1
|
de Latour RP, Kulasekararaj A, Dingli D, Wilson K, Wojciechowski P, Hakimi Z, Nazir J, Czech B, Hillmen P, Szer J. Anchored Indirect Treatment Comparison Finds Comparable Effects of Pegcetacoplan and Iptacopan in Paroxysmal Nocturnal Haemoglobinuria. Eur J Haematol 2025. [PMID: 40285403 DOI: 10.1111/ejh.14422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Paroxysmal nocturnal haemoglobinuria (PNH) is an ultra-rare, acquired non-malignant haematological disorder characterised by thrombosis risk, serious complications and debilitating symptoms in untreated patients. OBJECTIVE This anchored indirect treatment comparison (ITC) evaluated efficacy data between proximal complement 3 inhibitor (C3i) pegcetacoplan and factor B inhibitor, iptacopan, in patients with PNH previously treated with complement 5 inhibitors (C5i; eculizumab, ravulizumab). METHODS Respective pivotal studies provided patient-level trial data for pegcetacoplan (16-week PEGASUS [NCT03500549]) and published data for iptacopan (24-week APPLY PNH [NCT04558918]). Differences in study design, duration and statistical methods between PEGASUS and APPLY PNH necessitated the comparative analyses to be conducted on secondary measures based on haemoglobin (Hb) levels, absolute reticulocyte count (ARC), lactate dehydrogenase (LDH) levels, and patient-reported outcomes on the Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-Fatigue) scale scores. The availability of a common reference C5i treatment group in both PEGASUS and APPLY PNH studies allowed anchored ITC (Bucher method). Simulated treatment comparison (STC) assessed the robustness of the main analysis. RESULTS Overall, baseline characteristics of the populations in the PEGASUS and APPLY PNH studies were broadly comparable. Anchored ITC showed comparable outcomes (mean difference, [95% CI]) on change-from-baseline to end of study for pegcetacoplan versus C5i, and iptacopan versus C5i, respectively, across endpoints: Hb level (-0.49 g/dL [-1.78, 0.80]); ARC (-34.41 × 109/L [-90.02, 21.21]); LDH level (-115.16 U/L [-244.40, 14.01]); FACIT-Fatigue score (3.57 [-5.60, 12.73]). Finally, the STC produced results consistent with the main Bucher analyses across all clinical endpoints and patient-reported fatigue, providing similar point estimates and confidence intervals. CONCLUSION This anchored ITC, based on data from pivotal trials, did not indicate significant differences in clinical or patient-reported outcomes between pegcetacoplan and iptacopan in PNH treatment. The findings suggest that PNH treatment decisions should also consider individualised disease- and patient-related factors. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03500549.
Collapse
Affiliation(s)
- Regis Peffault de Latour
- French Reference Center for Aplastic Anemia and Paroxysmal Nocturnal Hemoglobinuria, Paris, France
| | - Austin Kulasekararaj
- Department of Haematological Medicine, King's College Hospital, National Institute of Health Research/Wellcome King's Clinical Research Facility, London, UK
| | - David Dingli
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, USA
| | - Koo Wilson
- Swedish Orphan Biovitrum AB (Sobi), Stokholm, Sweden
| | | | - Zalmai Hakimi
- Swedish Orphan Biovitrum AB (Sobi), Stokholm, Sweden
| | - Jameel Nazir
- Swedish Orphan Biovitrum AB (Sobi), Stokholm, Sweden
| | - Barbara Czech
- Swedish Orphan Biovitrum AB (Sobi), Stokholm, Sweden
| | | | - Jeff Szer
- Department of Clinical Haematology, Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Kulasekararaj A, Griffin M, Piatek C, Shammo J, Nishimura JI, Patriquin C, Schrezenmeier H, Barcellini W, Panse J, Gaya A, Patel Y, Liu P, Filippov G, Sicre de Fontbrune F, Risitano A, Lee JW. Long-term efficacy and safety of danicopan as add-on therapy to ravulizumab or eculizumab in PNH with significant EVH. Blood 2025; 145:811-822. [PMID: 39700502 PMCID: PMC11867134 DOI: 10.1182/blood.2024026299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 12/21/2024] Open
Abstract
ABSTRACT Complement C5 inhibitor treatment with ravulizumab or eculizumab for paroxysmal nocturnal hemoglobinuria (PNH) improves outcomes and survival. Some patients remain anemic due to clinically significant extravascular hemolysis (cs-EVH; hemoglobin [Hb] ≤9.5 g/dL and absolute reticulocyte count [ARC] ≥120 × 109/L). In the phase 3 ALPHA trial, participants received oral factor D inhibitor danicopan (150 mg 3 times daily) or placebo plus ravulizumab or eculizumab during the 12-week, double-blind treatment period 1 (TP1); those receiving placebo switched to danicopan during the subsequent 12-week, open-label TP2 and continued during the 2-year long-term extension (LTE). There were 86 participants randomized in the study, of whom 82 entered TP2, and 80 entered LTE. The primary end point was met, with Hb improvements from baseline at week 12 (least squares mean change, 2.8 g/dL) with danicopan. For participants switching from placebo to danicopan at week 12, improvements in mean Hb were observed at week 24. Similar trends were observed for the proportion of participants with ≥2 g/dL Hb increase, ARC, proportion of participants achieving transfusion avoidance, and Functional Assessment of Chronic Illness Therapy-Fatigue scale scores. Improvements were maintained up to week 72. No new safety signals were observed. The breakthrough hemolysis rate was 6 events per 100 patient-years. These long-term data demonstrate sustained efficacy and safety of danicopan plus ravulizumab/eculizumab for continued control of terminal complement activity, intravascular hemolysis, and cs-EVH in PNH. This trial was registered at www.clinicaltrials.gov as #NCT04469465.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Hemoglobinuria, Paroxysmal/drug therapy
- Hemoglobinuria, Paroxysmal/complications
- Hemoglobinuria, Paroxysmal/blood
- Male
- Female
- Middle Aged
- Adult
- Hemolysis/drug effects
- Double-Blind Method
- Aged
- Complement Inactivating Agents/therapeutic use
- Complement Inactivating Agents/adverse effects
- Drug Therapy, Combination
- Treatment Outcome
Collapse
Affiliation(s)
- Austin Kulasekararaj
- King's College Hospital NHS Foundation Trust, NIHR/Wellcome King’s Clinical Research Facility, King's College London, London, United Kingdom
| | - Morag Griffin
- Department of Haematology, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Caroline Piatek
- Jane Anne Nohl Division of Hematology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jamile Shammo
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Christopher Patriquin
- Division of Medical Oncology and Hematology, University of Toronto, Toronto, ON, Canada
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, University Hospital of Ulm, Ulm, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Ulm, Germany
| | - Wilma Barcellini
- Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jens Panse
- Department of Oncology, Hematology, Hemostaseology, and Stem Cell Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Aachen, Germany
| | - Anna Gaya
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Peng Liu
- Alexion, AstraZeneca Rare Disease, Boston, MA
| | | | - Flore Sicre de Fontbrune
- Centre de Référence Aplasie Médullaire, Service d'Hématologie Greffe, Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Antonio Risitano
- Federico II University of Naples, Naples, Italy
- AORN Moscati, Avellino, Italy
| | - Jong Wook Lee
- Hanyang University Seoul Hospital, Seoul, Republic of Korea
| |
Collapse
|
3
|
Wong RSM, Ho Jang J, Wong LLL, Kim JS, Rojnuckarin P, Goh YT, Ueda Y, Chou WC, Lee JW, Kanakura Y, Chiou TJ. Monitoring and Treatment of Paroxysmal Nocturnal Hemoglobinuria in Patients with Aplastic Anemia in Asia: An Expert Consensus. Int J Mol Sci 2024; 25:12160. [PMID: 39596227 PMCID: PMC11594386 DOI: 10.3390/ijms252212160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) clones can be identified in a significant proportion of patients with aplastic anemia (AA). Screening for PNH clones at the time of an AA diagnosis is recommended by national and international guidelines. In this report, an expert panel of physicians discusses current best practices and provides recommendations for managing PNH in patients with AA in the Asia-Pacific region. Plasma/serum lactate dehydrogenase (LDH) levels and reticulocyte count should be measured with every blood test. PNH clone size should be monitored regularly by flow cytometry, with on-demand testing in the event of a rise in LDH level ± reticulocyte count or development of symptoms such as thrombosis. Monitoring for PNH clones can guide the choice of initial AA treatment, although flow cytometry has resource implications which may present a challenge in some Asia-Pacific countries. The treatment of patients with both PNH and AA depends on which condition predominates; following PNH treatment guidelines if hemolysis is the main symptom and AA treatment guidelines if bone marrow failure is severe (regardless of whether hemolysis is mild or moderate). The expert panel's recommendations on the monitoring and treatment of PNH in patients with AA are practical for healthcare systems in the Asia-Pacific region.
Collapse
Affiliation(s)
- Raymond Siu Ming Wong
- Sir Y.K. Pao Centre for Cancer & Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Sha Tin, Hong Kong SAR, China
| | - Jun Ho Jang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 2066 Seobu-Ro, Suwon 16419, Republic of Korea
| | - Lily Lee Lee Wong
- Queen Elizabeth Hospital, 13a, Jalan Penampang, Kota Kinabalu 88200, Sabah, Malaysia
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50-1 Yonsei-ro, Sinchon-dong, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ponlapat Rojnuckarin
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, 1873 Rama IV Rd, Pathum Wan, Bangkok 10330, Thailand;
- Research Unit in Translational Hematology, Chulalongkorn University, 254 Phaya Thai Rd, Khwaeng Wang Mai, Pathum Wan, Bangkok 10330, Thailand
| | - Yeow-Tee Goh
- Department of Haematology, Singapore General Hospital, Outram Rd, Singapore 169608, Singapore
| | - Yasutaka Ueda
- Department of Hematology and Oncology, Graduate School of Medicine, Faculty of Medicine, Osaka University, 1-1 Yamadaoka, Suita, Osaka 565-0871, Japan;
| | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No.7, Chung Shan S. Rd. (Zhongshan S. Rd), Zhongzheng Dist., Taipei City 100225, Taiwan
| | - Jong Wook Lee
- Division of Hematology-Oncology, Hanyang University Seoul Hospital, 222-1 Wangsimni-ro, Seong-dong-gu, Seoul 04763, Republic of Korea
| | - Yuzuru Kanakura
- Department of Hematology, Sumitomo Hospital, 5-chōme-3-20 Nakanoshima, Kita Ward, Osaka 530-0005, Japan
| | - Tzeon-Jye Chiou
- Cancer Center, Division of Hematology and Oncology, Department of Medicine, Taipei Municipal Wanfang Hospital, Taipei Medical University, No. 111, Section 3, Xinglong Rd, Wenshan District, Taipei City 11696, Taiwan;
| |
Collapse
|
4
|
Kokoris S, Polyviou A, Evangelidis P, Grouzi E, Valsami S, Tragiannidis K, Gialeraki A, Tsakiris DA, Gavriilaki E. Thrombosis in Paroxysmal Nocturnal Hemoglobinuria (PNH): From Pathogenesis to Treatment. Int J Mol Sci 2024; 25:12104. [PMID: 39596172 PMCID: PMC11594924 DOI: 10.3390/ijms252212104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/04/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Paroxysmal Nocturnal Hemoglobinuria (PNH) constitutes a rare bone marrow failure syndrome characterized by hemolytic anemia, thrombotic events (TEs), and bone marrow aplasia of variable degrees. Thrombosis is one of the major clinical manifestations of the disease, affecting up to 40% of individuals with PNH. Venous thrombosis is more prevalent, affecting mainly unusual sites, such as intrabdominal and hepatic veins. TEs might be the first clinical manifestation of PNH. Complement activation, endothelial dysfunction, hemolysis, impaired bioavailability of nitric oxide, and activation of platelets and neutrophils are implicated in the pathogenesis of TEs in PNH patients. Moreover, a vicious cycle involving the coagulation cascade, complement system, and inflammation cytokines, such as interleukin-6, is established. Complement inhibitors, such as eculizumab and ravulizumab (C5 inhibitors), have revolutionized the care of patients with PNH. C5 inhibitors should be initiated in patients with PNH and thrombosis, while they constitute a great prophylactic measure for TEs in those individuals. Anticoagulants, such as warfarin and low-molecular-weight heparin, and, in selected cases, direct oral anticoagulants (DOACs) should be used in combination with C5 inhibitors in patients who develop TEs. Novel complement inhibitors are considered an alternative treatment option, especially for those who develop extravascular or breakthrough hemolysis when terminal inhibitors are administered.
Collapse
Affiliation(s)
- Styliani Kokoris
- Laboratory of Hematology and Blood Bank Unit, “Attikon” University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.K.); (A.G.)
| | - Antri Polyviou
- Department of Hematology and Lymphoma, BMT Unit, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Paschalis Evangelidis
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (K.T.)
| | - Elisavet Grouzi
- Department of Transfusion Service and Clinical Hemostasis, “Saint Savvas” Oncology Hospital, 11522 Athens, Greece;
| | - Serena Valsami
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Konstantinos Tragiannidis
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (K.T.)
| | - Argyri Gialeraki
- Laboratory of Hematology and Blood Bank Unit, “Attikon” University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.K.); (A.G.)
| | - Dimitrios A. Tsakiris
- Department of Hemostasis and Thrombosis, University of Basel, 4001 Basel, Switzerland;
| | - Eleni Gavriilaki
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (K.T.)
| |
Collapse
|
5
|
Hillmen P, Horneff R, Yeh M, Kolev M, Deschatelets P. Navigating the Complement Pathway to Optimize PNH Treatment with Pegcetacoplan and Other Currently Approved Complement Inhibitors. Int J Mol Sci 2024; 25:9477. [PMID: 39273426 PMCID: PMC11395449 DOI: 10.3390/ijms25179477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare and potentially life-threatening hematologic disorder caused by a somatic mutation in a relevant portion of hematopoietic stem cells. Mutation of the phosphatidylinositol glycan biosynthesis class A (PIGA) gene prevents the expression of cell-surface proteins, including the complement regulatory proteins CD55 and CD59. With decreased or a lack of CD55 and CD59 expression on their membranes, PNH red blood cells become susceptible to complement-mediated hemolysis (symptoms of which include anemia, dysphagia, abdominal pain, and fatigue), leading to thrombosis. State-of-the-art PNH treatments act by inhibiting the dysregulated complement at distinct points in the activation pathway: late at the C5 level (C5 inhibitors, eculizumab, ravulizumab, and crovalimab), centrally at the C3 level (C3/C3b inhibitors and pegcetacoplan), and early at the initiation and amplification of the alternative pathway (factor B inhibitor, iptacopan; factor D inhibitor, danicopan). Through their differing mechanisms of action, these treatments elicit varying profiles of disease control and offer valuable insights into the molecular underpinnings of PNH. This narrative review provides an overview of the mechanisms of action of the six complement inhibitors currently approved for PNH, with a focus on the C3/C3b-targeted therapy, pegcetacoplan.
Collapse
Affiliation(s)
- Peter Hillmen
- Apellis Pharmaceuticals, Inc., Waltham, MA 02451, USA
| | | | - Michael Yeh
- Apellis Pharmaceuticals, Inc., Waltham, MA 02451, USA
| | - Martin Kolev
- Apellis Pharmaceuticals, Inc., Waltham, MA 02451, USA
| | | |
Collapse
|
6
|
Horneff R, Czech B, Yeh M, Surova E. Three Years On: The Role of Pegcetacoplan in Paroxysmal Nocturnal Hemoglobinuria (PNH) since Its Initial Approval. Int J Mol Sci 2024; 25:8698. [PMID: 39201383 PMCID: PMC11354333 DOI: 10.3390/ijms25168698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare disease characterized by complement-mediated hemolysis and potentially life-threatening complications. Pegcetacoplan, an inhibitor of complement components C3 and C3b, was approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) in 2021. A recent expansion to its indication by the EMA has made pegcetacoplan available for the treatment of both complement inhibitor-naïve and -experienced patients with PNH who have hemolytic anemia, a similarly broad patient population as in the US. This approval was based on results from the Phase 3 PEGASUS study, where pegcetacoplan showed superiority over the C5 inhibitor eculizumab with regard to improving the hemoglobin level in patients with anemia despite eculizumab treatment, and the Phase 3 PRINCE study, where pegcetacoplan showed superiority over supportive care with regard to hemoglobin stabilization and improving the lactate dehydrogenase level in complement inhibitor-naïve patients. In light of this recent indication expansion by the EMA, this article describes how the strong efficacy of pegcetacoplan is linked to its mechanism of action, which provides broad hemolysis control over both intravascular and extravascular hemolysis to improve a range of disease markers and enhance patients' quality of life. Furthermore, additional data and learnings obtained from over 3 years of experience with pegcetacoplan are summarized, including long-term efficacy and safety results, real-world clinical experiences, pharmacokinetic characteristics, and extensive practical guidance for the first-to-market proximal complement inhibitor for PNH.
Collapse
Affiliation(s)
| | - Barbara Czech
- Swedish Orphan Biovitrum AB, 171 65 Stockholm, Sweden
| | - Michael Yeh
- Apellis Pharmaceuticals, Inc., Waltham, MA 02451, USA
| | - Elena Surova
- Swedish Orphan Biovitrum AB, 171 65 Stockholm, Sweden
| |
Collapse
|
7
|
Meuleman MS, Duval A, Grunenwald A, Rezola Artero M, Dermani M, Peliconi J, Revel M, Vieira-Martins P, Courbebaisse M, Parfait B, Lebeaux D, Friedlander G, Roumenina L, Chauvet S, Frémeaux-Bacchi V, Dragon-Durey MA. Usefulness and analytical performances of complement multiplex assay for measuring complement biomarkers in plasma. Clin Chim Acta 2024; 554:117750. [PMID: 38176523 DOI: 10.1016/j.cca.2023.117750] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
INTRODUCTION The complement system is involved in numerous diseases, through diverse mechanisms and degree of activation. With the emergence of complement targeting therapeutic, simple and accessible tools to evaluate the extent of complement activation are strongly needed. METHODS We evaluated two multiplex panels, measuring complement activation fragments (C4a, C3a, C5a, Bb, Ba, sC5b9) and intact components or regulators (C1q, C2, C3, C4, C5, FD, FP, FH, FI). The specificity of each measurement was assessed by using complement proteins depleted sera and plasma collected from patients with complement deficiencies. Normal values distribution was estimated using 124 plasma samples from healthy donors and complement activation profile was assessed in plasma collected from 31 patients with various complement-mediated disorders. RESULTS We observed good inter-assay variation. All tested protein deficiencies were accurately detected. We established assay-specific reference values for each analyte. Except for C3, C4 and C4a, the majority of the measurements were in good agreement with references methods or published data. CONCLUSION Our study substantiates the utility of the Complement Multiplex assay as a tool for measuring complement activation and deficiencies. Quantifying complement cleavage fragments in patients exhibiting classical or alternative pathway activation allowed evaluating the activation state of the whole cascade.
Collapse
Affiliation(s)
- Marie-Sophie Meuleman
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France; Laboratory of Immunology, Georges Pompidou European Hospital, APHP, Paris, France
| | - Anna Duval
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France; Department of Nephrology, Strasbourg University Hospital, Strasbourg, France
| | - Anne Grunenwald
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France; Department of Nephrology, Poissy Intercommunal Hospital, Poissy, France
| | - Mikel Rezola Artero
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France
| | - Mohamed Dermani
- Laboratory of Immunology, Georges Pompidou European Hospital, APHP, Paris, France
| | - Julie Peliconi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France
| | - Margot Revel
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France
| | - Paula Vieira-Martins
- Laboratory of Immunology, Georges Pompidou European Hospital, APHP, Paris, France
| | - Marie Courbebaisse
- Paris Cité University, Physiology Department, European Georges-Pompidou Hospital, APHP, INSERM U1151, Paris, France
| | - Béatrice Parfait
- Centre de Ressources Biologiques - site Cochin, Fédération des CRB/PRB, DMU BioPhyGen, AP-HP.Centre-Université Paris Cité, Hôpital Cochin, Paris, France
| | - David Lebeaux
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, 75015 Paris, France; Service de Microbiologie, Unité Mobile d'Infectiologie, AP-HP, Hôpital Européen Georges Pompidou, 20 rue Leblanc, 75015 Paris, France
| | | | - Lubka Roumenina
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France
| | - Sophie Chauvet
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France; Paris Cité University, Paris, France; Department of Nephrology, Georges Pompidou European Hospital, APHP, Paris, France
| | - Véronique Frémeaux-Bacchi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France; Laboratory of Immunology, Georges Pompidou European Hospital, APHP, Paris, France
| | - Marie-Agnès Dragon-Durey
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer Team, Paris, France; Laboratory of Immunology, Georges Pompidou European Hospital, APHP, Paris, France; Paris Cité University, Paris, France.
| |
Collapse
|
8
|
Lee J, Lee H, Kim S, Suh HS. Efficacy of complement inhibitors for patients with paroxysmal nocturnal hemoglobinuria: a systematic review and meta-analysis. Ther Adv Hematol 2023; 14:20406207231216080. [PMID: 38105771 PMCID: PMC10725119 DOI: 10.1177/20406207231216080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/19/2023] [Indexed: 12/19/2023] Open
Abstract
Background Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired hematological disease. The development of complement inhibitors such as eculizumab, ravulizumab, and pegcetacoplan has revolutionized the management of PNH, leading to improvements in overall survival and quality of life for patients. Objectives This systematic review aims to provide comprehensive evidence of the efficacy of complement inhibitors in relation to treatment duration. Design This is a systematic review and meta-analysis. Data sources and methods A thorough literature search was conducted in MEDLINE, EMBASE, and the Cochrane Library up to 3 May 2022. We included all prospective interventional studies including single-arm trials. The primary outcomes of interest were lactate dehydrogenase (LDH) levels, hemoglobin (Hb) concentrations, transfusion avoidance, and Functional Assessment of Chronic Illness Therapy Fatigue (FACIT-F) scores. Results Our study included a total of 27 studies, comprising 5 randomized controlled trials and 11 single-arm trials, with a total of 912 patients with PNH. We stratified the studies according to treatment duration, based on the most frequently reported period of 26 weeks. Our analysis showed that treatment-naïve patients who received complement inhibitors had a pooled estimate of a decrease in LDH levels from baseline by -1462.0 U/L (95% CI: -1735.6 to -1188.5) for treatment ⩽26 weeks and -1696.5 U/L (95% CI: -2122.7 to -1270.2) for treatment >26 weeks. The mean Hb levels were increased by 1.4 g/dL (95% CI: 0.5-2.3) and 1.9 g/dL (95% CI: 0.7-3.1) in each group. Treatment with any complement inhibitor prevented the need for transfusion in at least 50% of patients with PNH in all treatment periods. Clinically meaningful improvements in FACIT-F were observed both before and after 26 weeks, with a pooled estimate of 6.8 (95% CI: 6.0-7.6) and 9.5 (95% CI: 7.0-12.0), respectively. Conclusion Our findings suggest that complement inhibitors can result in positive treatment outcomes and sustained benefits for patients with PNH.
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory Innovation Through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Haeseon Lee
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory Innovation Through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Siin Kim
- College of Pharmacy, Woosuk University, Wanju-gun, Republic of Korea
| | - Hae Sun Suh
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory Innovation Through Science, Kyung Hee University, Seoul, Republic of Korea
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| |
Collapse
|
9
|
Oliver M, Patriquin CJ. Paroxysmal Nocturnal Hemoglobinuria: Current Management, Unmet Needs, and Recommendations. J Blood Med 2023; 14:613-628. [PMID: 38084255 PMCID: PMC10710797 DOI: 10.2147/jbm.s431493] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/30/2023] [Indexed: 10/16/2024] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an ultra-rare, acquired clonal abnormality, which renders hematopoietic cells exquisitely sensitive to complement-mediated destruction. Classical features of PNH include intravascular hemolytic anemia, increased thrombotic risk, and manifestations related to end-organ damage (eg fatigue, chest pain, dyspnea, renal failure, and pulmonary hypertension). With supportive care alone, mortality rate of patients with PNH is approximately 35%. The anti-C5 monoclonal antibody, eculizumab, was the first targeted therapy approved for PNH, and led to improved hemoglobin, quality of life, reduced transfusion need, reduced thrombosis, and greater overall survival. More recently, therapeutics such as longer acting anti-C5 (ravulizumab) and anti-C3 (pegcetacoplan) medications have been approved, along with other novel therapeutics in late-stage clinical trials. Biosimilars of eculizumab are also now available. Proximal inhibitors (against C3, factor B, and factor D) have shown significant improvements in hemoglobin and transfusion-avoidance in patients who remain anemic despite C5 inhibition. Despite these novel therapies, some unmet challenges remain, including management of breakthrough hemolysis, clinically significant iatrogenic extravascular hemolysis, optimal management in pregnancy, and infection risk mitigation as new targets in the complement system are blocked. In addition, the use of self-administered subcutaneous and oral therapies raises concerns around treatment adherence and the risks of uncontrolled terminal complement. Given the ultra-rare nature of PNH, development is underway of a centralized international registry to capture and analyze the data as they mature for various new therapies and characterize the clinical challenges related to PNH management.
Collapse
Affiliation(s)
- Monika Oliver
- Department of Medicine, University of Alberta; Division of Hematology, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Christopher J Patriquin
- Department of Medicine (Hematology), University of Toronto, Division of Medical Oncology & Hematology, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|