Network


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

Hotspot


Dive into the research topics where Giulia Pavani is active.

Publication


Featured researches published by Giulia Pavani.


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.


Nature Biotechnology | 2011

A zymogen-like factor Xa variant corrects the coagulation defect in hemophilia

Lacramioara Ivanciu; Raffaella Toso; Paris Margaritis; Giulia Pavani; Haein Kim; Alexander Schlachterman; Jianhua Liu; Valerie Clerin; Debra D. Pittman; Rosalind Rose-Miranda; Kathleen M. Shields; David V. Erbe; James F. Tobin; Valder R. Arruda; Rodney M. Camire

Effective therapies are needed to control excessive bleeding in a range of clinical conditions. We improve hemostasis in vivo using a conformationally pliant variant of coagulation factor Xa (FXaI16L) rendered partially inactive by a defect in the transition from zymogen to active protease. Using mouse models of hemophilia, we show that FXaI16L has a longer half-life than wild-type FXa and does not cause excessive activation of coagulation. Once clotting mechanisms are activated to produce its cofactor FVa, FXaI16L is driven to the protease state and restores hemostasis in hemophilic animals upon vascular injury. Moreover, using human or murine analogs, we show that FXaI16L is more efficacious than FVIIa, which is used to treat bleeding in hemophilia inhibitor patients. FXaI16L may provide an effective strategy to enhance blood clot formation and act as a rapid pan-hemostatic agent for the treatment of bleeding conditions.Effective therapies are needed to control excessive bleeding in a range of clinical conditions. We describe a surprisingly useful approach to improve hemostasis in vivo using a variant of coagulation factor Xa (FXaI16L). This conformationally pliant derivative is partially inactive due to a defect in transitioning from zymogen to protease 1,2. Using mouse models of hemophilia, we show that FXaI16L has a prolonged half-life, relative to wild-type FXa and does not cause excessive activation of coagulation. Once clotting mechanisms are activated to produce its cofactor FVa, FXaI16L is driven to the protease state and restores hemostasis in hemophilic animals upon vascular injury. Moreover, using human or murine analogs, we show that FXaI16L is more efficacious than FVIIa which is used to treat bleeding in hemophilia inhibitor patients3. Because of its underlying mechanism of action, FXaI16L may provide an effective strategy to enhance blood clot formation and act as a rapid pan-hemostatic agent for the treatment of bleeding conditions.


Blood | 2014

The endothelial protein C receptor enhances hemostasis of FVIIa administration in hemophilic mice in vivo

Giulia Pavani; Lacramioara Ivanciu; Armida Faella; Oscar A. Marcos-Contreras; Paris Margaritis

Recombinant activated human factor VII (rhFVIIa) is an established hemostatic agent in hemophilia, but its mechanism of action remains unclear. Although tissue factor (TF) is its natural receptor, rhFVIIa also interacts with the endothelial protein C receptor (EPCR) through its γ-carboxyglutamic acid (Gla) domain, with unknown hemostatic consequences in vivo. Here, we study whether EPCR facilitates rhFVIIa hemostasis in hemophilia using a mouse model system. Mouse activated FVII (mFVIIa) is functionally homologous to rhFVIIa, but binds poorly to mouse EPCR (mEPCR). We modified mFVIIa to gain mEPCR binding using 3 amino acid changes in its Gla domain (L4F/L8M/W9R). The resulting molecule mFVIIa-FMR specifically bound mEPCR in vitro and in vivo and was identical to mFVIIa with respect to TF affinity and procoagulant functions. In macrovascular injury models, hemophilic mice administered mFVIIa-FMR exhibited superior hemostatic activity compared with mFVIIa. This was abolished by blocking mEPCR and was absent in ex vivo whole blood coagulation assays, implicating a specific mFVIIa-FMR and endothelial mEPCR interaction. Because mFVIIa-FMR models the TF-dependent and EPCR binding properties of rhFVIIa, our data unmask a novel contribution of EPCR on the action of rhFVIIa administration in hemophilia, prompting the rational design of improved and safer rhFVIIa therapeutics.


Blood | 2011

Catalytic domain modification and viral gene delivery of activated factor VII confers hemostasis at reduced expression levels and vector doses in vivo

Paris Margaritis; Elise Roy; Armida Faella; Harre D. Downey; Lacramioara Ivanciu; Giulia Pavani; Shangzhen Zhou; Ralph M. Bunte; Katherine A. High

Catalytic domain variants of activated factor VII (FVIIa) with enhanced hemostatic properties are highly attractive for the treatment of bleeding disorders via gene-based therapy. To explore this in a hemophilic mouse model, we characterized 2 variants of murine activated FVII (mFVIIa-VEAY and mFVIIa-DVQ) with modified catalytic domains, based on recombinant human FVIIa (rhFVIIa) variants. Using purified recombinant proteins, we showed that murine FVIIa (mFVIIa) and variants had comparable binding to human and murine tissue factor (TF) and exhibited similar extrinsic coagulant activity. In vitro in the absence of TF, the variants showed a 6- to 17-fold enhanced proteolytic and coagulant activity relative to mFVIIa, but increased inactivation by antithrombin. Gene delivery of mFVIIa-VEAY resulted in long-term, effective hemostasis at 5-fold lower expression levels relative to mFVIIa in hemophilia A mice or in hemophilia B mice with inhibitors to factor IX. However, expression of mFVIIa-VEAY at 14-fold higher than therapeutic levels resulted in a progressive mortality to 70% within 6 weeks after gene delivery. These results are the first demonstration of the hemostatic efficacy of continuous expression, in the presence or absence of inhibitors, of a high-activity gene-based FVIIa variant in an animal model of hemophilia.


Blood | 2016

Sustained correction of FVII deficiency in dogs using AAV-mediated expression of zymogen FVII.

Oscar A. Marcos-Contreras; Shannon M. Smith; Dwight A. Bellinger; Robin A. Raymer; Elizabeth P. Merricks; Armida Faella; Giulia Pavani; Shangzhen Zhou; Timothy C. Nichols; Katherine A. High; Paris Margaritis

Factor VII (FVII) deficiency is a rare autosomal recessive bleeding disorder treated by infusion of fresh-frozen plasma, plasma-derived FVII concentrates and low-dose recombinant activated FVII. Clinical data suggest that a mild elevation of plasma FVII levels (>10% normal) results in improved hemostasis. Research dogs with a G96E missense FVII mutation (FVII-G96E) have <1% FVII activity. By western blot, we show that they have undetectable plasmatic antigen, thus representing the most prevalent type of human FVII deficiency (low antigen/activity). In these dogs, we determine the feasibility of a gene therapy approach using liver-directed, adeno-associated viral (AAV) serotype 8 vector delivery of a canine FVII (cFVII) zymogen transgene. FVII-G96E dogs received escalating AAV doses (2E11 to 4.95E13 vector genomes [vg] per kg). Clinically therapeutic expression (15% normal) was attained with as low as 6E11 vg/kg of AAV and has been stable for >1 year (ongoing) without antibody formation to the cFVII transgene. Sustained and supraphysiological expression of 770% normal was observed using 4.95E13 vg/kg of AAV (2.6 years, ongoing). No evidence of pathological activation of coagulation or detrimental animal physiology was observed as platelet counts, d-dimer, fibrinogen levels, and serum chemistries remained normal in all dogs (cumulative 6.4 years). We observed a transient and noninhibitory immunoglobulin G class 2 response against cFVII only in the dog receiving the highest AAV dose. In conclusion, in the only large-animal model representing the majority of FVII mutation types, our data are first to demonstrate the feasibility, safety, and long-term duration of AAV-mediated correction of FVII deficiency.


Journal of Thrombosis and Haemostasis | 2017

One amino acid in mouse factor VIIa defines its endothelial protein C receptor binding and modulates its EPCR‐dependent hemostatic activity in vivo

Giulia Pavani; S. M. Zintner; Lacramioara Ivanciu; J. C. Small; Katherine A. Stafford; J. H. Szeto; Paris Margaritis

Essentials The lack of factor (F) VIIa‐endothelial protein C receptor (EPCR) binding in mice is unresolved. A single substitution of Leu4 to Phe in mouse FVIIa (mFVIIa) enables its interaction with EPCR. mFVIIa with a Phe4 shows EPCR binding‐dependent enhanced hemostatic function in vivo vs. mFVIIa. Defining the FVIIa‐EPCR interaction in mice allows for further investigating its biology in vivo.


Molecular Therapy | 2018

Optimization of CRISPR/Cas9 Delivery to Human Hematopoietic Stem/Progenitor Cells for Therapeutic Genomic Rearrangements

Annalisa Lattanzi; Vasco Meneghini; Giulia Pavani; Fatima Amor; Sophie Ramadier; Tristan Felix; Chiara Antoniani; Cecile Masson; Olivier Alibeu; Ciaran Lee; Matthew H. Porteus; Gang Bao; Mario Amendola; Fulvio Mavilio; Annarita Miccio

Editing the β-globin locus in hematopoietic stem cells is an alternative therapeutic approach for gene therapy of β-thalassemia and sickle cell disease. Using the CRISPR/Cas9 system, we genetically modified human hematopoietic stem and progenitor cells (HSPCs) to mimic the large rearrangements in the β-globin locus associated with hereditary persistence of fetal hemoglobin (HPFH), a condition that mitigates the clinical phenotype of patients with β-hemoglobinopathies. We optimized and compared the efficiency of plasmid-, lentiviral vector (LV)-, RNA-, and ribonucleoprotein complex (RNP)-based methods to deliver the CRISPR/Cas9 system into HSPCs. Plasmid delivery of Cas9 and gRNA pairs targeting two HPFH-like regions led to high frequency of genomic rearrangements and HbF reactivation in erythroblasts derived from sorted, Cas9+ HSPCs but was associated with significant cell toxicity. RNA-mediated delivery of CRISPR/Cas9 was similarly toxic but much less efficient in editing the β-globin locus. Transduction of HSPCs by LVs expressing Cas9 and gRNA pairs was robust and minimally toxic but resulted in poor genome-editing efficiency. Ribonucleoprotein (RNP)-based delivery of CRISPR/Cas9 exhibited a good balance between cytotoxicity and efficiency of genomic rearrangements as compared to the other delivery systems and resulted in HbF upregulation in erythroblasts derived from unselected edited HSPCs.


Blood | 2012

A Novel Strategy to Circumvent Pre-Existing Humoral Immunity to AAV

Federico Mingozzi; Xavier M. Anguela; Giulia Pavani; Yifeng Chen; Robert J. Davidson; Daniel J. Hui; Christian Hinderer; Armida Faella; Crolann Howard; Alex Tai; Gregory M. Podsakoff; Margaret V. Ragni; Shangzhen Zhou; Etiena Basner-Tschakarjan; J. Fraser Wright; Katherine A. High


Blood | 2016

Induction of Fetal Hemoglobin Synthesis By Crispr/Cas9-Mediated Disruption of the β-Globin Locus Architecture

Chiara Antoniani; Vasco Meneghini; Annalisa Lattanzi; Giulia Pavani; Tristan Felix; Fatima Amor; Oriana Romano; Elisa Magrin; Leslie Weber; Thomas Cradick; Ante Sven Lundberg; Matthew H. Porteus; Mario Amendola; Marina Cavazzana; Fulvio Mavilio; Annarita Miccio


Blood | 2015

A Single Amino Acid in the Gla Domain Mouse FVIIa Allows Its Binding to the Endothelial Protein C Receptor and Enhances Its Coagulant Activity In Vivo

Giulia Pavani; Lacramioara Ivanciu; Katherine A. Stafford; Paris Margaritis

Collaboration


Dive into the Giulia Pavani's collaboration.

Top Co-Authors

Avatar

Paris Margaritis

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Lacramioara Ivanciu

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Armida Faella

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Katherine A. High

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

Katherine A. Stafford

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Alex Tai

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel J. Hui

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Etiena Basner-Tschakarjan

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge