Currently, we are at an enviable place in hemophilia treatment. Although full prophylaxis with standard half-life recombinant or plasma-derived factor concentrates has been definitively shown to be inadequate for full protection against bleeding and arthropathy, a number of novel therapies with improved hemostatic enhancement are clinically available or in promising clinical trials. In order to compare outcomes among a number of very efficacious therapies, it is necessary to have sensitive tools employed in long-term follow-up for several years for participants with no or minimal joint disease. The tool kit must be comprehensive, with outcomes of bleeding, factor level restoration or hemostatic capacity, joint structure, joint function, pain, quality of life, and patient satisfaction. This article reviews the history of prophylaxis, the promise of emerging therapies, and the sensitive tools used to assess long-term efficacy for joint structure and function.

Learning Objectives

  • Understand the role of novel therapies for hemophilia in the prevention of joint damage

  • Become familiar with the numerous tools useful for comparing outcomes of very efficacious therapies for hemophilia

  • Understand the emerging importance of patient-reported outcomes for assessing the efficacy of novel hemophilia therapies

Hemophilia A and B are genetic disorders resulting in deficiencies of factors VIII (FVIII) and IX (FIX), respectively. Historically, severe hemophilia was associated with crippling arthritis in 90% of affected individuals and with intracranial hemorrhage in up to 12%.1-3 

Dr Inge Marie Nilsson began prophylactic infusions of low-dose FVIII every 5 to 12 days in 1958 to prevent bleeding and subsequent joint damage.4  The safety and efficacy of prophylaxis were later established in a prospective randomized controlled trial, called the Joint Outcome Study (JOS), in which children were randomized prior to age 2.5 years to receive either 25 IU/kg of a standard half-life recombinant FVIII (rFVIII) every other day or treatment with rFVIII only at the time of clinical bleeding.5  By age 6 years, the children on prophylaxis had significantly less functional and structural joint disease, as determined by sensitive joint physical examination and magnetic resonance imaging (MRI), compared with children treated for bleeding only. Clinically recognized bleeding was also greatly decreased with prophylaxis; however, physical exam and MRI evidence of joint damage was present in some joints of children on early prophylaxis without clinical bleeding events, suggesting that subclinical bleeding on prophylaxis can promote joint damage.5  In the JOS continuation study that followed JOS participants to age 18 years, those on early prophylaxis showed evidence of progressive joint disease on MRI, although it was less than those who had started on prophylaxis after age 6. At age 6, 7% of those who had started prophylaxis prior to 2.5 years of age had osteochondral damage, and 27% had osteochondral or synovial abnormalities, while at age 18, 35% had osteochondral damage, and 86% had osteochondral or synovial abnormalities in at least 1 joint despite 92% treatment adherence.6 

Regarding quality of life, older adolescents on lifelong prophylaxis complained about the inconvenience of every-other-day venipuncture and lack of access to certain occupations, such as the military, but chronic pain and disability were not noted. In addition, no child on early prophylaxis developed intracranial or other life-threatening hemorrhage.6 

The JOS and its continuation clearly demonstrated that prophylaxis with a standard half-life rFVIII product was a significant improvement over episodic treatment for bleeding and arthropathy but was not adequate to completely prevent joint disease. Additionally, the JOS supported earlier German data that prophylaxis must be started very early in life,7  when the technical challenges of frequent venipuncture are greatest.

Efforts have been made to develop more effective products for prophylaxis of joint damage in hemophilia, as listed in Table 1.

Table 1.

Products supporting long-term prophylaxis that are enhanced over standard half-life factor products

1. Extended half-life recombinant factors supporting once-weekly administration 
FIX 
a. Alprolix, coagulation FIX (recombinant), Fc fusion protein, efmoroctocog alfa, Biogen Sanofi 
b. Idelvion, coagulation FIX (recombinant), albutrepenonacog alfa, albumin fusion protein, CSL Behring 
c. Rebinyn, coagulation FIX (recombinant), glycopegylated, nonacog beta pegol, Novo Nordisk 
FVIII 
d. Efanesoctocog alfa (BIVV001, rFVIIIc-VWF-XTEN), in clinical trials, Sobi and Sanofi 
2. FVIII mimetics (bivalent monoclonal antibodies that promote activation of FX in the absence of FVIII) 
a. Hemlibra, emicizumab, Genentech, Roche 
b. Mim8, Novo Nordisk, in clinical trials 
3. Hemostatic rebalancing agents 
a. Antithrombin siRNA, fitusiran, Sanofi, in clinical trials 
b. TFPI monoclonal antibodies 
i. Concizumab, humanized immunoglobulin G4 in clinical trials, Novo Nordisk 
ii. Marstacimab, humanized immunoglobulin G1 in clinical trials, Pfizer 
c. Lowering of protein C or protein S 
i. Discussed, not yet in clinical trials 
4. Gene therapy 
FIX 
a. Fidanacogene elaparvovec, SPK 9001, in clinical trials, Pfizer 
b. Etranacogene dezaparvovec, AMT-061, in clinical trials, uniQure 
c. scAAV2/8-LP1-hFIXcoAAV8, in clinical trials, St. Jude, University College London, and the Royal Free Hospital 
FVIII 
d. Valoctocogene roxaparvovec, 270-303, in clinical trials, BioMarin 
e. SPK-8011, in clinical trials, Spark Therapeutics 
1. Extended half-life recombinant factors supporting once-weekly administration 
FIX 
a. Alprolix, coagulation FIX (recombinant), Fc fusion protein, efmoroctocog alfa, Biogen Sanofi 
b. Idelvion, coagulation FIX (recombinant), albutrepenonacog alfa, albumin fusion protein, CSL Behring 
c. Rebinyn, coagulation FIX (recombinant), glycopegylated, nonacog beta pegol, Novo Nordisk 
FVIII 
d. Efanesoctocog alfa (BIVV001, rFVIIIc-VWF-XTEN), in clinical trials, Sobi and Sanofi 
2. FVIII mimetics (bivalent monoclonal antibodies that promote activation of FX in the absence of FVIII) 
a. Hemlibra, emicizumab, Genentech, Roche 
b. Mim8, Novo Nordisk, in clinical trials 
3. Hemostatic rebalancing agents 
a. Antithrombin siRNA, fitusiran, Sanofi, in clinical trials 
b. TFPI monoclonal antibodies 
i. Concizumab, humanized immunoglobulin G4 in clinical trials, Novo Nordisk 
ii. Marstacimab, humanized immunoglobulin G1 in clinical trials, Pfizer 
c. Lowering of protein C or protein S 
i. Discussed, not yet in clinical trials 
4. Gene therapy 
FIX 
a. Fidanacogene elaparvovec, SPK 9001, in clinical trials, Pfizer 
b. Etranacogene dezaparvovec, AMT-061, in clinical trials, uniQure 
c. scAAV2/8-LP1-hFIXcoAAV8, in clinical trials, St. Jude, University College London, and the Royal Free Hospital 
FVIII 
d. Valoctocogene roxaparvovec, 270-303, in clinical trials, BioMarin 
e. SPK-8011, in clinical trials, Spark Therapeutics 

Extended half-life factor products

During the first 5 years of the JOS, trough FVIII levels in half of the participants were less than 1%.5,8  Other studies have also shown rapid FVIII clearance in young children.9  The promise of full joint bleeding prevention seemed to rest on the ability to maintain higher trough levels; attention turned to extended half-life (EHL) products. Hermans and colleagues published a comparison of 8 pivotal trials including 900 participants using 4 EHL rFVIII products (rFVIIIFc, efmoroctocog alfa [Eloctate]; rurioctocog alfa pegol [Adynovate]; turoctocog alfa pegol [Esperoct]; damoctocog alfa pegol [Jivi]).10  In this study, median total annualized bleeding rates (ABRs) were reported at 1.3 to 1.9; achieved trough levels of 1% to 3% were judged insufficient to prevent joint bleeding or bleeding overall.10-14  Real-world experience using the same 4 EHL rFVIII products was reported by Dettoraki et al. It included 23 children, half 3 to 12 years old and half 13 to 18 years old, with pharmacokinetic studies performed on both standard and EHL FVIII products.15  On EHL FVIII, mean trough levels rose from 2.3% to 4.1%; mean ABR decreased from 1.0 to 0.3, and mean joint ABR decreased from 0.8 to 0.2. Dosing frequency decreased from every 2.3 to every 3.5 days. The results of Dettoraki et al are similar to the individual licensure trial reports.11,14 

A novel FVIII product in clinical trials, efanesoctocog alfa (BIVV001, rFVIIIc-VWF-XTEN), has been reported to increase the geometric mean FVIII half-life 4-fold to 42.5 hours using a dose of 65 IU/kg.16  At this dose, the plasma FVIII activity was above 50% for 4 days and was 17% at 7 days, suggesting potential joint protection with once-weekly dosing.

The EHL FIX products rFIX-FP (albutrepenonacog [Idelvion]), rFIXFc (coagulation FIX, Fc fusion protein [Alprolix]), and N9-GP (nonacog beta pegol [Rebinyn]) have achieved greater circulating plasma durations and levels. In the rFIXFc clinical trials, the geometric mean half-life was 82.1 hours (vs 33.8 hours standard rFIX) for adults and 68.6 hours in children under 12 years.17-18  In the pediatric study Kids-B-LONG, in which all children were on standard FIX prophylaxis 1 to 3 times per week prior to initiating rFIXFc prophylaxis weekly, the overall bleeding rate was similar (2.5 [0.0-5.0] vs 2.0 [0.0-3.1]; median [interquartile range; IQR] for standard FIX vs rFIXFc), with minimal joint bleeding on rFIXFc prophylaxis (0 [0-1.1]; median [IQR] joint ABR).18  rFIX-FP clinical trials showed a mean half-life of 92 hours in adults and 91.4 hours in children,19,20  with a similar median ABR in the pediatric studies (3.0 [1.0, 6.0], median [IQR] vs 3.12 [0.91, 5.91], median [IQR]) in standard vs rFIX-FP prophylaxis with fewer infusions and few joint bleeds (0.99 [0, 2.33], median [IQR]).20  Nonacog beta pegol (N9-GP, Rebinyn) is a recombinant FIX product in which the FIX gene is linked to a 40 kD PEG moiety at the FIX activation peptide. In phase 3 studies, 29 adults with moderate to severe hemophilia B given 40 IU/kg showed the product to have a single-dose half-life of 93 hours, while in 25 children it had a half-life of 70 hours in those younger than 7 years of age and 76 hours in those 7 to 11 years; trough levels were higher than 15%.21,22  The mean ABR was 1.04 (IQR, 0-4) in adults, with 67% of adults having no bleeding; mean ABR was 1.44 (95% CI, 0.92-2.26) in children. Although the study design and analytic methods to calculate half-life differed among these studies, in clinical practice these 3 EHL FIX products perform similarly, and most children have no spontaneous bleeding on once-weekly dosing. These long-acting products could be competitive with emerging nonfactor and gene therapy alternatives.23,24 

FVIII mimetics

The FVIII mimetic emicizumab has presented another major advance in replacement therapy for hemophilia. Emicizumab is a bivalent monoclonal antibody that binds FIXa and activates FX in the absence of FVIII. Bleeding outcomes for up to 3.5 years on emicizumab have been reported.25  Bleeding on weekly or every-other-week dosing was reduced to less than 1 per year. Bleeding rates continued to decrease over time, with a calculated median joint ABR of 0. To date, there are no long-term outcome data on joint structure and function in persons using emicizumab prophylaxis, although the HAVEN 7 study of infants starting on emicizumab does include joint structure and function evaluations.26  The FVIII mimetic currently known as Mim8 is in clinical trials.27 

Hemostatic rebalancing

Hemostatic rebalancing is an approach to maximize thrombin generation in persons with hemophilia by decreasing thrombin regulation. Although there are no currently licensed products for hemostasis rebalancing, clinical trials are in progress for a small-molecule mRNA silencer of antithrombin (fitusiran) as well as antibodies to tissue factor pathway inhibitor (TFPI; concizumab, marstacimab), with both showing promise.28  Additional potential targets also include protein C and protein S.29  The hemostatic rebalancing agents have the potential downside to be thrombogenic in high-risk situations, although thrombosis has been rare in the clinical trials to date. Fitusiran has the advantage that it can be overridden with antithrombin concentrates should the need arise. In the report of a phase 1 study, 17 adults with hemophilia A or B with inhibitors given 50 or 80  mg of fitusiran had an 82% (SD 2.2) and 87.4% (SD 0.7) reduction in antithrombin, respectively. Eleven of 17 participants had no bleeding during a mean observation period of 69.4 days. Thrombin generation was increased, but there were no clinical thromboses.30  Concuzimab phase 2 clinical trials have been reported with good safety profiles. The joint ABR decrease was greater for hemophilia A and B with inhibitor (mean ABR 2.7 and 3.8, respectively) than for hemophilia A without inhibitor (mean ABR 4.9). All participant groups showed an elevation of thrombin generation into the normal range and elevated prothrombin fragment 1 plus 2 (particularly in noninhibitor patients) with free TFPI suppression to approximately 25% of baseline.31  A phase 1b/2 clinical study of marstacimab has also been recently reported.32  Participants on marstacimab experienced an 86% to 90% reduction in ABR. Prothrombin fragment 1 plus 2 and dimerized plasmin fragment D increased without clinical thrombosis. Mildly elevated troponins in 3 participants were judged not to be related to marstacimab and resolved without a change in treatment. To date, the 3 hemostasis rebalancers appear to have similar safety and efficacy profiles, but the trials are not yet completed.

Gene therapy

Gene therapy holds the ultimate promise for prevention of joint disease by the continuous endogenous release of FIX or FVIII to normal levels. Although to date no FVIII or FIX gene therapy products are approved by the US Food and Drug Administration or European Medicines Agency, great progress has been made, and 3 trials are in the late stages. Table 2 displays gene therapies under investigation in the US and Europe. Biomarin's FVIII AAV-5 trial has reported 3-year outcomes.33  FVIII levels tended to decrease over the first year, with an apparent slower decline thereafter. At 3 years, 3 of 7 participants who received 6 × 1013  vector genomes (vg)/kg achieved an FVIII level of 20% to 36%, and 1 participant achieved 100%. Participants reported an almost complete cessation in clinical bleeding and FVIII infusions, even with low assayed plasma FVIII activity. A 2022 report of 132 participants in a phase 3 trial showed a median FVIIII activity of 23.5% at 3-year follow-up. The FIX trial of Spark Therapeutics reported 10 treated patients on a phase 1/2a study who achieved a mean FIX activity of 33.7% (range, 14-81), with a follow-up of 28 to 78 weeks.34  These patients experienced a reduction in ABR from 11.1 (range, 0-48) to 0.4 (range, 0-4). Nine of the 10 reported no bleeding events, and 8 of the 10 used no factor replacement. The UniQure trial, AMT-061, reported 3 patients with a mean 26-week level of 47% (range, 33.2%-57%).35  Both of these gene therapies as well as the St. Jude, University College London, and Royal Free Hospital trial utilize the Padua variant of FIX with increased activity relative to wild-type FIX. All of the gene therapies have shown variability in interindividual response. There are no long-term reports of joint structure or function following gene therapy. It is very important to document joint outcomes over time in these patients, as many reported no bleeding and no use of prophylaxis with a steady-state FVIII or FIX level less than 10%, and the occurrence or progression of arthropathy in the absence of clinical bleeding, clearly documented in the JOS, could occur in the absence of symptoms.

Table 2.

Gene therapy trials for hemophilia

SponsorVectorStatusFactor level achievedBleeding outcome
Gene therapy trials for hemophilia A 
BioMarin
(NCT03370913)70,71 
AAV-5, valoctogogene roxaparvovec Phase 3, 6 × 1013  vg N = 132; median FVIII 23% (R 11-61) 3 y: bleeding events decreased 84% 
Spark Therapeutics, Pfizer
(NCT03003533,
NCT03432520)72  
AAV-3 SPK-8011 Phase 1/2, 5 × 1011-2 × 1012  vg N = 18; 15/18 had mean OS FVIII 11+/− 6.8% at 52 wk 33.4 mo: bleeding events decreased 91.5% 
Gene therapy trials for hemophilia B 
Spark Therapeutics, Pfizer
(NCT02484092)34  
SPK-9001, AAV, hFIX, R338L (Padua) variant Phase 1/2a N = 10; mean FIX 33.7 +/− 18.5% at 49 wk 49 wk: bleeding events decreased 96% 
uniQure, CSL Behring
(NCT03489291)35,73 
AAV5-hFIX, etranacogene dezaparvovec (EtranaDez)
R338L (Padua) variant 
Phase 3,
AMT-061
HOPE-B,
2 × 1013  vg 
N = 53; mean FIX 36.9% at 18 mo 18 mo: bleeding events decreased 64%
 
Freeline,
St. Jude's, University College London, and Royal Free Hospital (NCT03641703)74,75 
AAV8 FLT-180a
verbrinacogene setparvovec
R338L (Padua) variant 
Phase 1/2,
3.84 × 1011- 1.28 × 1012  vg 
N = 10; mean FIX 67.4%
(R 9-260) at mean 26.6 mo 
Mean 26.6 mo: bleeding events decreased 76% 
SponsorVectorStatusFactor level achievedBleeding outcome
Gene therapy trials for hemophilia A 
BioMarin
(NCT03370913)70,71 
AAV-5, valoctogogene roxaparvovec Phase 3, 6 × 1013  vg N = 132; median FVIII 23% (R 11-61) 3 y: bleeding events decreased 84% 
Spark Therapeutics, Pfizer
(NCT03003533,
NCT03432520)72  
AAV-3 SPK-8011 Phase 1/2, 5 × 1011-2 × 1012  vg N = 18; 15/18 had mean OS FVIII 11+/− 6.8% at 52 wk 33.4 mo: bleeding events decreased 91.5% 
Gene therapy trials for hemophilia B 
Spark Therapeutics, Pfizer
(NCT02484092)34  
SPK-9001, AAV, hFIX, R338L (Padua) variant Phase 1/2a N = 10; mean FIX 33.7 +/− 18.5% at 49 wk 49 wk: bleeding events decreased 96% 
uniQure, CSL Behring
(NCT03489291)35,73 
AAV5-hFIX, etranacogene dezaparvovec (EtranaDez)
R338L (Padua) variant 
Phase 3,
AMT-061
HOPE-B,
2 × 1013  vg 
N = 53; mean FIX 36.9% at 18 mo 18 mo: bleeding events decreased 64%
 
Freeline,
St. Jude's, University College London, and Royal Free Hospital (NCT03641703)74,75 
AAV8 FLT-180a
verbrinacogene setparvovec
R338L (Padua) variant 
Phase 1/2,
3.84 × 1011- 1.28 × 1012  vg 
N = 10; mean FIX 67.4%
(R 9-260) at mean 26.6 mo 
Mean 26.6 mo: bleeding events decreased 76% 

AAV, adeno-associated virus; hFIX, human factor IX; OS, one stage FVIII activity assay; R, range.

Today there is no clear winner in the race for optimal prophylaxis of bleeding and arthropathy in hemophilia. Maintenance of a factor level within a reasonable hemostatic level with weekly injections is likely possible for 1 or more FVIII and FIX EHL products. Emicizumab has already achieved wide acceptance with the elimination of most spontaneous bleeding and a consistent hemostatic capacity of approximately 20% FVIII activity equivalence on chromogenic FVIII assay using human substrates.36  The hemostasis rebalancers continue in clinical trial, and interim data are encouraging. Finally, 1 or more gene therapies is expected to be presented for regulatory approval within the year.

All of the novel therapies decrease ABR to 1 or fewer per year, with median joint bleeding rates at or near 0. It is very difficult to distinguish novel therapy efficacy based upon joint or other bleeding rates. More subtle outcomes of joint dysfunction and pain caused by subclinical bleeding now assume primary importance.

The success of prophylaxis can be considered from multiple angles: achieving a normal factor level; decreasing the number of bleeds; decreasing the risk of catastrophic bleeding; improving joint health; improving quality of life; preventing or decreasing pain; and improving the ability to participate in employment, physical activity, sports, and life.37 Table 3 displays outcomes of importance for hemophilia prophylaxis. Outcome instruments for hemophilia prophylaxis have been recently reviewed.37  A wider community perspective is also essential. The cost of a successful prophylaxis therapy must be such that it can be disseminated to all affected persons and all national economies, without selecting recipients based upon extraordinary resources.

Table 3.

Monitoring the success of novel hemophilia therapies

1. Hemostasis restoration 
a. Bleeding (patient-reported bleeding, objective documentation with US, MRI) 
b. Factor level: one-stage and chromogenic assays 
c. Global assay of hemostasis: thrombin generation, thromboelastography, microfluidic assay 
2. Joint damage 
a. Sensitive physical exam, such as the HJHS 
b. Structural changes on ultrasound or MRI 
c. Functional impact on gait via gait lab assessment 
d. Bone health: bone density scanning, bone biomarkers 
3. Patient-reported outcomes 
a. Quality of life 
i. Generic: SF36, PROMIS measures, Peds QL 
ii. Hemophilia specific: PROBE61  and Canadian Hemophilia Outcomes-Kids Life Assessment Tool (CHO-KLAT) 
b. Pain 
i. McGill Pain Scale 
ii. Brief Pain Inventory 
iii. PROMIS 
iv. PROBE 
v. CHO-KLAT 
1. Hemostasis restoration 
a. Bleeding (patient-reported bleeding, objective documentation with US, MRI) 
b. Factor level: one-stage and chromogenic assays 
c. Global assay of hemostasis: thrombin generation, thromboelastography, microfluidic assay 
2. Joint damage 
a. Sensitive physical exam, such as the HJHS 
b. Structural changes on ultrasound or MRI 
c. Functional impact on gait via gait lab assessment 
d. Bone health: bone density scanning, bone biomarkers 
3. Patient-reported outcomes 
a. Quality of life 
i. Generic: SF36, PROMIS measures, Peds QL 
ii. Hemophilia specific: PROBE61  and Canadian Hemophilia Outcomes-Kids Life Assessment Tool (CHO-KLAT) 
b. Pain 
i. McGill Pain Scale 
ii. Brief Pain Inventory 
iii. PROMIS 
iv. PROBE 
v. CHO-KLAT 

US, ultrasound.

Hemostasis restoration

In assessing the effectiveness of hemophilia prophylaxis, factor level continues to be a critical outcome, as a large body of experience exists relating arthropathy risk to baseline or trough factor level. Gene therapy is evaluated, in large measure, by the level of factor achieved. Many of the novel factor products do not perform equivalently to standard factors in standard laboratory assays. Thus, a menu of assays, including 1-stage and chromogenic assays, are needed. Nonfactor products cannot be evaluated in specific factor assays, and thus global assays such as thromboelastogram, thrombin generation, and dynamic (eg, microfluidic assays) are required.

Joint damage

Because even short durations of blood within a joint can cause long-term damage, many efforts have focused on optimizing prophylaxis by finding joint damage early.38 

In the initial prophylaxis protocols of Sweden, which started in the late 1950s with long-term outcomes published in the 1990s, those who started on low-dose factor had joint changes visible on plain x-ray and physical exam, with those starting on higher prophylaxis doses showing less damage.39  However, the techniques used were less sensitive, and the imaging and physical exam scores used for those studies had a higher threshold for defining damage than current scores. For example, in the orthopedic joint scoring system used by Lofqvist et al, 1 point is attributed for a loss of range of motion of 11% in the ankle or knee (a loss of 15 degrees of flexion in the elbow), while in the Hemophilia Joint Health Score (HJHS) commonly used today, 1 point is attributed for a loss of range of motion of 1 to 4 degrees.40 

The early physical exam and imaging scores were critically important to begin to evaluate the success of hemophilia treatment, but they were limited because they were not able to detect early joint abnormalities, which are more likely to be improved with augmented prophylaxis than more severe joint damage.41  Part of the data safety monitoring plan for the JOS included the monitoring of hemophilia-related joint health every 6 months using physical examination, prompting the development of a more sensitive joint physical examination tool to detect very early joint changes.42  A sensitive MRI scale to image bone, cartilage, and soft tissue changes was also developed and validated for the JOS trial.43  Around that time, the International Prophylaxis Study Group was formed to develop, validate, optimize, and disseminate sensitive joint assessment tools.41,44-46 

The International Prophylaxis Study Group continues to refine the HJHS for reliability, sensitivity, and ease of training and use.47  However, more functional and dynamic assessments are required to detect very subtle early changes. Exciting advances in the application of gait analysis to hemophilia joint monitoring promise techniques that are more sensitive as well as more functional to detect early changes and plan interventions.48,49  The gold standard for detecting joint damage with imaging is MRI, but MRI is less accessible and more time-consuming and expensive. Ultrasound evaluation of joints is logistically much simpler and more accessible. The Hemophilia Early Arthropathy Detection with Ultrasound (HEAD-US) and Joint Tissue Activity and Damage Examination (JADE) scores have been developed to systematically evaluate joint damage and bleeding by ultrasound.50,51  To date there are no long-term outcome studies reported using ultrasound. Cross-sectional studies show concordance of HEAD-US with HJHS,52-54  although the HEAD-US was better at detecting structural changes and the HJHS was better at detecting functional changes, suggesting a role for each.53,54 

The standard outcome in the development of all new hemophilia medications is annualized bleeding rate. For many study participants, bleeding events can at times be difficult to define. For example, it is challenging to distinguish between arthritis pain and bleeding pain.55,56  Although both are critically important to the patient experience, and both should ideally be prevented, the implications are different in a clinical trial. In addition, clinically, not all bleeds are equal. An ankle bleed that is recognized immediately and treated with a single additional dose of factor is a very different patient experience compared to a more serious ankle bleed that takes months to fully resolve, but they are counted equally in clinical trials. Days without bleeding or days without pain may be the preferred outcome as prophylaxis methods continue to improve. Pain can only be prevented if joint damage is detected early and mitigated with physical therapy and augmentation of prophylaxis.

Patient outcomes are of increasing interest in assessing prophylactic efficacy. Ultimately, it is the patient who decides what his or her goals are in health care and how a particular therapy meets those goals. Relevant patient outcomes are displayed in Table 4. Quality-of-life assessments include those that are disease agnostic, such as the SF-36 and the Patient-Reported Outcomes Measurement Information System (PROMIS) measures57-59 ; those specific to children, such as the Peds-QL60 ; and those specific to hemophilia, such as Patient Reported Outcomes Burdens and Experiences (PROBE) and Canadian Hemophilia Outcomes-Kids' Life Assessment Tool (CHO-KLAT).61,62  We prefer the PROMIS measures because they were developed using item response theory in which computerized adaptive testing methods can be employed, shortening the number of questions needed to assess an outcome. Customized questionnaires can be created from existing item banks to focus measurement on particular aspects of an outcome that are important to a specific group of patients, and scores on a particular measure can be calculated even when some data points are missing. In addition, PROMIS measures are publicly available, and they provide standardized scores using a T-score metric based on normative data from the US general population, allowing comparisons of scores across domains and disease states.37 

Table 4.

Outcomes of optimal hemophilia care

a. Education 
b. Employment 
c. Participation: family, community 
d. Sports and leisure activities 
e. Days without bleeding 
f. Days without pain 
a. Education 
b. Employment 
c. Participation: family, community 
d. Sports and leisure activities 
e. Days without bleeding 
f. Days without pain 

Pain, chiefly joint pain, comprises the dominant adverse patient outcome of hemophilia.63  Pain intensity constitutes the severity of pain experienced, while pain interference denotes the impact of pain on one's life activity and behavior. There are several validated scales for quality of life as well as pain. The McGill Pain Scale as well as the Brief Pain Inventory have been used extensively in hemophilia.64,65  The PROMIS is a US National Institutes of Health initiative that led to the development of PRO instruments, collections of validated surveys to assess for a number of domains, including pain intensity, interference, and behavior.58,59  The PROBE questionnaire was developed for persons with hemophilia, with extensive patient engagement, and has been rigorously validated.61  PROBE covers both pain occurrence and interference using patient-derived definitions for acute and chronic pain and captures use of pain medication. The CHO-KLAT pediatric hemophilia outcomes score has been developed with significant input from hemophilia patients and has been cross-culturally validated.66  The PROMIS and PROBE validated instruments are now being used longitudinally and within hemophilia clinical trials.67-69 

Currently, we are at an enviable place in hemophilia treatment. Although full prophylaxis with standard half-life recombinant or plasma-derived factor concentrates has been definitively shown to be inadequate for full protection against bleeding and arthropathy, a number of novel therapies with improved hemostatic enhancement are clinically available or in promising clinical trials. In order to compare outcomes of very efficacious therapies, it is necessary to have sensitive tools, employed in long-term follow-up for several years, in participants with no or minimal joint disease. The tool kit must be comprehensive, with outcomes of bleeding, factor level restoration or hemostatic capacity, joint structure, joint function, pain, quality of life, and patient satisfaction.

Marilyn Jean Manco-Johnson: no competing financial interests to declare.

Beth Boulden Warren: consultancy: Novo Nordisk, Hema Biologics, Genentech.

Marilyn Jean Manco-Johnson: nothing to disclose.

Beth Boulden Warren: nothing to disclose.

1.
Aledort
LM
,
Haschmeyer
RH
,
Pettersson
H
.
A longitudinal study of orthopaedic outcomes for severe factor-VIII-deficient haemophiliacs. The Orthopaedic Outcome Study Group
.
J Intern Med
.
1994
;
236
(
4
):
391
-
399
.
doi:10.1111/j.1365-2796.1994.tb00815.x
.
2.
Nelson
MD
Jr
,
Maeder
MA
,
Usner
D
, et al.
Prevalence and incidence of intracranial haemorrhage in a population of children with haemophilia. The Hemophilia Growth and Development Study
.
Haemophilia
.
1999
;
5
(
5
):
306
-
312
.
doi:10.1046/j.1365-2516.1999.00338.x
.
3.
Klinge
J
,
Auberger
K
,
Auerswald
G
,
Brackmann
HH
,
Mauz-Körholz
C
,
Kreuz
W
.
Prevalence and outcome of intracranial haemorrhage in haemophiliacs —a survey of the paediatric group of the German Society of Thrombosis and Haemostasis (GTH)
.
Eur J Pediatr
.
1999
;
158
(
suppl 3
):
S162
-
S165
.
doi:10.1007/pl00014346
.
4.
Nilsson
IM
,
Berntorp
E
,
Löfqvist
T
,
Pettersson
H
.
Twenty-five years' experience of prophylactic treatment in severe haemophilia A and B
.
J Intern Med
.
1992
;
232
(
1
):
25
-
32
.
doi:10.1111/j.1365-2796.1992.tb00546.x
.
5.
Manco-Johnson
MJ
,
Abshire
TC
,
Shapiro
AD
, et al.
Prophylaxis versus episodic treatment to prevent joint disease in boys with severe hemophilia
.
N Engl J Med
.
2007
;
357
(
6
):
535
-
544
.
doi:10.1056/NEJMoa067659
.
6.
Warren
BB
,
Thornhill
D
,
Stein
J
, et al.
Young adult outcomes of childhood prophylaxis for severe hemophilia A: results of the Joint Outcome Continuation Study
.
Blood Adv
.
2020
;
4
(
11
):
2451
-
2459
.
doi:10.1182/bloodadvances.2019001311
.
7.
Kreuz
W
,
Escuriola-Ettingshausen
C
,
Funk
M
,
Schmidt
H
,
Kornhuber
B
.
When should prophylactic treatment in patients with haemophilia A and B start?—the German experience
.
Haemophilia
.
1998
;
4
(
4
):
413
-
417
.
doi:10.1046/j.1365-2516.1998.440413.x
.
8.
Warren
BB
,
Jacobson
L
,
Kempton
C
, et al
; Joint Outcome Study Group Investigators.
Factor VIII prophylaxis effects outweigh other hemostasis contributors in predicting severe haemophilia A joint outcomes
.
Haemophilia
.
2019
;
25
(
5
):
867
-
875
.
doi:10.1111/hae.13778
.
9.
Björkman
S
,
Oh
M
,
Spotts
G
, et al.
Population pharmacokinetics of recombinant factor VIII: the relationships of pharmacokinetics to age and body weight
.
Blood
.
2012
;
119
(
2
):
612
-
618
.
doi:10.1182/blood-2011-07-360594
.
10.
Hermans
C
,
Reding
MT
,
Astermark
J
,
Klamroth
R
,
Mancuso
ME
.
Clinical studies of extended-half-life recombinant FVIII products for prophylaxis in adults and children: a critical review from the physician's perspective
.
Crit Rev Oncol Hematol
.
2022
;
174
(
June
):
103678
.
doi:10.1016/j.critrevonc.2022.103678
.
11.
Klamroth
R
,
Windyga
J
,
Radulescu
V
, et al.
Rurioctocog alfa pegol PK-guided prophylaxis in hemophilia A: results from the phase 3 PROPEL study
.
Blood
.
2021
;
137
(
13
):
1818
-
1827
.
doi:10.1182/blood.2020005673
.
12.
Konkle
BA
,
Stasyshyn
O
,
Chowdary
P
, et al.
Pegylated, full-length, recombinant factor VIII for prophylactic and on-demand treatment of severe hemophilia A
.
Blood
.
2015
;
126
(
9
):
1078
-
1085
.
doi:10.1182/blood-2015-03-630897
.
13.
Mahlangu
J
,
Powell
JS
,
Ragni
MV
, et al
; A-LONG Investigators.
Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A
.
Blood
.
2014
;
123
(
3
):
317
-
325
.
doi:10.1182/blood-2013-10-529974
.
14.
Young
G
,
Mahlangu
J
,
Kulkarni
R
, et al.
Recombinant factor VIII Fc fusion protein for the prevention and treatment of bleeding in children with severe hemophilia A
.
J Thromb Haemost
.
2015
;
13
(
6
):
967
-
977
.
doi:10.1111/jth.12911
.
15.
Dettoraki
A
,
Michalopoulou
A
,
Mazarakis
M
, et al.
Clinical application of extended half-life factor VIII in children with severe haemophilia A
.
Haemophilia
.
2022
;
28
(
4
):
619
-
624
.
doi:10.1111/hae.14576
.
16.
Konkle
BA
,
Shapiro
AD
,
Quon
DV
, et al.
BIVV001 fusion protein as factor VIII replacement therapy for hemophilia A
.
N Engl J Med
.
2020
;
383
(
11
):
1018
-
1027
.
doi:10.1056/NEJMoa2002699
.
17.
Powell
JS
,
Pasi
KJ
,
Ragni
MV
, et al
; B-LONG Investigators.
Phase 3 study of recombinant factor IX Fc fusion protein in hemophilia B
.
N Engl J Med
.
2013
;
369
(
24
):
2313
-
2323
.
doi:10.1056/NEJMoa1305074
.
18.
Fischer
K
,
Kulkarni
R
,
Nolan
B
, et al.
Recombinant factor IX Fc fusion protein in children with haemophilia B (Kids B-LONG): results from a multicentre, non-randomised phase 3 study
.
Lancet Haematol
.
2017
;
4
(
2
):
e75
-
e82
.
doi:10.1016/S2352-3026(16)30193-4
.
19.
Santagostino
E
,
Negrier
C
,
Klamroth
R
, et al.
Safety and pharmacokinetics of a novel recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in hemophilia B patients
.
Blood
.
2012
;
120
(
12
):
2405
-
2411
.
doi:10.1182/blood-2012-05-429688
.
20.
Kenet
G
,
Chambost
H
,
Male
C
, et al
; PROLONG-9FP Investigator Study Group.
Long-acting recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in children. Results of a phase 3 trial
.
Thromb Haemost
.
2016
;
116
(
4
):
659
-
668
.
doi:10.1160/TH16-03-0179
.
21.
Collins
PW
,
Young
G
,
Knobe
K
, et al
; Paradigm 2 Investigators.
Recombinant long-acting glycoPEGylated factor IX in hemophilia B: a multinational randomized phase 3 trial
.
Blood
.
2014
;
124
(
26
):
3880
-
3886
.
doi:10.1182/blood-2014-05-573055
.
22.
Carcao
M
,
Zak
M
,
Abdul Karim
F
, et al.
Nonacog beta pegol in previously treated children with hemophilia B: results from an international open-label phase 3 trial
.
J Thromb Haemost
.
2016
;
14
(
8
):
1521
-
1529
.
doi:10.1111/jth.13360
.
23.
Mancuso
ME
,
Oldenburg
J
,
Boggio
L
, et al.
High adherence to prophylaxis regimens in haemophilia B patients receiving rIX-FP: evidence from clinical trials and real-world practice
.
Haemophilia
.
2020
;
26
(
4
):
637
-
642
.
doi:10.1111/hae.14018
.
24.
Ducore
JM
,
Miguelino
MG
,
Powell
JS
.
Alprolix (recombinant factor IX Fc fusion protein): extended half-life product for the prophylaxis and treatment of hemophilia B
.
Expert Rev Hematol
.
2014
;
7
(
5
):
559
-
571
.
doi:10.1586/17474086.2014.951322
.
25.
Callaghan
MU
,
Negrier
CG
,
Paz-Priel
I
, et al.
Long-term outcomes with emicizumab prophylaxis for hemophilia A with/without FVIII inhibitors from the HAVEN 1-4 studies
.
Blood
.
2020
;
37
(
16
):
2231
-
2242
.
doi:10.1182/blood.2020009217
.
26.
Clinicaltrials.gov
.
A study to evaluate the efficacy, safety, pharmacokinetics, and pharmacodynamics of subcutaneous emicizumab in participants from birth to 12 months of age with hemophilia a without inhibitors (HAVEN 7)
. https://clinicaltrials.gov/ct2/show/NCT04431726. Accessed
28
October
2022
.
27.
Lauritzen
B
,
Bjelke
M
,
Björkdahl
O
, et al.
A novel next-generation FVIIIa mimetic, Mim8, has a favorable safety profile and displays potent pharmacodynamic effects: results from safety studies in cynomolgus monkeys
.
J Thromb Haemost
.
2022
;
20
(
6
):
1312
-
1324
.
doi:10.1111/jth.15682
.
28.
Butterfield
JSS
,
Hege
KM
,
Herzog
RW
,
Kaczmarek
R
.
A molecular revolution in the treatment of hemophilia
.
Mol Ther
.
2020
;
28
(
4
):
997
-
1015
.
doi:10.1016/j.ymthe.2019.11.006
.
29.
Weyand
AC
,
Pipe
SW
.
New therapies for hemophilia
.
Blood
.
2019
;
133
(
5
):
389
-
398
.
doi:10.1182/blood-2018-08-872291
.
30.
Pasi
KJ
,
Rangarajan
S
,
Georgiev
P
, et al.
Targeting of antithrombin in hemophilia A or B with RNAi therapy
.
N Engl J Med
.
2017
;
377
(
9
):
819
-
828
.
doi:10.1056/NEJMoa1616569
.
31.
Shapiro
AD
,
Angchaisuksiri
P
,
Astermark
J
, et al.
Subcutaneous concizumab prophylaxis in hemophilia A and hemophilia A/B with inhibitors: phase 2 trial results
.
Blood
.
2019
;
134
(
22
):
1973
-
1982
.
doi:10.1182/blood.2019001542
.
32.
Mahlangu
JN
,
Lamas
JL
,
Morales
JC
, et al.
A phase 1b/2 clinical study of marstacimab, targeting human tissue factor pathway inhibitor, in haemophilia [published online 23 August 2022]
.
Br J Haematol
.
doi:10.1111/bjh.18420
.
33.
Pasi
KJ
,
Rangarajan
S
,
Mitchell
N
, et al.
Multiyear follow-up of AAV5-hFVIII-SQ gene therapy for hemophilia A
.
N Engl J Med
.
2020
;
382
(
1
):
29
-
40
.
doi:10.1056/NEJMoa1908490
.
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
.
2017
;
377
(
23
):
2215
-
2227
.
doi:10.1056/NEJMoa1708538
.
35.
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
.
2019
;
3
(
21
):
3241
-
3247
.
doi:10.1182/bloodadvances.2019000811
.
36.
Lenting
PJ
,
Denis
CV
,
Christophe
OD
.
Emicizumab, a bispecific antibody recognizing coagulation factors IX and X: how does it actually compare to factor VIII?
Blood
.
2017
;
130
(
23
):
2463
-
2468
.
doi:10.1182/blood-2017-08-801662
.
37.
Manco-Johnson
MJ
,
Warren
BB
,
Buckner
TW
,
Funk
SM
,
Wang
M
.
Outcome measures in haemophilia: beyond ABR (annualized bleeding rate)
.
Haemophilia
.
2021
;
27
(
suppl 3
):
87
-
95
.
doi:10.1111/hae.14099
.
38.
Hakobyan
N
,
Enockson
C
,
Cole
AA
,
Sumner
DR
,
Valentino
LA
.
Experimental haemophilic arthropathy in a mouse model of a massive haemarthrosis: gross, radiological and histological changes
.
Haemophilia
.
2008
;
14
(
4
):
804
-
809
.
doi:10.1111/j.1365-2516.2008.01689.x
.
39.
Löfqvist
T
,
Nilsson
IM
,
Berntorp
E
,
Pettersson
H
.
Haemophilia prophylaxis in young patients—a long-term follow-up
.
J Intern Med
.
1997
;
241
(
5
):
395
-
400
.
doi:10.1046/j.1365-2796.1997.130135000.x
.
40.
International Prophylaxis Study Group
. Group IPSGP. Hemophilia Joint Health Score (HJHS) 2.1 Instruction Manual. World Federation of Hemophilia; 2012. https://elearning.wfh.org/resource/hemophilia-joint-health-score-hjhs/. Accessed
28
October
2022
.
41.
Manco-Johnson
MJ
,
Pettersson
H
,
Petrini
P
, et al.
Physical therapy and imaging outcome measures in a haemophilia population treated with factor prophylaxis: current status and future directions
.
Haemophilia
.
2004
;
10
(
suppl 4
):
88
-
93
.
doi:10.1111/j.1365-2516.2004.00978.x
.
42.
Hacker
MR
,
Funk
SM
,
Manco-Johnson
MJ
.
The Colorado Haemophilia Paediatric Joint Physical Examination Scale: normal values and interrater reliability
.
Haemophilia
.
2007
;
13
(
1
):
71
-
78
.
doi:10.1111/j.1365-2516.2006.01387.x
.
43.
Nuss
R
,
Kilcoyne
RF
,
Geraghty
S
, et al.
MRI findings in haemophilic joints treated with radiosynoviorthesis with development of an MRI scale of joint damage
.
Haemophilia
.
2000
;
6
(
3
):
162
-
169
.
doi:10.1046/j.1365-2516.2000.00383.x
.
44.
Feldman
BM
,
Funk
S
,
Lundin
B
, et al
;
International Prophylaxis Study Group
.
Musculoskeletal measurement tools from the International Prophylaxis Study Group (IPSG)
.
Haemophilia
.
2008
;
14
(
suppl 3
):
162
-
169
.
doi:10.1111/j.1365-2516.2008.01750.x
.
45.
Lundin
B
,
Manco-Johnson
ML
,
Ignas
DM
, et al
;
International Prophylaxis Study Group
.
An MRI scale for assessment of haemophilic arthropathy from the International Prophylaxis Study Group
.
Haemophilia
.
2012
;
18
(
6
):
962
-
970
.
doi:10.1111/j.1365-2516.2012.02883.x
.
46.
Feldman
BM
,
Funk
SM
,
Bergstrom
BM
, et al.
Validation of a new pediatric joint scoring system from the International Hemophilia Prophylaxis Study Group: validity of the Hemophilia Joint Health Score
.
Arthritis Care Res (Hoboken)
.
2011
;
63
(
2
):
223
-
230
.
doi:10.1002/acr.20353
.
47.
St-Louis
J
,
Abad
A
,
Funk
S
, et al.
The Hemophilia Joint Health Score version 2.1 Validation in Adult Patients Study: a multicenter international study
.
Res Pract Thromb Haemost
.
2022
;
6
(
2
):
e12690
.
doi:10.1002/rth2.12690
.
48.
Warren
BB
,
Mah
J
,
Mah
N
, et al.
Early findings on the use of motion capture during simulated sports activities to better understand hemophilic arthropathy
.
Blood
.
2021
;
138
(
suppl 1
):
3203
.
doi:10.1182/blood-2021-151532
.
49.
Fukuchi
CA
,
Zorzi
AR
,
Fukuchi
RK
,
Ricciardi
JBS
,
Feldberg
G
,
Cliquet
A
Jr
.
Biomechanical gait analysis of an adult with severe hemophilia: a case report
.
Hematol Rep
.
2022
;
14
(
2
):
112
-
118
.
doi:10.3390/hematolrep14020017
.
50.
Martinoli
C
,
Della Casa Alberighi
O
,
Di Minno
G
, et al.
Development and definition of a simplified scanning procedure and scoring method for Haemophilia Early Arthropathy Detection with Ultrasound (HEAD-US)
.
Thromb Haemost
.
2013
;
109
(
6
):
1170
-
1179
.
doi:10.1160/TH12-11-0874
.
51.
Mesleh Shayeb
A
,
Barnes
RFW
,
Hanacek
C
, et al.
Quantitative measurements of haemophilic joint tissues by point-of-care musculoskeletal ultrasound: associations with clinical and functional joint outcome parameters
.
Haemophilia
.
2021
;
27
(
5
):
866
-
875
.
doi:10.1111/hae.14368
.
52.
Prasetyo
M
,
Moniqa
R
,
Tulaar
A
,
Prihartono
J
,
Setiawan
SI
.
Correlation between Hemophilia Early Arthropathy Detection with Ultrasound (HEAD-US) score and Hemophilia Joint Health Score (HJHS) in patients with hemophilic arthropathy
.
Plos One
.
2021
;
16
(
4
):
e0248952
.
doi:10.1371/journal.pone.0248952
.
53.
De la Corte-Rodriguez
H
,
Rodriguez-Merchan
EC
,
Alvarez-Roman
MT
,
Martin-Salces
M
,
Martinoli
C
,
Jimenez-Yuste
V
.
HJHS 2.1 and HEAD-US assessment in the hemophilic joints: how do their findings compare?
Blood Coagul Fibrinolysis
.
2020
;
31
(
6
):
387
-
392
.
doi:10.1097/MBC.0000000000000934
.
54.
Altisent
C
,
Martorell
M
,
Crespo
A
,
Casas
L
,
Torrents
C
,
Parra
R
.
Early prophylaxis in children with severe haemophilia A: clinical and ultrasound imaging outcomes
.
Haemophilia
.
2016
;
22
(
2
):
218
-
224
.
doi:10.1111/hae.12792
.
55.
Timmer
MA
,
Pisters
MF
,
de Kleijn
P
,
de Bie
RA
,
Fischer
K
,
Schutgens
RE
.
Differentiating between signs of intra-articular joint bleeding and chronic arthropathy in haemophilia: a narrative review of the literature
.
Haemophilia
.
2015
;
21
(
3
):
289
-
296
.
doi:10.1111/hae.12667
.
56.
Ceponis
A
,
Wong-Sefidan
I
,
Glass
CS
,
von Drygalski
A
.
Rapid musculoskeletal ultrasound for painful episodes in adult haemophilia patients
.
Haemophilia
.
2013
;
19
(
5
):
790
-
798
.
doi:10.1111/hae.12175
.
57.
Brazier
JE
,
Harper
R
,
Jones
NM
, et al.
Validating the SF-36 health survey questionnaire: new outcome measure for primary care
.
BMJ
.
1992
;
305
(
6846
):
160
-
164
.
doi:10.1136/bmj.305.6846.160
.
58.
Fries
JF
,
Bruce
B
,
Cella
D
.
The promise of PROMIS: using item response theory to improve assessment of patient-reported outcomes
.
Clin Exp Rheumatol
.
2005
;
23
(
suppl 5, pt 39
):
S53
-
S57
.
59.
Cella
D
,
Yount
S
,
Rothrock
N
, et al
;
PROMIS Cooperative Group
.
The Patient-Reported Outcomes Measurement Information System (PROMIS): progress of an NIH Roadmap cooperative group during its first two years
.
Med Care
.
2007
;
45
(
suppl 1, pt 5
):
S3
-
S11
.
doi:10.1097/01.mlr.0000258615.42478.55
.
60.
Varni
JW
,
Thompson
KL
,
Hanson
V
.
The Varni/Thompson Pediatric Pain Questionnaire, I: Chronic musculoskeletal pain in juvenile rheumatoid arthritis
.
Pain
.
1987
;
28
(
1
):
27
-
38
.
doi:10.1016/0304-3959(87)91056-6
.
61.
Chai-Adisaksopha
C
,
Skinner
MW
,
Curtis
R
, et al.
Psychometric properties of the Patient Reported Outcomes, Burdens and Experiences (PROBE) questionnaire
.
BMJ Open
.
2018
;
8
(
8
):
e021900
.
doi:10.1136/bmjopen-2018-021900
.
62.
Price
VE
,
Dover
S
,
Blanchette
VS
, et al.
Updating the Canadian Hemophilia Outcomes-Kids' Life Assessment Tool (CHO-KLAT) in the era of extended half-life clotting factor concentrates
.
Res Pract Thromb Haemost
.
2021
;
5
(
3
):
403
-
411
.
doi:10.1002/rth2.12498
.
63.
Forsyth
AL
,
Witkop
M
,
Lambing
A
, et al.
Associations of quality of life, pain, and self-reported arthritis with age, employment, bleed rate, and utilization of hemophilia treatment center and health care provider services: results in adults with hemophilia in the HERO study
.
Patient Prefer Adherence
.
2015
;
9
(29 October):
1549
-
1560
.
doi:10.2147/PPA.S87659
.
64.
Choinière
M
,
Melzack
R
.
Acute and chronic pain in hemophilia
.
Pain
.
1987
;
31
(
3
):
317
-
331
.
doi:10.1016/0304-3959(87)90161-8
.
65.
Buckner
TW
,
Batt
K
,
Quon
D
, et al.
Assessments of pain, functional impairment, anxiety, and depression in US adults with hemophilia across patient-reported outcome instruments in the Pain, Functional Impairment, and Quality of Life (P-FiQ) study
.
Eur J Haematol
.
2018
;
100
(
suppl 1
):
5
-
13
.
doi:10.1111/ejh.13027
.
66.
McCusker
PJ
,
Fischer
K
,
Holzhauer
S
, et al.
International cross-cultural validation study of the Canadian Haemophilia Outcomes: Kids' Life Assessment Tool
.
Haemophilia
.
2015
;
21
(
3
):
351
-
357
.
doi:10.1111/hae.12597
.
67.
Kempton
CL
,
Michaels Stout
M
,
Barry
V
, et al.
Validation of a new instrument to measure disease-related distress among patients with haemophilia
.
Haemophilia
.
2021
;
27
(
1
):
60
-
68
.
doi:10.1111/hae.14187
.
68.
Pinto
PR
,
Paredes
AC
,
Pedras
S
, et al.
Sociodemographic, clinical, and psychosocial characteristics of people with hemophilia in Portugal: findings from the First National Survey
.
TH Open
.
2018
;
2
(
1
):
e54
-
e67
.
doi:10.1055/s-0038-1624568
.
69.
Skinner
MW
,
Chai-Adisaksopha
C
,
Curtis
R
, et al.
The Patient Reported Outcomes, Burdens and Experiences (PROBE) Project: development and evaluation of a questionnaire assessing patient reported outcomes in people with haemophilia. Pilot Feasibility Stud.
2018
;
4
:
58
.
doi:10.1186/s40814-018-0253-0
.
70.
Pasi
KJ
,
Fischer
K
,
Ragni
M
, et al.
Long-term safety and sustained efficacy for up to 5 years of treatment with recombinant factor IX Fc fusion protein in subjects with haemophilia B: results from the B-YOND extension study
.
Haemophilia
.
2020
;
26
(
6
):
e262
-
e271
.
doi:10.1111/hae.14036
.
71.
Ozelo
MC
,
Mahlangu
J
,
Pasi
KJ
, et al
;
GENEr8-1 Trial Group
.
Valoctocogene roxaparvovec gene therapy for hemophilia A
.
N Engl J Med
.
2022
;
386
(
11
):
1013
-
1025
.
doi:10.1056/NEJMoa2113708
.
72.
George
LA
,
Monahan
PE
,
Eyster
ME
, et al.
Multiyear factor VIII expression after AAV gene transfer for hemophilia A
.
N Engl J Med
.
2021
;
385
(
21
):
1961
-
1973
.
doi:10.1056/NEJMoa2104205
.
73.
Miesbach
W
,
Leebeek
FW
,
Recht
M
, et al.
Final analysis from the pivotal phase 3 HOPE-B gene therapy trial: stable steady-state efficacy and safety of etranacogene dezaparvovec in adults with severe or moderately severe hemophilia B [abstract]
.
Haemophilia
.
2022
;
28
:
99
-
100
. Abstract PO143.
74.
Nathwani
AC
,
Reiss
UM
,
Tuddenham
EG
, et al.
Long-term safety and efficacy of factor IX gene therapy in hemophilia B
.
N Engl J Med
.
2014
;
371
(
21
):
1994
-
2004
.
doi:10.1056/NEJMoa1407309
.
75.
Chowdary
P
,
Shapiro
S
,
Makris
M
, et al.
Phase 1–2 trial of AAVS3 gene therapy in patients with hemophilia B
.
N Engl J Med
.
2022
;
387
(
3
):
237
-
247
.
doi:10.1056/nejmoa2119913
.