Skip to main content

Open Access 10.05.2024 | Original Research Article

Normalization of Hemoglobin, Lactate Dehydrogenase, and Fatigue in Patients with Paroxysmal Nocturnal Hemoglobinuria Treated with Pegcetacoplan

verfasst von: Brian P. Mulherin, Michael Yeh, Mohammed Al-Adhami, David Dingli

Erschienen in: Drugs in R&D

Abstract

Background and Objectives

We determined normalization rates for hemoglobin, lactate dehydrogenase (LDH), and fatigue in patients with paroxysmal nocturnal hemoglobinuria (PNH) treated with pegcetacoplan (PEG) in the PEGASUS (NCT03500549) and PRINCE (NCT04085601) phase III trials.

Methods

Enrolled patients had PNH and hemoglobin < 10.5 g/dL despite ≥ 3 months of eculizumab (ECU) [PEGASUS], or were complement component 5 (C5) inhibitor-naive, receiving supportive care only, with hemoglobin less than the lower limits of normal (LLN) [PRINCE]. Hematologic and fatigue normalization endpoints were hemoglobin greater than or equal to the LLN (females: 12 g/dL; males: 13.6 g/dL) in the absence of transfusion; LDH ≤226 U/L in the absence of transfusion; and Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue ≥ 43.6, the general population norm. Safety was assessed by investigators using standardized terms and definitions for seriousness and severity.

Results

Hemoglobin normalization occurred in 34.1% (14/41) of PEG-treated patients at Week 16 (randomized controlled period) in PEGASUS (vs. 0% [0/39] of ECU-treated patients) and in 45.7% (16/35) of PEG-treated patients at Week 26 in PRINCE (vs. 0% [0/18] of supportive care–treated patients). At Week 48 (open-label period) in PEGASUS, 24.4% of PEG-treated patients (PEG-to-PEG) and 30.8% of patients treated with ECU through Week 16 who switched to PEG through Week 48 (ECU-to-PEG) had hemoglobin normalization. Rates of LDH normalization in PEGASUS were 70.7% (PEG-treated patients) and 15.4% (ECU-treated patients) at Week 16, and 56.1% (PEG-to-PEG) and 51.3% (ECU-to-PEG) at Week 48. In PRINCE, 67.5% of PEG-treated patients at Week 26 had normalized LDH concentrations. Rates of FACIT-Fatigue score normalization in PEGASUS were 48.8% and 10.3% in PEG- and ECU-treated patients, respectively, at Week 16, and 34.1% and 51.3% in PEG-to-PEG- and ECU-to-PEG-treated patients, respectively, at Week 48. In PRINCE, 68.6% of PEG-treated patients and 11.1% of supportive care patients had FACIT-Fatigue score normalization at Week 26. PEG was safe and well tolerated. Injection site reactions, mostly mild, were the most common adverse event of special interest in PEG-treated patients in the PEGASUS randomized controlled period (36.6%) and in PRINCE (30.4%).

Conclusion

PEG is superior to ECU and supportive care in hemoglobin, LDH, and FACIT-Fatigue score normalization for patients with PNH and persistent anemia despite ≥3 months of treatment with ECU, and in C5 inhibitor–naive patients.

Clinical Trial Registration

The PEGASUS trial (NCT03500549) was registered on 18 August 2018, and the PRINCE trial (NCT04085601) was registered on 11 September 2019.
Key Points
Pegcetacoplan, the first complement component 3-targeted therapy for paroxysmal nocturnal hemoglobinuria (PNH), markedly improved outcomes for patients with PNH in two phase III clinical trials—PEGASUS and PRINCE.
This analysis examined the percentages of patients from PEGASUS and PRINCE who had clinical outcomes and fatigue scores that were not only improved but within the normal range after treatment with pegcetacoplan, eculizumab (a complement component 5 inhibitor), or supportive care (e.g., transfusions, corticosteroids, anticoagulants, supplements).
Pegcetacoplan was superior to both eculizumab and supportive care in normalization of hemoglobin concentrations, lactate dehydrogenase concentrations, and fatigue scores in patients with PNH who had anemia after at least 3 months of eculizumab treatment, and in patients who had never received a complement component 5 inhibitor.

1 Introduction

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired, life-threatening disease characterized by chronic complement-mediated hemolysis, thrombosis, bone marrow failure, and fatigue that impairs patients’ quality of life (QoL) [1, 2]. It is caused by acquired somatic mutations in the PIG-A gene in hematopoietic stem cells that prevents the complement inhibitors CD55 and CD59 from being expressed on red blood cells (RBCs) [24]. RBCs derived from PNH hematopoietic stem cells are highly susceptible to complement-mediated lysis [2].
Clinical manifestations of PNH include anemia, abdominal pain, erectile dysfunction, and dysphagia, secondary to intravascular hemolysis (for which elevated lactate dehydrogenase [LDH] is a biomarker) [2, 5]. Other disease manifestations include thrombotic events, renal insufficiency, and bone marrow failure, as well as debilitating symptoms, such as fatigue or dyspnea, that impair QoL [2, 5].
Terminal complement activation and subsequent intravascular hemolysis in PNH are inhibited by complement component 5 (C5) inhibitors (e.g., eculizumab and ravulizumab) [2, 68]. However, a substantial proportion of patients experience complement component 3 (C3)-mediated extravascular hemolysis during C5 inhibitor treatment [2, 9]. Up to 88% of patients receiving C5 inhibitors continue to have some degree of anemia due to residual intravascular hemolysis and emerging extravascular hemolysis, and up to 52% have required one or more RBC infusions within a 12-month period [2, 10]. In countries where complement inhibitors are not available, only supportive care (e.g., blood transfusions, prophylactic anticoagulants, corticosteroids, supplements) that manages PNH symptoms is available [11].
Pegcetacoplan is the first C3-targeting therapy for PNH and provides comprehensive hemolysis control [12, 13]. Pegcetacoplan studies in PNH have demonstrated rapidly improved clinical and fatigue parameters and a favorable safety profile [1417]. Previously reported results have not provided a comprehensive picture of the proportion of patients with PNH whose hematologic and quality-of-life outcomes improve to normal with pegcetacoplan treatment.
In the current analysis, we determined normalization rates for hemoglobin, LDH, and fatigue in patients with PNH treated with pegcetacoplan in two phase III trials: PEGASUS (pegcetacoplan vs. eculizumab in adults with hemoglobin < 10.5 g/dL despite ≥ 3 months of eculizumab therapy) and PRINCE (pegcetacoplan vs. supportive care only in complement inhibitor–naive patients) [14, 16, 17].

2 Methods

Patients enrolled in the PEGASUS trial [14] (NCT03500549) had PNH and anemia (hemoglobin < 10.5 g/dL) despite ≥ 3 months of eculizumab treatment. Patients enrolled in the PRINCE trial [16] (NCT04085601) were naive to complement inhibitor therapy (i.e., had not received treatment with any complement inhibitor [e.g., eculizumab, ravulizumab] within 3 months prior to screening), had hemoglobin concentrations below the lower limits of normal (females: < 12.0 g/dL; males: < 13.6 g/dL) and LDH concentrations ≥ 1.5 times the upper limit of normal (≥ 339 U/L), and received supportive care only (blood transfusions, corticosteroids, anticoagulants, and supplements [iron, folate, vitamin B12]) in countries where complement inhibitors were not approved or widely available. Patients receiving supportive care in the PRINCE study could switch to pegcetacoplan treatment if their hemoglobin concentration decreased ≥ 2 g/dL below their baseline concentration or if they had a qualifying thromboembolic event secondary to PNH. In both PEGASUS and PRINCE, specific objective criteria for transfusions (hemoglobin < 7 g/dL regardless of whether symptoms are present, or < 9 g/dL with symptoms) were applied regardless of treatment assignment.
Normalization of hematologic and fatigue endpoints were assessed in PEGASUS at Week 16 (randomized controlled period; pegcetacoplan and eculizumab groups), and Week 48 (open-label period; pegcetacoplan-to-pegcetacoplan and eculizumab-to-pegcetacoplan groups) and in PRINCE at Week 26 (pegcetacoplan and supportive care [control] groups). Normalization for each endpoint was defined as follows:
o
Hemoglobin greater than or equal to the lower limit of the sex-specific normal range (females: 12.0 g/dL; males: 13.6 g/dL).
 
o
LDH ≤226 U/L, the upper limit of normal.
 
o
Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-Fatigue) ≥43.6, the general US population norm [18].
 
Patients who received a transfusion (hemoglobin/LDH endpoints only), withdrew from the study, were lost to follow-up, or switched from control (supportive care only) to pegcetacoplan were considered not normalized. No additional statistical analysis was performed on the results from the PEGASUS and PRINCE trials.
Investigators reported treatment-emergent adverse events (TEAEs) by the Medical Dictionary for Regulatory Activities preferred terms through the end of the open-label period of PEGASUS and the end of PRINCE. The TEAEs of injection site reactions, infections, hemolytic disorders, thrombosis, and hypersensitivity were of special interest because of their relevance to PNH and potential relatedness to pegcetacoplan’s mechanism of action or route of administration. Event seriousness, severity, and relatedness to pegcetacoplan were determined by investigators. An adverse event (AE) was considered serious if the investigator viewed it as resulting in death, a life-threatening AE, inpatient hospitalization or prolongation of ongoing hospitalization, a persistent or significant incapacity or a substantial disruption of normal life functions, or a congenital anomaly or birth defect. Events that did not meet these criteria could have been considered serious if they jeopardized the patient’s health and required medical or surgical interventions to prevent one of the outcomes that defined a serious AE. The severity of an AE was defined as mild (i.e., resulted in mild or transient discomfort that did not limit or interfere with activities; did not require treatment), moderate (i.e., resulted in sufficient discomfort to limit or interfere with activities; may have required treatment), or severe (i.e., resulted in significant symptoms that prevented normal activities; may have required hospitalization or invasive intervention).
Original study protocols were designed and monitored in accordance with the ethical principles of Good Clinical Practice and the Declaration of Helsinki, and approved by an Institutional Review Board or independent Ethics Committee at each center [14, 16, 17]. Patients provided written informed consent before initiation of study-related procedures.

3 Results

3.1 Baseline Population and Characteristics

In PEGASUS, 41 patients were randomly assigned to receive pegcetacoplan and 39 to receive eculizumab during the 16-week randomized controlled period, after which 38 patients in the pegcetacoplan group continued and all 39 eculizumab-treated patients switched to pegcetacoplan during the 32-week open-label period. In PRINCE, 53 complement inhibitor–naive patients were randomized to pegcetacoplan treatment (n = 35; in addition to supportive care) or to continued supportive care (control; n = 18) for 26 weeks. Among the 18 patients in the supportive care group, 11 switched to pegcetacoplan treatment during the study.
Age was similar across treatment groups in both studies (PEGASUS, pegcetacoplan: mean [range] 50.2 [19–81] years; eculizumab: 47.3 [23–78] years; PRINCE, pegcetacoplan: mean [range] 42.2 [22–67] years; control/supportive care: 49.1 [20–74] years). Likewise, the percentage of female patients was similar across treatment groups in both studies: 66% and 56% in the pegcetacoplan and eculizumab groups, respectively, in PEGASUS, and 45.7% and 44.4% in the pegcetacoplan and supportive care groups, respectively, in PRINCE. In PEGASUS, the median (range) number of years since PNH diagnosis was slightly lower among pegcetacoplan-treated patients than eculizumab-treated patients: 6.0 (1–31) versus 9.7 (1–38), respectively, whereas in PRINCE, this variable was similar across treatment groups (pegcetacoplan: median [range] 3.4 [0.1–27.0] years; supportive care group: 4.7 [0.1–15.1] years).

3.2 Hemoglobin Normalization Rates

In both studies, normalization of hemoglobin concentrations was observed as early as Week 2 in a substantial percentage of pegcetacoplan-treated patients (PEGASUS: 43.9%; PRINCE: 22.9%). Hemoglobin normalization in the absence of transfusion was maintained in 34.1% of pegcetacoplan-treated patients at the end of the 16-week randomized controlled period in PEGASUS and in 45.7% of pegcetacoplan-treated patients at the end of the 26-week treatment period in PRINCE (Fig. 1). The median (range) time to hemoglobin normalization with pegcetacoplan was 4.1 (1.1–48.3) weeks in PEGASUS and 6.1 (2–42.1) weeks in PRINCE (note that some patients received pegcetacoplan longer than the trial end period in both PEGASUS and PRINCE due to coronavirus disease 2019 [COVID-19]-related shutdowns). No patients treated with eculizumab in PEGASUS or randomized to supportive care in PRINCE had hemoglobin normalization at the end of 16 or 26 weeks, respectively.
At the end of the open-label period in PEGASUS (Week 48), approximately one-quarter (24.4%) of patients treated with pegcetacoplan during both the 16-week randomized controlled period and the 32-week open-label period had hemoglobin normalization in the absence of transfusion. Among patients treated with eculizumab through Week 16 who then switched to pegcetacoplan through Week 48, 30.8% had normalized hemoglobin concentrations in the absence of transfusion (Fig. 1). Compared with a median (range) time to hemoglobin normalization of 4.1 (1.1–48.3) weeks for patients who received pegcetacoplan throughout PEGASUS, this was 3.2 (3.1–15.6) weeks for patients who switched from eculizumab to pegcetacoplan.

3.3 Lactate Dehydrogenase Normalization Rates

As early as Week 2, a substantial percentage of pegcetacoplan-treated patients in both studies had normalization of LDH concentrations in the absence of transfusion (PEGASUS: 87.8%; PRINCE: 51.4%). Normalized LDH concentrations in the absence of transfusion occurred in 70.7% of pegcetacoplan-treated patients at the end of the 16-week randomized controlled period in PEGASUS (vs. 15.4% of eculizumab-treated patients) and in 65.7% of pegcetacoplan-treated patients at the end of the 26-week treatment period in PRINCE (vs. none in the control group) (Fig. 2); the median (range) time to normalization with pegcetacoplan was 1 (1–29) day in PEGASUS and 18 (14–168) days (2.6 [2–24] weeks) in PRINCE.
At the end of the open-label period in PEGASUS (Week 48), more than half (56.1%) of patients treated with pegcetacoplan only during both the 16-week randomized controlled period and the 32-week open-label period had normalized LDH concentrations in the absence of transfusion, and 51.3% of patients treated with eculizumab through Week 16 who switched to pegcetacoplan through Week 48 had normalized LDH concentrations (Fig. 2). Normalization for eculizumab-to-pegcetacoplan patients occurred after a median (range) duration of 1 (1–22) day, similarly to 1 (1–29) day for pegcetacoplan-to-pegcetacoplan patients.

3.4 Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue Normalization Rates

In PEGASUS, almost half (48.8%) of pegcetacoplan-treated patients (vs. 10.3% of eculizumab-treated patients) had normalization of FACIT-Fatigue scores by the end of the 16-week randomized controlled period (Fig. 3), with a median (range) time to normalization of 2.3 (0.1–40.1) weeks for patients who received pegcetacoplan. In the PRINCE trial, 68.6% of pegcetacoplan-treated patients (vs. 11.1% of patients who received supportive care) had normalization of FACIT-Fatigue scores at the end of the 26-week treatment period; the median (range) time to normalization was 4.1 (0.1–16.4) weeks for patients in the pegcetacoplan group.
At the end of the open-label period in PEGASUS (Week 48), approximately one-third (34.1%) of patients treated with pegcetacoplan during both the 16-week randomized controlled period and the 32-week open-label period had FACIT-Fatigue score normalization (Fig. 3). Among patients treated with eculizumab through Week 16 who switched to pegcetacoplan through Week 48, approximately half (51.3%) had FACIT-Fatigue score normalization (median [range] time to normalization: 1.14 [0.14–5.14] weeks).

3.5 Adverse Events

In both the randomized controlled period of the PEGASUS and PRINCE studies, the overall percentage of patients with TEAEs was similar in both treatment groups (Table 1). Most TEAEs were mild or moderate in severity.
Table 1
Pegcetacoplan safety profile in the PEGASUS and PRINCE studies
PEGASUS
Randomized controlled period
Total OLP
PEG [n = 41]
ECU [n = 39]
PEG [n = 77]
Any TEAE
36 (87.8)
36 (92.3)
71 (92.2)
 PEG-related
14 (34.1)
34 (44.2)
Serious TEAEs
7 (17.1)
5 (12.8)
18 (23.4)
 PEG-related
1 (2.4)
4 (5.2)
TEAEs leading to study discontinuation
3 (7.3)
0
9 (11.7)
Drug-related TEAEs leading to study discontinuation
2 (4.9)
0
4 (5.2)
TEAEs of special interesta
 Injection site reactions
15 (36.6)
1 (2.6)
20 (26.0)
 Infections
12 (29.3)
11 (28.2)
43 (55.8)
 Hypersensitivity
5 (12.2)
2 (5.1)
14 (18.2)
 Hemolytic disorders
5 (12.2)
14 (35.9)
18 (23.4)
 Sepsis
0
0
3 (3.9)
 Thrombosis
0
0
2 (2.6)
PRINCE
Overall PEGb [N = 46]
Control [N = 18]
Any TEAE
33 (71.7)
12 (66.7)
 PEG-related
13 (28.3)
Serious TEAEs
4 (8.7)
3 (16.7)
 PEG-related
0
TEAEs leading to study discontinuation
0
TEAEs of special interesta
 Injection site reactions
14 (30.4)
 Infections
9 (19.6)
5 (27.8)
 Hypersensitivity
9 (19.6)
1 (5.6)
 Hemolytic disorders
0
0
 Thrombosis
0
0
 Sepsis
0
0
Data are expressed as n (%)
ECU eculizumab, OLP open-label period, PEG pegcetacoplan, PNH paroxysmal nocturnal hemoglobinuria, TEAE treatment-emergent adverse event
aInjection site reactions, infections, hemolytic disorders, thrombosis, sepsis, and hypersensitivity
bIncludes patients randomized to the control arm who had the option to switch to PEG if they demonstrated hemoglobin concentrations ≥ 2 g/dL below baseline measurement or presented with a qualifying thromboembolic event secondary to PNH
In the randomized controlled period of PEGASUS, 1 patient (2.4%) had a serious TEAE that investigators deemed related to pegcetacoplan. Three patients (7.3%), all receiving pegcetacoplan, had a TEAE that led to study discontinuation (hemolysis in all cases). Two patients (4.9%) had a pegcetacoplan-related TEAE that resulted in study discontinuation (hemolysis in both cases). No patients had a thrombotic event.
In the PRINCE trial, no pegcetacoplan-related TEAEs were considered serious. No TEAEs led to study discontinuation. No thrombotic events were reported in either treatment group.
Injection site reactions were the most common TEAE of special interest reported in pegcetacoplan-treated patients in both the PEGASUS randomized controlled period (36.6%) and the PRINCE trial (30.4%). Most reactions were mild, occurred early in the trials, and did not lead to any discontinuations.
In the open-label period of PEGASUS, TEAEs occurred in 71/77 patients (92.2%). Four patients (5.2%) had serious TEAEs that were pegcetacoplan-related. A total of 9 (11.7%) patients had a TEAE that led to study discontinuation, including 2 (2.6%) patients with hemolysis and 1 (1.3%) each with bone marrow failure, hemolytic anemia, acute myeloid leukemia, diffuse large B-cell lymphoma, intestinal ischemia, COVID-19, and hypersensitivity pneumonitis. Possible drug-related TEAEs leading to study discontinuation occurred in 4 patients (5.2%) during the open-label period (one each of hemolysis, hemolytic anemia, hypersensitivity pneumonitis, and mesenteric ischemia [the last one during the trial follow-up period]). Two patients (2.6%) had thrombotic events that were considered not related to pegcetacoplan.

4 Discussion

In this analysis of two phase III trials, PEGASUS and PRINCE, pegcetacoplan treatment rapidly resulted in sustained normalization of hemoglobin concentrations (in the absence of transfusions) and fatigue scores in a substantial percentage of patients with PNH compared with eculizumab or supportive care. Rates of LDH normalization (in the absence of transfusion), indicative of reduced intravascular hemolysis, were higher with pegcetacoplan versus eculizumab or supportive care. These results corroborate findings of rapidly improved clinical and fatigue parameters in pegcetacoplan studies in PNH [1417]. Across treatment groups, the median times to normalization among patients receiving pegcetacoplan were the shortest for LDH (1 day–2.5 weeks), followed by FACIT-Fatigue (2.3–4.1 weeks), and hemoglobin (4.1–6.1 weeks). PEGASUS patients normalized faster than PRINCE patients, and within PEGASUS, patients switching from eculizumab to pegcetacoplan normalized as fast (or slightly faster) as those who had been initially randomized to pegcetacoplan.
Studies of eculizumab and ravulizumab in PNH have demonstrated improvement in hemoglobin concentrations, although hemoglobin normalization occurs in only about 20–30% of patients treated with C5 inhibitors [19, 20]. Additionally, a recent survey-based study found high rates of persistent anemia, fatigue, and poor QoL among patients treated with C5 inhibitors [10]. The results of the current analyses demonstrate that pegcetacoplan fills unmet needs in the treatment of PNH by reducing rates of anemia and fatigue, and achieving better control of hemolysis compared with C5 inhibitors. In PEGASUS, hemoglobin, FACIT-Fatigue, and LDH normalization rates were markedly higher with pegcetacoplan versus eculizumab in patients who had anemia despite eculizumab treatment. This higher rate of LDH normalization with pegcetacoplan versus eculizumab (70.7% vs. 15.4%) may indicate that eculizumab was not sufficiently blocking the terminal complement pathway in these patients, resulting in residual intravascular hemolysis that was subsequently normalized with pegcetacoplan. In the complement inhibitor-naive patients in PRINCE, hemoglobin, FACIT-Fatigue, and LDH normalization rates were substantially higher with pegcetacoplan than with supportive care alone.
It is worth noting that although the hemoglobin normalization rates were markedly improved with pegcetacoplan, some patients did not have normalization. This could be explained by the bone marrow disorders, such as aplastic anemia, that are often present in patients with PNH, which limit the capacity for red blood production [21]. Future research that identifies common characteristics of patients without hemoglobin normalization, and the rates of normalization for patients with suboptimal bone marrow function, could identify patients who are likely to be less responsive to pegcetacoplan and develop appropriate treatment goals for these patients.
We observed that the percentages of patients with hemoglobin, LDH, and FACIT-Fatigue normalization decreased somewhat from week 16 to week 48 in eculizumab-experienced patients who were randomized to pegcetacoplan. The decreased normalization at week 48 could be an artifact of the analysis, in which patients who discontinued, died, or were censored because of transfusions were retained in the denominator, even though they could not have had normalization; this could have decreased the reported percentage of patients with normalization. It is also possible that the decreased normalization rates reflect variations in hemolysis that occurred spontaneously or because of complement-amplifying conditions (CACs; e.g., vaccinations, infections) [22]. CACs can reduce disease control because complement inhibitor therapy does not completely suppress the complement system, leaving room for hemolysis in times of high complement pathway activation [23]. Because this activation cannot always be avoided, the reasonable approach to address such hemolysis is to prevent CACs when possible and initiate an effective strategy to treat hemolysis events when they occur. Recent publications have shown that breakthrough hemolysis events in patients receiving pegcetacoplan can be well managed with temporary dosage increases [22, 23].
Despite the possibility of increased risk of encapsulated bacterial infections with broad complement inhibition [24], no cases of meningococcal infection were noted in either trial, and a single case of sepsis (biliary) possibly related to pegcetacoplan was reported in the 32-week open-label period in PEGASUS. Safety results in these studies indicate that pegcetacoplan was generally well tolerated.
This study had several limitations. This analysis was not designed to identify the percentage of patients who had normalization of all three endpoints (i.e., hemoglobin, LDH, and FACIT-Fatigue). Another limitation is that the normalization analyses included all patients, regardless of their eligibility for normalization, which could have resulted in artificially low normalization rates. In addition, a longer study duration would have provided more information about response durability. Future studies to identify clinical markers that predict normalization with pegcetacoplan and to discern differences in safety by normalization status could provide useful guidance for treatment decisions.

5 Conclusions

These results demonstrate that pegcetacoplan is superior to both eculizumab and supportive care in achieving normalization of hemoglobin, LDH, and FACIT-Fatigue scores in patients with PNH and persistent anemia despite at least 3 months of treatment with eculizumab, and in C5 inhibitor–naive patients with PNH.

Acknowledgments

The authors thank Jessica Savage, MD, for her early reviews and feedback on the manuscript. Writing assistance, provided by Angela Cimmino, PharmD, and Kathryn Fogarty, PhD (Kay Square Scientific, Newtown Square, PA, USA) was funded by Apellis Pharmaceuticals Inc., and Swedish Orphan Biovitrum AB.

Declarations

Funding

This study was sponsored by Apellis Pharmaceuticals, Inc., and Swedish Orphan Biovitrum AB. The sponsors also contributed to data interpretation and manuscript development. Writing assistance was funded by Apellis Pharmaceuticals, Inc., and Swedish Orphan Biovitrum AB.

Conflicts of Interest

Brian P. Mulherin has no disclosures to declare. Michael Yeh is an employee of Apellis and holds stock options. Mohammed Al-Adhami was an employee of Apellis at the time of the study. David Dingli serves on advisory boards with compensation from Alexion (AstraZeneca), Apellis, Janssen, Novartis, and Sanofi; and receives research support from K-36 Therapeutics.

Ethics Approval

Original study protocols were designed and monitored in accordance with the ethical principles of Good Clinical Practice and the Declaration of Helsinki, and approved by an Institutional Review Board or independent Ethics Committee at each center.
Patients provided written informed consent before the initiation of study-related procedures.
Participants consented to publication provided their confidentiality would be maintained, which it is in the data reported in this manuscript.

Data Availability

The datasets generated and/or analyzed during the current study are not publicly available but are available from the corresponding author on reasonable request.

Code Availability

Not applicable.

Author Contributions

All authors contributed to the conception and design of this analysis, the interpretation of data, and the critical revision of the manuscript; agree to be accountable for ensuring the integrity and accuracy of the work; and approved the final version of the manuscript.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Literatur
12.
Zurück zum Zitat EMPAVELI (pegcetacoplan). Package insert. Apellis Pharmaceuticals; 2023. EMPAVELI (pegcetacoplan). Package insert. Apellis Pharmaceuticals; 2023.
13.
Zurück zum Zitat ASPAVELI (pegcetacoplan). Summary of product characteristics. Apellis Pharmaceuticals; 2022. ASPAVELI (pegcetacoplan). Summary of product characteristics. Apellis Pharmaceuticals; 2022.
Metadaten
Titel
Normalization of Hemoglobin, Lactate Dehydrogenase, and Fatigue in Patients with Paroxysmal Nocturnal Hemoglobinuria Treated with Pegcetacoplan
verfasst von
Brian P. Mulherin
Michael Yeh
Mohammed Al-Adhami
David Dingli
Publikationsdatum
10.05.2024
Verlag
Springer International Publishing
Erschienen in
Drugs in R&D
Print ISSN: 1174-5886
Elektronische ISSN: 1179-6901
DOI
https://doi.org/10.1007/s40268-024-00463-9