Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Neurol., 18 June 2025

Sec. Experimental Therapeutics

Volume 16 - 2025 | https://doi.org/10.3389/fneur.2025.1603125

Viral vectors in neurodegenerative diseases: immune responses and therapeutic applications


Yifei Wang,,&#x;Yifei Wang1,2,3Siyu Mu,,&#x;Siyu Mu1,2,3Fangxi Liu,,
Fangxi Liu1,2,3*
  • 1Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
  • 2Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China
  • 3Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, Shenyang, China

Gene transfer-based therapies utilizing viral vectors have undergone remarkable advancements and hold significant promise in addressing neurodegenerative diseases in recent years, whose potential mechanisms include replacing or silencing pathogenic genes and delivering neurotrophic factors. Current preclinical research focuses on developing novel strategies in gene modification to combat neurodegenerative disorders. Numerous clinical trials involving viral vectors in the nervous system are either on-going or completed. Despite these advancements, progress in this field remains constrained by immune responses triggered by viral vectors, which can be triggered through innate and adaptive pathways. The present review will focus on the advances in the development and application of viral vector-based gene therapies for neurodegenerative diseases and summarize the related immune responses, aiming to provide a forward-looking perspective for this emerging arena.

1 Introduction

Neurodegenerative diseases (NDs) are a heterogeneous group of complex diseases characterized by neuronal loss and progressive degeneration of different areas in the nervous system (1). To date, no effective therapeutics have been developed to slow, halt, or prevent any NDs (2). As one of the therapeutic vectors widely studied, viral vectors are now routinely used as tools for studying gene function as well as developing gene-based therapies for a variety of diseases (3). Viral vectors take advantage of the ability of viruses to enter a cell, enabling direct intracellular gene product delivery (4, 5). However, increasing attention is being directed toward the immune responses induced by viral vector administration, along with the associated side effects. Although significant progress has been made over the past two decades in understanding innate and adaptive immune responses to natural infections with viral vectors or viral vector-mediated gene transfer using various types of vectors (6), further strategies are still required to mitigate the side effects caused by these immune responses.

In recent years, viral vector-mediated gene therapies have been increasingly explored as potential treatments for neurodegenerative diseases. Although preclinical and clinical data have demonstrated the relative safety and effectiveness of these in-vivo therapies across central nervous system (CNS) pathologies, challenges still remain in the progress of clinical translation (7). In this review, we will focus on preclinical studies using viral vector-mediated gene therapy for neurodegenerative diseases, as well as their clinical translation, and discuss the different types of immune responses elicited by viral vectors and how they translate into lack of efficacy or increased toxicity. Importantly, this review integrates advances in gene delivery with a detailed analysis of host immune reactions, highlighting the balance between therapeutic potential and immunological barriers in neurodegeneration.

2 Basic biology of viral vectors

Viral vectors are widely used tools for gene delivery, especially in the central nervous system (CNS). Common types include adeno-associated viruses (AAVs), retroviruses (RVs), adenoviruses (Ads), lentiviruses (LVs) (8). Viral vectors vary in their payload, cell tropism, immunogenicity and capacity and persistence of transgene induction (8). In this section, viral vectors commonly used will be discussed and key features of each vector are compared in Table 1.

Table 1
www.frontiersin.org

Table 1. Key features of viral vectors.

2.1 Adenoviruses (Ads)

Adenoviruses have a genome size of 35–40 kb (8), which possess a capsid that accommodates a 26–45 kb linear, double-stranded DNA genome (9). Ads effectively infect dividing cells and does not integrate into the host genome (8), and they bind to various cell surface proteins to facilitate their entry into the target cells (10). The type of tissue infected largely depends on the cell tropism of the virus. It is assumed that Ads infect a broad range of cell and tissue types, including neuronal cells, dendritic cells, and hepatocytes preferentially (8). This is mainly because most human cells express the primary adenovirus receptor and the secondary integrin receptors (11, 12). Different serotypes exhibit distinct cell tropisms. For instance, Ad5 shows strong liver tropism mainly through binding of its hexon protein to coagulation factor X (10). While Ad35, a group B adenovirus, uses CD46 as its primary cellular receptor, which is broadly expressed on dendritic and hematopoietic cells, facilitating preferential infection of immune cell types (13).

Since adenoviral vectors were initially used for brain cell transduction in the early 1990s (14), they have been applied in the treatment of cancer, Parkinson's disease, and Huntington's disease (1). Ads were the first DNA virus to enter rigorous therapeutic development, largely because of its well-defined biology, genetic stability, large transgene capacity and ease of large-scale production (9). However, Ad vectors can induce high-level innate inflammatory responses within the first 24 h of transduction (15). For instance, Schaack (15) reported that adenoviral early region proteins upregulate pro-inflammatory cytokines such as IL-6 and TNF-α, even in the absence of viral replication. In spinal cord injury models, Islam and Tom observed that Ad vector delivery induced notable microglial activation and neutrophil infiltration, highlighting the potential for both therapeutic and detrimental inflammatory effects (16). Besides, they also have limitations like short gene expression of 2 weeks to several months (transient) and high risk of cytopathic effects (8, 16). Additionally, high titters can result in organ damage and even mortality (16). An intravenous administration of Ads may induce acute liver injury, as has been reported in animal models (17). Still, these disadvantages can be corrected by designing and synthesizing other novel serotypes (8). For example, the third-generation adenovirus vectors, also referred to as the helper-dependent or gutless adenovirus, can significantly reduce in vivo immune response compared to the first- and second-generation Ads, although high transduction efficiency and tropism are maintained (18).

2.2 Adeno-associated viruses (AAVs)

Since AAVs were initially discovered as a contaminant in adenovirus preparations (19), their biological characteristics and applications have been widely studied. AAVs have a single-stranded DNA, with genome DNA payloads under 5 kb (8), and have a small (~25 nm) icosahedral capsid composed of three types of structural proteins, namely, VP1, VP2, and VP3 (20). AAVs have the ability to transduce dividing and nondividing cells, but do not integrate into the host genome (8). They are capable of expressing transgenes over several months in nondividing cells (9), and provide a relatively stable expression in dividing cells as well (21). In comparison to Ads, AAVs have a long duration of gene expression in vivo (22). The specificity of adeno-associated virus vector-based cell and tissue targeting is determined by the capsid proteins of AAVs (9). Compared with Ads, a similar cell tropism is shown by AAVs (8). AAVs have at least 12 natural serotypes and many artificial variants, each of which exhibits different cell tropisms in the nervous system (2326). For instance, AAV2 is one of the most extensively studied serotypes and primarily transduces neurons after direct injection into the brain parenchyma (23). AAV5 shows enhanced transduction in the striatum and cerebellum (23), while AAV8 and AAV9 are capable of crossing the blood–brain barrier (BBB) following systemic administration, thus enabling widespread CNS gene delivery (21). Notably, AAV9 preferentially targets neonatal neurons and adult astrocytes (24). AAV11 has recently been shown to enhance astrocyte-directed transduction and enable efficient retrograde labeling of projection neurons, therefore broadening its potential in circuit-specific interventions (22). These variations among AAV serotypes provide opportunities to tailor gene delivery strategies based on cell-type specificity and disease context.

AAV-mediated gene transfer has great potential as a therapeutic approach (27). Most of the currently developed AAV vectors are directed toward monogenic diseases (28), but they are also versatile and enable one to test for other CNS diseases (9). AAV vectors can lead to severe adverse effects due to off-target expression, such as hepatotoxicity (6), neurotoxicity (29), and even death in critical trials (3032). Using AAV vectors alone does not elicit a strong immune response similar to that elicited upon using other viruses (9). AAV vectors can also induce unwanted immune responses, while about 50% of humans may have neutralizing antibodies due to previous infections (33, 34). Although certain major obstacles limit the widespread application of AAV vectors, including limited insert size, inefficient transduction, narrowed disease case, and off-target responses, they hold great potential to revolutionize the clinical management of human diseases (9). Overall, AAVs offer a relatively safe and versatile platform for CNS gene therapy, though limitations such as pre-existing neutralizing antibodies and vector immunogenicity must be addressed in clinical applications.

2.3 Lentiviruses (LVs)

Lentiviruses have a single-stranded RNA genome and may incorporate constructs up to 9–10 kB in size (35, 36). Lentiviral (LV) vectors can infect both dividing and non-dividing cells, such as neurons (8, 37). While it also can preserve long-term and stable transgene induction (8), which is crucial for adolescents or pediatric patients (9). Genetic engineering makes it possible to produce LVs with specific integration sites for the increased safety of use (38). Unlike Ad or AAV vectors, LV vectors rarely generate neutralizing antibodies (36). Due to their relatively low-immunogenic characteristics (35), It is reported to have low Immunogenicity and elicit no pathogenic effects (39), suggesting that this vector is relatively safe in vivo. LVs are capable of infecting a broad range of host cell types, allowing their use for the treatment of Parkinson's disease (PD) Alzheimer's disease (AD) (1). They have also been demonstrated as efficient gene transfer vehicles for human solid tumor cells. For example, they enabled diphtheria toxin A delivery to suppress prostate cancer xenografts (39), mediated suicide gene therapy using HSV-tk in hepatocellular carcinoma (40), and outperformed retroviral vectors in transducing ovarian cancer cells (41). Although certain problems remain to be addressed, the safe and efficient LV vectors are nonetheless considered promising as a tool for human gene therapy (9). These findings highlight the potential of LV vectors for sustained and low-immunogenic gene delivery in chronic neurodegenerative conditions.

2.4 Other viral vectors

Another commonly used viral vector is retroviruses (RVs), which are single-stranded RNA and integrated into the genome of the host cell (42). They can carry up to 8–9 kb of foreign DNA for transduction (43, 44). RVs can stably integrate into the target cell genome (45), so they are useful for ex vivo delivery of somatic cells. RVs provide a stable and efficient expression of the transgene to patients (4). They have been used in early gene therapy trials for severe combined immunodeficiency (SCID), demonstrating long-term gene correction in hematopoietic stem cells (46). However, they have limitations including immunogenic problems and inability to transduce the nondividing cells (43). Herpes simplex viruses (HSVs) are also one of the recent virus candidates in gene delivery. HSVs are an enveloped virus with a double-stranded DNA (dsDNA) Genome (4). They can usually accommodate up to 40 kb of transgenic DNA (3). Because of its neurotropic features (47), they have strong tropism for neurons. For example, HSV vectors have been studied for the delivery of neurotrophic factors in models of chronic pain and epilepsy (48). HSVs may initiate strong inflammatory response (4), and result in transient gene expression in cells infected with them (49). Vesicular Stomatitis viruses (VSVs) contain a single stranded RNA genome (50). And their cargo capacity is approximately 6 kb (50). VSVs are reported to cause humoral and cellular immune response in previous studies (50, 51). These immunological features, along with their strong cytolytic activity, support their use as oncolytic agents and viral vectors in cancer virotherapy (52). These viral vectors expand the toolkit for gene delivery in the CNS, each offering unique advantages and limitations depending on the therapeutic context.

3 Viral vector delivery strategies for the nervous system

There are many ways to deliver viral vectors to the brain, each varying in efficiency, target region, and biodistribution. In this section, we will introduce traditional and novel delivery strategies and their advantages and disadvantages.

3.1 Intraparenchymal delivery

Intraparenchymal delivery is the most used viral vector delivery method with high delivery efficiency in studies for treating neurological diseases. It's an approach that enables direct delivery to the target structure, bypassing the blood-brain barrier, and usually accomplished via a stereotactic head frame or magnetic resonance imaging guidance (53, 54). While stereotactic guided injection is frequently used for intraparenchymal delivery, risk of surgical intervention and the restricted spread of transgene expression still needs to be concerned. A clinical study on Parkinson's disease patients (NCT00400634) reported severe side effects associated with surgical procedure (55). Intraparenchymal delivery typically affects only a limited range of brain regions, with highest vector distribution and downstream gene product activity around the injection site (56, 57).

A new technology called interventional MRI-guided convection-enhanced delivery (iMRI-CED) has been introduced in recent years, which is the gold standard for verifying accurate vector delivery in real time (58). Convection-enhanced delivery (CED) has been shown to improve specificity and delivery efficiency in a safe, reliable, targeted, and homogeneous manner across the blood-brain barrier (216219). Therefore, CED holds significant potential for broad application in the treatment of neurological diseases.

3.2 Intravenous delivery

Efficacy of intravenous delivery largely hinges on the ability of AAV to cross the BBB. Among the naturally existing AAVs, AAV9 is currently the most commonly used AAV serotype for CNS transduction in preclinical and clinical studies (59), which may be attributed to its active-transport mechanisms (60, 61). It has several advantages including non-invasive, widely distributed, and targeting both CNS and peripheral tissues. However, this approach also faces a few challenges. People who have been exposed to AAVs may pre-existing antibodies that can neutralize AAVs (6, 62). As it travels through the bloodstream, immune responses can diminish its efficacy (63). Besides, systemic toxicity such as off-target effects has also been found in previous studies (64, 65).

3.3 Intracerebralspinal fluid injection (intra-CSF)

Another method is intra-CSF injection including intra-cerebroventricular injection (ICV), intrathecal injection (IT), and intracisternal magna injection (ICM). ICV refers to direct administration into ventricles in the brain, allowing the AAV to permeate the barrier and spread widely across different brain regions (66, 67). IT delivers AAV via injection into the spinal canal through the lumbar puncture, resulting in broad coverage of the brain and spinal cord (68). ICM is a technique similar to IT, while its injection site is the cisterna magna (CM). Intra-CSF injection can bypass the BBB and spread widely within the CNS. Compared with intravenous delivery, intra-CSF injection minimizes systemic exposure and avoids neutralization by circulating antibodies. However, it has insufficient coverage, leading to limited access to deeper brain regions and the presence of surgical risks (6971).

3.4 Intranasal delivery

Intranasal delivery is an alternative and non-invasive option which can directly deliver viral vectors through the nose to the brain (72). Intranasal delivery has been used in clinical studies to deliver small-molecule agents to the brain, such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), thus treating neurodegenerative diseases (7375). Intranasal delivery is easy to perform and can be repeated, even by patients themselves (72). It is also capable of bypassing the BBB (72). However, intranasal delivery holds limitations. Although it minimizes systemic exposure (76), the cells along the delivery pathways, such as olfactory sensory neurons and trigeminal nerves, may also be transduced and cause off-target effects (72).

