probiologists.com
Open in
urlscan Pro
182.50.151.34
Public Scan
Submitted URL: https://list.probiologist.net/mautic/r/70287c366fe48e7c571d2d4c1?ct=YTo1OntzOjY6InNvdXJjZSI7YToyOntpOjA7czoxNDoiY2FtcGFpZ24uZX...
Effective URL: https://probiologists.com/Article/The-significance-of-triple-capsid-mutant-AAV8-for-treatment-of-Sanfilippo-Syndrome-Type-...
Submission: On January 10 via manual from IN — Scanned from FR
Effective URL: https://probiologists.com/Article/The-significance-of-triple-capsid-mutant-AAV8-for-treatment-of-Sanfilippo-Syndrome-Type-...
Submission: On January 10 via manual from IN — Scanned from FR
Form analysis
0 forms found in the DOMText Content
* Home * About Us * Journals * Contact Us ARCHIVES OF STEM CELL AND THERAPY ISSN : 2767-343X ISSN : 2767-343X * Home * Editorial Board * Submit Manuscript * Articles * In Press * Current Issue * Archives * Author Guidelines * Editorial Policies * For Editors * For Reviewers * Special Issues * Guidelines * Open Issues * Contact Commentary | Open Access | Volume 3 | Issue 1 | 2022 | DOI No.: 10.46439/stemcell.3.013 THE SIGNIFICANCE OF TRIPLE-CAPSID-MUTANT AAV8 FOR TREATMENT OF SANFILIPPO SYNDROME TYPE B Frederick Ashby, Coy Heldermon* University of Florida, College of Medicine, Gainesville, FL 32610-0278,USA *Corresponding Author: Coy Heldermon University of Florida, College of Medicine, Gainesville, FL 32610-0278,USA E-mail:Coy.heldermon@medicine.ufl.edu Received date: April 03, 2022; Accepted date: April 26, 2022 Citation: Ashby F, Heldermon C. The significance of triple-capsid-mutant AAV8 for treatment of Sanfilippo Syndrome Type B. Arch Stem Cell Ther.2022;3(1):11-17. Copyright: © 2022 Ashby F, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution,and reproduction in any medium,provided the original author and source are credited. ABSTRACT Sanfilippo Syndrome Type-B remains an untreatable childhood neurodegenerative disease with great burden for both patient and caregiver. Very few clinical trials have been undertaken to treat the disease, and none of these have yet yielded clinically obtainable products for patients. Caused by a simple enzyme function deficiency, Sanfilippo Syndrome Type-B has been considered a great prospect for gene-therapy interventions. Adeno-associated virus (AAV) remains a major choice for therapeutic gene delivery due to its relatively low-immunogenicity, versatility and tissue tropism. However, many clinical trials with AAV continue to use wild-type capsids, which in many cases are not able to reach stable transgene expression for long enough to be clinically effective in most cases. Previous research in AAV gene-therapy has created a litany of novel AAV capsids that can improve overall transduction efficiency far above that of wildtype AAV capsids. One such example is the triple-capsid mutant AAV8 (TCM8), which has been shown to exhibit transgene expression far superior to other capsids in Sanfilippo mouse models, specifically. Originally designed to bypass capsid ubiquitination intracellularly, mouse studies suggest this TCM8 vector outperforms both AAV5 and AAV9 when delivered to the central nervous system. This implies it as an ideal contender for an effective gene-therapy clinical trial candidate and has the potential to advance the progress of Sanfilippo Syndrome treatment. Here we provide commentary on the TCM8 vector and its context in the field of Sanfilippo Syndrome Type-B research. KEYWORDS Sanfilippo Syndrome, Mucopolysaccharidosis, Adeno-associated virus, Gene-therapy, AAV8, Capsid mutants SANFILIPPO SYNDROME TYPE-B Sanfilippo Syndrome, or Mucopolysaccharidosis (MPS) III, was first described in 1963 as an inherited condition of intellectual disability associated with significant mucopolysacchariduria, specifically of heparan sulfate [1]. The pathological elevation in mucopolysaccharides placed it in the same disease category as Hurler’s Syndrome (MPS I) and Hunter’s Syndrome (MPS II), however typically with less somatic manifestations and more central nervous system (CNS) dysfunction. MPS III is characterized on the cellular level by lysosomal distention, and grossly by organomegaly, coarsened facies and central nervous system degeneration [2]. Symptom onset typically happens in the first few years of life and can initially present as regression of developmental milestones and can be initially mistaken for isolated autism spectrum disorder (ASD). Recurrent ear, nose and throat infections are common in this disease, along with diarrhea and in some cases hearing loss [3]. After this initial phase, the next decade is typically characterized by behavioral problems, sleep disturbances and progressive cognitive and motor decline [4-8]. The final stage of the disease is typically characterized by severe CNS dysfunction, with seizures and coma [9,10]. MPS III Type A and B comprise over 80% of cases and have the most aggressive symptoms within the Sanfilippo Syndrome category with death commonly occurring within the first two decades of life. The most common cause of mortality is pneumonia, followed by cardiorespiratory failure [11]. The accumulation of heparan sulfate (HS), a sulfonated repeating-disaccharide glycosaminoglycan (GAG), in the context of MPS III occurs definitively from biallelic mutations affecting an enzyme involved in the degradation pathway. While each type is almost clinically indistinguishable, biochemically MPS III mutations affecting the SGSH gene (17q25.3) are categorized as Type A; while mutations affecting the NAGLU gene (17q21.2) are categorized as type B; mutations affecting the HGSNAT gene (8p11.21-p11.1) are Type C; and mutations affecting the GNS gene (12q14.3) are Type D[12] – with Type A and B commonly being the most severe. Of note, there remains an ARSG gene (17q24.2) in the HS degradation pathway, which if mutated would be referred to as Type E, however this has yet only been described in animal models [13]. All four types of confirmed human MPS III (A, B, C and D) have had causative mutations such as missense, nonsense and splicing along with small and large indels [13,14], illustrating the allelic heterogeneity of the disease. While promoter mutations and other gene-regulation-level mutations are not frequently described in MPS, promoter/3’-UTR mutations have been reported in MPS I [15]. Phenotypically, MPS III has a very wide range of severity depending on the degree of lost degradation function, due to either loss of respective enzyme expression, enzyme function or possibly a combination in rare circumstances. Due to this very simple pathophysiology, most MPS III therapies have the goal of increasing activity levels of the affected enzyme to ameliorate disease course. Enzyme Replacement Therapy (ERT) has been a reasonable proposal for treatment, along with gene-therapy and stem cell therapy, yet there are still no approved treatments for MPS IIIB. AAV GENE THERAPY Adeno-associated Virus (AAV), a replication deficient parvovirus, originally discovered as a contaminant [17,18], remains a popular choice for gene-therapy, in large part due to its relatively low immunogenicity, selective tissue tropism and overall versatility [19-21]. Seroprevalence studies suggest that over 90% of participants have been exposed to at least one serotype of AAV, with heavy variation between serotypes and populations [22-24]. Despite high seroprevalence, to date there have been no known diseases confirmed to be caused by the virus. However, there remains a long-standing debate on the relationship with AAV (wild or vector) and certain cancers [25,26]. The high prevalence of neutralizing antibodies to AAV in the general population remains a significant challenge to effective AAV treatments. Currently, there are three approved AAV gene-therapies (Table 1): AAV1-LPLS447X driven by cytomegalovirus (CMV) promoter for treatment of Lipoprotein Lipase Deficiency (LPLD) in 2012 [27]; AAV2-RPE65 driven by a CMV/Chicken- β-actin (CβA) hybrid promoter for treatment of inherited retinal dystrophy (RD) caused by biallelic RPE65 dysfunction in 2017 [28-30]; and self-complementary AAV9-SMN1 driven by a CMV/ CβA hybrid promoter for treatment of spinal muscular atrophy 1 (SMA1) in 2019 [31]. It is worth noting that each of these products utilizes wild-type AAV capsids. Currently, there are over a hundred active clinical trials involving AAV gene-therapy (ClinicalTrials.gov) treating a litany of diseases. Capsid Brand Name Generic Name Disease Trans-Gene Clinical Trial wtAAV1 Glybera® Alipogene tiparvovec LPLD CMVp-LPL(S447X) NCT02904772* wtAAV2 Luxturna® voretigene neparvovec- rzyl RD (RPE65 subtype) CMVp/CβAp-RPE65 NCT00999609 wtAAV9 Zolgensma® onasemnogene abeparvovec-xioi SMA1 CMVp/CβAp-scSMN1 NCT03381729 *Trial discontinued by company. Table 1: Currently Approved AAV Gene-therapies. CLINICAL TRIALS FOR MPS IIIB TREATMENT The clinicaltrial.gov registry currently only has 16 trials registered for Sanfilippo B (Table 2; Figure 1). Out of these 16 trials, 9 are observational with no treatment focus. The remaining 7 were focused on novel treatments. Four of these studies investigated two candidates for ERT: AX 250 (NCT03784287; NCT02754076) and SBC-103 (NCT02324049; NCT02618512), by both intravenous (IV) and intracerebroventricular (ICV) delivery methods. AX 250 is a chimeric fusion of recombinant human α-N-acetylglucosaminidase and truncated human insulin-like growth factor 2 (rhNAGLUIGF2), while SBC-103 is only a recombinant human α-Nacetylglucosaminidase (rhNAGLU). At least in the context of the study endpoints, such as developmental quotient (DQ), grey matter volume and age-equivalence (AE), SBC-103 did not appear to give adequate disease correction [32] when delivered IV (NCT02324049 or NCT02618512). However, it should be noted that family members of trial participants have claimed a noticeable improvement of non-endpoint metrics associated with both symptoms and quality of life, but these results have yet to be published in peer review. Meanwhile, the ICV trial of AX 250 is ongoing (NCT03784287). Two of the clinical trials for treatment of MPS IIIB investigated AAV gene therapy as a method. The first utilized a pseudotyped rAAV2/5- huNAGLU (wtAAV2 genome within a wtAAV5 capsid) administered intracerebrally (NCT03300453) by a 16-point intraparenchymal injection method in 4 patients with MPS IIIB. The patients were 20-53 months old at the time of treatment, with the youngest patient showing the best developmental outcomes overall, suggesting an optimal window for treatment [33]. However, it should be noted that a 5-year follow-up study found an escalating cell-mediated immune response in the cerebrospinal fluid (CSF) of study participants [34], underscoring the importance of recognizing the role of immunity in long-term treatment response [35]. The second AAV trial in MPS IIIB utilized rAAV9-CMVp-huNAGLU administered IV (NCT03315182). The study aimed to leverage the AAV9 serotype’s superior ability to cross the blood-brain barrier [36,37], which allows much less invasive administration. Continued development of this gene therapy product was recently discontinued by the company but had no reported significant adverse events. Additionally, the lentiviral-transduced autologous hematopoietic stem cell transplant based study that was just starting with Orchard Therapeutics was also discontinued by the company. This leaves MPS IIIB with no active trials using a gene therapy approach to curative therapy, reinforcing a need for development of approaches in this area. Figure 1: Current Clinical Trials in MPS IIIB: Out of the 16 clinical trials registered through the US National Libraries of Medicine (USNLM), 6 were randomized controlled trails (RTC’s). Only two of these trials tested rAAV treatments, and both utilized capsids from wtAAV serotypes. All data was obtained through the clinicaltrials.gov website. Study Name Study Type Study Identifier Intervention Study Sponsor Results Natural History Study of Patients With Mucopolysaccharidosis Type IIIB (MPS IIIB, Sanfilippo Syndrome Type B) Observational, Prospective, Cohort NCT01509768 N/A Shire Published in peer-review [57], completed 2013 Natural History Studies of Mucopolysaccharidosis III Observational, Prospective, Cohort NCT02037880 N/A Nationwide Children’s Hospital None provided, completed 2015 Natural History Study to Characterise the Course of Disease Progression in Participants With Mucopolysaccharidosis Type IIIB Observational, Prospective, Case- series NCT02293408 N/A Alexion Pharm. None provided, completed 2017 Neurobehavioral Phenotypes in MPS III Observational, Prospective, Cohort NCT01873911 N/A University of Minnesota; NIH U54NS065768 Published in peer-review [7,8], completed 2014 A Retrospective Chart Review of Deceased Patients With Mucopolysaccharidosis Type IIIB Observational, Retrospective, Case-series NCT02293382 N/A Alexion Pharm. None provided, completed 2015 A Study of Mucopolysaccharidosis Type IIIB (MPS IIIB) Observational, Prospective, Case- series NCT02493998 N/A Allievex Corp. None provided, completed 2019 Biomarker for Sanfilippo Type A-B-C-D Disease (BioSanfilippo) Observational, Prospective, Cohort NCT02298686 N/A CENTOGENE GmbH Rostock None provided, completed 2021 Evaluation of Blood Brain Barrier Integrity and Structural Abnormalities in MPS IIIB Patients Using Multimodal Magnetic Resonance Imaging Observational, Prospective, Case- series NCT02090179 N/A Alexion Pharm. None provided, completed 2016 A Treatment Study of Mucopolysaccharidosis Type IIIB (MPS IIIB) Interventional, Randomized Controlled Trial, Phase-1/2 NCT02754076 ERT, AX 250, IV Allievex Corp. None provided, completed 2020 A Treatment Extension Study of Mucopolysaccharidosis Type IIIB Interventional, Randomized Controlled Trial, Phase-2 NCT03784287 ERT, AX 250, ICV Allievex Corp. None provided, ongoing until 2025 Safety, Pharmacokinetics, and Pharmacodynamics/Efficacy of SBC- 103 in Mucopolysaccharidosis III, Type B (MPS IIIB) Interventional, Randomized Controlled Trial, Phase-1/2 NCT02324049 ERT, SBC-103, ICV Alexion Pharm. Published on USNLM, completed 2017 A Open Label Study in Previously Studied, SBC-103 Treatment Naïve MPS IIIB Subjects to Investigate the Safety, Pharmacokinetics, and Pharmacodynamics / Efficacy of SBC- 103 Administered Intravenously Interventional, Randomized Controlled Trial, Phase-1/2 NCT02618512 ERT, SBC-103, IV Alexion Pharm. Published on USNLM, Published in peer-review [32], completed 2018 Intracerebral Gene Therapy in Children With Sanfilippo Type B Syndrome Interventional, Randomized Controlled Trial, Phase-1/2 NCT03300453 Gene-therapy, rAAV2/5- hNAGLU UniQure Biopharma B.V. Published in peer-review [33], completed 2019 Gene Transfer Clinical Trial for Mucopolysaccharidosis (MPS) IIIB (MPSIIIB) Interventional, Randomized Controlled Trial, Phase-1/2 NCT03315182 Gene-therapy, rAAV9.CMV. hNAGLU, ABO-101 Abeona Therapeutics, Inc None provided, ongoing until 2022 A Long-term Follow-up Study of Patients With MPS IIIB Treated With ABO-101 Observational, Prospective, Cohort NCT04655911 Prior participation in ABO-101 Abeona Therapeutics, Inc None provided, ongoing until 2026 ERT: Enzyme Replacement Therapy; IV: Intravenous; ICV: Intracerebroventricular Table 2: Sanfilippo Type-B Clinical Trials Summary. DEVELOPMENT OF THE TCM8 VECTOR Efforts have been made for years to improve the effectiveness of AAV capsids in the context of human gene-therapy. While relatively effective in some trials, wild-type AAV capsids have not evolved to fill the niche they serve in gene-therapy treatment, but rather have evolved to propagate and survive in their natural host environment [38]. While AAV is weakly immunogenic, systemic AAV genetherapy typically requires very large vector loads to achieve desired effects, frequently exceeding 1013 vg/kg per patient. This can elicit complement activation with thrombocytopenia, humoral immunity within the liver and other organ injury, and in severe cases lifethreatening anaphylactic immune responses. Improving the efficiency of AAV vector can lower the dose required to achieve clinically significant outcomes, and thus reduce the risks to the patient while reducing the production cost of treatment. Vector efficiency has previously been improved preclinically by a combination of selective evolution of capsids [39-41], site-directed mutagenesis [42,43], capsid shuffling and chimerism [44-46], and even deep-learning simulation [47]. In order to identify better serotypes and methods for transduction of the brain, AAV5, AAV8, AAV9 and AAVrh10 were compared in wild type (WT) and MPS IIIB mouse models using a variety of injection methods, IV, thalamic, ventricular, ventral tegmental area or 6 intraparenchymal sites, to deliver a green fluorescent protein (GFP) reporter. In all mice, the broadest brain transduction was with the 6 intraparenchymal site injections. In contrast, the broadest transduction of WT brain marginally favored AAV9, however AAV8 clearly provided much broader and more intense brain transduction in MPS IIIB mice, and the other serotypes were similar between WT and MPS IIIB mice [48,49]. Building upon this disease specific enhanced brain tropism, the capsid modification principle was applied to AAV8 capsids. The triple-capsid mutant AAV8 (TCM8) proposed for treatment of Sanfilippo Syndrome type-B was originally designed to address the issue of capsid ubiquitination leading to proteasomal degradation of vectors post viral entry. Proteasomal degradation of capsids has been extensively reported to be a major cause of lost vector efficiency [40-54], so the next logical step in improving vector efficiency for many researchers has been to identify ways to circumvent this process. The process of ubiquitination is partially dependent on phosphorylation of amino acid residues on the target protein, therefore site-directed mutagenesis of exterior-facing serine, threonine or tyrosine residues to a sterically similar hydrophobic amino acid, such as phenylalanine for cyclic groups or valine for aliphatic groups, was shown to profoundly inhibit the ubiquitination process [42] without altering the capsid proteins’ function. The subsequent AAV8 capsids created included a double mutant (Y444 + 733F) and triple mutant (Y444 + 733F + T494V) that were designed based on these principles using X-ray crystallography to reference the exterior facing portions of the capsid. The triple-capsid mutant AAV8 had significantly stronger transduction efficiency compared to the double-mutant or wtAAV8, as measured by green-fluorescent protein (GFP) trans-gene expression in MPS IIIB mice [55]. Therefore, it was proposed as an optimal candidate to explore as a clinical therapy vector, specifically for MPS IIIB. Subsequently this AAV TCM8 vector was made with a codon-optimized NAGLU sequence insert and used for neonatal treatment of MPS IIIB mice with either the 6 intra-parenchymal injections or a single cisterna magna injection (Publication Currently Under Review). The treated mice demonstrate broad and high-level brain NAGLU activity and complete correction of functional and lifespan measures with either injection method demonstrating the clinical potential of the AAV TCM8 vector for correction of this disorder. CONCLUSION In summary, the results from the TCM8 studies in MPS IIIB overall suggest that this modified capsid vector may be capable of treating MPS IIIB patients with far greater efficiency than any treatments currently being investigated in humans. This is particularly apparent by the results published by Gilkes et al. 2016 [49], which suggest wtAAV8 is superior to the two other capsid serotypes currently under clinical trial (AAV5 & AAV9), at least in MPS IIIB mice. The subsequent follow-up by Gilkes et al. in 2021[55] show a profoundly increased transduction efficiency in TCM8 above wtAAV8, which would imply a commensurately higher efficiency than wtAAV5 & wtAAV9. Our follow-up studies using a therapeutic gene (Publication Currently Under Review) also show significant results. All of this is to suggest TCM8 may make an ideal candidate for clinical trial in MPS IIIB patients. One important limitation to the work done thus far relates to the phylogenetic divergence between humans and mice, and past animal studies in AAV have demonstrated the considerable effect this can have on AAV tissue tropism. One such example is the tropism of wtAAV8 for mouse hepatocytes without such a commensurate effect being observed in human hepatocytes [56], underscoring the need for humanized-liver mouse models in hepatocyte targeting rAAV studies and the consideration of humanized tissue models in-general. While it important to note that rAAV expression in a mouse model may or may not translate to humans well, currently the evidence suggests AAV TCM8 is a prime candidate for rAAV treatment of MPS IIIB. With the absence of a non-human primate MPS IIIB model, it is difficult to test TCM8 comparatively in primates preclinically, especially if this preference for TCM8 in the CNS is disease model dependent, as appears to be the case in the previous mouse studies.[49] Given the lack of current treatments or gene therapy clinical trials in this disease, the AAV TCM8-coNAGLU gene therapy vector is highly promising to advance treatment progress for MPS IIIB patients. ACKNOWLEDGEMENTS The Heldermon lab would like to acknowledge Sanfilippo Children’s Research Foundation, Sanfilippo Fundacja and Sanfilippo Initiative, Cure Sanfilippo, Lacerta Therapeutics, and NIH/NINDS R01NS102624. REFERENCES 1. Sanfilippo SJ. Mental retardation associated with acid mucopolysacchariduria (heparan sulfate type). J Pediatr. 1963;63:837-8. 2. Valstar MJ, Ruijter GJ, Van Diggelen OP, Poorthuis BJ, Wijburg FA. Sanfilippo syndrome: a mini-review. Journal of Inherited Metabolic Disease. 2008 Apr;31(2):240-52. 3. Cleary MA, Wraith JE. Management of mucopolysaccharidosis type III. Archives of Disease in Childhood. 1993 Sep;69(3):403-6. 4. Bax MC, Colville GA. Behaviour in mucopolysaccharide disorders. Archives of Disease in Childhood. 1995 Jul 1;73(1):77-81. 5. Fraser J, Wraith JE, Delatycki MB. Sleep disturbance in mucopolysaccharidosis type III (Sanfilippo syndrome): a survey of managing clinicians. Clinical Genetics. 2002 Nov;62(5):418-21. 6. Valstar MJ, Marchal JP, Grootenhuis M, Colland V, Wijburg FA. Cognitive development in patients with Mucopolysaccharidosis type III (Sanfilippo syndrome). Orphanet Journal of Rare Diseases. 2011 Dec;6(1):43. 7. Shapiro EG, Nestrasil I, Delaney KA, Rudser K, Kovac V, Nair N, et al. A prospective natural history study of mucopolysaccharidosis type IIIA. The Journal of Pediatrics. 2016 Mar 1;170:278-87. 8. Shapiro E, King K, Ahmed A, Rudser K, Rumsey R, Yund B, et al. The neurobehavioral phenotype in mucopolysaccharidosis type IIIB: an exploratory study. Molecular Genetics and Metabolism Reports. 2016 Mar 1;6:41-7. 9. Nidiffer FD, Kelly T. Developmental and degenerative patterns associated with cognitive, behavioural and motor difficulties in the Sanfilippo syndrome: an epidemiological study. Journal of Mental Deficiency Research. 1983 Sep 1;27:185-203. 10. Valstar MJ, Bruggenwirth HT, Olmer R, Wevers RA, Verheijen FW, Poorthuis BJ, et al. Mucopolysaccharidosis type IIIB may predominantly present with an attenuated clinical phenotype. Journal of Inherited Metabolic Disease: Official Journal of the Society for the Study of Inborn Errors of Metabolism. 2010 Dec;33(6):759-67. 11. Lavery C, Hendriksz CJ, Jones SA. Mortality in patients with Sanfilippo syndrome. Orphanet Journal of Rare Diseases. 2017 Dec;12(1):168. 12. Coutinho MF, Lacerda L, Alves S. Glycosaminoglycan storage disorders: a review. Biochemistry Research International. 2012 Oct;2012:1-16. 13. Fedele AO. Sanfilippo syndrome: causes, consequences, and treatments. The application of Clinical Genetics. 2015;8:269-81. 14. Gilkes JA, Heldermon CD. Mucopolysaccharidosis III (Sanfilippo Syndrome)-disease presentation and experimental therapies. Pediatric Endocrinology Reviews: PER. 2014 Sep 1;12:133-40. 15. Stenson PD, Mort M, Ball EV, Evans K, Hayden M, Heywood S, et al. The Human Gene Mutation Database: towards a comprehensive repository of inherited mutation data for medical research, genetic diagnosis and next-generation sequencing studies. Human Genetics. 2017 Jun;136(6):665-77. 16. Zelei T, Csetneki K, Voko Z, Siffel C. Epidemiology of Sanfilippo syndrome: results of a systematic literature review. Orphanet Journal of Rare Diseases. 2018 Dec;13(1):53. 17. Atchison RW, Casto BC, Hammon WM. Adenovirus-associated defective virus particles. Science. 1965 Aug 13;149(3685):754-6. 18. Hoggan MD, Blacklow NR, Rowe W. Studies of small DNA viruses found in various adenovirus preparations: physical, biological, and immunological characteristics. Proceedings of the National Academy of Sciences of the United States of America. 1966 Jun;55(6):1467-74. 19. Gonçalves MA. Adeno-associated virus: from defective virus to effective vector. Virology Journal. 2005 Dec;2(1):43. 20. Wang D, Tai PW, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nature Reviews Drug Discovery. 2019 May;18(5):358-78. 21. Calcedo R, Vandenberghe LH, Gao G, Lin J, Wilson JM. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. The Journal of Infectious Diseases. 2009 Feb 1;199(3):381-90. 22. Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus MF, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Human Gene Therapy. 2010 Jun 1;21(6):704-12. 23. Khatri A, Shelke R, Guan S, Somanathan S. Higher Seroprevalence of Anti-Adeno-Associated Viral Vector Neutralizing Antibodies Among Racial Minorities in the United States. Human Gene Therapy. 2022 Feb 2; hum.2021.243. 24. Srivastava A, Carter BJ. AAV infection: protection from cancer. Human Gene Therapy. 2017 Apr 1;28(4):323-7. 25. Nguyen GN, Everett JK, Kafle S, Roche AM, Raymond HE, Leiby J, et al. A long-term study of AAV gene therapy in dogs with hemophilia A identifies clonal expansions of transduced liver cells. Nature Biotechnology. 2021 Jan;39(1):47-55. 26. Gaudet D, Méthot J, Déry S, Brisson D, Essiembre C, Tremblay G, et al. Efficacy and long-term safety of alipogene tiparvovec (AAV1- LPLS447X) gene therapy for lipoprotein lipase deficiency: an openlabel trial. Gene Therapy. 2013 Apr;20(4):361-9. 27. Patel U, Boucher M, de Léséleuc L, Visintini S. Voretigene neparvovec: an emerging gene therapy for the Treatment of Inherited Blindness. In: CADTH Issues in Emerging Health Technologies. Canadian Agency for Drugs and Technologies in Health; 2016. 28. Ciulla TA, Hussain RM, Berrocal AM, Nagiel A. Voretigene neparvovec-rzyl for treatment of RPE65-mediated inherited retinal diseases: a model for ocular gene therapy development. Expert Opinion on Biological Therapy. 2020 Jun 2;20(6):565-78. 29. Amato A, Arrigo A, Aragona E, Manitto MP, Saladino A, Bandello F, et al. Gene Therapy in Inherited Retinal Diseases: An Update on Current State of the Art. Frontiers in Medicine. 2021;8:750586. 30. Stevens D, Claborn MK, Gildon BL, Kessler TL, Walker C. Onasemnogene abeparvovec-xioi: gene therapy for spinal muscular atrophy. Annals of Pharmacotherapy. 2020 Oct;54(10):1001-9. 31. Whitley CB, Vijay S, Yao B, Pineda M, Parker GJ, Rojas-Caro S, et al. Final results of the phase 1/2, open-label clinical study of intravenous recombinant human N-acetyl-a-d-glucosaminidase (SBC-103) in children with mucopolysaccharidosis IIIB. Molecular Genetics and Metabolism. 2019 Feb 1;126(2):131-8. 32. Tardieu M, Zérah M, Gougeon ML, Ausseil J, de Bournonville S, Husson B, et al. Intracerebral gene therapy in children with mucopolysaccharidosis type IIIB syndrome: an uncontrolled phase 1/2 clinical trial. The Lancet Neurology. 2017 Sep 1;16(9):712-20. 33. Deiva K, Ausseil J, de Bournonville S, Zérah M, Husson B, Gougeon ML, et al. Intracerebral Gene Therapy in Four Children with Sanfilippo B Syndrome: 5.5-Year Follow-Up Results. Human Gene Therapy. 2021 Oct 1;32(19-20):1251-9. 34. Gougeon ML, Poirier-Beaudouin B, Ausseil J, Zérah M, Artaud C, Heard JM, et al. Cell-mediated immunity to NAGLU transgene following intracerebral gene therapy in children with Mucopolysaccharidosis type IIIB syndrome. Frontiers in Immunology. 2021 May 10;12: 655478. 35. Fu H, DiRosario J, Killedar S, Zaraspe K, McCarty DM. Correction of neurological disease of mucopolysaccharidosis IIIB in adult mice by rAAV9 trans-blood–brain barrier gene delivery. Molecular Therapy. 2011 Jun 1;19(6):1025-33. 36. Murrey DA, Naughton BJ, Duncan FJ, Meadows AS, Ware TA, Campbell KJ, et al. Feasibility and safety of systemic rAAV9-h NAGLU delivery for treating mucopolysaccharidosis IIIB: toxicology, biodistribution, and immunological assessments in primates. Human Gene Therapy Clinical Development. 2014 Jun 1;25(2):72- 84. 37. Kotterman MA, Schaffer DV. Engineering adeno-associated viruses for clinical gene therapy. Nature Reviews Genetics. 2014 Jul;15(7):445-51. 38. Bartel MA, Weinstein JR, Schaffer DV. Directed evolution of novel adeno-associated viruses for therapeutic gene delivery. Gene Therapy. 2012 Jun;19(6):694-700. 39. Davidsson M, Wang G, Aldrin-Kirk P, Cardoso T, Nolbrant S, Hartnor M, et al. A systematic capsid evolution approach performed in vivo for the design of AAV vectors with tailored properties and tropism. Proceedings of the National Academy of Sciences. 2019 Dec 26;116(52):27053-62. 40. Byrne LC, Day TP, Visel M, Strazzeri JA, Fortuny C, Dalkara D, et al. In vivo–directed evolution of adeno-associated virus in the primate retina. JCI insight. 2020 May 21;5(10). 41. Zhong L, Li B, Mah CS, Govindasamy L, Agbandje-McKenna M, Cooper M, et al. Next generation of adeno-associated virus 2 vectors: point mutations in tyrosines lead to high-efficiency transduction at lower doses. Proceedings of the National Academy of Sciences. 2008 Jun 3;105(22):7827-32. 42. Li B, Ma W, Ling C, Van Vliet K, Huang LY, Agbandje-McKenna M, et al. Site-directed mutagenesis of surface-exposed lysine residues leads to improved transduction by AAV2, but not AAV8, vectors in murine hepatocytes in vivo. Human Gene Therapy Methods. 2015 Dec 1;26(6):211-20. 43. Li W, Asokan A, Wu Z, Van Dyke T, DiPrimio N, Johnson JS, et al. Engineering and selection of shuffled AAV genomes: a new strategy for producing targeted biological nanoparticles. Molecular Therapy. 2008 Jul 1;16(7):1252-60. 44. Herrmann AK, Bender C, Kienle E, Grosse S, El Andari J, Botta J, et al. A robust and all-inclusive pipeline for shuffling of adeno-associated viruses. ACS Synthetic Biology. 2018 Dec 4;8(1):194-206. 45. Viney L, Bürckstümmer T, Eddington C, Mietzsch M, Choudhry M, Henley T, et al. Adeno-associated virus (AAV) capsid chimeras with enhanced infectivity reveal a core element in the AAV genome critical for both cell transduction and capsid assembly. Journal of Virology. 2021 Jan 13;95(7):e02023-20. 46. Bryant DH, Bashir A, Sinai S, Jain NK, Ogden PJ, Riley PF, et al. Deep diversification of an AAV capsid protein by machine learning. Nature Biotechnology. 2021 Jun;39(6):691-6. 47. Gilkes JA, Bloom MD, Heldermon CD. Preferred transduction with AAV8 and AAV9 via thalamic administration in the MPS IIIB model: A comparison of four rAAV serotypes. Molecular Genetics and Metabolism Reports. 2016 Mar 1;6:48-54. 48. Gilkes JA, Bloom MD, Heldermon CD. Mucopolysaccharidosis IIIB confers enhanced neonatal intracranial transduction by AAV8 but not by 5, 9 or rh10. Gene Therapy. 2016 Mar;23(3):263-71. 49. Duan D, Yue Y, Yan Z, Yang J, Engelhardt JF. Endosomal processing limits gene transfer to polarized airway epithelia by adenoassociated virus. The Journal of Clinical Investigation. 2000 Jun 1;105(11):1573-87. 50. Douar AM, Poulard K, Stockholm D, Danos O. Intracellular trafficking of adeno-associated virus vectors: routing to the late endosomal compartment and proteasome degradation. Journal of Virology.2001 Feb 15;75(4):1824-33. 51. Ding W, Zhang LN, Yeaman C, Engelhardt JF. rAAV2 traffics through both the late and the recycling endosomes in a dose-dependent fashion. Molecular Therapy. 2006 Apr 1;13(4):671-82. 52. Dhungel BP, Bailey CG, Rasko JE. Journey to the Center of the Cell: Tracing the Path of AAV Transduction. Trends in Molecular Medicine. 2021 Feb 1;27(2):172-84. 53. Aslanidi GV, Rivers AE, Ortiz L, Song L, Ling C, Govindasamy L, et al. Optimization of the capsid of recombinant adeno-associated virus 2 (AAV2) Vectors: the Final Threshold?. PloS One. 2013 Mar 19;8(3):e59142. 54. Gilkes JA, Judkins BL, Herrera BN, Mandel RJ, Boye SL, Boye SE, et al. Site-specific modifications to AAV8 capsid yields enhanced brain transduction in the neonatal MPS IIIB mouse. Gene Therapy. 2021 Aug;28(7):447-55. 55. Vercauteren K, Hoffman BE, Zolotukhin I, Keeler GD, Xiao JW, Basner-Tschakarjan E, et al. Superior in vivo transduction of human hepatocytes using engineered AAV3 capsid. Molecular Therapy. 2016 Jun 1;24(6):1042-9. 56. Whitley CB, Cleary M, Mengel KE, Harmatz P, Shapiro E, Nestrasil I, et al. Observational prospective natural history of patients with Sanfilippo syndrome type B. The Journal of Pediatrics. 2018 Jun 1;197:198-206. ARCHIVES OF STEM CELL AND THERAPY Contents Register EDITORIAL BOARD Kartoosh Heydari Stanford Health Care USA Amanda Baracho Trindade Hill Max Planck University Brazil Himanshu Arora University of Miami USA TWITTER FEED PROBIOLOGISTS LLC. Our Locations * North America * 600N Broad Street 5 - 906, * Middletown, DE 19709, USA * Europe * 63-66 Hatton Garden, London, * England, EC1N 8LE * Contact Us * Tel : +1 302 261 2410 * Whatsapp : +1 302 261 2410 * contact@probiologists.com QUICK LINKS * Home Author Guidelines * About Us Policies * Journals Online Submission * Articles Contact US ©Copyright ProBiologists 2023