1
|
Keeler AM, Zhan W, Ram S, Fitzgerald KA, Gao G. The curious case of AAV immunology. Mol Ther 2025:S1525-0016(25)00211-4. [PMID: 40156190 DOI: 10.1016/j.ymthe.2025.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Immune responses to adeno-associated virus (AAV) have long been perplexing, from its first discovery to the latest clinical trials of recombinant AAV (rAAV) therapy. Wild-type AAV (wtAAV) does not cause any known disease, making it an ideal vector for gene therapy, as viral vectors retain virus-like properties. Although AAV stimulates only a mild immune response compared with other viruses, it is still recognized by the innate immune system and induces adaptive immune responses. B cell responses against both wtAAV and rAAV are robust and can hinder gene therapy applications and prevent redosing. T cell responses can clear transduced cells or establish tolerance against gene therapy. Immune responses to AAV gene therapy are influenced by many factors. Most clinical immunotoxicities that develop in response to gene therapies have emerged as higher doses of AAV vectors have been utilized and were not properly modeled in preclinical animal studies. Thus, several strategies have been undertaken to reduce or mitigate immune responses to AAV. While we have learned a considerable amount about how the immune system responds to AAV gene therapy since the discovery of AAV virus, it still remains a curious case that requires more investigation to fully understand.
Collapse
Affiliation(s)
- Allison M Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; NeroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Katherine A Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
2
|
Herzog RW, Kaczmarek R, High KA. Gene therapy for hemophilia - From basic science to first approvals of "one-and-done" therapies. Mol Ther 2025:S1525-0016(25)00217-5. [PMID: 40156189 DOI: 10.1016/j.ymthe.2025.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Realistic paths to gene therapy for the X-linked bleeding disorder hemophilia started to materialize in the mid 1990s, resulting in disease correction in small and large animal models. Out of a diversity of approaches, in vivo adeno-associated viral (AAV) gene transfer to hepatocytes emerged as the most promising strategy, eventually forming the basis for multiple advanced clinical trials and regulatory approval of two products for the treatment of hemophilia B (coagulation factor IX deficiency) and one for hemophilia A (factor VIII deficiency). Ideally, gene therapy is effective with a single administration, thus providing therapeutic factor levels over a period of years, without the need for frequent injections. Overcoming multiple obstacles, some not predicted by preclinical studies, sustained partial to complete correction of coagulation for several years to an entire decade has now been documented in patients, with observation ongoing. A hyperactive form of FIX improved efficacy in hemophilia B, and superior engineered variants of FVIII are emerging. Nonetheless, challenges remain, including pre-existing immunity to AAV capsids, toxicities, inter-patient variability in response to treatment, and difficulty in obtaining durable therapeutic expression of FVIII. In alternative approaches, in vivo gene editing and ex vivo gene therapies targeting hemopoietic cells are in development.
Collapse
Affiliation(s)
- Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Radoslaw Kaczmarek
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Katherine A High
- Laboratory of Blood and Vascular Biology, Rockefeller University, New York, NY, USA.
| |
Collapse
|
3
|
Young G. Induction of factor VIII tolerance by hemophilia gene transfer to eradicate factor VIII inhibitors. Blood Adv 2025; 9:265-269. [PMID: 39418639 PMCID: PMC11782813 DOI: 10.1182/bloodadvances.2024013000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
ABSTRACT Patients with hemophilia A can develop antifactor antibodies to factor VIII. The incidence is ∼30%, and such patients suffer worse morbidity and mortality. The only proven method to eradicate these inhibitors is via immune tolerance induction therapy, which consists of infusing factor VIII concentrates at regular intervals. This approach is effective ∼65% of the time, leaving at least a third of patients who develop inhibitors with this lifelong problem. Although emicizumab has greatly improved the quality of life of inhibitor patients, eradicating the inhibitor remains an important treatment goal. Animal models have shown the potential for gene therapy to induce tolerance. A recent abstract describing a study in humans demonstrated the potential for successful tolerance induction. This article will describe the rationale for using gene therapy to induce tolerance and provide this author's viewpoint on the importance and possible historic significance of attempting to eradicate inhibitors with this approach.
Collapse
Affiliation(s)
- Guy Young
- Cancer and Blood Disorders Institute, Children’s Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| |
Collapse
|
4
|
Tyravska Y, Nadeem T, Savchenko O, Bondarchuk O, Talabko Y. Immunohaemostasis and the significance of immune reactions in the regulation of blood coagulation. Eur J Microbiol Immunol (Bp) 2024; 14:392-404. [PMID: 39630220 PMCID: PMC11836649 DOI: 10.1556/1886.2024.00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction This study was conducted to determine the specific features of the mutual influence of the immune and haemostatic systems in immunohaemostasis, the role of immune reactions in the regulation of blood coagulation, and the efficacy of modern methods of treating thrombosis and bleeding. Methods The study analysed relevant scientific sources on immunology and haematology and identified the specific features of the blood clotting process and the role of immune reactions in it. Results The study found that the immune system influences the haematological system through the interaction of blood clotting factors, platelets, plasminogen, endothelial cells with immune cells. The haemostatic system influences the immune system through mechanisms to maintain immune tolerance and immune memory and the properties of clotting factors to activate the stimulation and migration of immune cells to the site of infection. Immune reactions regulate blood coagulation by activating platelets, regulating blood coagulation factors, affecting fibrinolysis, and immune tolerance. The process of platelet activation involves immune cells, immune complexes, and microbial components. The regulation of blood coagulation factors is influenced by the ability of immune cells to produce activators and inhibitors of these factors and to stimulate or slow down fibrinolysis. The immune system's maintenance of immune tolerance to blood components is regulated by mechanisms of immune response suppression, partial immune ignoring of certain blood elements, inhibition of activation of certain immune cells, apoptosis, and selection of immature T-lymphocytes. Treatment methods for patients at risk of thrombosis and bleeding include anticoagulation, antiplatelet, dual antiplatelet therapy, thrombectomy, endovascular methods, medical prophylaxis of bleeding, and coagulation monitoring. Conclusions The findings of this study suggest the significance of immune responses in the regulation of blood coagulation processes, and therefore they can be used in the development of immunotherapy methods for the treatment of thrombosis and bleeding.
Collapse
Affiliation(s)
- Yuliya Tyravska
- Department of Internal Medicine No. 4, Bogomolets National Medical University, Kyiv, Ukraine
| | - Tarana Nadeem
- Department of Internal Medicine No. 4, Bogomolets National Medical University, Kyiv, Ukraine
| | - Oleksandr Savchenko
- Department of Internal Medicine No. 4, Bogomolets National Medical University, Kyiv, Ukraine
| | - Oleksandr Bondarchuk
- Department of Internal Medicine No. 4, Bogomolets National Medical University, Kyiv, Ukraine
| | - Yuliia Talabko
- Department of Internal Medicine No. 4, Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
5
|
Samelson-Jones BJ, Doshi BS, George LA. Coagulation factor VIII: biological basis of emerging hemophilia A therapies. Blood 2024; 144:2185-2197. [PMID: 39088776 PMCID: PMC11600081 DOI: 10.1182/blood.2023023275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024] Open
Abstract
ABSTRACT Coagulation factor VIII (FVIII) is essential for hemostasis. After activation, it combines with activated FIX (FIXa) on anionic membranes to form the intrinsic Xase enzyme complex, responsible for activating FX in the rate-limiting step of sustained coagulation. Hemophilia A (HA) and hemophilia B are due to inherited deficiencies in the activity of FVIII and FIX, respectively. Treatment of HA over the last decade has benefited from an improved understanding of FVIII biology, including its secretion pathway, its interaction with von Willebrand factor in circulation, the biochemical nature of its FIXa cofactor activity, the regulation of activated FVIII by inactivation pathways, and its surprising immunogenicity. This has facilitated biotechnology innovations with first-in-class examples of several new therapeutic modalities recently receiving regulatory approval for HA, including FVIII-mimetic bispecific antibodies and recombinant adeno-associated viral (rAAV) vector-based gene therapy. Biological insights into FVIII also guide the development and use of gain-of-function FVIII variants aimed at addressing the limitations of first-generation rAAV vectors for HA. Several gain-of-function FVIII variants designed to have improved secretion are currently incorporated in second-generation rAAV vectors and have recently entered clinical trials. Continued mutually reinforcing advancements in the understanding of FVIII biology and treatments for HA are necessary to achieve the ultimate goal of hemophilia therapy: normalizing hemostasis and optimizing well-being with minimal treatment burden for all patients worldwide.
Collapse
Affiliation(s)
- Benjamin J. Samelson-Jones
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
- Division of Hematology, Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia PA
| | - Bhavya S. Doshi
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
- Division of Hematology, Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia PA
| | - Lindsey A. George
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
- Division of Hematology, Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia PA
| |
Collapse
|
6
|
Levy-Mendelovich S, Avishai E, Samelson-Jones BJ, Dardik R, Brutman-Barazani T, Nisgav Y, Livnat T, Kenet G. A Novel Murine Model Enabling rAAV8-PC Gene Therapy for Severe Protein C Deficiency. Int J Mol Sci 2024; 25:10336. [PMID: 39408666 PMCID: PMC11477312 DOI: 10.3390/ijms251910336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Severe protein C deficiency (SPCD) is a rare inherited thrombotic disease associated with high morbidity and mortality. In the current study, we established a viable murine model of SPCD, enabling preclinical gene therapy studies. By creating SPCD mice with severe hemophilia A (PROC-/-/F8-), the multi-month survival of SPCD mice enabled the exploration of recombinant adeno-associated viral vector-PC (rAAV8-PC) gene therapy (GT). rAAV8- PC (1012 vg/kg of AAV8-PC) was injected via the tail vein into 6-8-week-old PROC-/-/F8- mice. Their plasma PC antigen levels (median of 714 ng/mL, range 166-2488 ng/mL) and activity (303.5 ± 59%) significantly increased to the normal range after GT compared to untreated control animals. PC's presence in the liver after GT was also confirmed by immunofluorescence staining. Our translational research results provide the first proof of concept that an infusion of rAAV8-PC increases PC antigen and activity in mice and may contribute to future GT in SPCD. Further basic research of SPCD mice with prolonged survival due to the rebalancing of this disorder using severe hemophilia A may provide essential data regarding PC's contribution to specific tissues' development, local PC generation, and its regulation in inflammatory conditions.
Collapse
Affiliation(s)
- Sarina Levy-Mendelovich
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Einat Avishai
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Benjamin J. Samelson-Jones
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Division of Hematology, Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rima Dardik
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tami Brutman-Barazani
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yael Nisgav
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tami Livnat
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gili Kenet
- National Hemophilia Center, Thrombosis & Hemostasis Institute, Sheba Medical Center, Ramat Gan 52621, Israel; (E.A.); (R.D.); (T.B.-B.); (Y.N.); (T.L.); (G.K.)
- Amalia Biron Research Institute of Thrombosis & Hemostasis, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
7
|
Lam P, Zygmunt DA, Ashbrook A, Bennett M, Vetter TA, Martin PT. Dual FKRP/FST gene therapy normalizes ambulation, increases strength, decreases pathology, and amplifies gene expression in LGMDR9 mice. Mol Ther 2024; 32:2604-2623. [PMID: 38910327 PMCID: PMC11405156 DOI: 10.1016/j.ymthe.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/12/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
Recent clinical studies of single gene replacement therapy for neuromuscular disorders have shown they can slow or stop disease progression, but such therapies have had little impact on reversing muscle disease that was already present. To reverse disease in patients with muscular dystrophy, new muscle mass and strength must be rebuilt at the same time that gene replacement prevents subsequent disease. Here, we show that treatment of FKRPP448L mice with a dual FKRP/FST gene therapy packaged into a single adeno-associated virus (AAV) vector can build muscle strength and mass that exceed levels found in wild-type mice and can induce normal ambulation endurance in a 1-h walk test. Dual FKRP/FST therapy also showed more even increases in muscle mass and amplified muscle expression of both genes relative to either single gene therapy alone. These data suggest that treatment with single AAV-bearing dual FKRP/FST gene therapies can overcome loss of ambulation by improving muscle strength at the same time it prevents subsequent muscle damage. This design platform could be used to create therapies for other forms of muscular dystrophy that may improve patient outcomes.
Collapse
Affiliation(s)
- Patricia Lam
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Macey Bennett
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Tatyana A Vetter
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Paul T Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, and Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
8
|
Kaczmarek R, Samelson-Jones BJ, Herzog RW. Immune tolerance induction by hepatic gene transfer: First-in-human evidence. Mol Ther 2024; 32:863-864. [PMID: 38518767 PMCID: PMC11163209 DOI: 10.1016/j.ymthe.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/24/2024] Open
Affiliation(s)
- Radoslaw Kaczmarek
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benjamin J Samelson-Jones
- Department of Pediatrics, The Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
9
|
Coyle CW, Knight KA, Brown HC, George SN, Denning G, Branella GM, Childers KC, Spiegel PC, Spencer HT, Doering CB. Humanization and functional characterization of enhanced coagulation factor IX variants identified through ancestral sequence reconstruction. J Thromb Haemost 2024; 22:633-644. [PMID: 38016519 PMCID: PMC10922771 DOI: 10.1016/j.jtha.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Laboratory resurrection of ancient coagulation factor (F) IX variants generated through ancestral sequence reconstruction led to the discovery of a FIX variant, designated An96, which possesses enhanced specific activity independent of and additive to that provided by human p.Arg384Lys, referred to as FIX-Padua. OBJECTIVES The goal of the current study was to identify the amino acid substitution(s) responsible for the enhanced activity of An96 and create a humanized An96 FIX transgene for gene therapy application. METHODS Reductionist screening approaches, including domain swapping and scanning residue substitution, were used and guided by one-stage FIX activity assays. In vitro characterization of top candidates included recombinant high-purity preparation, specific activity determination, and enzyme kinetic analysis. Final candidates were packaged into adeno-associated viral (AAV) vectors and delivered to hemophilia B mice. RESULTS Five of 42 total amino acid substitutions in An96 appear sufficient to retain the enhanced activity of An96 in an otherwise human FIX variant. Additional substitution of the Padua variant further increased the specific activity 5-fold. This candidate, designated ET9, demonstrated 51-fold greater specific activity than hFIX. AAV2/8-ET9 treated hemophilia B mice produced plasma FIX activities equivalent to those observed previously for AAV2/8-An96-Padua, which were 10-fold higher than AAV2/8-hFIX-Padua. CONCLUSION Starting from computationally inferred ancient FIX sequences, novel amino acid substitutions conferring activity enhancement were identified and translated into an AAV-FIX gene therapy cassette demonstrating high potency. This ancestral sequence reconstruction discovery and sequence mapping refinement approach represents a promising platform for broader protein drug and gene therapy candidate optimization.
Collapse
Affiliation(s)
- Christopher W Coyle
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kristopher A Knight
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | - Gianna M Branella
- Cancer Biology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kenneth C Childers
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - P Clint Spiegel
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - H Trent Spencer
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Christopher B Doering
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
10
|
Gaddie CD, Senior KG, Chan C, Hoffman BE, Keeler GD. Upregulation of CD8 + regulatory T cells following liver-directed AAV gene therapy. Cell Immunol 2024; 397-398:104806. [PMID: 38244266 DOI: 10.1016/j.cellimm.2024.104806] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024]
Abstract
Liver-directed AAV gene therapy represents a unique treatment modality for a host of diseases. This is due, in part, to the induction of tolerance to transgene products. Despite the plethora of recognized regulatory cells in the body, there is currently a lack of literature supporting the induction of non-CD4+ regulatory cells following hepatic AAV gene transfer. In this work, we show that CD8+ regulatory T cells are up-regulated in PBMCs of mice following capsid only and therapeutic transgene AAV administration. Further, we demonstrate that hepatic AAV gene transfer results in a significant increase in CD8+ regulatory T cells following experimental autoimmune encephalomyelitis induction. Notably, this response occurred only in therapeutic vector treated animals, not capsid only controls. Understanding the role these cells play in treatment efficacy will result in the development of improved AAV vectors that take advantage of the full gamut of regulatory cells within the body.
Collapse
Affiliation(s)
- Cristina D Gaddie
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Kevin G Senior
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Christopher Chan
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Brad E Hoffman
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA; Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Geoffrey D Keeler
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
Valentino LA, Ozelo MC, Herzog RW, Key NS, Pishko AM, Ragni MV, Samelson-Jones BJ, Lillicrap D. A review of the rationale for gene therapy for hemophilia A with inhibitors: one-shot tolerance and treatment? J Thromb Haemost 2023; 21:3033-3044. [PMID: 37225021 DOI: 10.1016/j.jtha.2023.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/09/2023] [Accepted: 05/14/2023] [Indexed: 05/26/2023]
Abstract
The therapeutic landscape for people living with hemophilia A (PwHA) has changed dramatically in recent years, but many clinical challenges remain, including the development of inhibitory antibodies directed against factor VIII (FVIII) that occur in approximately 30% of people with severe hemophilia A. Emicizumab, an FVIII mimetic bispecific monoclonal antibody, provides safe and effective bleeding prophylaxis for many PwHA, but clinicians still explore therapeutic strategies that result in immunologic tolerance to FVIII to enable effective treatment with FVIII for problematic bleeding events. This immune tolerance induction (ITI) to FVIII is typically accomplished through repeated long-term exposure to FVIII using a variety of protocols. Meanwhile, gene therapy has recently emerged as a novel ITI option that provides an intrinsic, consistent source of FVIII. As gene therapy and other therapies now expand therapeutic options for PwHA, we review the persistent unmet medical needs with respect to FVIII inhibitors and effective ITI in PwHA, the immunology of FVIII tolerization, the latest research on tolerization strategies, and the role of liver-directed gene therapy to mediate FVIII ITI.
Collapse
Affiliation(s)
- Leonard A Valentino
- National Hemophilia Foundation, New York, New York, USA; Rush University, Chicago, Illinois, USA.
| | | | - Roland W Herzog
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nigel S Key
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | | | | | | | | |
Collapse
|
12
|
Nguyen NH, Jarvi NL, Balu-Iyer SV. Immunogenicity of Therapeutic Biological Modalities - Lessons from Hemophilia A Therapies. J Pharm Sci 2023; 112:2347-2370. [PMID: 37220828 DOI: 10.1016/j.xphs.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/25/2023]
Abstract
The introduction and development of biologics such as therapeutic proteins, gene-, and cell-based therapy have revolutionized the scope of treatment for many diseases. However, a significant portion of the patients develop unwanted immune reactions against these novel biological modalities, referred to as immunogenicity, and no longer benefit from the treatments. In the current review, using Hemophilia A (HA) therapy as an example, we will discuss the immunogenicity issue of multiple biological modalities. Currently, the number of therapeutic modalities that are approved or recently explored to treat HA, a hereditary bleeding disorder, is increasing rapidly. These include, but are not limited to, recombinant factor VIII proteins, PEGylated FVIII, FVIII Fc fusion protein, bispecific monoclonal antibodies, gene replacement therapy, gene editing therapy, and cell-based therapy. They offer the patients a broader range of more advanced and effective treatment options, yet immunogenicity remains the most critical complication in the management of this disorder. Recent advances in strategies to manage and mitigate immunogenicity will also be reviewed.
Collapse
Affiliation(s)
- Nhan H Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA; Currently at Truvai Biosciences, Buffalo, NY, USA
| | - Nicole L Jarvi
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
13
|
Mihaila RG. From a bispecific monoclonal antibody to gene therapy: A new era in the treatment of hemophilia A. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2023; 167:1-8. [PMID: 36413008 DOI: 10.5507/bp.2022.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/02/2022] [Indexed: 11/23/2022] Open
Abstract
The treatment of hemophilia A has progressed amazingly in recent years. Emicizumab, a bispecific-humanized monoclonal antibody, is able to improve coagulation by bridging activated factor IX and factor X. Emicizumab is administered subcutaneously and much less often compared to factor VIII products. It has low immunogenicity, does not require dose adjustment, and can be administered regardless of the presence of factor VIII inhibitors. Thrombin generation assays but not factor VIII activity are indicated to guide and monitor the treatment. Emicizumab has enabled the conversion of patients with severe forms into patients with milder forms of hemophilia A. It has reduced the number of bleeding episodes compared to both on-demand and prophylactic substitution therapy and has an excellent safety profile. Gene therapy can elevate factor VIII plasma levels for many years after a single treatment course, could offer long-term protection from bleeding episodes, and minimize or eliminate the need for substitutive treatment with factor VIII concentrates. Gene therapy can provoke an immune response, manifested by an increase in common liver enzymes, that require immunotherapy. Long term monitoring is necessary to identify possible adverse effects. Future objectives are: the development of an ideal viral vector, the possibility of its re-administration, the use of gene therapy in hemophiliac children, and determining whether it can be successfully used to induce immune tolerance to factor VIII ceteri paribus. The future will determine the place of each type of treatment and group of patients for which it is indicated.
Collapse
|
14
|
Abstract
In vivo gene therapy is rapidly emerging as a new therapeutic paradigm for monogenic disorders. For almost three decades, hemophilia A (HA) and hemophilia B (HB) have served as model disorders for the development of gene therapy. This effort is soon to bear fruit with completed pivotal adeno-associated viral (AAV) vector gene addition trials reporting encouraging results and regulatory approval widely anticipated in the near future for the current generation of HA and HB AAV vectors. Here we review the clinical development of AAV gene therapy for HA and HB and examine outstanding questions that have recently emerged from AAV clinical trials for hemophilia and other monogenic disorders.
Collapse
Affiliation(s)
- Benjamin J. Samelson-Jones
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Hematology and Raymond G. Perelman Center for Cellular and Molecular Therapeutics, the Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lindsey A. George
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Hematology and Raymond G. Perelman Center for Cellular and Molecular Therapeutics, the Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Luo L, Zheng Q, Chen Z, Huang M, Fu L, Hu J, Shi Q, Chen Y. Hemophilia a patients with inhibitors: Mechanistic insights and novel therapeutic implications. Front Immunol 2022; 13:1019275. [PMID: 36569839 PMCID: PMC9774473 DOI: 10.3389/fimmu.2022.1019275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022] Open
Abstract
The development of coagulation factor VIII (FVIII) inhibitory antibodies is a serious complication in hemophilia A (HA) patients after FVIII replacement therapy. Inhibitors render regular prophylaxis ineffective and increase the risk of morbidity and mortality. Immune tolerance induction (ITI) regimens have become the only clinically proven therapy for eradicating these inhibitors. However, this is a lengthy and costly strategy. For HA patients with high titer inhibitors, bypassing or new hemostatic agents must be used in clinical prophylaxis due to the ineffective ITI regimens. Since multiple genetic and environmental factors are involved in the pathogenesis of inhibitor generation, understanding the mechanisms by which inhibitors develop could help identify critical targets that can be exploited to prevent or eradicate inhibitors. In this review, we provide a comprehensive overview of the recent advances related to mechanistic insights into anti-FVIII antibody development and discuss novel therapeutic approaches for HA patients with inhibitors.
Collapse
Affiliation(s)
- Liping Luo
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qiaoyun Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Zhenyu Chen
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Medical Technology and Engineering College of Fujian Medical University, Fuzhou, Fujian, China
| | - Meijuan Huang
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianda Hu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qizhen Shi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti, Milwaukee, WI, United States
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, WI, United States
- Midwest Athletes Against Childhood Cancer (MACC) Fund Research Center, Milwaukee, WI, United States
| | - Yingyu Chen
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
16
|
Shao W, Sun J, Chen X, Dobbins A, Merricks EP, Samulski RJ, Nichols TC, Li C. Chimeric Mice Engrafted With Canine Hepatocytes Exhibits Similar AAV Transduction Efficiency to Hemophilia B Dog. Front Pharmacol 2022; 13:815317. [PMID: 35173619 PMCID: PMC8841897 DOI: 10.3389/fphar.2022.815317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated virus (AAV) mediated gene therapy has been successfully applied in clinical trials, including hemophilia. Novel AAV vectors have been developed with enhanced transduction and specific tissue tropism. Considering the difference in efficacy of AAV transduction between animal models and patients, the chimeric xenograft mouse model with human hepatocytes has unique advantages of studying AAV transduction efficiency in human hepatocytes. However, it is unclear whether the results in humanized mice can predict AAV transduction efficiency in human hepatocytes. To address this issue, we studied the AAV transduction efficacy in canine hepatocytes in both canine hepatocyte xenografted mice and real dogs. After administration of AAV vectors from different serotypes into canine hepatocyte xenograft mice, AAV8 induced the best canine hepatocyte transduction followed by AAV9, then AAV3, 7, 5 and 2. After administration of AAV/cFIX (cFIX-opt-R338L) vectors in hemophilia B dogs, consistent with the result in chimeric mice, AAV8 induced the highest cFIX protein expression and function, followed by AAV9 and then AAV2. These results suggest that mice xenografted with hepatocytes from different species could be used to predict the AAV liver transduction in real species and highlight this potential platform to explore novel AAV variants for future clinical applications.
Collapse
Affiliation(s)
- Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.,Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaojing Chen
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Amanda Dobbins
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Elizabeth P Merricks
- Department of Pathology and Laboratory Medicine and The Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Timothy C Nichols
- Department of Pathology and Laboratory Medicine and The Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
17
|
Chen N, Sun K, Chemuturi NV, Cho H, Xia CQ. The Perspective of DMPK on Recombinant Adeno-Associated Virus-Based Gene Therapy: Past Learning, Current Support, and Future Contribution. AAPS J 2022; 24:31. [PMID: 35102450 PMCID: PMC8817103 DOI: 10.1208/s12248-021-00678-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
Given the recent success of gene therapy modalities and the growing number of cell and gene-based therapies in clinical development across many different therapeutic areas, it is evident that this evolving field holds great promise for the unmet medical needs of patients. The recent approvals of Luxturna® and Zolgensma® prove that recombinant adeno-associated virus (rAAV)-based gene therapy is a transformative modality that enables curative treatment for genetic disorders. Over the last decade, Takeda has accumulated significant experience with rAAV-based gene therapies, especially in the early stage of development. In this review, based on the learnings from Takeda and publicly available information, we aim to provide a guiding perspective on Drug Metabolism and Pharmacokinetics (DMPK) substantial role in advancing therapeutic gene therapy modalities from nonclinical research to clinical development, in particular the characterization of gene therapy product biodistribution, elimination (shedding), immunogenicity assessment, multiple platform bioanalytical assays, and first-in-human (FIH) dose projection strategies. Graphical abstract ![]()
Collapse
Affiliation(s)
- Nancy Chen
- Takeda Development Center Americas, Inc. (TDCA), 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA.
| | - Kefeng Sun
- Takeda Development Center Americas, Inc. (TDCA), 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| | - Nagendra Venkata Chemuturi
- Takeda Development Center Americas, Inc. (TDCA), 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| | - Hyelim Cho
- Takeda Development Center Americas, Inc. (TDCA), 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| | - Cindy Q Xia
- Takeda Development Center Americas, Inc. (TDCA), 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
18
|
Braga LAM, Conte Filho CG, Mota FB. Future of genetic therapies for rare genetic diseases: what to expect for the next 15 years? THERAPEUTIC ADVANCES IN RARE DISEASE 2022; 3:26330040221100840. [PMID: 37180410 PMCID: PMC10032453 DOI: 10.1177/26330040221100840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/22/2022] [Indexed: 05/16/2023]
Abstract
Introduction Rare genetic diseases affect millions of people worldwide. Most of them are caused by defective genes that impair quality of life and can lead to premature death. As genetic therapies aim to fix or replace defective genes, they are considered the most promising treatment for rare genetic diseases. Yet, as these therapies are still under development, it is still unclear whether they will be successful in treating these diseases. This study aims to address this gap by assessing researchers' opinions on the future of genetic therapies for the treatment of rare genetic diseases. Methods We conducted a global cross-sectional web-based survey of researchers who recently authored peer-reviewed articles related to rare genetic diseases. Results We assessed the opinions of 1430 researchers with high and good knowledge about genetic therapies for the treatment of rare genetic diseases. Overall, the respondents believed that genetic therapies would be the standard of care for rare genetic diseases before 2036, leading to cures after this period. CRISPR-Cas9 was considered the most likely approach to fixing or replacing defective genes in the next 15 years. The respondents with good knowledge believed that genetic therapies would only have long-lasting effects after 2036, while those with high knowledge were divided on this issue. The respondents with good knowledge on the subject believed that non-viral vectors are more likely to be successful in fixing or replacing defective genes in the next 15 years, while most of the respondents with high knowledge believed viral vectors would be more successful. Conclusion Overall, the researchers who participated in this study expect that in the future genetic therapies will greatly benefit the treatment of patients with rare genetic diseases.
Collapse
Affiliation(s)
| | | | - Fabio Batista Mota
- Laboratory of Cellular Communication, Oswaldo
Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil, 4.365, Pavilhão 108,
Manguinhos, Rio de Janeiro RJ 21040-360, Brazil
| |
Collapse
|
19
|
Olgasi C, Borsotti C, Merlin S, Bergmann T, Bittorf P, Adewoye AB, Wragg N, Patterson K, Calabria A, Benedicenti F, Cucci A, Borchiellini A, Pollio B, Montini E, Mazzuca DM, Zierau M, Stolzing A, Toleikis P, Braspenning J, Follenzi A. Efficient and safe correction of hemophilia A by lentiviral vector-transduced BOECs in an implantable device. Mol Ther Methods Clin Dev 2021; 23:551-566. [PMID: 34853801 PMCID: PMC8606349 DOI: 10.1016/j.omtm.2021.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 11/18/2022]
Abstract
Hemophilia A (HA) is a rare bleeding disorder caused by deficiency/dysfunction of the FVIII protein. As current therapies based on frequent FVIII infusions are not a definitive cure, long-term expression of FVIII in endothelial cells through lentiviral vector (LV)-mediated gene transfer holds the promise of a one-time treatment. Thus, here we sought to determine whether LV-corrected blood outgrowth endothelial cells (BOECs) implanted through a prevascularized medical device (Cell Pouch) would rescue the bleeding phenotype of HA mice. To this end, BOECs from HA patients and healthy donors were isolated, expanded, and transduced with an LV carrying FVIII driven by an endothelial-specific promoter employing GMP-like procedures. FVIII-corrected HA BOECs were either directly transplanted into the peritoneal cavity or injected into a Cell Pouch implanted subcutaneously in NSG-HA mice. In both cases, FVIII secretion was sufficient to improve the mouse bleeding phenotype. Indeed, FVIII-corrected HA BOECs reached a relatively short-term clinically relevant engraftment being detected up to 16 weeks after transplantation, and their genomic integration profile did not show enrichment for oncogenes, confirming the process safety. Overall, this is the first preclinical study showing the safety and feasibility of transplantation of GMP-like produced LV-corrected BOECs within an implantable device for the long-term treatment of HA.
Collapse
Affiliation(s)
- Cristina Olgasi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Chiara Borsotti
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Simone Merlin
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Thorsten Bergmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97082 Würzburg, Germany
| | - Patrick Bittorf
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97082 Würzburg, Germany
| | - Adeolu Badi Adewoye
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, B15 2TT Birmingham, UK
| | - Nicholas Wragg
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST47QB Stoke-on-Trent, UK
| | | | | | | | - Alessia Cucci
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Alessandra Borchiellini
- Haematology Unit Regional Center for Hemorrhagic and Thrombotic Diseases, City of Health and Science University Hospital of Molinette, 10126 Turin, Italy
| | - Berardino Pollio
- Immune-Haematology and Transfusion Medicine, Regina Margherita Children Hospital, City of Health and Science University Hospital of Molinette, 10126 Turin, Italy
| | | | | | - Martin Zierau
- IMS Integrierte Management Systeme e. K., 64646 Heppenheim, Germany
| | - Alexandra Stolzing
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, LE113TU Loughborough, UK
- SENS Research Foundation, Mountain View, CA 94041, USA
| | | | - Joris Braspenning
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97082 Würzburg, Germany
| | - Antonia Follenzi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
20
|
Pipe SW, Gonen-Yaacovi G, Segurado OG. Hemophilia A Gene Therapy: Current and Next-Generation Approaches. Expert Opin Biol Ther 2021; 22:1099-1115. [PMID: 34781798 DOI: 10.1080/14712598.2022.2002842] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION : Hemophilia comprises a group of X-linked hemorrhagic disorders that result from a deficiency of coagulation factors. The disorder affects mainly males and leads to chronic pain, joint deformity, reduced mobility, and increased mortality. Current therapies require frequent administration of replacement clotting factors, but the emergence of alloantibodies (inhibitors) diminishes their efficacy. New therapies are being developed to produce the deficient clotting factors and prevent the emergence of inhibitors. AREAS COVERED : This article provides an update on the characteristics and disease pathophysiology of hemophilia A, as well as current treatments, with a special focus on ongoing clinical trials related to gene replacement therapies. EXPERT OPINION : Gene replacement therapies provide safe, durable, and stable transgene expression while avoiding the challenges of clotting factor replacement therapies in patients with hemophilia. Improving the specificity of the viral construct and decreasing the therapeutic dose are critical toward minimizing cellular stress, induction of the unfolded protein response, and the resulting loss of protein production in liver cells. Next-generation gene therapies incorporating chimeric DNA sequences in the transgene can increase clotting factor synthesis and secretion, and advance the efficacy, safety, and durability of gene replacement therapy for hemophilia A as well as other blood clotting disorders.
Collapse
|
21
|
Rapti K, Grimm D. Adeno-Associated Viruses (AAV) and Host Immunity - A Race Between the Hare and the Hedgehog. Front Immunol 2021; 12:753467. [PMID: 34777364 PMCID: PMC8586419 DOI: 10.3389/fimmu.2021.753467] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated viruses (AAV) have emerged as the lead vector in clinical trials and form the basis for several approved gene therapies for human diseases, mainly owing to their ability to sustain robust and long-term in vivo transgene expression, their amenability to genetic engineering of cargo and capsid, as well as their moderate toxicity and immunogenicity. Still, recent reports of fatalities in a clinical trial for a neuromuscular disease, although linked to an exceptionally high vector dose, have raised new caution about the safety of recombinant AAVs. Moreover, concerns linger about the presence of pre-existing anti-AAV antibodies in the human population, which precludes a significant percentage of patients from receiving, and benefitting from, AAV gene therapies. These concerns are exacerbated by observations of cellular immune responses and other adverse events, including detrimental off-target transgene expression in dorsal root ganglia. Here, we provide an update on our knowledge of the immunological and molecular race between AAV (the “hedgehog”) and its human host (the “hare”), together with a compendium of state-of-the-art technologies which provide an advantage to AAV and which, thus, promise safer and more broadly applicable AAV gene therapies in the future.
Collapse
Affiliation(s)
- Kleopatra Rapti
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany.,BioQuant Center, BQ0030, University of Heidelberg, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany.,BioQuant Center, BQ0030, University of Heidelberg, Heidelberg, Germany.,German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF) and German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Erkrankungen (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
22
|
Abstract
One approach to improve the utility of adeno-associated virus (AAV)-based gene therapy is to engineer the AAV capsid to 1) overcome poor transport through tissue barriers and 2) redirect the broadly tropic AAV to disease-relevant cell types. Peptide- or protein-domain insertions into AAV surface loops can achieve both engineering goals by introducing a new interaction surface on the AAV capsid. However, we understand little about the impact of insertions on capsid structure and the extent to which engineered inserts depend on a specific capsid context to function. Here, we examine insert-capsid interactions for the engineered variant AAV9-PHP.B. The 7-amino-acid peptide insert in AAV9-PHP.B facilitates transport across the murine blood-brain barrier via binding to the receptor Ly6a. When transferred to AAV1, the engineered peptide does not bind Ly6a. Comparative structural analysis of AAV1-PHP.B and AAV9-PHP.B revealed that the inserted 7-amino-acid loop is highly flexible and has remarkably little impact on the surrounding capsid conformation. Our work demonstrates that Ly6a binding requires interactions with both the PHP.B peptide and specific residues from the AAV9 HVR VIII region. An AAV1-based vector that incorporates a larger region of AAV9-PHP.B-including the 7-amino-acid loop and adjacent HVR VIII amino acids-can bind to Ly6a and localize to brain tissue. However, unlike AAV9-PHP.B, this AAV1-based vector does not penetrate the blood-brain barrier. Here we discuss the implications for AAV capsid engineering and the transfer of engineered activities between serotypes. Importance Targeting AAV vectors to specific cellular receptors is a promising strategy for enhancing expression in target cells or tissues while reducing off-target transgene expression. The AAV9-PHP.B/Ly6a interaction provides a model system with a robust biological readout that can be interrogated to better understand the biology of AAV vectors' interactions with target receptors. In this work, we analyzed the sequence and structural features required to successfully transfer the Ly6a receptor-binding epitope from AAV9-PHP.B to another capsid of clinical interest: AAV1. We found that AAV1- and AAV9-based vectors targeted to the same receptor exhibited different brain-transduction profiles. Our work suggests that, in addition to attachment-receptor binding, the capsid context in which this binding occurs is important for a vector's performance.
Collapse
|
23
|
Emerging Immunogenicity and Genotoxicity Considerations of Adeno-Associated Virus Vector Gene Therapy for Hemophilia. J Clin Med 2021; 10:jcm10112471. [PMID: 34199563 PMCID: PMC8199697 DOI: 10.3390/jcm10112471] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022] Open
Abstract
Adeno-associated viral (AAV) vector gene therapy has shown promise as a possible cure for hemophilia. However, immune responses directed against AAV vectors remain a hurdle to the broader use of this gene transfer platform. Both innate and adaptive immune responses can affect the safety and efficacy of AAV vector-mediated gene transfer in humans. These immune responses may be triggered by the viral capsid, the vector's nucleic acid payload, or other vector contaminants or excipients, or by the transgene product encoded by the vector itself. Various preclinical and clinical strategies have been explored to overcome the issues of AAV vector immunogenicity and transgene-related immune responses. Although results of these strategies are encouraging, more efficient approaches are needed to deliver safe, predictable, and durable outcomes for people with hemophilia. In addition to durability, long-term follow-up of gene therapy trial participants will allow us to address potential safety concerns related to vector integration. Herein, we describe the challenges with current methodologies to deliver optimal outcomes for people with hemophilia who choose to undergo AAV vector gene therapy and the potential opportunities to improve on the results.
Collapse
|
24
|
Sun H, Hodgkinson CP, Pratt RE, Dzau VJ. CRISPR/Cas9 Mediated Deletion of the Angiotensinogen Gene Reduces Hypertension: A Potential for Cure? Hypertension 2021; 77:1990-2000. [PMID: 33813849 PMCID: PMC9896968 DOI: 10.1161/hypertensionaha.120.16870] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/17/2021] [Indexed: 02/06/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hualing Sun
- Mandel Center and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710
| | - Conrad P. Hodgkinson
- Mandel Center and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710
| | - Richard E. Pratt
- Mandel Center and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710
| | - Victor J. Dzau
- Mandel Center and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
25
|
Samelson-Jones BJ, Guelcher C, Kuhn J, Butler R, Massey G, Guerrera MF, Raffini L. Real-world cost estimates of initiating emicizumab in US patients with haemophilia A. Haemophilia 2021; 27:591-598. [PMID: 34050689 DOI: 10.1111/hae.14347] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/21/2021] [Accepted: 05/08/2021] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Emicizumab is the first approved non-factor therapy for haemophilia A. It provides superior prophylactic bleeding control compared to other products in both patients with and patients without inhibitors. However, there is no real-world data about the monetary consequences of starting emicizumab. AIM To examine the estimated costs of starting emicizumab in a cohort of real-world haemophilia A patients with and without inhibitors. METHODS The cost of haemostatic therapy for 6 months before and after initiating emicizumab for participants in a multicentre observational study was calculated based on the type of product and dosing that was used for prophylaxis and treating breakthrough bleeds, the number of treated bleeds and the participant weight. RESULTS Ninety-two patients were included, 18 with an active inhibitor. The median age was 8.7 years. The median total cost for all patients decreased from $176,720 to $128,099 (p = .04) after initiating emicizumab, largely because of decrease in the total cost of high-cost outliers. The cost of prophylaxis and the total cost of bleeds also significantly decreased after starting emicizumab, both for patient with and patients without inhibitors. CONCLUSIONS Starting or switching to prophylaxis with emicizumab results in decreased costs for the treatment of patients with haemophilia A. This real-world data could inform on payer decisions as well as future cost-effective analysis.
Collapse
Affiliation(s)
| | - Christine Guelcher
- Children's National Hospital, George Washington University, Washington, DC, USA
| | - Jan Kuhn
- Virginia Commonwealth University, Richmond, VA, USA
| | - Regina Butler
- The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Gita Massey
- Virginia Commonwealth University, Richmond, VA, USA
| | - Michael F Guerrera
- Children's National Hospital, George Washington University, Washington, DC, USA
| | - Leslie Raffini
- The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
26
|
Abstract
Decades of preclinical and clinical studies developing gene therapy for hemophilia are poised to bear fruit with current promising pivotal studies likely to lead to regulatory approval. However, this recent success should not obscure the multiple challenges that were overcome to reach this destination. Gene therapy for hemophilia A and B benefited from advancements in the general gene therapy field, such as the development of adeno-associated viral vectors, as well as disease-specific breakthroughs, like the identification of B-domain deleted factor VIII and hyperactive factor IX Padua. The gene therapy field has also benefited from hemophilia B clinical studies, which revealed for the first time critical safety concerns related to immune responses to the vector capsid not anticipated in preclinical models. Preclinical studies have also investigated gene transfer approaches for other rare inherited bleeding disorders, including factor VII deficiency, von Willebrand disease, and Glanzmann thrombasthenia. Here we review the successful gene therapy journey for hemophilia and pose some unanswered questions. We then discuss the current state of gene therapy for these other rare inherited bleeding disorders and how the lessons of hemophilia gene therapy may guide clinical development.
Collapse
Affiliation(s)
- Valder R. Arruda
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania
| | - Jesse Weber
- Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin J. Samelson-Jones
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Lugin ML, Lee RT, Kwon YJ. Synthetically Engineered Adeno-Associated Virus for Efficient, Safe, and Versatile Gene Therapy Applications. ACS NANO 2020; 14:14262-14283. [PMID: 33073995 DOI: 10.1021/acsnano.0c03850] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Gene therapy directly targets mutations causing disease, allowing for a specific treatment at a molecular level. Adeno-associated virus (AAV) has been of increasing interest as a gene delivery vehicle, as AAV vectors are safe, effective, and capable of eliciting a relatively contained immune response. With the recent FDA approval of two AAV drugs for treating rare genetic diseases, AAV vectors are now on the market and are being further explored for other therapies. While showing promise in immune privileged tissue, the use of AAV for systemic delivery is still limited due to the high prevalence of neutralizing antibodies (nAbs). To avoid nAb-mediated inactivation, engineered AAV vectors with modified protein capsids, materials tethered to the capsid surface, or fully encapsulated in a second, larger carrier have been explored. Many of these engineered AAVs have added benefits, including avoided immune response, overcoming the genome size limit, targeted and stimuli-responsive delivery, and multimodal therapy of two or more therapeutic modalities in one platform. Native and engineered AAV vectors have been tested to treat a broad range of diseases, including spinal muscular atrophy, retinal diseases, cancers, and tissue damage. This review will cover the benefits of AAV as a promising gene vector by itself, the progress and advantages of engineered AAV vectors, particularly synthetically engineered ones, and the current state of their clinical translation in therapy.
Collapse
|
28
|
Arruda VR, Doshi BS. Gene Therapy for Hemophilia: Facts and Quandaries in the 21st Century. Mediterr J Hematol Infect Dis 2020; 12:e2020069. [PMID: 32952980 PMCID: PMC7485465 DOI: 10.4084/mjhid.2020.069] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023] Open
Abstract
Therapy for hemophilia has evolved in the last 40 years from plasma-based concentrates to recombinant proteins and, more recently, to non-factor therapeutics. Along this same timeline, research in adeno-associated viral (AAV) based gene therapy vectors has provided the framework for early phase clinical trials initially for hemophilia B (HB) and now for hemophilia A. Successive lessons learned from early HB trials have paved the way for current advanced phase trials. Nevertheless, questions linger regarding 1) the optimal balance of vector dose to transgene expression, 2) amount and durability of transgene expression required, and 3) long-term safety. Some trials have demonstrated unique findings not seen previously regarding transient elevation of liver enzymes, immunogenicity of the vector capsid, and loss of transgene expression. This review will provide an update on the clinical AAV gene therapy trials in hemophilia and address the questions above. A thoughtful and rationally approached expansion of gene therapy to the clinics would certainly be a welcome addition to the arsenal of options for hemophilia therapy. Further, the global impact of gene therapy could be vastly improved by expanding eligibility to different patient populations and to developing nations. With the advances made to date, it is possible to envision a shift from the early goal of simply increasing life expectancy to a significant improvement in quality of life by reduction in spontaneous bleeding episodes and disease complications.
Collapse
Affiliation(s)
- Valder R. Arruda
- Divsion of Hematology, Children’s Hospital of Philadelphia, Philadelphia PA USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia PA USA
| | - Bhavya S. Doshi
- Divsion of Hematology, Children’s Hospital of Philadelphia, Philadelphia PA USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA USA
| |
Collapse
|