Hemophilia B Gene Therapy Modelled in Null and Missense Mutant Mice: Kinetics of Transgenic Factor IX Expression Influences Tolerance to a Greater Extent Than Dose or Serotype of Adeno-Associated Virus (AAV) Vector.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5526-5526
Author(s):  
Paul E. Monahan ◽  
Tai-Ping Zhang ◽  
Da-Yun Jin ◽  
Tong Gui ◽  
Darrel W. Stafford

Abstract A major concern regarding the safety of gene therapy for protein deficiencies, including hemophilia, is the possibility of immune responses against the therapeutic transgenic protein. We have previously reported the use of hemophilic mice expressing defective missense human factor IX (hFIX) protein (Cross-reactive material positive, CRM +) or having a complete deletion of FIX product (CRM-) to examine the influence of the underlying mutation upon the risk of inhibitor development following intramuscular FIX gene therapy. When inhibitor antibodies have developed following adeno-associated virus (AAV) virus FIX gene therapy, the vector dose per injection site has been implicated as an important influence. To examine this, we treated CRM + missense R333Q-hFIX strain hemophilic mice with a single IM injection of AAV serotype2.hFIX. Doses of 1 x 1011 and 8 x 1011 vector genomes/animal resulted in increases of 50 ng/ml and 100 ng/ml hFIX, respectively, without inhibitor development. Identical vector doses given to the FIXKO CRM- strain resulted in inhibitors in all animals, predominantly IgG1 subclass, and zero circulating FIX. Using identical gene sequences and injection parameters, 1 x 1011 vg/animal of an AAV1 serotype hFIX vector was given to R333Q-hFIX mice and FIXKO mice. This resulted in physiologic hFIX levels in all animals at two weeks (1st timepoint examined), peaking at 3.9 μg/ml in FIXKO mice and 21.8 μg/ml in R333Q-hFIX mice. No mice developed inhibitors, despite the development of non-inhibitory IgG1 and IgG2 anti-factor IX. Despite subsequent challenge with repeated IV hFIX protein injections and with 1 x 1011 AAV2.hFIX, inhibitor antibodies could not be elicited in these animals. To examine the role of kinetics of trangene expression upon inhibitor development, stepwise decreases in the AAV1 vector dose were used, to try to reproduce the level of expression achieved in AAV2-treated animals that developed inhibitors [see Table]. At doses of 1 x 109 and 1 x 1010 AAV1hFIX, onset of expression was slow in R333Q-hFIX, averaging only10 ng/ml and 60 ng/ml above background at 2 weeks, and remaining below 300 ng/ml over months. Although lowering the vector dose of AAV1 reproduced the pattern of slow, low level transgene expression seen with the higher doses of AAV2, no inhibitors ever developed in the mice with CRM+ background. Nevertheless, FIXKO CRM- mice failed to achieve tolerance after the lower AAV1 doses. Neither the AAV1 serotype nor the vector particle number independently determined the inhibitor trigger. The results suggest the influence of the kinetics of onset and level of transgenic protein achieved are of primary importance in establishing tolerance in this application. Factor IX Expression and Inhibitor Formation Relative to Underlying FIX Mutation Mouse strain/Dose vector (vg/animal) FIX incr: 2 weeks (μg/ml) FIX incr: Peak (μg/ml) Bethesda Inhibitor (BIU range) R333Q, AAV2 1x10e11 0.01 0.05 0/5 mice (0 BIU) FIXKO, AAV2 1x10e11 0 0 5/5 mice (3–34 BIU) R333Q, AAV1 1x10e11 15.8 21.8 0/5 mice (0 BIU) R333Q, AAV1 1x10e10 0.06 0.25 0/4 mice (0 BIU) R333Q, AAV1 1x10e9 0.02 0.12 0/4 mice (0 BIU) FIXKO, AAV1 1x10e11 2.5 3.9 0/5 mice (0 BIU) FIXKO, AAV1 1x10e10 0.04 0.4 4/5 mice (2–16 BIU) FIXKO, AAV1 1x10e9 0 0 3/4 mice (2–6 BIU)

2000 ◽  
Vol 74 (8) ◽  
pp. 3555-3565 ◽  
Author(s):  
Ajay K. Malik ◽  
Paul E. Monahan ◽  
David L. Allen ◽  
Bing-Guan Chen ◽  
R. Jude Samulski ◽  
...  

ABSTRACT Recombinant adeno-associated virus (rAAV) vectors have been shown to be useful for efficient gene delivery to a variety of dividing and nondividing cells. Mechanisms responsible for the long-term, persistent expression of the rAAV transgene are not well understood. In this study we investigated the kinetics of rAAV-mediated human factor IX (hFIX) gene transfer into human primary myoblasts and myotubes. Transduction of both myoblasts and myotubes occured with a similar and high efficiency. After 3 to 4 weeks of transduction, rAAV with a cytomegalovirus (CMV) promoter showed 10- to 15-fold higher expression than that with a muscle-specific creatine kinase enhancer linked to β-actin promoter. Factor IX expression from transduced myoblasts as well as myotubes reached levels as high as approximately 2 μg of hFIX/106 cells/day. Southern blot analyses of high-molecular-weight (HMW) cellular genomic and Hirt DNAs isolated from rAAV/CMVhFIXm1-transduced cells showed that the conversion of single-stranded vector genomes to double-stranded DNA forms, but not the level of the integrated forms in HMW DNA, correlated with increasing expression of the transgene. Together, these results indicate that rAAV can transduce both proliferating and terminally differentiated muscle cells at about the same efficiency, that expression of transgenes increases linearly over their lifetime with no initial lag phase, and that increasing expression correlates with the appearance of double-stranded episomal rAAV genomes. Evidence showing that the rAAV virions can copackage hFIX, presumably nonspecifically, was also obtained.


Blood ◽  
2006 ◽  
Vol 107 (7) ◽  
pp. 2653-2661 ◽  
Author(s):  
Amit C. Nathwani ◽  
John T. Gray ◽  
Catherine Y. C. Ng ◽  
Junfang Zhou ◽  
Yunyu Spence ◽  
...  

AbstractTransduction with recombinant adeno-associated virus (AAV) vectors is limited by the need to convert its single-stranded (ss) genome to transcriptionally active double-stranded (ds) forms. For AAV-mediated hemophilia B (HB) gene therapy, we have overcome this obstacle by constructing a liver-restricted mini–human factor IX (hFIX) expression cassette that can be packaged as complementary dimers within individual AAV particles. Molecular analysis of murine liver transduced with these self-complementary (sc) vectors demonstrated rapid formation of active ds-linear genomes that persisted stably as concatamers or monomeric circles. This unique property resulted in a 20-fold improvement in hFIX expression in mice over comparable ssAAV vectors. Administration of only 1 × 1010 scAAV particles led to expression of hFIX at supraphysiologic levels (8I U/mL) and correction of the bleeding diathesis in FIX knock-out mice. Of importance, therapeutic levels of hFIX (3%-30% of normal) were achieved in nonhuman primates using a significantly lower dose of scAAV than required with ssAAV. Furthermore, AAV5-pseudotyped scAAV vectors mediated successful transduction in macaques with pre-existing immunity to AAV8. Hence, this novel vector represents an important advance for hemophilia B gene therapy.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 13-13
Author(s):  
Annette von Drygalski ◽  
Adam Giermasz ◽  
Giancarlo Castaman ◽  
Nigel S. Key ◽  
Susan U. Lattimore ◽  
...  

Background: Gene therapy for hemophilia offers the possibility to ameliorate disease severity to a mild or functionally curative state through a single administration. Etranacogene dezaparvovec (AMT-061) is an investigational gene therapy for hemophilia B comprising an adeno associated virus serotype 5 (AAV5) vector containing a codon-optimized Padua variant human factor IX (FIX) gene with liver specific promoter. Aims: We have previously shown a single dose of etranacogene dezaparvovec to provide sustained FIX activity into the mild-to normal range for up to 52 weeks post-dose in participants with severe or moderate-severe hemophilia B. This time, 2 years of follow-up data will be presented for the first time. Methods: A Phase 2b, open-label, single-dose, single-arm, multi-center trial (NCT03489291) in adult hemophilia B subjects. Interestingly, participants were not excluded based on neutralizing antibodies to AAV5. All subjects received a single intravenous dose of etranacogene dezaparvovec (2x1013 gc/kg) and will be followed for 5-years. The primary endpoint was FIX activity at Week 6. Secondary endpoints include e-diary recordings of bleeds and FIX concentrate use, laboratory parameters, joint health, patient reported outcomes, and adverse events (AEs). Results: All participants had FIX ≤1% (severe or moderately-severe FIX deficiency), required routine FIX prophylaxis, and had neutralizing activity to AAV5 at baseline. Following AMT-061 treatment, FIX activity increased rapidly to a mean of 31% at Week 6. At Week 52, mean FIX activity increased further to 41% with FIX activity levels of 50%, 31% and 41% in participants 1-3 respectively. There was no relationship between the presence of anti-AAV5 NAbs and response to etranacogene dezaparvovec. As of 52 weeks, there were no bleeds post-treatment and no requirement for FIX replacement aside from protocol-specified use for perioperative management in participant 3. There were no clinically significant elevations in liver enzymes and no participants required steroids related to the treatment. One participant experienced 2 mild AEs possibly related to treatment shortly after dosing (self-limiting headache and slightly elevated CRP). One patient had hip surgery due to worsening of pre-existing avascular necrosis deemed unrelated by investigator to etranacogene dezaparvovec and received FIX per protocol according to standard clinical practice. No participant developed inhibitors to FIX. Updated results to 2 years of follow-up will be presented with the main focus on FIX activity, FIX replacement therapy use and reported bleeds. Conclusions: Patients with AAV5 NAbs were included in the Phase 2b etranacogene dezaparvovec trial and have shown sustained FIX activity into the mild-to normal range. All participants were able to discontinue routine prophylaxis, and there have been no bleeds post-treatment with etranacogene dezaparvovec. Disclosures Giermasz: BioMarin: Consultancy, Research Funding, Speakers Bureau; Genentech/Roche: Consultancy, Research Funding, Speakers Bureau; uniQure: Consultancy, Research Funding; Sangamo Therapeutics: Research Funding; Bioverativ/Sanofi: Consultancy, Research Funding, Speakers Bureau. Castaman:Alexion: Honoraria; Roche: Consultancy, Honoraria, Speakers Bureau; CSL Behring: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Ablynx: Honoraria; Baxalta/Shire: Honoraria; Bayer: Honoraria; Uniqure: Honoraria, Membership on an entity's Board of Directors or advisory committees; Kedrion: Speakers Bureau; Werfen: Speakers Bureau; Sobi: Honoraria, Research Funding, Speakers Bureau; Novo Nordisk: Honoraria, Speakers Bureau. Key:Novo Nordisk: Other: Chair of Grants Committee; Takeda: Research Funding; Grifols: Research Funding; Uniqure: Consultancy. Miesbach:Bayer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; BioMarin Pharmaceutical Inc: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; UniQure: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Recht:Spark: Research Funding; Novo Nordisk: Consultancy, Other: personal fees, Research Funding; uniQure: Consultancy, Other: personal fees, Research Funding; Takeda: Consultancy, Other: personal fees, Research Funding; BioMarin: Research Funding; Pfizer: Consultancy, Other: personal fees; Genentech: Consultancy, Other: personal fees, Research Funding; CSL Behring: Consultancy, Other: personal fees. Gomez:Global Blood Therapeutics: Speakers Bureau. Gut:uniQure: Current Employment. Pipe:Siemens: Research Funding; Medical and Scientific Advisory Council to the National Hemophilia Foundation; Medical Advisory Board to World Federation of Hemophilia: Membership on an entity's Board of Directors or advisory committees; Apcintex, Bayer, BioMarin, Catalyst Biosciences, CSL Behring, HEMA Biologics, Freeline, Novo Nordisk, Pfizer, F. Hoffmann-La Roche Ltd/Genentech, Inc., Sangamo Therapeutics, Sanofi, Takeda, Spark Therapeutics, uniQure: Consultancy. OffLabel Disclosure: Etranacogene dezaparvovec (AMT-061) is an investigational gene therapy for hemophilia B comprising an adeno associated virus serotype 5 (AAV5) vector containing a codon-optimized Padua variant human factor IX (FIX) gene with liver specific promoter.


2000 ◽  
Vol 74 (4) ◽  
pp. 1761-1766 ◽  
Author(s):  
Marina Moskalenko ◽  
Lili Chen ◽  
Melinda van Roey ◽  
Brian A. Donahue ◽  
Richard O. Snyder ◽  
...  

ABSTRACT Recombinant adeno-associated virus type 2 (AAV) is a common vector used in human gene therapy protocols. We characterized the humoral immune response to AAV and observed that 80% of normal human subjects have anti-AAV antibodies and that 18% have neutralizing antibodies. To analyze the effect of neutralizing antibodies on AAV readministration, we attempted to deliver recombinant AAV expressing human factor IX (AAV-hFIX) intraportally into the livers of mice which had been preexposed to AAV and shown to harbor a neutralizing antibody response. While all naive control mice expressed hFIX following administration of AAV-hFIX, none of the mice with preexisting immunity expressed hFIX, even after transient immunosuppression at the time of the second administration with anti-CD4 or anti-CD40L antibodies. This suggests that preexisting immunity to AAV, as measured by a neutralizing antibody response, may limit AAV-mediated gene delivery. Using human sera in an enzyme-linked immunosorbent assay for AAV and a capsid peptide scan library to block antibody binding, we mapped seven regions of the AAV capsid containing immunogenic epitopes. Using pools of these peptides to inhibit the binding of neutralizing antibodies, we have identified a subset of six peptides which potentially reconstitute a single neutralizing epitope. This information may allow the design of reverse genetic approaches to circumvent the preexisting immunity that can be encountered in some individuals.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3181-3181
Author(s):  
John T. Gray ◽  
Andrew M. Davidoff ◽  
Amit C. Nathwani

Abstract Current approaches for gene therapy of hemophilia B (HB) are focused on liver targeted delivery of recombinant adeno-associated virus vectors (rAAV) encoding human Factor IX (hFIX). A major rate limiting step to efficient transduction of the liver is the need to convert the single stranded rAAV genome to a double stranded transcriptionally active form by the host cell. Recently described self-complementary AAV (scAAV) vectors, which can package AAV transgenes as DNA dimers when they are half the size of the wild type genome, bypass the need for second strand synthesis. Their substantially smaller packaging capacity has, however, limited their use for HB gene therapy. We have overcome this obstacle by designing a mini-human FIX expression cassette that is efficiently packaged as self-complementary DNA dimers in rAAV vectors. The key aspects of this novel expression cassette include a truncated regulatory element (LP1) consisting of the critical domains of human apolipoprotein E/C-I gene locus control region and the α1-antitrypsin (hAAT) promoter, and very small intron and polyadenylation elements from SV40. The hFIX 3′untranslated region has been deleted and the coding sequence was re-constructed (hFIXco) using a subset of codons most frequently found in highly expressed eukaryotic genes. The resulting expression cassette, which spanned 2.15 kbp, was cloned into an AAV-2 vector in which the right terminal resolution site had been deleted. Encapsidation with AAV-8 capsid proteins produced the scAAV-2/8 LP1-hFIXco vector in yields comparable to single stranded rAAV. Hirt analysis indicated that all vector genomes in murine liver after tail vein administration existed in a double stranded conformation. Stable plasma hFIX levels at 50% of physiologic (2867 ± 501 ng/ml) were achieved after tail vein administration of 2 x 109 scAAV-2/8 LP1-hFIXco vector/mouse. This is >500 fold higher than levels achieved with standard single stranded rAAV-2 vectors and >10 fold higher than levels achieved with a comparable single stranded vector pseudotyped with AAV-8 capsid proteins, confirming the superiority of scAAV vectors. Further increases in scAAV-2/8 LP1-hFIXco vector dose to 1 x 1011 particles/mouse resulted in a proportionately linear increase in stable plasma hFIX levels to over 100 μg/ml without any adverse toxicity. Importantly, transgene expression mediated by the LP1 promoter/enhancer appears to be strictly restricted to the liver. Evaluation of the efficacy and safety of scAAV-2/8 LP1-hFIXco in rhesus macaques is ongoing. However, based on the murine studies, our novel self complementary hFIX expressing vector offers a unique opportunity to mediate therapeutic gene transfer in humans without the need for higher vector doses. This has important safety implications and will therefore substantially improve the prospects of hemophilia gene therapy.


2005 ◽  
Vol 79 (24) ◽  
pp. 15238-15245 ◽  
Author(s):  
Alejandra E. Arbetman ◽  
Michael Lochrie ◽  
Shangzhen Zhou ◽  
Jennifer Wellman ◽  
Ciaran Scallan ◽  
...  

ABSTRACT Preexisting humoral immunity to adeno-associated virus (AAV) vectors may limit their clinical utility in gene delivery. We describe a novel caprine AAV (AAV-Go.1) capsid with unique biological properties. AAV-Go.1 capsid was cloned from goat-derived adenovirus preparations. Surprisingly, AAV-Go.1 capsid was 94% identical to the human AAV-5, with differences predicted to be largely on the surface and on or under the spike-like protrusions. In an in vitro neutralization assay using human immunoglobulin G (IgG) (intravenous immune globulin [IVIG]), AAV-Go.1 had higher resistance than AAV-5 (100-fold) and resistance similar to that of AAV-4 or AAV-8. In an in vivo model, SCID mice were pretreated with IVIG to generate normal human IgG plasma levels prior to the administration of AAV human factor IX vectors. Protein expression after intramuscular administration of AAV-Go.1 was unaffected in IVIG-pretreated mice, while it was reduced 5- and 10-fold after administration of AAV-1 and AAV-8, respectively. In contrast, protein expression after intravenous administration of AAV-Go.1 was reduced 7.1-fold, similar to the 3.8-fold reduction observed after AAV-8administration in IVIG-pretreated mice, and protein expression was essentially extinguished after AAV-2 administration in mice pretreated with much less IVIG (15-fold). AAV-Go.1 vectors also demonstrated a marked tropism for lung when administered intravenously in SCID mice. The pulmonary tropism and high neutralization resistance to human preexisting antibodies suggest novel therapeutic uses for AAV-Go.1 vectors, including targeting diseases such as cystic fibrosis. Nonprimate sources of AAVs may be useful to identify additional capsids with distinct tropisms and high resistance to neutralization by human preexisting antibodies.


2017 ◽  
Vol 1 (26) ◽  
pp. 2591-2599 ◽  
Author(s):  
Lindsey A. George

Abstract Concurrent with the development of recombinant factor replacement products, the characterization of the F9 and F8 genes over 3 decades ago allowed for the development of recombinant factor products and made the hemophilias a target disease for gene transfer. The progress of hemophilia gene therapy has been announced in 3 American Society of Hematology scientific plenary sessions, including the first “cure” in a large animal model of hemophilia B in 1998, first in human sustained vector-derived factor IX activity in 2011, and our clinical trial results reporting sustained vector-derived factor IX activity well into the mild or normal range in 2016. This progression to clinically meaningful success combined with numerous ongoing recombinant adeno-associated virus (rAAV)–mediated hemophilia gene transfer clinical trials suggest that the goal of gene therapy to alter the paradigm of hemophilia care may soon be realized. Although several novel therapeutics have recently emerged for hemophilia, gene therapy is unique in its potential for a one-time disease-altering, or even curative, treatment. This review will focus on the prior progress and current clinical trial investigation of rAAV-mediated gene transfer for hemophilia A and B.


Blood ◽  
1996 ◽  
Vol 87 (12) ◽  
pp. 5095-5103 ◽  
Author(s):  
G Hortelano ◽  
A Al-Hendy ◽  
FA Ofosu ◽  
PL Chang

A potentially cost-effective strategy for gene therapy of hemophilia B is to create universal factor IX-secreting cell lines suitable for implantation into different patients. To avoid graft rejection, the implanted cells are enclosed in alginate-polylysine-alginate microcapsules that are permeable to factor IX diffusion, but impermeable to the hosts' immune mediators. This nonautologous approach was assessed by implanting encapsulated mouse myoblasts secreting human factor IX into allogeneic mice. Human factor IX was detected in the mouse plasma for up to 14 days maximally at approximately 4 ng/mL. Antibodies to human factor IX were detected after 3 weeks at escalating levels, which were sustained throughout the entire experiment (213 days). The antibodies accelerated the clearance of human factor IX from the circulation of the implanted mice and inhibited the detection of human factor IX in the mice plasma in vitro. The encapsulated myoblasts retrieved periodically from the implanted mice up to 213 days postimplantation were viable and continued to secrete human factor IX ex vivo at undiminished rates, hence suggesting continued factor IX gene expression in vivo. Thus, this allogeneic gene therapy strategy represents a potentially feasible alternative to autologous approaches for the treatment of hemophilia B.


Blood ◽  
2002 ◽  
Vol 99 (8) ◽  
pp. 2670-2676 ◽  
Author(s):  
Jane D. Mount ◽  
Roland W. Herzog ◽  
D. Michael Tillson ◽  
Susan A. Goodman ◽  
Nancy Robinson ◽  
...  

Abstract Hemophilia B is an X-linked coagulopathy caused by absence of functional coagulation factor IX (FIX). Using adeno-associated virus (AAV)–mediated, liver-directed gene therapy, we achieved long-term (> 17 months) substantial correction of canine hemophilia B in 3 of 4 animals, including 2 dogs with an FIX null mutation. This was accomplished with a comparatively low dose of 1 × 1012 vector genomes/kg. Canine FIX (cFIX) levels rose to 5% to 12% of normal, high enough to result in nearly complete phenotypic correction of the disease. Activated clotting times and whole blood clotting times were normalized, activated partial thromboplastin times were substantially reduced, and anti-cFIX was not detected. The fourth animal, also a null mutation dog, showed transient expression (4 weeks), but subsequently developed neutralizing anti-cFIX (inhibitor). Previous work in the canine null mutation model has invariably resulted in inhibitor formation following treatment by either gene or protein replacement therapies. This study demonstrates that hepatic AAV gene transfer can result in sustained therapeutic expression in a large animal model characterized by increased risk of a neutralizing anti-FIX response.


Sign in / Sign up

Export Citation Format

Share Document