Network


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

Hotspot


Dive into the research topics where Gregory M. Podsakoff is active.

Publication


Featured researches published by Gregory M. Podsakoff.


Nature Medicine | 1999

Long-term correction of canine hemophilia B by gene transfer of blood coagulation factor IX mediated by adeno-associated viral vector

Roland W. Herzog; Edmund Y Yang; Linda B. Couto; J. Nathan Hagstrom; Dan Elwell; Paul A. Fields; Melissa Burton; Dwight A. Bellinger; Marjorie S. Read; Kenneth M. Brinkhous; Gregory M. Podsakoff; Timothy C. Nichols; Gary J. Kurtzman; Katherine A. High

Hemophilia B is a severe X-linked bleeding diathesis caused by the absence of functional blood coagulation factor IX, and is an excellent candidate for treatment of a genetic disease by gene therapy. Using an adeno-associated viral vector, we demonstrate sustained expression (>17 months) of factor IX in a large-animal model at levels that would have a therapeutic effect in humans (up to 70 ng/ml, adequate to achieve phenotypic correction, in an animal injected with 8.5 × 1012 vector particles/kg). The five hemophilia B dogs treated showed stable, vector dose-dependent partial correction of the whole blood clotting time and, at higher doses, of the activated partial thromboplastin time. In contrast to other viral gene delivery systems, this minimally invasive procedure, consisting of a series of percutaneous intramuscular injections at a single timepoint, was not associated with local or systemic toxicity. Efficient gene transfer to muscle was shown by immunofluorescence staining and DNA analysis of biopsied tissue. Immune responses against factor IX were either absent or transient. These data provide strong support for the feasibility of the approach for therapy of human subjects.


Science Translational Medicine | 2013

Overcoming Preexisting Humoral Immunity to AAV Using Capsid Decoys

Federico Mingozzi; Xavier M. Anguela; Giulia Pavani; Yifeng Chen; Robert J. Davidson; Daniel J. Hui; Mustafa Yazicioglu; Liron Elkouby; Christian Hinderer; Armida Faella; Carolann Howard; Alex Tai; Gregory M. Podsakoff; Shangzhen Zhou; Etiena Basner-Tschakarjan; John Fraser Wright; Katherine A. High

Capsid decoys enhance the efficacy of AAV vector transduction after systemic delivery in the presence of neutralizing antibodies. A Slight of Hand for Gene Therapy Gene therapy has been quite successful—in animal models. But when it comes to translating gene therapy to humans, there have only been a few shining successes. One limiting factor has been the vectors used. Adeno-associated virus (AAV) vectors are safe, noninvasive, and potentially effective; however, people who have been previously exposed to AAV have preexisting neutralizing antibodies that block gene delivery. Now, Mingozzi et al. trick these antibodies into binding empty viral capsid, overcoming their inhibitory effects. The authors hypothesized that introducing empty capsids along with the gene therapy vector would titrate out the neutralizing antibody response to AAV, allowing for successful gene therapy even in the presence of preexisting neutralizing antibodies. They found that varying the ratio of empty capsid to gene therapy vector could successfully inhibit the neutralizing antibody response in both human serum and a mouse model by serving as a decoy for antibody binding. The authors then mutated the receptor binding site of their capsid so that it could bind the neutralizing antibody but not target cells, further increasing the safety profile of this approach. These capsid decoys worked in a dose-dependent manner and were successful even with high antibody titers. What’s more, they were safe and effective in rhesus macaques. Although this approach remains to be tested in humans, tricking neutralizing antibodies with decoys may be the next step in advancing gene therapy in the clinic. Adeno-associated virus (AAV) vectors delivered through the systemic circulation successfully transduce various target tissues in animal models. However, similar attempts in humans have been hampered by the high prevalence of neutralizing antibodies to AAV, which completely block vector transduction. We show in both mouse and nonhuman primate models that addition of empty capsid to the final vector formulation can, in a dose-dependent manner, adsorb these antibodies, even at high titers, thus overcoming their inhibitory effect. To further enhance the safety of the approach, we mutated the receptor binding site of AAV2 to generate an empty capsid mutant that can adsorb antibodies but cannot enter a target cell. Our work suggests that optimizing the ratio of full/empty capsids in the final formulation of vector, based on a patient’s anti-AAV titers, will maximize the efficacy of gene transfer after systemic vector delivery.


Blood | 2012

Factor IX expression in skeletal muscle of a severe hemophilia B patient 10 years after AAV-mediated gene transfer.

George Buchlis; Gregory M. Podsakoff; Antonetta Radu; Sarah M. Hawk; Alan W. Flake; Federico Mingozzi; Katherine A. High

In previous work we transferred a human factor IX-encoding adeno-associated viral vector (AAV) into skeletal muscle of men with severe hemophilia B. Biopsy of injected muscle up to 1 year after vector injection showed evidence of gene transfer by Southern blot and of protein expression by IHC and immunofluorescent staining. Although the procedure appeared safe, circulating F.IX levels remained subtherapeutic (< 1%). Recently, we obtained muscle tissue from a subject injected 10 years earlier who died of causes unrelated to gene transfer. Using Western blot, IHC, and immunofluorescent staining, we show persistent factor IX expression in injected muscle tissue. F.IX transcripts were detected in injected skeletal muscle using RT-PCR, and isolated whole genomic DNA tested positive for the presence of the transferred AAV vector sequence. This is the longest reported transgene expression to date from a parenterally administered AAV vector, with broad implications for the future of muscle-directed gene transfer.


Molecular therapy. Methods & clinical development | 2015

AAV capsid CD8+ T-cell epitopes are highly conserved across AAV serotypes

Daniel J. Hui; Shyrie C Edmonson; Gregory M. Podsakoff; Gary C Pien; Lacramioara Ivanciu; Rodney M. Camire; Hildegund C.J. Ertl; Federico Mingozzi; Katherine A. High; Etiena Basner-Tschakarjan

Adeno-associated virus (AAV) has become one of the most promising vectors in gene transfer in the last 10 years with successful translation to clinical trials in humans and even market approval for a first gene therapy product in Europe. Administration to humans, however, revealed that adaptive immune responses against the vector capsid can present an obstacle to sustained transgene expression due to the activation and expansion of capsid-specific T cells. The limited number of peripheral blood mononuclear cells (PBMCs) obtained from samples within clinical trials allows for little more than monitoring of T-cell responses. We were able to identify immunodominant major histocompatibility complex (MHC) class I epitopes for common human leukocyte antigen (HLA) types by using spleens isolated from subjects undergoing splenectomy for non-malignant indications as a source of large numbers of lymphocytes and restimulating them with single AAV capsid peptides in vitro. Further experiments confirmed that these epitopes are naturally processed and functionally relevant. The design of more effective and less immunogenic AAV vectors, and precise immune monitoring of vector-infused subjects, are facilitated by these findings.


Cancer Genetics and Cytogenetics | 2016

Clonal evolution and clinical significance of copy number neutral loss of heterozygosity of chromosome arm 6p in acquired aplastic anemia

Marisol Betensky; Daria V. Babushok; Jacquelyn J. Roth; Philip J. Mason; Jaclyn A. Biegel; Tracy M. Busse; Yimei Li; Curt Lind; Anna Papazoglou; Dimitri Monos; Gregory M. Podsakoff; Monica Bessler; Timothy S. Olson

Acquired aplastic anemia (aAA) results from the T cell-mediated autoimmune destruction of hematopoietic stem cells. Factors predicting response to immune suppression therapy (IST) or development of myelodysplastic syndrome (MDS) are beginning to be elucidated. Our recent data suggest most patients with aAA treated with IST develop clonal somatic genetic alterations in hematopoietic cells. One frequent acquired abnormality is copy-number neutral loss of heterozygosity on chromosome 6p (6p CN-LOH) involving the human leukocyte antigen (HLA) locus. We hypothesized that because 6p CN-LOH clones may arise from selective pressure to escape immune surveillance through deletion of HLA alleles, the development of 6p CN-LOH may affect response to IST. We used single nucleotide polymorphism array genotyping and targeted next-generation sequencing of HLA alleles to assess frequency of 6p CN-LOH, identity of HLA alleles lost through 6p CN-LOH, and impact of 6p CN-LOH on response to IST. 6p CN-LOH clones were present in 11.3% of patients, remained stable over time, and were not associated with development of MDS-defining cytogenetic abnormalities. Notably, no patient with 6p CN-LOH treated with IST achieved a complete response. In summary, clonal 6p CN-LOH in aAA defines a unique subgroup of patients that may provide insights into hematopoietic clonal evolution.


PLOS ONE | 2015

Impaired Telomere Maintenance and Decreased Canonical WNT Signaling but Normal Ribosome Biogenesis in Induced Pluripotent Stem Cells from X-Linked Dyskeratosis Congenita Patients.

Bai-Wei Gu; Marisa Apicella; Jason A. Mills; Jian-Meng Fan; Dara Akosua Reeves; Deborah L. French; Gregory M. Podsakoff; Monica Bessler; Philip J. Mason

Dyskeratosis congenita (DC) is an inherited bone marrow failure syndrome characterized by the presence of short telomeres at presentation. Mutations in ten different genes, whose products are involved in the telomere maintenance pathway, have been shown to cause DC. The X-linked form is the most common form of the disease and is caused by mutations in the gene DKC1, encoding the protein dyskerin. Dyskerin is required for the assembly and stability of telomerase and is also involved in ribosomal RNA (rRNA) processing where it converts specific uridines to pseudouridine. DC is thought to result from failure to maintain tissues, like blood, that are renewed by stem cell activity, but research into pathogenic mechanisms has been hampered by the difficulty of obtaining stem cells from patients. We reasoned that induced pluripotent stem (iPS) cells from X-linked DC patients may provide information about the mechanisms involved. Here we describe the production of iPS cells from DC patients with DKC1 mutations Q31E, A353V and ΔL37. In addition we constructed “corrected” lines with a copy of the wild type dyskerin cDNA expressed from the AAVS1 safe harbor locus. We show that in iPS cells with DKC1 mutations telomere maintenance is compromised with short telomere lengths and decreased telomerase activity. The degree to which telomere lengths are affected by expression of telomerase during reprograming, or with ectopic expression of wild type dyskerin, is variable. The recurrent mutation A353V shows the most severe effect on telomere maintenance. A353V cells but not Q31E or ΔL37 cells, are refractory to correction by expression of wild type DKC1 cDNA. Because dyskerin is involved in both telomere maintenance and ribosome biogenesis it has been postulated that defective ribosome biogenesis and translation may contribute to the disease phenotype. Evidence from mouse and zebra fish models has supported the involvement of ribosome biogenesis but primary cells from human patients have so far not shown defects in pseudouridylation or ribosomal RNA processing. None of the mutant iPS cells presented here show decreased pseudouridine levels in rRNA or defective rRNA processing suggesting telomere maintenance defects account for most of the phenotype of X-linked DC. Finally gene expression analysis of the iPS cells shows that WNT signaling is significantly decreased in all mutant cells, raising the possibility that defective WNT signaling may contribute to disease pathogenesis.


British Journal of Haematology | 2014

Single nucleotide polymorphism array analysis of bone marrow failure patients reveals characteristic patterns of genetic changes.

Daria V. Babushok; Hongbo M. Xie; Jacquelyn J. Roth; Nieves Perdigones; Timothy S. Olson; Joshua D. Cockroft; Xiaowu Gai; Juan C. Perin; Yimei Li; Michele Paessler; Hakon Hakonarson; Gregory M. Podsakoff; Philip J. Mason; Jaclyn A. Biegel; Monica Bessler

The bone marrow failure syndromes (BMFS) are a heterogeneous group of rare blood disorders characterized by inadequate haematopoiesis, clonal evolution, and increased risk of leukaemia. Single nucleotide polymorphism arrays (SNP‐A) have been proposed as a tool for surveillance of clonal evolution in BMFS. To better understand the natural history of BMFS and to assess the clinical utility of SNP‐A in these disorders, we analysed 124 SNP‐A from a comprehensively characterized cohort of 91 patients at our BMFS centre. SNP‐A were correlated with medical histories, haematopathology, cytogenetic and molecular data. To assess clonal evolution, longitudinal analysis of SNP‐A was performed in 25 patients. We found that acquired copy number‐neutral loss of heterozygosity (CN‐LOH) was significantly more frequent in acquired aplastic anaemia (aAA) than in other BMFS (odds ratio 12·2, P < 0·01). Homozygosity by descent was most common in congenital BMFS, frequently unmasking autosomal recessive mutations. Copy number variants (CNVs) were frequently polymorphic, and we identified CNVs enriched in neutropenia and aAA. Our results suggest that acquired CN‐LOH is a general phenomenon in aAA that is probably mechanistically and prognostically distinct from typical CN‐LOH of myeloid malignancies. Our analysis of clinical utility of SNP‐A shows the highest yield of detecting new clonal haematopoiesis at diagnosis and at relapse.


Human Gene Therapy | 2013

Effects of immunosuppression on circulating adeno-associated virus capsid-specific T cells in humans.

Elizabeth M. Parzych; Hua Li; Xiangfan Yin; Qin Liu; Te-Lang Wu; Gregory M. Podsakoff; Katherine A. High; Matthew H. Levine; Hildegund C.J. Ertl

In humans adeno-associated virus (AAV)-mediated gene transfer is followed by expansion of AAV capsid-specific T cells, evidence of cell damage, and loss of transgene product expression, implicating immunological rejection of vector-transduced cells, which may be prevented by immunosuppressive drugs. We undertook this study to assess the effect of immunosuppression (IS) used for organ transplantation on immune responses to AAV capsid antigens. Recipients of liver or kidney transplants were tested before and 4 weeks after induction of IS in comparison with matched samples from healthy human adults and an additional cohort with comorbid conditions similar to those of the transplant patients. Our data show that transplant patients and comorbid control subjects have markedly higher frequencies of circulating AAV capsid-specific T cells compared with healthy adults. On average, IS resulted in a reduction of AAV-specific CD4⁺ T cells, whereas numbers of circulating CD8⁺ effector and central memory T cells tended to increase. Independent of the type of transplant or the IS regimens, the trend of AAV capsid-specific T cell responses after drug treatment varied; in some patients responses were unaffected whereas others showed decreases or even pronounced increases, casting doubt on the usefulness of prophylactic IS for AAV vector recipients.


Cancer Research | 2015

Abstract 2977: Most patients with acquired aplastic anemia develop clonal hematopoiesis early in disease

Daria V. Babushok; Nieves Perdigones; Juan C. Perin; Timothy S. Olson; Wenda Ye; Jacquelyn J. Roth; Curt Lind; Carine Cattier; Yimei Li; Helge Hartung; Michele Paessler; Dale Frank; Hongbo M. Xie; Tracy M. Busse; Shanna Cross; Gregory M. Podsakoff; Dimitrios Monos; Jaclyn A. Biegel; Philip J. Mason; Monica Bessler

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Clonal hematopoiesis is an expansion of hematopoietic stem cells, caused by somatic mutations or epigenetic changes that confer a growth advantage to the host cell. Although recently recognized as a phenomenon of aging, clonal hematopoiesis has been traditionally associated with pre-cancerous states and malignant transformation. Acquired aplastic anemia (AA), a non-neoplastic autoimmune blood disorder occurring in children and adults, has been associated with clonal hematopoietic disorders; transformation to myelodysplastic syndrome (MDS) or acute leukemia is a late complication in 10-15% of AA patients. Based on the association of AA with clonal disorders, we hypothesized that clonal hematopoiesis is a general phenomenon in AA, and can be seen in the majority of AA patients, including children. To evaluate somatic genetic changes in AA, we used a combination of single nucleotide polymorphism array (SNP-A) genotyping and comparative whole exome sequencing of paired bone marrow aspirates and skin in twenty nine patients with AA. All somatic mutations were validated by bi-directional Sanger sequencing. The median age of diagnosis was 14 years (range 1.5-65). Patients were analyzed at a median of 1.1 years from diagnosis. None of the patients had histopathological evidence of MDS at the time of analysis. Somatic mutations were identified in the majority of patients, including patients with pediatric-onset AA. Three patients (10%) had somatic loss-of-function mutations in HLA class I alleles. Although MDS-associated mutations were identified in 2 of 29 patients, the majority of mutations were not in genes associated with MDS and hematologic malignancies. Pathway analysis of mutated genes revealed an enrichment of genes in pathways of immunity and transcriptional regulation. Comparison of somatic mutations in AA to a patient with a 30-year history of AA who progressed to MDS revealed that, unlike in AA, which was characterized by diverse and frequently oligoclonal hematopoiesis, progression to MDS was associated with an expansion of a dominant clone carrying multiple classical mutations linked to malignancy: pathogenic mutations in SUZ12 (homozygous for the mutated region due to copy number-neutral loss of heterozygosity (CN-LOH) at the chromosomal region 17q11.2qter), ASXL1, RUNX1, and PHF6. In conclusion, our data show that clonal hematopoiesis emerges in the majority of patients with AA, including children and young adults, can be detected early in disease, and has a mutational spectrum largely distinct from MDS. Our results highlight that in the absence of morphologic features of myelodysplasia, the presence of clonal hematopoiesis with somatic mutations cannot be used to distinguish MDS from AA. Future longitudinal studies of clonal hematopoiesis in AA will help to explain differences in patients’ disease course, and will enable personalized treatment approaches in AA. Citation Format: Daria V. Babushok, Nieves Perdigones, Juan C. Perin, Timothy S. Olson, Wenda Ye, Jacquelyn J. Roth, Curt Lind, Carine Cattier, Yimei Li, Helge Hartung, Michele E. Paessler, Dale M. Frank, Hongbo M. Xie, Tracy M. Busse, Shanna Cross, Gregory M. Podsakoff, Dimitrios Monos, Jaclyn A. Biegel, Philip J. Mason, Monica Bessler. Most patients with acquired aplastic anemia develop clonal hematopoiesis early in disease. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2977. doi:10.1158/1538-7445.AM2015-2977


Proceedings of the National Academy of Sciences of the United States of America | 1996

Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein

Paul D. Kessler; Gregory M. Podsakoff; Xiaojuan Chen; Susan A. Mcquiston; Peter Colosi; Laura A. Matelis; Gary J. Kurtzman; Barry J. Byrne

Collaboration


Dive into the Gregory M. Podsakoff's collaboration.

Top Co-Authors

Avatar

Monica Bessler

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Philip J. Mason

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katherine A. High

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Jaclyn A. Biegel

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Timothy S. Olson

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Yimei Li

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Daria V. Babushok

Hospital of the University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Donald B. Kohn

University of California

View shared research outputs
Top Co-Authors

Avatar

Hongbo M. Xie

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge