Network


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

Hotspot


Dive into the research topics where Iram Khan is active.

Publication


Featured researches published by Iram Khan.


Science Translational Medicine | 2015

Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template

Blythe D. Sather; Guillermo S. Romano Ibarra; Karen Sommer; Gabrielle Curinga; Malika Hale; Iram Khan; Swati Singh; Yumei Song; Kamila Gwiazda; Jaya Sahni; Jordan Jarjour; Alexander Astrakhan; Thor A. Wagner; Andrew M. Scharenberg; David J. Rawlings

Therapeutic coding sequences can be targeted to the CCR5 locus of primary human T cells with high efficiency by using megaTAL nuclease and an AAV donor template. Delete and replace Newer gene-editing methods hold promise for correcting human disease but so far have been hampered by low efficiencies when used in primary cells. To address this issue, Sather et al. have devised a more effective way to both disrupt and replace the CCR5 locus in human T cells, a procedure that has already been shown to improve HIV clearance. Serotype 6 of an adeno-associated viral vector worked particularly well for delivery of megaTAL nucleases and homologous donor templates to primary human T cells, achieving efficient gene-editing rates and little toxicity. The megaTALs generate homology-directed repair (rather than previous efforts, which induce nonhomologous end-joining repair) and so was used for both deletion and accurate replacement of the CCR5 locus. The authors demonstrate that chimeric antigen receptors and an HIV fusion inhibitor inserted into the CCR5 locus ameliorate HIV infection in mice and show that their approach also works in CD34+ hematopoietic precursor cells. Genetic mutations or engineered nucleases that disrupt the HIV co-receptor CCR5 block HIV infection of CD4+ T cells. These findings have motivated the engineering of CCR5-specific nucleases for application as HIV therapies. The efficacy of this approach relies on efficient biallelic disruption of CCR5, and the ability to efficiently target sequences that confer HIV resistance to the CCR5 locus has the potential to further improve clinical outcomes. We used RNA-based nuclease expression paired with adeno-associated virus (AAV)–mediated delivery of a CCR5-targeting donor template to achieve highly efficient targeted recombination in primary human T cells. This method consistently achieved 8 to 60% rates of homology-directed recombination into the CCR5 locus in T cells, with over 80% of cells modified with an MND-GFP expression cassette exhibiting biallelic modification. MND-GFP–modified T cells maintained a diverse repertoire and engrafted in immune-deficient mice as efficiently as unmodified cells. Using this method, we integrated sequences coding chimeric antigen receptors (CARs) into the CCR5 locus, and the resulting targeted CAR T cells exhibited antitumor or anti-HIV activity. Alternatively, we introduced the C46 HIV fusion inhibitor, generating T cell populations with high rates of biallelic CCR5 disruption paired with potential protection from HIV with CXCR4 co-receptor tropism. Finally, this protocol was applied to adult human mobilized CD34+ cells, resulting in 15 to 20% homologous gene targeting. Our results demonstrate that high-efficiency targeted integration is feasible in primary human hematopoietic cells and highlight the potential of gene editing to engineer T cell products with myriad functional properties.


Blood | 2014

Rapamycin relieves lentiviral vector transduction resistance in human and mouse hematopoietic stem cells.

Cathy Wang; Blythe D. Sather; Xuefeng Wang; Jennifer E. Adair; Iram Khan; Swati Singh; Shanshan Lang; Amie B. Adams; Gabrielle Curinga; Hans Peter Kiem; Carol H. Miao; David J. Rawlings; Bruce E. Torbett

Transplantation of genetically modified hematopoietic stem cells (HSCs) is a promising therapeutic strategy for genetic diseases, HIV, and cancer. However, a barrier for clinical HSC gene therapy is the limited efficiency of gene delivery via lentiviral vectors (LVs) into HSCs. We show here that rapamycin, an allosteric inhibitor of the mammalian target of rapamycin complexes, facilitates highly efficient lentiviral transduction of mouse and human HSCs and dramatically enhances marking frequency in long-term engrafting cells in mice. Mechanistically, rapamycin enhanced postbinding endocytic events, leading to increased levels of LV cytoplasmic entry, reverse transcription, and genomic integration. Despite increasing LV copy number, rapamycin did not significantly alter LV integration site profile or chromosomal distribution in mouse HSCs. Rapamycin also enhanced in situ transduction of mouse HSCs via direct intraosseous infusion. Collectively, rapamycin strongly augments LV transduction of HSCs in vitro and in vivo and may prove useful for therapeutic gene delivery.


Nucleic Acids Research | 2014

Novel fluorescent genome editing reporters for monitoring DNA repair pathway utilization at endonuclease-induced breaks

Ryan Kuhar; Kamila Gwiazda; Olivier Humbert; Tyler Mandt; Joey Pangallo; Michelle Brault; Iram Khan; Nancy Maizels; David J. Rawlings; Andrew M. Scharenberg; Michael T. Certo

The creation of a DNA break at a specific locus by a designer endonuclease can be harnessed to edit a genome. However, DNA breaks may engage one of several competing repair pathways that lead to distinct types of genomic alterations. Therefore, understanding the contribution of different repair pathways following the introduction of a targeted DNA break is essential to further advance the safety and efficiency of nuclease-induced genome modification. To gain insight into the role of different DNA repair pathways in resolving nuclease-induced DNA breaks into genome editing outcomes, we previously developed a fluorescent-based reporter system, designated the Traffic Light Reporter, which provides a readout of gene targeting and gene disruption downstream of a targeted DNA double-strand break. Here we describe two related but novel reporters that extend this technology: one that allows monitoring of the transcriptional activity at the reporter locus, and thus can be applied to interrogate break resolution at active and repressed loci; and a second that reads out single-strand annealing in addition to gene targeting and gene disruption. Application of these reporters to assess repair pathway usage in several common gene editing contexts confirms the importance that chromatin status and initiation of end resection have on the resolution of nuclease-induced breaks.


Molecular Therapy | 2015

Intraosseous Delivery of Lentiviral Vectors Targeting Factor VIII Expression in Platelets Corrects Murine Hemophilia A

Xuefeng Wang; Simon Shin; Andy F J Chiang; Iram Khan; Dao Pan; David J. Rawlings; Carol H. Miao

Intraosseous (IO) infusion of lentiviral vectors (LVs) for in situ gene transfer into bone marrow may avoid specific challenges posed by ex vivo gene delivery, including, in particular, the requirement of preconditioning. We utilized IO delivery of LVs encoding a GFP or factor VIII (FVIII) transgene directed by ubiquitous promoters (a MND or EF-1α-short element; M-GFP-LV, E-F8-LV) or a platelet-specific, glycoprotein-1bα promoter (G-GFP-LV, G-F8-LV). A single IO infusion of M-GFP-LV or G-GFP-LV achieved long-term and efficient GFP expression in Lineage(-)Sca1(+)c-Kit(+) hematopoietic stem cells and platelets, respectively. While E-F8-LV produced initially high-level FVIII expression, robust anti-FVIII immune responses eliminated functional FVIII in circulation. In contrast, IO delivery of G-F8-LV achieved long-term platelet-specific expression of FVIII, resulting in partial correction of hemophilia A. Furthermore, similar clinical benefit with G-F8-LV was achieved in animals with pre-existing anti-FVIII inhibitors. These findings further support platelets as an ideal FVIII delivery vehicle, as FVIII, stored in α-granules, is protected from neutralizing antibodies and, during bleeding, activated platelets locally excrete FVIII to promote clot formation. Overall, a single IO infusion of G-F8-LV was sufficient to correct hemophilia phenotype for long term, indicating that this approach may provide an effective means to permanently treat FVIII deficiency.


Molecular Therapy | 2016

High Efficiency CRISPR/Cas9-mediated Gene Editing in Primary Human T-cells Using Mutant Adenoviral E4orf6/E1b55k “Helper” Proteins

Kamila Gwiazda; Alexandra Grier; Jaya Sahni; Stephen M Burleigh; Unja Martin; Julia G Yang; Nicholas A Popp; Michelle C Krutein; Iram Khan; Kyle Jacoby; Michael C. Jensen; David J. Rawlings; Andrew M. Scharenberg

Many future therapeutic applications of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 and related RNA-guided nucleases are likely to require their use to promote gene targeting, thus necessitating development of methods that provide for delivery of three components-Cas9, guide RNAs and recombination templates-to primary cells rendered proficient for homology-directed repair. Here, we demonstrate an electroporation/transduction codelivery method that utilizes mRNA to express both Cas9 and mutant adenoviral E4orf6 and E1b55k helper proteins in association with adeno-associated virus (AAV) vectors expressing guide RNAs and recombination templates. By transiently enhancing target cell permissiveness to AAV transduction and gene editing efficiency, this novel approach promotes efficient gene disruption and/or gene targeting at multiple loci in primary human T-cells, illustrating its broad potential for application in translational gene editing.


Nucleic Acids Research | 2014

Progressive engineering of a homing endonuclease genome editing reagent for the murine X-linked immunodeficiency locus

Yupeng Wang; Iram Khan; Sandrine Boissel; Jordan Jarjour; Joseph Pangallo; Summer B. Thyme; David Baker; Andrew M. Scharenberg; David J. Rawlings

LAGLIDADG homing endonucleases (LHEs) are compact endonucleases with 20–22 bp recognition sites, and thus are ideal scaffolds for engineering site-specific DNA cleavage enzymes for genome editing applications. Here, we describe a general approach to LHE engineering that combines rational design with directed evolution, using a yeast surface display high-throughput cleavage selection. This approach was employed to alter the binding and cleavage specificity of the I-Anil LHE to recognize a mutation in the mouse Bruton tyrosine kinase (Btk) gene causative for mouse X-linked immunodeficiency (XID)—a model of human X-linked agammaglobulinemia (XLA). The required re-targeting of I-AniI involved progressive resculpting of the DNA contact interface to accommodate nine base differences from the native cleavage sequence. The enzyme emerging from the progressive engineering process was specific for the XID mutant allele versus the wild-type (WT) allele, and exhibited activity equivalent to WT I-AniI in vitro and in cellulo reporter assays. Fusion of the enzyme to a site-specific DNA binding domain of transcription activator-like effector (TALE) resulted in a further enhancement of gene editing efficiency. These results illustrate the potential of LHE enzymes as specific and efficient tools for therapeutic genome engineering.


Molecular therapy. Methods & clinical development | 2017

Safe and Effective Gene Therapy for Murine Wiskott-Aldrich Syndrome Using an Insulated Lentiviral Vector

Swati Singh; Iram Khan; Socheath Khim; Brenda Seymour; Karen Sommer; Matthew Wielgosz; Zachary K. Norgaard; Hans-Peter Kiem; Jennifer E. Adair; Denny Liggitt; Arthur W. Nienhuis; David J. Rawlings

Wiskott-Aldrich syndrome (WAS) is a life-threatening immunodeficiency caused by mutations within the WAS gene. Viral gene therapy to restore WAS protein (WASp) expression in hematopoietic cells of patients with WAS has the potential to improve outcomes relative to the current standard of care, allogeneic bone marrow transplantation. However, the development of viral vectors that are both safe and effective has been problematic. While use of viral transcriptional promoters may increase the risk of insertional mutagenesis, cellular promoters may not achieve WASp expression levels necessary for optimal therapeutic effect. Here we evaluate a self-inactivating (SIN) lentiviral vector combining a chromatin insulator upstream of a viral MND (MPSV LTR, NCR deleted, dl587 PBS) promoter driving WASp expression. Used as a gene therapeutic in Was−/− mice, this vector resulted in stable WASp+ cells in all hematopoietic lineages and rescue of T and B cell defects with a low number of viral integrations per cell, without evidence of insertional mutagenesis in serial bone marrow transplants. In a gene transfer experiment in non-human primates, the insulated MND promoter (driving GFP expression) demonstrated long-term polyclonal engraftment of GFP+ cells. These observations demonstrate that the insulated MND promoter safely and efficiently reconstitutes clinically effective WASp expression and should be considered for future WAS therapy.


Molecular Therapy | 2017

Engineering protein-secreting plasma cells by homology-directed repair in primary human B cells

King Hung; Iana Meitlis; Malika Hale; Chun-Yu Chen; Swati Singh; Shaun W. Jackson; Carol H. Miao; Iram Khan; David J. Rawlings; Richard G. James

The ability to engineer primary human B cells to differentiate into long-lived plasma cells and secrete a de novo protein may allow the creation of novel plasma cell therapies for protein deficiency diseases and other clinical applications. We initially developed methods for efficient genome editing of primary B cells isolated from peripheral blood. By delivering CRISPR/CRISPR-associated protein 9 (Cas9) ribonucleoprotein (RNP) complexes under conditions of rapid B cell expansion, we achieved site-specific gene disruption at multiple loci in primary human B cells (with editing rates of up to 94%). We used this method to alter ex vivo plasma cell differentiation by disrupting developmental regulatory genes. Next, we co-delivered RNPs with either a single-stranded DNA oligonucleotide or adeno-associated viruses containing homologous repair templates. Using either delivery method, we achieved targeted sequence integration at high efficiency (up to 40%) via homology-directed repair. This method enabled us to engineer plasma cells to secrete factor IX (FIX) or B cell activating factor (BAFF) at high levels. Finally, we show that introduction of BAFF into plasma cells promotes their engraftment into immunodeficient mice. Our results highlight the utility of genome editing in studying human B cell biology and demonstrate a novel strategy for modifying human plasma cells to secrete therapeutic proteins.


Molecular Therapy | 2016

316. Successful Editing of the CD40LG Locus in Human Hematopoietic Stem Cells

Gabrielle Curinga; Sarah Leach; Swati Singh; Nick Hubbard; David Hagin; Karen Sommer; Iram Khan; Yumei Song; Troy R. Torgerson; Andy Scharenberg; David J. Rawlings

X-Linked Hyper-IgM Syndrome (X-HIGM) is a genetic disorder caused by mutations in CD40LG that result in the loss of functional CD40L protein on the T cell surface. CD40L is required for T cells to provide “help” to B cells during an immune challenge; thus X-HIGM patients have impaired immunoglobulin class-switching and somatic hypermutation, and suffer recurrent infections. Murine gene transfer and X-HIGM patient studies suggest that safe and effective gene therapies for this disorder need to replicate cell surface expression patterns of WT endogenous CD40L, as well as knock-out expression of the mutant CD40L; requirements that are unlikely to be achievable using viral gene replacement. We recently reported a gene editing approach combining mRNA-delivered TALEN (targeting just upstream of the coding sequence of CD40LG) with an rAAV donor template for homology directed repair, targeting a promoter-less CD40L cDNA to the ATG start codon of the endogenous allele. This approach restored regulated cell surface expression of CD40L to X-HIGM T cells, disrupted expression of the mutant protein, and resulted in T cells that induced B cell class-switching in vitro, thus demonstrating its potential as a T cell therapy for X-HIGM.Although the pathologies of X-HIGM are attributed mostly to the T cell defects, CD40L is expressed in most hematopoietic lineages. Besides providing a stable X-HIGM treatment, editing of autologous hematopoietic stem cells (HSC) would likely rescue regulated CD40L expression in all lineages. A selective advantage for gene corrected cells is not anticipated; however, patients with mixed donor chimerism post-transplantation have substantial improvements when as few as 10% of HSC have the WT allele. Here we report gene editing of CD40LG in adult human CD34+ PBSC at rates that are anticipated to provide such clinical benefit. Initial experiments using mRNA delivery of TALEN pairs targeting exon 1 of the CD40LG locus in human CD34+ cells demonstrated indel frequencies of >50%. To investigate the potential for HDR at the CD40LG locus in adult CD34+ cells, we combined delivery of the TALEN with an AAV6 donor template containing an MND promoter-GFP expression cassette flanked by 1 kb CD40LG homology arms. This donor template allowed us to track editing rates within the CD40LG locus (normally silent in HSCs) by flow cytometry. Using this co-delivery strategy, we consistently achieved editing rates of ~30% across multiple control human stem cell donors. Edited cells demonstrated minimal loss of viability, and expansion rates in culture equivalent to controls. Edited cells have been transplanted into immune deficient, NSG mice in order to track engraftment and differentiation potential. In parallel, we are currently evaluating editing rates using a more clinically relevant, promoter-less donor template encoding the CD40L cDNA. In summary, we have achieved clinically relevant rates of gene editing within the endogenous CD40LG locus in human HSC, setting the stage for additional work required to translate this approach into clinical application.


Molecular Therapy | 2016

569. Precision Editing of the WAS Locus via Homologous Recombination in Primary Human Hematopoietic Cells Mediated by Either TALEN or CRISPR/Cas Nucleases

Iram Khan; Yupeng Wang; Courtnee Clough; Swati Singh; King Hung; David J. Rawlings

Wiskott-Aldrich Syndrome (WAS) is an inherited primary immunodeficiency caused by mutations in the WAS gene which encodes a protein (WASp) that regulates the actin cytoskeleton in multiple hematopoietic cell lineages. Currently, allogeneic stem cell transplantation constitutes the only available cure for WAS, although phase I/II clinical trials using lentiviral gene therapy of autologous stem cells are currently underway and have shown improvement in immune system defects. Although effective, the gene therapy approach carries a risk of insertional mutagenesis, and unpredictable expression due to promoter choice as well as influences of the random integration site, including epigenetic status and the presence of neighboring transcription regulatory elements. As a refinement, our goal is to develop gene editing methodologies that would allow the specific targeting of a WASp cDNA to a position within the WAS locus allowing transcriptional regulation by the endogenous WAS promoter. To this end, we first designed and tested the cleavage efficiency of several guide RNAs (delivered as self-complementary AAV along with Cas9, delivered as mRNA), as well as candidate TALEN pairs (delivered as mRNAs), in primary human T cells. Using the T7 endonuclease assay, we identified candidate nucleases from both platforms (CRISPR/Cas9 and TALEN) that achieved a high Indel frequency: 73 and 85%, respectively. We next created a synthetic AAV6 donor template for homology-directed repair (HDR) that contained an MND promoter driven GFP cDNA with 1kb of WAS homology arms flanking it. When delivered with the TALEN or CRISPR/Cas9 nucleases, we observed stable integration of the GFP reporter within >25% of primary human T cells. Subsequently, we utilzed the identical reagents to target integration of the reporter cDNA into the WAS locus in adult human mobilized CD34+ cells, albeit at lower efficiencies. The off-target cleavage sites for TALENs identified using the Prognos software were amplified and sequenced, with no evidence of off-target cleavage observed at any of the predicted loci. Thus, we have generated genome editing tools that possess a high degree of specificity, providing the foundation for site-specific modification of the WAS locus as a therapeutic option.

Collaboration


Dive into the Iram Khan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew M. Scharenberg

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Swati Singh

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Karen Sommer

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Malika Hale

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jaya Sahni

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Blythe D. Sather

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Courtnee Clough

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Gabrielle Curinga

Seattle Children's Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge