Introduction: The study aimed to assess the safety, immunogenicity, and efficacy of long-term therapy with biosimilar of eculizumab (Elizaria®) in paroxysmal nocturnal hemoglobinuria (PNH) patients. Methods: The study included 30 patients with PNH who had completed previous clinical trials. Of these, 25 patients continued receiving the biosimilar product, and 5 patients switched from the originator product Soliris. The maximum duration of follow-up was 104 weeks, during which the investigational product was administered 52 times at a standard dose. Results: Throughout the study, the levels of lactate dehydrogenase, hemoglobin, reticulocytes, and PNH clone remained stable compared to baseline, regardless of the previous therapy (p > 0.05). There were no significant differences in the number of patients with chronic kidney disease at different visits, as well as in the number of patients who received donor red blood cell and platelet transfusions during the study (p > 0.05). There were 2 cases of adverse reactions reported in 2 patients (6.6%): elevated aspartate aminotransferase (3.3%) and alopecia (3.3%). Immunogenicity analysis showed no significant differences in the frequency of antidrug antibody detection compared to baseline (p > 0.05). Conclusion: The study findings confirm the long-term efficacy and safety of biosimilar in patients with PNH.

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare clonal hematopoietic stem cell disease that presents with hemolytic anemia, thrombosis, and smooth muscle dystonias, and in some cases is associated with bone marrow failure [1]. The disease is associated with the expansion of hematopoietic stem cell clones with a somatic mutation of the PIGA gene. This impairs the synthesis of the carbohydrate moiety of the glycosylphosphatidylinositol (GPI) anchor of GPI-anchored proteins, including membrane complement inhibitors CD55 and CD59 [2, 3]. The absence of CD59 on PNH red blood cells leads to uncontrolled formation of membrane attack complexes (MACs) [4, 5], which results in red blood cell hemolysis, release of hemoglobin into the plasma, nitric oxide (NO) consumption, and platelet activation [6, 7].

The inhibition of MAC formation using eculizumab, a humanized anticomplement component C5 antibody, remains the mainstay of treatment for PNH [7, 8]. Several studies have shown successful control of intravascular hemolysis and prevention of life-threatening complications both in clinical trials and in real-world clinical practice [9‒11].

Elizaria® developed as a biosimilar has been used in the Russian Federation since 2019 for the treatment of PNH and atypical hemolytic uremic syndrome [12]. The biosimilar product was developed in accordance with the regulatory requirements of the Eurasian Economic Union (EAEU) and the European Medicines Agency (EMA) [12‒15]. In the early stages of clinical development of biosimilar, phase I and Ib clinical trials were conducted to confirm sufficient safety and tolerability of the product, as well as to study the pharmacokinetic (PK) and pharmacodynamic (PD) properties of investigated product following its single-dose administration in healthy volunteers and as induction therapy in treatment-naive PNH patients [16]. A phase III clinical trial (NCT04463056 on clinicaltrial.gov) has demonstrated non-inferiority of the investigational biosimilar product to the originator product Soliris [17]. This clinical trial ECU-PNH-III-X (NCT04679103, clicaltrials.gov) was carried out to assess the long-term safety, efficacy, and immunogenicity of eculizumab biosimilar in patients with PNH.

Patients

An open-label, prospective, multicenter study of the long-term safety, immunogenicity, PKs, PDs, and efficacy of eculizumab biosimilar in patients who had previously completed the ECU-PNH-III and ECU-PNH-Ib clinical trials was conducted at 8 study sites (https://clinicaltrials.gov/study/NCT04671810). After signing the informed consent form, 30 patients with PNH aged 23–75 years were enrolled in the clinical trial. Prior to this study, 25 patients received Elizaria® (Group A), and 5 patients received Soliris® (Group B). All patients were vaccinated against meningococcal infection with a tetravalent vaccine (Neisseria meningitidis serotypes A, C, Y, and W135), and also agreed to use reliable methods of contraception throughout the study period and for 10 weeks after the last dose of the investigational product.

Patients with hypersensitivity to the investigational product and its components, as well as active ingredients and components of the meningococcal vaccine, patients with a history of infectious diseases caused by N. meningitidis, and patients with a concomitant disease or condition that the investigator believed could endanger the patient’s safety during the study, were excluded.

A patient could be excluded during the study for the following reasons: ≥2 events of grade 3 toxicity, or ≥1 event of grade 4 toxicity according to the CTCAE 4.03, which the investigator considered as associated with the investigational product; diagnosis of cancer; meningococcal infection; treatment with immunosuppressants, except for eculizumab, glucocorticosteroids, androgens, erythropoietin products, or adjusted-dose warfarin; refusal to participate or noncompliance with study procedures; significant deviations from the protocol (ver. 4.0 dated 13.07.2018). Any outcome data were collected for participants who could discontinue or deviate from intervention protocols.

Study Design

The open-label, prospective, multicenter study included the following periods: the end of study visit of previous trials (Switching Visit), which was the first visit of this study; the treatment period (up to 104 weeks); and the follow-up period (2 weeks). If the product became commercially available to an individual patient, the patient would withdraw from the study.

Study Treatment

The patients received the biosimilar at a maintenance dose of 900 mg every 2 weeks. The maximum duration of treatment was 104 weeks. During the study, patients received from 18 to 44 injections of the biosimilar, the average number of injections was 32.33 ± 6.74). In previous clinical trials, all patients were immunized with a meningococcal vaccine at least 14 days before the first eculizumab infusion or received preventive antibacterial therapy for at least 2 weeks after vaccination. Re-immunization during the clinical study was possible, if necessary.

Examination

The study procedures included physical examination, anthropometry, assessment of vital signs, electrocardiography, and Doppler echocardiography with an assessment of the mean pulmonary artery pressure and the degree of pulmonary hypertension. Blood samples were taken for red blood cell/granulocyte PNH clone tests (according to the high-sensitivity flow cytometry protocol [18]), complete blood count and blood chemistry with glomerular filtration rate (GFR) calculation, coagulation profile, and D-dimer test. Urine samples were taken for routine urinalysis.

The PK analysis (Cmin, which is the minimum product concentration at the end of the dosing interval) included the data of 5 patients from group B. Blood samples for the PK/PD studies were taken at visit 1 and after 2, 4, and 6 weeks of treatment, 5 min pre-dose. The analysis was performed using Octet® QKe System interferometer (Pall ForteBio). All calculations were performed automatically using Octet® Software, v.10.0 (Pall ForteBio). The analytical range for quantitative determination of eculizumab in serum samples was 6.25–1,200 μg/mL. The PD analysis included the measurement of membrane attack complex serum concentration (MAC, C5b-9) by enzyme-linked immunosorbent assay (MicroVue Complement SC5b-9 Plus EIA [Quidel, San Diego, CA, USA]) at the same time periods as for the PK analysis.

The therapy safety was assessed by the incidence and severity of adverse reactions (ARs) according to symptoms, physical examination, assessment of vital signs, electrocardiography, laboratory and instrumental findings, and patient diaries. All adverse events (AEs) reported in the study were analyzed by their association with the investigational products (definite, probable, or possible), system organ classes, preferred terms, and treatment groups. The AEs and ARs were coded using the Medical Dictionary for Regulatory Activities (MedDRA), version 21.0, and presented with the aggregation of preferred terms by System Organ Classes.

Immunogenicity was determined by the level of antidrug antibodies (ADAs), including the neutralizing activity of antibodies to eculizumab, by bridging enzyme-linked immunosorbent assay. The assay was carried out at Weeks 12, 24, 36, 48, 60, 72, 84, and 102 during the treatment period. The immunogenicity analysis was performed considering the total duration of therapy, including the results of previous studies. The analysis results before the first dose of the product in previous clinical studies (ECU-PNH-Ib and ECU-PNH-III) were considered as baseline.

Efficacy Parameters

Efficacy variables included changes in the levels of lactate dehydrogenase (LDH), hemoglobin, reticulocytes, and red blood cell/granulocyte PNH clone. In addition, the number of patients requiring blood transfusions was evaluated.

Statistical Methods

R 4.0.3 [19] was used for statistical analysis, which evaluated 2 patient populations. All patients enrolled in the study comprised the full analysis set (FAS). This population was used to assess the efficacy, safety, and immunogenicity, as well as to describe baseline characteristics, including patient demographics, anthropometric parameters, and underlying and concomitant diseases. The population for PK and PD analysis (PK/PD population) included all patients with data available to assess at least one of the prespecified PK/PD parameters. The analysis was performed in two groups of patients depending on the previous therapy: group A included patients who continued receiving the biosimilar, and group B included patients who switched from the originator product to the biosimilar product.

Qualitative attributes were described using the absolute number of observations, percentages, and two-sided 95% CIs for percentages. Nonparametric Mann-Whitney and Friedman tests were used to compare quantitative parameters between groups and changes in these parameters between visits in each group, respectively, since all study populations and groups were represented by small subsets.

Patients

The study enrolled 30 patients, of which 25 (83%) and 5 (17%) previously received biosimilar (group A) and originator (group B), respectively. All patients completed the study according to the protocol when the biosimilar became commercially available, each at their own visit. As a result, the data at the end of study visit were actually collected at several different time points. The PK/PD population accounted for 16.67% of the FAS population (5 patients) and included only patients from group B.

The median age of patients was 37 years (39 years in group A and 33 years in group B). The study included 12 male and 18 female patients. Patients in groups A and B were comparable in terms of all baseline characteristics (shown in Table 1).

Table 1.

Baseline characteristics of enrolled patients

VariableTotal (N = 30)Group A (N = 25)Group B (N = 5)p value
Age, years 
 Median 37 39 33 0.87 
 Q1; Q3 30; 57.5 30; 56 30; 60  
Female, n (%) 18 (60) 14 (56) 5 (80) 0.622 
Caucasian, na 30 25 1.000 
History of thrombosis, n (%) 6 (20) 6 (24) 0 (0) 0.553 
History of aplastic anemia, n (%) 7 (23.33) 6 (24) 1 (20) 1.000 
LDH, U/L (ULN: 250 U/L) 
 Median 320 324 271 0.21 
 Q1; Q3 235.75; 377.5 241; 390 185; 336  
Red blood cell PNH clone (II+III type), % 
 Median 66.35 65.1 67.6 0.78 
 Q1; Q3 48; 76.97 48.8; 77.1 54; 75.3  
Granulocyte PNH clone, % 
 Median 96.7 96.5 98.2 1.000 
 Q1; Q3 90.72; 99.25 89.5; 99.4 96.4; 99.1  
Hemoglobin, g/L 
 Median 97.5 94 110 0.21 
 Q1; Q3 85; 109 85; 105 103; 124  
Glomerular filtration rate, mL/min/1.732b 
 Median 91.65 90.3 116.2 0.21 
 Q1; Q3 77.68; 110.3 75.2; 106.2 108.5; 123.4  
Mean pulmonary artery pressure, mm Hg 
 Median 17 17 25 0.91 
 Q1; Q3 12.25; 24.5 12; 20 4; 30  
MAC, ng/mL 
 Median NA NA 70 NA 
 Q1; Q3 NA NA 56.5; 83.75  
Eculizumab Cmin, μg/mL 
  NA NA 141.45 NA 
  NA NA 90.12; 179.1  
VariableTotal (N = 30)Group A (N = 25)Group B (N = 5)p value
Age, years 
 Median 37 39 33 0.87 
 Q1; Q3 30; 57.5 30; 56 30; 60  
Female, n (%) 18 (60) 14 (56) 5 (80) 0.622 
Caucasian, na 30 25 1.000 
History of thrombosis, n (%) 6 (20) 6 (24) 0 (0) 0.553 
History of aplastic anemia, n (%) 7 (23.33) 6 (24) 1 (20) 1.000 
LDH, U/L (ULN: 250 U/L) 
 Median 320 324 271 0.21 
 Q1; Q3 235.75; 377.5 241; 390 185; 336  
Red blood cell PNH clone (II+III type), % 
 Median 66.35 65.1 67.6 0.78 
 Q1; Q3 48; 76.97 48.8; 77.1 54; 75.3  
Granulocyte PNH clone, % 
 Median 96.7 96.5 98.2 1.000 
 Q1; Q3 90.72; 99.25 89.5; 99.4 96.4; 99.1  
Hemoglobin, g/L 
 Median 97.5 94 110 0.21 
 Q1; Q3 85; 109 85; 105 103; 124  
Glomerular filtration rate, mL/min/1.732b 
 Median 91.65 90.3 116.2 0.21 
 Q1; Q3 77.68; 110.3 75.2; 106.2 108.5; 123.4  
Mean pulmonary artery pressure, mm Hg 
 Median 17 17 25 0.91 
 Q1; Q3 12.25; 24.5 12; 20 4; 30  
MAC, ng/mL 
 Median NA NA 70 NA 
 Q1; Q3 NA NA 56.5; 83.75  
Eculizumab Cmin, μg/mL 
  NA NA 141.45 NA 
  NA NA 90.12; 179.1  

MAC, membrane attack complex; Cmin, the minimum product concentration at the end of the dosing interval.

aThe information on race was provided by patients themselves.

bThe glomerular filtration rate was calculated using the CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration) formula.

The most common concomitant diseases in the study subjects were vascular diseases (50% of patients) and cardiac disorders (36.7% of patients). There were no significant differences in the manifestations of PNH and incidence of various comorbidities between the groups (p > 0.05).

PKs and PDs

In group B, where patients switched from the originator to the biosimilar prior to enrollment in this study, the mean serum concentration of eculizumab was 127.77 ± 63.33 μg/mL at visit 1, 128.62 ± 57.96 μg/mL at visit 2, 112.54 ± 48.84 μg/mL at visit 3, and 126.54 ± 62.73 μg/mL at visit 4. High variability in the product concentration was observed at all time points; the coefficient of variation was close to 50% but did not exceed this value (shown in Fig. 1).

Fig. 1.

Minimum product concentration at the end of the dosing interval and membrane attack complex concentration at first study visits. PK population (N = 5).

Fig. 1.

Minimum product concentration at the end of the dosing interval and membrane attack complex concentration at first study visits. PK population (N = 5).

Close modal

At the end of each dosing interval, the mean eculizumab concentration 5 min pre-dose at all visits exceeded 35 μg/mL, which is the minimum concentration sufficient for complete inhibition of intravascular hemolysis in patients with PNH [20, 21]. The PD analysis showed stable MAC levels in the blood at all visits after switching from the originator product to the biosimilar product. The concentration of MAC was 70.25 ± 34.86 ng/mL at visit 1, 57.03 ± 32.87 ng/mL at visit 2, 59.73 ± 29.08 ng/mL at visit 3, and 63.24 ± 31.44 at Visit 4 (p = 0.66) (shown in Fig. 1).

Efficacy

The median LDH activity ranged from 226 (197.8; 300) U/L to 320 (235.8; 377.5) U/L, and median changes in LDH activity from baseline (visit 1) ranged from −3.5 (−84.3; 130.8) U/L to −98.5 (−128.3; −68.8) U/L. The analysis of changes in LDH activity showed that the median LDH activity in the FAS population was stable throughout the study (p = 0.21). There was an insignificant increase in the LDH activity at week 64 due to acute respiratory viral infection in 1 patient, and at week 68 due to an exacerbation of chronic hepatitis in another patient. The intergroup comparison demonstrated no significant differences in LDH activity at each visit, and there were no significant changes in LDH activity from baseline (visit 1) (p > 0.05) (shown in Fig. 2).

Fig. 2.

Changes in serum LDH activity during the study (median with interquartile range).

Fig. 2.

Changes in serum LDH activity during the study (median with interquartile range).

Close modal

Both groups demonstrated comparable positive mean changes in the hemoglobin level at the end of the study, which were 3.7 ± 13.6 g/L in group A and 1 ± 2.5 g/L in group B (p = 0.36). There were no significant changes in the hemoglobin level at each visit compared to baseline (visit 1) both in the general population (p > 0.05) and in groups A (p = 0.090) and B (p = 0.54) (shown in Table 2).

Table 2.

Results of comparative assessment of efficacy endpoints

Efficacy criterionTotal in the FAS populationGroup AGroup Bp value
Change in LDH activity, U/L 10.43±407.4 (95% CI: [−135.35; 156.21]) 23.36±446.53 (95% CI: [−151.68; 198.4]) −54.2±27.14 (95% CI: [−77.99; −30.41]) 0.82 
Change in the hemoglobin level, g/L 3.2±12.5 (95% CI: [−1.3; 7.7]) 3.7±13.6 (95% CI: [−1.6; 9]) 1±2.5 (95% CI: [−1.2; 3.2]) 0.36 
Change in absolute reticulocyte count, 109/L −17.92±192.78 (95% CI: [−86.9; 51.06]) −23.96±203.76 (95% CI: [−103.83; 55.91]) 12.96±203.76 (95% CI: [−103.83; 55.91]) 0.87 
Change in glomerular filtration rate, mL/min/1.732−7.31±11.32 (95% CI: [−11.36; −3.26]) −6.32±11.12 (95% CI: [−10.68; −1.96]) −12.22±12.32 (95% CI: [−23.02; −1.42]) 0.59 
Red blood cell transfusions, N (%) 8/30 (26.67) 7/25 (28) 1/5 (20) 1.000 
Breakthrough hemolysis, N (%) 4/30 (13.3) 4/25 (16) 1,000 
Change in red blood cell PNH clone size, % −2.85±17.89 (95% CI: [9.25; 3.55]) −3.2±18.42 (95% CI: [−10.42; 4.02]) −1.08±16.73 (95% CI: [−15.74; 13.58]) 0.79 
Change in granulocyte PNH clone size, % −1.64±25.25 (95% CI: [−10.68; 7.4]) −1.81±27.69 (95% CI: [−12.66; 9.04]) −0.79±4.36 (95% CI: [−4.61; 3.03]) 0.65 
Efficacy criterionTotal in the FAS populationGroup AGroup Bp value
Change in LDH activity, U/L 10.43±407.4 (95% CI: [−135.35; 156.21]) 23.36±446.53 (95% CI: [−151.68; 198.4]) −54.2±27.14 (95% CI: [−77.99; −30.41]) 0.82 
Change in the hemoglobin level, g/L 3.2±12.5 (95% CI: [−1.3; 7.7]) 3.7±13.6 (95% CI: [−1.6; 9]) 1±2.5 (95% CI: [−1.2; 3.2]) 0.36 
Change in absolute reticulocyte count, 109/L −17.92±192.78 (95% CI: [−86.9; 51.06]) −23.96±203.76 (95% CI: [−103.83; 55.91]) 12.96±203.76 (95% CI: [−103.83; 55.91]) 0.87 
Change in glomerular filtration rate, mL/min/1.732−7.31±11.32 (95% CI: [−11.36; −3.26]) −6.32±11.12 (95% CI: [−10.68; −1.96]) −12.22±12.32 (95% CI: [−23.02; −1.42]) 0.59 
Red blood cell transfusions, N (%) 8/30 (26.67) 7/25 (28) 1/5 (20) 1.000 
Breakthrough hemolysis, N (%) 4/30 (13.3) 4/25 (16) 1,000 
Change in red blood cell PNH clone size, % −2.85±17.89 (95% CI: [9.25; 3.55]) −3.2±18.42 (95% CI: [−10.42; 4.02]) −1.08±16.73 (95% CI: [−15.74; 13.58]) 0.79 
Change in granulocyte PNH clone size, % −1.64±25.25 (95% CI: [−10.68; 7.4]) −1.81±27.69 (95% CI: [−12.66; 9.04]) −0.79±4.36 (95% CI: [−4.61; 3.03]) 0.65 

The reticulocyte count in the general population was stable throughout the study; there were no significant changes compared to visit 1 (p = 0.06). The intergroup comparison showed no significant differences in the reticulocyte count at each visit, as well as compared to baseline (visit 1) (p > 0.05). At the end of the study, both groups showed insignificant changes in the reticulocyte count: −1.48 ± 7.1% in group A and 0.45 ± 5.56% in group B (p = 1.00).

With regard to changes in PNH type II + III red blood cells and granulocytes compared to baseline values at screening, similar changes in mean values were observed in both groups. At the end of the study, groups A and B demonstrated an insignificant decrease in type II + III red blood cells, which was −3.2 ± 18.42% and −1.08 ± 16.73%, respectively (p = 0.79), as well as a decrease in the percentage of PNH granulocytes, which was −1.81 ± 27.69% in group A and −0.79 ± 4.36% in group B (p = 0.65).

Breakthrough hemolysis was recorded in 4 patients in group A (shown in Table 2). In 2 patients, it occurred after a respiratory infection. In another 2 patients, only darkening of urine was observed, which did not require treatment adjustment.

Eight (27%) patients received red blood cell transfusions from the start of therapy to the end of the study, with no significant differences between groups A (28%) and B (20%). No new thrombotic events were reported in the study.

Safety

During the study period, none of the patients developed a meningococcal infection or other infections potentially associated with long-term terminal complement inhibition. None of the evaluated patients had SAEs associated with hemolysis.

Among the reported 362 AEs, only two episodes in two patients had at least a possible association with the investigational product: alopecia in group A (4%) and elevated aspartate aminotransferase in Group B (20%) (shown in online suppl. Table 1; for all online suppl. material, see https://doi.org/10.1159/000542294). These ARs were mild (elevated aspartate aminotransferase) and moderate (alopecia) according to the NCI CTCAE 4.03 and did not require drug treatment.

Immunogenicity

Throughout the study, binding ADAs were detected in 7/30 (23.3%) patients: 6/25 (24%) in group A and 1/5 (20%) in group B. According to the immunogenicity analysis, there were no significant differences between the groups with regard to the frequency of ADA detection at any study visit (p > 0.05). The ADAs detected in patients of group A did not show neutralizing activity or affect the efficacy and safety parameters in these patients. Neutralizing antibodies were detected in 1 patient who received the originator product (group B) prior to enrollment in this study. However, the formation of neutralizing antibodies in this patient did not affect the efficacy and safety parameters.

Currently, data on the efficacy and safety of new biosimilars of eculizumab in PNH are being actively published [12, 22‒25]. However, the data on long-term therapy with biosimilars of eculizumab are limited by this study.

The stable LDH activity and hemoglobin levels observed in this study indicate long-term treatment efficacy due to a decrease in hemolytic activity. The persistent need for RBC transfusions is consistent with previously published data on the long-term use of the originator product Soliris (18–29%), including the population of Russian patients [10, 11].

According to the PK/PD analysis, mean eculizumab concentration 5 min pre-dose at all visits exceeded 35 μg/mL, which is the minimum concentration sufficient for complete inhibition of intravascular hemolysis in patients with PNH. At the same time, a persistent decrease in the mean serum MAC concentration confirms the efficacy of terminal complement activation control [21, 26]. The mean eculizumab and serum MAC concentrations were stable and showed no significant differences between visits after switching from the originator to the biosimilar in group B.

The overall safety profile of the investigational product was consistent with the findings of previous studies of originator [9, 10]. The percentage of patients with reported ARs and changes in laboratory and investigational findings during long-term therapy with biosimilar was comparable regardless of previous therapy with the biosimilar or originator.

The main limitation of this clinical study was the low number of PNH patients, as well as the orphan status of eculizumab, which made it difficult to include more patients in the study, and especially in group B, given the decreased use of the originator. In addition, the study did not implement the recently proposed criteria for response to complement inhibitor therapy [27]. The study did not evaluate the prevalence of extravascular C3-mediated hemolysis, which significantly affects the quality of the erythroid response during therapy with eculizumab in PNH patients [7, 27, 28].

The anti-C5 monoclonal antibody eculizumab has been used in the treatment of PNH for 20 years. Eculizumab has revolutionized treatment, controlling intravascular hemolysis and thrombosis occurrence, with improved long-term survival [7, 29]. The development of biosimilars of eculizumab for the treatment of PNH is increasing interest and becoming more significant [12].

Therefore, this clinical study demonstrates that long-term use of biosimilar of eculizumab in the treatment of PNH patients ensures successful control of hemolytic activity of the complement system, which is the main pathogenetic mechanism of the disease. The study also confirms the safety of biosimilar in long-term treatment of PNH patients, with a low percentage of ARs and cases of ADA formation. The findings of the ECU-PNH-III-X clinical study conducted as part of the clinical development of the biosimilar confirm its efficacy and safety for long-term use in patients with PNH.

This clinical study was approved by the Ministry of Health of Russia, Approval No. 205 dated 08.05.2018. The study protocol was reviewed and approved by Ethics Council at the Ministry of Health of the Russia; meeting number 165 dated 13.03.2018. This study protocol was reviewed and approved by Ethics Committees at each of the participating sites. This full list of participating site and Ethics Committees can be found at https://clinicaltrials.gov/search?term=NCT04679103.

The study was conducted in accordance with the ethical principles of the World Medical Association (WMA) Declaration of Helsinki (last revised in 2013) and the standards of the Good Clinical Practice (GCP) Guidelines of the International Conference on Harmonization of technical requirements for registration of pharmaceuticals for human use (ICH), designed to monitor compliance with the interests and safety of the patients. Personal information about potential and enrolled participants was collected and kept in strict confidence before, during and after the study. Written informed consent to participate in the study was obtained by the investigators from all participants before their participation in the study.

Alexander D. Kulagin: Alexion Pharmaceuticals, Inc: consultancy, research funding; JSC GENERIUM: consultancy, honoraria, research funding. Vadim V. Ptushkin: Alexion Pharmaceuticals, Inc: consultancy, research funding; JSC GENERIUM: research funding; Janssen: consultancy; AbbVie: consultancy; Roche: consultancy. Elena A. Lukina: JSC GENERIUM: research funding; Sanofi Genzyme: honoraria, membership on an entity’s Board of Directors or advisory committees; Shire: Honoraria, Membership on an entity’s Board of Directors or advisory committees. Other: travel reimbursement, research funding. Oksana А. Markova: JSC GENERIUM: employment. Eugene V. Zuev: JSC GENERIUM: employment.

The study was organized and conducted with the support of GENERIUM JSC.

Contribution: AD.K., V.V.P., and E.A.L. recruited patients, collected data, analyzed and interpreted the data, contributed to the manuscript, and approved the final version; I.L.D., A.V.K., T.S.K., E.Yu.K., N.V.M., T.A.M., O.U.К., E.G.A., and V.D.L. recruited patients, collected data, contributed to the manuscript, and approved the final version; O.A.M. and E.V.Z. developed the protocol, analyzed and interpreted the data, contributed to the manuscript, and approved the final version.

The data that support the findings of this study are not publicly available due to they containing information that could compromise the privacy of research participants but are available from the corresponding author Alexander Kulagin.

1.
Hill
A
,
DeZern
A
,
Kinoshita
T
,
Brodsky
RA
.
Paroxysmal nocturnal haemoglobinuria
.
Nat Rev Dis Primers
.
2017
;
3
:
17028
.
2.
Takeda
J
,
Miyata
T
,
Kawagoe
K
,
Iida
Y
,
Endo
Y
,
Fujita
T
, et al
.
Deficiency of the GPI anchor caused by a somatic mutation of the PIG-A gene in paroxysmal nocturnal hemoglobinuria
.
Cell
.
1993
;
73
(
4
):
703
11
.
3.
Pangburn
MK
,
Schreiber
RD
,
Müller-Eberhard
HJ
.
Deficiency of an erythrocyte membrane protein with complement regulatory activity in paroxysmal nocturnal hemoglobinuria
.
Proc Natl Acad Sci USA
.
1983
;
80
(
17
):
5430
4
.
4.
Hillmen
P
,
Muus
P
,
Duhrsen
U
,
Risitano
AM
,
Schubert
J
,
Luzzatto
L
, et al
.
Effect of the complement inhibitor eculizumab on thromboembolism in patients with paroxysmal nocturnal hemoglobinuria
.
Blood
.
2007
;
110
(
12
):
4123
8
.
5.
Rother
RP
,
Bell
L
,
Hillmen
P
,
Gladwin
MT
.
The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease
.
JAMA
.
2005
;
293
(
13
):
1653
62
.
6.
Hill
A
,
Kelly
R
,
Hillmen
P
.
Thrombosis in paroxysmal nocturnal hemoglobinuria
.
Blood
.
2013
;
121
(
25
):
4985
5105
.
7.
Peffault de Latour
R
,
Hosokawa
K
,
Risitano
AM
.
Hemolytic paroxysmal nocturnal hemoglobinuria: 20 years of medical progress
.
Semin Hematol
.
2022
;
59
(
1
):
38
46
.
8.
Brodsky
R
.
How I treat paroxysmal nocturnal hemoglobinuria
.
Blood
.
2021
;
137
(
10
):
1304
9
.
9.
Hillmen
P
,
Young
NS
,
Schubert
J
,
Brodsky
RA
,
Socié
G
,
Muus
P
, et al
.
The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria
.
N Engl J Med
.
2006
;
355
(
12
):
1233
43
.
10.
Hillmen
P
,
Muus
P
,
Roth
A
,
Elebute
MO
,
Risitano
AM
,
Schrezenmeier
H
, et al
.
Long-term safety and efficacy of sustained eculizumab treatment in patients with paroxysmal nocturnal haemoglobinuria
.
Br J Haematol
.
2013
;
162
(
1
):
62
73
.
11.
Kulagin
A
,
Klimova
O
,
Rudakova
T
,
Golubovskaya
I
,
Ivanova
M
,
Smirnova
A
, et al
.
Benefits and limitations of long-term eculizumab treatment for paroxysmal nocturnal hemoglobinuria (PNH): real-world data from large cohort study in Russia
.
Blood
.
2018
;
132
(
Suppl 1
):
2589
.
12.
Kulasekararaj
A
,
Brodsky
R
,
Kulagin
A
,
Jang
J
.
Biosimilars in rare diseases: a focus on paroxysmal nocturnal hemoglobinuria
.
Haematologica
.
2023
;
108
(
5
):
1232
43
.
13.
Rules for conducting research of biological medicines of the Eurasian Economic Union
. Astana. 2016. 714 p. (Electronic source) Available from: https://docs.eaeunionorg/docs/ru-ru/01411954/cncd_21112016_89 (accessed on September 11, 2020).
14.
Guideline on similar biological medicinal products containing biotechnology-derived proteins as active substance: non-clinical and clinical issues. EMEA/CHMP/BMWP/42832/2005 Rev1. 2015. 13c
. (Electronic source). URL Available from: https://www.ema.europa.eu/en/similar-biological-medicinal-products-containing-biotechnology-derived-proteins-active-substance-non
15.
Ivanov
R
,
Sekaryova
G
,
Kravtsova
O
,
Kudlay
D
,
Lukyanov
S
,
Tikhonova
I
, et al
.
Guidelines of research biosimilar drugs
.
Farmakokinetika i Farmakodinamika Pharmacokinet Pharmacodynamics
.
2014
;
1
:
21
36
.
16.
Ptushkin
VV
,
Kulagin
AD
,
Lukina
EA
,
Davydkin
IL
,
Konstantinova
TS
,
Shamrai
VS
, et al
.
Results of phase Ib open multicenter clinical trial of the safety, pharmacokinetics and pharmacodynamics of first biosimilar of eculizumab in untreated patients with paroxysmal nocturnal hemoglobinuria during induction of therapy
.
Ter Arkh
.
2020
;
92
(
7
):
77
84
.
17.
Kulagin
AD
,
Ptushkin
VV
,
Lukina
EA
,
Davydkin
IL
,
Korobkin
AV
,
Shamrai
VS
, et al
.
Randomized multicenter noninferiority phase III clinical trial of the first biosimilar of eculizumab
.
Ann Hematol
.
2021
;
100
(
11
):
2689
98
.
18.
Sipol
AA
,
Babenko
EV
,
Borisov
VI
,
Naumova
EV
,
Boyakova
EV
,
Yakunin
DI
, et al
.
An inter-laboratory comparison of PNH clone detection by high-sensitivity flow cytometry in a Russian cohort
.
Hematology
.
2015
;
20
(
1
):
31
8
.
19.
R Core Team
.
R: a language and environment for statistical computing (electronic resource)
.
R Foundation for Statistical Computing
;
2020
. URL Available from: https://en.R-project.org/ (accessed on June 17, 2021).
20.
US Food and Drug Administration (FDA)
.
Clinical pharmacology and biopharmaceutics review
.
Silver Spring
:
US FDA
;
2007
.
21.
European Medicines Agency (EMA)
.
Soliris: EPAR—scientific discussion
.
London
:
EMA
;
2007
.
22.
Jang
,
JH
,
Demichelis Gomez
R
,
Bumbea
H
,
Nogaieva
,
L
,
Wong
,
LLL
,
Lim
,
SM
, et al
.
P829: a phase III randomized clinical trial comparing SB12 (proposed eculizumab biosimilar) with reference eculizumab in patients with paroxysmal nocturnal hemoglobinuria
.
HemaSphere
.
2022
;
6
:
723
4
.
23.
Kulasekararaj
A
,
Lanza
F
,
Arvanitakis
A
,
Langemeijer
SM
,
Chonat
S
,
Chow
V
, et al
.
Efficacy and safety of biosimilar candidate ABP 959 as compared with eculizumab reference product in paroxysmal nocturnal hemoglobinuria
.
Blood
.
2022
;
140
(
Suppl 1
):
8660
2
.
24.
Kulagin
AD
,
Bakirov
BA
,
Davydkin
IL
,
Elykomov
IV
,
Konstantinova
TS
,
Korobkin
AV
, et al
.
The efficacy and safety of eculizumab biosimilar in patients with paroxysmal nocturnal hemoglobinuria. Results of a phase III randomized open-label comparative clinical trial
.
Ter Arkh
.
2021
;
93
(
11
):
1340
8
.
25.
Ptushkin
V
,
Arshanskaya
E
,
Vinogradova
O
,
Kudlay
D
,
Nikitin
E
.
The features of COVID-19’s course and the efficacy of the gam-COVID-vac vaccine in patients with paroxysmal nocturnal hemoglobinuria
.
Hematol Rep
.
2023
;
15
(
3
):
503
12
.
26.
Wijnsma
KL
,
ter Heine
R
,
Moes
DJAR
,
Langemeijer
S
,
Schols
SEM
,
Volokhina
EB
, et al
.
Pharmacology, pharmacokinetics and pharmacodynamics of eculizumab, and possibilities for an individualized approach to eculizumab
.
Clin Pharmacokinet
.
2019
;
58
(
7
):
859
74
.
27.
Risitano
AM
,
Peffault de Latour
R
.
How we(‘ll) treat paroxysmal nocturnal haemoglobinuria: diving into the future
.
Br J Haematol
.
2022
;
196
(
2
):
288
303
.
28.
Risitano
AM
,
Notaro
R
,
Marando
L
,
Serio
B
,
Ranaldi
D
,
Seneca
E
, et al
.
Complement fraction 3 binding on erythrocytes as additional mechanism of disease in paroxysmal nocturnal hemoglobinuria patients treated by eculizumab
.
Blood
.
2009
;
113
(
17
):
4094
100
.
29.
Kelly
RJ
,
Holt
M
,
Vidler
J
,
Arnold
LM
,
Large
J
,
Forrest
B
, et al
.
Treatment outcomes of complement protein C5 inhibition in 509 UK patients with paroxysmal nocturnal hemoglobinuria
.
Blood
.
2024
;
143
(
12
):
1157
66
.