Network


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

Hotspot


Dive into the research topics where Xavier M. Anguela is active.

Publication


Featured researches published by Xavier M. Anguela.


Nature | 2011

In vivo genome editing restores haemostasis in a mouse model of haemophilia

Hojun Li; Virginia Haurigot; Yannick Doyon; Tianjian Li; Sunnie Y. Wong; Anand S. Bhagwat; Nirav Malani; Xavier M. Anguela; Rajiv Sharma; Lacramiora Ivanciu; Samuel L. Murphy; Jonathan D. Finn; Fayaz R. Khazi; Shangzhen Zhou; David Paschon; Edward J. Rebar; Frederic D. Bushman; Philip D. Gregory; Michael C. Holmes; Katherine A. High

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


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 | 2015

In vivo genome editing of the albumin locus as a platform for protein replacement therapy.

Rajiv Sharma; Xavier M. Anguela; Yannick Doyon; Thomas Wechsler; Russell DeKelver; Scott Sproul; David Paschon; Jeffrey C. Miller; Robert J. Davidson; David A. Shivak; Shangzhen Zhou; Julianne Rieders; Philip D. Gregory; Michael C. Holmes; Edward J. Rebar; Katherine A. High

Site-specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: zinc finger nuclease (ZFN) -mediated site-specific integration of therapeutic transgenes within the albumin gene. By using adeno-associated viral (AAV) vector delivery in vivo, we achieved long-term expression of human factors VIII and IX (hFVIII and hFIX) in mouse models of hemophilia A and B at therapeutic levels. By using the same targeting reagents in wild-type mice, lysosomal enzymes were expressed that are deficient in Fabry and Gaucher diseases and in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and nongenetic diseases.


Blood | 2013

Robust ZFN-mediated genome editing in adult hemophilic mice

Xavier M. Anguela; Rajiv Sharma; Yannick Doyon; Jeffrey C. Miller; Hojun Li; Virginia Haurigot; Michelle Rohde; Sunnie Y. Wong; Robert J. Davidson; Shangzhen Zhou; Philip D. Gregory; Michael C. Holmes; Katherine A. High

Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.


The New England Journal of Medicine | 2017

Hemophilia B Gene Therapy with a High-Specific-Activity Factor IX Variant

Lindsey A. George; Spencer K. Sullivan; Adam Giermasz; John E.J. Rasko; Benjamin J. Samelson-Jones; Jonathan M. Ducore; Adam Cuker; Lisa M. Sullivan; Suvankar Majumdar; Jerome Teitel; Catherine E. McGuinn; Margaret V. Ragni; Alvin Luk; Daniel Hui; J. Fraser Wright; Yifeng Chen; Yun Liu; Katie Wachtel; Angela Winters; Stefan Tiefenbacher; Valder R. Arruda; Johannes C.M. van der Loo; Olga Zelenaia; Daniel Takefman; Marcus E. Carr; Linda B. Couto; Xavier M. Anguela; Katherine A. High

Background The prevention of bleeding with adequately sustained levels of clotting factor, after a single therapeutic intervention and without the need for further medical intervention, represents an important goal in the treatment of hemophilia. Methods We infused a single‐stranded adeno‐associated viral (AAV) vector consisting of a bioengineered capsid, liver‐specific promoter and factor IX Padua (factor IX–R338L) transgene at a dose of 5×1011 vector genomes per kilogram of body weight in 10 men with hemophilia B who had factor IX coagulant activity of 2% or less of the normal value. Laboratory values, bleeding frequency, and consumption of factor IX concentrate were prospectively evaluated after vector infusion and were compared with baseline values. Results No serious adverse events occurred during or after vector infusion. Vector‐derived factor IX coagulant activity was sustained in all the participants, with a mean (±SD) steady‐state factor IX coagulant activity of 33.7±18.5% (range, 14 to 81). On cumulative follow‐up of 492 weeks among all the participants (range of follow‐up in individual participants, 28 to 78 weeks), the annualized bleeding rate was significantly reduced (mean rate, 11.1 events per year [range, 0 to 48] before vector administration vs. 0.4 events per year [range, 0 to 4] after administration; P=0.02), as was factor use (mean dose, 2908 IU per kilogram [range, 0 to 8090] before vector administration vs. 49.3 IU per kilogram [range, 0 to 376] after administration; P=0.004). A total of 8 of 10 participants did not use factor, and 9 of 10 did not have bleeds after vector administration. An asymptomatic increase in liver‐enzyme levels developed in 2 participants and resolved with short‐term prednisone treatment. One participant, who had substantial, advanced arthropathy at baseline, administered factor for bleeding but overall used 91% less factor than before vector infusion. Conclusions We found sustained therapeutic expression of factor IX coagulant activity after gene transfer in 10 participants with hemophilia who received the same vector dose. Transgene‐derived factor IX coagulant activity enabled the termination of baseline prophylaxis and the near elimination of bleeding and factor use. (Funded by Spark Therapeutics and Pfizer; ClinicalTrials.gov number, NCT02484092.)


Human Molecular Genetics | 2016

Adeno-associated viral vectors for the treatment of hemophilia

Katherine A. High; Xavier M. Anguela

Gene transfer studies for the treatment of hemophilia began more than two decades ago. A large body of pre-clinical work evaluated a variety of vectors and target tissues, but by the start of the new millennium it became evident that adeno-associated viral (AAV)-mediated gene transfer to the liver held great promise as a therapeutic tool. The transition to the clinical arena uncovered a number of unforeseen challenges, mainly in the form of a human-specific immune response against the vector that poses a significant limitation in the application of this technology. While the full nature of this response has not been elucidated, long-term expression of therapeutic levels of factor IX is already a reality for a small number of patients. Extending this success to a greater number of hemophilia B patients remains a major goal of the field, as well as translating this strategy to clinical therapy for hemophilia A. This review summarizes the progress of AAV-mediated gene therapy for the hemophilias, along with its upcoming prospects and challenges.


Molecular Therapy | 2013

Preclinical evaluation of an anti-HCV miRNA cluster for treatment of HCV infection.

Xiao Yang; Katherine T. Marcucci; Xavier M. Anguela; Linda B. Couto

We developed a strategy to treat hepatitis C virus (HCV) infection by replacing five endogenous microRNA (miRNA) sequences of a natural miRNA cluster (miR-17-92) with sequences that are complementary to the HCV genome. This miRNA cluster (HCV-miR-Cluster 5) is delivered to cells using adeno-associated virus (AAV) vectors and the miRNAs are expressed in the liver, the site of HCV replication and assembly. AAV-HCV-miR-Cluster 5 inhibited bona fide HCV replication in vitro by up to 95% within 2 days, and the spread of HCV to uninfected cells was prevented by continuous expression of the anti-HCV miRNAs. Furthermore, the number of cells harboring HCV RNA replicons decreased dramatically by sustained expression of the anti-HCV miRNAs, suggesting that the vector is capable of curing cells of HCV. Delivery of AAV-HCV-miR-Cluster 5 to mice resulted in efficient transfer of the miRNA gene cluster and expression of all five miRNAs in liver tissue, at levels up to 1,300 copies/cell. These levels achieved up to 98% gene silencing of cognate HCV sequences, and no liver toxicity was observed, supporting the safety of this approach. Therefore, AAV-HCV-miR-Cluster 5 represents a different paradigm for the treatment of HCV infection.


Nature Biotechnology | 2016

An edible switch for gene therapy

Xavier M. Anguela; Katherine A. High

The expression of therapeutic transgenes in mice is made responsive to the amino acid content of the diet.


Molecular Therapy | 2016

52. Therapeutic Factor VIII Expression After AAV Delivery in Non-Human Primates

Xavier M. Anguela; Liron Elkouby; Raffaella Toso; Marti A. DiPietro; Robert J. Davidson; Katherine A. High; Denise E. Sabatino

Adeno-associated viral (AAV) vector delivery of factor VIII (FVIII) has been challenging due to its intrinsic properties that result in inefficient expression compared to similarly sized proteins. Early studies of AAV delivery in hemophilia A mice and dogs suggested that the therapeutic vector dose for FVIII will be higher than for factor IX. However, higher vector loads may induce stronger immune responses against capsid antigens, as evidenced in the clinical studies of AAV delivery for hemophilia B. The use of codon-optimization and novel FVIII variants with enhanced biological properties may provide strategies to increase FVIII expression or secretion to support clinical studies for hemophilia A. One published study has reported clinically relevant levels of hFVIII following AAV-hFVIII delivery in non-human primates (NHPs). This study utilized a hFVIII variant that included a 17 amino acid synthetic sequence within the 14 amino acid B-domain region that increased hFVIII expression compared to the parental B-domain deleted FVIII-SQ transgene (McIntosh, 2013). While this and other variants may increase expression after AAV delivery, the use of non-native FVIII sequences may also increase the risk of development of neutralizing antibodies to potential neoantigens. In order to generate an AAV-hFVIII vector capable of expressing therapeutic levels of FVIII at a clinically relevant vector dose without introduction of any neoantigens, 28 hFVIII-SQ sequences were generated and introduced into our optimized expression cassette containing a modified transthyretin (TTRm) promoter. The constructs were initially screened by hydrodynamic delivery of plasmid DNA which identified 11 candidates that expressed FVIII 2-7 fold higher than our first generation codon optimized construct, CO3. AAV vectors (n=9) were generated using a novel AAV capsid, Spark100, with the best performing FVIII constructs. Hem A/CD4 KO mice were administered the vectors alongside CO3 (4×10e12vg/kg). At 8 weeks post vector administration, 2/9 expressed hFVIII similar to CO3, 5/9 were 4-8 fold higher than CO3 while 2/9 (SPK-8003 and SPK-8005) were >10 fold more potent than CO3. SPK-8005 was then evaluated in a dose escalation study in cynomologus macaques (n=3/group) treated with 3 doses: 2×10e12, 5×10e12 and 1×10e13 vg/kg and compared to vehicle controls (n=2). At 2 weeks post AAV administration, average hFVIII levels in the low, mid and high dose cohorts were 12.7 ± 2.1, 22.6 ± 0.8 and 54.1 ± 15.6 percent of normal, respectively. By 3-4 weeks, hFVIII expression started to decline in most of the animals concomitant with generation of antibodies against human FVIII. Of note, this is an expected and well-described observation that occurs in immune competent animal models due to differences between human and endogenous FVIII protein sequences. The 2 macaques that did not develop anti-hFVIII antibodies had sustained FVIII expression through the last time point evaluated. Finally, no vector-related toxicity events were observed. In summary, extensive codon-optimization identified novel AAV-hFVIII constructs capable of achieving therapeutic FVIII levels in macaques at clinically relevant doses. To our knowledge, the hFVIII levels observed in this study are the highest reported in a large animal model after treatment with an AAV vector expressing an unmodified FVIII-SQ protein. These safety and efficacy results in NHPs support the use of SPK-8005 hepatic gene transfer for the potential treatment of hemophilia A.


Blood | 2016

Spk-9001: Adeno-Associated Virus Mediated Gene Transfer for Hemophilia B Achieves Sustained Mean Factor IX Activity Levels of >30% without Immunosuppression

Lindsey A. George; Spencer K. Sullivan; Adam Giermasz; Jonathan M. Ducore; Jerome Teitel; Adam Cuker; Lisa Sullivan; Suvankar Majumdar; Catherine E. McGuinn; Aline M. Galvão; Alvin Luk; J. Fraser Wright; Yifeng Chen; Daniel J. Hui; Katie Wachtel; Teresa Urich; Daniel Takefman; Linda B. Couto; Marcus E. Carr; Xavier M. Anguela; Katherine A. High

Collaboration


Dive into the Xavier M. Anguela's collaboration.

Top Co-Authors

Avatar

Katherine A. High

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Robert J. Davidson

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Shangzhen Zhou

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajiv Sharma

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Federico Mingozzi

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Hojun Li

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge