Gene Therapy Breakthrough: New Evidence Shows Promise for Hemophilia Cure

Introduction
The search for cures for hemophilia has reached a groundbreaking milestone. Gene therapy, once a futuristic concept, can now offer patients the possibility of near-normal blood clotting and a life largely free from frequent bleeding episodes. In clinical studies, gene therapy has reduced annual bleeding rates by up to 90%, offering a potential one-time treatment option for a condition that has long required lifelong care.
In a major advancement, the U.S. Food and Drug Administration (FDA) recently approved etranacogene dezaparvovec, the first gene therapy specifically for hemophilia B. This therapy uses a viral vector to deliver a healthy copy of the F9 gene directly to the liver, where FIX is normally produced. The goal? Enable the patient’s own body to produce sufficient clotting factor, reducing or even eliminating the need for external infusions.
Clinical trial results have been nothing short of remarkable:
- 96% of participants were able to discontinue regular FIX treatments.
- There was a 97% reduction in the use of FIX infusions overall.
- Many patients achieved FIX levels in or near the normal range, drastically reducing spontaneous bleeding episodes.
This article examines the latest evidence supporting gene therapy as a potential cure for hemophilia, including its mechanisms, clinical efficacy, and implications for future treatment approaches.
The Science Behind Gene Therapy for Hemophilia B
Hemophilia B is a rare, inherited bleeding disorder caused by mutations in the F9 gene, which encodes clotting factor IX (FIX). It affects about 1 in every 25,000 male births globally. In the United States alone, around 20,000 individuals live with some form of hemophilia, while worldwide cases exceed 400,000.
Patients with hemophilia B typically require frequent intravenous infusions of FIX to prevent or control bleeding. This lifelong therapy can be both physically demanding and financially burdensome, with annual treatment costs sometimes exceeding $450,000 per patient.
Unlike traditional treatments that require frequent intravenous infusions, gene therapy represents a potential one-time intervention aimed at correcting the underlying genetic defect. Understanding the scientific principles behind this approach reveals why gene therapy has emerged as a promising path toward cures for hemophilia.
How Hemophilia Disrupts Normal Blood Clotting
Hemophilia B occurs due to mutations in the F9 gene located on the X chromosome, which explains its predominance in males. This gene contains instructions for producing factor IX, a crucial protein in the intrinsic pathway of the blood coagulation cascade. When functioning normally, factor IX is synthesized by hepatocytes and circulates in plasma at concentrations of 4-5 µg/mL with a half-life of approximately 18-24 hours.
The severity of hemophilia B correlates directly with the amount of functional factor IX present in the bloodstream:
- Severe hemophilia – Less than 1% of normal factor IX activity; characterized by frequent spontaneous bleeds into joints and muscles
- Moderate hemophilia – 1-5% of normal factor IX activity; less frequent spontaneous bleeding but significant risk after minor trauma
- Mild hemophilia – 5-40% of normal factor IX activity; bleeding typically occurs only after significant injury
Since factor IX contributes to clot formation by catalyzing the conversion of factor X to activated factor X, its deficiency creates a critical gap in the coagulation cascade. Consequently, even minor injuries can lead to prolonged bleeding. Furthermore, spontaneous hemorrhage into joints (hemarthrosis) causes progressive joint damage, ultimately resulting in debilitating arthropathy if left untreated.
Viral Vectors as Gene Delivery Systems
Gene therapy relies on viral vectors, engineered viruses that deliver therapeutic genes to human cells. For hemophilia B, the preferred delivery vehicle is the adeno-associated virus (AAV), a non-pathogenic virus known for its ability to target liver cells (hepatocytes) with high efficiency and relatively low immune activation.
Several characteristics make hemophilia B an ideal candidate for gene therapy approaches:
- The therapeutic window is relatively wide – even modest increases in factor IX levels from <1% to >5% can substantially improve clinical outcomes
- Factor IX requires relatively low plasma concentrations for efficacy
- The F9 gene (approximately 1.6 kb) fits easily within viral vector capacity constraints
Initially, gene therapy trials employed various vector types, including retroviral, adenoviral, and plasmid vectors. At present, adeno-associated virus (AAV) vectors have emerged as the preferred platform due to their safety profile and efficacy in targeting post-mitotic tissues like the liver.
AAV Vectors: The Leading Platform for Hemophilia Gene Therapy
AAV vectors represent a revolutionary advancement in gene delivery for hemophilia B treatment. These vectors are derived from parvoviruses that are non-pathogenic in humans and possess several advantageous properties. Most importantly, they can efficiently transduce hepatocytes while displaying minimal immunogenicity compared to other viral vectors.
The structure of an AAV vector consists of an icosahedral protein capsid surrounding a genetic payload. This payload contains the therapeutic F9 gene flanked by inverted terminal repeats (ITRs), which are essential for both packaging the vector genome and forming episomal DNA after transduction. Once delivered to hepatocytes, these vectors primarily remain episomal (non-integrating), which reduces concerns about insertional mutagenesis.
Current AAV-based gene therapies for hemophilia B typically incorporate several key components:
- A codon-optimized F9 gene to enhance expression
- Liver-specific promoters (often derived from human α-1-antitrypsin with enhancer elements)
- The FIX-Padua variant (FIX-R338L), which provides approximately eightfold enhanced activity compared to wild-type FIX
The selection of specific AAV serotypes notably impacts treatment outcomes. While early studies utilized AAV2, over 100 naturally occurring AAV serotypes with varying tissue tropism now exist. For instance, the transition from AAV2 to AAV8 capsids demonstrated improved efficacy, as seen in trials by St. Jude Children’s Research Hospital and University College London.
Despite impressive advances, challenges remain in AAV-based gene therapy. Pre-existing neutralizing antibodies against AAV capsids can prevent successful transduction after systemic administration. Additionally, capsid-specific T-cell responses may lead to destruction of transduced hepatocytes, potentially reducing long-term expression. Scientists are actively developing strategies to mitigate these immune responses, including improved screening methods and refined vector designs.
The scientific foundations underlying gene therapy for hemophilia B continue to evolve, bringing us closer to effective cures for hemophilia with each technological advancement.
Clinical Evidence Supporting Gene Therapy Efficacy
Gene therapy has ushered in a new era in hemophilia treatment, moving beyond symptom management toward long-term disease modification—and potentially, a functional cure. A growing body of high-quality clinical trial data, including multiple Phase 1 through Phase 3 studies, provides robust evidence supporting the efficacy of gene therapy in both hemophilia A and B. These trials have consistently shown substantial and sustained improvements in bleeding outcomes, factor activity levels, and overall patient quality of life.
Reduction in Annual Bleeding Rates
The annualized bleeding rate (ABR) has emerged as a critical endpoint in hemophilia trials, providing a personalized measure of treatment efficacy beyond factor level measurements. Across various gene therapy clinical studies, ABR reductions have been dramatic and sustained:
- Etranacogene dezaparvovec (Hemgenix): Four years post-treatment, patients demonstrated a 90% decrease in bleeding rates compared to pre-treatment levels, with mean adjusted ABR falling to merely 0.4 bleeds per year. Notably, joint bleeds decreased from 2.34 per year during prophylaxis to less than 0.1 per year. In phase 3 trials, the therapy achieved statistical superiority with an ABR ratio of 0.36 compared to prior prophylaxis.
- Valoctocogene roxaparvovec: Phase 3 data showed an 84% reduction in mean ABR from 4.8 at baseline to 0.8 post-infusion (p<0.0001), with 80% of participants becoming bleed-free from week 5 onward. Long-term followup revealed mean FVIII activity of 9.8% and median activity of 5.6% at 6 years post-treatment.
- Giroctocogene fitelparvovec: Recent phase 3 AFFINE trial results indicated a statistically significant reduction in mean total ABR from 4.73 pre-treatment to 1.24 post-gene therapy (p=0.0040). Even more impressive, treated ABR showed a 98.3% reduction from 4.08 to just 0.07 (p<0.0001).
These improvements translate directly to patient quality of life. A University of Pennsylvania-led international Phase III trial documented a 71% average reduction in bleeding episodes after a single infusion of gene therapy, with more than half of the 45 participants experiencing no bleeds whatsoever after treatment.
Although individual variation exists, the data consistently reveals that gene therapy significantly outperforms traditional prophylactic factor replacement across all major clinical trials. In six comparative studies, bleeding reductions ranged from 64% to 96% in hemophilia B gene therapy and 83.8% to 91.5% in hemophilia A gene therapy.
Factor Level Restoration to Normal or Near-Normal Ranges
Beyond bleeding prevention, gene therapy has demonstrated remarkable capacity to restore coagulation factor activity to clinically meaningful levels:
For hemophilia B patients, etranacogene dezaparvovec produced mean factor IX activity of 37.4% at four years post-treatment—well above the 5% threshold considered sufficient to prevent spontaneous bleeding episodes. Similarly, at 18 months post-injection in the HOPE-B trial, average factor IX activity reached 34.3%, allowing 52 of 54 participants to discontinue prophylactic therapy.
In the hemophilia A context, early-phase trials of BioMarin’s gene therapy maintained factor VIII levels at 50% two years after infusion. The phase 3 valoctocogene roxaparvovec trial showed mean factor VIII activity increases of 41.9 IU/dL by week 52 after administration.
Interestingly, factor level restoration varies between products and sometimes exceeds normal ranges. The FDA-approved Roctavian for hemophilia A increased factor VIII activity above normal limits in some cases. Likewise, FLT180A produced factor IX activity up to 310% in one patient, though this extreme elevation was associated with thrombotic complications.
Regarding durability, the longest follow-up data (12-15 years) from four patients receiving AAV2-hFIX demonstrated sustained safety without hepatotoxicity or hepatocellular carcinoma. For fidanacogene elaparvovec, average factor IX activity remained at 19.8% after 5 years compared to 25.4% in the first year—showing some decline but still maintaining clinically significant levels.
Clinical observations also confirm practical benefits beyond laboratory measurements. Fidanacogene elaparvovec recipients who underwent surgery experienced no excess bleeding, thereby confirming that the restored clotting factors function effectively during physiological challenges.
Though individual responses vary considerably, these results collectively demonstrate gene therapy’s potential as not merely a treatment but possibly among the first actual cures for hemophilia.
FDA-Approved Gene Therapies for Hemophilia
Recent regulatory milestones have brought gene therapy from experimental status to approved treatment options for hemophilia patients. As of 2024, three gene therapies have received FDA approval, representing a paradigm shift in hemophilia management after decades of factor replacement therapy.
Etranacogene Dezaparvovec (Hemgenix): Mechanism and Approval
Etranacogene dezaparvovec, marketed as Hemgenix, made history as the first FDA-approved gene therapy for hemophilia when authorized in November 2022. This one-time treatment utilizes an adeno-associated virus serotype 5 (AAV5) vector carrying a codon-optimized DNA sequence of the gain-of-function Padua variant of human Factor IX controlled by a liver-specific promoter.
The therapy’s approval encompasses adult hemophilia B patients who:
- Currently use Factor IX prophylaxis therapy
- Have current or historical life-threatening hemorrhage
- Experience repeated, serious spontaneous bleeding episodes
Mechanistically, Hemgenix targets hepatocytes through intravenous administration, enabling these cells to produce the missing clotting factor. Following treatment, average Factor IX levels reached 42% after one year and stabilized at 37% after two years. Remarkably, in phase 3 clinical trials, 96% of patients who received Hemgenix discontinued prophylactic factor replacement and maintained this independence for at least two years.
The therapy’s approval was based on data from 57 men with severe or moderately severe hemophilia B. In the HOPE-B trial, a single dose increased factor IX activity to 39% at 6 months and 36.7% at 24 months after infusion. This translated to clinical benefits with 64% of respondents experiencing zero bleeds and an average of just 1.1 bleeds per year.
Undoubtedly, the high cost of Hemgenix—$3.5 million per one-time treatment—has generated discussion, yet this must be considered against lifetime factor replacement therapy costs.
Valoctocogene Roxaparvovec: Clinical Trial Results
The FDA approved valoctocogene roxaparvovec (Roctavian) in June 2023 for adults with severe hemophilia A lacking pre-existing antibodies to AAV5. Essentially, this adeno-associated virus vector-based gene therapy delivers a functional gene for clotting Factor VIII, enabling patients to produce this crucial protein independently.
Primary efficacy data came from the pivotal Phase 3 GENEr8-1 trial involving 134 adult men with severe hemophilia A. Four-year follow-up data revealed enduring clinical benefits:
- 73.6% of patients (81/110) experienced zero treated bleeds during year four
- Mean FVIII activity at year four measured 27.1 IU/dL by one-stage assay and 16.1 IU/dL by chromogenic assay
- Over the entire four-year period, mean annualized bleeding rate was 0.8 bleeds/year for treated bleeds
Correspondingly, treatment impact extended beyond bleeding control to quality of life improvements. Regardless of factor VIII levels achieved, patients reported significant enhancements in physical functioning, role functioning, and reduced consequences of bleeding.
Presently, long-term durability data continues to accumulate. Phase 2 results show that the majority of adults maintained bleed control seven years after infusion.
Fidanacogene Elaparvovec (BEQVEZ): Safety and Efficacy Profile
BEQVEZ (fidanacogene elaparvovec) received FDA approval for adult hemophilia B patients who are negative for neutralizing antibodies to AAV serotype Rh74var. This gene therapy uses an adeno-associated virus vector to deliver a high-activity FIX variant.
The BENEGENE-2 pivotal trial demonstrated BEQVEZ’s efficacy with:
- Mean annualized bleeding rate reduction from 4.5 pre-treatment to 2.5 post-administration
- Median ABR of zero (range 0-19) during the efficacy evaluation period
- Complete elimination of bleeds in 60% of patients compared to 29% in the prophylaxis arm
Regarding safety, BEQVEZ has displayed a favorable profile. Throughout follow-up of up to 6 years, investigators reported no thrombotic complications, factor IX inhibitors, or malignancies. Most adverse events occurred in the first year, with the most common being elevated transaminases (liver enzymes).
A noteworthy practical consideration for patients contemplating this therapy: those requiring corticosteroids due to elevated liver enzymes should receive gastric protection to prevent potential complications. Accordingly, safety monitoring includes regular laboratory assessments for liver function, FIX activity levels, and FIX inhibitors.
Generally, as with other gene therapies, BEQVEZ offers the potential to transform treatment by replacing frequent infusions with a one-time intervention, potentially providing lasting relief from the significant management burden associated with hemophilia B.
Comparing Gene Therapy to Traditional Treatments and Cures for Hemophilia
Traditional hemophilia treatments have provided crucial care for decades, yet come with substantial limitations that potential cures for hemophilia through gene therapy aim to address. The contrast between these approaches extends beyond clinical efficacy to quality of life and economic considerations.
Factor Replacement Therapy Limitations
Factor replacement therapy, the historical cornerstone of hemophilia management, presents several ongoing challenges:
- Treatment burden: Patients require frequent intravenous infusions, often several times weekly, creating a substantial mental and physical burden.
- Incomplete protection: Current prophylactic regimens, though effective, do not completely prevent joint disease over a lifetime.
- Inhibitor development: Approximately 30% of severe hemophilia A patients develop neutralizing antibodies against infused factor, significantly complicating treatment.
- Trough level issues: Between doses, factor levels typically hover between 1-4%, potentially allowing subclinical bleeding that contributes to long-term joint damage.
Beyond clinical limitations, patients face a lifetime of expensive, time-consuming treatments. Many patients report this ongoing regimen negatively impacts quality of life.
Non-Factor Therapies: Emicizumab and Beyond
In response to factor replacement limitations, non-factor therapies have emerged as an intermediate step between traditional treatments and gene therapy. Emicizumab (Hemlibra), a bispecific monoclonal antibody, revolutionized care by offering subcutaneous administration with equivalent safety and efficacy to traditional concentrates.
Non-factor therapies offer distinct advantages: they generate stable thrombin levels, feature longer half-lives, and effectively shift severe hemophilia toward milder phenotypes. Primarily, these treatments work through two mechanisms—either mimicking missing factors or rebalancing the coagulation system by inhibiting anticoagulant pathways.
Notably, emicizumab has captured substantial market share, with over 12,500 patients using it since 2018, generating $3.6 billion in sales in 2022. Nevertheless, gene therapy for hemophilia B has demonstrated superior outcomes by potentially eliminating the need for any routine prophylaxis.
Cost-Benefit Analysis Over Patient Lifetime
Despite gene therapies’ hefty upfront costs—ranging from $2-4 million per treatment—economic analyzes consistently show long-term value. One comprehensive study found gene therapy for severe hemophilia B to be more cost-effective than both on-demand and prophylactic factor replacement therapies, considering a $150,000/quality-adjusted life-year threshold.
The stark cost comparison becomes apparent when considering that prophylaxis with factor IX often costs $750,000 per year or more. Hence, gene therapy becomes economically advantageous typically within 3-5 years. Furthermore, sensitivity analyzes estimated gene therapy to be cost-effective in 92% of simulations.
Moreover, the impact extends beyond direct medical costs. Gene therapy eliminates the administration of prophylactic factor concentrate replacement, leading to substantial improvements in patient quality of life. In one modeled cohort of men with severe hemophilia A, projected cost savings ranged from $854 to $982 million over 10 years if just 10% received gene therapy.
First, outcomes-based payment agreements may help mitigate payer risks while facilitating patient access. Second, the durability of treatment effect remains critical to the economic equation—with models assuming at least 10 years of effectiveness to realize the projected cost advantages.
Safety Considerations in Hemophilia Gene Therapy
While gene therapies represent promising cures for hemophilia, their integration into clinical practice necessitates careful consideration of safety profiles. Current data reveals distinct safety patterns requiring specialized monitoring and management strategies.
Liver Enzyme Elevations and Management
Hepatic enzyme elevations represent the most common adverse event following hemophilia gene therapy administration. The prevalence varies significantly between hemophilia types:
- Hemophilia A recipients experience ALT elevations at rates of 71-89%
- Hemophilia B patients show lower incidence, ranging from 17-32%
Typically, these elevations occur within 8 weeks post-infusion, subsequently requiring prompt intervention with immunosuppressive therapy. Most trials employ oral corticosteroids (primarily prednisone/prednisolone) at initial doses around 60mg daily. Treatment duration varies considerably—hemophilia A patients often require prolonged immunosuppression (median 230 days) versus hemophilia B patients (median 78 days).
Importantly, elevated transaminases may correlate with reduced factor expression, yet prompt initiation of immunosuppression often preserves therapeutic benefit. In HOPE-B trial participants receiving etranacogene dezaparvovec, factor IX activity remained relatively stable throughout corticosteroid treatment cycles.
Neutralizing Antibodies: Impact on Treatment Efficacy
Pre-existing immunity against AAV vectors poses substantial challenges for gene therapy eligibility. Approximately 46.9-53.4% of hemophilia patients demonstrate neutralizing antibodies (NAbs) against common AAV serotypes. Furthermore, concurrent antibodies against multiple serotypes are common—around 38.2% of patients exhibit antibodies against all three major AAV serotypes.
Thereafter, most clinical trials establish eligibility cutoffs based on neutralizing antibody titers. Interestingly, etranacogene dezaparvovec maintains efficacy with pre-existing AAV5 neutralizing antibody titers ≤678, thereby expanding treatment accessibility. Most other therapies currently exclude patients with detectable NAbs, limiting patient eligibility substantially.
Long-Term Monitoring Requirements
Following gene therapy administration, patients require structured long-term monitoring. First-year surveillance typically includes:
- Weekly laboratory assessments for 12-26 weeks
- Thereafter transitioning to monthly or quarterly visits
- Regular monitoring of liver function, factor activity levels, and imaging studies
Beyond year one, monitoring generally continues with decreasing frequency—quarterly in year two, then semi-annually or annually thereafter. Monitoring should primarily focus on liver health, with some experts recommending hepatocellular carcinoma screening every six months.
Overall, current safety data reveals no evidence of insertional mutagenesis or treatment-related serious adverse events in approved gene therapies. Nevertheless, continued vigilance through patient registries like the WFH Gene Therapy Registry remains essential for thorough long-term safety assessment.
Future Directions in Hemophilia Gene Therapy Research
Beyond the currently approved gene therapies, several innovative approaches are under development to address remaining challenges in hemophilia treatment. These emerging technologies aim to overcome limitations of existing therapies, particularly focusing on improved durability, pediatric applications, and reaching patients with pre-existing immunity to AAV vectors.
CRISPR-Cas9 Gene Editing Approaches
CRISPR-Cas9 technology represents a significant advancement toward potential cures for hemophilia through permanent correction of gene mutations. Unlike AAV vectors that primarily remain episomal, CRISPR systems can edit the genome directly. Recent studies demonstrate promising results:
- Base editing with SpCas9-NG achieved twofold greater efficiency in repairing hemophilia B mutations compared to wild-type SpCas9
- The broad PAM flexibility of SpCas9-NG enables targeting approximately four times more mutation sites with recognition of just one G nucleotide
- Combination approaches using AAV8 vectors delivering wild-type FIX alongside lipid nanoparticles carrying Cas9 mRNA have achieved curative FIX levels in both murine and non-human primate models
Notably, mice dosed as neonates showed stable FIX levels, in contrast to animals treated with episomal AAV-FIX, which partially lost expression with growth. This advantage makes CRISPR particularly promising for pediatric patients.
Lentiviral Vector Development
Lentiviral vectors offer distinct advantages over AAV, especially regarding integration and neutralizing antibodies. Three clinical programs using integrating lentiviral vector gene therapy have recently initiated trials. First, a groundbreaking study targeting bone marrow stem cells demonstrated expression of FVIII protein in five hemophilia A patients with factor VIII activity ranging from 1.7 to 39.9 IU/dL and remained stable throughout follow-up.
Importantly, this approach could make gene therapy accessible to more patients, including children. Furthermore, lentiviral vectors targeting FVIII expression specifically to platelets (under ITGA2B promoter) show promise even for patients with inhibitors, as platelet-derived FVIII corrected hemostasis in animal models with pre-existing anti-FVIII.
Non-Viral Delivery Systems
Currently, non-viral gene delivery systems show considerable potential in overcoming immunogenicity concerns. Key advantages include:
- Reduced immunogenicity without pre-existing immunity limitations
- Transient expression of CRISPR-Cas9 leading to fewer off-target effects
- Potential for repeat dosing to achieve desired expression levels
Particularly promising is the piggyBac DNA insertion system, which enables delivery of large transgenes. When administered to hemophilia A mice, LNP-delivered piggyBac achieved sustained hFVIII expression of approximately 30% of normal levels over 7 months. More remarkably, immunocompetent adult hemophilia A mice showed dose-proportionate increases in FVIII activity after repeated administrations, reaching 96% of normal activity following three doses.
Yet challenges remain—non-viral LNP-based delivery has demonstrated lower efficacy when transitioning from small to large animal models. Additionally, the Super piggyBac transposase system shows nonspecific genome integration despite preference for intergenic regions.

Conclusion
Gene therapy stands as a transformative advancement in hemophilia treatment, demonstrating unprecedented success through multiple FDA-approved therapies. With robust safety profiles, long-term efficacy, and curative potential, it is reshaping treatment expectations. However, thoughtful implementation is essential—particularly in managing liver enzyme elevations, screening for pre-existing immunity, and committing to long-term monitoring.
Looking forward, emerging platforms such as CRISPR-Cas9, lentiviral vectors, and non-viral delivery systems promise to further enhance durability, safety, and accessibility. These innovations may extend gene therapy to pediatric populations, patients with inhibitors, and those previously excluded due to vector immunity.
By continuing to refine these therapies and invest in rigorous safety monitoring, gene therapy will likely become a cornerstone of hemophilia care—replacing chronic treatment with lasting freedom from disease.
Frequently Asked Questions:
FAQs
Q1. How effective is gene therapy in treating hemophilia? Gene therapy has shown remarkable efficacy in treating hemophilia, with clinical trials demonstrating up to 90% reduction in annual bleeding rates and restoration of clotting factor levels to 34-42% of normal ranges. Many patients have been able to discontinue regular factor replacement therapy after receiving gene therapy.
Q2. Are there FDA-approved gene therapies for hemophilia? Yes, there are FDA-approved gene therapies for hemophilia. Etranacogene dezaparvovec (Hemgenix) for hemophilia B, valoctocogene roxaparvovec (Roctavian) for hemophilia A, and fidanacogene elaparvovec (BEQVEZ) for hemophilia B have all received FDA approval as of 2024.
Q3. What are the main safety concerns with hemophilia gene therapy? The primary safety considerations for hemophilia gene therapy include liver enzyme elevations, which are more common in hemophilia A patients, and the potential impact of pre-existing neutralizing antibodies against the viral vectors used. Long-term monitoring is essential to assess ongoing safety and efficacy.
Q4. How does gene therapy compare to traditional hemophilia treatments? Gene therapy offers potential long-term benefits over traditional factor replacement therapies, including reduced treatment burden, improved quality of life, and potential cost-effectiveness over time. However, it comes with a high upfront cost and requires careful patient selection and monitoring.
Q5. What new approaches are being researched for hemophilia gene therapy? Future directions in hemophilia gene therapy research include CRISPR-Cas9 gene editing techniques, lentiviral vector development, and non-viral delivery systems. These approaches aim to improve treatment durability, expand eligibility to more patients, and potentially offer solutions for pediatric patients.