Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Aaron R. Cooper is active.

Publication


Featured researches published by Aaron R. Cooper.


Blood | 2015

Correction of the sickle-cell disease mutation in human hematopoietic stem/progenitor cells

Megan D. Hoban; Gregory J. Cost; Matthew C. Mendel; Zulema Romero; Michael L. Kaufman; Alok V. Joglekar; Michelle Ho; Dianne Lumaquin; David Gray; Georgia R. Lill; Aaron R. Cooper; Fabrizia Urbinati; Shantha Senadheera; Allen Zhu; Pei-Qi Liu; David Paschon; Lei Zhang; Edward J. Rebar; Andrew Wilber; Xiaoyan Wang; Philip D. Gregory; Michael C. Holmes; Andreas Reik; Roger P. Hollis; Donald B. Kohn

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the β-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the β-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Prostate cancer originating in basal cells progresses to adenocarcinoma propagated by luminal-like cells

Tanya Stoyanova; Aaron R. Cooper; Justin M. Drake; Xian Liu; Andrew J. Armstrong; Kenneth J. Pienta; Hong Zhang; Donald B. Kohn; Jiaoti Huang; Owen N. Witte; Andrew S. Goldstein

Significance This study determined that two histological phenotypes of cancer can arise from a common target cell. Whereas luminal cells are not efficient cells of origin, luminal-like tumor cells isolated from human prostate adenocarcinoma can serially propagate advanced disease. The relationship between the cells that initiate cancer and the cancer stem-like cells that propagate tumors has been poorly defined. In a human prostate tissue transformation model, basal cells expressing the oncogenes Myc and myristoylated AKT can initiate heterogeneous tumors. Tumors contain features of acinar-type adenocarcinoma with elevated eIF4E-driven protein translation and squamous cell carcinoma marked by activated beta-catenin. Lentiviral integration site analysis revealed that alternative histological phenotypes can be clonally derived from a common cell of origin. In advanced disease, adenocarcinoma can be propagated by self-renewing tumor cells with an androgen receptor-low immature luminal phenotype in the absence of basal-like cells. These data indicate that advanced prostate adenocarcinoma initiated in basal cells can be maintained by luminal-like tumor-propagating cells. Determining the cells that maintain human prostate adenocarcinoma and the signaling pathways characterizing these tumor-propagating cells is critical for developing effective therapeutic strategies against this population.


Journal of Clinical Investigation | 2013

β-globin gene transfer to human bone marrow for sickle cell disease

Zulema Romero; Fabrizia Urbinati; Sabine Geiger; Aaron R. Cooper; Jennifer Wherley; Michael L. Kaufman; Roger P. Hollis; Rafael Ruiz de Assin; Shantha Senadheera; Arineh Sahagian; Xiangyang Jin; Alyse Gellis; Xiaoyan Wang; David W. Gjertson; Satiro deOliveira; Pamela Kempert; Sally Shupien; Hisham Abdel-Azim; Mark C. Walters; Herbert J. Meiselman; Rosalinda B. Wenby; Theresa Gruber; Victor J. Marder; Thomas D. Coates; Donald B. Kohn

Autologous hematopoietic stem cell gene therapy is an approach to treating sickle cell disease (SCD) patients that may result in lower morbidity than allogeneic transplantation. We examined the potential of a lentiviral vector (LV) (CCL-βAS3-FB) encoding a human hemoglobin (HBB) gene engineered to impede sickle hemoglobin polymerization (HBBAS3) to transduce human BM CD34+ cells from SCD donors and prevent sickling of red blood cells produced by in vitro differentiation. The CCL-βAS3-FB LV transduced BM CD34+ cells from either healthy or SCD donors at similar levels, based on quantitative PCR and colony-forming unit progenitor analysis. Consistent expression of HBBAS3 mRNA and HbAS3 protein compromised a fourth of the total β-globin-like transcripts and hemoglobin (Hb) tetramers. Upon deoxygenation, a lower percentage of HBBAS3-transduced red blood cells exhibited sickling compared with mock-transduced cells from sickle donors. Transduced BM CD34+ cells were transplanted into immunodeficient mice, and the human cells recovered after 2-3 months were cultured for erythroid differentiation, which showed levels of HBBAS3 mRNA similar to those seen in the CD34+ cells that were directly differentiated in vitro. These results demonstrate that the CCL-βAS3-FB LV is capable of efficient transfer and consistent expression of an effective anti-sickling β-globin gene in human SCD BM CD34+ progenitor cells, improving physiologic parameters of the resulting red blood cells.


Molecular Therapy | 2016

CRISPR/Cas9-Mediated Correction of the Sickle Mutation in Human CD34+ cells

Megan D. Hoban; Dianne Lumaquin; Caroline Y. Kuo; Zulema Romero; Joseph Long; Michelle Ho; Courtney S. Young; Michelle Mojadidi; Sorel Fitz-Gibbon; Aaron R. Cooper; Georgia R. Lill; Fabrizia Urbinati; Beatriz Campo-Fernandez; Carmen Flores Bjurström; Matteo Pellegrini; Roger P. Hollis; Donald B. Kohn

Targeted genome editing technology can correct the sickle cell disease mutation of the β-globin gene in hematopoietic stem cells. This correction supports production of red blood cells that synthesize normal hemoglobin proteins. Here, we demonstrate that Transcription Activator-Like Effector Nucleases (TALENs) and the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system can target DNA sequences around the sickle-cell mutation in the β-globin gene for site-specific cleavage and facilitate precise correction when a homologous donor template is codelivered. Several pairs of TALENs and multiple CRISPR guide RNAs were evaluated for both on-target and off-target cleavage rates. Delivery of the CRISPR/Cas9 components to CD34+ cells led to over 18% gene modification in vitro. Additionally, we demonstrate the correction of the sickle cell disease mutation in bone marrow derived CD34+ hematopoietic stem and progenitor cells from sickle cell disease patients, leading to the production of wild-type hemoglobin. These results demonstrate correction of the sickle mutation in patient-derived CD34+ cells using CRISPR/Cas9 technology.


Molecular Therapy | 2014

Preclinical Demonstration of Lentiviral Vector-mediated Correction of Immunological and Metabolic Abnormalities in Models of Adenosine Deaminase Deficiency

Denise A. Carbonaro; Lin Zhang; Xiangyang Jin; Claudia Montiel-Equihua; Sabine Geiger; Marlene Carmo; Aaron R. Cooper; Lynette Fairbanks; Michael L. Kaufman; Nj Sebire; Roger P. Hollis; Michael P. Blundell; Shantha Senadheera; Pei Yu Fu; Arineh Sahaghian; Rebecca Chan; Xiaoyan Wang; Kenneth Cornetta; Adrian J. Thrasher; Donald B. Kohn; H. Bobby Gaspar

Gene transfer into autologous hematopoietic stem cells by γ-retroviral vectors (gRV) is an effective treatment for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). However, current gRV have significant potential for insertional mutagenesis as reported in clinical trials for other primary immunodeficiencies. To improve the efficacy and safety of ADA-SCID gene therapy (GT), we generated a self-inactivating lentiviral vector (LV) with a codon-optimized human cADA gene under the control of the short form elongation factor-1α promoter (LV EFS ADA). In ADA(-/-) mice, LV EFS ADA displayed high-efficiency gene transfer and sufficient ADA expression to rescue ADA(-/-) mice from their lethal phenotype with good thymic and peripheral T- and B-cell reconstitution. Human ADA-deficient CD34(+) cells transduced with 1-5 × 10(7) TU/ml had 1-3 vector copies/cell and expressed 1-2x of normal endogenous levels of ADA, as assayed in vitro and by transplantation into immune-deficient mice. Importantly, in vitro immortalization assays demonstrated that LV EFS ADA had significantly less transformation potential compared to gRV vectors, and vector integration-site analysis by nrLAM-PCR of transduced human cells grown in immune-deficient mice showed no evidence of clonal skewing. These data demonstrated that the LV EFS ADA vector can effectively transfer the human ADA cDNA and promote immune and metabolic recovery, while reducing the potential for vector-mediated insertional mutagenesis.


Stem Cells Translational Medicine | 2012

From Skin Biopsy to Neurons through a Pluripotent Intermediate Under Good Manufacturing Practice Protocols

Saravanan Karumbayaram; Peiyee Lee; Soheila F. Azghadi; Aaron R. Cooper; Michaela Patterson; Donald B. Kohn; April D. Pyle; Amander T. Clark; James A. Byrne; Jerome A. Zack; Kathrin Plath; William E. Lowry

The clinical application of human‐induced pluripotent stem cells (hiPSCs) requires not only the production of Good Manufacturing Practice‐grade (GMP‐grade) hiPSCs but also the derivation of specified cell types for transplantation under GMP conditions. Previous reports have suggested that hiPSCs can be produced in the absence of animal‐derived reagents (xenobiotics) to ease the transition to production under GMP standards. However, to facilitate the use of hiPSCs in cell‐based therapeutics, their progeny should be produced not only in the absence of xenobiotics but also under GMP conditions requiring extensive standardization of protocols, documentation, and reproducibility of methods and product. Here, we present a successful framework to produce GMP‐grade derivatives of hiPSCs that are free of xenobiotic exposure from the collection of patient fibroblasts, through reprogramming, maintenance of hiPSCs, identification of reprogramming vector integration sites (nrLAM‐PCR), and finally specification and terminal differentiation of clinically relevant cells. Furthermore, we developed a primary set of Standard Operating Procedures for the GMP‐grade derivation and differentiation of these cells as a resource to facilitate widespread adoption of these practices.


Stem Cell Research & Therapy | 2013

Generation and characterization of transgene-free human induced pluripotent stem cells and conversion to putative clinical-grade status

Jason P. Awe; Patrick Lee; Cyril Ramathal; Agustin Vega-Crespo; Jens Durruthy-Durruthy; Aaron R. Cooper; Saravanan Karumbayaram; William E. Lowry; Amander T. Clark; Jerome A. Zack; Vittorio Sebastiano; Donald B. Kohn; April D. Pyle; Martin G. Martin; Gerald S. Lipshutz; Patricia E. Phelps; Renee A. Reijo Pera; James A. Byrne

IntroductionThe reprogramming of a patient’s somatic cells back into induced pluripotent stem cells (iPSCs) holds significant promise for future autologous cellular therapeutics. The continued presence of potentially oncogenic transgenic elements following reprogramming, however, represents a safety concern that should be addressed prior to clinical applications. The polycistronic stem cell cassette (STEMCCA), an excisable lentiviral reprogramming vector, provides, in our hands, the most consistent reprogramming approach that addresses this safety concern. Nevertheless, most viral integrations occur in genes, and exactly how the integration, epigenetic reprogramming, and excision of the STEMCCA reprogramming vector influences those genes and whether these cells still have clinical potential are not yet known.MethodsIn this study, we used both microarray and sensitive real-time PCR to investigate gene expression changes following both intron-based reprogramming and excision of the STEMCCA cassette during the generation of human iPSCs from adult human dermal fibroblasts. Integration site analysis was conducted using nonrestrictive linear amplification PCR. Transgene-free iPSCs were fully characterized via immunocytochemistry, karyotyping and teratoma formation, and current protocols were implemented for guided differentiation. We also utilized current good manufacturing practice guidelines and manufacturing facilities for conversion of our iPSCs into putative clinical grade conditions.ResultsWe found that a STEMCCA-derived iPSC line that contains a single integration, found to be located in an intronic location in an actively transcribed gene, PRPF39, displays significantly increased expression when compared with post-excised stem cells. STEMCCA excision via Cre recombinase returned basal expression levels of PRPF39. These cells were also shown to have proper splicing patterns and PRPF39 gene sequences. We also fully characterized the post-excision iPSCs, differentiated them into multiple clinically relevant cell types (including oligodendrocytes, hepatocytes, and cardiomyocytes), and converted them to putative clinical-grade conditions using the same approach previously approved by the US Food and Drug Administration for the conversion of human embryonic stem cells from research-grade to clinical-grade status.ConclusionFor the first time, these studies provide a proof-of-principle for the generation of fully characterized transgene-free human iPSCs and, in light of the limited availability of current good manufacturing practice cellular manufacturing facilities, highlight an attractive potential mechanism for converting research-grade cell lines into putatively clinical-grade biologics for personalized cellular therapeutics.


Journal of Virological Methods | 2011

Highly efficient large-scale lentiviral vector concentration by tandem tangential flow filtration.

Aaron R. Cooper; Sanjeet Patel; Shantha Senadheera; Kathrin Plath; Donald B. Kohn; Roger P. Hollis

Large-scale lentiviral vector (LV) concentration can be inefficient and time consuming, often involving multiple rounds of filtration and centrifugation. This report describes a simpler method using two tangential flow filtration (TFF) steps to concentrate liter-scale volumes of LV supernatant, achieving in excess of 2000-fold concentration in less than 3h with very high recovery (>97%). Large volumes of LV supernatant can be produced easily through the use of multi-layer flasks, each having 1720 cm(2) surface area and producing ∼560 mL of supernatant per flask. Combining the use of such flasks and TFF greatly simplifies large-scale production of LV. As a demonstration, the method is used to produce a very high titer LV (>10(10)TU/mL) and transduce primary human CD34+ hematopoietic stem/progenitor cells at high final vector concentrations with no overt toxicity. A complex LV (STEMCCA) for induced pluripotent stem cell (iPSC) generation is also concentrated from low initial titer and used to transduce and reprogram primary human fibroblasts with no overt toxicity. Additionally, a generalized and simple multiplexed real-time PCR assay is described for lentiviral vector titer and copy number determination.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Long-term in vivo monitoring of mouse and human hematopoietic stem cell engraftment with a human positron emission tomography reporter gene

Melissa N. McCracken; Eric Gschweng; Evan Nair-Gill; Jami McLaughlin; Aaron R. Cooper; Mireille Riedinger; Donghui Cheng; Christopher Nosala; Donald B. Kohn; Owen N. Witte

Positron emission tomography (PET) reporter genes allow noninvasive whole-body imaging of transplanted cells by detection with radiolabeled probes. We used a human deoxycytidine kinase containing three amino acid substitutions within the active site (hdCK3mut) as a reporter gene in combination with the PET probe [18F]-L-FMAU (1-(2-deoxy-2-18fluoro-β-L-arabinofuranosyl)-5-methyluracil) to monitor models of mouse and human hematopoietic stem cell (HSC) transplantation. These mutations in hdCK3mut expanded the substrate capacity allowing for reporter-specific detection with a thymidine analog probe. Measurements of long-term engrafted cells (up to 32 wk) demonstrated that hdCK3mut expression is maintained in vivo with no counter selection against reporter-labeled cells. Reporter cells retained equivalent engraftment and differentiation capacity being detected in all major hematopoietic lineages and tissues. This reporter gene and probe should be applicable to noninvasively monitor therapeutic cell transplants in multiple tissues.


Journal of Clinical Investigation | 2017

Clinical efficacy of gene-modified stem cells in adenosine deaminase–deficient immunodeficiency

Kit L. Shaw; Elizabeth Garabedian; Suparna Mishra; Provaboti Barman; Alejandra Davila; Denise A. Carbonaro; Sally Shupien; Christopher Silvin; Sabine Geiger; Barbara Nowicki; E. Monika Smogorzewska; Berkley Brown; Xiaoyan Wang; Satiro N. De Oliveira; Yeong Choi; Alan K. Ikeda; Dayna Terrazas; Pei Yu Fu; Allen Yu; Beatriz Campo Fernandez; Aaron R. Cooper; Barbara C. Engel; Greg M. Podsakoff; Arumugam Balamurugan; Stacie M. Anderson; Linda M. Muul; G. Jayashree Jagadeesh; Neena Kapoor; John Tse; Theodore B. Moore

BACKGROUND. Autologous hematopoietic stem cell transplantation (HSCT) of gene-modified cells is an alternative to enzyme replacement therapy (ERT) and allogeneic HSCT that has shown clinical benefit for adenosine deaminase–deficient (ADA-deficient) SCID when combined with reduced intensity conditioning (RIC) and ERT cessation. Clinical safety and therapeutic efficacy were evaluated in a phase II study. METHODS. Ten subjects with confirmed ADA-deficient SCID and no available matched sibling or family donor were enrolled between 2009 and 2012 and received transplantation with autologous hematopoietic CD34+ cells that were modified with the human ADA cDNA (MND-ADA) &ggr;-retroviral vector after conditioning with busulfan (90 mg/m2) and ERT cessation. Subjects were followed from 33 to 84 months at the time of data analysis. Safety of the procedure was assessed by recording the number of adverse events. Efficacy was assessed by measuring engraftment of gene-modified hematopoietic stem/progenitor cells, ADA gene expression, and immune reconstitution. RESULTS. With the exception of the oldest subject (15 years old at enrollment), all subjects remained off ERT with normalized peripheral blood mononuclear cell (PBMC) ADA activity, improved lymphocyte numbers, and normal proliferative responses to mitogens. Three of nine subjects were able to discontinue intravenous immunoglobulin replacement therapy. The MND-ADA vector was persistently detected in PBMCs (vector copy number [VCN] = 0.1–2.6) and granulocytes (VCN = 0.01–0.3) through the most recent visits at the time of this writing. No patient has developed a leukoproliferative disorder or other vector-related clinical complication since transplant. CONCLUSION. These results demonstrate clinical therapeutic efficacy from gene therapy for ADA-deficient SCID, with an excellent clinical safety profile. TRIAL REGISTRATION. ClinicalTrials.gov NCT00794508. FUNDING. Food and Drug Administration Office of Orphan Product Development award, RO1 FD003005; NHLBI awards, PO1 HL73104 and Z01 HG000122; UCLA Clinical and Translational Science Institute awards, UL1RR033176 and UL1TR000124.

Collaboration


Dive into the Aaron R. Cooper's collaboration.

Top Co-Authors

Avatar

Donald B. Kohn

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaoyan Wang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zulema Romero

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Megan D. Hoban

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge