Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link

Disclaimer: Early release articles are not considered as final versions. Any changes will be reflected in the online version in the month the article is officially released.

Volume 31, Number 3—March 2025
Dispatch

Meningococcal Sepsis in Patient with Paroxysmal Nocturnal Hemoglobinuria during Pegcetacoplan Therapy

Author affiliation: Author affiliations: University of Helsinki, Helsinki, Finland (L. Starck, S. Meri); Helsinki University Central Hospital, Helsinki (V. Nummi, E. Poikonen, A.-E. Lehtinen, P. Kivelä, N. Friberg, J. Torvikoski, S. Meri); Finnish Institute for Health and Welfare, Helsinki (M. Toropainen)

Suggested citation for this article

Abstract

Complement C5 inhibitors bring an increased risk for Neisseria infections. A novel complement C3 inhibitor, pegcetacoplan, was recently approved to treat paroxysmal nocturnal hemoglobinuria, a condition commonly treated with complement C5 inhibitors. We present a case of meningococcal sepsis in a pegcetacoplan-treated patient with aplastic anemia and paroxysmal nocturnal hemoglobinuria.

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired disorder caused by a de novo mutation in the PIG-A gene. This mutation occurs in a hematopoietic stem cell and causes a clonal loss of the glycosylphosphatidylinositol anchor from the surfaces of affected blood cells (1). Complement decay-accelerating factor (2) and protectin (3) are bound to cell membranes by the glycosylphosphatidylinositol anchor. Thus, glycosylphosphatidylinositol-deficient blood cells are vulnerable to complement attack, leading to hemolytic anemia, thrombi, and other PNH-related complications.

Complement inhibition is a logical therapy for PNH. In 2007, the US Food and Drug Administration and the European Medicines Agency licensed an anti-C5 monoclonal antibody, eculizumab, and in 2018–2019, the longer-acting ravulizumab. Deficiencies of the terminal complement pathway increase the risk for Neisseria infections (4). C5 inhibitor decreases the ability of human blood to kill serogroup B meningococci despite vaccination (5); investigators have described sepsis caused by nonencapsulated N. meningitidis in a patient with PNH during ravulizumab therapy (6).

In 2021, pegcetacoplan was the first C3 inhibitor authorized by the US Food and Drug Administration and the European Medicines Agency. Phase III clinical trial data and postmarketing safety surveillance studies revealed no invasive N. meningitidis infections in patients (79). We describe invasive N. meningitidis infection in a patient with PNH during pegcetacoplan therapy.

Case Report

A 16-year-old girl received a diagnosis of aplastic anemia and PNH. The next year, her granulocyte PNH clone size increased from 1% to 98%. To address chronic, severe hemolysis, physicians prescribed ravulizumab therapy in April 2023. The patient received a dose of meningococcal conjugate vaccine (covering serogroups A, C, Y, and W-135and a dose of meningococcal group B vaccine (4CMenB) in April 2023; she received a dose of each again in June 2023, ≈8 months before meningococcal sepsis onset. In December 2023, physicians switched the patient’s therapy from ravulizumab to pegcetacoplan (1,080 mg 2×/wk) because of extravascular hemolysis; the last ravulizumab infusion was 78 days before the onset of her infection, corresponding to 1.5 times the half-life of ravulizumab. The patient’s medications for aplastic anemia included eltrombopag (125 mg 1×/d), cyclosporine (100 mg 2×/d), and folic acid (5 mg 1×/d). She also used lynestrenol. At a follow-up appointment in January 2024, the patient’s test results showed leukopenia: total leukocyte count of 1.8 × 109 cells/L (reference range 3.4–8.2 × 109 cells/L) and neutrophils at 0.60 × 109 cells/L (reference range 1.5–6.7 × 109 cells/L).

In February 2024, three days after a student celebration on an international cruise ship, the patient, then 18 years of age, visited the emergency department with signs of septic infection: occipital headache, fever 39.5°C, tachycardia 110 beat/min, and blood pressure 105/67 mm Hg. Physical examination revealed no petechiae; thorax radiograph and urinalysis results were unremarkable. We performed blood culture and started the patient on antimicrobial therapy (cefuroxime 1.5 g 3×/d). Tests showed a total leukocyte count of 2.5 × 109 cells/L and neutrophils at 1.90 × 109 cells/L. The patient’s hemoglobin level was 86 g/L (reference 117–155 g/L), and lactate dehydrogenase (LDH) was 398 U/L (reference 115–235 U/L) (Tables 1, 2).

During the first inpatient morning, the patient’s temperature peaked at 40.1°C, but she remained hemodynamically stable and in good consciousness. Her LDH increased to 463 U/L and hemoglobin decreased to 73 g/L. A transfusion of 1 unit of packed red blood cells was administered. Pegcetacoplan was continued at the same dose. Thromboprophylaxis with enoxaparin was initiated. Approximately 24 hours after admission, the patient’s blood culture sample became positive, with an unclear finding in gram-staining. A commercial multiplex-PCR array (Biofire FilmArray Blood Culture Identification Panel 2; bioMérieux, https://www.biomerieux.com) yielded no identification. Dosing of cefuroxime was increased to 4 times a day.

On the second day, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (VITEK MS; bioMérieux) identified N. meningitidis, but capsule agglutination was inconclusive. An in-house meningococcus-specific PCR remained negative. The multiplex PCR repeatedly returned a negative result for all targets. Cefuroxime was switched to ceftriaxone (2 g t2×/d), a medication more appropriate for treating meningococcal sepsis. Subsequent laboratory tests revealed a decrease in hemoglobin to 72 g/L and an increase in LDH to 726 U/L, indicating active hemolysis. She received another infusion of packed red blood cells.

Epidemiologic contact tracing and distribution of antimicrobial prophylaxis to close contacts commenced. The meningococcal isolate was sent to the Finnish Institute for Health and Welfare for verification, serogrouping, and characterization by whole-genome sequencing that included genes of the 4CMenB vaccine antigens. The isolate auto-agglutinated and was not groupable using a latex agglutination test (BioRad Laboratories, https://www.bio-rad.com).

On the third day of her hospital stay, the patient remained clinically stable, and her LDH level decreased to 619 U/L. On the fourth day, a slight drooping of the left side of her mouth was noticed. Magnetic resonance imaging of her brain revealed no abnormalities. The patient recovered fully and was discharged in good health after a 7-day course of intravenous antimicrobial drugs. She continued pegcetacoplan therapy for PNH. At discharge, the patient was prescribed amoxicillin/clavulanate (875/125 mg) to be started in duplicate in case of a sudden onset of fever at distance from hospital. On a follow-up visit for PNH, the patient gave informed consent for publication of a case report.

Whole-genome sequencing revealed a nonencapsulated N. meningitidis, harboring the capsule null locus (cnl) allele 2. The strain designation of the isolate was sequence type 198, clonal complex 198 (cnl: P1.18,25–1: F5–5: ST198 (cc198) (capsule group:PorA type:FetA type:sequence type [clonal complex]). The isolate contained neisserial heparin binding antigen peptide 10 and factor H binding protein peptide 94 but lacked neisserial adhesin A. The genetic Meningococcal Antigen Typing System (10) predicts isolate coverage by 4CMenB because of cross-reactive neisserial heparin binding antigen. Blocking of complement C3 probably hinders killing by both opsonophagocytosis and the complement terminal pathway (11), although there is also evidence for serum killing during C3 blockade through a C3 bypass pathway (12). The negative diagnostic PCR and latex agglutination test results were likely due to lack of capsular transport and biosynthesis genes, including the PCR-targeted ctrA (13).

Infections caused by nonencapsulated N. meningitidis are rare. The epidemiology of cnl invasive meningococci is difficult to ascertain. The 2022 European Centre for Disease Prevention and Control Surveillance Report for Invasive Meningococcal Disease (14) listed serotypes A, X, Z, 29E, nongroupable, and others in the same category (n = 63 in Europe in 2022). The US Centers for Disease Control and Prevention categorized cnl meningococci as nongroupables (n = 30) and other/unknowns (n = 40) in 2022. The PubMLST database (https://pubmlst.org/neisseria), however, received >100 reports of invasive infections by cnl meningococci, most belonging to the same strain designation as the isolate infecting our patient. As whole-genome sequencing becomes more common, allowing a more detailed characterization than the agglutination test, cnl and other nongroupable meningococci will likely become more frequently recognized and differentiated.

Conclusions

Our patient had aplastic anemia and associated PNH, with relative leukopenia and complement inhibition, increasing her susceptibility to meningococcal infections. Iatrogenic complement inhibition with pegcetacoplan and potential residual ravulizumab predisposed this patient to an unusual septic infection by nonencapsulated N. meningitidis, despite immunization with 4CMenB 10 months and 8 months earlier. We faced some clinical challenges in diagnosing and treating this patient. First, several meningococcal identification tests returned negative results. Second, because complement C3 inhibitor therapy hampers protection against meningococci conferred by 4CMenB immunization, patients who suffer severe infections must sometimes compromise continuity of complement inhibitor treatment. Finally, with evidence lacking about efficacy and safety of antimicrobial prophylaxis in complement inhibitor recipients (15), physicians lack guidance regarding appropriate prophylactic antimicrobial drugs after severe infections in patients with PNH. Our case sheds light on the obstacles involved in both diagnosing and treating neisserial infections in complement inhibitor recipients and demonstrates the importance of vigilance and rapid administration of effective antimicrobial drugs.

Dr. Starck is a researcher at the Department of Bacteriology and Immunology, University of Helsinki, Finland, whose primary interests are complement and bacterial research.

Top

Acknowledgment

This work was conducted as part of routine employment. S.M. is supported by the Sigrid Jusélius Foundation and the Helsinki University Hospital Funds (TYH2019311 and TYH202322).

Top

References

  1. 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;73:70311. DOIPubMedGoogle Scholar
  2. Nicholson-Weller  A, March  JP, Rosenfeld  SI, Austen  KF. Affected erythrocytes of patients with paroxysmal nocturnal hemoglobinuria are deficient in the complement regulatory protein, decay accelerating factor. Proc Natl Acad Sci U S A. 1983;80:506670. DOIPubMedGoogle Scholar
  3. Yamashina  M, Ueda  E, Kinoshita  T, Takami  T, Ojima  A, Ono  H, et al. Inherited complete deficiency of 20-kilodalton homologous restriction factor (CD59) as a cause of paroxysmal nocturnal hemoglobinuria. N Engl J Med. 1990;323:11849. DOIPubMedGoogle Scholar
  4. Fijen  CAP, Kuijper  EJ, Hannema  AJ, Sjöholm  AG, van Putten  JPM. Complement deficiencies in patients over ten years old with meningococcal disease due to uncommon serogroups. Lancet. 1989;2:5858. DOIPubMedGoogle Scholar
  5. Langereis  JD, van den Broek  B, Franssen  S, Joosten  I, Blijlevens  NMA, de Jonge  MI, et al. Eculizumab impairs Neisseria meningitidis serogroup B killing in whole blood despite 4CMenB vaccination of PNH patients. Blood Adv. 2020;4:361520. DOIPubMedGoogle Scholar
  6. Galli  N, Pettine  L, Panigada  M, Daprai  L, Suriano  G, Grancini  A, et al. Non-capsulated Neisseria meningitidis sepsis in a paroxysmal nocturnal hemoglobinuria patient treated with ravulizumab: case report and review of the literature. Front Immunol. 2023;14:1269325. DOIPubMedGoogle Scholar
  7. Hillmen  P, Szer  J, Weitz  I, Röth  A, Höchsmann  B, Panse  J, et al. Pegcetacoplan versus eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2021;384:102837. DOIPubMedGoogle Scholar
  8. de Castro  CM, Dingli  D, Al-Adhami  M, Mulherin  B, Yeh  M, Lallier  S, et al. A real-world evidence review of post-marketing safety and compliance with pegcetacoplan in patients with paroxysmal nocturnal hemoglobinuria (PNH) in the US. Blood. 2023;142(Supp 1):5647. DOIGoogle Scholar
  9. Patriquin  CJ, Bogdanovic  A, Griffin  M, Kelly  RJ, Maciejewski  JP, Mulherin  B, et al. Safety and efficacy of pegcetacoplan in adult patients with paroxysmal nocturnal hemoglobinuria over 48 weeks: 307 open-label extension study. Adv Ther. 2024;41:205069. DOIPubMedGoogle Scholar
  10. Muzzi  A, Brozzi  A, Serino  L, Bodini  M, Abad  R, Caugant  D, et al. Genetic Meningococcal Antigen Typing System (gMATS): A genotyping tool that predicts 4CMenB strain coverage worldwide. Vaccine. 2019;37:9911000. DOIPubMedGoogle Scholar
  11. Konar  M, Granoff  DM. Eculizumab treatment and impaired opsonophagocytic killing of meningococci by whole blood from immunized adults. Blood. 2017;130:8919. DOIPubMedGoogle Scholar
  12. Ispasanie  E, Muri  L, Schmid  M, Schubart  A, Thorburn  C, Zamurovic  N, et al. In vaccinated individuals serum bactericidal activity against B meningococci is abrogated by C5 inhibition but not by inhibition of the alternative complement pathway. Front Immunol. 2023;14:1180833. DOIPubMedGoogle Scholar
  13. Sirluck-Schroeder  I, Al-Rawahi  GN, Gadkar  V, Hoang  L, Tsang  R, Tilley  P. Limitation of ctrA as a target for Neisseria meningitidis identification and potential alternative targets. J Clin Microbiol. 2022;60:e0015222. DOIPubMedGoogle Scholar
  14. European Centre for Disease Prevention and Control. Invasive meningococcal disease—annual epidemiological report for 2022. Stockholm: The Centre; 2024 [cited 2024 Sep 5]. https://www.ecdc.europa.eu/en/publications-data/invasive-meningococcal-disease-annual-epidemiological-report-2022
  15. Crew  PE, McNamara  L, Waldron  PE, McCulley  L, Christopher Jones  S, Bersoff-Matcha  SJ. Antibiotic prophylaxis in vaccinated eculizumab recipients who developed meningococcal disease. J Infect. 2020;80:35071. DOIPubMedGoogle Scholar

Top

Tables

Top

Suggested citation for this article: Starck L, Nummi V, Poikonen E, Lehtinen A-E, Kivelä P, Friberg N, et al. Meningococcal sepsis in patient with paroxysmal nocturnal hemoglobinuria during pegcetacoplan therapy. Emerg Infect Dis. 2025 Mar [date cited]. https://doi.org/10.3201/eid3103.241182

DOI: 10.3201/eid3103.241182

Original Publication Date: February 21, 2025

Table of Contents – Volume 31, Number 3—March 2025

EID Search Options
presentation_01 Advanced Article Search – Search articles by author and/or keyword.
presentation_01 Articles by Country Search – Search articles by the topic country.
presentation_01 Article Type Search – Search articles by article type and issue.

Top

Comments

Please use the form below to submit correspondence to the authors or contact them at the following address:

Seppo Meri, Haartman-Institute, Haartmaninkatu 3 (PL 21), 00014 Helsinki, Finland

Send To

10000 character(s) remaining.

Top

Page created: January 22, 2025
Page updated: February 21, 2025
Page reviewed: February 21, 2025
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
file_external