - Abstract
The development of PD1/PDL1 targeting checkpoint inhibitors (CI) has transformed the oncology landscape, providing long term remissions in multiple indications. However, many tumor subtypes are resistant to checkpoint blockade therapy, and relapse remains a significant concern. Novel therapeutic approaches with the ability to overcome CI resistance are needed, and there is significant opportunity for therapies capable of additively or synergistically enhancing T-cell activation and recruitment when combined with CI. Adoptive transfer of NK cells from healthy donors has the potential to recruit T cells to the tumor microenvironment and augment T-cell activation at the tumor site. NK cells have both direct anti-tumor activity and the capacity to secrete inflammatory cytokines and chemokines upon activation, enabling the cells to play a unique and critical role in regulating anti-tumor T cell activity. We sought to determine whether FT500, an off-the-shelf NK cell product derived from a clonal master pluripotent cell line, could synergize with CI to relieve local immunosuppression and enhance T-cell activation and recruitment to the tumor site. FT500 is universally negative for cell surface PD1, and expression of PDL1 on tumor lines had no discernable effect on FT500 cytotoxicity. Similarly, addition of PDL1 blocking antibody had no effect on FT500 cytotoxicity or degranulation, suggesting that FT500 is inherently resistant to PDL1-PD1 mediated inhibition. Additionally, activation of FT500 induced the secretion of soluble factors capable of enhancing T-cell activation, as evidenced by increased upregulation of CD69. We hypothesized that FT500 might also enhance CI by promoting recruitment of T cells to the tumor site. Using conventional in vitro transwell migration assays, we found that FT500 produced soluble factors that promoted the migration of activated T cells. Additional profiling confirmed FT500 production of a range of chemokines, including CCL3, CCL4, CXCL10 and CCL22. Furthermore, using an in vivo recruitment model, FT500 was able to recruit T cells out of the circulation and into the peritoneal cavity. Similarly, utilizing a three-dimensional tumor spheroid model in vitro, infiltration of T cells into tumor spheroids was significantly enhanced when combined with FT500, suggesting that FT500 can enhance tumor infiltration of T cells. Our data suggest that FT500 is a potent producer of chemokines and can facilitate the recruitment of T cells to the tumor site. In addition to its direct cytotoxic potential, FT500 is also able to enhance T-cell activation, suggesting an ability to synergize with CI to reduce tumor burden. Together, our data provide evidence supporting the combination of FT500, an off-the-shelf NK cell cancer immunotherapy, with CI to overcome checkpoint blockade resistance. Citation Format: Ryan Bjordahl, Sajid Mahmood, Svetlana Gaidarova, Ramzey Abujarour, Raedun Clarke, Laurel Stokely, Paul Rogers, Moyar Ge, Megan Robinson, Betsy Rezner, Tom Tong Lee, Bahram Valamehr. FT500, an off-the-shelf NK cell cancer immunotherapy derived from a master pluripotent cell line, enhances T-cell activation and recruitment to overcome checkpoint blockade resistance [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 3576.
- Published
- 2018
29. Genome-wide gain-of-function screen identifies novel regulators of pluripotency
- Author
-
Ramzey Abujarour, Sheng Ding, and Jem A. Efe
- Subjects
Pluripotent Stem Cells ,Homeobox protein NANOG ,Embryonal Carcinoma Stem Cells ,Time Factors ,MKI67IP ,Rex1 ,Biology ,Transfection ,Mice ,Genes, Reporter ,Cell Line, Tumor ,Animals ,Humans ,Cell Lineage ,Promoter Regions, Genetic ,Induced pluripotent stem cell ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Homeodomain Proteins ,Intracellular Signaling Peptides and Proteins ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,RNA-Binding Proteins ,Nanog Homeobox Protein ,Cell Differentiation ,Cell Biology ,Molecular biology ,Embryonic stem cell ,Gene Knockdown Techniques ,Molecular Medicine ,RNA Interference ,Stem cell ,Nucleophosmin ,Developmental Biology - Abstract
Pluripotent stem cells are characterized by the capacity to self-renew and to differentiate into all the cell types of the body. To identify novel regulators of pluripotency, we screened cDNA libraries (>30,000 clones) in P19 embryonal carcinoma cells for factors that modulate the expression of a luciferase reporter driven by the promoter of the pluripotency master regulator Nanog. Ninety confirmed hits activated the reporter and 14 confirmed hits inhibited the reporter by more than two-fold. The identified hits were evaluated by gain- and loss-of-functions approaches. The reporter-activating hits Timp2, Hig2, and Mki67ip promoted embryonic stem (ES) cell self-renewal when episomally overexpressed in ES cells, whereas the reporter-inhibiting hits PU.1/Spi1, Prkaca, and Jun induced differentiation of ES cells. Conversely, the knockdown of the activating hits Timp2, Mki67ip, Esrrg, and Dusp7 in ES cells induced differentiation, whereas the knockdown of the reporter-inhibiting hit PU.1/Spi1 led to inhibition of differentiation. One of the novel hits, the RNA-binding protein Mki67ip was further characterized, and found to be overexpressed in ES cells and in early development and downregulated during differentiation. The knockdown of Mki67ip led to the differentiation of ES cells, decreased growth rate, reduction in pluripotency markers, and induction of lineage-specific markers. In addition, colocalization and coimmunoprecipitation experiments suggest that Mki67ip promotes ES cell self-renewal via a mechanism involving nucleophosmin, a multifunctional nucleolar protein upregulated in stem cells and cancer.
- Published
- 2010
30. Clinical Translation of Pluripotent Cell-Derived Off-the-Shelf Natural Killer Cell Cancer Immunotherapy
- Author
-
Stacey K. Moreno, Scott Wolchko, Frank Cichocki, Brian Groff, Svetlana Gaidarova, Ryan Bjordahl, Dan S. Kaufman, Bruce R. Blazar, Betsy Rezner, Ramzey Abujarour, Thomas H. Lee, Sarah Cooley, Greg Bonello, Stewart Abbot, Raedun Clarke, Paul Rogers, Jeffrey S. Miller, David H. McKenna, Darin Sumstad, Megan Robinson, Daniel Shoemaker, Weijie Lan, and Bahram Valamehr
- Subjects
education.field_of_study ,medicine.medical_treatment ,Immunology ,Antigen presentation ,Population ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Natural killer cell ,Cell therapy ,Cytokine ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,medicine ,Cancer research ,Induced pluripotent stem cell ,education - Abstract
Natural killer (NK) cells represent a lineage of immune cells capable of direct cytotoxicity against tumor cells and are a critical source of key inflammatory cytokines such as interferon (IFN)-γ and tumor necrosis factor (TNF). NK cell function is often impaired in the setting of cancer, reducing the effectiveness of the endogenous immune system. The unique biological attributes of NK cells, including multifaceted effector function, tumor cell recognition independent of antigen presentation and target cell selectivity independent of HLA-matching, has enabled NK cells from a donor to be adoptively transferred to a patient for the treatment of cancer. This safe and effective administration of donor NK cells to patients validates their potential for broad use as part of an off-the-shelf cancer immunotherapy strategy, including in combination with monoclonal antibody and checkpoint inhibitor therapies. We have previously shown that human induced pluripotent stem cells (hiPSC) can be clonally-selected, cryopreserved and banked, and that these master pluripotent cell lines (MPCLs) can be used to renewably generate large clonal populations of NK cells. The use of MPCLs represents a highly-promising, off-the-shelf approach to cell-based cancer immunotherapy, with the potential to overcome many of the challenges and limitations of patient-sourced and donor-derived cell therapies. However, to clinically and commercially enable this off-the-shelf strategy, it is essential to efficiently and reproducibly differentiate MPCLs to fully-functional NK cells using a robust and scalable process that meets regulatory requirements. Here we describe a novel paradigm for the manufacture of hiPSC-derived NK (iNK) cells consisting of a well-defined, small molecule-driven, staged protocol that enables clinical translation and is compatible with current good manufacturing practice (cGMP) requirements. The manufacturing protocol is currently being transferred from the laboratories of Fate Therapeutics to Molecular and Cellular Therapeutics at the University of Minnesota, which is a state-of-the-art GMP/GTP compliant, full-service developer and manufacturer of cell- and tissue-based products. iNK cell therapy manufacture consists of four unique steps including: 1) the derivation and master cell banking of a clonal pluripotent cell line (> 95% SSEA4+/TRA181+ hiPSCs); 2) differentiation of the clonal pluripotent cell line towards hematopoietic progenitor cells (enriched for > 80% CD34+ cells); 3) differentiation and expansion of iNKs (Figure 1A, approximately 1,000-fold expansion in 14 days); and 4) freeze and thaw of drug product, comprised of a sufficiently pure homogenous population of iNKs (Figure 1B, > 95% CD45+, > 90% CD56+, minimal CD3+ T cells). Importantly, testing at both the molecular and culture stages demonstrate that no hiPSCs exist in the final drug product (limit of detection 1 hiPSC in 1.25 million iNK cells). This novel manufacturing paradigm supports the generation of significant numbers of iNK cells: approximately 1 million-fold cell expansion is achieved in less than 50 days, such that a very small population of hiPSCs can readily produce 1x1012 iNK cells. We estimate that this represents hundreds of doses of drug product per each manufacturing run (Figure 1A). The iNK cells display markedly augmented effector function relative to ex vivo expanded primary peripheral blood or cord blood NK cells with respect to cytokine release (IFN-γ and TNF) and cellular cytotoxicity against various leukemic and solid tumor-derived target cells including K562, Raji, A549 and SKOV3 (Figure 1C). To enable centralized manufacturing, we established a freeze and thaw strategy that supports greater than 85% viability with a recovery of greater than 80% iNK cells at twenty-four hours post-thaw. Because the freeze process uses an infusible medium formulation, we demonstrated in vitro and in vivo that the iNK cells maintain their efficacy post-thaw and can be immediately infused into patients. The manufacturing data presented herein support the filing of an Investigational New Drug application for an off-the-shelf iNK cell therapy product to treat advanced hematologic and solid tumor malignancies alone or in combination with monoclonal antibody and checkpoint inhibitor therapies. Disclosures Bjordahl: Fate Therapeutics: Employment, Equity Ownership. Gaidarova: Fate Therapeutics Inc.: Employment, Equity Ownership. Rogers: Fate Therapeutics Inc.: Employment, Equity Ownership. Clarke: Fate Therapeutics Inc.: Employment, Equity Ownership. Groff: Fate Therapeutics Inc.: Employment. Moreno: Fate Therapeutics Inc.: Employment. Abujarour: Fate Therapeutics Inc.: Employment. Robinson: Fate Therapeutics Inc.: Employment. Bonello: Fate Therapeutics Inc.: Employment. Lee: Fate Therapeutics Inc.: Employment, Equity Ownership. Lan: Fate Therapeutics Inc.: Employment, Equity Ownership. Rezner: Fate Therapeutics, Inc.: Employment. Abbot: Fate Therapeutics Inc.: Employment. Wolchko: Fate Therapeutics Inc.: Employment. Kaufman: Fate Therapeutics: Consultancy, Research Funding. Valamehr: Fate Therapeutics: Employment, Equity Ownership. Miller: Oxis Biotech: Consultancy; Celegene: Consultancy; Fate Therapeutics: Consultancy, Research Funding.
- Published
- 2017
31. Generation of Human-Induced Pluripotent Stem Cells in the Absence of Exogenous Sox2
- Author
-
Sheng Ding, Hans R. Schöler, Jin Young Joo, Hongyan Zhou, Ramzey Abujarour, Tongxiang Lin, Saiyong Zhu, Alberto Hayek, Ergeng Hao, and Wenlin Li
- Subjects
Pluripotent Stem Cells ,Pyridines ,Cellular differentiation ,Cell Culture Techniques ,Kruppel-Like Transcription Factors ,Biology ,Article ,Glycogen Synthase Kinase 3 ,Kruppel-Like Factor 4 ,SOX2 ,Animals ,Humans ,Induced pluripotent stem cell ,Protein Kinase Inhibitors ,Cells, Cultured ,Induced stem cells ,SOXB1 Transcription Factors ,fungi ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,Cellular Reprogramming ,Embryo, Mammalian ,Embryonic stem cell ,Coculture Techniques ,Cell biology ,Pyrimidines ,KLF4 ,embryonic structures ,Molecular Medicine ,Stem cell ,Octamer Transcription Factor-3 ,Reprogramming ,Developmental Biology - Abstract
Induced pluripotent stem cell technology has attracted enormous interest for potential application in regenerative medicine. Here, we report that a specific glycogen synthase kinase 3 (GSK-3) inhibitor, CHIR99021, can induce the reprogramming of mouse embryonic fibroblasts transduced by only two factors, Oct4 and Klf4. When combined with Parnate (also named tranylcypromine), an inhibitor of lysine-specific demethylase 1, CHIR99021 can cause the reprogramming of human primary keratinocyte transduced with the two factors, Oct4 and Klf4. To our knowledge, this is the first time that human iPS cells have been generated from somatic cells without exogenous Sox2 expression. Our studies suggest that the GSK-3 inhibitor might have a general application to replace transcription factors in both mouse and human reprogramming. Disclosure of potential conflicts of interest is found at the end of this article.
- Published
- 2009
32. Generation of skeletal muscle cells from pluripotent stem cells: advances and challenges
- Author
-
Ramzey Abujarour and Bahram Valamehr
- Subjects
Cell type ,Satellite Cells, Skeletal Muscle ,Somatic cell ,Mini Review ,Cellular differentiation ,skeletal muscle cells ,Biology ,MyoD ,Cell and Developmental Biology ,pluripotent ,skeletal muscle ,Progenitor cell ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,satellite cells ,Myogenic Differentiation ,Cell Biology ,Anatomy ,Cell sorting ,Cell biology ,Myogenic Regulatory Factors ,lcsh:Biology (General) ,IPSC ,Myogenic regulatory factors ,pax7 ,Developmental Biology - Abstract
Human pluripotent stem cells (hPSCs) possess unlimited proliferative potential while maintaining the ability to differentiate into any cell type including skeletal muscle cells (SMCs). hPSCs are amenable to genetic editing and can be derived from patient somatic cells, and thus represent a promising option for cell therapies for the treatment of degenerative diseases such as muscular dystrophies. There are unresolved challenges however associated with the derivation and scale-up of hPSCs and generation of differentiated cells in large quantity and high purity. Reported myogenic differentiation protocols are long, require cell sorting and/or rely on ectopic expression of myogenic master regulators. More recent advances have been made with the application of small molecules to enhance the myogenic differentiation efficiency and the identification of more selective markers for the enrichment of myogenic progenitors with enhanced regenerative potential. Here we review the field of myogenic differentiation and highlight areas requiring further research.
- Published
- 2015
33. Abstract 3755: Renewable and genetically engineered natural killer cells for off-the-shelf adoptive cellular immunotherapy
- Author
-
Ramzey Abujarour, Svetlana Gaidarova, Stewart Abbot, Paul Rogers, Scott Wolchko, Bruce R. Blazar, Bahram Valamehr, Daniel Shoemaker, Brian Groff, Jeffrey S. Miller, Bruce Walcheck, Dave Robbins, Weijie Lan, Frank Cichocki, Raedun Clarke, Betsy Rezner, Thomas H. Lee, Sarah Cooley, Ryan Bjordahl, Greg Bonello, Stacey K. Moreno, and Matthieu Bauer
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,medicine.drug_class ,medicine.medical_treatment ,CD16 ,Biology ,Monoclonal antibody ,NKG2D ,Directed differentiation ,Immune system ,Oncology ,Cancer immunotherapy ,Immunology ,medicine ,Cancer research ,Induced pluripotent stem cell - Abstract
The unique attributes of a combinatorial tumor recognition system, diminished off-tumor cytotoxicity, and multifaceted effector function make natural killer (NK) cells a prime candidate for a universal approach to cancer immunotherapy. In addition, NK cells are the principal mediator of antibody-directed cellular cytotoxicity (ADCC). However, NK cell function is often impaired in the setting of cancer, reducing the effectiveness of the endogenous immune system and the therapeutic efficacy of monoclonal antibodies. To address the need for advanced and combinatorial cancer therapies, we developed a unique and effective strategy to create a renewable source of engineered “off-the-shelf” NK cells with augmented function, including enhanced ADCC and persistence. Key challenges associated with genetic editing, limited expansion, persistence and variability of peripheral blood (PB)-derived NK cells were overcome by utilizing our induced pluripotent stem cell (iPSC) technology as the unlimited starting material for the reproducible and consistent derivation of engineered NK cells. Through targeted transgene integration, we produced a clonal iPSC master cell line to continuously produce NK cells engineered to uniformly express a high affinity, non-cleavable version of CD16 (hnCD16-NK). In directed differentiation, the hnCD16-NK cells displayed homogeneous expression of CD16 (>95%) and a mature CD56+ NK cell phenotype, as exhibited by expression of KIR, NCRs, DNAM-1, and NKG2D. In contrast to endogenous CD16 expression, the engineered hnCD16 molecule was shown to be cleavage resistant upon NK cell activation (>95% CD16+ hnCD16-NK vs. Citation Format: Ryan Bjordahl, Frank Cichocki, Raedun Clarke, Svetlana Gaidarova, Brian Groff, Paul Rogers, Stacey Moreno, Ramzey Abujarour, Greg Bonello, Tom Lee, Weijie Lan, Matthieu Bauer, Dave Robbins, Betsy Rezner, Sarah Cooley, Bruce Walcheck, Stewart Abbot, Bruce Blazar, Scott Wolchko, Daniel Shoemaker, Jeffrey S. Miller, Bahram Valamehr. Renewable and genetically engineered natural killer cells for off-the-shelf adoptive cellular immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3755. doi:10.1158/1538-7445.AM2017-3755
- Published
- 2017
34. Abstract 609: Overcoming host histocompatibility barrier to create a renewable source of off-the-shelf effector lymphocytes for adoptive immunotherapy
- Author
-
Raedun Clarke, Michelle Burrascano, Ramzey Abujarour, Matthieu Bauer, Brian Groff, Ryan Bjordahl, Greg Bonello, Tom Tong Lee, Svetlana Gaidarova, Weijie Lan, Megan Robinson, Daniel Shoemaker, Stewart Abbot, Scott Wolchko, Bob Valamehr, and Jeffrey Sasaki
- Subjects
Cancer Research ,Adoptive cell transfer ,Lymphocyte ,Human leukocyte antigen ,Biology ,Haematopoiesis ,Immune system ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Cytotoxic T cell ,Induced pluripotent stem cell ,CD8 - Abstract
Encouraging clinical outcomes in autologous cellular immunotherapy have garnered hope and excitement. However, limitations of patient-derived cancer immunotherapies remain to be addressed to deliver reliable and efficacious therapies with broader applicability. Induced pluripotent stem cells (iPSCs) are a unique, renewable source for the continuous generation of cellular therapeutics and represent a highly promising approach for overcoming many of the limitations of autologous therapy. To advance the promise of iPSC technology as an “off-the-shelf” (OTS) source of cellular therapeutics, several considerations need to be addressed. Ensuring the persistence of allogeneic OTS therapies after adoptive cell transfer across histocompatibility barriers is a key requirement. Establishing a master cell line from genetically engineered clonal iPSC lines with the capacity to continuously generate homogenous populations of highly functional effector cells will also be necessary. Here we demonstrate a comprehensive approach for the generation of immune tolerant effector cells derived from a genetically engineered iPSC master cell line. We successfully combined deletion of classical human leukocyte antigen molecules with expression of immunosuppressive proteins to generate clonal iPSC lines with the ability to escape immune rejection. Utilizing in vitro quantitative live cell analysis we show that OTS-iPSCs elicit a significantly decreased cytotoxic response from both peripheral blood (PB)-NK cells (47.9 vs. 91.4% survival at 3:1 E:T ratio) and PB-T cells (>2.7-fold greater number of OTS-iPSC derived cells remaining at 88 hrs). Additionally, mixed lymphocyte reactions employing unfractionated PB mononuclear cells resulted in significantly decreased activation and proliferation of CD8+ T cells (63.4 vs. 29.6%), CD4+ T cells (70.9 vs. 17.3%) and NK cells (46.8 vs. 11.6%). In preclinical mouse models we demonstrate that OTS-iPSCs exhibit improved persistence in vivo. Bilateral engraftments were established in non-conditioned, fully immune-competent recipient mice using luciferized wildtype and OTS-iPSCs. Daily bioluminescence imaging revealed a significant increase in persistence of OTS-iPSCs during the 48-196 hour post injection window (>5.5 fold greater luminescence at 96 hrs). Using our potent chemically-defined stage-specific monolayer hematopoietic differentiation platform, we demonstrate that OTS-iPSC derived CD34 expressing hematopoietic cells are reproducibly scaled and readily give rise to functional lymphocytes carrying the engineered targeted modality in a homogenous manner (95 +/- 5%). The outlined preclinical data illustrate that iPSCs are an ideal renewable source for OTS hematopoietic cell-based immunotherapies and represent a potentially exponential advancement in adoptive immunotherapy. Citation Format: Raedun L. Clarke, Matthieu Bauer, Ryan Bjordahl, Jeffrey Sasaki, Brian Groff, Svetlana Gaidarova, Tom Tong Lee, Weijie Lan, Michelle Burrascano, Ramzey Abujarour, Greg Bonello, Megan Robinson, Stewart Abbot, Scott Wolchko, Daniel Shoemaker, Bob Valamehr. Overcoming host histocompatibility barrier to create a renewable source of off-the-shelf effector lymphocytes for adoptive immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 609. doi:10.1158/1538-7445.AM2017-609
- Published
- 2017
35. Pluripotent stem cell miRNAs and metastasis in invasive breast cancer
- Author
-
Stefano, Volinia, Gerard, Nuovo, Alessandra, Drusco, Stefan, Costinean, Ramzey, Abujarour, Caroline, Desponts, Michela, Garofalo, Raffaele, Baffa, Rami, Aeqilan, Kati, Maharry, Maria Elena, Sana, Maria Elena Sana Ramiro, Garzon, Gianpiero, Di Leva, Pierluigi, Gasparini, Paola, Dama, Jlenia, Marchesini, Marco, Galasso, Marco, Manfrini, Carlotta, Zerbinati, Fabio, Corrà, Timothy, Wise, Sylwia E, Wojcik, Maurizio, Previati, Flavia, Pichiorri, Nicola, Zanesi, Hansjuerg, Alder, Jeff, Palatini, Kay F, Huebner, Charles L, Shapiro, Massimo, Negrini, Andrea, Vecchione, Anne L, Rosenberg, Carlo M, Croce, and Ramiro, Garzon
- Subjects
Oncology ,Pluripotent Stem Cells ,Cancer Research ,medicine.medical_specialty ,Socio-culturale ,Breast Neoplasms ,Stem cell marker ,Article ,Metastasis ,Breast cancer ,stem cells ,Cancer stem cell ,Internal medicine ,medicine ,Humans ,Breast ,Induced pluripotent stem cell ,microRNA ,biology ,CD44 ,Carcinoma, Ductal, Breast ,medicine.disease ,MicroRNAs ,Lymphatic Metastasis ,Cancer cell ,biology.protein ,Neoplastic Stem Cells ,Female ,Stem cell - Abstract
Background The purpose of this study is to determine whether microRNA for pluripotent stem cells are also expressed in breast cancer and are associated with metastasis and outcome. Methods We studied global microRNA profiles during differentiation of human embryonic stem cells (n =26) and in breast cancer patients (n = 33) and human cell lines (n = 35). Using in situ hybridization, we then investigated MIR302 expression in 318 untreated breast cancer patients (test cohort, n = 22 and validation cohort, n = 296). In parallel, using next-generation sequencing data from breast cancer patients (n = 684), we assessed microRNA association with stem cell markers. All statistical tests were two-sided. Results In healthy tissues, the MIR302 (high)/MIR203 (low) asymmetry was exclusive for pluripotent stem cells. MIR302 was expressed in a small population of cancer cells within invasive ductal carcinoma, but not in normal breast (P < .001). Furthermore, MIR302 was expressed in the tumor cells together with stem cell markers, such as CD44 and BMI1. Conversely, MIR203 expression in 684 breast tumors negatively correlated with CD44 (Spearman correlation, Rho = -0.08, P = .04) and BMI1 (Rho = -0.11, P = .004), but positively correlated with differentiation marker CD24 (Rho = 0.15, P < .001). Primary tumors with lymph node metastasis had cancer cells showing scattered expression of MIR302 and widespread repression of MIR203. Finally, overall survival was statistically significantly shorter in patients with MIR302-positive cancer cells (P = .03). Conclusions In healthy tissues the MIR302(high)/MIR203(low) asymmetry was characteristic of embryonic and induced pluripotency. In invasive ductal carcinoma, the MIR302/MIR203 asymmetry was associated with stem cell markers, metastasis, and shorter survival.
- Published
- 2014
36. Genetic Engineering of Pluripotent Cells for the Continuous Derivation of Off-the-Shelf Effector Lymphocytes with Enhanced Therapeutic Persistence By Overcoming the Host Histocompatibility Barrier
- Author
-
Stewart Abbot, Scott Wolchko, Tom Tong Lee, Raedun Clarke, Matthieu Bauer, Michelle Burrascano, Greg Bonello, Svetlana Gaidarova, Ryan Bjordahl, Brian Groff, Bahram Valamehr, Weijie Lan, Ramzey Abujarour, Jeffrey Sasaki, Megan Robinson, and Daniel Shoemaker
- Subjects
education.field_of_study ,Effector ,medicine.medical_treatment ,Immunology ,Population ,Cell Biology ,Hematology ,Immunotherapy ,Human leukocyte antigen ,Biology ,Biochemistry ,Immune system ,Humanized mouse ,medicine ,Cancer research ,Induced pluripotent stem cell ,education ,Reprogramming - Abstract
Encouraging clinical outcomes in autologous cellular immunotherapy have garnered hope and excitement. However, considerable challenges and limitations of patient-derived cancer immunotherapies remain and need to be addressed in order to consistently deliver reliable and efficacious therapies with broadened applicability. Human induced pluripotent stem cells (hiPSCs) are a unique, renewable source for the continuous generation of cellular therapeutics for the treatment of hematological and non-hematological malignancies, and represent a highly promising approach for overcoming many of the limitations of autologous therapy. To advance the promise of hiPSC technology as an "off-the-shelf" source of cellular therapeutics, several considerations need to be addressed. Enabling cell transfer across histocompatibility barriers to permit persistence and therapeutic efficacy in an allogeneic setting is a key requirement. In addition to improving persistence, the ability to overcome histocompatibility barriers may facilitate multi-dosing regimens which may be a requirement in more advanced and complicated disease settings. Genetic incompatibilities between donor and recipient among the classical human leukocyte antigen (HLA) molecules is the leading cause of alloresponse by the host immune system and is currently mitigated by immunosuppressive strategies. Unfortunately, this treatment strategy is not only a stressful event for the patient but also damages the endogenous immune system, compromising the patient's ability to continue to fight the disease and opportunistic infections. Genetic editing of the HLA genes to generate histocompatible universal cell products is a viable opportunity that is currently being investigated. In addition to selective editing of unique genes to avoid a T cell mediated alloresponse, additional considerations such as natural killer (NK) cell-mediated rejection will need to be addressed. We have previously demonstrated that our proprietary reprogramming platform supports efficient and rapid derivation of clonal hiPSC lines with properties indicative of the naïve state of pluripotency. In addition to maintaining a homogeneous renewable population of hiPSCs, our platform is amenable to precise multi-gene and multi-loci targeted engineering at the single cell level, in both nuclease -dependent and -independent strategies. Furthermore, we have shown through small molecule-guided differentiation protocols, these highly-stable pluripotent cell lines can be banked and repeatedly tapped to consistently produce homogenous populations of immune cells with enhanced effector properties. Here we demonstrate a multi-faceted and comprehensive approach for the generation of immune tolerant hiPSCs and hiPSC-derived immune effector cells. We successfully combined deletion of classical HLA molecules with enforced expression of robust immunosuppressive proteins, including non-classical HLA molecules, to generate clonal hiPSC lines with the ability to escape immune rejection for "off-the-shelf" (OTS-hiPSCs) cellular immunotherapy. Utilizing in vitro real-time quantitative live cell analysis we determined that OTS-hiPSCs elicit a significantly decreased cytotoxic response from both activated peripheral blood (PB)-NK cells and primed PB-T cells compared to wildtype controls. Furthermore we demonstrate that OTS-hiPSCs exhibit improved persistence in xenograft studies in vivo. Bilateral teratomas were formed in a non-conditioned, fully immune-competent recipient mice using luciferized wildtype and OTS-hiPSCs. Daily bioluminescence imaging over a period of 7 days revealed a significant increase (>50 fold difference) in persistence of OTS-hiPSCs compared to wildtype hiPSCs during the 60-144 hour post injection window. Lastly we demonstrate that OTS-hiPSCs can successfully differentiate into functional effector lymphocytes using our potent chemically-defined monolayer hematopoietic differentiation platform. Our current studies focus on the functional characterization of OTS-hiPSC-derived effector lymphocytes in humanized mouse models and generating increased potency of OTS-hiPSC-derived effector lymphocytes through precise genetic engineering of antigen targeting and costimulatory proteins to create and optimized source of "off-the-shelf" cell-based immunotherapies. Disclosures Bauer: Fate Therapeutics: Employment. Clarke:Fate Therapeutics: Employment. Sasaki:Fate Therapeutics: Employment. Groff:Fate Therapeutics: Employment. Lee:Fate Therapeutics: Employment. Lan:Fate Therapeutics: Employment. Abujarour:Fate Therapeutics: Employment. Bonello:Fate Therapeutics: Employment. Burrascano:Fate Therapeutics: Employment. Robinson:Fate Therapeutics: Employment. Bjordahl:Fate Therapeutics, Inc: Employment. Gaidarova:Fate Therapeutics: Employment. Abbot:Fate Therapeutics: Employment. Wolchko:Fate Therapeutics: Employment. Shoemaker:Fate Therapeutics: Employment, Equity Ownership. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2016
37. Off-the-Shelf Natural Killer Cell Immunotherapy for Enhanced Antibody Directed Cellular Cytotoxicity
- Author
-
Greg Bonello, Matthieu Bauer, Svetlana Gaidarova, Ryan Bjordahl, Bahram Valamehr, Betsy Rezner, Scott Wolchko, Brian Groff, Raedun Clarke, Stacey K. Moreno, Ramzey Abujarour, Jeff Sasaki, Michelle Burrascano, Weijie Lan, Megan Robinson, Daniel Shoemaker, William Kim, Tom Tong Lee, David J. Robbins, Stewart Abbot, and Paul Rogers
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,medicine.medical_treatment ,Immunology ,Antigen presentation ,02 engineering and technology ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Biochemistry ,Chimeric antigen receptor ,0104 chemical sciences ,Natural killer cell ,Cell therapy ,medicine.anatomical_structure ,Cancer stem cell ,medicine ,Cancer research ,0210 nano-technology ,Induced pluripotent stem cell - Abstract
Natural Killer (NK) cells play a crucial role in immunosurveillance and form a first line of defense against cancer. In comparison to other lymphocytes, NK cells are unique in their capability to elicit tumoricidal responses without the need for antigen presentation or prior sensitization. Clinical data from bone marrow transplant and allogeneic NK immunotherapy suggest that MHC mismatch is advantageous in promoting graft-versus-leukemia without eliciting graft-versus-host, providing evidence that NK cells hold promisa as an allogeneic, universal immunotherapeutic. Further, the anti-tumor effect of many monoclonal antibodies is mediated through binding of the low-affinity Fc receptor CD16 on NK cells, which induces tumor cell killing through antibody-dependent cellular cytotoxicity (ADCC). Thus, NK cells represent a unique source of effector cells that can be combined with monoclonal antibodies, bispecific engagers or chimeric antigen receptors to direct tumor specificity and enhance cytotoxicity. Despite the significant potential of NK cell therapy, current clinical practices are limited by the need for large numbers of healthy NK cells, lack of in vivo persistence, and a burdensome manufacturing strategy that requires donor cell extraction, modulation, expansion and re-introduction per each patient. The ability to generate universally histocompatible and genetically-enhanced NK cells from continuously renewable human induced pluripotent stem cell (hiPSC) lines offers the potential to develop a true "off-the-shelf" cellular immunotherapy. While NK differentiation from hiPSC has been demonstrated, the clonal derivation of engineered hiPSCs to improve effector function has been challenging and the scalability and robustness of the differentiation method has been limited by skewed development towards primitive hematopoiesis and the cumbersome use of embryoid bodies. Here we highlight our "off-the-shelf" NK cell therapy preclinical program by demonstrating robust and highly scalable generation of functionally mature, genetically targeted and universally histocompatible NK cells. This program utilizes our previously described naïve hiPSC platform where we uniquely create clonal lines of precisely engineered, renewable hiPSCs and drive definitive hematopoiesis in a highly scalable manner. Because hiPSC differentiation is lineage directed, minimal cellular contamination is seen, including the lack of T and B cells, in the final product. Through precise genetic engineering of naïve hiPSC lines, we have engineered HLA-class I deficient NK cells uniformly expressing a high affinity, non-cleavable version of the Fc receptor CD16 (NcHaCD16-NK). The hiPSC-derived NcHaCD16-NKs display markers of maturity, including CD16, KIR, NCRs, and CD94. When compared to conventional cord blood and peripheral blood sourced NK cells, NcHaCD16-NKs exhibit superior cytotoxicity and production of effector cytokines in response to both solid and liquid tumor cell challenge in vitro. NcHaCD16-NKs exhibit augmented cytokine response following Fc-mediated stimulation, demonstrating function competence of the engineered CD16 construct. Because surface expression of CD16 is resistant to activation-induced shedding, NcHaCD16-NKs continuously maintain enhanced ADCC while retaining the capacity for general cytotoxicity. Importantly, the hiPSC-derived hematopoietic cells can be successfully cryopreserved and banked, serving as a highly-stable cell bank for subsequent therapeutic use. Preliminary data also shows NcHaCD16-NKs elicit preferred specificity for cancer stem cells as defined by expression of ALDH1 and surface markers such as CD24. In conclusion, the outlined preclinical data demonstrate the potential therapeutic utility of NK cells developed via precision genetic engineering of a renewable, scalable hiPSC platform, and highlights the therapeutic value of NcHaCD16-NKs as an ideal ADCC-mediated "off-the-shelf" NK cell-based immunotherapeutic product with augmented persistence, anti-tumor capacity and preclinical efficacy. Disclosures Bjordahl: Fate Therapeutics, Inc: Employment. Clarke:Fate Therapeutics: Employment. Gaidarova:Fate Therapeutics: Employment. Groff:Fate Therapeutics: Employment. Rogers:Fate Therapeutics, Inc: Employment. Moreno:Fate Therapeutics, Inc.: Employment, Equity Ownership. Abujarour:Fate Therapeutics, Inc.: Employment. Bonello:Fate Therapeutics, Inc.: Employment. Lee:Fate Therapeutics: Employment. Lan:Fate Therapeutics: Employment. Burrascano:Fate Therapeutics: Employment. Bauer:Fate Therapeutics: Employment. Robinson:Fate Therapeutics: Employment. Sasaki:Fate Therapeutics, Inc.: Employment. Kim:Fate Therapeutics, Inc.: Employment. Robbins:Fate Therapeutics: Employment, Equity Ownership. Rezner:Fate Therapeutics, Inc: Employment, Equity Ownership. Abbot:Fate Therapeutics: Employment. Wolchko:Fate Therapeutics: Employment. Shoemaker:Fate Therapeutics: Employment, Equity Ownership. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2016
38. Genetically Enhanced Pluripotent Stem Cell-Derived T Lymphocytes for Off-the-Shelf Cellular Immunotherapy
- Author
-
Stewart Abbot, Matthieu Bauer, Scott Wolchko, Heather Foster, Raedun Clarke, Bahram Valamehr, Brian Groff, Tom Tong Lee, Greg Bonello, Ramzey Abujarour, Weijie Lan, Michelle Burrascano, Jeffrey Sasaki, Megan Robinson, Daniel Shoemaker, and David J. Robbins
- Subjects
education.field_of_study ,T cell ,medicine.medical_treatment ,Immunology ,Population ,Lymphocyte differentiation ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,Biochemistry ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,medicine ,Progenitor cell ,Induced pluripotent stem cell ,education ,Reprogramming - Abstract
Cellular immunotherapies are poised to transform the treatment of cancer and immunological disorders. In the most promising setting to date, genetic modification to the T lymphocytes in the form of chimeric antigen receptors (CAR) has dramatically increased therapeutic efficacy with reported initial complete remission rates in acute lymphoblastic leukemia ranging between 80-100%. However, pressing challenges remain to be solved to ensure that engineered T-cell immunotherapies can be cost-effectively and consistently manufactured, and safely and reliably delivered at the scale necessary to support wide patient base commercialization. Human induced pluripotent stem cell (hiPSC) derived T lymphocytes represent a unique, renewable source of genetically engineered T cells for "off-the-shelf" immunotherapy. Through the precise genetic engineering at the hiPSC stage, clonal and uniform populations of modified cell lines can be banked and reliably tapped into on demand to generate highly efficacious T cells for therapeutic applications. Although great progress has been made, several challenges need to be addressed including the ability to enhance effector function through genome-engineering of persistence, targeting, histocompatibility and controlled safety mechanisms at the hiPSC juncture while retaining the capacity to efficiently and reproducibly generate the intricate stages of lymphocyte development in an accurate and scalable process. We have previously demonstrated that our proprietary reprogramming platform supports efficient and rapid derivation of clonal hiPSC lines with properties indicative of the naïve state of pluripotency. In addition to maintaining a homogeneous population of hiPSCs, our platform enables efficient multi-gene and multi-loci targeted engineering at a single cell level resulting in clonal population of pluripotent cell lines with desired genetic attributes. Here we will provide an update on our "off-the-shelf" T-cell immunotherapy preclinical program where engineered hiPSC lines are uniquely used as the renewable starting material. We will also highlight our novel differentiation platform to derive definitive hematopoietic progenitor cells termed hemogenic endothelium (HE); a well-defined, small molecule-driven, staged process that is currently being translated into cGMP (current good manufacturing practice) settings. The highly efficient differentiation system (on average >65% hiPSC to CD34 conversion) delivers approximately 100 CD34+ HE cells per each input hiPSC, representing a highly scalable process that is further expanded during lymphocyte differentiation and maturation. To validate that the iCD34+ HE is definitive in nature we demonstrate that during further hematopoietic differentiation the emerging CD43+ hematopoietic cells exhibit Notch dependency and high expression of key genes such as MYB and the HOXA cluster, found only in definitive hematopoietic progenitors. The hiPSC-derived HE exhibits multi-lineage potential and can be successfully cryopreserved and banked, serving as a highly-stable cell bank for subsequent therapeutic use. Through genetic modifications at the single cell hiPSC stage, we confer antigen-specificity via the expression of temporally inducible CARs as premature expression of CAR proteins during in vitro differentiation has been found to skew development towards innate-lymphoid like lineages. Utilizing our stage-specific hematopoietic differentiation platform we have identified the optimal developmental window to induce the expression of CAR proteins to maintain optimal differentiation towards functional effector lymphocytes. The hiPSC-derived engineered T lymphocytes are currently under preclinical investigation for in vitro and in vivo effector function including thymic rejuvenation, T cell repertoire repopulation, target specific recognition and enhanced killing potential. Preliminary data suggests that hiPSC-derived lymphocytes are functional and can home to their respective niche to support initial repopulation in vivo. Our study continues to support that naïve hiPSCs are an ideal renewal source for "off-the-shelf" hematopoietic cell-based immunotherapies and represent a potentially exponential advancement in adoptive T cell therapy. Disclosures Clarke: Fate Therapeutics: Employment. Groff:Fate Therapeutics: Employment. Sasaki:Fate Therapeutics: Employment. Bauer:Fate Therapeutics: Employment. Lee:Fate Therapeutics: Employment. Lan:Fate Therapeutics: Employment. Burrascano:Fate Therapeutics: Employment. Abujarour:Fate Therapeutics: Employment. Bonello:Fate Therapeutics: Employment. Robinson:Fate Therapeutics: Employment. Foster:Fate Therapeutics: Employment, Equity Ownership. Robbins:Fate Therapeutics: Employment, Equity Ownership. Wolchko:Fate Therapeutics: Employment. Shoemaker:Fate Therapeutics: Employment, Equity Ownership. Abbot:Fate Therapeutics: Employment. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2016
39. Efficient Site-Specific Multi-Gene Engineering of Renewable Pluripotent Cells for Generation of Off-the-Shelf Hematopoietic Immunotherapeutics
- Author
-
Miguel Meza, Weijie Lan, Tom Tong Lee, Bahram Valamehr, Chris Lynn, Raedun Clarke, Ramzey Abujarour, Greg Bonello, David J. Robbins, Stewart Abbot, Betsy Rezner, Christopher Truong, Megan Robinson, and Daniel Shoemaker
- Subjects
Genetics ,Cell type ,education.field_of_study ,Transgene ,Immunology ,Population ,Cell Biology ,Hematology ,Computational biology ,Biology ,Suicide gene ,Biochemistry ,Phenotype ,Haematopoiesis ,Directed differentiation ,Ectopic expression ,education - Abstract
Human induced pluripotent stem cells (hiPSCs) are a unique population of cells that can serve as an unlimited source for "off-the-shelf" cellular immunotherapeutics. Similar to master cell lines used in the manufacture of monoclonal antibodies, engineered hiPSC lines have the potential to serve as a renewable cell source for the consistent manufacture of homogeneous populations of effector cells for the treatment of thousands of patients. However, the creation of an effective master line is largely dependent on the ability to genetically edit hiPSCs in a precise, efficient and clonal manner. Furthermore, the genetically edited hiPSCs must maintain their inherent ability to continuously self-renew while retaining ability to express engineered modalities upon directed differentiation to the cell type of choice. We have previously reported the use of stage-specific media compositions to enable the footprint-free generation and long-term maintenance of single cell naïve hiPSCs with enhanced clonogenicity, an attribute critical for the derivation of engineered single cell-derived lines. Here we demonstrate the use of our naïve hiPSC platform to precisely introduce, in a site-specific manner, multiple genes into multiple safe harbor loci. By combining our single-cell naïve hiPSC platform with different nuclease-independent and -dependent strategies, we are able to generate large numbers of precisely engineered iPSC clones. The single cell-derived hiPSC clones were subsequently screened in a multiplexed fashion for successful multi-parameter engineering, maintained pluripotency and propensity for differentiation with lack of undesired phenotypes and genomic alterations. Using this approach, we derived individual clones containing a uniform population (>99%) of multi-engineered modalities consisting of tumor targeting, a controllable safety switch and a tracking marker. Moreover, we show that engineered modalities are expressed in undifferentiated and differentiated hiPSCs, including being expressed in >95% of both CD34 positive hematopoietic progenitor cells and CD56 positive natural killer cells. Furthermore, we have generated hiPSC clones with dual suicide genes (inducible Caspase 9 (iCasp9) and Herpes simplex virus thymidine kinase (HSV-TK)) targeted into two independent safe harbor loci, in both mono- and bi-allelic manner. The dual-targeted hiPSC clones were confirmed to have specific insertions into the predicted sites and were screened to exclude random insertions. Concurrent activation of both suicide genes led to complete elimination of engineered hiPSCs and no treatment-refractory cells were observed unlike the case when only one suicide gene was activated. In addition to robust targeted insertions, we were able to generate small insertions and deletions in up to 70% of naïve hiPSCs without selection and homozygous knockout of gene of interest in 100% of cells after selection. Finally, we will discuss efforts to temporally synchronize ectopic gene expression through endogenously regulated promoters by simultaneous endogenous gene disruption and transgene insertion. Overall, we show our naïve hiPSC platform is an ideal renewable source to efficiently generate, genetically engineer, isolate and bank clonally-derived homogenous population of pluripotent cells. These highly-stable pluripotent clonal banks can be repeatedly tapped to facilitate the consistent production of homogenous populations of potent, persistent, scalable and safer off-the-shelf cellular immunotherapeutics. Disclosures Abujarour: Fate Therapeutics: Employment. Lan:Fate Therapeutics: Employment. Lee:Fate Therapeutics: Employment. Bonello:Fate Therapeutics: Employment. Meza:Fate Therapeutics: Employment, Equity Ownership. Robinson:Fate Therapeutics: Employment. Clarke:Fate Therapeutics: Employment. Truong:Fate Therapeutics: Employment, Equity Ownership. Robbins:Fate Therapeutics: Employment, Equity Ownership. Rezner:Fate Therapeutics, Inc: Employment, Equity Ownership. Abbot:Fate Therapeutics: Employment. Shoemaker:Fate Therapeutics: Employment, Equity Ownership. Valamehr:Fate Therapeutics, Inc: Employment.
- Published
- 2016
40. Platform for induction and maintenance of transgene-free hiPSCs resembling ground state pluripotent stem cells
- Author
-
Megan Robinson, Thuy Le, David L. Robbins, Florin Vranceanu, Betsy Rezner, Peter Flynn, Tom Tong Lee, Michael J. Fitch, Bahram Valamehr, Amanda Medcalf, and Ramzey Abujarour
- Subjects
Pluripotent Stem Cells ,Transgene free ,Cellular differentiation ,Population ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Abnormal Karyotype ,Biology ,Biochemistry ,Genomic Instability ,Article ,Cell therapy ,Mice ,SOX2 ,Genetics ,Animals ,Cluster Analysis ,Humans ,Transgenes ,education ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Cells, Cultured ,Embryonic Stem Cells ,Chromosome Aberrations ,lcsh:R5-920 ,education.field_of_study ,Gene Expression Profiling ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Cellular Reprogramming ,Molecular biology ,Cell biology ,lcsh:Biology (General) ,Cell Transdifferentiation ,Female ,lcsh:Medicine (General) ,Reprogramming ,Developmental Biology ,Transcription Factors - Abstract
Summary Cell banking, disease modeling, and cell therapy applications have placed increasing demands on hiPSC technology. Specifically, the high-throughput derivation of footprint-free hiPSCs and their expansion in systems that allow scaled production remains technically challenging. Here, we describe a platform for the rapid, parallel generation, selection, and expansion of hiPSCs using small molecule pathway inhibitors in stage-specific media compositions. The platform supported efficient and expedited episomal reprogramming using just OCT4/SOX2/SV40LT combination (0.5%–4.0%, between days 12 and 16) in a completely feeder-free environment. The resulting hiPSCs are transgene-free, readily cultured, and expanded as single cells while maintaining a homogeneous and genomically stable pluripotent population. hiPSCs generated or maintained in the media compositions described exhibit properties associated with the ground state of pluripotency. The simplicity and robustness of the system allow for the high-throughput generation and rapid expansion of a uniform hiPSC product that is applicable to industrial and clinical-grade use., Graphical Abstract, Highlights • FRM supports highly efficient minimal factor episomal reprogramming • Clonal derivation of hiPSCs through high-resolution single-cell sorting • FMM supports long-term maintenance of genomically stable, transgene-free hiPSCs • Maintenance of homogeneous pluripotent culture resembling the ground state, Cell therapy and banking applications have placed increasing demands on hiPSC technology. Here, Flynn and colleagues demonstrate a simple multiplex selection and maintenance platform utilizing stage-specific small molecule media additives and flow cytometry sorting to derive minimal factor episomal hiPSCs that are genomically stable and resemble ground state pluripotent stem cells. This platform may serve as a path to generate clinically relevant hiPSCs.
- Published
- 2013
41. Optimized Surface Markers for the Prospective Isolation of High-Quality hiPSCs using Flow Cytometry Selection
- Author
-
Florin Vranceanu, Megan Robinson, Bahram Valamehr, Betsy Rezner, Ramzey Abujarour, and Peter Flynn
- Subjects
Homeobox protein NANOG ,Stage-Specific Embryonic Antigens ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Ki-1 Antigen ,Cell Separation ,Biology ,Article ,Flow cytometry ,Cell Line ,Mice ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,Homeodomain Proteins ,Multidisciplinary ,medicine.diagnostic_test ,Cluster of differentiation ,Teratoma ,Cell Differentiation ,Nanog Homeobox Protein ,Cell sorting ,Cellular Reprogramming ,Flow Cytometry ,Molecular biology ,Cell biology ,Cell culture ,Karyotyping ,Antigens, Surface ,Reprogramming ,Octamer Transcription Factor-3 - Abstract
hiPSC derivation and selection remains inefficient; with selection of high quality clones dependent on extensive characterization which is not amenable to high-throughput (HTP) approaches. We recently described the use of a cocktail of small molecules to enhance hiPSC survival and stability in single cell culture and the use of flow cytometry cell sorting in the HTP-derivation of hiPSCs. Here we report an enhanced protocol for the isolation of bona fide hiPSCs in FACS-based selection using an optimized combination of cell surface markers including CD30. Depletion of CD30(+) cells from reprogramming cultures almost completely abolished the NANOG and OCT4 positive sub-population, suggesting it is a pivotal marker of pluripotent cells. Combining CD30 to SSEA4 and TRA-1-81 in FACS greatly enhanced specificity and efficiency of hiPSC selection and derivation. The current method allows for the efficient and automated, prospective isolation of high-quality hiPSC from the reprogramming cell milieu.
- Published
- 2013
- Full Text
- View/download PDF
42. A novel platform to enable the high-throughput derivation and characterization of feeder-free human iPSCs
- Author
-
Bahram Valamehr, Ramzey Abujarour, Megan Robinson, Daniel Shoemaker, Thuy Ai Huyen Le, David J. Robbins, and Peter Flynn
- Subjects
Genetics ,Multidisciplinary ,Multiplex ,Feeder free ,Computational biology ,Copy-number variation ,Human Induced Pluripotent Stem Cells ,Biology ,Induced pluripotent stem cell ,Throughput (business) ,Article ,Genomic Stability - Abstract
Human induced pluripotent stem cells (hiPSCs) hold enormous potential, however several obstacles impede their translation to industrial and clinical applications. Here we describe a platform to efficiently generate, characterize and maintain single cell and feeder-free (FF) cultured hiPSCs by means of a small molecule cocktail media additive. Using this strategy we have developed an effective multiplex sorting and high-throughput selection platform where individual clonal hiPSC lines are readily obtained from a pool of candidate clones, expanded and thoroughly characterized. By promoting survival and self-renewal, the selected hiPSC clones can be rapidly expanded over multiple FF, single-cell passages while maintaining their pluripotency and genomic stability as demonstrated by trilineage differentiation, karyotype and copy number variation analysis. This study provides a robust platform that increases efficiency, throughput, scale and quality of hiPSC generation and facilitates the industrial and clinical use of iPSC technology.
- Published
- 2012
43. Human Induced Pluripotent Stem Cells Incorporating Safe Harbor Loci Integrated Inducible Suicide Systems for Use in the Application of Cellular Therapeutics
- Author
-
Betsy Rezner, Miguel Meza, David J. Robbins, Raedun Clarke, Xiaosong Huang, Tom Tong Lee, Megan Robinson, Daniel Shoemaker, Bahram Valamehr, Sarah Raynel, Weijie Lan, Parone Philippe Alessandro, and Ramzey Abujarour
- Subjects
Genetics ,Somatic cell ,Cellular differentiation ,Immunology ,Cell Biology ,Hematology ,Suicide gene ,Biology ,Biochemistry ,Viral vector ,Cell biology ,Cell therapy ,Epigenetics ,Expression cassette ,Induced pluripotent stem cell - Abstract
As the field of human induced pluripotent stem cell (hiPSC) research continues to advance, and as the clinical investigation of genetically-engineered hiPSC-derived cellular therapeutics begins to emerge, safety concerns relating to the administration of genetically-altered cells must be addressed and mitigated. A number of strategies including recombinant peptides, monoclonal antibodies, small molecule-modulated enzyme activity and gene-specific modifications have been explored to facilitate the selective elimination of aberrant cells. Of these, the application of genetically-encoded inducible "suicide" systems that can be rapidly activated by a specific non-toxic chemical inducer represents a very attractive, targeted approach for eliminating administered cells without damaging surrounding cells and tissues. Most previous studies have employed viral vectors and short promoters to stably introduce suicide-genes, including herpes complex virus thymidine kinase or inducible caspase-9 (iCasp9), into human cells. The use of viral vectors can lead to random integration events which can disrupt or activate disease-related genes, potentially causing deleterious effects. In addition, many artificial promoters and genome regions are prone to epigenetic silencing in both pluripotent and differentiated states, resulting in cells becoming unresponsive to suicide gene induction. Thus, it is of great importance to identify optimal integration sites and promoters in order to maintain functional suicide gene responses and facilitate the development of genetically-engineered cellular therapeutics. To effectively select and test suicide systems under the control of various promoters in combination with different safe harbor loci integration strategies, we took advantage of our proprietary hiPSC platform, which enables single cell passaging and high-throughput, 96-well plate-based flow cytometry sorting. In a single hiPSC per well manner, we utilized both nuclease-independent and nuclease-dependent strategies to efficiently and precisely integrate various suicide gene expression cassettes in AAVS1 or ROSA26 safe harbor loci. Several integration vectors, each containing a suicide gene expression cassette downstream of various exogenous and endogenous promoters, including endogenous AAVS1 or ROSA26, cytomegalovirus, elongation factor 1α, phosphoglycerate kinase, hybrid CMV enhancer/chicken β-actin and ubiquitin C promoters, were tested to systematically analyze and compare the activity of different suicide systems in both hiPSCs and hiPSC-derived differentiated cells. To conduct high-throughput analyses of these integration and expression strategies, we selected an iCasp9 suicide gene platform where rapid caspase-9 mediated cell death can be induced by small molecule chemical inducers of dimerization such as AP1903. Several endogenous promoters were found to drive persistent expression of iCasp9 during clonal expansion of hiPSCs, but the expression level was determined to be too low to effectively respond to AP1903 treatment. Expression of iCasp9 under the control of various exogenous promoters was lost during prolonged clonal expansion of hiPSCs, and failed to drive AP1903-induced cell death. However, one promoter maintained high levels of iCasp9 expression during the long-term clonal expansion of hiPSCs. Furthermore, these iCasp9-integrated clonal lines underwent rapid cell death in the presence of AP1903, and no residual cell survival was observed when cultures were allowed to recover in the absence of the dimerizing molecule. To test whether epigenetic landscape alterations would abrogate suicide gene-mediated response, hiPSC clones were differentiated into three somatic lineages in vitro and were found to be completely subject to AP1903-induced cell death. Clones were also specifically differentiated towards hematopoietic cells to demonstrate complete induction of cell death by AP1903 treatment. When injected into NSG mice to form teratomas, similar cell death-mediated response was observed in vivo. Notably, one hiPSC clone contained certain rare cells and did escape induced cell death, and this clone and these rare cells were characterized to assess the molecular mechanisms of escape. Our study describes novel findings toward designing optimal safety systems for integration into hiPSC-derived cellular therapies. Disclosures Huang: Fate Therapeutics Inc: Employment. Lan:Fate Therapeutics Inc: Employment. Parone:Fate Therapeutics Inc: Employment. Clarke:Fate Therapeutics Inc: Employment. Abujarour:Fate Therapeutics Inc: Employment. Robinson:Fate Therapeutics Inc: Employment. Meza:Fate Therapeutics Inc: Employment. Lee:Fate Therapeutics Inc: Employment. Shoemaker:Fate Therapeutics Inc: Employment. Valamehr:Fate Therapeutics Inc: Employment.
- Published
- 2015
44. A Platform for the Scalable Derivation of Genetically-Enhanced T and NK Lymphocytes from Naive Human Induced Pluripotent Stem Cells for Cancer Immunotherapy
- Author
-
Megan Robinson, Daniel Shoemaker, Sarah Raynel, William Kim, Bahram Valamehr, Newsha Sahaf, Xiaosong Huang, David J. Robbins, Brian Groff, Raedun Clarke, Lisa Guerrettaz, Betsy Rezner, and Ramzey Abujarour
- Subjects
education.field_of_study ,medicine.medical_treatment ,Immunology ,Population ,Cell Biology ,Hematology ,Embryoid body ,Biology ,Biochemistry ,Embryonic stem cell ,Cell biology ,Haematopoiesis ,Directed differentiation ,Cancer immunotherapy ,medicine ,Stem cell ,Induced pluripotent stem cell ,education - Abstract
Human induced pluripotent stem cell (hiPSC) technology enables the generation of a potentially unlimited source of therapeutically viable hematopoietic cells for the treatment of numerous hematological and non-hematological malignancies, and represents a highly promising approach for overcoming many of the challenges and limitations of patient-derived cancer immunotherapies. To advance the promise of hiPSC technology as an "off-the-shelf" source of hematopoietic cellular therapeutics, it is essential to be able to efficiently and reproducibly generate not only hematopoietic stem cells (HSCs) but also immune effector populations, including the diverse subsets of T and NK lymphoid cells, through a robust and scalable process. The in vitro derivation of HSCs and lymphocytes is complicated by the existence of at least two temporally and spatially distinct waves of blood cell formation during embryonic development: primitive and definitive. Primitive hematopoiesis initiates in the extraembryonic yolk sac and generates a transient and restricted hematopoietic repertoire consisting mainly of primitive erythroid and myeloid cells. Nascent HSCs only emerge later during the definitive wave from a specialized endothelial progenitor within the arterial vasculature termed hemogenic endothelium (HE). HE undergoes an endothelial-to-hematopoietic transition to give rise to HSCs, which then ultimately migrate to the bone marrow where they sustain multi-lineage hematopoiesis, including T and NK lymphoid cells, throughout adult life. Therefore the generation of HSCs and the formation of lymphoid effector cells from hiPSCs is dependent upon our ability to accurately recapitulate the intricate stages of early embryonic hematopoietic development towards the definitive program. While a limited number of studies have described the directed differentiation of hiPSCs to definitive HE in vitro, a major hurdle in utilizing hiPSCs for therapeutic purposes has been the requirement to initially co-culture such cells with murine-derived stromal cells in the presence of ill-defined serum-containing media in order to maintain pluripotency and induce differentiation. In addition, these protocols have employed an intermediate strategy consisting of embryoid body (EB) formation, which is difficult to scale and hindered by lack of reproducibility. We have previously demonstrated that our proprietary platform for robust and rapid derivation of clonal hiPSC lines, which utilizes small molecule reprograming and single cell selection strategies, generates cells with properties indicative of the naïve, or ground state of pluripotency. In addition to maintaining a homogeneous population of hiPSCs, our platform enables the genetic engineering of such pluripotent cells, at a single cell level, in both nuclease-dependent and -independent strategies. Here we describe a novel method for the generation of definitive HE from naïve hiPSCs in a scalable manner, void of an EB intermediate, under serum/feeder-free conditions. This platform represents a well-defined, small molecule-driven, staged protocol that can readily be translated to meet current good manufacturing practice (cGMP) requirements for the development of "off-the-shelf" hematopoietic cell-based immunotherapies. The derived HE population is definitive in nature as determine by Notch dependency and exhibits multi-lineage potential, as demonstrated through the formation of both T and NK lymphoid cells. HE generated by this protocol can be successfully cryopreserved and banked, serving as a highly-stable feedstock for subsequent derivation of various cell types for therapeutic use, including for T and NK cell-based immunotherapies. We have demonstrated that our proprietary, clinically-adaptable method for the large-scale production of definitive HE can efficiently give rise to a variety of lymphoid cell subsets. These derived lymphocytes, including NK cells, have been extensively characterized in vitro and in vivo, and we have demonstrated functionality through cytokine release and cellular cytotoxicity. Furthermore, through genetic modifications at the single cell hiPSC stage, tumor antigen-targeting and inducible caspase-mediated safety systems have been introduced into safe harbor loci to improve the specificity and safety profiles of hiPSC-derived T and NK cells for cancer immunotherapy applications. Disclosures Clarke: Fate Therapeutics Inc: Employment. Abujarour:Fate Therapeutics Inc: Employment. Robinson:Fate Therapeutics Inc: Employment. Huang:Fate Therapeutics Inc: Employment. Shoemaker:Fate Therapeutics Inc: Employment. Valamehr:Fate Therapeutics Inc: Employment.
- Published
- 2015
45. The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation
- Author
-
Arul M. Chinnaiyan, Ramzey Abujarour, Jindan Yu, Bin Zhao, Ian Lian, Sheng Ding, Kun-Liang Guan, Joungmok Kim, Mason A. Israel, Jiagang Zhao, Hideki Okazawa, and Lawrence S.B. Goldstein
- Subjects
Pluripotent Stem Cells ,Cellular differentiation ,Cell Cycle Proteins ,Biology ,Cell Line ,Mice ,Genetics ,Animals ,Humans ,Induced pluripotent stem cell ,Promoter Regions, Genetic ,Transcription factor ,Embryonic Stem Cells ,Adaptor Proteins, Signal Transducing ,Cell Differentiation ,YAP-Signaling Proteins ,Cellular Reprogramming ,Phosphoproteins ,Stem Cell Self-Renewal ,Embryonic stem cell ,Cell biology ,Transcription Coactivator ,Gene Knockdown Techniques ,Stem cell ,Reprogramming ,Developmental Biology ,Research Paper ,Protein Binding ,Transcription Factors - Abstract
Yes-associated protein (YAP) is a potent transcription coactivator acting via binding to the TEAD transcription factor, and plays a critical role in organ size regulation. YAP is phosphorylated and inhibited by the Lats kinase, a key component of the Hippo tumor suppressor pathway. Elevated YAP protein levels and gene amplification have been implicated in human cancer. In this study, we report that YAP is inactivated during embryonic stem (ES) cell differentiation, as indicated by decreased protein levels and increased phosphorylation. Consistently, YAP is elevated during induced pluripotent stem (iPS) cell reprogramming. YAP knockdown leads to a loss of ES cell pluripotency, while ectopic expression of YAP prevents ES cell differentiation in vitro and maintains stem cell phenotypes even under differentiation conditions. Moreover, YAP binds directly to promoters of a large number of genes known to be important for stem cells and stimulates their expression. Our observations establish a critical role of YAP in maintaining stem cell pluripotency.
- Published
- 2010
46. Gel mobilities of linking-number topoisomers and their dependence on DNA helical repeat and elasticity
- Author
-
Alexandre A. Vetcher, Ramzey Abujarour, Stephen D. Levene, Abbye E. McEwen, and Andreas Hanke
- Subjects
Electrophoresis, Agar Gel ,Topoisomer ,Gel electrophoresis of nucleic acids ,Rotation ,Chemistry ,DNA, Superhelical ,Organic Chemistry ,Biophysics ,Linking number ,Stereoisomerism ,Biochemistry ,Elasticity ,Article ,symbols.namesake ,Crystallography ,chemistry.chemical_compound ,Electrophoresis ,Motion ,Ionic strength ,symbols ,DNA supercoil ,DNA ,Writhe - Abstract
Agarose-gel electrophoresis has been used for more than thirty years to characterize the linking-number (Lk) distribution of closed-circular DNA molecules. Although the physical basis of this technique remains poorly understood, the gel-electrophoretic behavior of covalently closed DNAs has been used to determine the local unwinding of DNA by proteins and small-molecule ligands, characterize supercoiling-dependent conformational transitions in duplex DNA, and to measure helical-repeat changes due to shifts in temperature and ionic strength. Those results have been analyzed by assuming that the absolute mobility of a particular topoisomer is mainly a function of the integral number of superhelical turns, and thus a slowly varying function of plasmid molecular weight. In examining the mobilities of Lk topoisomers for a series of plasmids that differ incrementally in size over more than one helical turn, we found that the size-dependent agarose-gel mobility of individual topoisomers with identical values of Lk (but different values of the excess linking number, DeltaLk) vary dramatically over a duplex turn. Our results suggest that a simple semi-empirical relationship holds between the electrophoretic mobility of linking-number topoisomers and their average writhe in solution.
- Published
- 2010
47. A Chemical Platform for Improved Induction of Human iPS Cells
- Author
-
Tongxiang Lin, Ramzey Abujarour, Rajesh Ambasudhan, Xiangyi Lin, Wenlin Li, Ergeng Hao, Alberto Hayek, Simon Hilcove, Heung Sik Hahm, Sheng Ding, and Xu Yuan
- Subjects
Somatic cell ,Induced Pluripotent Stem Cells ,Diphenylamine ,MAP Kinase Kinase 1 ,Cell Differentiation ,Cell Biology ,Dioxoles ,Biology ,Fibroblasts ,Bioinformatics ,Biochemistry ,Article ,Trypsinization ,Cell biology ,Thiazoles ,Pyrimidines ,Transduction, Genetic ,Benzamides ,Humans ,Human Induced Pluripotent Stem Cells ,Induced pluripotent stem cell ,Molecular Biology ,Reprogramming ,Receptors, Transforming Growth Factor beta ,Biotechnology - Abstract
A cocktail of three small molecules improves the efficiency of reprogramming human fibroblasts to induced pluripotent stem cells and allows survival of the cells after trypsinization. The slow kinetics and low efficiency of reprogramming methods to generate human induced pluripotent stem cells (iPSCs) impose major limitations on their utility in biomedical applications. Here we describe a chemical approach that dramatically improves (200-fold) the efficiency of iPSC generation from human fibroblasts, within seven days of treatment. This will provide a basis for developing safer, more efficient, nonviral methods for reprogramming human somatic cells.
- Published
- 2009
48. p97 Is in a complex with cholera toxin and influences the transport of cholera toxin and related toxins to the cytoplasm
- Author
-
Ramzey Abujarour, Seema Dalal, Rockford K. Draper, and Phyllis I. Hanson
- Subjects
Cholera Toxin ,Cytoplasm ,Ricin ,Biology ,medicine.disease_cause ,Biochemistry ,Microbiology ,chemistry.chemical_compound ,Peptide Initiation Factors ,Chlorocebus aethiops ,medicine ,Animals ,Chemical Warfare Agents ,Molecular Biology ,Vero Cells ,Toxin ,Endoplasmic reticulum ,Cholera toxin ,Cell Biology ,medicine.disease ,Cholera ,Transport protein ,Protein Transport ,chemistry ,ADP-ribosylation ,Mutation - Abstract
Certain protein toxins, including cholera toxin, ricin, and Pseudomonas aeruginosa exotoxin A, are transported to the lumen of the endoplasmic reticulum where they retro-translocate across the endoplasmic reticulum membrane to enter the cytoplasm. The mechanism of retrotranslocation is poorly understood but may involve the endoplasmic reticulum-associated degradation pathway. The AAA ATPase p97 (also called valosin-containing protein) participates in the retro-translocation of cellular endoplasmic reticulum-associated degradation substrates and is therefore a candidate to participate in the retrotranslocation of protein toxins. To investigate whether p97 functions in toxin delivery to the cytoplasm, we measured the sensitivity to toxins of cells expressing either wild-type p97 or a dominant ATPase-defective p97 mutant under control of a tetracycline-inducible promoter. The rate at which cholera toxin and related toxins entered the cytoplasm was reduced in cells expressing the ATPase-defective p97, suggesting that the toxins might interact with p97. To detect interaction, the cholera toxin A chain was immunoprecipitated from cholera toxin-treated Vero cells, and co-immunoprecipitation of p97 was assessed by immunoblotting. The immunoprecipitates contained both cholera toxin A chain and p97, evidence that the two proteins are in a complex. Altogether, these results provide functional and structural evidence that p97 participates in the transport of cholera toxin to the cytoplasm.
- Published
- 2005
49. Evidence that the transport of ricin to the cytoplasm is independent of both Rab6A and COPI
- Author
-
Ramzey Abujarour, Rockford K. Draper, and Alice Chen
- Subjects
Models, Molecular ,endocrine system ,Cholera Toxin ,KDEL ,Golgi Apparatus ,CHO Cells ,Ricin ,Protein Sorting Signals ,medicine.disease_cause ,Endoplasmic Reticulum ,Coat Protein Complex I ,Shiga Toxin ,chemistry.chemical_compound ,symbols.namesake ,RAB6A ,Cricetulus ,Cricetinae ,Chlorocebus aethiops ,medicine ,Animals ,Vero Cells ,Cells, Cultured ,biology ,Endoplasmic reticulum ,Cholera toxin ,Shiga toxin ,Biological Transport ,Cell Biology ,COPI ,Golgi apparatus ,Biochemistry ,chemistry ,Microscopy, Fluorescence ,rab GTP-Binding Proteins ,biology.protein ,symbols - Abstract
Cholera toxin, Shiga toxin and ricin are examples of protein toxins that require retrograde transport from the Golgi complex into the endoplasmic reticulum (ER) to express their cytotoxic activities and different toxins appear to use different pathways of retrograde transport. Cholera toxin contains the mammalian retrograde targeting signal KDEL and is believed to exploit the coat protein I (COPI) and KDEL receptor-dependent pathway to go from the Golgi complex to the ER. Shiga toxin, however, has no KDEL sequence to specify its inclusion in COPI-coated retrograde vesicles and is believed to use a recently discovered COPI-independent and Rab6A-dependent retrograde pathway to enter the ER. Ricin, like Shiga toxin, does not contain a KDEL sequence and is therefore a candidate to use the COPI-independent and Rab6A-dependent pathway of retrograde transport to access the ER. We measured the effect of the GDP-restricted mutant of Rab6A (Rab6A-T27N) on the cytotoxic activity of ricin and found that expressing Rab6A-T27N in cells did not inhibit the cytotoxicity of ricin, suggesting that ricin enters the cytoplasm by a retrograde pathway that does not involve Rab6A. Moreover, ricin still intoxicated cells when Rab6A and COPI were simultaneously inhibited, implying that ricin requires neither Rab6A nor COPI to intoxicate cells.
- Published
- 2003
50. Induced pluripotent stem cells free of exogenous reprogramming factors
- Author
-
Sheng Ding and Ramzey Abujarour
- Subjects
Pluripotent Stem Cells ,Somatic cell ,Biology ,Cellular Reprogramming ,Molecular biology ,Cell biology ,Adult Stem Cells ,Mice ,Animals ,Humans ,Minireview ,Induced pluripotent stem cell ,Reprogramming ,Gene ,Adult stem cell - Abstract
The search continues to find ways of efficiently generating induced pluripotent stem cells free of vectors and transgenes., The development of novel approaches for reprogramming mouse and human somatic cells has enabled the generation of induced pluripotent stem cells that are free of exogenous genes.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.