Structured Abstract
Purpose of Review:
To assess the present status of gene therapy for osteoarthritis (OA).
Recent Findings:
An expanding list of cDNAs show therapeutic activity when introduced into the joints of animals with experimental models of OA. In vivo delivery with adenovirus or adeno-associated virus is most commonly used for this purpose. The list of encoded products includes cytokines, cytokine antagonists, enzymes, enzyme inhibitors, growth factors and non-coding RNA. Elements of CRISPR-Cas have also been delivered to mouse knees to ablate key genes. Several human trials have been initiated, using transgenes encoding transforming growth factor-β, interleukin-1 receptor antagonist, interferon-β, the NKX3.2 transcription factor or variant interleukin-10. The first of these, using ex vivo delivery with allogeneic chondrocytes, gained approval in Korea which was subsequently retracted. However, it is undergoing Phase III clinical trials in the US. The other trials are in Phase I or II. No gene therapy for OA has current marketing approval in any jurisdiction.
Summary:
Extensive pre-clinical data support the use of intra-articular gene therapy for treating OA. Translation is beginning to accelerate and six gene therapeutics are in clinical trials. Importantly, venture capital has begun to flow and at least seven companies are developing products. Significant progress in the future can be expected.
Keywords: Intra-articular therapy, synovium, cartilage, vector, clinical translation, clinical trial
Introduction
Although osteoarthritis (OA) has high heritability, variously estimated between 40% and 60% [1], this reflects the influences of a large number of genes, each of which has a small individual effect. A recent genome wide association meta-analysis, for example, identified 100 different loci associated with OA [2]. Thus, OA is clearly not a good candidate for traditional gene therapy that targets Mendelian disorders, where a single gene defect is responsible for a given disease. Nevertheless OA is a good candidate for gene therapies in which local gene transfer to the affected joint is used as a type of drug delivery system, where the active agent is the product encoded by the transgene [3].
Intra-articular therapy is an attractive strategy for OA because the disease primarily affects a limited number of weight-bearing joints and has no known important extra-articular sequelae. This approach delivers the therapeutic agent directly to the diseased joint, thus enhancing its effectiveness while greatly lowering the risk of adverse events and reducing costs. Local delivery by intra-articular therapy has become particularly important since the development of biologics. The latter, typically proteins, including antibodies, do not enter joints very efficiently following systemic delivery because of the sieving effect of the fenestrated endothelium of the sub-synovial blood vessels [4]. Although intra-articular injection obviates this limitation, there are two additional barriers to delivery: rapid efflux of the injected material via lymphatic drainage and, for drugs acting directly on chondrocytes, the dense extra-cellular matrix (ECM) of the cartilage [4]. No existing technology has succeeded in overcoming these barriers. In 1992 we proposed the novel approach of using gene delivery to target anti-arthritic proteins to joints [5]. The literature prior to 2018 has been comprehensively reviewed in reference 3. The present review summarizes the newer literature with an emphasis on clinical translation and clinical trials.
Gene Delivery to Joints by Intra-articular Injection
As reviewed extensively elsewhere [3], both in vivo and ex vivo strategies have been successful in delivering genes to joints in laboratory animals and, as described below, have been employed in human clinical trials. Reflecting the technology of the time, ex vivo delivery was the first strategy to be developed [6] and was used in the first delivery of a gene to a human joint [7]. However, with one notable exception (see below), this approach has been discontinued for clinical development with the advent of improved vectors allowing efficient in vivo delivery by intra-articular injection with extended periods of local transgene expression. Of the numerous vectors tested pre-clinically as vehicles of in vivo gene delivery to joints, adeno-associated virus (AAV), high-capacity (HC) adenovirus and plasmid DNA are in clinical trials. Self-complementing AAV is the only vector with the confirmed ability to achieve sustained, high levels of intra-articular transgene expression ([8] and Ghivizzani et al, unpublished). In addition, AAV is able to transduce chondrocytes in diseased joints more effectively than other viral vectors, most likely due to its substantially smaller capsid size. Additional important factors that determine the choice of vector are safety, the size of the transgene and its regulatory elements, ease and cost of manufacturing, the immune response to the vector and the ability to re-dose [9].
Preclinical Evaluation in Animal Models of Osteoarthritis
The choice of therapeutic transgene(s) for delivery to joints with OA remains controversial, which is not surprising given the complexity of OA, inexact understanding of its pathophysiology and its different phenotypes. A large number of cDNAs have been tested in animal models (Table 1).
Table 1:
Use of intra-articular gene therapy in animal models of osteoarthritis
| Transgene | Vector In/Ex vivo |
Model | Result | Reference |
|---|---|---|---|---|
| IL-1Ra | Retrovirus Ex vivo |
Dog ACL transection |
Reduced early loss of cartilage | [37] |
| IL-1Ra | Plasmid In vivo |
Rabbit partial meniscectomy |
Reduced osteophytes, cartilage loss | [38] |
| IL-1Ra | AAV In vivo |
Horse osteochondral chip |
Reduced lameness, cartilage loss, synovitis | [8] |
| IL-1Ra | Adenovirus | Horse osteochondral chip |
Reduced lameness, cartilage loss, synovitis | [36] |
| IL-1Ra | HC adenovirus In vivo |
Mouse Horse osteochondral chip |
Mouse: Reduced cartilage loss, osteophytes, hyperalgesia Horse: reduced lameness, cartilage loss, synovitis |
[10] |
| IL-1Ra | HC adenovirus In vivo |
Rat ACL transection |
Reduced cartilage loss | [11]* |
| Lubricin | HC adenovirus In vivo |
Mouse ACL transection |
Reduced cartilage loss | [13] |
| Lubricin | AAV In vivo |
Rabbit ACL transection |
Reduced cartilage loss | [14] |
| Lubricin and IL-1Ra | HC adenovirus In vivo |
Mouse DMM, CLT |
Stronger anti-OA effect than either gene alone | [15] |
| IL-1Ra, sTNFR1 alone, combined | Adenovirus In vivo |
Rabbit MCL transection, meniscectomy |
IL-1Ra, not sTNFR1, reduced cartilage loss. Combination stronger |
[16] |
| TGF-β1 | Retrovirus Ex vivo |
Rat MIA |
Reduced pain, cartilage loss | [17] |
| Variant IL-10 | Plasmid In vivo |
Canine Naturally occurring |
Reduced pain | [19]** |
| BMP-4 and sFlt-1 | Retrovirus Ex vivo |
Rat MIA |
Improved cartilage repair | [39] |
| γ-glutamyl carboxylase | Lentivirus In vivo |
Lapine ACL transection |
Reduced inflammation, cartilage loss | [40] |
| Kallistatin | Adenovirus In vivo |
Rat ACL transection |
Reduced cartilage loss | [41] |
| Cytostatin-C | AAV In vivo |
Rabbit ACL transection |
No effect | [42] |
| Thrombospondin-1 | Adenovirus In vivo |
Rat ACL transection |
Reduced inflammation, cartilage loss | [43] |
| Hemoxygenase-1 | AAV In vivo |
STR/ort Mouse Spontaneous |
No effect on cartilage loss | [44] |
| Dkk-1; Wnt10b | AAV In vivo |
Rat DMM | Dkk-1 reduced osteophytosis; Wnt10b no effect. | [45] |
| Ras homolog enriched in brain (RHEB) |
Adenovirus In vivo |
Mouse DMM | Reduced cartilage loss | [46] |
| CRISPR/cas vs NGF, IL-1β, MMP13 | AAV In vivo |
Mouse DMM | Ablating NGF reduced pain; IL-1β, MMP13 preserved structure | [20] |
| shRNA vs MIP-γ1 | Lentivirus In vivo |
Mouse ACL transection |
Reduced cartilage loss | [47] |
| miR-93–5p | Lentivirus In vivo |
Rat ACL transection |
Reduced cartilage loss | [48] |
| miR-140–5p | Lentivirus Ex vivo |
Rat DMM | Improved cartilage repair | [49] |
Abbreviations
ACL: Anterior cruciate ligament
BMP: Bone morphogenetic protein
Cas: CRISPR-associated protein
CLT: Cruciate ligament transection
CRISPR: Clustered Regularly Interspaced Short Palindromic Repeats
DMM: Destabilization of the medial meniscus
HC: High capacity
IL Interleukin
IL-1Ra: IL-1 receptor antagonist
MIA: Mono-iodoacetate
MIP: Macrophage inflammatory protein
MMP: Matrix metalloproteinase
NGF: Nerve growth factor
sFlt-1: Soluble fms-like tyrosine kinase-1
shRNA: short hairpin RNA
sTNFR: Soluble tumor necrosis factor-α receptor
TGF-β: Transforming growth factor-β
The interleukin-1 receptor antagonist (IL-1Ra) has been the most studied reflecting the status of IL-1 as a popular, albeit controversial, therapeutic target for OA. The most recent publications in this context report safety and efficacy in mice [10], rats [11*, 12] and horses [8, 10] using AAV and HC-adenovirus vectors. These data support two current phase I clinical trials, described in the next section.
The lubricating glycoprotein lubricin, also known as proteoglycan 4 or PRG4, has shown efficacy when delivered by HC-adenovirus (also known as helper-dependent or gutted adenovirus) as a transgene in mouse models of OA [13]. However, a recent paper using AAV delivery in a rabbit model described loss of efficacy when gene transfer was delayed by 2 weeks after surgery [14]. Stone et al. have reported that a combination of IL-1Ra and PRG4 cDNAs gives a stronger effect than either gene alone [15]. Likewise, a combination of IL-1Ra and soluble tumor necrosis-α receptor-1 (sTNFRI) cDNAs has shown greater efficacy than either alone in a rabbit model [16]. It is interesting that sTNFRI had no effect by itself, but potentiated the action of IL-1Ra.
A recent publication describes efficacy when injecting retrovirally transduced human chondrocytes over-expressing transforming growth factor-β1 into the knee joints of rats with OA induced by mono-iodoacetate (MIA) [17]. According to this paper, the xenografted human cells reduced pain and preserved cartilage by inducing macrophages in the joint to switch from a M1 to a M2 phenotype. This paper was published after clinical trials had started (see next section), with safety having been demonstrated in mice, rabbits and goats [18]. Later developments have cast doubt on the identity of the cells used in these studies (see next section).
Most pre-clinical development of arthritis gene therapy has focused on the use of viral vectors because transgene expression using non-viral delivery is low and transient, while many non-viral vectors are inflammatory. Nevertheless, Watkins et al [19**] recently reported success in delivering plasmid DNA encoding a long-acting variant of IL-10 to the stifle joints of dogs with naturally occurring OA. The treatment reduced pain and is now in human clinical trials.
Additional transgenes evaluated are listed in Table 1. One novel approach uses AAV to deliver elements of the CRISPR/Cas system to ablate specific genes whose products are involved in the intra-articular pathophysiology of OA [20]. In a mouse model of OA, ablation of nerve growth factor (NGF) reduced pain but enhanced structural deterioration of the joint. Ablating IL-1β or matrix metalloproteinse-13 (MMP-13) preserved structure but was less effective than NGF ablation in reducing pain [20].
Of the 24 pre-clinical studies listed in Table 1, 20 were performed in animal models that required surgical destabilization of the joint. While these models are likely to be relevant to post-traumatic OA, their relevance to idiopathic OA, the majority manifestation, is unclear [21]. Likewise, two of the listed studies used MIA, an inhibitor of glycolysis, to induce OA. Its relevance to human OA is also questionable, although it is frequently used as a pain model. It should also be noted that few of these experiments used large animal models of OA, which may be more reflective of human OA than experimental OA in rodents and rabbits.
Relatively little attention has been paid to regulation of transgene expression. Most studies have used a strong constitutive promoter, such as the cytomegalovirus (CMV) immediate early promoter/enhancer. Elements responsive to the transcription factor nuclear factor kappa B (NF-κB) have been most widely explored in the context of inducible transgene expression.
Most research has focused on intra-articular gene delivery. However, there has been recent interest in systemic delivery as a way of addressing systemic issues that engender OA. In this context AAV-mediated follistatin gene transfer, delivered via intra-venous injection to mice fed a high-fat diet, reduced body weight while also lowering levels of inflammatory mediators and diminishing susceptibility to OA induced by surgical destabilization of the medial meniscus (DMM) [22*]. Translating this interesting approach will run into the issues of manufacturing and cost illustrated by the example of Zolgensma, a systemic, AAV-based, in vivo gene therapy for spinal muscular atrophy that costs $2 million per dose [23]. For a common condition such as OA this may be unsupportable, but if the gene therapy also addresses obesity, metabolic syndrome and related conditions there could be room for debate. However, recent safety concerns when using high doses of AAV systemically will also be a major factor [24].
Clinical Trials in Osteoarthritis Gene Therapy
There have been several clinical trials of OA gene therapy (Table 2). Transgenes evaluated in these trials encode TGF-β1, interferon (IFN)-β, IL-1Ra, the NKX3.2 transcription factor and a variant of IL-10.
Table 2:
Clinical trials in the gene therapy of osteoarthritis
| Transgene | Vector (In vivo/Ex vivo) |
Phase | NCT identifier | Status | References |
|---|---|---|---|---|---|
| TGF-β1 | Retrovirus Ex vivo |
I, II, III |
02341391
02341378 02072070 01671072 00599248 03291470 03203330 |
Completed Completed Completed Completed Completed Recruiting Recruiting |
[25–27] |
| IFN-β# | AAV In vivo |
I | 02727764 | Active, not recruiting | [29]** |
| IL-1Ra | AAV In vivo |
I | 02790723 | Completed |
[30] |
| IL-1Ra | HC-Adenovirus In vivo |
I | 04119687 | Active, not recruiting | [31] |
| IL-10 | Plasmid In vivo |
I, II |
03477487
04124042 |
Completed Completed |
|
| NKX3.2 | AAV In vivo |
I |
04875754
05454566 |
Active, recruiting Not yet recruiting |
This trial targeted inflammatory hand arthritis, which could be caused by OA or RA
The first human trials delivered TGF-β1 to knee joints with OA in an ex vivo fashion using allogenic chondrocytes. Allogeneic cells obviate the inconvenience and costs associated with the use of culture expanded, autologous cells, but bring significant risk of immune rejection. The chondrocytes were derived from the digit of an infant with polydactyly and were retrovirally transduced to over-express TGF-β1. Because of the potential for insertional mutagenesis, prior to injection the transduced cells were irradiated at a dose that inhibited cell division, and thus the potential for malignancy, while permitting continued expression of TGF-β. Before injection, the transduced cells were mixed with untransduced chondrocytes from the same donor to sustain and amplify the effect.
One issue with ex vivo gene delivery to joints is the short intra-articular dwell time of the injected cells. Nevertheless, the primary endpoints of Phase I [25], II [26] and III [27] trials were met. These endpoints were mainly patient reported outcomes, but an MRI study suggested reduced disease progression and synovitis, while bone marrow lesions, meniscal damage and osteophytosis were unaffected [28]. In 2017 the South Korean authorities gave marketing approval to the therapy as the drug Invossa. This was the first gene therapeutic approved in Korea and the first for OA anywhere. This approval was revoked in 2019 when it came to light that the genetically modified cells were predominantly HEK293 cells, not chondrocytes. However, two Phase III trials have been allowed in the US (NCT03291470; NCT03203330) on the basis that the earlier US trials, unlike those in Korea, used HEK293 cells from the beginning. These trials are recruiting subjects with OA of the knee (Table 2) while the status of Invossa in Korea is being determined.
Other trials in OA use AAV for the intra-articular delivery of IFN-β, IL-1Ra or NKX3.2. Data from the IFN-β study have recently been published [29**]. AAV serotype 5 expressing IFN-β, under the transcriptional control of a NF-κB responsive promoter, was injected into joints of the hand with inflammatory hand arthritis; this could result from either OA or rheumatoid arthritis (RA). Although the trial was designed to dose 12 subjects, it was stopped after 4 subjects had been injected because of severe tenosynovitis in 3 of the 4 individuals. It is interesting that the 3 subjects whose adverse reactions led to discontinuation had OA, whereas the patient with only a mild reaction had RA. The adverse reactions were delayed, appearing between 4 and 12 days after injection of the vector, and persisted for 5 weeks to 4 months. They may have been related to the high dose of vector that was administered: 1.2 × 1012 viral genomes (vg) in the carpometacarpal joint and 0.6 × 1012 vg in the proximal interphalangeal joint. Patients were screened for neutralizing antibodies (Nab) against AAV5 to exclude those with pre-existing high titers. Nab to AAV5, but not IFN-β, were generated after injection of the virus without a detectable T-cell response to the vector. Shedding of virus was limited to transient, small increases in the blood and saliva
Data from the trials using AAV or HC-adenovirus to deliver IL-1Ra have not been published in the refereed literature, although each has been the subject of a recent abstract [30, 31]. The former uses AAV serotype 2.5 and a CMV promoter. Intra-articular injection of this vector produced no severe adverse events. One subject, who had pre-existing Nab to AAV2.5, experienced a mild/moderate knee effusion following injection which resolved with ice and rest [30]. Pre-existing neutralizing Nab to AAV2.5 have been measured previously in the synovial fluids and sera of patients with OA [32], but only 16% of patients had high titer Nab. Titers in synovial fluid correlated with those in sera, indicating that circulating levels of Nab could be measured to predict titers in synovial fluid.
Preliminary data from the first 5 subjects dosed with the HC-adenovirus vector encoding IL-1Ra suggest that the treatment is well tolerated with evidence of a clinical response in some patients [31]. The cassette delivered by the HC-adenovirus has a NF-κB inducible promoter to restrict transgene expression to inflammatory episodes. In this context it is interesting that higher levels of in vivo IL-1Ra expression are also seen with the AAV vector under inflammatory conditions [8], suggesting that the CMV promoter used in this vector also has an inducible component.
Recently two additional trials have been added to clinicaltrials.gov involving intra-articular injection of an AAV vector serotype 5.2 containing cDNA encoding the NKX3.2 transcription factor, which is responsible for maintaining the chondrocytic phenotype by preventing progression to hypertrophy [33]. Although little supporting information appears in the literature, one of these trials is actively recruiting OA patients (Table 2).
Finally, it is encouraging that testing of the IL-10 variant plasmid has progressed to a Phase II trial that is recorded as “completed” on ClinicalTrials.gov (NCT04124042). No outcome data are given, so it is not possible to evaluate the results. This variant of IL-10 does not appear to have been published in the scientific literature, but it may be one described in a US patent application that describes a combination of IL-10 with the IL-10R1 component of its receptor [34].
Perspectives
Gene therapy approaches to treating OA are now attracting serious attention. Several clinical trials have been initiated and one has completed Phase II; Phase III trials have been completed in Korea and are in progress in the US (Table 2). Venture capital is beginning to flow and there are at least seven companies developing gene therapeutics for OA. These advances auger well for accelerating development of the field.
Although considerable progress has been made in overcoming the technological hurdle of achieving long-term transgene expression in joints, there is no consensus on the choice of transgene and promoter. Constructs expressing more than one therapeutic gene product may have greater potency, and it is also worth considering combination therapy in which gene therapy is used in conjunction with other medical or surgical approaches. Because OA is a very heterogeneous disease, different types of OA may need to be matched with specific gene therapeutics, in which case a diagnostic test for patient selection would be of great value. In the case of IL-1Ra, for example, different haplotypes of the IL1RN gene predicting greater severity and quicker progression of disease could identify patients who respond best to IL-1Ra gene therapy [35*].
Key Points.
Advances in vector technology now permit in vivo gene delivery to joints with extended periods of transgene expression. The development of self-complementing AAV vectors has been particularly valuable in this regard.
A number of different cDNAs have shown therapeutic effects in animal models of OA using rodents and rabbits, but few have been evaluated in larger animals that may reflect better human joints with OA.
Six different gene therapeutics for the intra-articular treatment of OA are in clinical trials.
No gene therapy for OA is presently approved in any jurisdiction, but increasing investment from academia, biotechnology and venture capital groups provides optimism about future progress.
Acknowledgements
The authors’ work in this area has been supported by NIH grants R01 AR43623, R21 AR049606, R01 AR048566, R01 AR057422, R01 AR051085 and X01NS066865; DoD grants W81XWH-16-1-0540, W81XWH-14-1-0498 and W81XWH-19-1-0515.
Footnotes
Conflict of interest
The authors are co-founders of Genascence Corp.
References
- 1.Spector TD and MacGregor AJ, Risk factors for osteoarthritis: genetics. Osteoarthritis Cartilage, 2004. 12 Suppl A: p. S39–44. [DOI] [PubMed] [Google Scholar]
- 2.Boer CG, et al. , Deciphering osteoarthritis genetics across 826,690 individuals from 9 populations. Cell, 2021. 184(18): p. 4784–4818 e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Evans CH, Ghivizzani SC, and Robbins PD, Gene Delivery to Joints by Intra-Articular Injection. Hum Gene Ther, 2018. 29(1): p. 2–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Evans CH, Kraus VB, and Setton LA, Progress in intra-articular therapy. Nat Rev Rheumatol, 2014. 10(1): p. 11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bandara G, et al. , Gene transfer to synoviocytes: prospects for gene treatment of arthritis. DNA Cell Biol, 1992. 11(3): p. 227–31. [DOI] [PubMed] [Google Scholar]
- 6.Bandara G, et al. , Intraarticular expression of biologically active interleukin 1-receptor-antagonist protein by ex vivo gene transfer. Proc Natl Acad Sci U S A, 1993. 90(22): p. 10764–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Evans CH, et al. , Gene transfer to human joints: progress toward a gene therapy of arthritis. Proc Natl Acad Sci U S A, 2005. 102(24): p. 8698–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Watson Levings RS, et al. , Self-Complementary Adeno-Associated Virus-Mediated Interleukin-1 Receptor Antagonist Gene Delivery for the Treatment of Osteoarthritis: Test of Efficacy in an Equine Model. Hum Gene Ther Clin Dev, 2018. 29(2): p. 101–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Watson-Levings RS, et al. , Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front Bioeng Biotechnol, 2022. 10: p. 901317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Nixon AJ, et al. , Disease-Modifying Osteoarthritis Treatment With Interleukin-1 Receptor Antagonist Gene Therapy in Small and Large Animal Models. Arthritis Rheumatol, 2018. 70(11): p. 1757–1768. [DOI] [PubMed] [Google Scholar]
- 11. Senter R, et al. , Efficacy and Safety of FX201, a Novel Intra-Articular IL-1Ra Gene Therapy for Osteoarthritis Treatment, in a Rat Model . Hum Gene Ther, 2022. 33(9–10): p. 541–549. *This IND-enabling study laid the groundwork for a clinical trial in which HC-adenovirus is used to deliver IL-1Ra cDNA to the knee joints of patients with mid-stage OA.
- 12.Wang G, et al. , Safety and biodistribution assessment of sc-rAAV2.5IL-1Ra administered via intra-articular injection in a mono-iodoacetate-induced osteoarthritis rat model. Mol Ther Methods Clin Dev, 2016. 3: p. 15052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ruan MZ, et al. , Proteoglycan 4 expression protects against the development of osteoarthritis. Sci Transl Med, 2013. 5(176): p. 176ra34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Seol D, et al. , Intra-Articular Adeno-Associated Virus-Mediated Proteoglycan 4 Gene Therapy for Preventing Posttraumatic Osteoarthritis. Hum Gene Ther, 2022. 33(9–10): p. 529–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Stone A, et al. , Combinatorial Prg4 and Il-1ra Gene Therapy Protects Against Hyperalgesia and Cartilage Degeneration in Post-Traumatic Osteoarthritis. Hum Gene Ther, 2019. 30(2): p. 225–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wang HJ, et al. , Suppression of experimental osteoarthritis by adenovirus-mediated double gene transfer. Chin Med J (Engl), 2006. 119(16): p. 1365–73. [PubMed] [Google Scholar]
- 17.Lee H, et al. , TissueGene-C promotes an anti-inflammatory micro-environment in a rat monoiodoacetate model of osteoarthritis via polarization of M2 macrophages leading to pain relief and structural improvement. Inflammopharmacology, 2020. 28(5): p. 1237–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Noh MJ, et al. , Pre-clinical studies of retrovirally transduced human chondrocytes expressing transforming growth factor-beta-1 (TG-C). Cytotherapy, 2010. 12(3): p. 384–93. [DOI] [PubMed] [Google Scholar]
- 19. Watkins LR, et al. , Targeted interleukin-10 plasmid DNA therapy in the treatment of osteoarthritis: Toxicology and pain efficacy assessments. Brain Behav Immun, 2020. 90: p. 155–166. **This paper reports the use of plasmid DNA in the stifle joints of dogs with OA. The use of a large animal with naturally occurring OA adds authenticity to the study. It is remarkable how non-viral delivery induces a sustained analgesic effect.
- 20.Zhao L, et al. , Exploration of CRISPR/Cas9-based gene editing as therapy for osteoarthritis. Ann Rheum Dis, 2019. 78(5): p. 676–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Iijima H, et al. , Meta-analysis Integrated With Multi-omics Data Analysis to Elucidate Pathogenic Mechanisms of Age-Related Knee Osteoarthritis in Mice. J Gerontol A Biol Sci Med Sci, 2022. 77(7): p. 1321–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Tang R, et al. , Gene therapy for follistatin mitigates systemic metabolic inflammation and post-traumatic arthritis in high-fat diet-induced obesity. Sci Adv, 2020. 6(19): p. eaaz7492. *This paper reports a novel, systemic gene delivery approach to preventing OA. Rather than treating the joint directly, it counters obesity in these mice and thereby reduces weight, inflammation and susceptibility to post-traumatic OA. Although clinical translation may be problematic, the approach is interesting.
- 23.Nuijten M, Pricing Zolgensma - the world’s most expensive drug. J Mark Access Health Policy, 2022. 10(1): p. 2022353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mullard A, Gene therapy community grapples with toxicity issues, as pipeline matures. Nat Rev Drug Discov, 2021. 20(11): p. 804–805. [DOI] [PubMed] [Google Scholar]
- 25.Ha CW, et al. , Initial phase I safety of retrovirally transduced human chondrocytes expressing transforming growth factor-beta-1 in degenerative arthritis patients. Cytotherapy, 2012. 14(2): p. 247–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lee B, et al. , Results of a Phase II Study to Determine the Efficacy and Safety of Genetically Engineered Allogeneic Human Chondrocytes Expressing TGF-beta1. J Knee Surg, 2020. 33(2): p. 167–172. [DOI] [PubMed] [Google Scholar]
- 27.Kim MK, et al. , A Multicenter, Double-Blind, Phase III Clinical Trial to Evaluate the Efficacy and Safety of a Cell and Gene Therapy in Knee Osteoarthritis Patients. Hum Gene Ther Clin Dev, 2018. 29(1): p. 48–59. [DOI] [PubMed] [Google Scholar]
- 28.Guermazi A, et al. , Structural effects of intra-articular TGF-beta1 in moderate to advanced knee osteoarthritis: MRI-based assessment in a randomized controlled trial. BMC Musculoskelet Disord, 2017. 18(1): p. 461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Vrouwe JPM, et al. , Administration of an adeno-associated viral vector expressing interferon-beta in patients with inflammatory hand arthritis, results of a phase I/II study. Osteoarthritis Cartilage, 2022. 30(1): p. 52–60. **This provides data from one of the first in vivo gene therapy trials to include joints with OA. AAV serotype 5 was used to deliver IFN-g cDNA to the small joints of the hand. Although scheduled for 12 subjects, the trial was stopped after only 4 individuals had been dosed because of a severe inflammatory reaction to the vector.
- 30.Sellon S, et al. , A Phase I Clinical Trial of Osteoarthritis Gene Therapy (NCT02790723). Molecular Therapy, 2022. 30: p. 376. [Google Scholar]
- 31.Kelley S, et al. , Interim Data from the First-in-Human Phase 1 Trial of FX201, An Intra-Articular, Helper-Dependent Adenoviral Gene Therapy for Osteoarthritis - Safety, Tolerability, Biodistribution, and Preliminary Evaluation of Clinical Activity in 5 Patients. Molecular Therapy, 2021. 29: S1: p. 288. [Google Scholar]
- 32. Abdul TY, et al. , Prevalence of AAV2.5 neutralizing antibodies in synovial fluid and serum of patients with osteoarthritis. Gene Ther, 2022. *This provides the first measurement of pre-existing Nab to AAV serotype 2.5 in the sera and synovial fluids of patients with OA. Titers correlated between synovial fluid and serum, suggesting that serum measurements could be used conveniently to pre-screen subjects receiving AAV2.5-mediated gene therapy.
- 33.Caron MM, et al. , BAPX-1/NKX-3.2 acts as a chondrocyte hypertrophy molecular switch in osteoarthritis. Arthritis Rheumatol, 2015. 67(11): p. 2944–56. [DOI] [PubMed] [Google Scholar]
- 34.Forsayeth J, et al. , Methods and compositions to enhance the anti-inflammatory effects of interleukin-10. , U.P. Office, Editor 2019. [Google Scholar]
- 35. Attur M, et al. , Interleukin 1 receptor antagonist (IL1RN) gene variants predict radiographic severity of knee osteoarthritis and risk of incident disease. Ann Rheum Dis, 2020. 79(3): p. 400–407. *This paper provides additional evidence that certain haplotypes in the IL1RN gene can predict the course of OA and identify patients who are most likely to respond to IL-1Ra (IL1RN) gene therapy.
- 36.Frisbie DD, et al. , Treatment of experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene. Gene Ther, 2002. 9(1): p. 12–20. [DOI] [PubMed] [Google Scholar]
- 37.Pelletier JP, et al. , In vivo suppression of early experimental osteoarthritis by interleukin-1 receptor antagonist using gene therapy. Arthritis Rheum, 1997. 40(6): p. 1012–9. [DOI] [PubMed] [Google Scholar]
- 38.Fernandes J, et al. , In vivo transfer of interleukin-1 receptor antagonist gene in osteoarthritic rabbit knee joints: prevention of osteoarthritis progression. Am J Pathol, 1999. 154(4): p. 1159–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Matsumoto T, et al. , Cartilage repair in a rat model of osteoarthritis through intraarticular transplantation of muscle-derived stem cells expressing bone morphogenetic protein 4 and soluble Flt-1. Arthritis Rheum, 2009. 60(5): p. 1390–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fu X, et al. , Effects of GGCX overexpression on anterior cruciate ligament transection-induced osteoarthritis in rabbits. Mol Med Rep, 2018. 17(3): p. 3821–3828. [DOI] [PubMed] [Google Scholar]
- 41.Hsieh JL, et al. , Adenovirus-mediated kallistatin gene transfer ameliorates disease progression in a rat model of osteoarthritis induced by anterior cruciate ligament transection. Hum Gene Ther, 2009. 20(2): p. 147–58. [DOI] [PubMed] [Google Scholar]
- 42.Kyostio-Moore S, et al. , Overexpression of cystatin C in synovium does not reduce synovitis or cartilage degradation in established osteoarthritis. Arthritis Res Ther, 2015. 17: p. 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hsieh JL, et al. , Intraarticular gene transfer of thrombospondin-1 suppresses the disease progression of experimental osteoarthritis. J Orthop Res, 2010. 28(10): p. 1300–6. [DOI] [PubMed] [Google Scholar]
- 44.Kyostio-Moore S, et al. , Local gene delivery of heme oxygenase-1 by adeno-associated virus into osteoarthritic mouse joints exhibiting synovial oxidative stress. Osteoarthritis Cartilage, 2013. 21(2): p. 358–67. [DOI] [PubMed] [Google Scholar]
- 45.Mason JB, et al. , Wnt10b and Dkk-1 gene therapy differentially influenced trabecular bone architecture, soft tissue integrity, and osteophytosis in a skeletally mature rat model of osteoarthritis. Connect Tissue Res, 2017. 58(6): p. 542–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ashraf S, et al. , RHEB gene therapy maintains the chondrogenic characteristics and protects cartilage tissue from degenerative damage during experimental murine osteoarthritis. Osteoarthritis Cartilage, 2019. 27(10): p. 1508–1517. [DOI] [PubMed] [Google Scholar]
- 47.Shen PC, et al. , Lentiviral small hairpin RNA knockdown of macrophage inflammatory protein-1gamma ameliorates experimentally induced osteoarthritis in mice. Hum Gene Ther, 2013. 24(10): p. 871–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xue H, et al. , miR-93–5p attenuates IL-1beta-induced chondrocyte apoptosis and cartilage degradation in osteoarthritis partially by targeting TCF4. Bone, 2019. 123: p. 129–136. [DOI] [PubMed] [Google Scholar]
- 49.Geng Y, et al. , Intra-articular injection of hUC-MSCs expressing miR-140–5p induces cartilage self-repairing in the rat osteoarthritis. J Bone Miner Metab, 2020. 38(3): p. 277–288. [DOI] [PubMed] [Google Scholar]
