Abstract
On September 14–15, 2020, the Foundation Fighting Blindness convened a virtual workshop to discuss intraocular inflammation during viral vector-mediated gene therapy for inherited retinal diseases. The workshop's goals were to understand immune activation's nature and significance during ocular gene therapy, consider whether ocular inflammation limits gene therapy's potential, and identify knowledge gaps for future research. The event brought together a small group of experienced researchers in the field to present and discuss current data. Collectively, participants agreed that clinical, as well as subclinical, inflammation during ocular gene therapy is common. The severity of inflammation in both animal and clinical studies varied widely but is generally related to vector dose. Severe inflammation was associated with reduced gene therapy efficacy. However, the relationship between outcomes and subclinical inflammation, pre-existing antivector antibodies, or induced adaptive immune responses is still unclear. Uncertainties about the contribution of vector manufacturing issues to inflammation were also noted. Importantly, various immunosuppressive treatment protocols are being used, and this heterogeneity confounds conclusions about optimal strategies. Proposed near-term next steps include establishing an immunological consultant directory, establishing a data repository for pertinent animal and clinical data, and developing a larger meeting. Priority areas for future research include deeper understanding of immune activation during retinal diseases and during ocular gene therapy; better, harmonized application of animal models; and identifying best practices for managing gene therapy vector-related ocular inflammation.
Translational Relevance:
Subclinical or clinical inflammation often arises during ocular gene therapy with viral vectors. Understanding the biological bases and impacts on efficacy are important for clinical management and the improvement of future therapies.
Gene therapies predominantly use viral vectors for in vivo delivery of genes to augment or repair dysfunctional inherited genes. To date, more than 300 genes and loci have been implicated in causing inherited retinal diseases (IRDs), which are mostly monogenic, with varying inheritance patterns. Many of these single-gene defects may be amenable to gene therapy strategies, and efforts to develop gene-based treatments have been underway for more than 30 years. The approval of Kymriah, which uses ex vivo delivery of a chimeric antigen receptor via a lentiviral vector (Oxford BioMedica, Oxford, UK/Novartis, Basel, Switzerland) to patient T cells, validated the power of the approach. But clinical success of in vivo administration of a gene therapy has only come recently. The approval of Luxturna (voretigene neparvovec-rzyl) in 2017 to treat IRDs caused by biallelic pathogenic variants of the RPE65 gene was a major milestone. Subretinal administration of Luxturna delivers wild-type cDNA encoding RPE65 directly to the subretinal region of diseased eyes, thereby improving functional vision.
But development of gene therapies has also been accompanied by substantial risks and tragic setbacks. In 1999, Jesse Gelsinger died after administration of an adenoviral vector to treat an X-linked metabolic disease, ornithine transcarboxylase deficiency.
2 Mr. Gelsinger's death from multiple organ failure was attributed to severe antivector immune responses. In the early 2000s, several children being treated for X-linked severe combined immunodeficiency with a retroviral vector, which was derived from the Moloney Murine Leukemia Virus, had development of T-cell leukemias.
3 This outcome was likely due to oncogene activation at the site of retroviral integration. These past lessons served as cautionary examples and prompted significant changes to the viral vector platforms themselves.
The field's focus also shifted to ocular gene therapy, in part because of the relative “immune privilege” of the eye, the prevalence of monogenic diseases that could be effectively addressed, decreased vector quantities needed and manufacturing costs, and the limited systemic exposure and immune responses. The ultimate goal of treating IRDs in the eye by gene therapy is now to restore functional vision, although any therapy that slows or stops disease progression and improves vision is valuable. Approaches to correcting genetic defects now include gene augmentation, which introduces a wild-type copy of the affected gene to augment expression; gene correction, through various editing techniques to correct pathogenic variants or engineer out inappropriate splicing; and gene expression modification, using techniques such as genetic knockdown of dominant negative variants. The workshop focused on gene augmentation approaches.
Although gene therapies based on lentiviral and nonviral vectors delivery are in development, vectors derived from adeno-associated virus (AAV) are currently the most commonly used platform for delivery of ocular gene therapy.
4,5 Wild-type AAV is a small DNA virus with many naturally occurring capsid serotypes, which can be used directly for gene therapy or first intentionally modified. The virus is primarily non-integrating but persists as episomal DNA. The recombinant AAV vectors used for gene delivery have been engineered such that the only viral contents of the virion delivered to the patient are the two inverted terminal repeats (∼145 bases) required for viral formation that flank the gene of interest. The transgene is placed under the control of selected regulatory elements that include enhancers, promoters, introns, poly(A) signals, and posttranscriptional elements (e.g., the Woodchuck Hepatitis Virus Posttranscriptional Regulatory Element). The important properties of the resulting recombinant AAV vector are its ability to transduce cells by crossing cell membranes, trafficking through endosomes to the cell nucleus, and delivering the DNA cargo of choice. Furthermore, by taking advantage of the tissue-specific tropism of different AAV serotypes, tissue-specific promoters, and route of administration, AAV vectors can be directed to transduce and express only in the desired retinal cell-type(s).
AAV gene augmentation vectors are therefore, in effect, complex biologic drugs that consist of a formulation (viral capsid), which encapsulates a pre-pro-drug (DNA), which is transcribed inside host cells into a pro-drug (RNA), which is translated and processed into the final biologic drug (protein). The product developmental lifecycles for these complex biologics present multiple challenges in terms of optimizing each of these stages of the drug. Moreover, product development also presents the challenge of considering host biological responses. To the host cells and immune system, the vector formulation presents foreign DNA sequences, viral expression elements, and viral capsid proteins, which cells will recognize both as nonspecific danger signals (through pathogen-associated molecular patterns and damage-associated molecular patterns) and as foreign antigens. As a result, both nonspecific clinical inflammation and specific acquired immunity may neutralize the vector effect, reduce cell viability, and further fuel immune responses. The current clinical toolbox for handling inflammation and immune responses typically revolves around companion steroid therapy.
The workshop's goals were therefore to develop a deeper understanding of physiological ocular immunity and immune activation in response to gene therapy; to discuss the prevalence and significance of inflammation during gene therapy for eye disease; to consider whether ocular inflammation and immune responses limit gene therapy's potential; and to identify knowledge gaps for future research.
The second day of the workshop focused on human clinical experience from gene therapy trials. Presentations were structured to consider whether inflammation, or managing inflammation, interferes with treatment goals, namely to deliver the optimal formulation and amount of gene therapy vector that yields the best protein expression and efficacy.
Dr. Christine Kay summarized clinical observations gained from work using a variety of protocols. She noted that eyes with IRDs exist in a state of low-grade inflammation, and surgery itself is inflammatory. AAV treatment–related inflammation was typical after either subretinal or intravitreal injections and appears to be dose dependent. Across different clinical protocols, surgical techniques vary, dose and volume of AAV delivered varies, and the type and duration of presurgery immunosuppression varies. Technical differences such as the use of pre-blebs, bleb location, and volume of vector delivered may have substantial implications for interpreting dose-escalation studies. Anecdotally, Dr. Kay estimated that about 10% of gene therapy patients have significant clinically apparent inflammation that can be treated with either oral or periocular steroids, but a considerably larger percentage of patients appear to have subtle, subclinical inflammation. In some cases, this inflammation can be detected by fluorescein angiography and optical coherence tomography; loss of retinal function can be detected with microperimetry and occasionally by visual acuity measurements. Despite treatment, some inflammation can be long-lasting and appears to correlate with loss of efficacy.
On the basis of his experience, as well as literature reports, Dr. Tim Stout estimated that as much as 25% to 35% of patients have some degree of GTAU, more frequently following intravitreal injection than after subretinal injection. Dr. Stout's protocols typically use oral steroid pretreatment ranging from three to seven days before surgery, coupled with local steroid treatment during and after surgery. Whereas most inflammation seems to be transient and treatable, its impact on transgene expression has not been clear. A number of factors may promote inflammation, including viral dose and transduction kinetics. To better understand the role of dose, Drs. Stout and Violet Lin modeled the subretinal injection site mathematically to estimate the multiplicity of infection (MOI) of viral particles per cell in the retina. In their experience, surgical blebs are typically either a semisphere shape or a squashed-sphere shape; 300 µL macular blebs average about 14 mm in diameter, making contact with as many as 18 × 106 cells. Using bleb morphology and diameter coupled with assumptions about volume, cell density, and retinal dimensions, preliminary modeling results suggested that AAV MOIs could be quite large. Estimated MOIs ranged from about 9600:1 for rods to 212,000:1 for RPE cells. These calculations raised concerns about vector overtreatment and potential off-target effects.
Dose-escalation studies of intravitreal injection with GS010 (Lumevoq) for treating Leber hereditary optic neuropathy, described by Dr. José-Alain Sahel, did not use immunosuppression before surgery. Inflammation was seen in more than 90% of patients but was mostly mild and treatable; however, the use of high doses was limited by inflammation. Optogenetic trials of another product, GS030, to treat one form of retinitis pigmentosa did use immunosuppression at time of surgery; to date, mild but manageable intraocular inflammation has been observed. Patients have been monitored for serum anti-AAV antibodies at the time of treatment and for development of antibodies and T-cell responses, but so far immune status and treatment outcomes have not been related.
Dr. Kanmin Xue discussed clinical studies using subretinal injections of an AAV2 vector expressing REP1 to treat choroideremia and of an AAV8 vector expressing codon-optimized retinitis pigmentosa GTPase regulator gene (RPGR) to treat X-linked retinitis pigmentosa (
Table 1B). Some patients developed signs of retinal inflammation that were related to vector dose and resolved with treatment (
Table 2B). Because inflammation developed in the first patient to receive high-dose gene therapy for choroideremia, the standard perioperative immunosuppression regime was increased from seven days to 21 days. Close observation within the first three months after treatment was important, because signs of retinal inflammation may be subtle and correlate with fluctuations in macular function; timely intervention was important to improve clinical outcome. Laboratory studies suggested a potential role for hydroxychloroquine as an adjunct to suppress TLR9-mediated immune activation in the retina during innate immune responses after AAV gene therapy.
Dose-escalation studies of AAV8-RS1 to treat X-linked retinoschisis (
Table 1B) indicated that inflammation increased with dose, as presented by Dr. Paul Sieving. Appropriate delivery of this transgene, a secreted protein, via intravitreal vector injection is a complex and challenging process. Initial surgeries used oral prednisone pretreatment (
Table 2B). Although NAb responses among patients were variable, the sera of some patients contained substantial amounts of NAbs despite immunosuppression, and NAb levels appeared to track with inflammation. The most recent surgeries have therefore used a triple drug immunosuppression regimen (a combination of prednisone, cyclosporin, and mycophenolate mofetil).
Dr. Mark Shearman reviewed NHP and clinical studies (
Table 1B) of AAV vectors designed to treat X-linked retinoschisis, X-linked retinitis pigmentosa, and two forms of achromatopsia. NHP studies intentionally did not use steroid pretreatment, but some animals were treated with steroids at surgery or when needed. Across different NHP studies, inflammation increased with dose but was transient, as reflected mostly by in-life clinical evaluations. Outcomes appeared to be improved by purifying AAV vector preparations to remove empty capsids. The presence of pre-existing anti-AAV serum antibodies, or development of anti-AAV antibodies on injection, did not correspond with the extent of ocular inflammation and did not prohibit gene expression. Only anti-AAV capsid antibodies have been observed, not antibodies to the transgenes. Clinical studies have used steroid treatment at the time of surgery and for a period after vector administration, tapering over time; to date, mild to moderate inflammation has been observed, and a few patients had development of GTAU.
Dr. Ian MacDonald described his experience to date with an AAV2 vector expressing REP1 to treat choroideremia (
Table 1B), which used a 21-day steroid regimen, including 2 days’ pretreatment (
Table 2B). Despite pretreatment, five of six patients had some level of intraocular inflammation. One subject had significant loss of the central macular retinal pigment epithelium, as revealed by fundus autofluorescence. The same patient experienced a serious adverse event after the initial steroid treatment stopped. Hyperreflective deposits appeared within the retina, presumably cellular infiltrates; these bodies resolved slowly with a second course of systemic steroid treatment. Dr. MacDonald noted that these patients were being treated at a relatively late disease stage, which may in itself limit both safety and efficacy outcomes.
The workshop's discussions consistently suggested that, in both animals and people, ocular inflammation almost always accompanies gene therapy treatments by any route, and the degree of inflammation is correlated with dose. Beyond dose per se, other variables such as viral concentration, volume, and retinal surface area appear to be important. However, harmonizing these parameters between small-globe animals and people to study them is difficult. Even when inflammation was not clinically apparent, tissue and cellular changes in the retina were often found. Interpreting the significance of these changes is not straightforward.
While low levels of inflammation can be treated, severe inflammation both limits AAV dose and is consistently associated with reduced efficacy. Clearly a wide variety of different treatment protocols are being used before, during, and after gene therapy administration to suppress unwanted clinical inflammation, NAbs, or T cell responses. The heterogeneity in research and clinical protocols confounds conclusions about optimal strategies at this time. Identifying consensus protocols and best practices might help minimize study variables and allow harmonization, adding rigor to clinical studies. However, the amounts of cellular immune activation that could potentially reduce gene therapy efficacy are not clear, and correspondingly no evidence supports an approach to select the desired amounts of immunosuppression.
Immunosuppression regimens typically use various corticosteroids, locally or systemically, which is problematic; long-term steroid use, in particular, has undesirable consequences. The choice of steroid treatments or immunomodulatory therapy with cyclosporin A or other agents should be tailored in context, such as the nature of the patient's disease, age, and comorbidities, as well as the nature of the vector and route of administration. Using targeted biologics instead could be attractive, but the knowledge base to rationally choose targets and products is insufficient. This is an important area for future research. Some indication for using targeted immunomodulators, or perhaps biologics that inhibit lysosomal pathways and thus lysosomal TLR activation (such as through mechanistic target of rapamycin kinase (mTOR) inhibition), was supported by preclinical data presented on use of hydroxychloroquine.
The authors extend great appreciation to Leilla Kenny of the Foundation for all meeting logistic support, including coordinating, organizing, recording pre-workshop video presentations, and running the Zoom platform. We further thank Chris Adams for implementing both the private and public workshop websites and professionally editing the pre-workshop videos. We thank Amy Laster for assisting in the workshop organization and formatting the online surveys. Karen Elkins provided scientific writing and editing during the preparation of the manuscript.
The Foundation Fighting Blindness provided financial and logistic support for this event.
Authors are listed alphabetically, and all authors contributed equally.
Workshop organizers and lead authors
Harvard Medical School, Boston, MA, USA
UCL Institute of Ophthalmology, London, UK
University of Bristol, Bristol, UK
Hubble Therapeutics, Boston, MA, USA
University Hospital, Cologne, Germany
Foundation Fighting Blindness, Columbia, MD, USA
+The Foundation Fighting Blindness Ocular Gene Therapy Inflammation Working Group: Workshop presenters and discussants
Division of Experimental Retinal Therapies (Division of ExpeRTs), School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
Division of Experimental Retinal Therapies (Division of ExpeRTs), School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
Department of Pediatrics, University of Florida, Gainesville, FL, USA
Access BIO, Boyce, VA, USA
Harvard Medical School, Boston, MA, USA
Harvard Medical School, Boston, MA, USA
UCL Institute of Ophthalmology, London, UK
University of Bristol, Bristol, UK
University of California, San Francisco, San Francisco, CA, USA
Gyroscope Therapeutics, King of Prussia, PA, USA
University of Tübingen, Tübingen, Germany
University of California, Berkley, Berkley, CA, USA
Vitreoretinal Associates, Gainesville, FL, USA
University Hospital, Cologne, Germany
University of Alberta, Edmonton, Canada
University of Rochester, Rochester, NY, USA
University of Washington, Seattle, WA, USA
University of Pittsburgh School of Medicine, Pittsburg, PA, USA
University of California at Davis, Davis CA, USA
Baylor College of Medicine, Houston, TX, USA
Oxford Eye Hospital, Oxford, UK
Foundation Fighting Blindness
Disclosure: Y.K. Chan, Ally Therapeutics (C, I); A.D. Dick, MeiraGTx (F), Janssen Pharmaceuticals (F), Novartis (C), AbbVie Inc. (C), Active Bio (C), Affibody AB (C), Hubble Therapeutics (C); S.M. Hall, OcQuila Therapeutics (I), Hubble Therapeutics (I); T. Langmann, None; C.L. Scribner, None; B.C. Mansfield, the Foundation Fighting Blindness (E), Patient Advisory Council, AGTC (C)