ArticlePDF Available

Multifunctional CD40L: Pro- and anti-neoplastic activity

Authors:

Abstract and Figures

The CD40 ligand is a type I transmembrane protein that belongs to a tumor necrosis factor (TNF) superfamily. It is present not only on the surface of activated CD4+ T cells, B cells, blood platelets, monocytes, and natural killer (NK) cells but also on cancer cells. The receptor for ligand is constitutively expressed on cells, TNF family protein: CD40. The role of the CD40/CD40L pathway in the induction of body immunity, in inflammation, or in hemostasis has been well documented, whereas its involvement in neoplastic disease is still under investigation. CD40L ligand may potentiate apoptosis of tumor cells by activation of nuclear factor-κB (NF-κB), AP-1, CD95, or caspase-depended pathways and stimulate host immunity to defend against cancer. Although CD40L has a major contribution to anti-cancer activity, many reports point at its ambivalent nature. CD40L enhance release of strongly pro-angiogenic factor, vascular endothelial growth factor (VEGF), and activator of coagulation, TF, the level of which is correlated with tumor metastasis. CD40L involvement in the inhibition of tumor progression has led to the emergence of not only therapy using recombinant forms of the ligand and vaccines in the treatment of cancer but also therapy consisting of inhibiting platelets-main source of CD40L. This article is a review of studies on the ambivalent role of CD40L in neoplastic diseases.
Content may be subject to copyright.
REVIEW
Multifunctional CD40L: pro- and anti-neoplastic activity
Aleksandra Korniluk &Halina Kemona &
Violetta Dymicka-Piekarska
Received: 6 June 2014 /Accepted: 27 July 2014 /Published online: 13 August 2014
#The Author(s) 2014. This article is published with open access at Springerlink.com
Abstract The CD40 ligand is a type I transmembrane protein
that belongs to a tumor necrosis factor (TNF) superfamily. It is
present not only on the surface of activated CD4+ T cells, B
cells, blood platelets, monocytes, and natural killer (NK) cells
but also on cancer cells. The receptor for ligand is constitu-
tively expressed on cells, TNF family protein: CD40. The role
of the CD40/CD40L pathway in the induction of body immu-
nity, in inflammation, or in hemostasis has been well docu-
mented, whereas its involvement in neoplastic disease is still
under investigation. CD40L ligand may potentiate apoptosis
of tumor cells by activation of nuclear factor-κB(NF-κB),
AP-1, CD95, or caspase-depended pathways and stimulate
host immunity to defend against cancer. Although CD40L
has a major contribution to anti-cancer activity, many reports
point at its ambivalent nature. CD40L enhance release of
strongly pro-angiogenic factor, vascular endothelial growth
factor (VEGF), and activator of coagulation, TF, the level of
which is correlated with tumor metastasis. CD40L involve-
ment in the inhibition of tumor progression has led to the
emergence of not only therapy using recombinant forms of the
ligand and vaccines in the treatment of cancer but also therapy
consisting of inhibiting platelets-main source of CD40L. This
article is a review of studies on the ambivalent role of CD40L
in neoplastic diseases.
Keywords CD40L .CD40 .Inflammation .Apoptosis .
Cancer
Abbreviations
TNF Tumor necrosis factor
TRAF TNF receptor-associated factor
TAP Transporter associated with antigen processing
RANTES Regulated on activation, normal T cell expressed
and secreted
VEGF Vascular endothelial growth factor
FGF-2 Fibroblast growth factor 2
TF Tissue factor
APCs Antigen-presenting cells
DC Dendritic cells
NK cell Natural killer cells
GM-CSF Granulocyte-macrophage colony-stimulating
factor
CLL Chronic lymphocytic leukemia
MM Multiple myeloma
Introduction
The CD40 ligand (CD40L), also known as CD154, T-B
activating molecule (TBAM), tumor necrosis factor (TNF)-
related activation protein (TRAP), and gp39, is a type I
transmembrane protein, a member of the TNF superfamily,
together with lymphotoxin-α(LT-α, TNF-β), LT-β, FasL,
CD30L, CD27L, and 4-1BBL 9 [1]. The CD40L gene is
located on chromosome X (q26.3q27.1) [2], contains five
exons and four introns, and the protein that is produced
(29 kDa) via translation is built up of 261 amino acids. The
cell membrane presents the 3233-kDA form, which indicates
the posttranslation modification of the protein [3].
CD40L associated with cell membrane contains a globular
TNF-like extracellular domain, long extracellular domain
(ECD), short transmembrane domain (TMD), and a small
cytoplasmic intracellular domain (ICD). [3] The extracellular
structure of CD40L composed of beta sheet-alpha helix-beta
sheet that is arranged in the so-called Greek key motif is
specific to the TNF superfamily [4]. If CD40L is produced
A. Korniluk (*):H. Kemona :V. Dymicka-Piekarska
Department of Clinical Laboratory Diagnostics, Medical University
of Bialystok, Bialystok, Poland
e-mail: alex.korniluk@wp.pl
Tumor Biol. (2014) 35:94479457
DOI 10.1007/s13277-014-2407-x
as a single polypeptide chain, it can be found on the cell
surface as a homotrimeric complex. The long protein chain
is accompanied by two shorter forms of CD40L, which
through proteolytic hydrolysis are released from the cell mem-
brane to the circulation, where they form soluble trimmers,
sCD40L, having the receptor-binding ability [4,5]. Since both
the surface ligand and its soluble form have structural do-
mains, they show high biological activity. Due to the presence
of the KGD sequence (lysine-glycine-aspartic acid), they can
bind to the GPIIbIIIa receptor, and therefore, the ligand may
play a role in platelet activation and stimulate further release
of sCD40L [6,7]. The trimeric protein structure facilitates
signal transfer to the cell interior via binding to the receptor,
whereas the TNF-like domain allows ligand binding to its
main receptor-CD40. The presence of the ligand has been
observed, first of all, on the surface of activated CD4+ T cells,
but also on activated B cells and blood platelets, monocytes,
natural killer (NK) cells, adipose cells, and basophils during
inflammation [8]. It is assumed that over 95 % of CD40L
originates from blood platelets [9]. Henn et al. [9]haveshown
that CD40L is stored in platelet granules αand released after
their activation and degranulation.
The CD40 receptor is a transmembrane type I protein
belonging to the TNF family, encoded by the gene located
on chromosome 20 (q12q13.2). It is present on the cell
surface as a trimeric complex (4045 kD) [10]. Structurally,
CD40 contains a long ECD, transmembrane region, and short
C-terminal cytoplasmic fragment [11]. Since the cytoplasmic
part does not exhibit kinase activity, signals are transmitted
mainly through the ligand-dependent recruitment of adaptor
proteins of the TNF receptor-associated factor (TRAF) family
[12]. The CD40 receptor has been found on the surface of B
cells and on the membrane of the antigen-presenting cells
(APCs), epithelial cells, endothelial cells, smooth muscle
cells, fibroblasts, basophils, and blood platelets, where it is
constitutively expressed [13,14]. However, expression of the
receptor can be induced by the combination of TNF-αwith
interferon (IFN)-γ. These molecules significantly increased
de novo expression of CD40 on human endothelial and vas-
cular smooth muscle cells, and this process is mediated by the
simultaneous activation of nuclear factor-κB (NF-κB) by
TNF-αand STAT-1αby IFN-γ[15,16]. Activation of
CD40 receptor by CD40 ligand on epithelial cells leads to
the secretion of cytokines and chemokines by these cells,
whereas on fibroblasts and endothelial cells, it contributes to
their proliferation [17]. Interaction betweenCD40 and CD40L
can be inhibited by specific antibodies like 4D11, a novel fully
human anti-CD40 mAb which is produced by genetically
modified mice. Also, lucatumumab and Chi 220 are anti-
CD40 antibodies which inhibit ligation between receptor and
ligand [18].
Neither CD40L nor its receptor CD40 can function inde-
pendently, and only their interaction leads to the production of
intracellular signal that is responsible for enhanced humoral
and cellular response and for the cellular production of cyto-
kines, chemokines, and adhesion proteins [19].
CD40L in the immune response and inflammation
The first studies on the roleof CD40Lin the immune response
were performed when the ligand was detected on the surface
of T and B cells, confirming its involvement in cellular and
humoral immune response. The activation of B cells by
CD40L turned out to be essential in the process of lymphocyte
proliferation, differentiation, and maturation and in isotope
switching [10]. The interaction of CD40L with CD40 exerts
an effect on the production of cytokines (IL-6, IL-10, TNF-α,
LT-α), expression of adhesion molecules and co-stimulatory
receptors (intercellular adhesion molecule (ICAM), CD23,
B7.1/CD80, B7.2/CD86), as well as class I MHC, class II
MHC, and transporter associated with antigen processing
(TAP) present on B cells [20].
The significant role of the CD40L-CD40 system in the
immune response was confirmed with the discovery of
CD40L gene mutation, leading to the hyper-IgM syndrome
(HIGM). In these patients, overproduction of IgM antibodies
and lack of IgG, IgA, and IgE antibodies can be observed [3].
The binding of CD40L to CD40 present on endothelial
cells and fibroblasts increases the secretion of metalloprotein-
ases and activates the production of chemokines (IL-8, MCP-
1, macrophage inflammatory protein (MIP)-1α,RANTES)
and cytokines (IL-1, IL-6, IL-12, and TNF-α), which attract
lymphocytes to the site of inflammation and increase expres-
sion of adhesion molecules (ICAM-1, VCAM-1, and E-
selectin), responsible for the recruitment of monocytes and
lymphocytes, thus leading to their accumulation in the internal
vascular membrane [21].
Interaction between ligand and receptor causes overexpres-
sion of cycloxygenase 2 (COX-2) and prostaglandin E2
(PGE2) [22]. The soluble form of CD40L is able to activate
blood platelets and stimulate the release of β-thromboglobulin
(β-TG) and 5-hydroxytryptoamine from their granules,
whereas cell membrane CD40L causes the release of regulated
on activation, normal T cell expressed and secreted
(RANTES), the protein of strong pro-inflammatory proper-
ties, from the platelets [23,24].
The evidence for the involvement of the CD40/CD40L
pathway in inflammation includes highly elevated expression
of these molecules in inflammatory conditions of the colon,
such as Lesniewski-Crohn disease or ulcerative colitis [19].
The pro-inflammatory and pro-thrombotic activities of the
CD40/CD40L system have been also shown in diabetes
[25], atherosclerosis [26], and cardiovascular diseases [27].
High level of sCD40L can be a prognostic risk factor of death,
heart infarct, and recurrent angina in acute coronary
9448 Tumor Biol. (2014) 35:94479457
syndromes and future cardiovascular episodes in healthy
women [28]. It is also a reliable marker for the population of
patients with acute coronary syndrome at a high risk of heart
incidents [29].
CD40L in neoplastic disease
The role of the CD40/CD40L pathway in the induction of
body immunity, in inflammation, or in hemostasis has been
well documented, whereas its involvement in neoplastic dis-
ease is still controversial.
Studies on CD40L have revealed that it enhances anti-
neoplastic immune response of the body, inhibits tumor
growth, and induces apoptosis of cancer cells [30,31]. Subse-
quent reports have suggested that in many cancers, CD40
activation by its ligand results in a completely reverse situa-
tion, i.e., enhancement of tumor growth and progression
(Fig. 1)[32,33]. The effect induced by CD40L has appeared
to depend not only on the type of cells that show the receptor
expression but also on the strength of the signal transmitted by
the ligand. High signal (the cell has many CD40 molecules)
indicates apoptosis of cancer cells, whereas low signal (a small
number of receptors) CD40L promotes cancer growth [34].
FasL
INFγ
perforin
granzyme
c
a
n
c
e
r
p
r
o
c
o
a
g
u
l
a
n
t
,
T
F
INFγ, TNFα
+IL-2
TRAF
IKKα,β,γ
NEMO
IκB
NFκB
CD40-receptor
CD40-ligand
TF up-reg
ula
tion
pro
coagulant activit
y
CD80/86 CD28
INF-
γ
I
L-
4
I
gG
p
ro
du
c
t
ion
I
N
F
-
γ
,
I
L
-
1
2
A
ngioge
n
e
si
s
sCD4OL
VEGF
RAS
PI3K
Activation
IL-4
IL-10
INFγ
TRAF
JNK
p38MA PK
PKC/NFκB
TRAF
IκB/NFκB JNK /p38
NFκB AP-1
TRAF
JNK/AP-1
caspa se 9/
caspase 3
DC
TC
iNKTc
NKc
EC
CD 4+
CD 8+
B cells
Fig. 1 This figure illustrates some of the mechanisms by which CD40L
can influence on different cell functions and the processes by which these
altered cells can impact on host immunity in neoplastic diseases. CD40L
is present as a membrane-bound form (mCD40L), present first of all on T
lymphocytes (CD4+,CD8+,iNKTc,andNKc), and as a soluble form
(sCD40L) derived primarily from active platelets. Interaction of the
ligand with the receptor (CD40) may not only potentiate the anti-tumor
immunity but also promote the development of cancer. The binding of
CD40L with a receptor on dendritic cells (DC) leads to the expression of
co-stimulatory molecules necessary for the correct antigen presentation
and protects DC against apoptosis induced by factors derived from tumor
cells (TC). Moreover, CD40L/CD40 interaction enhances the prolifera-
tion and maturation of lymphocytes B (Bcells) and the production of
antibodies against the tumor. mCD40L present on various cells induces
apoptosis of tumor cells via the TRAF-JNK/AP-1-caspase-9/caspase-3
pathway. On the other hand, the activation of endothelial cells (EC)may
result in enhance TF pro-coagulant activity and high expression of VEGF,
main mediator of tumor angiogenesis
Tumor Biol. (2014) 35:94479457 9449
CD40L and cancer progression
CD40L has a major contribution to immunological activity;
however, many reports point at its ambivalent nature in neo-
plastic diseases. On one hand, the ligand activatesthe immune
system to combat the cancer, but on the other, it stimulates
tumor progression, growth, and metastasis formation.
Induction of angiogenesis by CD40L induces VEGF
production
Tumor growth and metastasis depend on angiogenesis and
lymphangiogenesis caused by different factors released from
host and tumor cells [35]. Binding of CD40L to CD40 present
on endothelial cells leads to the expression of strongly pro-
angiogenic factors, such as vascular endothelial growth factor
(VEGF) or fibroblast growth factor 2 (FGF-2) that can pro-
mote in vivo angiogenesis. [3638]. VEGF participates in
mobilization of endothelial stem cells, which take part in
formation of new blood vessels in tumor microenvironment
[35]. In addition, VEGF increases expression of tissue factor
(TF), which also promotes blood coagulation. Moreover,
VEGF induces monocyte chemotaxis and activation and also
impairs the immune system functions through the inhibition of
dendritic cell maturation [39]. In gastric cancer, ligation of
CD40 by CD40L causes up-expression of VEGF by PI3K
pathway [40]. CD40L/CD40 interaction also has been shown
to induce COX-2 expression in cells and subsequent VEGF
production, what was confirmed by Miura et al. [41]. Tai YT
et al. [42] suggested that in human multiple myeloma (MM)
cells, CD40 activation by CD40L can induce secretion of
VEGF by p53 pathway. In this case, VEGF stimulates IL-6
secretion in bone marrow stromal cells and thereby augments
paracrine IL-6-mediated MM cell growth. What is more,
Farahani et al. [43] shows that in cells from patients with
chronic lymphocytic leukemia (CLL), CD40L also can up-
regulate production of VEGF, which leads to CLL cell sur-
vival. This process depends, in part, on NF-κB activation.
CD40L-induced survival of malignant cells depends on com-
bined signaling by CD40 and VEGF receptor (VEGFR).
Inhibition of CD40L-induced production of VEGF and cyto-
kines (IL-6) and activation of signaling pathways, prolifera-
tion, and survival of CLL [44] and MM [42] cells is caused by
lucatumumab (HCD122). HCD122 binds to CD40 and block-
ade CD40/CD40L interactions that induce apoptosis and me-
diate antibody-depended cellular toxicity on lucatumumab-
bound CD40-expressed malignant cells. This antibody is cur-
rently in phase I/II clinical trials in CLL. VEGF can also
enhance cleavage of membrane-bound CD40L and cause
increased level of sCD40L. Inhibition of this process is pos-
sible through the use of bevacizumab-anti-VEGF antibody
[18].
Hematological malignancy
The activation of CD40 contributes to the increased survival
and resistance to chemotherapy of follicular lymphoma, hairy
cell leukemia, and CLL cells [4547]. It is suggested that the
co-stimulation of IL-4 and CD40L causes long-term prolifer-
ation of B cells and short-term proliferation and increased
percentage of coat cells in hairy cell leukemia [48]. Kato
et al. [49] showed that such interaction in diffuse non-
Hodgkins lymphoma depending on the type of cancer cell
line enhanced short- or long-term proliferation of cancer cells.
Research conducted on the established cell lines (GDLBGCB-
1 and GDLBGCB-2) and cells taken from the patients has
proved that lack of IL-4 or introduction of antibodies against
CD40L causes inhibition of cell proliferation. In some types
of CLL, cell proliferation does not depend on endogenous
CD40L, but on the stimulation with the ligand from tumor
microenvironment. Pham et al. [50] suggest that endogenous
CD40L which is present on aggressive B cells of lymphoma
binds to CD40 and the signal transferred into the cell activates
the NF-κB pathway.
The effect of CD40L on the action of many drugs has been
reported. The CD40L induces resistance to a number of anti-
cancer drugs, including doxorubicin or vinblastin. In non-
Hodgkin
s lymphoma, lack of reaction to drugs is caused by
caspase-independent and independent pathways [51], whereas
in breast cancer, only by the caspase-dependent route [52]. A
study conducted on transgenic mice showed that the admin-
istration of clopidogrel, a drug inhibiting platelet aggregation
and CD40L release, decreased tumor size [53]. This may
suggest that tumor growth inhibition by the drug is associated
with an indirect decrease in CD40L expression [51]. The
sCD40L/CD40 pathway activation in gastric cancer inhibited
Fas- and drug-dependent apoptosis and also increased the
motility of CD40-positive cancer cells, thus facilitating the
formation of metastases, what was confirmed in humans [54].
Rui Li et al. [55], who investigated the expression of CD40
and CD40L on gastric cancer cells from patients, suggest that
a simultaneous expression of the two molecules and perma-
nent activation of the CD40/CD40L pathway have a major
significance in neoplastic progression. CD40 activation in-
hibits apoptosis and promotes spread of transformed cells.
Likewise, in cancer of the urinary bladder, the activity of the
CD40/CD40L system protects cells against apoptosis and
increases their survival, which is associated with the inhibition
of CD95-dependent apoptosis [56]. The assessment of CD40
expression on the surface of pancreatic cancer cells revealed
that at stage I of the disease, the cells were CD40-negative,
whereas in patients with lymph node involvement and distant
metastases, the cells exhibited high expression of CD40. Also,
the serum level of sCD40L in these patients was significantly
elevated. The two molecules correlated with cancer stage,
which may indicate their involvement in the disease
9450 Tumor Biol. (2014) 35:94479457
development. The use of shrCD40L on pancreatic cancer cell
lines significantly inhibited their proliferation and enhanced
apoptosis, although the effect was most pronounced in the
cells showing high CD40 expression [57].
Solid tumors
Rosseli et al. [58] showed a high level of sCD40L in patients
with adenocarcinoma and squamous cell carcinoma of the
lungs, with the highest level of the ligand observed in patients
with distant metastases of the latter. Moreover, sCD40L
showed a positive correlation with the pro-thrombotic system
components, i.e., fragments of pro-thrombin 1 and 2 (F1+ 2),
and the thrombin/anti-thrombin complex (TATc). The re-
searchers forwarded the hypothesis that the pro-thrombotic
agents released from lung cancer cells stimulate platelet acti-
vation and release of sCD40L, which in turn interacts with
other types of CD40-positive cells, including tumor-
associated macrophage (TAM) cells and cancer cells of the
lungs. Also, Amirkhosravi et al. [59] suggested that the inter-
action of CD40L present on blood platelets with CD40 on
tumor cells may potentially promote activation of coagulation
on human melanoma cells A375 through the effect of in-
creased expression of TF.
Anti-neoplastic stimulation of immune response
Research on CD40L indicates that the inhibition of cancer
progression can be associated with the activation of two
mechanisms by the ligand, namely, stimulation of anti-
neoplastic immune response and induction of apoptosis of
transformed cells.
The enhancement of immune response is inextricably as-
sociated with the activation of antigen-presenting cells
(APCs), especially dendritic cells, monocytes, and B cells,
showing CD40 expression. Cancer cells release a number of
cytokines and soluble agents that have an immunosuppressive
effect and cause APC dysfunction [60]. Most data seem to
confirm a beneficial effect of CD40L/CD40 interaction on the
function of dendritic cells (DC). It has been proved that
CD40L/CD40 binding protects DC against apoptosis, by in-
ducing the expression of the anti-apoptotic molecules Bcl-2 or
serpin serine protease inhibitor 6 (SPI-6) [61,62]. The appli-
cation of recombinant CD154 therapy leads to the formation
of mature dendritic cells, increased expression of adhesion
and co-stimulatory molecules (ICAM-1, CD83, CD80/86),
and secretion of pro-inflammatory cytokines (TNF-α,IL-6,
IL-12) and chemokines (IL-8, MIP-1α), which confirms an
exceptionally important role of the ligand in the activation of
these cells [63]. CD40L, together with pro-inflammatory cy-
tokines and INF-γ, is indispensable for the presentation of
foreign antigens absorbed by dendritic cells (cross-priming) to
T cells. It has been suggested that IL-12 and INF-γregulate
the expression of CD154 on effector cells, whereas the ele-
vated level of the ligand may modulate NK cell cytotoxicity
[64]. The ligand binding to the receptor present on monocytes
and macrophages is associated with the activation of numer-
ous immune processes involving these cells [65].
The activation of CD40 by CD40L enhances the expres-
sion of TLR-9 on macrophages, whereas its stimulation by
sCD40L increases the activity of monocytes, especially in
cancer of the uterine cervix, which is related to the activation
of the NF-κB and MARK pathways [66]. Studies on the
stimulation of CD40 present on cancer cell lines in the blad-
der, pancreas, or breast through the recombinant CD40L have
shown increased expression of ICAM and Fas by cancer cells
and the production of IL-6, IL-8, GROα,GM-CSF,and
TNF-α. The incubation of transformed cells with the expres-
sion of CD40 and CD40L has been found to considerably
inhibit their proliferation, disturbances in the cell cycle, and
decreased cell viability [66]. Moreover, the administration of
anti-CD40 antibodies has been observed to activate macro-
phages, thus enabling them to inhibit proliferation of melano-
ma B-16 cells and to stimulate them to interferon production
[67]. CD40 binding to CD40L not only leads to the increased
production of cytokines by immune cells, but also stimulates
many types of cancers to release pro-inflammatory proteins.
Cells of Hodgkins lymphoma due to CD40 activation release
IL-8, IL-6, or TNF, which in consequence can increase Tcell-
dependent anti-neoplastic immune response [68]. Also, in
acute lymphoblastic leukemia, the activation of CD40 present
on cancer cells by CD40L enhances the secretion of MDC and
TARC; i.e., the proteins that play a role of chemoattractans for
CCR4+ T cells [69]. These lymphocytes possess CCR4 re-
ceptor on such chemokines as MPC-1, MIP-1, or RANTES.
The presence of this receptor also allows migration of lym-
phocytes to the skin. High percentage of CCR4+ T cells have
been observed in malignant skin lymphomas, such as mycosis
fungoides or Sezaryssyndrome[70].
CD40L activation-dependent apoptosis of cancer cells
Apart from its effect on the body immunity, the CD40L/CD40
system can inhibit cancer growth via enhanced apoptosis of
cancer cells. Interestingly, binding of CD40L by CD40 in-
duces the apoptosis of cancer cells, but not of the healthy ones.
CD40L prevents death of fibroblasts, dendritic cells, mono-
cytes or B cells, and enhances the death of transformed mes-
enchymal cells, epithelial cells, neurons, or hepatocytes. This
phenomenon is referred to as activation-induced cell death
(AICD) [31,71,72]. The ligand induces the apoptosis of
CD40-positive cancer cells. The presence of CD40 molecule
was first identified on cancer cells of urinary bladder.
Tumor Biol. (2014) 35:94479457 9451
Subsequent research showed CD40 expression on cancer cells
of the breast [73], ovary, intestines [74], liver [75], glioma
[17], nasopharynx [76], melanoma [77], or lymphoma.
As shown by literature data, cells with CD40 on their
surface very seldom express CD40L. Moreover, various can-
cer cell lines exhibit a varied level of CD40, which is associ-
ated with cancer stage and usually decreases with disease
progression [30]. The receptor present on the transformed
cells can bind to the cell membrane-bound ligand (mCD40L),
soluble CD40L (sCD40L), recombinant CS40L (srhCD40L),
and anti-CD40 antibody. The effect induced by the ligand-
receptor interaction depends on the type of cells on which the
molecules are located and on the form of the ligand that binds
to the receptor.
The CD40L/CD40 pathway may activate CD95-dependent
apoptosis, which has been proved in the research on neuro-
blastoma, and via the activation of caspase-8 that is induced
by recombinant CD40L administration [78]. Investigations of
normal and transformed epithelial cells of the urinary tract
indicate that the activation of cancer cells by mCD40L, but not
by sCD40L, leads to the growth and stabilization of TRAF-3
and causes activation of caspase-9 and caspase-3. In healthy
cells, CD40L does not induce apoptosis but leads to a decrease
in the activity of TRAF-3 and TRAF-2 [79]. The TRAF-2
factor exerts a positive effect on differentiation of B cells,
whereas TRAF-3 inhibits the growth and blockade of NF-κB
factor in cancer cells [79]. Other authors have investigated the
significance of CD40 expression on cancer cells of the urinary
tract. The incubation with CD40L leads to the apoptosis of
transformed cells, whereas decreased expression of CD40 on
these cells is associated with enhanced cancer progression
[80].
It has been shown that CD40L may enhance apoptosis of
solid tumor cells and transformed cells of blood cancers [17,
79].
Hematological malignancy
The CD40/CD40L interaction on human and murine B cell
lymphoma resulted in the arrest of cell cycle, which had a
major significance for the induction and maintenance of tumor
quiescent state. Also, in the case of Burkitt lymphoma, CD40
activation caused tumor growth inhibition, increased activity
of Fas, and enhanced apoptosis of tumor cells [46]. Tong et al.
[31] have demonstrated the expression of CD40 by MM cells
and blockage of their progression by the use of srCD40L
therapy. In turn, Young et al. [81] have suggested that the loss
of CD154 expression on the myeloma cells leads to the
ligand-dependent loss of epithelial growth regulation and
uncontrolled inflammations and infections, which may con-
tribute to the increased and uncontrolled susceptibility of
patients to the development of cancer. It has been also ob-
served that despite preserved expression of CD40 by oral
basal epithelial cells, the loss of CD154 expression may
contribute to epithelial metastasis [76]. Also, in patients with
CD40L gene mutation (HIMN), an increase was noted in the
incidence of cancer of the liver, pancreas, or bile ducts, which
seems to confirm the Young et al. theory [82].
Solid tumors
It is assumed that the level and type of the cytokines secreted
by cancer cells depend on whether the ligand is membrane-
bound or soluble. Numerous studies have shown that binding
of both mCD40L and sCD40L to the receptor CD40 leads to
the activation of NF-κB, although in the case of the transcrip-
tion factor AP-1, the pro-apoptotic cascade is induced only by
mCD40L [74]. The activation of NF-κBfactorstimulatesthe
release of IL-8, whereas AP-1 enhances the production of
GM-CSF [75].
The results obtained by Georgopoulos et al. [74] indicate
that the interaction between mCD40L and CD40 on colon
cancer cells leads to the formation of a strong pro-apoptotic
signal. Interestingly, the authors have revealed that this effect
is induced only by mCD40L binding. Apoptosis of colon
cancer cells was not observed when they were CD40-
negative and subject to the action of sCD40L [79,80].
mCD40L interaction with CD40-positive cancer cells was
accompanied by increased production of IL-8 by transformed
cells. It has also been shown that in the course of colon cancer,
mCD40L affects the expression of GM-CSF. Growth inhibi-
tion or death in carcinoma cells mediated by CD40 can be
accompanied also by induction of IL-6 production [74]. These
cytokines have an ambivalent character. On one hand, up-
regulated production of cytokines in response to CD40 acti-
vation can enhance the immunogenicity through augmented
presentation of tumor-associated antigens by the tumors and
increasing recruitment of immune cells into tumor sites, which
was observed after CD40L stimulation [83]. Enhance produc-
tion of IL-6, IL-8, TNF-α, and GM-CSF and corresponding
caspase activation can contribute to CD40-dependent tumor
growth inhibition [31]. On the other hand, prolonged produc-
tion of IL-6 and GM-CSF and activation NF-κB pathway can
exacerbate the inflammation and induce cancer progression
through intensifying angiogenesis and inhibition of trans-
formed cells apoptosis.
CD40L inhibits in vitro melanoma cell proliferation by
inducing their apoptosis or stimulating the production of
cytokines that activate the immune system (IL-6, IL-8,
TNF-α) by cancer cells [77]. In another study, two indepen-
dent teams assessed the role of the ligand and CD40 receptor
activation in breast cancer. The results reported by Hirano
et al. [84] suggest that the action of the CD40/CD40L pathway
may inhibit the growth and enhancement of apoptosis of
transformed cells. The use of genetically modified CD40L-
srhCD40L by these authors caused death of cancer cells via
9452 Tumor Biol. (2014) 35:94479457
Fas activation. In turn, Tong et al. [73]haverevealedthat
breast cancer cell apoptosis does not require the presence of
INF-γ, whichwas earlier suggested by Wingett et al. [85]. The
authors showed that modified sCD40L by binding on cancer
cells inhibits tumor growth, whereas the receptor-free trans-
formed cells do not undergo apoptosis.
Some authors believe that the CD40L/CD40 pathway may
inhibit cancer progression through only one mechanism, i.e.,
enhanced immune response or apoptosis induction. However,
most researchers favor the theory that anti-neoplastic activity
is only possible when both mechanisms by CD40L are in-
volved in anti-cancer therapy.
Since CD40L and CD40 molecules are strong
immunostimulators, they are considered useful in anti-
cancer therapy. Studies on novel methods of treatment
have been conducted on murine models, using anti-CD40
and anti-CD40L antibodies, recombinant CD40L, and
transformed viral vectors, carrying encoding sequences/
ligand genes or vaccines, to the production of which
dendritic cells containing tumor peptides and CD40L
gene are used [86,87].
The activation of antigen-presenting cells (APCs, i.e.,
CD4+ T cells and dendritic cells) by CD40L present on
CD4+T cells enhances the specific anti-cancer immune re-
sponse. Therefore, the major therapeutic purpose was target
activation of CD40, which leads to the intensified CD-
dependent T cell activation and the resulting effective anti-
cancer immune response. This assumption is accomplished
with the use of various methods, including agonistic antibod-
ies and recombinant forms of sCD40L. Currently, researchers
are widely interested in the transfer of the CD40L encoding
sequences to various cells, including cancer cells, fibroblasts,
or dendritic cells. The introduction of the CD40L encoding
sequence by means of viral vector (adenoviruses, retroviruses)
to cancer cells leads to the enhancement of the ligand expres-
sion and increased activity of dendritic cells that stay in
contact with cancer cells, which accelerates their maturation
and induces anti-cancer activity [88].
One of the cancer treatment strategies is to use CD40-
ligand/interleukin-2 vaccines. Chronic lymphocytic leukemia
(B-CLL) cells express many of tumor-associated and tumor-
specific antigens but lack co-stimulatory molecules, which are
required for effective antigen presentation. Up-regulating of
CD80, CD86, and CD54 can be caused by CD40L interaction
with CD40, and further immunostimulatory effect can be
enhance by IL-2 [89]. Biagi et al. [90] prepared autologous
B-CLL cells expressing human CD40 ligand (hCD40L) and
human interleukin-2 (hIL-2). Vaccine leads to CD4+ and
CD8+ effector cells reactive with autologous B-CLL cells
and consequently to immunomodulatory effect. In non-
Hodgkins lymphoma, cells after cultured with embryonic
lung fibroblasts were transducted by adenoviral vector with
Adh CD40L and Adh IL2. This combination caused enhanced
initial T cell activation and generation autologous T cells
against B-NHL cells [91].
Murine model research on urinary bladder cancer has
shown that gene therapy with the use of vectors that carry
the CD40L encoding sequence (AdCD40L) has high potential
in the inhibition of tumor growth. The introduction of
AdCD40L has led to enhanced expression of IL-12 and gen-
eration of specific all-systemic defense response against tu-
mor. At the same time, the development and function of
regulator T cells were suppressed in the lymph nodes [92].
Kipps et al. [93] proposed gene therapy for patients suffering
from chronic lymphoblastic leukemia. Leukemia cells were
transduced by Ad-CD154, which promoted antigen presenta-
tion and generation of autologic T cell response. This caused
the formation of cytotoxic T cells, specific to leukemia cells
[65]. Similar results have been reported by researchers who
studied the effect of gene therapy with the use of CD40L on
gastric cancers [94], non-small-cell lung cancer [95], breast
cancer [85], cancers of the uterine cervix, and the prostate
[96]. Studies on the new more effective and safer forms of
vectors that carry genes into the cells are still performed. For
instance, conditionally replicative oncolytic adenovirus
(AdEHCD40L) containing hybrid promoter ERE/HRE that
regulates transgenic gene CD40L has been proposed for the
treatment of breast cancer. The application of gene therapy has
led to the inhibition of tumor growth, enhanced apoptosis of
cancer cells, and turned out to be completely safe for the body
[97,98]. Vardouli et al. [99] assessed the effect of therapy
using recombinant non-replicating adenovirus, showing the
expression of CD40L, replication-defective recombinant ade-
novirus (RAd)-hCD40L (RAd vector expressing hCD40L) on
cancer cells. Their findings indicate that permanent activation
of CD40 by CD40L inhibits proliferation and enhances apo-
ptosis of cancer cells. Transduction of Rad-hCD40L to CD40-
positive cancer cells of the urinary bladder, uterine cervix, and
ovary inhibits strong proliferation of these cells [99].
CD40L genes can be transduced not only to cancer cells,
but also to the healthy ones in order to enhance their activity.
Kikuchi et al. [100] have forwarded the hypothesis that ge-
netically modified CD40L-positive dendritic cells will be able
to activate each other and enhance anti-cancer response after
being introduced to tumor microenvironment. Research
in vivo has confirmed the theory. The use of modified den-
dritic cells (AdmCD40L-modified syngenic DCs) increased
the immune response and suppressed tumor growth. In order
to increase the activity of antigen-presenting cells, therapy is
used with anti-CD40 antibodies (SGN-40). In MM, the use of
these antibodies significantly enhanced cell apoptosis and
decreased the expression of the receptor for IL-6 [101].
CD40L on cell membrane is known as a homotrimeric
molecule, which allows its binding to the receptor and activa-
tion of the signaling path. At present, an increasing number of
literaturedata suggest that such a structure is indispensable for
Tumor Biol. (2014) 35:94479457 9453
the ligand functioning, although the transmitted signal is too
weak to exert the maximum effect [102]. The modified
multimeric forms of CD40L considerably more strongly acti-
vate the receptor than the homotrimeric form does [103]. This
correlation was applied to CD40L therapies. Naito et al. [108]
have suggested an innovative preparation of human recombi-
nant CD40L (CD40L Tri), in which the ECD of CD40L is
connected to the trimeric motifof CD40L (CD40L-Tri) by a
long elastic peptide bond. CD40L-Tri considerably increases
the population of CD19 B cells and induces their transition
into antigen-presenting cells. The B cells activated by CD40L-
Tri can effectively stimulate the T cell-dependent response.
Also, the multimeric molecule, SP-D-CD154, being the fusion
of CD40L and lung surfactant protein D (SP-D), activates
proliferation of B cells more strongly than the CD40L-Tri
does [104].
Conclusions
The knowledge of the role of CD40L in normal functioning of
the immune system has initiated a numberof studies that have
revealed not only its involvement in the functioning of T and
B cells, but also its key role in the pathogenesis of such
diseases as atherosclerosis, cardiovascular disorders, and can-
cer. The latest research on the ligand has concentrated around
its practical therapeutic use. At present, the recombinant li-
gand is effectively used in the treatment of renal failure and
has very promising implications in the therapy for breast
cancer and pancreatic cancer. Moreover, new forms of the
modified ligand, vaccines that contain its gene, and
multimetric forms of the protein are produced. In a few years,
therapies that use CD40L are likely to be part of combined
therapy.
Conflicts of interest None.
Open Access This article is distributed under the terms of the Creative
Commons Attribution License which permits any use, distribution, and
reproduction in any medium, provided the original author(s) and the
source are credited.
References
1. Karpusas M, Hsu YM, Wang JH, Thompson J, Lederman S, Chess
L, et al. 2 Å crystal structure of an extracellular fragment of human
CD40 ligand. Structure. 1995;15:10319.
2. Villa A, Notarangelo LD, DiSanto JP, Macchi PP, Strina D, Frattini
A, et al. Organization of the human CD40L gene: implications for
molecular defects in X chromosome-linked hyper-IgM syndrome
and prenatal diagnosis. Proc Natl Acad Sci. 1994;91:21104.
3. van Kooten C, Banchereau J. CD40-CD40 ligand. JLB.
2000;67:127.
4. Hsu YM, Lucci J, Su L, Ehrenfels B, Garber E, Thomas D.
Heteromultimeric complexes of CD40 ligand are present on the cell
surface of human T lymphocytes. J Biol Chem. 1997;272:9115.
5. Graf D, Muller S, Korthauer U, van Kooten C, Weise C, Kroczek
RA. A solubleform of TRAP (CD40 ligand) is rapidly released after
T cell activation. Eur J Immunol. 1995;25:174954.
6. Prasad KS, Andre P, He M, Bao M, Manganello J, Phillips DR.
Soluble CD40 ligand induces beta3 integrin tyrosine phosphoryla-
tion and triggers platelet activation by outside-in signaling. Proc
Natl Acad Sci U S A. 2003;14:1236771.
7. Andre P, Prasad KS, Denis CV, He M, Papalia JM, Hynes RO, et al.
CD40L stabilizes arterial thrombi by a beta3 integrin-dependent
mechanism. Nat Med. 2002;8:24752.
8. Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ.
Molecular mechanism and function of CD40/CD40L engagement
in the immune system. Immunol Rev. 2009;229:15272.
9. Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Förster
R, Müller-Berghaus G, et al. CD40 ligand on activated
platelets triggers an inflammatory reaction of endothelial
cells. Nature. 1998;391:5914.
10. Kawabe T, Matsushima M, Hashimoto N, Imaizumi K, Hasegawa
Y. CD40/CD40 ligand interactions in immune responses and pul-
monary immunity. Nagoya J Med Sci. 2011;73:6978.
11. Loskoga AS, Eliopoulosb AG. The Janus faces of CD40 in cancer.
Semin Immunol. 2009;21:3017.
12. Bishop GA, Moore CR, Xie P, Stunz LL, Kraus ZJ. TRAF proteins
in CD40 signaling. Adv Exp Med Biol. 2007;597:13151.
13. Loskog A, Tötterman TH. CD40La multipotent molecule for
tumor therapy. Endocr Metab Immune Disord-Drug Targets.
2007;7:238.
14. Saluk-Juszczak J, Królewska K. Rola szlaku CD40/CD40L w
biologicznej aktywności płytek krwi. Część II. Przegląd
Menopauzalny. 2010;6:3715.
15. Krzesz R, Wagner AH, Cattaruzza M, Hecker M. Cytokine-
inducible CD40 gene expression in vascular smooth muscle cells
is mediated by nuclear factor κB and signal transducer and activa-
tion of transcription-1. FEBS Lett. 1999;453:1916.
16. Wagner AH, Gebauer M, Pollok-Kopp B, Hecker M. Cytokine-
inducible CD40 expression in human endothelial cells is mediated
by interferon regulatory factor-1. Blood. 2002;99:5205.
17. Dallman C, Johnson PW, Packham G. Differential regulation of cell
survival by CD40. Apoptosis. 2003;8:4553.
18. Srinivasan SK, Triemer HL, Kirk AD. Combination and adjuvant
therapies to facilitate the efficacy of costimulatory blockade. In:
Kaplan B, Burkhart GJ, Lakkis FG, editors. Immunotherapy in
transplantation: principles and practice. Oxford: Wiley-Blackwell;
2012. p. 40729.
19. Danese S, Sans M, Fiocchi C. The CD40/CD40L costimulatory
pathway in Inflammatory Bowel Disease. Gut. 2004;53:103543.
20. Khanna R, Cooper L, Kienzle N, Moss DJ, Burrows SR, Khanna
KK. Engagement of CD40 antigen with soluble CD40 ligand up-
regulates peptide transporter expression and restores endogenous
processing function in Burkitts lymphoma cells. J Immunol.
1997;159:57825.
21. Denese S, Fiocchi C. Platelet activation and the CD40/CD40L
ligand pathway: mechanisms and implications for human diseases.
Crit Rev Immunol. 2005;25:10321.
22. Zhang Y, Cao HJ, Graf B, Meekins H, Smith TJ, Phipps RP. CD40
engagement up-regulates cyclooxygenase-2 expression and prosta-
glandin E2 production in human lung fibroblasts. J Immunol.
1998;160:10537.
23. Inwald DP, McDowall A, Peters MJ, Callard RE, Klein NJ, et al.
CD40 is constitutively expressed on platelets and provides a novel
mechanism for platelet activation. Circ Res. 2003;92:9446.
9454 Tumor Biol. (2014) 35:94479457
24. Danese S, de la Motte C, Reyes BM, Sans M, Levine AD, Fiocchi
C. T-cells trigger CD40-dependent platelet activation and granular
RANTES release: a novel pathway for immune response amplifi-
cation. J Immunol. 2004;172:20115.
25. Jinchuan Y, Zonggui W, Jinming C, Li L, Xiantao K. Upregulation
of CD40-CD40 ligand system in patients with diabetes mellitus.
Clin Chim Acta. 2004;339:8590.
26. Lutgens E, Gorelik L, Daemen MJ, de Muinck ED, Grewal IS,
Koteliansky VE, et al. Requirement for CD154 in the progression of
atherosclerosis. Nat Med. 1999;5:13136.
27. Pamukcu B, Lip GYH, Snezhitskiy V, Shantsila E. The CD40-
CD40L system in cardiovascular disease. Ann Med. 2011;43:331
40.
28. Schönbeck U, Varo N, Libby P, Buring J, Ridker PM. Soluble
CD40L and cardiovascular risk in women. Circulation. 2001;104:
22668.
29. Heeschen C, Dimmeler S, Hamm CW, van den Brand MJ, Boersma
E, Zeiher AM, et al. Soluble CD40 ligand in acute coronary syn-
dromes. N Engl J Med. 2003;348:110411.
30. Bereznaya NM, Chekhun VF. Expression of CD40 andCD40L on
tumor cells: their interaction and new approach to immunotherapy.
Exp Oncol. 2007;29:112.
31. Tong AT, Stone MJ. Prospects for CD40-directed experimental
therapy of human cancer. Cancer Gene Ther. 2003;10:113.
32. Bergmann S, Pandolfi PP. Giving blood: a new role for CD40 in
tumorigenesis. J Exp Med. 2006;203:240912.
33. Murugaiyan G, Martin S, Saha B. CD40-induced counter current
conduits for tumor escape or elimination? Trends Immunol.
2007;28:46773.
34. Huang J, Jochems C, Talaie T, Anderson A, Jales A, Tsang KY, et al.
Elevated serum soluble CD40 ligand in cancer patients may play an
immunosuppressive role. Blood. 2012;120:30308.
35. Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis
in cancer. Vasc Health Risk Manag. 2006;2:2139.
36. French LE, Tschopp J. Defective death receptor signaling as
a cause of tumor immune escape. Semin Cancer Biol.
2002;12:515.
37. Somasundaram R, Jacob L, Swoboda R, Caputo L, Song H, Basak
S, et al. Inhibition of cytolytic T lymphocyte proliferation by autol-
ogous CD4+/CD25+ regulatory T cells in a colorectal carcinoma
patient is mediated by transforming growth factor-h. Cancer Res.
2002;62:526772.
38. Berger CL, Tigelaar R, Cohen J, Mariwalla K, Trinh J, Wang N,
et al. Cutaneous T-cell lymphoma: malignant proliferation of T-
regulatory cells. Blood. 2005;105:16407.
39. ŁuczyńskiW,Krawczuk-RybakM,Stasiak-BarmutaA.
Experimental and selected clinical aspects of active immunotherapy
in leukemia. Postepy Hig Med Dosw. 2006;60:37986.
40. Ning Y, Qian K, Qi C. Role of vascular endothelial growth factor
receptor in the pro-proliferation activity of CD40-CD40L in AGS
gastric cancer cells. Asian Biomed. 2010;4:797802.
41. Miura S, Tatsuguchi A, Wada K, Takeyama H, Shinji Y, Hiratsuka
T, et al. Cyclooxygenase-2-regulated vascular endothelial growth
factor release in gastric fibroblasts. Am J Physiol Gastrointest Liver
Physiol. 2004;287:44451.
42. Tai YT, Li X, Tong X, Santos D, Otsuki T, Catley L, et al. Human
anti-CD40 antagonist antibody triggers significant antitumor activ-
ity against human multiple myeloma. Cancer Res. 2005;65:5898
906.
43. Farahani M, Treweeke AT, Toh CH, Till KJ, Harris RJ, Cawley JC,
et al. Autocrine VEGF mediates the antiapoptotic effect of CD154
on CLL cells. Leukemia. 2005;19:52430.
44. Byrd JC, Kipps TJ, Flinn IW, Cooper M, Odenike O, Bendiske J,
et al. Phase I study of the anti-CD40 humanized monoclonal anti-
body lucatumumab (HCD122) in relapsed chronic lymphocytic
leukemia. Leuk Lymphoma. 2012;53:213642.
45. Kluin-Nelemans HC, Beverstock GC, Mollevanger P, Wessels HW,
Hoogendoorn E, Willemzeet R, et al. Proliferation and cytogenetic
analysis of hairy cell leukemia upon stimulation via the CD40
antigen. Blood. 1994;84:313441.
46. Schattner EJ, Mascarenhas J, Bishop J, Yoo DH, Chadburn A, Crow
MK, et al. CD4+ T-cell induction of Fas-mediated apoptosis in
Burkitts lymphoma B cells. Blood. 1996;88:137582.
47. Garrone P, Neidhardt EM, Garcia E, Galibert L, van Kooten C,
Banchereau J. Fas ligation induces apoptosis of CD40-activated
human B lymphocytes. J Exp Med. 1995;182:126573.
48. Castillo R, Mascarenhas J, Telford W, Chadburn A, Friedman SM,
Schattner EJ. Proliferative response of mantle cell lymphoma cells
stimulated by CD40 ligation and IL-4. Leukemia. 2000;14:2928.
49. Kato H, Kagami Y, Nakagawa M, Karnan S, Yatabe Y, Nakamura S,
et al. IL-4/CD40L co-stimulation induces long-term proliferation
for CD10-positive germinal centre anticancer activity B cell-like
diffuse large B-cell lymphoma. Open Leuk J. 2010;3:608.
50. Pham LV, Tamayo AT, Yoshimura LC, Lo P, TerryN, Reid PS, et al.
A CD40 signalosome anchored in lipid rafts leads to constitutive
activation of NF-kappaB and autonomous cell growth in B cell
lymphomas. Immunity. 2002;16:3750.
51. Voorzanger-Rousselot N, Blay JY. Coexpression of CD40 and
CD40L on B lymphoma and carcinoma cells: an autocrine anti-
apoptotic role. Leuk Lymphoma. 2004;45:123945.
52. Voorzanger-Rousselot N, Alberti L, Blay JY. CD40L induces mul-
tidrug resistance to apoptosis in breast carcinoma and lymphoma
cells through caspase independent and dependent pathways. BMC
Cancer. 2006;18:75.
53. Hermann A, Rauch BH, Braun M, Schror K, Weber AA. Platelet
CD40 ligand (CD40L)-subcellular localization, regulation of ex-
pression, and inhibition by clopidogrel. Platelets. 2001;12:7482.
54. Yamaguchi H, Tanaka F, Sadanaga N, Ohta M, Inoue H, Mori M.
Stimulation of CD40 inhibits Fas- or chemotherapy-mediated apo-
ptosis and increases cell motility in human gastric carcinoma cells.
Int J Oncol. 2003;23:1697702.
55. Li R, Chen W-C, Pang X-Q, Hua C, Li L, Zhang X-G. Expression of
CD40 and CD40L in gastric cancer tissue and its clinical signifi-
cance. Int J Mol Sci. 2009;10:390017.
56. Jakobson E, Jonsson G, Bjorck P, Paulie S. Stimulation of CD40 in
human bladder carcinoma cells inhibits anti-Fas/APO-1 (CD95)-
induced apoptosis. Int J Cancer. 1998;77:84953.
57. He S, Zhao H, Fei M, Wu Y, Wang L, Zhu X, et al.
Expression of the co-signaling molecules CD40-CD40L and
their growth inhibitory effect on pancreatic cancer in vitro.
Oncol Rep. 2012;28:2628.
58. Roselli M, Mineo TC, Basili S, Martini F, Mariotti S, Aloe S, et al.
Soluble CD40 ligand plasma levels in lung cancer. Clin Cancer Res.
2004;10:6104.
59. Amirkhosravi A, Amaya M, Desai H, Francis JL. Platelet-CD40
ligand interaction with melanoma cell and monocyte CD40 en-
hances cellular procoagulant activity. Blood Coagul Fibrinolysis.
2002;13:50512.
60. Pinzon-Charry A, Schmidt CW, Lopez JA. The key role of CD40
ligand in overcoming tumor-induced dendritic cell dysfunction.
Breast Cancer Res. 2006;8:402.
61. Esche C, GambottoA, Satoh Y, Gerein V, Robbins PD, Watkins SC,
et al. CD154 inhibits tumor-induced apoptosis in dendritic cells and
tumor growth. Eur J Immunol. 1999;29:214855.
62. Medema JP, Schuurhuis DH, Rea D, van Tongeren J, de Jong J, Bres
SA, et al. Expression of the serpin serine protease inhibitor 6
protects dendritic cells against cytotoxic T lymphocyteinduced
apoptosis. Differential modulation by T helper type 1 and type 2
cells. J Exp Med. 2001;194:65768.
63. Wurtzen PA, Nissen MH, Claesson MH. Maturation of den-
dritic cells by recombinant human CD40L-trimer leads to a
homogeneous cell population with enhanced surface marker
Tumor Biol. (2014) 35:94479457 9455
expression and increased cytokine production. Scand J
Immunol. 2001;53:57987.
64. Jyothi MD, Khar A. Regulation of CD40L expression on natural
killer cells by interleukin-12 and interferon gamma: its role in the
elicitation of an effective antitumor immune response. Cancer
Immunol Immunother. 2000;49:56372.
65. Hill SC, Youde SJ, Man S, Teale GR, Baxendale AJ, Hislop A, et al.
Activation of Cl carcinoma cells facilitates CTL responses and
augments chemotherapy-induced apoptosis. J Immunol. 2005;174:
4150.
66. Alexandroff AB, Jackson AM, Paterson T, Haley JL, Ross JA,
Longo DL, et al. Role for CD40-CD40 ligand interactions in the
immune response to solid tumours. Mol Immunol. 2000;37:51526.
67. Buhtoiarov IN, Lum HD, Berke G, Sondel PM, Rakhmilevich AL.
Synergistic activation of macrophages via CD40 and TLR9 results in
T cell independent antitumor effects. J Immunol. 2006;176:30918.
68. Küppers R. Molecular biology of Hodgkin lymphoma. Hematology
Am Soc Hematol Educ Program. 2009:491-6.
69.GhiaP,TransidicoP,VeigaJP,SchanielC,SallustoF,
Matsushima K, et al. Chemoattractants MDC and TARC
are secreted by malignant B-cell precursors following CD40
ligation and support the migration of leukemia-specific T
cells. Blood. 2001;98:53340.
70. Sokolowska-Wojdylo M, Wenzel J, Gaffal E, Lenz J, Speuser P,
Erdmann S, et al. Circulating clonal CLA(+) and CD4(+) T cells in
Sezary syndrome express the skin-homing chemokine receptors
CCR4 and CCR10 as well as the lymph node-homing chemokine
receptor CCR7. Br J Dermatol. 2005;152:25864.
71. Szocinski JL, Khaled AR, Hixon J, Halverson D, Funakoshi S,
Fanslow WC, et al. Activation-induced cell death of aggressive
histology lymphomas by CD40 stimulation: induction of bax.
Blood. 2002;100:21723.
72. Baxendale AJ, Dawson CW, Stewart SE, Mudaliar V, Reynolds G,
Gordon J, et al. Constitutive activation of the CD40 pathway pro-
motes cell transformation and neoplastic growth. Oncogene.
2005;24:791323.
73. Tong AT, Papayoti MH, Netto G, Armstrong DT, Ordonez G,
Lawson JM, et al. Growth-inhibitory effects of CD40 ligand
(CD154) and its endogenous expression in human breast cancer.
Clin Cancer Res. 2001;7:691703.
74. Georgopoulos NT, Merrick A, Scott N, Selby PJ, Melcher A,
Trejdosiewicz LK. CD40-mediated death and cytokine secretion
in colorectal cancer: a potential target for inflammatory tumour cell
killing. Int J Cancer. 2007;15:137381.
75. Schwabe RF, Schnabl B, Kweon YO, Brenner DA. CD40 activates
NF-jB and c-Jun N-terminal kinase and enhances chemokine secretion
on activated human hepatic stellate cells. J Immunol. 2001;166:68129.
76. Loro LL, Ohlsson M, Vintermyr OK, Liavaag PG, Jonsson R,
Johannessen AC. Maintained CD40 and loss of polarized CD40
ligand expression in oral squamous cell carcinoma. Anticancer Res.
2001;21:1137.
77. Von Leoprechting A, van der Bruggen P, Pahl HL, Aruffo A, Simon
JC. Stimulation of CD40 on immunogenic human malignant mela-
nomas augments their cytotoxic T lymphocyte-mediated lysis and
induces apoptosis. Cancer Res. 1999;59:128794.
78. Prigione I, Corrias MV, Airoldi I, Raffaghello L, Morandi F, Bocca
P, et al. Immunogenicity of human neuroblastoma. Ann N YAcad
Sci. 2004;1028:6980.
79. Georgopoulos NT, Steele LP, Thomson MJ, Selby PJ, Southgate J,
Trejdosiewicz LK. A novel mechanism of CD40-induced apoptosis
of carcinoma cells involving TRAF3 and JNK/AP-1 activation. Cell
Death Differ. 2006;13:1789801.
80. Bugajska U, Georgopoulos NT, Southgate J, Johnson PW, Graber P,
Gordon J, et al. The effects of malignant transformation on suscep-
tibility of human urothelial cells to CD40-mediated apoptosis. J Natl
Cancer Inst. 2002;94:138195.
81. Young LS, Eliopoulos AG, Gallagher NJ, Dawson CW. CD40 and
epithelial cells: across the great divide. Immunol Today. 1998;19:
5026.
82. Aruffo A, Farrington M, Hollenbaugh D, Li X, Milatovich A,
Nonoyama S, et al. The CD40 ligand, gp39, is defective in activated
T cells from patients with X-linked hyper-IgM syndrome. Cell.
1993;72:291300.
83. Law CL, Grewal IS. Therapeutic interventions targeting
CD40L (CD154) and CD40: the opportunities and chal-
lenges. Therapeutic targets of the TNF superfamily. Adv
Exp Med Biol. 2009;647:836.
84. Hirano A, Longo DL, Taub DD, Ferris DK, Young LS,
Eliopoulos AG, et al. Inhibition of human breast carcinoma
growth by a soluble recombinant human CD40 ligand.
Blood. 1999;93:29993007.
85. Wingett DG, Vestal RE, Forcier K, Hadjokas N, Nielson CP. CD40
is functionally expressed on human breast carcinomas: variable
inducibility by cytokines and enhancement of Fas-mediated apo-
ptosis. Breast Cancer Res Treat. 1998;50:2736.
86. Iida T, Shiba H, Misawa T, Ohashi T, Eto Y, Yanaga Y. Immunogene
therapy against colon cancer metastasis using an adenovirus vector
expressing CD40 ligand. Surgery. 2010;148:92535.
87. Feder-Mengus C, Schultz-Thater E, Oertli D, Marti WR, Heberer
M, Spagnoli GC, et al. Nonreplicating recombinant vaccinia virus
expressing CD40 ligand enhances APC capacity to stimulate spe-
cific CD4+ and CD8+ T cell responses. Hum Gene Ther. 2005;16:
34860.
88. Nakajima A, Kodama T, Morimoto S, Azuma M, Takeda K,
Oshima H, et al. Antitumor effect of CD40 ligand: elicitation of
local and systemic antitumor responses by IL-12 and B7. J
Immunol. 1998;161:19017.
89. Okur FV, Yvon E, Biagi E, Dotti G, Carrum G, Heslop H, et al.
Comparison of two CD40-ligand/interleukin-2 vaccines in patients
with chronic lymphocytic leukemia. Cytotherapy. 2011;13:112839.
90. Biagi E, Rousseau R, Yvon E, Schwartz M, Dotti G, Foster A, et al.
Responses to human CD40 ligand/human interleukin-2 autologous
cell vaccine in patients with B-cell chronic lymphocytic leukemia.
Clin Cancer Res. 2005;11:691623.
91. Takahashi S, Yotnda P, Rousseau R, Mei Z, Smith S, Rill D, et al.
Transgenic expression of CD40L and interleukin-2 induces an
autologous antitumor immune response in patients with non-
Hodgkins lymphoma. Cancer Gene Ther. 2001;8:37887.
92. Loskog AS, Fransson ME, Totterman TT. AdCD40L gene therapy
counteracts T regulatory cells and cures aggressive tumors in an
orthotopic bladder cancer model. Clin Cancer Res. 2005;11:881621.
93. Kipps TJ, Wierda WG, Cantwell MJ, Woods SJ, Rassenti LZ,
Prussak CE. CD40-ligand (CD154) gene therapy for chronic lym-
phocytic leukemia. Blood. 2000;96:291724.
94. Kim SW, Lee HS, Yoon SK, Chung WC, Cho YS, Jeong JJ, et al.
Expression of CD40 in gastric cancer and its effect on the apoptosis
of gastric cancer cells. Korean J Gastroenterol. 2003;42:27482.
95. Roselli M, Mineo TC, Basili S, Mariotti S, Martini F, Bellotti A,
et al. Vascular endothelial growth factor (VEGF-A) plasma levels in
non-small cell lung cancer: relationship with coagulation and plate-
let activation markers. Thromb Haemost. 2003;89:17784.
96. Dzojic H, Loskog A, Tötterman TH, Essand M. Adenovirus-
mediated CD40 ligand therapy induces tumor cell apoptosis and
systemic immunity in the TRAMP-C2 mouse prostate cancer mod-
el. Prostate. 2006;66:8318.
97. Gomes EM, Rodrigues MS, Phadke AP, Butcher LD, Starling C,
Chen S, et al. Antitumor activity of an oncolytic adenoviral-CD40
ligand (CD154) transgene construct in human breast cancer cells.
Clin Cancer Res. 2009;15:131725.
98. Hernandez-Alcoceba R, Pihalja M, Qian D, Clarke MF. New
oncolytic adenoviruses with hypoxia- and estrogen receptor-
regulated replication. Hum Gene Ther. 2002;13:173750.
9456 Tumor Biol. (2014) 35:94479457
99. Vardouli L, Lindqvist C, Vlahou K, Loskog AS, Eliopoulos AG.
Adenovirus delivery of human CD40 ligand gene confers direct
therapeutic effectson carcinomas. Cancer Gene Ther. 2009;16:848
60.
100. Kikuchi T, Moore MAS, Crystal RG. Dendritic cells modified to
express CD40 ligand elicit therapeutic immunity against preexisting
murine tumors. Blood. 2000;1:919.
101. Tai YT, Li XF, Catley L, Coffey R, Breitkreutz I, Bae J, et al.
Immunomodulatory drug lenalidomide (CC-5013, IMiD3) aug-
ments anti-CD40 SGN-40-induced cytotoxicity in human multiple
myeloma: clinical implications. Cancer Res. 2005;65:1171220.
102. Haswell LE, Glennie MJ, Al-Shamkhani A. Analysis of the oligo-
meric requirement for signaling by CD40 using soluble multimeric
forms of its ligand, CD154. Eur J Immunol. 2001;31:3094100.
103. Stone GW, Barzee S, Snarsky V, Kee K, Spina CA, Yu XF, et al.
Multimeric soluble CD40 ligand and GITR ligand as adjuvants for
human immunodeficiency virus DNA vaccines. J Virol. 2006;80:
176272.
104. Naito M, Hainz U, Burkhardt UE, Fu B, Ahove D, Stevenson KE,
et al. CD40L-Tri, a novel formulation of recombinant human
CD40L that effectively activates B cells. Cancer Immunol
Immunother. 2013;62:34757.
Tumor Biol. (2014) 35:94479457 9457
... Studies revealed that patients with type 2 diabetes have a significantly higher concentration of sCD40L than healthy individuals [23][24][25]. Receptor membrane ligand CD40L (CD154) is found on various cell surfaces, including B and T lymphocytes, natural killer cells, basophils, monocytes/macrophages, mastocytes, endothelial cells, dendritic cells, and platelets [26][27][28]. The interaction of its soluble form (sCD40L) with its receptor CD40 significantly increases the inflammation of the endothelium via the enhancement of IL-6 production, which also affects thrombocytopoiesis [4,26]. ...
... Receptor membrane ligand CD40L (CD154) is found on various cell surfaces, including B and T lymphocytes, natural killer cells, basophils, monocytes/macrophages, mastocytes, endothelial cells, dendritic cells, and platelets [26][27][28]. The interaction of its soluble form (sCD40L) with its receptor CD40 significantly increases the inflammation of the endothelium via the enhancement of IL-6 production, which also affects thrombocytopoiesis [4,26]. Additionally, sCD40L increases the expression of P-selectin [28]. ...
... Literature data indicate that besides TPO, IL-6, sCD40L, and sP-selectin [4,13,15,20,26,28], other factors can influence blood platelet activation and the MPV value [20]. Various factors have been identified, such as the percentage of glycated hemoglobin (HbA1c), fasting and postprandial blood glucose concentration, diabetes duration, platelet morphological parameters (platelet distribution width and large platelet ratio), retinopathy, and microalbuminuria, which have been indicated as factors influencing the MPV value in patients with type 2 diabetes [2,8,29,30]. ...
Article
Full-text available
Background Thromboembolic episodes, which are largely mediated by blood platelets, are prevalent chronic complications of diabetes. The mean platelet volume (MPV) serves as a marker for in vivo platelet activation. This study aimed to assess the factors influencing MPV in 106 patients with type 2 diabetes, compared with 59 non-diabetic individuals at a single center in Poland. Material/Methods We performed linear regression analysis, with MPV as the dependent variable and factors such as age, sex, thrombopoiesis-influencing cytokines, blood pressure, body mass index, glycosylated hemoglobin percentage, platelet count, large platelet count, lipid profile parameters, creatinine concentration, estimated glomerular filtration rate, treatment modalities, and comorbidities as independent variables. MPV was measured using the ADVIA 2120 hematology analyzer, with a reference range of 7–12 fL. Results The analysis revealed that in patients with type 2 diabetes, an increase in platelet count by 10×10³/μL resulted in a decrease in MPV by 0.05 (P<0.001), while an increase in large platelet count by 1×10³/μL led to an increase in MPV by 0.18 (P<0.001). Additionally, patients taking β-blockers or insulin had lower MPVs by 0.77 (P=0.008) and 5.63 (P<0.001), respectively, compared with those not on these medications. Conclusions This study delineates the relationship between MPV, platelet parameters, and treatment modalities in type 2 diabetes, paving the way for further research to elucidate underlying mechanisms and potential clinical applications.
... CD40 is a 48 kDa transmembrane cell surface glycoprotein belonging to the tumor necrosis factor receptor (TNFR) superfamily [7,8]. It is expressed in diverse cell types and can be detected on APCs, including dendritic cells (DCs), macrophages, B cells, and several other cell types such as neutrophils, endothelial cells, smooth muscle cells, fibroblasts, and epithelial cells [7][8][9][10]. CD40 is also present on the membranes of a wide range of malignant cells, including B cell malignancies and different carcinomas [8,11]. ...
... CD40 is also present on the membranes of a wide range of malignant cells, including B cell malignancies and different carcinomas [8,11]. The CD40 ligand (CD40L) is a transmembrane protein that is expressed primarily by activated T cells, but also by B cells, platelets, mast cells, macrophages, basophils, and natural killer (NK) cells [8,10]. Binding of CD40 to CD40L activates an intracellular signal transduction pathway that involves a series of adapter molecules known as TNFR activation factors (TRAF). ...
Article
Introduction: There is a need for new therapies that can enhance response rates and broaden the number of cancer indications where immunotherapies provide clinical benefit. CD40 targeting therapies provide an opportunity to meet this need by promoting priming of tumor-specific T cells and reverting the suppressive tumor microenvironment. This is supported by emerging clinical evidence demonstrating the benefits of immunotherapy with CD40 antibodies in combination with standard of care chemotherapy. Areas covered: This review is focused on the coming wave of next-generation CD40 agonists aiming to improve efficacy and safety, using new approaches and formats beyond monospecific antibodies. Further, the current understanding of the role of different CD40 expressing immune cell populations in the tumor microenvironment is reviewed. Expert opinion: There are multiple promising next-generation approaches beyond monospecific antibodies targeting CD40 in immuno-oncology. Enhancing efficacy is the most important driver for this development, and approaches that maximize the ability of CD40 to both remodel the tumor microenvironment and boost the anti-tumor T cell response provide great opportunities to benefit cancer patients. Enhanced understanding of the role of different CD40 expressing immune cells in the tumor microenvironment may facilitate more efficient clinical development of these compounds.
... However, some studies suggest the opposite result, indicating that CD40/CD40L may be closely associated with tumorigenesis [76]. CD40 is expressed on the surface of normal cells and cancer cells of the bladder, lung, and ovary [77][78][79] and is highly expressed in malignant hematological tumors [80]. CD40L is highly expressed in many cancers, but its tumorigenic functions in neoplastic disease remain controversial [81]. ...
Article
Full-text available
Allogeneic tumors are eradicated by host immunity; however, it is unknown how it is initiated until the report in Nature by Yaron Carmi et al. in 2015. Currently, we know that allogeneic tumors are eradicated by allogeneic IgG via dendritic cells. AlloIgG combined with the dendritic cell stimuli tumor necrosis factor alpha and CD40L induced tumor eradication via the reported and our proposed potential signaling pathways. AlloIgG triggers systematic immune responses targeting multiple antigens, which is proposed to overcome current immunotherapy limitations. The promising perspectives of alloIgG immunotherapy would have advanced from mouse models to clinical trials; however, there are only 6 published articles thus far. Therefore, we hope this perspective view will provide an initiative to promote future discussion.
... Although the interaction of CD40/ CD40L is manifested to regulate the proliferation and activation of B and T lymphocytes, it induces apoptosis in tumor cells, suggesting its role in generating a potential anti-tumor response [138,139]. The binding of CD40 to CD40L leads to the recruitment of TNFR-associated factors (TRAFs) to the cytoplasmic domain of CD40. ...
Article
Objective Recent scientific advances have expanded insight into the immune system and its response to malignant cells. In the past few years, immunotherapy has attained a hallmark for cancer treatment, especially for patients suffering from the advanced-stage disease. Modulating the immune system by blocking various immune checkpoint receptor proteins through monoclonal antibodies has improved cancer patients' survival rates. Methods The scope of this review spans from 1985 to the present day. Many journals, books, and theses have been used to gather data, as well as Internet-based information such as Wiley, PubMed, Google Scholar, ScienceDirect, EBSCO, SpringerLink, and Online electronic journals. Key findings Current review elaborates on the potential inhibitory and stimulatory checkpoint pathways which are emerged and have been tested in various preclinical models, clinical trials, and practices. Twenty-odd such significant checkpoints are identified and discussed in the present work. Conclusion A large number of ongoing studies reveal that combination therapies that target more than one signaling pathway may become effective in order to maximize efficacy and minimize toxicity. Moreover, these immunotherapy targets can be a part of integrated therapeutic strategies in addition to classical approaches. It may become a paradigm shift as a promising strategy for cancer treatment.
... The interaction of CD40 with its ligand (CD40L/ CD154) activates innate and adaptive immune responses involving T-and B-lymphocytes ( Supplementary Fig. S1) [4,5]. These pivotal CD40/CD40L-mediated pathways have been shown to play critical roles in promoting tumor progression, and inducing tumor cell apoptosis [6]. ...
Article
Full-text available
Mitazalimab is an agonistic human monoclonal antibody targeting CD40, a target for anti-tumor immunotherapy. This phase 1, dose-escalation study evaluated the safety, dose-limiting toxicities (DLTs), pharmacokinetic and pharmacodynamic profile of mitazalimab. Adults with advanced solid malignancies received mitazalimab intravenously once every-2-weeks. Dose-escalation was pursued with and without pre-infusion corticosteroids for mitigation of infusion-related reactions (IRRs). In all, 95 patients were enrolled in 7 cohorts (n = 50, 75–2000 µg/kg) with corticosteroids and in 5 cohorts (n = 45, 75–1200 µg/kg) without corticosteroids. Two patients experienced DLTs (transient Grade-3 headache; Grade-3 drug-induced liver injury [Hy's law]). The most frequently reported (≥ 25%) treatment-emergent adverse events were fatigue (44.2%), pyrexia (38.9%), pruritus (38.9%), chills (27.4%), and headache (26.3%). IRRs were reported in 51.6% of patients; pruritus (30.5%; with corticosteroids [36.0%], without corticosteroids [24.4%]) was the most frequent. Following the first infusions of 600 μg/kg and 2000 μg/kg, mitazalimab was rapidly cleared from the systemic circulation with mean terminal half-life of 11.9 and 24.1 h, respectively. Pharmacokinetics appeared to exhibit target-mediated drug disposition at the tested doses. Mitazalimab treatment induced higher levels of selected chemokines and transient reduction of B-cells, T-cells, and NK cells. One patient (renal cell carcinoma) displayed partial response lasting 5.6 months. Stable disease was reported by 35 (36.8%) patients, persisting for ≥ 6 months in 9 patients. Mitazalimab has a manageable safety profile with acceptable pharmacokinetic and pharmacodynamic properties. Future clinical development will evaluate combination with existing treatment options. Trial registration NCT02829099 (ClinicalTrials.gov; July 7, 2016).
... Costimulatory receptor CD40 is expressed on APCs, including B cells, macrophages, and DCs. It has been demonstrated that the association between CD40 and its ligand CD40L enhances the release of cytokines, accelerates MHC class II-dependent antigen presentation, increases T cell priming, and induces cancer cell death [116]. Zafar et al. (2018) reported that an adenoviral vector expressing CD40L (Ad3-hTERT-CMV-hCD40L) enhanced the activation of DCs, which in turn led to the induction of Th1 immune responses. ...
Article
Full-text available
Cancer is among the major leading causes of mortality globally, and chemotherapy is currently one of the most effective cancer therapies. Unfortunately, chemotherapy is invariably accompanied by dose-dependent cytotoxic side effects. Recently, genetically engineered adenoviruses emerged as an alternative gene therapy approach targeting cancers. This review focuses on the characteristics of genetically modified adenovirus and oncology clinical studies using adenovirus-mediated gene therapy strategies. In addition, modulation of the tumor biology and the tumor microenvironment as well as the immunological responses associated with adenovirus-mediate cancer therapy are discussed.
Article
Full-text available
Background Precursor plasma cell disorders such as monoclonal gammopathy of undetermined significance (MGUS) always precede the development of active malignancies such as multiple myeloma (MM). There is a need for novel biomarkers to identify those patients with such precursor plasma cell disorders who rapidly progress to MM. Plasma‐derived extracellular vesicles (EVs) may serve as a reservoir of potential biomarkers that can shed light on the pathogenesis and disease biology of MM. Methods This study isolated small EVs (SEVs) and large EVs (LEVs) from the platelet‐poor peripheral blood plasma of MGUS (n = 9) and MM (n = 12) patients using the size exclusion chromatography‐based method and evaluated their proteome using a label‐free proteomics workflow. Results In total, 2055 proteins were identified in SEVs, while 2794 proteins were identified in LEVs. The transferrin receptor (or CD71) protein was upregulated in both populations of EVs derived from MM patients compared to MGUS patients and was of prognostic significance. Similarly, three isoforms of serum amyloid A (SAA) protein, SAA1, SAA2, and SAA4, were also highly upregulated in SEVs within MM patients relative to MGUS patients. Finally, CD40 expression was also higher in the LEVs derived from MM patients than in MGUS patients. Conclusions This study demonstrates the feasibility of successfully isolating both SEVs and LEVs from the peripheral blood of patients with plasma cell disorders and quantifying protein biomarkers within these EVs that could be of prognostic and diagnostic interest.
Article
Full-text available
BACKGROUND: Colorectal cancer (CRC) is often associated with elevated platelet count (> 400 × 109/L), known as thrombocytosis. The role of CD40 ligand (CD40L), a member of the tumor necrosis factor family, is controversial in CRC. Circulating CD40L is higher in CRC, but its relationship with disease staging and local and distant metastasis is not clear. Although most of the circulating CD40L is produced by platelets, no previous study investigated its relationship with CRC-related thrombocytosis. AIM: To investigate the role of CD40L to predict the outcome of CRC and its relation to thrombocytosis. METHODS: A total of 106 CRC patients and 50 age and sex-matched control subjects were enrolled for the study. Anamnestic data including comorbidities and histopathological data were collected. Laboratory measurements were performed at the time of CRC diagnosis and 1.5 mo and at least 6 mo after the surgical removal of the tumor. Plasma CD40L and thrombopoietin were measured via enzyme-linked immunosorbent assay, while plasma interleukin-6 was measured via electrochemiluminescence immunoassay. Patient follow-ups were terminated on January 31, 2021. RESULTS: Plasma CD40L of CRC patients was tendentiously higher, while platelet count (P = 0.0479), interleukin-6 (P = 0.0002), and thrombopoietin (P = 0.0024) levels were significantly higher as opposed to the control subjects. Twelve of the 106 CRC patients (11.3%) had thrombocytosis. Significantly higher CD40L was found in the presence of distant metastases (P = 0.0055) and/or thrombocytosis (P = 0.0294). A connection was found between CD40L and platelet count (P = 0.0045), interleukin-6 (P = 0.0130), and thrombocytosis (P = 0.0155). CD40L was constant with the course of CRC, and all baseline differences persisted throughout the whole study. Both pre- and postoperative elevated platelet count, CD40L, and interleukin-6 level were associated with poor overall and disease-specific survival of patients. The negative effect of CD40L and interleukin-6 on patient survival remained even after the stratification by thrombocytosis. CONCLUSION: CD40L levels of CRC patients do not change with the course of the disease. The CD40L level is strongly correlated with platelet count, interleukin-6, thrombocytosis, and the presence of distant metastases.
Article
Full-text available
Background The combination of antiangiogenic agents with immune checkpoint inhibitors could potentially overcome immune suppression driven by tumor angiogenesis. We report results from a phase IB study of ziv-aflibercept plus pembrolizumab in patients with advanced solid tumors. Methods This is a multicenter phase IB dose-escalation study of the combination of ziv-aflibercept (at 2–4 mg/kg) plus pembrolizumab (at 2 mg/kg) administered intravenously every 2 weeks with expansion cohorts in programmed cell death protein 1 (PD-1)/programmed death-ligand 1(PD-L1)-naïve melanoma, renal cell carcinoma (RCC), microsatellite stable colorectal cancer (CRC), and ovarian cancer. The primary objective was to determine maximum tolerated dose (MTD) and recommended dose of the combination. Secondary endpoints included overall response rate (ORR) and overall survival (OS). Exploratory objectives included correlation of clinical efficacy with tumor and peripheral immune population densities. Results Overall, 33 patients were enrolled during dose escalation (n=3) and dose expansion (n=30). No dose-limiting toxicities were reported in the initial dose level. Ziv-aflibercept 4 mg/kg plus pembrolizumab 2 mg/kg every 2 weeks was established as the MTD. Grade ≥3 adverse events occurred in 19/33 patients (58%), the most common being hypertension (36%) and proteinuria (18%). ORR in the dose-expansion cohort was 16.7% (5/30, 90% CI 7% to 32%). Complete responses occurred in melanoma (n=2); partial responses occurred in RCC (n=1), mesothelioma (n=1), and melanoma (n=1). Median OS was as follows: melanoma, not reached (NR); RCC, 15.7 months (90% CI 2.5 to 15.7); CRC, 3.3 months (90% CI 0.6 to 3.4); ovarian, 12.5 months (90% CI 3.8 to 13.6); other solid tumors, NR. Activated tumor-infiltrating CD8 T cells at baseline (CD8+PD1+), high CD40L expression, and increased peripheral memory CD8 T cells correlated with clinical response. Conclusion The combination of ziv-aflibercept and pembrolizumab demonstrated an acceptable safety profile with antitumor activity in solid tumors. The combination is currently being studied in sarcoma and anti-PD-1-resistant melanoma. Trial registration number NCT02298959 .
Article
Full-text available
Background: CD40 is a type α-membrane protein of the tumor necrosis factor receptor super-family, and CD40-induced responses may mediate growth and angiogenesis in carcinoma cells. Objectives: Define the effect of CD40 ligation on AGS gastric cancer cell line and the role of vascular endothelial growth factor/vascular endothelial growth factor receptor (VEGF/VEGFR) signals in this process. Methods: We treated AGS cells with 1 μg/mL soluble CD40 ligand (sCD40L) with or without pre-incubation of either anti-VEGF mAb (MAB293) or VEGFR tyrosine kinase inhibitor (SU5416). We determined the growth effects by cell counts or [3H]-thymidine incorporation assay and VEGF levels in cell-free supernatant using enzyme-linked immunosorbent assays. Results: The engagement of CD40-induced AGS cells proliferation accompanied by a significant increase autocrine VEGF through PI3K activation (p <0.05), and exogenous VEGF alone had no effect on spontaneous cell growth. SU5416 with a concentration of 8 μM lead to a dramatic decrease in cell survival induced by sCD40L (p <0.05), whereas MAB293 did not have the similar effect (p >0.05). Conclusion: CD40-CD40L interaction promoted AGS cancer cell line proliferation through a VEGFR-dependent signal pathway in the presence of an internal autocrine loop.
Article
CD40 ligand (CD40L) is essential for the initiation of antigen-specific T-cell responses. This study is based on the hypothesis that dendritic cells (DCs) genetically modified ex vivo to express CD40L will enhance in vivo presentation of tumor antigen to the cellular immune system with consequent induction of antitumor immunity to suppress tumor growth. To examine this concept, subcutaneous murine tumors were injected with bone marrow-derived DCs that had been modified in vitro with an adenovirus (Ad) vector expressing murine CD40L (AdmCD40L). In B16 (H-2b, melanoma) and CT26 (H-2d, colon cancer) murine models, intratumoral injection of 2 × 106 AdmCD40L-modified DCs (CD40L-DCs) to established (day 8) subcutaneous tumors resulted in sustained tumor regression and survival advantage. This antitumor effect was sustained when the number of CD40L-DCs were reduced 10-fold to 2 × 105. Analysis of spleens from CD40L-DC–treated animals demonstrated that CD40L-DCs injected into the subcutaneous CT26 flank tumors migrated to the spleen, resulting in activation of immune-relevant processes. Consistent with this concept, intratumoral administration of CD40L-DCs elicited tumor-specific cytotoxic T-lymphocyte responses, and the transfer of spleen cells from CD40L-DC–treated mice efficiently protected naive mice against a subsequent tumor challenge. In a distant 2-tumor model of metastatic disease, an untreated B16 tumor in the right flank regressed in parallel with a left B16 tumor treated with direct injection of CD40L-DCs. These results support the concept that genetic modification of DCs with a recombinant CD40L adenovirus vector may be a useful strategy for directly activating DCs for cancer immunotherapy.
Article
Chronic lymphocytic leukemia (CLL) cells can be made to express recombinant CD40-ligand (CD154) by transduction with a replication-defective adenovirus vector (Ad-CD154). Ad-CD154–transduced and bystander leukemia cells become highly effective antigen-presenting cells that can induce CLL-specific autologous cytotoxic T lymphocytes in vitro. This study investigated the immunologic and clinical responses to infusion of autologous Ad-CD154-CLL cells in patients with CLL. After a one-time bolus infusion of autologous Ad-CD154–transduced leukemia cells, there was increased or de novo expression of immune accessory molecules on bystander, noninfected CLL cells in vivo. Treated patients also developed high plasma levels of interleukin-12 and interferon-γ, the magnitudes of which corresponded to absolute blood CD4+T-cell counts before therapy. On average, patients experienced a greater than 240% increase in absolute blood T-cell counts within 1 to 4 weeks of treatment. Moreover, treatment increased the numbers of leukemia-specific T cells, demonstrated by autologous ELISPOT assay and mixed lymphocyte reactions. These biologic effects were associated with reductions in leukemia cell counts and lymph node size. Treatment did not induce autoimmune thrombocytopenia or hemolytic anemia and no dose-limiting toxicity was observed. This approach may provide a novel and effective form of gene therapy for patients with this disease.
Article
Using the CD40 system, in vitro proliferation of hairy cell leukemia (HCL) was examined in 43 patients. In this culture system, cells were stimulated by interleukin-4 (IL-4) and anti-CD40 monoclonal antibodies (MoAbs) that were added in soluble form or were cross-linked via their Fc part using Fc gamma RII-transfected mouse fibroblast cells. Proliferation was induced and confirmed by 3H-thymidine incorporation in 14 cases and by the presence of metaphases in 42 cases. 3H-thymidine incorporation showed a heterogeneous pattern: cross-linking of anti- CD40 gave the highest proliferation in 8 cases; in 11 cases, stimulation with anti-CD40 MoAbs alone, without cross-linking also resulted in proliferation; the addition of IL-4 further enhanced 3H- thymidine incorporation in 5 cases, but suppressed this phenomenon in 5 other cases. The CD40 system proved to be very effective in obtaining cytogenetic data. With a success rate of 42 of 43 patients tested, we found clonal abnormalities in 8 cases (19%) and nonclonal abnormalities with involvement of one or two abnormal metaphases in another 7 cases. The chromosomes most frequently involved in the abnormal karyotypes, both structurally and numerically, were chromosomes 5, 7, and 14. By fluorescence-activated cell-sorting analysis of the cultured cells, and by immunophenotypic analysis of metaphase spreads, T-cell growth could be excluded and the HCL-lineage confirmed. Stimulation via the CD40 antigen is an excellent tool for growing hairy cell leukemia cells.
Article
CD40 is present on B cells, monocytes, dendritic cells, and endothelial cells, as well as a variety of neoplastic cell types, including carcinomas. CD40 stimulation by an antibody has previously been demonstrated to induce activation-induced cell death in aggressive histology human B-cell lymphoma cell lines. Therefore, we wanted to assess the effects of a recombinant soluble human CD40 ligand (srhCD40L) on human breast carcinoma cell lines. Human breast carcinoma cell lines were examined for CD40 expression by flow cytometry. CD40 expression could be detected on several human breast cancer cell lines and this could be augmented with interferon-γ. The cell lines were then incubated with a srhCD40L to assess effects on in vitro growth. srhCD40L significantly inhibited the proliferation of the CD40+ human breast cancer cell lines. This inhibition could also be augmented with interferon-γ. Viability was also affected and this was shown to be due to increased apoptosis of the cell lines in response to the ligand. Treatment of tumor-bearing mice was then performed to assess the in vivo efficacy of the ligand. Treatment of tumor-bearing SCID mice with the ligand resulted in significant increases in survival. Thus, CD40 stimulation by its ligand directly inhibits human breast carcinoma cells in vitro and in vivo. These results suggest that srhCD40L may be of clinical use to inhibit human breast carcinoma growth.
Article
We have recently demonstrated that murine and human tumors induce apoptosis of dendritic cells (DC). Here, we evaluated the effect of CD40 ligation on the survival of tumor-associated DC and tumor growth. Retroviral transduction of MC38 colon carcinoma cells with the CD154 gene resulted in inhibition of tumor growth. This effect was abrogated in IL-12 knockout mice. Immunohistochemical analysis revealed an increase in CD11c⁺ (N418) and CD8⁺ but not NLDC-145⁺ cells in CD154-transfected tumors in wild-type mice. This increase was less pronounced in IL-12-deficient mice. In vitro, overexpression of CD154 on tumor cells significantly decreased the level of tumor-induced DC apoptosis. Surprisingly, the CD154-induced protection of DC from tumor-induced apoptosis was IL-12 independent in vitro, suggesting an IL-12-dependent and an IL-12-independent mechanism of CD154-induced anti-tumor immunity. Thus, our data suggest a new strategy to improve immunotherapy of cancer by protecting DC from tumor-induced apoptosis.
Article
The main function of blood platelets is their haemostatic role, but recent evidence shows that processes of platelet activation may be sometimes a critical link between haemostasis and development of inflammation. The discovery of expression of CD40 and its ligand - CD40L on platelet surface revealed the participation of these cells in immune responses and inflammation. In many pathological processes related to cardiovascular disorders, e.g. changes connected with menopause, enhanced platelet activation is observed in atherosclerosis, diabetes and cancer, in which proinflammatory and prothrombotic CD40/CD40L pathway is involved. Signals that drive inflammatory events and are transmitted through CD40/CD40L pathway cause the inflammatory cell interactions leading to induction of innate immune response. This signal transduction is associated with the release of adhesion molecules, chemokines, cytokines, tissue factors, reactive oxygen species, growth factors and other proinflammatory mediators from activated cells. Hyperactivity of platelets associated with signal transduction by CD40/CD40L pathway is observed in many pathogenic processes, including thrombosis, diabetes, inflammation and cancers. The inhibition of CD40/CD40L pathway may provide alternative treatment for various diseases in the future.
Chapter
Many drugs are used to prevent allograft rejection. Most target T cells but inhibit pathways that are not exclusive to T cells and thus evoke off-target side effects. Many, particularly calcineurin inhibitors, hinder T cell receptor (TCR) signaling and thus inhibit T cells in a non-antigen-specific manner. Recently, agents targeting T cell costimulation have been developed. Costimulation molecules supplement TCR signaling and are necessary for full T cell activation. Costimulatory molecule-specific monoclonal antibodies and fusion proteins have been shown to prevent rejection in preclinical settings, and by sparing the TCR, their effects have retained antigen specificity. The CD28–CD80/86 and CD40–CD154 pathways are the most studied targets, and agents inhibiting these pathways have reached clinical trials. However, these agents cannot by themselves prevent clinical rejection. This chapter will review costimulation blockade-based agents particularly in regard to their use in combination with adjuvant therapies, including conventional immunosuppressants and agents inhibiting adhesion molecule function.
Article
Since CD40/CD40 ligand (CD40Lig) interactions are essential in vivo for the generation of germinal center B cells that express Fas (Apo-1/CD95), we explored whether CD40 engagement may modulate Fas expression and function on human B lymphocytes. Resting tonsil B cells, isolated by density gradient centrifugation, express either absent or low levels of Fas. They could be induced to promptly express Fas after ligation of their CD40, however, using either a recombinant human CD40Lig or a cross-linked anti-CD40 mAb. In contrast, engagement of the B cell antigen receptor by immobilized anti-kappa and -lambda antibodies did not turn on Fas expression. Addition of anti-Fas mAb CH11 inhibited the later phases of CD40-induced B cell growth as a result of apoptotic cell death. Furthermore, Fas ligation inhibited proliferation and Ig secretion of CD40-activated B cells in response to recombinant cytokines such as interleukin (IL)-2, IL-4, and IL-10, as well as a cytokine-rich supernatant of phytohemagglutinin-activated T cells, indicating that none of those B cell tropic factors were able to prevent the Fas-induced death. Taken together, the present results show that engagement of CD40 antigen on B cells induces Fas expression and sensitizes them to Fas-mediated apoptosis. The delayed functional response to Fas ligation after CD40 activation may represent a way to limit the size of a specific B cell clone that is generated during T-B cell interactions.
Article
Background: The tumor necrosis factor (TNF) superfamily of ligands and receptors mediates immune cell survival. Some members possess a death domain, a protein motif that functions to transmit apoptotic signals, whereas others, such as CD40, do not. CD40 is expressed by both normal and malignant epithelial cells. To investigate the functional significance of this expression, we studied the effects of ligation of CD40, Fas, and TNF receptors (TNFRs) on the proliferation and survival of normal and malignant human urothelial cells and urothelial cells with disabled p53 function. Methods: Normal and malignant human urothelial cells were cultured with soluble TNF family agonists (CD40 ligand [CD40L], TNF-α, anti-Fas antibody, or cocultured with mouse fibroblasts stably transfected with plasmids that caused the cells to constitutively express CD40L or CD32; cell proliferation was estimated by an [³H]thymidine incorporation assay, and apoptosis was determined by Annexin V staining and by a DNA fragmentation assay. Messenger RNA levels for CD40 and potential downstream effector molecules were quantified by polymerase chain reaction-based and ribonuclease protection assays, respectively, and nuclear factor (NF) κB nuclear translocation was detected by immunofluorescence. All statistical tests were two-sided. Results: Soluble trimeric CD40L inhibited the growth of normal and malignant urothelial cells but did not induce apoptosis. Cell surface-presented CD40L induced massive apoptosis in CD40-positive transitional cell carcinoma cells but not in normal urothelial cells. Normal cells underwent CD40L-mediated apoptosis only in the presence of other TNFR agonists. An agonistic anti-CD40 antibody presented on the surface of CD32-transfected fibroblasts also induced apoptosis in transitional cell carcinoma cells and in normal urothelial cells. Apoptotic responses of tumor (but not normal) cells to soluble agonists were enhanced by blocking protein synthesis. Karyotypically normal urothelial cells with disabled p53 function underwent apoptosis during coculture with CD40L-expressing fibroblasts alone but were not additionally sensitive to additional TNFR agonists. Conclusions: Susceptibility to CD40 ligation-induced apoptosis may be a novel mechanism for eliminating neoplastically transformed urothelial cells. Loss of CD40 expression may be an important adaptive mechanism for transitional cell carcinoma development and progression.