Haemophilia gene therapy - Wiley Online Library

7 downloads 0 Views 452KB Size Report
Mar 20, 2018 - successful gene therapy trials for haemophilia A or B relied on codon optimization to ...... Roth DA, Tawa NE Jr, O'Brien JM, Treco DA, Selden RF. Factor. VTTSG. .... George LA, Sullivan SK, Giermasz A, et al. Hemophilia B ...
Accepted: 20 March 2018 DOI: 10.1111/hae.13494

ORIGINAL ARTICLE

Haemophilia gene therapy: From trailblazer to gamechanger H. Evens1 | M. K. Chuah1,2 | T. VandenDriessche1,2 1 Department of Gene Therapy & Regenerative Medicine, Faculty of Medicine & Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium 2

Department of Cardiovascular Sciences, Center for Molecular & Vascular Biology, University of Leuven, Leuven, Belgium Correspondence Thierry VandenDriessche and Marinee K. Chuah, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel (VUB), Brussels Belgium. Emails: [email protected]; [email protected] Funding information FWO; Shire; Bayer; Pfizer; VUB-IOF-GEAR (GENEFIX); Strategic Research Project Grower; AFM; Willy Gepts Fund

Haemophilia is an attractive disease target for gene therapy that fostered the development of the field at large. The delivery of the clotting factor genes into the patients’ cells could be accomplished using different types of gene delivery vehicles or vectors. Adeno-­associated viral vectors (AAV) and lentiviral vectors represent some of the most promising gene delivery technologies that allow for a relatively efficient delivery of the therapeutic FVIII and FIX transgenes into the relevant target cells. To reduce the risks associated with insertional mutagenesis due to random vector integration, gene-­ editing approaches have also been considered based primarily on zinc finger nuclease (ZFN) and CRISPR/Cas. However, comprehensive analysis of off-­target effects is still required. It is particularly encouraging that relatively stable therapeutic FVIII or FIX expression levels were reached in severe haemophilia patients in recent clinical trials after liver-­directed AAV gene therapy. This success could be ascribed in part to improvements in vector design. In particular, clotting factor levels could be increased by codon optimization of coagulation factor transgenes. Alternatively, incorporation of a hyperactive gain-­of-­function R338L mutation (FIX Padua) in the FIX gene improved the overall efficacy. However, some patients still show transient liver toxicity, especially at high vector doses, possibly due to inflammatory immune responses, requiring the need for transient immunosuppression. The exact immune mechanisms are not fully understood, but may at least in some patients involve an AAV-capsid specific T cell response. Moreover, there is a need to identify the key factors that contribute to the interpatient variability in therapeutic efficacy and safety after gene therapy. KEYWORDS

adeno-associated viral vectors, coagulation, CRISPR, factor IX, factor IX Padua, factor VIII, haemophilia, lentiviral, zinc finger nuclease

1 | I NTRO D U C TI O N

FIX proteins can develop in some patients, requiring immune toler-

Current treatment for haemophilia is protein substitution therapy

ulation factors, several protein-­based next-­generation therapeutics

(PST) by administration of recombinant or plasma-­derived clotting

have been developed recently. These include PEGylation or fusion

factors. Although PST significantly improved the quality of life and

of the clotting factors with immunoglobulin domains or albumin.1

life expectancy of patients suffering from haemophilia, it has several

These products will reduce the administration frequency of recom-

disadvantages. First, it does not cure haemophilia, because patients

binant clotting factors and provide a new standard for treatment of

are still at risk of bleeding episodes despite the treatment. Second,

haemophilia patients. Despite these advances, the need to develop a

conventional clotting factors have a short half-­life, necessitating fre-

long-­term cure for haemophilia by gene therapy remains. 2

ance induction or bypass agents. To prolong the half-­life of the coag-

quent administration of factor VIII (FVIII) or factor IX (FIX). Finally,

Earlier reviews on the subject3-10 provide a comprehensive

neutralizing antibodies (“inhibitors”) against the recombinant FVIII or

description of all of the possible strategies that were tested in

50  |  wileyonlinelibrary.com/journal/hae © 2018 John Wiley & Sons Ltd

Haemophilia. 2018;24(Suppl. 6):50–59.

|

      51

EVENS et al.

preclinical animal studies and describe some of the earlier clinical

be used to boost mRNA levels of FVIII and FIX.18-20 Expression can

trials. In this review, we will highlight the most recent advances in

be further increased by combining these promoters with other reg-

gene therapy for haemophilia and address some of the outstanding

ulatory elements (e.g. hepatic control regions (HCR), enhancers).19

questions. We will highlight some of the key concepts and their clin-

Although regulatory elements are typically identified through trial-­

ical translation.

and-­ error, we have developed and experimentally validated an alternative approach to identify robust human tissue-­specific cis-­ regulatory modules (CRMs) by means of genomewide data mining.

2 | G E N E TH E R A PY

FVIII or FIX expression could be robustly increased in different gene therapy vectors when these CRMs were combined with existing

The ultimate aim of gene therapy for haemophilia is to achieve sus-

promoters. 21-23 While augmenting transgene expression, the risk

tained high-­level coagulation factor expression and a stable cor-

of insertional oncogenesis of integrating vectors was not increased

rection of the bleeding phenotype. Finding a cure for haemophilia

when using these CRMs, not even in tumour-­prone mouse models. 23

requires the delivery of the constructs to the appropriate target cells

Synthetic transcriptional enhancers can also be created de novo by

and the optimization of the coagulation FVIII or FIX expression cas-

randomly ligating synthetic oligonucleotides coding for hepatic tran-

sette. The therapeutic gene can be stably integrated into the target

scription factor binding sites and screening these in vitro in hepatic

cell chromosomes or stay in the nucleus as episomal DNA, depend-

cell lines. 24 Preclinical mouse models indicate that the cellular speci-

ing on the gene delivery vector used. One approach to achieve this is

ficity of the promoter and regulatory elements is important, because

to deliver the FVIII or FIX gene directly in vivo to nondividing post-

they can have consequences on the immune response directed

mitotic cells, like hepatocytes or skeletal muscle cells. An alternative

against the clotting factors (as discussed below). 24,25

approach is to deliver the clotting factor genes into stem/progenitor

To our knowledge, we were the first to explore the use codon-­

cells, like haematopoietic stem cells (HSCs). To achieve persisting

optimized coagulation factor genes more than 20 years ago 17 in an

expression in the HSC progeny, integrating vectors should be used.

attempt to augment their expression. The development of codon-­

Additionally, the induction of inhibitors by gene therapy should be

optimized FVIII and FIX transgenes led to an increase in protein ex-

avoided, or the therapy should induce FVIII or FIX-­specific immune

pression without changing any amino acid sequence of the FVIII or

tolerance, which already has been accomplished in preclinical stud-

FIX protein.18,21,26,27 It is therefore encouraging that all of the recent

ies.11-15 The induction of immune tolerance to the coagulation fac-

successful gene therapy trials for haemophilia A or B relied on codon

tors after gene therapy is dependent on several variables. These

optimization to boost expression levels. Preclinical studies revealed

include the vector design, target cells, transgene product or the mu-

that the outcome of the different codon optimization strategies may

tation causing the haemophilia disease phenotype.

vary, depending on the transgene and or the algorithm used. For example, typically codon optimization resulted in a twofold to three-

2.1 | Designing optimal FVIII and FIX expression cassettes

fold increase in FIX protein expression levels, whereas an increase in FVIII protein expression levels up to 44-­fold has been reported. 26 One of the potential advantages of codon optimization is that the

All gene therapy approaches focus on expressing the therapeutic

amino acids remain unchanged (i.e. synonymous mutations) which

protein at high levels while using a low dose of vector to reduce un-

may obviate immune concerns, as opposed to nonsynonymous mu-

wanted immune responses or toxicity related to the vector. Mainly

tations. However, this assumption has recently been challenged as

for FVIII this has been challenging, because of bottlenecks in ex-

synonymous mutations may inadvertently result in conformational

pression at the transcriptional, translational and post-­translational

changes in the FIX protein. 28

16

level.

