Skip to main content
Molecular Therapy. Nucleic Acids logoLink to Molecular Therapy. Nucleic Acids
. 2023 May 17;32:949–959. doi: 10.1016/j.omtn.2023.05.014

Rationale and strategies for the development of safe and effective optimized AAV vectors for human gene therapy

Arun Srivastava 1,
PMCID: PMC10244667  PMID: 37293185

Abstract

Recombinant adeno-associated virus (AAV) vectors have been, or are currently in use, in 332 phase I/II/III clinical trials in a number of human diseases, and in some cases, remarkable clinical efficacy has also been achieved. There are now three US Food and Drug Administration (FDA)-approved AAV “drugs,” but it has become increasingly clear that the first generation of AAV vectors are not optimal. In addition, relatively large vector doses are needed to achieve clinical efficacy, which has been shown to provoke host immune responses culminating in serious adverse events and, more recently, in the deaths of 10 patients to date. Thus, there is an urgent need for the development of the next generation of AAV vectors that are (1) safe, (2) effective, and (3) human tropic. This review describes the strategies to potentially overcome each of the limitations of the first generation of AAV vectors and the rationale and approaches for the development of the next generation of AAV serotype vectors. These vectors promise to be efficacious at significant reduced doses, likely to achieve clinical efficacy, thereby increasing the safety as well as reducing vector production costs, ensuring translation to the clinic with higher probability of success, without the need for the use of immune suppression, for gene therapy of a wide variety of diseases in humans.

Keywords: MT: Delivery Strategies, AAV vectors, gene expression, immune response, human-tropism, gene therapy

Graphical abstract

graphic file with name fx1.jpg


The first generation of recombinant AAV vectors have taken center stage in human gene therapy, but given serious adverse events and deaths in several patients, especially at high doses, they are not optimal. Thus, development of the next generation of AAV vectors that are safe and effective at lower doses is warranted.

Introduction

Adeno-associated virus (AAV), first discovered in 19651 and considered a “biological oddity,”2 continued to fascinate only a handful of investigators. Following determination of the complete nucleotide sequence,3 molecular cloning4,5 of the wild-type (WT) AAV genome, and the demonstration that the WT AAV possessed the remarkable ability of site-specific integration of the AAV DNA into the long arm of human chromosome 19 (19q13.3),6,7 although it must be noted that site-specific integration was observed in cell cultures in vitro, the first recombinant AAV vectors were subsequently developed by two independent groups.8,9 Further refinements followed,10,11 and interest in this vector system has continued to grow exponentially in the past two decades.12,13,14

The first generation of AAV vectors have been or are currently in use in 331 phase I/II/III clinical trials for a wide variety of human diseases, and in some cases, such as Leber’s congenital amaurosis,15,16,17 lipoprotein lipase deficiency,18 hemophilia B,19,20,21,22,23,24,25,26 aromatic L-amino acid decarboxylase deficiency,27 choroideremia,28 Leber hereditary optic neuropathy,29,30 hemophilia A,31,32,33,34 and spinal muscular atrophy,35 unexpected, remarkable clinical efficacy has also been achieved. Several AAV serotype vectors are now available, which have shown clinical efficacy in a number of human diseases in animal models.36,37 Thus far, three AAV “drugs”—Luxturna, Zolgensma, and Hemgenix—have been approved by the US Food and Drug Administration (FDA).38,39

Despite these remarkable achievements, it has become increasingly clear that the first generation of AAV vectors currently in use are not optimal. For example, despite their efficacy in animal models, these vectors have failed to show clinical efficacy in some cases. In addition, relatively large vector doses are needed to achieve clinical efficacy. The use of high doses has been shown to provoke host immune responses culminating in serious adverse events and, more recently, in the deaths of 10 patients to date. Thus, it has become increasingly clear that there is an urgent need for the development of the next generation of AAV vectors that are (1) safe, (2) effective, and (3) human tropic. Since AAV evolved as a virus, and not as a vector for the purposes of delivery of therapeutic genes, the host immune system cannot distinguish between AAV as a virus versus AAV as a vector. Thus, the use of AAV vectors composed of naturally occurring capsids is likely to induce immune responses, especially at high doses, since the host immune response is directly correlated with the AAV vector dose. Similarly, AAV as a virus does not express its own genes effectively since its single-stranded DNA genome is transcriptionally inactive. Most of the single-stranded AAV (ssAAV) vectors currently in use are also sub-optimal in expressing therapeutic genes. And finally, the tropisms of AAV vectors in animal models do not necessarily translate well in humans, and hence there is a need to identify and further develop human-tropic AAV vectors. This review describes the strategies to overcome each of the limitations of the first generation of AAV vectors and the rationale and approaches for the development of the next generation of AAV serotype vectors. These vectors promise to be efficacious at significant reduced doses, likely to achieve clinical efficacy, thereby increasing the safety as well as reducing vector production costs, ensuring translation to the clinic with higher probability of success, without the need for the use of immune suppression, for gene therapy of a wide variety of human diseases.

Problems associated with the first generation of AAV vectors

Despite their remarkable achievements, it has become increasingly clear that the first generation of AAV vectors that are currently in use in clinical trials in humans are not optimal. For example, in some cases, relatively large vector doses are needed to achieve clinical efficacy.22,26,31,32,34 It has also become increasingly clear that the currently available first generation of AAV vectors being used at high doses have been shown to provoke host immune responses culminating in serious adverse events and, more recently, in the deaths of 10 patients,40,41,42,43,44,45,46 as shown in Table 1.

Table 1.

Reported cases of deaths associated with the use of the first generation of AAV vectors

Sponsor Disease Serotype Vector dose (vg/kg) Number of patients
Audentes Therapeutics XLMTM AAV8 3 × 1014
1 × 1014
3
1
Lysogene MPS-IIIA AAVrh10 7 × 1012 1
Pfizer DMD AAV9 2 × 1014 1
Novartis SMA AAV9 1.1 × 1014 3
Cure Rare Disease DMD AAV9 1x1014 1

XLMTM, X-linked myotubular myopathy; MPSIIIA, mucopolysaccharidosis type IIIA; DMD, Duchenne muscular dystrophy; SMA, spinal muscular atrophy.

Furthermore, the use of the first generation of AAV serotype vectors has also been reported to lead to serious adverse events or has failed to reach the primary clinical endpoints.47,48,49 These details are provided in Table 2. Similarly, the use of shuffled AAV vectors derived from directed evolution has also led to serious adverse events that have been reported in gene therapy trials with AAV vectors derived from directed evolution50,51,52,53 (Table S1).

Table 2.

Adverse events/lack of efficacy reported with the use of the first generation of AAV vectors

Sponsor Disease Serotype Vector dose (vg/kg) Symptoms/outcome
Solid Biosciences DMD AAV9 3 × 1014 thrombocytopenia, renal failure, cardio-pulmonary insufficiency
Pfizer DMD AAV9 2 × 1014 acute kidney injury, atypical hemolytic uremic syndrome-like complement activation, thrombocytopenia
Sarepta Therapeutics DMD AAVrh74 2 × 1014 failed to reach primary clinical endpoint
Amicus Therapeutics BD AAV9 5 × 1013 failed to reach primary clinical endpoint

DMD, Duchenne muscular dystrophy; BD, Batten disease.

Taken together, the following conclusions can be drawn: (1) the use of the first generation of AAV vectors composed of naturally occurring capsids is likely to induce immune responses, especially at high doses, because the host immune system cannot distinguish between AAV as a virus versus AAV as a vector; (2) the host immune response is directly correlated with the AAV vector dose; and (3) because the WT AAV did not evolve for the purposes of delivery of therapeutic genes, recombinant AAV (rAAV) vectors composed of naturally occurring capsid are unlikely to be optimal in human clinical trials.54,55

Thus, it is clear that there is an urgent need for the development of the next generation of AAV vectors that are

  • Capable of high-efficiency transduction at lower doses,

  • Capable of mediating efficient transgene expression,

  • Less immunogenic,

  • Capable of obviating the need for immune suppression,

  • Capable of transducing primary human cells and tissues, and

  • More cost-effective.

Several elegant approaches have been employed by a number of investigators to overcome the limitations of the first generation of AAV vectors. These include the use of directed evolution,56,57,58,59,60 peptide insertions,61,62,63,64,65,66 DNA shuffling,67,68,69,70,71 rational design,72,73,74 ancestral vectors,75,76 chimeric vectors,77,78 dual vectors,79,80,81,82 protease activation,83,84 chemical modifications,85 and machine learning.86,87 A brief account of our strategies to achieve most, if not all, of the objectives outlined above follows.

Development of capsid-modified AAV vectors

Since AAV evolved as a virus, and not as a vector, the naturally occurring AAV and the first generation of rAAV vectors appear identical externally. Thus, the host immune system cannot distinguish between the two and targets both equally well. It could be argued that the two are not identical because the naturally occurring AAV contains the WT AAV DNA, whereas the rAAV vector contains a therapeutic gene. However, the immune system cannot “see” what is inside of the virus or the vector and, again, targets them both just the same. In other words, we want our immune system to work perfectly when it comes to viruses but not when we use them as vectors, which is unrealistic. It should be pointed out, however, that the vector genome has been shown to increase the Toll-like receptor 9-dependent immune response, at least in a murine models,88,89 and that immune response to transgene products can and does occur.90

We nonetheless reasoned that the vector has to be different from the virus. The strategy that led to the development of the next generation (“NextGen”) of AAV vectors was reported in 200891 and is described briefly as follows.

As depicted schematically in Figure 1A, the first generation of AAV2 vectors have 7 tyrosine (Y) residues that are surface exposed. These Y residues are targeted by cellular epidermal growth factor receptor protein tyrosine kinase (EGFR-PTK)92 such that a large fraction of incoming AAV vectors become phosphorylated, which serves as a signal for ubiquitination, followed by proteasome-mediated degradation,93,94,95 which negatively impacts not only on the transduction efficiency, but the broken down peptides also trigger a cytotoxic T cell response.96

Figure 1.

Figure 1

Intracellular fate of the first generation of AAV vectors

(A) Schematic representation of the first generation of AAV2 vectors that contain 7 surface-exposed tyrosine (Y) residues, undergo ① phosphorylation, ubiquitination, and proteasome-mediated degradation in the cytoplasm, and the broken down peptides trigger the MHC class I-mediated CD8+ cytotoxic T cell response. Site-directed mutagenesis ② of each of the Y residues to phenylalanine (F) residues leads to the next generation (“NextGen”) of AAV vectors, and the transduction efficiency of each Y-F mutant vector is increased, ranging between ∼2- and 11-fold. The combination of the three most efficient mutations ③ into one capsid results in the triple-mutant vector, the transduction efficiency of which is up to ∼30-fold higher than the first generation of AAV2 vectors. (B) Schematic representation of the life cycle of the first generation of AAV2 vectors that in addition to 7 Y residues, also contain 15 serine (S), 17 threonine (T), and 10 lysine (K) residues that are surface exposed and undergo phosphorylation and ubiquitination, followed by proteasome-mediated degradation in the cytoplasm. Site-directed mutagenesis each of these residues was also performed, and the transduction efficiencies of the resulting NextGen AAV vectors were documented to be significantly increased.

Each of the seven Y residues in AAV2 capsids were replaced them with phenylalanine (F) residues to generate 7 different Y-F AAV vectors. By changing just one amino acid on the capsid, it was observed that the transduction efficiency of these vectors could be improved ranging from between ∼2- and 11-fold in the mouse liver following tail vein injections.94 When the three most efficient mutations were combined into one capsid, the resulting triple-mutant vector was up to ∼30-fold more efficient than the conventional AAV2 vector in the mouse liver.97 More specifically, the triple-mutant AAV2 vectors were shown to mediate phenotypic correction in a mouse model of hemophilia B.97 Thus, it was concluded that the NextGen AAV2 vectors are more efficient than the conventional first generation of AAV2 vectors. Furthermore, the triple-mutant AAV2 vectors were documented to be less immunogenic since they minimize the cytotoxic T cell response by avoiding phosphorylation, ubiquitination, and proteasome-mediated degradation.91,96,98 The NextGen AAV2 vectors have also been used in a phase I/II clinical trial and have shown clinical efficacy in patients with Leber hereditary optic neuropathy (LHON), with no adverse events.29,30 More specifically, 14 legally blind patients were enrolled in this trial and were followed for up to 24 months. Thirteen of 14 patients showed improved visual acuity, and 1 patient lost vision because of the course of bilateral visual loss, which is characteristic of LHON.

Since 6 of the 7 tyrosine residues are also conserved in all AAV serotypes, mutagenesis of these residues also increases the transduction efficiencies of all AAV serotype vectors evaluated thus far.99 In addition to the 7 surface-exposed Y residues, AAV vectors also contain 15 serine (S), 17 threonine (T), and 10 lysine (K) residues that are surface exposed and can be phosphorylated (Y, S, T) or ubiquitinated (K), which leads to proteasome-mediated degradation, as illustrated in Figure 1B. Each of the 15 S residues,100 17 of the T residues,101 and 10 of the K residues102 were mutagenized. Combination of various permutations and combinations of Y-, S-, T-, and K-mutants led to the identification of the most efficient quadruple mutant (QM) that was observed to be ∼80-fold more efficient in the mouse liver.101

Thus, the NextGen AAV2 vectors overcome the first major limitation of first-generation AAV2 vectors. Since most, if not all, surface-exposed Y, S, T, and K residues are also highly conserved in all AAV serotypes, the transduction of these vectors can also be significantly increased.99

Development of genome-modified AAV vectors

The second major limitation of the first generation of AAV vectors is illustrated in Figure 2. The WT AAV and most of the rAAV vectors contain a ssDNA, which is a problem because there is no host cell RNA polymerase that can transcribe a ssDNA genome.

Figure 2.

Figure 2

Rate-limiting step of viral second-strand DNAsynthesis of the first generation of ssAAV vectors

(A) Schematic model of inhibition of AAV second-strand DNA synthesis by a cellular chaperone protein, FKBP52, phosphorylated forms of which bind to the single-stranded D-sequence at the 3′ end of the AAV-ITR, which is also the “packaging signal” of the AAV genome and is indispensable. (B) The D-sequence at the 5′ end of the AAV-ITR, on the other hand, is dispensable, deletion of which leads to generation X (“GenX”) AAV vectors, transgene expression from which is significantly higher than that from conventional ssAAV vectors.

AAV as a virus does not express its own genes efficiently since viral second-strand DNA synthesis is needed before gene expression can occur. The expectation that AAV as vectors express therapeutic genes to high levels is also unrealistic. Thus, it was reasoned that in the ssAAV vector, the ssDNA genome also needed to be modified to allow viral second-strand DNA synthesis to occur.

This problem has been known since 1997, when it was discovered that a tyrosine-phosphorylated cellular chaperone protein, FKBP52, binds to the single-stranded D-sequence (ssD-sequence) at the 3′ inverted rerminal repeat (ITR) and strongly inhibits AAV second-strand DNA synthesis.103 Deletion of the D-sequence at the 3′ ITR to prevent FKBP52 binding, thus leading to robust second-strand DNA synthesis, resulted in the failure for the AAV genome to undergo packaging, leading to the conclusion that the D-sequence at the 3′ end is the “packaging signal” for AAV104,105 and thus could not be deleted.106,107 However, it was subsequently observed that the deletion of the D-sequence at the 5′ end allowed AAV DNA to undergo successful packaging, which led to the development of the generation X (“GenX”) vectors, shown schematically in Figure 3B, the extent of the transgene expression from which was up to 8-fold higher than that from the conventional ssAAV vectors.108 Thus, GenX vectors overcome the second major limitation of the first generation of ssAAV vectors.

Figure 3.

Figure 3

Strategies for the development of capsid-, genome-, and capsid+genome-modified AAV serotype vectors

(A) Schematic structures of first generation, NextGen, ssAAV, GenX, and optimized (Opt) AAV2 vectors. (B) Schematic structures of Opt AAV2, Opt AAV3, and Opt AAV6 serotype vectors.

A few additional strategies involving the AAV-ITRs deployed by other investigators include the use of ITRs from different AAV serotypes. For example, Engelhardt and colleagues engineered more efficient vectors using AAV2 and AAV5 ITRs at opposite ends of the vector genome.109 Interestingly, the hybrid ITR-containing vectors were also more efficient in intermolecular and intramolecular homologous recombination and trans-splicing compared with those containing the homologous ITRs. In addition, the use of the hybrid-ITR vectors also improved the delivery of transgenes that exceed the AAV packaging capacity.110 Samulski and colleagues described the transcriptional activity of AAV ITRs from several different serotypes and identified three distinct levels of transcriptional activity independent of the promoter function.111 Recently, Duan and colleagues reported the development of CpG-free ITRs and documented that although the vector genome encapsidation was not affected, the vector yield was decreased by ∼3-fold.112

More recently, our own efforts have focused on the development of additional genome-modified AAV vectors, with which it has become feasible to achieve significantly enhanced transgene expression from ssAAV genomes. For example, we observed that the distal 10 nt in the AAV2 D-sequence share partial homology with the glucocorticoid response element (GRE) ½ binding site and that the AAV2 genomes in which the distal 10 nt were replaced with the 15-nt consensus full-length GRE site resulted in generation Y (GenY) AAV vectors that mediate up to ∼6-fold increased transgene expression.113 Similarly, we previously reported that AAV second-strand DNA synthesis is strongly inhibited by phosphorylated forms of a host cell chaperone protein, FKBP52, which binds to the D-sequence at the 3′ end,103 but it has not been possible to delete the D-sequence at the 3′ ITR as it serves as the “packaging signal” for the AAV genome.104,105,106,107 We recently also developed generation Z (GenZ) AAV vectors in which the proximal 10 nt in the D-sequence were replaced with random 10 nt, and one sequence was identified that allowed successful rescue, replication, and packaging of the AAV genome. This sequence was inserted in a rAAV2 genome, replacing the proximal 10 nt in the D-sequence at both ITRs. The extent of the transgene expression from the resulting GenZ ssAAV vectors is up to ∼20-fold higher than that from the WT ssAAV vectors.114 Thus, the GenZ ssAAV vectors not only overcome the problem of viral second-strand DNA synthesis, but they behave more like scAAV vectors but without the size limitation.

Development of optimized AAV vectors

The next obvious question was whether GenX genomes could be packaged into NextGen capsids to achieve even higher efficiency of transduction. As shown schematically in Figure 3A, the packaging of the D-sequence-deleted GenX genomes into NextGen capsids would be expected to lead to optimized (Opt) AAV vectors. Indeed, the development of not only Opt AAV2 vectors but also Opt AAV3 vectors, depicted schematically in Figure 5, was reported, and Opt AAV2 and Opt AAV3 serotype vectors were observed to be ∼20- to 30-fold more efficient than the corresponding NextGen AAV2 and AAV3 vectors, respectively.115 This strategy can and does work with several additional AAV serotype vectors that have been evaluated. Thus, it can be concluded that Opt AAV serotype vectors overcome both major limitations of the first generation of AAV serotype vectors.

Figure 5.

Figure 5

Schematic structures of the 10 most commonly used recombinant AAV serotype vectors

AAV3 and AAV6 vectors have been documented to be selectively tropic for high-efficiency transduction of primary human hepatocytes and primary human hematopoietic stem cells, respectively.

Development of strategies to evade the humoral response to AAV vectors

Beyond the cell-mediated immune response, the humoral response to AAV vectors also remains a major challenge since B cells generate anti-AAV antibodies, making repeat vector dosing difficult, and pre-existing neutralizing anti-AAV antibodies preclude patients from being enrolled in clinical trials. This problem has been addressed by the following three distinct strategies: first, by using anti-CD20 antibodies, transient B cell ablation has been used.116,117 Second, the partial sequence homology between the AAV D-sequence and the major histocompatibility complex (MHC) class II promoter has been exploited to generate AAV vectors that can downregulate the host cell MHC class II promoter function leading to suppression of B cell-helper T cell interaction, resulting in inhibition of differentiation and production of plasma cells that generate antibodies.118 And third, two independent groups have reported a promising approach to address this problem by using immunoglobulin G (IgG) antibody-degrading enzymes, IdeS119 and IdeZ.120 These strategies are depicted and summarized in Figure 4.121 More recently, an additional strategy has been described that utilizes neonatal Fc receptor inhibition to enable AAV gene therapy even in the presence of pre-existing humoral immunity.122

Figure 4.

Figure 4

Strategies for dampening the host humoral immune response to AAV vectors

① Transient depletion of B lymphocytes using CD20 antibody, ② AAV D-sequence-mediated suppression of MHC class II gene expression, and ③ transient degradation of pre-existing IgG antibodies using IgG-cleaving IdeS and IdeZ bacterial endopeptidases. Modified image from.121

Development of human-tropic AAV serotype vectors

Since the tropisms of AAV vectors in animal models do not necessarily translate well in humans, we next wished to identify AAV vectors that specifically and efficiency transduce primary human cell types. In this quest, two human-tropic AAV vectors have been identified that selectively and efficiently transduce primary human cell types. First, of the 10 most commonly used serotypes, AAV3 was observed to be by far the most efficient in transducing primary human hepatocytes,123,124 and second, AAV6 was identified to be the most efficient in transducing primary human hematopoietic stem cells.125,126 This is depicted schematically in Figure 5. Several other independent investigators have corroborated these observations for both AAV3 and AAV6 serotype vectors for primary human hepatocytes127,128,129,130,131 and primary human hematopoietic stem cells,132,133,134,135,136,137,138,139,140,141 respectively. NextGen AAV3 vectors have also been shown to be significantly more efficient in primary hepatocytes in “humanized” mice142,143,144 and NextGen AAV6 vectors more efficient in primary human hematopoietic stem cells.145,146

However, another important consideration is to develop NextGen AAV vectors that are less liver tropic and instead transduce other organs better for non-liver-specific diseases. This would reduce the risk of severe adverse events (SAEs) associated with liver transduction of most AAV serotype vectors upon systemic administration for non-liver-based diseases.40,41,42,43

Concerns regarding the use of high doses of AAV vectors

It has been known for more than four decades that infection by the WT AAV2 in infants and young children leads to no significant clinical sequalae.147 It has also become increasingly clear that the host immune response to AAV vectors directly correlates with the vector dose and that at a dose of 2 × 1011 vg/kg, AAV vectors are only mildly immunogenic.148 Thus, it was of interest to reexamine the total doses of AAV vectors that were, or are currently, being used in a number of clinical trials in humans. These details are provided in Table 3.

Table 3.

Total number of AAV particles/cell in the human body

Sponsor Disease Vector Dose (vg/kg) Total dose (quadrillioin) AAV particles/cell
uniQure HB AAV5 2 × 1013 1.4 ∼47
LogicBio Therapeutics MMA LB-001 5 × 1013 3.5 ∼117
BioMarin HA AAV5 6 × 1013 4.2 ∼140
Pfizer DMD AAV9 2 × 1014 14.0 ∼467
Audentes XLMTM AAV8 3 × 1014 21.0 ∼700

Based on the estimated ∼3 × 1013 (30 trillion) total number of cells in the human body.149 HB, hemophilia B; MMA, methylmalonic acidemia; HA, hemophilia A; DMD, Duchenne muscular dystrophy; XLMTM, X-linked myotubular myopathy.

As can been seen, the estimated total number of cells in a human body is ∼30 trillion.149 Thus, it is quite a testament to the remarkable safety of even the first generation of AAV vectors that humans are easily able to tolerate roughly ∼140× (4.2 quadrillions) of AAV vectors as there are number of cells in the body. However, serious adverse events and deaths have occurred when the vector doses exceed ∼467× (or higher) the number of cells in the body. Thus, again, the use of Opt AAV serotype vectors is likely to be less immunogenic and more effective at significantly lower doses.55

Conclusions

Based on the account presented above, the following conclusions can be drawn.

  • The use of the first generation of AAV serotype vectors composed of naturally occurring capsids is not optimal, given the induction of the host cell-mediated immune response, especially at high doses.

  • The use of the ssAAV genomes with WT ITRs is also not optimal, given the sub-optimal levels of transgene expression.

  • The use of NextGen, GenX, and, preferably, Opt AAV serotype vectors in all future clinical trials should be considered.

Epilogue

AAV, as a virus, did not evolve to be used as a vector for the purposes of delivery of therapeutic genes. Thus, the full potential of AAV as a vector will only be realized after its capsid is modified to evade the host immune response and its genome is modified to express optimal levels of the therapeutic transgene. More, the potential use of Opt AAV vectors at significantly reduced doses promises to achieve clinical efficacy, thereby increasing the safety as well as reducing vector production costs, ensuring translation to the clinic with higher probability of success, without the need for the use of immune-suppression, for gene therapy of human diseases. Specifically, Opt AAV serotype vectors are likely to be less immunogenic and more effective at lower doses, thus further increasing the safety as well as reducing vector production costs, ensuring translation to the clinic with a higher probability of success.150

Acknowledgments

I wish to thank my colleagues for their helpful comments and suggestions. This research was funded in part by Public Health Service grants R01 GM-119186 and R21 AR-081018 from the National Institutes of Health; a 2021 Global Hemophilia ASPIRE grant from Pfizer; and support from the Kitzman Endowment.

I would like to dedicate this article to the memory of Dr. Nicholas Muzyczka (1947-2023), the Father of AAV vectors.

Author contributions

The author conceptualized and wrote this review article.

Declaration of interests

The author is a cofounder of, and holds equity in, Lacerta Therapeutics. He is an inventor on several issued/filed patents on rAAV vectors that have been licensed to various gene therapy companies. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Footnotes

Supplemental information can be found online at https://doi.org/10.1016/j.omtn.2023.05.014.

Supplemental information

Document S1. Table S1
mmc1.pdf (735.9KB, pdf)
Document S2. Article plus supplemental information
mmc2.pdf (3.7MB, pdf)

References

  • 1.Atchison R.W., Casto B.C., Hammon W.M. Adenovirus-associated defective virus particles. Science. 1965;149:754–756. doi: 10.1126/science.149.3685.754. [DOI] [PubMed] [Google Scholar]
  • 2.Berns K.I. My life with adeno-associated virus: a long time spent studying a short genome. DNA Cell Biol. 2013;32:342–347. doi: 10.1089/dna.2013.2120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.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. doi: 10.1128/jvi.45.2.555-564.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Samulski R.J., Berns K.I., Tan M., Muzyczka N. Cloning of adeno-associated virus into pBR322: rescue of intact virus from the recombinant plasmid in human cells. Proc. Natl. Acad. Sci. USA. 1982;79:2077–2081. doi: 10.1073/pnas.79.6.2077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Laughlin C.A., Tratschin J.D., Coon H., Carter B.J. Cloning of infectious adeno-associated virus genomes in bacterial plasmids. Gene. 1983;23:65–73. doi: 10.1016/0378-1119(83)90217-2. [DOI] [PubMed] [Google Scholar]
  • 6.Kotin R.M., Siniscalco M., Samulski R.J., Zhu X.D., Hunter L., Laughlin C.A., McLaughlin S., Muzyczka N., Rocchi M., Berns K.I. Site-specific integration by adeno-associated virus. Proc. Natl. Acad. Sci. USA. 1990;87:2211–2215. doi: 10.1073/pnas.87.6.2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Samulski R.J., Zhu X., Xiao X., Brook J.D., Housman D.E., Epstein N., Hunter L.A. Targeted integration of adeno-associated virus (AAV) into human chromosome 19. EMBO J. 1991;10:3941–3950. doi: 10.1002/j.1460-2075.1991.tb04964.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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. doi: 10.1073/pnas.81.20.6466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tratschin J.D., West M.H., Sandbank T., Carter B.J. A human parvovirus, adeno-associated virus, as a eucaryotic vector: transient expression and encapsidation of the procaryotic gene for chloramphenicol acetyltransferase. Mol. Cell Biol. 1984;4:2072–2081. doi: 10.1128/mcb.4.10.2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Samulski R.J., Chang L.S., Shenk T. A recombinant plasmid from which an infectious adeno-associated virus genome can be excised in vitro and its use to study viral replication. J. Virol. 1987;61:3096–3101. doi: 10.1128/jvi.61.10.3096-3101.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Nahreini P., Woody M.J., Zhou S.Z., Srivastava A. Versatile adeno-associated virus 2-based vectors for constructing recombinant virions. Gene. 1993;124:257–262. doi: 10.1016/0378-1119(93)90402-o. [DOI] [PubMed] [Google Scholar]
  • 12.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. doi: 10.1038/s41573-019-0012-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Li C., Samulski R.J. Engineering adeno-associated virus vectors for gene therapy. Nat. Rev. Genet. 2020;21:255–272. doi: 10.1038/s41576-019-0205-4. [DOI] [PubMed] [Google Scholar]
  • 14.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. doi: 10.1016/j.ymthe.2022.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hauswirth W.W., Aleman T.S., Kaushal S., Cideciyan A.V., Schwartz S.B., Wang L., Conlon T.J., Boye S.L., Flotte T.R., Byrne B.J., et al. Treatment of Leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum. Gene Ther. 2008;19:979–990. doi: 10.1089/hum.2008.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bainbridge J.W.B., Smith A.J., Barker S.S., Robbie S., Henderson R., Balaggan K., Viswanathan A., Holder G.E., Stockman A., Tyler N., et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N. Engl. J. Med. 2008;358:2231–2239. doi: 10.1056/NEJMoa0802268. [DOI] [PubMed] [Google Scholar]
  • 17.Maguire A.M., Simonelli F., Pierce E.A., Pugh E.N., Jr., Mingozzi F., Bennicelli J., Banfi S., Marshall K.A., Testa F., et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N. Engl. J. Med. 2008;358:2240–2248. doi: 10.1056/NEJMoa0802315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Stroes E.S., Nierman M.C., Meulenberg J.J., Franssen R., Twisk J., Henny C.P., Maas M.M., Zwinderman A.H., Ross C., Aronica E., et al. Intramuscular administration of AAV1-lipoprotein lipase S447X lowers triglycerides in lipoprotein lipase-deficient patients. Arterioscler. Thromb. Vasc. Biol. 2008;28:2303–2304. doi: 10.1161/ATVBAHA.108.175620. [DOI] [PubMed] [Google Scholar]
  • 19.Nathwani A.C., Tuddenham E.G.D., Rangarajan S., Rosales C., McIntosh J., Linch D.C., Chowdary P., Riddell A., Pie A.J., Harrington C., et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N. Engl. J. Med. 2011;365:2357–2365. doi: 10.1056/NEJMoa1108046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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. doi: 10.1056/NEJMoa1407309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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. doi: 10.1056/NEJMoa1708538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Miesbach W., Meijer K., Coppens M., Kampmann P., Klamroth R., Schutgens R., Tangelder M., Castaman G., Schwäble J., Bonig H., et al. Gene therapy with adeno-associated virus vector 5-human factor IX in adults with hemophilia B. Blood. 2018;131:1022–1031. doi: 10.1182/blood-2017-09-804419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Konkle B.A., Walsh C.E., Escobar M.A., Josephson N.C., Young G., von Drygalski A., McPhee S.W.J., Samulski R.J., Bilic I., de la Rosa M., et al. BAX 335 hemophilia B gene therapy clinical trial results: potential impact of CpG sequences on gene expression. Blood. 2021;137:763–774. doi: 10.1182/blood.2019004625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chowdary P., Shapiro S., Makris M., Evans G., Boyce S., Talks K., Dolan G., Reiss U., Phillips M., Riddell A., et al. Phase 1-2 trial of AAVS3 gene therapy in patients with Hemophilia B. N. Engl. J. Med. 2022;387:237–247. doi: 10.1056/NEJMoa2119913. [DOI] [PubMed] [Google Scholar]
  • 25.Xue F., Li H., Wu X., Liu W., Zhang F., Tang D., Chen Y., Wang W., Chi Y., Zheng J., et al. Safety and activity of an engineered, liver-tropic adeno-associated virus vector expressing a hyperactive Padua factor IX administered with prophylactic glucocorticoids in patients with haemophilia B: a single-centre, single-arm, phase 1, pilot trial. Lancet. Haematol. 2022;9:e504–e513. doi: 10.1016/S2352-3026(22)00113-2. [DOI] [PubMed] [Google Scholar]
  • 26.Pipe S.W., Leebeek F.W.G., Recht M., Key N.S., Castaman G., Miesbach W., Lattimore S., Peerlinck K., Van der Valk P., et al. Gene therapy with etranacogene dezaparvovec for Hemophilia B. N. Engl. J. Med. 2023;388:706–718. doi: 10.1056/NEJMoa2211644. [DOI] [PubMed] [Google Scholar]
  • 27.Hwu W.L., Muramatsu S., Tseng S.H., Tzen K.Y., Lee N.C., Chien Y.H., Snyder R.O., Byrne B.J., Tai C.H., Wu R.M. Gene therapy for aromatic L-amino acid decarboxylase deficiency. Sci. Transl. Med. 2012;4:134ra161. doi: 10.1126/scitranslmed.3003640. [DOI] [PubMed] [Google Scholar]
  • 28.MacLaren R.E., Groppe M., Barnard A.R., Cottriall C.L., Tolmachova T., Seymour L., Clark K.R., During M.J., Cremers F.P.M., Black G.C.M., et al. Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial. Lancet. 2014;383:1129–1137. doi: 10.1016/S0140-6736(13)62117-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Feuer W.J., Schiffman J.C., Davis J.L., Porciatti V., Gonzalez P., Koilkonda R.D., Yuan H., Lalwani A., Lam B.L., Guy J. Gene therapy for leber hereditary optic neuropathy: initial results. Ophthalmology. 2016;123:558–570. doi: 10.1016/j.ophtha.2015.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Guy J., Feuer W.J., Davis J.L., Porciatti V., Gonzalez P.J., Koilkonda R.D., Yuan H., Hauswirth W.W., Lam B.L. Gene therapy for leber hereditary optic neuropathy: low- and medium-dose visual results. Ophthalmology. 2017;124:1621–1634. doi: 10.1016/j.ophtha.2017.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rangarajan S., Walsh L., Lester W., Perry D., Madan B., Laffan M., Yu H., Vettermann C., Pierce G.F., Wong W.Y., et al. AAV5-Factor VIII gene transfer in severe Hemophilia A. N. Engl. J. Med. 2017;377:2519–2530. doi: 10.1056/NEJMoa1708483. [DOI] [PubMed] [Google Scholar]
  • 32.Pasi K.J., Rangarajan S., Mitchell N., Lester W., Symington E., Madan B., Laffan M., Russell C.B., Li M., Pierce G.F., et al. Multiyear follow-up of AAV5-hFVIII-SQ gene therapy for Hemophilia A. N. Engl. J. Med. 2020;382:29–40. doi: 10.1056/NEJMoa1908490. [DOI] [PubMed] [Google Scholar]
  • 33.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. doi: 10.1056/NEJMoa2104205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mahlangu J., Kaczmarek R., von Drygalski A., Shapiro S., Chou S.-C., Ozelo M.C., Kenet G., Peyvandi F., Wang M., Madan B., et al. Two-year outcomes of valoctocogene roxaparvovec therapy for Hemophilia A. N. Engl. J. Med. 2023;388:694–705. doi: 10.1056/NEJMoa2211075. [DOI] [PubMed] [Google Scholar]
  • 35.Mendell J.R., Al-Zaidy S., Shell R., Arnold W.D., Rodino-Klapac L.R., Prior T.W., Lowes L., Alfano L., Berry K., Church K., et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N. Engl. J. Med. 2017;377:1713–1722. doi: 10.1056/NEJMoa1706198. [DOI] [PubMed] [Google Scholar]
  • 36.Gao G.P., Alvira M.R., Wang L., Calcedo R., Johnston J., Wilson J.M. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc. Natl. Acad. Sci. USA. 2002;99:11854–11859. doi: 10.1073/pnas.182412299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gao G., Vandenberghe L.H., Alvira M.R., Lu Y., Calcedo R., Zhou X., Wilson J.M. Clades of Adeno-associated viruses are widely disseminated in human tissues. J. Virol. 2004;78:6381–6388. doi: 10.1128/JVI.78.12.6381-6388.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Keeler A.M., Flotte T.R. Recombinant adeno-associated virus gene therapy in light of Luxturna (and Zolgensma and glybera): where are we, and how did we get here? Ann Rev Virol. 2019;6:601–621. doi: 10.1146/annurev-virology-092818-015530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.https://www.fda.gov/news-events/press-announcements/fda-approves-first-gene-therapy-treat-adults-hemophilia-b
  • 40.Wilson J.M., Flotte T.R. 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. [DOI] [PubMed] [Google Scholar]
  • 41.Shieh P.B., Bönnemann C.G., Müller-Felber W., Blaschek A., Dowling J.J., Kuntz N.L., Seferian A.M. Letter to the editor. Hum. Gene Ther. 2020;31:787. doi: 10.1089/hum.2020.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.https://lysogene.com/lysogene-provides-update-on-the-aavance-clinical-trial-evaluating-lys-saf302-in-patients-with-mps-iiia/
  • 43.Philippidis A. After patient death, FDA places hold on pfizer Duchenne muscular dystrophy gene therapy trial. Hum. Gene Ther. 2022;33:111–115. doi: 10.1089/hum.2022.29211.bfs. [DOI] [PubMed] [Google Scholar]
  • 44.Guillou J., de Pellegars A., Porcheret F., Frémeaux-Bacchi V., Allain-Launay E., Debord C., Denis M., Péréon Y., Barnérias C., Desguerre I., et al. Fatal thrombotic microangiopathy case following adeno-associated viral SMN gene therapy. Blood Adv. 2022;6:4266–4270. doi: 10.1182/bloodadvances.2021006419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.https://www.fiercepharma.com/pharma/two-deaths-after-novartis-zolgensma-bring-gene-therapys-liver-safety-spotlight-again
  • 46.https://www.genengnews.com/topics/genome-editing/gene-therapy/gene-therapy-briefs-cure-rare-disease-ceos-brother-died-in-clinical-trial/
  • 47.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. doi: 10.1016/j.ymthe.2020.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mullard A. Sarepta's DMD gene therapy falls flat. Nat. Rev. Drug Discov. 2021;20:91. doi: 10.1038/d41573-021-00010-0. [DOI] [PubMed] [Google Scholar]
  • 49.https://www.fiercebiotech.com/biotech/jpm-2022-amicus-axes-gene-therapy-program-for-type-batten-disease-advances-another
  • 50.https://www.globenewswire.com/news-release/2021/07/22/2267699/32452/en/Adverum-Provides-Update-on-ADVM-022-and-the-INFINITY-Trial-in-Patients-with-Diabetic-Macular-Edema.html
  • 51.https://sparktx.com/press_releases/spark-therapeutics-spk-8011-suggests-stable-and-durable-factor-viii-expression-in-largest-phase-1-2-gene-therapy-study-in-hemophilia-a-to-date/
  • 52.Philippidis A. Gene therapy briefs – FDA places clinical hold on 4D Molecular Therapeutics Fabry Program. Hum. Gene Ther. 2023;34:177–179. [Google Scholar]
  • 53.https://globalgenes.org/2022/02/02/fda-places-hold-on-logicbios-trial-of-lb-001-for-the-treatment-of-pediatric-patients-with-mma/
  • 54.Srivastava A. Adeno-associated virus: the naturally occurring virus versus the recombinant vector. Hum. Gene Ther. 2016;27:1–6. doi: 10.1089/hum.2015.29017.asr. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Srivastava A. AAV vectors: are they safe? Hum. Gene Ther. 2020;31:697–699. doi: 10.1089/hum.2020.187. [DOI] [PubMed] [Google Scholar]
  • 56.Maheshri N., Koerber J.T., Kaspar B.K., Schaffer D.V. Directed evolution of adeno-associated virus yields enhanced gene delivery vectors. Nat. Biotechnol. 2006;24:198–204. doi: 10.1038/nbt1182. [DOI] [PubMed] [Google Scholar]
  • 57.Perabo L., Endell J., King S., Lux K., Hallek M., Büning H. Combinatorial engineering of a gene therapy vector: directed evolution of adeno-associated virus. J. Gene Med. 2006;8:155–162. doi: 10.1002/jgm.849. [DOI] [PubMed] [Google Scholar]
  • 58.Grimm D., Lee J.S., Wang L., Desai T., Akache B., Storm T.A., Kay M.A. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 2008;82:5887–5911. doi: 10.1128/JVI.00254-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li W., Zhang L., Johnson J.S., Zhijian W., Grieger J.C., Ping-Jie X., Drouin L.M., Agbandje-McKenna M., Pickles R.J., Samulski R.J. Generation of novel AAV variants by directed evolution for improved CFTR delivery to human ciliated airway epithelium. Mol. Ther. 2009;17:2067–2077. doi: 10.1038/mt.2009.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Maguire C.A., Gianni D., Meijer D.H., Shaket L.A., Wakimoto H., Rabkin S.D., Gao G., Sena-Esteves M. Directed evolution of adeno-associated virus for glioma cell transduction. J. Neurooncol. 2010;96:337–347. doi: 10.1007/s11060-009-9972-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Perabo L., Büning H., Kofler D.M., Ried M.U., Girod A., Wendtner C.M., Enssle J., Hallek M. In vitro selection of viral vectors with modified tropism: the adeno-associated virus display. Mol. Ther. 2003;8:151–157. doi: 10.1016/s1525-0016(03)00123-0. [DOI] [PubMed] [Google Scholar]
  • 62.Müller O.J., Kaul F., Weitzman M.D., Pasqualini R., Arap W., Kleinschmidt J.A., Trepel M. Random peptide libraries displayed on adeno-associated virus to select for targeted gene therapy vectors. Nat. Biotechnol. 2003;21:1040–1046. doi: 10.1038/nbt856. [DOI] [PubMed] [Google Scholar]
  • 63.Varadi K., Michelfelder S., Korff T., Hecker M., Trepel M., Katus H.A., Kleinschmidt J.A., Müller O.J. Novel random peptide libraries displayed on AAV serotype 9 for selection of endothelial cell-directed gene transfer vectors. Gene Ther. 2012;19:800–809. doi: 10.1038/gt.2011.143. [DOI] [PubMed] [Google Scholar]
  • 64.Yu C.Y., Yuan Z., Cao Z., Wang B., Qiao C., Li J., Xiao X. A muscle-targeting peptide displayed on AAV2 improves muscle tropism on systemic delivery. Gene Ther. 2009;16:953–962. doi: 10.1038/gt.2009.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Weinmann J., Weis S., Sippel J., Tulalamba W., Remes A., El Andari J., Herrmann A.K., Pham Q.H., Borowski C., Hille S., et al. Identification of a myotropic AAV by massively parallel in vivo evaluation of barcoded capsid variants. Nat. Commun. 2020;11:5432. doi: 10.1038/s41467-020-19230-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tabebordbar M., Lagerborg K.A., Stanton A., King E.M., Ye S., Tellez L., Krunnfusz A., Tavakoli S., Widrick J.J., Messemer K.A., et al. Directed evolution of a family of AAV capsid variants enabling potent muscle-directed gene delivery across species. Cell. 2021;184:4919–4938.e22. doi: 10.1016/j.cell.2021.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Li W., Asokan A., Wu Z., Van Dyke T., DiPrimio N., Johnson J.S., Govindaswamy L., Agbandje-McKenna M., Leichtle S., Eugene Redmond D., Jr., et al. Engineering and selection of shuffled AAV genomes: a new strategy for producing targeted biological nanoparticles. Mol. Ther. 2008;16:1252–1260. doi: 10.1038/mt.2008.100. [DOI] [PubMed] [Google Scholar]
  • 68.Koerber J.T., Jang J.H., Schaffer D.V. DNA shuffling of adeno-associated virus yields functionally diverse viral progeny. Mol. Ther. 2008;16:1703–1709. doi: 10.1038/mt.2008.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yang L., Jiang J., Drouin L.M., Agbandje-McKenna M., Chen C., Qiao C., Pu D., Hu X., Wang D.Z., et al. A myocardium tropic adeno-associated virus (AAV) evolved by DNA shuffling and in vivo selection. Proc. Natl. Acad. Sci. USA. 2009;106:3946–3951. doi: 10.1073/pnas.0813207106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lisowski L., Dane A.P., Chu K., Zhang Y., Cunningham S.C., Wilson E.M., Nygaard S., Grompe M., Alexander I.E., Kay M.A. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature. 2014;506:382–386. doi: 10.1038/nature12875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Paulk N.K., Pekrun K., Zhu E., Nygaard S., Li B., Xu J., Chu K., Leborgne C., Dane A.P., Haft A., et al. Bioengineered AAV Capsids with combined high human liver transduction in vivo and unique humoral seroreactivity. Mol. Ther. 2018;26:289–303. doi: 10.1016/j.ymthe.2017.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lochrie M.A., Tatsuno G.P., Christie B., McDonnell J.W., Zhou S., Surosky R., Pierce G.F., Colosi P. Mutations on the external surfaces of adeno-associated virus type 2 capsids that affect transduction and neutralization. J. Virol. 2006;80:821–834. doi: 10.1128/JVI.80.2.821-834.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Gabriel N., Hareendran S., Sen D., Gadkari R.A., Sudha G., Selot R., Hussain M., Dhaksnamoorthy R., Samuel R., Srinivasan N., et al. Bioengineering of AAV2 capsid at specific serine, threonine, or lysine residues improves its transduction efficiency in vitro and in vivo. Hum. Gene Ther. Methods. 2013;24:80–93. doi: 10.1089/hgtb.2012.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sen D., Gadkari R.A., Sudha G., Gabriel N., Kumar Y.S., Selot R., Samuel R., Rajalingam S., Ramya V., Nair S.C., et al. Targeted modifications in adeno-associated virus serotype 8 capsid improves its hepatic gene transfer efficiency in vivo. Hum. Gene Ther. Methods. 2013;24:104–116. doi: 10.1089/hgtb.2012.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zinn E., Pacouret S., Khaychuk V., Turunen H.T., Carvalho L.S., Andres-Mateos E., Shah S., Shelke R., Maurer A.C., Plovie E., et al. In silico reconstruction of the viral evolutionary lineage yields a potent gene therapy vector. Cell Rep. 2015;12:1056–1068. doi: 10.1016/j.celrep.2015.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Santiago-Ortiz J., Ojala D.S., Westesson O., Weinstein J.R., Wong S.Y., Steinsapir A., Kumar S., Holmes I., Schaffer D.V. AAV ancestral reconstruction library enables selection of broadly infectious viral variants. Gene Ther. 2015;22:934–946. doi: 10.1038/gt.2015.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hauck B., Chen L., Xiao W. Generation and characterization of chimeric recombinant AAV vectors. Mol. Ther. 2003;7:419–425. doi: 10.1016/s1525-0016(03)00012-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Bowles D.E., Rabinowitz J.E., Samulski R.J. Marker rescue of adeno-associated virus (AAV) capsid mutants: a novel approach for chimeric AAV production. J. Virol. 2003;77:423–432. doi: 10.1128/JVI.77.1.423-432.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Duan D., Yue Y., Yan Z., Engelhardt J.F. A new dual-vector approach to enhance recombinant adeno-associated virus-mediated gene expression through intermolecular cis activation. Nat. Med. 2000;6:595–598. doi: 10.1038/75080. [DOI] [PubMed] [Google Scholar]
  • 80.Nakai H., Storm T.A., Kay M.A. Increasing the size of rAAV-mediated expression cassettes in vivo by intermolecular joining of two complementary vectors. Nat. Biotechnol. 2000;18:527–532. doi: 10.1038/75390. [DOI] [PubMed] [Google Scholar]
  • 81.Sun L., Li J., Xiao X. Overcoming adeno-associated virus vector size limitation through viral DNA heterodimerization. Nat. Med. 2000;6:599–602. doi: 10.1038/75087. [DOI] [PubMed] [Google Scholar]
  • 82.Maina N., Zhong L., Li X., Zhao W., Han Z., Bischof D., Aslanidi G., Zolotukhin S., Weigel-Van Aken K.A., Rivers A.E., et al. Optimization of recombinant adeno-associated viral vectors for human beta-globin gene transfer and transgene expression. Hum. Gene Ther. 2008;19:365–375. doi: 10.1089/hum.2007.173. [DOI] [PubMed] [Google Scholar]
  • 83.Guenther C.M., Brun M.J., Bennett A.D., Ho M.L., Chen W., Zhu B., Lam M., Yamagami M., Kwon S., Bhattacharya N., et al. Protease-activatable adeno-associated virus vector for gene delivery to damaged heart tissue. Mol. Ther. 2019;27:611–622. doi: 10.1016/j.ymthe.2019.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Tong J.G., Evans A.C., Ho M.L., Guenther C.M., Brun M.J., Judd J., Wu E., Suh J. Reducing off target viral delivery in ovarian cancer gene therapy using a protease-activated AAV2 vector platform. Mol. Ther. 2019;307:292–301. doi: 10.1016/j.jconrel.2019.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kelemen R.E., Mukherjee R., Cao X., Erickson S.B., Zheng Y., Chatterjee A. A precise chemical strategy to alter the receptor specificity of the adeno-associated virus. Angew. Chem. Int. Ed. Engl. 2016;55:10645–10649. doi: 10.1002/anie.201604067. [DOI] [PubMed] [Google Scholar]
  • 86.Ogden P.J., Kelsic E.D., Sinai S., Church G.M. Comprehensive AAV capsid fitness landscape reveals a viral gene and enables machine-guided design. Science. 2019;366:1139–1143. doi: 10.1126/science.aaw2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bryant D.H., Bashir A., Sinai S., Jain N.K., Ogden P.J., Riley P.F., Church G.M., Colwell L.J., Kelsic E.D. Deep diversification of an AAV capsid protein by machine learning. Nat. Biotechnol. 2021;39:691–696. doi: 10.1038/s41587-020-00793-4. [DOI] [PubMed] [Google Scholar]
  • 88.Martino A.T., Suzuki M., Markusic D.M., Zolotukhin I., Ryals R.C., Moghimi B., Ertl H.C.J., Muruve D.A., Lee B., Herzog R.W. The genome of self-complementary adeno-associated viral vectors increases Toll-like receptor 9-dependent innate immune responses in the liver. Blood. 2011;117:6459–6468. doi: 10.1182/blood-2010-10-314518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Herzog R.W., Cooper M., Perrin G.Q., Biswas M., Martino A.T., Morel L., Terhorst C., Hoffman B.E. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Arjomandnejad M., Dasgupta I., Flotte T.R., Keeler A.M. Immunogenicity of recombinant adeno-associated virus (AAV) vectors for gene transfer. BioDrugs. 2023;2:1–19. doi: 10.1007/s40259-023-00585-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhong L., Li B., Mah C.S., Govindasamy L., Agbandje-McKenna M., Cooper M., Herzog R.W., Zolotukhin I., Warrington K.H., Jr., Weigel-Van Aken K.A., et al. Next generation of adeno-associated virus 2 vectors: point mutations in tyrosines lead to high-efficiency transduction at lower doses. Proc. Natl. Acad. Sci. USA. 2008;105:7827–7832. doi: 10.1073/pnas.0802866105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Mah C., Qing K., Khuntirat B., Ponnazhagan S., Wang X.S., Kube D.M., Yoder M.C., Srivastava A. Adeno-associated virus type 2-mediated gene transfer: role of epidermal growth factor receptor protein tyrosine kinase in transgene expression. J. Virol. 1998;72:9835–9843. doi: 10.1128/jvi.72.12.9835-9843.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Duan D., Yue Y., Yan Z., Yang J., Engelhardt J.F. Endosomal processing limits gene transfer to polarized airway epithelia by adeno-associated virus. J. Clin. Invest. 2000;105:1573–1587. doi: 10.1172/JCI8317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhong L., Zhao W., Wu J., Li B., Zolotukhin S., Govindasamy L., Agbandje-McKenna M., Srivastava A. A dual role of EGFR protein tyrosine kinase signaling in ubiquitination of AAV2 capsids and viral second-strand DNA synthesis. Mol. Ther. 2007;15:1323–1330. doi: 10.1038/sj.mt.6300170. [DOI] [PubMed] [Google Scholar]
  • 95.Zhong L., Li B., Jayandharan G., Mah C.S., Govindasamy L., Agbandje-McKenna M., Herzog R.W., Weigel-Van Aken K.A., Hobbs J.A., et al. Tyrosine-phosphorylation of AAV2 vectors and its consequences on viral intracellular trafficking and transgene expression. Virology. 2008;381:194–202. doi: 10.1016/j.virol.2008.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mingozzi F., Maus M.V., Hui D.J., Sabatino D.E., Murphy S.L., Rasko J.E.J., Ragni M.V., Manno C.S., Sommer J., Jiang H., et al. CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat. Med. 2007;13:419–422. doi: 10.1038/nm1549. [DOI] [PubMed] [Google Scholar]
  • 97.Markusic D.M., Herzog R.W., Aslanidi G.V., Hoffman B.E., Li B., Li M., Jayandharan G.R., Ling C., Zolotukhin I., Ma W., et al. High-efficiency transduction and correction of murine hemophilia B using AAV2 vectors devoid of multiple surface-exposed tyrosines. Mol. Ther. 2010;18:2048–2056. doi: 10.1038/mt.2010.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Martino A.T., Basner-Tschakarjan E., Markusic D.M., Finn J.D., Hinderer C., Zhou S., Ostrov D.A., Srivastava A., Ertl H.C.J., Terhorst C., et al. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells. Blood. 2013;121:2224–2233. doi: 10.1182/blood-2012-10-460733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Berns K.I., Srivastava A. Next generation of adeno-associated virus vectors for gene therapy for human liver diseases. Gastroenterol. Clin. North Am. 2019;48:319–330. doi: 10.1016/j.gtc.2019.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Aslanidi G.V., Rivers A.E., Ortiz L., Govindasamy L., Ling C., Jayandharan G.R., Zolotukhin S., Agbandje-McKenna M., Srivastava A. High-efficiency transduction of human monocyte-derived dendritic cells by capsid-modified recombinant AAV2 vectors. Vaccine. 2012;30:3908–3917. doi: 10.1016/j.vaccine.2012.03.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Aslanidi G.V., Rivers A.E., Ortiz L., Song L., Ling C., Govindasamy L., Van Vliet K., Tan M., Agbandje-McKenna M., Srivastava A. Optimization of the capsid of recombinant adeno-associated virus 2 (AAV2) vectors: the final threshold? PLoS One. 2013;8:e59142. doi: 10.1371/journal.pone.0059142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Li B., Ma W., Ling C., Van Vliet K., Huang L.Y., Agbandje-McKenna M., Srivastava A., Aslanidi G.V. Site-directed mutagenesis of surface-exposed lysine residues leads to improved transduction by AAV2, but not AAV8, vectors in murine hepatocytes in vivo. Hum. Gene Ther. Methods. 2015;26:211–220. doi: 10.1089/hgtb.2015.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Qing K., Wang X.S., Kube D.M., Ponnazhagan S., Bajpai A., Srivastava A. Role of tyrosine phosphorylation of a cellular protein in adeno-associated virus 2-mediated transgene expression. Proc. Natl. Acad. Sci. USA. 1997;94:10879–10884. doi: 10.1073/pnas.94.20.10879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang X.S., Ponnazhagan S., Srivastava A. Rescue and replication signals of the adeno-associated virus 2 genome. J. Mol. Biol. 1995;250:573–580. doi: 10.1006/jmbi.1995.0398. [DOI] [PubMed] [Google Scholar]
  • 105.Wang X.S., Ponnazhagan S., Srivastava A. Rescue and replication of adeno-associated virus type 2 as well as vector DNA sequences from recombinant plasmids containing deletions in the viral inverted terminal repeats: selective encapsidation of viral genomes in progeny virions. J. Virol. 1996;70:1668–1677. doi: 10.1128/jvi.70.3.1668-1677.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wang X.S., Qing K., Ponnazhagan S., Srivastava A. Adeno-associated virus type 2 DNA replication in vivo: mutation analyses of the D sequence in viral inverted terminal repeats. J. Virol. 1997;71:3077–3082. doi: 10.1128/jvi.71.4.3077-3082.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ling C., Wang Y., Lu Y., Wang L., Jayandharan G.R., Aslanidi G.V., Li B., Cheng B., Ma W., Lentz T., et al. The adeno-associated virus genome packaging puzzle. J. Mol. Genet. Med. 2015;9:175. doi: 10.4172/1747-0862.1000175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ling C., Wang Y., Lu Y., Wang L., Jayandharan G.R., Aslanidi G.V., Li B., Cheng B., Ma W., Lentz T., et al. Enhanced transgene expression from recombinant single-stranded D-sequence-substituted adeno-associated virus vectors in human cell lines in vitro and in murine hepatocytes in vivo. J. Virol. 2015;89:952–961. doi: 10.1128/JVI.02581-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Yan Z., Zak R., Zhang Y., Engelhardt J.F. Inverted terminal repeat sequences are important for intermolecular recombination and circularization of adeno-associated virus genomes. J. Virol. 2005;79:364–379. doi: 10.1128/JVI.79.1.364-379.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Yan Z., Lei-Butters D.C.M., Zhang Y., Zak R., Engelhardt J.F. Hybrid adeno-associated virus bearing nonhomologous inverted terminal repeats enhances dual-vector reconstruction of minigenes in vivo. Hum. Gene Ther. 2007;18:81–87. doi: 10.1089/hum.2006.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Earley L.F., Conatser L.M., Lue V.M., Dobbins A.L., Li C., Hirsch M.L., Samulski R.J. Adeno-associated virus serotype-specific inverted terminal repeat sequence role in vector transgene expression. Hum. Gene Ther. 2020;31:151–162. doi: 10.1089/hum.2019.274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Pan X., Yue Y., Boftsi M., Wasala L.P., Tran N.T., Zhang K., Pintel D.J., Tai P.W.L., Duan D. Rational engineering of a functional CpG-free ITR for AAV gene therapy. Gene Ther. 2022;29:333–345. doi: 10.1038/s41434-021-00296-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Shoti K., Qing K., Keeler G.D., Byrne B.J., Srivastava A. Development of genome-modified generation Y (GenY) AAVrh74 vectors with improved transgene expression in a mouse skeletal muscle cell line and in primary human skeletal muscle cells. Mol. Ther. 2022;30:237. [Google Scholar]
  • 114.Qing K., Shoti J., Nath A., Keeler G.D., Srivastava A. Development of genome-modified generation Z (GenZ) single-stranded AAV vectors with improved transgene expression in human cells in vitro and in mouse liver in vivo. Mol. Ther. 2023;31:234–235. [Google Scholar]
  • 115.Ling C., Li B., Ma W., Srivastava A. Development of optimized AAV serotype vectors for high-efficiency transduction at further reduced doses. Hum. Gene Ther. Methods. 2016;27:143–149. doi: 10.1089/hgtb.2016.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Corti M., Elder M., Falk D., Lawson L., Smith B., Nayak S., Conlon T., Clément N., Erger K., Lavassani E., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Corti M., Cleaver B., Clément N., Conlon T.J., Faris K.J., Wang G., Benson J., Tarantal A.F., Fuller D., Herzog R.W., et al. Evaluation of readministration of a recombinant adeno-associated virus vector expressing acid alpha-glucosidase in pompe disease: preclinical to clinical planning. Hum. Gene Ther. Clin. Dev. 2015;26:185–193. doi: 10.1089/humc.2015.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kwon H.J., Qing K., Ponnazhagan S., Wang X.S., Markusic D.M., Gupte S., Boye S.E., Srivastava A. Adeno-associated virus D-sequence-mediated suppression of expression of a human major histocompatibility class II gene: implications in the development of adeno-associated virus vectors for modulating humoral immune response. Hum. Gene Ther. 2020;31:565–574. doi: 10.1089/hum.2020.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Leborgne C., Barbon E., Alexander J.M., Hanby H., Delignat S., Cohen D.M., Collaud F., Muraleetharan S., Lupo D., Silverberg J., et al. IgG-cleaving endopeptidase enables in vivo gene therapy in the presence of anti-AAV neutralizing antibodies. Nat. Med. 2020;26:1096–1101. doi: 10.1038/s41591-020-0911-7. [DOI] [PubMed] [Google Scholar]
  • 120.Elmore Z.C., Oh D.K., Simon K.E., Fanous M.M., Asokan A. Rescuing AAV gene transfer from neutralizing antibodies with an IgG-degrading enzyme. JCI Insight. 2020;5:e139881. doi: 10.1172/jci.insight.139881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.https://www.antibodiesinc.com/blogs/news/how-to-make-a-great-antibody.
  • 122.Horiuchi M., Hinderer C.J., Shankle H.N., Hayashi P.M., Chichester J.A., Kissel C., Bell P., Dyer C., Wilson J.M. Neonatal Fc receptor inhibition enables adeno-associated virus gene therapy despite pre-existing humoral immunity. Hum. Gene Ther. 2023;31 doi: 10.1089/hum.2022.216. [DOI] [PubMed] [Google Scholar]
