Network


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

Hotspot


Dive into the research topics where Nicholas Hubbard is active.

Publication


Featured researches published by Nicholas Hubbard.


The Journal of Allergy and Clinical Immunology | 2013

Diagnosis and evaluation of primary panhypogammaglobulinemia: A molecular and genetic challenge

Maaz S. Mohiuddin; Jordan K. Abbott; Nicholas Hubbard; Troy R. Torgerson; Hans D. Ochs; Erwin W. Gelfand

1. Review the target audience, learning objectives and author disclosures. 2. Complete the pre-test online at www.jacionline.org (click on the Online CME heading). 3. Follow the online instructions to read the full version of the article, including the clinical vignette and review components. 4. Complete the post-test. At this time, you will have earned 1.00 AMA PRA Category 1 CME Credit. 5. Approximately 4 weeks later you will receive an online assessment regarding your application of this article to your practice. Once you have completed this assessment, you will be eligible to receive 2 MOC Part II Self-Assessment credits from the American Board of Allergy and Immunology.


Blood Advances | 2018

Rapid immune reconstitution of SCID-X1 canines after G-CSF/AMD3100 mobilization and in vivo gene therapy.

Olivier Humbert; Frieda Chan; Yogendra S. Rajawat; Troy R. Torgerson; Christopher R. Burtner; Nicholas Hubbard; Daniel Humphrys; Zachary K. Norgaard; Patricia O’Donnell; Jennifer E. Adair; Grant D. Trobridge; Andrew M. Scharenberg; Peter J. Felsburg; David J. Rawlings; Hans-Peter Kiem

Hematopoietic stem-cell gene therapy is a promising treatment of X-linked severe combined immunodeficiency disease (SCID-X1), but currently, it requires recipient conditioning, extensive cell manipulation, and sophisticated facilities. With these limitations in mind, we explored a simpler therapeutic approach to SCID-X1 treatment by direct IV administration of foamy virus (FV) vectors in the canine model. FV vectors were used because they have a favorable integration site profile and are resistant to serum inactivation. Here, we show improved efficacy of our in vivo gene therapy platform by mobilization with granulocyte colony-stimulating factor (G-CSF) and AMD3100 before injection of an optimized FV vector incorporating the human phosphoglycerate kinase enhancerless promoter. G-CSF/AMD3100 mobilization before FV vector delivery accelerated kinetics of CD3+ lymphocyte recovery, promoted thymopoiesis, and increased immune clonal diversity. Gene-corrected T lymphocytes exhibited a normal CD4:CD8 ratio and a broad T-cell receptor repertoire and showed restored γC-dependent signaling function. Treated animals showed normal primary and secondary antibody responses to bacteriophage immunization and evidence for immunoglobulin class switching. These results demonstrate safety and efficacy of an accessible, portable, and translatable platform with no conditioning regimen for the treatment of SCID-X1 and other genetic diseases.


Molecular therapy. Methods & clinical development | 2018

Efficient Enrichment of Gene-Modified Primary T Cells via CCR5-Targeted Integration of Mutant Dihydrofolate Reductase

Biswajit Paul; Guillermo S. Romano Ibarra; Nicholas Hubbard; Teresa Einhaus; Alexander Astrakhan; David J. Rawlings; Hans-Peter Kiem; Christopher W. Peterson

Targeted gene therapy strategies utilizing homology-driven repair (HDR) allow for greater control over transgene integration site, copy number, and expression—significant advantages over traditional vector-mediated gene therapy with random genome integration. However, the relatively low efficiency of HDR-based strategies limits their clinical application. Here, we used HDR to knock in a mutant dihydrofolate reductase (mDHFR) selection gene at the gene-edited CCR5 locus in primary human CD4+ T cells and selected for mDHFR-modified cells in the presence of methotrexate (MTX). Cells were transfected with CCR5-megaTAL nuclease mRNA and transduced with adeno-associated virus containing an mDHFR donor template flanked by CCR5 homology arms, leading to up to 40% targeted gene insertion. Clinically relevant concentrations of MTX led to a greater than 5-fold enrichment for mDHFR-modified cells, which maintained a diverse TCR repertoire over the course of expansion and drug selection. Our results demonstrate that mDHFR/MTX-based selection can be used to enrich for gene-modified T cells ex vivo, paving the way for analogous approaches to increase the percentage of HIV-resistant, autologous CD4+ T cells infused into HIV+ patients, and/or for in vivo selection of gene-edited T cells for the treatment of cancer.


Molecular Therapy | 2016

284. Long-Term Therapeutic Immune Reconstitution in XSCID Canine Model via In Vivo Foamy Virus Delivery of Common Gamma Chain

Frieda Chan; Olivier Humbert; Christopher R. Burtner; Daniel Humphrys; Jennifer E. Adair; Grant Trobridge; Patricia O'Donnell; Nicholas Hubbard; Troy Torgerson; Andrew M. Scharenberg; David J. Rawlings; Peter J. Felsburg; Hans-Peter Kiem

X-linked combined immunodeficiency disease (XSCID) is caused by mutation in the common gamma chain, γC (interleukin-2 receptor subunit gamma, IL2RG) in both humans and canines. It is characterized by the inability of T-cell development leading to absence of T-cells in peripheral blood, lack of T-cell mediated immune response, low IgA and IgG levels, and early infant mortality. In the 1990s, human XSCID clinical trials utilizing gamma-retroviral vectors to deliver the IL2RG gene caused leukemia in 5 out of 20 patients due to vector integration in or near proto-oncogenes. Recent studies showed Foamy virus based vectors as an excellent alternative for in vivo gene-therapy because it is non-pathogenic in humans while exhibiting increased serum stability and favorable integration pattern. Previously, we have demonstrated CD3+ T-cell reconstitution in the canine model via intravenous injection of foamy virus expressing human elongation factor-1 alpha promoter (Ef1α)-yC. Unfortunately, the treated animals contained a low number of gene corrected progenitors at a sub-therapeutic level. Here, we achieved long-term therapeutic immune-reconstitution by intravenous delivery of a human phosphoglycerate kinase promoter (Pgk)-mediated γC foamy viral vector into XSCID neonatal canines. Long-term (2 years) post-injection follow-up demonstrated therapeutic levels of CD3+ T-cell expansion. Within the T-cell population, gene correction with Pgk-γC stabilized at ~80%. We validated T-cell functionality by using spectratyping analysis, which exhibited a diverse repertoire of receptor gene rearrangement. Retroviral integration site analysis (RIS) indicated polyclonal contribution to the reconstituted T-cells. Immunoglobulin ELISA assays showed that IgA and IgG levels in peripheral blood are comparable to normal healthy controls. We immunized the gene-corrected canine recipients with bacteriophage ϕx174 and confirmed production of specific IgG antibodies, showing the ability for isotype switching in B-lymphocytes. Currently, the gene-corrected canines exhibit comparable health and physical attributes to normal controls. Furthermore, semen from the gene-corrected male canine was used via artificial insemination to produce a litter of viable offsprings. In summary, our data demonstrate that Pgk-γC foamy viral vector delivered long-term therapeutic gene correction in a large-animal model for XSCID gene therapy. Most importantly, these results indicate that in vivo Pgk-γC foamy vector administration is a viable option for long-term immune reconstitution in future XSCID human clinical trials.


Molecular Therapy | 2016

40. Nuclease-Targeted Gene-Editing of FOXP3 in Primary T Cells Creates a Stable and Functional Treg Phenotype

Nicholas Hubbard; David Hagin; Karen Sommer; Yumei Song; Iram F. Khan; Andrew M. Scharenberg; David J. Rawlings; Troy R. Torgerson

Reprogramming T cells to adopt regulatory T cell (Treg) functions represents a promising strategy for treating autoimmune disorders and graft-versus-host disease. The development and maintenance of Treg cells is dependent on FOXP3 expression, which in turn is tightly controlled through epigenetic modification of intronic conserved non-coding sequences (CNS) surrounding the first coding exon. Two major challenges associated with FOXP3 gene-transfer approaches for Treg reprogramming are: achieving adequately high FOXP3 expression, and avoiding eventual gene silencing. We devised two gene editing strategies with the goal of driving stable expression of FOXP3 in primary human T cells. Using a combination of TALEN-mediated gene disruption and adeno-associated-virus (AAV) delivered donor repair templates, we introduced either an MND promoter upstream of the first coding exon, or we deleted intronic CNS implicated in transcriptional silencing. The donor templates were also designed to co-express FOXP3 with either GFP or EGFRt (truncated Epidermal Growth Factor receptor), to allow tracking and purification of edited cells. Both strategies resulted in T cells stably expressing FOXP3 at high levels (~60 %). Introduction of the MND promoter resulted in the greatest levels of cellular FOXP3 expression (MFI), and these cells showed phenotypic and functional changes consistent with Treg cells, including: surface marker expression (CD25high, CD127low, CTLA4high, LAG3high), suppression of cytokine production (IL-2, IL-17 and IFN-γ), and resistance to Rapamycin. Edited cells also suppressed the proliferation of stimulated T cells in vitro, demonstrating their effective ‘reprogramming’ towards a Treg lineage. Thus, our gene modification strategy allowed us to over-ride (using the MND promoter) or modulate (by deleting CNS elements) endogenous FOXP3 regulatory mechanisms to enforce stable, long-term FOXP3 expression in T cells that were not previously committed to the Treg lineage. This approach, used alone or in combination with selection for disease-relevant TCR specificity or with delivery of a chimeric antigen receptor, is likely to be broadly applicable for producing stable, functionally active Tregs for a range of future clinical applications.


Molecular Therapy | 2016

44. Novel Combination of megaTAL Nuclease-Driven Genome Engineering with a Drug Selection Cassette Increases Efficiency of HIV Gene Therapy

Biswajit Paul; Guillermo Romano; Kevin G. Haworth; Patrick Younan; Nicholas Hubbard; Donald Gisch; Iram Khan; Alexander Astrakhan; David J. Rawlings; Hans-Peter Kiem

Human Immunodeficiency Virus (HIV) infection remains a substantial health problem worldwide. The C-C chemokine receptor 5 (CCR5) serves as a co-receptor for HIV entry into CD4+ T cells and represent an alternative therapeutic target. Early clinical trials using CCR5-targeting zinc finger nucleases demonstrated transient control of HIV infection in the course of antiretroviral treatment interruption (Tebas, NEJM, 2014). Our current work improves on these advances by combining high level of CCR5 gene disruption with preferential selection of gene modified cells. The CCR5-targeting megaTAL combines a LAGLIDADG homing endonuclease scaffold with an eleven repeat transcription activator-like (TAL) effector array to achieve site specific DNA cleavage. This nuclease produces highly efficient CCR5 targeting in primary human CD4+ T cells in vitro (70-90% disruption). To test the protective effects of megaTAL treatment, primary human CD4+ T cells treated with CCR5-megaTAL were transplanted into NOD/SCID/γc-null (NSG) ‘humanized’ mice and challenged with HIV-1. We observed a 100-fold increase of megaTAL-treated cells compared to untreated controls during an active in vivo infection demonstrating the functionality of this approach. Based on the decline of CCR5 modified cells in the clinical trials to date, we hypothesized that we could improve maintenance of HIV resistant cells by expanding them either ex vivo or in vivo. We propose that coupling megaTAL nuclease treatment with drug selection will help us achieve therapeutically relevant levels of HIV-protected cells by enabling efficient selection only of CCR5-modified T-cells. The mutant human dihydrofolate reductase (mDHFR) chemoselection system has been used to render cells resistant to lymphotoxic concentrations of the drug methotrexate (MTX). We tested our experimental approach by transducing cells with lentiviral vector encoding a mDHFR cassette followed by chemoselection in MTX at 0.02uM. This approach resulted in a six fold enrichment of gene modified primary CD4+ T cells ex vivo. Previously we have shown that combining megaTAL treatment with adeno-associated virus (AAV) transduction produces very high rates of homology-driven repair (HDR) in primary human T cells. Hence, we combined megaTAL/AAV treatment to integrate the mutant DHFR into the CCR5 locus, producing a population of MTX-resistant CD4+ cells that also lack CCR5. Primary human CD4+ T cells were transfected with CCR5-megaTAL mRNA and transduced with AAV6 containing a mutant DHFR donor template flanked by 0.6kb CCR5 homology arms. They demonstrated a greater than five-fold enrichment in MTX compared to untreated controls ex vivo. Next, we have transplanted NSG mice with 1×106 gene modified cells/mouse to assess the therapeutic potential of our approach. Mice that engraft effectively will be treated with daily injections of 0, 0.5 and 2 mg/kg of MTX to monitor preferential selection and enrichment of our target cell population. Subsequent studies will assess the long term control of viremia in these mice following HIV challenge. In conclusion, the CCR5-megaTAL nuclease platform produces very high levels of gene-modified CD4+ T-cells and protects these cells from subsequent HIV infection in vivo. Furthermore, combining targeted integration and chemical selection results in the specific selection of gene modified primary human T cells. To our knowledge we are the first group to report MTX-mediated chemoselection and expansion of CD4+ T cells following targeted integration at the CCR5 locus.


Molecular Therapy | 2015

441. Direct Comparison of EF1α and PGK Promoters Reveals Superior Performance of the PGK Promoter for Expression of the Common Gamma Chain in a Canine Model of In Vivo Foamy Virus Gene Therapy for Severe Combined Immunodeficiency

Christopher R. Burtner; Humbert Olivier; Patricia O'Donnell; Nicholas Hubbard; Daniel Humphrys; Jennifer E. Adair; Grant D. Trobridge; Troy R. Torgerson; Andrew M. Scharenberg; David J. Rawlings; Peter J. Felsburg; Hans-Peter Kiem

Foamy viruses (FV) are ideal vectors for in vivo gene therapy because FV infection is non-pathogenic in people, FV are more resistant to serum inactivation than lentiviruses packaged with the vesicular stomatitis virus glycoprotein (VSV-G), and FV exhibit favorable proviral integration patterns compared to gammaretroviruses. In vivo gene therapy with a FV vector expressing the human common gamma chain (γC) under the short EF1α promoter results in a functional immune reconstitution in X-SCID dogs.


Journal of Clinical Immunology | 2013

A novel gain-of-function IKBA mutation underlies ectodermal dysplasia with immunodeficiency and polyendocrinopathy.

Lena F. Schimke; Nikolaus Rieber; Stacey Rylaarsdam; Otavio Cabral-Marques; Nicholas Hubbard; Anne Puel; Laura Kallmann; Stephanie Anover Sombke; Gundula Notheis; Hans Peter Schwarz; Birgit Kammer; Tomas Hökfelt; Reinald Repp; Capucine Picard; Jean-Laurent Casanova; Bernd H. Belohradsky; Michael H. Albert; Hans D. Ochs; Ellen D. Renner; Troy R. Torgerson


Blood | 2016

Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome

Nicholas Hubbard; David Hagin; Karen Sommer; Yumei Song; Iram Khan; Courtnee Clough; Hans D. Ochs; David J. Rawlings; Andrew M. Scharenberg; Troy R. Torgerson


Blood | 2016

Rapid Expansion of Gene-Marked Lymphocytes in X-SCID Dogs after AMD3100+G-CSF-Based Hematopoietic Stem/Progenitor Cell Mobilization and Intravenous Injection of a Common γ-Chain Expressing Foamy Viral Vector

Frieda Chan; Olivier Humbert; Troy R. Torgerson; Nicholas Hubbard; Patricia O'Donnell; Daniel Humphrys; Jennifer E. Adair; Peter J. Felsburg; Grant D. Trobridge; Andrew M. Scharenberg; David J. Rawlings; Hans-Peter Kiem

Collaboration


Dive into the Nicholas Hubbard's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew M. Scharenberg

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Humphrys

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer E. Adair

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Peter J. Felsburg

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Christopher R. Burtner

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Grant D. Trobridge

Washington State University

View shared research outputs
Top Co-Authors

Avatar

Olivier Humbert

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge