You are on page 1of 19

Seminar

Haemolytic uraemic syndrome


Mini Michael, Arvind Bagga, Sarah E Sartain, Richard J H Smith

Lancet 2022; 400: 1722–40 Haemolytic uraemic syndrome (HUS) is a heterogeneous group of diseases that result in a common pathology, thrombotic
Published Online microangiopathy, which is classically characterised by the triad of non-immune microangiopathic haemolytic anaemia,
October 19, 2022 thrombocytopenia, and acute kidney injury. In this Seminar, different causes of HUS are discussed, the most common
https://doi.org/10.1016/
being Shiga toxin-producing Escherichia coli HUS. Identifying the underlying thrombotic microangiopathy trigger can be
S0140-6736(22)01202-8
challenging but is imperative if patients are to receive personalised disease-specific treatment. The quintessential example
Division of Pediatric
Nephrology, Baylor College of
is complement-mediated HUS, which once carried an extremely high mortality but is now treated with anti-complement
Medicine, Texas Children’s therapies with excellent long-term outcomes. Unfortunately, the high cost of anti-complement therapies all but precludes
Hospital, Houston, TX, USA their use in low-income countries. For many other forms of HUS, targeted therapies are yet to be identified.
(M Michael MD); Department of
Pediatrics, All India Institute of
Medical Sciences, Introduction purpura from HUS through the use of PLASMIC or
New Delhi, India Haemolytic uraemic syndrome (HUS), comprising non- French scores is recommended by the International
(Prof A Bagga MD); immune microangiopathic haemolytic anaemia, throm­bo­ Society on Thrombosis and Haemostasis.4 Since
Pediatrics-Hematology/
cytopenia, and acute kidney injury, is associated with thrombotic thrombocytopenic purpura is a distinct disease
Oncology, Baylor College of
Medicine, Houston, TX, USA substantial acute and chronic morbidity in children and entity, its further discussion is beyond the scope of this
(S E Sartain MD); Department of adults. The unifying feature of this heterogeneous group Seminar. This Seminar focuses on the epidemiology,
Otolaryngology, Pediatrics and of diseases is endothelial damage, which leads to the pathophysiology, genetics, and clinical manifestations of
Molecular Physiology &
formation of microthrombi in vascular beds in multiple various types of HUS, and discusses approaches to their
Biophysics, The University of
Iowa, Iowa City, IA, USA organs, thrombocytopenia due to platelet consumption, diagnosis and treatment.
(Prof R J H Smith MD) and haemolysis caused by intravascular mechanical
Correspondence to: disruption of erythrocytes traversing the microthrombotic Classification and pathology
Dr Mini Michael, Division of fibrin network.1–3 Treatment for HUS is varied and depends The classification of HUS is challenging and constantly
Pediatric Nephrology,
on the underlying cause. Its early recognition is important, evolving. An aetiology-based classification is often used
Baylor College of Medicine,
Texas Children’s Hospital, both for comprehensive evaluation of the cause, and to (figure 1). Infection-associated HUS includes that
Houston, TX 77030, USA allow timely intervention with specific, effective treatment second­ ary to Shiga toxin-producing Escherichia coli
mmichael@bcm.edu when applicable. Age at presentation, clinical symptoms, (STEC), Streptococcus pneumoniae, and H1N1/influenza
and family history are important in the initial evaluation. A. Atypical HUS (aHUS) is broadly used for all other
Although thrombotic thrombocytopenic purpura can types, which can be primary or secondary. Primary aHUS
present with similar features, making it difficult to typically denotes the presence of an underlying
distinguish from HUS, the underlying cause differs. complement system defect (such as a mutation in a
Thrombotic thrombocytopenic purpura develops complement gene or the presence of factor H [FH]
secondary to acquired or congenital deficiency of von autoantibodies), as well as other causes such as
Willebrand protease, ADAMTS13 (a disintegrin and metabolism-associated HUS (cobalamin C disease), and
metalloproteinase with thrombospondin type 1 motifs, DGKE and WT1-associated HUS. The term secondary
member 13). Distin­ction of thrombotic thrombocytopenic HUS implies another underlying cause for disease, such
as infection, malignant hypertension, or complement-
amplifying conditions such as systemic lupus
Search strategy and selection criteria erythematosus, scleroderma, and anti­ phospholipid
We searched the Cochrane Library, MEDLINE, and Embase syndrome. Secondary HUS can also be associated with
between Jan 1, 2010, and April 30, 2021. We used the search co-existing conditions such as pregnancy, the use of
terms “haemolytic uremic syndrome”, “thrombotic particular drugs such as calcineurin inhibitors, oral
microangiopathy”, “shigatoxin”, “pneumococcus”, “factor H contraceptives, and quinine, malignancy, and stem-cell
autoantibody”, “drug induced TMA”, “transplant”, and solid organ transplantation. This primary versus
“WT1 mutation”, “DEGKE mutation”, “eculizumab”, secondary binary classification belies the fact that, even
and “ravulizumab” in combination with the terms in the presence of a complement abnormality, an
“pathophysiology”, “causes” and “treatment”. We largely initiating trigger (eg, infection) is the rule rather than the
selected publications from within the past 5 years but did not exception, and underscores the challenge associated with
exclude commonly referenced and highly regarded older determining the underlying cause of disease, which is
publications. We also searched the reference lists of articles clinically relevant because prompt treatment with a
identified by this search strategy and selected those we terminal complement inhibitor improves renal outcomes
judged relevant. Review articles and book chapters are cited of complement-driven aHUS.5–7 Because the response is
to provide readers with more details and more references dramatic and potentially life-saving, anti-comple­ment
than this Seminar has space for. therapy is often started immediately, and discon­tinued if
a non-complement cause for disease is later identified.

1722 www.thelancet.com Vol 400 November 12, 2022


Seminar

Pathological findings in HUS (appendix p 20) reflect Pathogenesis See Online for appendix
tissue responses to endothelial injury, including STEC produce two main types of Shiga toxins (Stx) known
endothelial swelling, subendothelial widening, and as Stx1 and Stx2, encoded by the stx1 and stx2 genes. Stx is
presence of luminal fibrin with entrapped fragmented composed of a single A subunit (30 kDa) and five B subunits
red cells; substantial luminal occlusion in glomerular (each 7 kDa); the former is biologically active and the latter
capillaries and arterioles can occur (figure 1). Ultra­ binds to the cell surface receptor. The majority of STEC
structural evaluation shows glomerular endothelial O157 harbour stx2. 9,18 Stx2 is regarded as more virulent
swelling, fenestral fusion, subendothelial expansion with than Stx1, with higher risk of causing HUS. The
electron-lucent material, and later reduplication of the mechanism of cytotoxicity and complement activation by
glomerular basement membrane (figure 2). Stx is shown in the appendix (pp 18–20). Stx produced by
bacterial infection of the large intestine binds the
Infection-associated HUS enterocyte, and crosses the brush border to enter the
Shiga toxin-associated HUS circulation. The pentameric B subunit of the toxin binds to
Incidence and epidemiology Gb3 and TLR4 expressed on circulating blood cells, causing
The most common infectious agent that causes HUS, activation of neutrophils, monocytes, platelets, and
especially in children younger than 5 years of age, is erythrocytes, inducing budding of microvesicles containing
STEC. Rarely, other Shiga toxin-producing bacteria the toxin within and on its surface. Micro­ vesicles are
including Shigella dysenteriae, Shigella flexneri, endowed with activated complement factors C3b and C9
Shigella sonnei and Citrobacter spp are isolated. STEC is a and tissue factor. The key pathogenic mechanism in STEC-
water-borne and food-borne pathogen that causes HUS is targeting by Stx or Stx associated with microvesicles
diarrhoea and haemorrhagic colitis.8 Diarrhoea or to Gb3-expressing endothelial cells, followed by docking
dysentery due to Shiga toxin-producing enteropathogens and release of toxin by microvesicles. The A1 subunit of the
is followed by HUS in 5–15% of cases, a proportion that toxin causes depurination of adenosine in a highly
can reach 20–30% during outbreaks. The predominant conserved region of the 28s ribosomal subunit of 60s
STEC serotype causing HUS is O157:H7, responsible for ribosomal RNA that alters its binding to tRNA, interrupting
around 2000 such cases annually worldwide.9,10 The most the synthesis of proteins and leading to cell death.19,20 Other
common non-O157 serogroups in Europe include mechanisms of cytotoxicity include increased synthesis of
O145, O146, O91, O103, and O26, and in the USA, chemokines (via the SDF-1–CXCR4–CXCR7 pathway) and
O121, O45, O26, O111, O103, and O145.11 The non-O157 adhesion molecules. Stx induces surface mobilisation of
organisms are a heterogeneous group with variable P-selectin that favours C3 activation and the assembly of
HUS risk (around 1%) and are more commonly
encountered in patients with diarrhoea or dysentery. 12
STEC-HUS is mostly sporadic, the incidence being Infection-associated HUS Complement-mediated aHUS
Shiga toxin-producing Hereditary: mutations of CFH,
highest in young children.13 The reason for its lower Escherichia coli HUS CFI, C3, CFB, MCP and THBD
incidence in adults might be due to the acquired Streptococcus pneumoniae Acquired: factor H
H1N1/influenza A autoantibody-associated HUS
presence of anti-Shiga toxin (Stx) antibodies or to higher
glomerular expression of the globotriaosylceramide Non-complement-
3 (Gb3) Stx receptor in children. STEC colonise cattle mediated aHUS
DGKE
and transmission is faecal-to-oral via undercooked Pregnancy-induced aHUS
WT1
contaminated beef, raw milk products, drinking water, G6PD
direct contact with animals, and person-
to-person spread. Contamination of leafy vegetables is Metabolism-associated
an important cause of STEC infection. 14 The HUS
Drug-induced aHUS
Cobalamin C disease
2011 German epidemic, caused by the O104:H4 serotype, Methionine synthase
was unique for several reasons: an enteroaggregative deficiency
E coli that acquired a prophage-encoding Stx2 was
responsible, mostly adults were affected, the typical Coagulation-mediated aHUS
Transplant-associated HUS Thrombomodulin
history of dysentery was absent, and HUS occurred Haematopoietic stem cell transplantation
in 20–22% of patients with frequent neurological thrombotic microangiopathy Secondary aHUS
Solid organ transplantation thrombotic Malignant hypertension
complications and high mortality (54 deaths in nearly microangiopathy Complement-amplifying conditions
4000 people infected).15 The STEC O80:H2 serotype that Antiphospholipid antibody
emerged in France and Switzerland was associated with Malignancy or cancer
severe HUS,16 while the recurrent outbreaks of
diarrhoeal infection in England in 2014–18 were
Figure 1: Aetiology-based classification of HUS
associated with STEC O55:H7, with early age at onset The centre circle shows light microscopy of renal biopsy findings of thrombotic microangiopathy: glomerular fresh
(median 4 years [range 6 months–69 years]) and HUS in fibrin thrombus (green arrow) with severe red blood cell congestion (haematoxylin and eosin stain
54% of patients. 17 20 × magnification). HUS=haemolytic uraemic syndrome. aHUS=atypical haemolytic uraemic syndrome.

www.thelancet.com Vol 400 November 12, 2022 1723


Seminar

ulcers on colonoscopy. Severe colonic involvement can


Neurological result in complications such as toxic megacolon and
Seizures, nystagmus, diplopia, hemiparesis,
headache, altered consciousness,
transmural necrosis with gastrointestinal perforation or
hallucinations, encephalopathy, and coma right-sided colonic strictures. After a median of 7 days,
when diarrhoea is resolving, around 5–15% of patients
develop HUS. The risk factors for developing HUS include
age younger than 5 years, high white blood cell count,
Ocular Gastrointestinal specific strains of STEC (O157:H7, O126, and O104:H4),
Loss of visual acuity, visual Diarrhoea, vomiting, and infection with organisms producing Stx2.27 Features of
scotomas, ocular pain, pancreatitis, cholelithiasis,
diplopia, and blurred transaminitis, hepatitis, HUS include fatigue, pallor, and oligoanuria, with or
vision and gastrointestinal without fluid overload, hypertension, and oedema. Most
bleeding
patients have mild disease, managed by optimised fluid
balance, nutrition support, and control of hypertension.
Dialysis is required in 30–60% of patients with severe
illness and oligoanuria. A proportion of patients (about
Cardiac 70%) require transfusion of red cells. Case series suggest
Pulmonary Left ventricular hypertrophy, that almost all adult patients have loose stools, and bloody
Pulmonary oedema, pulmonary hypertrophic cardiomyopathy,
haemorrhage, and pulmonary dilated cardiomyopathy, elevated diarrhoea is relatively less frequent than in children.28,29
embolism creatine kinase blood level, valve Cerebral involvement characterised by irritability,
insufficiency and intracardiac
thrombus
altered consciousness, and seizures, is seen in 10–25% of
cases.30,31 Other characteristics include pyramidal and
Figure 2: Extra-renal manifestations of atypical haemolytic uraemic syndrome extrapyramidal features, diplopia, dysphasia, and facial
The centre circle shows electron microscopy of renal biopsy findings of thrombotic microangiopathy: endothelial nerve palsy. Severe neurological involvement, mainly in
swelling (green star), fenestral fusion, and subendothelial expansion with reduplication of the glomerular the form of persistent encephalopathy, brain haemorrhage
basement membrane (green arrow) are considered evidence of endothelial injury (uranyl acetate and lead citrate, or infarction, and anoxic brain injury, is associated with
830 × magnification).
worse disease severity and higher mortality.32,33 The
neurological outcomes are satisfactory, with most patients
alternative pathway C3 convertase (C3bBb) on the cell showing complete recovery.31 Neurological findings are
surface. The anaphylatoxin C3a interacts with its more common in adults than in children.29
endothelial receptor (C3aR), potentiating P-selectin Pancreatic involvement with elevated blood levels of
expression and throm­bomodulin shedding with thrombus amylase and lipase may be seen in up to two-thirds of
formation. Shiga­toxin also binds ultra-large multimers of patients with STEC-HUS during the acute phase. Throm­
von Willebrand factor and reduces their cleavage by botic microangiopathy (TMA) might involve both the
ADAMTS13 which leads to further thrombosis. Elevated endocrine and exocrine pancreas; pancreatic inflammation
free haemoglobin concentrations during haemolysis with TMA and pancreatic necrosis has rarely been reported.34
further mediate endothelial injury by promoting oxidative A meta-analysis, including 1139 patients from 13 studies,
stress and a pro-thrombotic state. There is growing showed that the pooled incidence of diabetes during the
evidence of alternative complement pathway activation in acute stage of HUS was 3·2% (95% CI 1·3–5·1).35
STEC-HUS, with some patients demonstrating comple­ Later onset (3–6 months) of diabetes has also been
ment cleavage products, soluble C5b-9 (sC5b-9), and reported. Cardiovascular involvement with hypertension,
comple­ment-coated micro­vesicles.21–23 In vitro, Stx binds to congestive heart failure, pericardial effusion, depressed
short consensus repeats 6–8 and 18–20 of FH, impairing its myocardial function, and left ventricular hypertrophy is
complement regulatory effect. Children with STEC-HUS reported in children with STEC-HUS.32 In a case series,
and reduced levels of C3 are at risk for neurological 19 of 64 patients with STEC-HUS undergoing autopsy had
involvement and severe clinical mani­ festations.24 The cardiac involvement, including 15 with TMA affecting
patho­ genesis of neurological findings is unclear, and myocardial vessels, and two each with multiple haemorr­h­
includes the combination of Stx-induced vascular injury ages without TMA, and left ventricular infarction.36
and endothelial dysfunction, inflammation of neural cells
due to cytokine release, and direct binding of Stx to neural S pneumoniae-associated HUS
cells.25,26 Epidemiology
S pneumoniae-associated HUS (Sp-HUS), comprising
Clinical features 5% of all cases of HUS, is a serious complication of
Profuse bloody diarrhoea (haemorrhagic colitis) occurs in invasive pneumococcal disease.37,38 The incidence of
about 90% of patients around 3–8 days after ingestion of HUS after invasive pneumococcal disease is 0·4–0·6%,
contaminated food. Abdominal pain is substantial and but it is possibly underdiagnosed because no clear case
defecation is painful. Inflammation mainly affects the right definition or specific laboratory test exist, and it can be
colon, with notable oedema, easy bleeding, and longitudinal misdiagnosed as pneumococcal sepsis or disseminated

1724 www.thelancet.com Vol 400 November 12, 2022


Seminar

intravascular coagulation39,40 (appendix p 2). 41,42 A public test in patients with invasive pneumococcal infection
health surveillance study in the UK from 2006 to 2016 seems to be sensitive for Sp-HUS, but its specificity is
reported a reduction in the incidence of Sp-HUS unclear.
from 0·25 per 100 000 to 0·08 per 100 000 during that
time period associated with change in the pneumococcal Clinical features
conjugate vaccine from the seven-valent vaccine to the Compared with those affected by STEC-HUS, children
13-valent vaccine.43 Serotype 19A was most commonly affected by Sp-HUS are younger (aged <2 years) with severe
associated with HUS, probably due to enhanced renal or haematological disease or both, have a longer stay
expression of neuraminidase in the serotype. in hospital, and are more likely to have persistent kidney
dysfunction due to cortical necrosis41,53,54 (appendix p 2).
Pathophysiology Approximately 25% of patients have extrarenal
Pneumococcal neuraminidase cleaves terminal N-acetyl complications, including impaired liver function.54
neuraminic acid (sialic acid) from glycoproteins present
on erythrocytes, glomerular endothelial cells, renal Other infectious causes of HUS
epithelial cells and platelets, exposing the normally Other infectious causes of HUS have been well studied
hidden Thomsen-Friedenreich (T) antigen44 (appendix and reported in the literature. The pathogenesis of
pp 17–20). Preformed IgM antibodies react to exposed infectious disease-associated HUS is complex, and
T antigen on these cells, with initiation of a cascade of includes direct endothelial injury and complement
events resulting in Sp-HUS.44,45 A heavy bacterial load activation.55 The list of infectious causes is long and
leads to increased production of neura­minidase (which includes H1N1 influenza (production of neuraminidase
cleaves glycosidic linkages of neuraminic acid), making similar to Sp-HUS), HIV, cytomegalovirus, Epstein-Barr
Sp-HUS more likely. It has been hypothesised that virus, varicella zoster virus, parvovirus B19, malaria, and
complement plays a role in the development of Sp-HUS, dengue fever. Microvascular injury and TMA have also
although the precise mechanisms are not understood. been reported with SARS-CoV2 infection, due to its
Two recent studies identified genetic defects of comple­ direct effect on endothelial cells and complement
ment proteins that might be responsible for Sp-HUS,46,47 activation.56
while another study postulated that the loss of cell-
surface binding sites for FH occurs secondary to Primary aHUS or complement-mediated HUS
desialylation. Some pneumococcal serotypes (serotypes Epidemiology
2 and 3) directly bind to FH and inhibit its action, aHUS is an ultra-rare disease with an estimated annual
thereby leading to acquired dysregulation of incidence in the USA of around two cases per 1 000 000.37
complement.48,49 aHUS can occur at any age but onset during childhood is
more frequent than in adulthood and accounts for
Definition and diagnosis approximately 10% of cases in children, but most HUS
In 2002, the Canadian Pediatric Surveillance Program cases in adults. Autoantibodies to FH are responsible for
classified patients with invasive pneumococcal infection about 10% of cases of complement-mediated aHUS.3
and features of HUS into definite and possible cases This type of HUS is often referred to as DEAP-HUS
(appendix p 3). Renal biopsy is usually not required for the (deficiency of FHR1 plasma protein and anti-FH positive
diagnostic workup. Many experts use the peanut lectin HUS; appendix p 12).57
agglutination and direct Coombs tests as evidence of
T antigen activation to define Sp-HUS.41,42,50,51 Coagulation Pathophysiology
studies to rule out disseminated intravascular coagulation The pathophysiological consequences of aHUS result
are also included in the definitions.41,51 Invasive pneumo­ directly from damage due to uncontrolled activation of
coccal infection is diagnosed on culture, antigen detection, the alternative complement pathway. 58–60 A link between
or positive PCR from blood or physiologically sterile complement and aHUS was first reported in 1981 in two
biological fluid. brothers who had deficiency of FH; later, in 1998,
A modified Sp-HUS case definition41,42 is shown in the mutations in CFH were recognised to be associated with
appendix (p 3). Demonstration of T antigen presence by aHUS.61,62 Since then, mutations in several other
peanut agglutinin test is 86% sensitive and 57% specific complement genes have been identified.63 The alternative
for Sp-HUS or pneumococcal haemolytic anaemia, and pathway is most commonly implicated. A unique feature
has a positive predictive value of 76%.52 Since T antigen of this pathway is its continuous low-level spontaneous
is activated before the drop in haemoglobin or platelets, activation through a process of C3 hydrolysis known as
use of the peanut agglutinin test is recommended as tick-over. Tick-over drives the production of low levels of
soon as invasive pneumococcal disease is suspected. C3 convertase (C3bBb) and if complement is further
Although the positivity of direct Coombs test among activated, C5 convertase (C3bBbC3b) forms. C5 convertase
children with Sp-HUS varies, it has been reported to be cleaves C5 to generate C5b, which binds to C6, C7, C8, and
positive in up to 90% of patients.39,48 A positive Coombs multimers of C9 to produce C5b-9, the membrane attack

www.thelancet.com Vol 400 November 12, 2022 1725


Seminar

complex. As early as 30 min after assembly of the C5 convertases. CR1 and MCP act as cofactors for
membrane attack complex, cell lysis occurs. complement factor I-mediated cleavage of C3b. Clusterin
Complement damage to self-surfaces is prevented by and vitronectin are plasma proteins that prevent the
regulators of complement activation (RCA) proteins.64,65 insertion of terminal complement complexes into cell
The major RCA proteins in the pathogenesis of aHUS are membranes. The activity of the alternative pathway
FH, factor I (FI), and membrane cofactor protein (MCP, C3/C5 convertase can be enhanced by the only known
CD46; table 1). Dysregulation of the alternative pathway physiological positive complement regulator, properdin.
can arise from loss-of-function mutations in the genes
that encode these proteins or from hybrid genes that Genetics
encode FHR1–FH fusion proteins.71 Cross-talk between Approximately 60% of patients with aHUS carry a
the coagulation system and the complement cascade also mutation in a gene encoding a protein essential for
occurs and is important to the disease process72 (appendix alternative pathway function. Mutations in CFH are
pp 18–19). Complement activation occurs in a sequential most frequently identified and account for 25–30% of
manner by initiation of complement activation, formation cases.66,67 These mutations either reduce production of
and amplification of C3 convertase (C3bBb), formation FH, result in a null allele, or generate a protein with
and amplification of C5 convertase (C3bBbC3b), and the abnormal functionality. Most missense mutations that
assembly of the terminal complement complex or MAC impair FH function occur at the C-terminal region
(C5b-9). The self-amplifying process of the alternative of the protein, a site essential for attachment to
pathway is inhibited by multiple regulator molecules. cell membranes and protection of host cells from
FH is a fluid-phase inhibitor of the C3 convertase complement-mediated damage.60,73 Fusion proteins,
and competes with factor B for binding to C3b which include FH/FHR1, FH/FHR3 and FHR1/FH
and displaces Bb from the C3bBb convertase (decay- fusions, act as competitive antagonists of FH and also
accelerating activity). It acts as a cofactor for FI in the lead to complement dysregulation at the cell surface.71,74
cleavage of C3b to iC3b. Three membrane glycoproteins, MCP mutations are reported in about 9% of patients
C3b receptor (CR1, CD35), MCP (CD46), and decay with aHUS and lead to decreased expression of MCP on
accelerating factor (CD55), act as inhibitors of C3 and cell surfaces or impaired regulation of complement.68,75
C5 convertases, whereas protectin (CD59) inhibits the Mutations in CFI are reported in around 8% of patients.
assembly of the MAC. CR1 and delay-accelerating factor Less frequent are the gain-of-function mutations in
have decay-accelerating activity towards C3 and C3 (2–8%) and CFB (1–4%), both of which increase

Protein function Frequency End-stage renal disease in native kidney after first presentation or Risk of recurrence
of at 1 year in transplant
mutation kidney
French registry Italian registry* American Global registry
registry†
CFH Inhibits C3b binding, inhibits 20–30% Children: 47%; 31% 41% Children: 42%; 80–90%
production and accelerates decay adults: 58% (41% unknown) adults: 42%
of C3 convertase; cofactor for FI
MCP Cofactor for FI to cleave C3b and 10–15% Children: 0%; 6% 14% Children: 0%; 15–20%
C4b on surface of host cells adults: 63% (57% unknown) adults: 0%
C3 Generates C3a and 5–10% Children: 43%; 58% Unknown Children: 25%; 40–50%
C3b, constituents of adults: 63% adults: 11%
C3 convertase and C5 convertase
CFB Binds to C3 and forms C3 1–4% NA NA 33% NA 100%
convertase (14% unknown)
CFI Cleaves and inactivates C3b and 4–10% Children: 17%; 60% 33% Children: 0%; 70–80%
C4b in the presence of cofactors adults: 42% (33% unknown) adults: 33%
FH, MCP, and THBD
THBD Regulate FI-induced 3–5% NA 15% 25% NA Not reported
C3b inactivation (25% unknown)
Combined Depends on the combination of 2–12% NA NA 71% NA Combined CFH
mutation proteins affected (29% unknown) and MCP risk
haplotype: 30%
FHAA Affects FH function 4–6% NA 37% NA Children: 28%; High, if antibody
adults: 10% titre is high
FH=factor H. FHAA=factor H autoantibody. FI=factor I. MCP=membrane cofactor protein. NA=not available. THBD=thrombomodulin. *Overall outcome and was not
reported separately in children and adults. †Overall outcome; in many patients the outcome was unknown.

Table 1: Role of complement regulatory proteins and their mutation in atypical HUS66–70

1726 www.thelancet.com Vol 400 November 12, 2022


Seminar

alternative complement pathway activity. Thrombo­ others can have persistent visual deficits. Cardiac
modulin is a cofactor in the initiation of the protein C manifestations are identified in 7% of paediatric patients
anticoagulant pathway and also accelerates the inactivation with aHUS.79 In some patients with aHUS, hypertension
of C3b by FI. In-vitro studies have shown that THBD and fluid overload associated with acute kidney injury can
mutations result in dysregulation of the complement lead to cardiac dysfunction, but there are also reports of
system.76 About 12% of patients carry mutations in two or cardiac symptoms independent of these factors, suggesting
more genes implicated in aHUS. direct injury to the myocardial tissue and vasculature.82
Although familial disease (autosomal recessive or Peripheral vascular manifestations with skin changes,
dominant inheritance) is observed in about 20% of including cutaneous rash and peripheral gangrene, have
families,67,77 the absence of a positive family history been described in a few reported aHUS cases.83–85 Skin
does not preclude the possibility of genetic trans­ changes can occur in the absence of anaemia or
mission. Most aHUS-associated complement mutations thrombocytopenia, and therefore might be evidence of
segregate as heterozygous variants, meaning that the persistent comple­ment activation in the absence of other
mutation will be found in a parent. Penetrance is highly biochemical abnormalities typically associated with aHUS.
variable and approximates 50%. Factors that impact Gastro­intestinal manifestations can also occur: analysis of
penetrance include age, specific triggers (eg, infection or global registry of aHUS data by Schaefer and colleagues69
pregnancy), genetic burden (other gene variants) and reported gastrointestinal manifestations in a greater
circulating levels of regulatory proteins (especially FH), proportion of paediatric patients (47%) than adults (33%)
under­scoring the fact that aHUS is a multifactorial disease in the acute phase. However, only 10% of paediatric
that results from environmental events that initiate patients experienced gastrointestinal symptoms in a
endothelial damage and genetic factors (mutations and at- Turkish registry study.86 Pulmonary mani­festations were
risk polymorphisms) that affect disease progression. seen in a lower proportion of pediatric patients (12%) than
adult patients (20%) in the global registry of aHUS.69
Clinical features Careful monitoring of fluid and respiratory status is
Most patients with aHUS present acutely with the needed in patients with aHUS because respiratory
triad of TMA with non-immune haemolytic anaemia, failure secondary to pulmonary oedema necessitating
thrombocytopenia, and kidney dysfunct­ ion follow­ ing a mechanical ventilation is a serious and life-threating
triggering event. Less commonly, patients have indolent complication.
disease, with subclinical anae­mia, fluctuating throm­bo­
cytopenia, preserved kidney function, and non-s­ pecific Non-complement-mediated aHUS
symptoms such as malaise, fatigue, and anorexia, making Non-complement-mediated aHUS, including that caused
diagnosis difficult. aHUS renal manifes­tations are variable by DGKE and WT1 mutations, should be considered
and include acute kidney injury, nephrotic range when disease presentation occurs early, typically within
proteinuria resulting from glomerular basement the first 12 months of life, and also in patients who are
membrane damage, and hyper­tensive emergency. Patients resistant to complement inhibitor therapy (panel 1 and
with hypertensive emergency at presentation have poor appendix p 18). G6PD deficiency, an X-linked recessive
kidney outcomes and a high incidence of progressive disease due to mutations in G6PD, can in rare cases
kidney disease.78 mimic the diagnosis of HUS.101,102
Extrarenal manifestations occur in around 20% of
patients with aHUS, with the incidence of specific organ Metabolism-associated HUS
system complications ranging from a few case reports to The differential diagnosis of HUS includes metabolism-
50% of described patients (figure 2). Such complications associated conditions, the most common being cobala­min
can occur not only during the acute phase of aHUS, but C deficiency, an inborn error of vitamin B12 metabolism.
also later.66,79–81 Neurological symptoms are reported in Cobalamin G deficiency is less common but also
8–48% of cases: seizures can occur in the acute phase both can present as a TMA. Inherited as an autosomal recessive
in the presence and absence of hypertension. MRI changes disorder, cobalamin C disease is caused by mutations in
can help to distinguish between posterior reversible MMACHC, whereas cobalamin G is due to a mutation in
encepha­­lopathy syndrome with hypertension versus TMA- MTR. Notwith­­s­tanding their rarity, these diagnoses are
specific findings, which include hyper i­ntense lesions in important to make because targeted disease-specific
the posterior white matter, posterior cortex, deep white therapy is available, and therapy with eculizumab is not
matter, thalami, brainstem, and basal ganglia. Ocular effective. The pathophysiology, clinical features, and
involvement can be serious but is rare; reported ocular management of metabolism-associated HUS are
findings on fundoscopic examination include bilateral discussed in panel 2, figure 3, and the appendix (p 19).
flame-shaped intraretinal haemorrhage, optic disc oedema,
central retinal vein occlusions, and tortuosity of the retinal Transplant-associated HUS
vessels. Full recovery after initiation of therapy with HUS can occur after haematopoietic stem cell
eculizumab has been reported in some patients, whereas transplantation (HSCT) or, less commonly, after solid

www.thelancet.com Vol 400 November 12, 2022 1727


Seminar

Panel 1: Clinical characteristics of HUS due to non-complement causes


DGKE mutation-associated HUS cohort, including developmental delay, learning difficulties,
• Epidemiology and pathophysiology and autistic spectrum disorder87
• DGKE-mediated disease is exceedingly rare (0·009 cases
Management87,88,95
per million per year)87
• Supportive management
• Inherited as an autosomal recessive disease (homozygous or
• Brocklebank and colleagues87 described their experience in
compound heterozygous mutations)
15 patients with numerous relapses while on eculizumab
• DGKE encodes diacylglycerol kinase-epsilon (DGKε), an
• Renal half-life is approximately 20 years and those who
intracellular lipid kinase with high specificity for
progress to end-stage renal disease should undergo kidney
diacylglycerol conjugated with arachidonic acid in the
transplantation without the need for eculizumab and no
absence of DGKε; a shortage of phosphatidyl inositol
relapses reported in the transplant
bisphosphate compromises VEGFR2-dependent protein
kinase B (Akt) activation, perturbing the complex autocrine WT1 mutation-associated HUS96–100
signalling pathways required to maintain kidney • The WT1 gene encodes proteins that regulate progenitor
microvascular stability88–90 cells and their differentiation, especially those of the gonads,
• Renal biopsy demonstrates chronic thrombotic uteri, and kidneys
microangiopathy, often with foot process effacement88 • WT1 variant is pathogenic for Denys–Drash syndrome, a rare
• DGKE is highly expressed in the brain91 and studies in mice disorder characterised by early-onset nephrotic syndrome
with a targeted deletion of DGKE suggest a role in and renal failure, pseudo-hermaphroditism, and a high risk
modulating neuronal signalling pathways92–94 of Wilms’ tumour
• Pathogenesis of aHUS in WT1 mutation is not fully
Clinical presentation
understood but several reports suggest podocyte
• Patients with DGKE-mediated HUS present with a few unique
dysfunction leading to decreased VEGF, contributing to the
features that can offer clinical clues as to the underlying cause
development of renal thrombotic microangiopathy
• Typically presents within the first 12 months of life, with an
• There are reports of five cases of Denys–Drash syndrome
estimated 25% of atypical HUS cases in this age group due to
presenting with atypical HUS; all cases progressed to end-
DGKE mutations; the fractional contribution increases to 50%
stage renal disease despite treatment with plasma exchange,
if there is a positive history of familial disease88
eculizumab, or both, which led to testing for other gene
• Nephrotic syndrome is also common within 3–5 years of
mutations by whole-exome sequencing and diagnosis of
diagnosis, a noteworthy clinical finding because heavy
WT1 mutation; all received renal transplantation without
proteinuria is not typically seen with complement-mediated
recurrence of atypical HUS
HUS
• Brocklebank and colleagues reported a high incidence HUS=haemolytic uraemic syndrome.
(6 of 14 individuals) of developmental disorders in their

organ transplantation. HSCT-TMA is a severe compli­ in the setting of autoimmune disease is multifactorial.55,142–145
cation of both allogeneic and autologous tran­splantation. It is important to rule out thrombotic thrombocytopenia
Patients typically present 35–47 days following HSCT with purpura in patients with systemic lupus erythema­
the classic triad of HUS. Their pathophysiology, clinical tosus because these entities can sometimes occur
features and management are discussed in panel 3 and together146,147 Treatment of secondary HUS in the setting
the appendix (pp 10, 22). of autoimmunity might include anti-complement
medications, treatment of underlying conditions, or
Secondary HUS both.55,144,148–150
Secondary HUS refers to any disorder presenting
with HUS features and includes entities such as malig­ Malignancy-induced HUS
nant hypertension140 or autoimmune disorders such as Malignancy can induce HUS through a wide variety of
systemic lupus erythematosus, antiphospholipid anti­ mechanisms, including systemic microvascular meta­
body syndrome, or scleroderma. Whether these disorders stases, extensive bone marrow invasion, or secondary
cause HUS or have features that overlap with HUS is not necrosis.151 Increased thrombin generation can directly
entirely clear. In malignant hypertension, HUS features activate the complement system through C5 cleavage.72,152
may be secondary to hypertension; control of blood HUS syndromes have been reported with metastatic
pressure alone without anti-complement therapy might gastric and ovarian cancers, leukaemias, and lymph­
be sufficient to treat this disorder. However, complement- omas.153–157 Furthermore, many chemotherapeutic drugs
mediated HUS itself can present as a hypertensive have been associated with HUS (see the section on
emergency, confusing the diagnosis.141 HUS pathogenesis drug-induced HUS later). Treatment of the underlying

1728 www.thelancet.com Vol 400 November 12, 2022


Seminar

Panel 2: Clinical characteristics of patients with HUS associated with disorder of cobalamin metabolism
Epidemiology103,104 • Clinical features common to both cblC and cblG include
• Cobalamin C (cblC) disease, an inborn error of vitamin B12 feeding difficulties, failure to thrive, movement disorder,
metabolism, is the most common disorder of cobalamin abnormal muscle tone, developmental delay, megaloblastic
metabolism anaemia, neutropenia, venous thrombosis, and embolism.
• Inherited as an autosomal recessive disorder, cblC disease is Additionally, children with cblC disease might be small for
caused by mutations in MMACHC and cobalamin G (cblG) gestational age and have cardiomyopathy and stroke106,107
disease is due to methionine synthase deficiency caused by • When disease onset is in later childhood and adulthood, the
a mutation in MTR presentation is dominated by neurological manifestations
• cblC disease accounts for fewer than 0·5% of cases of such as ataxia, cognitive impairment, and psychosis108,109
secondary HUS • Inherited disorders of cobalamin metabolism should be
• cblG disease also presents with thrombotic considered in infants presenting with HUS, especially those
microangiopathy (8 cases reported ever) with a history of megaloblastic anaemia and any other
• Inherited disorders of cobalamin metabolism should be clinical features listed above
considered in infants younger than 1 year of age presenting • Normal vitamin B12 levels should not preclude further
with thrombotic microangiopathy because targeted testing for cobalamin defect
disease-specific therapy can be life-saving • Screening laboratory tests include: plasma homocysteine
levels, and plasma MMA and plasma amino acid
Pathophysiology
(methionine levels), which are required to diagnose a
• Suggested mechanism of HUS (appendix p 19) is
disorder of intracellular cobalamin metabolism
endothelial damage induced by hyperhomocysteinaemia,
• Confirm diagnosis by genetic testing: MTR mutation in cblG
impaired nitric oxide-dependent inhibition of platelet
and MMACHC mutation (>90 different pathogenic variants
aggregation, or induction of a procoagulant state of
have been identified) in cblC110
endothelium leading to microthrombi formation. Only a
minority of patients with cblC and cblG disease develop Management103,110–113
HUS, suggesting that other modifiers have a role • Stabilise patients in consultation with metabolic specialist
• The hallmarks are hyperhomocysteinaemia with low to treat acidosis, reversing catabolism and initiating
methionine in both cblC and cblG; high levels of parenteral hydroxycobalamin
methylmalonic acid (MMA) are found in cblC and low MMA in • Goal of treatment is to lower plasma homocysteine,
cblG disease maintain plasma methionine in the normal range, and
• Renal biopsy (not necessary for diagnosis) shows lower MMA if elevated (cblC)
vacuolated appearance of the glomerular basement • Hydroxycobalamin intramuscular form of vitamin B12
membrane more frequently in patients with cblC deficiency supplements the precursor of cobalamin metabolism
than in other thrombotic microangiopathy controls • Betaine acts as a methyl donor and helps in the conversion of
(in 6 of 7 patients with cblC deficiency vs 1 of 14 thrombotic homocysteine to methionine
microangiopathy controls; p<0·001)105 • Folic acid (or folinic acid/leucovorin which efficiently crosses
the blood–brain barrier) replenishes the folate cycle, and
Clinical presentation and diagnosis
improves plasma levels of homocysteine and methionine
• cblC and cblG disease most commonly present in early
infancy (within the first few months of life); cblC disease HUS=haemolytic uraemic syndrome. MMACHC=methylmalonic aciduria and homocystinu-
ria, cobalamin type C. MTR=methionine synthase.
can manifest at any age

cancer often results in resolution of HUS, but HUS can syndrome.161,162 The ratio of soluble fms-like tyrosine
recur with relapse of the malignancy.158 kinase-1 (sFlt-1) to placental growth factor is being
increasingly used to exclude pre-eclampsia or eclampsia
Pregnancy-associated aHUS and HELLP syndrome, and enables clinicians to search
Pregnancy-associated aHUS refers to TMA resulting from for other TMA causes, including pregnancy-associated
uncontrolled complement activation during pregnancy or aHUS.162,163 In women with pregnancy-associated aHUS,
post partum. It accounts for 7% of total aHUS cases and complement abnor­ malities including mutations in
up to 20% of all cases of aHUS in women.3,159,160 aHUS CFH (30%) and CFI (9%) have been demonstrated.164 In
should be suspected when, despite delivery, TMA fails to the pre-eculizumab era, a high proportion of cases of
improve and extends beyond 72 h, which is the expected pregnancy-associated aHUS (around 76%) progressed to
time for recovery in other causes of pregnancy-associated end-stage renal disease,159 but eculizumab has been shown
TMA, including eclampsia, pre-eclampsia, and HELLP to improve kidney outcomes.164 The optimal duration of
(haemolysis, elevated liver enzymes, low platelet count) eculizumab therapy is debatable; some experts propose its

www.thelancet.com Vol 400 November 12, 2022 1729


Seminar

Suspected HUS
Haemoglobin <10 g/dL, schistocytes ≥2%, lactate dehydrogenase >450 IU/L,
platelets <150 000/µL, acute kidney injury

Plasma exchange, Thrombotic Shiga toxin-producing STEC-HUS


immunosuppression thrombocytopenic purpura enterohemorrhagic Escherichia Supportive management
ADAMTS13 activity and coli (STEC)
ADAM- ADAMTS13 autoantibodies Stool or rectal swab culture, Shiga
TS13 Shiga toxin PCR or serology toxin-
activity positive
<10%
Parenteral hydroxocobalamine, Metabolism-associated HUS Streptococcus pneumoniae HUS Streptococcus pneumoniae-
oral folinic acid, and betaine Cobalamin C and cobalamin G Blood culture, chest x-ray, lumbar HUS
disease, homocystinaemia, low puncture, Coombs test, Cephalosporin and
methionine, high or low T-antigen testing vancomycin, washed red
methylmalonic acid blood cells, and supportive
Genetics: MMACHC and MTR management

Secondary HUS Infection-associated HUS


Systemic lupus erythematosus, Evaluate if clinically indicated
antiphospholipid syndrome,
malignant hypertension, drugs,
malignancy, transplant

Atypical HUS

C3, C4, factor H, factor I, CD46,


anti-factor H antibody
Genetics: CFH, CFI, CD46, THMD, C3,
CFB
MPLA: CFHR1-5, CFHR1/CFH

Supportive management. DGKE-HUS and WT1-HUS


No reported benefit of Early onset (<2 years of age).
eculizumab or plasma Whole-exome sequencing if not
already included in the genetic
panel

First-line eculizumab within 24 h of onset; if not available, start plasma exchange


with fresh frozen plasma or plasma infusion if plasma exchange not available

Anti-FH antibodies positive if >150 antibody units per mL or >1000 units/mL. Add
plasma exchange and immunosuppression if already started on eculizumab

Figure 3: Suggested diagnostic pathway and treatment for suspected HUS


HUS=haemolytic uraemic syndrome.

discontin­
uation after normalising haematological and causes of TMA with these agents are diverse and might be
kidney manifestations with monitoring for recurrence in dose-related. Drug-induced TMA should be suspected
subsequent pregnancies.165 when there is a sudden onset of acute kidney injury with
anuria that occurs within days of starting a new
Drug-induced HUS medication, although it can also be delayed. Management
Drug-induced TMA occurs due to immune-mediated or involves stopping the causative medication, although that
direct toxic effects of medications on the endothe­lium.166,167 alone might not be sufficient, and in patients with
TMA resulting from inhibitors of VEGF (anti- worsening acute kidney injury, other therapies such as
VEGF) involves injury to renal podocytes.168 Quinine- plasma exchange and eculizumab should be considered.
associated TMA develops secondary to quinine-dependent Im­prove­ment of TMA is likely, although some degree of
anti­
bodies against endothelial cells, platelets, and kidney injury can persist.55
leuk­o­cytes.169 Other examples include calcineurin in­hi­bi­
tors (cyclosporin and tacrolimus), mTOR inhibitors Diagnosis of HUS
(sirolimus and everolimus), vincristine, gemcita­­bine, and Early diagnosis is important because HUS is associated
oxalipla­tin.170,171 As opposed to anti-VEGF and quinine, the with acute morbidity and mortality, and delays in

1730 www.thelancet.com Vol 400 November 12, 2022


Seminar

Panel 3: Main clinical characteristics in patients with transplant-related HUS


Epidemiology114–120 • Heart, lungs, brain, and intestines can be affected.
• Thrombotic microangiopathy (TMA) occurring after • Diagnosis: tissue histology is gold standard, but often
haematopoietic stem cell transplantation (HSCT) or solid prohibitive. Diagnostic algorithms available combine clinical
organ transplantation (SOT). and laboratory markers in lieu of tissue diagnosis.
• HSCT-TMA: follows allogeneic or autologous SOT-TMA
transplantation with an incidence of 3–75% and a mortality • Thrombocytopenia and Coombs-negative haemolytic
rate of 15–90%. anaemia.
• SOT-TMA: follows renal, liver, cardiac, lung, or intestinal • Microangiopathy, including schistocytes on smear, elevated
transplantation, with an incidence of 0–33% and a mortality lactace dehydrogenase, and low haptoglobin often
rate of 25–60%. observed.
Pathophysiology121–123 • Microvascular thrombosis can manifest systemically or be
• Caused by systemic or local vascular endothelial injury restricted to the kidneys or the graft itself.
triggered by the transplantation process.
• Mechanisms of endothelial injury in HSCT-TMA: See the appendix (p 10) for differential diagnosis of transplant-
microthrombi formation due to activation of endothelial related HUS.
cells to produce a pro-coagulant state, antigen-presenting Management124–139
cells and lymphocytes, and complement. HSCT-TMA
• The so-called three hit hypothesis proposes that TMA • Address underlying or inciting factors and optimise anti-
develops after three insults: 1) underlying predisposition to hypertensive management.
endothelial injury; 2) pre-transplant conditioning regimen; • Plasmapheresis or eculizumab (neither is optimally
and 3) post-transplant factors causing persistent effective).
endothelial injury. • Other experimental therapies: conversin (C5 inhibitor),
• The appendix (p 10) lists the risk factors for HSCT-TMA and narsoplimab (MASP-2 inhibitor), rituximab, recombinant
biomarkers of endothelial injury. thrombomodulin, defibrotide, and daclizumab (anti-CD25).
• Mechanisms of endothelial injury in SOT-TMA:
transplantation surgery, immunosuppressive drugs SOT-TMA
(calcineurin inhibitors and mTOR inhibitors), acute • Trial alternative immunosuppression, implement supportive
rejection, infection, or relative deficiency of ADAMTS13. measures.
• Plasmapheresis or eculizumab (neither is optimally
Clinical presentation and diagnosis116,124–133 effective).
HSCT-TMA • Basiliximab (anti-CD25) is a potential experimental therapy.
• Classic triad of renal dysfunction (severe), microangiopathic
ADAMTS13=a disintegrin and metalloproteinase with thrombospondin type 1 motifs,
haemolytic anaemia, and thrombocytopenia. member 13. HUS=haemolytic uraemic syndrome.
• Hypertension an early finding, proteinuria often present.
• Schistocytes on peripheral blood smear is pathognomonic,
but might be absent.

diagnosis can lead to chronic multisystem morbidity. All typhus, and leptospirosis also need to be excluded.
types of HUS have similar presentation with some or all Patients with thrombotic thrombocytopenia purpura
features of the triad of non-immune microangio­pathic also show features of TMA and can be diagnosed by
haemolytic anaemia (haemoglobin <10 g/dL) and testing for ADAMTS13 activity. Once the afore­mentioned
fragmented red cells on peripheral smear (schistocytes causes of TMA have been excluded, the diagnosis of
≥2%) with either increased lactate dehydrogenase or HUS is made. Clinical and family history, age at
undetectable haptoglobin, throm­bocytopenia, and acute presentation, recent illnesses and associated symptoms
kidney injury. A direct Coombs test is an important guide subsequent evaluation. aHUS, especially
diagnostic step in differentiating HUS from other causes complement-mediated HUS, is a diagnosis of exclusion
of immune-mediated haemolysis. A systematic approach at presentation and therefore all potential causes of HUS
is necessary to evaluate all patients with suspected TMA, must be considered when evaluating a patient in the
with exclusion of systemic sepsis with or without acute setting.
consumptive coagulopathy (prolonged prothrombin Several diagnostic algorithms exist that use stepwise
time and partial thromboplastin time, low fibrinogen, approaches to identify the cause of HUS. A suggested
elevated D-dimer, and thrombocytopenia). In tropical diagnostic approach for a patient with suspected HUS is
countries, infections such as malaria, dengue fever, shown in figure 3 (see appendix p 13 for details).

www.thelancet.com Vol 400 November 12, 2022 1731


Seminar

Management of HUS extended-spectrum cephalosporin and vancomycin in


Supportive care for all types of HUS critically ill children with suspected invasive pneu­mococcal
Supportive care is similar in all patients with HUS disease. Because of concern that anti-T antigen antibodies
irrespective of the cause. Increased awareness and early might be present in blood products, dextran washing of
recognition of symptoms with advanced intensive care peripheral red blood cells before transfusion is
unit availability60,172 and dialysis care have improved recommended,45 and plasma-containing blood products
outcomes173 (appendix p 14). such as fresh frozen plasma are generally avoided.
Successful use of plasmapheresis specifically with albumin
Management of STEC-HUS to remove the anti-T antigen anti­ bodies has been
Epidemiological data have shown an association between reported;183,184 however, there is little systematic evidence
dehydration and adverse outcomes in patients with STEC that it improves outcomes. Because of evidence of
infection.174 Early use of isotonic fluids is recommended complement activation, there are few reports of eculizumab
in patients with dysentery, starting from onset of bloody use to treat severe cases of Sp-HUS, with improvement
diarrhoea to the day of onset of HUS and monitoring for in platelet counts and subjective improvement in neuro-
fluid overload in patients with acute kidney injury. The irritability.47,54 More evidence from controlled trials is
role of antibiotics, specifically cotrimoxazole and necessary before eculizumab can become standard
β-lactams, for STEC gastroenteritis is debated, since these therapy.
might induce expression and release of Stx and increase Improved awareness of Sp-HUS could help to modify
the risk of HUS.175,176 Tarr and Freedman reinforce the therapy appropriately and improve outcomes. Long-
importance of antibiotic avoidance in possible or definite term follow-up of renal function is indicated to detect
STEC infections because of an absence of evidence that late-onset end-stage renal disease. A sustained
any antibiotic class improves STEC infection versus no pneumococcal vaccination programme and vigilance for
antibiotic treatment, and because of growing evidence replacement serotypes will be the key for persistent
that antibiotics increase the risk of developing HUS when reduction in Sp-HUS cases worldwide.
used to treat early STEC infections.177 Medications that
slow gastric transit, including loperamide, diphenoxylate, Management of complement-mediated HUS
and opioids, should be avoided because they reduce the Complement C5 inhibitors
colonic excretion of STEC and might be associated with Eculizumab is a humanised monoclonal IgG antibody
an increased risk of HUS. The practice of plasma that binds to the C5 complement protein and blocks its
exchange varies, with no consensus regarding its use. cleavage, thereby preventing the formation of C5b-9,
Although used anecdotally in patients with refractory the membrane attack complex.185 Eculizumab therapy
illness and severe neurological symptoms, current reduces anaphylatoxin-induced inflammation and limits
evidence does not support plasma exchange or immuno­ the C5b-9 prothrombotic consequences of complement
adsorption.178 Eculizumab, a humanised monoclonal activation. Three trials, two in adole­scents and adults5,6
anti-C5 antibody that blocks the terminal complement and one in paediatric patients7 demon­strated the efficacy
pathway, has shown remarkable success in aHUS. at 26 weeks of weight-based dosing once every 2 weeks
However, analysis of the German STEC-HUS registry did (appendix p 4) for the treatment of aHUS, and long-term
not show a benefit of eculizumab in adults.179 Although extension data at 2 years confirmed these positive
one report did suggest benefit in three children with findings186 (table 2). Eculizumab was approved for the
neurological involvement,180 the overall evidence of treatment of aHUS both in the USA and in many
efficacy and safety of eculizu­ mab in STEC-HUS is European countries in 2011. Additional eculizumab
unclear.181 Two placebo-controlled randomised trials have efficacy and safety data are now available from nearly a
completed enrolment (ECUSTEC in the UK decade of registry and non-trial patient data.85,187
[ISRCTN89553116] and ECULISHU in France Ravulizumab, a humanised monoclonal antibody that
[NCT02205541]) to examine the therapeutic role of blocks terminal complement activation at C5, engineered
eculizumab in STEC-HUS, the results of which should from eculizumab, has an extended half-life and can be
provide more clear evidence of the utility and safety of its administered once every 8 weeks. It is effective in adults
use in STEC-HUS. Although animal models suggest that aged 18 years and older with aHUS.188 A paediatric study
administration of Shiga toxin-binding and neutralising (birth to 18 years) also demonstrated safety and efficacy
agents might influence the disease course, clinical studies when administered every 4–8 weeks based on body­
have not shown benefit of such drugs. Plasma infusions, weight189 (appendix p 4) Ravulizumab was approved for
heparin, urokinase, dipyridamole, and glucocorticoids do treatment of aHUS in the USA in 2019.
not ameliorate the disease course.182 Because dosing of ravulizumab has not been studied
with plasma, use of eculizumab is preferred in the
Management of Sp-HUS acute management of aHUS, especially when results
Therapy for Sp-HUS remains supportive as discussed of ADAMTS13 and FH autoantibody levels are
previously. Pneumococcal infection should be treated with pending. Eculizumab can be safely switched to

1732 www.thelancet.com Vol 400 November 12, 2022


Seminar

Children: pre-eculizumab era Children: Adults: pre-eculizumab era Adults: eculizumab era trials Adults: Eculizumab post-trial data
(registry data) eculizumab (registry data) ravulizumab (children)
era trials
French Italian Global Trial 3 French Italian Global Trial 1 Trial 2 Trial 4 Trial 5* Turkish registry86† Menne
registry registry registry (n=22)7 registry registry registry (n= 17)6 (n=20) 6 (n=41)5 (n=56)137 et al187 ‡
(n=89)66 (n=149)67 (n=851)84 (n=89)66 (n=149) 67 (n=851)69
First episode 17% ·· 15% ·· 46% ·· 22% ·· ·· ·· ·· ··
6 months ·· ·· ·· 9% ·· ·· ·· 6% 10% 15% 28% ·· ··
1 year 29% ·· 5% 9% 56% ·· 9% 6% 10% 15% ·· ··
2 years ·· ·· ·· ·· ·· ·· ·· 12% 10% ·· ·· All patients: 10% ··
Eculizumab-
treated patients:
5%
3 years ·· 48% 2% ·· ·· 67% 4% ·· ·· ·· ·· ·· ··
5 years 36% ·· 1% ·· 64% 7% ·· ·· ·· ·· ·· 8·1%
*34/47 patients at chronic kidney disease stage 5 and 7/47 at chronic kidney disease stage 4 at baseline and 13/47 (with chronic kidney disease stage 5 at last follow-up. †5 of the eculizumab-treated patients had
low glomerular filtration rate at the start of treatment; glomerular filtration rate at last follow-up (1·7 years) >90 mL/min per 1·73m2 in 69·6% patients (23/33) in the plasma-treated group and 66% (68/103) of
eculizumab-treated patients. ‡Prospective long-term follow-up of trials 1–5.

Table 2: Percentage of patients with atypical haemolytic uraemic syndrome who progressed to end-stage renal disease or died in the pre-eculizumab era, in eculizumab era trials, and
post-trial data

ravulizumab once a patient’s clinical status is stable. or lower doses are feasible options (appendix pp 7–8).
Meningococcal vaccination, prophylaxis, or both are Large multicentre prospective studies including biopsy-
important before starting complement 5 inhibitors proven remission with varying doses of both
(appendix p 15). eculizumab and ravulizumab are required to
standardise long-term management of aHUS in
Frequency and duration of anti-complement therapy patients with rare variants in complement genes who
The optimal duration of eculizumab treatment for are at a high risk of relapse.
aHUS is unknown. High cost, risk of infection, and
unknown long-term sequelae have raised the Plasma therapy
question of whether discontinuation of eculizumab is Plasma therapy, including plasma exchange or plasma
safe. Additionally, there are reports of hepatotoxicity infusions, was the first line of treatment for aHUS in the
with eculizumab.190,191 There are few retrospective series, pre-eculizumab era based on experience in patients with
mostly in adults, showing eculizumab discontinuation thrombotic thrombocytopenia purpura. No prospective
with close monitoring for relapse is feasible.192–195 In 2021, trials have been done, but registry data66–69 have shown
a prospective multicentre study in 55 patients renal outcome is poor in patients with aHUS treated with
with aHUS (including children and adults) in France plasma therapy (table 2). Although eculizumab has
showed that the risk of aHUS relapse after eculizumab replaced therapeutic plasma exchange as the first-line
discontinuation (with close monitoring, including urine treatment for aHUS in many countries, plasma exchange
dipstick for albuminuria and haematuria twice weekly) continues to be the standard first-line treatment option
was low when there was no detected complement gene for aHUS when eculizumab is inaccessible (see
variant,196 supporting results from other retrospective appendix p 16 for guideline) and for FH autoantibodies-
studies (appendix pp 5–6). associated HUS (appendix p 12).201 Plasma exchange
Based on the effect of eculizumab in patients with might also have a role in severe cases of STEC-HUS,
paroxysmal nocturnal haemoglobinuria, trials in potentially decreasing circulating cytokines, Stx toxin,
patients with aHUS aimed at reaching trough levels for and unusually large von Willebrand factor multimers that
eculizumab between 50 and 100 μg/mL, which is the contribute to endothelial injury, although little data exist
level considered necessary to block complement (CH50 to support its routine use.202
activity <10%). However, all patients were treated with
fixed doses based on their weight and therapeutic Transplantation
drug monitoring was not done.194 As a result, serum Eculizumab has dramatically improved the outcome of
eculizumab levels above this target were often reached, kidney transplantation in patients with aHUS; however, its
prompting clinical investigators to adapt the treatment use for all transplant recipients is questioned. To
schedule by either increasing the interval or decreasing personalise aHUS management after transplanta­ tion,
the dosage to maintain recommended trough levels, to Zuber and colleagues retrospectively stratified recurrence
maintain complement blockade, or both.197–200 Based on risk as high, moderate, and low, and found that eculizumab
these reports, extended dose intervals up to 4–6 weeks is strongly and independently associated with significantly

www.thelancet.com Vol 400 November 12, 2022 1733


Seminar

reduced recurrence risk (hazard ratio 0·05, p<0·001) and and more than 80% of dialysis-dependent patients are able
longer graft survival in patients in whom complement to discontinue dialysis within a median time of
genetics and a past medical history predict a high or 7 days (range 4–15 days). In initial prospective trials, more
moderate risk of post-transplant recurrence.203 This than 80% of patients with thrombocytopenia had
recommendation has been adopted by Kidney Disease normalised platelet counts by a median time of 7 days
Improving Global Outcomes204 (appendix p 9). Global (range 1–80 days). The median time for LDH normal­
registry analysis also found significantly better early graft isation was 48 days (range 1–153 days). If platelet count
function in patients who received prophylactic eculizumab normalisation is not observed by week 2, assessment of
treatment compared with those who received eculizumab adequacy of complement blockade (CH50 activity <10%)
post-transplant.205 Treatment duration remains contro­ and eculizumab levels (50–100 µg/mL) are important.
versial and information about eculizumab withdrawal Other complement functional assays are being
after transplantation is scarce. Living related kidney eval­ua­ted for monitoring of optimal complement blockade
donation carries an increased risk of de-novo aHUS in the (appendix p 11). When there is a poor haematological and
donor and recurrence in the recipient if the donor carries renal response, one must rule out the presence of a C5
an at-risk genetic variant.206 Therefore, potential donors polymorphism impairing binding of eculizumab to C5216
with evidence of abnormal alternative complement and/or consider non-complement-mediated HUS,
pathway activity should be excluded. Liver transplant including that caused by DGKE and WT1 mutations.
remains an option in patients with liver-derived comple­
ment protein abnormalities, in particular for renal Emerging therapies
transplant recipients with uncontrolled disease activity Crovalimab is a subcutaneous long-acting C5 inhibitor
despite eculizumab therapy. with sequential monoclonal antibody recycling technology
that binds efficiently to C5. Clinical trials are underway
Management of HSCT-TMA testing its use in pediatric (COMMUTE-p; NCT04958265)
At this time, there are no US Food and Drug Administration and adult (COMMUTE-a; NCT04861259) patients with
(FDA)-approved anti-complement agents for HSCT-TMA, aHUS. As in the case of eculizumab, newer complement
but they are becoming more commonly used. Various anti- blocking agents are being studied in patients with
complement therapies with different mechanisms of paroxysmal nocturnal haemoglobinuria with potential
action, including anti-C5 (eculizumab and conversin) and for use in aHUS. These new agents include pozelimab, a
mannan-binding lectin-associated serine protease-2 fully human monoclonal IgG4P antibody directed
inhibition (MASP-2, narsoplimab), have been evaluated as against C5 that has been shown to be a safe C5 inhibitor
potential therapies for HSCT-TMA (panel 3). with subcutaneous dosing in healthy volunteers
Eculizumab use in HSCT-TMA is mostly limited to (NCT03946748).
small retrospective cohorts.207–212 A prospective study on Several proximal complement protein inhibitors
its use in 64 paediatric patients showed a 64% response targeting C3, factor B, and factor D are being studied in
rate and 66% survival at 1 year.213 Eculizumab can be clinical trials for a variety of complement-mediated
considered a front-line therapy for HSCT-TMA, with diseases.217 Another novel therapeutic agent that has been
titration of dosing based on CH50 levels.211–213 Evidence on described as a potential option for complement-related
the use of ravulizumab for HSCT-TMA is scarce. There is disorders is a synthetic fusion protein, MFHR1, which is
one case report on the use of conversin in a child with composed of the regulatory components of CFH and
HSCT-TMA and a mutation in C5 that conferred CFHR1. MFHR1 modulates multiple levels of the
resistance to eculizumab,214 although the patient in this alternative complement pathway, including decay of the
report died from HSCT-TMA. C3 convertase and inhibition of the C5 convertase.218,219
Narsoplimab is a MASP-2 inhibitor that blocks When available for clinical use, these agents will be
the lectin pathway of complement. A study using narso­ important additions to the treatment options for
plimab in 28 adult patients with HSCT-TMA showed complement-mediated aHUS.
improvement in organ function in 74% (20/27), with a
median overall survival of 274 days (95% CI 103–not Summary
estimable). The drug was well tolerated with no safety HUS is a clinical condition comprised of a triad of non-
concerns.215 The FDA has yet to approve narsoplimab for immune microangiopathic haemolytic anaemia,
the treatment of HSCT-TMA, requesting more data after throm­bo­cyto­penia, and acute kidney injury. The unifying
results from the single-arm, open-label phase 2 trial215 feature of this heterogeneous group of diseases is
were submitted in mid-2021. endothelial damage. Although the classification of HUS is
constantly evolving, one based on aetiology is used
Monitoring response to eculizumab therapy and non- in this Seminar. Disease epidemiology, pathogenesis,
responders clinical presentation and management are discussed. The
The first indication of HUS recovery or response to most common type of HUS, especially in children
eculizumab therapy is normalisation of the platelet count, younger than 5 years, is STEC-HUS, treatment of which is

1734 www.thelancet.com Vol 400 November 12, 2022


Seminar

supportive. Complement-mediated HUS is extremely rare 12 Vishram B, Jenkins C, Greig DR, et al. The emerging importance of
and although it once carried very high morbidity and Shiga toxin-producing Escherichia coli other than serogroup O157 in
England. J Med Microbiol 2021; 70: 001375.
mortality, treatment with anti-complement therapies has 13 Joseph A, Cointe A, Mariani Kurkdjian P, Rafat C, Hertig A.
made end-stage renal disease and death unusual in Shiga toxin-associated hemolytic uremic syndrome: a narrative
high-income countries. Further studies are needed to review. Toxins 2020; 12: 67.
14 Marshall KE, Hexemer A, Seelman SL, et al. Lessons learned from a
determine the optimal frequency and duration of decade of investigations of shiga toxin-producing Escherichia coli
complement blockers, and guidelines for monitoring outbreaks linked to leafy greens, United States and Canada.
optimal complement blockade would be helpful. There Emerg Infect Dis 2020; 26: 2319–28.
15 Bielaszewska M, Mellmann A, Zhang W, et al. Characterisation of
are many other causes of HUS, including those associated the Escherichia coli strain associated with an outbreak of haemolytic
with mutations in non-complement genes and HUS that uraemic syndrome in Germany, 2011: a microbiological study.
develops in association with a primary disease process. Lancet Infect Dis 2011; 11: 671–76.
For many of these rarer presentations of HUS, our 16 Wijnsma KL, Schijvens AM, Rossen JWA, Kooistra-Smid AMDM,
Schreuder MF, van de Kar NCAJ. Unusual severe case of hemolytic
understanding of the underlying patho­ physiology is uremic syndrome due to Shiga toxin 2d-producing E. coli O80:H2.
incomplete and anti-complement is either not indicated Pediatr Nephrol 2017; 32: 1263–68.
or its role in treatment remains to be defined. An urgent 17 Sawyer C, Vishram B, Jenkins C, et al. Epidemiological
investigation of recurrent outbreaks of haemolytic uraemic
need remains to make affordable versions of anti- syndrome caused by Shiga toxin-producing Escherichia coli serotype
complement agents available in low-income countries. O55:H7 in England, 2014–2018. Epidemiol Infect 2021; 149: e108.
18 Ardissino G, Vignati C, Masia C, et al. Bloody diarrhea and shiga toxin-
Contributors
producing Escherichia coli hemolytic uremic syndrome in children:
All authors contributed equally. data from the ItalKid-HUS network. J Pediatr 2021; 237: 34–40.e1.
Declaration of interests 19 Lee MS, Yoon JW, Tesh VL. Editorial: recent advances in
MM declares no conflicts of interest related to this topic but has served as understanding the pathogenesis of shiga toxin-producing Shigella
site principal investigator for use of lumasiran for primary hyperoxaluria and Escherichia coli. Front Cell Infect Microbiol 2020; 10: 620703.
type 1 by Alnylam Pharmaceuticals. SES has received research funding 20 Menge C. Molecular biology of Escherichia coli Shiga toxins’ effects
from Alexion Pharmaceuticals to test pre-clinical complement agents in on mammalian cells. Toxins 2020; 12: 345.
murine models of complement-mediated disease. RJHS directs the 21 Buelli S, Zoja C, Remuzzi G, Morigi M. Complement activation
Molecular Otolaryngology and Renal Research Laboratories contributes to the pathophysiology of Shiga toxin-associated
(Iowa City, IA, USA) where genetic and complement testing for atypical hemolytic uremic syndrome. Microorganisms 2019; 7: 15.
HUS and the TMAs is done. AB declares no competing interests. 22 Westra D, Volokhina EB, van der Molen RG, et al. Serological and
genetic complement alterations in infection-induced and
Acknowledgments complement-mediated hemolytic uremic syndrome. Pediatr Nephrol
We thank Geetika Singh (All India Institute of Medical Sciences, 2017; 32: 297–309.
New Delhi, Delhi, India) for providing pathology slides of thrombotic 23 Noris M, Mescia F, Remuzzi G. STEC-HUS, atypical HUS and TTP
microangiopathy. are all diseases of complement activation. Nat Rev Nephrol 2012;
8: 622–33.
References
24 Netti GS, Santangelo L, Paulucci L, et al. Low C3 serum levels
1 Loirat C, Frémeaux-Bacchi V. Atypical hemolytic uremic syndrome.
predict severe forms of STEC-HUS with neurologic involvement.
Orphanet J Rare Dis 2011; 6: 60.
Front Med 2020; 7: 357.
2 Nester CM, Barbour T, de Cordoba SR, et al. Atypical aHUS: state of
25 Obata F, Tohyama K, Bonev AD, et al. Shiga toxin 2 affects the
the art. Mol Immunol 2015; 67: 31–42.
central nervous system through receptor globotriaosylceramide
3 Noris M, Remuzzi G. Atypical hemolytic-uremic syndrome. localized to neurons. J Infect Dis 2008; 198: 1398–406.
N Engl J Med 2009; 361: 1676–87.
26 Eisenhauer PB, Jacewicz MS, Conn KJ, et al. Escherichia coli Shiga
4 Zheng XL, Vesely SK, Cataland SR, et al. ISTH guidelines for the toxin 1 and TNF-alpha induce cytokine release by human cerebral
diagnosis of thrombotic thrombocytopenic purpura. microvascular endothelial cells. Microb Pathog 2004; 36: 189–96.
J Thromb Haemost 2020; 18: 2486–95.
27 Pifer R, Sperandio V. The interplay between the microbiota and
5 Fakhouri F, Hourmant M, Campistol JM, et al. Terminal enterohemorrhagic Escherichia coli. Microbiol Spectr 2014; 2: doi:
complement inhibitor eculizumab in adult patients with atypical 10.1128/microbiolspec.EHEC-0015-2013.
hemolytic uremic syndrome: a single-arm, open-label trial.
28 Travert B, Dossier A, Jamme M, et al. Shiga toxin-associated
Am J Kidney Dis 2016; 68: 84–93.
hemolytic uremic syndrome in adults, France, 2009–2017.
6 Legendre CM, Licht C, Muus P, et al. Terminal complement Emerg Infect Dis 2021; 27: 1876–85.
inhibitor eculizumab in atypical hemolytic-uremic syndrome.
29 Travert B, Rafat C, Mariani P, et al. Shiga toxin-associated hemolytic
N Engl J Med 2013; 368: 2169–81.
uremic syndrome: specificities of adult patients and implications
7 Greenbaum LA, Fila M, Ardissino G, et al. Eculizumab is a safe and for critical care management. Toxins 2021; 13: 306.
effective treatment in pediatric patients with atypical hemolytic
30 Costigan C, Raftery T, Carroll AG, et al. Neurological involvement in
uremic syndrome. Kidney Int 2016; 89: 701–11.
children with hemolytic uremic syndrome. Eur J Pediatr 2022;
8 Majowicz SE, Scallan E, Jones-Bitton A, et al. Global incidence of 181: 501–12.
human Shiga toxin-producing Escherichia coli infections and deaths:
31 Nathanson S, Kwon T, Elmaleh M, et al. Acute neurological
a systematic review and knowledge synthesis. Foodborne Pathog Dis
involvement in diarrhea-associated hemolytic uremic syndrome.
2014; 11: 447–55.
Clin J Am Soc Nephrol 2010; 5: 1218–28.
9 Alconcher LF, Balestracci A, Coccia PA, et al. Hemolytic uremic
32 Khalid M, Andreoli S. Extrarenal manifestations of the hemolytic
syndrome associated with Shiga toxin-producing Escherichia coli
uremic syndrome associated with Shiga toxin-producing Escherichia
infection in Argentina: update of serotypes and genotypes and their
coli (STEC HUS). Pediatr Nephrol 2019; 34: 2495–507.
relationship with severity of the disease. Pediatr Nephrol 2021;
36: 2811–17. 33 Brown CC, Garcia X, Bhakta RT, Sanders E, Prodhan P. Severe
acute neurologic involvement in children with hemolytic-uremic
10 Tack DM, Kisselburgh HM, Richardson LC, et al. Shiga toxin-
syndrome. Pediatrics 2021; 147: e2020013631.
producing Escherichia coli outbreaks in the United States, 2010–2017.
Microorganisms 2021; 9: 1529. 34 Andreoli S, Bergstein J. Exocrine and endocrine pancreatic
insufficiency and calcinosis after hemolytic uremic syndrome.
11 Valilis E, Ramsey A, Sidiq S, DuPont HL. Non-O157 Shiga toxin-
J Pediatr 1987; 110: 816–17.
producing Escherichia coli–a poorly appreciated enteric pathogen:
systematic review. Int J Infect Dis 2018; 76: 82–87.

www.thelancet.com Vol 400 November 12, 2022 1735


Seminar

35 Suri RS, Clark WF, Barrowman N, et al. Diabetes during diarrhea- 60 Campistol JM, Arias M, Ariceta G, et al. An update for atypical
associated hemolytic uremic syndrome: a systematic review and haemolytic uraemic syndrome: diagnosis and treatment.
meta-analysis. Diabetes Care 2005; 28: 2556–62. A consensus document. Nefrologia 2015; 35: 421–47.
36 Gallo EG, Gianantonio CA. Extrarenal involvement in 61 Thompson RA, Winterborn MH. Hypocomplementaemia due
diarrhoea-associated haemolytic-uraemic syndrome. Pediatr Nephrol to a genetic deficiency of beta 1H globulin. Clin Exp Immunol 1981;
1995; 9: 117–19. 46: 110–9.
37 Constantinescu AR, Bitzan M, Weiss LS, et al. Non-enteropathic 62 Warwicker P, Goodship TH, Donne RL, et al. Genetic studies into
hemolytic uremic syndrome: causes and short-term course. inherited and sporadic hemolytic uremic syndrome. Kidney Int
Am J Kidney Dis 2004; 43: 976–82. 1998; 53: 836–44.
38 Mizusawa Y, Pitcher LA, Burke JR, Falk MC, Mizushima W. Survey 63 George JN, Nester CM. Syndromes of thrombotic microangiopathy.
of haemolytic-uraemic syndrome in Queensland 1979–1995. N Engl J Med 2014; 371: 1847–48.
Med J Aust 1996; 165: 188–91. 64 Thurman JM, Holers VM. The central role of the alternative
39 Copelovitch L, Kaplan BS. Streptococcus pneumoniae-associated complement pathway in human disease. J Immunol 2006;
hemolytic uremic syndrome. Pediatr Nephrol 2008; 23: 1951–56. 176: 1305–10.
40 Cabrera GR, Fortenberry JD, Warshaw BL, Chambliss CR, 65 Ricklin D. Manipulating the mediator: modulation of the alternative
Butler JC, Cooperstone BG. Hemolytic uremic syndrome associated complement pathway C3 convertase in health, disease and therapy.
with invasive Streptococcus pneumoniae infection. Pediatrics 1998; Immunobiology 2012; 217: 1057–66.
101: 699–703. 66 Fremeaux-Bacchi V, Fakhouri F, Garnier A, et al. Genetics and
41 Copelovitch L, Kaplan BS. Streptococcus pneumoniae-associated outcome of atypical hemolytic uremic syndrome: a nationwide
hemolytic uremic syndrome: classification and the emergence of French series comparing children and adults. Clin J Am Soc Nephrol
serotype 19A. Pediatrics 2010; 125: e174–82. 2013; 8: 554–62.
42 Loupiac A, Elayan A, Cailliez M, et al. Diagnosis of Streptococcus 67 Noris M, Caprioli J, Bresin E, et al. Relative role of genetic
pneumoniae-associated hemolytic uremic syndrome. complement abnormalities in sporadic and familial aHUS and their
Pediatr Infect Dis J 2013; 32: 1045–49. impact on clinical phenotype. Clin J Am Soc Nephrol 2010; 5: 1844–59.
43 Makwana A, Sheppard C, Fry NK, Ladhani SN. Pneumococcal- 68 Maga TK, Nishimura CJ, Weaver AE, Frees KL, Smith RJ. Mutations
related hemolytic uremic syndrome in the United Kingdom: national in alternative pathway complement proteins in American patients
surveillance, 2006–2016. Pediatr Infect Dis J 2019; 38: e254–59. with atypical hemolytic uremic syndrome. Hum Mutat 2010;
44 Klein PJ, Bulla M, Newman RA, et al. Thomsen-Friedenreich 31: E1445–60.
antigen in haemolytic-uraemic syndrome. Lancet 1977; 2: 1024–25. 69 Schaefer F, Ardissino G, Ariceta G, et al. Clinical and genetic
45 Crookston KP, Reiner AP, Cooper LJ, Sacher RA, Blajchman MA, predictors of atypical hemolytic uremic syndrome phenotype and
Heddle NM. RBC T activation and hemolysis: implications for outcome. Kidney Int 2018; 94: 408–18.
pediatric transfusion management. Transfusion 2000; 40: 801–12. 70 Bresin E, Rurali E, Caprioli J, et al. Combined complement gene
46 Gilbert RD, Nagra A, Haq MR. Does dysregulated complement mutations in atypical hemolytic uremic syndrome influence clinical
activation contribute to haemolytic uraemic syndrome secondary to phenotype. J Am Soc Nephrol 2013; 24: 475–86.
Streptococcus pneumoniae? Med Hypotheses 2013; 81: 400–03. 71 Valoti E, Alberti M, Tortajada A, et al. A novel atypical hemolytic
47 Szilágyi A, Kiss N, Bereczki C, et al. The role of complement in uremic syndrome-associated hybrid CFHR1/CFH gene encoding
Streptococcus pneumoniae-associated haemolytic uraemic syndrome. a fusion protein that antagonizes factor H-dependent complement
Nephrol Dial Transplant 2013; 28: 2237–45. regulation. J Am Soc Nephrol 2015; 26: 209–19.
48 Spinale JM, Ruebner RL, Kaplan BS, Copelovitch L. Update on 72 Krisinger MJ, Goebeler V, Lu Z, et al. Thrombin generates
Streptococcus pneumoniae associated hemolytic uremic syndrome. previously unidentified C5 products that support the terminal
Curr Opin Pediatr 2013; 25: 203–08. complement activation pathway. Blood 2012; 120: 1717–25.
49 Gómez Delgado I, Corvillo F, Nozal P, et al. Complement genetic 73 Ferreira VP, Pangburn MK, Cortés C. Complement control protein
variants and FH desialylation in S. pneumoniae-haemolytic uraemic factor H: the good, the bad, and the inadequate. Mol Immunol 2010;
syndrome. Front Immunol 2021; 12: 641656. 47: 2187–97.
50 Ariceta G, Besbas N, Johnson S, et al. Guideline for the 74 Eyler SJ, Meyer NC, Zhang Y, Xiao X, Nester CM, Smith RJ. A novel
investigation and initial therapy of diarrhea-negative hemolytic hybrid CFHR1/CFH gene causes atypical hemolytic uremic
uremic syndrome. Pediatr Nephrol 2009; 24: 687–96. syndrome. Pediatr Nephrol 2013; 28: 2221–25.
51 Brandt J, Wong C, Mihm S, et al. Invasive pneumococcal disease 75 Caprioli J, Noris M, Brioschi S, et al. Genetics of HUS: the impact
and hemolytic uremic syndrome. Pediatrics 2002; 110: 371–76. of MCP, CFH, and IF mutations on clinical presentation, response
52 Huang DT, Chi H, Lee HC, Chiu NC, Huang FY. T-antigen to treatment, and outcome. Blood 2006; 108: 1267–79.
activation for prediction of pneumococcus-induced hemolytic 76 Delvaeye M, Noris M, De Vriese A, et al. Thrombomodulin
uremic syndrome and hemolytic anemia. Pediatr Infect Dis J 2006; mutations in atypical hemolytic-uremic syndrome. N Engl J Med
25: 608–10. 2009; 361: 345–57.
53 Waters AM, Kerecuk L, Luk D, et al. Hemolytic uremic syndrome 77 Sellier-Leclerc AL, Fremeaux-Bacchi V, Dragon-Durey MA, et al.
associated with invasive pneumococcal disease: the United Differential impact of complement mutations on clinical
Kingdom experience. J Pediatr 2007; 151: 140–44. characteristics in atypical hemolytic uremic syndrome.
54 Banerjee R, Hersh AL, Newland J, et al. Streptococcus pneumoniae- J Am Soc Nephrol 2007; 18: 2392–400.
associated hemolytic uremic syndrome among children in North 78 El Karoui K, Boudhabhay I, Petitprez F, et al. Impact of
America. Pediatr Infect Dis J 2011; 30: 736–39. hypertensive emergency and rare complement variants on the
55 Palma LMP, Sridharan M, Sethi S. Complement in secondary presentation and outcome of atypical hemolytic uremic syndrome.
thrombotic microangiopathy. Kidney Int Rep 2021; 6: 11–23. Haematologica 2019; 104: 2501–11.
56 Noris M, Benigni A, Remuzzi G. The case of complement 79 Fidan K, Göknar N, Gülhan B, et al. Extra-renal manifestations of
activation in COVID-19 multiorgan impact. Kidney Int 2020; atypical hemolytic uremic syndrome in children. Pediatr Nephrol
98: 314–22. 2018; 33: 1395–403.
57 Zipfel PF, Mache C, Müller D, et al. DEAP-HUS: deficiency of 80 Formeck C, Swiatecka-Urban A. Extra-renal manifestations of atypical
CFHR plasma proteins and autoantibody-positive form of hemolytic hemolytic uremic syndrome. Pediatr Nephrol 2019; 34: 1337–48.
uremic syndrome. Pediatr Nephrol 2010; 25: 2009–19. 81 Johnson S, Stojanovic J, Ariceta G, et al. An audit analysis of a
58 Franchini M. Atypical hemolytic uremic syndrome: from diagnosis guideline for the investigation and initial therapy of diarrhea
to treatment. Clin Chem Lab Med 2015; 53: 1679–88. negative (atypical) hemolytic uremic syndrome. Pediatr Nephrol
2014; 29: 1967–78.
59 Noris M, Bucchioni S, Galbusera M, et al. Complement factor
H mutation in familial thrombotic thrombocytopenic purpura with 82 Vilalta R, Lara E, Madrid A, Chocron S, Muñoz M, Casquero A,
ADAMTS13 deficiency and renal involvement. J Am Soc Nephrol Nieto J. Long-term eculizumab improves clinical outcomes in atypical
2005; 16: 1177–83. hemolytic uremic syndrome. Pediatr Nephrol 2012; 27: 2323–26.

1736 www.thelancet.com Vol 400 November 12, 2022


Seminar

83 Kaplan BS, Garcia CD, Chesney RW, Segar WE, Giugno K, Chem R. 106 Rosenblatt DS, Aspler AL, Shevell MI, Pletcher BA, Fenton WA,
Peripheral gangrene complicating idiopathic and recessive Seashore MR. Clinical heterogeneity and prognosis in combined
hemolytic uremic syndromes. Pediatr Nephrol 2000; 14: 985–89. methylmalonic aciduria and homocystinuria (cblC).
84 Ozel A, Calişkan U, Gücer S. Peripheral gangrene complicating J Inherit Metab Dis 1997; 20: 528–38.
hemolytic uremic syndrome in a child. Pediatr Nephrol 2003; 107 Lerner-Ellis JP, Anastasio N, Liu J, et al. Spectrum of mutations in
18: 465–67. MMACHC, allelic expression, and evidence for genotype-phenotype
85 Ardissino G, Tel F, Testa S, et al. Skin involvement in atypical correlations. Hum Mutat 2009; 30: 1072–81.
hemolytic uremic syndrome. Am J Kidney Dis 2014; 63: 652–55. 108 Tsai AC, Morel CF, Scharer G, et al. Late-onset combined
86 Besbas N, Gulhan B, Soylemezoglu O, et al. Turkish pediatric homocystinuria and methylmalonic aciduria (cblC) and
atypical hemolytic uremic syndrome registry: initial analysis of neuropsychiatric disturbance. Am J Med Genet A 2007;
146 patients. BMC Nephrol 2017; 18: 6. 143A: 2430–34.
87 Brocklebank V, Kumar G, Howie AJ, et al. Long-term outcomes and 109 Thauvin-Robinet C, Roze E, Couvreur G, et al. The adolescent
response to treatment in diacylglycerol kinase epsilon nephropathy. and adult form of cobalamin C disease: clinical and molecular
Kidney Int 2020; 97: 1260–74. spectrum. J Neurol Neurosurg Psychiatry 2008; 79: 725–28.
88 Lemaire M, Frémeaux-Bacchi V, Schaefer F, et al. Recessive 110 Sloan JL, Carrillo N, Adams D, Venditti CP. Disorders of
mutations in DGKE cause atypical hemolytic-uremic syndrome. intracellular cobalamin metabolism. In: Adam MP, Mirzaa GM,
Nat Genet 2013; 45: 531–36. Pagon RA, et al, eds. GeneReviews® [Internet]. Seattle, WA:
89 Ozaltin F, Li B, Rauhauser A, et al. DGKE variants cause a University of Washington, 2018.
glomerular microangiopathy that mimics membranoproliferative 111 Craig SA. Betaine in human nutrition. Am J Clin Nutr 2004;
GN. J Am Soc Nephrol 2013; 24: 377–84. 80: 539–49.
90 Liu D, Ding Q, Dai DF, et al. Loss of diacylglycerol kinase ε causes 112 Vidal-Alaball J, Butler CC, Cannings-John R, et al. Oral vitamin B12
thrombotic microangiopathy by impairing endothelial VEGFA versus intramuscular vitamin B12 for vitamin B12 deficiency.
signaling. JCI Insight 2021; 6: e146959. Cochrane Database Syst Rev 2005; 3: CD004655.
91 Shulga YV, Topham MK, Epand RM. Regulation and functions of 113 Ardissino G, Perrone M, Tel F, et al. Late onset cobalamin disorder
diacylglycerol kinases. Chem Rev 2011; 111: 6186–208. and hemolytic uremic syndrome: a rare cause of nephrotic
92 Tang W, Bunting M, Zimmerman GA, McIntyre TM, Prescott SM. syndrome. Case Rep Pediatr 2017; 2017: 2794060.
Molecular cloning of a novel human diacylglycerol kinase highly 114 Ponticelli C, Banfi G. Thrombotic microangiopathy after kidney
selective for arachidonate-containing substrates. J Biol Chem 1996; transplantation. Transpl Int 2006; 19: 789–94.
271: 10237–41. 115 Jodele S, Davies SM, Lane A, et al. Diagnostic and risk criteria for
93 Rodriguez de Turco EB, Tang W, Topham MK, et al. Diacylglycerol HSCT-associated thrombotic microangiopathy: a study in children
kinase epsilon regulates seizure susceptibility and long-term and young adults. Blood 2014; 124: 645–53.
potentiation through arachidonoyl-inositol lipid signaling. 116 Gavriilaki E, Sakellari I, Batsis I, et al. Transplant-associated
Proc Natl Acad Sci USA 2001; 98: 4740–45. thrombotic microangiopathy: incidence, prognostic factors,
94 Milne SB, Ivanova PT, Armstrong MD, et al. Dramatic differences morbidity, and mortality in allogeneic hematopoietic cell
in the roles in lipid metabolism of two isoforms of diacylglycerol transplantation. Clin Transplant 2018; 32: e13371.
kinase. Biochemistry 2008; 47: 9372–79. 117 Schoettler M, Lehmann L, Li A, Ma C, Duncan C. Thrombotic
95 Sánchez Chinchilla D, Pinto S, Hoppe B, et al. Complement microangiopathy following pediatric autologous hematopoietic cell
mutations in diacylglycerol kinase-ε-associated atypical hemolytic transplantation: a report of significant end-organ dysfunction in
uremic syndrome. Clin J Am Soc Nephrol 2014; 9: 1611–19. eculizumab-treated survivors. Biol Blood Marrow Transplant 2019;
96 Denys P Malvaux P, Van Den Berghe H, Tanghe W, Proesmans W. 25: e163–68.
Association of an anatomo-pathological syndrome of male 118 Tolbert VP, Dvorak CC, Golden C, et al. Risk factors for transplant-
pseudohermaphroditism, Wilms’ tumor, parenchymatous associated thrombotic microangiopathy after autologous
nephropathyand XX/XY mosaicism. Arch Fr Pediatr 1967; hematopoietic cell transplant in high-risk neuroblastoma.
24: 729–39. Biol Blood Marrow Transplant 2019; 25: 2031–39.
97 Manivel JC, Sibley RK, Dehner LP. Complete and incomplete 119 Epperla N, Li A, Logan B, et al. Incidence, risk factors for and
Drash syndrome: a clinicopathologic study of five cases of a outcomes of transplant-associated thrombotic microangiopathy.
dysontogenetic-neoplastic complex. Hum Pathol 1987; 18: 80–89. Br J Haematol 2020; 189: 1171–81.
98 Sherbotie JR, van Heyningen V, Axton R, Williamson K, Finn LS, 120 Caires RA, Marques ID, Repizo LP, et al. De novo thrombotic
Kaplan BS. Hemolytic uremic syndrome associated with Denys- microangiopathy after kidney transplantation: clinical features,
Drash syndrome. Pediatr Nephrol 2000; 14: 1092–97. treatment, and long-term patient and graft survival. Transplant Proc
99 Alge JL, Wenderfer SE, Hicks J, et al. Hemolytic uremic syndrome 2012; 44: 2388–90.
as the presenting manifestation of WT1 mutation and Denys-Drash 121 Jodele S, Laskin BL, Dandoy CE, et al. A new paradigm: diagnosis
syndrome: a case report. BMC Nephrol 2017; 18: 243. and management of HSCT-associated thrombotic
100 Cheng C, Chen L, Wen S, Lin Z, Jiang X. Case report: Denys– microangiopathy as multi-system endothelial injury. Blood Rev
Drash syndrome with WT1 causative variant presenting as 2015; 29: 191–204.
atypical hemolytic uremic syndrome. Front Pediatr 2020; 122 Kobayashi T, Wada H, Usui M, et al. Decreased ADAMTS13 levels
8: 605889. in patients after living donor liver transplantation. Thromb Res 2009;
101 Walsh PR, Johnson S, Brocklebank V, Salvatore J, Christian M, 124: 541–45.
Kavanagh D. Glucose-6-phosphate dehydrogenase deficiency 123 Pereboom IT, Adelmeijer J, van Leeuwen Y, Hendriks HG, Porte RJ,
mimicking atypical hemolytic uremic syndrome. Am J Kidney Dis Lisman T. Development of a severe von Willebrand factor/
2018; 71: 287–90. ADAMTS13 dysbalance during orthotopic liver transplantation.
102 Luzzatto L, Ally M, Notaro R. Glucose-6-phosphate dehydrogenase Am J Transplant 2009; 9: 1189–96.
deficiency. Blood 2020; 136: 1225–40. 124 Young JA, Pallas CR, Knovich MA. Transplant-associated
103 Mullikin D, Pillai N, Sanchez R, et al. Megaloblastic anemia thrombotic microangiopathy: theoretical considerations and a
progressing to severe thrombotic microangiopathy in patients with practical approach to an unrefined diagnosis.
disordered vitamin B. J Pediatr 2018; 202: 315–19.e2. Bone Marrow Transplant 2021; 56: 1805–17.
104 Bayer G, von Tokarski F, Thoreau B, et al. Etiology and outcomes of 125 Verbiest A, Pirenne J, Dierickx D. De novo thrombotic
thrombotic microangiopathies. Clin J Am Soc Nephrol 2019; microangiopathy after non-renal solid organ transplantation.
14: 557–66. Blood Rev 2014; 28: 269–79.
105 Lemoine M, François A, Grangé S, et al. Cobalamin C deficiency 126 Dvorak CC, Higham C, Shimano KA. Transplant-associated
induces a typical histopathological pattern of renal arteriolar and thrombotic microangiopathy in pediatric hematopoietic cell
glomerular thrombotic microangiopathy. Kidney Int Rep 2018; transplant recipients: a practical approach to diagnosis and
3: 1153–62. management. Front Pediatr 2019; 7: 133.

www.thelancet.com Vol 400 November 12, 2022 1737


Seminar

127 Wirtschafter E, VanBeek C, Linhares Y. Bone marrow transplant- 148 Cavero T, Rabasco C, López A, et al. Eculizumab in secondary
associated thrombotic microangiopathy without peripheral blood atypical haemolytic uraemic syndrome. Nephrol Dial Transplant
schistocytes: a case report and review of the literature. 2017; 32: 466–74.
Exp Hematol Oncol 2018; 7: 14. 149 Tinti MG, Carnevale V, Inglese M, et al. Eculizumab in refractory
128 Jodele S, Hirsch R, Laskin B, Davies S, Witte D, Chima R. catastrophic antiphospholipid syndrome: a case report and
Pulmonary arterial hypertension in pediatric patients with systematic review of the literature. Clin Exp Med 2019; 19: 281–88.
hematopoietic stem cell transplant-associated thrombotic 150 Devresse A, Aydin S, Le Quintrec M, et al. Complement activation
microangiopathy. Biol Blood Marrow Transplant 2013; 19: 202–07. and effect of eculizumab in scleroderma renal crisis. Medicine 2016;
129 Sartain S, Shubert S, Wu MF, et al. Therapeutic plasma exchange does 95: e4459.
not improve renal function in hematopoietic stem cell transplantation- 151 Thomas MR, Scully M. Microangiopathy in cancer: causes,
associated thrombotic microangiopathy: an institutional experience. consequences, and management. Cancer Treat Res 2019;
Biol Blood Marrow Transplant 2019; 25: 157–62. 179: 151–58.
130 Ho VT, Cutler C, Carter S, et al. Blood and marrow transplant 152 Huber-Lang M, Sarma JV, Zetoune FS, et al. Generation of C5a in
clinical trials network toxicity committee consensus summary: the absence of C3: a new complement activation pathway. Nat Med
thrombotic microangiopathy after hematopoietic stem cell 2006; 12: 682–87.
transplantation. Biol Blood Marrow Transplant 2005; 11: 571–75. 153 Brain MC, Azzopardi JG, Baker LR, Pineo GF, Roberts PD,
131 Ruutu T, Barosi G, Benjamin RJ, et al. Diagnostic criteria for Dacie JV. Microangiopathic haemolytic anaemia and mucin-
hematopoietic stem cell transplant-associated microangiopathy: forming adenocarcinoma. Br J Haematol 1970; 18: 183–93.
results of a consensus process by an International Working Group. 154 Lohrmann HP, Adam W, Heymer B, Kubanek B. Microangiopathic
Haematologica 2007; 92: 95–100. hemolytic anemia in metastatic carcinoma. Report of eight cases.
132 Cho BS, Yahng SA, Lee SE, et al. Validation of recently proposed Ann Intern Med 1973; 79: 368–75.
consensus criteria for thrombotic microangiopathy after allogeneic 155 Antman KH, Skarin AT, Mayer RJ, Hargreaves HK, Canellos GP.
hematopoietic stem-cell transplantation. Transplantation 2010; Microangiopathic hemolytic anemia and cancer: a review. Medicine
90: 918–26. 1979; 58: 377–84.
133 Jodele S, Dandoy CE, Myers KC, et al. New approaches in the 156 Coppola A, Davi G, De Stefano V, Mancini FP, Cerbone AM,
diagnosis, pathophysiology, and treatment of pediatric Di Minno G. Homocysteine, coagulation, platelet function,
hematopoietic stem cell transplantation-associated thrombotic and thrombosis. Semin Thromb Hemost 2000; 26: 243–54.
microangiopathy. Transfus Apher Sci 2016; 54: 181–90.
157 Ishida Y, Yamashita K, Sasaki H, et al. Activation of complement
134 Fujiwara H, Maeda Y, Sando Y, et al. Treatment of thrombotic system in adult T-cell leukemia (ATL) occurs mainly through lectin
microangiopathy after hematopoietic stem cell transplantation with pathway: a serum proteomic approach using mass spectrometry.
recombinant human soluble thrombomodulin. Transfusion 2016; Cancer Lett 2008; 271: 167–77.
56: 886–92.
158 Weitz IC. Thrombotic microangiopathy in cancer.
135 Goodship THJ, Pinto F, Weston-Davies WH, et al. Use of the Semin Thromb Hemost 2019; 45: 348–53.
complement inhibitor coversin to treat HSCT-associated TMA.
159 Fakhouri F, Roumenina L, Provot F, et al. Pregnancy-associated
Blood Adv 2017; 1: 1254–58.
hemolytic uremic syndrome revisited in the era of complement
136 Yeates L, Slatter MA, Bonanomi S, et al. Use of defibrotide to treat gene mutations. J Am Soc Nephrol 2010; 21: 859–67.
transplant-associated thrombotic microangiopathy: a retrospective
160 Dashe JS, Ramin SM, Cunningham FG. The long-term
study of the Paediatric Diseases and Inborn Errors Working Parties
consequences of thrombotic microangiopathy (thrombotic
of the European Society of Blood and Marrow Transplantation.
thrombocytopenic purpura and hemolytic uremic syndrome) in
Bone Marrow Transplant 2017; 52: 762–64.
pregnancy. Obstet Gynecol 1998; 91: 662–68.
137 Wolff D, Wilhelm S, Hahn J, et al. Replacement of calcineurin
161 Fakhouri F, Vercel C, Frémeaux-Bacchi V. Obstetric nephrology:
inhibitors with daclizumab in patients with transplantation-
AKI and thrombotic microangiopathies in pregnancy.
associated microangiopathy or renal insufficiency associated with
Clin J Am Soc Nephrol 2012; 7: 2100–06.
graft-versus-host disease. Bone Marrow Transplant 2006; 38: 445–51.
162 Fakhouri F, Scully M, Provôt F, et al. Management of thrombotic
138 Togashi J, Sugawara Y, Akamatsu N, et al. A single-center
microangiopathy in pregnancy and postpartum: report from an
experience of the use of interleukin-2 receptor antagonists for
international working group. Blood 2020; 136: 2103–17.
various situations in liver transplant recipients. Transplant Proc
2014; 46: 739–43. 163 Nikuei P, Rajaei M, Roozbeh N, et al. Diagnostic accuracy of
sFlt1/PlGF ratio as a marker for preeclampsia. BMC Pregnancy
139 Wilson CH, Brown AL, White SA, Goodship TH, Sheerin NS,
Childbirth 2020; 20: 80.
Manas DM. Successful treatment of de novo posttransplant
thrombotic microangiopathy with eculizumab. Transplantation 2011; 164 Amari Chinchilla K, Vijayan M, Taveras Garcia B, Jim B.
92: e42–43. Complement-mediated disorders in pregnancy.
Adv Chronic Kidney Dis 2020; 27: 155–64.
140 Cremer A, Amraoui F, Lip GY, et al. From malignant hypertension
to hypertension-MOD: a modern definition for an old but still 165 Kumar D, King M, Jim B, Acharya A. Recurrent case of pregnancy-
dangerous emergency. J Hum Hypertens 2016; 30: 463–66. induced atypical haemolytic uremic syndrome (P-aHUS).
BMJ Case Rep 2019; 12: bcr-2018-226571.
141 Timmermans SAME, Wérion A, Damoiseaux JGMC, Morelle J,
Reutelingsperger CP, van Paassen P. Diagnostic and risk factors for 166 Chatzikonstantinou T, Gavriilaki M, Anagnostopoulos A,
complement defects in hypertensive emergency and thrombotic Gavriilaki E. An update in drug-induced thrombotic
microangiopathy. Hypertension 2020; 75: 422–30. microangiopathy. Front Med 2020; 7: 212.
142 Babar F, Cohen SD. Thrombotic microangiopathies with 167 Zakarija A, Bennett C. Drug-induced thrombotic microangiopathy.
rheumatologic involvement. Rheum Dis Clin North Am 2018; Semin Thromb Hemost 2005; 31: 681–90.
44: 635–49. 168 Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and
143 Thurman JM, Frazer-Abel A, Holers VM. The evolving landscape renal thrombotic microangiopathy. N Engl J Med 2008; 358: 1129–36.
for complement therapeutics in rheumatic and autoimmune 169 Page EE, Little DJ, Vesely SK, George JN. Quinine-induced
diseases. Arthritis Rheumatol 2017; 69: 2102–13. thrombotic microangiopathy: a report of 19 patients.
144 Noris M, Remuzzi G. Genetics of immune-mediated glomerular Am J Kidney Dis 2017; 70: 686–95.
diseases: focus on complement. Semin Nephrol 2017; 37: 447–63. 170 Saleem R, Reese JA, George JN. Drug-induced thrombotic
145 Turrent-Carriles A, Herrera-Félix JP, Amigo MC. Renal involvement microangiopathy: an updated systematic review, 2014–2018.
in antiphospholipid syndrome. Front Immunol 2018; 9: 1008. Am J Hematol 2018; 93: E241–43.
146 Fayyaz A, Igoe A, Kurien BT, et al. Haematological manifestations 171 Al-Nouri ZL, Reese JA, Terrell DR, Vesely SK, George JN.
of lupus. Lupus Sci Med 2015; 2: e000078. Drug-induced thrombotic microangiopathy: a systematic review of
published reports. Blood 2015; 125: 616–18.
147 Li J, Jiang JJ, Wang CY, et al. Clinical features and prognosis of
patients with thrombotic thrombocytopenic purpura associated with 172 Campistol JM, Arias M, Ariceta G, et al. An update for atypical
systemic lupus erythematosus: a review of 25 cases. Ital J Pediatr haemolytic uraemic syndrome: diagnosis and treatment.
2019; 45: 55. A consensus document. Nefrologia 2013; 33: 27–45.

1738 www.thelancet.com Vol 400 November 12, 2022


Seminar

173 Azoulay E, Knoebl P, Garnacho-Montero J, et al. Expert 194 Wijnsma KL, Duineveld C, Wetzels JFM, van de Kar NCAJ.
statements on the standard of care in critically ill adult patients Eculizumab in atypical hemolytic uremic syndrome: strategies
with atypical hemolytic uremic syndrome. Chest 2017; toward restrictive use. Pediatr Nephrol 2019; 34: 2261–77.
152: 424–34. 195 Merrill SA, Brittingham ZD, Yuan X, Moliterno AR, Sperati CJ,
174 Grisaru S, Xie J, Samuel S, et al. Associations between hydration Brodsky RA. Eculizumab cessation in atypical hemolytic uremic
status, intravenous fluid administration, and outcomes of patients syndrome. Blood 2017; 130: 368–72.
infected with Shiga toxin-producing Escherichia coli: a systematic 196 Fakhouri F, Fila M, Hummel A, et al. Eculizumab discontinuation
review and meta-analysis. JAMA Pediatr 2017; 171: 68–76. in children and adults with atypical hemolytic-uremic syndrome:
175 Freedman SB, Xie J, Neufeld MS, et al. Shiga toxin-producing a prospective multicenter study. Blood 2021; 137: 2438–49.
Escherichia coli infection, antibiotics, and risk of developing hemolytic 197 Cugno M, Gualtierotti R, Possenti I, et al. Complement functional
uremic syndrome: a meta-analysis. Clin Infect Dis 2016; 62: 1251–58. tests for monitoring eculizumab treatment in patients with atypical
176 Kakoullis L, Papachristodoulou E, Chra P, Panos G. Shiga toxin- hemolytic uremic syndrome. J Thromb Haemost 2014; 12: 1440–48.
induced haemolytic uraemic syndrome and the role of antibiotics: 198 Ardissino G, Tel F, Sgarbanti M, et al. Complement functional tests
a global overview. J Infect 2019; 79: 75–94. for monitoring eculizumab treatment in patients with atypical
177 Tarr PI, Freedman SB. Why antibiotics should not be used to treat hemolytic uremic syndrome: an update. Pediatr Nephrol
Shiga toxin-producing Escherichia coli infections. 2018; 33: 457–61.
Curr Opin Gastroenterol 2022; 38: 30–38. 199 Gatault P, Brachet G, Ternant D, et al. Therapeutic drug monitoring
178 Schwartz J, Padmanabhan A, Aqui N, et al. Guidelines on the use of of eculizumab: rationale for an individualized dosing schedule.
therapeutic apheresis in clinical practice-evidence-based approach MAbs 2015; 7: 1205–11.
from the writing committee of the American Society for Apheresis: 200 Volokhina E, Wijnsma K, van der Molen R, et al. Eculizumab
the seventh special issue. J Clin Apher 2016; 31: 149–62. dosing regimen in atypical HUS: possibilities for individualized
179 Kielstein JT, Beutel G, Fleig S, et al. Best supportive care and treatment. Clin Pharmacol Ther 2017; 102: 671–78.
therapeutic plasma exchange with or without eculizumab in Shiga- 201 Padmanabhan A, Connelly-Smith L, Aqui N, et al. Guidelines on
toxin-producing E. coli O104:H4 induced haemolytic-uraemic the use of therapeutic apheresis in clinical practice – evidence-
syndrome: an analysis of the German STEC-HUS registry. based approach from the Writing Committee of the American
Nephrol Dial Transplant 2012; 27: 3807–15. Society for Apheresis: the eighth special issue. J Clin Apher 2019;
180 Lapeyraque AL, Malina M, Fremeaux-Bacchi V, et al. Eculizumab in 34: 171–354.
severe Shiga-toxin-associated HUS. N Engl J Med 2011; 364: 2561–63. 202 Igarashi T, Ito S, Sako M, et al. Guidelines for the management and
181 Mahat U, Matar RB, Rotz SJ. Use of complement monoclonal investigation of hemolytic uremic syndrome. Clin Exp Nephrol 2014;
antibody eculizumab in Shiga toxin producing Escherichia coli 18: 525–57.
associated hemolytic uremic syndrome: a review of current 203 Zuber J, Frimat M, Caillard S, et al. Use of highly individualized
evidence. Pediatr Blood Cancer 2019; 66: e27913. complement blockade has revolutionized clinical outcomes after
182 Michael M, Elliott EJ, Ridley GF, Hodson EM, Craig JC. Interventions kidney transplantation and renal epidemiology of atypical hemolytic
for haemolytic uraemic syndrome and thrombotic thrombocytopenic uremic syndrome. J Am Soc Nephrol 2019; 30: 2449–63.
purpura. Cochrane Database Syst Rev 2009; 2009: CD003595. 204 Goodship TH, Cook HT, Fakhouri F, et al. Atypical hemolytic
183 Hopkins CK, Yuan S, Lu Q, Ziman A, Goldfinger D. A severe case uremic syndrome and C3 glomerulopathy: conclusions from
of atypical hemolytic uremic syndrome associated with a “Kidney Disease: Improving Global Outcomes” (KDIGO)
pneumococcal infection and T activation treated successfully with controversies conference. Kidney Int 2017; 91: 539–51.
plasma exchange. Transfusion 2008; 48: 2448–52. 205 Siedlecki AM, Isbel N, Vande Walle J, James Eggleston J, Cohen DJ,
184 Weintraub L, Ahluwalia M, Dogra S, Uehlinger J, Skversky A, Global aHUS Registry. Eculizumab use for kidney transplantation
Vasovic L. Management of streptococcal pneumoniae-induced in patients with a diagnosis of atypical hemolytic uremic syndrome.
hemolytic uremic syndrome: a case report. Clin Nephrol Case Stud Kidney Int Rep 2019; 4: 434–46.
2014; 2: 9–17. 206 Kurup M, Mandelbrot D, Garg N, Singh T. Living related donor
185 Rother RP, Rollins SA, Mojcik CF, Brodsky RA, Bell L. Discovery kidney transplantation in atypical HUS: when should it be
and development of the complement inhibitor eculizumab for the considered? Kidney 2021; 360: 524–27.
treatment of paroxysmal nocturnal hemoglobinuria. Nat Biotechnol 207 de Fontbrune FS, Galambrun C, Sirvent A, et al. Use of eculizumab
2007; 25: 1256–64. in patients with allogeneic stem cell transplant-associated
186 Licht C, Greenbaum LA, Muus P, et al. Efficacy and safety of thrombotic microangiopathy: a study from the SFGM-TC.
eculizumab in atypical hemolytic uremic syndrome from 2-year Transplantation 2015; 99: 1953–59.
extensions of phase 2 studies. Kidney Int 2015; 87: 1061–73. 208 Vasu S, Wu H, Satoskar A, et al. Eculizumab therapy in adults with
187 Menne J, Delmas Y, Fakhouri F, et al. Outcomes in patients with allogeneic hematopoietic cell transplant-associated thrombotic
atypical hemolytic uremic syndrome treated with eculizumab in a microangiopathy. Bone Marrow Transplant 2016; 51: 1241–44.
long-term observational study. BMC Nephrol 2019; 20: 125. 209 Rudoni J, Jan A, Hosing C, Aung F, Yeh J. Eculizumab for transplant-
188 Rondeau E, Scully M, Ariceta G, et al. The long-acting C5 inhibitor, associated thrombotic microangiopathy in adult allogeneic stem cell
ravulizumab, is effective and safe in adult patients with atypical transplant recipients. Eur J Haematol 2018; 101: 389–98.
hemolytic uremic syndrome naïve to complement inhibitor 210 Bohl SR, Kuchenbauer F, von Harsdorf S, et al. Thrombotic
treatment. Kidney Int 2020; 97: 1287–96. microangiopathy after allogeneic stem cell transplantation:
189 Tanaka K, Adams B, Aris AM, et al. The long-acting C5 inhibitor, a comparison of eculizumab therapy and conventional therapy.
ravulizumab, is efficacious and safe in pediatric patients with Biol Blood Marrow Transplant 2017; 23: 2172–77.
atypical hemolytic uremic syndrome previously treated with 211 Jodele S, Dandoy CE, Myers KC, et al. New approaches in the
eculizumab. Pediatr Nephrol 2021; 36: 889–98. diagnosis, pathophysiology, and treatment of pediatric
190 Hayes W, Tschumi S, Ling SC, Feber J, Kirschfink M, Licht C. hematopoietic stem cell transplantation-associated thrombotic
Eculizumab hepatotoxicity in pediatric aHUS. Pediatr Nephrol 2015; microangiopathy. Transfus Apher Sci 2016; 54: 181–90.
30: 775–81. 212 Jodele S, Fukuda T, Vinks A, et al. Eculizumab therapy in children
191 Oruc A, Ayar Y, Vuruskan BA, et al. Hepatotoxicity associated with with severe hematopoietic stem cell transplantation-associated
eculizumab in a patient with atypical hemolytic uremic syndrome. thrombotic microangiopathy. Biol Blood Marrow Transplant 2014;
Nefrologia 2018; 38: 448–50. 20: 518–25.
192 Ardissino G, Testa S, Possenti I, et al. Discontinuation of 213 Jodele S, Dandoy CE, Lane A, et al. Complement blockade for TA-
eculizumab maintenance treatment for atypical hemolytic uremic TMA: lessons learned from a large pediatric cohort treated with
syndrome: a report of 10 cases. Am J Kidney Dis 2014; 64: 633–7. eculizumab. Blood 2020; 135: 1049–57.
193 Fakhouri F, Fila M, Provôt F, et al. Pathogenic variants in 214 Goodship THJ, Pinto F, Weston-Davies WH, et al. Use of the
complement genes and risk of atypical hemolytic uremic syndrome complement inhibitor coversin to treat HSCT-associated TMA.
relapse after eculizumab discontinuation. Clin J Am Soc Nephrol Blood Adv 2017; 1: 1254–58.
2017; 12: 50–59.

www.thelancet.com Vol 400 November 12, 2022 1739


Seminar

215 Khaled SK, Claes K, Goh YT, et al. Narsoplimab, a mannan-binding 218 Top O, Parsons J, Bohlender LL, et al. Recombinant production of
lectin-associated serine protease-2 inhibitor, for the treatment of MFHR1, a novel synthetic multitarget complement inhibitor,
adult hematopoietic stem-cell transplantation-associated thrombotic in moss bioreactors. Front Plant Sci 2019; 10: 260.
microangiopathy. J Clin Oncol 2022; 40: 2447–57. 219 Michelfelder S, Fischer F, Wäldin A, et al. The MFHR1 fusion
216 Nishimura J, Yamamoto M, Hayashi S, et al. Genetic variants in C5 protein is a novel synthetic multitarget complement inhibitor with
and poor response to eculizumab. N Engl J Med 2014; 370: 632–39. therapeutic potential. J Am Soc Nephrol 2018; 29: 1141–53.
217 Griffin M, Kelly R, Pike A. A review of the treatment landscape in
paroxysmal nocturnal haemoglobinuria: where are we now and Copyright © 2022 Elsevier Ltd. All rights reserved.
where are we going? Ther Adv Rare Dis 2020; 1: 1–12.

1740 www.thelancet.com Vol 400 November 12, 2022

You might also like