Next Article in Journal
A Novel Class of Human ADAM8 Inhibitory Antibodies for Treatment of Triple-Negative Breast Cancer
Next Article in Special Issue
Transcription Factor Binding Site in Promoter Determines the Pattern of Plasmid-Based Transgene Expression In Vivo
Previous Article in Journal
Investigation of Photodynamic Therapy Promoted by Cherenkov Light Activated Photosensitizers—New Aspects and Revelations
Previous Article in Special Issue
Sucrose Treatment Enhances the Electrotransfer of DNA by Activating Phospholipase A2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Delivery of DNA-Based Therapeutics for Treatment of Chronic Diseases

by
Carleigh Sussman
,
Rachel A. Liberatore
and
Marek M. Drozdz
*
RenBio Inc., Long Island City, New York, NY 11101, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2024, 16(4), 535; https://doi.org/10.3390/pharmaceutics16040535
Submission received: 9 March 2024 / Revised: 2 April 2024 / Accepted: 8 April 2024 / Published: 13 April 2024

Abstract

:
Gene therapy and its role in the medical field have evolved drastically in recent decades. Studies aim to define DNA-based medicine as well as encourage innovation and the further development of novel approaches. Gene therapy has been established as an alternative approach to treat a variety of diseases. Its range of mechanistic applicability is wide; gene therapy has the capacity to address the symptoms of disease, the body’s ability to fight disease, and in some cases has the ability to cure disease, making it a more attractive intervention than some traditional approaches to treatment (i.e., medicine and surgery). Such versatility also suggests gene therapy has the potential to address a greater number of indications than conventional treatments. Many DNA-based therapies have shown promise in clinical trials, and several have been approved for use in humans. Whereas current treatment regimens for chronic disease often require frequent dosing, DNA-based therapies can produce robust and durable expression of therapeutic genes with fewer treatments. This benefit encourages the application of DNA-based gene therapy to manage chronic diseases, an area where improving efficiency of current treatments is urgent. Here, we provide an overview of two DNA-based gene therapies as well as their delivery methods: adeno associated virus (AAV)-based gene therapy and plasmid DNA (pDNA)-based gene therapy. We will focus on how these therapies have already been utilized to improve treatment of chronic disease, as well as how current literature supports the expansion of these therapies to treat additional chronic indications in the future.

1. Introduction

Traditional approaches of medicine and surgery have helped to treat many patients; however, there is a wide scope of indications for which conventional treatment is either inefficient or burdensome to the patient. Such indications, often associated with an underlying genetic cause, require multiple rounds of intervention over an extended period of time, making them chronic by nature. Current prescribed treatments for such indications might modulate the course of the disease or improve symptoms, but they are typically lifelong and have very involved dosing regimens. DNA-based medicines have the potential to make the treatment of such chronic conditions more manageable.
For decades, gene therapy has been an attractive approach to overcome diseases with a known underlying genetic cause [1]. The aim of gene therapy is to manipulate or modify the body’s gene expression for therapeutic benefit [1]. Approaches to gene therapy typically involve inactivating a disease-causing gene, replacing a damaged gene with a healthy copy, or introducing a new or modified gene to combat the disease [1]. Impressively, benefits provided by gene therapy can include complete curing from a disease [1]. Applications of gene therapy can be beneficial when tackling and managing chronic indications that possess multiple underlying causes.
Historically, a variety of DNA-based products have been established [1,2,3,4]. Such products include, but are not limited to, viral vectors [5,6,7,8,9,10] and pDNA [11,12]. In the case of viral vectors, viruses are modified to eliminate pathogenic attributes, typically including any ability to replicate, then are used as a vehicle to deliver therapeutic genes into cells [5,8]. Similarly, pDNA can be designed to carry therapeutic genes, but pDNA requires an additional delivery component to efficiently enter cells [11]. In both cases, these therapies utilize the host cell as the “factory” to produce the therapeutic protein [2,3,13]. Not only is the format of the therapeutic gene important (viral vectors vs. plasmids), but the delivery method has also been shown to impact efficiency of the approach [14,15,16,17]. While viral vectors are effective in introducing the transgene into a target cell with basic injection [8], pDNA requires an additional delivery method to facilitate efficient uptake by the target cells [14,15,16,17]. Electroporation (EP) is a delivery technique that has been investigated for this purpose and has shown potential when it comes to improving the therapeutic benefit of pDNA treatments (pDNA/EP) [14,15] (Table 1).
Here, we will review the recent advances in the treatment of chronic diseases with DNA-based medicines with a focus on adeno-associated virus (AAV) and pDNA-based therapies. We will discuss the benefits and limitations of both, highlight the potential of EP for delivery of pDNA-based therapies, and elaborate on how the field may shape in the near future.

2. Adeno-Associated Virus-Based Gene Therapy

2.1. Attributes of Adeno-Associated Virus-Based Gene Therapy

AAV was discovered in the 1960s as a contaminant in a preparation of simian adenovirus and is defined as a non-enveloped, replication-defective member of the Parvoviridae virus family [5]. AAVs have small icosahedral capsids containing DNA and require co-infection with a helper virus for lytic growth [18,19], and thus pathogenicity [19]. Lower levels of pathogenicity when compared to other viruses [19], along with additional advantages such as the ability to transduce both dividing and non-dividing cells [20,21], make AAV an attractive candidate for gene delivery. The first AAV proof-of-concept study was published in 1984 [22], in which Hermonat and Muzyczka employed AAV to deliver a neomycin resistance gene to mammalian cells in culture [22]. In the decades following, AAV continued to be evaluated for its ability to deliver genes to cells and has made significant strides in reaching important milestones in the clinic [23,24]. Advances culminated in the approval of the first AAV therapy, Glybera, by the European Medicines Agency in 2012 [25] and subsequently by the US Food and Drug Administration in 2017. Although this AAV therapy has recently been discontinued, its withdrawal was due to the cost of treatment rather than any ineffectiveness of the therapy itself [26].
The AAV viral genome of ~4.8 kb is composed of linear single-stranded DNA with inverted terminal repeats (ITRs) at both ends [20,27]. The viral genetic material between the ITRs can be replaced with therapeutic genetic material [8]. While an attractive vector, the packaging capacity of ~4.8 kb is a limiting factor [8], making vector design involving larger genes more difficult, and sometimes impossible. Exceeding packaging capacity can have negative effects on the therapy. Repercussions include lower expression efficiency [28,29], and in severe cases, truncation of the transgene, thereby completely abolishing efficacy of the therapy [28]. Subceeding packaging capacity has also shown similar negative effects of reducing expression efficiency [29]. Another consideration when designing an efficient form of AAV gene therapy is the selection of a suitable AAV serotype. There are 13 natural AAV serotypes that are more commonly reported on, with that number likely to grow [18,30]. In addition to the 13 natural serotypes, novel hybrid AAV vectors have been developed and are still being developed [30,31]. Each serotype, regardless of natural or novel identity, has specific receptor and tissue tropisms, as reviewed by Issa et al. [27] (Table 2), with engineered serotypes being generated to modulate such tropisms [32,33,34]. With this in mind, serotype selection is critical for maximizing efficiency of the therapy [18,30,35].
The capacity to select from an array of serotypes for therapy design can be advantageous [30,35], but the ubiquitous nature of AAV infections has the potential to negate the benefits. Since AAVs are endemic and non-pathogenic [19], many individuals do not realize they have been exposed to AAVs. Globally, studies have seen the highest level of serotype positivity (seroprevalence or seropositivity) being associated with AAV2, and lowest associated with AAV5 [36,37]. Exposure to AAVs can correlate with pre-existing immunity to various serotypes, limiting success of treatments [36,38,39]. Testing for pre-existing antibodies against AAVs has been introduced as a strategy to determine treatment eligibility to avoid such issues [38,39,40]. By similar biological mechanisms, repeated exposure to the same AAV therapy can result in immunity to treatment, even if no pre-existing seropositivity was detected [41,42,43,44,45]. The body’s inevitable immune response to AAV treatment, regardless of pre-existing seropositivity, makes repeat treatments with AAV gene therapy challenging [42,43]. A meta-analysis of more than 200 AAV clinical trials found that almost 50% of trials administered immunosuppressants in conjunction with AAV delivery to avoid immune response to treatment [46]. With immunity proven to play a fundamental role in efficiency of treatment, methods to improve AAV technology to avoid such responses are currently being investigated [47,48]. Strategies addressing immunity, as well as other limitations of AAV, include, but are not limited to, optimizing dosing regimen [49], further engineering of AAV capsids [50,51], and vector development such as self-complementary AAVs (scAAVs) [51,52,53,54].

2.2. AAV-Based Therapy in the Context of Chronic Disease

While there is clearly room for improvement [55,56], AAV still harbors many advantages that make it an attractive treatment for various indications, especially for targeting monogenic recessive diseases [8,23].
Hemophilia is one of the many chronic indications that has become a target for AAV-based gene therapy [57,58,59,60]. Hemophilia is a rare inherited disorder in which one’s blood does not clot properly due to insufficient clotting factors [61,62]. There are two types of hemophilia. Hemophilia A is classified as a lack or total absence of clotting factor 8 (FVIII) [61,63], and hemophilia B is classified as a shortage or absence of clotting factor 9 (FIX) [63,64]. The standard of care for hemophilia includes prophylactic intravenous (IV) injections of the missing factor, or injection of antibodies that bridge the biological pathway necessary to restore the function of the clotting signaling cascade [61,65]. Management of hemophilia requires frequent treatments, often requiring administration multiple times per week [61]. In some cases, additional doses must be given following blunt injuries to prevent uncontrolled bleeding [61]. Such frequent administration is costly and time consuming, making it difficult for the patient to adhere to treatment regimens [66]. Therefore, a gene therapy for hemophilia, in which the necessary clotting factors would be continuously produced in the patient’s body, is an attractive alternative [60].
With one AAV therapy addressing hemophilia A already approved for use in humans [67], and others in the clinic (ClinicalTrials.Gov; NCT03432520), there is enormous excitement surrounding AAV-based hemophilia treatments [68]. For treatment of hemophilia A, Spark Therapeutics tested its therapy using AAV serotype 3 [60], SPK-8011 (Dirloctocogene Samoparvovec), through an open label phase 1/2 trial (ClinicalTrials.Gov; NCT03003533) as well as a long-term study (ClinicalTrials.Gov; NCT03432520). These studies confirmed the safety profile of the therapy and also evaluated the expression pattern of FVIII in the years following treatment [69]. A single IV infusion of Dirloctocogene Samoparvovec was jointly administered with glucocorticoids [69] and resulted in sustained FVIII expression in 16 of 18 participants. Expression of FVIII lasted more than one year post-therapy, and in many, was stably maintained for more than two years [69]. These data highlight the tremendous benefits that AAV therapy can provide over standard treatments; compared to standard treatments for hemophilia, AAV requires fewer interventions to yield the same therapeutic effects. Of the 18 participants, the two that did not respond to the therapy experienced an immune response to the AAV, despite the administration of glucocorticoids [69].
Spark Therapeautics has also made strides with another AAV gene therapy, SPK-9001 (Fidanacogene Elaparvovec), for hemophilia B [70,71]. This AAV therapy, utilizing recombinant AAV serotype rh74 [72], has advanced through similar phase 1/2 trials (ClinicalTrials.Gov; NCT02484092, ClinicalTrials.Gov; NCT03307980). Data obtained showed that one-time IV infusion of Fidanacogene Elaparvovec almost completely eliminated the need for exogenous FIX treatment, with the follow-up being one year after administration [70]. Based on the success of earlier trials, a phase 3 trial is ongoing to track long-term expression and response to the AAV therapy, with a completion date set in 2030 (ClinicalTrials.Gov; NCT03861273). Positive results published in studies and trials emphasize the promise of AAV-based gene therapy for treatment of hemophilia. This success also suggests establishing a wider applicability of AAV therapy as treatment.
There are various other chronic indications where AAV gene therapy could prove advantageous. One such example is Wilson’s Disease. Wilson’s Disease is a rare genetic disease caused by an inherited mutation in the ATP7B gene, which encodes for a copper transporting ATPase mainly expressed in hepatocytes [73,74]. Mutations affecting ATPase cause an accumulation of copper in the body [73,74]. Abnormally high levels of copper can cause damage to organs if left untreated, making it of high priority to regulate these levels [73,74]. Current treatment for Wilson’s Disease is the prescription of chelating agents to manage and remove excess copper from the blood [73,74]. Lifelong treatment is necessary to prevent additional copper buildup [73,74]. Severity of the disease combined with the frequent dosing to manage symptoms suggest Wilson’s Disease would be an attractive candidate for AAV therapies. Indeed, AAV therapies that deliver a functional form of the ATP7B gene to the liver have already been tested in mouse models and were successful in correcting the disease [75,76,77]. Success in mouse models shifted focus to translate these accomplishments in the clinic. Vivet Therapeutics has begun a phase 1/2 trial (ClinicalTrials.Gov; NCT04537377) to observe the safety profile of its AAV-based gene therapy VTX-801, of serotype AAV-Anc80 [78], for Wilson’s Disease. The study aims to deliver functional variants of the ATP7B gene to the liver, and the therapy has already proven efficacious in mice [77]. This clinical trial is still currently recruiting and aims to be completed by the first quarter of 2029.
Gaucher Disease is another chronic indication well-suited to AAV-based gene therapy. Gaucher disease is an autosomal recessive disorder caused by a mutation in the GBA gene [79,80,81], resulting in a lack of the enzyme glucocerebrosidase (GCase) [81]. This enzyme is required to break down lipids in the body. GCase enzyme deficiency results in accumulation of fat in organs such as the spleen and liver [81], leading to a myriad of symptoms including enlarged organs, easy bruising, fatigue, and other potentially more serious neurological effects [81,82]. Currently, more conventional treatment options include enzyme replacement therapy (ERT), which is typically administered every 2 weeks via IV infusions [83]. Additionally, substrate reduction therapy (SRT) may be used in specific patient populations, also requiring daily oral pills [83]. Regardless of treatment, common therapies prescribed to manage symptoms of Gaucher disease are lifelong and demand frequent dosing [84]. Freeline Therapeutics is trying to address the inefficiencies of traditional treatment with its AAV therapy FLT201 (also known as GALILEO-1), which is of AAVS3 serotype [46] and designed to deliver a functional GBA transgene. This therapy is currently at the tail end of a phase 1 clinical trial to assess safety, tolerability, and efficacy. The trial is planned to conclude by the end of 2024 (ClinicalTrialsGov; NCT05324943).
It is quite clear that AAV therapies have the potential to offer improved treatments of many chronic diseases. With multiple AAV-based gene therapies in various stages in the clinic (ClinicalTrialsGov; NCT04669535, ClinicalTrialsGov; NCT05603312, ClinicalTrialsGov; NCT05568719, ClinicalTrialsGov; NCT00229736), we will likely continue to see the expansion of this approach as groups pursue it to address additional chronic indications. However, genetic cargo design restrictions and immunogenicity associated with AAV are obstacles that persist, emphasizing aspects of this therapy that require further optimization. AAV-based therapy may provide efficacious treatment for a number of monogenic diseases that require a single gene replacement. However, it may not be suitable for every chronic condition, particularly those with a multifactorial etiology.

3. Use of pDNA in Gene Therapy

Delivery of naked DNA, such as pDNA, as a therapeutic has gained attention in recent years because of advantages that it offers, especially when compared to other therapies [11,85]. pDNA has demonstrated a strong safety profile in multiple clinical trials [86,87,88,89], confirming high levels of tolerability and minimal toxicity following administration [89]. While AAV therapy has shown cases of genotoxicity as well as unintended integration into the genome across many species [90,91,92,93], these problems have not been reported when delivering pDNA [94,95,96]. Indeed, studies have shown that the risk of integration following delivery of pDNA is negligible [97,98,99]. Unlike the case for AAV, the lack of immune responses directed against pDNA permits repeat dosing [42,43]. The ability to administer this therapy more than once, without the risk of inducing immunity to the therapy and compromising expression, makes pDNA a safe and powerful option for chronic indications where multiple treatments may be necessary.
In addition to its inherent safety characteristics, another advantage of using pDNA is its flexibility when it comes to vector design. Whereas AAV therapies have more strict sequence length limitations [28,29], pDNA has a larger capacity and is better positioned to deliver longer transgenes [100], enabling pDNA-based treatments to target a larger pool of indications [100]. This greater capacity can also lend itself to inclusion of various factors that may help further increase transgene expression [11]. For example, tissue-specific promoters have been shown to help increase efficiency and specificity of treatment by selectively expressing DNA cargo in cells where the specific promoter is active [101]. These, as well as various other modifications, are relatively easy to incorporate into pDNA sequences.
Aside from the efficacy, safety, and design of gene therapy, a critical consideration needs to be given to the manufacturing and stability of the therapeutic. Large-scale production of AAV is difficult, costly, and time consuming [102,103]. On the other hand, the manufacturing of pDNA avoids most of these disadvantages (Figure 1). The process is relatively inexpensive, scalable, and quick [11]. Additionally, pDNA is more stable than AAV at a wider range of temperatures [104,105]; thus, pDNA therapies do not suffer from some of the cold-chain requirements and stability concerns that apply to AAV delivery systems.
While pDNA delivery as a method of DNA-based therapy offers several benefits, it is not without limitations. The presence of backbone elements required for large-scale production, such as antibiotic resistance genes or other bacteria-derived sequences, might have negative effects on the patient as well as on expression of the encoded transgene [100,106,107,108]. These can, however, be addressed by incorporating technologies such as minicircle DNA, or fully synthetic linear DNAs, that eliminate bacterial elements and encode only the transgene expression cassette [109,110,111,112].
Although pDNA presents several benefits, direct injection of pDNA does not inherently lead to high and sustained levels of gene expression [113,114]. Therefore, pDNA-based therapy requires an alternative method of delivery to facilitate cellular uptake as well as increase stability and durability of transgene expression.

4. Electroporation as a Method for pDNA Delivery

4.1. Characteristics of Electroporation

To address the need for high and durable transgene expression levels, different delivery techniques have been developed, including liposomes, particle-mediated gene transfer (‘gene gun’), and EP [115,116,117]. While there has been a lot of excitement surrounding liposomes, nanoparticles, and their derivatives, when delivering nucleic acids, most of the literature focuses on use of these systems for delivery of RNA as opposed to pDNA [118,119]; studies connecting nanoparticles for delivery of pDNA are scarce [120], and most are still in early stages. By comparison, there is an extensive literature outlining EP as a technique previously used in animals, making it an attractive approach for delivery of pDNA in humans [14,121,122].
EP can be used to deliver pDNA to various target tissues. Intradermal EP is often used for DNA vaccines [123], and direct, intratumoral EP is occasionally used in oncology [124,125,126], but the most common target for delivery of the pDNA is skeletal muscle. Injection of pDNA in the muscle is accompanied by short electrical pulses, thereby creating an electrical field which leads to an accumulation of charge across the cell membrane [127]. Past a certain threshold, transient pore formation occurs. Smaller pores stabilize and can increase in size to allow larger molecules, such as pDNA, to enter the cell [127]. Under the right electrical conditions, this permeabilization is temporary and reversible, and after the electrical pulses are complete, the membrane seals within minutes [128,129,130]. By creating pores to facilitate movement of DNA, EP significantly increases DNA uptake by cells, therefore increasing the total expression yield when compared to injections without EP [16,123,131]. By injecting intramuscularly, pDNA/EP converts the muscle cells into therapeutic-producing cells. After intramuscular injection and cell electroporation, the expressed transgene (therapeutic protein) is secreted by the muscle cells and taken up into systemic circulation. Thus, delivery of the protein therapeutic becomes systemic. Although EP of pDNA has been beneficial in boosting transgene expression in smaller species, challenges persist regarding its scalability for human use. Optimized pDNA/EP protocols that are able to achieve robust levels of expression in mice do not always reflect what is achievable in larger animals [132,133]. This lack of translation is due to inherent differences in species anatomy and a much higher dilutional effect in larger animals. Compared to humans, the systemic circulation in mice is confined to lesser volumes; the blood volume in which the expressed transgene can be diluted once entering circulation is 3000-4000-fold smaller in mice than in humans [132,133]. While numerous studies have been conducted to develop more optimized protocols of pDNA/EP in larger species [132,134,135], further research is required to continue bridging the gap between delivery in smaller animals and humans.

4.2. Approaching Chronic Indications Using pDNA/EP

Studies, predominantly in the DNA vaccine field, have demonstrated that in addition to EP being safe and well tolerated, it is also effective in increasing stability and durability of pDNA transgene expression [123,136,137,138]. Such expression characteristics are ideal when evaluating treatments for chronic diseases, as higher and durable expression profiles allow for fewer treatment interventions while remaining efficacious (Figure 2).
While AAV therapies have made significant progress in hemophilia treatment, the issues of immunogenicity persist [69]. Such concerns have not been reported with pDNA-based gene therapies, suggesting pDNA/EP could be a more favorable approach for hemophilia treatments. Studies in immunocompetent mice investigating pDNA/EP of plasmids encoding human FIX (for treatment of hemophilia B) showed high plasma levels of the encoded protein lasting for at least 2 months [139]. Studies have shown similar techniques of pDNA/EP with plasmids encoding FIX in larger animals, such as dogs, suggesting this gene therapy is scalable [140]. Promising results across multiple animal models indicate the benefits of this alternative approach and suggest the therapy can be translated to humans suffering from this indication.
The short half-life of clotting factors (FVIII and FIX) dictates frequent administration when delivered as recombinant proteins. This limitation can be overcome with pDNA/EP gene therapy by enabling continuous production and release of these factors into circulation. This characteristic of pDNA/EP-based therapy could be beneficial to another blood disorder called chronic neutropenia. Chronic neutropenia is a condition where one exhibits low levels of neutrophils in the blood over an extended period of time [141,142]. Severe chronic neutropenia is treated with recombinant granulocyte colony stimulating factor (G-CSF), which is very efficacious in almost all cases [141,142,143]. However, the short half-life of G-CSF of about 3.5–3.8 h in serum means that the current standard of care often requires daily injections, an approach that is both costly and inconvenient for patients [144,145]. To address this, many attempts have been made to extend the half-life of G-CSF [145,146], and while moderately successful, these improvements do not considerably change the treatment regimen. EP of pDNA presents a potential alternative approach to address these challenges. With studies showing robust and durable expression of genes encoded by pDNA through pDNA/EP [123], there is an opportunity to concurrently increase expression levels as well as improve the pharmacokinetic profile of G-CSF, allowing for significantly less frequent dosing.
Hormone deficiency disorders, such as Addison’s disease, represent other chronic conditions that are driven by lack of a protein factor in circulation. Addison’s disease is characterized by insufficient hormonal production by the adrenal gland [147,148]. Current treatments for Addison’s disease are lifelong, consisting of daily oral pills taken to replace the missing hormones [147,148]. Studies in pigs showed that plasmids encoding growth hormone-releasing hormone (GHRH) delivered using pDNA/EP [149] stimulated growth hormone (GH) secretion over at least 2 months with a single treatment. Applying pDNA/EP to the treatment of Addison’s disease could potentially produce comparable outcomes, reflecting the success seen in pigs and thus reducing frequency of treatments necessary to manage the disease. While this study highlights the potential translation of a novel approach to treat Addison’s disease, it also demonstrates the versatility of pDNA/EP, emphasizing its ability to deliver diverse payloads.
In line with the adaptability of DNA-based medicines, pDNA/EP can also enhance the delivery of monoclonal antibodies for treating chronic diseases [15]. Multiple in vivo studies have demonstrated effective and durable delivery of DNA-based monoclonal antibodies using pDNA/EP [150,151,152], and phase 1 clinical trials are already in progress to evaluate the safety profile of this approach [151] (ClinicalTrialsGov; NCT05293249).
Chronic viral infections, a subset of chronic diseases, like those caused by the human immunodeficiency virus (HIV), could benefit from DNA-based methods for delivering antibodies. When left untreated, HIV leads to acquired immunodeficiency syndrome (AIDS) [153,154]. Although substantial progress has been made in developing treatments for HIV in recent decades, allowing people who have been diagnosed to live relatively normal lives, treatments are continuous and administered frequently [153,154]. Most antiretroviral therapies (ART) require daily treatment, comprised of one or more drugs [153,154] that potently decrease the viral load [154,155]. While treatment helps resolve symptoms and decreases chances of transmission, it does not cure HIV [154,155,156]. As a result, treatment needs to be maintained for life, making the dosing regimen of daily pills even more strenuous and time consuming. For patients where daily oral ART has accomplished viral suppression, treatment regimens can be switched to longer-acting injectable therapy [156,157]. This longer-acting therapy is administered once every 2 months and does not need to be taken with any other HIV medications [156,157]. However, these longer-acting treatments still require relatively frequent administration, making pDNA/EP an attractive alternative approach. Studies in mice and non-human primates (NHP) have demonstrated that DNA-encoded broadly neutralizing antibodies against HIV can be efficiently delivered with EP [158]. The DNA-encoded antibodies delivered by EP not only showed stable expression in mice for almost a year, but were also efficacious in NHPs against multiple HIV strains, highlighting potential advantages of pDNA/EP as an approach for treatment and possible prevention of this chronic disease [158].
Similarly, chronic inflammatory conditions represent a large disease area that could benefit from pDNA/EP treatment. For example, plaque psoriasis is a chronic, relapsing, immune inflammatory disorder, characterized by patches of dry, itchy, raised skin covered with a whitish buildup of dead cells [159,160]. Many biologics used to manage symptoms of plaque psoriasis are monoclonal antibodies. Dosing frequencies range from 4 to 8 weeks on average and require lifelong administration to remain efficacious [160,161]. These biologics could be delivered with pDNA/EP, potentially just once or twice a year, facilitating a durable and long-lasting expression of encoded antibodies to greatly improve the dosing regimen.

5. Concluding Remarks

In recent years, substantial progress has been made in the domain of DNA-based therapy, confirming its capacity to act as an alternative to conventional treatments [1]. AAV-based therapy has come a long way, with several therapies already FDA-approved, and many others being currently evaluated in clinical trials [18]. However, limitations of AAV remain, including complications due to immunity and large-scale manufacturing difficulties [42,103]. Continued work towards developing methods that avoid these obstacles is critical. Doing so will help to increase the therapeutic impact of AAV-based gene therapy and is of utmost importance in addressing chronic indications where multiple rounds of AAV might be necessary.
pDNA-based therapy is another DNA-based approach that has gained attention. pDNA demonstrates a strong safety profile and has low immunogenicity [11,151]. However, injection of pDNA on its own is incapable of producing expression levels that are high and stable enough to treat many indications [113,114]. Implementation of EP as a delivery method has helped to abrogate these drawbacks, enabling high, robust, and durable expression of pDNA-encoded therapeutics in vivo across many species [14,121,132]. Durable expression profiles support the use of this approach to address many chronic indications. In multiple disease models, pDNA/EP-based treatments have shown success, yielding robust expression and requiring fewer interventions to maintain therapeutic levels [139,149,158]. While success across various species suggests translatability to humans, scalability of pDNA/EP treatments requires further investigation to ensure efficient production of the biologic at therapeutic levels [132,133,134].
The pDNA/EP approach to gene therapy has proven its versatility, with the ability to deliver a diverse range of cargo. With previous success comes the hope that this approach will improve the landscape of chronic disease treatments, with one potential avenue being Glucagon-like peptide-1 (GLP-1) receptor agonists currently used to treat type 2 diabetes and obesity [162,163,164]. The current treatment necessitates weekly administration to maintain therapeutic efficacy, highlighting the potential of pDNA/EP as an appealing approach to revamp the dosing schedule for these prevalent chronic conditions. [162,163,164].
With approaches being investigated to address current limitations, an as new data emerge encouraging expansion into new indications, we can feel confident that DNA-based therapies will continue to flourish.

Author Contributions

C.S., R.A.L. and M.M.D. wrote and edited the manuscript, which all authors reviewed and approved for publication. All authors contributed to the article and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank the RenBio team for providing feedback on the manuscript and fruitful discussions.

Conflicts of Interest

C.S., R.A.L. and M.M.D. are employees of RenBio and as such receive salary and benefits, including ownership of stock and stock options, from the company. The company had no role in the design of the studies included in this review; in the collection, analyses, or interpretation of data; or in the decision to publish the results reviewed here.

References

  1. Bulaklak, K.; Gersbach, C.A. The once and future gene therapy. Nat. Commun. 2020, 11, 5820. [Google Scholar] [CrossRef] [PubMed]
  2. Wirth, T.; Parker, N.; Ylä-Herttuala, S. History of gene therapy. Gene 2013, 525, 162–169. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, D.; Gao, G. State-of-the-Art Human Gene Therapy: Part I. Gene Delivery Technologies. Discov. Med. 2014, 18, 67–77. [Google Scholar] [PubMed]
  4. Ma, C.-C.; Wang, Z.-L.; Xu, T.; He, Z.-Y.; Wei, Y.-Q. The approved gene therapy drugs worldwide: From 1998 to 2019. Biotechnol. Adv. 2020, 40, 107502. [Google Scholar] [CrossRef] [PubMed]
  5. Atchison, R.W.; Casto, B.C.; Hammon, W.M. Adenovirus-Associated Defective Virus Particles. Science 1965, 149, 754–756. [Google Scholar] [CrossRef] [PubMed]
  6. Flotte, T.R.; Afione, S.A.; Conrad, C.; McGrath, S.A.; Solow, R.; Oka, H.; Zeitlin, P.L.; Guggino, W.B.; Carter, B.J. Stable in vivo expression of the cystic fibrosis transmembrane conductance regulator with an adeno-associated virus vector. Proc. Natl. Acad. Sci. USA 1993, 90, 10613–10617. [Google Scholar] [CrossRef] [PubMed]
  7. Wagner, H.J.; Weber, W.; Fussenegger, M. Synthetic Biology: Emerging Concepts to Design and Advance Adeno-Associated Viral Vectors for Gene Therapy. Adv. Sci. 2021, 8, 2004018. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, D.; Tai, P.W.L.; Gao, G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat. Rev. Drug Discov. 2019, 18, 358–378. [Google Scholar] [CrossRef] [PubMed]
  9. Lundstrom, K. Viral Vectors in Gene Therapy: Where Do We Stand in 2023? Viruses 2023, 15, 698. [Google Scholar] [CrossRef] [PubMed]
  10. Bulcha, J.T.; Wang, Y.; Ma, H.; Tai, P.W.L.; Gao, G. Viral vector platforms within the gene therapy landscape. Signal Transduct. Target. Ther. 2021, 6, 53. [Google Scholar] [CrossRef] [PubMed]
  11. Hardee, C.L.; Arévalo-Soliz, L.M.; Hornstein, B.D.; Zechiedrich, L. Advances in Non-Viral DNA Vectors for Gene Therapy. Genes 2017, 8, 65. [Google Scholar] [CrossRef] [PubMed]
  12. Fynan, E.F.; Webster, R.G.; Fuller, D.H.; Haynes, J.R.; Santoro, J.C.; Robinson, H.L. DNA vaccines: Protective immunizations by parenteral, mucosal, and gene-gun inoculations. Proc. Natl. Acad. Sci. USA 1993, 90, 11478–11482. [Google Scholar] [CrossRef] [PubMed]
  13. Vaughan, E.E.; DeGiulio, J.V.; Dean, D.A. Intracellular Trafficking of Plasmids for Gene Therapy: Mechanisms of Cytoplasmic Movement and Nuclear Import. Curr. Gene Ther. 2006, 6, 671–681. [Google Scholar] [CrossRef] [PubMed]
  14. Widera, G.; Austin, M.; Rabussay, D.; Goldbeck, C.; Barnett, S.W.; Chen, M.; Leung, L.; Otten, G.R.; Thudium, K.; Selby, M.J.; et al. Increased DNA Vaccine Delivery and Immunogenicity by Electroporation In Vivo. J. Immunol. 2000, 164, 4635–4640. [Google Scholar] [CrossRef] [PubMed]
  15. Pagant, S.; Liberatore, R.A. In Vivo Electroporation of Plasmid DNA: A Promising Strategy for Rapid, Inexpensive, and Flexible Delivery of Anti-Viral Monoclonal Antibodies. Pharmaceutics 2021, 13, 1882. [Google Scholar] [CrossRef]
  16. Aihara, H.; Miyazaki, J. Gene transfer into muscle by electroporation in vivo. Nat. Biotechnol. 1998, 16, 867–870. [Google Scholar] [CrossRef] [PubMed]
  17. Mathiesen, I. Electropermeabilization of skeletal muscle enhances gene transfer in vivo. Gene Ther. 1999, 6, 508–514. [Google Scholar] [CrossRef] [PubMed]
  18. Pupo, A.; Fernández, A.; Low, S.H.; François, A.; Suárez-Amarán, L.; Samulski, R.J. AAV vectors: The Rubik’s cube of human gene therapy. Mol. Ther. 2022, 30, 3515–3541. [Google Scholar] [CrossRef] [PubMed]
  19. Meier, A.F.; Fraefel, C.; Seyffert, M. The Interplay between Adeno-Associated Virus and Its Helper Viruses. Viruses 2020, 12, 662. [Google Scholar] [CrossRef] [PubMed]
  20. Maurer, A.C.; Weitzman, M.D. Adeno-Associated Virus Genome Interactions Important for Vector Production and Transduction. Hum. Gene Ther. 2020, 31, 499–511. [Google Scholar] [CrossRef] [PubMed]
  21. Schultz, B.R.; Chamberlain, J.S. Recombinant adeno-associated virus transduction and integration. Mol. Ther. 2008, 16, 1189–1199. [Google Scholar] [CrossRef] [PubMed]
  22. Hermonat, P.L.; Muzyczka, N. Use of adeno-associated virus as a mammalian DNA cloning vector: Transduction of neomycin resistance into mammalian tissue culture cells. Proc. Natl. Acad. Sci. USA 1984, 81, 6466–6470. [Google Scholar] [CrossRef]
  23. Zhao, Z.; Anselmo, A.C.; Mitragotri, S. Viral vector-based gene therapies in the clinic. Bioeng. Transl. Med. 2022, 7, e10258. [Google Scholar] [CrossRef] [PubMed]
  24. Mendell, J.R.; Al-Zaidy, S.A.; Rodino-Klapac, L.R.; Goodspeed, K.; Gray, S.J.; Kay, C.N.; Boye, S.L.; Boye, S.E.; George, L.A.; Salabarria, S.; et al. Current Clinical Applications of In Vivo Gene Therapy with AAVs. Mol. Ther. 2021, 29, 464–488. [Google Scholar] [CrossRef] [PubMed]
  25. Ylä-Herttuala, S. Endgame: Glybera Finally Recommended for Approval as the First Gene Therapy Drug in the European Union. Mol. Ther. 2012, 20, 1831–1832. [Google Scholar] [CrossRef] [PubMed]
  26. Senior, M. After Glybera’s withdrawal, what’s next for gene therapy? Nat. Biotechnol. 2017, 35, 491–492. [Google Scholar] [CrossRef] [PubMed]
  27. Srivastava, A.; Lusby, E.W.; Berns, K.I. Nucleotide sequence and organization of the adeno-associated virus 2 genome. J. Virol. 1983, 45, 555–564. [Google Scholar] [CrossRef] [PubMed]
  28. Wu, Z.; Yang, H.; Colosi, P. Effect of Genome Size on AAV Vector Packaging. Mol. Ther. 2010, 18, 80–86. [Google Scholar] [CrossRef]
  29. Dong, J.Y.; Fan, P.D.; Frizzell, R.A. Quantitative analysis of the packaging capacity of recombinant adeno-associated virus. Hum. Gene Ther. 1996, 7, 2101–2112. [Google Scholar] [CrossRef]
  30. Issa, S.S.; Shaimardanova, A.A.; Solovyeva, V.V.; Rizvanov, A.A. Various AAV Serotypes and Their Applications in Gene Therapy: An Overview. Cells 2023, 12, 785. [Google Scholar] [CrossRef]
  31. Muzyczka, N.; Warrington, K.H. Custom Adeno-Associated Virus Capsids: The Next Generation of Recombinant Vectors with Novel Tropism. Hum. Gene Ther. 2005, 16, 408–416. [Google Scholar] [CrossRef] [PubMed]
  32. White, A.F.; Mazur, M.; Sorscher, E.J.; Zinn, K.R.; Ponnazhagan, S. Genetic Modification of Adeno-Associated Viral Vector Type 2 Capsid Enhances Gene Transfer Efficiency in Polarized Human Airway Epithelial Cells. Hum. Gene Ther. 2008, 19, 1407–1414. [Google Scholar] [CrossRef]
  33. Huang, Q.; Chen, A.T.; Chan, K.Y.; Sorensen, H.; Barry, A.J.; Azari, B.; Zheng, Q.; Beddow, T.; Zhao, B.; Tobey, I.G.; et al. Targeting AAV vectors to the central nervous system by engineering capsid-receptor interactions that enable crossing of the blood-brain barrier. PLoS Biol. 2023, 21, e3002112. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, L.; Rossi, A.; Lange, L.; Meumann, N.; Koitzsch, U.; Christie, K.; Nesbit, M.A.; Moore, C.B.T.; Hacker, U.T.; Morgan, M.; et al. Capsid Engineering Overcomes Barriers toward Adeno-Associated Virus Vector-Mediated Transduction of Endothelial Cells. Hum. Gene Ther. 2019, 30, 1284–1296. [Google Scholar] [CrossRef] [PubMed]
  35. Zincarelli, C.; Soltys, S.; Rengo, G.; Rabinowitz, J.E. Analysis of AAV Serotypes 1–9 Mediated Gene Expression and Tropism in Mice After Systemic Injection. Mol. Ther. 2008, 16, 1073–1080. [Google Scholar] [CrossRef] [PubMed]
  36. Dhungel, B.P.; Winburn, I.; Pereira, C.d.F.; Huang, K.; Chhabra, A.; Rasko, J.E.J. Understanding AAV vector immunogenicity: From particle to patient. Theranostics 2024, 14, 1260–1288. [Google Scholar] [CrossRef] [PubMed]
  37. Klamroth, R.; Hayes, G.; Andreeva, T.; Gregg, K.; Suzuki, T.; Mitha, I.H.; Hardesty, B.; Shima, M.; Pollock, T.; Slev, P.; et al. Global Seroprevalence of Pre-existing Immunity Against AAV5 and Other AAV Serotypes in People with Hemophilia A. Hum. Gene Ther. 2022, 33, 432–441. [Google Scholar] [CrossRef] [PubMed]
  38. Schulz, M.; Levy, D.I.; Petropoulos, C.J.; Bashirians, G.; Winburn, I.; Mahn, M.; Somanathan, S.; Cheng, S.H.; Byrne, B.J. Binding and neutralizing anti-AAV antibodies: Detection and implications for rAAV-mediated gene therapy. Mol. Ther. 2023, 31, 616–630. [Google Scholar] [CrossRef]
  39. Pipe, S.; Leebeek, F.W.G.; Ferreira, V.; Sawyer, E.K.; Pasi, J. Clinical Considerations for Capsid Choice in the Development of Liver-Targeted AAV-Based Gene Transfer. Mol. Ther. Methods Clin. Dev. 2019, 15, 170–178. [Google Scholar] [CrossRef] [PubMed]
  40. Mendell, J.R.; Connolly, A.M.; Lehman, K.J.; Griffin, D.A.; Khan, S.Z.; Dharia, S.D.; Quintana-Gallardo, L.; Rodino-Klapac, L.R. Testing preexisting antibodies prior to AAV gene transfer therapy: Rationale, lessons and future considerations. Mol. Ther. Methods Clin. Dev. 2022, 25, 74–83. [Google Scholar] [CrossRef] [PubMed]
  41. Mingozzi, F.; High, K.A. Immune responses to AAV in clinical trials. Curr. Gene Ther. 2007, 7, 316–324. [Google Scholar] [CrossRef] [PubMed]
  42. Arjomandnejad, M.; Dasgupta, I.; Flotte, T.R.; Keeler, A.M. Immunogenicity of Recombinant Adeno-Associated Virus (AAV) Vectors for Gene Transfer. BioDrugs 2023, 37, 311–329. [Google Scholar] [CrossRef] [PubMed]
  43. Ertl, H.C.J. Immunogenicity and toxicity of AAV gene therapy. Front. Immunol. 2022, 13, 975803. [Google Scholar] [CrossRef] [PubMed]
  44. Manno, C.S.; Pierce, G.F.; Arruda, V.R.; Glader, B.; Ragni, M.; Rasko, J.J.E.; Ozelo, M.C.; Hoots, K.; Blatt, P.; Konkle, B.; et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat. Med. 2006, 12, 342–347. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, Y.; Shao, W. Innate Immune Response to Viral Vectors in Gene Therapy. Viruses 2023, 15, 1801. [Google Scholar] [CrossRef] [PubMed]
  46. Shen, W.; Liu, S.; Ou, L. rAAV immunogenicity, toxicity, and durability in 255 clinical trials: A meta-analysis. Front. Immunol. 2022, 13, 1001263. [Google Scholar] [CrossRef] [PubMed]
  47. Kreppel, F.; Hagedorn, C. Capsid and Genome Modification Strategies to Reduce the Immunogenicity of Adenoviral Vectors. Int. J. Mol. Sci. 2021, 22, 2417. [Google Scholar] [CrossRef] [PubMed]
  48. Muhuri, M.; Maeda, Y.; Ma, H.; Ram, S.; Fitzgerald, K.A.; Tai, P.W.L.; Gao, G. Overcoming innate immune barriers that impede AAV gene therapy vectors. J. Clin. Investig. 2021, 131, e143780. [Google Scholar] [CrossRef] [PubMed]
  49. Kishimoto, T.K.; Samulski, R.J. Addressing high dose AAV toxicity—‘one and done’ or ‘slower and lower’? Expert. Opin. Biol. Ther. 2022, 22, 1067–1071. [Google Scholar] [CrossRef] [PubMed]
  50. He, X.; Urip, B.A.; Zhang, Z.; Ngan, C.C.; Feng, B. Evolving AAV-delivered therapeutics towards ultimate cures. J. Mol. Med. 2021, 99, 593–617. [Google Scholar] [CrossRef] [PubMed]
  51. Li, C.; Samulski, R.J. Engineering adeno-associated virus vectors for gene therapy. Nat. Rev. Genet. 2020, 21, 255–272. [Google Scholar] [CrossRef] [PubMed]
  52. Shitik, E.M.; Shalik, I.K.; Yudkin, D.V. AAV-based vector improvements unrelated to capsid protein modification. Front. Med. 2023, 10, 1106085. [Google Scholar] [CrossRef] [PubMed]
  53. Kolesnik, V.V.; Nurtdinov, R.F.; Oloruntimehin, E.S.; Karabelsky, A.V.; Malogolovkin, A.S. Optimization strategies and advances in the research and development of AAV-based gene therapy to deliver large transgenes. Clin. Transl. Med. 2024, 14, e1607. [Google Scholar] [CrossRef] [PubMed]
  54. Nathwani, A.C.; Reiss, U.M.; Tuddenham, E.G.D.; Rosales, C.; Chowdary, P.; McIntosh, J.; Della Peruta, M.; Lheriteau, E.; Patel, N.; Raj, D.; et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N. Engl. J. Med. 2014, 371, 1994–2004. [Google Scholar] [CrossRef] [PubMed]
  55. Colella, P.; Ronzitti, G.; Mingozzi, F. Emerging Issues in AAV-Mediated In Vivo Gene Therapy. Mol. Ther. Methods Clin. Dev. 2017, 8, 87–104. [Google Scholar] [CrossRef] [PubMed]
  56. Domenger, C.; Grimm, D. Next-generation AAV vectors—Do not judge a virus (only) by its cover. Hum. Mol. Genet. 2019, 28, R3–R14. [Google Scholar] [CrossRef] [PubMed]
  57. Rodríguez-Merchán, E.C.; De Pablo-Moreno, J.A.; Liras, A. Gene Therapy in Hemophilia: Recent Advances. Int. J. Mol. Sci. 2021, 22, 7647. [Google Scholar] [CrossRef] [PubMed]
  58. Lisowski, L.; Staber, J.M.; Wright, J.F.; Valentino, L.A. The intersection of vector biology, gene therapy, and hemophilia. Res. Pract. Thromb. Haemost. 2021, 5, e12586. [Google Scholar] [CrossRef] [PubMed]
  59. Leebeek, F.W.G.; Miesbach, W. Gene therapy for hemophilia: A review on clinical benefit, limitations, and remaining issues. Blood 2021, 138, 923–931. [Google Scholar] [CrossRef] [PubMed]
  60. Castaman, G.; Di Minno, G.; De Cristofaro, R.; Peyvandi, F. The Arrival of Gene Therapy for Patients with Hemophilia A. Int. J. Mol. Sci. 2022, 23, 10228. [Google Scholar] [CrossRef] [PubMed]
  61. Sarmiento Doncel, S.; Díaz Mosquera, G.A.; Cortes, J.M.; Agudelo Rico, C.; Meza Cadavid, F.J.; Peláez, R.G. Haemophilia A: A Review of Clinical Manifestations, Treatment, Mutations, and the Development of Inhibitors. Hematol. Rep. 2023, 15, 130–150. [Google Scholar] [CrossRef] [PubMed]
  62. Berntorp, E.; Fischer, K.; Hart, D.P.; Mancuso, M.E.; Stephensen, D.; Shapiro, A.D.; Blanchette, V. Haemophilia. Nat. Rev. Dis. Primers 2021, 7, 45. [Google Scholar] [CrossRef] [PubMed]
  63. Castaman, G.; Matino, D. Hemophilia A and B: Molecular and clinical similarities and differences. Haematologica 2019, 104, 1702–1709. [Google Scholar] [CrossRef]
  64. Dolan, G.; Benson, G.; Duffy, A.; Hermans, C.; Jiménez-Yuste, V.; Lambert, T.; Ljung, R.; Morfini, M.; Zupančić Šalek, S. Haemophilia B: Where are we now and what does the future hold? Blood Rev. 2018, 32, 52–60. [Google Scholar] [CrossRef]
  65. Marchesini, E.; Morfini, M.; Valentino, L. Recent Advances in the Treatment of Hemophilia: A Review. Biol. Targets Ther. 2021, 15, 221–235. [Google Scholar] [CrossRef] [PubMed]
  66. Makris, M. Prophylaxis in haemophilia should be life-long. Blood Transfus. 2012, 10, 165–168. [Google Scholar] [CrossRef] [PubMed]
  67. Blair, H.A. Valoctocogene Roxaparvovec: First Approval. Drugs 2022, 82, 1505–1510. [Google Scholar] [CrossRef]
  68. Nathwani, A.C. Gene therapy for hemophilia. Hematol. Am. Soc. Hematol. Educ. Program. 2022, 2022, 569–578. [Google Scholar] [CrossRef]
  69. George, L.A.; Monahan, P.E.; Eyster, M.E.; Sullivan, S.K.; Ragni, M.V.; Croteau, S.E.; Rasko, J.E.J.; Recht, M.; Samelson-Jones, B.J.; MacDougall, A.; et al. Multiyear Factor VIII Expression after AAV Gene Transfer for Hemophilia A. N. Engl. J. Med. 2021, 385, 1961–1973. [Google Scholar] [CrossRef]
  70. George, L.A.; Sullivan, S.K.; Giermasz, A.; Rasko, J.E.J.; Samelson-Jones, B.J.; Ducore, J.; Cuker, A.; Sullivan, L.M.; Majumdar, S.; Teitel, J.; et al. Hemophilia B Gene Therapy with a High-Specific-Activity Factor IX Variant. N. Engl. J. Med. 2017, 377, 2215–2227. [Google Scholar] [CrossRef]
  71. George, L.A.; Sullivan, S.K.; Rasko, J.E.J.; Giermasz, A.; Samelson-Jones, B.J.; Ducore, J.M.; Teitel, J.M.; McGuinn, C.E.; Runowski, A.R.; Wright, F.; et al. Efficacy and Safety in 15 Hemophilia B Patients Treated with the AAV Gene Therapy Vector Fidanacogene Elaparvovec and Followed for at Least 1 Year. Blood 2019, 134, 3347. [Google Scholar] [CrossRef]
  72. Frenzel, L.; Kavakli, K.; Klamroth, R.; Chiou, S.-S.; Shapiro, A.D.; Sun, P.; Fuiman, J.; McKay, J.; Fang, A.F.; Biondo, F.; et al. Characterizing a Cohort of Patients with Hemophilia B Treated with Fidanacogene Elaparvovec from the Phase 3 Benegene-2 Study Who Returned to Factor IX Prophylaxis. Blood 2023, 142, 2257. [Google Scholar] [CrossRef]
  73. Dev, S.; Kruse, R.L.; Hamilton, J.P.; Lutsenko, S. Wilson Disease: Update on Pathophysiology and Treatment. Front. Cell Dev. Biol. 2022, 10, 871877. [Google Scholar] [CrossRef] [PubMed]
  74. Shribman, S.; Poujois, A.; Bandmann, O.; Czlonkowska, A.; Warner, T.T. Wilson’s disease: Update on pathogenesis, biomarkers and treatments. J. Neurol. Neurosurg. Psychiatry 2021, 92, 1053–1061. [Google Scholar] [CrossRef] [PubMed]
  75. Greig, J.A.; Nordin, J.M.L.; Smith, M.K.; Ashley, S.N.; Draper, C.; Zhu, Y.; Bell, P.; Buza, E.L.; Wilson, J.M. A Gene Therapy Approach to Improve Copper Metabolism and Prevent Liver Damage in a Mouse Model of Wilson Disease. Hum. Gene Ther. Clin. Dev. 2019, 30, 29–39. [Google Scholar] [CrossRef]
  76. Murillo, O.; Luqui, D.M.; Gazquez, C.; Martinez-Espartosa, D.; Navarro-Blasco, I.; Monreal, J.I.; Guembe, L.; Moreno-Cermeño, A.; Corrales, F.J.; Prieto, J.; et al. Long-term metabolic correction of Wilson’s disease in a murine model by gene therapy. J. Hepatol. 2016, 64, 419–426. [Google Scholar] [CrossRef] [PubMed]
  77. Padula, A.; Petruzzelli, R.; Philbert, S.A.; Church, S.J.; Esposito, F.; Campione, S.; Monti, M.; Capolongo, F.; Perna, C.; Nusco, E.; et al. Full-length ATP7B reconstituted through protein trans-splicing corrects Wilson disease in mice. Mol. Ther. Methods Clin. Dev. 2022, 26, 495–504. [Google Scholar] [CrossRef] [PubMed]
  78. Baruteau, J.; Brunetti-Pierri, N.; Gissen, P. Liver-directed gene therapy for inherited metabolic diseases. J. Inherit. Metab. Dis. 2024, 47, 9–21. [Google Scholar] [CrossRef] [PubMed]
  79. Baris, H.N.; Cohen, I.J.; Mistry, P.K. Gaucher disease: The metabolic defect, pathophysiology, phenotypes and natural history. Pediatr. Endocrinol. Rev. PER 2014, 12 (Suppl. 1), 72–81. [Google Scholar] [PubMed]
  80. Rosenbloom, B.E.; Weinreb, N.J. Gaucher disease: A comprehensive review. In Advances in Gaucher Disease: Basic and Clinical Perspectives; Future Medicine Ltd.: London, UK, 2013; pp. 26–49. [Google Scholar]
  81. Nalysnyk, L.; Rotella, P.; Simeone, J.C.; Hamed, A.; Weinreb, N. Gaucher disease epidemiology and natural history: A comprehensive review of the literature. Hematology 2017, 22, 65–73. [Google Scholar] [CrossRef] [PubMed]
  82. Roshan Lal, T.; Sidransky, E. The Spectrum of Neurological Manifestations Associated with Gaucher Disease. Diseases 2017, 5, 10. [Google Scholar] [CrossRef] [PubMed]
  83. Gary, S.E.; Ryan, E.; Steward, A.M.; Sidransky, E. Recent advances in the diagnosis and management of Gaucher disease. Expert Rev. Endocrinol. Metab. 2018, 13, 107–118. [Google Scholar] [CrossRef] [PubMed]
  84. Davidson, B.A.; Hassan, S.; Garcia, E.J.; Tayebi, N.; Sidransky, E. Exploring Genetic Modifiers of Gaucher Disease: The Next Horizon. Hum. Mutat. 2018, 39, 1739–1751. [Google Scholar] [CrossRef] [PubMed]
  85. Ramamoorth, M.; Narvekar, A. Non Viral Vectors in Gene Therapy—An Overview. J. Clin. Diagn. Res. 2015, 9, GE01–GE06. [Google Scholar] [CrossRef] [PubMed]
  86. Comerota, A.J.; Throm, R.C.; Miller, K.A.; Henry, T.; Chronos, N.; Laird, J.; Sequeira, R.; Kent, C.K.; Bacchetta, M.; Goldman, C.; et al. Naked plasmid DNA encoding fibroblast growth factor type 1 for the treatment of end-stage unreconstructible lower extremity ischemia: Preliminary results of a phase I trial. J. Vasc. Surg. 2002, 35, 930–936. [Google Scholar] [CrossRef] [PubMed]
  87. Disis, M.L.; Guthrie, K.A.; Liu, Y.; Coveler, A.L.; Higgins, D.M.; Childs, J.S.; Dang, Y.; Salazar, L.G. Safety and Outcomes of a Plasmid DNA Vaccine Encoding the ERBB2 Intracellular Domain in Patients with Advanced-Stage ERBB2-Positive Breast Cancer: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol. 2023, 9, 71–78. [Google Scholar] [CrossRef] [PubMed]
  88. Khobragade, A.; Bhate, S.; Ramaiah, V.; Deshpande, S.; Giri, K.; Phophle, H.; Supe, P.; Godara, I.; Revanna, R.; Nagarkar, R.; et al. Efficacy, safety, and immunogenicity of the DNA SARS-CoV-2 vaccine (ZyCoV-D): The interim efficacy results of a phase 3, randomised, double-blind, placebo-controlled study in India. Lancet 2022, 399, 1313–1321. [Google Scholar] [CrossRef] [PubMed]
  89. Schalk, J.A.C.; Mooi, F.R.; Berbers, G.A.M.; van Aerts, L.A.G.J.M.; Ovelgönne, H.; Kimman, T.G. Preclinical and Clinical Safety Studies on DNA Vaccines. Hum. Vaccines 2006, 2, 45–53. [Google Scholar] [CrossRef] [PubMed]
  90. Donsante, A.; Miller, D.G.; Li, Y.; Vogler, C.; Brunt, E.M.; Russell, D.W.; Sands, M.S. AAV Vector Integration Sites in Mouse Hepatocellular Carcinoma. Science 2007, 317, 477. [Google Scholar] [CrossRef] [PubMed]
  91. Chandler, R.J.; LaFave, M.C.; Varshney, G.K.; Trivedi, N.S.; Carrillo-Carrasco, N.; Senac, J.S.; Wu, W.; Hoffmann, V.; Elkahloun, A.G.; Burgess, S.M.; et al. Vector design influences hepatic genotoxicity after adeno-associated virus gene therapy. J. Clin. Investig. 2015, 125, 870–880. [Google Scholar] [CrossRef] [PubMed]
  92. Nakai, H.; Montini, E.; Fuess, S.; Storm, T.A.; Grompe, M.; Kay, M.A. AAV serotype 2 vectors preferentially integrate into active genes in mice. Nat. Genet. 2003, 34, 297–302. [Google Scholar] [CrossRef] [PubMed]
  93. Lozier, J.N.; Csako, G.; Mondoro, T.H.; Krizek, D.M.; Metzger, M.E.; Costello, R.; Vostal, J.G.; Rick, M.E.; Donahue, R.E.; Morgan, R.A. Toxicity of a First-Generation Adenoviral Vector in Rhesus Macaques. Hum. Gene Ther. 2002, 13, 113–124. [Google Scholar] [CrossRef] [PubMed]
  94. Sheets, R.L.; Stein, J.; Manetz, T.S.; Andrews, C.; Bailer, R.; Rathmann, J.; Gomez, P.L. Toxicological Safety Evaluation of DNA Plasmid Vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile Virus Is Similar Despite Differing Plasmid Backbones or Gene-Inserts. Toxicol. Sci. 2006, 91, 620–630. [Google Scholar] [CrossRef] [PubMed]
  95. Sheets, R.L.; Stein, J.; Manetz, T.S.; Duffy, C.; Nason, M.; Andrews, C.; Kong, W.-P.; Nabel, G.J.; Gomez, P.L. Biodistribution of DNA Plasmid Vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile Virus Is Similar, without Integration, despite Differing Plasmid Backbones or Gene Inserts. Toxicol. Sci. 2006, 91, 610–619. [Google Scholar] [CrossRef] [PubMed]
  96. Patel, A.; Bah, M.A.; Weiner, D.B. In Vivo Delivery of Nucleic Acid-Encoded Monoclonal Antibodies. BioDrugs 2020, 34, 273–293. [Google Scholar] [CrossRef] [PubMed]
  97. Manam, S.; Ledwith, B.J.; Barnum, A.B.; Troilo, P.J.; Pauley, C.J.; Harper, L.B.; Griffiths, T.G., II; Niu, Z.; Denisova, L.; Follmer, T.T.; et al. Plasmid DNA Vaccines: Tissue Distribution and Effects of DNA Sequence, Adjuvants and Delivery Method on Integration into Host DNA. Intervirology 2001, 43, 273–281. [Google Scholar] [CrossRef] [PubMed]
  98. Kang, K.K.; Choi, S.M.; Choi, J.H.; Lee, D.S.; Kim, C.Y.; Ahn, B.O.; Kim, B.M.; Kim, W.B. Safety Evaluation of GX-12, a New HIV Therapeutic Vaccine: Investigation of Integration into the Host Genome and Expression in the Reproductive Organs. Intervirology 2003, 46, 270–276. [Google Scholar] [CrossRef] [PubMed]
  99. Vilalta, A.; Mahajan, R.K.; Hartikka, J.; Rusalov, D.; Martin, T.; Bozoukova, V.; Leamy, V.; Hall, K.; Lalor, P.; Rolland, A.; et al. I. Poloxamer-Formulated Plasmid DNA-Based Human Cytomegalovirus Vaccine: Evaluation of Plasmid DNA Biodistribution/Persistence and Integration. Hum. Gene Ther. 2005, 16, 1143–1150. [Google Scholar] [CrossRef] [PubMed]
  100. Gill, D.R.; Pringle, I.A.; Hyde, S.C. Progress and Prospects: The design and production of plasmid vectors. Gene Ther. 2009, 16, 165–171. [Google Scholar] [CrossRef] [PubMed]
  101. Zheng, C.; Baum, B.J. Evaluation of Promoters for Use in Tissue-Specific Gene Delivery. In Gene Therapy Protocols; Doux, J.M., Ed.; Humana Press: Totowa, NJ, USA, 2008; pp. 205–219. [Google Scholar]
  102. van der Loo, J.C.M.; Wright, J.F. Progress and challenges in viral vector manufacturing. Hum. Mol. Genet. 2016, 25, R42–R52. [Google Scholar] [CrossRef] [PubMed]
  103. Jiang, Z.; Dalby, P.A. Challenges in scaling up AAV-based gene therapy manufacturing. Trends Biotechnol. 2023, 41, 1268–1281. [Google Scholar] [CrossRef] [PubMed]
  104. Matange, K.; Tuck, J.M.; Keung, A.J. DNA stability: A central design consideration for DNA data storage systems. Nat. Commun. 2021, 12, 1358. [Google Scholar] [CrossRef] [PubMed]
  105. Howard, D.B.; Harvey, B.K. Assaying the Stability and Inactivation of AAV Serotype 1 Vectors. Hum. Gene Ther. Methods 2017, 28, 39–48. [Google Scholar] [CrossRef] [PubMed]
  106. Mairhofer, J.; Grabherr, R. Rational Vector Design for Efficient Non-viral Gene Delivery: Challenges Facing the Use of Plasmid DNA. Mol. Biotechnol. 2008, 39, 97–104. [Google Scholar] [CrossRef] [PubMed]
  107. Williams, J.A.; Carnes, A.E.; Hodgson, C.P. Plasmid DNA Vaccine vector design: Impact on efficacy, safety and upstream production. Biotechnol. Adv. 2009, 27, 353–370. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, Z.-Y.; Riu, E.; He, C.-Y.; Xu, H.; Kay, M.A. Silencing of Episomal Transgene Expression in Liver by Plasmid Bacterial Backbone DNA Is Independent of CpG Methylation. Mol. Ther. 2008, 16, 548–556. [Google Scholar] [CrossRef] [PubMed]
  109. Darquet, A.-M.; Rangara, R.; Kreiss, P.; Schwartz, B.; Naimi, S.; Delaère, P.; Crouzet, J.; Scherman, D. Minicircle: An improved DNA molecule for in vitro and in vivo gene transfer. Gene Ther. 1999, 6, 209–218. [Google Scholar] [CrossRef] [PubMed]
  110. Kay, M.A.; He, C.-Y.; Chen, Z.-Y. A robust system for production of minicircle DNA vectors. Nat. Biotechnol. 2010, 28, 1287–1289. [Google Scholar] [CrossRef] [PubMed]
  111. Barreira, M.; Kerridge, C.; Jorda, S.; Olofsson, D.; Neumann, A.; Horton, H.; Smith-Moore, S. Enzymatically amplified linear dbDNATM as a rapid and scalable solution to industrial lentiviral vector manufacturing. Gene Ther. 2023, 30, 122–131. [Google Scholar] [CrossRef] [PubMed]
  112. Vandermeulen, G.; Marie, C.; Scherman, D.; Préat, V. New Generation of Plasmid Backbones Devoid of Antibiotic Resistance Marker for Gene Therapy Trials. Mol. Ther. 2011, 19, 1942–1949. [Google Scholar] [CrossRef] [PubMed]
  113. Dean, D.A. Nonviral gene transfer to skeletal, smooth, and cardiac muscle in living animals. Am. J. Physiol.-Cell Physiol. 2005, 289, C233–C245. [Google Scholar] [CrossRef] [PubMed]
  114. Young, J.L.; Dean, D.A. Nonviral gene transfer strategies for the vasculature. Microcirculation 2002, 9, 35–49. [Google Scholar] [CrossRef] [PubMed]
  115. Mali, S. Delivery systems for gene therapy. Indian. J. Hum. Genet. 2013, 19, 3–8. [Google Scholar] [CrossRef] [PubMed]
  116. Sung, Y.K.; Kim, S.W. Recent advances in the development of gene delivery systems. Biomater. Res. 2019, 23, 8. [Google Scholar] [CrossRef]
  117. Du, X.; Wang, J.; Zhou, Q.; Zhang, L.; Wang, S.; Zhang, Z.; Yao, C. Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv. 2018, 25, 1516–1525. [Google Scholar] [CrossRef]
  118. Thi, T.T.H.; Suys, E.J.A.; Lee, J.S.; Nguyen, D.H.; Park, K.D.; Truong, N.P. Lipid-Based Nanoparticles in the Clinic and Clinical Trials: From Cancer Nanomedicine to COVID-19 Vaccines. Vaccines 2021, 9, 359. [Google Scholar] [CrossRef] [PubMed]
  119. Jerzykiewicz, J.; Czogalla, A. Polyethyleneimine-Based Lipopolyplexes as Carriers in Anticancer Gene Therapies. Materials 2022, 15, 179. [Google Scholar] [CrossRef]
  120. Wang, Y.; Wang, R.; Wu, S.; An, J.; Liang, Y.; Hou, L.; Zhang, Z. Self-responsive co-delivery system for remodeling tumor intracellular microenvironment to promote PTEN-mediated anti-tumor therapy. Nanoscale 2020, 12, 9392–9403. [Google Scholar] [CrossRef]
  121. Mir, L.M.; Bureau, M.F.; Gehl, J.; Rangara, R.; Rouy, D.; Caillaud, J.-M.; Delaere, P.; Branellec, D.; Schwartz, B.; Scherman, D. High-efficiency gene transfer into skeletal muscle mediated by electric pulses. Proc. Natl. Acad. Sci. USA 1999, 96, 4262–4267. [Google Scholar] [CrossRef] [PubMed]
  122. Rosazza, C.; Meglic, S.H.; Zumbusch, A.; Rols, M.-P.; Miklavcic, D. Gene Electrotransfer: A Mechanistic Perspective. Curr. Gene Ther. 2016, 16, 98–129. [Google Scholar] [CrossRef]
  123. Todorova, B.; Adam, L.; Culina, S.; Boisgard, R.; Martinon, F.; Cosma, A.; Ustav, M.; Kortulewski, T.; Le Grand, R.; Chapon, C. Electroporation as a vaccine delivery system and a natural adjuvant to intradermal administration of plasmid DNA in macaques. Sci. Rep. 2017, 7, 4122. [Google Scholar] [CrossRef] [PubMed]
  124. Han, M.; Nguyen, B.; Lee, J.Y.; Browning, E.; Zhang, J.; Mukhopadhyay, A.; Gujar, R.; Salazar, J.; Hermiz, R.; Svenson, L.; et al. Intratumoral Electroporation of Plasmid Encoded IL12 and Membrane-Anchored Anti-CD3 Increases Systemic Tumor Immunity. Mol. Cancer Res. MCR 2022, 20, 983–995. [Google Scholar] [CrossRef] [PubMed]
  125. Bhatia, S.; Longino, N.V.; Miller, N.J.; Kulikauskas, R.; Iyer, J.G.; Ibrani, D.; Blom, A.; Byrd, D.R.; Parvathaneni, U.; Twitty, C.G.; et al. Intratumoral Delivery of Plasmid IL12 Via Electroporation Leads to Regression of Injected and Noninjected Tumors in Merkel Cell Carcinoma. Clin. Cancer Res. 2020, 26, 598–607. [Google Scholar] [CrossRef] [PubMed]
  126. Greaney, S.K.; Algazi, A.P.; Tsai, K.K.; Takamura, K.T.; Chen, L.; Twitty, C.G.; Zhang, L.; Paciorek, A.; Pierce, R.H.; Le, M.H.; et al. Intratumoral Plasmid IL12 Electroporation Therapy in Patients with Advanced Melanoma Induces Systemic and Intratumoral T-cell Responses. Cancer Immunol. Res. 2020, 8, 246–254. [Google Scholar] [CrossRef] [PubMed]
  127. Young, J.L.; Dean, D.A. Chapter Three—Electroporation-Mediated Gene Delivery. In Advances in Genetics; Huang, L., Liu, D., Wagner, E., Eds.; Academic Press: Cambridge, MA, USA, 2015; pp. 49–88. [Google Scholar]
  128. Bhattacharya, S.; Silkunas, M.; Gudvangen, E.; Mangalanathan, U.; Pakhomova, O.N.; Pakhomov, A.G. Ca2+ dependence and kinetics of cell membrane repair after electropermeabilization. Biochim. Biophys. Acta BBA-Biomembr. 2022, 1864, 183823. [Google Scholar] [CrossRef] [PubMed]
  129. Silkunas, M.; Silkuniene, G.; Pakhomov, A.G. Real-time imaging of individual electropores proves their longevity in cells. Biochem. Biophys. Res. Commun. 2024, 695, 149408. [Google Scholar] [CrossRef] [PubMed]
  130. Wegener, J.; Keese, C.R.; Giaever, I. Recovery of Adherent Cells after In Situ Electroporation Monitored Electrically. BioTechniques 2002, 33, 348–357. [Google Scholar] [CrossRef] [PubMed]
  131. Peng, B.; Zhao, Y.; Lu, H.; Pang, W.; Xu, Y. In vivo plasmid DNA electroporation resulted in transfection of satellite cells and lasting transgene expression in regenerated muscle fibers. Biochem. Biophys. Res. Commun. 2005, 338, 1490–1498. [Google Scholar] [CrossRef] [PubMed]
  132. Hollevoet, K.; Thomas, D.; Compernolle, G.; Vermeire, G.; De Smidt, E.; De Vleeschauwer, S.; Smith, T.R.F.; Fisher, P.D.; Dewilde, M.; Geukens, N.; et al. Clinically relevant dosing and pharmacokinetics of DNA-encoded antibody therapeutics in a sheep model. Front. Oncol. 2022, 12, 1017612. [Google Scholar] [CrossRef]
  133. Andrews, C.D.; Huang, Y.; Ho, D.D.; Liberatore, R.A. In vivo expressed biologics for infectious disease prophylaxis: Rapid delivery of DNA-based antiviral antibodies. Emerg. Microbes Infect. 2020, 9, 1523–1533. [Google Scholar] [CrossRef]
  134. Hollevoet, K.; De Vleeschauwer, S.; De Smidt, E.; Vermeire, G.; Geukens, N.; Declerck, P. Bridging the Clinical Gap for DNA-Based Antibody Therapy Through Translational Studies in Sheep. Hum. Gene Ther. 2019, 30, 1431–1443. [Google Scholar] [CrossRef] [PubMed]
  135. McNee, A.; Smith, T.R.F.; Holzer, B.; Clark, B.; Bessell, E.; Guibinga, G.; Brown, H.; Schultheis, K.; Fisher, P.; Ramos, S.; et al. Establishment of a Pig Influenza Challenge Model for Evaluation of Monoclonal Antibody Delivery Platforms. J. Immunol. 2020, 205, 648–660. [Google Scholar] [CrossRef] [PubMed]
  136. Mau, T.; Amin, M.R.; Belafsky, P.C.; Best, S.R.; Friedman, A.D.; Klein, A.M.; Lott, D.G.; Paniello, R.C.; Pransky, S.M.; Saba, N.F.; et al. Interim Results of a Phase 1/2 Open-Label Study of INO-3107 for HPV-6 and/or HPV-11-Associated Recurrent Respiratory Papillomatosis. Laryngoscope 2023, 133, 3087–3093. [Google Scholar] [CrossRef] [PubMed]
  137. Xu, Z.; Wise, M.C.; Choi, H.; Perales-Puchalt, A.; Patel, A.; Tello-Ruiz, E.; Chu, J.D.; Muthumani, K.; Weiner, D.B. Synthetic DNA delivery by electroporation promotes robust in vivo sulfation of broadly neutralizing anti-HIV immunoadhesin eCD4-Ig. eBioMedicine 2018, 35, 97–105. [Google Scholar] [CrossRef] [PubMed]
  138. Edupuganti, S.; De Rosa, S.C.; Elizaga, M.; Lu, Y.; Han, X.; Huang, Y.; Swann, E.; Polakowski, L.; Kalams, S.A.; Keefer, M.; et al. Intramuscular and Intradermal Electroporation of HIV-1 PENNVAX-GP® DNA Vaccine and IL-12 Is Safe, Tolerable, Acceptable in Healthy Adults. Vaccines 2020, 8, 741. [Google Scholar] [CrossRef] [PubMed]
  139. Bettan, M.; Emmanuel, F.; Darteil, R.; Caillaud, J.-M.; Soubrier, F.; Delaere, P.; Branelec, D.; Mahfoudi, A.; Duverger, N.; Scherman, D. High-Level Protein Secretion into Blood Circulation after Electric Pulse-Mediated Gene Transfer into Skeletal Muscle. Mol. Ther. 2000, 2, 204–210. [Google Scholar] [CrossRef] [PubMed]
  140. Fewell, J.G.; MacLaughlin, F.; Mehta, V.; Gondo, M.; Nicol, F.; Wilson, E.; Smith, L.C. Gene Therapy for the Treatment of Hemophilia B Using PINC-Formulated Plasmid Delivered to Muscle with Electroporation. Mol. Ther. 2001, 3, 574–583. [Google Scholar] [CrossRef] [PubMed]
  141. Dale, D.C.; Bolyard, A.A. An update on the diagnosis and treatment of chronic idiopathic neutropenia. Curr. Opin. Hematol. 2017, 24, 46. [Google Scholar] [CrossRef]
  142. Rezaei, N.; Moazzami, K.; Aghamohammadi, A.; Klein, C. Neutropenia and Primary Immunodeficiency Diseases. Int. Rev. Immunol. 2009, 28, 335–366. [Google Scholar] [CrossRef] [PubMed]
  143. Spiekermann, K.; Roesler, J.; Emmendoerffer, A.; Elsner, J.; Welte, K. Functional features of neutrophils induced by G-CSF and GM-CSF treatment: Differential effects and clinical implications. Leukemia 1997, 11, 466–478. [Google Scholar] [CrossRef]
  144. Link, H. Current state and future opportunities in granulocyte colony-stimulating factor (G-CSF). Support. Care Cancer 2022, 30, 7067–7077. [Google Scholar] [CrossRef] [PubMed]
  145. Theyab, A.; Alsharif, K.F.; Alzahrani, K.J.; Oyouni, A.A.A.; Hawsawi, Y.M.; Algahtani, M.; Alghamdi, S.; Alshammary, A.F. New insight into strategies used to develop long-acting G-CSF biologics for neutropenia therapy. Front. Oncol. 2023, 12, 1026377. [Google Scholar] [CrossRef] [PubMed]
  146. Nikravesh, F.Y.; Shirkhani, S.; Bayat, E.; Talebkhan, Y.; Mirabzadeh, E.; Sabzalinejad, M.; Aliabadi, H.A.M.; Nematollahi, L.; Ardakani, Y.H.; Sardari, S. Extension of human GCSF serum half-life by the fusion of albumin binding domain. Sci. Rep. 2022, 12, 667. [Google Scholar] [CrossRef] [PubMed]
  147. Kumar, R.; Wassif, W.S. Adrenal insufficiency. J. Clin. Pathol. 2022, 75, 435–442. [Google Scholar] [CrossRef] [PubMed]
  148. Barthel, A.; Benker, G.; Berens, K.; Diederich, S.; Manfras, B.; Gruber, M.; Kanczkowski, W.; Kline, G.; Kamvissi-Lorenz, V.; Hahner, S.; et al. An Update on Addison’s Disease. Exp. Clin. Endocrinol. Diabetes 2019, 127, 165–175. [Google Scholar] [CrossRef] [PubMed]
  149. Draghia-Akli, R.; Ellis, K.M.; Hill, L.A.; Malone, P.B.; Fiorotto, M.L. High-efficiency growth hormone-releasing hormone plasmid vector administration into skeletal muscle mediated by electroporation in pigs. FASEB J. 2003, 3, 526–528. [Google Scholar] [CrossRef] [PubMed]
  150. Cuypers, M.-L.; Geukens, N.; Hollevoet, K.; Declerck, P.; Dewilde, M. Exploring the Fate of Antibody-Encoding pDNA after Intramuscular Electroporation in Mice. Pharmaceutics 2023, 15, 1160. [Google Scholar] [CrossRef] [PubMed]
  151. Kim, H.; Danishmalik, S.N.; Hwang, H.; Sin, J.-I.; Oh, J.; Cho, Y.; Lee, H.; Jeong, M.; Kim, S.-H.; Hong, H.J. Gene therapy using plasmid DNA-encoded anti-HER2 antibody for cancers that overexpress HER2. Cancer Gene Ther. 2016, 23, 341–347. [Google Scholar] [CrossRef] [PubMed]
  152. Parzych, E.M.; Du, J.; Ali, A.R.; Schultheis, K.; Frase, D.; Smith, T.R.F.; Cui, J.; Chokkalingam, N.; Tursi, N.J.; Andrade, V.M.; et al. DNA-delivered antibody cocktail exhibits improved pharmacokinetics and confers prophylactic protection against SARS-CoV-2. Nat. Commun. 2022, 13, 5886. [Google Scholar] [CrossRef] [PubMed]
  153. Simon, V.; Ho, D.D.; Abdool Karim, Q. HIV/AIDS epidemiology, pathogenesis, prevention, and treatment. Lancet 2006, 368, 489–504. [Google Scholar] [CrossRef]
  154. Becerra, J.C.; Bildstein, L.S.; Gach, J.S. Recent Insights into the HIV/AIDS Pandemic. Microb. Cell 2016, 3, 451–475. [Google Scholar] [CrossRef] [PubMed]
  155. Kemnic, T.R.; Gulick, P.G. HIV Antiretroviral Therapy. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  156. Gandhi, R.T.; Bedimo, R.; Hoy, J.F.; Landovitz, R.J.; Smith, D.M.; Eaton, E.F.; Lehmann, C.; Springer, S.A.; Sax, P.E.; Thompson, M.A.; et al. Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2022 Recommendations of the International Antiviral Society–USA Panel. JAMA 2023, 329, 63–84. [Google Scholar] [CrossRef] [PubMed]
  157. Smith, G.H.R.; Henry, W.K.; Podzamczer, D.; Masiá, M.D.M.; Bettacchi, C.J.; Arasteh, K.; Jaeger, H.; Khuong-Josses, M.-A.; Montes-Ramírez, M.L.; Stellbrink, H.-J.; et al. Efficacy, Safety, and Durability of Long-Acting Cabotegravir and Rilpivirine in Adults with Human Immunodeficiency Virus Type 1 Infection: 5-Year Results from the LATTE-2 Study. Open Forum Infect. Dis. 2021, 8, ofab439. [Google Scholar] [CrossRef] [PubMed]
  158. Wise, M.C.; Xu, Z.; Tello-Ruiz, E.; Beck, C.; Trautz, A.; Patel, A.; Elliott, S.T.C.; Chokkalingam, N.; Kim, S.; Kerkau, M.G.; et al. In vivo delivery of synthetic DNA–encoded antibodies induces broad HIV-1–neutralizing activity. J. Clin. Investig. 2020, 130, 827–837. [Google Scholar] [CrossRef] [PubMed]
  159. Greb, J.E.; Goldminz, A.M.; Elder, J.T.; Lebwohl, M.G.; Gladman, D.D.; Wu, J.J.; Mehta, N.N.; Finlay, A.Y.; Gottlieb, A.B. Psoriasis. Nat. Rev. Dis. Primers 2016, 2, 16082. [Google Scholar] [CrossRef] [PubMed]
  160. Armstrong, A.W.; Read, C. Pathophysiology, Clinical Presentation, and Treatment of Psoriasis: A Review. JAMA 2020, 323, 1945–1960. [Google Scholar] [CrossRef] [PubMed]
  161. Bellinato, F.; Gisondi, P.; Girolomoni, G. Latest Advances for the Treatment of Chronic Plaque Psoriasis with Biologics and Oral Small Molecules. Biologics 2021, 15, 247–253. [Google Scholar] [CrossRef] [PubMed]
  162. Nauck, M.A.; Quast, D.R.; Wefers, J.; Meier, J.J. GLP-1 receptor agonists in the treatment of type 2 diabetes—State-of-the-art. Mol. Metab. 2021, 46, 101102. [Google Scholar] [CrossRef] [PubMed]
  163. Collins, L.; Costello, R.A. Glucagon-like Peptide-1 Receptor Agonists. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  164. Yao, H.; Zhang, A.; Li, D.; Wu, Y.; Wang, C.-Z.; Wan, J.-Y.; Yuan, C.-S. Comparative effectiveness of GLP-1 receptor agonists on glycaemic control, body weight, and lipid profile for type 2 diabetes: Systematic review and network meta-analysis. BMJ 2024, 384, e076410. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Manufacturing of DNA-based therapies. Large-scale production of AAV-based therapies (black arrows) requires several steps, many of which need optimization, before the product is ready to be administered to the patient. These steps demand a great amount of resources, making the process very expensive, cumbersome, and time consuming. Conversely, producing pDNA (indicated by red dotted lines) on a large scale involves fewer processes, making it faster and more cost-effective. Created with https://BioRender.com (accessed on 13 March 2024).
Figure 1. Manufacturing of DNA-based therapies. Large-scale production of AAV-based therapies (black arrows) requires several steps, many of which need optimization, before the product is ready to be administered to the patient. These steps demand a great amount of resources, making the process very expensive, cumbersome, and time consuming. Conversely, producing pDNA (indicated by red dotted lines) on a large scale involves fewer processes, making it faster and more cost-effective. Created with https://BioRender.com (accessed on 13 March 2024).
Pharmaceutics 16 00535 g001
Figure 2. Benefits of pDNA/EP therapy compared to traditional standard of care. pDNA/EP yields robust and stable expression of DNA-encoded transgenes, allowing for fewer treatments than the current standard of care with drugs/biologics delivered via injection or orally. Continuous dosing regimens require more resources to meet the demands of the therapy. Created with https://BioRender.com (accessed on 13 March 2024).
Figure 2. Benefits of pDNA/EP therapy compared to traditional standard of care. pDNA/EP yields robust and stable expression of DNA-encoded transgenes, allowing for fewer treatments than the current standard of care with drugs/biologics delivered via injection or orally. Continuous dosing regimens require more resources to meet the demands of the therapy. Created with https://BioRender.com (accessed on 13 March 2024).
Pharmaceutics 16 00535 g002
Table 1. Comparison of pDNA/EP and AAV DNA-based therapies. Characteristics described highlight a list of important factors that should be considered when contemplating which therapy is most suitable for a given indication. The number of asterisks indicates the strength of a given characteristic (arbitrary scale).
Table 1. Comparison of pDNA/EP and AAV DNA-based therapies. Characteristics described highlight a list of important factors that should be considered when contemplating which therapy is most suitable for a given indication. The number of asterisks indicates the strength of a given characteristic (arbitrary scale).
FeaturepDNA/EP Gene TherapyAAV Gene Therapy
DurabilityMonths to yearsMonths to many years
Clinical safety of technology****
Redosable*****
Large genetic payload******
OpportunitySystemic activity, Genetic disease, Chronic diseaseSpecific tissue target, Genetic disease, Chronic disease
Large scale manufacturing*****
Freedom from cold chain*****
Table 2. Tissue tropism for 13 natural AAV serotypes in non-human primates and humans.
Table 2. Tissue tropism for 13 natural AAV serotypes in non-human primates and humans.
Natural AAV Serotype Tissue Tropisms
Tissue TypeAAV Serotype
Skeletal MuscleAAV1, AAV2, AAV6, AAV7, AAV8, AAV9, AAV12
Cardio-myocytesAAV1, AAV4, AAV6, AAV7, AAV8, AAV9
Endothelial Vascular Smooth MusclesAAV1, AAV5, AAV7
Inner Ear CellsAAV3
Retinal CellsAAV1, AAV2, AAV4, AAV5, AAV7, AAV8, AAV9, AAV10
CNSAAV1, AAV2, AAV4, AAV5, AAV7, AAV9, AAV10, AAV11
Airway EpitheliaAAV4, AAV5, AAV6, AAV9, AAV10, AAV12
HepatocyesAAV2, AAV3, AAV5, AAV7, AAV8, AAV9, AAV10, AAV11
Salivary GlandsAAV12
Pancreatic CellsAAV8, AAV9, AAV10
Small Intestine CellsAAV10, AAV11
Colon CellsAAV10
Lymph NodesAAV10, AAV11
Leydig CellsAAV9
Adrenal GlandsAAV10, AAV11
Renal TissueAAV2, AAV4, AAV8, AAV9, AAV10, AAV11
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sussman, C.; Liberatore, R.A.; Drozdz, M.M. Delivery of DNA-Based Therapeutics for Treatment of Chronic Diseases. Pharmaceutics 2024, 16, 535. https://doi.org/10.3390/pharmaceutics16040535

AMA Style

Sussman C, Liberatore RA, Drozdz MM. Delivery of DNA-Based Therapeutics for Treatment of Chronic Diseases. Pharmaceutics. 2024; 16(4):535. https://doi.org/10.3390/pharmaceutics16040535

Chicago/Turabian Style

Sussman, Carleigh, Rachel A. Liberatore, and Marek M. Drozdz. 2024. "Delivery of DNA-Based Therapeutics for Treatment of Chronic Diseases" Pharmaceutics 16, no. 4: 535. https://doi.org/10.3390/pharmaceutics16040535

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop