ArticlePDF AvailableLiterature Review

Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes

MDPI
Cancers
Authors:

Abstract and Figures

Simple Summary Despite initially responding to treatment, many ovarian cancers recur because of tumor cell heterogeneity, chemoresistance, and the cancer-promoting and immunosuppressive tumor microenvironment. Recurrent tumors account for the reduced overall and progression-free survival of patients with ovarian cancer. Ovarian cancer is commonly accompanied by thrombocytosis and thrombotic events, which implies that platelets may participate in cancer progression via their association with cancer cells and the tumor microenvironment. This review focuses on platelets’ interactions with cellular and acellular components of the tumor microenvironment, including endothelial cells, mesenchymal stem cells, adipocytes, pericytes, immune cells, and extracellular matrix elements, and discusses how these interactions support the proliferation and metastasis of ovarian cancer cells. It also provides an overview of potential therapeutic strategies that obstruct platelets’ protumor effects by reprogramming the tumor microenvironment. Abstract Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Content may be subject to copyright.
Citation: Oncul, S.; Cho, M.S.
Interactions between Platelets and
Tumor Microenvironment
Components in Ovarian Cancer and
Their Implications for Treatment and
Clinical Outcomes. Cancers 2023,15,
1282. https://doi.org/10.3390/
cancers15041282
Academic Editor: David Wong
Received: 13 January 2023
Revised: 7 February 2023
Accepted: 13 February 2023
Published: 17 February 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
cancers
Review
Interactions between Platelets and Tumor Microenvironment
Components in Ovarian Cancer and Their Implications for
Treatment and Clinical Outcomes
Selin Oncul and Min Soon Cho *
Section of Benign Hematology, Department of Pulmonary Medicine, The University of Texas MD Anderson
Cancer Center, Houston, TX 77030, USA
*Correspondence: mscho@mdanderson.org; Tel.: +1-713-792-3987
Simple Summary:
Despite initially responding to treatment, many ovarian cancers recur because of
tumor cell heterogeneity, chemoresistance, and the cancer-promoting and immunosuppressive tumor
microenvironment. Recurrent tumors account for the reduced overall and progression-free survival
of patients with ovarian cancer. Ovarian cancer is commonly accompanied by thrombocytosis
and thrombotic events, which implies that platelets may participate in cancer progression via their
association with cancer cells and the tumor microenvironment. This review focuses on platelets’
interactions with cellular and acellular components of the tumor microenvironment, including
endothelial cells, mesenchymal stem cells, adipocytes, pericytes, immune cells, and extracellular
matrix elements, and discusses how these interactions support the proliferation and metastasis of
ovarian cancer cells. It also provides an overview of potential therapeutic strategies that obstruct
platelets’ protumor effects by reprogramming the tumor microenvironment.
Abstract:
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and
wound healing after blood vessel injury, are also involved in pathological conditions, including
cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated
with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer
and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets.
The communication between activated platelets, cancer cells, and the tumor microenvironment is via
various platelet membrane proteins or mediators released through degranulation or the secretion of
microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote
ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions
between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the
tumor microenvironment influence ovarian cancer progression. It also presents novel potential
therapeutic approaches toward this gynecological cancer.
Keywords:
ovarian cancer; platelet; tumor microenvironment; metastasis; angiogenesis; thrombosis;
immune system
1. Introduction
Ovarian cancer is the fifth most common cause of death among women and the most
lethal gynecological malignancy in the United States [
1
]. Epithelial ovarian cancer, which
accounts for 90% of ovarian cancers, is further categorized into serous, endometrioid, muci-
nous, and clear cell types, in addition to other rare or non-specified subgroups [
2
]. The most
prevalent type of epithelial ovarian cancer, high-grade serous ovarian cancer (HGSOC),
originates in the fallopian tubes or ovarian surface epithelium and disseminates to the
ovaries and peritoneum [
3
]. HGSOC, which carries a poor prognosis, is often aggressive,
diagnosed in its late stages, and has a high capacity for metastasis and ascites formation [
2
].
Cancers 2023,15, 1282. https://doi.org/10.3390/cancers15041282 https://www.mdpi.com/journal/cancers
Cancers 2023,15, 1282 2 of 37
Non-epithelial ovarian cancers (e.g., germ cell and sex cord–stromal) constitute approxi-
mately 10% of all ovarian cancers and are generally less aggressive than epithelial ovarian
cancer [
4
]. Risk factors for ovarian cancer include genetic alterations, including germline
mutations of breast cancer genes 1 and 2 (BRCA1 and BRCA2) [
5
] and alternative mutations
of DNA damage repair-associated genes such as BRIP1,RAD51, and ATM/ATR [
6
,
7
]; Lynch
syndrome [
8
]; and various environmental factors such as hormone replacement therapy [
9
],
not giving birth and/or not breastfeeding [
10
], being overweight [
11
], and frequent tobacco
smoking [
12
]. The conventional therapy for ovarian cancer is surgical excision of the tumor
along with neoadjuvant or adjuvant platinum- or taxane-based chemotherapy [
13
]. Addi-
tional treatments include using agents that target proangiogenic factors, poly (ADP-ribose)
polymerase (PARP), and immune system components [3].
Despite marked improvements in the efficacy of treatments, ovarian cancer is asso-
ciated with high mortality because of late diagnosis due to its anatomical location and
mostly indistinguishable symptoms, high recurrence rate [
2
], primary or acquired resis-
tance to chemotherapy [
3
,
4
], and tumor heterogeneity [
5
]. Thus, more specific and effective
approaches are in demand to inhibit tumor growth and prolong patient progression-free
survival and overall survival. Because the tumor microenvironment (TME) is a princi-
pal contributor to cancer progression and poor therapeutic response [
6
,
7
], targeting TME
components holds excellent potential for more effective cancer management.
The TME comprises cancer cells; cancer stem cells (CSCs); stromal cells such as
fibroblasts; endothelial cells; immune cells; and proteins of the extracellular matrix (ECM)
such as collagen and fibronectin [
8
]. Crosstalk among these components constitutes a
complex signaling network that promotes malignancy and metastasis [
9
]. Accumulating
evidence indicates that TME has a role in the development and progression of ovarian
cancer [
10
,
11
], and many studies manipulate TME in the treatment of ovarian cancer [
12
,
13
].
One of the components of TME in ovarian cancer is extravasated platelets [
14
,
15
]. Mem-
brane proteins on activated platelets, including P-selectin [
16
], GPIb
α
[
17
], GPIIb/IIIa [
18
],
and C-type lectin-like receptor 2 (CLEC-2) [
19
], mediate the binding of platelets to various
elements in the TME. The released contents of granules from activated platelets, such as
growth hormones [
20
], cytokines [
21
], and chemokines [
22
], also affect the TME. Addition-
ally, platelet microparticles [
23
], mitochondria [
24
], and nucleic acids [
25
,
26
] can reshape
the TME response in cancer. The major molecules on platelets or in their granules are
listed in Table 1. We also summarize the protumorigenic and antitumorigenic influences
of various platelet-associated molecules in the TME in Table 2. The interactions between
platelets and the TME components of ovarian cancer are illustrated in Figure 1.
Healthy individuals have platelet counts of between 150,000 and 450,000 per micro-
liter of blood. In contrast, roughly one-third of newly diagnosed ovarian cancer patients
have platelet counts exceeding 450,000 per microliter [
27
]. In patients with ovarian cancer,
thrombocytosis is an adverse prognostic factor associated with elevated serum carcinoma
antigen 125 (CA-125) levels, advanced disease stage, and poor clinical outcomes [
27
,
28
],
as well as the diminished efficacy of secondary cytoreductive surgery [
29
] and chemother-
apy [
30
]. Aside from contributing to the formation of venous thromboembolisms, platelets
contribute to cancer progression via distinct mechanisms, including increasing prolifera-
tion [
31
], epithelial–mesenchymal transition (EMT) [
32
], and anoikis resistance in cancer
cells [
33
]; promoting the formation of the premetastatic niche and metastasis [
33
]; en-
hancing angiogenesis [
27
] and the integrity of tumor vasculature [
34
]; inducing immune
tolerance [35]; and reducing the impact of chemotherapy [30].
Cancers 2023,15, 1282 3 of 37
Cancers 2023, 15, x FOR PEER REVIEW 4 of 38
junctional adhesion molecule; LPA: lysophosphatidic acid; MCP-1: monocyte chemoattractant pro-
tein 1; MIP: migration inhibitory protein; MMP: matrix metalloproteinase; NAP-2: neutrophil-acti-
vating peptide 2; PAF: platelet-activating factor; PAI-1: plasminogen activator inhibitor 1; PAR: pro-
tease-activated receptor; PD-L1: programmed death ligand 1; PDGF: platelet-derived growth factor;
PECAM-1: platelet endothelial cell adhesion molecule 1; PF4: platelet factor 4; PGD2: prostaglandin
D2; PGE2: prostaglandin E2; PGI2: prostaglandin I2; polyP: polyphosphate; RANTES: regulated
upon activation and normal T cell expressed and secreted; S1P: sphingosine 1-phosphate; SDF-1:
stromal-derived factor 1; TGF-β: transforming growth factor-beta; TIMP: tissue inhibitor of metal-
loproteinases; TLR: Toll-like receptor; TNF-α: tumor necrosis factor-alpha; TxA2: thromboxane A2;
uPA: urokinase-type plasminogen; VEGF: vascular endothelial growth factor; vWF: von Willebrand
factor.
Figure 1. The tumor microenvironment (TME) of ovarian cancer. Tumor cell-induced activated
platelets engage with endothelial cells, pericytes, mesenchymal stem cells (MSCs), cancer-associated
fibroblasts (CAFs), adipocytes, immune cells, and extracellular matrix (ECM) elements through di-
rect interaction or by releasing various modulatory factors and platelet microparticles. Platelets pro-
mote the extravasation, differentiation, and activation of these cells, which, in turn, contribute to the
hyper-responsiveness of platelets and thrombosis in a positive feedback loop.
This review focuses on the interactions between platelets and TME components, the
contribution of these interactions to the progression of ovarian cancer, and the plausible
approaches to interrupting this interwork to improve the prognosis of patients with ovar-
ian cancer.
2. Interactions of Platelets with TME Compartments: Endothelial Cells, Pericytes, and
Cancer-Associated Fibroblasts
2.1. Interactions with Endothelial Cells
Figure 1.
The tumor microenvironment (TME) of ovarian cancer. Tumor cell-induced activated
platelets engage with endothelial cells, pericytes, mesenchymal stem cells (MSCs), cancer-associated
fibroblasts (CAFs), adipocytes, immune cells, and extracellular matrix (ECM) elements through direct
interaction or by releasing various modulatory factors and platelet microparticles. Platelets promote
the extravasation, differentiation, and activation of these cells, which, in turn, contribute to the
hyper-responsiveness of platelets and thrombosis in a positive feedback loop.
Table 1. The major components reside on activated platelets or are released from platelet granules.
Location General Function Examples References
Surface
molecules
Integrins α
2
β
1 (GPIa/IIa),
α
5
β
1,
α
6
β
1,
α
L
β
2 (ICAM-2),
αIIbβ3 (GPIIb/IIIa), αVβ3[36]
Selectins P-selectin (CD62P), CLEC-2 [37,38]
Leucine-rich repeat
receptors GPIb-IX-V, TLR1, TLR2, TLR4, TLR6, MMPs [36,39,40]
ADP receptors P2Y1, P2Y12 [41]
Thrombin receptors PAR1, PAR4, GPIbα[42,43]
Tetraspanins CD63, CD9, CD53 [36]
Prostaglandin receptors PGD2 and PGE2 receptors [44]
Prostacyclin receptors PGI2 receptors [44]
Thromboxane receptors TxA2 receptors [45]
Lipid receptors PAF and LPA receptors [46,47]
Ig receptors GPVI, FcγRIIA (CD32), FcεRI (CD23) [48,49]
JAMs JAM-1, JAM-2, JAM-3, PECAM-1 (CD31) [50,51]
Tyrosine kinase receptors Thrombopoietin, leptin, insulin,
PDGF receptors [36]
Immune checkpoints PD-L1, GITRL, OX40L [5254]
Other receptors
Serotonin receptors, GPIV (CD36), IAP (CD47)
complement receptors, CD40, CD40L (CD154) [5559]
Cancers 2023,15, 1282 4 of 37
Table 1. Cont.
Location General Function Examples References
α-granules
Adhesion molecules vWF, αIIbβ3 (GPIIb/IIIa), αVβ3, P-selectin
(CD62P), fibrinogen, fibronectin,
thrombospondin [36,60,61]
Proangiogenic factors VEGF, angiopoietin-1, SDF-1 (CXCL12), S1P,
TGF-β, IL-6, PF4 (CXCL4) [61]
Angiostatic factors Endostatin, angiostatin, thrombospondin-1 [56,61]
Growth factors VEGF, PDGF, EGF, FGF, HGF,
IGF-1, CTGF, TGF-β[6164]
Coagulation-associated components Prothrombin, fibrinogen, factor V, factor VIII,
factor XI, protein S [61,65,66]
Fibrinolytic factors α2-macroglobulin, uPA, PAI-1 [61]
MMPs MMP-1, MMP-2, MMP-3, MMP-9 [67,68]
Metalloproteinases ADAM-10, ADAM-17, ADAMTS-13 [69]
TIMPs TIMP-1, TIMP-2, TIMP-4 [69]
Inflammamodulatory molecules
CXCL1, PF4 (CXCL4), CXCL5, CXCL7
(NAP-2), IL-1β, IL-6, IL-8 (CXCL8), SDF-1
(CXCL12), CCL2 (MCP-1), CCL3 (MIP-1α),
CCL5 (RANTES), CCL7, PAF, LPA, TGF-β,
TNF-α, GM-CSF
[61,7075]
Immunologic molecules Complement factors, IgG, IgA,
IgM, thymosin-β4[61,7678]
Other components Albumin, α1-antitrypsin,
HMWK [61]
δ-granules
Nucleotides ADP, ATP, GDP, GTP [79]
Bioactive amines Serotonin, histamine, epinephrine [60]
Ions Calcium, magnesium, phosphate,
pyrophosphate [61]
Polyphosphates Polyphosphate (polyP) [60]
Lysosomes
Proteases Cathepsin D/E, carboxypeptidase A/B,
glycohydrolases, collagenase, elastase [79,80]
Phosphatases Acid phosphatase [79]
Phospholipases Phospholipase A [61]
ADAM: a disintegrin and metalloproteinase; CCL: chemokine (C-C motif) ligand; CD40L: CD40 ligand;
CLEC-2: C-type lectin-like receptor 2; CTGF: connective tissue growth factor; CXCL: chemokine (C-X-C mo-
tif) ligand; EGF: epidermal growth factor; FGF: fibroblast growth factor; GITRL: glucocorticoid-induced tu-
mor necrosis factor receptor-related protein ligand; GM-CSF: granulocyte-monocyte colony-stimulating factor;
GP: glycoprotein; HGF: hepatocyte growth factor; HMWK: high-molecular-weight kininogen; IAP: integrin-
associated protein; ICAM-2: intercellular adhesion molecule 2; Ig: immunoglobulin; IGF-1: insulin-like growth
factor 1; IL: interleukin; JAM: junctional adhesion molecule; LPA: lysophosphatidic acid; MCP-1: monocyte
chemoattractant protein 1; MIP: migration inhibitory protein; MMP: matrix metalloproteinase; NAP-2: neutrophil-
activating peptide 2; PAF: platelet-activating factor; PAI-1: plasminogen activator inhibitor 1; PAR: protease-
activated receptor; PD-L1: programmed death ligand 1; PDGF: platelet-derived growth factor; PECAM-1: platelet
endothelial cell adhesion molecule 1; PF4: platelet factor 4; PGD2: prostaglandin D2; PGE2: prostaglandin E2;
PGI2: prostaglandin I2; polyP: polyphosphate; RANTES: regulated upon activation and normal T cell expressed
and secreted; S1P: sphingosine 1-phosphate; SDF-1: stromal-derived factor 1; TGF-
β
: transforming growth factor-
beta; TIMP: tissue inhibitor of metalloproteinases; TLR: Toll-like receptor; TNF-
α
: tumor necrosis factor-alpha;
TxA2: thromboxane A2; uPA: urokinase-type plasminogen; VEGF: vascular endothelial growth factor; vWF: von
Willebrand factor.
This review focuses on the interactions between platelets and TME components, the
contribution of these interactions to the progression of ovarian cancer, and the plausi-
ble approaches to interrupting this interwork to improve the prognosis of patients with
ovarian cancer.
Cancers 2023,15, 1282 5 of 37
2. Interactions of Platelets with TME Compartments: Endothelial Cells, Pericytes, and
Cancer-Associated Fibroblasts
2.1. Interactions with Endothelial Cells
2.1.1. In Angiogenesis
Tumor angiogenesis involves degradation of the vascular endothelial matrix, the pro-
liferation and migration of endothelial cells, the branching of endothelial cells to generate
vascular rings, and the establishment of new basement membranes [
81
]. Tumor blood
vessels, which tend to be erratic, branched, and leaky, are dissimilar to normal blood vessels
in terms of shape, integrity, and permeability. Moreover, perivascular cells are reduced in
number and are less likely to be associated with endothelial cells [
82
,
83
]. Blood flow in
tumor-associated vessels is inconsistent and may lead to maladjusted circulation [
82
,
84
].
Consequently, tumors cannot receive adequate oxygen and nutrients, and discharge excess
carbon dioxide and other metabolites generated by the glycolytic pathway. The TME
becomes more hypoxic, acidic, and ischemic [
85
]. In addition, the hyperpermeability of the
tumor vasculature enhances extravascular clotting, fibrin gel clot formation, and endothe-
lial and stromal cell expansion [
86
]. Angiogenesis is a poor prognostic factor in ovarian
cancer [
87
], and antiangiogenic therapeutics demonstrate a moderate effect on overall and
progression-free survival [88,89].
Platelets preferentially attach to tumor-associated vessels rather than normal vascula-
ture, amplifying the delivery of tumorigenic mediators to the TME [
90
]. Tumor cell-induced
platelet activation (TCIPA) leads to the translocation of P-selectin (also known as CD62P), a
cell adhesion molecule stored in the
α
-granules [
37
], to the platelet surface. The binding
of P-selectin to the P-selectin glycoprotein ligand (PSGL-1) on leukocytes governs leuko-
cyte rolling in activated endothelial cells [
91
] and the generation of platelet—cancer cell
complexes [
92
]. Adhesion molecules, including integrins, von Willebrand factor (vWF), fib-
rinogen, fibronectin, and coagulation factors, and several members of the a disintegrin and
metalloproteinase (ADAM) protein family accommodate the activation, tethering, rolling,
and firm adhesion of platelets to endothelial cells [
93
]. Activated platelets degranulate and
release various factors that affect angiogenesis. More than 30 components associated with
platelets that influence angiogenesis have been described [
94
]. Platelets generate angiostatic
factors such as endostatin, angiostatin, and thrombospondin-1 (TSP-1), and angiogenic fac-
tors including vascular endothelial growth factor (VEGF), angiopoietin-1, stromal-derived
factor 1 (SDF-1, also known as the chemokine (C-X-C motif) ligand [CXCL]12), sphingosine
1-phosphate (S1P), transforming growth factor-beta (TGF-
β
), interleukin (IL)-6, and platelet
factor 4 (PF4; also known as CXCL4) [
61
]. Platelet-derived growth factor (PDGF) supports
the function of cancer-associated fibroblasts (CAFs), vascular pericytes, and smooth muscle
cells in angiogenesis [
95
]. Platelets also support the recruitment of endothelial progenitor
cells (EPCs) [
96
]. Platelet integrin GPIIb/IIIa promotes endothelial cell proliferation and
function [
97
]. The activation of platelets and the release of their granular content, such
as angiopoietin-1 and serotonin, prevent intratumoral bleeding [
98
]. ATP released from
the
δ
-granules of platelets activates endothelial P2Y
2
receptors, causing the retraction of
endothelial cells and promoting the transendothelial migration of cancer cells (intravasation
and extravasation) and metastasis [
99
]. Platelet microparticles increase the expression of
matrix metalloproteinases (MMPs) on endothelial cells [
100
], assisting in the generation of
new vessels [61].
The co-localization of GPIIb (CD41), platelet endothelial cell adhesion molecule-1
(PECAM-1; also known as CD31), and VEGF in ovarian cancer tissues suggests the in-
volvement of platelets in angiogenesis and tumor growth [
101
]. An increased level of
VEGF can be considered a biomarker of ovarian cancer [
102
] and an indicator of advanced
disease, ascites formation, metastasis, and reduced survival [
103
,
104
]. Moreover, the levels
of PDGF-BB and VEGF were found to be positively correlated in the TME and ascites, and
the pharmacological inhibition of their receptors increased the efficacy of chemotherapy in
patients with ovarian cancer [
105
]. The co-localization of regulator of G-protein signaling 5
(RGS5), a signal transduction molecule upregulated in endothelial cells in the TME, with
Cancers 2023,15, 1282 6 of 37
PECAM-1 and PDGF receptor (PDGFR)-
β
, has been reported in various types of cancer,
including ovarian cancer [
106
]. The participation of activated platelets in angiogenesis is
displayed in Figure 2.
Cancers 2023, 15, x FOR PEER REVIEW 6 of 38
Figure 2. The pro-angiogenic role of platelets in cancer. The interaction between platelets, endothe-
lial cells, and pericytes supports the extravasation of immune cells, mesenchymal stem cells (MSCs),
endothelial precursor cells (EPCs), and tumor cells. Platelets also release proangiogenic factors that
promote new blood vessel formation and facilitate tumor growth. The newly formed cancer-associ-
ated blood vessels are branched, leaky, and less supported by pericytes. Insufficient oxygen and
nutrient supplies lead to hypoxic and necrotic areas in the tumor. Platelet-targeting strategies can
restrict neoangiogenesis.
Platelet GPIb-IX receptor complex, a receptor for vWF, and GPIIb/IIIa, the receptor
for fibrinogen, promote platelet aggregation [107] and adhesion to endothelial cells. Ad-
ditionally, platelet P-selectin and GPIIb assist in the adhesion of platelets to cancer cells.
Hence, platelets assist in the clinging of cancer cells to the endothelium and metastasis
[108,109]. Targeting platelet surface proteins might show therapeutic benefits in cancer.
Antiplatelet agent-directed platelet inhibition diminishes the proliferative capability of
ovarian cancer cells [31]. In addition, focal adhesion kinase (FAK) promotes platelet infil-
tration into the TME, and targeting FAK suppresses ovarian tumor growth. Dual therapy
using antiplatelet agents and antiangiogenic drugs prevents rebound tumor growth after
discontinuing antiangiogenic agents [20].
Figure 2.
The pro-angiogenic role of platelets in cancer. The interaction between platelets, endothelial
cells, and pericytes supports the extravasation of immune cells, mesenchymal stem cells (MSCs),
endothelial precursor cells (EPCs), and tumor cells. Platelets also release proangiogenic factors
that promote new blood vessel formation and facilitate tumor growth. The newly formed cancer-
associated blood vessels are branched, leaky, and less supported by pericytes. Insufficient oxygen
and nutrient supplies lead to hypoxic and necrotic areas in the tumor. Platelet-targeting strategies
can restrict neoangiogenesis.
Platelet GPIb-IX receptor complex, a receptor for vWF, and GPIIb/IIIa, the receptor for
fibrinogen, promote platelet aggregation [
107
] and adhesion to endothelial cells. Addition-
ally, platelet P-selectin and GPIIb assist in the adhesion of platelets to cancer cells. Hence,
platelets assist in the clinging of cancer cells to the endothelium and metastasis [
108
,
109
].
Targeting platelet surface proteins might show therapeutic benefits in cancer. Antiplatelet
agent-directed platelet inhibition diminishes the proliferative capability of ovarian cancer
Cancers 2023,15, 1282 7 of 37
cells [
31
]. In addition, focal adhesion kinase (FAK) promotes platelet infiltration into the
TME, and targeting FAK suppresses ovarian tumor growth. Dual therapy using antiplatelet
agents and antiangiogenic drugs prevents rebound tumor growth after discontinuing
antiangiogenic agents [20].
2.1.2. In Lymphangiogenesis
Lymphangiogenesis is the formation of new lymphatic vessels and occurs during
embryonic development and in pathological conditions involving inflammation and tumor
metastasis [
110
]. Platelets are essential for the proper partitioning of blood and lym-
phatic vessels during development, mainly by coordinating endothelial cells’ expansion,
relocation, and tube formation. This phenomenon occurs following the engagement of
platelet CLEC-2 with its ligand podoplanin on lymphatic endothelial cells [
111
]. Although
platelets do not necessarily contribute to the maintenance of the separation of the two
circulatory systems post-development in normal conditions, in certain situations, including
wound healing or tumor growth, platelets again participate in lymphangiogenesis [
112
].
Platelets stimulate lymphangiogenesis by secreting proangiogenic factors such as VEGF,
angiopoietin-1, PDGF, and insulin-like growth factor 1 (IGF-1) [
61
,
113
], and through the
interaction of CLEC-2 and podoplanin [111].
In patients with ovarian cancer, the upregulation of lymphangiogenic markers is
associated with more aggressive disease and shorter overall survival [
114
]. Podoplanin
overexpression in the malignant stroma of ovarian cancer patients predicts lymphatic
spread and poor clinical outcomes [
115
]. Blocking podoplanin—CLEC-2 contact between
ovarian cancer cells and platelets forestalls lymph vessel proliferation [
116
]. Likewise,
VEGF and PDGF released by platelets promote lymph vessel generation in epithelial
ovarian cancer [
117
]. Treatment with antiangiogenic agents might attenuate lymphangio-
genesis in ovarian cancer [
118
]. Notably, inhibiting the TGF-
β
signaling cascade prevents
lymphangiogenesis and subsequent VEGF-mediated ascites generation in ovarian cancer
patients [119].
2.2. Interactions with Pericytes
Pericytes are perivascular cells embedded in the basement membrane surrounding
the microvasculature [
120
]. Pericytes might prevent the intravasation of cancer cells and
metastasis [
121
]; however, they might also facilitate micrometastasis by supporting the
formation of tumor vasculature [
122
]. Tumor vessels have atypical coverage of pericytes,
whose contact with endothelial cells is disrupted [
122
]. The tumor vasculature has an excess
of pericytes that loosely interact with endothelial cells, deteriorating the integrity of the
vessels and resulting in hemorrhage [
123
]. In ovarian cancer, abnormal pericyte numbers
and expression signatures are associated with tumor growth, aggressive metastasis, and
poor clinical outcomes [124].
Podoplanin, which is highly expressed on pericytes, mediates platelet binding to
pericytes via CLEC-2 [
125
]. Furthermore, platelet-derived TGF-
β
, angiopoietin, and PDGF
also stimulate pericyte differentiation, colonization, and their interaction with endothelial
cells [
64
]. TGF-
β
strongly influences the proliferation of pericytes [
126
] and their associa-
tion with endothelial cells through the TGF-
β
—matrix protein axis [
127
]. The activation
of the TGF-
β
signaling cascade impacts the density and lumen size of tumor microves-
sels [
127
]. Angiopoietin overexpression is linked with pericyte impairment and tumor
vessel instability [
128
]. Blocking TGF-
β
or angiopoietin signaling inhibits tumor growth
and neovascularization [
127
,
129
]. PDGF released from platelets and other cells [
130
] is
essential for pericytes recruitment and function during tumor neoangiogenesis. Preventing
PDGF isoforms from binding to their receptors and hindering angiogenesis with antiangio-
genic agents such as bevacizumab may interfere with the incorporation of pericytes into
new blood vessels [131].
Cancers 2023,15, 1282 8 of 37
TSP-1 is a matricellular glycoprotein with antiangiogenic properties that counters the
proliferative effects of growth factors on endothelial cells [
132
,
133
]. TSP-1 is released from
the granules of activated platelets, prompting platelet aggregation and tethering [
56
]. In
ovarian cancer, binding of the TSP-1 three type 1 repeats (3TSR) domain of TSP-1 to GPIV
(CD36) normalizes the tumor vasculature and exhibits antitumor function [
134
]. Treating
patients with 3TSR in combination with chemotherapeutics [
135
] or oncolytic viruses [
136
]
can improve the efficacy of anticancer therapies. 3TSR alone, or fused with the Fc region
of human IgG1 for improved stability, increases the number of pericyte-covered blood
vessels, reduces the proliferative capacity of endothelial cells, and contributes to vascular
normalization [134].
Platelets release IL-6 [
73
] and also trigger IL-6 secretion from tumor cells by releasing
several factors, such as lysophosphatidic acid (LPA) [
137
]. The protumorigenic cytokine
IL-6 is significantly elevated in ovarian cancer patients with confirmed thrombocytosis [
27
].
High IL-6 levels promote neoangiogenesis with abnormal pericyte coating. Anti-VEGF and
anti-IL-6 agents reduce vessel sprouting and leakiness of the vasculature by reinstating the
pericyte lining [
138
]. Combining chemotherapeutic agents and pazopanib, a multitargeted
tyrosine kinase inhibitor, can help restore pericyte coverage and restrict tumor microvessel
density [
139
] in patients with ovarian cancer. The antiplatelet action of pazopanib [
140
]
might further inhibit tumor growth and angiogenesis [20].
2.3. Interactions with Cancer-Associated Fibroblasts
Fibroblasts are a heterogeneous population of connective tissue cells with a presum-
ably mesenchymal origin [
141
]. Fibroblasts can differentiate into particular mesenchymal
cell types, including osteoblasts, adipocytes, and chondrocytes [
142
]. In the TME, CAFs
promote disease progression by releasing various molecules; rearranging the ECM to facili-
tate cancer cell motility, invasion, and EMT; and stimulating angiogenesis, tumor growth,
and metastasis. CAFs modulate the function of immune cells and the metabolism of cancer
cells to promote tumor survival [
143
]. CAFs also release extracellular vesicles that support
cancer progression [
144
] and chemoresistance [
145
]. Although fibroblasts play a role in
tumorigenesis, they may also restrict tumor development by activating the tumoricidal
immune response or consolidating the ECM to prevent tumor dissemination [146].
Extravasated platelets promote EMT by releasing mediators such as TGF-
β
, SDF-1,
and PDGF. The same mediators also induce the differentiation, migration, and proliferation
of CAFs [
147
,
148
]. PDGF and TGF-
β
induce mesenchymal stem cell (MSC) differentiation
into CAFs [
149
]. Integrin
α
11 is a CAF marker, and its expression is related to myofibrob-
last differentiation and ECM alteration. CAFs expressing integrin
α
11 and PDGFR-
β
are
associated with poor clinical outcomes in ovarian cancers and other malignancies [
150
].
Platelet-originated CLEC-2 induces the migration and proliferation of CAFs in the TME [
15
].
The binding of platelet-derived CLEC-2 to podoplanin on CAFs and cancer cells promote tu-
mor growth and venous thrombosis in patients with ovarian cancer [
125
,
151
]. LPA derived
from ovarian cancer cells promotes the differentiation of fibroblasts into CAFs through
a hypoxia-inducible factor 1 alpha (HIF-1
α
)-dependent mechanism [
152
]. LPA activates
platelets [
153
], which, in turn, release LPA [
137
]. Moreover, LPA promotes the secretion of
VEGF and SDF-1 from MSCs, further supporting ovarian cancer progression [
154
]. Under
oxidative stress, platelets release their mitochondria, which are picked up by MSCs [
24
].
Mitochondria originating in platelets and engulfed by MSCs promote fatty acid synthesis
and ATP production and stimulate the release of angiogenic components, such as VEGF
and hepatocyte growth factor (HGF), from MSCs [24].
CAFs originating from MSCs release platelet-activating factor (PAF), promoting
platelet activation and aggregation [
155
], which further supports ovarian cancer progres-
sion and induces ovarian cancer development through the PAF/PAF receptor signaling
pathway [
34
]. Exosomes from ovarian cancer cells induce the generation of CAFs from
MSCs in the tumor stroma [
156
]. CAF-released IL-6 causes EMT in ovarian cancer cells, tu-
mor growth, and ECM reorganization, mainly by mediating STAT3 phosphorylation [
157
].
Cancers 2023,15, 1282 9 of 37
The increased concentrations of IL-6 in the stroma or ascites can activate platelet function
and aggregation and lead to thrombosis [158].
Most patients with metastatic ovarian cancer have peritoneal dissemination, which
indicates a poor prognosis. It starts with the emigration of cancer cells into the peritoneal
fluid, forming floating masses that attach to peritoneal mesothelial cells throughout the
peritoneal cavity [
159
]. Alternatively, the cancer cells can initiate an inflammatory reaction
in the peritoneal stroma, promoting the generation of a fibrin mesh that can be used for
adhesion to the peritoneal surface. Fibrin mesh can also potentiate the colonization of
fibroblasts and endothelial cells. Fibroblasts that differentiate into CAFs promote ovarian
cancer invasion through the upregulation of several markers such as alpha-smooth muscle
actin (
α
-MA), PDGFR, and podoplanin [
151
]. A subset of CAFs originating from mesothe-
lial cells through mesothelial-to-mesenchymal transition (MMT) contribute to peritoneal
metastasis [
160
]. Ovarian cancer cells that have been relocated to the peritoneal cavity
promote ascites, and the ascitic fluid contains various cytokines and growth factors [
161
].
TGF-
β
derived from ascites and activated platelets is one of the main stimulating factors
for MMT [
160
,
162
]. In addition, tissue factor (TF), present in high amounts in ascites,
cancer cell masses, and cancer cell-derived microparticles, induces thrombin generation,
which activates platelets and produces fibrin [
163
]. Activated platelets further increase the
expression of TF, prompting ovarian cancer migration [
164
]. The activation of mesothelial
cells by TGF-
β
released from platelets is partially responsible for ECM remodeling during
metastasis [165].
Like fibroblasts, mesothelial cells and adipocytes in the omentum and peritoneum
can be prompted by cancer cells to differentiate into CAFs [
166
]. Periostin is a secretory
protein that is overexpressed by stromal fibroblasts in multiple cancers, including ovarian
cancer, and its overexpression is associated with poor clinical outcomes in patients with
epithelial ovarian cancer. TGF-
β
modulates periostin expression and promotes ovarian
cancer growth and chemotherapy resistance [
167
]. Similarly, the aberrant expression and
release of connective tissue growth factor (CTGF), a stromal factor, induces the colonization
and peritoneal adhesion of ovarian cancer cells [
168
]. Platelets store a large quantity of
CTGF [
63
], suggesting that platelet activation and the release of CTGF may participate in
ovarian cancer seeding in the peritoneum.
Inhibitors of the TGF-
β
signaling pathway can diminish CAFs’ function in ovarian
cancer [
169
]. Anti-VEGF therapy can inhibit ovarian cancer progression, metastasis, and
malignant ascites formation promoted by the release of VEGF from CAFs, along with vari-
ous other cell types [
170
]; however, synchronous anti-PDGF treatment might be necessary
to target CAFs resistant to VEGF-neutralizing agents [
171
]. It has been reported that aspirin
therapy suppresses chemotherapy-induced CAF formation in colorectal cancer [
172
] and
the impact of CAFs on ovarian cancer.
3. Interplay of Platelets with the Tumor Immune Microenvironment
3.1. Interplay with Tumor-Associated Neutrophils
Most inflammatory cells in solid tumors are tumor-associated neutrophils (TANs)
that promote or inhibit tumor growth and angiogenesis, depending on various circum-
stances. In the initial stages of tumor growth, TANs tend to exert tumoricidal functions
(N1 neutrophils), whereas in later stages of tumor growth, they become protumorigenic
(N2 neutrophils). Permissive N2 neutrophils enhance the proliferation and metastasis
of malignant cells by releasing growth factors (e.g., TGF-
β
and VEGF), MMPs, and re-
active oxygen species (ROS) [
173
]. In addition, they can suppress antitumor immune
responses [
174
]. Neutrophils assist cancer cells in escaping the antitumor immune response
through the overexpression of immune checkpoint molecules, including programmed
death ligand 1 (PD-L1) [
175
], glucocorticoid-induced tumor necrosis factor receptor-related
protein-ligand (GITRL) [
176
], and V-domain Ig suppressor of T cell activation (VISTA) [
177
].
In patients with ovarian cancer, elevated neutrophil counts, neutrophil-lymphocyte ratios
(NLRs), and platelet counts indicate a poor prognosis and shorter overall survival [
178
,
179
].
Cancers 2023,15, 1282 10 of 37
Adhesion molecules on the surface of neutrophils include L-selectin (CD62L), PSGL-
1, macrophage-1 antigen (Mac-1; also known as CD11b/CD18), and leukocyte function-
associated antigen 1 (LFA-1; also known as CD11a/CD18), which are essential for the rolling
of neutrophils on the endothelium, as well as their firm adhesion and transendothelial
migration [
180
]. Inflammation promotes the expression of surface adhesion molecules on
platelets and endothelial cells. Platelets help recruit neutrophils to the tissues via direct
or indirect interaction. Neutrophils and platelets interact mainly through the binding of
P-selectin to PSGL-1 [
181
]. The binding of GPIb on platelets to Mac-1 [
182
] on neutrophils,
and that of the intercellular adhesion molecule 2 (ICAM-2) on endothelial cells to LFA-
1 [
183
] on neutrophils, provide additional adhesive interactions under flow conditions. At
baseline, Mac-1 has low expression on neutrophils in the peripheral blood; however, it is
significantly upregulated in inflammation [
184
]. Neutrophils in the peripheral blood of
ovarian cancer patients have higher levels of Mac-1.
In ovarian cancer patients, neutrophils overexpress Mac-1, which improves the ad-
hesion of neutrophils to endothelial cells and modifies the endothelium for improved
cancer cell migration [
185
]. Of note, the PSGL-1–L-selectin interaction is also important
for neutrophil adherence to endothelium [
186
]. The interaction between cancer cells and
neutrophils through Mac-1 and L-selectin can promote cancer cell migration through blood
vessels [
184
,
187
]. Other interactions, including those between CD40–CD40 ligands (CD40L,
also known as CD154) [
188
] and between platelet PSGL-1 and neutrophil L-selectin [
189
],
might also support neutrophil interactions in the TME.
Platelets and neutrophils communicate indirectly through intermediary molecules.
For instance, platelet GPIIb/IIIa and neutrophil Mac-1 both bind to fibrinogen [
190
]. In
addition, platelets’ secretion of chemokines and cytokines, such as PF4 [
70
], CXCL7 (also
known as neutrophil-activating peptide 2 (NAP-2)) [
71
], and TGF-
β
[
191
], following their
interaction with cancer cells reinforces neutrophils’ attachment to the endothelium and
their transendothelial migration. In ovarian cancer, IL-8 (CXCL8) recruits neutrophils to
the TME [
192
]. IL-8 released from the activated platelets [
193
] or platelet-activated cancer
cells [
194
] can help increase the number of neutrophils in the TME. IL-8 receptor CXCR2
is upregulated in the neutrophils of patients with ovarian cancer compared to healthy
individuals [
195
]. Moreover, the elevated concentration of IL-8 in peritoneal lavage in
ovarian cancer is associated with shorter overall survival [
196
]. Cytokines and chemokines,
such as granulocyte colony-stimulating factor (G-CSF), granulocyte-monocyte colony-
stimulating factor (GM-CSF), CXCL1, CXCL2 (also known as migration inhibitory protein
2 alpha (MIP-2
α
)), CXCL5, and CCL3 (chemokine (C-C motif) ligand 3) (MIP-1
α
) recruit
neutrophils to the TME [
197
]. TGF-
β
and G-CSF are important for neutrophil differentiation
into the N2 phenotype [
173
]; thus, platelet-derived TGF-
β
[
198
] and GM-CSF [
75
] create an
immunotolerant environment for ovarian cancer.
Neutrophil extracellular trap (NET) formation (NETosis) is a defense mechanism
against inflammation [199]. The activated neutrophils secrete antimicrobial proteases and
release chromatins and cytosolic and granular proteins, such as myeloperoxidase and
neutrophil elastase, to trap and kill pathogens [
200
]. The presence of lipopolysaccharide
(LPS) activates platelets in endotoxemia, and hyperactive platelets bind to adherent neu-
trophils via Toll-like receptor (TLR) 4, causing neutrophil activation and NETosis. LPS is
not adept at directly stimulating NET formation [
40
]. In return, NETs trigger thrombosis
because histone proteins and DNA released from neutrophils, in addition to NET-linked
cathepsin G, activate platelets [
201
]. NETs bind to factor XII or P-selectin to initiate blood
coagulation [
202
]. Neutrophil and the NET-dependent release of neutrophil elastase cleaves
E-cadherin and enhances EMT in ovarian cancer cells [
203
]. Cathepsin G and neutrophil
elastase activate platelets via GPIIb/IIIa and induce platelet aggregation in the presence of
exogenous fibrinogen [204].
Mitochondrial DNA (mtDNA) and formylated peptides are among the mitochondrial
damage-associated molecular patterns discharged from various cells. These molecules
stimulate neutrophil function via the adhesion of TLR9 and formylated peptide receptors
Cancers 2023,15, 1282 11 of 37
and promote NETosis [
205
]. NETs support cancer-related thrombosis through factors such
as extracellular chromatin and tissue factor [
206
]. NETs also play a role in ovarian cancer
cell invasion and premetastatic niche formation in the omentum [
207
]. The amount of
platelet-derived mtDNA and the number of microparticles in ascites correlate with poor
clinical outcomes in ovarian cancer patients [
208
]. The fibrin deposits in the TME further
assist in ovarian cancer invasion by providing an adhesion surface for platelets, promot-
ing their aggregation, and stimulating monocyte differentiation into tumor-associated
macrophage (TAM)-like cells [
209
]. The pharmacological suppression of NETosis-related
genes, including the protein arginine deiminase 4 (PAD4) gene, which encodes a histone
protein-citrullinating enzyme, can delay ovarian cancer cell invasion of the peritoneal
cavity [207].
3.2. Interplay with Tumor-Associated Macrophages
Mononuclear cells extravasate into tumor tissues and differentiate into macrophages.
Macrophages polarize into two distinct subtypes that have pro- or anti-inflammatory char-
acteristics. Classically activated macrophages, also termed M1 macrophages, are stimulated
by molecules such as LPS, interferon-gamma (IFN-
γ
), tumor necrosis factor-alpha (TNF-
α
),
and GM-CSF, as well as by target pathogens and tumor cells. In contrast, alternatively
activated macrophages (M2 macrophages) are induced by molecules such as TGF-
β
, IL-
10, and macrophage colony-stimulating factor (M-CSF), and display protumorigenic and
immunomodulatory features [210].
In the ovarian cancer TME, a population of TAMs from both the M1 and M2 pheno-
types is observed [
211
]. As cancer progresses, TAMs polarize into M2 macrophages via
the upregulation of multiple markers, including CD163, CD206, PD-L1, and arginase 1
(ARG1) [
210
]. epithelial ovarian cancers support M2 polarization by releasing mediators
such as M-CSF [
212
]. The number of TAMs escalates as the tumor spreads, so much so
that half of the cells in the peritoneal TME and ascites can be TAMs with tolerogenic
functions [
213
,
214
]. M2 macrophages in ascites induce the transcoelomic metastasis of
ovarian cancer cells. M2 macrophages secrete multiple mediators that induce ovarian
cancer spheroid formation, encourage attachment to the metastasis site, and escape from
the cytolytic immune response [
215
,
216
]. An elevated M2/M1 macrophage ratio correlates
with reduced overall survival in ovarian cancer patients [217].
Platelet—monocyte aggregates are more potent markers for platelet activation than the
expression of P-selectin [
218
]. The activated macrophages release cytokines such as TNF-
α
,
IL-1
β
, IL-6, and IL-8 [
219
]. The interaction between monocytes and platelets enhances the
procoagulant effect of macrophages and ROS generation. Activated platelets adhere to
monocytes via P-selectin and promote monocyte function and ROS release [
220
]. A high
concentration of ROS differentiates TAMs to the M2 phenotype, and this effect is annihilated
when macrophages are polarized to the M1-like phenotype, rather than M2 [221].
The upregulation of platelet-derived TGF-
β
, the subsequent overexpression of cy-
clooxygenase 2 (COX-2), and the generation of prostaglandin E2 (PGE2) in monocytes
modulate CD16 levels. The activation of CD16 on monocytes induces M2 polarization [
222
].
COX-2-positive TAMs increase cancer cell proliferation and metastasis because the PGE2
generated by COX-2 activates the protein kinase C/A and TGF-
β
signaling pathways [
223
].
COX-2 expression is elevated in macrophages that infiltrate epithelial ovarian cancer tu-
mors [
224
], and the resulting increased levels of PGE2 support tolerogenic M2 polarization
and ovarian cancer cell chemoresistance [225].
The platelet-directed differentiation of monocytes into the immunogenic phenotype via
IL-1
β
and IL-8 potentiates NF-
κ
B signaling and supports thrombosis [
226
]. Macrophages
with high levels of podoplanin expression migrate toward vascular endothelial cells, bind to
CLEC-2 on the surface of platelets, activate platelets [
38
,
227
], and promote thrombosis [
38
,
227
].
Additionally, podoplanin is upregulated in ovarian cancer cells, and extracellular vesicles
released from these cells increase the likelihood of thrombosis [125].
Cancers 2023,15, 1282 12 of 37
Platelet microparticles regulate the function of macrophages recruited to the inflam-
mation site by chemotactic components such as PF4, TGF-
β
, and PDGF-
β
[
23
]. These
microparticles can drive macrophages toward the protumorigenic phenotype by suppress-
ing TNF-
α
, CCL4, and M-CSF production and increasing their phagocytic efficacy [
228
].
Conversely, platelet microparticles may direct macrophage activation, which can inhibit
tumor growth through cytokines such as TNF-related apoptosis-inducing ligand (TRAIL),
CCL2 (also known as monocyte chemoattractant protein 1 (MCP-1)) and IL-8 [229].
Cancer cells and activated monocytes can release extracellular vesicles containing
TF that contribute to venous thromboembolism [
163
,
230
]. Moreover, the coincubation of
platelets with extracellular vesicles upregulates the TF activity of monocytes [
231
]. Cancer
cell-derived extracellular vesicles also trigger platelet activation and cause the release
of procoagulant platelet microparticles [
232
]. The extracellular vesicles of platelets with
phosphatidylserine attach to monocytes and monocyte extracellular vesicles and promote
their procoagulant effects [
233
,
234
]. TAMs express the coagulation factors II, V, VII, and X,
stimulating thrombin formation and platelet activation [
235
]. The TF-dependent throm-
bin formation promotes several cellular interactions in the TME [
236
]. For instance, the
TF-related activation of protease-activated receptor (PAR)-2 and integrin ligation enhance
tumor growth and angiogenesis [
237
]. Moreover, TF supports the intravasation and in-
travascular survival of cancer cells, and the formation of metastatic niches, to provide an
environment for the interaction of monocytes and macrophages with tumor-associated
microthrombi [
238
]. The thrombin receptors PAR1, PAR4, and GPIb
α
on the surface of
platelets can facilitate metastasis [
42
,
43
]. By disrupting the TF-accompanying pathways,
PAR inhibitors might diminish the proliferation and metastasis of ovarian cancer cells [
237
].
The network between platelets, cancer cells, endothelial cells, and smooth muscle
cells attracts monocytes into the early metastatic niches through chemokines such as
CCL2 and CCL5 (known as regulated on activation, normal T cell expressed and secreted
(RANTES)) [
238
]. The CCL5-induced attraction of monocytes to metastatic cancer cells
indicates that CCL5 is indirectly critical to the organization of metastasis rather than
directly acting on cancer cells [
239
]. Similarly, platelets’ secretion of SDF-1 is decisive in the
recruitment of macrophages to tumors and in the migration of CXCR4
+
cancer cells [
240
]. In
addition, SDF-1 mediates the migration of TAMs into the hypoxic TME and the polarization
of monocytes toward the tolerogenic phenotype [
241
]. The blockade of the SDF-1—CXCR4
interaction, combined with anti-programmed death 1 (PD-1)-based immunotherapy, results
in a higher M1/M2 ratio, which is associated with better clinical outcomes in patients with
ovarian cancer [242].
PAFR activation stimulates macrophages to differentiate toward the M2 phenotype,
whereas the absence of PAFR drives tumor-infiltrating macrophages to demonstrate an-
titumor properties [
243
]. PAFR ligands support tumor development by reprogramming
the TAM phenotype and increasing the frequency of M2 macrophages rather than M1
macrophages. This specific polarization of macrophages through apoptotic cells results in a
tolerogenic environment that supports tumor growth [
243
]. Thus, PAFR antagonists may
be a therapeutic option for patients with ovarian cancer [244].
Ovarian cancer cell-derived M-CSF stimulates the polarization of macrophages to
the M2 subtype, which supports EMT and the peritoneal metastasis of ovarian cancer
cells [
216
]. M-CSF can also trigger platelet aggregation [
245
] and provoke thrombosis in
cancer patients. M2 macrophages release high concentrations of epidermal growth factor
(EGF) and induce angiogenesis and the invasion and metastasis of cancer cells through the
activation of an EGF receptor (EGFR) [
246
]. EGF stimulation facilitates the activation of
platelets and the release of IL-1
β
[
72
]. Moreover, EGF-triggered ovarian cancer cells secrete
PAF and contribute to the hyper-responsiveness of platelets. The blockade of EGFR might
abolish EGF’s promotion of ovarian cancer progression [247].
Cancers 2023,15, 1282 13 of 37
In the ovarian cancer TME, LPA is produced from phospholipids by TAM- and platelet-
derived phospholipase A2 (PLA2) and autotaxin [
248
,
249
]. LPA promotes ovarian cancer
growth and metastasis through diverse networks [
250
,
251
]. It also induces platelet activa-
tion [
252
]; thus, the pharmacological inhibition of LPA and the application of antiplatelet
agents might interfere with ovarian cancer development [249,253].
3.3. Interplay with T Cells
3.3.1. CD4+Helper T Cells
CD4
+
helper T (Th) cells are major contributors to adaptive immunity. Their antitu-
mor effect can be exerted directly by targeting tumor cells or indirectly by triggering the
expansion of cytotoxic CD8
+
T cells [
254
]. An increased percentage of tumor-infiltrating
lymphocytes (TILs) is associated with prolonged survival in patients with ovarian can-
cer [255,256].
Platelets may affect CD4
+
T cells directly through physical interaction [
257
] or indi-
rectly through the secretion of soluble mediators [
258
], extracellular vesicles, and mito-
chondria [
259
,
260
]. Activated platelets form complexes with CD4
+
T cells and mediate the
adhesion of lymphocytes to the endothelium under flow conditions, and to the ECM [
261
].
T cells cannot bind to fibronectin efficiently without the participation of platelets [
262
].
CD40–CD40L, P-selectin–PSGL-1, and ICAM-2–LFA-1 interactions are partly responsible
for the construction of platelet–CD4
+
T cell complexes and the rolling and firm adhesion of
T cells under flow [
262
]. CD4
+
T cells’ CD40L binds to platelets’ CD40 [
58
] and stimulates
CCL5 secretion [
263
]. The soluble CD40L (sCD40L) binds to CD40 on CD4
+
T cells, causing
CD4
+
T cells to undergo apoptosis following the suppressed expression of IL-2, Bcl-2, and
Bcl-xL [
264
]. Platelets, rather than T cells, are the predominant source of sCD40L in can-
cer [
59
]. Antiplatelet reagents might restore the antitumor immune response by reducing
sCD40L levels and preventing CD4+T cell apoptosis [59].
The interaction between platelet P-selectin and T cell PSGL-1 recruits T cells to the
activated endothelium at the site of inflammation or injury. Memory CD4
+
T cells form com-
plexes with platelets, which may exacerbate thrombo-inflammation [
265
]. In conjunction
with its role in mediating cell adhesion [
266
], the upregulation of PSGL-1 on effector T cells
may alter their function. PSGL-1 engagement can induce T cell exhaustion by inhibiting
ERK/Akt signaling and IL-2 secretion, and increasing PD-1 expression. In this context,
PSGL-1 ligation can reduce the antitumor effect of CD4+and CD8+T cells [267].
Platelets and CD4
+
T cells form complexes that can trigger immune-related adverse
events in cancer patients treated with chemotherapeutic agents or immune checkpoint
inhibitors. The elevated levels of PD-L1 in platelets can inhibit CD4
+
and CD8
+
T cells [
52
].
Moreover, platelet PD-L1 may increase the overall expression of PD-L1 in tumors after
platelet adhesion to cancer cells [
268
], suggesting that adding an antiplatelet agent can
improve the efficacy of anti-PD-L1 immunotherapy [
269
]. On the other hand, other studies
show that platelets induce the expression of PD-L1 in cancer cells [
14
,
270
], and reduc-
ing platelet counts via an antiplatelet agent treatment can minimize the effectiveness of
immunotherapy [
14
]. In addition, platelet-derived TGF-
β
reduces the efficacy of T cell
recruitment via bispecific antibody (BsAb)-based immunotherapy [
271
] in ovarian can-
cer [
272
]. Immune checkpoint inhibitors administered with VEGF inhibitors have been
shown to improve the efficacy of BsAbs in ovarian cancer [273].
3.3.2. Regulatory T Cells
Regulatory T cells (Tregs), which constitute 5% to 7% of CD4
+
T cells, induce immune
tolerance by cytokines such as TGF-
β
and IL-10, and receptors such as cytotoxic T lym-
phocyte antigen 4 (CTLA-4), CD39, and CD73 [
274
]. Consequently, Tregs exert protumor
effects by providing a permissive environment for tumor growth. In the early stages of
ovarian cancer, the Th17-related immune response is more dominant, whereas a persistent
shift toward Tregs is witnessed in the later stages [
275
]. Tregs are recruited and activated
by the hypoxic conditions in ovarian cancer TME [
276
] and are abundant in ascites. Their
Cancers 2023,15, 1282 14 of 37
presence has been linked to unfavorable clinical outcomes [
277
,
278
]. Meanwhile, high
CD8
+
T cell/Treg and CD4
+
T cell/Treg ratios are indicators of a better prognosis [
279
].
Interestingly, a positive correlation between increased numbers of CD8
+
T cells and Tregs
in the TME has been observed in multiple patients with ovarian cancer, but this immune
profile has not influenced patients’ overall survival [280].
Treg-derived PSGL-1 is pivotal in regulating the immune response, since this glyco-
protein plays a part in hampering T cell–DC conjugation and subsequent T cell expansion
and activation [
281
]. In contrast, P-selectin is upregulated in CD4
+
non-Tregs and is critical
for Treg differentiation and activity because it functions in signaling cascades involving Syk
kinases and CCL5 [
263
,
282
,
283
]. P-selectin–PSGL-1 binding might potentiate the influence
of costimulatory components such as CD40L [
284
]. The engagement of CD40 with CD40L
is important for the development and survival of Tregs [
285
], and CD40–CD40L activates
Tregs and platelets in a positive feedback loop [
263
]. CD40 or CD40L deficiency reduces
the number of Tregs without affecting the total number of T cells [286].
Moreover, P-selectin plays an important role in recruiting Tregs to tumors and repress-
ing effector CD8
+
T cells [
16
]. IL-4 stimulates endothelial cells’ expression of P-selectin
and the recruitment of activated platelets expressing P-selectins to the TME, which causes
positive feedback in immunosuppression [287].
Platelet releasate stimulates Treg activity, which, in the short-term, increases and
subsequently inhibits Th1/Th17 responses. The mediators secreted by platelets (e.g., PF4
and TGF-
β
) and by platelet-activated Tregs downregulate Th1 polarization [
288
]. Platelet
deficiency leads to reduced Treg activation through several pathways related to TLR4
and TNF-
α
. The adherence of platelet-derived TNF-
α
to TNFR2 on Tregs activates these
cells [
74
]. In the tumor-associated ascites of patients with ovarian cancer, IL-6 stimulates
Tregs’ expression of TNFR2 [
289
]. IL-6 generated by platelets [
73
] supports the function of
Tregs in ascites, creating an immunosuppressive environment. TNFR antagonists might
restrict Treg expansion in the TME and hinder ovarian cancer cell proliferation [290].
TGF-
β
is required to generate Tregs, and platelets, as an important source of TGF-
β
,
are known to promote Treg activity. Platelet TGF-
β
modulates the expression of granzyme
B, IFN-
γ
, and IL-2 to promote Treg development [
291
]. Thus, platelet hyperactivity in
malignancies may create a favorable environment for Treg expansion [
292
]. Moreover,
platelet-derived growth factors, including IGF-1 [
62
] and PDGF [
293
], also foster Treg
proliferation. The inhibition of the TGF-
β
-directed network can mitigate an increased
number of Tregs and support cytotoxic T cell expansion while reducing tumor and ascites
volumes in patients with ovarian cancer [
294
]. Similarly, the blockade of IGF [
295
] and
PDGF [296] receptors might restore immunosurveillance for malignant ovarian cells.
PF4 suppresses the overall expansion of CD4
+
T cells and Th1 polarization while
promoting the proliferation of Tregs. The binding of PF4 to its receptor, CXCR3, escalates
mitochondrial transcription factor A (TFAM) expression, mitochondrial biogenesis, and
ATP and ROS generation. The increased concentrations of ATP and ROS increase T-bet
and Foxp3 expression, whichmodulates Th1 and Treg differentiation, respectively [
297
].
Moreover, thromboxane A2 (TxA2) secreted from platelets inhibits the activity of CD4
+
T
cells [
298
]. PF4 also heterodimerizes with other platelet-derived chemokines, including
CCL2, CCL5, CXCL7, SDF-1, and TxA2. The chemokine pairing aggravates chronic inflam-
mation in cancer; as a result, preventing chemokine pairing might provide a therapeutic
advantage [299,300].
CXCR4 and SDF-1 are upregulated in ovarian cancer cells [
301
]. Platelets express
CXCR4 and SDF-1, and activated platelets generate SDF-1 [
302
,
303
]. Hypoxia increases
the expression of CXCR4 on Tregs and supports immunosuppression [
304
]. Blocking the
binding of SDF-1 to its receptor, CXCR4, increases the CD8
+
T cell/Treg ratio and reduces
the intraperitoneal metastasis of ovarian cancer cells [
305
]. Tregs with high immune
checkpoint expression support the immunoregulatory function of these cells in ovarian
cancer [
306
]. The Treg
hi
phenotype is distinguished by elevated expression levels of
forkhead box P3 (Foxp3), PD-1, 4-1BB, inducible T cell co-stimulator (ICOS), and CD25.
Cancers 2023,15, 1282 15 of 37
The infiltration of these Tregs into tumors is associated with intercepted antitumor immune
response [
306
]. Ovarian tumors display the Treg
hi
phenotype [
278
,
306
], which may explain
ovarian cancer patients’ unsatisfactory responses to immune checkpoint inhibitors. In
many cancers, PD-L1-containing platelet microparticles adhere to PD-1 on Tregs and
induce immune tolerance [
307
]. The simultaneous inhibition of the CXCR4—SDF-1 and
PD-1—PD-L1 pathways and the depletion of effector Tregs may enhance the antitumor
immune response and reduce mortality in ovarian cancer patients [242,308].
Platelet microparticles prevent Treg differentiation to proinflammatory T cells that gen-
erate IL-17 and IFN-
γ
[
309
]. This mechanism occurs when the P-selectin on microparticles
adheres to PSGL-1 on CCR6
+
HLA-DR
+
memory-like Tregs, which are the progenitors of
Th17-like cells and stabilizers of the inflammatory state [
309
,
310
]. The reduction in IFN-
γ
release after PF4–CXCR3 engagement [311] and in communication among Tregs upon the
binding of platelet microparticles [312] support this mechanism.
3.3.3. CD8+T Cells
CD8
+
T cells, also termed cytotoxic or cytolytic T lymphocytes (CTLs), play a cardinal
role in protecting the host from infections. Unlike acute infections, chronic inflammation
and cancer may involve CD8
+
T cells. CD8
+
T cells can become exhausted and develop
reduced functionality. Exhausted CD8
+
T cells are distinguished by their persistent over-
expression of immune checkpoints [
313
]. Functional cytotoxic TILs improve survival in
ovarian cancer patients [
314
], and the recruitment of CD8
+
T cells into the peritoneal cavity
is associated with better prognostic scores in these patients [315].
Platelets interact with CD8
+
T cells and decrease their cytolytic impact on cancer cells.
The abundance and size of platelet—T cell aggregates in cancer patients are increased
compared to healthy individuals. High levels of these aggregates are regarded as cancer
biomarkers [
316
]. Platelets in platelet—T cell aggregates express high levels of P-selectin,
and platelets may be more prone to adhering to PSGL-1 on CD8
+
T cells than on the surface
of CD4
+
T cells [
316
]. PSGL-1 is involved in the migration of T cells to the inflammation site
and secondary lymphoid tissues [
317
]. However, in chronic inflammation, PSGL-1 signaling
limits T cell survival and promotes CD8
+
T cell exhaustion because of the upregulation of
immune checkpoint molecules such as PD-1, B- and T-lymphocyte attenuator (BTLA), T
cell immunoglobulin mucin 3 (TIM-3), and lymphocyte activation gene 3 (LAG-3), and the
inhibition of immunostimulatory cytokines such as IL-2. The suppression of PSGL-1 may
prevent the exhaustion of cytotoxic T cells and reduce tumor growth [
267
]. However, this
effect may occur not through P-selectin—PSGL-1 ligation but through TCR signaling [
267
]
or the binding to other ligands [318].
TGF-
β
suppresses CD8
+
T cells and can promote cancer cell proliferation [
319
] and
metastasis [
320
]. The activation of latent TGF-
β
through the TGF-
β
/glycoprotein A repeti-
tions predominant (GARP) axis in the TME via platelets may inhibit the antitumor effect
of T cells [
321
]. The activation of platelets encourages tumor growth, at least partly, by
supporting the immunosuppressive TME and minimizing the adaptive immune response.
Therefore, the inhibition of platelets may increase the effects of immune checkpoint in-
hibitors [
322
]. In addition, bioengineered PD-1-expressing or antineoplastic agent-carrying
platelets may accumulate in tumors and block PD-L1 on cancer cells to help exhausted
CD8
+
T cells recover their functionality [
323
]. CD8
+
T Tregs, which are known to reduce the
expansion of CD4
+
T cells and follicular helper T cells, are enriched in ovarian tumors [
324
].
Indeed, CD8
+
CD25
+
Foxp3
+
CTLA-4
+
T cells have been detected in patients with ovarian
cancer and have increased in number as the tumor stage increases [
324
,
325
]. The increased
levels of TGF-
β
expand CD8
+
Tregs in the ovarian cancer TME, mainly through p38 MAPK.
The inhibition of TGF-
β
and the p38 signaling cascade can impede ovarian cancer progres-
sion [
326
]. In addition, various other mediators, including IFN-
γ
and IL-2, support the
regulatory functions of CD8
+
T cells [
324
]. In contrast, TGF-
β
upregulates CD103 (also
known as integrin
α
E
β
7 (ITGAE)) on CD8
+
T cells infiltrating the tumor and malignant
Cancers 2023,15, 1282 16 of 37
ascites, and by supporting the habitation of these cytotoxic T cells in the TME, they may
represent a prognostic advantage in patients with ovarian cancer [327].
Thrombin exerts its protumorigenic properties through platelet activation and the
subsequent release of various mediators involving VEGF and TGF-
β
[
198
,
328
]. These medi-
ators can further modify the immune response toward a tolerogenic state [
198
,
329
]. Specific
drugs, such as dabigatran etexilate, when given concurrently with potentially thrombogenic
antineoplastic agents, repress thrombin and thus benefit patients with ovarian cancer. This
treatment regimen may reduce the number of immunoregulatory myeloid cells and incite
CD8+cytotoxic T cell function in ascites [330].
Similarly, platelet-derived PF4 stimulates the differentiation of monocytes into myeloid-
derived suppressor cells (MDSCs), which suppresses the activity of CD8
+
T cells. This shift
can result in advanced metastatic disease and poor overall survival [
21
]. An antiplatelet
agent-based therapy regimen can promote the expansion of CD8
+
T cells, and the admin-
istration of antiplatelet agents with an anti-VEGF or anti-PD-1 antibody can resensitize
tumors to immune cells [331,332].
3.4. Interplay with B Cells
B cells are immune cells that produce antibodies during initial exposure to antigens
and memory cells and that mediate a subsequent antibody response after re-exposure to
the same antigens. Mature naïve B lymphocytes in the peripheral lymphoid structure
are exposed to antigens, selectively expand, hypermutate to generate antibodies that
better fit specific antigens, and differentiate into plasma cells or transform into memory
B cells [
333
]. Antibodies produced by B cells may recognize antigens on malignant cells
or other components of the TME, tag cancer cells for destruction, and lead to immune
cell- and antibody-mediated cytotoxicity [
334
]. On the other hand, cancer cells or their
extracellular vesicles modify the immune system, promote the generation of regulatory
B cells (Bregs) [
335
], and evade the antineoplastic impact of B cells [
336
]. Moreover,
circulating antigen–antibody complexes activate the complement system and platelets,
supporting angiogenesis and metastasis [
337
339
]. B cells may have prognostic significance
in ovarian cancer patients and have been shown to correlate with worse [
340
] or better [
341
]
clinical outcomes. High numbers of IL-10-positive Bregs have been detected in ascites of
ovarian cancer patients, and their numbers are correlated with the percentage of Tregs. The
recruitment of Bregs and Tregs is associated with advanced-stage and aggressive ovarian
cancer [335].
The binding of CD40L to CD40 on resting B cells stimulates their differentiation to
plasma cells and memory B cells. Platelets and platelet microparticles express CD40L
and can trigger B cells’ production of antibodies, although not at the same level as the
T cell-dependent isotype-switched antibodies [
342
]. Through CD40L, platelets enable B
cell isotype conversion and enhance germinal center formation [
342
]. B cells stimulated
through CD40 and TLR9 restrict the differentiation of monocytes into mature dendritic cells
(DCs) and diminish DC-dependent T cell proliferation and function [
343
]. CD40–CD40L
contact is linked to the activation of Bregs and tumor growth via the suppression of IL-10
and TGF-
β
1 release [
344
]. In contrast, CD40L released from B cells binds platelet CD40 and
activates platelets, causing the expression of P-selectin, the secretion of
α
- and
γ
-granules,
the transformation of platelet morphology, and the upregulation of GPIIb/IIIa [58].
In addition to platelets and Tregs, peripheral B cells also express GARP, which releases
TGF-
β
and can induce immune tolerance [
345
]. TGF-
β
signaling stimulates the apoptosis
of B cells [
345
], suppresses immunoglobulin release, and impedes surface immunoglobulin
expression on activated B lymphocytes [
346
]. In addition, TGF-
β
promotes the class
switch to IgA during the differentiation of B cells into plasma cells [
347
]. Although IgA is
primarily associated with defense against pathogens on mucosal surfaces, IgG is involved
in adaptive and memory immune responses. Accordingly, the trend toward IgA instead
of IgG leads to a curtailed cytotoxic immune response in tissue [
348
]. Bregs’ production
of excess TGF-
β
can further propel the tolerogenic immune response by hastening the
Cancers 2023,15, 1282 17 of 37
differentiation of cytotoxic T cells into Tregs [
349
]. Thus, platelet-derived TGF-
β
may
contribute to immunosuppressive conditions in tumor progression.
PAF, an inflammatory phospholipid produced mainly by platelets, immune cells, and
endothelial cells [350], plays a role in B cell activation, expansion, and Ig production. PAF
receptor is upregulated on B cells by several cytokines, including IL-4 and TGF-
β
. The
binding of PAF to its receptors on B cells activates these cells [351].
The adhesion molecules on platelets that support the interaction between platelets
and immune cells are summarized in Figure 3.
Cancers 2023, 15, x FOR PEER REVIEW 18 of 38
Figure 3. The molecules that mediate the interaction between platelets and immune cells in the tu-
mor immune microenvironment (TIME). Activated platelets and immune cells in the TME com-
municate through various receptors and ligands or secreted modulatory factors, and these interac-
tions can be targeted for the therapy of ovarian cancer.
Table 2. The influence of various platelet-associated molecules on TME in cancer.
Hallmark Platelet Constituents Prognostic Role References
Pro-
tumorigenic
Endostatin, angiostatin, TSP-1, angi-
opoietin-1, VEGF, PDFG, HGF, FGF,
SDF-1, S1P, TGF-β, IL-6
Stimulation of angiogenesis [61,90]
Figure 3.
The molecules that mediate the interaction between platelets and immune cells in the tumor
immune microenvironment (TIME). Activated platelets and immune cells in the TME communicate
through various receptors and ligands or secreted modulatory factors, and these interactions can be
targeted for the therapy of ovarian cancer.
Cancers 2023,15, 1282 18 of 37
Table 2. The influence of various platelet-associated molecules on TME in cancer.
Hallmark Platelet Constituents Prognostic Role References
Pro-
tumorigenic
Endostatin, angiostatin, TSP-1,
angiopoietin-1, VEGF, PDFG,
HGF, FGF, SDF-1, S1P,
TGF-β, IL-6
Stimulation of angiogenesis [61,90]
TGF-β, angiopoietin, PDGF,
CLEC-2, IL-6
Proliferation, differentiation, and
irregularity of pericytes [64,125,126,128,138]
Platelet microparticles Overexpression of MMPs on
endothelial cells [100]
P-selectin, GPIIb Transendothelial migration of
cancer cells [108,109]
VEGF, angiopoietin-1, PDGF,
IGF-1, CLEC-2
Stimulation of lymphangiogenesis
[61,111,113]
TGF-β, SDF-1, PDGF, CLEC-2
Stimulation and differentiation,
proliferation, and migration
of CAFs
[15,147149]
TGF-β, G-CSF
Differentiation of neutrophils into
N2-like phenotype [173]
IL-8, G-CSF, GM-CSF, CXCL1,
CXCL2,
CXCL5, MIP-1α
Recruitment of neutrophils
into TME [192,197]
factor XII, P-selectin, GPIIb/IIIa
Platelet interaction with NETs and
subsequent platelet aggregation [202,204]
mtDNA, P-selectin NETosis [202,205]
P-selectin, TGF-β, COX-2, PGE2,
PF4, PAF, PDGF Activation of TAMs [220222]
CCL2, CCL5, RANTES, SDF-1 Migration of TAMs into TME [238,241]
CD40, CCL5, TxA2 Apoptosis and inhibition of
CD4+T helper cells [264,298]
PF4, TGF-β, P-selectin Suppression of Th1 and Th17
differentiation [288,297,309,310]
P-selectin Stimulation of T cell exhaustion [267]
PD-L1, TGF-β
Inhibition of CD4+and
CD8+T cell
antitumor functions
[52,319321,326]
CD40L, P-selectin, TNF-α, IL-6,
IGF-1, PDGF, SDF-1
Activation, proliferation, and
migration of Tregs
[16,62,73,74,263,285,293,
302,303]
PF4 Differentiation of monocytes
into MDSCs [21]
CD40L Expansion of Bregs [344]
Platelet microparticles Overexpression of MMPs [61,100]
CXCL5, CXCL7, TGF-β, TSP-1,
P2Y12, COX-1, TxA2
Formation of pre-metastatic
niches [352]
Cancers 2023,15, 1282 19 of 37
Table 2. Cont.
Hallmark Platelet Constituents Prognostic Role References
Anti-
tumorigenic
Endostatin, angiostatin, TSP-1 Inhibition of angiogenesis [56,61]
TRAIL, CCL2, MCP-1, IL-8 Activation of tumoricidal
macrophages [229]
Platelet microparticles Enhancement of the phagocytic
capacity of macrophages [228]
TGF-β, EGF
Upregulation of PD-1 in
cancer cells
and increase in the effectiveness
of immunotherapy
[14,270]
TGF-β, CD40L Activation and infiltration of
CD8+T cells into the tumor [327,353]
CD40L
Differentiation of resting B cells
into plasma cells and generation
of antibodies
[342]
OX40L Infiltration of immune cells
into TME [54]
CCL: chemokine (C-C motif) ligand; CD40L: CD40 ligand; CLEC-2: C-type lectin-like receptor 2; COX: cy-
clooxygenase; CXCL: chemokine (C-X-C motif) ligand; EGF: epidermal growth factor; FGF: fibroblast growth
factor; G-CSF: granulocyte colony-stimulating factor; GM-CSF: granulocyte-monocyte colony-stimulating factor;
HGF: hepatocyte growth factor; IGF-1: insulin-like growth factor 1; IL: interleukin; MCP-1: monocyte chemoat-
tractant protein 1; MIP-1
α
: migration inhibitory protein 1-alpha; mtDNA: mitochondrial DNA; PAF: platelet-
activating factor; PD-L1: programmed death ligand 1; PDGF: platelet-derived growth factor; PF4: platelet factor 4;
PGE2: prostaglandin E2; RANTES: regulated upon activation and normal T cell expressed and secreted;
S1P: sphingosine 1-phosphate; SDF-1: stromal-derived factor 1; TGF-
β
: transforming growth factor-beta;
TNF-
α
: tumor necrosis factor-alpha; TRAIL: TNF-related apoptosis-inducing ligand; TSP-1: thrombospondin 1;
TxA2: thromboxane A2; VEGF: vascular endothelial growth factor.
4. Discussion and Perspectives
The involvement of platelets in cancer progression through the modulation of tu-
mor growth, angiogenesis, metastasis, and chemoresistance has been recognized for
decades [
354
]. Tumor cells activate platelets, and platelets alter cancer cells and the TME
and their interaction [
64
]. As a result, improving our understanding of platelets’ role in
cancer would have a diagnostic, prognostic, and therapeutic impact.
Ovarian cancer is one of the many types of cancer in which platelets interact with
immune and nonimmune cellular components of the TME, including endothelial cells,
pericytes, CAFs, neutrophils, macrophages, and T and B lymphocytes. The adhesion
molecules on the platelets mediate the platelets’ binding to endothelial cells and extravasa-
tion [
355
357
]. The proangiogenic factors released by activated platelets, including VEGF
and angiopoietin-1, facilitate neovascularization, lymphangiogenesis [
61
], and remodeling
of the ECM [
100
], required for tumor growth and metastasis. The binding of platelet
CLEC-2 to podoplanin on pericytes [
125
] and the release of platelet-derived mediators,
such as TGF-
β
and PDGF, promote pericyte accumulation and interaction with endothe-
lial cells [
64
]. Thus, platelets, through pericytes, contribute to vessel density [
127
] and
integrity [
128
,
129
]. Platelets [
147
,
148
], platelet microparticles [
24
], and platelet-derived
mitochondria [
24
] contribute to the transformation of fibroblasts to CAFs, which promotes
the peritoneal dissemination of ovarian cancer cells [
160
] and the formation of malignant
ascites [358].
Platelets communicate with immune cells and induce a tolerogenic state [
359
]. Platelets
bind neutrophils [
181
183
,
188
,
189
] and facilitate neutrophil recruitment to the TME. Cy-
tokines and chemokines released from platelets promote neutrophil migration to the
TME [
360
]. Neutrophils activate platelets and promote thrombosis by releasing prothrom-
botic mediators and generating NETs [
360
,
361
]. Platelet–macrophage interaction favors
Cancers 2023,15, 1282 20 of 37
immunosuppressive macrophage polarization [
222
,
362
]. Activated monocytes, in turn,
promote coagulation and thrombosis [
230
]. Platelets promote the exhaustion of CD4
+
T
helper and CD8
+
cytotoxic T cells [
321
] in conjunction with Tregs and Bregs [
363
]. More-
over, platelets promote the expression of immune checkpoint molecules on ovarian cancer
cells, which further supports cancer progression [
14
] and can affect patients’ responses to
immunotherapies [307,364].
Thrombocytosis is responsible for the approximately 8-fold elevated risk of ovarian
cancer occurrence and 5-fold mortality rate in ovarian cancer patients [
28
]. An elevated
platelet count is significantly associated with poor progression-free and overall survival
of patients with localized and advanced cancer stages [
365
]. Moreover, ovarian cancer
patients with high platelet numbers respond less to chemotherapy and demonstrate shorter
treatment-free intervals caused by increased resistance [
366
,
367
]. This makes platelet
count an important parameter for the preoperative assessment of pelvic mass [
368
] and in
evaluating the response to therapy in ovarian cancer patients [369].
Antiplatelet reagents that have been extensively studied in cardiovascular disease [
370
,
371
]
can beconsidered for potential therapeutic goals in cancers, including ovarian cancer. Agents
that hinder platelet functions, including aspirin, ADP receptor antagonists (e.g., ticagrelor
and clopidogrel), thrombin receptor-targeting molecules, inhibitors of GP Ib-IX, P-selectin,
and CLEC-2, can be used as complementary therapeutics in ovarian cancer. Although they
have arole in preventing cancer progression [
372
374
], long-term usage of antiplatelet agents
such as aspirin, clopidogrel, and ticagrelor is associated with increased bleeding risk [
375
,
376
].
The blockade of CLEC-2 can be a beneficial alternative for patients with elevated bleeding
susceptibility, since this approach a lower tendency to cause hemorrhage due to its inhibition
of platelet activation without substantially affecting aggregation [
227
,
377
]. Including agents
that target EGFR, PDGFR, and FGFR in anti-VEGF therapy can also improve the efficacy of
cancer treatment by rewiring multiple cellular pathways and minimizing drug resistance in
ovarian and other cancers [
89
,
378
,
379
]. Apart from suppressing the viability, progression, and
dissemination of tumor cells, platelet-targeting compounds interfere with platelet-induced
angiogenesis, thrombosis, ascites formation, and immunosuppression. Multiple clinical trials
have assessed the impact of antiplatelet agents, alone or with chemotherapeutic drugs, on
preventing or treating ovarian cancer. Significantly, platelet counts or platelet-derived molecules
can beused as diagnostic, prognostic, or predictive biomarkers [380].
Of note, thrombocytopenia, a side-effect of cancer treatment, is a common phe-
nomenon in cancer patients [
381
]. Platelet transfusion and platelet-stimulating agent treat-
ment are clinically exploited applications to recover the platelet counts of cancer patients
with immune thrombocytopenia and those who have undergone chemotherapy or radio-
therapy. On the other hand, thrombocytosis supports tumor growth and chemoresistance
by enhancing platelet-directed alterations in cancer cells and TME components [
30
,
382
].
Therefore, extra caution must be taken during the treatment of thrombocytopenia to avoid
promoting cancer progression.
5. Conclusions
Paraneoplastic thrombocytosis in ovarian cancer is due to IL-6 originating from can-
cer cells and consequent thrombopoietin released from the liver. Thrombocytosis and
activated platelets can be detected in advanced diseases, and might be associated with
cancer-induced venous thrombosis and reduced overall survival. Besides cancer cells,
platelets interact with other components of the TME. Antiplatelet strategies might interfere
with neoangiogenesis, lymphangiogenesis, ECM remodeling, and the immunosuppressive
profile of the TME. In addition, antiplatelet agents may reduce the formation of malignant
ascites and premetastatic niches. Future studies must examine platelet—TME interaction
and plan specific treatments for ovarian and other cancers.
Cancers 2023,15, 1282 21 of 37
Author Contributions:
Conceptualization, M.S.C.; initial draft preparation, S.O. and M.S.C.; draft
review and editing, S.O. and M.S.C.; visualization, S.O. and M.S.C.; supervision, M.S.C.; project
administration, M.S.C. All authors have read and agreed to the published version of the manuscript.
Funding:
This work was supported by an Early Career Investigator Grant from the Ovarian Cancer
Research Alliance (ECIG-P-2018–600065); the Ovarian Cancer Research Fund in honor of Liza Chance
from the Foundation for Women’s Cancer; and the National Institutes of Health (P50 CA217685)
(all to M.S.C.).
Acknowledgments:
The authors thank Vahid Afshar-Kharghan, Section of Benign Hematology,
Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center) for his
critical reading and valuable comments on the manuscript. Figures were created using Biorender.com.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2022,72, 7–33. [CrossRef] [PubMed]
2.
Jelovac, D.; Armstrong, D.K. Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J. Clin.
2011
,61,
183–203. [CrossRef] [PubMed]
3.
Etemadmoghadam, D.; Defazio, A.; Beroukhim, R.; Mermel, C.; George, J.; Getz, G.; Tothill, R.; Okamoto, A.; Raeder, M.B.;
Harnett, P.; et al. Integrated Genome-Wide DNA Copy Number and Expression Analysis Identifies Distinct Mechanisms of
Primary Chemoresistance in Ovarian Carcinomas. Clin. Cancer Res. 2009,15, 1417–1427. [CrossRef] [PubMed]
4. Patch, A.-M.; Christie, E.L.; Etemadmoghadam, D.; Garsed, D.W.; George, J.; Fereday, S.; Nones, K.; Cowin, P.; Alsop, K.; Bailey,
P.J.; et al. Whole–genome characterization of chemoresistant ovarian cancer. Nature 2015,521, 489–494. [CrossRef]
5.
Jiménez-Sánchez, A.; Cybulska, P.; Mager, K.L.; Koplev, S.; Cast, O.; Couturier, D.-L.; Memon, D.; Selenica, P.; Nikolovski, I.;
Mazaheri, Y.; et al. Unraveling tumor–immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of
chemotherapy. Nat. Genet. 2020,52, 582–593. [CrossRef]
6.
Jin, M.-Z.; Jin, W.-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther.
2020,5, 166. [CrossRef]
7.
Jordan, K.R.; Sikora, M.J.; Slansky, J.E.; Minic, A.; Richer, J.K.; Moroney, M.R.; Hu, J.; Wolsky, R.J.; Watson, Z.L.; Yamamoto, T.M.;
et al. The Capacity of the Ovarian Cancer Tumor Microenvironment to Integrate Inflammation Signaling Conveys a Shorter
Disease-free Interval. Clin. Cancer Res. 2020,26, 6362–6373. [CrossRef]
8.
CChen, F.; Zhuang, X.; Lin, L.; Yu, P.; Wang, Y.; Shi, Y.; Hu, G.; Sun, Y. New horizons in tumor microenvironment biology:
Challenges and opportunities. BMC Med. 2015,13, 2498. [CrossRef] [PubMed]
9.
Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman,
W.H.; et al. The clinical role of the TME in solid cancer. Br. J. Cancer 2019,120, 45–53. [CrossRef]
10.
Corvigno, S.; Burks, J.K.; Hu, W.; Zhong, Y.; Jennings, N.B.; Fleming, N.D.; Westin, S.N.; Fellman, B.; Liu, J.; Sood, A.K. Immune
microenvironment composition in high-grade serous ovarian cancers based on BRCA mutational status. J. Cancer Res. Clin. Oncol.
2021,147, 3545–3555. [CrossRef]
11.
Kreuzinger, C.; Geroldinger, A.; Smeets, D.; Braicu, E.I.; Sehouli, J.; Koller, J.; Wolf, A.; Darb-Esfahani, S.; Joehrens, K.; Vergote, I.;
et al. A Complex Network of Tumor Microenvironment in Human High-Grade Serous Ovarian Cancer. Clin. Cancer Res.
2017
,23,
7621–7632. [CrossRef]
12.
Ukita, M.; Hamanishi, J.; Yoshitomi, H.; Yamanoi, K.; Takamatsu, S.; Ueda, A.; Suzuki, H.; Hosoe, Y.; Furutake, Y.; Taki, M.; et al.
CXCL13-producing CD4
+
T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. J. Clin. Investig.
2022,7, e157215. [CrossRef] [PubMed]
13.
Taylor, S.E.; Chan, D.K.; Yang, D.; Bruno, T.; Lieberman, R.; Siddiqui, J.; Soong, T.R.; Coffman, L.; Buckanovich, R.J. Shifting the
Soil: Metformin Treatment Decreases the Protumorigenic Tumor Microenvironment in Epithelial Ovarian Cancer. Cancers
2022
,
14, 2298. [CrossRef] [PubMed]
14.
Cho, M.S.; Lee, H.; Gonzalez-Delgado, R.; Li, D.; Sasano, T.; Carlos-Alcalde, W.; Ma, Q.; Liu, J.; Sood, A.K.; Afshar-Kharghan, V.
Platelets Increase the Expression of PD-L1 in Ovarian Cancer. Cancers 2022,14, 2498. [CrossRef] [PubMed]
15.
Miyashita, T.; Tajima, H.; Gabata, R.; Okazaki, M.; Shimbashi, H.; Ohbatake, Y.; Okamoto, K.; Nakanuma, S.; Sakai, S.; Makino, I.;
et al. Impact of Extravasated Platelet Activation and Podoplanin-positive Cancer-associated Fibroblasts in Pancreatic Cancer
Stroma. Anticancer. Res. 2019,39, 5565–5572. [CrossRef] [PubMed]
16.
Nasti, T.H.; Bullard, D.C.; Yusuf, N. P-selectin enhances growth and metastasis of mouse mammary tumors by promoting
regulatory T cell infiltration into the tumors. Life Sci. 2015,131, 11–18. [CrossRef]
17.
Yamaguchi, T.; Fushida, S.; Kinoshita, J.; Okazaki, M.; Ishikawa, S.; Ohbatake, Y.; Terai, S.; Okamoto, K.; Nakanuma, S.; Makino, I.;
et al. Extravasated platelet aggregation contributes to tumor progression via the accumulation of myeloid-derived suppressor
cells in gastric cancer with peritoneal metastasis. Oncol. Lett. 2020,20, 1879–1887. [CrossRef]
Cancers 2023,15, 1282 22 of 37
18.
Lonsdorf, A.S.; Krämer, B.F.; Fahrleitner, M.; Schönberger, T.; Gnerlich, S.; Ring, S.; Gehring, S.; Schneider, S.W.; Kruhlak, M.J.;
Meuth, S.G.; et al. Engagement of
α
IIb
β
3 (GPIIb/IIIa) with
ανβ
3 Integrin Mediates Interaction of Melanoma Cells with Platelets.
J. Biol. Chem. 2012,287, 2168–2178. [CrossRef]
19.
Shirai, T.; Inoue, O.; Tamura, S.; Tsukiji, N.; Sasaki, T.; Endo, H.; Satoh, K.; Osada, M.; Sato-Uchida, H.; Fujii, H.; et al. C-type
lectin-like receptor 2 promotes hematogenous tumor metastasis and prothrombotic state in tumor-bearing mice. J. Thromb.
Haemost. 2016,15, 513–525. [CrossRef]
20.
Haemmerle, M.; Bottsford-Miller, J.; Pradeep, S.; Taylor, M.L.; Choi, H.-J.; Hansen, J.M.; Dalton, H.J.; Stone, R.L.; Cho, M.S.; Nick,
A.M.; et al. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal. J. Clin. Investig.
2016,126, 1885–1896. [CrossRef]
21.
Joseph, R.; Soundararajan, R.; Vasaikar, S.; Yang, F.; Allton, K.L.; Tian, L.; Hollander, P.D.; Isgandarova, S.; Haemmerle, M.; Mino,
B.; et al. CD8+T cells inhibit metastasis and CXCL4 regulates its function. Br. J. Cancer 2021,125, 176–189. [CrossRef]
22.
Plantureux, L.; Mège, D.; Crescence, L.; Carminita, E.; Robert, S.; Cointe, S.; Brouilly, N.; Ezzedine, W.; Dignat-George, F.; Dubois,
C.; et al. The Interaction of Platelets with Colorectal Cancer Cells Inhibits Tumor Growth but Promotes Metastasis. Cancer Res.
2020,80, 291–303. [CrossRef] [PubMed]
23.
Pavlovic, N.; Rani, B.; Gerwins, P.; Heindryckx, F. Platelets as Key Factors in Hepatocellular Carcinoma. Cancers
2019
,11,
1022. [CrossRef] [PubMed]
24.
Levoux, J.; Prola, A.; Lafuste, P.; Gervais, M.; Chevallier, N.; Koumaiha, Z.; Kefi, K.; Braud, L.; Schmitt, A.; Yacia, A.; et al. Platelets
Facilitate the Wound-Healing Capability of Mesenchymal Stem Cells by Mitochondrial Transfer and Metabolic Reprogramming.
Cell Metab. 2021,33, 283–299. [CrossRef]
25.
Sibilano, M.; Tullio, V.; Adorno, G.; Savini, I.; Gasperi, V.; Catani, M.V. Platelet-Derived miR-126-3p Directly Targets AKT2
and Exerts Anti-Tumor Effects in Breast Cancer Cells: Further Insights in Platelet-Cancer Interplay. Int. J. Mol. Sci.
2022
,23,
5484. [CrossRef] [PubMed]
26.
Ye, B.; Li, F.; Chen, M.; Weng, Y.; Qi, C.; Xie, Y.; Zhang, Q.; Ding, H.; Zhang, J.; Gao, X. A panel of platelet-associated circulating
long non-coding RNAs as potential biomarkers for colorectal cancer. Genomics 2021,114, 31–37. [CrossRef] [PubMed]
27.
Stone, R.L.; Nick, A.M.; McNeish, I.A.; Balkwill, F.; Han, H.D.; Bottsford-Miller, J.; Rupaimoole, R.; Armaiz-Pena, G.N.; Pecot, C.V.;
Coward, J.; et al. Paraneoplastic Thrombocytosis in Ovarian Cancer. N. Engl. J. Med. 2012,366, 610–618. [CrossRef] [PubMed]
28.
Allensworth, S.; Langstraat, C.; Martin, J.; Lemens, M.; McGree, M.; Weaver, A.; Dowdy, S.; Podratz, K.; Bakkum-Gamez, J.
Evaluating the prognostic significance of preoperative thrombocytosis in epithelial ovarian cancer. Gynecol. Oncol.
2013
,130,
499–504. [CrossRef]
29.
Cohen, J.G.; Tran, A.-Q.; Rimel, B.; Cass, I.; Walsh, C.S.; Karlan, B.Y.; Li, A.J. Thrombocytosis at secondary cytoreduction for
recurrent ovarian cancer predicts suboptimal resection and poor survival. Gynecol. Oncol. 2014,132, 556–559. [CrossRef]
30.
Bottsford-Miller, J.; Choi, H.-J.; Dalton, H.J.; Stone, R.L.; Cho, M.S.; Haemmerle, M.; Nick, A.M.; Pradeep, S.; Zand, B.; Previs,
R.A.; et al. Differential Platelet Levels Affect Response to Taxane-Based Therapy in Ovarian Cancer. Clin. Cancer Res.
2015
,21,
602–610. [CrossRef]
31.
Cho, M.S.; Bottsford-Miller, J.; Vasquez, H.G.; Stone, R.; Zand, B.; Kroll, M.H.; Sood, A.K.; Afshar-Kharghan, V. Platelets increase
the proliferation of ovarian cancer cells. Blood 2012,120, 4869–4872. [CrossRef] [PubMed]
32.
Guo, Y.; Cui, W.; Pei, Y.; Xu, D. Platelets promote invasion and induce epithelial to mesenchymal transition in ovarian cancer cells
by TGF-βsignaling pathway. Gynecol. Oncol. 2019,153, 639–650. [CrossRef] [PubMed]
33.
Haemmerle, M.; Taylor, M.L.; Gutschner, T.; Pradeep, S.; Cho, M.S.; Sheng, J.; Lyons, Y.M.; Nagaraja, A.S.; Dood, R.L.;
Wen, Y.; et al. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat. Commun.
2017
,8,
310. [CrossRef] [PubMed]
34. Davis, A.N.; Afshar-Kharghan, V.; Sood, A.K. Platelet Effects on Ovarian Cancer. Semin. Oncol. 2014,41, 378–384. [CrossRef]
35.
Cho, M.S.; Gonzalez-Pagan, O.; Pinto, K.C.; Sood, A.; Afshar-Kharghan, V. The Inhibition of Platelets Restore Anti-Tumor Immune
Response to Ovarian Cancer and Its Therapeutic Implication. Blood 2018,132, 3698. [CrossRef]
36.
Saboor, M.; Ayub, Q.; Ilyas, S. Moinuddin Platelet receptors; an instrumental of platelet physiology. Pak. J. Med. Sci.
2013
,29,
891–896. [CrossRef]
37.
McEver, R.P.; Beckstead, J.H.; Moore, K.L.; Marshall-Carlson, L.; Bainton, D.F. GMP-140, a platelet alpha-granule membrane
protein, is also synthesized by vascular endothelial cells and is localized in Weibel-Palade bodies. J. Clin. Investig.
1989
,84,
92–99. [CrossRef]
38.
Rayes, J.; Lax, S.; Wichaiyo, S.; Watson, S.K.; Di, Y.; Lombard, S.; Grygielska, B.; Smith, S.W.; Skordilis, K.; Watson, S.P. The
podoplanin-CLEC-2 axis inhibits inflammation in sepsis. Nat. Commun. 2017,8, 2239. [CrossRef]
39.
Cognasse, F.; Nguyen, K.A.; Damien, P.; McNicol, A.; Pozzetto, B.; Hamzeh-Cognasse, H.; Garraud, O. The inflammatory role of
platelets via their TLRs and Siglec receptors. Front. Immunol. 2015,6, 83. [CrossRef]
40.
Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair,
G.D.; et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med.
2007
,13,
463–469. [CrossRef]
41.
Cho, M.S.; Noh, K.; Haemmerle, M.; Li, D.; Park, H.; Hu, Q.; Hisamatsu, T.; Mitamura, T.; Mak, S.L.C.; Kunapuli, S.; et al. Role of
ADP receptors on platelets in the growth of ovarian cancer. Blood 2017,130, 1235–1242. [CrossRef] [PubMed]
Cancers 2023,15, 1282 23 of 37
42.
Camerer, E.; Qazi, A.A.; Duong, D.N.; Cornelissen, I.; Advincula, R.; Coughlin, S.R. Platelets, protease-activated receptors, and
fibrinogen in hematogenous metastasis. Blood 2004,104, 397–401. [CrossRef]
43.
Jain, S.; Zuka, M.; Liu, J.; Russell, S.; Dent, J.; Guerrero, J.A.; Forsyth, J.; Maruszak, B.; Gartner, T.K.; Felding-Habermann,
B.; et al. Platelet glycoprotein Ib
α
supports experimental lung metastasis. Proc. Natl. Acad. Sci. USA
2007
,104,
9024–9028. [CrossRef] [PubMed]
44.
Tynan, S.S.; Andersen, N.H.; Wills, M.T.; Harker, L.A.; Hanson, S.R. On the multiplicity of platelet prostaglandin receptors.
Prostaglandins 1984,27, 683–696. [CrossRef] [PubMed]
45.
Paul, B.Z.S.; Jin, J.; Kunapuli, S.P. Molecular Mechanism of Thromboxane A2-induced Platelet Aggregation. J. Biol. Chem.
1999
,
274, 29108–29114. [CrossRef]
46.
Burgers, J.A.; Akkerman, J.W.N. Regulation of the receptor for platelet-activating factor on human platelets. Biochem. J.
1993
,291,
157–161. [CrossRef]
47.
Motohashi, K.; Shibata, S.; Ozaki, Y.; Yatomi, Y.; Igarashi, Y. Identification of lysophospholipid receptors in human platelets: The
relation of two agonists, lysophosphatidic acid and sphingosine 1-phosphate. FEBS Lett. 2000,468, 189–193. [CrossRef]
48.
Tsuji, M.; Ezumi, Y.; Arai, M.; Takayama, H. A Novel Association of Fc Receptor
γ
-Chain with Glycoprotein VI and Their
Co-expression as a Collagen Receptor in Human Platelets. J. Biol. Chem. 1997,272, 23528–23531. [CrossRef]
49.
Hasegawa, S.; Pawankar, R.; Suzuki, K.; Nakahata, T.; Furukawa, S.; Okumura, K.; Ra, C. Functional expression of the high
affinity receptor for IgE (FcepsilonRI) in human platelets and its’ intracellular expression in human megakaryocytes. Blood
1999
,
93, 2543–2551. [CrossRef]
50.
Santoso, S.; Sachs, U.J.H.; Kroll, H.; Linder, M.; Ruf, A.; Preissner, K.T.; Chavakis, T. The Junctional Adhesion Molecule 3 (JAM-3)
on Human Platelets is a Counterreceptor for the Leukocyte Integrin Mac-1. J. Exp. Med. 2002,196, 679–691. [CrossRef]
51.
Newman, D.K.; Hamilton, C.; Newman, P.J. Inhibition of antigen-receptor signaling by Platelet Endothelial Cell Adhesion
Molecule-1 (CD31) requires functional ITIMs, SHP-2, and p56lck. Blood 2001,97, 2351–2357. [CrossRef] [PubMed]
52.
Hinterleitner, C.; Strähle, J.; Malenke, E.; Hinterleitner, M.; Henning, M.; Seehawer, M.; Bilich, T.; Heitmann, J.; Lutz, M.; Mattern,
S.; et al. Platelet PD-L1 reflects collective intratumoral PD-L1 expression and predicts immunotherapy response in non-small cell
lung cancer. Nat. Commun. 2021,12, 7005. [CrossRef] [PubMed]
53.
Zhou, Y.; Heitmann, J.S.; Clar, K.L.; Kropp, K.N.; Hinterleitner, M.; Engler, T.; Koch, A.; Hartkopf, A.D.; Zender, L.; Salih,
H.R.; et al. Platelet-expressed immune checkpoint regulator GITRL in breast cancer. Cancer Immunol. Immunother.
2021
,70,
2483–2496. [CrossRef] [PubMed]
54.
Rittig, S.M.; Lutz, M.S.; Clar, K.L.; Zhou, Y.; Kropp, K.N.; Koch, A.; Hartkopf, A.D.; Hinterleitner, M.; Zender, L.; Salih, H.R.; et al.
Controversial Role of the Immune Checkpoint OX40L Expression on Platelets in Breast Cancer Progression. Front. Oncol.
2022
,12,
917834. [CrossRef]
55.
Dale, G.L.; Friese, P.; Batar, P.; Hamilton, S.F.; Reed, G.L.; Jackson, K.W.; Clemetson, K.J.; Alberio, L. Stimulated platelets use
serotonin to enhance their retention of procoagulant proteins on the cell surface. Nature
2002
,415, 175–179. [CrossRef] [PubMed]
56.
Isenberg, J.S.; Romeo, M.J.; Yu, C.; Nghiem, K.; Monsale, J.; Rick, M.E.; Wink, D.A.; Frazier, W.A.; Roberts, D.D. Thrombospondin-
1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling. Blood
2008
,111,
613–623. [CrossRef]
57.
Hamzeh-Cognasse, H.; Damien, P.; Chabert, A.; Pozzetto, B.; Cognasse, F.; Garraud, O. Platelets and Infections-Complex
Interactions with Bacteria. Front. Immunol. 2015,6, 82. [CrossRef]
58.
Inwald, D.P.; McDowall, A.; Peters, M.J.; Callard, R.E.; Klein, N.J. CD40 Is Constitutively Expressed on Platelets and Provides a
Novel Mechanism for Platelet Activation. Circ. Res. 2003,92, 1041–1048. [CrossRef]
59.
Huang, J.; Jochems, C.; Talaie, T.; Anderson, A.; Jales, A.; Tsang, K.Y.; Madan, R.A.; Gulley, J.L.; Schlom, J. Elevated serum soluble
CD40 ligand in cancer patients may play an immunosuppressive role. Blood 2012,120, 3030–3038. [CrossRef]
60.
Fitch-Tewfik, J.L.; Flaumenhaft, R. Platelet Granule Exocytosis: A Comparison with Chromaffin Cells. Front. Endocrinol.
2013
,4,
77. [CrossRef]
61.
Wojtukiewicz, M.Z.; Sierko, E.; Hempel, D.; Tucker, S.C.; Honn, K.V. Platelets and cancer angiogenesis nexus. Cancer Metastasis
Rev. 2017,36, 249–262. [CrossRef] [PubMed]
62.
Kim, S.; Garcia, A.; Jackson, S.P.; Kunapuli, S.P. Insulin-like growth factor-1 regulates platelet activation through PI3-K
α
isoform.
Blood 2007,110, 4206–4213. [CrossRef] [PubMed]
63.
Kubota, S.; Kawata, K.; Yanagita, T.; Doi, H.; Kitoh, T.; Takigawa, M. Abundant Retention and Release of Connective Tissue
Growth Factor (CTGF/CCN2) by Platelets. J. Biochem. 2004,136, 279–282. [CrossRef] [PubMed]
64.
Haemmerle, M.; Stone, R.L.; Menter, D.G.; Afshar-Kharghan, V.; Sood, A.K. The Platelet Lifeline to Cancer: Challenges and
Opportunities. Cancer Cell 2018,33, 965–983. [CrossRef] [PubMed]
65. Walsh, P.N. Platelets and coagulation proteins. Fed. Proc. 1981,40, 2086–2091. [PubMed]
66.
Stavenuiter, F.; Davis, N.F.; Duan, E.; Gale, A.J.; Heeb, M.J. Platelet protein S directly inhibits procoagulant activity on platelets
and microparticles. Thromb. Haemost. 2013,109, 229–237. [CrossRef]
67.
Trivedi, V.; Boire, A.; Tchernychev, B.; Kaneider, N.C.; Leger, A.J.; O’Callaghan, K.; Covic, L.; Kuliopulos, A. Platelet Matrix
Metalloprotease-1 Mediates Thrombogenesis by Activating PAR1 at a Cryptic Ligand Site. Cell 2009,137, 332–343. [CrossRef]
68.
Sheu, J.R.; Fong, T.H.; Liu, C.M.; Shen, M.Y.; Chen, T.L.; Chang, Y.; Lu, M.S.; Hsiao, G. Expression of matrix metalloproteinase-9 in
human platelets: Regulation of platelet activation in
in vitro
and
in vivo
studies. Br. J. Pharmacol.
2004
,143, 193–201. [CrossRef]
Cancers 2023,15, 1282 24 of 37
69.
Santos-Martínez, M.J.; Medina, C.; Jurasz, P.; Radomski, M.W. Role of metalloproteinases in platelet function. Thromb. Res.
2008
,
121, 535–542. [CrossRef]
70.
Lishko, V.K.; Yakubenko, V.P.; Ugarova, T.P.; Podolnikova, N.P. Leukocyte integrin Mac-1 (CD11b/CD18,
α
M
β
2, CR3) acts as a
functional receptor for platelet factor 4. J. Biol. Chem. 2018,293, 6869–6882. [CrossRef]
71.
Walz, A.; Dewald, B.; von Tscharner, V.; Baggiolini, M. Effects of the neutrophil-activating peptide NAP-2, platelet ba-
sic protein, connective tissue-activating peptide III and platelet factor 4 on human neutrophils. J. Exp. Med.
1989
,170,
1745–1750. [CrossRef] [PubMed]
72.
Chen, R.; Jin, G.; Li, W.; McIntyre, T.M. Epidermal Growth Factor (EGF) Autocrine Activation of Human Platelets Promotes EGF
Receptor–Dependent Oral Squamous Cell Carcinoma Invasion, Migration, and Epithelial Mesenchymal Transition. J. Immunol.
2018,201, 2154–2164. [CrossRef] [PubMed]
73.
Chen, Y.; Zhong, H.; Zhao, Y.; Luo, X.; Gao, W. Role of platelet biomarkers in inflammatory response. Biomark. Res.
2020
,
8, 28. [CrossRef]
74.
Bock, M.; Bergmann, C.B.; Jung, S.; Kalbitz, M.; Relja, B.; Huber-Wagner, S.; Biberthaler, P.; van Griensven, M.; Hanschen, M. The
posttraumatic activation of CD4
+
T regulatory cells is modulated by TNFR2- and TLR4-dependent pathways, but not by IL-10.
Cell. Immunol. 2018,331, 137–145. [CrossRef]
75.
Raiden, S.; Schettini, J.; Salamone, G.; Trevani, A.; Vermeulen, M.; Gamberale, R.; Giordano, M.; Geffner, J. Human Platelets
Produce Granulocyte-Macrophage Colony-Stimulating Factor and Delay Eosinophil Apoptosis. Lab. Investig.
2003
,83,
589–598. [CrossRef]
76.
Kim, H.; Conway, E.M. Platelets and Complement Crosstalk in Early Atherogenesis. Front. Cardiovasc. Med.
2019
,
6, 131. [CrossRef] [PubMed]
77.
George, J.N.; Saucerman, S. Platelet Igg, Iga, Igm, and Albumin: Correlation of Platelet and Plasma Concentrations in Normal
Subjects and in Patients with Itp or Dysproteinemia. Blood 1988,72, 362–365. [CrossRef]
78.
Huff, T.; Otto, A.M.; Müller, C.S.G.; Meier, M.; Hannappel, E. Thymosin
β4
is released from human blood platelets and attached
by factor XIIIa (transglutaminase) to fibrin and collagen. FASEB J. 2002,16, 691–696. [CrossRef]
79. de Jong, J.S.S.G.; Dekker, L.R.C. Platelets and Cardiac Arrhythmia. Front. Physiol. 2010,1, 166. [CrossRef]
80. Chesney, C.M.; Harper, E.; Colman, R.W. Human Platelet Collagenase. J. Clin. Investig. 1974,53, 1647–1654. [CrossRef]
81.
Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol.
Life Sci. 2020,77, 1745–1770. [CrossRef]
82.
Nagy, J.A.; Chang, S.-H.; Dvorak, A.M.; Dvorak, H.F. Why are tumour blood vessels abnormal and why is it important to know.
Br. J. Cancer 2009,100, 865–869. [CrossRef] [PubMed]
83.
Klein, D. The Tumor Vascular Endothelium as Decision Maker in Cancer Therapy. Front. Oncol.
2018
,8, 367. [CrossRef] [PubMed]
84.
Chaplin, D.J.; Olive, P.L.; Durand, R.E. Intermittent blood flow in a murine tumor: Radiobiological effects. Cancer Res.
1987
,47,
597–601. [PubMed]
85.
Fitzgerald, G.; Soro-Arnaiz, I.; de Bock, K. The Warburg Effect in Endothelial Cells and its Potential as an Anti-angiogenic Target
in Cancer. Front. Cell Dev. Biol. 2018,6, 100. [CrossRef] [PubMed]
86.
Dvorak, H.F.; Brown, L.F.; Detmar, M.; Dvorak, A.M. Vascular permeability factor/vascular endothelial growth factor, microvas-
cular hyperpermeability, and angiogenesis. Am. J. Pathol. 1995,146, 1029–1039. [PubMed]
87.
Perren, T.J.; Swart, A.M.; Pfisterer, J.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.;
Kurzeder, C.; et al. A Phase 3 Trial of Bevacizumab in Ovarian Cancer. N. Engl. J. Med. 2011,365, 2484–2496. [CrossRef]
88.
Burger, R.A.; Sill, M.W.; Monk, B.J.; Greer, B.E.; Sorosky, J.I. Phase II Trial of Bevacizumab in Persistent or Recurrent Ep-
ithelial Ovarian Cancer or Primary Peritoneal Cancer: A Gynecologic Oncology Group Study. J. Clin. Oncol.
2007
,25,
5165–5171. [CrossRef] [PubMed]
89.
Choi, H.-J.; Pena, G.N.A.; Pradeep, S.; Cho, M.S.; Coleman, R.L.; Sood, A.K. Anti-vascular therapies in ovarian cancer: Moving
beyond anti-VEGF approaches. Cancer Metastasis Rev. 2014,34, 19–40. [CrossRef]
90.
Kisucka, J.; Butterfield, C.E.; Duda, D.G.; Eichenberger, S.C.; Saffaripour, S.; Ware, J.; Ruggeri, Z.M.; Jain, R.K.; Folkman, J.;
Wagner, D.D. Platelets and platelet adhesion support angiogenesis while preventing excessive hemorrhage. Proc. Natl. Acad. Sci.
USA 2006,103, 855–860. [CrossRef]
91.
Martins, P.D.C.; García-Vallejo, J.-J.; van Thienen, J.V.; Fernandez-Borja, M.; van Gils, J.M.; Beckers, C.; Horrevoets, A.J.; Hordijk,
P.L.; Zwaginga, J.-J. P-Selectin Glycoprotein Ligand-1 Is Expressed on Endothelial Cells and Mediates Monocyte Adhesion to
Activated Endothelium. Arter. Thromb. Vasc. Biol. 2007,27, 1023–1029. [CrossRef] [PubMed]
92.
Gong, L.; Cai, Y.; Zhou, X.; Yang, H. Activated Platelets Interact with Lung Cancer Cells Through P-Selectin Glycoprotein
Ligand-1. Pathol. Oncol. Res. 2012,18, 989–996. [CrossRef] [PubMed]
93.
Sang, Y.; Roest, M.; de Laat, B.; de Groot, P.G.; Huskens, D. Interplay between platelets and coagulation. Blood Rev.
2020
,46,
100733. [CrossRef]
94.
Yan, M.; Lesyk, G.; Radziwon-Balicka, A.; Jurasz, P. Pharmacological Regulation of Platelet Factors That Influence Tumor
Angiogenesis. Semin. Oncol. 2014,41, 370–377. [CrossRef]
95.
Raica, M.; Cimpean, A.M. Platelet-Derived Growth Factor (PDGF)/PDGF Receptors (PDGFR) Axis as Target for Antitumor and
Antiangiogenic Therapy. Pharmaceuticals 2010,3, 572–599. [CrossRef] [PubMed]
Cancers 2023,15, 1282 25 of 37
96.
Langer, H.; May, A.E.; Daub, K.; Heinzmann, U.; Lang, P.; Schumm, M.; Vestweber, D.; Massberg, S.; Schönberger, T.; Pfisterer,
I.; et al. Adherent Platelets Recruit and Induce Differentiation of Murine Embryonic Endothelial Progenitor Cells to Mature
Endothelial Cells In Vitro. Circ. Res. 2006,98, e2–e10. [CrossRef]
97.
Trikha, M.; Zhou, Z.; Timar, J.; Raso, E.; Kennel, M.; Emmell, E.; Nakada, M.T. Multiple roles for platelet GPIIb/IIIa and
alphavbeta3 integrins in tumor growth, angiogenesis, and metastasis. Cancer Res. 2002,62, 2824–2833.
98.
Ho-Tin-Noé, B.; Goerge, T.; Cifuni, S.M.; Duerschmied, D.; Wagner, D.D. Platelet Granule Secretion Continuously Prevents
Intratumor Hemorrhage. Cancer Res. 2008,68, 6851–6858. [CrossRef]
99.
Schumacher, D.; Strilic, B.; Sivaraj, K.K.; Wettschureck, N.; Offermanns, S. Platelet-Derived Nucleotides Promote Tumor-Cell
Transendothelial Migration and Metastasis via P2Y2 Receptor. Cancer Cell 2013,24, 130–137. [CrossRef]
100.
Sun, C.; Feng, S.-B.; Cao, Z.-W.; Bei, J.-J.; Chen, Q.; Xu, X.-J.; Zhou, Z.; Yu, Z.-P.; Hu, H.-Y. Up-Regulated Expression of Matrix
Metalloproteinases in Endothelial Cells Mediates Platelet Microvesicle-Induced Angiogenesis. Cell. Physiol. Biochem.
2017
,41,
2319–2332. [CrossRef]
101.
Yuan, L.; Liu, X. Platelets are associated with xenograft tumor growth and the clinical malignancy of ovarian cancer through an
angiogenesis-dependent mechanism. Mol. Med. Rep. 2014,11, 2449–2458. [CrossRef] [PubMed]
102.
Artini, P.G.; Ruggiero, M.; Monteleone, P.; Carpi, A.; Cristello, F.; Cela, V.; Genazzani, A.R. Vascular endothelial growth factor and
its soluble receptor in benign and malignant ovarian tumors. Biomed. Pharmacother. 2008,62, 373–377. [CrossRef] [PubMed]
103.
Byrne, A.T.; Ross, L.; Holash, J.; Nakanishi, M.; Hu, L.; Hofmann, J.I.; Yancopoulos, G.D.; Jaffe, R.B. Vascular endothelial growth
factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascular remodeling in an ovarian cancer model. Clin.
Cancer Res. 2003,9, 5721–5728. [PubMed]
104.
Li, L.; Wang, L.; Zhang, W.; Tang, B.; Zhang, J.; Song, H.; Yao, D.; Tang, Y.; Chen, X.; Yang, Z.; et al. Correlation of serum VEGF
levels with clinical stage, therapy efficacy, tumor metastasis and patient survival in ovarian cancer. Anticancer. Res.
2004
,24,
1973–1979. [PubMed]
105.
Matei, D.; Kelich, S.; Cao, L.; Menning, N.; Emerson, R.E.; Rao, J.; Jeng, M.H.; Sledge, G.W. PDGF BB induces VEGF secretion in
ovarian cancer. Cancer Biol. Ther. 2007,6, 1951–1959. [CrossRef] [PubMed]
106.
Silini, A.; Ghilardi, C.; Figini, S.; Sangalli, F.; Fruscio, R.; Dahse, R.; Pedley, R.B.; Giavazzi, R.; Bani, M. Regulator of G-
protein signaling 5 (RGS5) protein: A novel marker of cancer vasculature elicited and sustained by the tumor’s proangiogenic
microenvironment. Cell. Mol. Life Sci. 2011,69, 1167–1178. [CrossRef]
107.
Bergmeier, W.; Piffath, C.L.; Goerge, T.; Cifuni, S.M.; Ruggeri, Z.M.; Ware, J.; Wagner, D.D. The role of platelet adhesion receptor
GPIb
α
far exceeds that of its main ligand, von Willebrand factor, in arterial thrombosis. Proc. Natl. Acad. Sci. USA
2006
,103,
16900–16905. [CrossRef]
108.
Borsig, L.; Wong, R.; Feramisco, J.; Nadeau, D.R.; Varki, N.M.; Varki, A. Heparin and cancer revisited: Mechanistic connections
involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proc. Natl. Acad. Sci. USA
2001
,98, 3352–3357. [CrossRef]
109.
Zhang, N.; Zhang, W.-J.; Cai, H.-Q.; Liu, H.-L.; Peng, L.; Li, C.-H.; Ye, L.-Y.; Xu, S.-Q.; Yang, Z.-H.; Lou, J.-N. Platelet adhesion and
fusion to endothelial cell facilitate the metastasis of tumor cell in hypoxia-reoxygenation condition. Clin. Exp. Metastasis
2010
,28,
1–12. [CrossRef]
110.
Stacker, S.A.; Williams, S.P.; Karnezis, T.; Shayan, R.; Fox, S.B.; Achen, M.G. Lymphangiogenesis and lymphatic vessel remodelling
in cancer. Nat. Rev. Cancer 2014,14, 159–172. [CrossRef]
111.
Suzuki-Inoue, K. Essential
in vivo
roles of the platelet activation receptor CLEC-2 in tumour metastasis, lymphangiogenesis and
thrombus formation. J. Biochem. 2011,150, 127–132. [CrossRef] [PubMed]
112.
Lim, L.; Bui, H.; Farrelly, O.; Yang, J.; Li, L.; Enis, D.; Ma, W.; Chen, M.; Oliver, G.; Welsh, J.D.; et al. Hemostasis stimulates
lymphangiogenesis through release and activation of VEGFC. Blood 2019,134, 1764–1775. [CrossRef] [PubMed]
113.
Da, M.-X.; Wu, Z.; Tian, H.-W. Tumor Lymphangiogenesis and Lymphangiogenic Growth Factors. Arch. Med. Res.
2008
,39,
365–372. [CrossRef]
114.
Sopo, M.; Anttila, M.; Muukkonen, O.-T.; Ylä-Herttuala, S.; Kosma, V.-M.; Keski-Nisula, L.; Sallinen, H. Microvessels in Epithelial
Ovarian Tumors: High Microvessel Density Is a Significant Feature of Malignant Ovarian Tumors. Anticancer. Res.
2020
,40,
6923–6931. [CrossRef]
115.
Kitano, H.; Kageyama, S.-I.; Hewitt, S.M.; Hayashi, R.; Doki, Y.; Ozaki, Y.; Fujino, S.; Takikita, M.; Kubo, H.; Fukuoka, J.
Podoplanin Expression in Cancerous Stroma Induces Lymphangiogenesis and Predicts Lymphatic Spread and Patient Survival.
Arch. Pathol. Lab. Med. 2010,134, 1520–1527. [CrossRef]
116.
Bianchi, R.; Fischer, E.; Yuen, D.; Ernst, E.; Ochsenbein, A.M.; Chen, L.; Otto, V.I.; Detmar, M. Mutation of Threonine 34 in Mouse
Podoplanin-Fc Reduces CLEC-2 Binding and Toxicity in Vivo While Retaining Anti-lymphangiogenic Activity. J. Biol. Chem.
2014
,
289, 21016–21027. [CrossRef]
117.
Zhao, R.-W.; Yang, S.-H.; Cai, L.-Q.; Zhang, J.; Wang, J.; Wang, Z.-H. Roles of vascular endothelial growth factor and platelet-
derived growth factor in lymphangiogenesis in epithelial ovarian carcinoma. Zhonghua Fu Chan Ke Za Zhi 2009,44, 760–764.
118.
Krishnapriya, S.; Sidhanth, C.; Manasa, P.; Sneha, S.; Bindhya, S.; Nagare, R.P.; Ramachandran, B.; Vishwanathan, P.; Murhekar,
K.; Shirley, S.; et al. Cancer stem cells contribute to angiogenesis and lymphangiogenesis in serous adenocarcinoma of the ovary.
Angiogenesis 2019,22, 441–455. [CrossRef]
119.
Liao, S.; Liu, J.; Lin, P.; Shi, T.; Jain, R.K.; Xu, L. TGF-
β
Blockade Controls Ascites by Preventing Abnormalization of Lymphatic
Vessels in Orthotopic Human Ovarian Carcinoma Models. Clin. Cancer Res. 2011,17, 1415–1424. [CrossRef] [PubMed]
Cancers 2023,15, 1282 26 of 37
120.
Sweeney, M.; Ayyadurai, S.; Zlokovic, B.V. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat.
Neurosci. 2016,19, 771–783. [CrossRef]
121.
Xian, X.; Håkansson, J.; Ståhlberg, A.; Lindblom, P.; Betsholtz, C.; Gerhardt, H.; Semb, H. Pericytes limit tumor cell metastasis. J.
Clin. Investig. 2006,116, 642–651. [CrossRef] [PubMed]
122.
Raza, A.; Franklin, M.J.; Dudek, A.Z. Pericytes and vessel maturation during tumor angiogenesis and metastasis. Am. J. Hematol.
2010,85, 593–598. [CrossRef] [PubMed]
123.
Bergers, G.; Song, S. The role of pericytes in blood-vessel formation and maintenance. Neuro-Oncology
2005
,7, 452–464. [CrossRef]
124.
Sinha, D.; Chong, L.; George, J.; Schlueter, H.; Mönchgesang, S.; Mills, S.; Jason Australian Ovarian Cancer Study Group; Parish,
C.R.; Bowtell, D.D.; Kaur, P. Pericytes Promote Malignant Ovarian Cancer Progression in Mice and Predict Poor Prognosis in
Serous Ovarian Cancer Patients. Clin. Cancer Res. 2016,22, 1813–1824. [CrossRef] [PubMed]
125.
Sasano, T.; Gonzalez-Delgado, R.; Muñoz, N.M.; Carlos-Alcade, W.; Cho, M.S.; Sheth, R.A.; Sood, A.K.; Afshar-Kharghan, V.
Podoplanin promotes tumor growth, platelet aggregation, and venous thrombosis in murine models of ovarian cancer. J. Thromb.
Haemost. 2021,20, 104–114. [CrossRef]
126.
Armulik, A.; Genové, G.; Betsholtz, C. Pericytes: Developmental, Physiological, and Pathological Perspectives, Problems, and
Promises. Dev. Cell 2011,21, 193–215. [CrossRef]
127.
Zonneville, J.; Safina, A.; Truskinovsky, A.M.; Arteaga, C.L.; Bakin, A.V. TGF-
β
signaling promotes tumor vasculature by
enhancing the pericyte-endothelium association. BMC Cancer 2018,18, 670. [CrossRef]
128.
Zhang, L.; Yang, N.; Park, J.-W.; Katsaros, D.; Fracchioli, S.; Cao, G.; O’Brien-Jenkins, A.; Randall, T.C.; Rubin, S.C.; Coukos,
G. Tumor-derived vascular endothelial growth factor up-regulates angiopoietin-2 in host endothelium and destabilizes host
vasculature, supporting angiogenesis in ovarian cancer. Cancer Res. 2003,63, 3403–3412.
129.
Sallinen, H.; Anttila, M.; Gröhn, O.; Koponen, J.; Hämäläinen, K.; Kholova, I.; Kosma, V.-M.; Heinonen, S.; Alitalo, K.; Ylä-
Herttuala, S. Cotargeting of VEGFR-1 and -3 and angiopoietin receptor Tie2 reduces the growth of solid human ovarian cancer in
mice. Cancer Gene Ther. 2010,18, 100–109. [CrossRef]
130.
Heldin, C.-H.; Westermark, B. Mechanism of Action and In Vivo Role of Platelet-Derived Growth Factor. Physiol. Rev.
1999
,79,
1283–1316. [CrossRef]
131.
Lu, C.; Shahzad, M.M.; Moreno-Smith, M.; Lin, Y.; Jennings, N.B.; Allen, J.K.; Landen, C.N.; Mangala, L.S.; Armaiz-Pena, G.N.;
Schmandt, R.; et al. Targeting pericytes with a PDGF-B aptamer in human ovarian carcinoma models. Cancer Biol. Ther.
2010
,9,
176–182. [CrossRef] [PubMed]
132.
Taraboletti, G.; Roberts, D.; Liotta, L.A.; Giavazzi, R. Platelet thrombospondin modulates endothelial cell adhesion, motility, and
growth: A potential angiogenesis regulatory factor. J. Cell Biol. 1990,111, 765–772. [CrossRef]
133.
Bagavandoss, P.; Wilks, J. Specific inhibition of endothelial cell proliferation by thrombospondin. Biochem. Biophys. Res. Commun.
1990,170, 867–872. [CrossRef]
134.
Matuszewska, K.; Kortenaar, S.T.; Pereira, M.; Santry, L.A.; Petrik, D.; Lo, K.-M.; Bridle, B.W.; Wootton, S.K.; Lawler, J.; Petrik,
J. Addition of an Fc-IgG induces receptor clustering and increases the
in vitro
efficacy and
in vivo
anti-tumor properties of
the thrombospondin-1 type I repeats (3TSR) in a mouse model of advanced stage ovarian cancer. Gynecol. Oncol.
2021
,164,
154–169. [CrossRef] [PubMed]
135.
Russell, S.; Duquette, M.; Liu, J.; Drapkin, R.; Lawler, J.; Petrik, J. Combined therapy with thrombospondin-1 type I repeats (3TSR)
and chemotherapy induces regression and significandy improves survival in a preclinical model of advanced stage epithelial
ovarian cancer. FASEB J. 2014,29, 576–588. [CrossRef]
136.
Matuszewska, K.; Santry, L.A.; van Vloten, J.P.; Auyeung, A.W.K.; Major, P.P.; Lawler, J.; Wootton, S.K.; Bridle, B.W.; Petrik, J.
Combining Vascular Normalization with an Oncolytic Virus Enhances Immunotherapy in a Preclinical Model of Advanced-Stage
Ovarian Cancer. Clin. Cancer Res. 2019,25, 1624–1638. [CrossRef] [PubMed]
137.
Boucharaba, A.; Serre, C.-M.; Grès, S.; Saulnier-Blache, J.S.; Bordet, J.-C.; Guglielmi, J.; Clézardin, P.; Peyruchaud, O. Platelet-
derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J. Clin. Investig.
2004
,114,
1714–1725. [CrossRef]
138.
Gopinathan, G.; Milagre, C.; Pearce, O.M.; Reynolds, L.E.; Hodivala-Dilke, K.; Leinster, D.A.; Zhong, H.; Hollingsworth,
R.E.; Thompson, R.; Whiteford, J.R.; et al. Interleukin-6 Stimulates Defective Angiogenesis. Cancer Res.
2015
,75,
3098–3107. [CrossRef] [PubMed]
139.
Merritt, W.M.; Nick, A.M.; Carroll, A.R.; Lu, C.; Matsuo, K.; Dumble, M.; Jennings, N.; Zhang, S.; Lin, Y.G.; Spannuth, W.A.; et al.
Bridging the Gap between Cytotoxic and Biologic Therapy with Metronomic Topotecan and Pazopanib in Ovarian Cancer. Mol.
Cancer Ther. 2010,9, 985–995. [CrossRef]
140.
Tullemans, B.M.E.; Nagy, M.; Sabrkhany, S.; Griffioen, A.W.; Egbrink, M.G.A.O.; Aarts, M.; Heemskerk, J.W.M.; Kuijpers, M.J.E.
Tyrosine Kinase Inhibitor Pazopanib Inhibits Platelet Procoagulant Activity in Renal Cell Carcinoma Patients. Front. Cardiovasc.
Med. 2018,5, 142. [CrossRef]
141.
Tracy, L.E.; Minasian, R.A.; Caterson, E. Extracellular Matrix and Dermal Fibroblast Function in the Healing Wound. Adv. Wound
Care 2016,5, 119–136. [CrossRef] [PubMed]
142.
Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.;
Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer
2020
,20,
174–186. [CrossRef] [PubMed]
Cancers 2023,15, 1282 27 of 37
143. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016,16, 582–598. [CrossRef] [PubMed]
144.
Luga, V.; Zhang, L.; Viloria-Petit, A.M.; Ogunjimi, A.A.; Inanlou, M.R.; Chiu, E.; Buchanan, M.; Hosein, A.N.; Basik, M.; Wrana,
J.L. Exosomes Mediate Stromal Mobilization of Autocrine Wnt-PCP Signaling in Breast Cancer Cell Migration. Cell
2012
,151,
1542–1556. [CrossRef]
145.
Hu, Y.; Yan, C.; Mu, L.; Huang, K.; Li, X.; Tao, D.; Wu, Y.; Qin, J. Fibroblast-derived exosomes contribute to chemoresistance
through priming cancer stem cells in colorectal cancer. PLoS ONE 2015,10, e0125625. [CrossRef]
146.
Ronca, R.; van Ginderachter, J.; Turtoi, A. Paracrine interactions of cancer-associated fibroblasts, macrophages and endothelial
cells: Tumor allies and foes. Curr. Opin. Oncol. 2018,30, 45–53. [CrossRef]
147.
Kojima, Y.; Acar, A.; Eaton, E.N.; Mellody, K.T.; Scheel, C.; Ben-Porath, I.; Onder, T.T.; Wang, Z.C.; Richardson, A.L.; Weinberg,
R.A.; et al. Autocrine TGF-
β
and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting
mammary stromal myofibroblasts. Proc. Natl. Acad. Sci. USA 2010,107, 20009–20014. [CrossRef]
148.
Anderberg, C.; Li, H.; Fredriksson, L.; Andrae, J.; Betsholtz, C.; Li, X.; Eriksson, U.; Pietras, K. Paracrine Signaling by Platelet-
Derived Growth Factor-CC Promotes Tumor Growth by Recruitment of Cancer-Associated Fibroblasts. Cancer Res.
2009
,69,
369–378. [CrossRef]
149.
Aoto, K.; Ito, K.; Aoki, S. Complex formation between platelet-derived growth factor receptor
β
and transforming growth
factor
β
receptor regulates the differentiation of mesenchymal stem cells into cancer-associated fibroblasts. Oncotarget
2018
,9,
34090–34102. [CrossRef]
150.
Zeltz, C.; Alam, J.; Liu, H.; Erusappan, P.M.; Hoschuetzky, H.; Molven, A.; Parajuli, H.; Cukierman, E.; Costea, D.-E.; Lu, N.; et al.
α
11
β
1 Integrin is Induced in a Subset of Cancer-Associated Fibroblasts in Desmoplastic Tumor Stroma and Mediates In Vitro Cell
Migration. Cancers 2019,11, 765. [CrossRef]
151.
Fukagawa, D.; Sugai, T.; Osakabe, M.; Suga, Y.; Nagasawa, T.; Itamochi, H.; Sugiyama, T. Protein expression patterns in
cancer-associated fibroblasts and cells undergoing the epithelial-mesenchymal transition in ovarian cancers. Oncotarget
2018
,9,
27514–27524. [CrossRef]
152.
Radhakrishnan, R.; Ha, J.H.; Jayaraman, M.; Liu, J.; Moxley, K.M.; Isidoro, C.; Sood, A.K.; Song, Y.S.; Dhanasekaran, D.N. Ovarian
cancer cell-derived lysophosphatidic acid induces glycolytic shift and cancer-associated fibroblast-phenotype in normal and
peritumoral fibroblasts. Cancer Lett. 2018,442, 464–474. [CrossRef] [PubMed]
153.
