ArticlePDF AvailableLiterature Review

How Does Complement Affect Hematological Malignancies: From Basic Mechanisms to Clinical Application

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Complement, as a central immune surveillance system, can be activated within seconds upon stimulation, thereby displaying multiple immune effector functions. However, in pathologic scenarios (like in tumor progression), activated complement can both display protective effects to control tumor development and passively promotes the tumor growth. Clinical investigations show that patients with several hematological malignancies often display abnormal level of specific complement components, which in turn modulates complement activation or deregulated cascade. In the past decades, complement-dependent cytotoxicity and complement-dependent cell-mediated phagocytosis were fully approved to display vital roles in monoclonal antibody-based immunotherapies, especially in therapies against hematological malignancies. However, tumor-mediated complement evasion presents a big challenge for such a therapy. This review aims to provide an integrative overview on the roles of the complement in tumor promotion, highlights complement mediated effects on antibody-based immunotherapy against distinct hematological tumors, hopefully provides a theoretical basis for the development of complement-based cancer targeted therapies.
Content may be subject to copyright.
How Does Complement Affect
Hematological Malignancies: From
Basic Mechanisms to Clinical
Application
Shanshan Luo
1
, Moran Wang
1
, Huafang Wang
1
, Desheng Hu
1,2
, Peter F. Zipfel
3,4
*
and Yu Hu
1
*
1
Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,
China,
2
Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China,
3
Department of Infection Biology, Leibniz Institute for
Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany,
4
Faculty of Biological Sciences,
Friedrich Schiller University, Jena, Germany
Complement, as a central immune surveillance system, can be activated within seconds
upon stimulation, thereby displaying multiple immune effector functions. However, in
pathologic scenarios (like in tumor progression), activated complement can both display
protective effects to control tumor development and passively promotes the tumor
growth. Clinical investigations show that patients with several hematological
malignancies often display abnormal level of specic complement components, which
in turn modulates complement activation or deregulated cascade. In the past decades,
complement-dependent cytotoxicity and complement-dependent cell-mediated
phagocytosis were fully approved to display vital roles in monoclonal antibody-based
immunotherapies, especially in therapies against hematological malignancies. However,
tumor-mediated complement evasion presents a big challenge for such a therapy. This
review aims to provide an integrative overview on the roles of the complement in tumor
promotion, highlights complement mediated effects on antibody-based immunotherapy
against distinct hematological tumors, hopefully provides a theoretical basis for the
development of complement-based cancer targeted therapies.
Keywords: complement, hematological malignancies, immune evasion, tumor progression, immunotherapy
INTRODUCTION
Complement was initially identied more than 100 years ago due to a result of its bactericidal
activity complementaryto the action of immunoglobulins and the role in phagocytosis of cellular
debris (1,2). As an essential part of innate immunity and an evolutionary old system, complement is
highly conserved among a wide variety of species, emphasizing its importance in immune defense
throughout evolution (3). Complement-dependent cytotoxicity (CDC) and complement-dependent
cell-mediated phagocytosis (CDCP) were fully approved to display vital roles in maintaining
homeostasis, ghting against infection and even in monoclonal antibody-based immunotherapies,
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936101
Edited by:
Junji Xing,
Houston Methodist Research Institute,
United States
Reviewed by:
Lubka T. Roumenina,
INSERM U1138 Centre de Recherche
des Cordeliers (CRC), France
Yifan Wang,
University of Texas Southwestern
Medical Center, United States
*Correspondence:
Peter F. Zipfel
peter.zipfel@leibniz-hki.de
Yu Hu
dr_huyu@126.com
These authors have contributed
equally to this work
Specialty section:
This article was submitted to
Molecular Innate Immunity,
a section of the journal
Frontiers in Immunology
Received: 11 August 2020
Accepted: 02 October 2020
Published: 29 October 2020
Citation:
Luo S, Wang M, Wang H, Hu D,
Zipfel PF and Hu Y (2020) How Does
Complement Affect Hematological
Malignancies: From Basic
Mechanisms to Clinical Application.
Front. Immunol. 11:593610.
doi: 10.3389/fimmu.2020.593610
REVIEW
published: 29 October 2020
doi: 10.3389/fimmu.2020.593610
especially in therapies against hematological malignancies.
Dysfunction of each step of the whole cascade disrupts
homeostasis and ultimately leads to severe diseases, such as
tissue damage, autoimmune diseases, infection and tumor
progression (4). Clinical investigations show that patients with
hematological malignancies often display abnormal level of
complement components, which in turn modulates
complement activation.
Hematologic malignancies are complex groups of disorders,
which becomes more and more into peoplesfocussincethey
are often diagnosed in clinic. Although the general cure rate of
hematologic malignancies has been greatly improved and some
types have even high cure rates now, great challenges still exist
due to the large number of disease subtypes and high
heterogeneity. According to the 2018 global cancer statistics,
the incidence and mortality of non-Hodgkin lymphoma (NHL)
ranks rst in hematological neoplasms, followed by leukemia,
multiple myeloma (MM), and Hodgkin lymphoma (HL) (5).
The causes of the diseases are various, but the host immune
conditions and tumor virulence factors are the main
determinants which closely correlated with the disease state.
The management of hematologic malignancies has traditionally
relied on chemotherapy and/or radiotherapy regimens to
control the tumor progressionandprolongthelifespan(6).
However, over the past two decades, signicant progress has
been made for the development of monoclonal antibody (mAb)
therapies for hematologic malignancies. For example, anti-
CD20 mAbs, mainly rituximab (RTX) or obinutuzumab,
combined with chemotherapy agents have been approved for
treatment of diffuse large B-cell lymphoma (DLBCL), follicular
lymphoma (FL) and chronic lymphocytic leukemia (CLL),
respectively (7,8). Complement-dependent cytotoxicity and
complement-dependent cell-mediated phagocytosis display
vital roles in mAb-based immunotherapies. However, tumor-
mediated complement evasion might be a big challenge for such
atherapy.
In this review, we will rst summarize complement
activation, function, and regulation, present an overview of
the double edgedroles of complement in tumor progression,
then provide a deep insight into the roles of complement in
hematological malignancies, and further discuss complement
mediated effects on antibody-based immunotherapy against
hematological tumors.
COMPLEMENT ACTIVATION, FUNCTION
AND REGULATION
The complement system comprises more than 60 different
components which include main components, various activation
products, effector components, regulators, and several surface
bound complement receptors. Soluble complement exists in the
body uids, displaying multiple immune effector functions.
Complement activation occurs in a sequential manner via three
different pathways and consists of four main steps: initiation, C3
convertase formation and amplication, C5 convertase formation,
and the assembly of the terminal complement complex (TCC),
also known as membrane attack complex (MAC). The alternative
pathway (AP) is initiated spontaneously and constantly. The lectin
pathway (LP) is activated uponbinding of mannose-binding lectin
to mannan and carbohydrate structures on microbial surfaces. The
classical pathway (CP) is activated via antigenantibody
complexes or by C-reactive protein (4,9). Activation of all three
pathways results in the generation of C3 convertases that cleave C3
into C3a and C3b, followed by C5 convertase formation that
cleaves C5 into C5a and C5b, and the generation of TCC (3,4)
(Figure 1).
The cleavage product C3b binds to target surfaces, where it
acts as opsonin and mediates recognition and phagocytosis by
host immune effector cells (10,11). C3a and C5a function as
anaphylatoxins which initiate inammation. Furthermore, C3a
also has antimicrobial activity by binding to the cell surface of
microbes and induces membrane perturbations and release of
extracellular material (12). In addition, complement also
functions as a link between innate and adaptive immunity. C3
synthesis by myeloid cells, a relatively minor source of
complement, provides a critical function during the induction
of humoral B responses to peripheral herpes simplex virus
infection. Further, macrophages derived from bone marrow
produce sufcient C4 to restore the humoral response to virus
infection in C4-decient animals, demonstrating local
complement C3 and C4 production are required to enable
efcient B cell responses (13,14). Immune responses of T cell
to Listeria monocytogens were impaired in the absence of C3
(15). Besides that, complement activation products C5a/C3a and
its receptors (C5aR/C3aR) have a clear role in directly and
indirectly promoting T cell activation and proliferation and as
such, promoting allograft rejection, autoimmunity, and ghting
infection (1618).
Due to its potency and the damaging effects, many
complement components are engaged in regulation (Figure
1). Complement regulators function at all levels of the cascade
and are classied into two major classes: uid phase regulators
and membrane-integral complement receptors, such as Factor
H, Factor H like protein 1 (FHL-1), C4b binding protein
(C4BP), C1 inhibitor (C1INH), complement Factor H related
protein 1 (CFHR1), CFHR2, CFHR3, CFHR4, CFHR5, as well
as complement receptor 1 (CR1), CR2, CR3, CR4, CD46,
CD55, CD59, CRIg, vitronectin, clusterin as well as
carboxypeptidase N (2,19,20). Being highly regulated by
these regulators, complement forms an important, central
immune defense line and mediates cell integrity and tissue
homeostasis. Additionally, beyond complement regulation,
several of the above-mentioned regulators have additional
activities, such as mediating cell adhesion and extracellular
matrix interaction, or linking the complement cascade with
other important physiological networks (e.g. the coagulation
cascade) (21). However, due to the complement dysfunction,
many disease pathologies including tumor progression, tissue
damage, autoimmune diseases and infection may take
place (22).
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936102
ROLE OF COMPLEMENT IN TUMOR
PROGRESSION: THE TWO SIDES
OF THE COIN
Complement Mediated Anti-Tumor Effects
The complement system is a double-edged sword in tumor
development since complement activation is not only involved
in anti-tumor cytotoxicity and immune responses, but
also promotes tumor development directly and indirectly.
Regarding to its anti-tumor side, complement, upon
activation, displays various controlling effects (e.g. C3b/iC3b
mediated phagocytosis and TCC mediated cell lysis) on various
tumor cells including both solid tumors and hematological
malignancies (23). For example, upon treatment of CLL,
complement activation was initiated by anti-CD20
monoclonal antibody RTX, thereby displaying efcient CDC
and CDCP to clear the tumor cells (24). However, in order to
block the toxic effects of activated complement, and to survive,
tumor cells, similar to infectious microbes utilize multiple
evasion strategies to actively escape complement attack and
immune surveillance. For example, lung, ovarian, glial and
hematological tumor cells show enhanced expression and
surface binding of soluble regulators including Factor H,
FHL-1, FHR1, FHR-4, FHR5, and C4BP. These up-regulated/
bound complement regulators further display the cofactor
activity, which function together with factor I to block
complement activation at the level of C3 convertase, thereby
leading to complement evasion (2529). Similarly, the
membrane-bound complement inhibitors (e.g. CD46, CD55,
and CD59) are up-regulated in various primary tumors and
FIGURE 1 | Complement activation, effector function and regulation. Complement system is activated by three different pathways, then merged at the level of C3
cleavage, followed by C5 convertase formation and generation of terminal complement complex. Upon activation, different activation products are generated, which
display multiple immune effector functions. The whole system is tightly self-controlled by different regulators.
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936103
tumor lines to evade the complement attack (30,31). Serglycin is
another endogenous complement inhibitor secreted by human
MM cell lines. Serglycin can inhibit both the classical and lectin
pathwaysactivation by direct interaction with C1q and
mannose-binding lectin, thereby blocking complement
mediated immune effector effects on MM cells (32).
Complement Mediated Promotion
of Tumor Growth
Besides the controlling effect, over-activation of complement also
promotes tumor growth through the pro-inammatory
properties of effector compounds, which is in line with
established cancer-promoting effects of chronic infections (33).
Chou et al. reported that local production and activation of
complement effector compounds were distinctly important for
promoting tumor growth, while the systemic production of
complement by liver did not affect cancer (34).
C3aC3aR and C5aC5aR Mediated
Proliferative Effects
The proliferative effect on tumors is directly correlated to
the activation of complement cascade, as derived by C3a and
C5a via C3aR and C5aR1 mediated PI3K/AKT signaling
pathway (34). Shu et al. reported that C3a-C3aR signaling via
PI3K/AKT pathway in carcinoma associated broblasts
facilitated the metastasis of breast cancer. Targeting C3aR
signaling was shown to be a potential anti-metastasis
strategy for breast cancer therapy (35). Upon signaling by C5a
stimulation, C5aR1 expressed tumor cells underwent cytoskeletal
rearrangement (e.g. lopodia formation, membrane rufing) and
furthermore released matrix metalloproteinase, which lead to
increased tumor cell motility and invasiveness both in vitro and
in vivo (36). Another report showed that C5aR1 signaling
induced myeloid-derived suppressor cells to produce larger
amounts of reactive oxygen species and reactive nitrogen
species, which inhibited CD8+ T cell mediated anti-tumor
activity, thereby leading to tumor growth (37). Vadrevu et al.
showed that in a lung cancer metastatic model, C5aR1 blockage
resulted in decreased lung metastasis due to reduced TGF-b and
IL-10 production by myeloid-derived suppressor cells since both
TGF-b and IL-10 induced regulatory T-cell generation and
facilitated an immunosuppressive Th2-based T cell responses
(38). Further in vivo analysis showed that C5aR1 signaling
promotes melanoma growth by promoting inltration of
immunosuppressive leukocyte populations into the tumor
microenvironment, whereas C5aR2 has a more restricted but
benecial role in limiting tumor growth, further proving the
double-edgedrole of complement activation in tumor
promotion (39).
Other Complement Components
Mediated Effects
Besides the complement receptors mediated effects, different
components of the main cascade also display onco-progression
ability, such as C1q, C3 and C5b-9. Bioinformatics analysis by
Mangogna et al. showed that high levels of C1q have a favorable
prognostic index in basal-like breast cancer for disease-free
survival, and in HER2-positive breast cancer for overall
survival (40). C1q acts in the tumor microenvironment as a
cancer-promoting factor in a complement dependent as well as
independent manner. In clear-cell renal cell carcinoma, tumor
associated macrophages produce high densities of C1q, which
together with tumor cell expressed C1r, C1s, C4, and C3 initiates
CP activation. The activation products as well as tumor
associated macrophages-derived C1q further promotes an
immunosuppressed microenvironment characterized by high
expression of immune checkpoints (e.g. programmed cell death
protein (PD)-1, Lag-3, programmed cell death ligand 1 (PD-L1),
and PD-L2), thereby fueling tumor progression. Mice decient in
C1q, C3, and C4 displayed decreased tumor growth (41).
Yuan et al. reported that local complement C3 overexpression
activated JAK2/STAT3 pathway and promoted gastric cancer
progression. Further clinical investigation showed that C3 and
C3a expression was markedly enhanced in gastric cancer
tissues at both mRNA and protein levels compared with those
in paired non-tumorous tissues. High C3 deposition was
identied as an independent prognostic factor of poor 5-year
overall survival, suggesting that local C3 deposition in the tumor
microenvironment is a relevant immune signature for predicting
prognosis of gastric cancer (42). In vivo data showed that the
knockdown of C3 suppressed hepatic stellate cells-promoted
hepatocellular carcinoma development (43).
Further, sublytic C5b-9 displays tumor-promoting properties
by activating signal transduction pathways (e.g., Gi protein/
PI3K/Akt kinase and Ras/Raf1/ERK1) and modulating the
activation of cancer-related transcription factors, while shielding
malignant cells from apoptosis (44). Such complement promoted
tumor progression also actively involved in cutaneous squamous
cell carcinoma, which was nicely reviewed by Pilvi Riihilä
et al. (45).
Based on these roles of complement in promoting cancer
progression, more and more complement proteins are becoming
potential candidates for cancer targeted therapy and numerous
new anti-complement drugs are under clinical development (46).
For example, C3aR and C5aR are recently classied as a new class
of immune checkpoint receptor in cancer immunotherapy (47).
In addition, humanized soluble CR1-Fc fusion protein were
generated to target C3b/C4b and its therapeutic effect was
conferred in a colitis-associated colorectal cancer model and
orthotopic 4T1 breast cancer model (48).
COMPLEMENT IN HEMATOLOGICAL
MALIGNANCIES
The number of patients with hematological malignancies is
increasing on a daily basis. Many patients with hematologic
malignancies display abnormal levels of complement components,
possibly as a result of the tumor avoiding complement surveillance
(Figure 2).Inthispart,wewillsummarizetheroleofcomplement
in hematological malignancy conditions.
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936104
Complement in Multiple Myeloma
MM is a hematological malignant tumor characterized by clonal
proliferation of plasma cells that produce M protein,
accompanied by various types of impaired immune function.
C1q, being one of the main components in CP, seems to be a
potential marker of the immunodeciency status in MM
patients. Statistically, the mean serum level of C1q in MM
patients is lower than that in the control group. At different
stages of MM, C1q level also changes dynamically. When the
disease is in remission, C1q is back to a normal level. However,
with disease re-progressed, C1q level in the plasma decrease
again, which indicates that MM cells, by controlling/consuming
C1q, to some extent modulate the CP activation (49). Further,
MM cells recruit complement regulator Factor H to their surface.
Surface bound Factor H mediates factor I to cleave C3b, thereby
down-regulating complement activation. Consistent with the
above ndings regarding complement deciency, evidence
shows that MM patients are at high risk of bacterial infections.
No matter with normal or elevated level of C3, all sera from MM
patients have a defect in C3b binding to Streptococcus
penumoniae. Experimental data showed that addition of
normal human serum to serum from MM patients restored the
C3b binding ability to S. pneumoniae. However, adding anti-S.
pneumoniae antibodies to MM serum did not rescue C3b
deposition to any S. pneumoniae types. Such a scenario
indicates that serum from MM patients has a defect in C3
activation, also explains why the MM patients have increased
susceptibility to S. pneumoniae infections (50). Further, a recent
clinical investigation showed that serumC3 and C4levels in MM
patients were signicantly higher than those in healthy people.
Among the MM patients, serum C3 and C4 levels positively
correlated with diseases severity. However, the mechanism how
serum C3 and C4 promote MM development is not yet
clear (51).
Ficolins (Ficolin 1, 2, 3) are complement lectin pathway
defense factors, which are able to distinguish between
self,abnormal selfand non-self, thereby contributing to
the elimination of abnormal selfand non-selfby direct
opsonization and/or initiation of complement activation
through the lectin pathway. Lower level of colin-1 and colin-2
as well as polymorphisms of FCN1 and FCN2 genes were detected
in patients diagnosed as MM, compared with control group,
which leads to the higher hospital infections because of limited
complement activation (52). The same group reported that in the
early time a higher frequency of mannose-binding lectin
deciency-associated genotypes among MM patients was
detected compared with controls (53).
Complement in Leukemia
Leukemia is another type of life-threatening hematological
malignancy. Experimental data show that leukemia cell lines
together with clonogenic blasts from both CLL and acute
myeloid leukemia (AML) patients respond strongly to C3 and
C5 cleavage fragments in form of chemotaxis and increased
adhesion. C3aR and C5aR were also detected at the mRNA and
protein level in both human malignant hematopoietic cell lines
and patient blasts upon stimulation by C3a and C5a (54).
Further, abnormal C5 pattern approved in 42% CLL patients
since C5 protein was detected as double bands accompanied by a
lower molecular band when analyzed by Western blot, indicating
that C5 cleavage/C5 activation happened, whereas C5 from the
healthy controls and other CLL patients showed only one normal
FIGURE 2 | Complement evasion of hematological malignancies. Different types of hematological malignancies utilize different strategies for complement evasion.
In MM patients, serum levels of C1q are down-regulated, which mediates CP inactivation. MM cells recruit Factor H to their surface, thereby down-regulating AP
activation. Up-regulation of CD46 and CD55 inhibits complement activation. Down-regulation and variation of colin1, 2 lead to LP inactivation. In CLL patients,
tumor cells altered C5 pattern to modify CP activation. Variation of C9 correlates with EFS of FL, and variation of C7 correlates with EFS of DLBCL.
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936105
band (55). In comparison to patients with normal C5 pattern,
patients with abnormal C5 pattern have increased basal levels of
sC5b-9 and C5a, while sC5b-9 levels after CP activation were
signicantly decreased by 48%, suggesting CP activation was
impaired. Such a scenario suggests a link between the pattern of
C5 and CP activation (55) in CLL patients. In addition, a defect
of C3b deposition on bacterial surface was detected although the
serum concentrations of C3 are normal in patients with CLL.
Mixing normal serum with serum from CLL patients restored
C3b binding to bacterial surface, suggesting a defect in either the
activation or activity of C3 in CLL serum, which likely accounts
for the increased incidence of infections in these patients (56).
Further, signicant decrease of serum C1 and C4 levels was
found in CLL patients, which correlated with the abnormal
hemolytic activities. Meanwhile, a complement prole
characteristic of acquired C1-IHN deciency was observed in
all tested patients, which indicates that the depression of the CP
activity is a frequently occurring feature in CLL patients (57).
Moreover, different patterns of complement proteins were
observed in different types of leukemia. In AML patients, each
complement parameter tested was elevated as compared to the
control values (sera of healthy blood donors). The extent of C3
activation and C3 splitting were correlated with disease severity
while immunoglobin level remained consistently high and not
varied much between different types of leukemia. Similar results
were observed in acute lymphocytic leukemia patients although
the differences were less marked. In chronic myelocytic
leukemia, the CP and AP activities behave differently:
activities of CP and serum C4 levels were signicantly
elevated, whereas activity of AP as well as serum C3 and
factor B concentration were not signicantly different from
the control groups (58).
CR1 is expressed by erythrocytes and most leukocytes. sCR1,
a soluble form, is shed from the leukocytes and found in plasma,
which is identied as a powerful inhibitor of complement. Sadallah
et al. reported sCR1 in the plasma of leukemia patients increased
up to levels producing measurable complement inhibition, which
is a possible complement evasion strategy utilized by leukemia
cells (59).
Complement in Non-Hodgkin Lymphoma
The most common types of NHL are DLBCL and FL. Germline
mutations in complement genes have been associated with
susceptibility to infections and autoimmune diseases,
conditions that are associated with NHL risk (60). For
example, genetic variations of complement genes (e.g., MASP2,
C5 and C9) are strongly associated with the development of NHL
(60,61). Charbonneau et al. also found that variations in C9 and
complement regulatory genes (e.g., Factor H, CFHR1, CFHR5,
CD46, and CD55) were associated with the event-free survival
(EFS) of FL, while C7 variations were associated with EFS of
DLBCL (62). Thus, patients with different genotypes of
complement components respond differently to complement
attack and to antibody-based immunotherapy. Factor H,
CFHR1 and CFHR5 polymorphisms have a stronger impact on
EFS of FL patients who were treated with anti-CD20 antibody,
while CD46 and CD55 polymorphisms had a stronger impact on
EFS in FL patients who did not receive initial treatment. In
addition, C1qA polymorphism seems to be a biomarker for
predicting rst-line response to R-CHOP regimen in DLBCL
patients. In a retrospective analysis, DLBCL patients with C1qA
homozygote A allele showed higher complete remission rates
and longer overall survival after receiving R-CHOP regimen with
an unknown mechanism (63). Further, over-expression of
complement regulators, such as CD46, CD55 and CD59 is one
of the main strategies utilized by NHL cell lines for complement
evasion (64). All these data suggest a potential role of the
complement system in NHL progression.
The Co-Expression of Complement Genes
by Hematological Malignancies and Solid
Tumors
Many complement genes are co-expressed by different types
of tumors. Roumenina et al. reported that there is strong
heterogeneity in expression among different complement genes;
however, not much difference existed between cancer types when
comparing the transcription levels of 50 complement related genes
within 30 types of tumors (mainly solid tumors) (65). When we
further compared the co-expression prole of complement genes
between hematological malignancies and solid tumors using the
available data, both DLBCL and AML have higher expression level
of complement CFD gene. Moreover, similar to most of the solid
tumors, DLBCL also shows higher expression of C3,ITGB2,as
well as genes of the classical pathway (i.e. C1QA,C1QB,C1QC,
C1R,C1S,andC2), while the C4BP, lectin pathway genes (i.e.
MBL2, FCN,andMASP2), and the terminal pathway genes (i.e C6,
C8. and C9) are poorly expressed. However, different transcription
proles of the highly expressed genes are observed for the AML.
Besides CFD,FCN1, C1RL, C3AR1, and C5AR1 are strongly
expressed, the rests including C3 are all poorly expressed
(Supplementary Figure 1,Supplementary Table 1). In
addition, Roumenina L. and colleagues suggested that the co-
expression of complement genes by the tumors confers poor or
favorable prognosis or had no impact depending on the cancer
type. In this scenario, both DLBC and AML fall in the group of no
impact on the prognosisbased on our further analysis
(Supplementary Figure 2,Supplementary Table 2).
ANTIBODY-BASED IMMUNOTHERAPY
AGAINST HEMATOLOGICAL
MALIGNANCIES
Different Types of Immunotherapies for
the Treatment of Hematological
Malignancies
Immunotherapy is becoming the mainstream for many types of
hematological. malignancies. Hematological malignancies are
derived from immune cells and are sittingin immune
microenvironment, thereby having many opportunities to
interact with resident immune cells, local antibodies,
or complement system. This provides unique opportunities
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936106
for immunotherapy. Currently, immunotherapy against
hematological malignancies involves two major approaches: T
cell therapies and antibody therapies. For T cell therapies,
chimeric antigen receptor T (CAR-T) cell therapy is
particularly promising for hematologic malignancies, garnering
two FDA approvals using autologous cells in 2017 (66,67), one
for the treatment of pediatric ALL and the other for adult
patients with advanced lymphomas (68). The main point for
CAR-T therapy is generating engineered T cell by transfection of
mRNA for CAR domain expression which typically consists an
antigen-binding domain, a hinge that connects the scFv to a
transmembrane domain and a signaling domain composed of
CD3z. Antibody therapies include mAb and bispecic antibody.
Blockade of PD-L1/PD-1 interaction has brought about another
advance in immunotherapy for hematological malignancies. The
clinical outcomes of anti-PD-1 mAbs on Hodgkins lymphoma
are particularly impressive (69,70). Combining mAb and CAR-T
cell therapy, bispecic antibodies (BsAbs) were further
developed, which contains one tumor antigen binding side and
another side for binding and activating T cells. Due to the
structure specicity, BsAbs can bridge T cells and target cells,
thereby redirecting and activating T cell at sites of tumor cells
(71). Currently, FBTA05 (Lymphomun) is a heterodimeric BsAb
that recognizes CD20 and CD3, which has been used as
monotherapy or followed by donor lymphocyte infusion in the
treatment of CLL, high grade NHL, ALL, post-transplant
lympho- proliferative disease. In addition, CD123- and CD33-
specic BsAbs have been evaluated in clinical trials for patients
with AML (72).
Current Existing Monoclonal Antibodies
Against Hematological Malignancies
Over the past two decades, the use of mAb and molecules derived
from them has achieved considerable attention and success,
establishing this mode of therapy as important therapeutic
strategy in many cancers, especially in hematological
malignancies. Among mAbs used for the treatment of
hematological malignancies, anti-CD20 is the most routinely
used and well characterized mAb for the treatment of the CD20-
positive NHL and chronic CLL with major therapeutic
advances (73). RTX, ofatumumab, ublituximab, veltuzumab,
ocaratuzumab as well as tositumomab, obinutuzumab are
currently available types. In addition, 97% of patients with
classical HL typically exhibit an overexpression of PD-L1 due
to the alteration in chromosome 9p24.1 (74). Therefore, the PD-
1/PD-L1 axis is a good target for mAbs to kill tumor cells in HL.
Nivolumab, a human IgG4 mAb, blocks the interaction of PD-L1
and PD-L2 by binding to the PD-1 receptor on activated immune
cells, which was already approved by the FDA in 2016 for the
treatment of relapsed or progressed HL (75). The anti-CD33 (e.g.
immunotoxin, gemtuzumab, and ozogamicin) and anti-CD52
mAbs (alemtuzumab) are approved for treatment of relapsed
AML in older patients and B-cell CLL. IGN523, targeting CD90
on the surface of malignant hematological cells (e.g. AML) is
currently being evaluated in a Phase I clinical trial for AML (76).
In addition, monoclonal antibodies targeting CD4, CD19, CD20,
CD22, CD23, CD25, CD45, CD66, and CD122 are also under
investigation in the clinic for the treatment of leukemia (77).
Breakthrough has also been made in targeting surface
molecules expressed by MM cells, such as daratumumab,
isatuximab, MOR202 as well as SAR650984 (different
generations of humanized anti-CD38 monoclonal antibodies)
(78), and elotuzumab, a humanized anti-signaling lymphocytic
activation molecule family member 7 mAb. Among these mAbs,
daratumumab and elotuzumab have been approved in the
treatment of relapsed or refractory MM patients who received
at least three prior therapies including proteasome inhibitors and
immunomodulatory drugs (79). Investigational mAbs targeting
CD138, CD56, CD40, CD74, BAFF, BCMA, GRP78, IGF-1R,
and ICAM-1 on the surface of MM cells are pre-clinically
developed, and several of them are in clinical trials (80).
Effect of Complement on mAb-Based
Immunotherapies
CDC and CDCP display a vital role in mAb-based
immunotherapies (81). Approved for the treatment of
hematological malignancies, most mAbs make use of
complement in their mechanism of action. Upon application
of mAbs, the complement pathways need to be fully effective to
achieve better clinic efcacy. However, complement deciencies,
the over-expression of membrane complement regulatory proteins
(e.g. CD55, CD59, and CD46) and uid phase inhibitors (e.g.
CFH, CFHR5, and C4BP) in the tumor microenvironment often
cause resistance and non-responsiveness to mAb treatment (64,
8284).
Di Gaetano et al. reported that the C1q-decient mice
exhibited a defective response to RTX in a lymphoma tumor
mouse model. The work delineates the importance of CDC as an
important effector mechanism in immunotherapy instead of
ADCC as such an effect was not detected after depletion of
either natural killer cells or granulocyte cells (85). Further, CLL
patients undergoing complement deciencies are suspected of
limiting anti-CD20 mAb efcacy in vivo (83). In CLL, upon
administration of RTX or ofatumumab, complement is quickly
consumed (86,87). C1 and C4 levels were below normal in more
than 50% of the sera tested from CLL patients (57). Only when
concurrent administration of fresh frozen plasma that RTX
therapeutic activity can be restored in CLL patients.
Polymorphisms of C1qA, C5, and C9 were often detected in
FL and DLBCL patients, which may also limit complement
activation, thereby affecting the clinical response to RTX (88).
Furthermore, complement regulators (CD55, CD59 and
Factor H) limited RTX efcacy in CLL patients via down-
regulating CDC (89). Functional block of these regulators
signicantly increased the susceptibility of primary CLL cells to
anti-CD20 mAb. In FL and DLBCL patients, CD46, CD55,
CD59, CFH, CFHR1, and CFHR5 gene expression likely affects
the clinical response and duration of response to RTX therapy
(90). Like CLL and NHL, MM cells also utilize complement
regulators (CD55, CD59, and CFH) to block CDC, thereby
resisting anti-CD38 mediated-antitumor immunotherapy (91).
The serum levels of serglycin are elevated in patients with MM
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936107
compared to healthy controls. Upon mAb treatment, serglycin
protects MM cells from complement attack by blocking CDC,
thereby promoting survival of malignant cells (32). Due to the
different expression levels of each regulator/inhibitor of
individual patients, mAb efcacy varies from patient to patient.
All in all, these comprehensive investigations that were
performed in vitro studies and in mouse models as well as
analyses from clinical patients provide key insights that functional
complement is important for the mAb-based anti-tumor effects
on hematological malignancies (62,63). Down-regulation or
inhibition of complement activation by hematological tumors is a
big challenge for mAb-based immunotherapy.
Improvement of Clinical Efciency of mAb-
Based Immunotherapies
To improve the clinical efciency of mAbs, new strategies should
be developed to initiate efcient complement activation by (1)
mAb modications (such as changing the target epitopes, Fc
mutation, and immunoglobulin G subclass switching), (2) the
control of membrane and soluble complement inhibitors, and (3)
the concurrent administration of fresh frozen plasma during
mAb therapy (84,92).
Despite the clinical success achieved with RTX, incomplete
treatment responses and emergence of resistance represent
important limitations, suggesting further improvements of
anti-CD20 mAb efcacy are required. Many novel anti-
CD20 antibodies are under development either by changing
the target CD20 epitope or by altering the Fc region to
enhance immune effector cell activity (ADCC, ADCP). For
example, compared with RTX, ofatumumab exhibits increased
CDC by binding to a different CD20 epitope (93), while by Fc
alterations, obinutuzumab has increased ADCC, reduced CDC,
and enhanced direct non-apoptotic cell death (94,95). CDC
induction by obinutuzumab is 10 to 100-fold less than by the
RTX and ofatumumab (93), resulting in a further-increased
capacity to bind and activate natural killer cells in the presence
of complement (96). In fact, Fc region engineering includes
modifying the amino acid sequence or the glycosylation
pattern, which allows enhancing both CDC and ADCC
effector functions. Using this technique, four variants of RTX
were generated as a native IgG1, a variant carrying the EFTAE
modication (S267E/H268F/S324T/G236A/I332E) for enhanced
CDC as well as glyco-engineered, non-fucosylated derivatives of
both to boost ADCC. Antibodies with EFTAE modication were
more efcacious in inducing CDC than antibodies with wild-type
sequences due to enhanced C1q binding. Meanwhile, non-
fucosylated variants had an enhanced afnity to FcgRIIIA and
improved ADCC activity (97).
So far several types of anti-CD20 mAbs have been developed,
such as RTX, ofatumumab (OFA), ublituximab, veltuzumab,
ocaratuzumab as well as tositumomab and obinutuzumab.
These variant anti-CD20 mAbs display different clinical
efciency because of CD20-binding characteristics and ability
to induce CDC as well as ADCC (94). Structural analysis of
CD20 by RougeL. et al. nicely explains why some anti-CD20s are
more efcient complement activators than others. The authors
show that CD20 exists as a compact double-barrel dimer, which
can be bound by two RTX antigen-binding fragments (Fabs).
Each of the dimerized CD20 consists two parts, one as a
composite epitope and the other one as an extensive
homotypic Fab: Fab interface. RTX, by cross-linking CD20
into circular assemblies forms a structural model for
complement recruitment, thereby leading to stronger
complement activation (98).KumarA.andhiscolleagues
further used the cryo-electron microscopy to identify
structures of full-length CD20 complexed either with
prototypical type I (RTX and OFA) or type II (Obinutuzumab)
mAbs. Their data showed that type I complexes function as
molecular seeds to increase local concentration of mAbs, thereby
effectively activating complement upon binding to CD20, which
is similar to what RougeL. et al. reported. However, type
II complexes are unable to recruit additional mAbs and
complement components, thereby failing to cause efcient
complement activation. In addition, compared to RTX, OFA
activates complement more potently because of the sharper
binding angle of Fab
OFA
, suggesting that concatenation of
IgG
OFA
seeding complexes may bring their Fc domains in
closer proximity, further facilitating their oligomerization (99).
Human IgG3 activates complement most efciently among
the IgG subclasses. An IgG3 switch variant of RTX induced
better CDC even on low CD20 expressing cells compared with its
parental IgG1 counterpart (100). Further, the shorter serum half-
life of IgG3 can be rescued by the introduction of the R435H
mutation, resulting in a potent mAb for CDC (101). IgG1/IgG3
chimera targeting CD20 showed stronger C1q binding, increased
CDC capacity and more efcient B cell depletion in cynomolgus
monkeys compared with the isotype matched parental mAbs
(102,103).
Furthermore, application of mAb together with inhibitors to
mCRP as an adjuvant can achieve higher efciency. The ongoing
TABLE 1 | Complement-related therapies for treatment of hematological malignancies.
Therapies Functions Targeted molecules Molecular nature References
rILYd4 blocking CD59 regulatory function CD59 30 amino acid fragments (104,105)
hSCR18-20 blocking Factor H regulatory function Factor H recombinant protein (106,107)
MB-59 blocking CD59 regulatory function CD59 mini antibody (108)
MB-55 blocking CD55 regulatory function CD55 mini antibody (108)
mAb A247 blocking Factor I function Factor I neutrolize antibody (102,109)
Sorafenib decreasing the expression of complement regulatory Proteins unknown an oral compound (110)
ATRA decreasing the expression of complement regulatory proteins unknown Metabolic intermediates of vitamin A (91)
ATRA, all-trans retinoic acid.
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936108
clinical trials of complement-related therapies for treatment of
hematological malignancies are summarized in Table 1.
Recombinant ILYd4, a novel CD59 inhibitor, effectively
enhances the RTX-mediated CDC effect on RTX-sensitive RL-
7 lymphoma cells and RTX-induced resistant RR51.2 cells.
Meanwhile, recombinant ILYd4 also enhances the effect of
RTX and anti-CD24 mAb on the refractory MM cell line
ARH-77 (104,105). Similarly, sorafenib potentiates RTX and
ofatumumab efcacy in CLL and HL patients by decreasing the
expression of complement regulatory proteins. Such effect of
sorafenib has been investigated in more than 500 clinical trials
with promising activity and good patientstolerance (110).
Alternatively, mCRP function could be blocked by specic
neutralizingantibodies. Blockage of CD55 and to a lesser
extent of CD59 with specic antibodies in vitro signicantly
increased CDC of B lymphoma cells by RTX (111). However, the
use of intact anti-mCRP antibodies in vivo may lead to CDC on
healthy host cells. Mini-antibodies (MB-55, MB-59), composed
of single-chain variable fragments to CD55 and CD59 and the
human hinge-CH2CH3 domains of IgG1, did not induce CDC
themselves, but increased RTX mediated CDC by twofold in
vitro (108). Their application in an in vivo model of human
CD20 positive B cell lymphoma in SCID mice markedly
increased survival upon RTX treatment (102).
Moreover, neutralization of sCRPs is another way to enhance
CDC. In combination with ofatumumab or RTX, human
recombinant Factor H-derived short-consensus repeat 1820
(hSCR1820) increased susceptibility of primary CLL cells to
CDC by abrogating Factor H function on the surface of CLL cells
(106,107). The decay of C3b to iC3b is strongly mediated by
Factor I, for which most of the described CRPs exhibit cofactor
function. Application of RTX or ofatumumab together with a
neutralizing mAb against Factor I (mAb A247) increased CDC
on CD20-expressing cell lines and primary CLL samples (102,
109). In addition, all-trans retinoic acid was reported to up-
regulate CD38 expression level and down-regulate CD55 and
CD59 level in daratumab-resistant MM cells, thereby enhancing
the CDC effect on MM cells (91).
A clinical investigation by Xu et al. showed that application of
fresh frozen plasma together with RTX is an effective measure to
regain CDC effect upon the treatment of udarabine refractory
CLL patients. Twenty-two patients were treated with two units of
fresh frozen plasma followed with RTX as a single agent, repeated
every 12 weeks with a total of four courses of the combined fresh
frozen plasma and RTX treatment. Sixteen patients (72.7%)
responded to treatment, and seven (31.8%) achieved a complete
remission. Three (13.6%) of them had no evidence of minimal
residual disease after treatment (112). Such a clinical investigation
suggests that the concurrent administration of fresh frozen plasma
during mAb therapy is an optional measure to increase the mAb-
induced clinical efcacy.
CONCLUSIONS
This overview not only serves a fundamental understanding
of the roles of complement in tumor progression and in mAb-
based immunotherapy, but importantly, also highlights
potential therapeutic targets/measures to improve the clinical
efcacy of mAbs against hematological malignancies and
further extrapolates this knowledge to other tumor related
diseases. However, the mechanisms how complement affects
hematological malignanciesdevelopment and which strategy
increases mAbsefcacy most clinically relevant are still elusive
and need further investigation.
AUTHOR CONTRIBUTIONS
SL designed the outline of the manuscript, wrote, and reviewed
the manuscript. MW wrote and reviewed the manuscript. HW
revised the manuscript. DH wrote and reviewed the manuscript.
YH and PZ discussed the topic and outlines of the manuscript
and reviewed the text. All authors contributed to the article and
approved the submitted version.
FUNDING
This work was supported by the National Natural Science
Foundations of China (No. 81601747, 82070136 to SL and No.
31770983, 81974249 to DH).
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found online
at: https://www.frontiersin.org/articles/10.3389/mmu.2020.
593610/full#supplementary-material
REFERENCES
1. Ehrlich P, Sachs H. Ueber die Vielheit der Complemente des Serums. Berl
Klin Wschr (2020) 39:2979.
2. Zipfel PF, Skerka C. Complement regulators and inhibitory proteins. Nat
Rev Immunol (2009) 9(10):72940. doi: 10.1038/nri2620
3. Walport MJ. Complement. First of two parts. N Engl J Med (2001) 344
(14):105866. doi: 10.1056/NEJM200104053441406
4. Walport MJ. Complement. Second of two parts. N Engl J Med (2001) 344
(15):11404. doi: 10.1056/NEJM200104123441506
5. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global
cancer statistics 2018: GLOBOCAN estimates of incidence and mortality
worldwide for 36 cancers in 185 countries. CA: Cancer J Clin (2018) 68
(6):394424. doi: 10.3322/caac.21492
6. Kansara RR, Speziali C. Immunotherapy in hematologic malignancies. Curr
Oncol (Toronto Ont) (2020) 27(Suppl 2):S124s131. doi: 10.3747/co.27.5117
7. Chaudhari K, Rizvi S, Syed BA. Non-Hodgkin lymphoma therapy
landscape. Nat Rev Drug Discov (2019) 18(9):6634. doi: 10.1038/d41573-
019-00051-6
8. Byrd JC, Jones JJ, Woyach JA, Johnson AJ, Flynn JM. Entering the era of
targeted therapy for chronic lymphocyticleukemia:impactonthepracticing
clinician. JClinOncol(2014) 32(27):303947. doi: 10.1200/JCO.2014.55.8262
9. Mihlan M, Stippa S, Jozsi M, Zipfel PF. Monomeric CRP contributes to
complement control in uid phase and on cellular surfaces and increases
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 5936109
phagocytosis by recruiting factor H. Cell Death Diff (2009) 16(12):163040.
doi: 10.1038/cdd.2009.103
10. van Lookeren Campagne M, Wiesmann C, Brown EJ. Macrophage
complement receptors and pathogen clearance. Cell Microbiol (2007) 9
(9):2095102. doi: 10.1111/j.1462-5822.2007.00981.x
11. Luo S, Hartmann A, Dahse HM, Skerka C, Zipfel PF. Secreted pH-regulated
antigen 1 of Candida albicans blocks activation and conversion of
complement C3. J Immunol (2010) 185(4):216473. doi: 10.4049/jimmunol.
1001011
12. Malmsten M, Schmidtchen A. Antimicrobial C3abiology, biophysics, and
evolution. Adv Exp Med Biol (2007) 598:14158. doi: 10.1007/978-0-387-
71767-8_11
13. Verschoor A, Brockman MA, Gadjeva M, Knipe DM, Carroll MC. Myeloid
C3 determines induction of humoral responses to peripheral herpes simplex
virus infection. J Immunol (2003) 171(10):536371. doi: 10.4049/jimmunol.
171.10.5363
14. Gadjeva M, Verschoor A, Brockman MA, Jezak H, Shen LM, Knipe DM,
et al. Macrophage-derived complement component C4 can restore humoral
immunity in C4-decient mice. J Immunol (2002) 169(10):548995. doi:
10.4049/jimmunol.169.10.5489
15. Verschoor A, Neuenhahn M, Navarini AA, Graef P, Plaumann A, Seidlmeier
A, et al. A platelet-mediated system for shuttling blood-borne bacteria to
CD8alpha+ dendritic cells depends on glycoprotein GPIb and complement
C3. Nat Immunol (2011) 12(12):1194201. doi: 10.1038/ni.2140
16. Li Q, Peng Q, Xing G, Li K, Wang N, Farrar CA, et al. Deciency of C5aR
prolongs renal allograft survival. J Am Soc Nephrol JASN (2010) 21(8):1344
53. doi: 10.1681/ASN.2009090977
17. Kemper C, Atkinson JP. T-cell regulation: with complements from innate
immunity. Nat Rev Immunol (2007) 7(1):918. doi: 10.1038/nri1994
18. Clarke EV, Tenner AJ. Complement modulation of T cell immune responses
during homeostasis and disease. J Leukocyte Biol (2014) 96(5):74556. doi:
10.1189/jlb.3MR0214-109R
19. Mohlin FC, Blom AM. Purication and functional characterization of C4b-
binding protein (C4BP). Methods Mol Biol (2014) 1100:16976. doi:
10.1007/978-1-62703-724-2_14
20. Zipfel PF, Skerka C, Hellwage J, Jokiranta ST, Meri S, Brade V, et al. Factor H
family proteins: on complement, microbes and human diseases. Biochem Soc
Trans (2002) 30(Pt 6):9718. doi: 10.1042/bst0300971
21. Lupu F, Keshari RS, Lambris JD, Coggeshall KM. Crosstalk between the
coagulation and complement systems in sepsis. Thromb Res (2014) 133
Suppl 1(0 1):S2831. doi: 10.1016/j.thromres.2014.03.014
22. Seya T. [Cell-associated complement regulatory proteins and their relation to
disease processes]. Rinsho Byori Japan J Clin Pathol (1991) 39(12):131724.
23. Wang SY, Weiner G. Complement and cellular cytotoxicity in antibody
therapy of cancer. Expert Opin Biol Ther (2008) 8(6):75968. doi: 10.1517/
14712598.8.6.759
24. Robak T, Robak E. New anti-CD20 monoclonal antibodies for the treatment
of B-cell lymphoid malignancies. BioDrugs Clin Immunother Biopharmaceut
Gene Ther (2011) 25(1):1325. doi: 10.2165/11539590-000000000-00000
25. Fedarko NS, Fohr B, Robey PG, Young MF, Fisher LW. Factor H binding to
bone sialoprotein and osteopontin enables tumor cell evasion of
complement-mediated attack. J Biol Chem (2000) 275(22):1666672. doi:
10.1074/jbc.M001123200
26.YoonYH,HwangHJ,SungHJ,HeoSH,KimDS,HongSH,etal.
Upregulation of Complement Factor H by SOCS-1/3(-)STAT4 in Lung
Cancer. Cancers (2019) 11(4):47186. doi: 10.3390/cancers11040471
27. Seguin-Devaux C, Plesseria JM, Verschueren C, Masquelier C, Iserentant G,
Fullana M, et al. FHR4-based immunoconjugates direct complement-
dependent cytotoxicity and phagocytosis towards HER2-positive cancer
cells. Mol Oncol (2019) 13(12):253153. doi: 10.1002/1878-0261.12554
28. Martin M, Gottsater A, Nilsson PM, Mollnes TE, Lindblad B, Blom AM.
Complement activation and plasma levels of C4b-binding protein in critical
limb ischemia patients. J Vasc Surg (2009) 50(1):1006. doi: 10.1016/
j.jvs.2008.12.033
29. Junnikkala S, Hakulinen J, Jarva H, Manuelian T, Bjorge L, Butzow R, et al.
Secretion of soluble complement inhibitors factor H and factor H-like
protein (FHL-1) by ovarian tumour cells. Br J Cancer (2002) 87(10):1119
27. doi: 10.1038/sj.bjc.6600614
30. Chen J, Ding P, Li L, Gu H, Zhang X, Zhang L, et al. CD59 Regulation by
SOX2 Is Required for Epithelial Cancer Stem Cells to Evade Complement
Surveillance. Stem Cell Rep (2017) 8(1):14051. doi: 10.1016/j.stemcr.2016.
11.008
31. Yin X, Shen C, Yin Y, Cai Z, Wang J, Zhao Z, et al. Overexpression of
CD55 correlates with tumor progression and poor prognosis in gastric
stromal tumors. OncoTargets Ther (2019) 12:470312. doi: 10.2147/
OTT.S195182
32. Skliris A, Happonen KE, Terpos E, Labropoulou V, Børset M, Heinegård D,
et al. Serglycin inhibits the classical and lectin pathways of complement via
its glycosaminoglycan chains: implications for multiple myeloma. Eur J
Immunol (2011) 41(2):43749. doi: 10.1002/eji.201040429
33. Reis ES, Mastellos DC, Ricklin D, Mantovani A, Lambris JD. Complement in
cancer: untangling an intricate relationship. Nat Rev Immunol (2018) 18
(1):518. doi: 10.1038/nri.2017.97
34. Cho MS, Vasquez HG, Rupaimoole R, Pradeep S, Wu S, Zand B, et al.
Autocrine effects of tumor-derived complement. Cell Rep (2014) 6(6):1085
95. doi: 10.1016/j.celrep.2014.02.014
35. Shu C, Zha H, Long H, Wang X, Yang F, Gao J, et al. C3a-C3aR signaling
promotes breast cancer lung metastasis via modulating carcinoma associated
broblasts. J Exp Clin Cancer Res (2020) 39(1):11. doi: 10.1186/s13046-019-
1515-2
36. Nitta H, Wada Y, Kawano Y, Murakami Y, Irie A, Taniguchi K, et al.
Enhancement of human cancer cell motility and invasiveness by
anaphylatoxin C5a via aberrantly expressed C5a receptor (CD88). Clin
Cancer Res (2013) 19(8):200413. doi: 10.1158/1078-0432.CCR-12-1204
37. Markiewski MM, DeAngelis RA, Benencia F, Ricklin-Lichtsteiner SK,
Koutoulaki A, Gerard C, et al. Modulation of the antitumor immune
response by complement. Nat Immunol (2008) 9(11):122535. doi:
10.1038/ni.1655
38. Vadrevu SK, Chintala NK, Sharma SK, Sharma P, Cleveland C, Riediger L,
et al. Complement c5a receptor facilitates cancer metastasis by altering T-cell
responses in the metastatic niche. Cancer Res (2014) 74(13):345465. doi:
10.1158/0008-5472.CAN-14-0157
39. Nabizadeh JA, Manthey HD, Panagides N, Steyn FJ, Lee JD, Li XX, et al. C5a
receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse
model of melanoma. FASEB J (2019) 33(10):1106071. doi: 10.1096/
fj.201800980RR
40. Mangogna A, Agostinis C, Bonazza D, Belmonte B, Zacchi P, Zito G, et al. Is
the Complement Protein C1q a Pro- or Anti-tumorigenic Factor?
Bioinformatics Analysis Involving Human Carcinomas. Front Immunol
(2019) 10:865. doi: 10.3389/mmu.2019.00865
41. Roumenina LT, Daugan MV, NoeR, Petitprez F, Vano YA, Sanchez-Salas R,
et al. Tumor Cells Hijack Macrophage-Produced Complement C1q to
Promote Tumor Growth. Cancer Immunol Res (2019) 7(7):1091105. doi:
10.1158/2326-6066.CIR-18-0891
42. Yuan K, Ye J, Liu Z, Ren Y, He W, Xu J, et al. Complement C3
overexpression activates JAK2/STAT3 pathway and correlates with gastric
cancer progression. J Exp Clin Cancer Res (2020) 39(1):9. doi: 10.1186/
s13046-019-1514-3
43. Xu Y, Huang Y, Xu W, Zheng X, Yi X, Huang L, et al. Activated Hepatic
Stellate Cells (HSCs) Exert Immunosuppressive Effects in Hepatocellular
Carcinoma by Producing Complement C3. OncoTargets Ther (2020)
13:1497505. doi: 10.2147/OTT.S234920
44. Vlaicu SI, Tatomir A, Rus V, Rus H. Role of C5b-9 and RGC-32 in Cancer.
Front Immunol (2019) 10:1054. doi: 10.3389/mmu.2019.01054
45. Riihilä P, Nissinen L, Knuutila J, Rahmati Nezhad P, Viiklepp K, Kähäri VM.
Complement System in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci
(2019) 20(14):3350570. doi: 10.3390/ijms20143550
46. Kleczko EK, Kwak JW, Schenk EL, Nemenoff RA. Targeting the
Complement Pathway as a Therapeutic Strategy in Lung Cancer. Front
Immunol (2019) 10:954. doi: 10.3389/mmu.2019.00954
47. Wang Y, Zhang H, He YW. The Complement Receptors C3aR and C5aR
Are a New Class of Immune Checkpoint Receptor in Cancer
Immunotherapy. Front Immunol (2019) 10:1574. doi: 10.3389/mmu.
2019.01574
48. Wang H, Li Y, Shi G, Wang Y, Lin Y, Wang Q, et al. A Novel Antitumor
Strategy: Simultaneously Inhibiting Angiogenesis and Complement by
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 59361010
Targeting VEGFA/PIGF and C3b/C4b. Mol Ther Oncol (2020) 16:209. doi:
10.1016/j.omto.2019.12.004
49. Yang R, Huang J, Ma H, Li S, Gao X, Liu Y, et al. Is complement C1q a
potential marker for tumor burden and immunodeciency in multiple
myeloma? Leukemia Lymphoma (2019) 60(7):18128. doi: 10.1080/
10428194.2018.1543883
50. Cheson BD, Walker HS, Heath ME, Gobel RJ, Janatova J. Defective binding
of the third component of complement (C3) to Streptococcus pneumoniae
in multiple myeloma. Blood (1984) 63(4):94957. doi: 10.1182/blood.
V63.4.949.949
51. Li CJ, Xia HL, Wu YM, Ding G, Xu DD. [Signicance of Detecting Serum
Complement C3 and C4 in Patients with Multiple Myeloma]. Zhongguo Shi
Yan Xue Ye Xue Za Zhi (2019) 27(2):4726.
52. Swierzko AS, Michalski M, Sokolowska A, Nowicki M, Szala-Pozdziej A,
Eppa L, et al. Associations of Ficolins With Hematological Malignancies in
Patients Receiving High-Dose Chemotherapy and Autologous
Hematopoietic Stem Cell Transplantations. Front Immunol (2019)
10:3097. doi: 10.3389/mmu.2019.03097
53. Swierzko AS, Michalski M, Sokolowska A, Nowicki M, Eppa L, Szala-
Pozdziej A, et al. The Role of Complement Activating Collectins and
Associated Serine Proteases in Patients With Hematological Malignancies,
Receiving High-Dose Chemotherapy, and Autologous Hematopoietic Stem
Cell Transplantations (Auto-HSCT). Front Immunol (2018) 9:2153. doi:
10.3389/mmu.2018.02153
54. Lenkiewicz A, Bujko K, Brzezniakiewicz-Janus K, Xu B, Ratajczak MZ. The
Complement Cascade as a Mediator of Human Malignant Hematopoietic
Cell Trafcking. Front Immunol (2019) 10:1292. doi: 10.3389/mmu.
2019.01292
55. Michelis R, Tadmor T, Barhoum M, Shehadeh M, Shvidel L, Aviv A, et al. A
C5a-Immunoglobulin complex in chronic lymphocytic leukemia patients is
associated with decreased complement activity. PLoS One (2019) 14(1):
e0209024. doi: 10.1371/journal.pone.0209024
56. Heath ME, Cheson BD. Defective complement activity in chronic
lymphocytic leukemia. Am J Hematol (1985) 19(1):6373. doi: 10.1002/
ajh.2830190109
57. Füst G, Czink E, Minh D, Miszlay Z, Varga L, Hollan SR. Depressed classical
complement pathway activities in chronic lymphocytic leukaemia. Clin Exp
Immunol (1985) 60(3):48995.
58. Minh DQ, Czink E, Mod A, Fust G, Hollan SR. Serial complement
measurements in patients with leukaemia. Clin Lab Haematol (1983) 5
(1):2334. doi: 10.1111/j.1365-2257.1983.tb00493.x
59. Sadallah S, Lach E, Schwarz S, Gratwohl A, Spertini O, Schifferli JA. Soluble
complement receptor 1 is increased in patients with leukemia and after
administration of granulocyte colony-stimulating factor. J Leukocyte Biol
(1999) 65(1):94101. doi: 10.1002/jlb.65.1.94
60. Cerhan JR, Novak AJ, Fredericksen ZS, Wang AH, Liebow M, Call TG, et al.
Risk of non-Hodgkin lymphoma in association with germline variation in
complement genes. Br J Haematol (2009) 145(5):61423. doi: 10.1111/
j.1365-2141.2009.07675.x
61. Hu W, Bassig BA, Xu J, Zheng T, Zhang Y, Berndt SI, et al. Polymorphisms
in pattern-recognition genes in the innate immunity system and risk of non-
Hodgkin lymphoma. Environ Mol Mutagen (2013) 54(1):727. doi: 10.1002/
em.21739
62. Charbonneau B, Maurer MJ, Fredericksen ZS, Zent CS, Link BK, Novak AJ,
et al. Germline variation in complement genes and event-free survival in
follicular and diffuse large B-cell lymphoma. Am J Hematol (2012) 87
(9):8805. doi: 10.1002/ajh.23273
63. Racila E, Link BK, Weng WK, Witzig TE, Ansell S, Maurer MJ, et al. A
polymorphism in the complement component C1qA correlates with
prolonged response following rituximab therapy of follicular lymphoma.
Clin Cancer Res (2008) 14(20):6697703. doi: 10.1158/1078-0432.CCR-08-
0745
64. Dzietczenia J, Wrobel T, Mazur G, Poreba R, Jazwiec B, Kuliczkowski K.
Expression of complement regulatory proteins: CD46, CD55, and CD59
and response to rituximab in patients with CD20+ non-Hodgkins
lymphoma. Med Oncol (2010) 27(3):7436. doi: 10.1007/s12032-009-
9278-9
65. Roumenina LT, Daugan MV, Petitprez F, Sautès-Fridman C, Fridman WH.
Context-dependent roles of complement in cancer. Nat Rev Cancer (2019)
19(12):698715. doi: 10.1038/s41568-019-0210-0
66. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA,
et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-
Cell Lymphoma. N Engl J Med (2017) 377(26):253144.
67. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al.
Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic
Leukemia. NEnglJMed(2018) 378(5):43948. doi: 10.1056/NEJMoa
1709866
68. Mochel JP, Ekker SC, Johannes CM, Jergens AE, Allenspach K, Bourgois-
Mochel A, et al. CAR T Cell Immunotherapy in Human and Veterinary
Oncology: Changing the Odds Against Hematological Malignancies. AAPS J
(2019) 21(3):50. doi: 10.1208/s12248-019-0322-1
69. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al.
PD-1 blockade with nivolumab in relapsed or refractory Hodgkinslymphoma.
N Engl J Med (2015) 372(4):3119. doi: 10.1056/NEJMoa1411087
70. Chen R, Zinzani PL, Fanale MA, Armand P, Johnson NA, Brice P, et al.
Phase II Study of the Efcacy and Safety of Pembrolizumab for Relapsed/
Refractory Classic Hodgkin Lymphoma. J Clin Oncol (2017) 35(19):212532.
doi: 10.1200/JCO.2016.72.1316
71. Viardot A, Bargou R. Bispecic antibodies in haematological malignancies.
Cancer Treat Rev (2018) 65:8795. doi: 10.1016/j.ctrv.2018.04.002
72. Velasquez MP, Bonifant CL, Gottschalk S. Redirecting T cells to
hematological malignancies with bispecic antibodies. Blood (2018) 131
(1):308. doi: 10.1182/blood-2017-06-741058
73. Okroj M, Eriksson I, Osterborg A, Blom AM. Killing of CLL and NHL cells
by rituximab and ofatumumab under limited availability of complement.
Med Oncol (2013) 30(4):759. doi: 10.1007/s12032-013-0759-5
74. Ray A, Das DS, Song Y, Richardson P, Munshi NC, Chauhan D, et al.
Targeting PD1-PDL1 immune checkpoint in plasmacytoid dendritic cell
interactions with T cells, natural killer cells and multiple myeloma cells.
Leukemia (2015) 29(6):14414. doi: 10.1016/j.ctrv.2015.07.003
75. Jelinek T, Mihalyova J, Kascak M, Duras J, Hajek R. PD-1/PD-L1 inhibitors
in haematological malignancies: update 2017. Immunology (2017) 152
(3):35771. doi: 10.1038/leu.2015.11
76. Hayes GM, Chinn L, Cantor JM, Cairns B, Levashova Z, Tran H, et al.
Antitumor activity of an anti-CD98 antibody. Int J Cancer (2015) 137
(3):71020. doi: 10.1111/imm.12788
77. Morris JC, Waldmann TA. Antibody-based therapy of leukaemia. Expert Rev
Mol Med (2009) 11:e29. doi: 10.1002/ijc.29415
78. van de Donk NW, Janmaat ML, Mutis T, Lammerts van Bueren JJ, Ahmadi
T, Sasser AK, et al. Monoclonal antibodies targeting CD38 in hematological
malignancies and beyond. Immunol Rev (2016) 270(1):95112. doi: 10.1017/
S1462399409001215
79. Abramson HN. Monoclonal Antibodies for the Treatment of Multiple
Myeloma: An Update. Int J Mol Sci (2018) 19(12):392454. doi: 10.1111/
imr.12389
80.NishidaH,YamadaT.Monoclonal Antibody Therapies in Multiple
Myeloma: A Challenge to Develop Novel Targets. JOncol(2019)
2019:6084012. doi: 10.3390/ijms19123924
81. Lee CH, Romain G, Yan W, Watanabe M, Charab W, Todorova B, et al. IgG
Fc domains that bind C1q but not effector Fcgreceptors delineate the
importance of complement-mediated effector functions. Nat Immunol
(2017) 18(8):88998. doi: 10.1155/2019/6084012
82. Guc D, Canpinar H, Kucukaksu C, Kansu E. Expression of complement
regulatory proteins CR1, DAF, MCP and CD59 in haematological
malignancies. Eur J Haematol (2000) 64(1):39. doi: 10.1038/ni.3770
83. Middleton O, Cosimo E, Dobbin E, McCaig AM, Clarke C, Brant AM, et al.
Complement deciencies limit CD20 monoclonal antibody treatment
efcacy in CLL. Leukemia (2015) 29(1):10714. doi: 10.1034/j.1600-
0609.2000.80097.x
84. Bordron A, Bagacean C, Tempescul A, Berthou C, Bettacchioli E, Hillion S,
et al. Complement System: a Neglected Pathway in Immunotherapy. Clin
Rev Allergy Immunol (2020) 58(2):15571. doi: 10.1038/leu.2014.146
85. Di Gaetano N, Cittera E, Nota R, Vecchi A, Grieco V, Scanziani E, et al.
Complement activation determines the therapeutic activity of rituximab in
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 59361011
vivo. J Immunol (Baltimore Md 1950) (2003) 171(3):15817. doi: 10.1007/
s12016-019-08741-0
86. Kennedy AD, Beum PV, Solga MD, DiLillo DJ, Lindorfer MA, Hess CE, et al.
Rituximab infusion promotes rapid complement depletion and acute CD20
loss in chronic lymphocytic leukemia. J Immunol (Baltimore Md 1950)
(2004) 172(5):32808. doi: 10.4049/jimmunol.171.3.1581
87. Tempescul A, Bagacean C, Riou C, Bendaoud B, Hillion S, Debant M, et al.
Ofatumumab capacity to deplete B cells from chronic lymphocytic
leukaemia is affected by C4 complement exhaustion. Eur J Haematol
(2016) 96(3):22935. doi: 10.4049/jimmunol.172.5.3280
88. Klepsh A, Gilles L, Ioannis K, Rachmilewitz EA, Schattner A. Enhancing
the action of rituximab in chronic lymphocytic leukemia by adding fresh
frozen plasma: complement/rituximab interactions & clinical results in
refractory CLL. Ann N Y Acad Sci (2009) 1173:86573. doi: 10.1111/
ejh.12573
89. Golay J, Lazzari M, Facchinetti V, Bernasconi S, Borleri G, Barbui T, et al.
CD20 levels determine the in vitro susceptibility to rituximab and
complement of B-cell chronic lymphocytic leukemia: further regulation by
CD55 and CD59. Blood (2001) 98(12):33839. doi: 10.1111/j.1749-
6632.2009.04803.x
90. Weng WK, Levy R. Expression of complement inhibitors CD46, CD55, and
CD59 on tumor cells does not predict clinical outcome after rituximab
treatment in follicular non-Hodgkin lymphoma. Blood (2001) 98(5):13527.
doi: 10.1182/blood.V98.12.3383
91. Nijhof IS, Casneuf T, van Velzen J, van Kessel B, Axel AE, Syed K, et al.
CD38 expression and complement inhibitors affect response and resistance
to daratumumab therapy in myeloma. Blood (2016) 128(7):95970. doi:
10.1182/blood.V98.5.1352
92. Taylor RP, Lindorfer MA. Cytotoxic mechanisms of immunotherapy:
Harnessing complement in the action of anti-tumor monoclonal
antibodies. Semin Immunol (2016) 28(3):30916. doi: 10.1182/blood-2016-
03-703439
93. Herter S, Herting F, Mundigl O, Waldhauer I, Weinzierl T, Fauti T, et al.
Preclinical activity of the type II CD20 antibody GA101 (obinutuzumab)
compared with rituximab and ofatumumab in vitro and in xenograft models.
Mol Cancer Ther (2013) 12(10):203142. doi: 10.1016/j.smim.2016.03.003
94. Illidge T, Klein C, Sehn LH, Davies A, Salles G, Cartron G. Obinutuzumab in
hematologic malignancies: lessons learned to date. Cancer Treat Rev (2015)
41(9):78492. doi: 10.1158/1535-7163.MCT-12-1182
95. Cheney CM, Stephens DM, Mo X, Raq S, Butchar J, Flynn JM, et al.
Ocaratuzumab, an Fc-engineered antibody demonstrates enhanced
antibody-dependent cell-mediated cytotoxicity in chronic lymphocytic
leukemia. mAbs (2014) 6(3):74955. doi: 10.4161/mabs.28282
96. Kern DJ, James BR, Blackwell S, Gassner C, Klein C, Weiner GJ. GA101
induces NK-cell activation and antibody-dependent cellular cytotoxicity
more effectively than rituximab when complement is present. Leukemia
Lymphoma (2013) 54(11):25005. doi: 10.3109/10428194.2013.781169
97. Wirt T, Rosskopf S, Rösner T, Eichholz KM, Kahrs A, Lutz S, et al. An Fc
Double-Engineered CD20 Antibody with Enhanced Ability to Trigger
Complement-Dependent Cytotoxicity and Antibody-Dependent Cell-
Mediated Cytotoxicity. Transfusion Med Hemother Ofzielles Organ Der
Deutschen Gesellschaft Fur Transfusionsmedizin Und Immunhamatol (2017)
44(5):292300. doi: 10.1159/000479978
98. RougeL, Chiang N, Steffek M, Kugel C, Croll TI, Tam C, et al. Structure of
CD20 in complex with the therapeutic monoclonal antibody rituximab. Sci
(N Y NY) 2020 (6483) 367:122430. doi: 10.1126/science.aaz9356
99. Kumar A, Planchais C, Fronzes R, Mouquet H, Reyes N. Binding
mechanisms of therapeutic antibodies to human CD20. Sci (N Y NY) 2020
Aug 14 (6505) 369:7939. doi: 10.1126/science.abb8008
100. Rösner T, Derer S, Kellner C, Dechant M, Lohse S, Vidarsson G, et al. An
IgG3 switch variant of rituximab mediates enhanced complement-dependent
cytotoxicity against tumour cells with low CD20 expression levels. Br J
Haematol (2013) 161(2):2826. doi: 10.1111/bjh.12209
101. Stapleton NM, Andersen JT, Stemerding AM, Bjarnarson SP, Verheul RC,
Gerritsen J, et al. Competition for FcRn-mediated transport gives rise to
short half-life of human IgG3 and offers therapeutic potential. Nat Commun
(2011) 2:599. doi: 10.1038/ncomms1608
102. Meyer S, Leusen JH, Boross P. Regulation of complement and modulation of
its activity in monoclonal antibody therapy of cancer. mAbs (2014) 6
(5):113344. doi: 10.4161/mabs.29670
103. Natsume A, In M, Takamura H, Nakagawa T, Shimizu Y, Kitajima K, et al.
Engineered antibodies of IgG1/IgG3 mixed isotype with enhanced cytotoxic
activities. Cancer Res (2008) 68(10):386372. doi: 10.1158/0008-5472.CAN-
07-6297
104. You T, Hu W, Ge X, Shen J, Qin X. Application of a novel inhibitor of human
CD59 for the enhancement of complement-dependent cytolysis on cancer
cells. Cell Mol Immunol (2011) 8(2):15763. doi: 10.1038/cmi.2010.35
105. Wu L, Su S, Liu F, Xu T, Wang X, Huang Y, et al. Removal of the tag from
His-tagged ILYd4, a human CD59 inhibitor, signicantly improves its
physical properties and its activity. Curr Pharm Design (2012) 18
(27):418796. doi: 10.2174/138161212802430486
106. Hörl S, Banki Z, Huber G, Ejaz A, Müllauer B, Willenbacher E, et al.
Complement factor H-derived short consensus repeat 18-20 enhanced
complement-dependent cytotoxicity of ofatumumab on chronic
lymphocytic leukemia cells. Haematologica (2013) 98(12):193947. doi:
10.1038/bcj.2015.27
107. Hörl S, Banki Z, Huber G, Ejaz A, Windisch D, Muellauer B, et al. Reduction
of complement factor H binding to CLL cells improves the induction of
rituximab-mediated complement-dependent cytotoxicity. Leukemia (2013)
27(11):22008. doi: 10.1182/blood.V95.12.3900
108. Ziller F, Macor P, Bulla R, Sblattero D, Marzari R, Tedesco F. Controlling
complement resistance in cancer by using human monoclonal antibodies
that neutralize complement-regulatory proteins CD55 and CD59. Eur J
Immunol (2005) 35(7):217583. doi: 10.1002/eji.200425920
109. Lindorfer M, Beum P, Taylor R. CD20 mAb-Mediated Complement
Dependent Cytotoxicity of Tumor Cells is Enhanced by Blocking the
Action of Factor I. Antibodies (2013) 2(4):598616. doi: 10.3324/
haematol.2013.089615
110. Dwojak M, Bobrowicz M, Bil J, Bojarczuk K, Pyrzynska B, Siernicka M, et al.
Sorafenib improves rituximab and ofatumumab efcacy by decreasing the
expression of complement regulatory proteins. Blood Cancer J (2015) 5(4):
e300. doi: 10.1038/leu.2013.169
111. Golay J, Zaffaroni L, Vaccari T, Lazzari M, Borleri GM, Bernasconi S, et al.
Biologic response of B lymphoma cells to anti-CD20 monoclonal antibody
rituximab in vitro: CD55 and CD59 regulate complement-mediated cell lysis.
Blood (2000) 95(12):39008. doi: 10.1182/blood.V95.12.3900.012k14_
3900_3908
112. Xu W, Miao KR, Zhu DX, Fang C, Zhu HY, Dong HJ, et al. Enhancing the
action of rituximab by adding fresh frozen plasma for the treatment of
udarabine refractory chronic lymphocytic leukemia. Int J Cancer (2011) 128
(9):2192201. doi: 10.1002/ijc.25560
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2020 Luo, Wang, Wang, Hu, Zipfel and Hu. This is an open-access
article distributed under the terms of the Creative Commons Attribution License
(CC BY). The use,distribution or reproduction in other forums is permitted, provided
the original author(s) and the copyright owner(s) are credited and that the original
publicationin this journal is cited, in accordancewith acceptedacademic practice. No
use, distribution or reproduction is permitted which does not comply with these terms.
Luo et al. Hematological Malignancies and Complement
Frontiers in Immunology | www.frontiersin.org October 2020 | Volume 11 | Article 59361012
... Upon complement activation, it attacks the cells with antigens, inducing cell damage and loss of function in these cells. The antibody involved in the complement fixation reaction is primarily IgG, which contains a complement binding site (the Fab) in the Fc segment (CH2 region) of its heavy chains (11)(12)(13). Of the three immunological approaches mentioned above, the most critical challenge is to obtain antibodies against cell surface specific antigens. ...
Article
Full-text available
Sex determination is the developmental assignment that results from genetic factors. The sexual characters were the specific manifestations of male and female individuals under stimulation of sexual hormonal production. The fusion of an oocyte with an X chromosome-bearing sperm will lead to a female (XX), while fusion with a Y chromosome-bearing sperm will develop into a male (XY) in mammals. Sexing technology has been developed to fertilize eggs with sorted sperm, producing offspring of the desired sex. Sperm sorting enables the sex pre-determination of offspring via in vitro fertilization (IVF) or artificial insemination (AI) in domestic animals. Flow cytometric sorting of X and Y sperm is widely considered the most applied method for sperm sorting and has been commercially applied in cattle. However, a non-invasive, immunological method for screening X and Y sperm is considered to be a feasible approach. This review summarizes the current knowledge and techniques of sperm immunological sorting, including the preparation of antibodies, application of immunomodulators, and immunoisolation. Additionally, we focus on identifying sex-specifically expressed proteins in X and Y sperm through proteomic analysis, and verifying the sex-specific proteins using experimental techniques. Furthermore, several housekeeping proteins as loading control were discussed in immunoblotting of sperm proteins. Immunological sorting of X and Y sperm could provide a convenient, cost-effective, and highly efficient technique that can improve economic benefits and achieve an advanced level of sexing technology. This review provides insight into immunological sorting of sperm and the pre-determination of sex in farm animals.
... Furthermore, the complement system plays a complex role in the occurrence and development of tumors, functioning as a double-edged sword (1,2). Patients frequently present abnormal levels of certain complement components, while synthesis sites also demonstrate higher diversity and heterogeneity (3,4). Complement components play a role in anti-tumor immunity through the complement dependent cytotoxicity (CDC) pathway, antibodydependent cell-dependent cytotoxicity (ADCC), and antibody dependent cell mediated phagocytosis (ADCP), which can be considered the favorable side of the double-edged sword (5). ...
Article
Full-text available
The domain of cancer treatment has persistently been confronted with significant challenges, including those associated with recurrence and drug resistance. The complement system, which serves as the foundation of the innate immune system, exhibits intricate and nuanced dual characteristics in the evolution of tumors. On the one hand, the complement system has the capacity to directly inhibit cancer cell proliferation via specific pathways, thereby exerting a beneficial anti-tumor effect. Conversely, the complement system can also facilitate the establishment of an immune escape barrier for cancer cells through non-complement-mediated mechanisms, thereby protecting them from eradication. Concurrently, the complement system can also be implicated in the emergence of drug resistance through a multitude of complex mechanisms, directly or indirectly reducing the efficacy of therapeutic interventions and facilitating the progression of cancer. This paper analyses the role of the complement system in tumors and reviews recent research advances in the mechanisms of tumor immune tolerance and drug resistance.
... The membrane attack complex (Membrane attack complex, MAC) is the final substance formed by the three pathways of the complement system MAC cannot only lyse infected cells but also kill cancer cells and inhibit cancer growth, while complement regulatory proteins (Complement regulatory proteins, CRPs) prevent the complement over-activation and reduce the efficiency of MAC production [86]. The results of a mouse model of non-small cell lung cancer showed that inhibition of CRP by immunomodulators could slow down cancer growth in mice, further confirming that the complement system is involved in the regulation of cancer growth [87]. ...
Article
Full-text available
Patients with malignant tumors are prone to present hypercoagulability of blood and form thrombosis, and its pathogenesis is complex involving various factors from clinical and histopathological to genetic influences. Current studies on the potential mechanism of blood hypercoagulability in patients with malignant tumors focus on the following aspects but are not limited: (1) tumor cells release coagulant-promoting substances, (2) tumor cells interact with the fibrinolytic system, (3) tumor cell-mediated platelet activation, (4) tumor-associated complement activation, and (5) genetic factors and clinical factors. Especially, the pathogenesis of blood hypercoagulability is in-depth analyzed covering tumor cells' release of procoagulant substances, the interplay of cancer cells and fibrinolytic system, platelet activation mediated by cancer cells, cancer-associated complement activation, and the action of genetic and clinical factors. We review the pathogenesis of blood hypercoagulability in patients with malignant tumors, which will assist in the research and development of new drugs and providing theoretical support for the formulation of the best treatment plan for patients, to prolong the survival of patients.
... Conversely, the EV RNAs exclusive to the B-ALL samples we investigated include C3, a complement protein, which is a serum effector of innate immunity. High C3 levels indicate inflammation, which may be associated with many hematological malignancies (73). Other top genes include ODAD4, which is associated with ALL (74), LINC02568, which is linked to squamous carcinoma (75) and MRPS31, which is a driver of carcinoma (76). ...
Article
Full-text available
Pediatric B-acute lymphoblastic leukemia (B-ALL) is a disease of abnormally growing B lymphoblasts. Here we hypothesized that extracellular vesicles (EVs), which are nanosized particles released by all cells (including cancer cells), could be used to monitor B-ALL severity and progression by sampling plasma instead of bone marrow. EVs are especially attractive as they are present throughout the circulation regardless of the location of the originating cell. First, we used nanoparticle tracking analysis to compare EVs between non-cancer donor (NCD) and B-ALL blood plasma; we found that B-ALL plasma contains more EVs than NCD plasma. We then isolated EVs from NCD and pediatric B-ALL peripheral blood plasma using a synthetic peptide-based isolation technique (Vn96), which is clinically amenable and isolates a broad spectrum of EVs. RNA-seq analysis of small RNAs contained within the isolated EVs revealed a signature of differentially packaged and exclusively packaged RNAs that distinguish NCD from B-ALL. The plasma EVs contain a heterogenous mixture of miRNAs and fragments of long non-coding RNA (lncRNA) and messenger RNA (mRNA). Transcripts packaged in B-ALL EVs include those involved in negative cell cycle regulation, potentially suggesting that B-ALL cells may use EVs to discard gene sequences that control growth. In contrast, NCD EVs carry sequences representative of multiple organs, including brain, muscle, and epithelial cells. This signature could potentially be used to monitor B-ALL disease burden in pediatric B-ALL patients via blood draws instead of invasive bone marrow aspirates.
... The majority of studies concerning CLL focus on dysfunctions of the adaptive immune response, particularly T-cell exhaustion and regulatory T-cellderived immunosuppression [9]. Additionally, defects in the complement system and hypogammaglobulinemia were identified as major mechanisms of immune system dysregulation that reduce the outcome of applied therapies [10,11]. Consequently, CLL patients do not respond effectively to vaccines and suffer from infections that significantly increase CLL mortality rates [12]. ...
Article
Full-text available
Background Impaired neutrophil activity is an important issue in chronic lymphocytic leukemia (CLL), as it contributes to a dysfunctional immune response leading to life-threatening infections in patients. Some features typical of CLL neutrophils, e.g., the B-cell-supportive secretion profile, have already been described. However, most of these studies were performed on cells isolated from peripheral blood. It is still unclear which molecular factors and cell types are involved in shaping neutrophil function and phenotype in the CLL microenvironment. Since regulatory T cells (Treg) play an important role in CLL progression and influence the activity of neutrophils, we investigated the crosstalk between Treg and neutrophils in the spleen using a murine model of CLL. Methods In this work, we used an Eµ-TCL1 mouse model of human CLL. For our in vivo and ex vivo experiments, we inoculated wild-type mice with TCL1 leukemic cells isolated from Eµ-TCL1 transgenic mice and then monitored disease progression by detecting leukemic cells in peripheral blood. We analyzed both the phenotype and activity of neutrophils isolated from the spleens of TCL1 leukemia-bearing mice. To investigate the interrelation between Treg and neutrophils in the leukemia microenvironment, we performed experiments using TCL1-injected DEREG mice with Treg depletion or RAG2KO mice with adoptively transferred TCL1 cells alone or together with Treg. Results The obtained results underline the plasticity of the neutrophil phenotype, observed under the influence of leukemic cells alone and depending on the presence of Treg. In particular, Treg affect the expression of CD62L and IL-4 receptor in neutrophils, both of which are crucial for the function of these cells. Additionally, we show that Treg depletion and IL-10 neutralization induce changes in the leukemia microenvironment, partially restoring the “healthy” phenotype of neutrophils. Conclusions Altogether, the results indicate that the crosstalk between Treg and neutrophils in CLL may play an important role in CLL progression by interfering with the immune response.
Article
Introduction: Acute lymphoblastic leukemia (ALL) is the second most common acute leukemia in adults and the 5-year survival remains low. Methods: We analyzed the gene expression profiles of the complement and coagulation cascades pathway (CCCP) in 998 bone marrow (BM) and 122 peripheral blood (PB) samples of ALL patients and healthy individuals obtained from the TCGA database and evaluated their clinical significance in terms of being diagnostic and prognostic biomarkers. Results: We identified 18 CCCP genes (SERPINA1, C5AR1, F5, CD55, PLAUR, C3AR1, THBD, CD59, PLAU, VWF, CFD, F13A1, C1QA, C1QB, C1QC, A2M, SERPINE1 and CR2) differentially expressed in the BM samples of ALL patients compared to healthy individuals. The expression levels of CD55, F13A1 and CR2 in BM were linked with the overall survival of ALL patients. While in PB only 11 CCCP genes (e.g., SERPINA1, C5AR1, F5, PLAUR, C3AR1, THBD, CFD, F13A1, C1QA, SERPINE1, and CR2) were differentially expressed and F13A1 was significantly associated with ALL patient survival. Machine learning enabled us to predict ALL using the CCCP genes and the accuracy can reach 0.9701 and 0.9167 using the BM and PB, respectively. Furthermore, using single-cell RNA sequencing, we found that the differential expression of CCCP genes was found with diversity in the BM-derived immune cells of ALL patients. Conclusion: Our findings suggest that the CCCP genes may play a key role in the progression of ALL and can be used as potential therapeutic targets and diagnostic markers.
Article
Purpose: To describe a case of endophthalmitis caused by Haemophilus influenzae, an encapsulated bacterium, after intravitreal (IVT) injection of pegcetacoplan. Methods: A single retrospective case was evaluated. Results: An 88-year-old woman with a history of diabetes mellitus, on dialysis for chronic kidney disease, hypertension, hypothyroidism, diffuse large B-cell lymphoma, and geographic atrophy in both eyes received an IVT injection of pegcetacoplan in both eyes. She subsequently developed endophthalmitis in the right eye caused by H influenzae. Further bloodwork showed Proteus mirabilis bacteremia. Conclusions: Pegcetacoplan, a complement inhibitor, may be associated with intraocular infections with encapsulated organisms, especially in immunocompromised patients.
Article
Infection is a major cause of treatment-related morbidity and mortality in pediatric acute lymphoblastic leukemia (ALL). Most children with ALL who develop life-threatening bacterial infections do so during induction therapy. We describe a rare case of ALL presenting simultaneously with Streptococcus agalactiae group B Streptococcus bacteremia and meningitis in a 3-year-old girl. She received appropriate antimicrobial therapy and a 2-drug early induction regimen consisting of vincristine and dexamethasone, leading to slow neurologic recovery and a favorable initial response to anti-neoplastic therapy as evidenced by minimal residual disease of 1.12% on day 15 of induction.
Article
The complement cascade is an ancient and highly conserved arm of the immune system. The accumulating evidence highlights elevated activity of the complement cascade in cancer microenvironment and emphasizes its effects on the immune, cancer and cancer stroma cells, pointing to a role in inflammation-mediated etiology of neoplasms. The role the cascade plays in development, progression and relapse of solid tumors is increasingly recognized, however its role in hematological malignancies, especially those of myeloid origin, has not been thoroughly assessed and remains obscure. As the role of inflammation and autoimmunity in development of myeloid malignancies is becoming recognized, in this review we focus on summarizing the links that have been identified so far for complement cascade involvement in the pathobiology of myeloid malignancies. Complement deficiencies are primary immunodeficiencies that cause an array of clinical outcomes including an increased risk of a range of infectious as well as local or systemic inflammatory and thrombotic conditions. Here, we discuss the impact that deficiencies in complement cascade initiators, mid- and terminal- components and inhibitors have on the biology of myeloid neoplasms. The emergent conclusions indicate that the links between complement cascade, inflammatory signaling and the homeostasis of hematopoietic system exist, and efforts should continue to detail the mechanistic involvement of complement cascade in the development and progression of myeloid cancers.
Article
Full-text available
The management of hematologic malignancies has traditionally relied on chemotherapy regimens, many of which are still in use today. However, with advancements in the knowledge of tumour pathophysiology, therapies are continually evolving. Monoclonal antibodies against specific targets on tumour cells are now widely used to treat hematologic malignancies, either in combination with chemotherapy or as single agents. Rituximab, a monoclonal antibody against the CD20 antigen, is a good example of successful monoclonal antibody therapy that has improved outcomes for patients with B cell non-Hodgkin lymphomas. Monoclonal antibodies are now being used against the immune checkpoints that function to inhibit T cell activation and subsequent tumour eradication by those cytotoxic T cells. Such therapies enhance T cell-mediated tumour eradication and are widely successful in treating patients with solid tumours such as malignant melanoma. Now, they are slowly finding their place in the management of hematologic neoplasms. Even though, currently, immune checkpoint inhibitors are used for relapsed or refractory hematologic neoplasms, trials are ongoing to evaluate their role in frontline treatment. Our review focuses on the current use of immunotherapies in various hematologic malignancies.
Article
Full-text available
Objective Hepatic stellate cells (HSCs) are the important players in liver cirrhosis and liver cancer. They also act as critical mediators of immunosuppression in hepatocellular carcinoma (HCC). In this study, we hypothesized that HSCs promote HCC progression via C3. Methods C3 in HSCs was knocked down using a shRNA retroviral plasmid. The conditioned medium from HSCs or shC3 HSCs (knockdown of C3 by shRNA in HSCs) was collected to detect their effects on bone marrow (BM) and T cells (including expansion and apoptosis) in vitro, and in an HCC in situ model in mice. Results We found that HSCs promoted T-cell apoptosis and decreased their proliferation, inhibited dendritic cell (DC) maturation, and induced myeloid-derived suppressor cell (MDSC) expansion through the C3 pathway in vitro. In addition, the knockdown of C3 suppressed HSC-promoted HCC development in the orthotopic transplantation tumor model of HCC in mice. Conclusion These findings provide more insights into the immunomodulatory roles of HSCs in HCC progression and indicate that modulation of the C3 pathway might be a novel therapeutic approach for liver cancer.
Article
Full-text available
A prospective study of 312 patients [194 with multiple myeloma (MM) and 118 with lymphomas (LYMPH)] receiving high-dose chemotherapy and autologous hematopoietic stem cell transplantation (auto-HSCT) was conducted. Ficolins are innate immune defense factors, able to distinguish between “self” “abnormal self,” and “non-self” and contribute to the elimination of the last two by direct opsonization and/or initiation of complement activation via the lectin pathway. Concentrations of ficolin-1, ficolin-2, and ficolin-3 in serially taken serum samples were determined as were the polymorphisms of the corresponding (FCN1, FCN2, and FCN3) genes. Serum samples were collected before conditioning chemotherapy, before HSCT, and once weekly post-HSCT (four to five samples in total); some patients were also sampled at 1 and/or 3 months post-transplantation. The control group (C) consisted of 267 healthy unrelated individuals. Median ficolin-1 and ficolin-2 (but not ficolin-3) levels in MM patients' sera taken before chemotherapy were lower (and correspondingly frequencies of the lowest concentrations were higher) compared with controls. That appeared to be associated with the malignant disease itself rather than with post-HSCT complications (febrile neutropenia, infections accompanied, or not with bacteremia). Higher frequencies of the FCN1 genotype G/A-C/C-G/G (corresponding to polymorphisms at positions −542, −144, and +6658, respectively) and FCN2 gene heterozygosity for the −857 C>A polymorphism were found among patients diagnosed with MM compared with the C group. Furthermore, FCN2 G/G homozygosity (−557 A>G) was found more frequently and heterozygosity G/T at +6424 less frequently among LYMPH patients than among the healthy subjects. Heterozygosity for +1637delC mutation of the FCN3 gene was more common among patients diagnosed with lymphomas who experienced hospital infections. Although no evidence for an association of low ficolin-1 or ficolin-2 with infections during neutropenia following chemotherapy before HSCT was found, we observed a possible protective effect of ficolins during follow-up.
Article
Full-text available
Background: Mounting evidence suggests that complement components promote tumor progression via modulating immune suppression, angiogenesis, or tumor cell proliferation. However, the role of C3a-C3aR signaling in regulating lung metastasis of breast cancer remains unknown. Methods: We performed various ex-vivo and in-vivo assays. Genetic and pharmacological C3aR blockade models were applied to investigate the role of C3a-C3aR in metastasis of breast cancer. Results: C3a-C3aR signaling in CAFs facilitates the metastasis of breast cancer. Mechanically, C3a-C3aR signaling augments pro-metastatic cytokine secretion and extracellular matrix components expression of CAFs via the activation of PI3K-AKT signaling. Genetic or pharmacological blockade of C3aR signaling effectively inhibited lung metastasis of breast cancer in mouse models. Conclusions: C3a-C3aR signaling in CAFs facilitates the metastasis of breast cancer. Targeting C3aR signaling is a potential anti-metastasis strategy for breast cancer therapy.
Article
Full-text available
Background: Localized C3 deposition is a well-known factor of inflammation. However, its role in oncoprogression of gastric cancer (GC) remains obscured. This study aims to explore the prognostic value of C3 deposition and to elucidate the mechanism of C3-related oncoprogression for GC. Methods: From August to December 2013, 106 GC patients were prospectively included. The regional expression of C3 and other effectors in gastric tissues were detected by WB, IHC, qRT-PCR and other tests. The correlation of localized C3 deposition and oncologic outcomes was determined by 5-year survival significance. Human GC and normal epithelial cell lines were employed to detect a relationship between C3 and STAT3 signaling pathway in vitro experiments. Results: C3 and C3a expression were markedly enhanced in GC tissues at both mRNA and protein levels compared with those in paired nontumorous tissues. According to IHC C3 score, 65 (61.3%) and 41 (38.7%) patients had high and low C3 deposition, respectively. C3 deposition was negatively correlated with plasma levels of C3 and C3a (both P < 0.001) and positively correlated with pathological T and TNM stages (both P < 0.001). High C3 deposition was identified as an independent prognostic factor of poor 5-year overall survival (P = 0.045). In vitro C3 administration remarkably enhanced p-JAK2/p-STAT3 expression in GC cell lines but caused a reduction of such activation when pre-incubated with a C3 blocker. Importantly, C3 failed to activate such signaling in cells pre-treated with a JAK2 inhibitor. Conclusions: Localized C3 deposition in the tumor microenvironment is a relevant immune signature for predicting prognosis of GC. It may aberrantly activate JAK2/STAT3 pathway allowing oncoprogression. Trial registration: ClinicalTrials.gov, NCT02425930, Registered 1st August 2013.
Article
Full-text available
Therapeutic antibodies targeting vascular endothelial growth factor (VEGF) have become a critical regimen for tumor therapy, but the efficacy of monotherapy is usually limited by drug resistance and multiple angiogenic mechanisms. Complement proteins are becoming potential candidates for cancer-targeted therapy based on their role in promoting cancer progression and angiogenesis. However, the antitumor abilities of simultaneous VEGF and complement blockade were unknown. We generated a humanized soluble VEGFR-Fc fusion protein (VID) binding VEGFA/PIGF and a CR1-Fc fusion protein (CID) targeting C3b/C4b. Both VID and CID had good affinities to their ligands and showed effective bioactivities. In vitro, angiogenesis effects induced by VEGF and hemolysis induced by complement were inhibited by VID and CID, respectively. Further, VID and CID confer a synergetic therapeutic effect in a colitis-associated colorectal cancer (CAC) model and an orthotopic 4T1 breast cancer model. Mechanically, combination therapy inhibited tumor angiogenesis, cell proliferation, and MDSC infiltration in the tumor microenvironment and promoted tumor cell apoptosis. Our study offers a novel therapeutic strategy for anti-VEGF-resistant tumors and chronic-inflammation-associated tumors.
Article
Full-text available
The treatment options in multiple myeloma (MM) has changed dramatically over the past decade with the development of novel agents such as proteasome inhibitors (PIs); bortezomib and immunomodulatory drugs (IMiDs); thalidomide, and lenalidomide which revealed high efficacy and improvement of overall survival (OS) in MM patients. However, despite these progresses, most patients relapse and become eventually refractory to these therapies. Thus, the development of novel, targeted immunotherapies has been pursued aggressively. Recently, next-generation PIs; carfilzomib and ixazomib, IMiD; pomalidomide, histone deacetylase inhibitor (HDADi); panobinostat and monoclonal antibodies (MoAbs); and elotuzumab and daratumumab have emerged, and especially, combination of mAbs plus novel agents has led to dramatic improvements in the outcome of MM patients. The field of immune therapies has been accelerating in the treatment of hematological malignancies and has also taken center stage in MM. This review focuses on an overview of current status of novel MoAb therapy including bispecific T-cell engager (BiTE) antibody (BsAb), antibody-drug conjugate (ADC), and chimeric antigen receptor (CAR) T cells, in relapsed or refractory MM (RRMM). Lastly, investigational novel MoAb-based therapy to overcome immunotherapy resistance in MM is shown.
Article
Strength in numbers Human cluster of differentiation 20 (CD20) is expressed on malignant B cells and is the target of therapeutic antibodies used in cancer immunotherapy. Kumar et al. now present structures that explain why so-called type I antibodies efficiently activate the complement pathway to kill cells, whereas type II antibodies do not. Type I antibodies each bind to two CD20 dimers and form clusters that facilitate binding to a component of the complement pathway. The second-generation type I antibody ofatumumab has molecular features that make it more efficient at clustering than first-generation rituximab. By contrast, the type II antibody obinutuzumab interacts with just one CD20 dimer and cannot form higher-order assemblies. Understanding these mechanisms will inform the design of next-generation immunotherapies. Science , this issue p. 793
Article
Clustering for the kill Cluster of differentiation 20 (CD20) is a membrane protein that defines most B cell populations and is the target of therapeutic antibodies to treat malignancies and autoimmune disorders. Rougé et al. present the structure of CD20 bound to the antibody rituximab that activates the complement system to kill B cells. CD20 forms a dimer and each monomer binds one rituximab antigen-binding fragment (Fab) to give 2:2 stoichiometry. The compact packing between Fab arms and CD20 gives rise to circular assemblies with a diameter similar to that of antibody hexamers known to recruit the first component of the complement cascade. Science , this issue p. 1224
Article
The tumour microenvironment (TME) highly influences the growth and spread of tumours, thus impacting the patient’s clinical outcome. In this context, the complement system plays a major and complex role. It may either act to kill antibody-coated tumour cells, support local chronic inflammation or hamper antitumour T cell responses favouring tumour progression. Recent studies demonstrate that these opposing effects are dependent upon the sites of complement activation, the composition of the TME and the tumour cell sensitivity to complement attack. In this Review, we present the evidence that has so far accrued showing a role for complement activation and its effects on cancer control and clinical outcome under different TME contexts. We also include a new analysis of the publicly available transcriptomic data to provide an overview of the prognostic value of complement gene expression in 30 cancer types. We argue that the interplay of complement components within each cancer type is unique, governed by the properties of the tumour cells and the TME. This concept is of critical importance for the design of efficient therapeutic strategies aimed at targeting complement components and their signalling.