Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal, complement-mediated hemolytic anemia associated with bone marrow failure and thrombophilia.1  Clonal expansion of a hematopoietic stem cell with a somatic mutation in PIGA underlies virtually all cases. The PIGA gene product is required to make glycosylphosphatidylinositol-anchored proteins (GPI-AP). CD55 and CD59 are complement regulatory proteins that are decreased or absent on PNH blood cells; their absence explains the chronic alternative pathway of complement-mediated hemolysis and acute paroxysms often triggered by classical complement-amplifying conditions such as infection or surgery. These paroxysms exacerbate hemoglobinuria (release of free hemoglobin) and smooth muscle dystonia due to tissue depletion of nitric oxide via free hemoglobin scavenging.2  Thrombophilia in PNH is multifactorial, but recent data suggest that the release of ADP, which is associated with intravascular hemolysis, leads to strong platelet activation.3 

Clinical manifestations of PNH are manifold, and the classical presentation characterized by intermittent bouts of hemoglobinuria is observed in less than 35% of patients.4  Some patients are asymptomatic and are diagnosed following the detection of mild anemia, neutropenia, and thrombocytopenia on routine blood counts. Bone marrow failure consistent with moderate or severe aplastic anemia is a common presentation. Other cases manifest with smooth muscle dystonias (e.g., erectile dysfunction, dysgeusia, esophageal spasm, or abdominal pain), and both microvascular and macrovascular forms of thrombosis are frequently observed.5,6  Thrombosis predominantly involves the abdominal (portal, hepatic, splenic, mesenteric) and cerebral veins; dermal vein thrombosis, deep venous thrombosis, and pulmonary emboli are less common.7  Arterial thrombosis is rare. Fatigue and dyspnea often out of proportion to the degree of anemia are common. Regardless of the presenting symptoms, most patients will have laboratory evidence of intravascular hemolysis (normocytic or macrocytic anemia with a lactate dehydrogenase [LDH] level 1.5 to 10 times the upper limit of normal, elevated indirect bilirubin, and low haptoglobin), mild to moderate bone marrow suppression, and fatigue.

The diagnostic evaluation for PNH should include a complete blood count (CBC), differential, reticulocyte count, biochemical profile, lactate dehydrogenase levels, d-dimer, and peripheral blood flow cytometry for PNH.8,9  If the CBC suggests underlying bone marrow failure, a bone marrow evaluation to rule out acquired aplastic anemia is indicated. Imaging of the abdominal veins and head is indicated if the patient presents with localized pain symptoms in these areas, especially when d-dimer levels are elevated. Hemolytic or classical PNH is diagnosed in patients with a large PNH granulocyte clone (usually >50%) based on flow cytometry, an LDH level greater than 1.5 times the upper limit of normal, a reticulocyte count greater than 120,000, and relatively well-preserved peripheral blood counts. In the setting of aplastic anemia, PNH is diagnosed based on the presence of a PNH clone (usually less than 20%) in conjunction with a hypocellular marrow, pancytopenia, and low corrected reticulocyte count. In such cases, the LDH level is often less than 1.5 times the upper limit of normal. Patients with subclinical PNH usually present with asymptomatic cytopenias, very mild hemolysis, and a PNH granulocyte clone of 0.1 to 10%. Although several classification schemes for PNH have been established, all are imperfect given the dynamic nature of the disease. In patients with PNH who have underlying aplastic anemia and those with subclinical PNH, the disease may evolve to hemolytic PNH over weeks to years, while hemolytic PNH can also evolve to aplastic anemia.

Observation is appropriate for patients with asymptomatic or minimally symptomatic PNH.10  Thrombosis is an urgent indication for complement inhibition. Patients with bothersome symptoms (fatigue, dyspnea, pain, etc.) due to a large PNH clone and evidence of intravascular hemolysis should also be treated. For patients with a PNH clone whose symptoms are primarily caused by underlying acquired severe aplastic anemia, the disease should be managed with immunosuppressive therapy or allogeneic bone marrow transplantation.11  Ravulizumab, a C5 inhibitor, is my firstline agent of choice for classical PNH due to its rapid onset of action, long half-life, good safety profile, and ability to mitigate classical pathway-amplifying triggers of complement.12  C5 inhibition is highly effective in preventing thrombosis.13,14  However, a drawback of all C5 inhibitors is that roughly 20% of patients continue to require intermittent red cell transfusions and have symptomatic anemia due to extravascular hemolysis.15  Upstream complement inhibitors that target the alternative complement pathway at C3 (pegcetacoplan),16  factor B (iptacopan),17  or factor D (danicopan)18  are now available. For patients with PNH who have symptomatic anemia despite C5 inhibition, both pegcetacoplan and iptacopan alone have been shown to be superior to remaining on C5 inhibition. Both drugs can restore hemoglobin to normal or near-normal levels without the need for transfusions in most cases. Pegcetacoplan is administered via subcutaneous infusion at least twice a week; iptacopan is administered orally twice a day. Danicopan is also an oral complement inhibitor that raises hemoglobin to near-normal levels in patients with PNH who exhibit persistent extravascular hemolysis and anemia despite C5 inhibition; however, it is approved as an “add-on” to C5 inhibitors. Danicopan is taken three times a day. All patients on complement inhibitors should remain current on Neisseria vaccinations, and individuals with high-risk disease should receive antibiotic prophylaxis.

Upstream complement inhibitors that inhibit factors C3, B, and D will increasingly be used to manage hemolytic PNH. At baseline, they may prove to be better than C5 inhibitors in reducing hemolysis. A caveat is that these agents have short half-lives and may leave patients vulnerable to severe hemolysis in the setting of classical pathway amplifiers (infection, surgery, pregnancy, etc.) and missed doses. More data on the use of these drugs in preventing thrombosis are needed.

Twenty years ago, PNH was a life-threatening, debilitating disease due to complement-dependent thrombosis and severe anemia. Fortunately, we now have a variety of options. Complement inhibition saves lives and improves quality of life. As the optimal sequence of these drugs continues to evolve, clinicians must consider the efficacy, safety, cost, and ease of administration.

Dr. Brodsky has received research funding from Alexion and AstraZeneca Rare Disease and is the section editor (red blood cell disorders) for UptoDate.

1
Hill
A
,
DeZern
AE
,
Kinoshita
T
, et al
.
Paroxysmal nocturnal haemoglobinuria
.
Nat Rev Dis Primers
.
2017
;
3
:
17028
. PubMed
2
Rother
RP
,
Bell
L
,
Hillmen
P
, et al
.
The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: A novel mechanism of human disease
.
JAMA
.
2005
;
293
(
13
):
1653
1662
.
3
Mannes
M
,
Pechtl
V
,
Hafner
S
, et al
.
Complemen and platelets: Prothrombotic cell activation requires membrane attack complex-induced release of danger signals
.
Blood Adv
.
2023
;
7
(
20
):
6367
6380
.
4
Schrezenmeier
H
,
Muus
P
,
Socié
G
, et al
.
Baseline characteristics and disease burden in patients in the international paroxysmal nocturnal hemoglobinuria registry
.
Haematologica
.
2014
;
99
(
5
):
922
929
.
5
Hillmen
P
,
Lewis
SM
,
Bessler
M
, et al
.
Natural history of paroxysmal nocturnal hemoglobinuria
.
N Engl J Med
.
1995
;
333
(
19
):
1253
1259
.
6
Moyo
VM
,
Mukhina
GL
,
Garrett
ES
, et al
.
Natural history of paroxysmal nocturnal haemoglobinuria using modern diagnostic assays
.
Br J Haematol
.
2004
;
126
(
1
):
133
138
.
7
Hill
A
,
Kelly
RJ
,
Hillmen
P
.
Thrombosis in paroxysmal nocturnal hemoglobinuria
.
Blood
.
2013
;
121
(
25
):
4985
4996
.
8
Brodsky
RA
,
Mukhina
GL
,
Li
S
, et al
.
Improved detection and characterization of paroxysmal nocturnal hemoglobinuria using fluorescent aerolysin
.
Am J Clin Pathol
.
2000
;
114
(
3
):
459
466
.
9
Borowitz
MJ
,
Craig
FE
,
DiGiuseppe
JA
, et al
.
Guidelines for the diagnosis and monitoring of paroxysmal nocturnal hemoglobinuria and related disorders by flow cytometry
.
Cytometry B Clin Cytom
.
2010
;
78
(
4
):
211
230
.
10
Brodsky
RA
.
How I treat paroxysmal nocturnal hemoglobinuria
.
Blood
.
2009
;
113
(
26
):
6522
6527
.
11
DeZern
AE
,
Jones
RJ
,
Brodsky
RA
.
Eculizumab bridging before bone marrow transplant for marrow failure disorders is safe and does not limit engraftment
.
Biol Blood Marrow Transplant
.
2018
;
24
(
12
):
e26
-
30
.
12
Kulasekararaj
AG
,
Hill
A
,
Langemeijer
S
, et al
.
One-yea outcomes from a phase 3 randomized trial of ravulizumab in adults with paroxysmal nocturnal hemoglobinuria who received prior eculizumab
.
Eur J Haematol
.
2021
;
106
(
3
):
389
397
.
13
Hillmen
P
,
Muus
P
,
Dührsen
U
, et al
.
Effect of the complement inhibitor eculizumab on thromboembolism in patients with paroxysmal nocturnal hemoglobinuria
.
Blood
.
2007
;
110
(
12
):
4123
4128
.
14
Gerber
GF
,
DeZern
AE
,
Chaturvedi
S
, et al
.
A 15-year single institution experience of anticoagulation management in paroxysmal nocturnal hemoglobinuria patients on terminal complement inhibition with history of thromboembolism
.
Am J Hematol
.
2022
;
97
(
2
):
E59
E62
.
15
Brodsky
RA
,
Peffault de Latour
R
,
Rottinghaus
ST
, et al
.
Characterizatio of breakthrough hemolysis events observed in the phase 3 randomized studies of ravulizumab versus eculizumab in adults with paroxysmal nocturnal hemoglobinuria
.
Haematologica
.
2020
;
106
(
1
):
230
.
16
Hillmen
P
,
Szer
J
,
Weitz
I
, et al
.
Pegcetacopla versus eculizumab in paroxysmal nocturnal hemoglobinuria
.
N Engl J Med
.
2021
;
384
(
11
):
1028
1037
.
17
Risitano
AM
,
Roth
A
,
Soret
J
, et al
.
Addition of iptacopan, an oral factor B inhibitor, to eculizumab in patients with paroxysmal nocturnal haemoglobinuria and active haemolysis: An open-label, single-arm, phase 2, proof-of-concept trial
.
Lancet Haematol
.
2021
;
8
(
5
):
e344
-
e354
.
18
Kulesekararaj
A
,
Risitano
AM
,
Maciejewski
JP
, et al
.
Phas 2 study of danicopan in paroxysmal nocturnal hemoglobinuria patients with an inadequate response to eculizumab
.
Blood
.
2021
;
138
(
20
):
1928
1938
.