3.5 FUS-BBBO

Beyond invasive approaches, focused ultrasound with microbubble-mediated BBB opening (FUS-BBBO) is an emerging non-invasive approach for viral vector delivery in treating neurological diseases. It has the potential to overcome existing delivery limitations by providing a means of non-invasive, site-specific gene transfer to the brain, offering advantages such as improved safety and targeted delivery (7781). Various administration routes have been explored, including intravenous, intranasal, and local injection, which have expanded its applicability for gene therapy across multiple neurodegenerative disorders (8184). Recent studies have demonstrated the successful application of FUS-BBBO in various neurodegenerative and neurological conditions. For example, AAV has been used in models of Alzheimer's disease and Huntington's disease, while HSV has been applied in glioblastoma (8385). Importantly, FUS-BBBO may reduce systemic immune activation by enabling lower vector doses, thereby potentially lowering the risk of pre-existing neutralizing antibodies and allowing for safer repeated administration (83). Despite these advantages, FUS-BBBO has its challenges. One primary concern is still the possibility of off-target side effects and toxicity due to the intravenous administration (59). In summary, FUS-BBBO offers a promising delivery strategy that enhances brain-targeted gene therapy while potentially mitigating immune-related complications.

4 Immune responses against AAV injection

As the most widely studied and used viral vectors in the field of therapeutic application to neurodegenerative diseases, AAV injection was found to trigger both innate and adaptive immune responses in the current preclinical studies. As mentioned above, immune responses were also reported in other viral vectors after transduction (15, 35, 8688). In this section, we would like to focus only on the immunogenicity of AAV vectors after administration and have further discussion. Figure 1 has summarized the main mechanisms of immune responses. Responses triggered by brain are less likely to be strong, given its immune-privileged nature (6). The responses are most likely linked to adaptive responses in most humans, except for young children who are less likely to have immune memory to AAV capsid (6).

Figure 1
www.frontiersin.org

Figure 1. Mechanism of immune responses against AAV-mediated gene transfer. The graph shows the different type of immune responses that are elicited upon injection of AAV vectors. Toxicities are underlined and listed next to the components of the immune responses that contribute. PAMP, Pathogen-Associated Molecular Pattern; MDA5, Melanoma Differentiation-Associated protein 5; CpG, unmethylated CpG motifs; DCs, dendritic cells; dRNA, double stranded RNA; MHC, Major Histocompatibility Complex. Created in BioRender. Wang, Y. (2025) https://BioRender.com/4dt5yfl.

4.1 Innate immune responses

Innate immunity is the first barrier against pathogens as it mounts rapidly and does not require specific adaptation to the pathogens (62). Its response depends on the recognition of pathogen-associated molecular patterns (PAMPs), which are carried by viruses or viral vectors (89). Both the capsid and the DNA components of AAV may contribute to the activation of innate immunity, resulting in adverse reactions and immune toxicity.

In the context of AAV-mediated gene transfer, one mechanism is considered to trigger inflammatory responses is a PAMP within the capsid proteins binding to Toll-like receptor 2 (TLR2) (90), positively expressed on myeloid dendritic cells. Another mechanism involves the AAV double-stranded DNA (dsDNA) genome, in particular its unmethylated CpG sequences, recognized by Toll-like receptor 9 (TLR9) (9194). TLR9 engagement increased antigen presentation on class I major histocompatibility complex (MHC-I) and further associated with enhanced activation of AAV-specific CD8+ T cell (9496). Further CpG Depletion of Vector Genome in animal models has shown that a reduction of the vectors' CpG motifs blunts CD8+ T cell responses without modifying B cell responses (97, 98). In addition to the vector DNA genome, double stranded RNA (dsRNA) may also participate in the induction of innate immunity from long-term AAV transduction, which could interact with melanoma differentiation-associated protein 5 (MDA5) (99). It is well known that MDA5 is cytoplasmic viral RNA sensor serving as a cytoplasmic pattern recognition receptor (PRR) and ubiquitously expressed. MDA5 is capable of activating type I interferons (type I IFNs) signaling pathways after virus infection, leading to the expression of type I IFNs (99). Besides, it is also found that AAV vectors activate type I IFNs expression in plasmacytoid dendritic cells (pDCs) of human and murine (93). TLR9 can interact with myeloid differentiation primary response gene 88 (MyD88) for downstream signaling, leading to type I IFNs production in pDCs and further promoting activation of CD8+T cells (93, 96). pDCs recognize AAV genomes via TLR9 and release type I interferons, which in turn activate conventional dendritic cells to prime capsid-specific CD8+ T cells—highlighting the interplay between innate sensing and adaptive immune activation (62). These studies underscore the important role of type I IFNs in bridging innate and adaptive immunity following AAV administration.

4.2 Adaptive immune responses

An adaptive immune response requires a longer time than innate immunity and is considered the second line of defense against pathogens. It is characterized by antigen-specific and the ability to eliminate pathogens while generating an immunological memory (62). AAV-specific adaptive responses can be induced by previous natural infection or they can be stimulated or recalled by AAV gene transfer.

After AAV vector administration, T and B lymphocytes are activated following molecular recognition of an antigen presented by antigen-presenting cells (APCs) (62). Cytotoxic CD8+T cell responding via MHC class I to AAV capsid protein can affect therapeutic efficacy (94, 95, 100), possibly by clearing AAV-transduced cells thus inducing inflammation in the target tissue (101, 102). It is also reported that increased AAV-antigen presented on capsid-derived MHC class I together with higher CD8+T cells activation in dose-dependent in vitro experiments (103, 104). The concurrent presentation of CD4+ T helper cells activated by capsid-derived MHC class II facilitate both humoral and cell-mediated immune responses (105). AAV-specific B cell responses induced by previous natural infections can neutralize the viral vectors before they can deliver their payload, thus precluding a successful AAV-mediated gene transfer (6). Efforts have made to avoid its neutralization by modification to the vector's capsid (106). However, it showed limited success as many of the antibodies bind to domains that are crucial for transduction (106, 107). We can see that T cells play an important part in adaptive Immune responses against viral vectors. Liver toxicity caused by T cells directed to epitopes, which bind with low affinity to MHC class I molecules, is more worthy of our attention. Therefore, cellular responses, in particular the T cells, are needed to further study and make a breakthrough.

4.3 Immune responses against the transgene product

Anti-transgene immune responses were documented in the clinical trials, mostly after intramuscular delivery of AAV vectors (108, 109). For example, evidence of T cell-mediated anti-transgene cytotoxic T cell responses was documented in a phase I/II trial of intramuscular gene transfer in Duchenne muscular dystrophy patients (108). The risk of antibody responses to the transgene product is influenced by many factors, including the route of vector administration, specific vector design, types of viral vectors, and vector dose. Additional host factors may include specific aspects such as tissue inflammation, target tissue and the host genetic background (105). Clinical experience indicates that immune responses directed against AAV vectors are dose dependent (110, 111). Compared to Ad and LV vectors, AAV vectors have relatively weak and transient innate response and least efficient inducer of CD8+ T cells (105). The clinical data presented suggest that disease-specific conditions, such as the ongoing inflammation in muscle dystrophies (108), are likely to increase transgene immunogenicity after gene transfer.

4.3.1 Therapeutic strategies for immune responses against AAV injection

Therapies are mostly administered only once in studies nowadays, with repeated dosing precluded. According to clinical trials, AAV vectors administration leads to the development of anti-AAV IgG and NAbs (112), which could be a potential reason for preventing vector readministration. Nevertheless, preclinical studies also show that administration of immunomodulatory regimens (113) or B-cell depletion prior to gene transfer (114) can effectively block humoral immune responses against AAV vectors. These encouraging results leading to attempts in a clinical trial testing rituximab, a B-cell depleting monoclonal antibody targeting CD20 and reducing antibody responses, in combination with rapamycin (an immunosuppressant) in humans as a strategy to enable vector readministration (NCT02240407). Induction of antibodies to a transgene product could be very harmful as such antibodies could complicate traditional protein therapy. An animal study showed that, immunosuppressants such as rapamycin, which inhibits mTOR signaling to reduce T cell proliferation and inflammation. Ibrutinib inhibits Bruton's tyrosine kinase (BTK), thereby suppressing B cell activation and antibody production. Both drugs, when given alone, reduce primary antibody responses against AAV capsid (115), blunting recall responses and reducing numbers of circulating antibody-secreting plasma cells, and the combination of therapy is more effective. In a clinical trial, about a third of the patients who received Onasemnogene abeparvovec, an FDA approved AAV9 vector therapy for SMA, showed liver damage associated with an inflammatory reaction comprised mainly of CD8+ T cells, but all patients recovered after treatment with steroids, which broadly suppress immune activity and are especially effective in controlling CD8+ T cell-mediated cytotoxicity (116). However, in some cases, some patients fail to respond and still reject the AAV-transduced cells (117, 118). Besides, immunosuppression would be required for lengthy periods of time or even for the lifespan of the gene transfer recipients. Taken together, these findings emphasize the importance of different immunomodulatory strategies to improve gene therapy outcomes, including pharmacological immunosuppressants (such as rapamycin and ibrutinib) and biological agents targeting B cells (such as rituximab). Figure 2A is provided to visually summarize these therapeutic approaches.

Figure 2
www.frontiersin.org

Figure 2. Immunomodulatory strategies and hepatic immune tolerance. (A) Major immunosuppressive approaches used to reduce anti-transgene immune responses, categorized by their main cellular targets. (B) Liver's role in immune tolerance mediated by key resident cells such as Kupffer cells and liver sinusoidal endothelial cells (LSECs). Created in BioRender. Wang, Y. (2025) https://BioRender.com/r8hrxah.

4.3.2 Hepatic immune tolerance in AAV-mediated gene therapy

Moreover, hepatic gene transfer is getting more attention in the field of immune tolerance. The liver's inherent tolerogenic nature, due to constant exposure to non-self antigens, makes it an ideal target for inducing systemic immune tolerance through gene therapy, preventing uncontrolled immune activation (119). Studies have shown that liver-directed AAV gene transfer can not only reduce systemic immune responses but also eradicate pre-existing antibodies against therapeutic proteins, such as factor VIII in hemophilia A models, highlighting the liver's role in establishing durable immune tolerance (62, 120). Therefore, liver becomes a preferred target organ for gene therapy not only for liver-specific diseases but also for disorders that require systemic delivery (121). Studies using AAV-mediated gene transfer to liver in mice and primate models indicated that hepatocyte-restricted transgene expression can induce a robust, antigen-specific peripheral tolerance (122, 123). Tolerance is induced through a combination of mechanisms. One mechanism is that Kupffer cells, a type of antigen presenting cells (APCs) resident in the liver, seem to have a less mature phenotype compared to other professional APCs (124, 125), thus leading to poor T cell-activation. Besides, Kupffer cells secret the anti-inflammatory cytokine IL-10, contributing to the suppression of CD8+ T cell responses (126, 127). Another important factor is liver sinusoidal endothelial cells (LSECs), who also act as professional APCs (128, 129). LSECs promote tolerance through the induction of T regulatory cells (Tregs), whose depletion in mice observed increased transgene immunogenicity (130, 131). Additional mechanisms include CD4+T cell anergy (132), T cell degradation (133) were proposed in the establishment and maintenance of liver tolerance. These key cellular mechanisms are visually summarized in Figure 2B. Overall, these findings highlight the complex interplay between innate and adaptive immunity in response to AAV vectors. Understanding how early innate signals shape long-term adaptive outcomes is essential for designing safer and more effective gene therapies. Substantially more studies are needed to address adverse immune responses.

5 Preclinical and clinical studies of viral vector delivery for neurodegenerative diseases treatment

In recent years, many preclinical studies and clinical trials on the application of viral vector delivery for neurodegenerative diseases have been conducted. We detail the specific pathogens and therapeutic genes involved in each disease and discuss the outcomes achieved by each way. Table 2 provides a comprehensive summary of clinical studies on neurodegenerative diseases. It is worth mentioning that, while this summary provides a broad perspective, viral vector-based gene therapy has been expanded to address diseases beyond the scope discussed here.

Table 2
www.frontiersin.org

Table 2. Summary of clinical studies on neurodegenerative diseases.

5.1 Alzheimer's disease (AD)

As a progressive neurodegenerative disorder, the earliest phase of Alzheimer's disease happens in parallel with accumulating amyloid β, inducing the spread of tau pathology. More than 40 Alzheimer's disease-associated genetic risk loci already have been identified, of which the APOE alleles have the strongest association with the disease (134). Preclinical and clinical studies have explored various strategies through viral vector delivery for the treatment of AD, including risky genes and related pathways.

5.1.1 Targeting APOE

The common apolipoprotein E (APOE) alleles (ε4, ε3, and ε2) are important genetic risk factors for late-onset Alzheimer's disease. The primary physiological function of apoE is to mediate lipid transport in the brain and periphery (135). However, pathogenically, apoE seeds amyloid-β (Aβ) plaques in the brain with apoE4 driving earlier and more abundant amyloids (135). Studies have shown that the APOE4 allele is the strongest genetic risk factor for late-onset AD (136), increasing the risk up to 15-fold in homozygous individuals (137), whereas APOE2 is associated with a reduced risk and delayed onset of AD (138). In the brain, apoE is produced mostly by astrocytes. In one study, AAV8-GFAP-apoE, which is astrocyte-specific, was injected into apoE3-targeted replacement (apoE3-TR) or apoE4-targeted replacement (apoE4-TR) mice. In the apoE4-TR background, apoE4 decreases apoE lipidation and enhances Aβ accumulation, whereas apoE2 has the opposite effects (139). This indicates the therapeutic potential of APOE2 in the treatment of AD. In another study, AAV gene delivery of APOE2 using an AAV vector rescues the detrimental effects of APOE4 on brain amyloid pathology (140). Furthermore, Günaydin et al. have recently found that AAVrh10 delivery of novel APOE2-Christchurch (APOE2Ch) variant can better suppress amyloid and Tau pathology in “amyloid mice” and “tau mice” than APOE2 (141), which makes APOE2Ch variant a promising therapy for APOE4-associated AD. A clinical trial (NCT03634007) using AAVrh10hAPOE2 (LX1001) to treat patients with APOE4 homozygote Alzheimer's Disease was completed last year, including 15 patients. A recent interim report from the Phase 1/2 trial, showed favorable biomarker responses and good tolerability in APOE4 homozygous Alzheimer's patients, supporting continued clinical development, with further results eagerly anticipated to assess long-term efficacy.

5.1.2 Expressing neurotrophic factors

Neurotrophic factors, including BDNF and NGF, have been shown to exert neuroprotective functions for AD (142). Microglia activation and reactive oxygen species (ROS) inhibition may exert neuroprotective effects against Aβ-induced neurotoxicity through NGF/TrkA signaling (143, 144). In a preclinical study, AAV2-NGF(CERE-110) was shown to be neuroprotective and neurorestorative to basal forebrain cholinergic neurons in the rat fimbria-fornix lesion and aged rat models (145). As compared to AAV2 vector, AAV2/5 vectors consistently showed higher transduction efficiency. Nagahara et al. used lentiviral gene delivery of NGF to the aged primate basal forebrain restoring cholinergic neuronal markers significantly (146). These experiments conducted in both rats and monkeys, led to the initiation of a Phase I clinical study to evaluate the safety and efficacy of NGF in Alzheimer's disease subjects. A pilot phase 1 clinical trial on 49 AD participants, AAV2-NGF was safe and well-tolerated through 24 months, but did not improve cognition (147). Further analysis of this trial revealed that it was not able to induce the cholinergic pathways which were needed for the improvement of cognition in AD patients, due to the limited spread of the injected AAV2- vectors coupled with incorrect stereotactic targeting (147). More research may be needed to find more suitable vectors to improve AAV transfection efficiency in the future.

BDNF is the most abundant neurotrophic factor in the adult brain, and its levels decline in the entorhinal cortex in AD (148). BDNF is usually combined with TrkB (149), and the BDNF/TrkB signaling promotes the accumulation of amyloid precursor protein (APP) thereby inhibiting amyloid cleavage and reducing Aβ production (150). MRI-guided infusion of AAV2-BDNF to the entorhinal cortex of the non-human primate resulted in safe and accurate targeting and distribution of BDNF to both the entorhinal cortex and the hippocampus (151). This encouraging result was further translated to a phase 1 study on early Alzheimer's Disease and mild cognitive Impairment (NCT05040217), which aims to assess the safety and distribution of AAV2-BDNF, potentially offering a disease-modifying intervention by restoring neurotrophic support to affected brain regions. This trial is currently ongoing, with surgical treatments for the Alzheimer's cohort expected to be completed by December 2027. These studies highlight the therapeutic potential of NGF and BDNF gene delivery in AD, although limitations in vector spread and targeting underscore the need for optimized delivery strategies in future clinical applications.

5.1.3 Expressing TERT

Telomerase reverse transcriptase (TERT) is the enzyme responsible for maintenance of the length of telomeres (152). TERT haploinsufficiency decreases BDNF and increases amyloid-β (Aβ) precursor in murine brain (153). While increased levels of TERT in AD mouse models results in reduced Aβ accumulation, improved spine morphology, and preserved cognitive function. As an attractive target widely explored in cancer research, telomerase has gained attention for its role in neurodegeneration. A preclinical study investigating the modulation of TERT in both adult and old mice, delays physiological aging and extends longevity through AAV9-TERT gene therapy (154). Paving the way for AD patients in a phase I clinical trial currently underway, both IV and IT delivery of AAV encoding TERT are being evaluated (NCT04133454), aiming to assess safety and transgene expression in patients with mild-to-moderate AD.

5.2 Parkinson's disease (PD)

Parkinson's disease (PD) is one of the common neurodegenerative diseases in middle-aged and elderly people, characterized by loss of dopaminergic neurons in the substantia nigra (SN). The focal nature of SN pathology in PD has been long considered amenable to gene therapy. 3–5% of Parkinson's disease is explained by genetic causes linked to known Parkinson's disease genes, whereas 90 genetic risk variants collectively explain 16–36% of the heritable risk of polygenic Parkinson's disease (47), leading to approaches that can target genes directly linked to the disease.

5.2.1 Expressing AADC

Aromatic L-amino acid decarboxylase (AADC) is an enzyme that is crucial for the synthesis of dopamine from its precursor, L-DOPA. The absence of AADC reduces dopamine synthesis in the brain. Enhanced L-DOPA to dopamine conversion leads to restored motor function and a measurable behavioral outcome in animal models of Parkinson's disease by AAV-AADC transduction (155). After infusion into parkinsonian nonhuman primate (NHP) putamen, AADC transgene expression remained unchanged at the 8-year point with no signs of neuroinflammation and other side effects (156). Transitioning from preclinical models to clinical trials, the AAV-AADC gene therapy has also shown encouraging outcomes. Magnetic resonance imaging-guided phase 1 trial of AAV2-hAADC(VY-AADC01) gene therapy for Parkinson's disease have demonstrated safety, stable expression for up to 3 years, and modest improvement in symptoms (157159) (NCT01973543). In another clinical trial(NCT00627588 and NCT01856439), ProSavin, a lentiviral vector-based AADC gene therapy delivering all three rate-limiting enzymes (TH, AADC, and GCH1) were administered to patients with advanced Parkinson's disease, motor scores of Unified Parkinson's Disease Rating Scale (UPDRS) were significantly improved at 6-month follow-up (160).

It is worth noting that, AADC gene therapy has also been used in patients with AADC deficiency. Delivery of AAV2-hAADC to patients with AADC deficiency has been proven to be safe and effective in several clinical trials (161, 162). These encouraging results have led eladocagene exuparvovec (AAV2-AADC) to be the first FDA-approved gene therapy for the treatment of AADC deficiency. These clinical experiences in AADC deficiency have also informed dosing strategies and delivery approaches for Parkinson's disease applications. Together, these findings support the continued development of AAV2-AADC gene therapy for both Parkinson's disease and AADC deficiency.

5.2.2 Expressing neurotrophic factors

Neurotrophic factors like GDNF and NRTN support neurons' growth, survival, and differentiation. GDNF, in particular, is essential for supporting dopaminergic neuron survival (163166). Preclinical studies in both rodent and primate models have shown that AAV2-GDNF delivery to the substantia nigra and striatum increases dopaminergic neuron survival and improves motor function. In MPTP-treated rats, AAV-GDNF preserved striatal synaptic plasticity and reduced rotational asymmetry (165). In parkinsonian monkeys, AAV-GDNF enhanced dopamine activity and improved motor coordination without inducing toxicity (164, 166). Based on these findings, a Phase 1 clinical trial (NCT01621581) involving 25 patients reported that AAV2-GDNF delivery was safe and well tolerated across three escalating doses. Although the trial confirmed consistent GDNF expression, it did not provide conclusive evidence of clinical efficacy. An ongoing study (NCT04167540) is expected to further evaluate therapeutic benefit and optimal dosing. Together, these results highlight the translational potential of GDNF gene therapy for Parkinson's disease, while also emphasizing the need for further clinical validation.

Neurturin (NRTN), a member of the GDNF family of neurotrophic factors with known potential to protect and restore the function of dopaminergic substantia nigra neurons. Striatal delivery of AAV2-NRTN (CERE-120) to aged rhesus monkeys enhances activity of the dopaminergic nigrostriatal system, as indicated by increased 18F-fluorodopa uptake and tyrosine hydroxylase expression (167). CERE-120 is also proven to provide structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys, leading to improvements in motor function (168). However, AAV2-neurturin delivery to the putamen and substantia nigra bilaterally in PD patients showed no significant benefit over sham surgery, possibly due to impaired retrograde transport in advanced disease stages (169). These findings highlight the discrepancy between preclinical and clinical outcomes, emphasizing the importance of timing and delivery strategies in gene therapy for PD.

5.2.3 Expressing GAD

Glutamic acid decarboxylase (GAD) is an enzyme catalyzing the synthesis of GABA. GABA depletion may contribute to increased motor symptoms (170, 171), and non-motor symptoms are also related to the dysfunction of the GABAergic pathway (170).

In preclinical studies, AAV-mediated GAD expression in the subthalamic nucleus (STN) of rat and NHP models ameliorates parkinsonian behavioral phenotype (172, 173), Additionally, it demonstrated neuroprotective effects on nigral dopamine neurons, suggesting broader disease-modifying potential (172). These encouraging preclinical outcomes also lead to following phase I clinical trials (NCT00195143), in which AAV-GAD gene therapy for Parkinson's disease has proven to be safe and tolerable (174). In a phase 2 randomized trial (NCT00643890), patients in the AAV2-GAD group exhibited significantly greater improvement in motor function, as measured by UPDRS Part III scores, at both 6 and 12 months compared to the sham group (54, 175). Further study of the mechanism underlying this trail has found that only the patients who received GAD gene therapy developed a unique treatment-dependent polysynaptic brain circuit, termed as the GAD-related pattern (GADRP), which could be useful in future clinical trials for isolating true treatment responses (176). These results support the potential of GAD gene therapy not only to alleviate motor symptoms but also to offer insight into circuit-level changes associated with therapeutic efficacy.

5.2.4 Targeting disease gene: GBA1

Variants in the GBA1 gene, which encodes lysosomal acid glucocerebrosidase (GCase), are among the most common genetic risk factors for Parkinson's disease and are associated with earlier onset, faster disease progression, and more severe non-motor symptoms (177). In preclinical studies, rAAV9-GBA1 was unilaterally delivered to the substantia nigra pars compacta (SNpc) in mice and NHPs. This treatment led to increased GCase activity, promoted clearance of alpha-synuclein aggregates, and enhanced survival of dopaminergic neurons, indicating potential disease-modifying effects in PD (178, 179).

Building on these findings, a Phase 1/2 clinical study (NCT04127578) using AAV9-GBA1 (LY3884961) delivered via intra-cisterna magna injection is currently ongoing to assess its safety, tolerability, and biological activity in GBA1-associated PD patients. These developments suggest that targeting GBA1 by AAV-mediated gene delivery may offer a promising therapeutic strategy for a well-defined genetic subgroup of PD, although clinical efficacy remains to be validated.

5.3 Huntington disease (HD)

Huntington's disease is a progressive, fatal, neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin gene(HTT), which encodes an abnormally long polyglutamine (polyQ) repeat in the huntingtin protein (180). The mutant HTT protein causes selective neuronal degeneration, especially in the striatum and cortex (180). Therefore, directly targeting the HTT gene using viral vectors offers a rational and potentially disease-modifying therapeutic approach.

5.3.1 Targeting HTT

As a monogenic inheritance of Huntington's disease, HTT is an appealing candidate for its gene therapy. Besides, miRNA dysregulation has been consistently reported in HD patients, transgenic HD mice, and in in vitro experimental models (181184). It was also found that downregulation was dominant in the abnormal miRNA expressions (183). Consequently, delivering microRNAs that silence the mutant HTT gene has been widely studied in preclinical studies. Spronck et al. (185) performed intrastriatal injection of an AAV expressing a miRNA targeting humans HTT (AAV5-miHTT) at different doses in Q175 knock-in mice, reduced mutant HTT protein levels, and improved motor performance. Fukuoka et al. (186) introduced AAV-miR132 into the striatum of R6/2 mice, enhancing synaptic plasticity, and improving neuronal function. Keeler et al. (187) performed bilateral intrastriatal injection of AAV9-GFP-miRHtt in Q140/Q140 knock-in mice and enhanced striatal neuron survival. The animal models mentioned above are commonly used transgenetic models of Huntington's disease, and all of them are able to exhibit significant HD-like phenotypes. These strategies showed a decrease in mutant huntingtin mRNA and protein levels, lowered the mutant huntingtin aggregates in striatum and cortex, improved performance in behavioral tests, and slowed down disease progression. From the bench to the clinic, rAAV5-miHTT (AMT-130) is currently being investigated in Phase 1/2 clinical trials (NCT04120493, NCT05243017) for adults with early manifest HD. This trial involves stereotactic intrastriatal delivery and aims to evaluate safety, biodistribution, and preliminary efficacy. Together, these findings underscore the potential of AAV-mediated HTT-lowering therapy to slow disease progression in HD, especially when initiated in the early stages.

5.3.2 Expressing CYP46A1

Increasing evidence suggests that CYP46A1, a gene encoding a member of the cytochrome P450 superfamily enzymes, has a role in the pathogenesis and progression of neurodegenerative disorders, and that increasing its levels in the brain is neuroprotective (188, 189). This approach is grounded in its potential to modulate cholesterol catabolism, leading to the conversion of cholesterol into 24 (S)-hydroxy-cholesterol (24S-OHC), a brain-specific oxysterol involved in cholesterol turnover. In preclinical studies, AAV-CYP46A1 infection in model mice for Huntington's disease restored cholesterol homeostasis, restored cholesterol homeostasis, enhanced cholesterol turnover, and normalized neuronal function (190, 191). Moreover, this intervention increased levels of lanosterol and desmosterol, which were found to protect striatal neurons expressing Exp-HTT from death in vitro (191). These findings support the hypothesis that modulating cholesterol metabolism may counteract HD pathogenesis by restoring neuronal homeostasis and enhancing cell survival. A Phase 1 trial (NCT05541627) of intracerebral bilateral injections of AAVrh10.CAG.hCYP46A1 (AB-1001) within the striatum in early manifest patients is currently ongoing.

5.4 Frontotemporal dementia (FTD)

GRN mutations cause frontotemporal dementia (GRN-FTD) due to deficiency in progranulin (PGRN), a lysosomal and secreted protein with unclear function, which plays roles in lysosomal homeostasis, neuronal survival, and inflammation regulation (192). Deficiency of the gene GRN results in gangliosidosis in frontotemporal dementia, causes neurodegeneration through lysosomal dysfunction, defects in autophagy, and neuroinflammation (193, 194).

Preclinical studies performed AAV-driven expression of progranulin to animal model of frontotemporal dementia due to GRN mutations (195, 196). Results have shown that it not only increased progranulin levels in the cerebrospinal fluid, but also normalized histological and biochemical markers of progranulin deficiency, including reduced lipofuscin accumulation and improved lysosomal enzyme activity (195, 196). These studies support the translation of GRN gene therapy for FTD from preclinical studies into the clinic. Interim results of a phase 1/2 trial showed preliminary insights into the safety and bioactivity of PR006 (AAV90-GRN), demonstrating increased CSF progranulin and reduced neurofilament light chain (NfL), a biomarker of neurodegeneration, while its long-term safety and potential efficacy remain to be confirmed in ongoing studies (NCT04408625, NCT04747431) (195).

5.5 Other neurodegenerative diseases

In other neurodegenerative diseases, GDNF can also be used in patients with Multiple System Atrophy (MSA), a rare and rapidly progressive disease involving multiple brain systems, as it has a trophic effect on the Purkinje cells (197). A Phase 1 clinical trial delivering AAV2-GDNF to the putamen in patients with either a possible or probable diagnosis of MSA has been ongoing since 2023 to evaluate its safety and potential clinical effect (NCT04680065). Amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease affecting motor neurons, has also been a target of experimental GDNF-based therapies. Delivering a retroviral vector encoding GDNF into hind limb muscles of rodent models of ALS increased motor neuron survival and delayed the “ALS-related” decline in performance on motor tests, demonstrating functional benefits in preclinical models (198). Injection of AAV-GDNF showed evidence of retrograde transport of GDNF to motor neurons and impeded neurodegeneration in transgenic models of ALS (199). In addition to GDNF, preclinical studies using AAV-IGF1 in rodent models of ALS have also shown a significant reduction in motor neuron loss and improvement in behavior, particularly in motor function and survival extension in some models (200202). However, further clinical translational studies are needed to assess the safety and efficacy of these approaches in humans.

