Foreword From Editor-in-Chief Shaji Kumar, MD

Hematologic disorders encompass myriad conditions ranging from the mundane to the exotic, often broadly grouped into benign or malignant — though the distinction has increasingly become blurred with the recognition of premalignant conditions that straddle the divide between benign and malignant blood disorders.

For a long time, the malignant conditions have enjoyed the limelight for a variety of reasons, not the least being the fear associated with the “C” word and the perceived mortality associated with many of the disorders. At the same time, the rapid strides that have been made in understanding the disease mechanisms and increasing treatment options for malignant disorders have kept them in the spotlight, attracting further interest and focus. However, what is often not perceived is the tremendous impact of non-malignant blood disorders on patients due to a combination of the overall burden, the chronic symptomatology associated with the disease as well as its treatment, and the economic burden resulting from these chronic conditions.

While these disorders are often thought of as “benign,” many of these conditions are anything but. For example, many of the hemoglobinopathies, while not fatal, significantly impact quality of life, present tremendous economic burden to society, cause much suffering for those affected, and most importantly reduce the life span of the affected individuals. Disorders of coagulation can lead to significant morbidity through their clinical manifestations and their sequelae, whether they are excessive bleeding or clotting. With the debate over what to call these disorders (i.e., classical, nonmalignant, or benign), as highlighted by the letter to the editor from Dr. Steven Lentz and the response from Dr. Robert Brodsky, we want to introduce a new series titled “Not So Benign” to draw the attention of the hematology community to these less appreciated disorders and increase awareness of the biology as well as treatment approaches. We start the series off with an article from Drs. Surbhi Shah and Ronald Go, who provide a detailed overview of cold agglutinin disease, an uncommon but serious disorder that can be a challenging diagnosis, and the potential treatment options. Thankfully, therapeutic advances have finally caught up with these disorders and have seen tremendous progress over the past few years, such as gene therapy for sickle cell disease. We anticipate that these advances will one day make them truly benign, but much work needs to be done to get there.

Cold agglutinin disease (CAD) was originally described by Helmut Schubothe in 1966 as a rare disorder that is mediated by cold antibodies causing complement-dependent immune hemolysis.1  This disorder contributes to 10 to 15% of autoimmune hemolytic anemia (AIHA) cases.2 

Patients can have primary CAD in the context of immunoglobulin M (IgM) monoclonal protein in the serum; recently it has been characterized as a distinct clonal B-cell lymphoproliferative disorder.3  Secondary form, or cold agglutinin syndrome (CAS), is typically associated with an infection such as Mycoplasma pneumoniae or Epstein-Barr virus.4, 5 

CAD is mediated by monoclonal IgM immunoglobulin with kappa light chain restriction in 90% of cases.6  The IgM remains unbound from the red cell surface when the blood circulates in the body core but as it makes its way toward the peripheral circulation, particularly in acral areas, IgM transiently binds to the I antigen binding site on the red cell membrane. It then fixes the complement on the surface of the red cell, activating the complement cascade (C3 binding). Upon the return of blood to the core of the body, IgM dissociates and the C3b-coated cell is subsequently phagocytosed by receptor-specific macrophages predominantly present in the liver. This process leads to extravascular hemolysis.7, 8  (Figure 1)

Figure 1

Immune-initiated, complement-mediated erythrocyte destruction in cold agglutinin disease (CAD) and cold agglutinin syndrome (CAS).

Berentsen S, et al. Biomed Res Int. 2015;2015:363278.

Figure 1

Immune-initiated, complement-mediated erythrocyte destruction in cold agglutinin disease (CAD) and cold agglutinin syndrome (CAS).

Berentsen S, et al. Biomed Res Int. 2015;2015:363278.

Close modal

Patients with CAD typically present in their 70s, with a median age around 67 years. Females have a higher propensity of developing this disorder.6, 9  The most common presentation is anemia, with hemoglobin ranging from 4 to 10 g/dL. Severe anemia is present in approximately one-third to one-fourth of patients.10 

The diagnosis is typically made secondary to symptoms of hemolytic anemia such as fatigue or cold-induced agglutination in the capillary circulation, which causes acrocyanosis, livedo reticularis, livedo racemosa, or even disabling Raynaud’s phenomenon.6  Patients with CAD can develop life-threatening hemolysis in the setting of febrile infectious complications or surgical intervention, both of which can exacerbate hemolysis.11, 12 

The hallmark of CAD is agglutination of red cells on a blood smear. Biochemical evidence of hemolysis, with a direct antiglobulin test strongly positive for complement C3d and a cold agglutinin titer above 64, clinches the diagnosis of CAD.13 

Laboratory results that help with the diagnosis of CAD are a complete blood count, which demonstrates varying degrees of anemia with elevated mean corpuscular volume (this can be related to reactive reticulocytosis or agglutination), reticulocyte count, lactate dehydrogenase, total and indirect bilirubin, and low haptoglobin. A direct antiglobulin test is the key to a CAD diagnosis; the test will be strongly positive for C3 and also positive for immunoglobulin G (IgG) in up to 20% of cases. In most cases, serum electrophoresis with immunofixation will show monoclonal IgM kappa class antibodies. Given the temperature sensitivity of the antibodies, the processing and handling of specimens becomes critically important.14 

B-cell clonality has been highlighted by several researchers as a peculiar clonal rearrangement of the heavy or light chain genes; this peculiarity differentiates this clonal B-cell process from what was then thought to be lymphoplasmacytic lymphoma (LPL) or Waldenström macroglobulinemia. Furthermore, the most prominent feature of CAD is absence of the MYD88 L265P mutation, which is seen in 90% cases of LPL. This mutation could not be demonstrated in CAD-associated lymphoproliferative disease. Clonal rearrangement of IGKV3-20 gene sequences in highly homogeneous CDR3 regions is seen in more than half of cases. Recurrent mutations in KMT2D and CARD11 were also seen.15 

Further evaluation is necessary to rule out CAS secondary to infectious etiology and to determine if any monoclonal protein is detected.16 

In the past, pharmacotherapy for CAD was reserved for patients with severe hemolysis due to poor response to the available therapeutic options such as steroids,17  azathioprine,6  and interferon.18  Cold avoidance and prompt treatment of infections, as well as warmed transfusions, were the cornerstones of therapy for patients with compensated low-grade hemolysis.19 

With significant advances in pharmacotherapy in past decades, patients with symptomatic mild-to-moderate anemia or cold-induced circulatory symptoms should be considered for therapy. Recombinant erythropoietin can be used for supportive therapy.20 

B-cell–directed chemoimmunotherapy is the cornerstone for management of primary CAD. Table 1 describes the choices and outcomes.

Table 1

B-cell–directed chemo-immunotherapy.

Initial/relapse settingPhase III?NOverall response rateMedian time to responseMedian duration of responseFDA approval?
Rituximab13  No 20-32 45-54% 2 months 10 months No 
Rituximab + prednisone24  No 56% 0.5 months N/A No 
Rituximab + fludarabine25  No 29 76% 4 months >66 months No 
Rituximab + bendamustine26  No 45 71% 1.9 months >88 months No 
Chlorambucil6, 13  No 19-37 16-46% N/A 11 months No 
Bortezomib27  No 19 32% N/A 16 months No 
Ibrutinib28  No 10 100% 1 month 9 months No 
Initial/relapse settingPhase III?NOverall response rateMedian time to responseMedian duration of responseFDA approval?
Rituximab13  No 20-32 45-54% 2 months 10 months No 
Rituximab + prednisone24  No 56% 0.5 months N/A No 
Rituximab + fludarabine25  No 29 76% 4 months >66 months No 
Rituximab + bendamustine26  No 45 71% 1.9 months >88 months No 
Chlorambucil6, 13  No 19-37 16-46% N/A 11 months No 
Bortezomib27  No 19 32% N/A 16 months No 
Ibrutinib28  No 10 100% 1 month 9 months No 

However, slow-to-achieve and suboptimal responses to chemoimmunotherapy (RR of 50 to 76%) led to the investigation of whether complement modulation could help achieve faster response rates (Table 2).

Table 2

Complement-directed therapy.

Initial/relapse settingPhase III?NOverall response rateTime to median responseMedian duration of responseFDA approval?
Eculizumab29  No 13 54% N/A NA No 
Sutimlimab21  Yes 22 73% 0.3 months NA Yes 
Initial/relapse settingPhase III?NOverall response rateTime to median responseMedian duration of responseFDA approval?
Eculizumab29  No 13 54% N/A NA No 
Sutimlimab21  Yes 22 73% 0.3 months NA Yes 

Sutimlimab is a humanized monoclonal antibody against C1 and the first agent approved by the U.S. Food and Drug Administration (FDA) for the management of CAD. Röth and colleagues published results from the open-label, single-arm, phase III CARDINAL study where sutimlimab demonstrated more rapid clinical and laboratory improvements in comparison to placebo.21  This study was followed by the randomized, placebo-controlled phase III CADENZA trial, in which sutimlimab demonstrated reduced hemolysis, anemia, and fatigue compared with placebo, with minimal adverse events.22  Unfortunately, the response to the drug is limited to the duration of therapy and it does not improve the symptoms related to Raynaud’s phenomenon.23 

Figure 2 outlines our approach to management of CAD.

Figure 2

A summary of our approach to the management of CAD.

Figure 2

A summary of our approach to the management of CAD.

Close modal

Other agents are under investigation, such as ANX005, a humanized monoclonal antibody that inhibits C1q, which is being evaluated in a safety trial with healthy volunteers (ClinicalTrials.gov: NCT03010046). APL-2 is a compstatin-based pegylated cyclic peptide inhibitor of C3; it prevents the formation of C3b. It is currently under evaluation for safety, tolerability, and efficacy in patients with warm autoimmune hemolytic anemia (AIHA) or CAD (ClinicalTrials.gov: NCT03226678).

CAD is a complement-driven rare AIHA with an underlying B-cell clonal disorder. B-cell–directed chemoimmunotherapy leads to slow-to-achieve responses but can offer long-term remissions. With the recent FDA approval of complement-directed therapy, prompt improvement is made possible. These options have opened the conversation for sequencing or combining these therapies. It might be pragmatic to combine both therapies, where complement-directed therapy helps in early control of hemolysis while the immune-ablative effect of B-cell– directed therapy is awaited. Further studies are needed to assess this approach.

Dr. Shah and Dr. Go indicated no relevant conflicts of interest.

1
Schubothe
H
.
The cold hemagglutinin disease
.
Semin Hematol
.
1966
;
3
(
1
):
27
47
.
2
Sokol
RJ
,
Hewitt
S
,
Stamps
BK
.
Autoimmune haemolysis: an 18-year study of 865 cases referred to a regional transfusion centre
.
Br Med J (Clin Res Ed)
.
1981
;
282
(
6281
):
2023
2027
.
3
Randen
U
,
Trøen
G
,
Tierens
A
, et al
.
Primary cold agglutinin-associated lymphoproliferative disease: a B-cell lymphoma of the bone marrow distinct from lymphoplasmacytic lymphoma
.
Haematologica
.
2014
;
99
(
3
):
497
504
.
4
Berentsen
S
.
New insights in the pathogenesis and therapy of cold agglutinin-mediated autoimmune hemolytic anemia
.
Front Immunol
.
2020
(
11
):
590
.
5
Jäger
U
,
Barcellini
W
,
Broome
CM
, et al
.
Diagnosis and treatment of autoimmune hemolytic anemia in adults: recommendations from the First International Consensus Meeting
.
Blood Rev
.
2020
(
41
):
100648
.
6
Berentsen
S
,
Ulvestad
E
,
Langholm
R
, et al
.
Primary chronic cold agglutinin disease: a population based clinical study of 86 patients
.
Haematologica
.
2006
;
91
(
4
):
460
466
.
7
Ulvestad
E
,
Berentsen
S
,
K
,
Shammas
FV
.
Clinical immunology of chronic cold agglutinin disease
.
Eur J Haematol
.
1999
;
63
(
4
):
259
266
.
8
affe
CJ
,
Atkinson
JP
,
Frank
MM
.
The role of complement in the clearance of cold agglutinin-sensitized erythrocytes in man
.
J Clin Invest
.
1976
;
58
(
4
):
942
949
.
9
Berentsen
S
.
Complement activation and inhibition in autoimmune hemolytic anemia: focus on cold agglutinin disease
.
Semin Hematol
.
2018
;
55
(
3
):
141
149
.
9
Petz
LD
.
Cold antibody autoimmune hemolytic anemias
.
Blood Rev
.
2008
;
22
(
1
):
1
15
.
10
Berentsen
S
,
Barcellini
W
,
D’Sa
S
, et al
.
Cold agglutinin disease revisited: a multinational, observational study of 232 patients
.
Blood
.
2020
;
136
(
4
):
480
488
.
11
Ulvestad
E
.
Paradoxical haemolysis in a patient with cold agglutinin disease
.
Eur J Haematol
.
1998
;
60
(
2
):
93
100
.
12
Ulvestad
E
,
Berentsen
S
,
Mollnes
TE
Acute phase haemolysis in chronic cold agglutinin disease
.
Scand J Immunol
.
2001
;
54
(
1–2
):
239
242
.
13
Swiecicki
PL
,
Hegerova
LT
,
Gertz
MA
.
Cold agglutinin disease
.
Blood
.
2013
;
122
(
7
):
1114
1121
.
14
Berentsen
S
,
Röth
A
,
Randen
U
,
Jilma
B
,
Tjønnfjord
GE
.
Cold agglutinin disease: current challenges and future prospects
.
J Blood Med
.
2019
10
93
103
.
15
Małecka
A
,
Trøen
G
,
Tierens
A
, et al
.
Frequent somatic mutations of KMT2D (MLL2) and CARD11 genes in primary cold agglutinin disease
.
Br J Haematol
.
2018
;
183
(
5
):
838
842
.
16
Go
RS
,
Winters
JL
,
Kay
NE
.
How I treat autoimmune hemolytic anemia
.
Blood
.
2017
;
129
(
22
):
2971
2979
.
17
Berentsen
S
.
Complement activation and inhibition in autoimmune hemolytic anemia: focus on cold agglutinin disease
.
Semin Hematol
.
2018
;
55
(
3
):
141
149
.
18
Hillen
HF
,
Bakker
SJ
.
Failure of interferon-alpha-2b therapy in chronic cold agglutinin disease
.
Eur J Haematol
.
1994
;
53
(
4
):
242
243
.
19
Hill
QA
,
Stamps
R
,
Massey
E
, et al
.
The diagnosis and management of primary autoimmune haemolytic anaemia
.
Br J Haematol
.
2017
;
176
(
3
):
395
411
.
20
Fattizzo
B
,
Michel
M
,
Zaninoni
A
, et al
.
Efficacy of recombinant erythropoietin in autoimmune hemolytic anemia: a multicenter international study
.
Haematologica
.
2021
;
106
(
2
):
622
625
.
21
Röth
A
,
Barcellini
W
,
D’Sa
S
, et al
.
Sutimlimab in cold agglutinin disease
.
N Engl J Med
.
2021
;
384
(
14
):
1323
1334
.
22
Röth
A
,
Berentsen
S
,
Barcellini
W
, et al
.
Sutimlimab in patients with cold agglutinin disease: results of the randomized placebo-controlled phase 3 CADENZA trial
.
Blood
.
2022
;
140
(
9
):
980
991
.
23
Jäger
U
,
D’Sa
S
,
Schörgenhofer
C
, et al
.
Inhibition of complement C1s improves severe hemolytic anemia in cold agglutinin disease: a first-in-human trial
.
Blood
.
2019
;
133
(
9
):
893
901
.
24
Barcellini
W
.
New insights in the pathogenesis of autoimmune hemolytic anemia
.
Transfus Med Hemother
.
2015
;
42
(
5
):
287
293
.
25
Berentsen
S
,
Randen
U
,
Vågan
AM
, et al
.
High response rate and durable remissions following fludarabine and rituximab combination therapy for chronic cold agglutinin disease
.
Blood
.
2010
;
116
(
17
):
3180
3184
.
26
Berentsen
S
,
Randen
U
,
Oksman
M
, et al
.
Bendamustine plus rituximab for chronic cold agglutinin disease: results of a Nordic prospective multicenter trial
.
Blood
.
2017
;
130
(
4
):
537
541
.
27
Rossi
G
,
Gramegna
D
,
Paoloni
F
, et al
.
Short course of bortezomib in anemic patients with relapsed cold agglutinin disease: a phase 2 prospective GIMEMA study
.
Blood
.
2018
;
132
(
5
):
547
550
.
28
Jalink
M
,
Berentsen
S
,
Castillo
JJ
, et al
.
Effect of ibrutinib treatment on hemolytic anemia and acrocyanosis in cold agglutinin disease/cold agglutinin syndrome
.
Blood
.
2021
;
138
(
20
):
2002
2005
.
29
Röth
A
,
Araten
DJ
,
Larratt
L
, et al
.
Beneficial effects of eculizumab regardless of prior transfusions or bone marrow disease: Results of the International Paroxysmal Nocturnal Hemoglobinuria Registry
.
Eur J Haematol
.
2020
;
105
(
5
):
561
570
.