Summary
Factor IX deficiency (hemophilia B) is less common than factor VIII deficiency (hemophilia A) and innovations in therapy for hemophilia B have generally lagged behind those for hemophilia A. Recently the first sustained correction of the hemophilia bleeding phenotype by clotting factor gene therapy has been described using recombinant adeno-associated virus (AAV) to deliver factor IX. Despite this success, many individuals with hemophilia B, including children, men with active hepatitis, and individuals who have pre-existing natural immunity to AAV are not eligible for the current iteration of hemophilia B gene therapy. In addition, recent advances in recombinant factor IX protein engineering have led some hemophilia treaters to reconsider the urgency of genetic cure. Current clinical and preclinical approaches to advancing AAV-based and alternative approaches to factor IX gene therapy are considered in the context of current demographics and treatment of the hemophilia B population.
Keywords: Factor IX, hemophilia B, dependovirus, genetic therapy, clinical trial
Factor IX gene therapy and the landscape of treatment for hemophilia B
Hemophilia B, resulting from deficient activity of clotting factor IX, accounts for 20% of hemophilia, while 80% of hemophilia results from deficient factor VIII activity (hemophilia A). Throughout most of the last half century, hemophilia B therapies have developed more slowly and often have been less available than treatments for hemophilia A. The description in 1964 of cryoprecipitate as a plasma preparation rich in factor VIII was a transformative moment in the care of hemophilia A, but large volume plasma infusion remained the only source for replacement factor IX. Widespread availability of plasma-derived factor IX concentrates was delayed almost a decade after factor VIII concentrates became available in the 1970s. In response to the tragic contamination of plasma-derived factor concentrates with hepatitis and HIV, recombinant FVIII was generated and licensed by 1992; hemophilia B patients waited until 1997 in the U.S. (and much later in some countries) for the licensure of recombinant factor IX (rFIX), the last real innovation in hemophilia B care.
Then in a one month period between December, 2011–January, 2012, reports appeared that two goals had been achieved by new factor IX therapies: 1) the successful, persistent expression of factor IX by human gene therapy [1] and 2) the demonstration in a human clinical trial several-fold extension of factor IX circulating half-life via recombinant protein engineering [2]. Hemophilia A still awaits these successes. In the interim between January 2012 and now, the licensure of a second recombinant factor IX (biosimilar rFIX concentrate having the wild type FIX molecular sequence and structure) expands product choice for conventional hemophilia B replacement therapy. During the same period three different approaches to modify the wild type factor IX protein have yielded comparable, >3-fold prolongations in the factor IX circulating half-life in human trials: the recombinant expression of factor IX monomer protein genetically fused to the constant region of immunoglobulin(rFIXFc); the recombinant expression of factor IX monomer protein genetically fused to albumin; and the modification of recombinant factor IX via the addition of a 40k polyethylene glycol moiety to the N-glycan acceptor sites at FIXAsn157 or FIXAsn167 (as reviewed by Kaufman and Powell) [3]. One rFIX fusion protein now is licensed (rFIXFc) and a U.S. Food and Drug Administration Biologic License Application has been submitted for the second (rIX-FP). This review limits its scope to considering how insights gained from three pioneering factor IX gene therapy trials have informed the objectives and design of several ongoing and planned trials of hemophilia B gene therapy. Hemophilia gene therapy’s first clinical success is reviewed in the context of an evolving era of choice for men with severe hemophilia B, while also considering the unique characteristics of this population (see Table 1) that currently limit the broad clinical translation of liver-directed adeno-associated virus (AAV) vector and other gene delivery approaches.
Table 1.
Proportion of adult male hemophilia B population (%) | ||
---|---|---|
Severe Hemophilia B (FIX:C <1%) 1 | 35.7% | |
Factor IX Inhibitor (+) 2 | 2% | |
Factor IX Inhibitor (−) 2 | 98% | |
Proportion of Severe Hemophilia B Population (%) | ||
Adult men (Total) | Adult men <30 years old | |
Factor IX infused on regular prophylactic schedule? 3 | 24.7% | 37.0% |
Annualized Joint Hemorrhage Rate ≥ 10 | 38% | 30% |
Hepatitis B seropositive | 51.2% | 5.7% |
Hepatitis C seropositive 4 | 77% | 41.4% |
Chronically elevated liver transaminases? | 18.5% | 4.8% |
If Hepatitis C antibody (+):Ever received anti-viral therapy for hepatitis C? | ||
No | 76% | 74% |
Yes | 24% | 26% |
If yes, Sustained viral response to therapy? | 54% | 61% |
HIV/HCV co-infection | 20% | 1.3% |
N/A = Not available.
Sources used to compile the table include national data collected and verified through 2011 [54] [59] [60] [61]. Shaded characteristics are exclusionary criteria for all current Hemophilia B gene therapy clinical trials.
Current gene therapy trials for hemophilia B typically include individuals with ≤ 2% factor IX and clinical history of frequent hemorrhage. In addition to the 791 adult men with <1% activity in the U.S., an additional 165 men potentially meet inclusion criteria based on baseline factor IX activity 1–2%.
The U.S. Centers for Disease Control and Prevention Universal Data Collection captures current and new incident inhibitors but does not capture individuals with a history of inhibitor who have no current inhibitor titer. History of factor IX inhibitor is exclusionary for current gene therapy trials.
Categorized as prophylaxis if, during the year preceding the visit, factor infusions were used on a regular schedule to prevent any and all bleeding and this therapy was expected to continue indefinitely.
Data are not available on the proportion of HCV-exposed (seropositive) individuals who have active disease as demonstrated by PCR of HCV RNA; in addition to individuals having a sustained viral response to HCV therapy, at least 20% of HCV-infected individuals may spontaneously clear HCV.
The first success: Adeno-associated virus vectors for the expression of factor IX
The first and only successful approach for human gene transfer of a coagulation factor used an adeno-associated virus (AAV) as the vector to deliver a human factor IX cDNA to the liver, the natural site of factor IX production [1] [4]. The wild type (wt) AAV is a small single-stranded DNA virus that is non-pathogenic and replication defective. Most individuals experience an asymptomatic exposure to wt AAV during childhood and mount a humoral immune response, which often results in persistent titers of anti-AAV antibodies sufficient to neutralize subsequent infection by wt AAV and by recombinant AAV vectors [5] [6] [7]. All coding sequences of the 4680 nt wt AAV genome are removed during the generation of AAV gene therapy vectors, adding further to their safety. Following transduction of the target cell, the recombinant AAV (rAAV) therapeutic gene sequences are retained primarily as concatameric episomes and uncommonly integrate into the host genomic DNA.
Following the initial recognition that rAAV vectors transduce a variety of relatively end-differentiated, non-dividing cells and tissues, the earliest attempts to correct hemophilia B targeted muscle expression. The capacity for intramuscular (I.M.) injection of AAV.FIX vectors to direct human factor IX expression and improve coagulation was demonstrated in mouse and dog models of hemophilia B [8] [9] [10] [11]. A Phase I clinical trial of AAV.FIX delivered to skeletal muscle commenced, sponsored by Avigen, Inc. Eight subjects were treated with a single strand rAAV serotype 2 vector following a single-dose, dose-escalation design via multiple direct I.M. injections. There were no safety concerns but also no sustained circulating factor IX activity was achieved [12]. Biopsy of injected skeletal muscle at 2–10 months after administration revealed persistence of the therapeutic transgene and factor IX protein not only intracellularly but also extensively bound to extracellular matrix collagen IV surrounding myocytes[13] [14]. Skeletal muscle from one subject examined at 10 years after AAV gene delivery demonstrated local persistence of vector-derived FIX mRNA transcripts and FIX protein [15]. Recently, large animal studies have validated a strategy to achieve more efficient and uniform transduction of skeletal muscle by avoiding multiple local percutaneous I.M. injections and delivering rAAV.FIX to the entire vascular bed of limb muscles by using a peripheral transvenular approach [16].
AAV serotype 2 factor IX gene delivery to the natural site of factor IX expression in the liver was explored in mice and found to generate greater transgenic factor IX activity than equivalent vector doses delivered to the muscle; at least a part of the difference results from less efficient post-translational modification of factor IX in muscle when compared to the natural organ of synthesis of the protein. An unusual complication of severe hemophilia B, occurring in about 2–4% of patients treated with factor IX protein infusions, is the development of factor IX-specific IgG that neutralizes the procoagulant activity of factor IX (“factor IX inhibitors”). Not only is factor IX therapy rendered ineffective by this loss of “tolerance” of factor IX by the patient’s immune system (to which the factor IX appears to be a neoantigen) but potentially life-threatening anaphylactic reactions may be triggered by factor IX exposure. In the context of gene therapy, liver transduction in animals did not lead to loss of tolerance and factor IX inhibitor formation as readily as muscle gene delivery [10]. Multiple investigations have reproduced the observation that the tolerance of clotting factor following hepatic transduction by AAV does not result from anergy or immunologic ignorance of the potentially neoantigenic clotting factor, but results from the induction of tolerogenic regulatory T lymphocytes [11] [17] [18]. Convincing preclinical data for the liver-directed AAV approach was generated by showing that null mutation hemophilia B dogs having a high risk for factor IX inhibitor formation produced persistent circulating factor IX activity following hepatic delivery of AAV2.FIX, experienced decreased bleeding, and did not form factor IX inhibitors [12].
Avigen sponsored a human gene therapy trial delivering an AAV2.FIX construct with a liver specific promoter directly into the hepatic artery, reported by Manno [19]. At two lower doses no safety concerns or factor IX expression were observed, prompting escalation to the planned highest dose of 2 × 1012 Vector Genomes (VG)/kg. Circulating factor IX activity peaked at 11.8% at 2 weeks after vector infusion, however factor IX then declined starting around 4 weeks after the AAV2 infusion, coincident with an ~ 10 times normal elevation of the liver transaminases ALT and AST. Peripheral blood T cells from a subsequent subject who received AAV2.FIX and experienced a less profound transaminitis suggested the most likely etiology was a cell-mediated adaptive immune response directed toward the liver[19] [20]. Following successful FIX gene delivery to hepatocytes, the capsid of the vector is degraded intracellularly and capsid-derived peptides are presented on the surface of the transduced hepatocyte in the context of MHC class I molecules. At relatively higher exposures of AAV capsid, memory T cell recognition of AAV capsid peptides then may trigger a cytotoxic lymphocyte (CTL)-mediated elimination of AAV-transduced hepatocytes.
Several strategies were subsequently incorporated into AAV.FIX vectors to increase the efficiency of liver transduction, so that the vector particle dose and potential immune recognition of AAV could be minimized. The rhesus macaque is the natural host for AAV8 and the seroprevalence of AAV8-neutralizing antibodies (NAb) in humans is lower than AAV2 NAb [6] [7] [5]. AAV8 transduces liver more efficiently than AAV2 and with less off-target distribution in organs outside the liver, allowing infusion by peripheral vein rather than the invasive hepatic artery route [21]. A rate-limiting step in AAV transduction, which is the need to convert single strand AAV into a double-stranded configuration to serve as a template for transcription, was overcome by using self-complementary (sc) AAV vectors, increasing transduction efficiency [22] [23] [24]. Conservative nucleotide changes leading to codon optimization also yielded robust increases in factor IX expression [17] [24].
In 2011 St Jude Children’s Research Hospital and the University College London (UCL/SJCRH) investigators reported the first hemophilia trial to achieve persistent clotting factor expression [1] [4]. Following the single scAAV8.FIX codon-optimized vector administration all ten subjects in the trial decreased markedly their use of exogenous factor IX protein infusions. Steady state expression of factor IX activity for the six subjects treated at the highest dose (2 × 1012 VG/kg) maintained a mean of 5.1 ±1.7%, measured at least a year after vector administration. At this high dose, however, 4 of 6 subjects developed AAV capsid-specific T lymphocyte responses at 7 to 10 weeks after vector administration. The pro-inflammatory T effector cell responses were temporally associated with transient alanine aminotransferase (ALT) increases to 1.5–4 times the upper limit of normal in two subjects; two additional subjects demonstrated an increase above their individual baseline, although the ALT remained in the normal range. Corticosteroid therapy was initiated promptly in response to the transaminase changes and continued for 8–12 weeks. Despite a decline in factor IX expression in these four subjects some factor IX expression has been maintained in all subjects, which is an encouraging improvement over the outcomes in the ssAAV2 liver trial. The first subject treated has expressed 2% factor IX steadily for more than 4 years of follow up.
Ongoing and planned hemophilia B clinical trials using the adeno-associated virus vector
The UCL/SJCRH study investigators describe the hepatic inflammation seen in their high dose study subjects as a manageable toxicity. Nevertheless, the complication limits the ability to offer the therapy to adults with hemophilia who are active carriers of hepatitis or to increase the vector dose to achieve truly normal hemostasis (e.g. to achieve baseline factor activity of ≥15% that is expected to prevent all joint bleeding) [25]. Subsequent to the 2011 report, three AAV8.FIX trials have commenced with the goal of achieving greater factor IX expression without toxicity. The UCL/SJCRH clinical vector contained, in addition to the therapeutic scAAV8.FIXco, empty AAV8 capsids carrying no FIX gene [26]. These contaminating empty AAV capsids comprised 80% of the vector preparation and were a potential trigger for the dose-dependent CTL response directed against AAV. The AAV vector manufacturing process at the University of North Carolina at Chapel Hill Vector Core Facility incorporates empty capsid removal steps. Vector made at this facility containing less than 10% empty capsids is being used in a trial sponsored by Baxter Healthcare, testing a scAAV8.FIX vector designated BAX335 [27, 28]. Purification steps that remove empty capsids are now employed by the UCL/SJCRH group, resulting in a 90% “full” capsid vector stock [29]. Each of these two groups is currently enrolling subjects to test whether scAAV8 vector having minimal empty capsid contamination will permit dose escalation without stimulating cell-mediated immunity.
Investigators at the Children’s Hospital of Philadelphia (CHOP) proposed an alternative approach to the empty capsids, theorizing that the defective particles may have aided AAV transduction in the UCL/SJCRH trial. Mingozzi and colleagues suggested that empty capsids titrated into therapeutic AAV vector preparations could act as “decoys” in the eventuality that circulating AAV NAb were present, thereby allowing therapeutic virus to evade neutralization and transduce liver more efficiently. This strategy was to be incorporated into a single-stranded AAV8.FIX trial [30]. A clinical trial initiated by these investigators (sponsored by Spark Therapeutics) has yet to be reported [31].
Given that a vector dose-dependent immune response is the only toxicity of AAV.FIX observed to date in humans, and the fact that capsid epitopes from empty capsids that enter cells may contribute to the CTL response, the CHOP investigators next introduced further mutations at amino acids 585/588 of their decoy capsids to disrupt the AAV2 binding for heparin sulfate (HS), the receptor for AAV on hepatocytes [30]. The investigators showed in mice that these mutations prevented receptor-mediated hepatocyte entry by the decoy empty capsids, presumably eliminating the ability of the hepatocyte to present capsid-derived peptides and trigger a hepatocyte-targeted CTL response. The use of excess HS-non-binding (585/588 mut) decoy capsids has not been tested in human application. One concern is that Kern and colleagues have previously shown that, although hepatocyte entry is lost in mutant rAAV585 capsids, entry into cardiac muscle is maintained or increased, and so the potential exists that the CTL could be redirected from liver to myocardium [32].
An alternative approach to achieve higher factor IX activity without increasing vector exposure, now validated in preclinical work, is to employ a gain-of-function variant of factor IX. Structure-function studies of the catalytic domain of factor IX in the Stafford laboratory revealed that substitution of alanine for arginine at amino acid 338 in the catalytic domain increased the specific activity of factor IX [33], and Simioni and colleagues subsequently characterized a family that expressed a hyperactive factor IX as a result of a leucine substitution at this same amino acid (FIXR338L) [34]. Multiple groups have now demonstrated in hemophilia B animal models that the incorporation of FIXR338L (Factor IX Padua) into AAV vectors for transduction of liver [35] or muscle [16], or into lentiviral vectors for transduction of liver [36] [37] leads to the expression of factor IX protein having at least 6–8 times normal specific activity without an increased incidence of thrombosis. Crudele and colleagues recently reported that AAV8-mediated hepatic expression of FIXR338L was associated with the development of factor IX tolerance in FIX inhibitor-prone hemophilia B dogs, as well as eradication of the pre-existing factor IX inhibitor in one dog [17]. Preclinical data has been reported recently that supported the initiation in 2013 of a human trial of the scAAV8FIXR338L codon-optimized BAX335 vector (the vector introduced above), with trial results pending [35].
Spark Therapeutics and Dimension Therapeutics each have promised Phase 1 trials for hemophilia B using proprietary AAV capsids; additional details are not publicly available. Uniqure has initiated a European Phase 1/2 trial using the identical factor IX expression cassette (LP1-hFIXco) used in the UCL/SJCRH trial, packaged within an AAV serotype 5 capsid instead of AAV serotype 8 and delivered via peripheral vein. Interestingly, the LP1-hFIXco expression construct pseudotyped with AAV5 and with AAV8 has previously been compared in non-human primates by Nathwani and colleagues [21]. Although the studies demonstrated comparable liver transduction and factor IX expression using AAV5- or AAV8-pseudotyped vectors, the two serotypes were not interchangeable. Following peripheral vein administration, when compared to scAAV8-LP1-hFIXco, the expression was slower from AAV5-pseudotyped sc-LP1-hFIXco, the clearance of vector genomes from plasma more delayed, and the off-target transduction of spleen, kidney and testis were greater for the serotype 5-pseudotyped vector. The investigators’ conclusion was that scAAV8 capsid proteins are preferable for liver transduction via peripheral vein but that scAAV5-pseudotyped vectors should be effective for treating individuals with pre-existing immunity to AAV8 or AAV2 [21]. Should pre-existing AAV8 NAbs limit enrollment in clinical trials, the Uniqure AAV5-pseudotyped vector might prove advantageous. A recent study of prevalence of naturally-occurring antibodies against AAV serotypes conducted in a healthy European population documented neutralizing antibody seroprevalence of 59% for AAV2, 19% for AAV8 and 3% for AAV5 [7]. Higher seroprevalence of AAV5 NAbs has been reported in other populations [38] [39], and the lack of concensus regarding a standardized methodology for NAb measurement complicates comparison between studies. The only AAV NAb study of a similar size evaluated in a hemophilia population is a pediatric study showing that by age 5–6 years boys with hemophilia had NAb seroprevalence of 27% for AAV2, 13% for AAV8 and 15% for AAV5 [6]. Uniqure has presented in abstract form a safety and dose-finding study in non-human primates to justify the trial design [40]. Five subjects will be treated with AAV5-hFIX 5.0 × 1012 VG/kg, followed by an escalation to 2.0 × 1013 VG/kg to treat the final five subjects.
Sangamo BioSciences in collaboration with Shire AG has announced plans for a human trial of factor IX gene editing using AAV to deliver zinc finger nuclease (ZFN) technology. The planned trial is based on work from the research group of High, in collaboration with Sangamo, who used a hemophilia B mouse model, engineered to carry a human nonsense mutant Y155stop F9 minigene inserted in the ROSA26 locus (hF9mut mice), to examine ZFN gene correction in vivo. The delivery of AAV8 ZFN to target intron 1 of the F9 gene along with a second AAV8 donor template vector with arms of homology flanking F9 exons 2–8 to neonatal mice (i.e. mice undergoing rapid growth of the liver) as well as adult mice resulted in apparent ZFN-induced double strand breaks in host DNA and homology-directed repair of the F9 gene [41]. Co-delivery of AAV8 expressing the hF9-specific ZFN and AAV8 expressing the corrective partial F9 cDNA was able to direct sustained expression averaging 23% of normal human factor IX in the adult hF9mut mouse model.
Although gene editing is an exciting direction for the field, several caveats exist with the work described to date. Off-target cleavage by the ZFN vector remains a concern (for example, treatment of WT littermates lacking the ZFN target site nevertheless led to expression of 1% normal human factor IX) [42]. Rates of off-target cleavage differ between ZFN and more recently developed nuclease systems that systems that are finding application in hemophilia gene and cell therapy [43], such as transcriptional activator-like effector nucleases (TALENS) and the two-component CRISPR (clustered regularly interspaced short palindromic repeat)– Cas9 (CRISPR-associated nuclease 9). Off-target rates depend not only on the nuclease reagent but also how long and at what level the nuclease is expressed and how many likely off-target sites exist in the genome from the outset. The AAV8.ZFN F9-targeting dual vector strategy described above provided for constitutive, rather than transient, expression of the ZFN by AAV vector transduced cells. This creates the potential for generation of an immune response against the ZFN (as a foreign protein), as well as the possibility for lasting genomic instability due to long-term low-level ZFN-mediated genomic damage. Furthermore, although ZFN-mediated gene editing is expected to introduce permanent gene correction (when compared to an episomally located AAV-delivered factor IX transgene), the proposed approach did not circumvent the need for doses of AAV vector that have been immunogenic in the previous hemophilia trials; the equivalent of 2.4 × 1013 VG/kg of the combined AAV vectors was used to promote gene correction in the murine model (i.e. ten times greater vg/kg dose than the dose associated with transaminitis in the Avigen and UCL/SJCRH trials). For human clinical application, Sangamo has developed an alternative approach in which F9 gene insertion is targeted downstream of the highly active albumin promoter, potentially driving robust factor IX production while requiring fewer vector particles/integration events than were required in the mouse studies. Such an approach may not require a nuclease to promote site-specific recombination [44].
An unknown regarding the ultimate potential for AAV gene therapy to provide a cure for hemophilia B is how long transgene expression from this non-integrating vector will persist, particularly in tissues like the liver that are felt to undergo some degree of turnover (albeit at a slow rate). The F9 transgene integration status and the persistent, stable expression of AAV-delivered factor IX from the livers of the null-mutation dogs has been extensively characterized at 8 years of follow up and subsequent reports confirm phenotypic correction for >10 years [45] [11]. Likewise, ongoing hepatic factor IX expression for greater than 8 years continues to be followed in non-human primates [4]. Consistent with the large animal experience, the first subjects treated on the UCL/SJCRH trial continue to demonstrate stable factor IX expression for greater than four years and counting.
Preclinical Development: Lentivirus vectors for liver gene therapy
Lentiviruses (LVs) efficiently transduce the relatively quiescent hepatocytes along with multiple cells of the liver. These include antigen-presenting cells, and a major task for adapting lentiviruses for hemophilia gene therapy has been to avoid immune responses to the potentially neoantigenic factor IX and factor VIII. In regard to factor IX, restricting gene expression to the liver with the use of liver-specific promoters and opposing gene expression in APCs via the incorporation of hematopoietic-specific microRNA target sequence (miR142-3p) achieved factor IX expression in and secretion from the liver in both mouse and dog models of hemophilia B [46] [47]. Moreover, lentivirus-directed liver-restricted factor IX expression appears to actively promote tolerance induction via the induction of CD4+ CD25+ Foxp3+regulatory T cells (Tregs) [47]. Tolerance induction and eradication of pre-existing factor IX inhibitors has recently been demonstrated following liver-restricted LV.FIX transduction [18].
Given somewhat less robust factor IX expression from LV (when compared to several serotypes of AAV) much recent emphasis has been on LV development for the task of delivering the large F8 gene, which is very difficult to accommodate in tiny AAV vectors [47] [48]. Investigation of the concern that LV genomic integration could cause genotoxicity is the subject of active investigation, and appears to be considerably lower risk than has been observed with the use of gamma retroviral vectors [46]. Nevertheless, several groups have addressed the concern of potential genotoxic risk by engineering lentiviral vectors with inactivating mutations in the integrase to create integration-defective lentiviral vectors. Although the transgene expression is reduced by loss of integration, optimization of the vectors and incorporation of gain-of-function FIXR338L transgene has achieved disease correction in hemophilic mice, while maintaining the potential for factor IX tolerance induction by hepatic expression [46] [36] [37] [47].
Preclinical Development: Cell-based approaches
Lentivirus-mediated delivery of the F9 gene has also been employed in a cell-based approach that results in circulating platelet delivery of factor IX to maintain hemostasis [49]. Adapting an approach that has demonstrated phenotypic correction in large animal models of hemophilia A and Glanzmann Thrombasthenia [50], a LV construct expressing human factor IX under the control of the platelet-specific glycoprotein IIb gene (αIIb) promoter was used to transduce FIX−/− mouse hematopoietic stem cells (HSC) ex vivo. Recipient FIX−/− mice were conditioned for HSC transfer with either a myeloablative (1100 cGy) or a nonmyeloablative (660 cGy) dose of total body irradiation 24 hours prior to infusion of the LV-transduced HSCs. Although negligible factor IX activity was measurable in plasma, human factor IX expression was present in cells of megakaryocyte lineage, co-localized with Von Willebrand Factor, provided partial hemostatic protection during a bleeding challenge, induced tolerance to human factor IX in FIX−/− mice, and could be transferred via bone marrow transplant to secondary recipient FIX−/− mice. Moreover, the nonmyeloablative conditioning was adequate to achieve all of these outcomes. Interestingly, an LV strategy employing a platelet-specific factor VIII expression cassette has recently been demonstrated to transduce hematopoietic stem cells and lead to correction of the hemophilia A bleeding phenotype in mice following direct intraosseous LV delivery with no requirement for a bone marrow conditioning regimen [48]. The latter approach has not been explored for the correction of hemophilia B.
Severe hemophilia B: Goals and Options
It has been stated many times that hemophilia B is an ideal disease model for the investigation of gene therapy approaches [47] [1]. It is worthwhile to examine the target patient population of adult men with severe factor IX deficiency and consider whether gene delivery in its current application is the ideal therapeutic approach and what patient-related factors may determine whether and how gene therapy translates from a primarily academic effort into wider clinical application.
An unprecedented time of new treatment options for men with severe hemophilia B approaches. An examination of the self-reported outcomes of men with severe factor IX deficiency captured by the U.S. surveillance instrument (Table 1) reveals that a third of men report potentially damaging high rates of joint bleeding (adjusted joint bleeding rate ≥10/yr), while the adoption of regular factor IX prophylaxis is low. (Table 1) Adherence to prophylactic infusions is an unrelenting demand whether the goal is to reduce bleeding or prevent it altogether. Hemophilia patients and physicians have recently been presented with three studies taking different approaches to maintaining joint health. One study compared on-demand therapy with unmodified recombinant factor IX (rFIX, Benefix®, Pfizer) using either a conventional approach to factor IX prophylaxis (50 IU/kg) twice weekly or a higher dose once-weekly (100 IU/kg) regimen. There were no treatment-related safety concerns. The mean annualized bleeding rates (ABR) were 35.1, 2.6, and 4.6 bleeding events/year for the on-demand, the twice-weekly therapy and the once-weekly therapy. The rationale for conventional twice weekly therapy is to maintain measurable (>1% activity) trough factor IX concentrations throughout a week; the higher once-weekly dose would be expected to result in some portion of the week with unmeasurable circulating factor IX, Nevertheless, there was no statistical difference between the ABR for the two prophylaxis approaches (P=0.22) [51]. These study results and others justify investigation of the possibility that factor IX bound in extracirculatory sites may contribute to hemostasis [52] [53]. Understanding the amount and the physiologic relevance, if any, of the factor IX distributed in extracirculatory sites could be relevant for understanding the total endogenous factor IX expression required from gene therapy.
A separate study compared recombinant factor IX Fc fusion protein (rFIXFc, Alprolix®, Biogen Idec), a newly licensed extended circulating half-life factor IX, infused on demand for bleeding events, compared to either 1) rFIXFc prophylaxis infused weekly and starting at a dose of 50 IU/kg or 2) prophylaxis infused at a dose of 100 IU/kg every ten days. Trough factor IX activity levels were obtained regularly and the dose of the lower prophylaxis dose was adjusted during the trial to maintain trough plasma factor IX activity 1–3%; the interval of the higher prophylaxis dose was adjusted to target the same trough levels. The median annualized bleeding rates were 17.7, 3.0, and 1.4 bleeding events/year for the on-demand, the lower dose, weekly therapy and the higher dose, less frequent therapy, with no statistical difference between the two prophylaxis approaches (P=0.22) [51]. There were no treatment-related safety concerns. The third recent study to consider alongside the rFIX and the rFIXFc studies is the UCL/SJCRH gene therapy trial, in which six subjects received the highest dose of 2 × 1012 VG/kg of scAAV2/8-LP1-hFIXco vector. This dose resulted in mean steady state plasma factor IX activity of 2.9–7.2% for the six subjects, a median annual bleeding rate of 1.0 episodes, and a relative 94% reduction of bleeding compared to before gene therapy [4]. Laboratory monitoring was intensive for the first four months of the trial and then tapered. Four of six participants received 8 to 12 weeks of corticosteroids to treat vector-associated hepatic inflammation. These examples of choices and outcomes are offered as a reminder that progress in gene therapy occurs in the context of constant shifts in hemophilia care standards and community outlook.
To illustrate an example of a developed world country, Table 1 compiles available data for the population of men with severe hemophilia B in the United States (keeping in mind that 80% of men with hemophilia B live in developing countries and have no access to factor IX therapy). While there are approximately 1000 men in the U.S. who could be eligible for gene therapy trials based on factor IX activity ≤2 % of normal (≤2 U/dl), the relative and the absolute exclusionary criteria that apply to liver-directed AAV.FIX trials (ongoing or planned) considerably narrow the eligible population.
The hepatocyte-directed immune response that has been elicited by AAV capsid epitopes at the most therapeutic vector doses drastically limits the eligibility of men over the age of ~ 30 years due to the high hepatitis B virus (HBV) and hepatitis C virus (HCV) carrier prevalence in this population. Almost 80% of the adult severe hemophilia B population in the United States, for instance, is seropositive following exposure to HCV. Rates of active carrier state (HCV PCR +) are not available for the hemophilia population. Although it is likely that spontaneous clearance of HCV may occur in as many as 20% of HCV exposures, at the start of the current decade most HCV Ab+ hemophilic men in the U.S. had never received therapy to attempt to eliminate the HCV carrier state. The last two years have witnessed the advent of new and well tolerated antiviral agents that are effective against even the most historically resistant strains of HCV. It can be hoped that eliminating active HCV will be a major accomplishment in hemophilia care in the next decade. Men who have a sustained HCV clearance response following therapy and no evidence of liver failure are eligible for AAV.FIX therapy and one consequence of eliminating the HCV carrier state would be to broaden the eligible population for systemic gene delivery. Alternatively, continued development of gene therapy targeting non-hepatic tissues (e.g. targeting muscles, joints, hematopoietic stem cells including platelets) may present the safest approaches in this large subset of individuals with hepatitis. [16] [53] [48] [49].
Although it is uncommon for inhibitors to complicate hemophilia B, the history of a factor IX inhibitor antibody is exclusionary for all current gene therapy trials [54]. As discussed above, multiple small and large animal studies have demonstrated that steady state endogenous factor IX expression via gene therapy may promote tolerance via the induction of factor-specific regulatory T cells (e.g. platelet-targeted gene expression) or even eradicate pre-existing factor IX inhibitors (as has been demonstrated via hepatic expression following AAV or lentivirus factor IX gene delivery) [55] [56] [18] [49]. Clinically, factor IX inhibitors are often associated with hypersensitivity reactions, and there are no reliable predictors of the risk or timing of the reactions. A deliberate attempt to reverse a factor IX inhibitor by constitutively expressing factor IX using an expression cassette that cannot be silenced involves some risk of triggering hypersensitivity that cannot be controlled. It is intriguing to consider combining formal factor IX desensitization or anti-IgE monoclonal antibody therapy with liver-directed gene therapy as a potential therapy for the very difficult clinical complication of allergy-associated factor IX inhibitors.
An additional immunologic consideration already mentioned is the growing understanding that pre-existing NAbs against AAV may cross-neutralize multiple serotypes of AAV and abrogate vector transduction at very low titers (e.g. neutralizing titers of 1:5 to 1:10) [57]. Older prevalence studies of AAV NAbs often defined as a lower limit of detection titers of 1:20 or higher, and methods of detecting neutralizing antibodies are not uniform between laboratories, so that the scope of AAV NAb interference remains unclear. Standardization of methods will be critical as the nascent field of hemophilia gene therapy grows beyond isolated, primarily academic trials and into multiple concurrent industry-supported trials. Implementation of a recently reported reference standard for the titering of AAV serotype 8 viral vectors is an important first step in being able to rationally compare dose-response and dose-toxicity results between clinical trials [58]. Currently children are excluded from gene therapy trials for diseases that do not cause death or great morbidity in childhood. It has been shown that once infants lose maternally transferred AAV NAb, there is a window around one year of life when essentially 0% of infants have AAV NAb [5]. Should liver-directed AAV gene therapy prove to be safe, to support factor IX tolerance and concerns about NAbs (and perhaps by extension also memory lymphocyte-driven CTL response)in infants are minimal, then broadening the application of gene therapy to children could be advantageous.
Following decades of limited choices regarding hemophilia B therapy, patients and their doctors now may consider variety of protein and (in the context of clinical trials) gene therapies. While gene therapy involves the most uncertain risks, extended therapeutic benefit has now been demonstrated in a human clinical trial. With multiple additional trials ongoing or commencing, one can expect that additional knowledge and experience will facilitate the discussion between patients, families and their doctors about how gene therapy may fit into the therapeutic options for hemophilia B.
Acknowledgments
PEM has been supported during this work by NIH NHLBI 1RC3HL103396 and NIH NHLBI P01-HL66973 and NIH NHLBI P01-HL112761. Tong Gui gave support with manuscript preparation and lively conversation. Andrew M. Scharenberg and Josh C. Grieger contributed technical detail.
Footnotes
Addendum: PEM reviewed the literature and wrote this paper in its entirety.
Disclosures: Paul E. Monahan receives research support through the UNC from Asklepios and Novo Nordisk and has received research support in the past from Baxter Healthcare, Novo Nordisk, Pfizer, and Prolor. He holds patents which have been licensed by UNC to Asklepios, for which he receives royalties. He has received payment for consultation, services and speaking for Asklepios, for Chatham LLC and for Baxter Healthcare, and has in addition consulted for Bayer, CSL Behring, Novo Nordisk and Pfizer.
References
- 1.Nathwani AC, Tuddenham EG, Rangarajan S, Rosales C, McIntosh J, Linch DC, Chowdary P, Riddell A, Pie AJ, Harrington C, O’Beirne J, Smith K, Pasi J, Glader B, Rustagi P, Ng CY, Kay MA, Zhou J, Spence Y, Morton CL, et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia b. N Engl J Med. 2011;365:2357–65. doi: 10.1056/NEJMoa1108046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Shapiro AD, Ragni MV, Valentino LA, Key NS, Josephson NC, Powell JS, Cheng G, Thompson AR, Goyal J, Tubridy KL, Peters RT, Dumont JA, Euwart D, Li L, Hallen B, Gozzi P, Bitonti AJ, Jiang H, Luk A, Pierce GF. Recombinant factor ix-fc fusion protein (rfixfc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia b patients. Blood. 2013;119:666–72. doi: 10.1182/blood-2011-07-367003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kaufman RJ, Powell JS. Molecular approaches for improved clotting factors for hemophilia. Blood. 2013;122:3568–74. doi: 10.1182/blood-2013-07-498261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Nathwani AC, Reiss UM, Tuddenham EG, Rosales C, Chowdary P, McIntosh J, Della Peruta M, Lheriteau E, Patel N, Raj D, Riddell A, Pie J, Rangarajan S, Bevan D, Recht M, Shen YM, Halka KG, Basner-Tschakarjan E, Mingozzi F, High KA, et al. Long-term safety and efficacy of factor ix gene therapy in hemophilia b. N Engl J Med. 2014;371:1994–2004. doi: 10.1056/NEJMoa1407309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Calcedo R, Morizono H, Wang L, McCarter R, He J, Jones D, Batshaw ML, Wilson JM. Adeno-associated virus antibody profiles in newborns, children, and adolescents. Clin Vaccine Immunol. 2011;18:1586–8. doi: 10.1128/CVI.05107-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Li C, Narkbunnam N, Samulski RJ, Asokan A, Hu G, Jacobson LJ, Manco-Johnson MJ, Monahan PE. Neutralizing antibodies against adeno-associated virus examined prospectively in pediatric patients with hemophilia. Gene Ther. 2012;19:288–94. doi: 10.1038/gt.2011.90. [DOI] [PubMed] [Google Scholar]
- 7.Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, Masurier C. Prevalence of serum igg and neutralizing factors against adeno-associated virus (aav) types 1, 2, 5, 6, 8, and 9 in the healthy population: Implications for gene therapy using aav vectors. Hum Gene Ther. 2010;21:704–12. doi: 10.1089/hum.2009.182. [DOI] [PubMed] [Google Scholar]
- 8.Lin HF, Maeda N, Smithies O, Straight DL, Stafford DW. A coagulation factor ix-deficient mouse model for human hemophilia b. Blood. 1997;90:3962–6. [PubMed] [Google Scholar]
- 9.Herzog RW, Hagstrom JN, Kung SH, Tai SJ, Wilson JM, Fisher KJ, High KA. Stable gene transfer and expression of human blood coagulation factor ix after intramuscular injection of recombinant adeno-associated virus. Proc Natl Acad Sci U S A. 1997;94:5804–9. doi: 10.1073/pnas.94.11.5804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Monahan PE, Samulski RJ, Tazelaar J, Xiao X, Nichols TC, Bellinger DA, Read MS, Walsh CE. Direct intramuscular injection with recombinant aav vectors results in sustained expression in a dog model of hemophilia. Gene Ther. 1998;5:40–9. doi: 10.1038/sj.gt.3300548. [DOI] [PubMed] [Google Scholar]
- 11.Nichols TC, Whitford MH, Arruda VR, Stedman HH, Kay MA, High KA. Translational data from adeno-associated virus-mediated gene therapy of hemophilia b in dogs. Hum Gene Ther Clin Dev. 2015;26:5–14. doi: 10.1089/humc.2014.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Manno CS, Chew AJ, Hutchison S, Larson PJ, Herzog RW, Arruda VR, Tai SJ, Ragni MV, Thompson A, Ozelo M, Couto LB, Leonard DG, Johnson FA, McClelland A, Scallan C, Skarsgard E, Flake AW, Kay MA, High KA, Glader B. Aav-mediated factor ix gene transfer to skeletal muscle in patients with severe hemophilia b. Blood. 2003;101:2963–72. doi: 10.1182/blood-2002-10-3296. [DOI] [PubMed] [Google Scholar]
- 13.Cheung WF, van den Born J, Kuhn K, Kjellen L, Hudson BG, Stafford DW. Identification of the endothelial cell binding site for factor ix. Proc Natl Acad Sci U S A. 1996;93:11068–73. doi: 10.1073/pnas.93.20.11068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Jiang H, Pierce GF, Ozelo MC, de Paula EV, Vargas JA, Smith P, Sommer J, Luk A, Manno CS, High KA, Arruda VR. Evidence of multiyear factor ix expression by aav-mediated gene transfer to skeletal muscle in an individual with severe hemophilia b. Mol Ther. 2006;14:452–5. doi: 10.1016/j.ymthe.2006.05.004. [DOI] [PubMed] [Google Scholar]
- 15.Buchlis G, Podsakoff GM, Radu A, Hawk SM, Flake AW, Mingozzi F, High KA. Factor ix expression in skeletal muscle of a severe hemophilia b patient 10 years after aav-mediated gene transfer. Blood. 2012;119:3038–41. doi: 10.1182/blood-2011-09-382317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Finn JD, Nichols TC, Svoronos N, Merricks EP, Bellenger DA, Zhou S, Simioni P, High KA, Arruda VR. The efficacy and the risk of immunogenicity of fix padua (r338l) in hemophilia b dogs treated by aav muscle gene therapy. Blood. 2012;120:4521–3. doi: 10.1182/blood-2012-06-440123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Crudele JM, Finn JD, Siner JI, Martin NB, Niemeyer GP, Zhou S, Mingozzi F, Lothrop CD, Jr, Arruda VR. Aav liver expression of fix-padua prevents and eradicates fix inhibitor without increasing thrombogenicity in hemophilia b dogs and mice. Blood. 2015;125:1553–61. doi: 10.1182/blood-2014-07-588194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Annoni A, Cantore A, Della Valle P, Goudy K, Akbarpour M, Russo F, Bartolaccini S, D’Angelo A, Roncarolo MG, Naldini L. Liver gene therapy by lentiviral vectors reverses anti-factor ix pre-existing immunity in haemophilic mice. EMBO Mol Med. 2013;5:1684–97. doi: 10.1002/emmm.201302857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, Ozelo MC, Hoots K, Blatt P, Konkle B, Dake M, Kaye R, Razavi M, Zajko A, Zehnder J, Rustagi PK, Nakai H, Chew A, Leonard D, Wright JF, et al. Successful transduction of liver in hemophilia by aav-factor ix and limitations imposed by the host immune response. Nat Med. 2006;12:342–7. doi: 10.1038/nm1358. [DOI] [PubMed] [Google Scholar]
- 20.Mingozzi F, High KA. Immune responses to aav vectors: Overcoming barriers to successful gene therapy. Blood. 2013;122:23–36. doi: 10.1182/blood-2013-01-306647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Nathwani AC, Gray JT, McIntosh J, Ng CY, Zhou J, Spence Y, Cochrane M, Gray E, Tuddenham EG, Davidoff AM. Safe and efficient transduction of the liver after peripheral vein infusion of self-complementary aav vector results in stable therapeutic expression of human fix in nonhuman primates. Blood. 2007;109:1414–21. doi: 10.1182/blood-2006-03-010181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.McCarty DM, Monahan PE, Samulski RJ. Self-complementary recombinant adeno-associated virus (scaav) vectors promote efficient transduction independently of DNA synthesis. Gene Ther. 2001;8:1248–54. doi: 10.1038/sj.gt.3301514. [DOI] [PubMed] [Google Scholar]
- 23.Nathwani AC, Gray JT, Ng CY, Zhou J, Spence Y, Waddington SN, Tuddenham EG, Kemball-Cook G, McIntosh J, Boon-Spijker M, Mertens K, Davidoff AM. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor ix expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood. 2006;107:2653–61. doi: 10.1182/blood-2005-10-4035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wu Z, Sun J, Zhang T, Yin C, Yin F, Van Dyke T, Samulski RJ, Monahan PE. Optimization of self-complementary aav vectors for liver-directed expression results in sustained correction of hemophilia b at low vector dose. Mol Ther. 2008;16:280–9. doi: 10.1038/sj.mt.6300355. [DOI] [PubMed] [Google Scholar]
- 25.den Uijl IE, Fischer K, Van Der Bom JG, Grobbee DE, Rosendaal FR, Plug I. Analysis of low frequency bleeding data: The association of joint bleeds according to baseline fviii activity levels. Haemophilia. 2011;17:41–4. doi: 10.1111/j.1365-2516.2010.02383.x. [DOI] [PubMed] [Google Scholar]
- 26.Allay JA, Sleep S, Long S, Tillman DM, Clark R, Carney G, Fagone P, McIntosh JH, Nienhuis AW, Davidoff AM, Nathwani AC, Gray JT. Good manufacturing practice production of self-complementary serotype 8 adeno-associated viral vector for a hemophilia b clinical trial. Hum Gene Ther. 2011;22:595–604. doi: 10.1089/hum.2010.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.BaxterHealthcare. Clinical trial/us nih. Nct01687608: Open-label single ascending dose of adeno-associated virus serotype 8 factor IX gene therapy in adults with hemophilia b. [updated January 20, 2015]. Available from: https://clinicaltrials.gov/ct2/show/NCT01687608?term=hemophilia+B+gene+therapy&rank=2.
- 28.Grieger JC, Samulski RJ. Adeno-associated virus vectorology, manufacturing, and clinical applications. Methods Enzymol. 2012;507:229–54. doi: 10.1016/B978-0-12-386509-0.00012-0. [DOI] [PubMed] [Google Scholar]
- 29.St Jude Children’s Research Hospital. Clinical trial/us nih. Nct00979238: Dose-escalation study of a self complementary adeno-associated viral vector for gene transfer in hemophilia b. [updated January 15, 2015]. Available from: https://clinicaltrials.gov/ct2/show/NCT00979238?term=hemophilia+B+gene+therapy&rank=2.
- 30.Mingozzi F, Anguela XM, Pavani G, Chen Y, Davidson RJ, Hui DJ, Yazicioglu M, Elkouby L, Hinderer CJ, Faella A, Howard C, Tai A, Podsakoff GM, Zhou S, Basner-Tschakarjan E, Wright JF, High KA. Overcoming preexisting humoral immunity to aav using capsid decoys. Sci Transl Med. 2013;5:194ra2. doi: 10.1126/scitranslmed.3005795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.SparkTherapeutics. Clinical trial/us nih. Nct01620801: Hemophilia b gene therapy-spark in 2012. [updated October, 2014]. Available from: https://clinicaltrials.gov/ct2/show/NCT01620801?term=hemophilia+B+gene+therapy&rank=1.
- 32.Kern A, Schmidt K, Leder C, Muller OJ, Wobus CE, Bettinger K, Von der Lieth CW, King JA, Kleinschmidt JA. Identification of a heparin-binding motif on adeno-associated virus type 2 capsids. J Virol. 2003;77:11072–81. doi: 10.1128/JVI.77.20.11072-11081.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chang J, Jin J, Lollar P, Bode W, Brandstetter H, Hamaguchi N, Straight DL, Stafford DW. Changing residue 338 in human factor ix from arginine to alanine causes an increase in catalytic activity. J Biol Chem. 1998;273:12089–94. doi: 10.1074/jbc.273.20.12089. [DOI] [PubMed] [Google Scholar]
- 34.Simioni P, Tormene D, Tognin G, Gavasso S, Bulato C, Iacobelli NP, Finn JD, Spiezia L, Radu C, Arruda VR. X-linked thrombophilia with a mutant factor ix (factor ix padua) N Engl J Med. 2009;361:1671–5. doi: 10.1056/NEJMoa0904377. [DOI] [PubMed] [Google Scholar]
- 35.Monahan PE, Sun J, Gui T, Hu G, Hannah WB, Wichlan DG, Wu Z, Grieger JC, Li C, Suwanmanee T, Stafford DW, Booth CJ, Samulski JJ, Kafri T, McPhee SW, Samulski RJ. Employing a gain-of-function factor ix variant r338l to advance the efficacy and safety of hemophilia b human gene therapy: Preclinical evaluation supporting an ongoing adeno-associated virus clinical trial. Hum Gene Ther. 2015;26:1–13. doi: 10.1089/hum.2014.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Suwanmanee T, Hu G, Gui T, Bartholomae CC, Kutschera I, von Kalle C, Schmidt M, Monahan PE, Kafri T. Integration-deficient lentiviral vectors expressing codon-optimized r338l human fix restore normal hemostasis in hemophilia b mice. Mol Ther. 2014;22:567–74. doi: 10.1038/mt.2013.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cantore A, Nair N, Della Valle P, Di Matteo M, Matrai J, Sanvito F, Brombin C, Di Serio C, D’Angelo A, Chuah M, Naldini L, Vandendriessche T. Hyperfunctional coagulation factor ix improves the efficacy of gene therapy in hemophilic mice. Blood. 2012;120:4517–20. doi: 10.1182/blood-2012-05-432591. [DOI] [PubMed] [Google Scholar]
- 38.Liu Q, Huang W, Zhang H, Wang Y, Zhao J, Song A, Xie H, Zhao C, Gao D, Wang Y. Neutralizing antibodies against aav2, aav5 and aav8 in healthy and hiv-1-infected subjects in china: Implications for gene therapy using aav vectors. Gene Ther. 2014;21:732–8. doi: 10.1038/gt.2014.47. [DOI] [PubMed] [Google Scholar]
- 39.van der Marel S, Comijn EM, Verspaget HW, van Deventer S, van den Brink GR, Petry H, Hommes DW, Ferreira V. Neutralizing antibodies against adeno-associated viruses in inflammatory bowel disease patients: Implications for gene therapy. Inflammatory bowel diseases. 2011;17:2436–42. doi: 10.1002/ibd.21673. [DOI] [PubMed] [Google Scholar]
- 40.Nijmeijer B, Petry H, Spronck L, Van Der Kruijssen C, Petersen J, Salmon F. Safety and efficacy of an adeno associated virus-based vector carrying the human clotting factor ix gene (aav5-hfix) as studied in non-human primates. Oral and poster abstracts. Blood. 2014;124(21):4800. Available from: https://ash.confex.com/ash/2014/webprogram/Paper71927.html. [Google Scholar]
- 41.Li H, Haurigot V, Doyon Y, Li T, Wong SY, Bhagwat AS, Malani N, Anguela XM, Sharma R, Ivanciu L, Murphy SL, Finn JD, Khazi FR, Zhou S, Paschon DE, Rebar EJ, Bushman FD, Gregory PD, Holmes MC, High KA. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature. 2011;475:217–21. doi: 10.1038/nature10177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Anguela XM, Sharma R, Doyon Y, Miller JC, Li H, Haurigot V, Rohde ME, Wong SY, Davidson RJ, Zhou S, Gregory PD, Holmes MC, High KA. Robust zfn-mediated genome editing in adult hemophilic mice. Blood. 2015;122:3283–7. doi: 10.1182/blood-2013-04-497354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Park CY, Kim J, Kweon J, Son JS, Lee JS, Yoo JE, Cho SR, Kim JH, Kim JS, Kim DW. Targeted inversion and reversion of the blood coagulation factor 8 gene in human ips cells using talens. Proc Natl Acad Sci U S A. 2014;111:9253–8. doi: 10.1073/pnas.1323941111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Barzel A, Paulk NK, Shi Y, Huang Y, Chu K, Zhang F, Valdmanis PN, Spector LP, Porteus MH, Gaensler KM, Kay MA. Promoterless gene targeting without nucleases ameliorates haemophilia b in mice. Nature. 2015;517:360–4. doi: 10.1038/nature13864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Niemeyer GP, Herzog RW, Mount J, Arruda VR, Tillson DM, Hathcock J, van Ginkel FW, High KA, Lothrop CD., Jr Long-term correction of inhibitor-prone hemophilia b dogs treated with liver-directed aav2-mediated factor ix gene therapy. Blood. 2009;113:797–806. doi: 10.1182/blood-2008-10-181479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Cantore A, Ranzani M, Bartholomae CC, Volpin M, Valle PD, Sanvito F, Sergi LS, Gallina P, Benedicenti F, Bellinger D, Raymer R, Merricks E, Bellintani F, Martin S, Doglioni C, D’Angelo A, VandenDriessche T, Chuah MK, Schmidt M, Nichols T, et al. Liver-directed lentiviral gene therapy in a dog model of hemophilia b. Sci Transl Med. 2015;7:277ra28. doi: 10.1126/scitranslmed.aaa1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chuah MK, Evens H, VandenDriessche T. Gene therapy for hemophilia. J Thromb Haemost. 2013;11(Suppl 1):99–110. doi: 10.1111/jth.12215. [DOI] [PubMed] [Google Scholar]
- 48.Wang X, Shin SC, Chiang AF, Khan I, Pan D, Rawlings DJ, Miao CH. Intraosseous delivery of lentiviral vectors targeting factor viii expression in platelets corrects murine hemophilia a. Mol Ther. 2015 doi: 10.1038/mt.2015.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen Y, Schroeder JA, Kuether EL, Zhang G, Shi Q. Platelet gene therapy by lentiviral gene delivery to hematopoietic stem cells restores hemostasis and induces humoral immune tolerance in fix(null) mice. Mol Ther. 2013;22:169–77. doi: 10.1038/mt.2013.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Du LM, Nurden P, Nurden AT, Nichols TC, Bellinger DA, Jensen ES, Haberichter SL, Merricks E, Raymer RA, Fang J, Koukouritaki SB, Jacobi PM, Hawkins TB, Cornetta K, Shi Q, Wilcox DA. Platelet-targeted gene therapy with human factor viii establishes haemostasis in dogs with haemophilia a. Nature communications. 2013;4:2773. doi: 10.1038/ncomms3773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Valentino LA, Rusen L, Elezovic I, Smith LM, Korth-Bradley JM, Rendo P. Multicentre, randomized, open-label study of on-demand treatment with two prophylaxis regimens of recombinant coagulation factor ix in haemophilia b subjects. Haemophilia. 2014;20:398–406. doi: 10.1111/hae.12344. [DOI] [PubMed] [Google Scholar]
- 52.Feng D, Stafford KA, Broze GJ, Stafford DW. Evidence of clinically significant extravascular stores of factor ix. J Thromb Haemost. 2013;11:2176–8. doi: 10.1111/jth.12421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sun J, Hakobyan N, Valentino LA, Feldman BL, Samulski RJ, Monahan PE. Intra-articular factor ix protein or gene replacement protects against development of hemophilic synovitis in the absence of circulating factor ix. Blood. 2008;112:4532–41. doi: 10.1182/blood-2008-01-131417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Puetz J, Soucie JM, Kempton CL, Monahan PE. Prevalent inhibitors in haemophilia b subjects enrolled in the universal data collection database. Haemophilia. 2013;20:25–31. doi: 10.1111/hae.12229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Markusic DM, Hoffman BE, Perrin GQ, Nayak S, Wang X, LoDuca PA, High KA, Herzog RW. Effective gene therapy for haemophilic mice with pathogenic factor ix antibodies. EMBO Mol Med. 2013;5:1698–709. doi: 10.1002/emmm.201302859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Crudele JM, Finn JD, Siner JI, Martin NB, Niemeyer GP, Zhou S, Mingozzi F, Lothrop CD, Jr, Arruda VR. Aav liver expression of fix-padua prevents and eradicates fix inhibitor without increasing thrombogenicity in hemophilia b dogs and mice. Blood. 2015 doi: 10.1182/blood-2014-07-588194. ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang L, Calcedo R, Bell P, Lin J, Grant RL, Siegel DL, Wilson JM. Impact of pre-existing immunity on gene transfer to nonhuman primate liver with adeno-associated virus 8 vectors. Hum Gene Ther. 2011;22:1389–401. doi: 10.1089/hum.2011.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ayuso E, Blouin V, Lock M, McGorray S, Leon X, Alvira MR, Auricchio A, Bucher S, Chtarto A, Clark KR, Darmon C, Doria M, Fountain W, Gao G, Gao K, Giacca M, Kleinschmidt J, Leuchs B, Melas C, Mizukami H, et al. Manufacturing and characterization of a recombinant adeno-associated virus type 8 reference standard material. Hum Gene Ther. 2014;25:977–87. doi: 10.1089/hum.2014.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.U.S. Center for disease control and prevention. Report on the universal data collection program. 2015 [updated February 15]. Available from: http://www.cdc.gov/ncbddd/blooddisorders/udc/documents/report-udcprogram_january2005-december-2009.
- 60.Powell JS, Apte S, Chambost H, Hermans C, Jackson S, Josephson NC, Mahlangu JN, Ozelo MC, Peerlinck K, Pasi J, Perry D, Ragni MV, Wang X, Jiang H, Li S, Cristiano LM, Innes A, Nugent K, Brennan A, Luk A, et al. Long-acting recombinant factor ix fc fusion protein (rfixfc) for perioperative management of subjects with haemophilia b in the phase 3 b-long study. Br J Haematol. 2015;168:124–34. doi: 10.1111/bjh.13112. [DOI] [PubMed] [Google Scholar]
- 61.Soucie JM, Richardson LC, Evatt BL, Linden JV, Ewenstein BM, Stein SF, Leissinger C, Manco-Johnson M, Sexauer CL Hemophilia Surveillance System Project I. Risk factors for infection with hbv and hcv in a large cohort of hemophiliac males. Transfusion (Paris) 2001;41:338–43. doi: 10.1046/j.1537-2995.2001.41030338.x. [DOI] [PubMed] [Google Scholar]