Skip to main content

REVIEW article

Front. Immunol., 13 April 2021
Sec. Vaccines and Molecular Therapeutics
This article is part of the Research Topic AAV Gene Therapy: Immunology and Immunotherapeutics View all 14 articles

T Cell-Mediated Immune Responses to AAV and AAV Vectors

  • Wistar Institute, Philadelphia, PA, United States

Adeno-associated virus (AAV)-mediated gene transfer has benefited patients with inherited diseases, such as hemophilia B, by achieving long-term expression of the therapeutic transgene. Nevertheless, challenges remain due to rejection of AAV-transduced cells, which in some, but not all, patients can be prevented by immunosuppression. It is assumed that CD8+ T cells induced by natural infections with AAVs are recalled by the AAV vector’s capsid and upon activation eliminate cells expressing the degraded capsid antigens. Alternatively, it is feasible that AAV vectors, especially if given at high doses, induce de novo capsid- or transgene product-specific T cell responses. This chapter discusses CD8+ T cell responses to AAV infections and AAV gene transfer and avenues to prevent their activation or block their effector functions.

Introduction

The goal of gene therapy is to permanently replace a missing or faulty gene and thereby through sustained production of the transgene product achieve a functional cure. Various methods have been explored to insert genes in situ into specific cells (1, 2). One of the most promising gene transfer vectors are AAV vectors, which in initial preclinical studies achieved sustained expression of their transgene product in mice (3), dogs (4), and nonhuman primates (5) without any overt serious adverse events. In humans clinical trials targeting Leber’s congenital amaurosis, a congenital form of blindness, by small doses of AAV injected into the subretinal space reported long-term improvement of vision (6, 7). In contrast, the first clinical trial for hepatic AAV-mediated transfer of factor (F)IX for correction of hemophilia B accomplished initial increases in F.IX levels, which were followed a few weeks later by a subclinical transaminitis and loss of F.IX (8). Additional studies showed that patients developed concomitantly with rises in liver enzymes circulating CD8+ T cells to AAV capsid antigens (9). This led to the still valid but nevertheless unproven hypothesis that patients had AAV-capsid-specific memory CD8+ T cells, which were reactivated by the gene transfer and then eliminated the vector-transduced hepatocytes (10).

This opened a slurry of pre-clinical experiments that aimed to recapitulate the findings of the clinical trial. Although the animal experiments allowed the field to gain valuable knowledge of the intricacies of anti-AAV capsid T and B cell responses (1113), in the end the studies confirmed what we have known for long – mice are not humans (14) and neither mice nor larger animals are overly informative about the presumably immune-mediated rejection of AAV-transduced cells.

Clinical AAV-mediated gene transfer trials by reducing vector doses and using various immunosuppressive regimens at least in part overcame immunological barriers and achieved treatment benefits or even cures for their patients (15, 16). Nevertheless, transfer of genes with high doses of AAV remains a crapshoot especially in 2020/21 during a global pandemic with a potentially fatal virus that is especially dangerous for immunocompromised humans (17). Immune responses to AAV gene transfer are complex involving both the innate and adaptive immune systems. Here we discuss what is known from pre-clinical models as well as clinical trials about CD8+ T cells to AAV gene transfer.

AAV Virus and Immune Responses to Natural Infections

AAVs are single-stranded DNA viruses of the parvovirus family. As dependoviruses they only replicate in presence of a helper virus such as an adenovirus. AAVs do not cause any known disease. The ~4,700 base pair long AAV genome, which is flanked by inverse terminal repeats (ITRs), has two open reading frames, one for rep proteins needed for viral replication, and the other for the capsid proteins vp1, vp2 and vp3, which are produced by differential splicing and therefore only differ in their N-terminus (18). Capsid proteins distinguish serotypes of AAV. Thus far 12 human serotypes of AAV have been identified (19). They differ in their tropism (20) and in the prevalence, with which they circulate in humans (21). AAV genomes persist mainly episomally in the nucleus of infected cells although they can integrate into a specific site of human chromosome 19 (22).

Humans, who become naturally infected with AAVs, mount adaptive immune responses, which presumably are in part driven by innate responses to the helper virus (23). Prevalence rates of neutralizing antibodies to different serotypes of AAVs, which serve as indicators for previous infections, vary in part depending on age and country of residency (21, 2431). Some studies report strikingly different prevalence rates even when they tested similar populations. This likely reflects that AAV neutralization assays are not standardized and therefore differ in their sensitivity. Overall trends are similar. Prevalence rates of neutralizing antibodies to AAV increase with age and they are higher for AAV2 or AAV8 than for example AAV5 or AAV6.

T cell responses have been studied less well. We reported that about 50% of healthy human adults have detectable frequencies of circulating AAV capsid-specific CD8+ and/or CD4+ T cells when tested by intracellular cytokine staining (ICS); 50% of these CD8+ T cells belong to the central memory subsets and 25% each to the effector and effector memory subsets. AAV capsid-specific CD4+ T cells belong mainly to the central memory subset (32). Non-human primates tested by the same method showed that 5 out of 6 have AAV capsid-specific CD8+ T cells while 6/6 have CD4+ T cells of that specificity. In monkeys, CD8+ T cells are strongly biased towards effector cells (32). For these assays we used a peptide panel that reflected the capsid sequence of AAV2 but would like to point out that many of the T cell epitopes are highly conserved. Nevertheless, unlike in humans AAV-mediated gene transfer achieves long-lasting transgene product expression in nonhuman primates, which may reflect that their T cells potentially due to high levels of persisting AAVs are functionally exhausted (32). Overall, not only prevalence but also frequencies of AAV capsid-specific T cells are higher in non-human primates than in humans. Testing additional non-human primates by an ELISPOT assay, which is the assay that is primarily being used by gene therapists to monitor T cell responses to AAV capsid upon AAV-mediated gene transfer, showed lower prevalence rates of AAV capsid-specific T cells of ~50% (32).

Using a proliferation and cytokine secretion assays another group reported that peripheral blood mononuclear cells (PBMCs) of less than 10% of humans mount a response (29) although it should be pointed out that these assays lack sensitivity. Another group using ELISPOT assays as well as ICS showed with either assay that ~30% of health human adults respond to AAV1 capsid (33). A study using a very sensitive method based on pre-selection of AAV8-specific CD8+ T cells with a specific tetramer showed that all tested humans have circulating effector memory CD8+ T cells against AAV8 capsid (34). Human circulating AAV capsid-specific CD8+ T cells are functional, they secrete cytokines (32, 34) and lyse target cells expressing their cognate antigen (33). T cell epitopes are conserved between several AAV serotypes (9) and several studies reported no correlations between antibody and CD8+ T cell responses (32, 35). One study showed that peripheral blood mononuclear cells from AAV2 seronegative donors mount a robust IFN-g-secreting natural killer cell response to in vitro culture with an AAV capsid peptides while those from seropositive individuals showed activation of tumor necrosis factor-α producing CD8+ T cells (36).

Overall, these data demonstrate that AAV infections are highly prevalent and cause sustained immunological memory that can presumably be recalled upon re-infection or transfer of an AAV vector.

AAV Vectors

Production and purification methods for AAV vectors are well established (37). In AAV vectors the viral genes but for the inverted terminal repeats (ITRs) are replaced with an expression cassette for a therapeutic protein. A variety of promoters have been used, some of which drive ubiquitous expression while others are specific for selected cell types. For some applications, such as hemophilia B, a variant transgene with improved functions compared to protein encoded by the wild-type gene has been used to allow for dose-sparing.

AAV vectors are in general generated by triple transfection of a cell line, such as HEK 293 cells, which carry the E1 gene of adenovirus. One plasmid expresses additional adenoviral genes to promote AAV production. A second plasmid carries the AAV cap and rep genes. The AAV2 rep gene is used for most AAV vectors while the cap gene determines the serotype of the vector. The third plasmid carries the transgene expression cassette flanked by the ITRs, again most commonly of AAV2. Vectors are then released from the transfected cells and purified by various methods such as gradient centrifugation, column purification and others (38). The type of purification may affect levels of empty AAV particles within the preparation, which in turn can influence the induction of immune responses or reduce the inhibitory effects of AAV neutralizing antibodies.

AAV vectors can also be produced in the baculovirus expression system, which is more amenable for scale-up than mammalian expression systems (39). AAVs produced in mammalian cells or insect cells show differences in post-translational modification, genome methylation and levels and types of host cell contaminations which affect their immunogenicity and their performance in clinical trials (40, 41).

CD8+ T Cells

CD8+ T cells are uniquely capable to eliminate virus-infected or vector-transduced cells by direct lysis mediated by the release of perforin and granzyme. They also secrete anti-viral cytokines such as interferon (IFN)-γ. Activation of naïve CD8+ T cells, which reside in lymphatic tissues and circulate in blood, requires presentation of antigen-derived peptides bound to major histocompatibility complex (MHC) class I molecules by professional mature antigen-presenting cells. Peptides can be generated from de novo synthesized misfolded proteins that upon degradation by proteasomes are transported by the transporter associated with antigen presentation (TAP) into the endoplasmic reticulum (ER), where they bind to MHC class I molecules, which are then transported to the cell surface. This classical presentation pathway would apply to vector-encoded transgene products. Antigen-presenting cells are able to cross-present protein such as those of the AAV capsid that are taken up by pinocytosis, phagocytosis or endocytosis. In the so-called cytosolic pathway, the particles are degraded in phagosomes and peptides are released into the cytoplasm from where they can be transported into the ER; there they can bind to MHC molecules (42). In the vacuolic pathway, proteins are degraded within endosomes. They escape into the cytoplasm upon acidification of the endosomes or upon reactive oxygen species-mediated lipid peroxidation of endosomal membranes (43).

The antigenic peptides displayed by MHC class I antigens on the cell surface bind to T cell receptors, which triggers a signaling cascade that through the adaptor molecule zeta-chain-associated protein kinase 70 (ZAP70) induces activation of calcineurin leading to the activation of nuclear factor of activated T-cells (NFAT). Full activation of T cells furthermore requires interactions with co-stimulators most commonly CD80 and CD86 or CD40 on antigen-presenting cells, which interact with CD28 or CD40L on T cells, respectively. This amplifies T cell receptor signaling and through phosphoinositide 3-kinase (PI3K) induces the mechanistic target of rapamycin (mTOR)/protein kinase B (Akt) pathway which modifies the T cells’ metabolism to provide energy and building blocks for rapid proliferation. Dendritic cells, the main cell type that presents antigens to naïve T cells, are immature when they are released from bone marrow. At this stage they do not express co-stimulators and are therefore unable to activate an effector CD8+ T cell response but rather induce tolerance. Maturation of dendritic cells into profession antigen-presenting cells is driven by pathogen-associated molecular patterns (PAMPs), such as CpG motifs within the genome, which are common in bacteria and viruses but largely absent in mammalian cells. PAMPs interact with pathogen recognition receptors (PRR) such as Toll-like receptors (TLRs) and others expressed in different cellular compartments (44). Binding of a PAMP to a PRR causes activation of numerous pathways, such as the nuclear factor kappa B (NK-κB) and interferon regulatory factor (IRF)3 pathways. Induction of these pathways, which can also be activated by type I interferons (IFN) or members of the tumor necrosis factor (TNF) family, involves a number of molecules such as TIR domain containing adaptor protein (TIRAP), myeloid differentiation primary response 88 (MyD88), inhibitor of NF-κB kinase (IKK)-γ, or interleukin-1 receptor-associated kinase (IRAK)-4, all of which can be targeted by drugs to block inflammatory responses. Once NF-κB or IRF-3 are activated they induce pro-inflammatory cytokine responses, which initiate or increase production of molecules that are essential for antigen processing and presentation. Upon stimulation, CD8+ T cell proliferate very rapidly and then migrate to sites of infection where they assume effector functions. Recognition of foreign antigen is exquisitely sensitive and can be triggered by as few as 2-3 MHC-peptide complexes on the surface of a cell (45). CD8+ T cell differentiation requires help from CD4+ T cells (46) belonging to the T helper (Th)1 subset. Once the antigen has been removed most of the effector CD8+ T cells die, some will differentiate into memory cells, which can be recalled rapidly. Re-activation of memory CD8+ T cells does not require professional antigen presenting cells and is less dependent on co-stimulation. Effector CD8+ T cells can differentiate into different type of memory cells, i.e., effector, central memory or tissue resident memory T cells. Effector memory CD8+ T cells circulate. They do not proliferate extensively after re-exposure to antigen and can assume functions instantly. Over time in absence of antigen they differentiate into central memory cells or die. Central memory CD8+ T cells reside in lymphatic tissues. They do not exhibit functions. Upon reencounter of their antigen they proliferate vigorously before they assume effector functions; this may take several days. Central memory CD8+ T cells are maintained at steady numbers potentially throughout the lifespan of an individual. Tissue resident memory CD8+ T cells are also very long-lived but they remain at sites of previous infections. Upon local reinfection they can immediately release cytolytic enzymes and initiate an inflammatory reaction. If the antigen is not removed but continues to persist at high levels, T cells will differentiate towards exhaustion by gradually losing function, increasing expression of co-inhibitors such as programmed cell death protein 1(PD-1) and eventually undergoing apoptosis (47).

How does this apply to AAV vectors? To activate naïve CD8+ T cells one would expect that the vector would be phagocytosed by immature dendritic cells. PAMPs within the vector genome or on the capsid would interact with PRRs on or within the cells. This would trigger an inflammatory reaction and maturation of the dendritic cells, which would then migrate to draining lymph nodes. Within cells antigens encoded by the AAV vector would enter the classical presentation pathway while antigens of the AAV capsid would be processed and presented by either the cytosolic or vacuolic pathway. Dendritic cells presenting antigen bound to MHC class II molecules would activate specific CD4+ T cells which would then facilitate stimulation followed by expansion of CD8+ T cells by antigen displayed by MHC class I antigen. Subsequently T cells would migrate out of lymph nodes and circulate till they find their cognate antigen. Cells displaying this antigen would be killed rapidly within minutes and then the T cell would find its next target. This process would continue till all of the antigen is removed, which in case of AAV particles may take months. Memory CD8+ T cells generated in response to a natural infection can be stimulated by cells other than dendritic cells and depending on subset they can act immediately once they see their antigen displayed on MHC class I. If they belong to the tissue resident memory CD8+ T cell subset they may not even proliferate, which raises the question if screening for increases in circulating T cells is adequate to predict immune-mediated rejection of AAV-transduced cells. Due to higher numbers of precursors memory T cell responses are more potent and come up more rapidly.

It is assumed that the increases of circulating AAV capsid specific CD8+ T cells reflect recall of memory cells that had initially been activated by a natural infection. This may be the case for some patients but activation of naïve CD8+ T cells should not be ruled out. The very slow increases of AAV capsid-specific T cells in some AAV vector recipients would be more typical for primary than secondary responses. As stimulation requirements and thereby sensitivity to immunosuppressive drugs differ for naïve and memory CD8+ T cells further studies are needed to elucidate what T cell subsets respond to AAV gene transfer.

CD8+ T Cell Responses to AAV Vectors

Viral vectors can induce CD8+ T cell responses to their own antigens as well as to a transgene product. In the case of AAV vectors, which have been stripped of genes that encode AAV proteins, any effector T cell response to the viral proteins would be limited to the time frame till all the vector particles have been completely degraded. T cell responses to the transgene product on the other hand could continue till all antigen-producing cells have been removed or till immunosuppressive mechanisms such as T cell exhaustion or regulatory T cells turn off the T cells.

Initial studies reported that AAV vectors did not induced CD8+ T cell responses to the transgene product and this was attributed to lack of activation of innate responses, which resulted in immunological ignorance (48). Additional studies contradicted these results and reported that AAV vectors can induce transgene product-specific CD8+ T cell responses in experimental animals (4951). It was also shown that AAV vectors elicit albeit weak and transient innate responses that are largely driven by TLR9 activation through CpG motifs within the vector genome (52) or TLR2 activation by capsid components (53). Naturally immunogenic transgene products, such as antigens from another pathogens, induce depending on the vector’s serotype and it’s genome structure such robust immune responses that AAV vectors were explored as vaccine carriers (50, 51). The magnitude of the immune responses depends on vector dose, the AAV serotype, the transgene, the type of promoter, the target tissue and the vectors’ genome structure (54). Self-antigens, such as F. IX with point mutations, are non-immunogenic while the same mutant F.IX in a mouse with a genetic F.IX deletion induces cellular and humeral responses (55). Comparing vectors with single-stranded and double-stranded DNA genomes, showed that the latter are more immunogenic presumably by inducing more potent innate responses (56). Some studies showed that CD8+ T cell responses induced by an AAV vector-encoded transgene are defective in mice: T cells do not proliferate upon re-exposure to their antigen in vivo, they only produce low levels of cytokines and they fail to protect against a surrogate pathogen (57, 58). Others showed that the effectiveness of transgene product-specific hepatic CD8+ T cell responses is dependent on vector dose; intermediate doses of vector lead to a delayed CD8+ T cell response that eliminates antigen-producing hepatocytes. High doses of vector induce multiple immunosuppressive pathways that block induction of transgene product-specific CD8+ T cells (59).

AAV vectors can induce capsid-specific CD8+ T cell responses, especially if highly immunogenic T cell epitopes are incorporated into the capsid (60, 61). This process is likely driven by cross-presentation of capsid proteins and not only requires plasmacytoid and conventional dendritic cells but also help from CD4+ T cells (62, 63). Nevertheless, in mice specific CD8+ T cells induced by AAV gene transfer fail to eliminate AAV-transduced cells (64), which can be achieved by adaptive transfer of ex vivo expanded capsid-specific CD8+ T cells suggesting defects at the level of T cell differentiation in vivo that could be overcome in tissue culture (65).

Although they fail to reject AAV-transduced cells, mice have been useful to study the duration of capsid degradation, which in the end dictates how long gene transfer recipients are at risk to lose treatment benefit due to AAV capsid-specific CD8+ T cells. Experiments using proliferation of capsid-specific CD8+ T cells, which were adoptively transferred into AAV-injected mice as a read-out, showed that T cells proliferated in their hosts even if transferred 6 months after AAV injection, which is reflective of the very slow degradation of AAV capsid (66).

Clinical trials using different serotypes of AAV vectors over a large range of doses have been completed, are ongoing or planned for a number of diseases. For ocular diseases such as choroiderma, an X-linked form of progressive vision loss (67), achromatopsia or color blindness (68), X-linked retinitis pigmentosa (69), or Leber’s congenital amaurosis (70) AAV vectors encoding the therapeutic protein are injected at modest doses into the subretinal space, which similar to the central nervous system is an immunoprivileged site (71) that contains high levels of transforming growth factor (TGF)-ß and is shielded by a physical barrier from blood. In addition, parts of the eye, such as the ocular chamber, actively induce immune tolerance through a process called anterior chamber-associated immune deviation (ACAID)  (72). It should be noted though that this pathway does not induce systemic tolerance after subretinal injection of an AAV vector (73). While some trials for correction of ocular diseases reported stable transgene product expression for years without evidence for induction of T cell responses (74, 75), others observed stimulation of adaptive immune responses combined in some cases with loss of therapeutic benefits (76). Pre-clinical studies indicate that induction of immune responses to ocular injection of AAV vectors depends on vector dose, the promoter, route of application and the transgene (77), which may in part explain discrepancies of results.

Many AAV-mediated gene transfer trials focus on hemophilia where circulating coagulation factors offer an easy read-out for transgene product expression. Vectors that were or are being explored for hemophilia B include AAV8, AAV5, AAVrh10 or AAVs with genetically engineered capsids expressing either wild-type F.IX or the 5 time more potent F. IX Padua variant. Vectors either contain a single-stranded or self-complementary genome. They were or are given at doses ranging from 2x1011 to 2 x 1012 vg/kg achieving post-infusion levels of F.IX depending on the trial and the vector dose from ~1-40% of normal. Many trials use codon-optimized vectors to increase expression and/or vectors in which most CpG motifs are modified to minimize TLR9 activation. Some trials, such as one using AAV5 expressing the Padua variant of F.IX failed to observe any post-infusion transaminitis (78), which would be indicative of an anti-AAV immune response while others using the same vector reported transient increases in liver enzymes, which were not accompanied by detectible T cell responses (79). Using an AAV vector with a bioengineered capsid expressing F.IX Padua or a self-complementary AAV8 vector expressing wild-type F.IX other groups reported in some patients increases in transaminases combined with increases of circulating AAV capsid-specific T cells (80, 81). Data have been published for one hemophilia A trials, which used a baculovirus-derived AAV5 vector. The study reported increases in liver enzymes but failed to detect concomitant rises in capsid-specific circulating T cells (80). Circulating FVIII levels tended to decrease gradually by 2-3 years after gene transfer most likely reflecting hepatocyte turn-over rather than an immune-mediated rejection (16). A number of additional trials that are ongoing for correction of hemophilia A have reported therapeutic benefits (82) but not yet released potential problems with immune-mediated rejection.

Overall transfer to the liver, a unique microenvironment that favors immunosuppression (83), can induce in a dose-dependent manner capsid-specific CD8+ T cell responses, which have been implicated to eliminate AAV-transduced cells. Thus far transgene product-specific CD8+ T cells have not yet been observed upon hepatic transfer of AAV vectors. This may in part reflect that trials mainly enrolled patients with point mutations in their coagulation factors, which destroy their biological activity but fail to prevent induction of immunological tolerance. Most patients had also received factor replacement therapy, which would further promote tolerance. It would be expected that AAV vectors will induce F.VIII- or F.IX-specific T cell responses in patients with large deletion mutations although such responses might potentially be dampened or blocked by concomitant induction of regulatory T cells (59).

For treatment of other diseases AAV vectors are given into the muscle. For example, for treatment of Pompe disease, caused by glycogen storage in muscle and motor neurons due to lack of lysosomal alpha-glucosidase, an AAV9 vector was injected at a high dose into the diaphragm in children with progressive respiratory failure requiring ventilation and enzyme replacement therapy (84). T cell responses to the vector or transgene product were not detected. Several trials are exploring intramuscular injection of AAV vectors expressing dystrophin for treatment of Duchenne’s muscular dystrophy (85, 86) or a1-antitrypsin (AAT) deficiency (87, 88). An AAT trial using an AAV-1 vector reported that vector-derived AAT levels were sustained for several months although all treatment recipients developed T cell responses to the capsid proteins of AAV1 and 1 of the 9 patients had at one timepoint a positive T cell response to AAT. A follow-up study conducted about a year later showed sustained presence of the AAV genome in the injected muscle and a marked reduction in inflammatory cells in year 1 compared to months 3 biopsy samples. A substantial portion of the muscle infiltrating lymphocytes were regulatory T cells suggesting that they had suppressed vector-induced effector T cell responses and thereby prevented loss of AAV-transduced muscle cells (87, 88). A further follow-up study conducted 5 years after gene transfer detected Tregs and AAV capsid-specific CD8+ T cells within the injected muscle (89).

For AAV-mediated correction of Duchenne’s muscular dystrophy one trial reported that patients developed no or only very weak T cell responses to the viral capsid but instead some generated robust transgene product-specific CD8+ T cell responses (90, 91). AAV1-mediated transfer of the alpha-sarcoglycan gene for correction of limb-girdle muscular dystrophy reported a detectable AAV capsid-specific T cell response in 3 patients. In one patient this response came up very rapidly on day 2 after gene transfer and may have contributed to his loss of AAV-transduced muscle cells (92). Additional studies are needed to further determine the risk induction of T cells upon intramuscular injection of AAV vectors. It will be especially important to further elucidate the role of regulatory T cells in preventing immune-mediated destruction of AAV-transduced muscle cells.

AAV vectors are directly infused into the central nervous system to treat neurological diseases such as Alzheimer (93), Parkinson’s disease (9497), infantile neuronal ceroid lipofuscinosis (98), Canavan disease, a N-acetylaspartate storage disease of the brain caused by mutations of the aspartoacylase gene (99) and others. Studies did not assess AAV capsid or transgene product-specific T cell responses following AAV gene transfer. Some trials analyzed serum antibody responses to AAV and reported that they increased in some patients suggesting that enough vectors had leaked from the injection sites to trigger a peripheral B cell response.

Considering that the central nervous system is an immunoprivileged site that lacks the lymphatic structures needed for activation of immune responses it is unlikely that enough vector will leak into the periphery to activate AAV capsid or transgene-product-specific CD8+ T cells which could then cross the blood brain barriers and attack vector-transduced cells. Nevertheless, it would be prudent to monitor patients for AAV-induced T cell responses.

Intracoronary application of an AAV vector expressing SERCA2a to modulate calcium metabolism reported transient increases in AAV capsid-specific T cell frequencies in 1 out of 9 patients without any clinical consequences or further changes in blood chemistry (100). Intravenous application of an AAV9 vector expressing the survival motor neuron 1 gene for treatment of spinal muscular atrophy reported increases in liver enzymes in some of their patients (101). As T cell responses were not analyzed, the etiology of this observation remains uncertain.

As one would expect, AAV vectors induce adaptive immune responses especially if given at high doses to peripheral sites. It remains unclear how common T cells to AAV capsid or the encoded transgene product interfere with sustained therapeutic benefits as many trials still fail to test for T cell responses. Injections of even large doses of AAV does not inevitably lead to detectable T cell responses with the obvious caveat that T cell assays have sensitivity limits and may not reveal small responses. Furthermore, the activity of tissue resident memory CD8+ T cells may not be spotted by monitoring increases in circulating AAV capsid- or transgene product-specific T. cells. It will be important for gene therapist that base their therapeutics on AAV to further define factors that promote CD8+ T cell responses. Vector dose and route of administration clearly play roles. CpG content within the vector genome may affect responses as was shown in mice (98). The capsid itself may have an effect. Age can have an effect; younger people tend to mount better immune responses, but older people are more likely to have immunological memory to AAVs. The underlying disease especially if it causes local inflammatory reaction can affect treatment outcome. The HLA type of the gene therapy recipient will affect if and how many epitopes of the capsid can be recognized by T cells. The type of the transgene and its similarity to endogenous proteins will determine if the host is tolerant or responsive.

After the initial AAV gene transfer trials for hemophilia B indicated that AAV-induced CD8+ T cells may cause loss of cells producing F.IX (8), one of the next trials very carefully monitored serum F.IX and transaminase levels following AAV gene transfer. The two patients that received the highest vector dose showed by weeks 7 or 9 modest subclinical increases in aspartate aminotransferase and alanine aminotransferase and decreases in F.IX. They were treated for 9 or 4 weeks, respectively with prednisolone, which reduced transaminase levels and stopped further declines in F.IX (5). Steroids, such as prednisolone, are in general well tolerated if given for a short period of time. They are widely used in transplantation medicine, to treat auto-immune diseases or conditions caused by overwhelming inflammatory responses. They affect multiple aspects of T cell responses resulting in reduced cytokine production and decreased proliferation. Specifically, they dampen T cell activation by reducing phosphorylation of key signaling molecules of the T cell receptor (102). They induced production of anti-inflammatory cytokines and they promote proliferation of regulatory T cells (103). Although steroids are not suitable as a monotherapy to prevent transplant rejection they have been used successfully by Nathwani (5) and others (79, 104, 105) to prevent loss of AAV-transduced cells upon hepatic gene transfer. Nevertheless, in other trials steroids did not avert gradual loss of the transgene product (106108). The optimal regimen for steroid therapy remains to be established (109). While most trials carefully monitored serum enzyme levels and started steroid treatment once transaminases increased others gave steroids to all patients immediately or shortly after treatment. The former approach is not only cumbersome as it requires weekly testing but also carries the risk that treatment could be initiated too late. The latter approach has the disadvantage that patients, who would not mount immune-mediated rejection are treated unnecessarily. It also remains unclear how long steroids should be given and if this time frame depends on the type of vector or differs for each patient. As steroids have not always prevented loss of transgene-expressing cells additional immunosuppressives need to be explored. In a trial with an AAV1 vector for correction of lipoprotein lipase deficiency, a 12 week course of methylprednisolone together with cyclosporin and mycophenolate mofetil was started shortly before AAV transfer; the drugs did not prevent increases in AAV capsid-specific T cells, but T cells appeared to be functionally impaired and failed to achieve removal of vector-transduced muscle cells (110). Cyclosporin inhibits T cell activation by blocking signaling through NFAT transcription factors, which are regulators of CD8+ T cell functions (111) as well as the NF-κB and activator protein 1 (AP-1) pathways. Cyclosporin is also being explored in pre-clinical models for dampening of AAV capsid-specific neutralizing antibody responses (112). Nevertheless, cyclosporin depending of timing of antigen exposure versus drug treatment was shown in a cardiac allograft rat model to prevent activation of regulatory T cells (113), while in other systems cyclosporin augmented the activity of this immunosuppressive cell subset (112, 113). Mycophenolate mofetil inhibits inosine monophosphate dehydrogenase, the rate-limiting enzyme for synthesis of guanosine nucleotides which are essential for cell cycle progression of proliferating lymphocytes (114).

Other drugs that are being considered are TLR antagonists or drugs that target downstream molecules of PRR signaling to inhibit activation of the inflammatory responses that are essential to drive activation of naïve T cells (115). Such drugs could block numerous steps of PRR activation pathways such as TIRAP, MyD88, IKK-γ, NF-κB, or IRAK-4. Inflammatory responses could also be dampened by blocking TNF-α signaling through monoclonal antibodies such as Remicade, Humira, Cimzia, the receptor fusion protein Enbre or the small molecule inhibitor pentoxifylline (116). Drugs are also available to block type I IFN signaling by inhibiting IRF-3 or JAK1 which would affect T cell maturation (117). All of these drugs would impair primary T cell responses but may not block recall responses. Induction of CD4+ T cells could be inhibited by preventing antigen presentation through the MHC class II pathway through chloroquine, which prevents acidification of endosomes (118), cyclosporin A and tacrolimus, which both inhibit calcineurin (119) or blockers of the lysosomal protease cathepsin S, such as morpholinurea-leucine-homophenylalanine-vinylsulfone-phenyl (120). Some of the drugs such as cyclosporin A and tacrolimus also block MHC class I antigen presentation, which can also be inhibited by proteasome inhibitors such as lactacystin (121) or inhibitors of TAP (122). T cell activation is inhibited by some of the drugs that are already being used such as steroids, cyclosporin A and mycophenolate mofetil. Others could be tried. These include belatacept, a fusion protein of CTLA-4 and the Fc portion of IgG-1, which binds CD80 and CD86 and thereby blocks CD28 signaling (123). Dapirolizumab pegol is an antibody against CD40L, which blocks another co-stimulatory pathway. This drug is currently undergoing phase III trials for treatment of systemic lupus erythematosus  (124, 125). Rapamycin was tested preclinically and was shown to block humoral and cellular immune responses to AAV and to increase induction of regulatory CD4+ T cells (126128). Rapamycin is an mTOR inhibitor that blocks cell cycle progression. When used during T cell activation it prevents generation of effector CD8+ T cells but promotes memory formation (129).

Immunosuppression although potentially essential for some patients to ensure therapeutic benefits of AAV-mediate gene transfer comes at a cost. Immunosuppressive drugs have side effects that are unrelated to inhibition of immune responses. To name a few, prednisolone can cause intestinal ulcers, tacrolimus can result in headaches and muscle pain, belatacept has been linked to intestinal problems as has mycophenolate mofetil. Immunosuppression by its very nature robs an individual of its ability to fight off pathogens thus heightening the risks of more serious infections associated with more severe disease and increased shedding of the pathogen.

Summary

The immune system evolved to respond to components of pathogens including those of viral vectors. This system, which is essential for survival of an organism, is built on redundancy to counter rapidly mutating pathogens that have come up with multiple ways to dodge immune-mediated destruction. Modifying AAV vectors by removing parts that induce inflammatory responses or provide epitopes for T cell recognition may at best blunt responses. Nevertheless, modern medicine has developed a multitude of drugs that prevent activation of the immune system to overcome rejection of organ transplants. This in turn provides blueprints for drugs that can effectively block destructive T cell responses. Organs have been transplanted successfully since 1954 while AAV was not discovered until the mid-1960s. It then took another 30 years before an AAV vector was tried in a human gene therapy trial. By 2008, AAV-mediated gene therapy reported clinical benefits for a congenital blindness and then by 2011 systemic AAV transfer to the liver showed clinical benefits for hemophilia B patients. AAV-mediated gene transfer has thus progressed from its infancy to a stage of adolescence where hopefully remaining problems such as immune-mediated rejection can be solved in the near future.

Author Contributions

The author confirms being the sole contributor of this work and has approved it for publication.

Funding

Wistar Institute.

Conflict of Interest

The author has in the past been the recipient of a Sponsored Research Agreement by Spark Therapeutics.

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Friedmann T. A brief history of gene therapy. Nat Genet (1992) 2:93–8. doi: 10.1038/ng1092-93

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Kaufmann KB, Büning H, Galy A, Schambach A, Grez M. Gene therapy on the move. EMBO Mol Med (2013) 5:1642–61. doi: 10.1002/emmm.201202287

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Snyder RO, Miao CH, Patijn GA, Spratt SK, Danos O, Nagy D, et al. Persistent and therapeutic concentrations of human factor IX in mice after hepatic gene transfer of recombinant AAV vectors. Nat Genet (1997) 16:270–6. doi: 10.1038/ng0797-270

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Niemeyer GP, Herzog RW, Mount J, Arruda VR, Tillson DM, Hathcock J, et al. Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood (2009) 113:797–806. doi: 10.1182/blood-2008-10-181479

PubMed Abstract | CrossRef Full Text | Google Scholar

5. McIntosh J, Lenting PJ, Rosales C, Lee D, Rabbanian S, Raj D, et al. Therapeutic levels of FVIII following a single peripheral vein administration of rAAV vector encoding a novel human factor VIII variant. Blood (2013) 121:3335–44. doi: 10.1182/blood-2012-10-462200

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Maguire AM, Simonelli F, Pierce EA, Pugh EN, Mingozzi F, Bennicelli J, et al. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N Engl J Med (2008) 358:2240–8. doi: 10.1056/NEJMoa0802315

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Bennett J, Wellman J, Marshall KA, McCague S, Ashtari M, DiStefano-Pappas J, et al. Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial. Lancet (2016) 388:661–72. doi: 10.1016/S0140-6736(16)30371-3

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med (2006) 12:342–7. doi: 10.1038/nm1358

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Mingozzi F, Maus MV, Hui DJ, Sabatino DE, Murphy SL, Rasko JEJ, et al. CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat Med (2007) 13:419–22. doi: 10.1038/nm1549

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Ertl HCJ, High KA. Impact of AAV Capsid-Specific T-Cell Responses on Design and Outcome of Clinical Gene Transfer Trials with Recombinant Adeno-Associated Viral Vectors: An Evolving Controversy. Hum Gene Ther (2017) 28:328–37. doi: 10.1089/hum.2016.172

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Martino AT, Markusic DM. Immune response mechanisms against AAV vectors in animal models. Mol Ther Method Clin Dev (2020) 17:198–208. doi: 10.1016/j.omtm.2019.12.008

CrossRef Full Text | Google Scholar

12. Shirley JL, de Jong YP, Terhorst C, Herzog RW. Immune responses to viral gene therapy vectors. Mol Ther (2020) 28:709–22. doi: 10.1016/j.ymthe.2020.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Li H, Lin S-W, Giles-Davis W, Li Y, Zhou D, Xiang ZQ, et al. A preclinical animal model to assess the effect of pre-existing immunity on AAV-mediated gene transfer. Mol Ther (2009) 17:1215–24. doi: 10.1038/mt.2009.79

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Herzog RW. Immune responses to AAV capsid: are mice not humans after all? Mol Ther (2007) 15:649–50. doi: 10.1038/sj.mt.6300123

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Nathwani AC, Tuddenham EGD, Rangarajan S, Rosales C, McIntosh J, Linch DC, et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med (2011) 365:2357–65. doi: 10.1056/NEJMoa1108046

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Pasi KJ, Rangarajan S, Mitchell N, Lester W, Symington E, Madan B, et al. Multiyear follow-up of AAV5-hFVIII-SQ gene therapy for hemophilia A. N Engl J Med (2020) 382:29–40. doi: 10.1056/NEJMoa1908490

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Lai Q, Spoletini G, Bianco G, Graceffa D, Agnes S, Rossi M, et al. SARS-CoV2 and immunosuppression: A double-edged sword. Transpl Infect Dis (2020) 22. doi: 10.1111/tid.13404

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Muzyczka N, Berns KI. Parvoviridae: the viruses and their replication. In: Knipe DM, Howley PM, Griffen DE, Lamb RA, Martin MA, Roizman B, Straus SE, editors. Fields Virology, vol. 2. Southborough, MA: Lippincott Williams & Wilkins (2001). p. 2327–59.

Google Scholar

19. Schmidt M, Voutetakis A, Afione S, Zheng C, Mandikian D, Chiorini JA. Adeno-associated virus type 12 (AAV12): a novel AAV serotype with sialic acid- and heparan sulfate proteoglycan-independent transduction activity. J Virol (2008) 82:1399–406. doi: 10.1128/JVI.02012-07

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Pillay S, Zou W, Cheng F, Puschnik AS, Meyer NL, Ganaie SS, et al. Adeno-associated virus (AAV) serotypes have distinctive interactions with domains of the cellular AAV receptor. J Virol (2017) 91(18):e00391-17. doi: 10.1128/JVI.00391-17

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther (2010) 21:704–12. doi: 10.1089/hum.2009.182

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Nakai H, Yant SR, Storm TA, Fuess S, Meuse L, Kay MA. Extrachromosomal recombinant adeno-associated virus vector genomes are primarily responsible for stable liver transduction in vivo. J Virol (2001) 75:6969–76. doi: 10.1128/JVI.75.15.6969-6976.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Chéneau C, Kremer EJ. Adenovirus—Extracellular Protein Interactions and Their Impact on Innate Immune Responses by Human Mononuclear Phagocytes. Viruses (2020) 12:1351. doi: 10.3390/v12121351

CrossRef Full Text | Google Scholar

24. Kotin RM, Siniscalco M, Samulski RJ, Zhu XD, Hunter L, Laughlin CA, et al. Site-specific integration by adeno-associated virus. Proc Natl Acad Sci USA (1990) 87:2211–5. doi: 10.1073/pnas.87.6.2211

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Halbert CL, Miller AD, Mcnamara S, Emerson J, Gibson RL, Ramsey B, et al. Prevalence of neutralizing antibodies against adeno-associated virus (AAV) Types 2, 5, and 6 in cystic fibrosis and normal populations: implications for gene therapy using AAV vectors. Hum Gene Ther (2006) 17:440–7. doi: 10.1089/hum.2006.17.440

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Daniel HDJ, Kumar S, Kannangai R, Lakshmi KM, Agbandje-McKenna M, Coleman KE, et al. Prevalence of AAV3 capsid binding and neutralizing antibodies in healthy and individuals with hemophilia B from India. Hum Gene Ther (2020). doi: 10.1089/hum.2020.258

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Mimuro J, Mizukami H, Shima M, Matsushita T, Taki M, Muto S, et al. The prevalence of neutralizing antibodies against adeno-associated virus capsids is reduced in young Japanese individuals: Prevalence of Antibodies Against AAV. J Med Virol (2014) 86:1990–7. doi: 10.1002/jmv.23818

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Calcedo R, Vandenberghe LH, Gao G, Lin J, Wilson JM. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J Infect Dis (2009) 199:381–90. doi: 10.1086/595830

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Chirmule N, Propert K, Magosin S, Qian Y, Qian R, Wilson J. Immune responses to adenovirus and adeno-associated virus in humans. Gene Ther (1999) 6:1574–83. doi: 10.1038/sj.gt.3300994

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Ling C, Wang Y, Feng Y, Zhang Y, Li J, Hu X, et al. Prevalence of neutralizing antibodies against liver-tropic adeno-associated virus serotype vectors in 100 healthy Chinese and its potential relation to body constitutions. J Integr Med (2015) 13:341–6. doi: 10.1016/S2095-4964(15)60200-X

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Liu Q, Huang W, Zhang H, Wang Y, Zhao J, Song A, et al. Neutralizing antibodies against AAV2, AAV5 and AAV8 in healthy and HIV-1-infected subjects in China: implications for gene therapy using AAV vectors. Gene Ther (2014) 21:732–8. doi: 10.1038/gt.2014.47

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Li H, Lasaro MO, Jia B, Lin SW, Haut LH, High KA, et al. Capsid-specific T-cell responses to natural infections with adeno-associated viruses in humans differ from those of nonhuman primates. Mol Ther (2011) 19:2021–30. doi: 10.1038/mt.2011.81

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Veron P, Leborgne C, Monteilhet V, Boutin S, Martin S, Moullier P, et al. Humoral and cellular capsid-specific immune responses to adeno-associated virus type 1 in randomized healthy donors. J Immunol (2012) 188:6418–24. doi: 10.4049/jimmunol.1200620

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Vandamme C, Xicluna R, Hesnard L, Devaux M, Jaulin N, Guilbaud M, et al. Tetramer-based enrichment of preexisting anti-AAV8 CD8+ T cells in human donors allows the detection of a TEMRA subpopulation. Front Immunol (2020) 10:3110. doi: 10.3389/fimmu.2019.03110

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Sabatino DE, Mingozzi F, Hui DJ, Chen H, Colosi P, Ertl HCJ, et al. Identification of mouse AAV capsid-specific CD8+ T cell epitopes. Mol Ther (2005) 12:1023–33. doi: 10.1016/j.ymthe.2005.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kuranda K, Jean-Alphonse P, Leborgne C, Hardet R, Collaud F, Marmier S, et al. Exposure to wild-type AAV drives distinct capsid immunity profiles in humans. J Clin Invest (2018) 128:5267–79. doi: 10.1172/JCI122372

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discovery (2019) 18:358–78. doi: 10.1038/s41573-019-0012-9

CrossRef Full Text | Google Scholar

38. Qu W, Wang M, Wu Y, Xu R. Scalable Downstream Strategies for Purification of Recombinant Adeno- Associated Virus Vectors in Light of the Properties. Curr Parmaceut Biotecnol (2015) 16:684–95. doi: 10.2174/1389201016666150505122228

CrossRef Full Text | Google Scholar

39. Sandro Q, Relizani K, Benchaouir R. AAV Production Using Baculovirus Expression Vector System. In: Manfredsson FP, Benskey MJ, editors. Viral Vectors for Gene Therapy Methods in Molecular Biology. New York: NY: Springer New York (2019). p. 91–9. doi: 10.1007/978-1-4939-9065-8_5

CrossRef Full Text | Google Scholar

40. Rumachik NG, Malaker SA, Poweleit N, Maynard LH, Adams CM, Leib RD, et al. Methods Matter: Standard Production Platforms for Recombinant AAV Produce Chemically and Functionally Distinct Vectors. Mol Ther - Methods Clin Dev (2020) 18:98–118. doi: 10.1016/j.omtm.2020.05.018

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Hervas-Stubbs S, Rueda P, Lopez L, Leclerc C. Insect baculoviruses strongly potentiate adaptive immune responses by inducing type I IFN. J Immunol (2007) 178:2361–9. doi: 10.4049/jimmunol.178.4.2361

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Kovacsovics-Bankowski M. Rock KL. A phagosome-to-cytosol pathway for exogenous antigens presented on MHC class I molecules. Science (1995) 267:243–6. doi: 10.1126/science.7809629

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Merzougui N, Kratzer R, Saveanu L. van Endert P. A proteasome-dependent, TAP-independent pathway for cross-presentation of phagocytosed antigen. EMBO Rep (2011) 12:1257–64. doi: 10.1038/embor.2011.203

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Akira S, Hemmi H. Recognition of pathogen-associated molecular patterns by TLR family. Immunol Lett (2003) 85:85–95. doi: 10.1016/S0165-2478(02)00228-6

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Purbhoo MA, Irvine DJ, Huppa JB, Davis MM. T cell killing does not require the formation of a stable mature immunological synapse. Nat Immunol (2004) 5:524–30. doi: 10.1038/ni1058

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Sun JC, Bevan MJ. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science (2003) 300:339–42. doi: 10.1126/science.1083317

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Wherry EJ. T cell exhaustion. Nat Immunol (2011) 12:492–9. doi: 10.1038/ni.2035

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Jooss K, Yang Y, Fisher KJ, Wilson JM. Transduction of Dendritic Cells by DNA Viral Vectors Directs the Immune Response to Transgene Products in Muscle Fibers. J Virol (1998) 72:4212–23. doi: 10.1128/JVI.72.5.4212-4223.1998

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Wu T, Töpfer K, Lin S-W, Li H, Bian A, Zhou XY, et al. Self-complementary AAVs Induce More Potent Transgene Product-specific Immune Responses Compared to a Single-stranded Genome. Mol Ther (2012) 20:572–9. doi: 10.1038/mt.2011.280

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Manning WC, Paliard X, Zhou S, Pat Bland M, Lee AY, Hong K, et al. Genetic immunization with adeno-associated virus vectors expressing herpes simplex virus type 2 glycoproteins B and D. J Virol (1997) 71:7960–2. doi: 10.1128/JVI.71.10.7960-7962.1997

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Logan GJ, Wang L, Zheng M, Cunningham SC, Coppel RL, Alexander IE. AAV vectors encoding malarial antigens stimulate antigen-specific immunity but do not protect from parasite infection. Vaccine (2007) 25:1014–22. doi: 10.1016/j.vaccine.2006.09.072

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Rogers GL, Martino AT, Aslanidi GV, Jayandharan GR, Srivastava A, Herzog RW. Innate Immune Responses to AAV Vectors. Front Microbiol (2011) 2:194. doi: 10.3389/fmicb.2011.00194

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Hösel M, Broxtermann M, Janicki H, Esser K, Arzberger S, Hartmann P, et al. Toll-like receptor 2-mediated innate immune response in human nonparenchymal liver cells toward adeno-associated viral vectors. Hepatology (2012) 55:287–97. doi: 10.1002/hep.24625

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Mays LE, Wilson JM. The Complex and Evolving Story of T cell Activation to AAV Vector-encoded Transgene Products. Mol Ther (2011) 19:16–27. doi: 10.1038/mt.2010.250

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Cao O, Hoffman BE, Moghimi B, Nayak S, Cooper M, Zhou S, et al. Impact of the Underlying Mutation and the Route of Vector Administration on Immune Responses to Factor IX in Gene Therapy for Hemophilia B. Mol Ther (2009) 17:1733–42. doi: 10.1038/mt.2009.159

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Martino AT, Suzuki M, Markusic DM, Zolotukhin I, Ryals RC, Moghimi B, et al. The genome of self-complementary adeno-associated viral vectors increases Toll-like receptor 9–dependent innate immune responses in the liver. Blood (2011) 117:6459–68. doi: 10.1182/blood-2010-10-314518

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Lin S-W, Hensley SE, Tatsis N, Lasaro MO, Ertl HCJ. Recombinant adeno-associated virus vectors induce functionally impaired transgene product-specific CD8+ T cells in mice. J Clin Invest (2007) 117:3958–70. doi: 10.1172/JCI33138

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Lin J, Zhi Y, Mays L, Wilson JM. Vaccines based on novel adeno-associated virus vectors elicit aberrant CD8+ T-cell responses in mice. J Virol (2007) 81:11840–9. doi: 10.1128/JVI.01253-07

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Kumar SRP, Hoffman BE, Terhorst C, de Jong YP, Herzog RW. The Balance between CD8+ T Cell-Mediated Clearance of AAV-Encoded Antigen in the Liver and Tolerance Is Dependent on the Vector Dose. Mol Ther (2017) 25:880–91. doi: 10.1016/j.ymthe.2017.02.014

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Li H, Murphy SL, Giles-Davis W, Edmonson S, Xiang Z, Li Y, et al. Pre-existing AAV capsid-specific CD8+ T cells are unable to eliminate AAV-transduced hepatocytes. Mol Ther (2007) 15:792–800. doi: 10.1038/sj.mt.6300090

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Li C, Hirsch M, DiPrimio N, Asokan A, Goudy K, Tisch R, et al. Cytotoxic-T-lymphocyte-mediated elimination of target cells transduced with engineered adeno-associated virus type 2 vector in vivo. J Virol (2009) 83:6817–24. doi: 10.1128/JVI.00278-09

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Rogers GL, Shirley JL, Zolotukhin I, Kumar SRP, Sherman A, Perrin GQ, et al. Plasmacytoid and conventional dendritic cells cooperate in crosspriming AAV capsid-specific CD8+ T cells. Blood (2017) 129:3184–95. doi: 10.1182/blood-2016-11-751040

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Shirley JL, Keeler GD, Sherman A, Zolotukhin I, Markusic DM, Hoffman BE, et al. Type I IFN Sensing by cDCs and CD4+ T Cell Help Are Both Requisite for Cross-Priming of AAV Capsid-Specific CD8+ T Cells. Mol Ther (2020) 28:758–70. doi: 10.1016/j.ymthe.2019.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Wang L, Figueredo J, Calcedo R, Lin J, Wilson JM. Cross-Presentation of Adeno-Associated Virus Serotype 2 Capsids Activates Cytotoxic T Cells But Does Not Render Hepatocytes Effective Cytolytic Targets. Hum Gene Ther (2007) 18:185–94. doi: 10.1089/hum.2007.001

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Martino AT, Basner-Tschakarjan E, Markusic DM, Finn JD, Hinderer C, Zhou S, et al. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells. Blood (2013) 121:2224–33. doi: 10.1182/blood-2012-10-460733

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Wu T-L, Li H, Faust SM, Chi E, Zhou S, Wright F, et al. CD8+ T cell recognition of epitopes within the capsid of adeno-associated virus 8-based gene transfer vectors depends on vectors’ genome. Mol Ther (2014) 22:42–51. doi: 10.1038/mt.2013.218

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Clinical Trial. Choroideremia Gene Therapy Clinical Trial. (2015). Available at: https://clinicaltrials.gov/ct2/show/NCT02553135.

Google Scholar

68. Clinical Trial. Gene Therapy for Achromatopsia (CNGB3) (CNGB3). (2016). Available at: https://clinicaltrials.gov/ct2/show/NCT03001310.

Google Scholar

69. Clinical Trial. Gene Therapy for X-linked Retinitis Pigmentosa (XLRP) Retinitis Pigmentosa GTPase Regulator (RPGR). Available at: https://www.clinicaltrials.gov/ct2/show/NCT03252847.

Google Scholar

70. Clinincal Trial. Long-Term Follow-Up Gene Therapy Study for Leber Congenital Amaurosis OPTIRPE65 (Retinal Dystrophy Associated With Defects in RPE65). (2016). Available at: https://clinicaltrials.gov/ct2/show/NCT02946879.

Google Scholar

71. Taylor AW. Ocular Immune Privilege and Transplantation. Front Immunol (2016) 7:37. doi: 10.3389/fimmu.2016.00037

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Katagiri K, Zhang-Hoover J, Mo JS, Stein-Streilein J, Streilein JW. Using Tolerance Induced Via the Anterior Chamber of the Eye to Inhibit Th2-Dependent Pulmonary Pathology. J Immunol (2002) 169:84–9. doi: 10.4049/jimmunol.169.1.84

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Bellodi-Privato M, Le Meur G, Aubert D, Mendes-Madera A, Pichard V, Rolling F, et al. AAV gene transfer to the retina does not protect retrovirally transduced hepatocytes from the immune response. J Mol Med (2004) 82:403–10. doi: 10.1007/s00109-004-0537-0

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Maguire AM, Russell S, Wellman JA, Chung DC, Yu Z-F, Tillman A, et al. Efficacy, Safety, and Durability of Voretigene Neparvovec-rzyl in RPE65 Mutation–Associated Inherited Retinal Dystrophy. Ophthalmology (2019) 126:1273–85. doi: 10.1016/j.ophtha.2019.06.017

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Maguire AM, High KA, Auricchio A, Wright JF, Pierce EA, Testa F, et al. Age-dependent effects of RPE65 gene therapy for Leber’s congenital amaurosis: a phase 1 dose-escalation trial. Lancet (2009) 374:1597–605. doi: 10.1016/S0140-6736(09)61836-5

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Dimopoulos IS, Hoang SC, Radziwon A, Binczyk NM, Seabra MC, MacLaren RE, et al. Two-Year Results After AAV2-Mediated Gene Therapy for Choroideremia: The Alberta Experience. Am J Ophthal (2018) 193:130–42. doi: 10.1016/j.ajo.2018.06.011

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Bucher K, Rodríguez-Bocanegra E, Dauletbekov D, Fischer MD. Immune responses to retinal gene therapy using adeno-associated viral vectors – Implications for treatment success and safety. Prog Retin Eye Res (2020) 100915. doi: 10.1016/j.preteyeres.2020.100915

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Von Drygalski A, Giermasz A, Castaman G, Key NS, Lattimore S, Leebeek FWG, et al. Etranacogene dezaparvovec (AMT-061 phase 2b): normal/near normal FIX activity and bleed cessation in hemophilia B. Blood Adv (2019) 3:3241–7. doi: 10.1182/bloodadvances.2019000811

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Miesbach W, Meijer K, Coppens M, Kampmann P, Klamroth R, Schutgens R, et al. Gene therapy with adeno-associated virus vector 5–human factor IX in adults with hemophilia B. Blood (2018) 131:1022–31. doi: 10.1182/blood-2017-09-804419

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Rangarajan S, Walsh L, Lester W, Perry D, Madan B, Laffan M, et al. AAV5–Factor VIII Gene Transfer in Severe Hemophilia A. N Engl J Med (2017) 377:2519–30. doi: 10.1056/NEJMoa1708483

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Nathwani AC, Reiss UM, Tuddenham EGD, Rosales C, Chowdary P, McIntosh J, et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med (2014) 371:1994–2004. doi: 10.1056/NEJMoa1407309

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Butterfield JSS, Hege KM, Herzog RW. Kaczmarek R. A Molecular Revolution in the Treatment of Hemophilia. Mol Ther (2020) 28:997–1015. doi: 10.1016/j.ymthe.2019.11.006

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Levitsky J. Does the Liver Provide Immunosuppressive Advantage? Clin Liver Dis (2019) 13:180–3. doi: 10.1002/cld.817

CrossRef Full Text | Google Scholar

84. Smith BK, Collins SW, Conlon TJ, Mah CS, Lawson LA, Martin AD, et al. Phase I/II trial of adeno-associated virus-mediated alpha-glucosidase gene therapy to the diaphragm for chronic respiratory failure in Pompe disease: initial safety and ventilatory outcomes. Hum Gene Ther (2013) 24:630–40. doi: 10.1089/hum.2012.250

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Clinical Trial. Clinical Intramuscular Gene Transfer Trial of rAAVrh74.MCK.Micro-Dystrophin to Patients With Duchenne Muscular Dystrophy. (2015). Available at: https://clinicaltrials.gov/ct2/show/NCT02376816.

Google Scholar

86. Clinincal Trial. Gene Transfer Clinical Trial to Deliver rAAVrh74.MCK.GALGT2 for Duchenne Muscular Dystrophy. (2017). Available at: https://clinicaltrials.gov/ct2/show/NCT03333590.

Google Scholar

87. Flotte TR, Trapnell BC, Humphries M, Carey B, Calcedo R, Rouhani F, et al. Phase 2 Clinical Trial of a Recombinant Adeno-Associated Viral Vector Expressing α 1 -Antitrypsin: Interim Results. Hum Gene Ther (2011) 22:1239–47. doi: 10.1089/hum.2011.053

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Mueller C, Chulay JD, Trapnell BC, Humphries M, Carey B, Sandhaus RA, et al. Human Treg responses allow sustained recombinant adeno-associated virus–mediated transgene expression. J Clin Invest (2013) 123:5310–8. doi: 10.1172/JCI70314

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Gernoux G, Gruntman AM, Blackwood M, Zieger M, Flotte TR, Mueller C. Muscle-Directed Delivery of an AAV1 Vector Leads to Capsid-Specific T Cell Exhaustion in Nonhuman Primates and Humans. Mol Ther (2020) 28:747–57. doi: 10.1016/j.ymthe.2020.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Mendell JR, Campbell K, Rodino-Klapac L, Sahenk Z, Shilling C, Lewis S, et al. Dystrophin Immunity in Duchenne’s Muscular Dystrophy. N Engl J Med (2010) 363:1429–37. doi: 10.1056/NEJMoa1000228

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Bowles DE, McPhee SW, Li C, Gray SJ, Samulski JJ, Camp AS, et al. Phase 1 Gene Therapy for Duchenne Muscular Dystrophy Using a Translational Optimized AAV Vector. Mol Ther (2012) 20:443–55. doi: 10.1038/mt.2011.237

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Mendell JR, Rodino-Klapac LR, Rosales XQ, Coley BD, Galloway G, Lewis S, et al. Sustained alpha-sarcoglycan gene expression after gene transfer in limb-girdle muscular dystrophy, type 2D. Ann Neurol (2010) 68:629–38. doi: 10.1002/ana.22251

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Rafii MS, Tuszynski MH, Thomas RG, Barba D, Brewer JB, Rissman RA, et al. for the AAV2-NGF Study Team. Adeno-Associated Viral Vector (Serotype 2)–Nerve Growth Factor for Patients With Alzheimer Disease: A Randomized Clinical Trial. JAMA Neurol (2018) 75:834. doi: 10.1001/jamaneurol.2018.0233

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Marks WJ, Ostrem JL, Verhagen L, Starr PA, Larson PS, Bakay RA, et al. Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2–neurturin) to patients with idiopathic Parkinson’s disease: an open-label, phase I trial. Lancet Neurol (2008) 7:400–8. doi: 10.1016/S1474-4422(08)70065-6

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Christine CW, Starr PA, Larson PS, Eberling JL, Jagust WJ, Hawkins RA, et al. Safety and tolerability of putaminal AADC gene therapy for Parkinson disease. Neurology (2009) 73:1662–9. doi: 10.1212/WNL.0b013e3181c29356

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Lawlor PA, et al. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson’s disease: an open label, phase I trial. Lancet (2007) 369:2097–105. doi: 10.1016/S0140-6736(07)60982-9

PubMed Abstract | CrossRef Full Text | Google Scholar

97. LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty AW, Eskandar EN, et al. AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol (2011) 10:309–19. doi: 10.1016/S1474-4422(11)70039-4

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Worgall S, Sondhi D, Hackett NR, Kosofsky B, Kekatpure MV, Neyzi N, et al. Treatment of Late Infantile Neuronal Ceroid Lipofuscinosis by CNS Administration of a Serotype 2 Adeno-Associated Virus Expressing CLN2 cDNA. Hum Gene Ther (2008) 19:463–74. doi: 10.1089/hum.2008.022

PubMed Abstract | CrossRef Full Text | Google Scholar

99. McPhee SWJ, Janson CG, Li C, Samulski RJ, Camp AS, Francis J, et al. Immune responses to AAV in a phase I study for Canavan disease. J Gene Med (2006) 8:577–88. doi: 10.1002/jgm.885

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Jaski BE, Jessup ML, Mancini DM, Cappola TP, Pauly DF, Greenberg B, et al. Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID Trial), a First-in-Human Phase 1/2 Clinical Trial. J Cardiac Fail (2009) 15:171–81. doi: 10.1016/j.cardfail.2009.01.013

CrossRef Full Text | Google Scholar

101. Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med (2017) 377:1713–22. doi: 10.1056/NEJMoa1706198

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Van Laethem F, Baus E, Smyth LA, Andris F, Bex F, Urbain J, et al. Glucocorticoids Attenuate T Cell Receptor Signaling. J Exp Med (2001) 193:803–14. doi: 10.1084/jem.193.7.803

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Bereshchenko O, Coppo M, Bruscoli S, Biagioli M, Cimino M, Frammartino T, et al. GILZ promotes production of peripherally induced Treg cells and mediates the crosstalk between glucocorticoids and TGF-β signaling. Cell Rep (2014) 7:464–75. doi: 10.1016/j.celrep.2014.03.004

PubMed Abstract | CrossRef Full Text | Google Scholar

104. George LA, Sullivan SK, Giermasz A, Rasko JEJ, Samelson-Jones BJ, Ducore J, et al. Hemophilia B gene therapy with a high-specific-activity factor IX variant. N Engl J Med (2017) 377:2215–27. doi: 10.1056/NEJMoa1708538

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Pipe S, Stine K, Rajasekhar A, Everington T, Poma A, Crombez E, et al. 101HEMB01 Is a Phase 1/2 Open-Label, Single Ascending Dose-Finding Trial of DTX101 (AAVrh10FIX) in Patients with Moderate/Severe Hemophilia B That Demonstrated Meaningful but Transient Expression of Human Factor IX (hFIX). Blood (2017) 130:3331. doi: 10.1182/blood.V130.Suppl_1.3331.3331

CrossRef Full Text | Google Scholar

106. Monahan PE, Walsh CE, Powell JS. Update on a phase 1/2 open-label trial of BAX335, an adeno-associated virus 8 (AAV8) vector-based gene therapy program for hemophilia B. J Thromb Haemost (2015) 13(Suppl 2):87. doi: 10.1111/jth.12957

CrossRef Full Text | Google Scholar

107. Calcedo R, Kuri-Cervantes L, Peng H, Qin Q, Boyd S, Schneider M, et al. Immune Responses in 101HEMB01, a Phase 1/2 Open-Label, Single Ascending Dose-Finding Trial of DTX101 (AAVrh10FIX) in Patients with Severe Hemophilia. Blood (2017) 130:3333. doi: 10.1182/blood.V130.Suppl_1.3333.3333

CrossRef Full Text | Google Scholar

108. High KA, George LA, Eyster ME, Sullivan SK, Ragni MV, Croteau SE, et al. A Phase 1/2 Trial of Investigational Spk-8011 in Hemophilia a Demonstrates Durable Expression and Prevention of Bleeds. Blood (2018) 132:487–7. doi: 10.1182/blood-2018-99-115495

CrossRef Full Text | Google Scholar

109. Samelson-Jones BJ, Finn JD, Favaro P, Wright JF, Arruda VR. Timing of Intensive Immunosuppression Impacts Risk of Transgene Antibodies after AAV Gene Therapy in Nonhuman Primates. Mol Ther Method Clin Dev (2020) 17:1129–38. doi: 10.1016/j.omtm.2020.05.001

CrossRef Full Text | Google Scholar

110. Ferreira V, Twisk J, Kwikkers K, Aronica E, Brisson D, Methot J, et al. Immune responses to intramuscular administration of alipogene tiparvovec (AAV1-LPL(S447X)) in a phase II clinical trial of lipoprotein lipase deficiency gene therapy. Hum Gene Ther (2014) 25:180–8. doi: 10.1089/hum.2013.169

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Matsuda S, Koyasu S. Mechanisms of action of cyclosporine. Immunopharmacology (2000) 47:119–25. doi: 10.1016/S0162-3109(00)00192-2

PubMed Abstract | CrossRef Full Text | Google Scholar

112. McIntosh JH, Cochrane M, Cobbold S, Waldmann H, Nathwani SA, Davidoff AM, et al. Successful attenuation of humoral immunity to viral capsid and transgenic protein following AAV-mediated gene transfer with a non-depleting CD4 antibody and cyclosporine. Gene Ther (2012) 19:78–85. doi: 10.1038/gt.2011.64

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Kawai M, Kitade H, Mathieu C, Waer M, Pirenne J. Inhibitory and Stimulatory Effects of Cyclosporine A on the Development of Regulatory T Cells In Vivo. Transplantation (2005) 79:1073–7. doi: 10.1097/01.TP.0000153505.73700.32

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology (2000) 47:85–118. doi: 10.1016/S0162-3109(00)00188-0

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Dolasia K, Bisht MK, Pradhan G, Udgata A, Mukhopadhyay S. TLRs/NLRs: Shaping the landscape of host immunity. Int Rev Immunol (2018) 37:3–19. doi: 10.1080/08830185.2017.1397656

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Davignon J-L, Rauwel B, Degboé Y, Constantin A, Boyer J-F, Kruglov A, et al. Modulation of T-cell responses by anti-tumor necrosis factor treatments in rheumatoid arthritis: a review. Arthrit Res Ther (2018) 20. doi: 10.1186/s13075-018-1725-6

CrossRef Full Text | Google Scholar

117. Kole A, He J, Rivollier A, Silveira DD, Kitamura K, Maloy KJ, et al. Type I IFNs Regulate Effector and Regulatory T Cell Accumulation and Anti-Inflammatory Cytokine Production during T Cell–Mediated Colitis. J Immunol (2013) 191:2771–9. doi: 10.4049/jimmunol.1301093

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Martinez GP, Zabaleta ME, Di Giulio C, Charris JE, Mijares MR. The Role of Chloroquine and Hydroxychloroquine in Immune Regulation and Diseases. Curr Parmaceut Des (2020) 26:4467–85. doi: 10.2174/1381612826666200707132920

CrossRef Full Text | Google Scholar

119. Schreiber SL, Crabtree GR. The mechanism of action of cyclosporin A and FK506. Immunol Today (1992) 13:136–42. doi: 10.1016/0167-5699(92)90111-J

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Fujii H, Ivison SM, Shimizu H, Kajiwara R, Kariminia A, Yan M, et al. Inhibition of Cathepsin S Reduces Allogeneic T Cell Priming but Not Graft-versus-Host Disease Against Minor Histocompatibility Antigens. Biol Blood Marrow Transplant (2012) 18:546–56. doi: 10.1016/j.bbmt.2011.11.027

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Song P-H, Xie H-Y, Zheng S-S, Wu J. The Inhibitory Role of Lactacystin and β-Lactacystin on T-cell Activation and Proliferation. Acta Biochim Biophys Sin (2004) 36:123–7. doi: 10.1093/abbs/36.2.123

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Rock KL, Yeh ET, Gramm CF, Haber SI, Reiser H, Benacerraf B. TAP, a novel T cell-activating protein involved in the stimulation of MHC-restricted T lymphocytes. J Exp Med (1986) 163:315–33. doi: 10.1084/jem.163.2.315

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Larsen CP, Pearson TC, Adams AB, Tso P, Shirasugi N, Strobert ME, et al. Rational Development of LEA29Y (belatacept), a High-Affinity Variant of CTLA4-Ig with Potent Immunosuppressive Properties. Am J Transplant (2005) 5:443–53. doi: 10.1111/j.1600-6143.2005.00749.x

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Clinical Trial. A Study to Evaluate the Efficacy and Safety of Dapirolizumab Pegol in Study Participants With Moderately to Severely Active Systemic Lupus Erythematosus (PHOENYCS GO). (2020). Available at: https://clinicaltrials.gov/ct2/show/NCT04294667.

Google Scholar

125. Clowse MEB, Wallace DJ, Furie RA, Petri MA, Pike MC, Leszczyński P, et al. Efficacy and Safety of Epratuzumab in Moderately to Severely Active Systemic Lupus Erythematosus: Results From Two Phase III Randomized, Double-Blind, Placebo-Controlled Trials: EFFICACY AND SAFETY OF EPRATUZUMAB IN SLE. Arthrit Rheumatol (2017) 69:362–75. doi: 10.1002/art.39856

CrossRef Full Text | Google Scholar

126. Nayak S, Sarkar D, Perrin GQ, Moghimi B, Hoffman BE, Zhou S, et al. Prevention and Reversal of Antibody Responses Against Factor IX in Gene Therapy for Hemophilia B. Front Microbiol (2011) 2:244. doi: 10.3389/fmicb.2011.00244

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Meliani A, Boisgerault F, Hardet R, Marmier S, Collaud F, Ronzitti G, et al. Antigen-selective modulation of AAV immunogenicity with tolerogenic rapamycin nanoparticles enables successful vector re-administration. Nat Commun (2018) 9. doi: 10.1038/s41467-018-06621-3

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Fanigliulo D, Lazzerini PE, Capecchi PL, Ulivieri C, Baldari CT, Laghi-Pasini F. Clinically-relevant cyclosporin and rapamycin concentrations enhance regulatory T cell function to a similar extent but with different mechanisms: An in-vitro study in healthy humans. Int Immunopharmacol (2015) 24:276–84. doi: 10.1016/j.intimp.2014.12.021

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, et al. mTOR regulates memory CD8 T-cell differentiation. Nature (2009) 460:108–12. doi: 10.1038/nature08155

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: CD8+ T cells, memory, immunosuppression, effector functions, animal models, clinical trials

Citation: Ertl HCJ (2021) T Cell-Mediated Immune Responses to AAV and AAV Vectors. Front. Immunol. 12:666666. doi: 10.3389/fimmu.2021.666666

Received: 10 February 2021; Accepted: 22 March 2021;
Published: 13 April 2021.

Edited by:

Nicole K. Paulk, University of California, San Francisco, United States

Reviewed by:

Ronzitti Giuseppe, Genethon, France
Romain Hardet, INSERM U1234 Physiopathologie, Autoimmunité, Maladies Neuromusculaires et THErapies Régénératrices (PANTHER), France

Copyright © 2021 Ertl. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Hildegund C. J. Ertl, ertl@wistar.org

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.