  • 123.Glushakova L.G., Lisankie M.J., Eruslanov E.B., Ojano-Dirain C., Zolotukhin I., Liu C., Srivastava A., Stacpoole P.W. AAV3-mediated transfer and expression of the pyruvate dehydrogenase E1 alpha subunit gene causes metabolic remodeling and apoptosis of human liver cancer cells. Mol. Genet. Metab. 2009;98:289–299. doi: 10.1016/j.ymgme.2009.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Ling C., Lu Y., Kalsi J.K., Jayandharan G.R., Li B., Ma W., Cheng B., Gee S.W.Y., McGoogan K.E., Govindasamy L., et al. Human hepatocyte growth factor receptor is a cellular coreceptor for adeno-associated virus serotype 3. Hum. Gene Ther. 2010;21:1741–1747. doi: 10.1089/hum.2010.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Song L., Kauss M.A., Kopin E., Chandra M., Ul-Hasan T., Miller E., Jayandharan G.R., Rivers A.E., Aslanidi G.V., Ling C., et al. Optimizing the transduction efficiency of capsid-modified AAV6 serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy. 2013;15:986–998. doi: 10.1016/j.jcyt.2013.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Song L., Li X., Jayandharan G.R., Wang Y., Aslanidi G.V., Ling C., Zhong L., Gao G., Yoder M.C., Ling C., et al. High-efficiency transduction of primary human hematopoietic stem cells and erythroid lineage-restricted expression by optimized AAV6 serotype vectors in vitro and in a murine xenograft model in vivo. PLoS One. 2013;8:e58757. doi: 10.1371/journal.pone.0058757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Handa A., Muramatsu S.I., Qiu J., Mizukami H., Brown K.E. Adeno-associated virus (AAV)-3-based vectors transduce haematopoietic cells not susceptible to transduction with AAV-2-based vectors. J. Gen. Virol. 2000;81:2077–2084. doi: 10.1099/0022-1317-81-8-2077. [DOI] [PubMed] [Google Scholar]
  • 128.Biswas M., Marsic D., Li N., Zou C., Gonzalez-Aseguinolaza G., Zolotukhin I., Kumar S.R.P., Rana J., Butterfield J.S.S., Kondratov O., et al. Engineering and in vitro selection of a novel AAV3B variant with high hepatocyte tropism and reduced seroreactivity. Mol. Ther. Methods Clin. Dev. 2020;19:347–361. doi: 10.1016/j.omtm.2020.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Purohit N., Jain A., Mathews V., Jayandharan G.R. Molecular characterization of novel Adeno-associated virus variants infecting human tissues. Virus Res. 2019;272:197716. doi: 10.1016/j.virusres.2019.197716. [DOI] [PubMed] [Google Scholar]
  • 130.Ito M., Takino N., Nomura T., Kan A., Muramatsu S.I. Engineered adeno-associated virus 3 vector with reduced reactivity to serum antibodies. Sci. Rep. 2021;11:9322. doi: 10.1038/s41598-021-88614-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Yin L., Keeler G.D., Zhang Y., Hoffman B.E., Ling C., Qing K., Srivastava A. AAV3-miRNA vectors for growth suppression of human hepatocellular carcinoma cells in vitro and human liver tumors in a murine xenograft model in vivo. Gene Ther. 2021;28:422–434. doi: 10.1038/s41434-020-0140-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Martin R.M., Ikeda K., Cromer M.K., Uchida N., Nishimura T., Romano R., Tong A.J., Lemgart V.T., Camarena J., Pavel-Dinu M., et al. Highly efficient and marker-free genome editing of human pluripotent stem cells by CRISPR-Cas9 RNP and AAV6 donor-mediated homologous recombination. Cell Stem Cell. 2019;24:821–828.e5. doi: 10.1016/j.stem.2019.04.001. [DOI] [PubMed] [Google Scholar]
  • 133.Houghton B.C., Panchal N., Haas S.A., Chmielewski K.O., Hildenbeutel M., Whittaker T., Mussolino C., Cathomen T., Thrasher A.J., Booth C. Genome editing with TALEN, CRISPR-Cas9 and CRISPR-Cas12a in combination with AAV6 homology donor restores T cell function for XLP. Front. Genome Ed. 2022;4:828489. doi: 10.3389/fgeed.2022.828489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Haltalli M.L.R., Wilkinson A.C., Rodriguez-Fraticelli A., Porteus M. Hematopoietic stem cell gene editing and expansion: state-of-the-art technologies and recent applications. Exp. Hematol. 2022;107:9–13. doi: 10.1016/j.exphem.2021.12.399. [DOI] [PubMed] [Google Scholar]
  • 135.Cromer M.K., Vaidyanathan S., Ryan D.E., Curry B., Lucas A.B., Camarena J., Kaushik M., Hay S.R., Martin R.M., Steinfeld I., et al. Global transcriptional response to CRISPR/Cas9-AAV6-Based genome editing in CD34+ hematopoietic stem and progenitor cells. Mol. Ther. 2018;26:2431–2442. doi: 10.1016/j.ymthe.2018.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wang J., Exline C.M., DeClercq J.J., Llewellyn G.N., Hayward S.B., Li P.W.L., Shivak D.A., Surosky R.T., Gregory P.D., Holmes M.C., et al. Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat. Biotechnol. 2015;33:1256–1263. doi: 10.1038/nbt.3408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Yang H., Qing K., Keeler G.D., Yin L., Mietzsch M., Ling C., Hoffman B.E., Agbandje-McKenna M., Tan M., Wang W., et al. Enhanced transduction of human hematopoietic stem cells by AAV6 vectors: implications in gene therapy and genome editing. Mol. Ther. Nucleic Acids. 2020;20:451–458. doi: 10.1016/j.omtn.2020.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Wilkinson A.C., Dever D.P., Baik R., Camarena J., Hsu I., Charlesworth C.T., Morita C., Nakauchi H., Porteus M.H. Cas9-AAV6 gene correction of beta-globin in autologous HSCs improves sickle cell disease erythropoiesis in mice. Nat. Commun. 2021;12:686. doi: 10.1038/s41467-021-20909-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Bak R.O., Dever D.P., Reinisch A., Cruz Hernandez D., Majeti R., Porteus M.H. Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6. Elife. 2017;6:e27873. doi: 10.7554/eLife.27873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Dudek A.M., Porteus M.H. AAV6 is superior to clade F AAVs in stimulating homologous recombination-based genome editing in human HSPCs. Mol. Ther. 2019;27:1701–1705. doi: 10.1016/j.ymthe.2019.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Romero Z., Lomova A., Said S., Miggelbrink A., Kuo C.Y., Campo-Fernandez B., Hoban M.D., Masiuk K.E., Clark D.N., Long J., et al. Editing the sickle cell disease mutation in human hematopoietic stem cells: comparison of endonucleases and homologous donor templates. Mol. Ther. 2019;27:1389–1406. doi: 10.1016/j.ymthe.2019.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Vercauteren K., Hoffman B.E., Zolotukhin I., Keeler G.D., Xiao J.W., Basner-Tschakarjan E., High K.A., Ertl H.C., Rice C.M., Srivastava A., et al. Superior in vivo transduction of human hepatocytes using engineered AAV3 capsid. Mol. Ther. 2016;24:1042–1049. doi: 10.1038/mt.2016.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Brown H.C., Doering C.B., Herzog R.W., Ling C., Markusic D.M., Spencer H.T., Srivastava A., Srivastava A. Development of a clinical candidate AAV3 vector for gene therapy of Hemophilia B. Hum. Gene Ther. 2020;31:1114–1123. doi: 10.1089/hum.2020.099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kumar S.R.P., Xie J., Hu S., Ko J., Huang Q., Brown H.C., Srivastava A., Markusic D.M., Doering C.B., Spencer H.T., et al. Coagulation factor IX gene transfer to non-human primates using engineered AAV3 capsid and hepatic optimized expression cassette. Mol. Ther. Methods Clin. Dev. 2021;23:98–107. doi: 10.1016/j.omtm.2021.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ling C., Bhukhai K., Yin Z., Tan M., Yoder M.C., Leboulch P., Payen E., Srivastava A. High-efficiency transduction of primary human hematopoietic stem/progenitor cells by AAV6 vectors: strategies for overcoming donor-variation and implications in genome editing. Sci. Rep. 2016;6:35495. doi: 10.1038/srep35495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yang H., Qing K., Keeler G.D., Yin L., Mietzsch M., Ling C., Hoffman B.E., Agbandje-McKenna M., Tan M., Wang W., et al. Enhanced transduction of human hematopoietic stem cells by AAV6 vectors: implications in gene therapy and genome editing. Mol. Ther. Nucleic Acids. 2020;20:451–458. doi: 10.1016/j.omtn.2020.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Blacklow N.R., Hoggan M.D., Sereno M.S., Brandt C.D., Kim H.W., Parrott R.H., Chanock R.M. A seroepidemiologic study of Adenovirus-associated virus infection in infants and children. Am. J. Epidemiol. 1971;94:359–366. doi: 10.1093/oxfordjournals.aje.a121331. [DOI] [PubMed] [Google Scholar]
  • 148.Zaiss A.K., Cotter M.J., White L.R., Clark S.A., Wong N.C.W., Holers V.M., Bartlett J.S., Muruve D.A. Complement is an essential component of the immune response to adeno-associated virus vectors. J. Virol. 2008;82:2727–2740. doi: 10.1128/JVI.01990-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Sender R., Fuchs S., Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14:e1002533. doi: 10.1371/journal.pbio.1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.https://www.genengnews.com/topics/genome-editing/looking-toward-the-future-of-cell-gene-therapies/.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Document S1. Table S1
mmc1.pdf (735.9KB, pdf)
Document S2. Article plus supplemental information
mmc2.pdf (3.7MB, pdf)

Articles from Molecular Therapy. Nucleic Acids are provided here courtesy of The American Society of Gene & Cell Therapy

RESOURCES