In addition to adult-onset neurodegenerative disorders, recent clinical advancements in CNS-targeted gene therapy have been largely driven by rare monogenic pediatric diseases that share progressive neurodegenerative characteristics. Among them, spinal muscular atrophy (SMA) has become a flagship example of CNS-targeted gene therapy, with the FDA-approved AAV9-based Zolgensma demonstrating significant clinical efficacy. In the pivotal trial NCT02122952, systemic administration of Zolgensma, an AAV9–mediated gene therapy targeting the SMN1 gene, led to marked improvements in motor function and survival in SMA1 patients (203), findings further supported by a comparative study with a natural history cohort (204). The success of Zolgensma was further confirmed in the phase III SPR1NT trials, where presymptomatic infants with two or three copies of SMN2 achieved age-appropriate motor milestones, such as sitting and walking, underscoring the benefits of early intervention (205207). However, the application of AAV9-based gene therapy also presents immunological challenges. In clinical trials including STR1VE and SPR1NT, immune-mediated adverse events such as transient hepatotoxicity and thrombocytopenia were commonly reported. These responses are likely due to host reactions against the AAV9 capsid or transgene product. To manage this, all patients received prophylactic corticosteroids (typically prednisolone), with liver function and hematologic parameters closely monitored and treatment adjusted accordingly (205, 207). Beyond SMA, Gene therapies for mucopolysaccharidoses (MPS, e.g., NCT03580083) and Rett syndrome (e.g., NCT06856759) are actively being explored. While most treatments remain in early or mid-stage trials with limited clinical efficacy so far, these efforts provide important proofs of concept for CNS-targeted gene therapy. Their progress offers valuable lessons on delivery strategies and immune response management, paving the way for future applications in adult neurodegenerative diseases.

6 Discussion

Immune responses-mediated toxicity caused by viral vectors affects the efficacy of gene therapy. Although immunosuppression has been used successfully to blunt some of the viral vector-induced immune responses, it has failed in other cases. Thus, the design of more effective and less immunogenic viral vectors is needed, for example by incorporating DNA-depleted AAV capsids (empty capsids) (208, 209). In addition, further optimized viral vectors could aim to improve the efficiency of transduction and the stability and persistence of gene expression, thus significantly expanding the application scope of viral vectors. With the rapid development of gene therapy, gene editing technologies have played a crucial role. Viral vectors are able to precisely deliver gene editing tools, such as CRISPR-Cas9, to target cells, particularly in in vivo experiments (210, 211). In this way, researchers can precisely manipulate specific genes, such as knocking out harmful genes, inserting functional ones or repairing mutations. This not only broadens the potential applications of gene editing technology but also plays an important role in basic research and disease treatment. Traditional strategies to deliver viral vectors lack spatial precision and thus cannot target regionally defined neural circuits. New and non-invasive methods, like FUS-BBBO, are increasingly being adopted and more novel delivery methods are to be explored.

As the safety and efficacy of virus as a vector are established, future studies examining vector-mediated delivery of different molecules for treating different neurodegenerative diseases is to be proved. The promising results from preclinical studies and the encouraging data from ongoing clinical trials provide a hopeful outlook for the application of gene therapy in neurological diseases. Still, significant challenges need to be resolved before gene therapy for neurodegenerative diseases becomes widely accepted. In some diseases, although some diseases have reproducibly positive data in preclinical models, clinical trials didn't show significant results. It remains to be determined if lack of success in clinical trials reflects a lack of biological effect of intervention, limitations of the technical delivery methods or failure to reverse the course of the advanced disease at the time of intervention. For neurodegenerative diseases, the disease may have progressed to a point with limited possibilities of intervention, that's why early diagnosis is crucial for attaining a better outcome. Since available clinical trials have limited follow-up data, future research should focus on long-term studies to better understand the efficacy and safety of these therapies.

7 Conclusion

In summary, we can see immune responses after vector administration and immune system-mediated toxicity continue to challenge the success of gene transfer by viral vectors especially when high doses are required to correct the targeted genetic disease. Some novel approaches, such as hepatic gene transfer, can mitigate immune toxicity to a certain extent. Given that numerous mechanisms and molecules have been extensively studied as potential protective interventions for neurodegenerative diseases, viral vector-mediated gene therapies, particularly those utilizing AAV vectors, have emerged as a highly promising approach in both preclinical and clinical studies. Before gene therapy for neurodegenerative diseases can be widely adopted, significant hurdles must be overcome by improving evidence-based strategies, mitigating immune-related adverse effects, and conducting larger clinical trials with longer follow-up periods.

Author contributions

YW: Writing – original draft, Writing – review & editing. SM: Writing – original draft, Writing – review & editing. FL: Supervision, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

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

Generative AI statement

The author(s) declare that no Gen AI was used in the creation of this manuscript.

Publisher's note

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.

References

1. Cheng Y-J, Lin C-H, Lane H-Y. From menopause to neurodegeneration-molecular basis and potential therapy. Int J Mol Sci. (2021) 22:8654. doi: 10.3390/ijms22168654

PubMed Abstract | Crossref Full Text | Google Scholar

2. Agnello L, Ciaccio M. Neurodegenerative diseases: from molecular basis to therapy. Int J Mol Sci. (2022) 23:12854. doi: 10.3390/ijms232112854

PubMed Abstract | Crossref Full Text | Google Scholar

3. Lim ST, Airavaara M, Harvey BK. Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res. (2022) 61:14–26. doi: 10.1016/j.phrs.2009.10.002

PubMed Abstract | Crossref Full Text | Google Scholar

4. Ghosh S, Brown AM, Jenkins C, Campbell K. Viral vector systems for gene therapy: a comprehensive literature review of progress and biosafety challenges. Appl Biosaf. (2020) 25:7–18. doi: 10.1177/1535676019899502

PubMed Abstract | Crossref Full Text | Google Scholar

5. Zhao Z, Anselmo AC, Mitragotri S. Viral vector-based gene therapies in the clinic. Bioeng Transl Med. (2022) 7:e10258. doi: 10.1002/btm2.10258

PubMed Abstract | Crossref Full Text | Google Scholar

6. Ertl HCJ. Immunogenicity and toxicity of AAV gene therapy. Front Immunol. (2022) 13:975803. doi: 10.3389/fimmu.2022.975803

PubMed Abstract | Crossref Full Text | Google Scholar

7. Patel RV, Nanda P, Richardson RM. Neurosurgical gene therapy for central nervous system diseases. Neurotherapeutics. (2024) 21:e00434. doi: 10.1016/j.neurot.2024.e00434

PubMed Abstract | Crossref Full Text | Google Scholar

8. Ingusci S, Verlengia G, Soukupova M, Zucchini S, Simonato M. Gene therapy tools for brain diseases. Front Pharmacol. (2019) 10:724. doi: 10.3389/fphar.2019.00724

PubMed Abstract | Crossref Full Text | Google Scholar

9. Li X, Le Y, Zhang Z, Nian X, Liu B, Yang X. Viral vector-based gene therapy. Int J Mol Sci. (2023) 24:7736. doi: 10.3390/ijms24097736

PubMed Abstract | Crossref Full Text | Google Scholar

10. Waddington SN, McVey JH, Bhella D, Parker AL, Barker K, Atoda H, et al. Adenovirus serotype 5 hexon mediates liver gene transfer. Cell. (2008) 132:397–409. doi: 10.1016/j.cell.2008.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

11. Stasiak AC, Stehle T. Human adenovirus binding to host cell receptors: a structural view. Med Microbiol Immunol. (2020) 209:325–33. doi: 10.1007/s00430-019-00645-2

PubMed Abstract | Crossref Full Text | Google Scholar

12. Lee CS, Bishop ES, Zhang R, Yu X, Farina EM, Yan S, et al. Adenovirus-mediated gene delivery: potential applications for gene and cell-based therapies in the new era of personalized medicine. Genes Dis. (2017) 4:43–63. doi: 10.1016/j.gendis.2017.04.001

PubMed Abstract | Crossref Full Text | Google Scholar

13. Gaggar A, Shayakhmetov DM, Lieber A. CD46 is a cellular receptor for group B adenoviruses. Nat Med. (2003) 9:1408–12. doi: 10.1038/nm952

PubMed Abstract | Crossref Full Text | Google Scholar

14. Le Gal La Salle G, Robert JJ, Berrard S, Ridoux V, Stratford-Perricaudet LD, Perricaudet M, et al. An adenovirus vector for gene transfer into neurons and glia in the brain. Science. (1993) 259:988–90. doi: 10.1126/science.8382374

PubMed Abstract | Crossref Full Text | Google Scholar

15. Schaack J. Induction and inhibition of innate inflammatory responses by adenovirus early region proteins. Viral Immunol. (2005) 18:79–88. doi: 10.1089/vim.2005.18.79

PubMed Abstract | Crossref Full Text | Google Scholar

16. Islam A, Tom VJ. The use of viral vectors to promote repair after spinal cord injury. Exp Neurol. (2022) 354:114102. doi: 10.1016/j.expneurol.2022.114102

PubMed Abstract | Crossref Full Text | Google Scholar

17. The promise and potential hazards of adenovirus gene therapy - PubMed. Available online at: https://pubmed.ncbi.nlm.nih.gov/11302979/ (accessed May 30, 2025).

Google Scholar

18. Alba R, Bosch A, Chillon M. Gutless adenovirus: last-generation adenovirus for gene therapy. Gene Ther. (2005) 12:S18–27. doi: 10.1038/sj.gt.3302612

PubMed Abstract | Crossref Full Text | Google Scholar

19. Adeno-associated Adeno-associated virus at 50: a golden anniversary of discovery, research, and gene therapy success–a personal perspective - PubMed. Available online at: https://pubmed.ncbi.nlm.nih.gov/25807962/ (accessed May 30, 2025).

Google Scholar

20. McIntosh NL, Berguig GY, Karim OA, Cortesio CL, De Angelis R, Khan AA, et al. Comprehensive characterization and quantification of adeno associated vectors by size exclusion chromatography and multi angle light scattering. Sci Rep. (2021) 11:3012. doi: 10.1038/s41598-021-82599-1

PubMed Abstract | Crossref Full Text | Google Scholar

21. Kaplitt MG, Xiao X, Samulski RJ, Li J, Ojamaa K, Klein IL, et al. Long-term gene transfer in porcine myocardium after coronary infusion of an adeno-associated virus vector. Ann Thorac Surg. (1996) 62:1669–76. doi: 10.1016/S0003-4975(96)00946-0

PubMed Abstract | Crossref Full Text | Google Scholar

22. Wang AY, Peng PD, Ehrhardt A, Storm TA, Kay MA. Comparison of adenoviral and adeno-associated viral vectors for pancreatic gene delivery in vivo. Hum Gene Ther. (2004) 15:405–13. doi: 10.1089/104303404322959551

PubMed Abstract | Crossref Full Text | Google Scholar

23. Cearley CN, Wolfe JH. Transduction characteristics of adeno-associated virus vectors expressing cap serotypes 7, 8, 9, and Rh10 in the mouse brain. Mol Ther. (2006) 13:528–37. doi: 10.1016/j.ymthe.2005.11.015

PubMed Abstract | Crossref Full Text | Google Scholar

24. Han Z, Luo N, Ma W, Liu X, Cai Y, Kou J, et al. AAV11 enables efficient retrograde targeting of projection neurons and enhances astrocyte-directed transduction. Nat Commun. (2023) 14:3792. doi: 10.1038/s41467-023-39554-7

PubMed Abstract | Crossref Full Text | Google Scholar

25. Burger C, Gorbatyuk OS, Velardo MJ, Peden CS, Williams P, Zolotukhin S, et al. Recombinant AAV viral vectors pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. Mol Ther. (2004) 10:302–17. doi: 10.1016/j.ymthe.2004.05.024

PubMed Abstract | Crossref Full Text | Google Scholar

26. Foust KD, Nurre E, Montgomery CL, Hernandez A, Chan CM, Kaspar BK. Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nat Biotechnol. (2009) 27:59–65. doi: 10.1038/nbt.1515

PubMed Abstract | Crossref Full Text | Google Scholar

27. High KA, Anguela XM. Adeno-associated viral vectors for the treatment of hemophilia. Hum Mol Genet. (2016) 25:R36–41. doi: 10.1093/hmg/ddv475

PubMed Abstract | Crossref Full Text | Google Scholar

28. Brommel CM, Cooney AL, Sinn PL. Adeno-associated virus-based gene therapy for lifelong correction of genetic disease. Hum Gene Ther. (2020) 31:985–95. doi: 10.1089/hum.2020.138

PubMed Abstract | Crossref Full Text | Google Scholar

29. Crunkhorn S. Reducing AAV vector associated neurotoxicity. Nat Rev Drug Discov. (2021) 20:20. doi: 10.1038/d41573-020-00209-7

PubMed Abstract | Crossref Full Text | Google Scholar

30. High-dose AAV gene therapy deaths. Nat Biotechnol. (2020) 38:910. doi: 10.1038/s41587-020-0642-9

PubMed Abstract | Crossref Full Text | Google Scholar

31. Hinderer C, Katz N, Buza EL, Dyer C, Goode T, Bell P, et al. Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an adeno-associated virus vector expressing human. SMN Hum Gene Ther. (2018) 29:285–98. doi: 10.1089/hum.2018.015

PubMed Abstract | Crossref Full Text | Google Scholar

32. Wilson JM, Flotte TR. Moving forward after two deaths in a gene therapy trial of myotubular myopathy. Hum Gene Ther. (2020) 31:695–696. doi: 10.1089/hum.2020.182

PubMed Abstract | Crossref Full Text | Google Scholar

33. Vandamme C, Adjali O, Mingozzi F. Unraveling the complex story of immune responses to AAV vectors trial after trial. Hum Gene Ther. (2017) 28:1061–74. doi: 10.1089/hum.2017.150

PubMed Abstract | Crossref Full Text | Google Scholar

34. Louis Jeune V, Joergensen JA, Hajjar RJ, Weber T. Pre-existing anti-adeno-associated virus antibodies as a challenge in AAV gene therapy. Hum Gene Ther Methods. (2013) 24:59–67. doi: 10.1089/hgtb.2012.243

PubMed Abstract | Crossref Full Text | Google Scholar

35. Abordo-Adesida E, Follenzi A, Barcia C, Sciascia S, Castro MG, Naldini L, et al. Stability of lentiviral vector-mediated transgene expression in the brain in the presence of systemic antivector immune responses. Hum Gene Ther. (2005) 16:741–51. doi: 10.1089/hum.2005.16.741

PubMed Abstract | Crossref Full Text | Google Scholar

36. Kalidasan V, Ng WH, Ishola OA, Ravichantar N, Tan JJ, Das KT. A guide in lentiviral vector production for hard-to-transfect cells, using cardiac-derived c-kit expressing cells as a model system. Sci Rep. (2021) 11:19265. doi: 10.1038/s41598-021-98657-7

PubMed Abstract | Crossref Full Text | Google Scholar

37. Brown BD. A shot in the bone corrects a genetic disease. Mol Ther. (2015) 23:614–5. doi: 10.1038/mt.2015.38

PubMed Abstract | Crossref Full Text | Google Scholar

38. Nowak I, Madej M, Secemska J, Sarna R, Strzalka-Mrozik B. Virus-based biological systems as next-generation carriers for the therapy of central nervous system diseases. Pharmaceutics. (2023) 15:1931. doi: 10.3390/pharmaceutics15071931

PubMed Abstract | Crossref Full Text | Google Scholar

39. Zheng J, Chen D, Chan J, Yu D, Ko E, Pang S. Regression of prostate cancer xenografts by a lentiviral vector specifically expressing diphtheria toxin A. Cancer Gene Ther. (2003) 10:764–70. doi: 10.1038/sj.cgt.7700629

PubMed Abstract | Crossref Full Text | Google Scholar

40. Gerolami R, Uch R, Faivre J, Garcia S, Hardwigsen J, Cardoso J, et al. Herpes simplex virus thymidine kinase-mediated suicide gene therapy for hepatocellular carcinoma using HIV-1-derived lentiviral vectors. J Hepatol. (2004) 40:291–7. doi: 10.1016/j.jhep.2003.10.019

PubMed Abstract | Crossref Full Text | Google Scholar

41. Indraccolo S, Habeler W, Tisato V, Stievano L, Piovan E, Tosello V, et al. Gene transfer in ovarian cancer cells: a comparison between retroviral and lentiviral vectors. Cancer Res. (2002) 62:6099–107.

PubMed Abstract | Google Scholar

42. Osega CE, Bustos FJ, Arriagada G. From entry to the nucleus: how retroviruses commute. Annu Rev Virol. (2024) 11:89–104. doi: 10.1146/annurev-virology-100422-023502

PubMed Abstract | Crossref Full Text | Google Scholar

43. Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res. (2012) 1:27. doi: 10.4103/2277-9175.98152

PubMed Abstract | Crossref Full Text | Google Scholar

44. Sreeramoju P, Libutti SK. Strategies for targeting tumors and tumor vasculature for cancer therapy. Adv Genet. (2010) 69:135–52. doi: 10.1016/S0065-2660(10)69015-3

PubMed Abstract | Crossref Full Text | Google Scholar

45. Vile RG, Russell SJ. Retroviruses as vectors. Br Med Bull. (1995) 51:12–30. doi: 10.1093/oxfordjournals.bmb.a072941

PubMed Abstract | Crossref Full Text | Google Scholar

46. Hacein-Bey-Abina S, von Kalle C, Schmidt M, Le Deist F, Wulffraat N, McIntyre E, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. (2003) 348:255–6. doi: 10.1056/NEJM200301163480314

PubMed Abstract | Crossref Full Text | Google Scholar

47. Liu W, Liu Z, Liu L, Xiao Z, Cao X, Cao Z, et al. A novel human foamy virus mediated gene transfer of GAD67 reduces neuropathic pain following spinal cord injury. Neurosci Lett. (2008) 432:13–8. doi: 10.1016/j.neulet.2007.11.054

PubMed Abstract | Crossref Full Text | Google Scholar

48. Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, et al. Gene therapy for pain: results of a phase I clinical trial. Ann Neurol. (2011) 70, 207–212. doi: 10.1002/ana.22446

PubMed Abstract | Crossref Full Text | Google Scholar

49. Lachmann RH. Herpes simplex virus-based vectors. Int J Exp Pathol. (2004) 85:177–90. doi: 10.1111/j.0959-9673.2004.00383.x

PubMed Abstract | Crossref Full Text | Google Scholar

50. Fathi A, Dahlke C, Addo MM. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum Vaccin Immunother. (2019) 15:2269–85. doi: 10.1080/21645515.2019.1649532

PubMed Abstract | Crossref Full Text | Google Scholar

51. Simon JK, Kennedy SB, Mahon BE, Dubey SA, Grant-Klein RJ, Liu K, et al. Immunogenicity of rVSVΔG-ZEBOV-GP Ebola vaccine (ERVEBO®) in African clinical trial participants by age, sex, and baseline GP-ELISA titer: a post hoc analysis of three Phase 2/3 trials. Vaccine. (2022) 40:6599–606. doi: 10.1016/j.vaccine.2022.09.037

PubMed Abstract | Crossref Full Text | Google Scholar

52. Abdelmageed AA, Dewhurst S, Ferran MC. Employing the oncolytic vesicular stomatitis virus in cancer virotherapy: resistance and clinical considerations. Viruses. (2024) 17:16. doi: 10.3390/v17010016

PubMed Abstract | Crossref Full Text | Google Scholar

53. Fiandaca MS, Varenika V, Eberling J, McKnight T, Bringas J, Pivirotto P, et al. Real-time MR imaging of adeno-associated viral vector delivery to the primate brain. Neuroimage. (2009) 47:T27–35. doi: 10.1016/j.neuroimage.2008.11.012

PubMed Abstract | Crossref Full Text | Google Scholar

54. Niethammer M, Tang CC, LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, et al. Long-term follow-up of a randomized AAV2-GAD gene therapy trial for Parkinson's disease. JCI Insight. (2017) 2:e90133. doi: 10.1172/jci.insight.90133

PubMed Abstract | Crossref Full Text | Google Scholar

55. Marks WJ, Jr Bartus RT, Siffert J, Davis CS, Lozano A, Boulis N, et al. Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomized, controlled trial. Lancet Neurol. (2010) 9:1164–72. doi: 10.1016/S1474-4422(10)70254-4

PubMed Abstract | Crossref Full Text | Google Scholar

56. Gong J, Liu Y, Chung TH, Xu L, Lund TC, Chang LJ. Intracerebral lentiviral ABCD1 gene therapy in an early disease onset ALD mouse model. Gene Ther. (2023) 30:18–30. doi: 10.1038/s41434-022-00355-0

PubMed Abstract | Crossref Full Text | Google Scholar

57. Hocquemiller M, Castelein RM. AAVrh10 Vector Corrects Disease Pathology in MPS IIIA Mice and Achieves Widespread Distribution of SGSH in Large Animal Brains. Mol Ther Methods Clin Dev. (2020) 17:174–87. doi: 10.1016/j.omtm.2019.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

58. Sudhakar V, Richardson RM. Gene therapy for neurodegenerative diseases. Neurotherapeutics. (2019) 16:166–75. doi: 10.1007/s13311-018-00694-0

PubMed Abstract | Crossref Full Text | Google Scholar

59. Adeno-associated virus as a delivery vector for gene therapy of human diseases - PubMed. Available online at: https://pubmed.ncbi.nlm.nih.gov/38565561/ (accessed May 30, 2025).

Google Scholar

60. Weber-Adrian D, Heinen S, Silburt J, Noroozian Z, Aubert I. The human brain endothelial barrier: transcytosis of AAV9, transduction by AAV2: an editorial highlight for “Trafficking of adeno-associated virus vectors across a model of the blood-brain barrier; a comparative study of transcytosis and transduction using primary human brain endothelial cells.” J Neurochem. (2017) 140:192–4. doi: 10.1111/jnc.13898

PubMed Abstract | Crossref Full Text | Google Scholar

61. Merkel SF, Andrews AM, Lutton EM, Mu D, Hudry E, Hyman BT, et al. Trafficking of adeno-associated virus vectors across a model of the blood-brain barrier; a comparative study of transcytosis and transduction using primary human brain endothelial cells. J Neurochem. (2017) 140:216–30. doi: 10.1111/jnc.13861

PubMed Abstract | Crossref Full Text | Google Scholar

62. Ronzitti G, Gross D-A, Mingozzi F. Human immune responses to adeno-associated virus (AAV) vectors. Front Immunol. (2020) 11:670. doi: 10.3389/fimmu.2020.00670

PubMed Abstract | Crossref Full Text | Google Scholar

63. Mingozzi F, High KA. Immune responses to AAV in clinical trials. Curr Gene Ther. (2007) 7:316–24. doi: 10.2174/156652307782151425

PubMed Abstract | Crossref Full Text | Google Scholar

64. Shevtsova Z, Malik JMI, Michel U, Bähr M, Kügler S. Promoters and serotypes: targeting of adeno-associated virus vectors for gene transfer in the rat central nervous system in vitro and in vivo. Exp Physiol. (2005) 90:53–9. doi: 10.1113/expphysiol.2004.028159

PubMed Abstract | Crossref Full Text | Google Scholar

65. McLean JR, Smith GA, Rocha EM, Hayes MA, Beagan JA, Hallett PJ, et al. Widespread neuron-specific transgene expression in brain and spinal cord following synapsin promoter-driven AAV9 neonatal intracerebroventricular injection. Neurosci Lett. (2014) 576:73–8. doi: 10.1016/j.neulet.2014.05.044

PubMed Abstract | Crossref Full Text | Google Scholar

66. Chakrabarty P, Rosario A, Cruz P, Siemienski Z, Ceballos-Diaz C, Crosby K, et al. Capsid serotype and timing of injection determines AAV transduction in the neonatal mice brain. PLoS One. (2013) 8:e67680. doi: 10.1371/journal.pone.0067680

PubMed Abstract | Crossref Full Text | Google Scholar

67. Kim J-Y, Grunke SD, Levites Y, Golde TE, Jankowsky JL. Intracerebroventricular viral injection of the neonatal mouse brain for persistent and widespread neuronal transduction. J Vis Exp. (2014) 51863.

PubMed Abstract | Google Scholar

68. Ohno K, Castelein RM. Kinetics and MR-based monitoring of AAV9 vector delivery into cerebrospinal fluid of nonhuman primates. Mol Ther Methods Clin Dev. (2019) 13:47–54. doi: 10.1016/j.omtm.2018.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

69. Bey K, Deniaud J, Dubreil L, Joussemet B, Cristini J, Ciron C, et al. Intra-CSF AAV9 and AAVrh10 Administration in nonhuman primates: promising routes and vectors for which neurological diseases? Mol Ther Methods Clin Dev. (2020) 17:771–84. doi: 10.1016/j.omtm.2020.04.001

PubMed Abstract | Crossref Full Text | Google Scholar

70. Kordasiewicz HB, Stanek LM, Wancewicz EV, Mazur C, McAlonis MM, Pytel KA, et al. Sustained therapeutic reversal of Huntington's disease by transient repression of huntingtin synthesis. Neuron. (2012) 74:1031–44. doi: 10.1016/j.neuron.2012.05.009

PubMed Abstract | Crossref Full Text | Google Scholar

71. Katz N, Goode T, Hinderer C, Hordeaux J, Wilson JM. Standardized method for intra-cisterna magna delivery under fluoroscopic guidance in nonhuman primates. Hum Gene Ther Methods. (2018) 29:212–9. doi: 10.1089/hgtb.2018.041

PubMed Abstract | Crossref Full Text | Google Scholar

72. Ye D, Chukwu C, Yang Y, Hu Z, Chen H. Adeno-associated virus vector delivery to the brain: Technology advancements and clinical applications. Adv Drug Deliv Rev. (2024) 211:115363. doi: 10.1016/j.addr.2024.115363

PubMed Abstract | Crossref Full Text | Google Scholar

73. Qi B, Yang Y, Cheng Y, Sun D, Wang X, Khanna R, et al. Nasal delivery of a CRMP2-derived CBD3 adenovirus improves cognitive function and pathology in APP/PS1 transgenic mice. Mol Brain. (2020) 13:58. doi: 10.1186/s13041-020-00596-3

PubMed Abstract | Crossref Full Text | Google Scholar

74. Capsoni S, Cattaneo A. Getting into the brain: the intranasal approach to enhance the delivery of nerve growth factor and its painless derivative in Alzheimer's Disease and Down Syndrome. Front Neurosci. (2022) 16:773347. doi: 10.3389/fnins.2022.773347

PubMed Abstract | Crossref Full Text | Google Scholar

75. de Bellis A, de Bellis M, Aloe L. Long-term non-invasive treatment via intranasal administration of nerve growth factor protects the human brain in frontotemporal dementia associated with corticobasal syndrome: a pilot study. J Alzheimers Dis Rep. (2018) 2:67–77. doi: 10.3233/ADR-180055

PubMed Abstract | Crossref Full Text | Google Scholar

76. Gadenstaetter AJ, Schmutzler L, Grimm D, Landegger LD. Intranasal application of adeno-associated viruses: a systematic review. Transl Res. (2022) 248:87–110. doi: 10.1016/j.trsl.2022.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

77. Thévenot E, Jordão JF, O'Reilly MA, Markham K, Weng YQ, Foust KD, et al. Targeted delivery of self-complementary adeno-associated virus serotype 9 to the brain, using magnetic resonance imaging-guided focused ultrasound. Hum Gene Ther. (2012) 23:1144–55. doi: 10.1089/hum.2012.013

PubMed Abstract | Crossref Full Text | Google Scholar

78. Wang S, Olumolade OO, Sun T, Samiotaki G, Konofagou EE. Noninvasive, neuron-specific gene therapy can be facilitated by focused ultrasound and recombinant adeno-associated virus. Gene Ther. (2015) 22:104–10. doi: 10.1038/gt.2014.91

PubMed Abstract | Crossref Full Text | Google Scholar

79. Wang S, Kugelman T, Buch A, Herman M, Han Y, Karakatsani ME, et al. Non-invasive, Focused Ultrasound-Facilitated Gene Delivery for Optogenetics. Sci Rep. (2017) 7:39955. doi: 10.1038/srep39955

PubMed Abstract | Crossref Full Text | Google Scholar

80. Szablowski JO, Lee-Gosselin A, Lue B, Malounda D, Shapiro MG. Acoustically targeted chemogenetics for the non-invasive control of neural circuits. Nat Biomed Eng. (2018) 2:475–84. doi: 10.1038/s41551-018-0258-2

PubMed Abstract | Crossref Full Text | Google Scholar

81. Alonso A, Reinz E, Leuchs B, Kleinschmidt J, Fatar M, Geers B, et al. Focal delivery of AAV2/1-transgenes Into the rat brain by localized ultrasound-induced BBB opening. Mol Ther Nucleic Acids. (2013) 2:e73. doi: 10.1038/mtna.2012.64

PubMed Abstract | Crossref Full Text | Google Scholar

82. Ye D, Yuan J, Yang Y, Yue Y, Hu Z, Fadera S, et al. Incisionless targeted adeno-associated viral vector delivery to the brain by focused ultrasound-mediated intranasal administration. EBioMedicine. (2022) 84:104277. doi: 10.1016/j.ebiom.2022.104277

PubMed Abstract | Crossref Full Text | Google Scholar

83. Owusu-Yaw BS, Zhang Y, Garrett L, Yao A, Shing K, Batista AR, et al. Focused Ultrasound-Mediated Disruption of the Blood-Brain Barrier for AAV9 Delivery in a Mouse Model of Huntington's Disease. Pharmaceutics. (2024) 16:710. doi: 10.3390/pharmaceutics16060710

PubMed Abstract | Crossref Full Text | Google Scholar

84. Blesa J, Pineda-Pardo JA, Inoue KI, Gasca-Salas C, Balzano T, Del Rey NL, et al. BBB opening with focused ultrasound in nonhuman primates and Parkinson's disease patients: targeted AAV vector delivery and PET imaging. Sci Adv. (2023) 9:eadf4888. doi: 10.1126/sciadv.adf4888

PubMed Abstract | Crossref Full Text | Google Scholar

85. Erickson NJ, Stavarache M, Tekedereli I, Kaplitt MG, Markert JM. Herpes simplex oncolytic viral therapy for malignant glioma and mechanisms of delivery. World Neurosurg. (2025) 194:123595. doi: 10.1016/j.wneu.2024.123595

PubMed Abstract | Crossref Full Text | Google Scholar

86. Sáez-Cirión A, Manel N. Immune Responses to Retroviruses. Annu Rev Immunol. (2018) 36, 193–220. doi: 10.1146/annurev-immunol-051116-052155

PubMed Abstract | Crossref Full Text | Google Scholar

87. Rollier CS, Spencer AJ, Sogaard KC, Honeycutt J, Furze J, Bregu M, et al. Modification of Adenovirus vaccine vector-induced immune responses by expression of a signaling molecule. Sci Rep. (2020) 10:5716. doi: 10.1038/s41598-020-61730-8

PubMed Abstract | Crossref Full Text | Google Scholar

88. Zhang Y, Bourgine M, Wan Y, Song J, Li Z, Yu Y, et al. Therapeutic vaccination with lentiviral vector in HBV-persistent mice and two inactive HBsAg carriers. J Hepatol. (2024) 80:31–40. doi: 10.1016/j.jhep.2023.09.019

PubMed Abstract | Crossref Full Text | Google Scholar

89. Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. (2011) 30:16–34. doi: 10.3109/08830185.2010.529976

PubMed Abstract | Crossref Full Text | Google Scholar

90. 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

91. Herzog RW, Cooper M, Perrin GQ, Biswas M, Martino AT, Morel L, et al. Regulatory T cells and TLR9 activation shape antibody formation to a secreted transgene product in AAV muscle gene transfer. Cell Immunol. (2019) 342:103682. doi: 10.1016/j.cellimm.2017.07.012

PubMed Abstract | Crossref Full Text | Google Scholar

92. 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

93. Zhu, J., Huang, X., Yang, Y. The TLR9-MyD88 pathway is critical for adaptive immune responses to adeno-associated virus gene therapy vectors in mice. J Clin Invest. (2009) 119:2388–2398. doi: 10.1172/JCI37607

PubMed Abstract | Crossref Full Text | Google Scholar

94. 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

95. Xiang Z, Kurupati RK, Li Y, Kuranda K, Zhou X, Mingozzi F, et al. The effect of CpG sequences on capsid-specific CD8+ T cell responses to AAV vector gene transfer. Mol Ther. (2020) 28:771–83. doi: 10.1016/j.ymthe.2019.11.014

PubMed Abstract | Crossref Full Text | Google Scholar

96. Rogers GL, Suzuki M, Zolotukhin I, Markusic DM, Morel LM, Lee B, et al. Unique Roles of TLR9- and MyD88-dependent and -independent pathways in adaptive immune responses to AAV-mediated gene transfer. J Innate Immun. (2015) 7:302–14. doi: 10.1159/000369273

PubMed Abstract | Crossref Full Text | Google Scholar

97. Bertolini TB, Shirley JL, Zolotukhin I, Li X, Kaisho T, Xiao W, et al. Effect of CpG depletion of vector genome on CD8+ T cell responses in AAV gene therapy. Front Immunol. (2021) 12:672449. doi: 10.3389/fimmu.2021.672449

PubMed Abstract | Crossref Full Text | Google Scholar

98. Faust SM, Bell P, Cutler BJ, Ashley SN, Zhu Y, Rabinowitz JE, et al. CpG-depleted adeno-associated virus vectors evade immune detection. J Clin Invest. (2013) 123:2994–3001. doi: 10.1172/JCI68205

PubMed Abstract | Crossref Full Text | Google Scholar

99. Shao W, Earley LF, Chai Z, Chen X, Sun J, He T, et al. Double-stranded RNA innate immune response activation from long-term adeno-associated virus vector transduction. JCI Insight. (2018) 3:e120474. doi: 10.1172/jci.insight.120474

PubMed Abstract | Crossref Full Text | Google Scholar

100. Hui DJ, Edmonson SC, Podsakoff GM, Pien GC, Ivanciu L, Camire RM, et al. AAV capsid CD8+ T-cell epitopes are highly conserved across AAV serotypes. Mol Ther Methods Clin Dev. (2015) 2:15029. doi: 10.1038/mtm.2015.29

PubMed Abstract | Crossref Full Text | Google Scholar

101. Palaschak B, Marsic D, Herzog RW, Zolotukhin S, Markusic DM. An Immune-Competent Murine Model to Study Elimination of AAV-Transduced Hepatocytes by Capsid-Specific CD8+ T Cells. Mol Ther Methods Clin Dev. (2017) 5:142–52. doi: 10.1016/j.omtm.2017.04.004

PubMed Abstract | Crossref Full Text | Google Scholar

102. Nathwani AC, Rosales C, McIntosh J, Rastegarlari G, Nathwani D, Raj D, et al. Long-term safety and efficacy following systemic administration of a self-complementary AAV vector encoding human FIX pseudotyped with serotype 5 and 8 capsid proteins. Mol Ther. (2011) 19:876–85. doi: 10.1038/mt.2010.274

PubMed Abstract | Crossref Full Text | Google Scholar

103. Finn JD, Hui D, Downey HD, Dunn D, Pien GC, Mingozzi F, et al. Proteasome inhibitors decrease AAV2 capsid derived peptide epitope presentation on MHC class I following transduction. Mol Ther. (2010) 18:135–42. doi: 10.1038/mt.2009.257

PubMed Abstract | Crossref Full Text | Google Scholar

104. Pien GC, Basner-Tschakarjan E, Hui DJ, Mentlik AN, Finn JD, Hasbrouck NC, et al. Capsid antigen presentation flags human hepatocytes for destruction after transduction by adeno-associated viral vectors. J Clin Invest. (2009) 119:1688–95. doi: 10.1172/JCI36891

PubMed Abstract | Crossref Full Text | Google Scholar

105. 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

106. Selot R, Arumugam S, Mary B, Cheemadan S, Jayandharan G. R. Optimized AAV rh10 vectors that partially evade neutralizing antibodies during hepatic gene transfer. Front Pharmacol. (2017) 8:441. doi: 10.3389/fphar.2017.00441

PubMed Abstract | Crossref Full Text | Google Scholar

107. Tse LV, Klinc KA, Madigan VJ, Castellanos Rivera RM, Wells LF, Havlik LP, et al. Structure-guided evolution of antigenically distinct adeno-associated virus variants for immune evasion. Proc Natl Acad Sci U S A. (2017) 114:E4812–21. doi: 10.1073/pnas.1704766114

PubMed Abstract | Crossref Full Text | Google Scholar

108. 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

109. Calcedo R, Somanathan S, Qin Q, Betts MR, Rech AJ, Vonderheide RH, et al. Class I-restricted T-cell responses to a polymorphic peptide in a gene therapy clinical trial for α-1-antitrypsin deficiency. Proc Natl Acad Sci U S A. (2017) 114:1655–9. doi: 10.1073/pnas.1617726114

PubMed Abstract | Crossref Full Text | Google Scholar

110. Mingozzi F, Meulenberg JJ, Hui DJ, Basner-Tschakarjan E, Hasbrouck NC, Edmonson SA, et al. AAV-1-mediated gene transfer to skeletal muscle in humans results in dose-dependent activation of capsid-specific T cells. Blood. (2009) 114:2077–86. doi: 10.1182/blood-2008-07-167510

PubMed Abstract | Crossref Full Text | Google Scholar

111. Nathwani AC, Tuddenham EG, 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

112. Murphy SL, Li H, Mingozzi F, Sabatino DE, Hui DJ, Edmonson SA, et al. Diverse IgG subclass responses to adeno-associated virus infection and vector administration J Med Virol. (2009) 81:65–74. doi: 10.1002/jmv.21360

PubMed Abstract | Crossref Full Text | Google Scholar

113. 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) 9:4098. doi: 10.1038/s41467-018-06621-3

PubMed Abstract | Crossref Full Text | Google Scholar

114. Corti M, Elder M, Falk D, Lawson L, Smith B, Nayak S, et al. B-Cell Depletion is protective against Anti-AAV capsid immune response: a human subject case study. Mol Ther Methods Clin Dev. (2014) 1:14033. doi: 10.1038/mtm.2014.33

PubMed Abstract | Crossref Full Text | Google Scholar

115. Xiang Z, Kuranda K, Quinn W, Chekaoui A, Ambrose R, Hasanpourghai M, et al. The effect of rapamycin and ibrutinib on antibody responses to adeno-associated virus vector-mediated gene transfer. Hum Gene Ther. (2022) 33:614–24. doi: 10.1089/hum.2021.258

PubMed Abstract | Crossref Full Text | Google Scholar

116. Day JW, Mendell JR, Mercuri E, Finkel RS, Strauss KA, Kleyn A, et al. Clinical trial and postmarketing safety of Onasemnogene Abeparvovec Therapy. Drug Saf. (2021) 44:1109–19. doi: 10.1007/s40264-021-01107-6

PubMed Abstract | Crossref Full Text | Google Scholar

117. 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.

Google Scholar

118. 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. doi: 10.1182/blood-2018-99-115495

Crossref Full Text | Google Scholar

119. Keeler GD, Markusic DM, Hoffman BE. Liver induced transgene tolerance with AAV vectors. Cell Immunol. (2019) 342:103728. doi: 10.1016/j.cellimm.2017.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

120. Samelson-Jones BJ, Arruda VR. Translational Potential of Immune Tolerance Induction by AAV Liver-Directed Factor VIII Gene Therapy for Hemophilia A. Front Immunol. (2020) 11:618. doi: 10.3389/fimmu.2020.00618

PubMed Abstract | Crossref Full Text | Google Scholar

121. LoDuca PA, Hoffman BE, Herzog RW. Hepatic gene transfer as a means of tolerance induction to transgene products. Curr Gene Ther. (2009) 9:104–14. doi: 10.2174/156652309787909490

PubMed Abstract | Crossref Full Text | Google Scholar

122. Mingozzi F, Hasbrouck NC, Basner-Tschakarjan E, Edmonson SA, Hui DJ, Sabatino DE, et al. Modulation of tolerance to the transgene product in a nonhuman primate model of AAV-mediated gene transfer to liver. Blood. (2007) 110:2334–41. doi: 10.1182/blood-2007-03-080093

PubMed Abstract | Crossref Full Text | Google Scholar

123. Mingozzi F, Liu YL, Dobrzynski E, Kaufhold A, Liu JH, Wang Y, et al. Induction of immune tolerance to coagulation factor IX antigen by in vivo hepatic gene transfer. J Clin Invest. (2003) 111:1347–56. doi: 10.1172/JCI16887

PubMed Abstract | Crossref Full Text | Google Scholar

124. Pillarisetty VG, Shah AB, Miller G, Bleier JI, DeMatteo RP. Liver dendritic cells are less immunogenic than spleen dendritic cells because of differences in subtype composition. J Immunol. (2004) 172:1009–17. doi: 10.4049/jimmunol.172.2.1009

PubMed Abstract | Crossref Full Text | Google Scholar

125. You Q, Cheng L, Kedl RM, Ju C. Mechanism of T cell tolerance induction by murine hepatic Kupffer cells. Hepatology. (2008) 48:978–90. doi: 10.1002/hep.22395

PubMed Abstract | Crossref Full Text | Google Scholar

126. Knolle P, Schlaak J, Uhrig A, Kempf P, Meyer zum Büschenfelde KH, Gerken G. Human Kupffer cells secrete IL-10 in response to lipopolysaccharide (LPS) challenge. J Hepatol. (1995) 22:226–9. doi: 10.1016/0168-8278(95)80433-1

PubMed Abstract | Crossref Full Text | Google Scholar

127. Liu J, Yu Q, Wu W, Huang X, Broering R, Werner M, et al. TLR2 Stimulation Strengthens Intrahepatic Myeloid-Derived Cell-Mediated T Cell Tolerance through Inducing Kupffer Cell Expansion and IL-10 Production. J Immunol. (2018) 200:2341–51. doi: 10.4049/jimmunol.1700540

PubMed Abstract | Crossref Full Text | Google Scholar

128. Burghardt S, Claass B, Erhardt A, Karimi K, Tiegs G. Hepatocytes induce Foxp3+ regulatory T cells by Notch signaling. J Leukoc Biol. (2014) 96:571–7. doi: 10.1189/jlb.2AB0613-342RR

PubMed Abstract | Crossref Full Text | Google Scholar

129. Burghardt S, Erhardt A, Claass B, Huber S, Adler G, Jacobs T, et al. Hepatocytes contribute to immune regulation in the liver by activation of the Notch signaling pathway in T cells. J Immunol. (2013) 191:5574–82. doi: 10.4049/jimmunol.1300826

PubMed Abstract | Crossref Full Text | Google Scholar

130. Breous E, Somanathan S. & Wilson, J. M BALB/c mice show impaired hepatic tolerogenic response following AAV gene transfer to the liver. Mol Ther. (2010) 18:766–74. doi: 10.1038/mt.2009.301

PubMed Abstract | Crossref Full Text | Google Scholar

131. Cao O, Dobrzynski E, Wang L, Nayak S, Mingle B, Terhorst C, et al. Induction and role of regulatory CD4+CD25+ T cells in tolerance to the transgene product following hepatic in vivo gene transfer. Blood. (2007) 110:1132–40. doi: 10.1182/blood-2007-02-073304

PubMed Abstract | Crossref Full Text | Google Scholar

132. Dobrzynski E, Mingozzi F, Liu YL, Bendo E, Cao O, Wang L, et al. Induction of antigen-specific CD4+ T-cell anergy and deletion by in vivo viral gene transfer. Blood. (2004) 104:969–77. doi: 10.1182/blood-2004-03-0847

PubMed Abstract | Crossref Full Text | Google Scholar

133. Benseler V, Warren A, Vo M, Holz LE, Tay SS, Le Couteur DG, et al. Hepatocyte entry leads to degradation of autoreactive CD8 T cells. Proc Natl Acad Sci U S A. (2011) 108:16735–40. doi: 10.1073/pnas.1112251108

PubMed Abstract | Crossref Full Text | Google Scholar

134. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, et al. Alzheimer's disease Lancet. (2021) 397:1577–90. doi: 10.1016/S0140-6736(20)32205-4

PubMed Abstract | Crossref Full Text | Google Scholar

135. Raulin AC, Doss SV, Trottier ZA, Ikezu TC, Bu G, Liu CC. ApoE in Alzheimer's disease: pathophysiology and therapeutic strategies. Mol Neurodegener. (2022) 17:72. doi: 10.1186/s13024-022-00574-4

PubMed Abstract | Crossref Full Text | Google Scholar

136. Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A. (1993) 90:1977–81. doi: 10.1073/pnas.90.5.1977

PubMed Abstract | Crossref Full Text | Google Scholar

137. Yamazaki Y, Zhao N, Caulfield TR, Liu C-C, Bu G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. (2019) 15:501–18. doi: 10.1038/s41582-019-0228-7

PubMed Abstract | Crossref Full Text | Google Scholar

138. Corder EH, Saunders AM, Risch NJ, Strittmatter WJ, Schmechel DE, Gaskell PC, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet. (1994) 7:180–4. doi: 10.1038/ng0694-180

PubMed Abstract | Crossref Full Text | Google Scholar

139. Hu J, Liu CC, Chen XF, Zhang YW, Xu H, Bu G. Opposing effects of viral mediated brain expression of apolipoprotein E2 (apoE2) and apoE4 on apoE lipidation and Aβ metabolism in apoE4-targeted replacement mice. Mol Neurodegener. (2015) 10:6. doi: 10.1186/s13024-015-0001-3

PubMed Abstract | Crossref Full Text | Google Scholar

140. Zhao L, Gottesdiener AJ, Parmar M, Li M, Kaminsky SM, Chiuchiolo MJ, et al. Intracerebral adeno-associated virus gene delivery of apolipoprotein E2 markedly reduces brain amyloid pathology in Alzheimer's disease mouse models. Neurobiol Aging. (2016) 44:159–72. doi: 10.1016/j.neurobiolaging.2016.04.020

PubMed Abstract | Crossref Full Text | Google Scholar

141. Günaydin C, Sondhi D, Kaminsky SM, Lephart HC, Leopold PL, Hackett NR, et al. AAVrh10 delivery of novel APOE2-Christchurch variant suppresses amyloid and Tau pathology in Alzheimer's disease mice. Mol Ther. (2024) 32:4303–18. doi: 10.1016/j.ymthe.2024.11.003

PubMed Abstract | Crossref Full Text | Google Scholar

142. Wei M, Wu T, Chen N. Bridging neurotrophic factors and bioactive peptides to Alzheimer's disease. Aging Res Rev. (2024) 94:102177. doi: 10.1016/j.arr.2023.102177

PubMed Abstract | Crossref Full Text | Google Scholar

143. Su R, Su W, Jiao, Q. NGF protects neuroblastoma cells against β-amyloid-induced apoptosis via the Nrf2/HO-1 pathway. FEBS Open Bio. (2019) 9:2063–71. doi: 10.1002/2211-5463.12742

PubMed Abstract | Crossref Full Text | Google Scholar

144. Rizzi C, Tiberi A, Giustizieri M, Marrone MC, Gobbo F, Carucci NM, et al. NGF steers microglia toward a neuroprotective phenotype. Glia. (2018) 66:1395–416. doi: 10.1002/glia.23312

PubMed Abstract | Crossref Full Text | Google Scholar

145. Bishop KM, Hofer EK, Mehta A, Ramirez A, Sun L, Tuszynski M, et al. Therapeutic potential of CERE-110 (AAV2-NGF): targeted, stable, and sustained NGF delivery and trophic activity on rodent basal forebrain cholinergic neurons. Exp Neurol. (2008) 211:574–84. doi: 10.1016/j.expneurol.2008.03.004

PubMed Abstract | Crossref Full Text | Google Scholar

146. Nagahara AH, Bernot T, Moseanko R, Brignolo L, Blesch A, Conner JM, et al. Long-term reversal of cholinergic neuronal decline in aged non-human primates by lentiviral NGF gene delivery. Exp Neurol. (2009) 215:153–9. doi: 10.1016/j.expneurol.2008.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

147. Castle MJ, Baltanás FC, Kovacs I, Nagahara AH, Barba D, Tuszynski MH. Postmortem analysis in a clinical trial of AAV2-NGF gene therapy for Alzheimer's Disease identifies a need for improved vector delivery. Human Gene Therapy. (2020) 31:415. doi: 10.1089/hum.2019.367

PubMed Abstract | Crossref Full Text | Google Scholar

148. Potential therapeutic uses of BDNF in neurological and psychiatric disorders - PubMed. Available online at: https://pubmed.ncbi.nlm.nih.gov/21358740/ (accessed May 30, 2025).

Google Scholar

149. Ginsberg SD, Malek-Ahmadi MH, Alldred MJ, Chen Y, Chen K, Chao MV, et al. Brain-derived neurotrophic factor (BDNF) and TrkB hippocampal gene expression are putative predictors of neuritic plaque and neurofibrillary tangle pathology. Neurobiol Dis. (2019) 132:104540. doi: 10.1016/j.nbd.2019.104540

PubMed Abstract | Crossref Full Text | Google Scholar

150. Xia Y, Wang ZH, Liu P, Edgington-Mitchell L, Liu X, Wang XC, et al. TrkB receptor cleavage by delta-secretase abolishes its phosphorylation of APP, aggravating Alzheimer's disease pathologies. Mol Psychiatry. (2021) 26:2943–63. doi: 10.1038/s41380-020-00863-8

PubMed Abstract | Crossref Full Text | Google Scholar

151. Nagahara AH, Wilson BR, Ivasyk I, Kovacs I, Rawalji S, Bringas JR, et al. MR-guided delivery of AAV2-BDNF into the entorhinal cortex of non-human primates. Gene. Ther. (2018) 25:104–14. doi: 10.1038/s41434-018-0010-2

PubMed Abstract | Crossref Full Text | Google Scholar

152. Zvereva MI, Shcherbakova DM, Dontsova OA. Telomerase: structure, functions, and activity regulation. Biochemistry. (2010) 75:1563–83. doi: 10.1134/S0006297910130055

PubMed Abstract | Crossref Full Text | Google Scholar

153. Shim HS, Horner JW, Wu CJ, Li J, Lan ZD, Jiang S, et al. Telomerase reverse transcriptase preserves neuron survival and cognition in Alzheimer's Disease Models. Nat Aging. (2021) 1:1162–74. doi: 10.1038/s43587-021-00146-z

PubMed Abstract | Crossref Full Text | Google Scholar

154. Bernardes de Jesus B, Vera E, Schneeberger K, Tejera AM, Ayuso E, Bosch F, et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol Med. (2012) 4:691–704. doi: 10.1002/emmm.201200245

PubMed Abstract | Crossref Full Text | Google Scholar

155. Sánchez-Pernaute R, Harvey-White J, Cunningham J, Bankiewicz KS. Functional effect of adeno-associated virus mediated gene transfer of aromatic L-amino acid decarboxylase into the striatum of 6-OHDA-lesioned rats. Mol Ther. (2001) 4:324–30. doi: 10.1006/mthe.2001.0466

PubMed Abstract | Crossref Full Text | Google Scholar

156. Hadaczek P, Eberling JL, Pivirotto P, Bringas J, Forsayeth J, Bankiewicz KS. Eight years of clinical improvement in MPTP-lesioned primates after gene therapy with AAV2-hAADC. Mol Ther. (2010) 18:1458–61. doi: 10.1038/mt.2010.106

PubMed Abstract | Crossref Full Text | Google Scholar

157. Christine CW, Richardson RM, Van Laar AD, Thompson ME, Fine EM, Khwaja OS, et al. Safety of AADC gene therapy for moderately advanced Parkinson Disease: three-year outcomes from the PD-1101 trial. Neurology. (2022) 98:e40–e50. doi: 10.1212/WNL.0000000000012952

PubMed Abstract | Crossref Full Text | Google Scholar

158. Christine CW, Bankiewicz KS, Van Laar AD, Richardson RM, Ravina B, Kells AP, et al. Magnetic resonance imaging-guided phase 1 trial of putaminal AADC gene therapy for Parkinson's disease. Ann Neurol. (2019) 85:704–14. doi: 10.1002/ana.25450

PubMed Abstract | Crossref Full Text | Google Scholar

159. Nutt JG, Curtze C, Hiller A, Anderson S, Larson PS, Van Laar AD, et al. Aromatic L-Amino Acid Decarboxylase Gene Therapy Enhances Levodopa Response in Parkinson's Disease. Mov Disord. (2020) 35:851–8. doi: 10.1002/mds.27993

PubMed Abstract | Crossref Full Text | Google Scholar

160. Palfi S., et al. Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial. Lancet. (2014) 383:1138–46.

PubMed Abstract | Google Scholar

161. Pearson TS, Gupta N, San Sebastian W, Imamura-Ching J, Viehoever A, Grijalvo-Perez A, et al. Gene therapy for aromatic L-amino acid decarboxylase deficiency by MR-guided direct delivery of AAV2-AADC to midbrain dopaminergic neurons. Nat Commun. (2021) 12:4251. doi: 10.1038/s41467-021-24524-8

PubMed Abstract | Crossref Full Text | Google Scholar

162. Tai CH, Lee NC, Chien YH, Byrne BJ, Muramatsu SI, Tseng SH, et al. Long-term efficacy and safety of eladocagene exuparvovec in patients with AADC deficiency. Mol Ther. (2022) 30:509–18. doi: 10.1016/j.ymthe.2021.11.005

PubMed Abstract | Crossref Full Text | Google Scholar

163. Ayanlaja AA, Zhang B, Ji G, Gao Y, Wang J, Kanwore K, et al. The reversible effects of glial cell line-derived neurotrophic factor (GDNF) in the human brain. Semin Cancer Biol. (2018) 53:212–22. doi: 10.1016/j.semcancer.2018.07.005

PubMed Abstract | Crossref Full Text | Google Scholar

164. Su X, Kells AP, Huang EJ, Lee HS, Hadaczek P, Beyer J, et al. Safety evaluation of AAV2-GDNF gene transfer into the dopaminergic nigrostriatal pathway in aged and parkinsonian rhesus monkeys. Hum Gene Ther. (2009) 20:1627–40. doi: 10.1089/hum.2009.103

PubMed Abstract | Crossref Full Text | Google Scholar

165. Chen YH, Harvey BK, Hoffman AF, Wang Y, Chiang YH, Lupica CR. MPTP-induced deficits in striatal synaptic plasticity are prevented by glial cell line-derived neurotrophic factor expressed via an adeno-associated viral vector. FASEB J. (2008) 22:261–75. doi: 10.1096/fj.07-8797com

PubMed Abstract | Crossref Full Text | Google Scholar

166. Eberling JL, Kells AP, Pivirotto P, Beyer J, Bringas J, Federoff HJ, et al. Functional effects of AAV2-GDNF on the dopaminergic nigrostriatal pathway in parkinsonian rhesus monkeys. Hum Gene Ther. (2009) 20:511–8. doi: 10.1089/hum.2008.201

PubMed Abstract | Crossref Full Text | Google Scholar

167. Herzog CD, Dass B, Holden JE, Stansell J, 3rd Gasmi M, Tuszynski MH, et al. Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys. Mov Disord. (2007) 22:1124–32. doi: 10.1002/mds.21503

PubMed Abstract | Crossref Full Text | Google Scholar

168. Kordower JH, Herzog CD, Dass B, Bakay RA, Stansell J, 3rd Gasmi M, et al. Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann Neurol. (2006) 60:706–15. doi: 10.1002/ana.21032

PubMed Abstract | Crossref Full Text | Google Scholar

169. Warren Olanow C, Bartus RT, Baumann TL, Factor S, Boulis N, Stacy M, et al. Gene delivery of neurturin to putamen and substantia nigra in Parkinson disease: a double-blind, randomized, controlled trial. Ann Neurol. (2015) 78:248–57. doi: 10.1002/ana.24436

PubMed Abstract | Crossref Full Text | Google Scholar

170. Alharbi B, Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Alharbi H, Alexiou A, et al. Role of GABA pathway in motor and non-motor symptoms in Parkinson's disease: a bidirectional circuit. Eur J Med Res. (2024) 29:205. doi: 10.1186/s40001-024-01779-7

PubMed Abstract | Crossref Full Text | Google Scholar

171. van Nuland AJM, den Ouden HEM, Zach H, Dirkx MFM, van Asten JJA, Scheenen TWJ, et al. GABAergic changes in the thalamocortical circuit in Parkinson's disease. Hum Brain Mapp. (2020) 41:1017–29. doi: 10.1002/hbm.24857

PubMed Abstract | Crossref Full Text | Google Scholar

172. Luo J, Kaplitt MG, Fitzsimons HL, Zuzga DS, Liu Y, Oshinsky ML, et al. Subthalamic GAD gene therapy in a Parkinson's disease rat model. Science. (2002) 298:425–9. doi: 10.1126/science.1074549

PubMed Abstract | Crossref Full Text | Google Scholar

173. Emborg ME, Carbon M, Holden JE, During MJ, Ma Y, Tang C, et al. Subthalamic glutamic acid decarboxylase gene therapy: changes in motor function and cortical metabolism. J Cereb Blood Flow Metab. (2007) 27:501–9. doi: 10.1038/sj.jcbfm.9600364

PubMed Abstract | Crossref Full Text | Google Scholar

174. 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

175. 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, randomized trial. Lancet Neurol. (2011) 10:309–19. doi: 10.1016/S1474-4422(11)70039-4

PubMed Abstract | Crossref Full Text | Google Scholar

176. Niethammer M, Tang CC, Vo A, Nguyen N, Spetsieris P, Dhawan V, et al. Gene therapy reduces Parkinson's disease symptoms by reorganizing functional brain connectivity. Sci Transl Med. (2018) 10:eaau0713. doi: 10.1126/scitranslmed.aau0713

PubMed Abstract | Crossref Full Text | Google Scholar

177. Giladi N, Alcalay RN, Cutter G, Gasser T, Gurevich T, Höglinger GU, et al. Safety and efficacy of venglustat in GBA1-associated Parkinson's disease: an international, multicentre, double-blind, randomized, placebo-controlled, phase 2 trial. Lancet Neurol. (2023) 22:661–71. doi: 10.1016/S1474-4422(23)00205-3

PubMed Abstract | Crossref Full Text | Google Scholar

178. Sucunza D, Rico AJ, Roda E, Collantes M, González-Aseguinolaza G, Rodríguez-Pérez AI, et al. Glucocerebrosidase Gene Therapy Induces Alpha-Synuclein Clearance and Neuroprotection of Midbrain Dopaminergic Neurons in Mice and Macaques. Int J Mol Sci. (2021) 22:4825. doi: 10.3390/ijms22094825

PubMed Abstract | Crossref Full Text | Google Scholar

179. Rocha EM, Smith GA, Park E, Cao H, Brown E, Hayes MA, et al. Glucocerebrosidase gene therapy prevents α-synucleinopathy of midbrain dopamine neurons. Neurobiol Dis. (2015) 82:495–503. doi: 10.1016/j.nbd.2015.09.009

PubMed Abstract | Crossref Full Text | Google Scholar

180. Ross CA, Tabrizi SJ. Huntington's disease: from molecular pathogenesis to clinical treatment. Lancet Neurol. (2011) 10:83–98. doi: 10.1016/S1474-4422(10)70245-3

PubMed Abstract | Crossref Full Text | Google Scholar

181. Packer AN, Xing Y, Harper SQ, Jones L, Davidson BL. The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington's disease. J Neurosci. (2008) 28:14341–6. doi: 10.1523/JNEUROSCI.2390-08.2008

PubMed Abstract | Crossref Full Text | Google Scholar

182. Martí E, Pantano L, Bañez-Coronel M, Llorens F, Miñones-Moyano E, Porta S, et al. A myriad of miRNA variants in control and Huntington's disease brain regions detected by massively parallel sequencing. Nucleic Acids Res. (2010) 38:7219–35. doi: 10.1093/nar/gkq575

PubMed Abstract | Crossref Full Text | Google Scholar

183. Lee ST, Chu K, Im WS, Yoon HJ, Im JY, Park JE, et al. Altered microRNA regulation in Huntington's disease models. Exp Neurol. (2011) 227:172–9. doi: 10.1016/j.expneurol.2010.10.012

PubMed Abstract | Crossref Full Text | Google Scholar

184. Dysregulation of REST-regulated coding and non-coding RNAs in a cellular model of Huntington's disease - PubMed. Available online at: https://pubmed.ncbi.nlm.nih.gov/23145961/ (accessed May 30, 2025).

Google Scholar

185. Spronck EA, Brouwers CC, Vallès A, de Haan M, Petry H, van Deventer SJ, et al. AAV5-miHTT gene therapy demonstrates sustained huntingtin lowering and functional improvement in huntington disease mouse models. Mol Ther Methods Clin Dev. (2019) 13:334–43. doi: 10.1016/j.omtm.2019.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

186. Fukuoka M, Takahashi M, Fujita H, Chiyo T, Popiel HA, Watanabe S, et al. Supplemental treatment for Huntington's Disease with miR-132 that is deficient in Huntington's Disease brain. Mol Ther Nucleic Acids. (2018) 11:79–90. doi: 10.1016/j.omtn.2018.01.007

PubMed Abstract | Crossref Full Text | Google Scholar

187. Keeler AM, Sapp E, Chase K, Sottosanti E, Danielson E, Pfister E, Cellular Cellular analysis of silencing the Huntington's Disease gene using AAV9 mediated delivery of artificial micro RNA into the Striatum of Q140/Q140 mice. J Huntingtons Dis. doi: 10.3233/JHD-160215

PubMed Abstract | Crossref Full Text | Google Scholar

188. Kreilaus F, Spiro AS, McLean CA, Garner B, Jenner AM. Evidence for altered cholesterol metabolism in Huntington's disease post mortem brain tissue Neuropathol Appl Neurobiol. (2016) 42:535–46. doi: 10.1111/nan.12286

PubMed Abstract | Crossref Full Text | Google Scholar

189. Leoni V, Mariotti C, Tabrizi SJ, Valenza M, Wild EJ, Henley SM, et al. Plasma 24S-hydroxycholesterol and caudate MRI in pre-manifest and early Huntington's disease. Brain. (2008) 131:2851–9. doi: 10.1093/brain/awn212

PubMed Abstract | Crossref Full Text | Google Scholar

190. Kacher R, Lamazière A, Heck N, Kappes V, Mounier C, Despres G, et al. CYP46A1 gene therapy deciphers the role of brain cholesterol metabolism in Huntington's disease. Brain. (2019) 142:2432–50. doi: 10.1093/brain/awz174

PubMed Abstract | Crossref Full Text | Google Scholar

191. Boussicault L, Alves S, Lamazière A, Planques A, Heck N, Moumné L, et al. CYP46A1, the rate-limiting enzyme for cholesterol degradation, is neuroprotective in Huntington's disease. Brain. (2016) 139:953–70. doi: 10.1093/brain/awv384

PubMed Abstract | Crossref Full Text | Google Scholar

192. Logan T, Simon MJ, Rana A, Cherf GM, Srivastava A, Davis SS, et al. Rescue of a lysosomal storage disorder caused by Grn loss of function with a brain penetrant progranulin biologic. Cell. (2021) 184.

Google Scholar

193. Huang M, Modeste E, Dammer E, Merino P, Taylor G, Duong DM, et al. Network analysis of the progranulin-deficient mouse brain proteome reveals pathogenic mechanisms shared in human frontotemporal dementia caused by GRN mutations. Acta Neuropathol Commun. (2020) 8:163. doi: 10.1186/s40478-020-01037-x

PubMed Abstract | Crossref Full Text | Google Scholar

194. Boland S, Swarup S, Ambaw YA, Malia PC, Richards RC, Fischer AW, et al. Deficiency of the frontotemporal dementia gene GRN results in gangliosidosis. Nat Commun. (2022) 13:5924. doi: 10.1038/s41467-022-33500-9

PubMed Abstract | Crossref Full Text | Google Scholar

195. Sevigny J, Uspenskaya O, Heckman LD, Wong LC, Hatch DA, Tewari A, et al. Progranulin AAV gene therapy for frontotemporal dementia: translational studies and phase 1/2 trial interim results. Nat Med. (2024) 30:1406–15. doi: 10.1038/s41591-024-02973-0

PubMed Abstract | Crossref Full Text | Google Scholar

196. Hinderer C, Miller R, Dyer C, Johansson J, Bell P, Buza E, et al. Adeno-associated virus serotype 1-based gene therapy for FTD caused by GRN mutations. Ann Clin Transl Neurol. (2020) 7:1843–53. doi: 10.1002/acn3.51165

PubMed Abstract | Crossref Full Text | Google Scholar

197. Kawamoto Y, Nakamura S, Matsuo A, Akiguchi I. Glial cell line-derived neurotrophic factor-like immunoreactivity in the cerebella of normal subjects and patients with multiple system atrophy. Acta Neuropathol. (2000) 100:131–7. doi: 10.1007/s004010050004

PubMed Abstract | Crossref Full Text | Google Scholar

198. Mohajeri MH, Figlewicz DA, Bohn MC. Intramuscular grafts of myoblasts genetically modified to secrete glial cell line-derived neurotrophic factor prevent motoneuron loss and disease progression in a mouse model of familial amyotrophic lateral sclerosis. Hum Gene Ther. (1999) 10:1853–66. doi: 10.1089/10430349950017536

PubMed Abstract | Crossref Full Text | Google Scholar

199. Wang LJ, Lu YY, Muramatsu S, Ikeguchi K, Fujimoto K, Okada T, et al. Neuroprotective effects of glial cell line-derived neurotrophic factor mediated by an adeno-associated virus vector in a transgenic animal model of amyotrophic lateral sclerosis. J Neurosci. (2002) 22:6920–8. doi: 10.1523/JNEUROSCI.22-16-06920.2002

PubMed Abstract | Crossref Full Text | Google Scholar

200. Kaspar BK, Lladó J, Sherkat N, Rothstein JD, Gage FH. Retrograde viral delivery of IGF-1 prolongs survival in a mouse ALS model. Science. (2003) 301:839–42. doi: 10.1126/science.1086137

PubMed Abstract | Crossref Full Text | Google Scholar

201. Dodge JC, Haidet AM, Yang W, Passini MA, Hester M, Clarke J, et al. Delivery of AAV-IGF-1 to the CNS extends survival in ALS mice through modification of aberrant glial cell activity. Mol Ther. (2008) 16:1056–64. doi: 10.1038/mt.2008.60

PubMed Abstract | Crossref Full Text | Google Scholar

202. Franz CK, Federici T, Yang J, Backus C, Oh SS, Teng Q, et al. Intraspinal cord delivery of IGF-I mediated by adeno-associated virus 2 is neuroprotective in a rat model of familial. ALS Neurobiol Dis. (2009) 33:473–81. doi: 10.1016/j.nbd.2008.12.003

PubMed Abstract | Crossref Full Text | Google Scholar

203. 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

204. Al-Zaidy SA, Kolb SJ, Lowes L, Alfano LN, Shell R, Church KR, et al. AVXS-101 (Onasemnogene Abeparvovec) for SMA1: comparative study with a prospective natural history cohort. J Neuromuscul Dis. (2019) 6:307–17. doi: 10.3233/JND-190403

PubMed Abstract | Crossref Full Text | Google Scholar

205. Strauss KA, Farrar MA, Muntoni F, Saito K, Mendell JR, Servais L, et al. Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med. (2022) 28:1381–9. doi: 10.1038/s41591-022-01866-4

PubMed Abstract | Crossref Full Text | Google Scholar

206. Mercuri E, Baranello G, Masson R, Boespflug-Tanguy O, Bruno C, Scoto M, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy type 1 (STR1VE-EU): an open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol. (2021) 20:832–41. doi: 10.1136/jnnp-2022-ABN.2

PubMed Abstract | Crossref Full Text | Google Scholar

207. Strauss KA, Farrar MA, Muntoni F, Saito K, Mendell JR, Servais L, et al. Onasemnogene abeparvovec for presymptomatic infants with three copies of SMN2 at risk for spinal muscular atrophy: the Phase III SPR1NT trial. Nat Med. (2022) 28:1390–7. doi: 10.1038/s41591-022-01867-3

PubMed Abstract | Crossref Full Text | Google Scholar

208. McColl-Carboni A, Dollive S, Laughlin S, Lushi R, MacArthur M, Zhou S, et al. Analytical characterization of full, intermediate, and empty AAV capsids. Gene Ther. (2024) 31:285–94. doi: 10.1038/s41434-024-00444-2

PubMed Abstract | Crossref Full Text | Google Scholar

209. Yarawsky AE, Zai-Rose V, Cunningham HM, Burgner 2nd JW, DeLion, MT, Paul LN. AAV analysis by sedimentation velocity analytical ultracentrifugation: beyond empty and full capsids. Eur Biophys J. (2023) 52:353–66. doi: 10.1007/s00249-023-01646-z

PubMed Abstract | Crossref Full Text | Google Scholar

210. Bhujbal S, Bhujbal R, Giram P. An overview: CRISPR/Cas-based gene editing for viral vaccine development. Expert Rev Vaccines. (2022) 21:1581–93. doi: 10.1080/14760584.2022.2112952

PubMed Abstract | Crossref Full Text | Google Scholar

211. Hunker AC, Soden ME, Krayushkina D, Heymann G, Awatramani R, Zweifel LS. Conditional single vector CRISPR/SaCas9 viruses for efficient mutagenesis in the adult mouse nervous system. Cell Rep. (2020) 30:4303–4316.e6. doi: 10.1016/j.celrep.2020.02.092

PubMed Abstract | Crossref Full Text | Google Scholar

212. Rafii MS, Tuszynski MH, Thomas RG, Barba D, Brewer JB, Rissman RA, et al. Adeno-associated viral vector (Serotype 2)-nerve growth factor for patients with Alzheimer Disease: a randomized clinical trial. JAMA Neurol. (2018) 75:834–41. doi: 10.1001/jamaneurol.2018.0233

PubMed Abstract | Crossref Full Text | Google Scholar

213. De BP, Cram S, Lee H, Rosenberg JB, Sondhi D, Crystal RG, et al. Assessment of residual full-length SV40 Large T antigen in clinical-grade Adeno-associated virus vectors produced in 293T Cells Hum Gene. Ther. (2023) 34:697–704. doi: 10.1089/hum.2023.032

PubMed Abstract | Crossref Full Text | Google Scholar

214. Bartus RT, Baumann TL, Siffert J, Herzog CD, Alterman R, Boulis N, et al. Safety/feasibility of targeting the substantia nigra with AAV2-neurturin in Parkinson patients Neurology. (2013) 80:1698–701. doi: 10.1212/WNL.0b013e3182904faa

PubMed Abstract | Crossref Full Text | Google Scholar

215. Marks WJ, Jr 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

216. Fiandaca MS, Forsayeth JR, Dickinson PJ, Bankiewicz KS. Image-guided convection-enhanced delivery platform in the treatment of neurological diseases. Neurotherapeutics. (2008) 5:123–7. doi: 10.1016/j.nurt.2007.10.064

PubMed Abstract | Crossref Full Text | Google Scholar

217. Richardson RM, Bankiewicz KS, Christine CW, Van Laar AD, Gross RE, Lonser R, et al. Data-driven evolution of neurosurgical gene therapy delivery in Parkinson's disease. J Neurol Neurosurg Psychiatry. (2020) 91:1210–8. doi: 10.1136/jnnp-2020-322904

PubMed Abstract | Crossref Full Text | Google Scholar

218. Richardson RM, Kells AP, Rosenbluth KH, Salegio EA, Fiandaca MS, Larson PS, et al. Interventional MRI-guided putaminal delivery of AAV2-GDNF for a planned clinical trial in Parkinson's disease. Mol Ther. (2011) 19:1048–57. doi: 10.1038/mt.2011.11

PubMed Abstract | Crossref Full Text | Google Scholar

219. Samaranch L, Blits B, San Sebastian W, Hadaczek P, Bringas J, Sudhakar V, et al. MR-guided parenchymal delivery of adeno-associated viral vector serotype 5 in non-human primate brain. Gene Ther. (2017) 24:253–61. doi: 10.1038/gt.2017.14

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: viral vector, immune responses, neurodegenerative disease, gene therapy, AAV

Citation: Wang Y, Mu S and Liu F (2025) Viral vectors in neurodegenerative diseases: immune responses and therapeutic applications. Front. Neurol. 16:1603125. doi: 10.3389/fneur.2025.1603125

Received: 01 April 2025; Accepted: 30 May 2025;
Published: 18 June 2025.

Edited by:

Ulises Gomez-Pinedo, Health Research Institute of Hospital Clínico San Carlos, Spain

Reviewed by:

Jordi Bové, Vall d'Hebron Research Institute (VHIR), Spain
Doddy Denise Ojeda-Hernández, Complutense University of Madrid, Spain
Andrew C. Mercer, ReGenX Biosciences, United States

Copyright © 2025 Wang, Mu and Liu. 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: Fangxi Liu, bGZ4bGl1ZmFuZ3hpQDE2My5jb20=

These authors have contributed equally to this work

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.