1
|
Li W, Sun J, Feng S, La Rosa A, Zhang P, Wu EY, Loeser R, Li C. Secreted PD-L1 alleviates inflammatory arthritis in mice through local and systemic AAV gene therapy. Front Immunol 2025; 16:1527858. [PMID: 39963137 PMCID: PMC11830590 DOI: 10.3389/fimmu.2025.1527858] [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: 11/13/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) primarily affects the joints but can also affect multiple organs and profoundly impacts patients' ability to carry out daily activities, mental health, and life expectancy. Current treatments for RA are limited in terms of duration, efficacy, and adverse effects. PD-L1 is a checkpoint protein that plays important roles in immune regulation and has been implicated in the initiation and progression of multiple autoimmune diseases. Method In a previous study, we demonstrated that intra-articular injection with adeno-associated virus (AAV) vectors encoding wild type PD-L1 improved local inflammation in the joint in the collagen-induced arthritis (CIA) mouse model of RA. To further improve efficacy, we explored AAV-mediated delivery of the soluble PD-L1 (sPD-L1) to CIA mice. Result After intra-articular injection of AAV6 vectors expressing the optimal isoform of sPD-L1 (shPD-L1), more potency was observed when compared to wild type PD-L1, with a lower dose of AAV6/shPD-L1 needed for arthritis improvement. To study the therapeutic effect of systemic expression of sPD-L1, we administered AAV8/shPD-L1 gene therapy in CIA mice via retro-orbital injection and found significant improvements in joint inflammation and paw swelling, exhibiting similar phenotypes to that in naïve mice. The levels of total immunoglobulin and anti-collagen specific antibodies were lower in AAV8/shPD-L1 treated CIA mice than those in controls. The levels of pro-inflammatory cytokines in blood were also significantly decreased in shPD-L1 treated mice. Additionally, T cell apoptosis rates in the spleen showed a 2-fold increase in treated mice. Finally, we investigated the therapeutic effect of AAV/shPD-L1 via intramuscular injection. After injection of AAV6/shPD-L1, decreased paw swelling, reduced joint inflammation, and lower levels of pro-inflammatory cytokines in blood were achieved. The therapeutic effect of shPD-L1 was dose dependent via intramuscular treatment with AAV vectors. Conclusion In conclusion, the findings in this study suggest that intra-articular injection of AAV vectors encoding sPD-L1 results in greater therapeutic benefit on arthritis, and systemic AAV/sPD-L1 is able to block the development of inflammatory arthritis with inhibition of the systemic immune response, underlining the potential of gene therapy with systemic delivery of shPD-L1 via AAV vectors in RA.
Collapse
Affiliation(s)
- Wenjun Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susi Feng
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ariana La Rosa
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Panli Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eveline Y. Wu
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Richard Loeser
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, 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
|
2
|
Defois A, Bon N, Mével M, Deniaud D, Maugars Y, Guicheux J, Adjali O, Vinatier C. Gene therapies for osteoarthritis: progress and prospects. JOURNAL OF CARTILAGE & JOINT PRESERVATION 2024; 4:100186. [DOI: 10.1016/j.jcjp.2024.100186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Li W, Sun J, Feng SL, Wang F, Miao MZ, Wu EY, Wallet S, Loeser R, Li C. Intra-articular delivery of AAV vectors encoding PD-L1 attenuates joint inflammation and tissue damage in a mouse model of rheumatoid arthritis. Front Immunol 2023; 14:1116084. [PMID: 36936967 PMCID: PMC10021025 DOI: 10.3389/fimmu.2023.1116084] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Objective Rheumatoid arthritis (RA) is the most common form of autoimmune inflammatory arthritis. Intra-articular gene delivery to block proinflammatory cytokines has been studied in pre-clinical models and human clinical trials. It has been demonstrated that the level of programmed death-ligand 1 (PD-L1) is associated with rheumatoid arthritis (RA). This study examined the therapeutic role of PD-L1 by intra-articular delivery via adeno-associated virus (AAV) vectors in the mouse collagen-induced arthritis (CIA) model. Methods Mice were intra-articularly injected with AAV5 vectors encoding human PD-L1 on day 0 and immunized with bovine type II collagen to induce CIA simultaneously. On day 49 post AAV administration, joints were collected for histo-pathological and cytokine analysis. Additionally, the systemic impacts of intra-articular injection of AAV5/PD-L1 vectors were also studied. To study the therapeutic effect of PD-L1, AAV5/PD-L1 vectors were administered into the joints of RA mice on day 21. Results After administration of AAV5/PD-L1 vectors, strong PD-L1 expression was detected in AAV transduced joints. Joints treated with PD-L1 at the time of arthritis induction exhibited significantly less swelling and improved histopathological scores when compared to untreated joints. Additionally, the infiltration of T cells and macrophages was decreased in joints of CIA mice that received AAV5/PD-L1 vectors (P<0.05). The levels of pro-inflammatory cytokines, including IL-1, IL-6, IL-17 and TNFα, were lower in AAV5/PD-L1 treated than untreated joints (P<0.05). Furthermore, the administration of AAV5/PD-L1 vectors into the joints of CIA mice did not impact serum cytokine levels and the antibody titers to type II collagen. Biodistribution of AAV vectors after intra-articular injection showed undetectable AAV genomes in other tissues except for a low level in the liver. Similar to the results of AAV5/PD-L1 vector administration on day 0, decreased joint swelling and lower histopathological damage were observed in joints treated with AAV5/PD-L1 vectors on day 21. Conclusion The results from this study demonstrate that local AAV mediated PD-L1 gene delivery into the joints is able to prevent the development and block the progression of arthritis in CIA mice without impacting systemic immune responses. This study provides a novel strategy to effectively treat inflammatory joint diseases using local AAV gene therapy by interference with immune checkpoint pathways.
Collapse
Affiliation(s)
- Wenjun Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Junjiang Sun
- Division of Chemical Biology and Medicinal Chemistry, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susi Liu Feng
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Feng Wang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael Z. Miao
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eveline Y. Wu
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shannon Wallet
- Department of oral biology, University of Florida, Gainesville, FL, United States
| | - Richard Loeser
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, 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
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
4
|
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
|
5
|
Vrouwe JPM, Meulenberg JJM, Klarenbeek NB, Navas-Cañete A, Reijnierse M, Ruiterkamp G, Bevaart L, Lamers RJ, Kloppenburg M, Nelissen RGHH, Huizinga TWJ, Burggraaf J, Kamerling IMC. Administration of an adeno-associated viral vector expressing interferon-β in patients with inflammatory hand arthritis, results of a phase I/II study. Osteoarthritis Cartilage 2022; 30:52-60. [PMID: 34626797 DOI: 10.1016/j.joca.2021.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/17/2021] [Accepted: 09/30/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Inflammatory hand arthritis (IHA) results in impaired function. Local gene therapy with ART-I02, a recombinant adeno-associated virus (AAV) serotype 5 vector expressing interferon (IFN)-β, under the transcriptional control of nuclear factor κ-B responsive promoter, was preclinically shown to have favorable effects. This study aimed to investigate the safety and tolerability of local gene therapy with ART-I02 in patients with IHA. METHODS In this first-in-human, dose-escalating, cohort study, 12 IHA patients were to receive a single intra-articular (IA) injection of ART-I02 ranging 0.3 × 1012-1.2 × 1013 genome copies in an affected hand joint. Adverse events (AEs), routine safety laboratory and the clinical course of disease were periodically evaluated. Baseline- and follow-up contrast enhanced magnetic resonance images (MRIs), shedding of viral vectors in bodily fluids, and AAV5 and IFN-β immune responses were evaluated. A data review committee provided safety recommendations. RESULTS Four patients were enrolled. Long-lasting local AEs were observed in 3 patients upon IA injection of ART-I02. The AEs were moderate in severity and could be treated conservative. Given the duration of the AEs and their possible or probable relation to ART-I02, no additional patients were enrolled. No systemic treatment emergent AEs were observed. The MRIs reflected the AEs by (peri)arthritis. No T-cell response against AAV5 or IFN-β, nor IFN-β antibodies could be detected. Neutralizing antibody titers against AAV5 raised post-dose. CONCLUSION Single IA doses of 0.6 × 1012 or 1.2 × 1012 ART-I02 vector genomes were administered without systemic side effects or serious AEs. However, local tolerability was insufficient for continuation. TRIAL REGISTRATION NCT02727764.
Collapse
Affiliation(s)
- J P M Vrouwe
- Centre for Human Drug Research, Zernikedreef 8, Leiden, 2333 CL, the Netherlands; Leiden University Medical Center (LUMC), Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - J J M Meulenberg
- Department of Oncology, Arthrogen B.V., Meibergdreef 45, Amsterdam, 1005BA, the Netherlands
| | - N B Klarenbeek
- Centre for Human Drug Research, Zernikedreef 8, Leiden, 2333 CL, the Netherlands; Leiden University Medical Center, Department of Internal Medicine, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - A Navas-Cañete
- Leiden University Medical Center, Department of Radiology, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands.
| | - M Reijnierse
- Leiden University Medical Center, Department of Radiology, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - G Ruiterkamp
- Department of Oncology, Arthrogen B.V., Meibergdreef 45, Amsterdam, 1005BA, the Netherlands
| | - L Bevaart
- Department of Oncology, Arthrogen B.V., Meibergdreef 45, Amsterdam, 1005BA, the Netherlands
| | - R J Lamers
- Department of Oncology, Arthrogen B.V., Meibergdreef 45, Amsterdam, 1005BA, the Netherlands
| | - M Kloppenburg
- Leiden University Medical Center, Department of Rheumatology, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - R G H H Nelissen
- Leiden University Medical Center, Department of Orthopaedics, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - T W J Huizinga
- Leiden Academic Centre for Drug Research, PO box 9500, Leiden, 2300 RA, the Netherlands
| | - J Burggraaf
- Centre for Human Drug Research, Zernikedreef 8, Leiden, 2333 CL, the Netherlands; Leiden University Medical Center, Department of Internal Medicine, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands; Leiden Academic Centre for Drug Research, PO box 9500, Leiden, 2300 RA, the Netherlands
| | - I M C Kamerling
- Centre for Human Drug Research, Zernikedreef 8, Leiden, 2333 CL, the Netherlands; Leiden University Medical Center, Department of Infectious Diseases, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands.
| |
Collapse
|
6
|
Candidates for Intra-Articular Administration Therapeutics and Therapies of Osteoarthritis. Int J Mol Sci 2021; 22:ijms22073594. [PMID: 33808364 PMCID: PMC8036705 DOI: 10.3390/ijms22073594] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) of the knee is a disease that significantly decreases the quality of life due to joint deformation and pain caused by degeneration of articular cartilage. Since the degeneration of cartilage is irreversible, intervention from an early stage and control throughout life is important for OA treatment. For the treatment of early OA, the development of a disease-modifying osteoarthritis drug (DMOAD) for intra-articular (IA) injection, which is attracting attention as a point-of-care therapy, is desired. In recent years, the molecular mechanisms involved in OA progression have been clarified while new types of drug development methods based on gene sequences have been established. In addition to conventional chemical compounds and protein therapeutics, the development of DMOAD from the new modalities such as gene therapy and oligonucleotide therapeutics is accelerating. In this review, we have summarized the current status and challenges of DMOAD for IA injection, especially for protein therapeutics, gene therapy, and oligonucleotide therapeutics.
Collapse
|
7
|
Zhang F, Yan X, Li M, Hua B, Xiao X, Monahan PE, Sun J. Exploring the Potential Feasibility of Intra-Articular Adeno-Associated Virus-Mediated Gene Therapy for Hemophilia Arthropathy. Hum Gene Ther 2020; 31:448-458. [PMID: 32079420 DOI: 10.1089/hum.2019.355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hemophilia arthropathy (HA) represents the majority of morbidity in severe hemophilia patients, especially in resource-limited countries. Adeno-associated virus (AAV)-mediated gene therapy is showing promise for managing hemophilia. However, patients with neutralizing antibodies (NAbs) against AAV, and inhibitors to clotting factors, are excluded from such therapy. This study explored the feasibility of AAV-mediated local gene therapy for HA. Factor VIII knockout (FVIII-/-) mice, with or without a FVIII inhibitor, were subjected to hemarthrosis induction and treated with either intravenous (IV) or intraarticular (IA) recombinant human factor VIII (rhFVIII). To investigate whether rhFVIII carried the risk to develop a FVIII inhibitor, FVIII-/- mice were treated with three doses of IV or IA rhFVIII and inhibitor development was measured. In patients with established HA requiring synovial fluid aspiration, plasma, and synovial fluid were collected and measured for anti-AAV capsid IgG (serotypes 1-9 and 843) and NAbs for AAV843. IA rhFVIII provided better protection from synovitis compared with IV rhFVIII, with or without the FVIII inhibitor. While IV rhFVIII led to all FVIII-/- mice developing an FVIII inhibitor (n = 31, median 4.9 Bethesda units [BU]/mL), only 50% of the mice developed a FVIII inhibitor by IA administration, and at a lower titer (median 0.55 BU/mL). In hemophilia patients, total anti-AAV IgG was lowest for AAV4 and AAV5, both in plasma and synovial fluid. Anti-AAV IgGs in synovial fluid for most samples were lower or similar to the plasma levels. These results show that direct IA rhFVIII administration yields better protection against bleeding-induced joint damage, even in the presence of an inhibitor antibody. IA rhFVIII delivery carried a lower risk of FVIII inhibitor formation compared with IV FVIII. The anti-AAV antibody level in synovial fluid was similar or lower than the plasma level, supporting the feasibility of local gene therapy for managing HA.
Collapse
Affiliation(s)
- Feixu Zhang
- School of Bioengineering, East China University of Science and Technology, Shanghai, China.,School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiaobo Yan
- Department of Hematology, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Min Li
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Baolai Hua
- Department of Hematology, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xiao Xiao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina.,Harold R. Roberts Comprehensive Hemophilia Diagnosis and Treatment Center, University of North Carolina, Chapel Hill, North Carolina.,Spark Therapeutics, Philadelphia, Pennsylvania
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina.,Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
8
|
Moss KL, Jiang Z, Dodson ME, Linardi RL, Haughan J, Gale AL, Grzybowski C, Engiles JE, Stefanovski D, Robinson MA, Ortved KF. Sustained Interleukin-10 Transgene Expression Following Intra-Articular AAV5-IL-10 Administration to Horses. Hum Gene Ther 2019; 31:110-118. [PMID: 31773987 DOI: 10.1089/hum.2019.195] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Joint trauma leads to post-traumatic inflammation with upregulation of inflammatory cytokines and degradative enzymes. If severe enough, this response can lead to irreversible post-traumatic osteoarthritis. Interleukin-10 (IL-10), a cytokine with potent anti-inflammatory effects, has been shown to have chondroprotective effects. A gene therapy approach using a vector to overexpress IL-10 in the joint represents a feasible method of delivering sustained high doses of IL-10 to post-traumatic joints. We hypothesized that an AAV5 vector overexpressing IL-10 would result in rapid and sustained IL-10 expression following direct intra-articular injection and that this increase would not be reflected in systemic circulation. In addition, we hypothesized that intra-articular AAV5-IL-10 injection would not induce a local inflammatory response. Twelve horses were assigned to either treatment (AAV5-IL-10-injected) or control (PBS-injected) groups. Middle carpal joints were injected with 1012 vector genomes/joint or phosphate-buffered saline (PBS) alone (3 mL). Serial synovial fluid samples were analyzed for inflammatory changes, IL-10 concentration, and vector genome copy number. Serum samples were also analyzed for IL-10 concentration and vector genome copy number. Synovial membrane was collected on day 84. Synovial fluid IL-10 was significantly increased within 48 h of AAV5-IL-10 injection and remained increased, compared to PBS-injected joints, until day 84. Serum IL-10 was not different between groups. Vector administration did not cause a significant synovial inflammatory response. Vector genomes were detectable in the plasma, synovial fluid, and synovial membrane of AAV5-IL-10-injected horses only. IL-10 has the potential to modulate the articular inflammatory response, thereby protecting cartilage from degradation and osteoarthritis. This study demonstrates the feasibility and efficiency of intra-articular AAV5-IL-10, and future studies investigating the chondroprotective effects of IL-10 in inflamed joints in vivo are warranted.
Collapse
Affiliation(s)
- Kaitlyn L Moss
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Zibin Jiang
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Michael E Dodson
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Renata L Linardi
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Joanne Haughan
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Alexis L Gale
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Cara Grzybowski
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Julie E Engiles
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Darko Stefanovski
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Mary A Robinson
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania.,Pennsylvania Equine Toxicology & Research Center, West Chester University, West Chester, Pennsylvania
| | - Kyla F Ortved
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| |
Collapse
|
9
|
Affiliation(s)
- Christopher H Evans
- Departments of Physical Medicine and Rehabilitation, Orthopaedic Surgery, Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
10
|
Watson Levings RS, Broome TA, Smith AD, Rice BL, Gibbs EP, Myara DA, Hyddmark EV, Nasri E, Zarezadeh A, Levings PP, Lu Y, White ME, Dacanay EA, Foremny GB, Evans CH, Morton AJ, Winter M, Dark MJ, Nickerson DM, Colahan PT, Ghivizzani SC. Gene Therapy for Osteoarthritis: Pharmacokinetics of Intra-Articular Self-Complementary Adeno-Associated Virus Interleukin-1 Receptor Antagonist Delivery in an Equine Model. HUM GENE THER CL DEV 2019; 29:90-100. [PMID: 29869540 DOI: 10.1089/humc.2017.142] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Toward the treatment of osteoarthritis (OA), the authors have been investigating self-complementary adeno-associated virus (scAAV) for intra-articular delivery of therapeutic gene products. As OA frequently affects weight-bearing joints, pharmacokinetic studies of scAAV gene delivery were performed in the joints of the equine forelimb to identify parameters relevant to clinical translation in humans. Using interleukin-1 receptor antagonist (IL-1Ra) as a secreted therapeutic reporter, scAAV vector plasmids containing codon-optimized cDNA for equine IL-1Ra (eqIL-1Ra) were generated, which produced eqIL-1Ra at levels 30- to 50-fold higher than the native sequence. The most efficient cDNA was packaged in AAV2.5 capsid, and following characterization in vitro, the virus was injected into the carpal and metacarpophalangeal joints of horses over a 100-fold dose range. A putative ceiling dose of 5 × 1012 viral genomes was identified that elevated the steady-state eqIL-1Ra in the synovial fluids of injected joints by >40-fold over endogenous levels and was sustained for at least 6 months. No adverse effects were seen, and eqIL-1Ra in serum and urine remained at background levels throughout. Using the 5 × 1012 viral genome dose of scAAV, and green fluorescent protein as a cytologic marker, the local and systemic distribution of vector and transduced cells following intra-articular injection scAAV.GFP were compared in healthy equine joints and in those with late-stage, naturally occurring OA. In both cases, 99.7% of the vector remained within the injected joint. Strikingly, the pathologies characteristic of OA (synovitis, osteophyte formation, and cartilage erosion) were associated with a substantial increase in transgenic expression relative to tissues in healthy joints. This was most notable in regions of articular cartilage with visible damage, where foci of brilliantly fluorescent chondrocytes were observed. Overall, these data suggest that AAV-mediated gene transfer can provide relatively safe, sustained protein drug delivery to joints of human proportions.
Collapse
Affiliation(s)
| | - Ted A Broome
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Andrew D Smith
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Brett L Rice
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Eric P Gibbs
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - David A Myara
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - E Viktoria Hyddmark
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Elham Nasri
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Ali Zarezadeh
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Padraic P Levings
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Yuan Lu
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Margaret E White
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - E Anthony Dacanay
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Gregory B Foremny
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| | - Christopher H Evans
- 3 Rehabilitation Medicine Research Center, Mayo Clinic , Rochester, Minnesota
| | - Alison J Morton
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Mathew Winter
- 4 Department of Small Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Michael J Dark
- 5 Department of Infectious Diseases and Pathology, University of Florida , Gainesville, Florida
| | - David M Nickerson
- 6 Department of Statistics and Actuarial Science, University of Central Florida , Orlando, Florida
| | - Patrick T Colahan
- 2 Department of Large Animal Clinical Sciences, University of Florida , Gainesville, Florida
| | - Steven C Ghivizzani
- 1 Department of Orthopedics and Rehabilitation, University of Florida , Gainesville, Florida
| |
Collapse
|
11
|
Evans CH, Ghivizzani SC, Robbins PD. Gene Delivery to Joints by Intra-Articular Injection. Hum Gene Ther 2019; 29:2-14. [PMID: 29160173 DOI: 10.1089/hum.2017.181] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Most forms of arthritis are incurable, difficult to treat, and a major cause of disability in Western countries. Better local treatment of arthritis is impaired by the pharmacokinetics of the joint that make it very difficult to deliver drugs to joints at sustained, therapeutic concentrations. This is especially true of biologic drugs, such as proteins and RNA, many of which show great promise in preclinical studies. Gene transfer provides a strategy for overcoming this limitation. The basic concept is to deliver cDNAs encoding therapeutic products by direct intra-articular injection, leading to sustained, endogenous synthesis of the gene products within the joint. Proof of concept has been achieved for both in vivo and ex vivo gene delivery using a variety of vectors, genes, and cells in several different animal models. There have been a small number of clinical trials for rheumatoid arthritis (RA) and osteoarthritis (OA) using retrovirus vectors for ex vivo gene delivery and adeno-associated virus (AAV) for in vivo delivery. AAV is of particular interest because, unlike other viral vectors, it is able to penetrate deep within articular cartilage and transduce chondrocytes in situ. This property is of particular importance in OA, where changes in chondrocyte metabolism are thought to be fundamental to the pathophysiology of the disease. Authorities in Korea have recently approved the world's first arthritis gene therapy. This targets OA by the injection of allogeneic chondrocytes that have been transduced with a retrovirus carrying transforming growth factor-β1 cDNA. Phase III studies are scheduled to start in the United States soon. Meanwhile, two additional Phase I trials are listed on Clinicaltrials.gov , both using AAV. One targets RA by transferring interferon-β, and the other targets OA by transferring interleukin-1 receptor antagonist. The field is thus gaining momentum and promises to improve the treatment of these common and debilitating diseases.
Collapse
Affiliation(s)
- Christopher H Evans
- 1 Rehabilitation Medicine Research Center, Mayo Clinic , Rochester, Minnesota
| | - Steven C Ghivizzani
- 2 Department of Orthopedics and Rehabilitation, University of Florida College of Medicine , Gainesville, Florida
| | - Paul D Robbins
- 3 Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, Florida
| |
Collapse
|
12
|
Kyostio-Moore S, Berthelette P, Cornell CS, Nambiar B, Figueiredo MD. Hyaluronic acid synthase-2 gene transfer into the joints of Beagles by use of recombinant adeno-associated viral vectors. Am J Vet Res 2018; 79:505-517. [DOI: 10.2460/ajvr.79.5.505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Grol MW, Lee BH. Gene therapy for repair and regeneration of bone and cartilage. Curr Opin Pharmacol 2018; 40:59-66. [PMID: 29621661 DOI: 10.1016/j.coph.2018.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/12/2018] [Indexed: 12/28/2022]
Abstract
Gene therapy refers to the use of viral and non-viral vectors to deliver nucleic acids to tissues of interest using direct (in vivo) or transduced cell-mediated (ex vivo) approaches. Over the past few decades, strategies have been adopted to express therapeutic transgenes at sites of injury to promote or facilitate repair of bone and cartilage. Targets of interest have typically included secreted proteins such as growth factors and anti-inflammatory mediators; however, work has also begun to focus intracellularly on signaling components, transcription factors and small, regulatory nucleic acids such as microRNAs (miRNAs). In recent years, a number of single therapeutic gene approaches (termed 'monotherapies') have proven effective in preclinical models of disease, and several are being evaluated in clinical trials. In particular, an ex vivo TGF-β1 gene therapy was approved in Korea in 2017 for treatment of moderate-to-severe osteoarthritis (OA). The ability to utilize viral vectors for context-specific and combinatorial gene therapy is also being investigated, and these strategies are likely to be important in more robustly addressing the complexities of tissue repair and regeneration in skeletal disease. In this review, we provide an overview of viral gene therapies being developed for treatment of bone and cartilage pathologies, with an emphasis on emerging combinatorial strategies as well as those targeting intracellular mediators such as miRNAs.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
14
|
Zhang C, Zhou X, Yao T, Tian Z, Zhou D. Precision Fluorescent Labeling of an Adeno-Associated Virus Vector to Monitor the Viral Infection Pathway. Biotechnol J 2018; 13:e1700374. [DOI: 10.1002/biot.201700374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 12/04/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Chuanling Zhang
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Xueying Zhou
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Tianzhuo Yao
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Zhenyu Tian
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| |
Collapse
|
15
|
Bellavia D, Veronesi F, Carina V, Costa V, Raimondi L, De Luca A, Alessandro R, Fini M, Giavaresi G. Gene therapy for chondral and osteochondral regeneration: is the future now? Cell Mol Life Sci 2018; 75:649-667. [PMID: 28864934 PMCID: PMC11105387 DOI: 10.1007/s00018-017-2637-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022]
Abstract
Gene therapy might represent a promising strategy for chondral and osteochondral defects repair by balancing the management of temporary joint mechanical incompetence with altered metabolic and inflammatory homeostasis. This review analysed preclinical and clinical studies on gene therapy for the repair of articular cartilage defects performed over the last 10 years, focussing on expression vectors (non-viral and viral), type of genes delivered and gene therapy procedures (direct or indirect). Plasmids (non-viral expression vectors) and adenovirus (viral vectors) were the most employed vectors in preclinical studies. Genes delivered encoded mainly for growth factors, followed by transcription factors, anti-inflammatory cytokines and, less frequently, by cell signalling proteins, matrix proteins and receptors. Direct injection of the expression vector was used less than indirect injection of cells, with or without scaffolds, transduced with genes of interest and then implanted into the lesion site. Clinical trials (phases I, II or III) on safety, biological activity, efficacy, toxicity or bio-distribution employed adenovirus viral vectors to deliver growth factors or anti-inflammatory cytokines, for the treatment of osteoarthritis or degenerative arthritis, and tumour necrosis factor receptor or interferon for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Daniele Bellavia
- Rizzoli Orthopedic Institute, Bologna, Italy.
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy.
| | - F Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - V Carina
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - V Costa
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - L Raimondi
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - A De Luca
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - R Alessandro
- Biology and Genetics Unit, Department of Biopathology and Medical Biotechnology, University of Palermo, Palermo, Italy
| | - M Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - G Giavaresi
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
16
|
Ru Q, Li W, Wang X, Zhang S, Chen L, Zhang Y, Ge Y, Zu Y, Liu Y, Zheng D. Preclinical study of rAAV2-sTRAIL: pharmaceutical efficacy, biodistribution and safety in animals. Cancer Gene Ther 2017; 24:251-258. [PMID: 28429751 DOI: 10.1038/cgt.2017.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 02/02/2017] [Accepted: 03/07/2017] [Indexed: 12/31/2022]
Abstract
The recombinant sTRAIL has been in clinical trial for various human malignancies. However, the half-life time of sTRAIL is very short, which might be an important factor influencing its clinical efficacy for cancer therapy. We previously reported the recombinant adeno-associated virus (AAV)-encoding sTRAIL95-281-mediated sTRAIL expression in vivo up to 8 months and suppressed tumor growth markedly in mouse xenografts. In the present study, we further evaluated the clinical potency for cancer gene therapy and the safety in mouse and non-human primates. The mouse models with HCT-116, NCI-H460 and BEL-7402 cancers were injected intraperitoneally with a single dose of 1.0 × 1011, 1.0 × 1010 and 1.0 × 109 vg of rAAV2-sTRAIL95-281 virus, respectively. The cynomolgus monkeys were injected (i.m.) with a single dose of rAAV2-sTRAIL95-281 of 1 × 1011, 3 × 1011 and 1 × 1012 vg, corresponding to 6-, 20- and 60-fold of intended use dosage for humans, respectively. The efficacy, pharmacology and toxicity of rAAV-sTRAIL in the animals were analyzed accordingly. The tumor inhibitory rates reached 44-76%, 48-52% and 55-74% in the three tumor models, respectively, and they had no influence on mouse spontaneous activity. Administration (s.c.) of a single dose of rAAV2-sTRAIL95-281 virus of 1.0 × 109 or 1.0 × 1010 vg in mice with implanted tumor led to mainly distribution in the spleen, liver, implanted tumor, blood, injected site of muscle and bone marrow. Two weeks later, there was no rAAV2-sTRAIL95-281 detected in blood and bone marrow, and it significantly decreased in other tissues and organs and then gradually cleared away in 4-12 weeks after administration. There was no rAAV2-sTRAIL accumulation in the animal's body and no influence on the body weights. Administration (i.v.) did not cause animal death, and no dose-related abnormal clinical symptoms were found in the mice. There were no abnormal tissue and organ found in all animals. Long-term toxicity test in cynomolgus monkeys did not cause rAAV2-sTRAIL95-281-related toxic and side effects, except that anti-AAV and anti-sTRAIL antibodies were generated. In conclusion, these data demonstrated that administration of rAAV2-sTRAIL95-281 in mice and in cynomolgus monkeys is safe without obvious toxic and side effects to the animals, and throw light on pharmacokinetics and safety in human clinical trials for cancer gene therapy.
Collapse
Affiliation(s)
- Q Ru
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - W Li
- National Institutes for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing, China
| | - X Wang
- National Institutes for Food and Drug Control, National Center for Safety Evaluation of Drugs, Beijing, China
| | - S Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - L Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Y Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Y Ge
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Y Zu
- Obio Technology (Shanghai) Corp. Ltd, Shanghai, China
| | - Y Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - D Zheng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Cucchiarini M. New cell engineering approaches for cartilage regenerative medicine. Biomed Mater Eng 2017; 28:S201-S207. [DOI: 10.3233/bme-171642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, D-66421 Homburg/Saar, Germany
| |
Collapse
|
18
|
Rey-Rico A, Cucchiarini M. Recent tissue engineering-based advances for effective rAAV-mediated gene transfer in the musculoskeletal system. Bioengineered 2017; 7:175-88. [PMID: 27221233 DOI: 10.1080/21655979.2016.1187347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Musculoskeletal tissues are diverse and significantly different in their ability to repair upon injury. Current treatments often fail to reproduce the natural functions of the native tissue, leading to an imperfect healing. Gene therapy might improve the repair of tissues by providing a temporarily and spatially defined expression of the therapeutic gene(s) at the site of the injury. Several gene transfer vehicles have been developed to modify various human cells and tissues from musculoskeletal system among which the non-pathogenic, effective, and relatively safe recombinant adeno-associated viral (rAAV) vectors that have emerged as the preferred gene delivery system to treat human disorders. Adapting tissue engineering platforms to gene transfer approaches mediated by rAAV vectors is an attractive tool to circumvent both the limitations of the current therapeutic options to promote an effective healing of the tissue and the natural obstacles from these clinically adapted vectors to achieve an efficient and durable gene expression of the therapeutic sequences within the lesions.
Collapse
Affiliation(s)
- Ana Rey-Rico
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany
| | - Magali Cucchiarini
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany
| |
Collapse
|