Medical Policy:
08.01.065-001
Topic:
Gene Therapies for Congenital Hemophilia A or B (Hemgenix, Roctavian)
Section:
Injections
Effective Date:
November 1, 2025
Issued Date:
August 19, 2025
Last Revision Date:
September 2024
Annual Review:
August 2026
 
 

Populations

Interventions

Comparators

Outcomes

 

Individuals:

Who are adults with congenital hemophilia B who currently use factor IX prophylaxis therapy, or have current or historical life- threatening hemorrhage, or have repeated, serious spontaneous bleeding episodes

 

 

 

 

 

 

 

Interventions of interest are:

Etranacogene dezaparvovec- drlb

 

 

 

 

 

 

Comparators of interest are:

Continued medical management (prophylactic exogenous factor replacement)

 

 

 

Relevant outcomes include:

Disease-specific survival

Change in disease status

Quality of life Resource utilization Treatment-related mortality Treatment-related morbidity

 

 

 

 

 

Individuals:

Who are adults with congenital severe hemophilia A

 

 

 

 

 

Interventions of interest are:

Valoctocogene roxaparvovec- rvox

 

 

 

 

Comparators of interest are:

Continued medical management (prophylactic exogenous factor replacement)

 

Relevant outcomes include:

Disease-specific survival

Change in disease status

Quality of life Resource utilization Treatment-related mortality Treatment-related morbidity

 

Summary

Description

Most commonly, hemophilia is a recessive X-linked congenital bleeding disorder that predominantly affects males caused by deficiency of coagulation factor VIII (hemophilia A) or factor IX (hemophilia B). Deficiency or absence of clotting factor results in impaired hemostasis, prolonged bleeding, and rebleeding. Etranacogene dezaparvovec-drlb (Hemgenix), and Valoctocogene roxaparvovec-rvox (Roctavian)are gene therapies designed to deliver a copy of the gene for the clotting factor using adeno-associated virus vector. Etranacogene dezaparvovec-drlb (Hemgenix) is designed to deliver a copy of the gene encoding the Padua variant of human coagulation factor IX (hFIX Padua). Valoctocogene roxaparvovec-rvox (Roctavian)is designed to deliver a functional copy of transgene encoding the B-domain deleted SQ form of human coagulation factor VIII.

Summary of Evidence

For individuals who are adults with congenital hemophilia B who currently use factor IX prophylaxis therapy, or have current or historical life-threatening hemorrhage, or have repeated, serious spontaneous bleeding episodes who receive Etranacogene dezaparvovec-drlb (Hemgenix), the evidence includes a single, prospective, single- arm study. Relevant outcomes are disease-specific survival, change in disease status, quality of life, resource utilization, treatment-related mortality and morbidity. The pivotal, open-label, phase III single-arm HOPE-B study enrolled 54 adult males with severe (factor IX <1%) or moderately severe (factor IX 1% to 2%) hemophilia. Of the 54 participants, 53 were included in the efficacy analysis. The estimated mean annualized bleeding rate (ABR) during months 7 to 18 after treatment with Etranacogene dezaparvovec-drlb (Hemgenix) was 1.9 bleeds/year (95% confidence interval [CI], 1.0 to 3.4) compared with an estimated mean ABR of 4.1 (95% CI, 3.2 to 5.4) during the lead-in period. The ABR ratio (months 7 to 18 post-treatment/lead-in) was 0.46 (95% CI, 0.26 to 0.81) demonstrating non-inferiority of ABR during months 7 to 18 compared to the lead-in period. The ABR represents an appropriate clinical benefit endpoint for individuals with hemophilia B and the evidence of clinical benefit was demonstrated by reduction of bleeds in the efficacy evaluable period post treatment. Limitations include uncontrolled study design, limited sample size, and relatively short follow-up which is inadequate to assess durability of treatment effect and safety, especially those adverse events that are potentially rare or have delayed onset. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals who are adults with congenital hemophilia A who receive valoctocogene roxaparvovec-rvox, the evidence includes a single, prospective, single-arm study. Relevant outcomes are disease-specific survival, change in disease status, quality of life, resource utilization, treatment-related mortality and morbidity. In the pivotal, open-label, phase III single-arm study, 134 study participants received a single intravenous infusion of valoctocogene roxaparvovec-rvox. Of the 134 participants, 112 were included in the efficacy analysis. The mean ABR after treatment with Valoctocogene roxaparvovec-rvox (Roctavian)was 2.6 bleeds/year compared with a mean ABR of 5.4 during the lead-in period yielding a mean difference of -2.8 (95% CI, -4.3 to 1.2) bleeds/year. This was within the pre-specified non-inferiority margin of 3.5. The ABR represents an appropriate clinical benefit endpoint for individuals with hemophilia A and the evidence of clinical benefit was demonstrated by reduction of bleeds during the post-treatment period. However, factor levels declined over time and therefore benefits of Valoctocogene roxaparvovec-rvox (Roctavian)could be relatively short-lived. According to the label, a total of 5 participants (4%) did not respond and 17 (15%) lost response to treatment over a median time of 2.3 years (range: 1.0 to 3.3). In the directly enrolled population with a longer follow-up, a total of 1 participant (5%) did not respond and 6 (27%) lost response to treatment over a median time of 3.6 years (range: 1.2 to 4.3). Limitations include uncontrolled study design, limited sample size, and relatively short follow-up. There is considerable uncertainty about the long-term net benefits of Valoctocogene roxaparvovec-rvox (Roctavian)compared with factor VIII prophylaxis. It is not yet clear that the initial increase in factor VIII levels will be maintained for decades. In addition, there are uncertainties about the long-term impact of the therapy on liver function and the risk for hepatocellular carcinoma as limited sample size is prone to uncertainty around the estimates for adverse events. Some serious harms are likely rare occurrences and as such may not be observed in small trials. Long-term follow- up (>15 years) is required to establish precision around durability of the treatment effect and safety. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

This policy is designed to address medical guidelines that are appropriate for the majority of individuals with a particular disease, illness, or condition. Each person's unique clinical circumstances may warrant individual consideration, based on review of applicable medical records.

Policy Position Coverage is subject to the specific terms of the member's benefit plan.

Etranacogene dezaparvovec-drlb (Hemgenix)

Etranacogene dezaparvovec-drlb (Hemgenix) is considered medically necessary for individuals if they meet criteria 1 through 11:

1.  18 years of age or older.

2.  Assigned male at birth.

3.  Severe or moderately severe hemophilia B as defined by a plasma factor IX (FIX) activity level ≤2%.

4.  Currently receiving FIX prophylaxis.

5.  Current or historical life-threatening hemorrhage OR repeated, serious spontaneous bleeding episodes.

6.  No history of FIX inhibitors or a positive screen results of ≥0.6 Bethesda Units (BU) using the Nijmegen- Bethesda assay.

 

7.  A baseline liver health assessment including enzyme testing [alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and total bilirubin] AND a hepatic ultrasound and elastography.

8.  No history of receiving gene therapy or under consideration for treatment for another gene therapy for hemophilia B.

9.  HIV negative or a controlled HIV infection.

10.  No active hepatitis B and/or hepatitis C infection.

Etranacogene dezaparvovec-drlb (Hemgenix) is considered investigational when the above criteria are not met.

Etranacogene dezaparvovec-drlb (Hemgenix) is considered investigational for all other indications.

Repeat treatment with Etranacogene dezaparvovec-drlb (Hemgenix) is considered

investigational.

Valoctocogene roxaparvovec-rvox

Valoctocogene roxaparvovec-rvox (Roctavian)is considered medically necessary for individuals if they meet criteria 1 through 11 :

1.  18 years of age or older.

2.  Assigned male at birth.

3.  Severe or moderately severe hemophilia A as defined by residual factor VIII (FVIII) levels ≤1 IU/dL.

4.  Currently receiving FVIII prophylaxis.

5.  No history of FVIII inhibitors or a positive screen results of ≥0.6 BU using the Nijmegen-Bethesda assay.

6.  No detectable pre-existing antibodies to the adeno-associated virus serotype 5 (AAV5) capsid.

7.  A baseline liver health assessment including but not limited to ALT.

8.  Educated regarding alcohol abstinence and concomitant use of certain medications (e.g., isotretinoin, efavirenz).

9.  No history of receiving gene therapy or under consideration for treatment for another gene therapy for hemophilia A.

10.  HIV negative or controlled HIV infection.

11. No active hepatitis B and/or hepatitis C infection.

Valoctocogene roxaparvovec-rvox (Roctavian)is considered investigational when the above criteria are not met. Valoctocogene roxaparvovec-rvox (Roctavian)is considered investigational for all other indications.

Repeat treatment with Valoctocogene roxaparvovec-rvox (Roctavian)is considered investigational.

 

POLICY GUIDELINES

Etranacogene dezaparvovec-drlb (Hemgenix)

 

Recommended Dose

 

The minimum recommended dose is 2 x 1013 genome copies (gc) per kg of body weight.

Dosing Limits

 

1 injection per lifetime.

Other Considerations

 

Etranacogene dezaparvovec-drlb (Hemgenix) was not studied in individuals assigned female at birth.

A baseline liver health assessment is recommended before starting treatment with Etranacogene dezaparvovec-drlb (Hemgenix). In cases of radiological liver abnormalities and/or sustained liver enzyme elevations, the prescriber is recommended to consider a consultation with a hepatologist to assess eligibility.

Where feasible, the individual should receive periodical monitoring for hepatotoxicity, hepatocellular carcinogenicity, factor IX (FIX) activity, and FIX inhibitors.

 

Valoctocogene roxaparvovec-rvox

Recommended Dose

 

The minimum recommended dose is 6 X 1013 vector genomes (vg) per kg of body weight.

Dosing Limits

 

1 injection per lifetime.

Other Considerations

 

Valoctocogene roxaparvovec-rvox (Roctavian)was not studied in individuals assigned female at birth.

It is recommended that prescribers perform regular alanine aminotransferase (ALT) testing at a certain frequency to monitor for elevations. Elevated liver enzymes, especially elevated ALT, may indicate immune - mediated hepatotoxicity and may be associated with a decline in factor VIII (FVIII) activity.

It is also recommended that prescribers monitor FVIII activity at the same frequency of ALT monitoring unless there are other clinical factors requiring additional monitoring (e.g., FVIII activity ≤5 IU/dL and evidence of bleeding). It may take several weeks after the Valoctocogene roxaparvovec-rvox (Roctavian)infusion before valoctocogene roxaparvovec-rvox-derived FVIII activity rises to a level sufficient for prevention of spontaneous bleeding episodes. Therefore, continued routine prophylaxis support with exogenous FVIII or other hemostatic products used in the management of hemophilia A may be needed during the first few weeks after infusion. After those initial weeks post-infusion, individuals should no longer require prophylaxis support with exogenous FVIII or other hemostatic products.

The use of the adeno-associated virus (AAV) vector DNA may carry the theoretical risk of hepatocellular carcinoma. It is recommended that prescribers monitor individual with risk factors for hepatocellular carcinoma with regular liver ultrasound and alpha-fetoprotein testing for 5 years after administration.

HCPCS

J1411

Injection, Etranacogene dezaparvovec-drlb (Hemgenix), per therapeutic dose

 

J1412

Injection, valoctocogene roxaparvovec-rvox, per mL, containing nominal 2 × 10^13 vector genomes

 

C9399

Unclassified drugs or biologicals

 

J3490

Unclassified drugs

 

J3590

Unclassified biologics




 

D66

Hereditary factor VIII deficiency

 

D67

Hereditary factor IX deficiency

 

D68.311

Acquired hemophilia



Reference to Our Policy Information Guidelines

BACKGROUND

Congenital Hemophilia

Most commonly, hemophilia is an inherited X-linked recessive congenital disorder that predominantly affects males caused by deficiency of coagulation factor VIII (FVIII; hemophilia A) and factor IX (FIX; hemophilia B). In Hemophilia A, variants in the FVIII gene lead to the associated impairment of the normal coagulation cascade.1, In hemophilia B, variant in the F9 gene results in deficiency or functional defectiveness of FIX.2,3,

Hemophilia affects more than 1.2 million individuals (mostly males) worldwide.4, Hemophilia A is more common than hemophilia B. Typically, the reported incidence of hemophilia A is approximately 1 in 4000 to 1 in 5000 live male births while incidence of hemophilia B has been reported to occur in approximately 1 in 15,000 to 1 in 30,000 live male births. Approximately one-third to half have severe disease (FIX activity <1% of normal).4,5,The exact prevalence of hemophilia in the United States (US) is not known but is estimated to be around 33,000 based on data during the period 2012 to 2018.6,Approximately 77% of all hemophiliacs in the US have hemophilia A, of which 60% may have severe disease. The estimated incidence of hemophilia A in the US is 1:5000 live male births. This translates to approximately 400 infants born each year with hemophilia

A. There is no clear effect of geography itself on incidence or prevalence. All races and ethnic groups are

equally affected.7,8,9,World Federation of Hemophilia (WFH) data from 1998 to 2006 indicate a global trend of increased prevalence of hemophilia A in approximately 80% of surveyed countries.10, Potential contributing factors include increased survival, improved diagnostic capabilities, a broader use of national registries and migration from areas with limited access to healthcare to areas with better access. The estimated number of prevalent cases of hemophilia B in the US is between 6300 and 7600 as of 2018.11, Reported prevalence rate of hemophilia B was estimated at 3.7 per 100,000 male population while the incidence rate was estimated at

5.3 per 100,000 male births, or 1 case per 19,283 live male births. Worldwide, there are approximately 33,000 people living with hemophilia B as of 2020.12,

The severity of hemophilia has generally been defined by factor levels.13, Severity based on factor levels does not perfectly correlate with any individual’s clinical severity, but no other classification system is widely accepted.14, Disease severity using factor level classifications is summarized in Table 1. Individuals with more severe hemophilia are more likely to have spontaneous bleeding, severe bleeding, and an earlier age of first bleeding episode, which can begin as early as birth. Those with severe disease, are at risk for potentially life threatening bleeding episodes and debilitating long-term complications.1, Individuals with severe hemophilia typically experience frequent, spontaneous bleeds (1 to 2 times per week) in their muscles or joints.15, Repeated, spontaneous bleeding in the joints (hemarthrosis) results in joint inflammation and damage to joint cartilage and synovium leading to hemophilic arthropathy.16, According to 1 study, hemophilic arthropathy was observed in >90% of those with severe hemophilia before the age of 30 years.17, Severe hemophilia is almost exclusively a disease of males, although females can be affected in some rare cases (eg, compound heterozygosity; skewed lyonization; X chromosome loss). In contrast, mild hemophilia has been reported in up to one-quarter of female carriers who are heterozygotes. Most commonly, hemophilia is inherited. However, sporadic disease (without a positive family history, presumed due to a new variant) is also common. Studies have demonstrated that sporadic causes account for as much as 55% of cases of severe hemophilia A and 43% of cases of severe hemophilia B.18, In moderate and mild hemophilia A and B, approximately 30% are sporadic cases.

Table 1. Hemophilia Severity, Factor Levels and Symptoms15,

 

Severity of Hemophiliaa

Clotting Factor levels

Symptoms

 

 

Mild

 

5% to 40% of normal

 

Might bleed for a long time after surgery, dental extraction, or a very bad injury

Rarely bleeds unless injured (rarely has spontaneous bleeding)

 

 

 

Moderate

 

 

1% to 5% of normal

 

Might bleed for a long time after surgery, a bad injury, or dental work

Might bleed for no clear reason (occasional spontaneous bleeding)

 

Severe

 

Below 1% of normal

 

Bleed often into the joints and sometimes the muscles Can bleed for no obvious reason (spontaneous bleeding)

 

a Severity of hemophilia is measured in percentage of normal factor activity in the blood, or in number of international units (IU) per milliliter (mL) of whole blood. The normal range of clotting factor VIII or IX in the blood is 40% to 150%. People with factor activity levels of less than 40% are considered to have hemophilia. Some people’s bleeding pattern does not match their baseline level. In these cases, the phenotypic severity (bleeding symptoms) is more important than the baseline level of factor in deciding upon treatment options.

 

Diagnosis

Hemophilia should be suspected in individuals who present with a history of easy bruising; “spontaneous” bleeding (i.e., bleeding for no apparent/known reason), particularly into the joints, muscles, and soft tissues; excessive bleeding following trauma or surgery. Diagnosis is made by assessing the patient’s personal and family history of bleeding and is confirmed through screening tests, including a complete blood count test and a blood coagulation tests, typically activated partial thromboplastin clotting time (aPTT) and a prothrombin time (PT) test.19, Both tests measure the length of time it takes for blood to clot and are important in identifying the potential cause of bleeding; the aPTT test assesses the clotting ability of factors VIII, IX, XI and XII while the PT assay tests for factors I, II, V, VII and X.20,6, In the event of an abnormal aPTT result, diagnosis of hemophilia A or B is established by the following criteria:

Diagnosis of hemophilia A requires confirmation of a factor VIII activity level below 40% of normal (below 0.40 international units [IU]/mL), or, in some circumstances where the factor VIII activity level is

≥40 percent, a pathogenic variant in the F8 gene. A normal von Willebrand factor antigen (VWF:Ag)

should also be documented to eliminate of the possibility of some forms of von Willebrand disease.

 Diagnosis of hemophilia B requires confirmation of a FIX activity level below 40% of normal, or, in some circumstances where the FIX activity level is ≥40%, a pathogenic variant in the F9 gene. Newborns have a lower normal range of FIX activity; the normal newborn range should be used as a reference when evaluating factor levels in newborns.

Genetic testing is recommended to identify the specific disease-causing gene mutation and evaluate the risk of inhibitor development.19, Diagnosis is usually at a younger age among patients with the severe (≤2 years) or moderate (<5 to 6 years) form of the disorder compared with those with mild disease who are typically diagnosed later in life or in adulthood.8,

Current Treatment

Factor replacement therapy is provided via 1 of 2 modalities: prophylaxis (regular replacement) or on demand (episodic). Prophylaxis is primary (before a bleeding event has occurred) or secondary (a bleeding event has occurred), and continuous or intermittent (eg, for a few months at a time). Individuals with hemophilia, particularly those with severe hemophilia, can be affected by development of inhibitors (antibodies that develop in response to exogenous administration of exogenous factors). In a 13-year US longitudinal study of individuals with hemophilia, 11% to 17% of those with severe hemophilia and 3% of individuals with mild hemophilia developed inhibitors during follow-up.21, The median age of inhibitor development for those with severe hemophilia A was 3 years or less in developed countries, and was approximately 30 years in those with moderate-to-mild hemophilia, often following intensive FVIII exposure with surgery.1, Development of inhibitors is also associated with increased mortality. A retrospective analysis of Centers for Disease Control and Prevention (CDC) surveillance data in individuals with severe hemophilia A reported that odds of death among the subgroup with inhibitors was 70% higher than among the subgroup without inhibitors (p<.01).22, In a retrospective claims analysis conducted in the Netherlands, all-cause mortality rates among individuals with non-severe hemophilia A were 5 times higher in the subgroup with inhibitors when compared with the subgroup without inhibitors.23, Several factor preparations are available for prophylaxis, some prepared from human plasma, some prepared using recombinant technology including some with modifications to extend the half-life of the therapy. An updated table is maintained by the of the Medical and Scientific Advisory Council (MASAC) of the National Hemophilia Foundation (NHF) in the United States (www.hemophilia.org).

Regulatory Status

 

On November 22, 2022, Etranacogene dezaparvovec-drlb (Hemgenix) (Hemgenix; CSL Behring) was approved by the U.S. Food and Drug Administration (FDA) for the treatment of adults with Hemophilia B (congenital FIX deficiency) who currently use FIX prophylaxis therapy, or have current or historical life-threatening hemorrhage, or have repeated, serious spontaneous bleeding episodes.

On June 29, 2023, Valoctocogene roxaparvovec-rvox (Roctavian)(Roctavian; BioMarin Pharmaceutical Inc.) was approved by the U.S. FDA for the treatment of adults with severe hemophilia A (congenital factor VIII deficiency with factor VIII activity < 1 IU/dL) without pre-existing antibodies to adeno-associated virus serotype 5 detected by an FDA-approved test.


Professional Statements and Societal Positions Guidelines

RATIONALE

This evidence review was created in January 2023 and with a search of the PubMed database through une 27, 2024.

Evidence reviews assess the clinical evidence to determine whether the use of a technology improves the net health outcome. Broadly defined, health outcomes are length of life, quality of life, and ability to function including benefits and harms. Every clinical condition has specific outcomes that are important to patients and to managing the course of that condition. Validated outcome measures are necessary to ascertain whether a condition improves or worsens; and whether the magnitude of that change is clinically significant. The net health outcome is a balance of benefits and harms.

To assess whether the evidence is sufficient to draw conclusions about the net health outcome of a technology, 2 domains are examined: the relevance and the quality and credibility. To be relevant, studies must represent one or more intended clinical use of the technology in the intended population and compare an effective and appropriate alternative at a comparable intensity. For some conditions, the alternative will be supportive care or surveillance. The quality and credibility of the evidence depend on study design and conduct, minimizing bias and confounding that can generate incorrect findings. The randomized controlled trial (RCT) is preferred to assess efficacy; however, in some circumstances, nonrandomized studies may be adequate. Randomized controlled trials are rarely large enough or long enough to capture less common adverse events and long-term effects. Other types of studies can be used for these purposes and to assess generalizability to broader clinical populations and settings of clinical practice.

Promotion of greater diversity and inclusion in clinical research of historically marginalized groups (e.g., People of Color [African-American, Asian, Black, Latino and Native American]; LGBTQIA (Lesbian, Gay, Bisexual, Transgender, Queer, Intersex, Asexual); Women; and People with Disabilities [Physical and Invisible]) allows policy populations to be more reflective of and findings more applicable to our diverse members. While we also strive to use inclusive language related to these groups in our policies, use of gender-specific nouns (e.g., women, men, sisters, etc.) will continue when reflective of language used in publications describing study populations.

Congenital Hemophilia B

 

Clinical Context and Therapy Purpose

 

The purpose of gene therapy in adults who have congenital hemophilia B is to provide a treatment option that is an improvement on existing therapies. Potential benefits of this therapy may include the following:

A novel mechanism of action or approach that may allow successful treatment of many individuals for whom other available treatments are not available or have failed or have yielded sub-optimal response.

 

 Reduced treatment complexity such as avoidance of repeated intravenous infusion or subcutaneous injections.

The following PICO was used to select literature to inform this review.

Populations

 

The relevant population of interest is individuals who are adults with congenital hemophilia B who currently use factor IX (FIX) prophylaxis therapy, or have current or historical life-threatening hemorrhage, or have repeated, serious spontaneous bleeding episodes.

Interventions

 

The therapy being considered is Etranacogene dezaparvovec-drlb (Hemgenix) and fidanacogene elaparvovec-dzkt. Etranacogene dezaparvovec-drlb (Hemgenix) is an adeno-associated virus (AAV) based gene therapy using a non- replicating recombinant AAV5 containing a codon-optimized DNA sequence of the gain-of-function Padua variant of human FIX (variant R338L), under control of a liver-specific promotor 1 (LP1).

Comparators

 

Life-long prophylaxis with exogenous factor replacement therapy is currently being used to manage individuals with congenital hemophilia B.

Outcomes

 

The general outcomes of interest are disease-specific survival, change in disease status, health status measures, quality of life, resource utilization, treatment-related mortality and treatment-related morbidity. Relevant outcome measures in alphabetical order are summarized in Table 2.

Table 2. Health Outcome Measures Relevant to Hemophilia B

 

Outcome

Description

Relevance

 

 

 

 

 

Annualized bleeding rate

 

 

 

Applicants of other FDA approved products have relied on ABR to demonstrate clinical benefit.8,

 

Although it is a direct assessment of clinical benefit, it has limitations in that it is a relatively infrequent event in patients on prophylactic factor regimens and the decision by a patient to treat a possible bleeding episode is usually somewhat subjective.8,

 

 

 

 

 

 

Factor activity levels

 

Factor activity levels is an objective endpoint.

For gene therapies, functional and structural differences between the transgene and normal factor protein may present a challenge in understanding the relationship between numerically identical factor activity levels or levels that are within the normal range for the transgene protein and normal factor protein.8,

 

 

To consider factor activity level as a validated surrogate endpoint, clinical data to assess and quantitate the relationship between steady-state factor activity levels and bleeding outcomes in patients with severe hemophilia are critical.8,

 

 

 

 

 

 

 

Haem-A-QoL

 

Patient-reported outcome questionnaire validated for hemophilia; contains 6 domains: Consequences of Bleeding, Emotional Impact, Physical Functioning, Role Functioning, Treatment Concern, and Worry.24,

Subscale and total scores range from 0 to 100.

41 total items scored from 0 to 5 with higher scores indicating better health-related quality of life or less impairment for a particular subscale.

 

 

 

 

 

The Haem-A-QoL has been validated for this population and trials.25,

 

 

 

 

 

 

Joint Activity/Bleeding

 

 

 

 

 

Joint examination using imaging measures such as x-rays, MRI, and ultrasound.

 

No validated scales or instruments that quantify joint damage have been identified. As per ISTH, target joints are major joints with ≥3 bleeding events within 6 months; bleeding in target joints is considered to be resolved if there are ≤2 bleeding events within 12 months.13,

Resource Utilization

 

Repeat intravenous infusions are burdensome and can results in non- adherence.

 

It is estimated that more than 94% of direct healthcare costs for patients with severe hemophilia A are attributable to the cost of FVIII replacement

 

 

 

 

therapy.26,27, Reduction in quantity as well as frequency of exogenous factor FVIII replacement is desirable.

ABR: annualized bleeding rate; FDA: Food and Drug Administration; FVII: factor VIII; ISTH: International Society on Thrombosis and Hemostasis; MRI: magnetic resonance imaging; QoL: quality of life.

 

Study Selection Criteria

 

Methodologically credible studies were selected using the following principles:

To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs;

In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.

To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.

 Consistent with a 'best available evidence approach,' within each category of study design, studies with larger sample sizes and longer durations were sought.

 Studies with duplicative or overlapping populations were excluded.

Etranacogene dezaparvovec-drlb (Hemgenix) Review of Evidence

The clinical development program is summarized in Table 3 and consists of 3 interventional studies (AMT-060- 01, AMT-061-01 and AMT-061-021). All 3 interventional studies are single-arm, open-label trials. Of these, the first 2 studies, AMT-060-01 and AMT-061-01 were phase I/II studies and are not reviewed in detail. The key trial for Etranacogene dezaparvovec-drlb (Hemgenix) is the phase III Hope-B trial (AMT-061-021) that includes 54 participants and is reviewed in detail.

Table 3. Clinical Development Program for Etranacogene dezaparvovec-drlb (Hemgenix)

 

Study

NCT No

Status

Study Dates

Objective

Sample Size

Follow-Up

 

CT-AMT- 060-01

 

NCT02396342

 

Completed and published28,

 

2015 to

2021

To evaluate the long-term safety and efficacy of AMT-060 comprising an AAV5 vector carrying a codon- optimized wild-type F9 transgene

 

10

 

5 years

 

 

CT-AMT- 061-01

 

 

 

NCT03489291

 

 

Ongoing and interim results published29,30,

 

 

2018 to

2023

To confirm the safety and preliminary efficacy endpoints of AMT-061 (Etranacogene dezaparvovec-drlb (Hemgenix)), with a modified F9 transgene encoding the naturally occurring hyperactive mutation, FIX-Padua, in place of wild- type F9

 

 

 

3

 

 

 

5 years

CT-AMT- 061-02 (HOPE- B)

 

NCT03569891

 

Ongoing

 

2018 to

2025

To evaluate the efficacy, and confirm the safety, of Etranacogene dezaparvovec-drlb (Hemgenix) (as a progression of AMT-060).

 

54

 

5 years

 

AAV5: adeno-associated virus serotype 5; FIX-Padua: Padua variant of coagulation Factor IX; NCT: national clinical trial.

 

Nonrandomized Studies

 

Study characteristics and baseline patient characteristics and results are summarized in Table 4 and 5, respectively. The prospective, open-label, single-dose, single arm, multi-national study enrolled adult males aged 19 to 75 years, with severe or moderately severe Hemophilia B. Study design involved a prospective lead-in period of at least 6 months with the intent to receive standard of care routine FIX prophylaxis. Study participants then received a single intravenous dose of Etranacogene dezaparvovec-drlb (Hemgenix) and were then followed up monthly until month 12, and then at 6-month intervals until year 5. The study is on-going. For the efficacy evaluation for the U.S. Food and Drug Administration (FDA) approval, data up to 18 months post- treatment were used.31, Of the 54 study participants, 53 completed at least 18 months of follow-up. One participant, aged 75 with numerous cardiovascular and urologic risk factors, died of urosepsis and cardiogenic shock at month 15 post-dose (at age 77 years) unrelated to treatment. Another participant received around 10% of the intended dose due to an infusion-related hypersensitivity reaction.

The primary efficacy outcome was a non-inferiority test of annualized bleeding rate (ABR) during months 7 to

18 after treatment compared with ABR during the lead-in period. All bleeding episodes, regardless of investigator assessment, were counted. Participants were allowed to continue prophylaxis during months 0 to

6. Results are summarized in Table 6. The ABR ratio (months 7 to 18 post-treatment/lead-in) was 0.46 [95% confidence interval [CI], 0.26 to 0.81] and meets the success criterion where the upper bound of the CI is less than 1.8 demonstrating non-inferiority of ABR during months 7 to 18 compared to the lead-in period. Two study participants were not able to stop routine prophylaxis after treatment with Etranacogene dezaparvovec-drlb (Hemgenix). During months 7 to 18, an additional participant received prophylaxis from days 396 to 534 [approximately 20 weeks].32,

In AAV vector–based gene therapies such as Etranacogene dezaparvovec-drlb (Hemgenix), pre-existing anti-AAV neutralizing antibodies may impede transgene expression at desired therapeutic levels. In the clinical studies, an unvalidated clinical trial assay was utilized to assess pre-existing anti-AAV5 neutralizing antibodies. There were 21 participants with positive neutralizing antibodies to AAV5. These neutralizing antibody titers were measured at baseline prior to infusion of the gene therapy product. The neutralizing antibody titers were in the range of 1:8.5 to 3212. According to the FDA reviewer, overall, there is limited data for participants with positive neutralizing antibody (NAb) titers. One participant with the highest titer of 1:3212, failed treatment, continued on routine prophylaxis with multiple bleeding episodes. The FDA reviewers observed no clear correlation of positive neutralizing antibodies titers and efficacy.31, Nine participants with higher ABRs post treatment compared to baseline included participants with and without neutralizing antibodies. The FDA reviewers noted that 4 participants with positive neutralizing antibodies had much higher ABRs compared to those with negative neutralizing antibodies. According to the prescribing label, individuals who intend to receive treatment with Etranacogene dezaparvovec-drlb (Hemgenix) are encouraged to enroll in a study that evaluates the effect of pre-existing anti-AAV5 neutralizing antibodies on the risk of bleeding.

The most common adverse reactions (incidence ≥5%) were elevated alanine aminotransferase (ALT), headache, blood creatine kinase elevations, flu-like symptoms, infusion-related reactions, fatigue, malaise and elevated aspartate aminotransferase (AST). One death was reported due to cardiogenic shock and was deemed unrelated to treatment.32, Nine participants were treated with corticosteroids for ALT elevation of either >2 times upper limit of normal (ULN; n=8) or >2 x baseline value (n=1). Participants with ALT elevation had approximately 44% lower mean FIX activity at month 18 compared to those that did not have ALT elevation. Study participants (17% or 9/53) that were treated with corticosteroid for ALT elevations exhibited approximately 63% lower mean FIX activity at month 18 compared to those who did not receive corticosteroid coadministration. Participants were treated for 51 to 130 days. ALT elevation is likely the result of T-cell response toward capsid proteins and may cause the lower FIX activity as noted. All participants discontinued steroid use prior to week 26, and no other form of immunosuppression was used in this study.33, As per the prescribing label, integration of liver-targeting AAV vector DNA into the genome may carry the theoretical risk of hepatocellular carcinoma development. As per the label, for individuals with preexisting risk factors (e.g., cirrhosis, advanced hepatic fibrosis, hepatitis B or C, non-alcoholic fatty liver disease, chronic alcohol

 

consumption, non-alcoholic steatohepatitis, and advanced age), regular (e.g., annual) liver ultrasound and alpha-fetoprotein testing should be performed following treatment.32,

Table 4. Summary of Key Nonrandomized Trial

 

Study

Study Type

Country

Dates

Participants

Treatment

Follow-Up

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

HOPE- B32,33,

Open- label, single- arm

US, UK

and EU

2018-

2025

Inclusion

Adult males with severe (FIX <1%) or moderately severe (FIX 1% to 2%) hemophilia Received continuous FIX protein prophylaxis for ≥2 months

>150 previous exposure days of treatment with FIX protein

Exclusion

 

History of FIX inhibitors or positive FIX inhibitor test at screening

Positive HIV test at screening, not controlled with anti- viral therapy

Active infection with hepatitis B or C virus at screening History of hepatitis B or C exposure, currently controlled by antiviral therapy at the end of the lead-in phase Previous gene therapy treatment

 

Screening period of approximately 4 weeks, followed by a ≥26-week lead- in phase with FIX prophylaxis, during which they were evaluated for bleeding events.

After the lead-in period, patients received a single dose of 2 x 1013 gc/kg of etranacogene dezaparvovec- drlb.

78 weeks (intended duration of study 5 years)

EU: European Union; FIX: factor IX; US: United States; UK: United Kingdom.

 

 

Table 5. Summary of Baseline Demographics and Disease Characteristics

 

Patient Characteristics in (HOPE-B)32,33,

N=54

Age, mean (SD, min to max), years

41.5 (15.8, 19 to 75)

Race, n (%)

 

White

40 (74.1)

Black

1 (1.9)

Asian

2 (3.7)

Other

6 (11.1)

Severity of hemophilia B at diagnosis, n (%)

 

Severe (FIX <1%)

44 (81.5)

Moderately severe (FIX ≥1% and ≤2%)

10 (18.5)

Positive HIV status, n (%)

3 (5.6)

Prior hepatitis B infection, n (%)

9 (16.7)

Prior hepatitis C infection, n (%)

31 (57.4)

Pre-screening FIX treatment (n, %)

 

Extended half-life

31 (57.4)

Standard half-life

23 (42.6)

Detectable NAbs at baseline, n (%)

21 (38.8)

Participants with zero bleeds at lead-in, n (%)

14 (25.9%)

 

FIX: factor IX; NAbs: neutralizing antibodies; SD: standard deviation.

 

 

Table 6. Summary of Results

 

 

Outcomes (HOPE-B)32,

 

Pre-Study Perioda

Post-Study Period (months 7 to 18b after Etranacogene dezaparvovec-drlb (Hemgenix) treatment)

Bleeding Related Outcome (Primary)

 

 

All Bleeds

136

96c

Follow-up time (person-year)

33

52

Mean adjusted ABR (95% CI)d

4.1 (3.2 to 5.4)

1.9 (1.0 to 3.4)

Individuals with bleeds

40 (74%)

20 (37%)

Individuals with zero bleeds

14 (26%)

34 (63%)

Observed spontaneous bleed count (proportion of total bleeds)e

50 (37%)

14 (26%)

Observed joint bleed count (proportion of total bleeds)e

77 (57%)

19 (35%)

Mean spontaneous ABR

1.52

0.44

Mean traumatic ABR

2.09

0.62

Number of bleeding episodes

136 (including 118 treated episodes)

54 (including 30 bleeding episodes)

Secondary Outcomes33,

 

 

FIX Activity (One-stage activated partial thromboplastin time-based assay), mean (±SD)

 

1.19 (0.39)

Month 6: 39.0 (18.7)

Month 12: 41.5 (21.7)

Month 18: 36.9 (21.4)

 

 

FIX Activity (chromogenic assay), mean (± SD), range

 

 

Not reported

Month 6: 38.95% ± 18.72, 8.2 to

97.1%)

Month 12: 41.48% ± 21.71, 5.9 to

113.0%)

Month 18: 36.90% ± 21.40, 4.5 to

122.9%)

 

ABR: annualized bleeding rate; CI: confidence interval; FIX: factor IX; SD: standard deviation.

a During the observational lead-in period individuals used their individualized approach to factor IX prophylaxis derived prior to enrollment in the study, rather than a standardized approach to factor IX prophylaxis. Not all individuals complied with their prescribed prophylaxis regimen during the lead-in period.

b Efficacy evaluation started from month 7 after Etranacogene dezaparvovec-drlb (Hemgenix) treatment, to allow factor IX expression to reach a steady state.

c An ABR of 20 was imputed for the period when three individuals were on continuous prophylaxis.

d Non-inferiority comparison and mean ABR estimates were based on a repeated measures generalized estimating equations negative binomial regression model.

e For spontaneous and joint bleed counts, no imputation was done for the three individuals receiving continuous prophylaxis during months 7 to 18.

 

The purpose of the study limitations tables (see Tables 7 and 8) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence and provides the conclusions on the sufficiency of evidence supporting the position statement. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of Etranacogene dezaparvovec-drlb (Hemgenix) in these racial groups. The FDA reviewer noted higher ABRs (during months 7 to 18 compared with the lead-in period) among 14 non-white study participants compared to white study participants.33, Because of the uncontrolled study design, limited sample size and relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of Etranacogene dezaparvovec-drlb (Hemgenix) compared with FIX prophylaxis. It is not yet clear that the initial increase in FIX levels will be maintained for decades. In addition, there are uncertainties about the long-term impact of the therapy on liver function and the risk for hepatocellular carcinoma. The small sample size creates uncertainty around the estimates of adverse events. Some serious harms are likely rare occurrences and as such may not be observed in small trials. Long-term follow-up (>15 years) is required to establish precision around durability of the treatment effect and safety.

 

Table 7. Study Relevance Limitations

 

Study

Populationa

Interventionb

Comparatorc

Outcomesd

Duration of Follow- upe

 

 

HOPE-B 32,33,

4. Enrolled populations do not reflect relevant diversity

 

 

 

1.  Not sufficient duration for benefit

2.  Not sufficient duration for harms

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.

a Population key: 1. Intended use population unclear; 2. Study population is unclear; 3. Study population not representative of intended use; 4, Enrolled populations do not reflect relevant diversity; 5. Other.

b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.

c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.

d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.

e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

 

 

Table 8. Study Design and Conduct Limitations

 

Study

Allocationa

Blindingb

Selective Reportingc

Data Completenessd

Powere

Statisticalf

 

 

 

 

 

 

HOPE-B 32,33,

 

1.  Participants not randomly allocated

2. Allocation not concealed

3. Allocation

concealment unclear

4.  Inadequate

control for selection bias

1.  Participants or study staff not blinded

2.  Outcome assessors not blinded

3.  Outcome assessed by treating physician

4.  Outcomes not assessed centrally

 

 

1.  Power calculations not reported

2.  Power not calculated for primary outcome

3.  Power not based on clinically important difference

 

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.

a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias; 5. Other.

b Blinding key: 1. Participants or study staff not blinded; 2. Outcome assessors not blinded; 3. Outcome assessed by treating physician; 4. Other.

c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication; 4. Other.

d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials); 7. Other.

e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference;

4. Other.

f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated; 5. Other.

 

Section Summary: Etranacogene dezaparvovec-drlb (Hemgenix) for Congenital Hemophilia B

The evidence for use of Etranacogene dezaparvovec-drlb (Hemgenix) for congenital hemophilia B consists of a single study. In the pivotal, open-label, phase III single-arm HOPE-B study, 54 study participants received a single intravenous infusion of Etranacogene dezaparvovec-drlb (Hemgenix). Of the 54 participants, 53 were included in the efficacy analysis. The estimated mean ABR during months 7 to 18 after treatment with Etranacogene dezaparvovec-drlb (Hemgenix) was 1.9 bleeds/year (95% CI: 1.0 to 3.4) compared with an estimated mean ABR of 4.1

 

(95% CI:, 3.2 to 5.4) during the lead-in period. The ABR ratio (months 7 to 18 post-treatment/lead-in) was 0.46

(95% CI, 0.26 to 0.81) demonstrating non-inferiority of annualized bleeding rate during months 7 to 18 compared to the lead-in period. The ABR represents an appropriate clinical benefit endpoint for individuals with hemophilia B and the evidence of clinical benefit was demonstrated by reduction of bleeds in the efficacy evaluable period post treatment. Limitations include uncontrolled study design, limited sample size and relatively short follow-up. There is considerable uncertainty about the long-term net benefits of Etranacogene dezaparvovec-drlb (Hemgenix) compared with FIX prophylaxis. It is not yet clear that the initial increase in FIX levels will be maintained for decades. In addition, there are uncertainties about the long-term impact of the therapy on liver function and the risk for hepatocellular carcinoma as limited sample size is prone to uncertainty around the estimates for adverse events. Some serious harms are likely rare occurrences and as such may not be observed in small trials. Long-term follow-up (>15 years) is required to establish precision around durability of the treatment effect and safety.

Congenital Hemophilia A

 

Clinical Context and Therapy Purpose

 

The purpose of gene therapy in adults who have congenital severe hemophilia A is to provide a treatment option that is an improvement on existing therapies. Potential benefits of this therapy may include the following:

A novel mechanism of action or approach that may allow successful treatment of many individuals for whom other available treatments are not available or have failed or have yielded sub-optimal response

 Reduced treatment complexity such as avoidance of repeated intravenous infusion or subcutaneous injections.

The following PICO was used to select literature to inform this review.

Populations

 

The relevant population of interest is individuals who are adults with congenital severe hemophilia A.

Interventions

 

The therapy being considered is valoctocogene roxaparvovec-rvox, an AAV5 mediated gene therapy designed to deliver a functional copy of a transgene encoding the B-domain deleted SQ form of human coagulation factor VIII (hFVIII-SQ). Transcription of this transgene occurs within the liver, using a liver-specific promoter, which results in the expression of hFVIII-SQ. The expressed hFVIII-SQ replaces the missing coagulation factor VIII needed for effective hemostasis.

Comparators

 

Life-long prophylaxis with exogenous factor replacement therapy is currently being used to manage individuals with congenital severe hemophilia A.

Outcomes

 

The general outcomes of interest are disease-specific survival, change in disease status, health status measures, quality of life, resource utilization, treatment-related mortality and treatment-related morbidity. Relevant outcome measures in alphabetical order are summarized in Table 1.

Study Selection Criteria

 

Methodologically credible studies were selected using the following principles:

To assess efficacy outcomes, comparative controlled prospective trials were sought, with a preference for RCTs;

In the absence of such trials, comparative observational studies were sought, with a preference for prospective studies.

To assess long-term outcomes and adverse events, single-arm studies that capture longer periods of follow-up and/or larger populations were sought.

 Consistent with a 'best available evidence approach,' within each category of study design, studies with larger sample sizes and longer durations were sought.

 Studies with duplicative or overlapping populations were excluded.

Valoctocogene roxaparvovec-rvox (Roctavian) Review of Evidence

The clinical development program is summarized in Table 15 and consists of 2 interventional studies (301 and 201). Both are single-arm, open-label trials. Of these, study 201 is a phase I/II study and is not reviewed in detail. The key trial for Valoctocogene roxaparvovec-rvox (Roctavian)is the phase III trial (study 301) that includes 134 participants and is reviewed in detail.

Table 15. Clinical Development Program for Valoctocogene Roxaparvovec-rvox

 

Study

NCT No

Phase

Study Population

Status

Study Dates

Design

Sample Size

Follow- Up

 

BMN 270-

301

 

 

NCT03370913

 

 

3

Adult males with hemophilia A and residual FVIII levels

≤1 IU/dL

Ongoing (results published at 1 year follow-up37, and 2 year follow-up38,)

 

 

2017-

Ongoing

Open- Label, Single- Arm Study

 

 

134

 

52 weeks (efficacy analysis)

 

BMN 270-

201

 

 

NCT02576795

 

 

2

Adult males with hemophilia A and residual FVIII levels

≤1 IU/dL

Ongoing (results published at 1 year follow-up39, and 3 year follow-up40,)

 

 

2015-

ongoing

Open- Label, Single- Arm Study

 

 

15

 

52 weeks (efficacy analysis)

FVIII: Factor VIII; IU: international units; NCT: national clinical trial

 

Nonrandomized Studies

 

Study characteristic and baseline patient characteristics and results are summarized in Tables 16 and 17, respectively. The prospective, open-label, single-dose, single arm, multi-national study enrolled adult males 18 years and older with severe hemophilia A (endogenous factor VIII [FVIII] level ≤1 IU/dL) as evidenced by their medical history. Study design involved a prospective lead-in period of at least 6 months with the intent to receive standard of care routine factor prophylaxis along with bleeding events. Of the 134 participants who received valoctocogene roxaparvovec-rvox, 112 had baseline ABR data prospectively collected during a period of at least 6 months on FVIII prophylaxis prior to receiving gene therapy (rollover population). The remaining

22 participants had baseline ABR collected retrospectively (directly enrolled population). All patients are intended to be followed for 5 years. The study is on-going. For the efficacy evaluation for the U.S. FDA approval, all patients were followed for at least 3 years.

The primary efficacy outcome was a non-inferiority test of difference in ABR in the efficacy evaluation period. All bleeding episodes were counted. Participants were allowed to continue prophylaxis if needed. Results are summarized in Table 18. The mean ABR after treatment and pre-treatment while patients were on FVIII prophylaxis in the rollover population (N=112) was 2.6 bleeds/year versus 5.4 bleeds/year. The mean difference in ABR was -2.8 (95% CI, -4.3 to -1.2) bleeds/year. The non-inferiority analysis met the pre-specified margin of 3.5. According to the label, a total of 5 participants (4%) did not respond and 17 (15%) lost response to treatment over a median time of 2.3 years (range: 1.0 to 3.3). In the directly enrolled population with a longer follow-up, a total of 1 participant (5%) did not respond and 6 (27%) lost response to treatment over a median time of 3.6 years (range: 1.2 to 4.3).

The most common adverse reactions (incidence ≥5%) were nausea, fatigue, headache, infusion-related reactions, vomiting, and abdominal pain. Most common laboratory abnormalities (incidence ≥10%) were ALT, AST, lactate dehydrogenase (LDH), creatine phosphokinase (CPK), FVIII activity levels, gamma-glutamyl transferase (GGT) and bilirubin above ULN. Transaminitis is presumed to occur due to immune-mediated injury of transduced hepatocytes and may reduce the therapeutic efficacy of AAV-vector based gene therapy. Most ALT elevations occurred within the first year following administration of gene therapy, especially within the first 26 weeks, were low-grade and resolved. The median time (range) to the first ALT elevation (defined as ALT ≥1.5 x baseline or above ULN) was 7 weeks (0.4 to 159 weeks) and the median duration (range) was 4 weeks (0.1 to 135 weeks). Some ALT elevations were associated with a decline in factor VIII activity. As per the prescribing label, integration of liver-targeting AAV vector DNA into the genome may carry the theoretical risk of hepatocellular carcinoma development. As per the label, for individuals with preexisting risk factors (e.g., cirrhosis, advanced hepatic fibrosis, hepatitis B or C, non-alcoholic fatty liver disease, chronic alcohol consumption, non-alcoholic steatohepatitis, and advanced age), regular (e.g., annual) liver ultrasound and alpha-fetoprotein testing should be performed following treatment.

Table 16. Summary of Key Nonrandomized Trial

 

Study

Study Type

Country

Dates

Participants

Treatment

Follow- Up

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Study 301

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Open- label, single- arm

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Global

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

2017-

2024

 

Inclusion

Males ≥18 years of age with severe hemophilia A and residual FVIII levels ≤1 IU/dL. On prophylactic FVIII replacement therapy for at least 12 months prior to study entry. No previous documented history of a detectable FVIII inhibitor.

Exclusion

Detectable pre-existing antibodies to the AAV5 capsid. Any evidence of active infection or any immunosuppressive disorder, including HIV infection. Active infection, chronic or active hepatitis B or C, immunosuppressive disorder including HIV.

Stage 3 or 4 liver fibrosis, cirrhosis, liver function test abnormalities, history of thrombosis or thrombophilia, serum creatinine ≥1.4 mg/dL, and active malignancy.

Primary endpoint

Non-inferiority test of the difference in ABR in the efficacy evaluation perioda compared with baseline period in the rollover population.

Non-inferiority margin was 3.5 bleeds per year.

 

 

 

 

 

 

 

 

Single intravenous dose of 6 X 1013 vg/kg body weight of valoctocogene roxaparvovec- rvox

Of the 134

participants, 112 patients had ABR data prospectively collected for at least 6 months (rollover population); for remaining 22 participants baseline ABR data was collected retrospectively (directly enrolled population).

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

5 years

AAV5; adeno-associated virus serotype 5; ABR; annualized bleeding rate; FVIII: factor VIII; IU: international units.

a All bleeding episodes, regardless of treatment, were counted towards ABR. The efficacy evaluation period started from study day 33 (week 5) or the end of FVIII prophylaxis including a washout period after treatment with gene therapy, whichever was later, and ended when a participant completed the study, had the last visit, or withdrew or was lost to follow-up from the study, whichever was the earliest.

 

 

Table 17. Summary of Baseline Demographics and Disease Characteristics

 

Patient Characteristics in Study 301

N=134

Age, median (min to max), years

30 (18 to 70)

Race, n (%)

 

White

72%

Asian

14%

Black

11%

Positive HIV status, n (%)

1%

Prior hepatitis B infection, n (%)

15%

Prior hepatitis C infection, n (%)

31%

 

HIV: human immunodeficiency virus

 

 

Table 18. Summary of Results

 

Outcomes (Study 301)

Pre-Study Period (n=112)

Post-Study Period (n=112)

Median (range) follow-up duration in years

0.6 (0.5 to 1.3)

3.0 (1.7 to 3.7)

Follow-up duration in person-years

78.3

342.8

Bleeding Related Outcomes (Primary)

 

 

Mean (SD) ABR in bleeds/year

5.4 (6.9)

2.6 (6.2)a

Median (min to max) ABR in bleeds/year

3.3 (0 to 34.6)

0.3 (0 to 35.0)a

Observed spontaneous bleed count (proportion of total bleeds)

176 (42%)

179 (41%)

Observed joint bleed count (proportion of total bleeds)

240 (57%)

195 (45%)

Secondary Outcomes (Factor VIII Activity Thresholds)

 

Chromogenic assay

Year 1 (n=111), n (%)

Year 2 (n=99), n (%)

Year 3 (n=97), n (%)

>150 IU/dL

6 (5%)

2 (2%)

2 (2%)

40 to ≤150 IU/dL

37 (33%)

14 (14%)

9 (9%)

15 to <40 IU/dL

37 (33%)

27 (28%)

23 (24%)

5 to <15 IU/dL

18 (16%)

33 (34%)

35 (36%)

3 to <5 IU/dL

3 (3%)

10 (10%)

8 (8%)

<3 IU/dL

10 (9%)

12 (12%)

19 (20%)

One-stage clotting assay, n (%)

 

 

 

>150 IU/dL

12 (11%)

5 (5%)

4 (4%)

40 to ≤150 IU/dL

44 (40%)

25 (25%)

17 (18%)

15 to <40 IU/dL

37 (33%)

36 (36%)

36 (37%)

5 to <15 IU/dL

10 (9%)

20 (20%)

26 (27%)

1 to <5 IU/dL

6 (5%)

11 (11%)

12 (12%)

<1 IU/dL

2 (2%)

2 (2%)

2 (2%)

ABR: annualized bleeding rate; IU: international units; Min: Minimum; Max: Maximum; SD: standard deviation.

a A total of 13 participants (12%) had used FVIII replacement products or emicizumab during the efficacy evaluation period for prophylaxis, with a

 

median start time at 2.3 (range: 0.1 to 3.3) years. An ABR of 35 was imputed for the periods when these patients were on prophylaxis.

 

The purpose of the study limitations tables (Tables 19 and 20) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence and provides the conclusions on the sufficiency of evidence supporting the position statement. The limited representation of African American, Asian, and Hispanic individuals makes it challenging to reach conclusions about the efficacy of Valoctocogene roxaparvovec-rvox (Roctavian)in these racial groups. The FDA reviewer noted a trend of lower FVIII activity levels in Black participants within the study population. Given the small sample size, the limited number of sites enrolling Black participants relative to the total population, the existence of potential confounding factors, and multiple post hoc analyses, this trend was insufficient to allow meaningful conclusions about the differences in response rates based on race or other factors influencing FVIII expression following Valoctocogene roxaparvovec-rvox (Roctavian)infusion. Despite differences in FVIII activity levels, ABR, and annualized FVIII usage was similar across races. Because of the uncontrolled study design, limited sample size and relatively short follow- up, there is still considerable uncertainty about the long-term net benefits of Valoctocogene roxaparvovec-rvox (Roctavian)compared with factor prophylaxis. It is not yet clear that the initial increase in factor levels will be maintained for decades. In addition, there are uncertainties about the long-term impact of the therapy on liver function and the risk for hepatocellular carcinoma. The small sample size creates uncertainty around the estimates of adverse events. Some serious harms are likely rare occurrences and as such may not be observed in small trials. Long-term follow-up (>15 years) is required to establish precision around durability of the treatment effect and safety.

Table 19. Study Relevance Limitations

 

Study

Populationa

Interventionb

Comparatorc

Outcomesd

Duration of Follow-upe

 

Study 301

 

4. Enrolled populations do not reflect relevant diversity

 

 

 

1. Not sufficient duration for benefit

2. Not sufficient duration

for harms

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.

a Population key: 1. Intended use population unclear; 2. Study population is unclear; 3. Study population not representative of intended use; 4, Enrolled populations do not reflect relevant diversity; 5. Other.

b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.

c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.

d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.

e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

 

 

Table 20. Study Design and Conduct Limitations

 

Study

Allocationa

Blindingb

Selective Reportingc

Data Completenessd

Powere

Statisticalf

 

 

 

 

Study 301

1. Participants not randomly allocated

2. Allocation not concealed

3. Allocation

concealment unclear

4. Inadequate

control for selection bias

1.  Participants or study staff not blinded

2.  Outcome assessors not blinded

3.  Outcome assessed by treating physician

4.  Outcomes not assessed centrally

 

 

 

1.  Power calculations not reported

2.  Power not calculated for primary outcome

3.  Power not based on clinically important difference

 

The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.

a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for

 

selection bias; 5. Other.

b Blinding key: 1. Participants or study staff not blinded; 2. Outcome assessors not blinded; 3. Outcome assessed by treating physician; 4. Other.

c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication; 4. Other.

d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials); 7. Other.

e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference;

4. Other.

f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated; 5. Other.

 

Section Summary: Valoctocogene roxaparvovec-rvox

The evidence for use of Valoctocogene roxaparvovec-rvox (Roctavian)for congenital hemophilia A consists of a single study. In the pivotal, open-label, phase III single-arm study, 134 study participants received a single intravenous infusion of valoctocogene roxaparvovec-rvox. Of the 134 participants, 112 were included in the efficacy analysis. The mean ABR after treatment with Valoctocogene roxaparvovec-rvox (Roctavian)was 2.6 bleeds/year compared with a mean ABR of 5.4 during the lead-in period yielding a mean difference of -2.8 (95% CI, -4.3 to

-1.2) bleeds/year. This was within pre-specified non-inferiority margin of 3.5. The ABR represents an appropriate clinical benefit endpoint for individuals with hemophilia A, and the evidence of clinical benefit was demonstrated by reduction of bleeds during the post-treatment period. However, factor levels declined over time, and therefore benefits of Valoctocogene roxaparvovec-rvox (Roctavian)could be relatively short-lived. According to the label, a total of 5 participants (4%) did not respond and 17 (15%) lost response to treatment over a median time of 2.3 years (range: 1.0 to 3.3). In the directly enrolled population with a longer follow-up, a total of 1 participant (5%) did not respond and 6 (27%) lost response to treatment over a median time of 3.6 years (range: 1.2 to 4.3). Limitations include uncontrolled study design, limited sample size, and relatively short follow-up. There is considerable uncertainty about the long-term net benefits of valoctocogene roxaparvovec- rvox compared with FVIII prophylaxis. It is not yet clear that the initial increase in FVIII levels will be maintained for decades. In addition, there are uncertainties about the long-term impact of the therapy on liver function and the risk for hepatocellular carcinoma as limited sample size is prone to uncertainty around the estimates for adverse events. Some serious harms are likely rare occurrences and as such may not be observed in small trials. Long-term follow-up (>15 years) is required to establish precision around durability of the treatment effect and safety.

REFERENCES

1.  Srivastava A, Brewer AK, Mauser-Bunschoten EP, et al. Guidelines for the management of hemophilia. Haemophilia. Jan 2013; 19(1): e1-47. PMID 22776238

2.  Goodeve AC. Hemophilia B: molecular pathogenesis and mutation analysis. J Thromb Haemost. Jul 2015; 13(7): 1184-95. PMID 25851415

3.  Li T, Miller CH, Payne AB, et al. The CDC Hemophilia B mutation project mutation list: a new online

resource. Mol Genet Genomic Med. Nov 2013; 1(4): 238-45. PMID 24498619

4.  Iorio A, Stonebraker JS, Chambost H, et al. Establishing the Prevalence and Prevalence at Birth of Hemophilia in Males: A Meta-analytic Approach Using National Registries. Ann Intern Med. Oct 15 2019; 171(8): 540-546. PMID 31499529

5.  Carcao MD. The diagnosis and management of congenital hemophilia. Semin Thromb Hemost. Oct 2012; 38(7): 727-34. PMID 23011791

6.  Centers for Disease Control and Prevention (CDC). Data & Statistics on Hemophilia. Available at

https://www.cdc.gov/ncbddd/hemophilia/data.html. Accessed July 5, 2024.

7.  Mansouritorghabeh H. Clinical and laboratory approaches to hemophilia a. Iran J Med Sci. May 2015; 40(3): 194-205. PMID 25999618

8.  U.S. Food and Drug Administration. Guidance for Industry: Human Gene Therapy for Hemophilia. https://www.fda.gov/media/113799/download. Accessed July 5, 2024.

9.  National Organization for Rare Disorders. Hemophilia A. Available at https://rarediseases.org/rare-

diseases/hemophilia-a/. Accessed July 1, 2024.

10.  Stonebraker JS, Bolton-Maggs PH, Soucie JM, et al. A study of variations in the reported haemophilia A prevalence around the world. Haemophilia. Jan 2010; 16(1): 20-32. PMID 19845775

11. Soucie JM, Miller CH, Dupervil B, et al. Occurrence rates of haemophilia among males in the United States based on surveillance conducted in specialized haemophilia treatment centres. Haemophilia. May 2020; 26(3): 487-493. PMID 32329553

12.  World Federation of Hemophilia. Report on the Annual Global Survey 2020. October 2021; https://www1.wfh.org/publications/files/pdf-2045.pdf. Accessed July 5, 2024.

13.  Blanchette VS, Key NS, Ljung LR, et al. Definitions in hemophilia: communication from the SSC of the ISTH. J Thromb Haemost. Nov 2014; 12(11): 1935-9. PMID 25059285

14.  Pavlova A, Oldenburg J. Defining severity of hemophilia: more than factor levels. Semin Thromb Hemost. Oct 2013; 39(7): 702-10. PMID 24026911

15.  World Federation of Hemophilia (WHF). Hemophilia- How Severe is Hemophilia? Available at

https://wfh.org/about-bleeding-disorders/#hemophilia. Accessed July 5, 2024.

16.  Melchiorre D, Manetti M, Matucci-Cerinic M. Pathophysiology of Hemophilic Arthropathy. J Clin Med. Jun 25 2017; 6(7). PMID 28672826

17.  Jiang C, Zhao Y, Feng B, et al. Simultaneous bilateral total knee arthroplasty in patients with end-stage hemophilic arthropathy: a mean follow-up of 6 years. Sci Rep. Jan 25 2018; 8(1): 1608. PMID 29371670

 

18.  Kasper CK, Lin JC. Prevalence of sporadic and familial haemophilia. Haemophilia. Jan 2007; 13(1): 90-

2. PMID 17212731

19.  Srivastava A, Santagostino E, Dougall A, et al. WFH Guidelines for the Management of Hemophilia, 3rd edition. Haemophilia. Aug 2020; 26 Suppl 6: 1-158. PMID 32744769

20.  Kitchen S, McCraw A, Echenagucia M. Diagnosis of Hemophilia and Other Bleeding Disorders: A Laboratory Manual. 2nd Edition. 2010; http://www1.wfh.org/publication/files/pdf-1283.pdf. Accessed July 7, 2024.

21.  Mazepa MA, Monahan PE, Baker JR, et al. Men with severe hemophilia in the United States: birth cohort analysis of a large national database. Blood. Jun 16 2016; 127(24): 3073-81. PMID 26983851

22.  Walsh CE, Soucie JM, Miller CH. Impact of inhibitors on hemophilia A mortality in the United States. Am

J Hematol. May 2015; 90(5): 400-5. PMID 25616111

23.  Eckhardt CL, Loomans JI, van Velzen AS, et al. Inhibitor development and mortality in non-severe hemophilia A. J Thromb Haemost. Jul 2015; 13(7): 1217-25. PMID 25912309

24.  Rentz A, Flood E, Altisent C, et al. Cross-cultural development and psychometric evaluation of a patient-

reported health-related quality of life questionnaire for adults with haemophilia. Haemophilia. Sep 2008; 14(5): 1023-34. PMID 18665853

25.  von Mackensen S, Eldar-Lissai A, Auguste P, et al. Measurement properties of the Haem-A-QoL in

haemophilia clinical trials. Haemophilia. May 2017; 23(3): 383-391. PMID 28026074

26.  Zhou ZY, Koerper MA, Johnson KA, et al. Burden of illness: direct and indirect costs among persons with hemophilia A in the United States. J Med Econ. Jun 2015; 18(6): 457-65. PMID 25660324

27.  O'Hara J, Hughes D, Camp C, et al. The cost of severe haemophilia in Europe: the CHESS study. Orphanet J Rare Dis. May 31 2017; 12(1): 106. PMID 28569181

28.  Miesbach W, Meijer K, Coppens M, et al. Gene therapy with adeno-associated virus vector 5-human

factor IX in adults with hemophilia B. Blood. Mar 01 2018; 131(9): 1022-1031. PMID 29246900

29.  Von Drygalski A, Giermasz A, Castaman G, et al. Etranacogene dezaparvovec (AMT-061 phase 2b): normal/near normal FIX activity and bleed cessation in hemophilia B. Blood Adv. Nov 12 2019; 3(21): 3241-3247. PMID 31698454

30.  von Drygalski A, Gomez E, Giermasz A, et al. Stable and durable factor IX levels in hemophilia B patients over 3 years post etranacogene dezaparvovec gene therapy. Blood Adv. Dec 09 2022. PMID 36490302

31.  Summary           Basis           for           Regulatory           Action-Hemgenix.          Available      at https://www.fda.gov/media/164094/download. Accessed July 10, 2024.

32.  Prescribing Label Hemgenix (Etranacogene dezaparvovec-drlb (Hemgenix)) suspension, for intravenous infusion.

Available at https://labeling.cslbehring.com/PI/US/Hemgenix/EN/Hemgenix-Prescribing-Information.pdf. Accessed July 5, 2024.

33.  Summary           Basis           for           Regulatory           Action-Hemgenix.                          Available      at

https://www.fda.gov/media/164094/download. Accessed July 10, 2024.

 

34.  George LA, Sullivan SK, Giermasz A, et al. Hemophilia B Gene Therapy with a High-Specific-Activity Factor IX Variant. N Engl J Med. Dec 07 2017; 377(23): 2215-2227. PMID 29211678

35.  Ozelo MC, Mahlangu J, Pasi KJ, et al. Valoctocogene Roxaparvovec Gene Therapy for Hemophilia A. N Engl J Med. Mar 17 2022; 386(11): 1013-1025. PMID 35294811

36.  Mahlangu J, Kaczmarek R, von Drygalski A, et al. Two-Year Outcomes of Valoctocogene Roxaparvovec

Therapy for Hemophilia A. N Engl J Med. Feb 23 2023; 388(8): 694-705. PMID 36812433

37.  Rangarajan S, Walsh L, Lester W, et al. AAV5-Factor VIII Gene Transfer in Severe Hemophilia A. N Engl J Med. Dec 28 2017; 377(26): 2519-2530. PMID 29224506

38.  Pasi KJ, Rangarajan S, Mitchell N, et al. Multiyear Follow-up of AAV5-hFVIII-SQ Gene Therapy for

Hemophilia A. N Engl J Med. Jan 02 2020; 382(1): 29-40. PMID 31893514

39.  Gene Therapy for Hemophilia B and An Update on Gene Therapy for Hemophilia A: Effectiveness and Value. Final Evidence Report Posted December 22, 2022. Institute for Clinical and Economic Review. Available                                                       at                             https://icer.org/wp- content/uploads/2022/05/ICER_Hemophilia_Final_Report_12222022.pdf. Accessed July 5, 2024.

40.  National Hemophilia Foundation (NHF). MASAC Recommendations Concerning Products Licensed for the Treatment of Hemophilia and Other Bleeding Disorders (MASAC Document 263). Revised August 2020.                       https://www.hemophilia.org/healthcare-professionals/guidelines-oncare/            masac- documents/masac-document-263-masac-recommendations-concerning-productslicensed-                       for-the- treatment-of-hemophilia-and-other-bleeding-disorders. Accessed July 5, 2024.


Place of Service: Inpatient/Outpatient


The policy position applies to all commercial lines of business




Links