Rationally designed transgenes and expression cassettes are

FVIII expression levels can be enhanced by deleting the B-­

needed to overcome these challenges. If improvements can be made

domain (FVIIIΔB), because it increased FVIII mRNA levels compared

at each of these levels, this could eventually lead to substantial dose

to when the full-­length FVIII cDNA was used. 29

benefits for haemophilia patients in clinical trials. Instead of using

However, when the B-­domain is deleted, post-­translation in-

the full-­length transgenes of FVIII and FIX, cDNAs of these genes

tracellular FVIII trafficking is compromised because of the loss of

are used in gene therapy. To improve expression levels, sometimes

critical glycosylated residues.30 This defect can be counteracted by

17,18

introns are integrated in the constructs,

which are likely to fa-

reintroducing a 17-­ amino acid synthetic B-­ domain–derived poly-

cilitate extra-­nuclear transport of the transcripts and subsequent

peptide into the FVIIIΔB cDNA, presumably reconstituting these

expression.

asparagine-­linked oligosaccharides, increasing secreted FVIII pro-

For haemophilia gene therapy, the use of the endogenous FVIII

tein levels around 3-­fold. 27 Another possibility is to incorporate a

and FIX promoters would seem attractive to mimic natural expres-

furin cleavage site within the B-­domain at position R1645H that is

sion as much as possible. However, these may not be very robust

similar to the canine sequence (HHQR vs. RHQR), which increases

and hence not very suitable to maximize expression levels in the

FVIII expression in haemophilia A mice around twofold.31 Porcine-­

face of limiting gene transfer efficiencies. Instead, tissue-­specific

human hybrid FVIIIΔB transgenes could also increase FVIII pro-

heterologous promoters, like transthyretin or α1-­antitrypsin, can

tein levels.32 However, overexpression of FVIII above 200% could

|

EVENS et al.

52      

potentially lead to cellular stress, which possibly increases the risk of inhibitor development.33 However, we showed that FVIII levels

2.3 | Preclinical and clinical studies

that are up to 75-­fold increased compared to physiological levels fol-

To achieve sustained, lifelong therapeutic coagulation factor expres-

lowing liver-­directed gene therapy did not result in liver toxicity or

sion without any toxicity or immune responses, a safe and efficient

transaminitis.34

gene therapy system is needed. Several well-­suited vectors have

The efficacy of gene therapy vectors could also be improved using hyperfunctional clotting factor genes. We have shown that

been developed for haemophilia gene therapy, each with their own advantages and limitations.

incorporation of a single R338L gain-­of-­function point mutation in the FIX gene leads to an increase in FIX activity up to eightfold using liver-­directed gene therapy. 21,35 This specific mutation (also known

2.3.1 | Nonviral vectors

as FIX-­Padua) was first discovered in patients suffering from throm-

Nonviral gene transfer generally relies on DNA-­b ased chemical

bophilia.36,37 It is a very suitable candidate to significantly boost

or physical transfection methods. Unlike viral vectors, nonviral

efficiency of haemophilia B gene therapy, with minimal change of

vectors in general do not lead to adaptive immune responses.

the FIX sequence. Independent studies have confirmed the func-

However, DNA can give rise to innate immune responses, typically

14,38,39

after gene ther-

related to Toll-­like receptor (TLR) signalling pathways. Compared

apy, and other mutations to enhance FIX activity have also been

to viral vector-­ m ediated gene therapy, the efficiency of gene

explored.40,41

transfer is in general lower in nonviral gene therapy. Additionally,

tional augmentation of this point mutation

the FVIII or FIX expression is typically short-­term after nonviral transfection, except when a selection is used on ex vivo trans-

2.2 | Target cells for haemophilia gene therapy

fected cells

There are several different cell types that are attractive targets to 3,42

47

or when transposons or integrases are used. 23,53-55

In a phase I clinical trial for haemophilia A, autologous fibroblasts

As FVIII and FIX proteins are

were electroporated with FVIII-­expressing plasmids, after which

naturally generated by liver sinusoidal endothelial cells (LSECs) and

these were selectively expanded and implanted into the omentum

hepatocytes, respectively, the liver is an obvious target organ for

of patients. Despite the fact that there were no adverse effects,

gene therapy for haemophilia. Both viral and nonviral gene therapy

the therapeutic efficiency was relatively limited and FVIII expres-

treat haemophilia by gene therapy.

systems targeting LSECs and hepatocytes have been explored.

43

sion was not sustained. 56

One of the most important features of gene therapy targeting the hepatic microenvironment is that immune tolerance towards the transgene product could be induced, which may depend partly on

2.3.2 | Adenoviral vectors

regulatory T-­cell induction.44-46 In addition to LSECs, other ECs have

The use of adenoviral vectors to treat haemophilia has been ex-

also been explored for haemophilia gene therapy, including blood

plored. However, in the late 1990s, the adenoviral vector trial for

outgrowth endothelial cells (BOECs),47,48 and strategies to enhance

ornithine transcarbamylase deficiency failed, and this research has

their engraftment and persistence after transplantation are being

been halted.57 After systemic administration, there is a risk of un-

explored further.49

controllable inflammatory reactions that can possibly be lethal,

Another target for haemophilia gene therapy is the skeletal muscle cell. These cells are able to perform all the required post-­

which is a main drawback for adenoviral gene therapy. We and others have shown that supraphysiological coagula-

translational modifications for a functional FIX protein and also have

tion factor levels can be reached after high-­c apacity adenoviral

a relatively robust secretion capacity. Whereas FIX can be easily se-

gene therapy with vectors without any viral genes and encoding

creted by muscle cells into the blood circulation, this is not the case

for the FVIII or FIX gene, without evident toxicity and inflamma-

for FVIII, possibly because of the relatively large size of the FVIII

tion in mouse and dog models of haemophilia. 34,58,59 In severe

protein particularly when a complex is formed with von Willebrand

haemophilia A patients, a phase I clinical trial was performed, in

factor (vWF). It would appear that the risk of provoking a transgene-­

which a high-­c apacity adenoviral vector encoding for the FVIII

directed immune response is higher following muscle-­directed gene

gene was administered systemically. The first treated patient

transfer, unlike in liver-­directed approaches.50 Nevertheless, early

seemed to express low FVIII levels above the 1% threshold.

clinical trials revealed that sustained FIX expression was detected

However, the trial was discontinued after that patient showed

in muscle biopsies in patients up to 10 years after muscle-­directed

signs of a transient inflammatory response with haematologic

gene therapy, despite the lack of circulating FIX levels.51

and liver abnormalities due to the vector infusion. It is therefore

Finally, haematopoietic stem cells (HSCs) are also investigated

necessary to minimize the interaction between adenoviral vec-

as haemophilia gene therapy targets to express the clotting factor

tors and the innate immune system, e.g. by localizing the delivery

genes in various haematopoietic lineages. FVIII and FIX can be se-

of the vectors. 60 Based on these concerns and given the success

creted into the blood circulation directly following HSC-­directed

of AAV-­b ased gene therapies, it seems unlikely that adenoviral

gene transfer, especially using HSC-­ derived erythrocytes, mega-

vectors would become a realistic option for haemophilia any time

karyocytes and their platelet progeny as a delivery platform.52

soon.

|

      53

EVENS et al.

in correction of the haemophilic phenotype in both mouse and dog

2.3.3 | γ-­Retroviral and lentiviral vectors

haemophilia models.52,74,75 The primary response to vascular injury

γ-­Retroviral vectors have the property to stably integrate into the

is mediated by activated blood platelets; therefore, the storage of

DNA of actively dividing cells. We demonstrated for the first time

FVIII protein within platelets could offer a locally inducible treat-

that gene therapy could cure haemophilia A in 1999 in neonatal hae-

ment for haemophilia A. Interestingly, even in presence of high-­titre

mophilia A mice, leading to lifelong FVIII expression.61 As neonatal

inhibitory antibodies after lentiviral platelet-­directed FVIII expres-

FVIII-­deficient mice have a rapid hepatocyte turn-­over, they are

sion, correction of the disease phenotype in haemophilia A mice

permissive for γ-­retroviral transduction. Subsequently, this was also

could be accomplished.73,76,77

verified in a neonatal dog model of haemophilia A and haemophilia B.

62,63

Cytotoxic preconditioning regimens, like busulfan, are needed to

Because of the necessity for cell division for this therapy, a

enable engraftment of HSCs and to create a bone marrow “niche.”

phase I clinical trial in adult haemophilia A patients resulted in low

This cannot be achieved without side effects; therefore, it is import-

FVIII levels.64,65 To overcome this problem, lentiviral vectors can be

ant to assess the risk/benefit ratio of HSC-­based gene therapy for

used, because they are capable of transducing quiescent nondivid-

haemophilia. One of the methods that has been looked into to avoid

ing hepatocytes, resulting in relatively efficient adult liver transduc-

myeloablative preconditioning is the delivery of lentiviral FVIII vec-

tion.66,67 Lentiviral vectors can not only transduce hepatocytes,

tors directly into the bone marrow. This method led to in situ trans-

but also transduce professional antigen-­presenting cells (APCs, e.g.

duction of bone marrow and HSCs and the resulting FVIII production

Kupffer cells) and LSECs.68 If FVIII or FIX is inadvertently expressed

in platelets derived from HSCs.78 An alternative option is to use B

in APCs, the risk of inhibitory antibody development is increased,

cells, which are capable of yielding therapeutic levels of FIX pro-

which prevents long-­term expression and makes the gene therapy

tein in vivo in immunodeficient mice receiving xeno-­transplantation

unsuccessful.69,70 In recent studies, it has been shown that residual

without necessitating myeloablative preconditioning.79

+

FIX expression in CD11b plasmacytoid dendritic cells (DC) could

The use of γ-­ retroviral and lentiviral vectors could result in

contribute to this immune reaction, while expression of FIX in con-

the risk of insertional oncogenesis caused by random genomic in-

ventional DCs or LSECs could instead contribute to immune toler-

tegration, which is one of its main safety concerns.67 This risk can

15

The immune tolerance that is induced by hepatic FVIII or FIX

be affected by the vector design or the presence of transcription-

gene transfer by lentiviral vectors typically requires regulatory T-­cell

ance.

ally active long terminal repeats (LTRs). This transcriptional activity

induction.

25,46

The use of hepatocyte-­specific promoters can avoid

can be reduced by removing around 400 bp in the LTR region (ie

expression in APCs and in this way result in long-­term FIX expres-

self-­inactivating vector, SIN). Combined with using a promoter in an

sion.69,70 In some circumstances, it was necessary to use micro-­RNA

internal position, this can markedly reduce the risk of insertional on-

regulated expression cassettes to tweak the specificity of expres-

cogenesis. Transducing HSCs or hepatocytes using lentiviral vectors

15,24,35,71

in tumour-­prone mouse models has confirmed that these SIN vec-

We have shown that introducing a codon-­optimized hyperac-

tors are relatively safe.72,80 This suggests that the risk of insertional

tive FIX-­R338L (Padua) gene with either integration-­defective or

oncogenesis can be significantly reduced, even in tumour-­ prone

integration-­competent lentiviral vectors in haemophilia B mice could

models, by merely changing the vector design. This risk can be fur-

sion to realize long-­term expression and immune tolerance.

significantly augment FIX activity.

35

A subsequent study in hae-

ther minimized by integration-­defective lentiviral vectors (IDLVs)

mophilia B dogs showed that liver-­directed lentiviral gene transfer

that contain an inactivation mutation in the integrase. In comparison

resulted in sustained therapeutic levels of FIX protein, which could

with conventional integrase-­competent lentiviral vectors, there was

pave the way for possible clinical translation,72 provided that inac-

a reduction in transgene expression levels.71 However, we showed

tivation of lentiviral vectors by human complement could be min-

that when the FIX-­R338L Padua variant was incorporated, relatively

imized. Most individuals have not been pre-­exposed to lentiviral

robust levels of FIX activity could be achieved even when IDLVs

vector components, contrary to AAV vectors; therefore, there is no

35

pre-­existing vector-­specific immunity, which is an advantage when

eventually contribute to a decline in transgene expression remains

using lentiviral vectors. However, lentiviral vectors can still induce

uncertain at this point.

were employed. Whether the slow hepatocyte turn-­over would

innate immune responses and the corresponding increase in pro-­ inflammatory cytokine production, because they are able to efficiently interact with professional APCs.70 To deliver FVIII or FVIII to HSC and other stem/progenitor

2.3.4 | Adeno-­associated viral vectors (AAV) Haemophilia B

cell populations, γ-­retroviral and lentiviral vectors have been em-

Stable FIX expression could be reached after muscle-­directed gene

ployed (reviewed in 66, 67, 73). When HSCs were transduced with

transfer in murine or canine models for haemophilia B that carry a

γ-­retroviral or lentiviral vectors containing the FVIII or FIX gene and

missense mutation.81,82 However, when haemophilia B dogs have a

transplanted into myeloablated haemophilic mice, this resulted in

FIX null mutation, the induction of high-­titre inhibitors after muscle-­

phenotypic correction and immune tolerance induction to the cog-

directed gene therapy limits phenotypic correction.83 The intravas-

nate transgene products. Using this method, the expression of FVIII

cular delivery of AAV-­FIX vectors to the muscle, under transient

and FIX could be directed specifically to platelets, which resulted

immune suppression could augment the therapeutic efficacy up

|

EVENS et al.

54      

to 10-­fold.84 Not only the underlying FIX gene mutation, but also

species.103,104 One advantage of the AAV8 serotype is that it shows

vector dose and local FIX antigen doses in the transduced muscle

less cross-­reactivity with pre-­existing antibodies against AAV2. Also,

determine the risk of inhibitor formation, which could be avoided

preclinical studies have indicated that the uptake of the capsid by

by transient immune suppression or by reducing the vector dose

dendritic cells could be decreased compared to AAV2 vectors, which

per site (reviewed in 85). These preclinical studies were the starting

could reduce T-­cell activation.105 Expression levels varied between

point of a phase I clinical trial by Dr. High and Dr. Kay and colleagues,

1 and 6% of normal FIX levels over a period of around 3 years, which

in which severe haemophilia B patients with an underlying missense

had not been shown previously. All 6 patients in the group receiv-

mutation in the FIX gene were administered with AAV-­FIX by intra-

ing the highest dose (2 × 1012 vg/kg) reached expression levels up

muscular injections at multiple sites.

86

Even though low or subthera-

to 5% of normal FIX. These patients displayed more than 90% less

peutic circulating FIX levels were reached, the FIX expression could

bleeding episodes and prophylactic treatment with FIX concentrate.

be locally detected in the muscle up to 10 years after injection.51 Because of the requirement of repeated muscle injections and

AAV-­specific T-­cell responses were also observed in this clinical trial, though not in all subjects.

also the risk of inhibitor development, liver-­directed AAV-­mediated

The same vector design was used in a more recent clinical trial

gene therapy was explored as another option. Besides AAV2, other

(UniQure; NCT02396342), although here AAV5 was used instead of

naturally occurring serotypes, like AAV5, AAV8 and AAV9, variants

AAV8. Approximately 4% of FIX expression levels was achieved in

of capsids acquired by evolution or selection87,88 or capsids contain-

the patients that received a dose of 5 × 1012 vg/kg of the AMT-­060

could be used. Preclinical studies in

vector.106 When a higher dose was given (2 × 1013 vg/kg) to another

haemophilia mouse or dog models and also in nonhuman primates

patient cohort, the FIX expression levels improved to almost 7%.

have shown that sustained therapeutic FIX expression levels can be

These patients showed a reduction in spontaneous bleeding rates

reached.18,20,70,90-95 Even in haemophilia B dogs that harbour a FIX

and FIX prophylactic treatment. Eight of 9 patients receiving pro-

null mutation and are prone to develop inhibitors, phenotypic cor-

phylactic treatment before gene therapy treatment did no longer

rection could be accomplished after liver-­directed AAV2-­FIX gene

need prophylaxis. Additionally, no AAV-­ specific T-­ cell responses

transfer.93 This is consistent with the clinical correlates showing that

were observed during this trial.

ing specific point mutations

89

subjects with CRM-­(cross-reacting material negative)/null mutations

Hence, to achieve comparable FIX levels as what had been re-

did not develop inhibitors after liver-­directed gene therapy.96,97 This

ported in the UCL/St Jude’s trial, it would appear that a 2.5-­to 10-­

suggests that liver-­directed gene therapy is able to reduce the risk of

fold higher vector dose was used in the UniQure trial. However, it

inhibitor development, possibly as a result of regulatory T-­cell (Treg)

remains challenging to draw definite conclusions from these trials

induction.44,98

in the absence of any head-­to-­head comparison and the low pa-

Because of these successful liver-­directed gene therapy studies,

tient numbers. Additionally, there might be other confounding vari-

a phase I clinical trial was carried out using AAV vectors containing

ables that could have an impact on the outcome of the trial, such

the FIX gene in severe haemophilia B patients. In this trial, promising

as variability in viral production methods, the purity of the vectors,

therapeutic FIX levels of almost 12% were reached.99 Unfortunately,

patient-­ specific features. Remarkably, transaminase levels were

FIX expression was only transient and there was evidence of vector

raised in patients in both the AAV5 and AAV8 trials, which could

dose-­dependent hepatotoxicity that coincided with elevated plasma

also be detected in the initial AAV2 trial.99 Consequently, patients

transaminases. This observation could be explained by the “T-­cell hy-

were given a transient immunosuppressive treatment with glucocor-

pothesis.” This implied that AAV capsid-­derived antigenic peptides

ticoids to attempt to block unwanted immune responses. Although

were possibly presented in association with MHC class I proteins

there is indirect evidence backing up the “T-­cell hypothesis” men-

by the transduced hepatocytes to T cells, as a possible explanation

tioned above, not all patients in the AAV8 trial with an increased

for the ELISPOT results obtained from PBMCs from the treated pa-

AAV capsid-­specific T-­cell response had elevated liver enzyme lev-

tients.99,100 Even though there was only modest antigen presenta-

els. Furthermore, other patients in both the AAV5 and AAV8 trial

tion, the levels could have been high enough to mark the hepatocytes

showed the opposite effect: elevated liver enzyme levels without

for T-­ cell–mediated destruction. Interestingly, this AAV-­ specific

an AAV capsid-­specific T-­cell response. This implies that the pres-

T-­cell response was not seen in the various preclinical haemophilia

ence of vector-­specific T cells in the circulation might not necessar-

models (murine, canine) nor in nonhuman primates, although several

ily be related to the immune rejection of hepatocytes transduced

efforts have been made to simulate this observation 101,102

with AAV, possibly because the liver microenvironment can have an

Another liver-­directed gene therapy clinical trial was performed

effect on the local T-­cell response. Another possibility is that the

at University College London (UCL)/St Jude’s Hospital Memphis by

elevated liver enzyme levels are not caused by the AAV-­specific T-­

Dr. Nathwani and colleagues, in which patients suffering from se-

cell response or the response was not strong enough to clear the

vere haemophilia B were intravenously administered with an AAV8

transduce hepatocytes. These controversies could eventually be

gene therapy vector containing a codon-­optimized FIX gene driven

elucidated by mouse models that are able to simulate these immune

by a liver-­specific promoter.73,96 AAV8 is able to increase transduc-

responses or by subsequent clinical trials.102

tion in hepatocytes compared to other serotypes, at least in mice,

The previously discussed AAV2, AAV5 and AAV8 trials indicate that

but this does not automatically mean it can be translated to higher

it is still important to further enhance the efficiency of these vectors,

|

      55

EVENS et al.

so lower, and thus safer, effective vector doses could be adminis-

In another more recent study, an AAV8 vector with a 5.2 kb ex-

tered, which do not result in transaminitis. The use of a hyperfunc-

pression cassette encoding for a codon-­optimized B-­domain deleted

tional codon-­optimized FIX-­R338L Padua variant led to a substantial

FVIII transgene was created. The incorporation of a 17-­amino acid

dose advantage with no evidence of an increased risk of thrombosis or

peptide (V3), which contained glycosylation residues, in the FVIIIΔB

immunogenicity in comparison with wild-­type FIX after liver-­directed

sequence could boost FVIII expression around threefold. In haemo-

Other studies have validated these

philia A mice treated with 2 × 1012 vg/kg, supraphysiological hFVIII

14,37,39

results, also in canine models by Dr. Arruda and colleagues.

expression levels (732 ± 162% of normal) could be obtained. In a non-

Therefore, the hyperfunctional FIX-­R338L Padua variant is a very in-

human primate model receiving the same dose, stable hFVIII expres-

teresting treatment option for haemophilia B gene therapy.

sion levels of 15 ± 4% were observed. Three macaques that received

gene therapy in mouse models.

21,35

In two independent recent clinical trials, the FIX-­ R338L

a higher dose did reach human FVIII expression levels of above 100%,

Padua gene was introduced and showed promising results. The

but this resulted in the development of neutralizing anti-­F VIII anti-

first trial (Shire; NCT01687608) had patients treated with a self-­

bodies, which could be eliminated by transient immunosuppression. It

complementary AAV8 vector containing a codon-­o ptimized FIX-­

is not yet known whether this V3 peptide increases the immunogenic

R338L Padua variant (BAX335). In this trial 3 dosing cohorts were

risk in patients. A clinical trial is ongoing to address this.

performed, treating 7 patients in total. Two of the patients showed

Recently, a phase I/II clinical trial was performed (BioMarin;

transient FIX activity levels of more than 50%. One of the patients

NCT02576795), in which 9 patients were treated with an AAV5 vec-

from the medium dose cohort (1 × 1012 vg/kg) showed persistent

tor expressing FVIIIΔB 114 based on encouraging results in primates

FIX levels. None of the patients had developed antibodies against

and haemophilic mice.115 Seven of those patients were treated with

FIX. The second trial (Spark Therapeutics/Pfizer; NCT02484092)

the highest dose of 6 × 1013 vg/kg after a dose escalation study.

97

resulted in a more consistent response in the patients.

The

Six of 7 patients treated with the highest dose of 6 × 1013 vg/

single-­ s tranded AAV vector (SPK-­ 9 001) encoded for a codon-­

kg showed a gradual increase in FVIII activity levels and reached

optimized FIX-­F338L Padua variant and had an alternative mu-

more than 50% of normal FVIII activity levels after 20 weeks, when

tated capsid. Fourteen weeks after receiving the initial dose of

steady-­state levels were attained. After 52 weeks, a mean FVIII ac-

5 × 1011 vg/kg, the 10 participants reached a steady-­s tate FIX ac-

tivity level of 93 ± 48% was achieved for the patients receiving the

tivity level of around 33.7 ± 18.5% without evidence of inhibitory

highest dose. In four of the patients, levels of more than 150% of

antibodies. The annualized bleeding rate of the patients dropped

normal FVIII activity levels were reached, with peaks ranging from

from 11.1 to 0.4 events after SPK-­9 001 administration. Not only

201 to 349% of normal. The six patients of the highest dose that

the annualized bleeding rate, but also the annualized infusion rate

were previously on FVIII prophylaxis showed a reduction in annual-

dropped from 67.5 to 1.2 after vector administration, rendering

ized bleeding rate from 16 to 1 event per year after treatment. The

further FIX prophylaxis unnecessary for these patients. However,

median annualized FVIII infusion rate dropped from 138 infusions

2 of the patients showed a transient elevation of liver enzymes

per year to 2 after gene transfer and was actually 0 since week 2.

and a decrease in FIX activity levels, which could indicate the pres-

However, the patients in the high-­dose cohort showed increased

ence of a capsid-­specific immune response. Oral corticosteroids

transaminase levels, but these were asymptomatic. These patients

prevented a further reduction of FIX activity levels.

did require glucocorticoid treatment after the gene therapy treatment, which was tapered off successfully for all patients. During

Haemophilia A

the 1-­year follow-­up, there was no indication of the presence of

One of the limiting steps for haemophilia AAV-­based gene therapy is

FVIII inhibitors or T-­cell–mediated immune responses to the AAV5

the packaging constraint of these vectors, because of the relatively

capsid. In this trial, the vector doses were substantially higher than

large size of the FVIII transgene. In general, the required vector dose

previously used in AAV-­based haemophilia trials, which causes chal-

was relatively high and combining the FVIII transgene with a potent

lenges during vector manufacturing.

hepatocyte-­specific promoter in a single vector remained challenging. It is preferred to use a single-­chain vector over a dual-­chain vector, which separately expresses the heavy and light chain.107-110 To

3 | CO N C LU S I O N S A N D PE R S PEC TI V E S

increase the efficiency of haemophilia A AAV-­based gene therapy, small regulatory elements can be used to drive the expression of

The recent results of haemophilia clinical trials yielded the most

a B-­domain deleted from of FVIII, codon optimizing the FVIII gene

encouraging results to date, showing sustained FIX and FVIII

and/or integrating specific mutations in the FVIII cDNA. 27,31,110-113

expression levels and a reduced bleeding rate and recombinant

Treatment of haemophilia A dogs with AAV2, AAV6 and AAV8 vec-

coagulation factor use, without requiring further prophylaxis.

tors containing the cFVIII gene resulted in persistent FVIII expres-

In particular, the improvements of vector designs or using im-

sion at a therapeutic level for more than 3 years, without antibody

proved clotting factors (codon-­o ptimized or hyperfunctional vari-

development or toxicities. However, in the haemophilia A dogs,

ants) were essential to make advancements in haemophilia gene

there was no difference in liver transduction efficiency between the

therapy. However, patients expressing FIX or FVIII in the lower

different serotypes, in contrast to mice.

range are still at risk of traumatic bleeding. Vector-­induced liver

|

EVENS et al.

56      

inflammation remains a challenge that is not fully understood but could be controlled by transient immune suppression. Although vector capsid-­s pecific T-­cell immune responses can be detected in at least some patients, it is likely that a more complicated picture will emerge whereby other immune mechanisms are involved that account for the transaminitis and/or the decline in transgene expression. This may possibly be due to innate immune responses that could be related—at least in part—to the presence

AC K N OW L E D G E M E N T S We thank the members of the Department of Gene Therapy & Regenerative Medicine and all our collaborators for their various contributions to some of the work presented in this review. We also wish to thank FWO, Shire, Bayer, Pfizer, VUB-­IOF-­GEAR (GENEFIX), Strategic Research Project Grower, AFM, Willy Gepts Fund for providing financial support.

of CpG motifs in the vector backbone. In the long run, it would be preferred to adapt the vector to the patient, rather than the other way around, and obviate the need for immune suppression altogether. In addition, controversies about the potential risk of insertional oncogenesis associated with the use of AAV vectors remain.116-123 It is particularly encouraging that preclinical studies in

D I S C LO S U R E S The authors have patent applications and granted patents in the field of gene therapy for haemophilia. They also received industrial grants and/or consultancies from various companies involved in gene therapy for haemophilia (Shire, Bayer, Pfizer).

haemophilia mice, dogs or nonhuman primates did not reveal any increased tumorigenic risk after AAV gene transfer. Nevertheless, in certain animal models, an increase in tumour incidence was apparent that seems to have been associated with the AAV treatment. Several confounding variables appear to be associated with this increased risk, including the vector dose, the vector design and the presence of strong enhancers, the mouse model and age of the recipient mouse. Interestingly, epidemiological studies revealed that Clonal integration of wild-­type adeno-­associated virus type 2 (AAV2) fragments corresponding to the 3′ untranslated region was found in 11 of 193 hepatocellular carcinomas. These AAV2 integrations occurred in known cancer driver genes, namely CCNA2, TERT, CCNE1, TNFSF10 and KMT2B, leading to their overexpression, consistent with the presence of an enhancer in the 3′ UTR.123 Nevertheless, efficient site-­specific integrating vectors based on designer zinc finger nucleases, CRISPR/Cas9 or nuclease-­free targeting approaches into “safe harbour” loci could potentially minimize this risk.124-127 These nucleases induce a double-­s trand break at the desired locus, increasing the efficiency of directed homologous recombination at least 10 000 to 100 000-­fold. These gene-­editing approaches had led to sustained clotting factor levels in haemophilic mice by either correcting the mutated clotting factor gene or by integrating a functional gene copy in a safe harbour locus. This opens up new perspectives to possibly treat paediatric subjects by gene editing even in the face of hepatocyte proliferation. Nevertheless, the immune ramifications of expressing designer nucleases and the possible off-­t arget effects following gene editing of homologous gene sequences in the genome would need to be addressed. Ultimately, it will be necessary to develop gene therapy strategies that could also be employed in patients with pre-­existing immunity to the vectors or even with high-­t itre inhibitors or a higher risk of inhibitor development than in the initial patient cohorts in these early phase I/II trials. Eventually pivotal trials are needed for marketing authorization approval by the regulatory authorities. Only then will gene therapy make a real difference for patients suffering from haemophilia and their families.

REFERENCES 1. Hartmann J, Croteau SE. 2017 Clinical trials update: innovations in hemophilia therapy. Am J Hematol. 2016;91:1252‐1260. 2. VandenDriessche T, Chuah MK. Hemophilia gene therapy: ready for prime time? Hum Gene Ther. 2017;28:1013‐1023. 3. Chuah MK, Nair N, VandenDriessche T. Recent progress in gene therapy for hemophilia. Hum Gene Ther. 2012;23:557‐565. 4. High KA, Anguela XM. Adeno-­ associated viral vectors for the treatment of hemophilia. Hum Mol Genet. 2016;25: R36‐R41. 5. Monahan PE. Gene therapy in an era of emerging treatment options for hemophilia B. J Thromb Haemost. 2015;13(Suppl 1):S151‐S160. 6. Nienhuis AW, Nathwani AC, Davidoff AM. Gene therapy for hemophilia. Mol Ther. 2017;25:1163‐1167. 7. Pierce GF, Pipe SW. A cornucopia of therapies under study for hemophilia. Mol Ther. 2017;25:2429‐2430. 8. High KA. Gene therapy for hemophilia: the clot thickens. Hum Gene Ther. 2014;25:915‐922. 9. Nathwani AC, Davidoff AM, Tuddenham EGD. Advances in gene therapy for hemophilia. Hum Gene Ther. 2017;28:1004‐1012. 10. Arruda VR, Doshi BS, Samelson-Jones BJ. Novel approaches to hemophilia therapy: successes and challenges. Blood. 2017;130:2251‐2256. 11. Annoni A, Cantore A, Della Valle P, et al. Liver gene therapy by lentiviral vectors reverses anti-­factor IX pre-­existing immunity in haemophilic mice. EMBO Mol Med. 2013;5:1684‐1697. 12. Markusic DM, Hoffman BE, Perrin GQ, et al. Effective gene therapy for haemophilic mice with pathogenic factor IX antibodies. EMBO Mol Med. 2013;5:1698‐1709. 13. Finn JD, Ozelo MC, Sabatino DE, et al. Eradication of neutralizing antibodies to factor VIII in canine hemophilia A after liver gene therapy. Blood. 2010;116:5842‐5848. 14. Crudele JM, Finn JD, Siner JI, et al. AAV liver expression of FIX-­ Padua prevents and eradicates FIX inhibitor without increasing thrombogenicity in hemophilia B dogs and mice. Blood. 2015;125:1553‐1561. 15. Merlin S, Cannizzo ES, Borroni E, et al. A novel platform for immune tolerance induction in hemophilia A mice. Mol Ther. 2017;25:1815‐1830. 16. Kaufman RJ, Pipe SW, Tagliavacca L, Swaroop M, Moussalli M. Biosynthesis, assembly and secretion of coagulation factor VIII. Blood Coagul Fibrinolysis. 1997;8(Suppl 2):S3‐S14. 17. Chuah MK, VandenDriessche T, Morgan RA. Development and analysis of retroviral vectors expressing human factor VIII

EVENS et al.

as a potential gene therapy for hemophilia A. Hum Gene Ther. 1995;6:1363‐1377. 18. Wu Z, Sun J, Zhang T, et al. Optimization of self-­complementary AAV vectors for liver-­ directed expression results in sustained correction of hemophilia B at low vector dose. Mol Ther. 2008;16:280‐289. 19. Miao CH, Ohashi K, Patijn GA, et al. Inclusion of the hepatic locus control region, an intron, and untranslated region increases and stabilizes hepatic factor IX gene expression in vivo but not in vitro. Mol Ther. 2000;1:522‐532. 20. Wang L, Takabe K, Bidlingmaier SM, Ill CR, Verma IM. Sustained correction of bleeding disorder in hemophilia B mice by gene therapy. Proc Natl Acad Sci U S A. 1999;96:3906‐3910. 21. Nair N, Rincon MY, Evens H, et al. Computationally designed liver-­ specific transcriptional modules and hyperactive factor IX improve hepatic gene therapy. Blood. 2014;123:3195‐3199. 22. Chuah MK, Petrus I, De Bleser P, et al. Liver-­specific transcriptional modules identified by genome-­wide in silico analysis enable efficient gene therapy in mice and non-­human primates. Mol Ther. 2014;22:1605‐1613. 23. Di Matteo M, Samara-Kuko E, Ward NJ, et al. Hyperactive piggyBac transposons for sustained and robust liver-­targeted gene therapy. Mol Ther. 2014;22:1614‐1624. 24. Brown BD, Cantore A, Annoni A, et al. A microRNA-­regulated lentiviral vector mediates stable correction of hemophilia B mice. Blood. 2007;110:4144‐4152. 25. Annoni A, Brown BD, Cantore A, Sergi LS, Naldini L, Roncarolo MG. In vivo delivery of a microRNA-­regulated transgene induces antigen-­specific regulatory T cells and promotes immunologic tolerance. Blood. 2009;114:5152‐5161. 26. Ward NJ, Buckley SM, Waddington SN, et al. Codon optimization of human factor VIII cDNAs leads to high-­level expression. Blood. 2011;117:798‐807. 27. McIntosh J, Lenting PJ, Rosales C, et al. Therapeutic levels of FVIII following a single peripheral vein administration of rAAV vector encoding a novel human factor VIII variant. Blood. 2013;121:3335‐3344. 28. Simhadri VL, Hamasaki-Katagiri N, Lin BC, et al. Single synonymous mutation in factor IX alters protein properties and underlies haemophilia B. J Med Genet. 2017;54:338‐345. 29. Pittman DD, Alderman EM, Tomkinson KN, Wang JH, Giles AR, Kaufman RJ. Biochemical, immunological, and in vivo functional characterization of B-­ domain-­ deleted factor VIII. Blood. 1993;81:2925‐2935. 30. Miao HZ, Sirachainan N, Palmer L, et al. Bioengineering of coagulation factor VIII for improved secretion. Blood. 2004;103:3412‐3419. 31. Siner JI, Iacobelli NP, Sabatino DE, et al. Minimal modification in the factor VIII B-­domain sequence ameliorates the murine hemophilia A phenotype. Blood. 2013;121:4396‐4403. 32. Doering CB, Denning G, Dooriss K, et al. Directed engineering of a high-­expression chimeric transgene as a strategy for gene therapy of hemophilia A. Mol Ther. 2009;17:1145‐1154. 33. Lange AM, Altynova ES, Nguyen GN, Sabatino DE. Overexpression of factor VIII after AAV delivery is transiently associated with cellular stress in hemophilia A mice. Mol Ther Methods Clin Dev. 2016;3:16064. 34. Chuah MK, Schiedner G, Thorrez L, et al. Therapeutic factor VIII levels and negligible toxicity in mouse and dog models of hemophilia A following gene therapy with high-­c apacity adenoviral vectors. Blood. 2003;101:1734‐1743. 35. Cantore A, Nair N, Della Valle P, et al. Hyperfunctional coagulation factor IX improves the efficacy of gene therapy in hemophilic mice. Blood. 2012;120:4517‐4520. 36. Simioni P, Tormene D, Tognin G, et al. X-­ linked thrombophilia with a mutant factor IX (factor IX Padua). N Engl J Med. 2009;361:1671‐1675.

|

      57

37. VandenDriessche T, Chuah MK. Hyperactive Factor IX Padua: a game-­ changer for hemophilia gene therapy. Mol Ther. 2018;26:14‐16. 38. Finn JD, Nichols TC, Svoronos N, et al. The efficacy and the risk of immunogenicity of FIX Padua (R338L) in hemophilia B dogs treated by AAV muscle gene therapy. Blood. 2012;120:4521‐4523. 39. Monahan PE, Sun J, Gui T, et al. 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;26:69‐81. 40. Schuettrumpf J, Herzog RW, Schlachterman A, Kaufhold A, Stafford DW, Arruda VR. Factor IX variants improve gene therapy efficacy for hemophilia B. Blood. 2005;105:2316‐2323. 41. Kao CY, Yang SJ, Tao MH, Jeng YM, Yu IS, Lin SW. Incorporation of the factor IX Padua mutation into FIX-­Triple improves clotting activity in vitro and in vivo. Thromb Haemost. 2013;110:244‐256. 42. Chuah MK, Collen D, VandenDriessche T. Gene therapy for hemophilia. J Gene Med. 2001;3:3‐20. 43. Kren BT, Unger GM, Sjeklocha L, et al. Nanocapsule-­delivered Sleeping Beauty mediates therapeutic Factor VIII expression in liver sinusoidal endothelial cells of hemophilia A mice. J Clin Invest. 2009;119:2086‐2099. 44. Mingozzi F, Liu YL, Dobrzynski E, et al. Induction of immune tolerance to coagulation factor IX antigen by in vivo hepatic gene transfer. J Clin Invest. 2003;111:1347‐1356. 45. Cao O, Loduca PA, Herzog RW. Role of regulatory T cells in tolerance to coagulation factors. J Thromb Haemost. 2009;7(Suppl 1):88‐91. 46. Matsui H, Shibata M, Brown B, et al. A murine model for induction of long-­term immunologic tolerance to factor VIII does not require persistent detectable levels of plasma factor VIII and involves contributions from Foxp3 +  T regulatory cells. Blood. 2009;114:677‐685. 47. Lin Y, Chang L, Solovey A, Healey JF, Lollar P, Hebbel RP. Use of blood outgrowth endothelial cells for gene therapy for hemophilia A. Blood. 2002;99:457‐462. 48. Matsui H, Shibata M, Brown B, et al. Ex vivo gene therapy for hemophilia A that enhances safe delivery and sustained in vivo factor VIII expression from lentivirally engineered endothelial progenitors. Stem Cells. 2007;25:2660‐2669. 49. Ozelo MC, Vidal B, Brown C, et al. Omental implantation of BOECs in hemophilia dogs results in circulating FVIII antigen and a complex immune response. Blood. 2014;123:4045‐4053. 50. Wang L, Cao O, Swalm B, Dobrzynski E, Mingozzi F, Herzog RW. Major role of local immune responses in antibody formation to factor IX in AAV gene transfer. Gene Ther. 2005;12:1453‐1464. 51. Buchlis G, Podsakoff GM, Radu A, et al. Factor IX expression in skeletal muscle of a severe hemophilia B patient 10 years after AAV-­mediated gene transfer. Blood. 2012;119:3038‐3041. 52. Montgomery RR, Shi Q. Platelet and endothelial expression of clotting factors for the treatment of hemophilia. Thromb Res. 2012;129(Suppl 2):S46‐S48. 53. Yant SR, Meuse L, Chiu W, Ivics Z, Izsvak Z, Kay MA. Somatic integration and long-­ term transgene expression in normal and haemophilic mice using a DNA transposon system. Nat Genet. 2000;25:35‐41. 54. Mates L, Chuah MK, Belay E, et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat Genet. 2009;41:753‐761. 55. VandenDriessche T, Ivics Z, Izsvak Z, Chuah MK. Emerging potential of transposons for gene therapy and generation of induced pluripotent stem cells. Blood. 2009;114:1461‐1468. 56. Roth DA, Tawa NE Jr, O’Brien JM, Treco DA, Selden RF. Factor VTTSG. Nonviral transfer of the gene encoding coagulation

|

58      

factor VIII in patients with severe hemophilia A. N Engl J Med. 2001;344:1735‐1742. 57. Raper SE, Yudkoff M, Chirmule N, et al. A pilot study of in vivo liver-­ directed gene transfer with an adenoviral vector in partial ornithine transcarbamylase deficiency. Hum Gene Ther. 2002;13:163‐175. 58. Ehrhardt A, Kay MA. A new adenoviral helper-­dependent vector results in long-­term therapeutic levels of human coagulation factor IX at low doses in vivo. Blood. 2002;99:3923‐3930. 59. Brown BD, Shi CX, Powell S, Hurlbut D, Graham FL, Lillicrap D. Helper-­ dependent adenoviral vectors mediate therapeutic factor VIII expression for several months with minimal accompanying toxicity in a canine model of severe hemophilia A. Blood. 2004;103:804‐810. 60. Brunetti-Pierri N, Stapleton GE, Law M, et al. Efficient, long-­term hepatic gene transfer using clinically relevant HDAd doses by balloon occlusion catheter delivery in nonhuman primates. Mol Ther. 2009;17:327‐333. 61. VandenDriessche T, Vanslembrouck V, Goovaerts I, et al. Long-­ term expression of human coagulation factor VIII and correction of hemophilia A after in vivo retroviral gene transfer in factor VIII-­ deficient mice. Proc Natl Acad Sci U S A. 1999;96:10379‐10384. 62. Xu L, Gao C, Sands MS, et al. Neonatal or hepatocyte growth factor-­ potentiated adult gene therapy with a retroviral vector results in therapeutic levels of canine factor IX for hemophilia B. Blood. 2003;101:3924‐3932. 63. Xu L, Nichols TC, Sarkar R, McCorquodale S, Bellinger DA, Ponder KP. Absence of a desmopressin response after therapeutic expression of factor VIII in hemophilia A dogs with liver-­directed neonatal gene therapy. Proc Natl Acad Sci U S A. 2005;102:6080‐6085. 64. Powell JS, Ragni MV, White GC II, et al. Phase 1 trial of FVIII gene transfer for severe hemophilia A using a retroviral construct administered by peripheral intravenous infusion. Blood. 2003;102:2038‐2045. 65. VandenDriessche T. Challenges and progress in gene therapy for hemophilia A. Blood. 2003;102:1938‐1939. 66. Matrai J, Chuah MK, VandenDriessche T. Preclinical and clinical progress in hemophilia gene therapy. Curr Opin Hematol. 2010;17:387‐392. 67. Matrai J, Chuah MK, VandenDriessche T. Recent advances in lentiviral vector development and applications. Mol Ther. 2010;18:477‐490. 68. VandenDriessche T, Thorrez L, Naldini L, et al. Lentiviral vectors containing the human immunodeficiency virus type-­1 central polypurine tract can efficiently transduce nondividing hepatocytes and antigen-­presenting cells in vivo. Blood. 2002;100:813‐822. 69. Follenzi A, Battaglia M, Lombardo A, Annoni A, Roncarolo MG, Naldini L. Targeting lentiviral vector expression to hepatocytes limits transgene-­specific immune response and establishes long-­ term expression of human antihemophilic factor IX in mice. Blood. 2004;103:3700‐3709. 70. Vandendriessche T, Thorrez L, Acosta-Sanchez A, et al. Efficacy and safety of adeno-­associated viral vectors based on serotype 8 and 9 vs. lentiviral vectors for hemophilia B gene therapy. J Thromb Haemost. 2007;5:16‐24. 71. Matrai J, Cantore A, Bartholomae CC, et al. Hepatocyte-­t argeted expression by integrase-­ defective lentiviral vectors induces antigen-­ specific tolerance in mice with low genotoxic risk. Hepatology. 2011;53:1696‐1707. 72. Cantore A, Ranzani M, Bartholomae CC, et al. Liver-­directed lentiviral gene therapy in a dog model of hemophilia B. Sci Transl Med. 2015;7:277ra28. 73. Vandendriessche T, Chuah MK. Clinical progress in gene therapy: sustained partial correction of the bleeding disorder in patients suffering from severe hemophilia B. Hum Gene Ther. 2012;23:4‐6.

EVENS et al.

74. Shi Q, Wilcox DA, Fahs SA, et al. Lentivirus-­mediated platelet-­ derived factor VIII gene therapy in murine haemophilia A. J Thromb Haemost. 2007;5:352‐361. 75. Du LM, Nurden P, Nurden AT, et al. Platelet-­t argeted gene therapy with human factor VIII establishes haemostasis in dogs with haemophilia A. Nat Commun. 2013;4:2773. 76. Kuether EL, Schroeder JA, Fahs SA, et al. Lentivirus-­ mediated platelet gene therapy of murine hemophilia A with pre-­existing anti-­factor VIII immunity. J Thromb Haemost. 2012;10:1570‐1580. 77. Shi Q, Schroeder JA, Kuether EL, Montgomery RR. The important role of von Willebrand factor in platelet-­derived FVIII gene therapy for murine hemophilia A in the presence of inhibitory antibodies. J Thromb Haemost. 2015;13:1301‐1309. 78. Wang X, Shin SC, Chiang AF, et al. Intraosseous delivery of lentiviral vectors targeting factor VIII expression in platelets corrects murine hemophilia A. Mol Ther. 2015;23:617‐626. 79. Levy C, Fusil F, Amirache F, et al. Baboon envelope pseudotyped lentiviral vectors efficiently transduce human B cells and allow active factor IX B cell secretion in vivo in NOD/SCIDgammac−/− mice. J Thromb Haemost. 2016;14:2478‐2492. 80. Montini E, Cesana D, Schmidt M, et al. The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. J Clin Invest. 2009;119:964‐975. 81. Herzog RW, Yang EY, Couto LB, et al. Long-­term correction of canine hemophilia B by gene transfer of blood coagulation factor IX mediated by adeno-­associated viral vector. Nat Med. 1999;5:56‐63. 82. Herzog RW, Hagstrom JN, Kung SH, et al. 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;94:5804‐5809. 83. Herzog RW, Mount JD, Arruda VR, High KA, Lothrop CD Jr. Muscle-­ directed gene transfer and transient immune suppression result in sustained partial correction of canine hemophilia B caused by a null mutation. Mol Ther. 2001;4:192‐200. 84. Arruda VR, Stedman HH, Haurigot V, et al. Peripheral transvenular delivery of adeno-­associated viral vectors to skeletal muscle as a novel therapy for hemophilia B. Blood. 2010;115:4678‐4688. 85. Wang L, Herzog RW. AAV-­mediated gene transfer for treatment of hemophilia. Curr Gene Ther. 2005;5:349‐360. 86. Kay MA, Manno CS, Ragni MV, et al. Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector. Nat Genet. 2000;24:257‐261. 87. Grimm D, Lee JS, Wang L, et al. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-­associated viruses. J Virol. 2008;82:5887‐5911. 88. Lisowski L, Dane AP, Chu K, et al. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature. 2014;506:382‐386. 89. Zhong L, Li B, Mah CS, et al. Next generation of adeno-­associated virus 2 vectors: point mutations in tyrosines lead to high-­ efficiency transduction at lower doses. Proc Natl Acad Sci U S A. 2008;105:7827‐7832. 90. Snyder RO, Miao CH, Patijn GA, et al. Persistent and therapeutic concentrations of human factor IX in mice after hepatic gene transfer of recombinant AAV vectors. Nat Genet. 1997;16:270‐276. 91. Snyder RO, Miao C, Meuse L, et al. Correction of hemophilia B in canine and murine models using recombinant adeno-­associated viral vectors. Nat Med. 1999;5:64‐70. 92. Wang L, Nichols TC, Read MS, Bellinger DA, Verma IM. Sustained expression of therapeutic level of factor IX in hemophilia B dogs by AAV-­mediated gene therapy in liver. Mol Ther. 2000;1:154‐158. 93. Mount JD, Herzog RW, Tillson DM, et al. Sustained phenotypic correction of hemophilia B dogs with a factor IX null mutation by liver-­directed gene therapy. Blood. 2002;99:2670‐2676.

|

      59

EVENS et al.

94. Nathwani AC, Davidoff AM, Hanawa H, et al. Sustained high-­level expression of human factor IX (hFIX) after liver-­t argeted delivery of recombinant adeno-­associated virus encoding the hFIX gene in rhesus macaques. Blood. 2002;100:1662‐1669. 95. Nathwani AC, Gray JT, Ng CY, et al. 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;107:2653‐2661. 96. Nathwani AC, Tuddenham EG, Rangarajan S, et al. Adenovirus-­ associated virus vector-­mediated gene transfer in hemophilia B. N Engl J Med. 2011;365:2357‐2365. 97. George LA, Sullivan SK, Giermasz A, et al. Hemophilia B gene therapy with a high-­specific-­activity factor IX variant. N Engl J Med. 2017;377:2215‐2227. 98. Dobrzynski E, Fitzgerald JC, Cao O, Mingozzi F, Wang L, Herzog RW. Prevention of cytotoxic T lymphocyte responses to factor IX-­expressing hepatocytes by gene transfer-­induced regulatory T cells. Proc Natl Acad Sci U S A. 2006;103:4592‐4597. 99. Manno CS, Pierce GF, Arruda VR, et al. Successful transduction of liver in hemophilia by AAV-­Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12:342‐347. 100. Mingozzi F, Maus MV, Hui DJ, et al. CD8(+) T-­ cell responses to adeno-­ associated virus capsid in humans. Nat Med. 2007;13:419‐422. 101. Pien GC, Basner-Tschakarjan E, Hui DJ, et al. Capsid antigen presentation flags human hepatocytes for destruction after transduction by adeno-­ associated viral vectors. J Clin Invest. 2009;119:1688‐1695. 102. Martino AT, Basner-Tschakarjan E, Markusic DM, et al. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-­specific CD8 +  T cells. Blood. 2013;121:2224‐2233. 103. Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. Novel adeno-­ associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A. 2002;99:11854‐11859. 104. Gao G, Vandenberghe LH, Alvira MR, et al. Clades of Adeno-­ associated viruses are widely disseminated in human tissues. J Virol. 2004;78:6381‐6388. 105. Vandenberghe LH, Wang L, Somanathan S, et al. Heparin binding directs activation of T cells against adeno-­associated virus serotype 2 capsid. Nat Med. 2006;12:967‐971. 106. Miesbach W, Meijer K, Coppens M, et al. Gene therapy with adeno-­associated virus vector 5-­human factor IX in adults with hemophilia B. Blood 2017;131:1022‐1031. 107. Sarkar R, Xiao W, Kazazian HH Jr. A single adeno-­associated virus (AAV)-­murine factor VIII vector partially corrects the hemophilia A phenotype. J Thromb Haemost. 2003;1:220‐226. 108. Sarkar R, Tetreault R, Gao G, et al. Total correction of hemophilia A mice with canine FVIII using an AAV 8 serotype. Blood. 2004;103:1253‐1260. 109. Scallan CD, Lillicrap D, Jiang H, et al. Sustained phenotypic correction of canine hemophilia A using an adeno-­associated viral vector. Blood. 2003;102:2031‐2037. 110. Sabatino DE, Lange AM, Altynova ES, et al. Efficacy and safety of long-­term prophylaxis in severe hemophilia A dogs following liver gene therapy using AAV vectors. Mol Ther. 2011;19:442‐449.

111. Jiang H, Lillicrap D, Patarroyo-White S, et al. Multiyear therapeutic benefit of AAV serotypes 2, 6, and 8 delivering factor VIII to hemophilia A mice and dogs. Blood. 2006;108:107‐115. 112. Nguyen GN, George LA, Siner JI, et al. Novel factor VIII variants with a modified furin cleavage site improve the efficacy of gene therapy for hemophilia A. J Thromb Haemost. 2017;15:110‐121. 113. Brown HC, Wright JF, Zhou S, et al. Bioengineered coagulation factor VIII enables long-­term correction of murine hemophilia A following liver-­ directed adeno-­ associated viral vector delivery. Mol Ther Methods Clin Dev. 2014;1:14036. 114. Rangarajan S, Walsh L, Lester W, et al. AAV5-­factor VIII gene transfer in severe hemophilia A. N Engl J Med. 2017;377:2519‐2530. 115. Bunting S, Zhang L, Xie L, et al. Gene therapy with BMN 270 results in therapeutic levels of FVIII in mice and primates and normalization of bleeding in hemophilic mice. Mol Ther. 2018;26:496‐509. 116. Donsante A, Miller DG, Li Y, et al. AAV vector integration sites in mouse hepatocellular carcinoma. Science. 2007;317:477. 117. Li H, Malani N, Hamilton SR, et al. Assessing the potential for AAV vector genotoxicity in a murine model. Blood. 2011;117:3311‐3319. 118. Nault JC, Datta S, Imbeaud S, et al. Recurrent AAV2-­related insertional mutagenesis in human hepatocellular carcinomas. Nat Genet. 2015;47:1187‐1193. 119. Berns KI, Byrne BJ, Flotte TR, et al. Adeno-­associated virus type 2 and hepatocellular carcinoma? Hum Gene Ther. 2015;26:779‐781. 120. Buning H, Schmidt M. Adeno-­associated Vector Toxicity-­To Be or Not to Be? Mol Ther. 2015;23:1673‐1675. 121. Chandler RJ, LaFave MC, Varshney GK, et al. Vector design influences hepatic genotoxicity after adeno-­associated virus gene therapy. J Clin Invest. 2015;125:870‐880. 122. Chandler RJ, Sands MS, Venditti CP. Recombinant adeno-­ associated viral integration and genotoxicity: insights from animal models. Hum Gene Ther. 2017;28:314‐322. 123. Logan GJ, Dane AP, Hallwirth CV, et al. Identification of liver-­ specific enhancer-­promoter activity in the 3′ untranslated region of the wild-­t ype AAV2 genome. Nat Genet. 2017;49:1267. 124. Li H, Haurigot V, Doyon Y, et al. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature. 2011;475:217‐221. 125. Anguela XM, Sharma R, Doyon Y, et al. Robust ZFN-­mediated genome editing in adult hemophilic mice. Blood. 2013;122:3283‐3287. 126. Sharma R, Anguela XM, Doyon Y, et al. In vivo genome editing of the albumin locus as a platform for protein replacement therapy. Blood. 2015;126:1777‐1784. 127. Park CY, Kim DH, Son JS, et al. Functional correction of large factor VIII gene chromosomal inversions in hemophilia a patient-­ derived iPSCs using CRISPR-­C as9. Cell Stem Cell. 2015;17:213‐220.

How to cite this article: Evens H, Chuah MK, VandenDriessche T. Haemophilia gene therapy: From trailblazer to gamechanger. Haemophilia. 2018;24(Suppl. 6):50‐59. https://doi.org/10.1111/ hae.13494