Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

IJMCM Review Article

Winter 2020, Vol 9, No 1


DOI: 10.22088/IJMCM.BUMS.9.1.33

Factor VIII: Perspectives on Immunogenicity and Tolerogenic


Strategies for Hemophilia A Patients

Sheyda Khalilian, Majid Motovali-Bashi, Halimeh Rezaie

Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science
and Technology, University of Isfahan, Iran.

A major complication in treating hemophilia A is the development of neutralizing antibodies (inhibitors) against
therapeutic administered factor VIII (FVIII), which occurs in approximately 20-30% of patients with severe
disease. These inhibitors render FVIII replacement therapy ineffective and increase the morbidity and mortality
risk. The currently accepted method to eradicate inhibitors is immune tolerance induction (ITI), and frequent
intensive administration of FVIII until inhibitor titers drop. Current ITI protocols are extremely costly and not
effective in all patients. During the last decade, many types of research have been accomplished to clarify the
mechanisms that mediate immune tolerance induction. Novel experimental therapies including monoclonal
antibodies, viral vector-mediated gene therapy, regulatory T cell induction using immunosuppressive drugs, and
nanoparticle-based immune modulation show promising results in hemophilia A clinical trials. This review
focuses on treatment options towards the anti-FVIII immune responses and current novel therapies in clinical
trials.

Key words: Factor VIII, hemophilia A, inhibitors, immune tolerance

H emophilia A is an X-linked disorder which is


associated with recurrent bleeding in affected
patients. The prevalence of hemophilia A is 1:5000
has restricted its application. Inhibitor development
predominantly occurs in approximately 30% of all
patients with severe hemophilia A and 5% of all
of live male birth without any genetic predispo- patients with mild hemophilia A (4). A series of
sition (1). This life-threatening disorder is caused both genetic and non-genetic factors are involved in
by a functional defect in plasma coagulation factor FVIII inhibitor development. Mutations at specific
VIII (FVIII) in human. For many years, regions of F8 gene (hot spots, heavy chain coding
administration of this protein as a plasma or regions) are associated with delayed or absent pro-
recombinant product has been used as a treatment coagulant activity. Patients with large deletions and
for hemophilia (2, 3). non-sense mutations should lack tolerance to all of
Alternative transfused FVIII has been a the epitopes in FVIII, and patients carrying small
practical treatment; however, developing antibodies deletions/insertions and missense mutations are
against this protein, which play a role as inhibitors, associated with mild or moderately severe


Corresponding author: Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and
Technology, University of Isfahan, Iran. Email: mbashi881@gmail.com
This work is published as an open access article distributed under the terms of the Creative Commons Attribution 4.0 License
(http://creativecommons.org/licenses/by-nc/4). Non-commercial uses of the work are permitted, provided the original work is properly
cited.
Khalilian SH et al.

hemophilia A. Findings from a series of studies domain protein comprises a heavy chain (A1-A2-B
showed a significant association between the HLA domains) and a light chain (A3-C1-C2 domains)
class II molecules and inhibitors development. (15).
However, the strength of this association depends FVIII endures several post-translational
on the genetic background of different populations modifications, including a high glycosylation
(5). For instance, Hosseini et al. found a strong process before becoming ready to circulate as a
association between HLA-DRB1*01:01 allele and a prepared glycoprotein. The heterodimer FVIII a
lower risk of developing inhibitor in Iran (6). high affinity (kd~0.3 nM) for von Willebrand factor
Actually, due to the development of these inhibitor (VWF), which is a chaperone molecule acting
antibodies in responder patients, the FVIII through the blood circulation (16).
replacement therapy is not an efficient and cost- The VWF-FVIII complex is formed through
effective treatment (7). Consequently, ineffective non-covalent interactions between A3, C1 and C2
treatment may lead to worsening of the situation domains of FVIII and the D’D3 domains of VWF.
through bleeding and consequent high morbidity Actually, VWF is essential for the maintenance of
rate increases disability and decreases quality of the heterodimer structural stability of FVIII, and
life (8). increases the stability of the interaction between the
Given the significant burden of inhibitors on FVIII heavy and light chains and also increases the
both patient's health and health-care costs, many FVIII half-life. In addition, the interaction of VWF
efforts have been made to prevent anti-FVIII with FVIII is beneficial for primary hemostasis, so
antibodies. Immune tolerance induction (ITI) is the it increases FVIII involvement in vascular injury.
most successful approach which consists of Activated FVIII (FVIIIa) is released from
intensive high dose FVIII treatment (9-11). VWF following proteolytic cleavage and release of
Although ITI has been effective in 60-80% of the B-domain. (17, 18). The formation of FVIIIa is
the patients, and is known as a preventive treatment crucial for the coagulation cascade due to the
for complications following the inhibitor important role of FVIIIa as a cofactor for factor IXa
development, it is extremely expensive and it often protein. In fact, intrinsic Xase complex, which is
takes several years in to achieve an effective essential for thrombin formation is normally
tolerance (12, 13). Accordingly, there is still a generated as a result of the factor IXa function with
compelling need for cost-effective treatment with the aid of its cofactor, FVIIIa. Endogenous FVIII
the rapid and productive clinical results in inducing has a half-life of 12–16 h, after which FVIII is
tolerance against FVIII. (14). eliminated by the liver and probably also by the
In this review, we discuss the characteristics spleen. Of note, the life cycle of therapeutically
of factor FVIII structure, and the pathophysiology administered FVIII is similar to endogenous FVIII.
of inhibitor formation. In addition, several novel However, the interaction between endogenous
approaches to modulate the immune response and VWF and FVIII normally takes place in the
induce tolerance are described. circulation instead of the liver (19, 20).
Factor VIII structure and function Molecular and clinical aspects of inhibitor
FVIII is a non-covalent heterodimer protein, formation
which is encoded by the F8 gene. The immature Definition of an inhibitor
FVIII protein consists of 2351 amino acids (aa) The immune system in responder patients
comprising a mature protein of 2332 aa and a signal produces polyclonal high-affinity IgG antibodies
protein of 19 aa. As Figure 1 shows, this multi- against FVIII which are known as FVIII inhibitors.

Int J Mol Cell Med Winter 2020; Vol 9 No 1 34


Inhibitor formation and strategies for hemophilia A treatment

This is a T cell-dependent immune response in over time (24). In addition, some anti-FVIII
which associated innate and adaptive immune cells, antibodies have the ability to play a role as a
including antigen-presenting cells (APCs), B and T- hydrolytic enzyme and inactivate the FVIII (25).
helper lymphocytes are involved (21). According to the kinetics and severity of
Although FVIII protein contains many inhibitory functions, anti-FVIII antibodies have
functional epitopes which can potentially interact been classified to two groups, including type I and
with specific inhibitors, antibodies targeting the A2 II. Type I inhibitors are able to thoroughly
and/or C2 domains of FVIII are the most frequent inactivate FVIII based on their second-order
(22). kinetics (dose-dependent linear inhibition). They
Recognition of the above-mentioned epitopes are more common in patients with severe
by antibody gives rise to the blockade in functional hemophilia. Conversely, type II inhibitors, due to
domains of the FVIII protein, which are normally their complex kinetics, inactivate FVIII in an
used to interact with FIX, phospholipid and VWF incomplete way, and are commonly developed in
factor interaction sites (23). Different targets of patients with mild hemophilia or who develop
FVIII epitope can be recognized by the inhibitors at acquired FVIII inhibitor (26).
the same time, and these epitope targets can change Immune response to FVIII

Fig. 1. Factor VIII structure and domain organization. A: linear arrangement as well as activation cleavage sites of the FVIII domains
A1, A2, B, A3, C1, C2 are shown; B: three dimensional crystal structure of B-domain depleted human factor VIII was created using RCSB
PDB viewer.

35 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

Recently, many review studies have discussed due to glycosylation patterns depending on their
several aspects of the immune response against cell expression system and covalent modifications
FVIII which can be the result of considerable to increase the circulating half-life of the protein
advances in this area (27, 28). FVIII proteins, at the (Fc fusion, PEGylation etc.) (35, 36).
first encounter with the immune system, are As both humoral and cellular arms of the
predominantly recognized and captured by immune system are involved in an effective
professional antigen presenting cells (APCs) immune response, T cell activation gives rise to B
including dendritic cells (DCs), macrophages, and cell differentiation, proliferation, and class
B cells. Following internalization of FVIII in to switching. As a consequence, B cell activation
APCs, it undergoes proteolytic cleavage in the results in producing large amounts of FVIII
endosomal compartments. Afterwards, small antibodies that block the FVIII function. Moreover,
peptides derived from a large FVIII protein are memory B cells and specialized plasma cells are
presented on the surface of APC through major also ready to intensify the battle (22). During the
histocompatibility complex (MHC) class II. The secondary immune response, T cell and B cell
MHCII-peptide complex is then recognized by activities are intensified, and higher amounts of
naïve T cells present in the lymph nodes (29). To specific antibodies which have already undergone
effectively respond to the FVIII peptides, naive T the affinity maturation process are produced from
cells should be differentiated into specialized plasma cells (Figure 2). Most of indentified
+
effector CD4 T cells. In addition to the MHCII- antibodies against FVIII epitopes in hemophilia
peptide complex, there are several co-stimulatory patients are immunoglobulin G which IgG1 and
molecules engaged in the effector T cell IgG4 are the most prevalent subclasses. IgG4
development. CD40 and CD80/86 (B7-1/B7-2) production needs a further class switching process
molecules on the surface of APCs come into a that proves the involvement of T cells in the
strong interaction with CD40 ligand (CD40L) and formation of an effective immune response against
+
CD28 on the surface of naïve helper T cells (CD4 ), FVIII (37).
resulting in the secretion of cytokines by both DCs As it has been shown in murine hemophilia
and T cells (30) Interestingly, when human DCs are models, the presence of co-stimulatory interactions
cultured with FVIII, DCs do not mature (31). The and signals is very important to form a protective
cause of this difference in in vitro and in vivo immune response. Actually, the lack of a functional
immune system recognition of FVIII is unclear, but co-stimulatory signal gives rise to anergy or
probably the environmental differences, including apoptosis of the T cells. A defective immunologic
danger signals resulting from trauma, surgery, synapse in the absence of co-stimulatory
severe/recurrent bleeds, infection or vaccination interactions produces tolerogenic T cells that do not
may influence the risk of immune reaction towards react with the antigens presented by the APCs
administered FVIII in hemophilia A patients (32, (38, 39).
33). The physical characteristics of the FVIII Immune tolerance induction
antigen, such as post-translational modifications or ITI is the most prevalent method exploited to
physical aggregation in high administered doses, prevent complications related to FVIII infusion
may have a significant effect on its immunogenicity (11, 40). As mentioned before, frequent exposure
(34). Differences in the intrinsic or extrinsic of the immune cells to FVIII antigens in a non-
property of the antigen, could affect the immune inflammatory environment can lead the immune
response against administered FVIII. This may be system response toward anergy and tolerance How-.

Int J Mol Cell Med Winter 2020; Vol 9 No 1 36


Inhibitor formation and strategies for hemophilia A treatment

Fig. 2. Primary and secondary immunization in response to FVIII. A: primary response; upon the first exposure to FVIII, this protein is
internalized by DCs or other APCs; then the protein is presented to naive CD4 + T cells which results in its activation in the presence of co-
stimulatory signals. The activated T cell activates naive B cells that expand and differentiate into FVIII plasma cells, secreting anti-FVIII
IgM antibodies, or FVIII memory B cells. B: Secondary response; in this type of immunization, FVIII memory B cells act as APCs and
activate memory T cells, and then memory B cells will differentiate into FVIII plasma cells that secrete anti-FVIII IgG antibodies. DC:
dendritic cell; APC: antigen presenting cell; MHC: major histocompatibility complex; TCR: T-cell receptor; CD40L: CD40 ligand.

-ever, the mechanism of this tolerance induction is However, due to the high cost, moderate success
not completely understood. Through the ITI rate, long duration and inconvenience of daily
method, regular administration of FVIII makes the infusions, ITI therapy is not a method to be used
immune cells to ignore FVIII antigens and develop extensively. In order to bring better results, all of
a tolerogenic response against it (41). To reach the these complications highlight the need for further
most practical ITI treatment, many different modifications (44).
regimens such as encompassing protocols with There are three mechanisms which all
variations in FVIII dose and using additional contribute in inducing immune tolerance:
immunosuppressive agents have been tested. From 1. Development of monoclonal antibodies (mAbs)
all of them, Bonn, Van Creveld, and Malmö ITI as the specific immuno-suppressant
protocols are mostly accepted and used, and 2. Anergy of effector T-cells due to the induction of
underwent various modifications. It has been shown Tregs using chronic exposition of FVIII
that the immune modulation effectively increases 3. Inhibition of memory B cells differentiation into
the result of the treatment (42). plasma cells using high FVIII concentrations
Elucidating the mechanisms leading to Development of monoclonal antibodies (mAbs)
functional immune tolerance and specific reduction A recent investigation has shown that mAbs
or elimination of inhibitor responses are the can be used as specific immunosuppressive agents
common destinations of all investigations on ITI that appear to be both more effective and more
therapy to develop a promising treatment (43). selective in facilitating immune tolerance induction,

37 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

and they are generally well tolerated by recipients. The mechanism involved in this process is
Actually, these immunosuppressive agents pointing increasing immunomodulatory cytokine TGF- β
at specific targets have less toxicity in comparison that is a key factor in decreasing cellular immune
with the immunosuppressive agents that target recognition of FVIII antigen. Consequently, a long-
many aspects of the immune system. Hence, they term tolerance was formed against FVIII, and the
have considerably lower side effects, and can be inhibitor development was controlled. Besides,
better tolerated by the recipients. There have been administration of anti-CD3 alone diminished pre-
many studies on FVIII knockout mice using mAbs existing antibodies against FVIII (54).
affecting a myriad of immunological pathways. The Regulatory CD4+CD25+ T cells play a
outcome has shown successful tolerance induction, prominent role in homeostasis of T cells. Actually,
especially when co-administered with antigen (45- they balance the cellular immune response by
47). Several investigations have displayed that suppressing effector T cells after a protective
blockade of co-stimulatory molecules’ responses, immune response against an antigen. T cell
including CD40/CD40L and B7/CD28 can hemostasis through Treg activity is very important
effectively diminish the inhibitor development in to protect the body against complications followed
hemophilia A mice models. It has been shown that by an uncontrolled immune response including
mAbs against CD40L prevented anti-FVIII autoimmunity or alloimmune responses. This
antibodies induction, with suppression of FVIII characteristic of regulatory T cells has been
specific T-cell responses in FVIII-primed exploited to induce a long-term immunomodulatory
hemophilia A mice (48, 49). In addition, blockade response against FVIII (55). In fact, a higher
of B7/CD28 using specific antibodies against number or percentage of CD4+FOXP3+ Tregs in
CTLA4 (CTLA4-Ig) effectively obstruct the both protein replacement and/or gene therapy
inhibitor development in a mouse model of settings are used to control inhibitor formation even
hemophilia A (50). in patients with a measurable pre-existing inhibitor
Another study on FVIII-plasmid treated titer (56, 57). These regulatory cells have been
hemophilia A mice revealed that dual blockade of shown to effectively restrict both cellular and
CD40/CD40L and B7/CD28 pathways using humoral immunity through suppressing cytotoxic
combined anti-CD40L and CTLA4-Ig obstruct both CD8+ T cell activity and antibody production by B
associated co-stimulatory signals in a synergic cells in mice and non-human primates, respectively
manner, and effective long-term tolerance was (58, 59). In addition, Fas/FasL interaction and T
obtained against FVIII (51, 52). cell depletion were also studied in Fas-deficient
T cell anergy and Treg induction mice to highlight the importance of Treg activity in
In addition to co-stimulatory molecules, mAbs the formation of a robust immune tolerance (60).
targeting pan T cell markers, especially CD3 have Regulatory T cells use different mechanisms
been shown to effectively increase T cell apoptosis. to prevent differentiation of T cells into effector
Apparently, T cell depletion decreases the cellular cells and promote their conversion into Tregs. They
immune response against the antigen of interest can either directly contact the target cells or
(53). indirectly reduce their activity by producing
As a study on hemophilia A mice revealed, immunomodulatory elements comprising IL-10
that anti-CD3 treatment, concomitant with FVIII- and/or TGF-β cytokines (61). It has been shown
plasmid injection prevent inhibitory antibodies and that using antigen together with the mTOR inhibitor
persistent FVIII expression levels was achieved. Rapamycin can effectively reduce the effector T

Int J Mol Cell Med Winter 2020; Vol 9 No 1 38


Inhibitor formation and strategies for hemophilia A treatment

cells and increase the regulatory lymphocytes tolerance against FVIII protein, which was
instead (Figure 3-A) (62). generally due to their ability to convert from FVIII-

IL-2/IL-2 mAb complexes are a good specific CD4+ CD25 conventional T cells into
alternative method to enrich Treg in vivo and rapid Tregs, in the presence of TGF-β1 (64, 65).
+ + +
formation of CD4 CD25 FOXP3 Tregs. These Therefore, producing a higher number of
Tregs are highly effective as a pre-treatment for regulatory T cells with specificity to FVIII through
preventing autoimmunity or transplant rejection due redirecting antigen-specificity through TCR or
to their strong suppressive activity (63). IL-2/IL-2 chimeric antigen receptor (CAR) gene transfer to
mAb complexes were also used in a hemophilia A Treg is extensively attractive (Figure 3-B). The
mice models and caused a considerable reduction in investigations showed that the expression of a
FVIII inhibitor titer to either FVIII replacement single human TCR, using ex vivo retroviral gene
therapy or plasmid-mediated gene therapy. It has transfer, can negatively affect CD4 + T cell
been proven that these Tregs induce a lasting and B cell activity, and also suppress hemophilia A

Fig. 3. Significant approaches for inducing tolerance and eliminating inhibitor formation in hemophilia A. A: selective deletion of
CD4+ T-helper cells and Tregs induction occur upon administration of Rapamycin as an immuno-suppressant; B: CAR Tregs are engineered
using fusion of TCR signaling molecules and scFV domains. CAR Tregs become activated through recognition of FVIII presented by DCs.
These pathways result in CD4+ T cells suppression and APC toleration; C: hepatic lentiviral gene transfer is known as a way for tolerance
induction which results in programmed cell death of CD4 + T-helper cells and the Treg induction. CAR: chimeric antigen receptor; scFV:
single-chain variable fragment; Treg: regulatory T cell.

39 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

mice expressing human HLA (66). Actually, the (Rituximab) is a monoclonal IgG1 which is
induced regulatory T cells predominantly react extensively used as immunomodulatory agent. This
against only a single epitope. However, this antibody can react with CD20 markers on the
recognition can lead to a general suppression surface of follicular and marginal zone B cells, and
against the FVIII molecule entirely. Due to highly deplete them via apoptosis. Hence, using a single
extensive HLA differences, the production of Treg dose of this immunomodulatory antibody followed
against every single immuno-dominant epitope in by daily high-dose FVIII intravenous injections in
FVIII protein requires many TCR clones. Hence, FVIII primed-mice showed a significant reduction
the CAR approach came to streamline general Treg in the inhibitors. On the other hand, using
production, which can be used for all patients as it Rituximab could affect T cells, so that increased
is not MHC restricted, and consequently antigen levels of regulatory T cells were observed in the
recognition and cell signaling by CAR Tregs spleen after pretreatment with anti-CD20. It is
happens in the absence of APCs (67, 68). CAR T worthy to note that decreased levels of B cell
cells are predominantly produced through immune response against FVIII lasted for three
introducing antigen recognition variable region months after Rituximab administration (75).
(single-chain variable fragment) antibody domains A dual therapy through which both T and B
fused to primary and co-stimulatory signaling cells are targeted have been shown to bring a
molecules and specialized to recognize surface promising treatment to control inhibitor
antigens (69, 70). However, studies have development in hemophilia A patients.
demonstrated that experimentally produced FVIII- Accordingly, Biswas et al. demonstrated that a
specific human CAR Tregs can provoke a combination of murine anti-CD20 IgG2a antibody
suppressive response against soluble FVIII and and Rapamycin effectively decrease both humoral
decrease the inhibitor development both in vitro and cellular immune responses against FVIII, and
and in vivo in hemophilia A mice (71). However, resulted in decreased FVIII inhibitor formation in
the exact mechanism is not completely understood. mice (76). The major side effect of both T and B
In fact, using immunotherapy methods with the aid cells depletion is the risk of severe infections,
of genetically engineered CAR T cells in leukemia which needs to be taken into consideration when
patients broaden the scientists’ perspectives about evaluating the risks and benefits of this therapy.
possible options of exploiting FVIII-specific CAR Therefore, in the meantime, some degree of
Tregs to induce immuno-suppression in hemophilia humoral immunity can be retained in patients by the
A patients (72, 73). The presence of APCs is administration of intravenous immunoglobulin
essential for in vitro suppression, highlighting the (IVIG) (76).
importance of cell surface interactions. A New mechanistic insights in inducing immun-
controversial question regarding this issue is ologic tolerance
whether engineered CAR Treg can suppress B cells Hepatic gene therapy
directly or not. It is important to consider in vivo Blocking elements for co-stimulatory
stability, durability of suppression and safety prior molecules and inducing transient
to translation of the approach. immunosuppression are not the only approaches
B cell depletion therapy used to solve the inhibitor problem. Administration
In addition to T cell depletion, B cells have of viral expression vectors which convey
also been targeted due to their prominent role in hydrodynamic naked DNA can also be used as a
producing inhibitor antibodies (74). Anti-CD20 good strategy to form a tolerogenic environment in

Int J Mol Cell Med Winter 2020; Vol 9 No 1 40


Inhibitor formation and strategies for hemophilia A treatment

order to hamper the inhibitor formation. This tolerance, including lower innate immune response,
approach often causes rapid, but often evanescent inflammatory environment, and professional APCs
production of functional clotting factors. However, formation in the liver, hepatic gene transfer is an
vectors and transgenes are still non-self to the appropriate destination for different types of AAV
immune system, and provoke a protective immune vectors with the lowest danger of protective
response, especially in the type of innate immunity immune reactions (Figure3-C) (89-91).The large
(55, 61, 77). capacity of lentiviral vectors (LV) in gene transfer,
In order to treat hemophilia B, in vivo gene and high potential of liver-directed gene transfer in
transfer to the liver was successfully performed the formation of an effective immune tolerance has
using adeno-associated virus (AAV) vector, which proven this approach as a practical option to treat
is a genomic DNA covered by a protein capsid (78). hemophilia A patients through gene therapy. The
As the result was successful, several liver-directed innate immune system can effectively recognize LV
AAV vectors have been performed to find a through toll like receptor 7 and 9, and innate pro-
practical treatment for hemophilia A (79). AAV inflammatory cytokines, especially type I
vectors can be found as single-stranded DNA or interferon, are produced following this recognition.
self-complementary DNA genome, which are In addition, a myriad of APCs capture and present
usually obtained from non-pathogenic parvoviruses LV antigens on their surfaces, which recruits
(80). As these vectors do not contain viral coding adaptive immune responses through T cell
sequences, they can efficiently convey genomic activation and antibody production against the
information in the form of DNA in vivo. It is transgene product (92, 93).
important to know that the vector capacity is It is noteworthy that specific microRNA
restricted (~5 kb) so; unessential parts of the (miRNA) sequences should be established in gene
interested gene should be omitted (81). As the B transfer expression cassettes due to the different
domain of FVIII does not affect the proper activity possible profiles of miRNAs expression in various
of FVIII, B domain-deleted FVIII (BDD-FVIII) is tissues. Actually, specific miRNA sequences (miR-
usually packaged in the vector and also used in 142-3p) along with tissue-specific promoters
recombinant FVIII products (82, 83). considerably decreases the number of APCs with
To minimize the detrimental adaptive immune transgene expression on their surface (94, 95).
reactions, it is an effective way to use tissue- Despite the fact that transcriptional and post-
specific promoters. As these promoters usually translational engineering of the LV can result in
decrease the presentation of transgenic elements in lower hepatocytes expression of transgenic
the target tissue and by APCs, they can strikingly products, exploiting tissue-specific promoters with
diminish the unwanted activation of the immune specific miRNAs has demonstrated an effective
system along with keeping their effective gene hemophilia A and B phenotype development, and
expression. Accordingly, there are superb options to increased tolerance induction during treatment
be used in vectors for gene therapy (84-86). Liver (96, 97).
sinusoidal endothelial cells (LSECs) are Recently, Merlin et al. has tried to restrict the
predominantly responsible of FVIII secretion into expression of transgenic FVIII products to certain
circulation, and it makes the liver to be considered cell types including LSECs and myeloid cells. To
as a key target for gene therapy in patients suffering achieve transgene expression only in certain cell
from hemophilia A (87, 88). Due to the ideal types, cell-specific promoters such as endothelial-
characteristics of liver for the induction of immune specific promoter cadherin 5 type II, also known as

41 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

vascular endothelial cadherin (VEC) were widely an antigen have been studied for over 50 years, and
used as the cell-specific promoters in a lentiviral the promising outcomes showed immune anergy
vector (LV)-mediated gene therapy through which against several food allergens in human (79, 102,
the immune response was restrained in a mouse 103). In addition, this approach has been shown to
model. In addition, miRT-142.3p and miRT-122 be effective in decreasing autoimmune reactions in
were also used as selective miRNA target several autoimmune disorders such as experimental
sequences (miRTs) to improve specificity and autoimmune encephalomyelitis, diabetes, and
achieve a silent expression of FVIII by interested rheumatoid arthritis in animal models (104-106).
cells comprising hematopoietic cells, endothelial This also applied to induce a desirable
cells, and hepatocytes (98). Moreover, the results of tolerance in hemophilia A patient. A study on
their studies on LV construct containing the mucosal tolerance performed by Rawle et al.
myeloid-specific CD11b promoter, which demonstrated that oral or nasal administration of the
recognizes miR-126, a miRNA that is immunogenic FVIII C2 domain (FVIII-C2) in mice
predominantly observed in endothelial cells and is able to relatively induce tolerance against this
plasmacytoid dendritic cells (pDCs), showed that part even after encountering full-length FVIII
this specific promoter restricts the expression of (107).
transgenes to the macrophages and conventional In spite of the partial success in inducing
dendritic cells (cDCs) in the liver and spleen. Mice tolerance, there was still the lack of a cost-effective
that received the vector intravenously showed only technology to produce high amounts of FVIII
5–6% of FVIII normal activity. This amount of antigens, and protect it against digestive enzymes in
activity lasted up to 1 year after vector delivery. the stomach, which guaranteed proper delivery of
Although the FVIII normal activity was not the administered antigen to the immune system
considerably high in injected mice, lack of antibody agents in the gut. Recent achievements in producing
formation even following recombinant human genetically engineered plants have helped to
FVIII use was promising (99). Therefore, achieve human therapeutic proteins,
expression at the physiologic site of synthesis biopharmaceuticals, and edible vaccines by using
(liver) can enhance efficacy and safety, and may the chloroplast of crop plants (108, 109).
help overcome the anti-FVIII immune response Furthermore, previous study showed that
problem, resulting in long-term correction of administration of frozen tobacco leaves in which C2
hemophilia A. domain or the heavy chain of human BDD-FVIII
Oral tolerance induction were expressed. The result indicated a considerable
Most of the food antigens usually do not reduction in inhibitor formation within two
provoke the immune reaction, and induce immune different strains of hemophilia A mice. Biological
tolerance (100, 101). Hence, target antigens can be encapsulated antigens were orally administered
administered orally to promote tolerogenic immune twice per week and one month before starting
response and be safe from immune recognition as traditional replacement therapy. It was shown that
dangerous agents. This approach omits the risk of not only the allergic reactions against FIX in
genetic manipulation for the host and is free from hemophilia B mice were abolished, but also pre-
an extensive use of immunosuppressive drugs or existing FVIII inhibitors increasingly were
expensive cell therapies. The systemic diminished in hemophilia A mice after using this
immunological unresponsiveness or hypo- method (110-112).
responsiveness resulted from oral administration of In fact, chloroplast genomic tools were developed

Int J Mol Cell Med Winter 2020; Vol 9 No 1 42


Inhibitor formation and strategies for hemophilia A treatment

in order to determine the ribosomal locations which controlled are coming into focus. All developments
give rise to optimization of codon usage (109, 113). in this area can lead to unleashing new approaches
The major advantage of this application is that to practically treat or prevent hemophilia A
genetically engineered plants can express proteins complications.
which contain the antigen of interest, and in large One of the promising strategies for the
amounts with a low price. This results in achieving treatment of genetic disorders in utero is maternal
therapeutic proteins potentially able to induce antigen transfer through which the antigen of
tolerance in oral administration (114). interest is expressed to the immature immune
Future perspectives in tolerogenic strategies for system of the fetus, and antigen-specific tolerance
hemophilia A treatment may happen in the absence of pre-existing
Different aspects of the inhibitor development antibodies with high probability (115). It was
in hemophilia A, and whether they might be shown that hemophilia A mice, which were intrave-

Table 1. Tolerance inducing protocols.


Tolerance protocol Mechanism of action Outcomes References
Blockade of co-stimulatory
pathways
CTLA4-immunoglobulin Blockade of the B7/CD28 Prevention of the inhibitor 50
(CTLA4-Ig) interaction formation in hemophilia A
mice
CTLA4-Ig + anti-CD40L Dual blockade of CD40/ Long-term tolerance to FVIII (51, 52)
CD40L and B7/ CD28 induction in F8-plasmid
pathways treated hemophilia A mice
T-cell depletion
FVIII plasmid + Anti-CD3 Transforming growth factor-β Long-term tolerance to FVIII (54)
levels and the generation of
adaptive FVIII-specific Tregs
Treg induction
IL-2/IL-2 mAb complexes Rapid expansion of Suppression of inhibitor (63-65)
CD4+CD25+FOXP3+ Treg formation to either FVIII
replacement therapy or
plasmid-mediated gene
therapy of FVIII
Chimeric antigen receptor FVIII-specific suppression by Suppression of the antibody (66)
(CAR) gene transfer FOXP3+ Treg formation in vitro and in vivo
in hemophilia A mice
B-cell depletion
Anti-CD20 (Rituximab) Depletion of the follicular B Remaining of the FVIII- (75)
cells and increasing the specific hypo responsive state
regulatory T cells in the spleen
Gene therapy
Lentiviral vector -mediated Restricting the expression of Exhibition of an average of 5– (98)
gene therapy included FVIII transgene to liver 6% of normal FVIII activity,
miRNA target sequences sinusoidal endothelial cells which is stable for up to 1 year
(miRTs) and myeloid cells after vector delivery
Oral tolerance induction
Lettuce encapsulated clotting Bioencapsulation and Acceleration of the decline of (109)
factor targeting of antigen to pre-existing FVIII inhibitors in
immune system, induction of hemophilia A mice
Tregs

43 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

-nously injected by Fc fusions of FVIII A2 and C2 Conclusion


domains could successfully transfer the antigen to In conclusion, many experimental investi-
their fetus via the neonatal Fc receptor. Besides, gations have been performed to evaluate the
after repeated administrations, an effective immune effectiveness of inducing tolerogenic responses to
tolerance was also observed in the offspring of overcome the main complication of hemophilia A
injected females in comparison with those that their patients, which is the anti FVIII inhibitor
mother hadnot been receiving antigen before (116, development (Table 1). Considering all the pros and
117). These new achievements in maternal antigen cons, these approaches follow common destinations
transfer broaden our perspective about other comprising cost-effectivity, durability, and time-
strategies to develop antigen-specific regulatory T efficiency of the treatment. Time will tell which of
cells obtained from thymus or periphery. Although these approaches may become the best clinical
it would be a good strategy, a large number of therapy in the future.
antigens will be needed to achieve successful Acknowledgments
antigen transfer to a fetus that limits its application. This research received no specific grant from
In replacement therapy, development of Fc- any funding agency in the public, commercial, or
conjugated FVIII has taken many efforts to produce not-for-profit sectors.
more efficient FVIII with a greater half-life (118). Conflict of Interest
Due to the importance of these molecules in the There is no conflict of interest to declare.
development of the immune tolerance, many
attempts have focused on tolerogenic properties of References
Fc sequences (118). In the era of novel therapies, 1. Soucie JM, Evatt B, Jackson D. Occurrence of hemophilia in
such as Emicizumab, a humanized monoclonal the United States. Am J Hematol 1998;59:288-94.
bispecific antibody that mimics the role of FVIII in 2. Weyand AC, Pipe SW. New therapies for hemophilia. Blood
the coagulation cascade and promotes thrombin 2019;133:389-98.
generation, the management of hemophilia A 3. Gouw SC, van den Berg HM, Fischer K, et al. Intensity of
patients who experience anaphylaxis to replacement factor VIII treatment and inhibitor development in children with
therapy is becoming easier and may obviate the severe hemophilia A: the RODIN study. Blood 2013;121:
need for ITI. 4046-55.
During the last decade, a nanoparticle 4. Wight J, Paisley S. The epidemiology of inhibitors in
approach has been developed to provide an haemophilia A: a systematic review. Haemophilia 2003;9:
alternative to engineer cellular therapies for 418-35.
tolerance; these kinds of nanoparticles have also 5. Margaglione M, Intrieri M. Genetic Risk Factors and Inhibitor

been used for drug delivery and vaccine Development in Hemophilia: What Is Known and Searching for
development (119). Such nanoparticles can contain the Unknown. Semin Thromb Hemost 2018;44:509-16.
drugs, and are delivered with the target antigen and 6. Hosseini S, Arabi S, Yari F, et al. HLA-DRB1* 01: 01, but
presumably are taken up by tolerogenic APCs, and not HLA-DRB1: 1503 or HLA-DRB1* 11, is associated with
induce Tregs (120, 121). The use of Rapamycin- decreased inhibitor risk in Iranian hemophilia A patients.
containing nanoparticles for tolerance was Transfus Apher Sci 2019;58:669-73.
successfully used by Zhang et al. for FVIII (122). 7. Coppola A, Di Minno MN, Santagostino E. Optimizing
Several other approaches, in addition to the above management of immune tolerance induction in patients with
strategies, are being developed to induce tolerance severe haemophilia A and inhibitors: towards evidence-based

to FVIII. approaches. Br J Haematol 2010;150:515-28.

Int J Mol Cell Med Winter 2020; Vol 9 No 1 44


Inhibitor formation and strategies for hemophilia A treatment

8. Jimenez-Yuste V, Kruse-Jarres R, Rangarajan S, et al. The 21. Batsuli G, Deng W, Healey JF, et al. High-affinity,
evolving role and future relevance of plasma-derived therapies in noninhibitory pathogenic C1 domain antibodies are present in

the management of bleeding disorders. Thromb Haemost patients with hemophilia A and inhibitors. Blood
2016;116 Suppl 1:S1. 2016;128:2055-67.
9. Carpenter SL, Khair K, Gringeri A, et al. Prophylactic 22. Minno GD, Santagostino E, Pratt K, et al. New predictive

bypassing agent use before and during immune tolerance approaches for ITI treatment. Haemophilia 2014;20 Suppl 6:
induction in patients with haemophilia A and inhibitors to FVIII. 27-43.
Haemophilia 2018;24:570-7. 23. Ibrahim UA, Ahmed SG. Determinants and modifiers of

10. Napolitano M, Kessler CM. Hemophilia A and hemophilia bleeding phenotypes in haemophilia-A: General and tropical
B. Consultative Hemostasis and Thrombosis: Elsevier; 2019. p. perspectives. Egypt J Med Hum Genet 2018;19:171-8.
39-58. 24. Fulcher CA, de Graaf Mahoney S, Roberts JR, et al.

11. Sherman A, Biswas M, Herzog RW. Tolerance induction in Localization of human factor FVIII inhibitor epitopes to two
hemophilia: innovation and accomplishments. Curr Opin polypeptide fragments. Proc Natl Acad Sci U S A 1985;82:
Hematol 2018;25:365-72. 7728-32.
12. Wight J, Paisley S, Knight C. Immune tolerance induction in 25. Lacroix-Desmazes S, Bayry J, Misra N, et al. The prevalence
patients with haemophilia A with inhibitors: a systematic review. of proteolytic antibodies against factor VIII in hemophilia A. N
Haemophilia 2003;9:436-63. Engl J Med 2002;346:662-7.
13. Valentino LA, Kempton CL, Kruse-Jarres R, et al. US 26. Ghosh K, Shetty S. Immune response to FVIII in hemophilia
Guidelines for immune tolerance induction in patients with A: an overview of risk factors. Clin Rev Allergy Immunol
haemophilia a and inhibitors. Haemophilia 2015;21:559-67. 2009;37:58-66.
14. Rocino A, Cortesi PA, Scalone L, et al. Immune tolerance 27. van Helden PM, Van Haren SD, Fijnvandraat K, et al. Factor
induction in patients with haemophilia a and inhibitors: VIII-specific B cell responses in haemophilia A patients with
effectiveness and cost analysis in an European Cohort (The inhibitors. Haemophilia 2010;16:35-43.
ITER Study). Haemophilia 2016;22:96-102. 28. Scott DW, Pratt KP, Miao CH. Progress toward inducing
15. Gitschier J, Wood WI, Goralka TM, et al. Characterization of immunologic tolerance to factor VIII. Blood 2013;121:4449-56.
the human factor VIII gene. Nature 1984;312:326-30. 29. Lacroix-Desmazes S, Navarrete AM, Andre S, et al.
16. Randi AM, Smith KE, Castaman G. von Willebrand factor Dynamics of factor VIII interactions determine its immunologic
regulation of blood vessel formation. Blood 2018;132:132-40. fate in hemophilia A. Blood 2008;112:240-9.
17. Kaufman RJ, Pipe SW. Regulation of factor VIII expression 30. Lovgren KM, Sondergaard H, Skov S, et al. Non-genetic risk
and activity by von Willebrand factor. Thromb Haemost factors in haemophilia A inhibitor management - the danger
1999;82:201-8. theory and the use of animal models. Haemophilia 2016;22:
18. Metzner H, Schulte S, Weimer T. Covalent complex of von 657-66.

Willebrand Factor and factor VIII, compositions, and uses 31. Tieu P, Chan A, Matino D. Molecular Mechanisms of
relating thereto. Google Patents; 2018. Inhibitor Development in Hemophilia. Mediterr J Hematol Infect
19. Susree M, Anand M. Importance of Initial Concentration of Dis 2020;12:e2020001.
Factor VIII in a Mechanistic Model of In Vitro Coagulation. 32. Varthaman A, Lacroix-Desmazes S. Pathogenic immune
Acta Biotheor 2018;66:201-12. response to therapeutic factor VIII: exacerbated response or
20. Toole JJ, Pittman DD, Orr EC, et al. A large region failed induction of tolerance? Haematologica 2019;104:236-44.
(approximately equal to 95 kDa) of human factor VIII is 33. Lacroix-Desmazes S, Voorberg J, Lillicrap D, et al.
dispensable for in vitro procoagulant activity. Proc Natl Acad Tolerating Factor VIII: Recent Progress. Front Immunol
Sci U S A 1986;83:5939-42. 2019;10:2991.

45 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

34. Scott DW, Pratt KP. Factor VIII: Perspectives on splenic germinal centers and anti‐factor VIII inhibitor titer. Eur J
Immunogenicity and Tolerogenic Strategies. Front Immunol Immunol 2000;30:2548-54.

2019;10:3078. 49. Nadler SG, Tamura JK, Price L, et al. Methods of treating
35. Wroblewska A, Reipert BM, Pratt KP, et al. Dangerous immune diseases by administering antibody polypeptides that
liaisons: how the immune system deals with factor VIII. J specifically bind CD40L. Google Patents; 2019.

Thromb Haemost 2013;11:47-55. 50. Scott DW. Inhibitors - cellular aspects and novel approaches
36. Peyvandi F, Mannucci PM, Garagiola I, et al. A Randomized for tolerance. Haemophilia 2014;20 Suppl 4:80-6.
Trial of Factor VIII and Neutralizing Antibodies in Hemophilia 51. Rogers GL, Herzog RW. Gene therapy for hemophilia. Front

A. N Engl J Med 2016;374:2054-64. Biosci (Landmark Ed) 2015;20:556-603.


37. Reipert BM. B-cell memory against factor VIII. Cell 52. Miao CH, Ye P, Thompson AR, et al. Immunomodulation of
Immunol 2016;301:49-58. transgene responses following naked DNA transfer of human

38. Batsuli G, Ito J, Mercer R, et al. Anti-C1 domain antibodies factor VIII into hemophilia A mice. Blood 2006;108:19-27.
that accelerate factor VIII clearance contribute to antibody 53. Lai JD, Georgescu MT, Hough C, et al. To clear or to fear:
pathogenicity in a murine hemophilia A model. J Thromb An innate perspective on factor VIII immunity. Cell Immunol
Haemost 2018;16:1779-88. 2016;301:82-9.
39. Qian J, Collins M, Sharpe AH, et al. Prevention and 54. Peng B, Ye P, Rawlings DJ, et al. Anti-CD3 antibodies
treatment of factor VIII inhibitors in murine hemophilia A. modulate anti–factor VIII immune responses in hemophilia A
Blood 2000;95:1324-9. mice after factor VIII plasmid-mediated gene therapy. Blood
40. Krishnamoorthy S, Liu T, Drager D, et al. Recombinant 2009;114:4373-82.
factor VIII Fc (rFVIIIFc) fusion protein reduces immunogenicity 55. Herzog RW, Kuteyeva V, Saboungi R, et al. Reprogrammed
and induces tolerance in hemophilia A mice. Cell Immunol CD4(+) T Cells That Express FoxP3(+) Control Inhibitory
2016;301:30-9. Antibody Formation in Hemophilia A Mice. Front Immunol
41. Waters B, Lillicrap D. The molecular mechanisms of 2019;10:274.
immunomodulation and tolerance induction to factor VIII. J 56. Wang X, Terhorst C, Herzog RW. In vivo induction of
Thromb Haemost 2009;7:1446-56. regulatory T cells for immune tolerance in hemophilia. Cell
42. Kreuz W. The role of VWF for the success of immune Immunol 2016;301:18-29.
tolerance induction. Thromb Res 2008;122:S7-S12. 57. Clemente-Casares X, Blanco J, Ambalavanan P, et al.
43. Koeberl DD. Immunomodulating gene therapy. Google Expanding antigen-specific regulatory networks to treat
Patents; 2015. autoimmunity. Nature 2016;530:434-40.
44. Lai JD, Lillicrap D. Factor VIII inhibitors: advances in basic 58. Dobrzynski E, Fitzgerald JC, Cao O, et al. Prevention of
and translational science. Int J Lab Hematol 2017;39:6-13. cytotoxic T lymphocyte responses to factor IX-expressing
45. De Groot AS, Moise L, McMurry JA, et al. Activation of hepatocytes by gene transfer-induced regulatory T cells. Proc

natural regulatory T cells by IgG Fc-derived peptide Natl Acad Sci U S A 2006;103:4592-7.
"Tregitopes". Blood 2008;112:3303-11. 59. Mingozzi F, Hasbrouck NC, Basner-Tschakarjan E, et al.
46. Algiman M, Dietrich G, Nydegger UE, et al. Natural Modulation of tolerance to the transgene product in a nonhuman
antibodies to factor VIII (anti-hemophilic factor) in healthy primate model of AAV-mediated gene transfer to liver. Blood
individuals. Proc Natl Acad Sci U S A 1992;89:3795-9. 2007;110:2334-41.
47. Hausl C, Ahmad RU, Sasgary M, et al. High-dose factor VIII 60. Cao O, Dobrzynski E, Wang L, et al. Induction and role of
inhibits factor VIII-specific memory B cells in hemophilia A regulatory CD4+ CD25+ T cells in tolerance to the transgene
with factor VIII inhibitors. Blood 2005;106:3415-22. product following hepatic in vivo gene transfer. Blood
48. Qian J, Burkly LC, Smith EP, et al. Role of CD154 in the 2007;110:1132-40.

secondary immune response: the reduction of pre‐existing 61. Ohkura N, Kitagawa Y, Sakaguchi S. Development and

Int J Mol Cell Med Winter 2020; Vol 9 No 1 46


Inhibitor formation and strategies for hemophilia A treatment

maintenance of regulatory T cells. Immunity 2013;38:414-23. 2014;1.


62. Nayak S, Cao O, Hoffman BE, et al. Prophylactic immune 75. Zhang AH, Skupsky J, Scott DW. Effect of B-cell depletion

tolerance induced by changing the ratio of antigen-specific using anti-CD20 therapy on inhibitory antibody formation to
effector to regulatory T cells. J Thromb Haemost 2009;7: human FVIII in hemophilia A mice. Blood 2011;117:2223-6.
1523-32. 76. Biswas M, Rogers GL, Sherman A, et al. Combination

63. Webster KE, Walters S, Kohler RE, et al. In vivo expansion therapy for inhibitor reversal in haemophilia A using monoclonal
of T reg cells with IL-2–mAb complexes: induction of resistance anti-CD20 and rapamycin. Thromb Haemost 2017;117:33-43.
to EAE and long-term acceptance of islet allografts without 77. Doshi BS, Arruda VR. Gene therapy for hemophilia: what

immunosuppression. J Exp Med 2009;206:751-60. does the future hold? Ther Adv Hematol 2018;9:273-93.
64. Liu CL, Ye P, Yen BC, et al. In vivo expansion of regulatory 78. Nathwani AC, Reiss UM, Tuddenham EG, et al. Long-term
T cells with IL-2/IL-2 mAb complexes prevents anti-factor VIII safety and efficacy of factor IX gene therapy in hemophilia B. N

immune responses in hemophilia A mice treated with factor VIII Engl J Med 2014;371:1994-2004.
plasmid-mediated gene therapy. Mol Ther 2011;19:1511-20. 79. Sherman A, Biswas M, Herzog RW. Innovative Approaches
65. Liu CL, Ye P, Lin J, et al. Long-term tolerance to factor VIII for Immune Tolerance to Factor VIII in the Treatment of
is achieved by administration of interleukin-2/interleukin-2 Hemophilia A. Front Immunol 2017;8:1604.
monoclonal antibody complexes and low dosages of factor VIII. 80. Annoni A, Gregori S, Naldini L, et al. Modulation of
J Thromb Haemost 2014;12:921-31. immune responses in lentiviral vector-mediated gene transfer.
66. Kim YC, Zhang AH, Su Y, et al. Engineered antigen-specific Cell Immunol 2019;342:103802.
human regulatory T cells: immunosuppression of FVIII-specific 81. Pickar AK, Gersbach CA. Gene therapies for hemophilia hit
T- and B-cell responses. Blood 2015;125:1107-15. the mark in clinical trials. Nat Med 2018;24:121-2.
67. Wang X, Riviere I. Clinical manufacturing of CAR T cells: 82. Nathwani A, McIntosh J, Tuddenham E, et al. Methods of
foundation of a promising therapy. Mol Ther Oncolytics delivering factor VIII encoding nucleic acid sequences. Google
2016;3:16015. Patents; 2018.
68. Levine BL, Miskin J, Wonnacott K, et al. Global 83. Doering CB, Denning G, Shields JE, et al. Preclinical
Manufacturing of CAR T Cell Therapy. Mol Ther Meth Clin Development of a Hematopoietic Stem and Progenitor Cell
Dev 2017;4:92-101. Bioengineered Factor VIII Lentiviral Vector Gene Therapy for
69. Gomes-Silva D, Ramos CA. Cancer Immunotherapy Using Hemophilia A. Hum Gene Ther 2018;29:1183-201.
CAR-T Cells: From the Research Bench to the Assembly Line. 84. Vetrini F, Ng P. Gene therapy with helper-dependent
Biotechnol J 2018;13. adenoviral vectors: current advances and future perspectives.
70. Pan D, Buning H, Ling C. Rational Design of Gene Therapy Viruses 2010;2:1886-917.
Vectors. Mol Ther Methods Clin Dev 2019;12:246-7. 85. Falkner F-G, Horling F, Lengler J, et al. Viral vectors
71. Yoon J, Schmidt A, Zhang AH, et al. FVIII-specific human encoding recombinant FVIII variants with increased expression

chimeric antigen receptor T-regulatory cells suppress T- and B- for gene therapy of hemophilia A. Google Patents; 2019.
cell responses to FVIII. Blood 2017;129:238-45. 86. Matsui H, Shibata M, Brown B, et al. A murine model for
72. Parvathaneni K, Abdeladhim M, Pratt KP, et al. Hemophilia induction of long-term immunologic tolerance to factor VIII
A inhibitor treatment: the promise of engineered T-cell therapy. does not require persistent detectable levels of plasma factor VIII
Transl Res 2017;187:44-52. and involves contributions from Foxp3+ T regulatory cells.
73. Bluestone JA, Tang Q. Treg cells-the next frontier of cell Blood 2009;114:677-85.
therapy. Science 2018;362:154-5. 87. Arruda VR. The search for the origin of factor VIII synthesis
74. Corti M, Elder M, Falk D, et al. B-Cell Depletion is and its impact on therapeutic strategies for hemophilia A.
Protective Against Anti-AAV Capsid Immune Response: A Haematologica 2015;100:849-50.

Human Subject Case Study. Mol Ther Methods Clin Dev 88. Yadav N, Kanjirakkuzhiyil S, Ramakrishnan M, et al. Factor

47 Int J Mol Cell Med Winter 2020; Vol 9 No 1


Khalilian SH et al.

VIII can be synthesized in hemophilia A mice liver by bone 102. Andorf S, Purington N, Block WM, et al. Anti-IgE
marrow progenitor cell-derived hepatocytes and sinusoidal treatment with oral immunotherapy in multifood allergic

endothelial cells. Stem Cells Dev 2012;21:110-20. participants: a double-blind, randomised, controlled trial. Lancet
89. Sack BK, Herzog RW, Terhorst C, et al. Development of Gastroenterol Hepatol 2018;3:85-94.
Gene Transfer for Induction of Antigen-specific Tolerance. Mol 103. Chipps BE, Ciaccio CE, Rosén K, et al. Real-world

Ther Methods Clin Dev 2014;1:14013. attitudes among allergists/immunologists regarding oral
90. Mingozzi F, Liu YL, Dobrzynski E, et al. Induction of immunotherapy and preferred terminology. J Allergy Clin
immune tolerance to coagulation factor IX antigen by in vivo Immunol Pract 2019;7:721-3. e9.

hepatic gene transfer. J Clin Invest 2003;111:1347-56. 104. Weiner MD, Howard L. Oral tolerance for the treatment of
91. Kumar SR, Markusic DM, Biswas M, et al. Clinical autoimmune diseases. Annu Rev Med 1997;48:341-51.
development of gene therapy: results and lessons from recent 105. Weiner HL, da Cunha AP, Quintana F, et al. Oral tolerance.

successes. Mol Ther Methods Clin Dev 2016;3:16034. Immunol Rev 2011;241:241-59.
92. Agudo J, Ruzo A, Kitur K, et al. A TLR and non-TLR 106. Buerth C, Mausberg AK, Heininger MK, et al. Oral
mediated innate response to lentiviruses restricts hepatocyte Tolerance Induction in Experimental Autoimmune
entry and can be ameliorated by pharmacological blockade. Mol Encephalomyelitis with Candida utilis Expressing the
Ther 2012;20:2257-67. Immunogenic MOG35-55 Peptide. PLoS One 2016;11:
93. Alfranca A, Campanero MR, Redondo JM. New Methods for e0155082.
Disease Modeling Using Lentiviral Vectors. Trends Mol Med 107. Rawle FE, Pratt KP, Labelle A, et al. Induction of partial
2018;24:825-37. immune tolerance to factor VIII through prior mucosal exposure
94. Brown BD, Cantore A, Annoni A, et al. A microRNA- to the factor VIII C2 domain. J Thromb Haemost 2006;4:2172-9.
regulated lentiviral vector mediates stable correction of 108. Daniell H. Expression of human interferon in transgenic
hemophilia B mice. Blood 2007;110:4144-52. chloroplasts. Google Patents; 2017.
95. Cantore A, Ranzani M, Bartholomae CC, et al. Liver- 109. Kwon KC, Sherman A, Chang WJ, et al. Expression and
directed lentiviral gene therapy in a dog model of hemophilia B. assembly of largest foreign protein in chloroplasts: oral delivery
Sci Transl Med 2015;7:277ra28. of human FVIII made in lettuce chloroplasts robustly suppresses
96. Borsotti C, Follenzi A. New technologies in gene therapy for inhibitor formation in haemophilia A mice. Plant Biotechnol J
inducing immune tolerance in hemophilia A. Expert Rev Clin 2018;16:1148-60.
Immunol 2018;14:1013-9. 110. Sherman A, Su J, Lin S, et al. Suppression of inhibitor
97. Jobson J, Brown BD. Micromanaging Tolerance in formation against FVIII in a murine model of hemophilia A by
Hemophilia A Gene Therapy. Mol Ther 2017;25:1739-40. oral delivery of antigens bioencapsulated in plant cells. Blood
98. Merlin S, Cannizzo ES, Borroni E, et al. A Novel Platform 2014;124:1659-68.
for Immune Tolerance Induction in Hemophilia A Mice. Mol 111. Wang X, Su J, Sherman A, et al. Plant-based oral tolerance

Ther 2017;25:1815-30. to hemophilia therapy employs a complex immune regulatory


99. Agudo J, Ruzo A, Tung N, et al. The miR-126-VEGFR2 axis response including LAP+CD4+ T cells. Blood 2015;125:
controls the innate response to pathogen-associated nucleic 2418-27.
acids. Nat Immunol 2014;15:54-62. 112. Daniell H, Roland H. Administration of plant expressed
100. Wang X, Sherman A, Liao G, et al. Mechanism of oral oral tolerance agents. Google Patents; 2018.
tolerance induction to therapeutic proteins. Adv Drug Deliv Rev 113. Kwon KC, Chan HT, Leon IR, et al. Codon Optimization to
2013;65:759-73. Enhance Expression Yields Insights into Chloroplast
101. Hussey Freeland DM, Fan-Minogue H, Spergel JM, et al. Translation. Plant Physiol 2016;172:62-77.
Advances in food allergy oral immunotherapy: toward tolerance. 114. Kwon KC, Daniell H. Low-cost oral delivery of protein

Curr Opin Immunol 2016;42:119-23. drugs bioencapsulated in plant cells. Plant Biotechnol J 2015:13:

Int J Mol Cell Med Winter 2020; Vol 9 No 1 48


Inhibitor formation and strategies for hemophilia A treatment

1017-22. encapsulated antigen and exhibit strong local immune activation


115. Sagar R, Götherström C, David AL, et al. Fetal stem cell without inducing systemic cytokine release. Vaccine

transplantation and gene therapy. Best Pract Res Clin Obstet 2014;32:2882-95.
Gynaecol 2019. 120. Maldonado RA, LaMothe RA, Ferrari JD, et al. Polymeric
116. Jennewein MF, Abu-Raya B, Jiang Y, et al. Transfer of synthetic nanoparticles for the induction of antigen-specific

maternal immunity and programming of the newborn immune immunological tolerance. Proc Natl Acad Sci U S A
system. Semin Immunopathol 2017;39:605-13. 2015;112:E156-65.
117. Gupta N, Culina S, Meslier Y, et al. Regulation of immune 121. Getts DR, Shea LD, Miller SD, et al. Harnessing

responses to protein therapeutics by transplacental induction of T nanoparticles for immune modulation. Trends Immunol
cell tolerance. Sci Transl Med 2015;7:275ra21. 2015;36:419-27.
118. Carcao M, Shapiro A, Staber JM, et al. Recombinant factor 122. Zhang AH, Rossi RJ, Yoon J, et al. Tolerogenic

VIII Fc fusion protein for immune tolerance induction in patients nanoparticles to induce immunologic tolerance: Prevention and
with severe haemophilia A with inhibitors-A retrospective reversal of FVIII inhibitor formation. Cell Immunol
analysis. Haemophilia 2018;24:245-52. 2016;301:74-81.
119. Ilyinskii PO, Roy CJ, O'Neil CP, et al. Adjuvant-carrying
synthetic vaccine particles augment the immune response to

49 Int J Mol Cell Med Winter 2020; Vol 9 No 1

You might also like