Rother, E.; Brandl, R.; Baker, D.L.; Goyal, P.; Gebhard, H.; Tigyi, G.; Siess, W. Subtype-Selective Antagonists of Lysophospha-
tidic Acid Receptors Inhibit Platelet Activation Triggered by the Lipid Core of Atherosclerotic Plaques. Circulation
2003
,108,
741–747. [CrossRef] [PubMed]
154.
Jeon, E.S.; Heo, S.C.; Lee, I.H.; Choi, Y.J.; Park, J.H.; Choi, K.U.; Park, Y.; Suh, D.-S.; Yoon, M.-S.; Kim, J.H. Ovarian cancer-derived
lysophosphatidic acid stimulates secretion of VEGF and stromal cell-derived factor-1
α
from human mesenchymal stem cells. Exp.
Mol. Med. 2010,42, 280–293. [CrossRef] [PubMed]
155.
Chignard, M.; Le Couedic, J.P.; Tence, M.; Vargaftig, B.B.; Benveniste, J. The role of platelet-activating factor in platelet aggregation.
Nature 1979,279, 799–800. [CrossRef] [PubMed]
156.
Cho, J.A.; Park, H.; Lim, E.H.; Kim, K.H.; Choi, J.S.; Lee, J.H.; Shin, J.W.; Lee, K.W. Exosomes from ovarian cancer cells induce
adipose tissue-derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor-supporting
myofibroblasts. Gynecol. Oncol. 2011,123, 379–386. [CrossRef] [PubMed]
157.
So, K.A.; Min, K.J.; Hong, J.H.; Lee, J.-K. Interleukin-6 expression by interactions between gynecologic cancer cells and human
mesenchymal stem cells promotes epithelial-mesenchymal transition. Int. J. Oncol. 2015,47, 1451–1459. [CrossRef]
158.
Gastl, G.; Plante, M.; Finstad, C.L.; Wong, G.Y.; Federici, M.G.; Bander, N.H.; Rubin, S.C. High IL-6 levels in ascitic fluid correlate
with reactive thrombocytosis in patients with epithelial ovarian cancer. Br. J. Haematol. 1993,83, 433–441. [CrossRef]
159.
Shield, K.; Ackland, M.L.; Ahmed, N.; Rice, G.E. Multicellular spheroids in ovarian cancer metastases: Biology and pathology.
Gynecol. Oncol. 2009,113, 143–148. [CrossRef]
160.
Rynne-Vidal, A.; Au-Yeung, C.L.; Jiménez-Heffernan, J.A.; Perez-Lozano, M.-L.; Cremades-Jimeno, L.; Bárcena, C.; Cristobal, I.;
Fernández-Chacón, C.; Yeung, T.L.; Mok, S.C.; et al. Mesothelial-to-mesenchymal transition as a possible therapeutic target in
peritoneal metastasis of ovarian cancer. J. Pathol. 2017,242, 140–151. [CrossRef]
161.
Matte, I.; Lane, D.; Laplante, C.; Rancourt, C.; Piché, A. Profiling of cytokines in human epithelial ovarian cancer ascites. Am. J.
Cancer Res. 2012,2, 566–580. [PubMed]
162.
Yan, D.; Liu, X.; Xu, H.; Guo, S.-W. Mesothelial Cells Participate in Endometriosis Fibrogenesis through Platelet-Induced
Mesothelial-Mesenchymal Transition. J. Clin. Endocrinol. Metab. 2020,105, e4124–e4147. [CrossRef] [PubMed]
163.
Steidel, C.; Ender, F.; Rody, A.; von Bubnoff, N.; Gieseler, F. Biologically Active Tissue Factor-Bearing Larger Ectosome-Like
Extracellular Vesicles in Malignant Effusions from Ovarian Cancer Patients: Correlation with Incidence of Thrombosis. Int. J. Mol.
Sci. 2021,22, 790. [CrossRef]
164.
Orellana, R.; Kato, S.; Erices, R.; Bravo, M.L.; Gonzalez, P.; Oliva, B.; Cubillos, S.; Valdivia, A.; Ibañez, C.; Brañes, J.; et al. Platelets
enhance tissue factor protein and metastasis initiating cell markers, and act as chemoattractants increasing the migration of
ovarian cancer cells. BMC Cancer 2015,15, 290. [CrossRef]
165.
Malacrida, B.; Nichols, S.; Maniati, E.; Jones, R.; Delanie-Smith, R.; Roozitalab, R.; Tyler, E.J.; Thomas, M.; Boot, G.; Mackerodt, J.;
et al. A human multi-cellular model shows how platelets drive production of diseased extracellular matrix and tissue invasion.
Iscience 2021,24, 102676. [CrossRef] [PubMed]
Cancers 2023,15, 1282 28 of 37
166.
Motohara, T.; Masuda, K.; Morotti, M.; Zheng, Y.; El-Sahhar, S.; Chong, K.Y.; Wietek, N.; Alsaadi, A.; Carrami, E.M.; Hu, Z.; et al.
An evolving story of the metastatic voyage of ovarian cancer cells: Cellular and molecular orchestration of the adipose-rich
metastatic microenvironment. Oncogene 2018,38, 2885–2898. [CrossRef] [PubMed]
167.
Ryner, L.; Guan, Y.; Firestein, R.; Xiao, Y.; Choi, Y.; Rabe, C.; Lu, S.; Fuentes, E.; Huw, L.-Y.; Lackner, M.R.; et al. Upregulation of
Periostin and Reactive Stroma Is Associated with Primary Chemoresistance and Predicts Clinical Outcomes in Epithelial Ovarian
Cancer. Clin. Cancer Res. 2015,21, 2941–2951. [CrossRef]
168.
Moran-Jones, K.; Gloss, B.S.; Murali, R.; Chang, D.K.; Colvin, E.K.; Jones, M.D.; Yuen, S.; Howell, V.M.; Brown, L.M.; Wong,
C.W.; et al. Connective tissue growth factor as a novel therapeutic target in high grade serous ovarian cancer. Oncotarget
2015
,6,
44551–44562. [CrossRef]
169.
Yeung, T.-L.; Leung, C.S.; Wong, K.-K.; Samimi, G.; Thompson, M.S.; Liu, J.; Zaid, T.M.; Ghosh, S.; Birrer, M.J.; Mok, S.C. TGF-
β
Modulates Ovarian Cancer Invasion by Upregulating CAF-Derived Versican in the Tumor Microenvironment. Cancer Res.
2013
,
73, 5016–5028. [CrossRef]
170.
Ghoneum, A.; Afify, H.; Salih, Z.; Kelly, M.; Said, N. Role of tumor microenvironment in ovarian cancer pathobiology. Oncotarget
2018,9, 22832–22849. [CrossRef]
171.
Crawford, Y.; Kasman, I.; Yu, L.; Zhong, C.; Wu, X.; Modrusan, Z.; Kaminker, J.; Ferrara, N. PDGF-C Mediates the Angiogenic
and Tumorigenic Properties of Fibroblasts Associated with Tumors Refractory to Anti-VEGF Treatment. Cancer Cell
2009
,15,
21–34. [CrossRef] [PubMed]
172.
Wawro, M.E.; Sobierajska, K.; Ciszewski, W.M.; Niewiarowska, J. Nonsteroidal Anti-Inflammatory Drugs Prevent Vincristine-
Dependent Cancer-Associated Fibroblasts Formation. Int. J. Mol. Sci. 2019,20, 1941. [CrossRef] [PubMed]
173.
Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor Associated Neutrophils. Their Role in Tumorigenesis, Metastasis, Prognosis
and Therapy. Front. Oncol. 2019,9, 1146. [CrossRef] [PubMed]
174.
Emmons, T.R.; Giridharan, T.; Singel, K.L.; Khan, A.N.H.; Ricciuti, J.; Howard, K.; Toro, S.L.S.-D.; Debreceni, I.L.; Aarts, C.E.;
Brouwer, M.C.; et al. Mechanisms Driving Neutrophil-Induced T-cell Immunoparalysis in Ovarian Cancer. Cancer Immunol. Res.
2021,9, 790–810. [CrossRef]
175.
Shang, A.; Wang, W.; Gu, C.; Chen, C.; Zeng, B.; Yang, Y.; Ji, P.; Sun, J.; Wu, J.; Lu, W.; et al. Long non-coding RNA HOTTIP
enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in
neutrophils. J. Exp. Clin. Cancer Res. 2019,38, 1–15. [CrossRef] [PubMed]
176.
Hanabuchi, S.; Watanabe, N.; Wang, Y.-H.; Wang, Y.-H.; Ito, T.; Shaw, J.; Cao, W.; Qin, F.X.-F.; Liu, Y.-J. Human plasmacytoid
predendritic cells activate NK cells through glucocorticoid-induced tumor necrosis factor receptor-ligand (GITRL). Blood
2006
,
107, 3617–3623. [CrossRef]
177.
Lines, J.L.; Pantazi, E.; Mak, J.; Sempere, L.F.; Wang, L.; O’Connell, S.; Ceeraz, S.; Suriawinata, A.A.; Yan, S.; Ernstoff, M.S.; et al.
VISTA Is an Immune Checkpoint Molecule for Human T Cells. Cancer Res. 2014,74, 1924–1932. [CrossRef]
178.
Zhou, M.; Li, L.; Wang, X.; Wang, C.; Wang, D. Neutrophil-to-Lymphocyte Ratio and Platelet Count Predict Long-Term Outcome
of Stage IIIC Epithelial Ovarian Cancer. Cell. Physiol. Biochem. 2018,46, 178–186. [CrossRef]
179. Komura, N.; Mabuchi, S.; Yokoi, E.; Kozasa, K.; Kuroda, H.; Sasano, T.; Matsumoto, Y.; Kimura, T. Comparison of clinical utility
between neutrophil count and neutrophil–lymphocyte ratio in patients with ovarian cancer: A single institutional experience and
a literature review. Int. J. Clin. Oncol. 2017,23, 104–113. [CrossRef]
180.
Williams, M.R.; Azcutia, V.; Newton, G.; Alcaide, P.; Luscinskas, F.W. Emerging mechanisms of neutrophil recruitment across
endothelium. Trends Immunol. 2011,32, 461–469. [CrossRef]
181.
Larsen, E.; Celi, A.; Gilbert, G.E.; Furie, B.C.; Erban, J.K.; Bonfanti, R.; Wagner, D.D.; Furie, B. PADGEM protein: A receptor that
mediates the interaction of activated platelets with neutrophils and monocytes. Cell 1989,59, 305–312. [CrossRef] [PubMed]
182.
Simon, D.I.; Chen, Z.; Xu, H.; Li, C.Q.; Dong, J.-F.; McIntire, L.V.; Ballantyne, C.M.; Zhang, L.; Furman, M.I.; Berndt, M.C.;
et al. Platelet Glycoprotein Ib
α
Is a Counterreceptor for the Leukocyte Integrin Mac-1 (Cd11b/Cd18). J. Exp. Med.
2000
,192,
193–204. [CrossRef] [PubMed]
183.
Kuijper, P.H.M.; Torres, H.I.G.; Lammers, J.-W.J.; Sixma, J.J.; Koenderman, L.; Zwaginga, J.J. Platelet Associated Fibrinogen and
ICAM-2 Induce Firm Adhesion of Neutrophils under Flow Conditions. Thromb. Haemost.
1998
,80, 443–448. [CrossRef] [PubMed]
184.
Bednarska, K.; Klink, M.; Wilczy´nski, J.R.; Szyłło, K.; Malinowski, A.; Sułowska, Z.; Nowak, M. Heterogeneity of the Mac-1
expression on peripheral blood neutrophils in patients with different types of epithelial ovarian cancer. Immunobiology
2016
,221,
323–332. [CrossRef]
185.
Klink, M.; Jastrzembska, K.; Nowak, M.; Bednarska, K.; Szpakowski, M.; Szyllo, K.; Sulowska, Z. Ovarian Cancer Cells Modulate
Human Blood Neutrophils Response to Activation In Vitro. Scand. J. Immunol. 2008,68, 328–336. [CrossRef]
186.
Stadtmann, A.; Germena, G.; Block, H.; Boras, M.; Rossaint, J.; Sundd, P.; Lefort, C.; Fisher, C.I.; Buscher, K.; Gelschefarth, B.; et al.
The PSGL-1–L-selectin signaling complex regulates neutrophil adhesion under flow. J. Exp. Med.
2013
,210, 2171–2180. [CrossRef]
187.
Di Wu, Q.; Wang, J.H.; Condron, C.; Bouchier-Hayes, D.; Redmond, H.P. Human neutrophils facilitate tumor cell transendothelial
migration. Am. J. Physiol. Physiol. 2001,280, C814–C822. [CrossRef]
188.
Vanichakarn, P.; Blair, P.; Wu, C.; Freedman, J.E.; Chakrabarti, S. Neutrophil CD40 enhances platelet-mediated inflammation.
Thromb. Res. 2008,122, 346–358. [CrossRef]
Cancers 2023,15, 1282 29 of 37
189.
Frenette, P.S.; Denis, C.V.; Weiss, L.; Jurk, K.; Subbarao, S.; Kehrel, B.; Hartwig, J.H.; Vestweber, D.; Wagner, D.D. P-Selectin
Glycoprotein Ligand 1 (Psgl-1) Is Expressed on Platelets and Can Mediate Platelet–Endothelial Interactions in Vivo. J. Exp. Med.
2000,191, 1413–1422. [CrossRef]
190.
Weber, C.; Springer, A.T. Neutrophil accumulation on activated, surface-adherent platelets in flow is mediated by interaction
of Mac-1 with fibrinogen bound to alphaIIbbeta3 and stimulated by platelet-activating factor. J. Clin. Investig.
1997
,100,
2085–2093. [CrossRef]
191.
Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated
neutrophil pheno-type by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009,16, 183–194. [CrossRef] [PubMed]
192.
Yang, M.; Zhang, G.; Wang, Y.; He, M.; Xu, Q.; Lu, J.; Liu, H.; Xu, C. Tumour-associated neutrophils orchestrate intratumoural
IL-8-driven immune evasion through Jagged2 activation in ovarian cancer. Br. J. Cancer 2020,123, 1404–1416. [CrossRef]
193.
Singh, A.; Bisht, P.; Bhattacharya, S.; Guchhait, P. Role of Platelet Cytokines in Dengue Virus Infection. Front. Cell. Infect. Microbiol.
2020,10, 561366. [CrossRef] [PubMed]
194.
Johnson, K.E.; Ceglowski, J.R.; Roweth, H.G.; Forward, J.A.; Tippy, M.D.; El-Husayni, S.; Kulenthirarajan, R.; Malloy, M.W.;
Machlus, K.R.; Chen, W.Y.; et al. Aspirin inhibits platelets from reprogramming breast tumor cells and promoting metastasis.
Blood Adv. 2019,3, 198–211. [CrossRef]
195.
Micheli, D.C.; Jammal, M.P.; Martins-Filho, A.; Côrtes, J.R.X.D.M.; de Souza, C.N.; Nomelini, R.S.; Murta, E.F.C.; Tavares-Murta,
B.M. Serum cytokines and CXCR2: Potential tumour markers in ovarian neoplasms. Biomarkers 2020,25, 474–482. [CrossRef]
196.
Rodrigues, I.S.S.; Martins-Filho, A.; Micheli, D.C.; de Lima, C.A.; Tavares-Murta, B.M.; Murta, E.F.C.; Nomelini, R.S. IL-6 and IL-8
as Prognostic Factors in Peritoneal Fluid of Ovarian Cancer. Immunol. Investig. 2019,49, 510–521. [CrossRef]
197.
Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer
2016
,16,
431–446. [CrossRef] [PubMed]
198.
Metelli, A.; Wu, B.X.; Riesenberg, B.; Guglietta, S.; Huck, J.D.; Mills, C.; Li, A.; Rachidi, S.; Krieg, C.; Rubinstein, M.P.; et al.
Thrombin contributes to cancer immune evasion via proteolysis of platelet-bound GARP to activate LTGF-
β
.Sci. Transl. Med.
2020,12, eaay4860. [CrossRef] [PubMed]
199.
Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil
extracellular traps kill bacteria. Science 2004,303, 1532–1535. [CrossRef] [PubMed]
200. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2017,18, 134–147. [CrossRef]
201.
Elaskalani, O.; Abdol Razak, N.B.; Metharom, P. Neutrophil extracellular traps induce aggregation of washed human platelets
independently of extracellular DNA and histones. Cell Commun. Signal. 2018,16, 24. [CrossRef]
202.
Etulain, J.; Martinod, K.; Wong, S.L.; Cifuni, S.M.; Schattner, M.; Wagner, D.D. P-selectin promotes neutrophil extracellular trap
formation in mice. Blood 2015,126, 242–246. [CrossRef] [PubMed]
203.
Mayer, C.; Darb-Esfahani, S.; Meyer, A.-S.; Hübner, K.; Rom, J.; Sohn, C.; Braicu, I.; Sehouli, J.; Hänsch, G.M.; Gaida, M.M.
Neutrophil Granulocytes in Ovarian Cancer—Induction of Epithelial-To-Mesenchymal-Transition and Tumor Cell Migration. J.
Cancer 2016,7, 546–554. [CrossRef]
204.
Si-Tahar, M.; Pidard, D.; Balloy, V.; Moniatte, M.; Kieffer, N.; van Dorsselaer, A.; Chignard, M. Human Neutrophil Elastase
Proteolytically Activates the Platelet Integrin
α
IIb
β
3 through Cleavage of the Carboxyl Terminus of the
α
IIb Subunit Heavy
Chain. J. Biol. Chem. 1997,272, 11636–11647. [CrossRef]
205.
Itagaki, K.; Kaczmarek, E.; Lee, Y.T.; Tang, I.T.; Isal, B.; Adibnia, Y.; Sandler, N.; Grimm, M.J.; Segal, B.H.; Otter-
bein, L.E.; et al. Mitochondrial DNA Released by Trauma Induces Neutrophil Extracellular Traps. PLoS ONE
2015
,10,
e0120549. [CrossRef] [PubMed]
206.
Demers, M.; Krause, D.S.; Schatzberg, D.; Martinod, K.; Voorhees, J.R.; Fuchs, T.A.; Scadden, D.T.; Wagner, D.D. Cancers
predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl. Acad. Sci.
USA 2012,109, 13076–13081. [CrossRef] [PubMed]
207. Lee, W.; Ko, S.Y.; Mohamed, M.S.; Kenny, H.A.; Lengyel, E.; Naora, H. Neutrophils facilitate ovarian cancer premetastatic niche
formation in the omentum. J. Exp. Med. 2018,216, 176–194. [CrossRef] [PubMed]
208.
Singel, K.; Grzankowski, K.S.; Khan, A.N.M.N.H.; Grimm, M.J.; D’Auria, A.C.; Morrell, K.; Eng, K.H.; Hylander, B.; Mayor, P.C.;
Emmons, T.R.; et al. Mitochondrial DNA in the tumour microenvironment activates neutrophils and is associated with worse
outcomes in patients with advanced epithelial ovarian cancer. Br. J. Cancer 2018,120, 207–217. [CrossRef]
209.
Zhang, T.; Ma, Z.; Wang, R.; Wang, Y.; Wang, S.; Cheng, Z.; Xu, H.; Jin, X.; Li, W.; Wang, X. Thrombin facilitates invasion of
ovarian cancer along peritoneum by inducing monocyte differentiation toward tumor-associated macrophage-like cells. Cancer
Immunol. Immunother. 2010,59, 1097–1108. [CrossRef]
210. Duan, Z.; Luo, Y. Targeting macrophages in cancer immunotherapy. Signal Transduct. Target. Ther. 2021,6, 127. [CrossRef]
211.
Reinartz, S.; Schumann, T.; Finkernagel, F.; Wortmann, A.; Jansen, J.M.; Meissner, W.; Krause, M.; Schwörer, A.; Wagner, U.;
Müller-Brüsselbach, S.; et al. Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma:
Correlation of CD163 expression, cytokine levels and early relapse. Int. J. Cancer 2013,134, 32–42. [CrossRef]
212.
Kawamura, K.; Komohara, Y.; Takaishi, K.; Katabuchi, H.; Takeya, M. Detection of M2 macrophages and colony-stimulating factor
1 expression in serous and mucinous ovarian epithelial tumors. Pathol. Int. 2009,59, 300–305. [CrossRef] [PubMed]
213.
Gupta, V.; Yull, F.; Khabele, D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers
2018
,
10, 366. [CrossRef] [PubMed]
Cancers 2023,15, 1282 30 of 37
214.
Yin, M.; Shen, J.; Yu, S.; Fei, J.; Zhu, X.; Zhao, J.; Zhai, L.; Sadhukhan, A.; Zhou, J. Tumor-Associated Macrophages (TAMs): A
Critical Activator in Ovarian Cancer Metastasis. OncoTargets Ther. 2019,12, 8687–8699. [CrossRef] [PubMed]
215.
Burleson, K.M.; Casey, R.C.; Skubitz, K.M.; Pambuccian, S.E.; Oegema, T.R., Jr.; Skubitz, A.P.N. Ovarian carcinoma as-
cites spheroids adhere to extracellular matrix components and mesothelial cell monolayers. Gynecol. Oncol.
2004
,93,
170–181. [CrossRef]
216.
Long, L.; Hu, Y.; Long, T.; Lu, X.; Tuo, Y.; Li, Y.; Wang, M.; Ke, Z. Tumor-associated macrophages induced spheroid formation
by CCL18-ZEB1-M-CSF feedback loop to promote transcoelomic metastasis of ovarian cancer. J. Immunother. Cancer
2021
,9,
e003973. [CrossRef]
217.
Zhang, M.; He, Y.; Sun, X.; Li, Q.; Wang, W.; Zhao, A.; Di, W. A high M1/M2 ratio of tumor-associated macrophages is associated
with extended survival in ovarian cancer patients. J. Ovarian Res. 2014,7, 19. [CrossRef]
218.
Michelson, A.D.; Barnard, M.R.; Krueger, L.A.; Valeri, C.R.; Furman, M.I. Circulating Monocyte-Platelet Aggregates Are a More
Sensitive Marker of In Vivo Platelet Activation Than Platelet Surface P-Selectin. Circulation 2001,104, 1533–1537. [CrossRef]
219.
Duque, G.A.; Descoteaux, A. Macrophage Cytokines: Involvement in Immunity and Infectious Diseases. Front. Immunol.
2014
,5,
491. [CrossRef]
220.
Nagata, K.; Tsuji, T.; Todoroki, N.; Katagiri, Y.; Tanoue, K.; Yamazaki, H.; Hanai, N.; Irimura, T. Activated platelets induce
superoxide anion release by monocytes and neutrophils through P-selectin (CD62). J. Immunol.
1993
,151, 3267–3273. [CrossRef]
221.
Griess, B.; Mir, S.; Datta, K.; Teoh-Fitzgerald, M. Scavenging reactive oxygen species selectively inhibits M2 macrophage
polarization and their pro-tumorigenic function in part, via Stat3 suppression. Free Radic. Biol. Med.
2019
,147,
48–60. [CrossRef] [PubMed]
222.
Lee, S.J.; Yoon, B.R.; Kim, H.Y.; Yoo, S.-J.; Kang, S.W.; Lee, W.-W. Activated Platelets Convert CD14
+
CD16
-
Into CD14
+
CD16
+
Monocytes with Enhanced Fc
γ
R-Mediated Phagocytosis and Skewed M2 Polarization. Front. Immunol.
2021
,11,
611133. [CrossRef]
223.
Reader, J.; Holt, D.; Fulton, A. Prostaglandin E2 EP receptors as therapeutic targets in breast cancer. Cancer Metastasis Rev.
2011
,
30, 449–463. [CrossRef] [PubMed]
224.
Klimp, A.H.; Hollema, H.; Kempinga, C.; van der Zee, A.G.; de Vries, E.G.; Daemen, T. Expression of cyclooxygenase-2
and inducible nitric oxide synthase in human ovarian tumors and tumor-associated macrophages. Cancer Res.
2001
,61,
7305–7309. [PubMed]
225.
Dijkgraaf, E.M.; Heusinkveld, M.; Tummers, B.; Vogelpoel, L.T.; Goedemans, R.; Jha, V.; Nortier, J.W.; Welters, M.J.; Kroep, J.R.;
van der Burg, S.H. Chemotherapy Alters Monocyte Differentiation to Favor Generation of Cancer-Supporting M2 Macrophages
in the Tumor Microenvironment. Cancer Res. 2013,73, 2480–2492. [CrossRef]
226.
Mussbacher, M.; Salzmann, M.; Brostjan, C.; Hoesel, B.; Schoergenhofer, C.; Datler, H.; Hohensinner, P.; Basílio, J.; Petzelbauer,
P.; Assinger, A.; et al. Cell Type-Specific Roles of NF-
κ
B Linking Inflammation and Thrombosis. Front. Immunol.
2019
,
10, 85. [CrossRef]
227.
Suzuki-Inoue, K. Platelets and cancer-associated thrombosis: Focusing on the platelet activation receptor CLEC-2 and podoplanin.
Blood 2019,134, 1912–1918. [CrossRef]
228.
Laffont, B.; Corduan, A.; Rousseau, M.; Duchez, A.C.; Lee, C.H.C.; Boilard, E.; Provost, P. Platelet microparticles reprogram
macrophage gene expression and function. Thromb. Haemost. 2016,115, 311–323. [CrossRef]
229.
Olingy, C.E.; Dinh, H.Q.; Hedrick, C.C. Monocyte heterogeneity and functions in cancer. J. Leukoc. Biol.
2019
,106,
309–322. [CrossRef]
230.
Deng, C.; Wu, S.; Zhang, L.; Yang, M.; Lin, Q.; Xie, Q.; Ding, H.; Lian, N.; Gao, S.; Huang, Y.; et al. Role of monocyte tissue factor
on patients with non-small cell lung cancer. Clin. Respir. J. 2017,12, 1125–1133. [CrossRef]
231.
Hell, L.; Däullary, T.; Burghart, V.; Mauracher, L.-M.; Grilz, E.; Moser, B.; Kramer, G.; Schmid, J.; Ay, C.; Pabinger, I.; et al.
Extracellular Vesicle-Associated Tissue Factor Activity in Prostate Cancer Patients with Disseminated Intravascular Coagulation.
Cancers 2021,13, 1487. [CrossRef] [PubMed]
232.
Zhang, C.; Yang, Z.; Zhou, P.; Yu, M.; Li, B.; Liu, Y.; Jin, J.; Liu, W.; Jing, H.; Du, J.; et al. Phosphatidylserine-exposing tumor-derived
microparticles exacerbate coagulation and cancer cell transendothelial migration in triple-negative breast cancer. Theranostics
2021,11, 6445–6460. [CrossRef] [PubMed]
233.
Montoro-García, S.; Shantsila, E.; Hernández-Romero, D.; Jover, E.; Valdés, M.; Marin, F.; Lip, G.Y.H. Small-size platelet
microparticles trigger platelet and monocyte functionality and modulate thrombogenesis via P-selectin. Br. J. Haematol.
2014
,166,
571–580. [CrossRef] [PubMed]
234.
Vasina, E.M.; Cauwenberghs, S.; Feijge, M.A.H.; Heemskerk, J.W.M.; Weber, C.; Koenen, R.R. Microparticles from apoptotic
platelets promote resident macrophage differentiation. Cell Death Dis. 2011,2, e211. [CrossRef] [PubMed]
235.
Ádány, R.; Kappelmayer, J.; Berényi, E.; Szegedi, A.; Fábián, E.; Muszbek, L. Factors of the Extrinsic Pathway of Blood Coagulation
in Tumour Associated Macrophages. Thromb. Haemost. 1989,62, 850–855. [CrossRef]
236.
Ruf, W.; Mueller, B. Thrombin Generation and the Pathogenesis of Cancer. Semin. Thromb. Hemost.
2006
,32, 061–068. [CrossRef]
237.
Chanakira, A.; Westmark, P.R.; Ong, I.M.; Sheehan, J.P. Tissue factor-factor VIIa complex triggers protease activated receptor
2-dependent growth factor release and migration in ovarian cancer. Gynecol. Oncol. 2017,145, 167–175. [CrossRef]
Cancers 2023,15, 1282 31 of 37
238.
Gil-Bernabé, A.M.; Ferjanˇciˇc, Š.; Tlalka, M.; Zhao, L.; Allen, P.D.; Im, J.H.; Watson, K.; Hill, S.A.; Amirkhosravi, A.; Francis, J.L.;
et al. Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and
premetastatic niche establishment in mice. Blood 2012,119, 3164–3175. [CrossRef]
239.
Läubli, H.; Spanaus, K.-S.; Borsig, L. Selectin-mediated activation of endothelial cells induces expression of CCL5 and promotes
metastasis through recruitment of monocytes. Blood 2009,114, 4583–4591. [CrossRef]
240.
Ratajczak, M.Z.; Zuba-Surma, E.; Kucia, M.; Reca, R.; Wojakowski, W.; Ratajczak, J. The pleiotropic effects of the SDF-1–CXCR4
axis in organogenesis, regeneration and tumorigenesis. Leukemia 2006,20, 1915–1924. [CrossRef]
241.
Sánchez-Martín, L.; Estecha, A.; Samaniego, R.; Sánchez-Ramón, S.; Vega, M.Á.; Sánchez-Mateos, P. The chemokine CXCL12
regulates monocyte-macrophage differentiation and RUNX3 expression. Blood 2011,117, 88–97. [CrossRef] [PubMed]
242.
Zeng, Y.; Li, B.; Liang, Y.; Reeves, P.M.; Qu, X.; Ran, C.; Liu, Q.; Callahan, M.V.; Sluder, A.E.; Gelfand, J.A.; et al. Dual
blockade of CXCL12-CXCR4 and PD-1–PD-L1 pathways prolongs survival of ovarian tumor–bearing mice by prevention of
immunosuppression in the tumor microenvironment. FASEB J. 2019,33, 6596–6608. [CrossRef]
243.
Junior, I.A.D.S.; Stone, S.C.; Rossetti, R.M.; Jancar, S.; Lepique, A.P. Modulation of Tumor-Associated Macrophages (TAM)
Phenotype by Platelet-Activating Factor (PAF) Receptor. J. Immunol. Res. 2017,2017, 5482768. [CrossRef]
244.
Deuster, E.; Hysenaj, I.; Kahaly, M.; Schmoeckel, E.; Mayr, D.; Beyer, S.; Kolben, T.; Hester, A.; Kraus, F.; Chelariu-Raicu, A.;
et al. The Platelet-Activating Factor Receptor’s Association with the Outcome of Ovarian Cancer Patients and Its Experimental
Inhibition by Rupatadine. Cells 2021,10, 2337. [CrossRef]
245.
Spiel, A.; Bartko, J.; Schwameis, M.; Firbas, C.; Siller-Matula, J.M.; Schuetz, M.; Weigl, M.; Jilma, B. Increased platelet aggre-
gation and
in vivo
platelet activation after granulocyte colony-stimulating factor administration. Thromb. Haemost.
2011
,105,
655–662. [CrossRef]
246.
Zeng, X.-Y.; Xie, H.; Yuan, J.; Jiang, X.-Y.; Yong, J.-H.; Zeng, D.; Dou, Y.-Y.; Xiao, S.-S. M2-like tumor-associated macrophages-
secreted EGF promotes epithelial ovarian cancer metastasis via activating EGFR-ERK signaling and suppressing lncRNA LIMT
expression. Cancer Biol. Ther. 2019,20, 956–966. [CrossRef]
247.
Yu, Y.; Zhang, M.; Zhang, X.; Cai, Q.; Hong, S.; Jiang, W.; Xu, C. Synergistic effects of combined platelet-activating factor receptor
and epidermal growth factor receptor targeting in ovarian cancer cells. J. Hematol. Oncol. 2014,7, 39. [CrossRef]
248.
Reinartz, S.; Lieber, S.; Pesek, J.; Brandt, D.T.; Asafova, A.; Finkernagel, F.; Watzer, B.; Nockher, W.A.; Nist, A.; Stiewe, T.; et al.
Cell type-selective pathways and clinical associations of lysophosphatidic acid biosynthesis and signaling in the ovarian cancer
microenvironment. Mol. Oncol. 2018,13, 185–201. [CrossRef]
249.
Leblanc, R.; Lee, S.-C.; David, M.; Bordet, J.-C.; Norman, D.D.; Patil, R.; Miller, D.; Sahay, D.; Ribeiro, J.; Clézardin, P.; et al.
Interaction of platelet-derived autotaxin with tumor integrin
α
V
β
3 controls metastasis of breast cancer cells to bone. Blood
2014
,
124, 3141–3150. [CrossRef] [PubMed]
250.
Feng, Y.; Xiao, M.; Zhang, Z.; Cui, R.; Jiang, X.; Wang, S.; Bai, H.; Liu, C.; Zhang, Z. Potential interaction between lysophosphatidic
acid and tumor-associated macrophages in ovarian carcinoma. J. Inflamm. 2020,17, 23. [CrossRef]
251.
Onallah, H.; Catane, L.J.; Tropé, C.G.; Falkenthal, T.E.H.; Reich, R.; Davidson, B. Activity and clinical relevance of autotaxin and
lysophosphatidic acid pathways in high-grade serous carcinoma. Virchows Arch. 2018,473, 463–470. [CrossRef]
252.
Khandoga, A.L.; Fujiwara, Y.; Goyal, P.; Pandey, D.; Tsukahara, R.; Bolen, A.; Guo, H.; Wilke, N.; Liu, J.; Valentine, W.J.; et al.
Lysophosphatidic acid-induced platelet shape change revealed through LPA
1–5
receptor-selective probes and albumin. Platelets
2008,19, 415–427. [CrossRef]
253.
Rogers, L.C.; Davis, R.R.; Said, N.; Hollis, T.; Daniel, L.W. Blocking LPA-dependent signaling increases ovarian cancer cell death
in response to chemotherapy. Redox Biol. 2018,15, 380–386. [CrossRef]
254.
Wang, W.; Zou, W.; Liu, J.R. Tumor-infiltrating T cells in epithelial ovarian cancer: Predictors of prognosis and biological basis of
immunotherapy. Gynecol. Oncol. 2018,151, 1–3. [CrossRef]
255.
Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger,
K.; Liebman, M.N.; et al. Intratumoral T Cells, Recurrence, and Survival in Epithelial Ovarian Cancer. N. Engl. J. Med.
2003
,348,
203–213. [CrossRef]
256.
Pinto, M.P.; Balmaceda, C.; Bravo, M.L.; Kato, S.; Villarroel, A.; Owen, G.I.; Roa, J.C.; Cuello, M.A.; Ibañez, C. Patient inflammatory
status and CD4
+
/CD8
+
intraepithelial tumor lymphocyte infiltration are predictors of outcomes in high-grade serous ovarian
cancer. Gynecol. Oncol. 2018,151, 10–17. [CrossRef]
257.
Starossom, S.C.; Veremeyko, T.; Yung, A.W.; Dukhinova, M.; Au, C.; Lau, A.Y.; Weiner, H.L.; Ponomarev, E.D. Platelets
Play Differential Role During the Initiation and Progression of Autoimmune Neuroinflammation. Circ. Res.
2015
,117,
779–792. [CrossRef]
258.
Gerdes, N.; Zhu, L.; Ersoy, M.; Hermansson, A.; Hjemdahl, P.; Hu, H.; Hansson, G.K.; Li, N. Platelets regulate CD4
+
T-cell
differentiation via multiple chemokines in humans. Thromb. Haemost. 2011,106, 353–362. [CrossRef]
259.
Eken, C.; Sadallah, S.; Amicarella, F.; Iezzi, G.; Schifferli, J.A. Ectosomes released by platelets induce differentiation of CD4
+
T
cells into T regulatory cells. Thromb. Haemost. 2014,112, 1219–1229. [CrossRef]
260.
Yu, H.; Hu, W.; Song, X.; Zhao, Y. Immune Modulation of Platelet-Derived Mitochondria on Memory CD4
+
T Cells in Humans.
Int. J. Mol. Sci. 2020,21, 6295. [CrossRef]
Cancers 2023,15, 1282 32 of 37
261.
Vitkovsky, Y.; Brill, G.; Koltakov, A.; Farzam, N.; Varon, D.; Bank, I.; Solpov, A.; Shenkman, B.; Savion, N. Platelets enhance CD4
+
lymphocyte adhesion to extracellular matrix under flow conditions: Role of platelet aggregation, integrins, and non-integrin
receptors. Thromb. Haemost. 2006,95, 815–821. [CrossRef]
262.
Shenkman, B.; Brill, G.; Solpov, A.; Vitkovsky, Y.; Kuznik, B.; Koltakov, A.; Kotev-Emeth, S.; Savion, N.; Bank, I. CD4
+
lymphocytes
require platelets for adhesion to immobilized fibronectin in flow: Role of
β
1 (CD29)-,
β
2 (CD18)-related integrins and non-integrin
receptors. Cell. Immunol. 2006,242, 52–59. [CrossRef] [PubMed]
263.
Danese, S.; de la Motte, C.; Reyes, B.M.R.; Sans, M.; Levine, A.D.; Fiocchi, C. Cutting Edge: T Cells Trigger CD40-Dependent
Platelet Activation and Granular RANTES Release: A Novel Pathway for Immune Response Amplification. J. Immunol.
2004
,172,
2011–2015. [CrossRef] [PubMed]
264.
Blair, P.J.; Riley, J.L.; Harlan, D.M.; Abe, R.; Tadaki, D.K.; Hoffmann, S.C.; White, L.; Francomano, T.; Perfetto, S.J.; Kirk, A.D.; et al.
Cd40 Ligand (Cd154) Triggers a Short-Term Cd4
+
T Cell Activation Response That Results in Secretion of Immunomodulatory
Cytokines and Apoptosis. J. Exp. Med. 2000,191, 651–660. [CrossRef]
265.
Salas-Perdomo, A.; Miró-Mur, F.; Urra, X.; Justicia, C.; Gallizioli, M.; Zhao, Y.; Brait, V.H.; Laredo, C.; Tudela, R.; Hidalgo, A.;
et al. T Cells Prevent Hemorrhagic Transformation in Ischemic Stroke by P-Selectin Binding. Arter. Thromb. Vasc. Biol.
2018
,38,
1761–1771. [CrossRef]
266.
Hirata, T.; Merrill-Skoloff, G.; Aab, M.; Yang, J.; Furie, B.C.; Furie, B. P-Selectin Glycoprotein Ligand 1 (Psgl-1) Is a Physiological
Ligand for E-Selectin in Mediating T Helper 1 Lymphocyte Migration. J. Exp. Med. 2000,192, 1669–1676. [CrossRef]
267.
Tinoco, R.; Carrette, F.; Barraza, M.L.; Otero, D.C.; Magaña, J.; Bosenberg, M.W.; Swain, S.L.; Bradley, L.M. PSGL-1 Is an Immune
Checkpoint Regulator that Promotes T Cell Exhaustion. Immunity 2016,44, 1190–1203. [CrossRef]
268.
Zaslavsky, A.B.; Adams, M.P.; Cao, X.; Maj, T.; Choi, J.E.; Stangl-Kremser, J.; Patel, S.; Putelo, A.; Lee, S.K.; Nallandhighal, S.; et al.
Platelet PD-L1 suppresses anti-cancer immune cell activity in PD-L1 negative tumors. Sci. Rep. 2020,10, 19296. [CrossRef]
269.
Zhou, Z.; Zhang, B.; Zai, W.; Kang, L.; Yuan, A.; Hu, Y.; Wu, J. Perfluorocarbon nanoparticle-mediated platelet inhibition promotes
intratumoral infiltration of T cells and boosts immunotherapy. Proc. Natl. Acad. Sci. USA 2019,116, 11972–11977. [CrossRef]
270.
Guo, Q.; Malloy, M.W.; Roweth, H.G.; McAllister, S.S.; Italiano, J.J.E.; Battinelli, E.M. Platelets upregulate tumor cell programmed
death ligand 1 in an epidermal growth factor receptor-dependent manner in vitro. Blood Adv. 2022,6, 5668–5675. [CrossRef]
271.
Lutz, M.S.; Klimovich, B.; Maurer, S.; Heitmann, J.S.; Märklin, M.; Zekri, L.; Jung, G.; Salih, H.R.; Hinterleitner, C. Platelets subvert
antitumor efficacy of T cell-recruiting bispecific antibodies. J. Immunother. Cancer 2022,10, e003655. [CrossRef] [PubMed]
272.
Crawford, A.; Haber, L.; Kelly, M.P.; Vazzana, K.; Canova, L.; Ram, P.; Pawashe, A.; Finney, J.; Jalal, S.; Chiu, D.; et al. A Mucin 16
bispecific T cell–engaging antibody for the treatment of ovarian cancer. Sci. Transl. Med.
2019
,11, eaau7534. [CrossRef] [PubMed]
273.
Yeku, O.O.; Rao, T.D.; Laster, I.; Kononenko, A.; Purdon, T.J.; Wang, P.; Cui, Z.; Liu, H.; Brentjens, R.J.; Spriggs, D. Bispecific T-Cell
Engaging Antibodies Against MUC16 Demonstrate Efficacy Against Ovarian Cancer in Monotherapy and in Combination With
PD-1 and VEGF Inhibition. Front. Immunol. 2021,12, 663379. [CrossRef] [PubMed]
274.
Raffin, C.; Vo, L.T.; Bluestone, J.A. Treg cell-based therapies: Challenges and perspectives. Nat. Rev. Immunol.
2019
,20,
158–172. [CrossRef]
275.
Fialová, A.; Partlová, S.; Sojka, L.; Hromádková, H.; Brtnický, T.; Fuˇcíková, J.; Kocián, P.; Rob, L.; Bart ˚u ˇnková, J.; Špíšek, R.
Dynamics of T-cell infiltration during the course of ovarian cancer: The gradual shift from a Th17 effector cell response to a
predominant infiltration by regulatory T-cells. Int. J. Cancer 2012,132, 1070–1079. [CrossRef]
276.
Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.-P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al.
Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 2011,475, 226–230. [CrossRef]
277.
Giuntoli, R.L., 2nd; Webb, T.J.; Zoso, A.; Rogers, O.; Diaz-Montes, T.P.; Bristow, R.E.; Oelke, M. Ovarian Cancer-Associated Ascites
Demonstrates Altered Immune Environment: Implications for Antitumor Immunity. Anticancer. Res. 2009,29, 2875–2884.
278.
Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow,
M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival.
Nat. Med. 2004,10, 942–949. [CrossRef]
279.
Knutson, K.L.; Maurer, M.J.; Preston, C.C.; Moysich, K.B.; Goergen, K.; Hawthorne, K.M.; Cunningham, J.M.; Odunsi, K.;
Hartmann, L.C.; Kalli, K.R.; et al. Regulatory T cells, inherited variation, and clinical outcome in epithelial ovarian cancer. Cancer
Immunol. Immunother. 2015,64, 1495–1504. [CrossRef]
280.
Sun, J.; Yan, C.; Xu, D.; Zhang, Z.; Li, K.; Li, X.; Zhou, M.; Hao, D. Immuno-genomic characterisation of high-grade serous ovarian
cancer reveals immune evasion mechanisms and identifies an immunological subtype with a favourable prognosis and improved
therapeutic efficacy. Br. J. Cancer 2022,126, 1570–1580. [CrossRef]
281.
Angiari, S.; Rossi, B.; Piccio, L.; Zinselmeyer, B.H.; Budui, S.; Zenaro, E.; della Bianca, V.; Bach, S.D.; Scarpini, E.; Bolomini-Vittori,
M.; et al. Regulatory T Cells Suppress the Late Phase of the Immune Response in Lymph Nodes through P-Selectin Glycoprotein
Ligand-1. J. Immunol. 2013,191, 5489–5500. [CrossRef] [PubMed]
282.
Kohm, A.P.; Miller, S.D. Role of ICAM-1 and P-selectin expression in the development and effector function of CD4
+
CD25
+
regulatory
T cells. J. Autoimmun. 2003,21, 261–271. [CrossRef] [PubMed]
283.
Urzainqui, A.; Serrador, J.M.; Viedma, F.; Yáñez-Mó, M.; Rodríguez, A.; Corbí, A.L.; Alonso-Lebrero, J.L.; Luque, A.; Deckert, M.;
Vázquez, J.; et al. ITAM-Based Interaction of ERM Proteins with Syk Mediates Signaling by the Leukocyte Adhesion Receptor
PSGL-1. Immunity 2002,17, 401–412. [CrossRef]
Cancers 2023,15, 1282 33 of 37
284.
Rupp, M.-C.; Bergmann, C.B.; Jung, S.; Bock, M.; Biberthaler, P.; Heimann, L.; Hanschen, M. The posttraumatic response of CD4
+
regulatory T cells is modulated by direct cell-cell contact via CD40L- and P-selectin-dependent pathways. Central Eur. J. Immunol.
2021,46, 283–294. [CrossRef] [PubMed]
285.
Guiducci, C.; Valzasina, B.; Dislich, H.; Colombo, M.P. CD40/CD40L interaction regulates CD4
+
CD25
+
T reg homeostasis through
dendritic cell-produced IL-2. Eur. J. Immunol. 2005,35, 557–567. [CrossRef]
286.
Renshaw, B.R.; Fanslow, W.C.; Armitage, R.J.; Campbell, K.A.; Liggitt, D.; Wright, B.; Davison, B.L.; Maliszewski, C.R. Humoral
immune responses in CD40 ligand-deficient mice. J. Exp. Med. 1994,180, 1889–1900. [CrossRef] [PubMed]
287.
Yao, L.; Pan, J.; Setiadi, H.; Pate, K.D.; McEver, R.P. Interleukin 4 or oncostatin M induces a prolonged increase in P-selectin
mRNA and protein in human endothelial cells. J. Exp. Med. 1996,184, 81–92. [CrossRef] [PubMed]
288.
Legein, B.; Temmerman, L.; Biessen, E.A.L.; Lutgens, E. Inflammation and immune system interactions in atherosclerosis. Cell.
Mol. Life Sci. 2013,70, 3847–3869. [CrossRef]
289.
Kampan, N.C.; Madondo, M.T.; McNally, O.M.; Stephens, A.N.; Quinn, M.A.; Plebanski, M. Interleukin 6 Present in Inflammatory
Ascites from Advanced Epithelial Ovarian Cancer Patients Promotes Tumor Necrosis Factor Receptor 2-Expressing Regulatory T
Cells. Front. Immunol. 2017,8, 1482. [CrossRef]
290.
Torrey, H.; Butterworth, J.; Mera, T.; Okubo, Y.; Wang, L.; Baum, D.; Defusco, A.; Plager, S.; Warden, S.; Huang, D.; et al. Targeting
TNFR2 with antagonistic antibodies inhibits proliferation of ovarian cancer cells and tumor-associated T
regs
.Sci. Signal.
2017
,10,
eaaf8608. [CrossRef]
291.
Haribhai, D.; Luo, X.; Chen, J.; Jia, S.; Shi, L.; Schroeder, J.A.; Weiler, H.; Aster, R.H.; Hessner, M.J.; Hu, J.; et al. TGF-
β
1 along with
other platelet contents augments Treg cells to suppress anti-FVIII immune responses in hemophilia A mice. Blood Adv.
2016
,1,
139–151. [CrossRef] [PubMed]
292.
Zimmer, N.; Krebs, F.K.; Zimmer, S.; Mitzel-Rink, H.; Kumm, E.J.; Jurk, K.; Grabbe, S.; Loquai, C.; Tuettenberg, A. Platelet-Derived
GARP Induces Peripheral Regulatory T Cells—Potential Impact on T Cell Suppression in Patients with Melanoma-Associated
Thrombocytosis. Cancers 2020,12, 3653. [CrossRef] [PubMed]
293.
Re, S.L.; Lecocq, M.; Uwambayinema, F.; Yakoub, Y.; Delos, M.; Demoulin, J.-B.; Lucas, S.; Sparwasser, T.; Renauld, J.-C.; Lison, D.;
et al. Platelet-Derived Growth Factor–Producing CD4
+
Foxp3
+
Regulatory T Lymphocytes Promote Lung Fibrosis. Am. J. Respir.
Crit. Care Med. 2011,184, 1270–1281. [CrossRef] [PubMed]
294.
Roane, B.M.; Meza-Perez, S.; Katre, A.A.; Goldsberry, W.N.; Randall, T.D.; Norian, L.A.; Birrer, M.J.; Arend, R.C. Neutralization
of TGF
β
Improves Tumor Immunity and Reduces Tumor Progression in Ovarian Carcinoma. Mol. Cancer Ther.
2021
,20,
602–611. [CrossRef]
295.
Yahya, M.A.; Sharon, S.M.; Hantisteanu, S.; Hallak, M.; Bruchim, I. The Role of the Insulin-Like Growth Factor 1 Pathway
in Immune Tumor Microenvironment and Its Clinical Ramifications in Gynecologic Malignancies. Front. Endocrinol.
2018
,9,
297. [CrossRef]
296.
Camorani, S.; Passariello, M.; Agnello, L.; Esposito, S.; Collina, F.; Cantile, M.; di Bonito, M.; Ulasov, I.V.; Fedele, M.; Zannetti, A.;
et al. Aptamer targeted therapy potentiates immune checkpoint blockade in triple-negative breast cancer. J. Exp. Clin. Cancer Res.
2020,39, 180. [CrossRef]
297.
Liu, C.Y.; Battaglia, M.; Lee, S.H.; Sun, Q.-H.; Aster, R.H.; Visentin, G.P. Platelet Factor 4 Differentially Modulates CD4+CD25+
(Regulatory) versus CD4+CD25(Nonregulatory) T Cells. J. Immunol. 2005,174, 2680–2686. [CrossRef]
298.
Kabashima, K.; Murata, T.; Tanaka, H.; Matsuoka, T.; Sakata, D.; Yoshida, N.; Katagiri, K.; Kinashi, T.; Tanaka, T.; Miyasaka, M.;
et al. Thromboxane A2 modulates interaction of dendritic cells and T cells and regulates acquired immunity. Nat. Immunol.
2003
,
4, 694–701. [CrossRef]
299.
von Hundelshausen, P.; Agten, S.M.; Eckardt, V.; Blanchet, X.; Schmitt, M.M.; Ippel, H.; Neideck, C.; Bidzhekov, K.; Leberzammer,
J.; Wichapong, K.; et al. Chemokine interactome mapping enables tailored intervention in acute and chronic inflammation. Sci.
Transl. Med. 2017,9, eaah6650. [CrossRef]
300.
Tan, S.; Zhang, J.; Sun, Y.; Gisterå, A.; Sheng, Z.; Malmström, R.E.; Hou, M.; Peng, J.; Ma, C.; Liao, W.; et al. Platelets enhance
CD4
+
central memory T cell responses via platelet factor 4-dependent mitochondrial biogenesis and cell proliferation. Platelets
2021,33, 360–370. [CrossRef]
301.
Figueras, A.; Alsina-Sanchís, E.; Lahiguera, Á.; Abreu, M.; Muinelo-Romay, L.; Moreno-Bueno, G.; Casanovas, O.; Graupera, M.;
Matias-Guiu, X.; Vidal, A.; et al. A Role for CXCR4 in Peritoneal and Hematogenous Ovarian Cancer Dissemination. Mol. Cancer
Ther. 2018,17, 532–543. [CrossRef] [PubMed]
302.
Chatterjee, M.; Rath, D.; Gawaz, M. Role of chemokine receptors CXCR4 and CXCR7 for platelet function. Biochem. Soc. Trans.
2015,43, 720–726. [CrossRef] [PubMed]
303.
Chatterjee, M.; I von Ungern-Sternberg, S.N.; Seizer, P.; Schlegel, F.; Büttcher, M.; Sindhu, N.A.; Müller, S.; Mack, A.; Gawaz,
M. Platelet-derived CXCL12 regulates monocyte function, survival, differentiation into macrophages and foam cells through
differential involvement of CXCR4–CXCR7. Cell Death Dis. 2015,6, e1989. [CrossRef] [PubMed]
304.
Yan, M.; Jene, N.; Byrne, D.; Millar, E.K.A.; O’Toole, S.A.; McNeil, C.M.; Bates, G.J.; Harris, A.L.; Banham, A.H.; Sutherland,
R.L.; et al. Recruitment of regulatory T cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor
prognosis in basal-like breast cancers. Breast Cancer Res. 2011,13, R47. [CrossRef]
Cancers 2023,15, 1282 34 of 37
305.
Righi, E.; Kashiwagi, S.; Yuan, J.; Santosuosso, M.; Leblanc, P.; Ingraham, R.; Forbes, B.; Edelblute, B.; Collette, B.; Xing, D.; et al.
CXCL12/CXCR4 Blockade Induces Multimodal Antitumor Effects That Prolong Survival in an Immunocompetent Mouse Model
of Ovarian Cancer. Cancer Res. 2011,71, 5522–5534. [CrossRef]
306.
Toker, A.; Nguyen, L.T.; Stone, S.C.; Yang, S.C.; Katz, S.R.; Shaw, P.A.; Clarke, B.A.; Ghazarian, D.; Al-Habeeb, A.; Easson, A.; et al.
Regulatory T Cells in Ovarian Cancer Are Characterized by a Highly Activated Phenotype Distinct from that in Melanoma. Clin.
Cancer Res. 2018,24, 5685–5696. [CrossRef]
307.
Battaglia, A.; Piermattei, A.; Buzzonetti, A.; Pasciuto, T.; Zampetti, N.; Fossati, M.; Angelico, G.; Iacobelli, V.; Nero, C.; Iannucci, V.;
et al. PD-L1 Expression on Circulating Tumour-Derived Microvesicles as a Complementary Tool for Stratification of High-Grade
Serous Ovarian Cancer Patients. Cancers 2021,13, 5200. [CrossRef]
308.
Kamada, T.; Togashi, Y.; Tay, C.; Ha, D.; Sasaki, A.; Nakamura, Y.; Sato, E.; Fukuoka, S.; Tada, Y.; Tanaka, A.; et al. PD-1
+
regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl. Acad. Sci. USA
2019
,116,
9999–10008. [CrossRef]
309.
Dinkla, S.; van Cranenbroek, B.; van der Heijden, W.A.; He, X.; Wallbrecher, R.; Dumitriu, I.E.; van der Ven, A.J.; Bosman,
G.J.C.G.M.; Koenen, H.J.P.M.; Joosten, I. Platelet microparticles inhibit IL-17 production by regulatory T cells through P-selectin.
Blood 2016,127, 1976–1986. [CrossRef]
310.
Ayyoub, M.; Deknuydt, F.; Raimbaud, I.; Dousset, C.; Leveque, L.; Bioley, G.; Valmori, D. Human memory FOXP3
+
Tregs secrete
IL-17 ex vivo and constitutively express the T
H
17 lineage-specific transcription factor ROR
γ
t. Proc. Natl. Acad. Sci. USA
2009
,
106, 8635–8640. [CrossRef]
311.
Fleischer, J.; Grage-Griebenow, E.; Kasper, B.; Heine, H.; Ernst, M.; Brandt, E.; Flad, H.-D.; Petersen, F. Platelet Factor 4 Inhibits
Proliferation and Cytokine Release of Activated Human T Cells. J. Immunol. 2002,169, 770–777. [CrossRef] [PubMed]
312.
Forlow, S.B.; McEver, R.P.; Nollert, M.U. Leukocyte-leukocyte interactions mediated by platelet microparticles under flow. Blood
2000,95, 1317–1323. [CrossRef] [PubMed]
313.
Johnston, R.J.; Comps-Agrar, L.; Hackney, J.; Yu, X.; Huseni, M.; Yang, Y.; Park, S.; Javinal, V.; Chiu, H.; Irving, B.; et al.
The Immunoreceptor TIGIT Regulates Antitumor and Antiviral CD8 + T Cell Effector Function. Cancer Cell
2014
,26,
923–937. [CrossRef]
314.
Osorio Ovarian Tumor Tissue Analysis (OTTA) Consortium; Goode, E.L.; Block, M.S.; Kalli, K.R.; Vierkant, R.A.; Chen, W.; Fogarty,
Z.C.; Gentry-Maharaj, A.; Toloczko, A.; Hein, A.; et al. Dose-Response Association of CD8
+
Tumor-Infiltrating Lymphocytes and
Survival Time in High-Grade Serous Ovarian Cancer. JAMA Oncol. 2017,3, e173290. [CrossRef] [PubMed]
315.
Lieber, S.; Reinartz, S.; Raifer, H.; Finkernagel, F.; Dreyer, T.; Bronger, H.; Jansen, J.M.; Wagner, U.; Worzfeld, T.; Müller, R.;
et al. Prognosis of ovarian cancer is associated with effector memory CD8
+
T cell accumulation in ascites, CXCL9 levels and
activation-triggered signal transduction in T cells. Oncoimmunology 2018,7, e1424672. [CrossRef]
316.
Meikle, C.K.; Meisler, A.J.; Bird, C.M.; Jeffries, J.A.; Azeem, N.; Garg, P.; Crawford, E.L.; Kelly, C.A.; Gao, T.Z.; Wuescher, L.M.;
et al. Platelet-T cell aggregates in lung cancer patients: Implications for thrombosis. PLoS ONE 2020,15, e0236966. [CrossRef]
317.
Veerman, K.M.; Williams, M.J.; Uchimura, K.; Singer, M.S.; Merzaban, J.S.; Naus, S.; Carlow, D.A.; Owen, P.; Rivera-Nieves, J.;
Rosen, S.D.; et al. Interaction of the selectin ligand PSGL-1 with chemokines CCL21 and CCL19 facilitates efficient homing of T
cells to secondary lymphoid organs. Nat. Immunol. 2007,8, 532–539. [CrossRef]
318.
Tinoco, R.; Neubert, E.N.; Stairiker, C.J.; Henriquez, M.L.; Bradley, L.M. PSGL-1 Is a T Cell Intrinsic Inhibitor That Regulates
Effector and Memory Differentiation and Responses During Viral Infection. Front. Immunol. 2021,12, 677824. [CrossRef]
319.
Zhang, Q.; Yang, X.J.; Kundu, S.D.; Pins, M.; Javonovic, B.; Meyer, R.; Kim, S.-J.; Greenberg, N.M.; Kuzel, T.; Meagher, R.; et al.
Blockade of transforming growth factor-
β
signaling in tumor-reactive CD8
+
T cells activates the antitumor immune response
cycle. Mol. Cancer Ther. 2006,5, 1733–1743. [CrossRef]
320.
Marcheteau, E.; Farge, T.; Pérès, M.; Labrousse, G.; Tenet, J.; Delmas, S.; Chusseau, M.; Duprez-Paumier, R.; Franchet, C.; Dalenc,
F.; et al. Thrombospondin-1 Silencing Improves Lymphocyte Infiltration in Tumors and Response to Anti-PD-1 in Triple-Negative
Breast Cancer. Cancers 2021,13, 4059. [CrossRef]
321.
Rachidi, S.; Metelli, A.; Riesenberg, B.; Wu, B.X.; Nelson, M.H.; Wallace, C.; Paulos, C.M.; Rubinstein, M.P.; Garrett-Mayer,
E.; Hennig, M.; et al. Platelets subvert T cell immunity against cancer via GARP-TGF
β
axis. Sci. Immunol.
2017
,2,
eaai7911. [CrossRef] [PubMed]
322.
Riesenberg, B.P.; Ansa-Addo, E.A.; Gutierrez, J.; Timmers, C.D.; Liu, B.; Li, Z. Cutting Edge: Targeting Thrombocytes to
Rewire Anticancer Immunity in the Tumor Microenvironment and Potentiate Efficacy of PD-1 Blockade. J. Immunol.
2019
,203,
1105–1110. [CrossRef] [PubMed]
323.
Zhang, X.; Wang, J.; Chen, Z.; Hu, Q.; Wang, C.; Yan, J.; Dotti, G.; Huang, P.; Gu, Z. Engineering PD-1-Presenting Platelets for
Cancer Immunotherapy. Nano Lett. 2018,18, 5716–5725. [CrossRef] [PubMed]
324.
Zhang, S.; Ke, X.; Zeng, S.; Wu, M.; Lou, J.; Wu, L.; Huang, P.; Huang, L.; Wang, F.; Pan, S. Analysis of CD8
+
Treg cells in patients
with ovarian cancer: A possible mechanism for immune impairment. Cell. Mol. Immunol. 2015,12, 580–591. [CrossRef]
325.
Napoletano, C.; Bellati, F.; Landi, R.; Pauselli, S.; Marchetti, C.; Visconti, V.; Sale, P.; Liberati, M.; Rughetti, A.; Frati, L.; et al.
Ovarian cancer cytoreduction induces changes in T cell population subsets reducing immunosuppression. J. Cell. Mol. Med.
2010
,
14, 2748–2759. [CrossRef]
Cancers 2023,15, 1282 35 of 37
326.
Wu, M.; Chen, X.; Lou, J.; Zhang, S.; Zhang, X.; Huang, L.; Sun, R.; Huang, P.; Wang, F.; Pan, S. TGF-
β
1 con-
tributes to CD8
+
Treg induction through p38 MAPK signaling in ovarian cancer microenvironment. Oncotarget
2016
,7,
44534–44544. [CrossRef] [PubMed]
327.
Webb, J.R.; Wick, D.A.; Nielsen, J.S.; Tran, E.; Milne, K.; McMurtrie, E.; Nelson, B.H. Profound elevation of CD8
+
T cells expressing
the intraepithelial lymphocyte marker CD103 (
α
E/
β
7 Integrin) in high-grade serous ovarian cancer. Gynecol. Oncol.
2010
,118,
228–236. [CrossRef]
328.
Nierodzik, M.L.; Karpatkin, S. Thrombin induces tumor growth, metastasis, and angiogenesis: Evidence for a thrombin-regulated
dormant tumor phenotype. Cancer Cell 2006,10, 355–362. [CrossRef]
329.
Kopp, H.-G.; Placke, T.; Salih, H.R. Platelet-Derived Transforming Growth Factor-
β
Down-Regulates NKG2D Thereby Inhibiting
Natural Killer Cell Antitumor Reactivity. Cancer Res. 2009,69, 7775–7783. [CrossRef]
330.
Alexander, E.T.; Minton, A.R.; Peters, M.C.; van Ryn, J.; Gilmour, S.K. Thrombin inhibition and cisplatin block tumor progression
in ovarian cancer by alleviating the immunosuppressive microenvironment. Oncotarget 2016,7, 85291–85305. [CrossRef]
331.
Motz, G.T.; Santoro, S.P.; Wang, L.-P.; Garrabrant, T.; Lastra, R.R.; Hagemann, I.S.; Lal, P.; Feldman, M.D.; Benencia, F.; Coukos,
G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med.
2014
,20,
607–615. [CrossRef] [PubMed]
332.
Zelenay, S.; van der Veen, A.G.; Böttcher, J.P.; Snelgrove, K.J.; Rogers, N.; Acton, S.E.; Chakravarty, P.; Girotti, M.R.;
Marais, R.; Quezada, S.A.; et al. Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell
2015
,162,
1257–1270. [CrossRef]
333. Melchers, F. Checkpoints that control B cell development. J. Clin. Investig. 2015,125, 2203–2210. [CrossRef] [PubMed]
334.
Petitprez, F.; de Reyniès, A.; Keung, E.Z.; Chen, T.W.-W.; Sun, C.-M.; Calderaro, J.; Jeng, Y.-M.; Hsiao, L.-P.; Lacroix, L.; Bougoüin,
A.; et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020,577, 556–560. [CrossRef]
335.
Wei, X.; Jin, Y.; Tian, Y.; Zhang, H.; Wu, J.; Lu, W.; Lu, X. Regulatory B cells contribute to the impaired antitumor immunity in
ovarian cancer patients. Tumor Biol. 2015,37, 6581–6588. [CrossRef]
336.
Capello, M.; Vykoukal, J.V.; Katayama, H.; Bantis, L.E.; Wang, H.; Kundnani, D.L.; Aguilar-Bonavides, C.; Aguilar, M.; Tripathi,
S.C.; Dhillon, D.S.; et al. Exosomes harbor B cell targets in pancreatic adenocarcinoma and exert decoy function against
complement-mediated cytotoxicity. Nat. Commun. 2019,10, 254. [CrossRef] [PubMed]
337.
de Visser, K.E.; Korets, L.V.; Coussens, L.M. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte
dependent. Cancer Cell 2005,7, 411–423. [CrossRef]
338.
Miao, S.; Shu, D.; Zhu, Y.; Lu, M.; Zhang, Q.; Pei, Y.; He, A.-D.; Ma, R.; Zhang, B.; Ming, Z.-Y. Cancer cell-derived immunoglobulin
G activates platelets by binding to platelet FcγRIIa. Cell Death Dis. 2019,10, 87. [CrossRef] [PubMed]
339.
van de Veen, W.; Globinska, A.; Jansen, K.; Straumann, A.; Kubo, T.; Verschoor, D.; Wirz, O.F.; Castro-Giner, F.; Tan, G.;
Rückert, B.; et al. A novel proangiogenic B cell subset is increased in cancer and chronic inflammation. Sci. Adv.
2020
,6,
eaaz3559. [CrossRef] [PubMed]
340.
Dong, H.P.; Elstrand, M.B.; Holth, A.; Silins, I.; Berner, A.; Trope, C.G.; Davidson, B.; Risberg, B. Nk- and B-Cell Infiltration
Correlates with Worse Outcome in Metastatic Ovarian Carcinoma. Am. J. Clin. Pathol. 2006,125, 451–458. [CrossRef]
341.
Nielsen, J.S.; Sahota, R.A.; Milne, K.; Kost, S.E.; Nesslinger, N.J.; Watson, P.H.; Nelson, B.H. CD20+ Tumor-Infiltrating Lymphocytes
Have an Atypical CD27
Memory Phenotype and Together with CD8
+
T Cells Promote Favorable Prognosis in Ovarian Cancer.
Clin. Cancer Res. 2012,18, 3281–3292. [CrossRef]
342.
Elzey, B.D.; Grant, J.F.; Sinn, H.W.; Nieswandt, B.; Waldschmidt, T.J.; Ratliff, T.L. Cooperation between platelet-derived CD154
and CD4+T cells for enhanced germinal center formation. J. Leukoc. Biol. 2005,78, 80–84. [CrossRef]
343.
Morva, A.; Lemoine, S.; Achour, A.; Pers, J.-O.; Youinou, P.; Jamin, C. Maturation and function of human dendritic cells are
regulated by B lymphocytes. Blood 2012,119, 106–114. [CrossRef] [PubMed]
344.
Shao, Y.; Lo, C.M.; Ling, C.C.; Liu, X.B.; Ng, K.T.-P.; Chu, A.C.Y.; Ma, Y.Y.; Li, C.X.; Fan, S.T.; Man, K. Regulatory B cells accelerate
hepatocellular carcinoma progression via CD40/CD154 signaling pathway. Cancer Lett.
2014
,355, 264–272. [CrossRef] [PubMed]
345.
Wallace, C.H.; Wu, B.X.; Salem, M.; Ansa-Addo, E.A.; Metelli, A.; Sun, S.; Gilkeson, G.; Shlomchik, M.J.; Liu, B.;
Li, Z. B lymphocytes confer immune tolerance via cell surface GARP-TGF-
β
complex. J. Clin. Investig.
2018
,3,
e99863. [CrossRef] [PubMed]
346.
Kehrl, J.H.; Thevenin, C.; Rieckmann, P.; Fauci, A.S. Transforming growth factor-beta suppresses human B lymphocyte Ig
production by inhibiting synthesis and the switch from the membrane form to the secreted form of Ig mRNA. J. Immunol.
1991
,
146, 4016–4023. [CrossRef] [PubMed]
347. Cerutti, A. The regulation of IgA class switching. Nat. Rev. Immunol. 2008,8, 421–434. [CrossRef]
348.
Bulk, J.V.D.; de Miranda, N.F.; Dijke, P.T. Therapeutic targeting of TGF-
β
in cancer: Hacking a master switch of immune
suppression. Clin. Sci. 2021,135, 35–52. [CrossRef]
349.
Olkhanud, P.B.; Damdinsuren, B.; Bodogai, M.; Gress, R.E.; Sen, R.; Wejksza, K.; Malchinkhuu, E.; Wersto, R.P.; Biragyn, A.
Tumor-Evoked Regulatory B Cells Promote Breast Cancer Metastasis by Converting Resting CD4
+
T Cells to T-Regulatory Cells.
Cancer Res. 2011,71, 3505–3515. [CrossRef]
350.
Gill, P.; Jindal, N.L.; Jagdis, A.; Vadas, P. Platelets in the immune response: Revisiting platelet-activating factor in anaphylaxis. J.
Allergy Clin. Immunol. 2015,135, 1424–1432. [CrossRef]
Cancers 2023,15, 1282 36 of 37
351.
Denizot, Y.; Guglielmi, L.; Donnard, M.; Trimoreau, F. Platelet-activating Factor and Normal or Leukaemic Haematopoiesis. Leuk.
Lymphoma 2003,44, 775–782. [CrossRef] [PubMed]
352.
Braun, A.; Anders, H.-J.; Gudermann, T.; Mammadova-Bach, E. Platelet-Cancer Interplay: Molecular Mechanisms and New
Therapeutic Avenues. Front. Oncol. 2021,11, 665534. [CrossRef] [PubMed]
353.
Ma, C.; Fu, Q.; Diggs, L.P.; McVey, J.C.; McCallen, J.; Wabitsch, S.; Ruf, B.; Brown, Z.; Heinrich, B.; Zhang, Q.; et al. Platelets control
liver tumor growth through P2Y12-dependent CD40L release in NAFLD. Cancer Cell
2022
,40, 986–998.e5. [CrossRef] [PubMed]
354.
Menter, D.G.; Tucker, S.C.; Kopetz, S.; Sood, A.K.; Crissman, J.D.; Honn, K.V. Platelets and cancer: A casual or causal relationship:
Revisited. Cancer Metastasis Rev. 2014,33, 231–269. [CrossRef]
355.
Gawaz, M.; Neumann, F.-J.; Dickfeld, T.; Reininger, A.; Adelsberger, H.; Gebhardt, A.; Schömig, A. Vitronectin Receptor (
αvβ3
)
Mediates Platelet Adhesion to the Luminal Aspect of Endothelial Cells. Circulation 1997,96, 1809–1818. [CrossRef]
356.
Bombeli, T.; Schwartz, B.R.; Harlan, J.M. Adhesion of Activated Platelets to Endothelial Cells: Evidence for a GPIIbIIIa-dependent
Bridging Mechanism and Novel Roles for Endothelial Intercellular Adhesion Molecule 1 (ICAM-1),
α
v
β
3 Integrin, and GPIb
α
.J.
Exp. Med. 1998,187, 329–339. [CrossRef]
357.
Langer, H.; May, A.E.; Bültmann, A.; Gawaz, M. ADAM15 is an adhesion receptor for platelet GPIIb-IIIa and induces platelet
activation. Thromb. Haemost. 2005,94, 555–561. [CrossRef]
358.
Tan, D.S.; Agarwal, R.; Kaye, S.B. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol.
2006
,7,
925–934. [CrossRef]
359.
Schmied, L.; Höglund, P.; Meinke, S. Platelet-Mediated Protection of Cancer Cells from Immune Surveillance—Possible Implica-
tions for Cancer Immunotherapy. Front. Immunol. 2021,12, 640578. [CrossRef]
360. Schlesinger, M. Role of platelets and platelet receptors in cancer metastasis. J. Hematol. Oncol. 2018,11, 125. [CrossRef]
361.
Olsson, A.-K.; Cedervall, J. NETosis in Cancer—Platelet–Neutrophil Crosstalk Promotes Tumor-Associated Pathology. Front.
Immunol. 2016,7, 373. [CrossRef] [PubMed]
362.
Linke, B.; Schreiber, Y.; Picard-Willems, B.; Slattery, P.; Nüsing, R.M.; Harder, S.; Geisslinger, G.; Scholich, K. Activated Platelets
Induce an Anti-Inflammatory Response of Monocytes/Macrophages through Cross-Regulation of PGE
2
and Cytokines. Mediat.
Inflamm. 2017,2017, 1463216. [CrossRef] [PubMed]
363.
Li, X.; Zhong, H.; Bao, W.; Boulad, N.; Evangelista, J.; Haider, M.A.; Bussel, J.; Yazdanbakhsh, K. Defective regulatory B-cell
compartment in patients with immune thrombocytopenia. Blood 2012,120, 3318–3325. [CrossRef] [PubMed]
364.
Rolfes, V.; Idel, C.; Pries, R.; Plötze-Martin, K.; Habermann, J.; Gemoll, T.; Bohnet, S.; Latz, E.; Ribbat-Idel, J.; Franklin, B.S.; et al.
PD-L1 is expressed on human platelets and is affected by immune checkpoint therapy. Oncotarget
2018
,9, 27460–27470. [CrossRef]
365.
Ye, Q.; Cheng, J.; Ye, M.; Liu, D.; Zhang, Y. Association of pretreatment thrombocytosis with prognosis in ovarian cancer: A
systematic review and meta-analysis. J. Gynecol. Oncol. 2019,30, e5. [CrossRef]
366.
Canzler, U.; Lück, H.-J.; Neuser, P.; Sehouli, J.; Burges, A.; Harter, P.; Schmalfeldt, B.; Aminossadati, B.; Mahner, S.; Kommoss, S.;
et al. Prognostic role of thrombocytosis in recurrent ovarian cancer: A pooled analysis of the AGO Study Group. Arch. Gynecol.
Obstet. 2020,301, 1267–1274. [CrossRef]
367.
Nakao, S.; Minaguchi, T.; Itagaki, H.; Hosokawa, Y.; Shikama, A.; Tasaka, N.; Akiyama, A.; Ochi, H.; Matsumoto, K.; Satoh, T.
Pretreatment thrombocytosis as an independent predictive factor for chemoresistance and poor survival in epithelial ovarian
cancer. J. Ovarian Res. 2020,13, 55–59. [CrossRef]
368.
Watrowski, R.; Heinze, G.; Jäger, C.; Forster, J.; Zeillinger, R. Usefulness of the preoperative platelet count in the diagnosis of
adnexal tumors. Tumor Biol. 2016,37, 12079–12087. [CrossRef]
369.
Hufnagel, D.H.; Cozzi, G.D.; Crispens, M.A.; Beeghly-Fadiel, A. Platelets, Thrombocytosis, and Ovarian Cancer Prognosis:
Surveying the Landscape of the Literature. Int. J. Mol. Sci. 2020,21, 8169. [CrossRef]
370.
Naito, R.; Miyauchi, K.; Yasuda, S.; Kaikita, K.; Akao, M.; Ako, J.; Matoba, T.; Nakamura, M.; Hagiwara, N.; Kimura, K.; et al.
Rivaroxaban Monotherapy vs Combination Therapy with Antiplatelets on Total Thrombotic and Bleeding Events in Atrial
Fibrillation with Stable Coronary Artery Disease. JAMA Cardiol. 2022,7, 787–794. [CrossRef]
371.
Anadani, M.; de Havenon, A.; Henninger, N.; Kuohn, L.; Mac Grory, B.; Furie, K.L.; Kim, A.S.; Easton, J.D.; Johnston, S.C.; Yaghi,
S. Antiplatelet Use and Ischemic Stroke Risk in Minor Stroke or Transient Ischemic Attack: A Post Hoc Analysis of the POINT
Trial. Stroke 2021,52, e773–e776. [CrossRef] [PubMed]
372.
Cooke, N.M.; Spillane, C.D.; Sheils, O.; O’Leary, J.; Kenny, D. Aspirin and P2Y12 inhibition attenuate platelet-induced ovarian
cancer cell invasion. BMC Cancer 2015,15, 627. [CrossRef]
373.
Denslow, A.; ´
Switalska, M.; Jarosz, J.; Papiernik, D.; Porshneva, K.; Nowak, M.; Wietrzyk, J. Clopidogrel in a combined therapy
with anticancer drugs—Effect on tumor growth, metastasis, and treatment toxicity: Studies in animal models. PLoS ONE
2017
,12,
e0188740. [CrossRef]
374.
Wright, J.R.; Chauhan, M.; Shah, C.; Ring, A.; Thomas, A.L.; Goodall, A.H.; Adlam, D. The TICONC (Ticagrelor-Oncology) Study.
JACC CardioOncol. 2020,2, 236–250. [CrossRef] [PubMed]
375.
Cuzick, J.; Thorat, M.A.; Bosetti, C.; Brown, P.H.; Burn, J.; Cook, N.R.; Ford, L.G.; Jacobs, E.J.; Jankowski, J.A.; La Vecchia,
C.; et al. Estimates of benefits and harms of prophylactic use of aspirin in the general population. Ann. Oncol.
2014
,26,
47–57. [CrossRef] [PubMed]
376.
Hwang, B.-O.; Park, S.-Y.; Cho, E.S.; Zhang, X.; Lee, S.K.; Ahn, H.-J.; Chun, K.-S.; Chung, W.-Y.; Song, N.-Y. Platelet CLEC2-
Podoplanin Axis as a Promising Target for Oral Cancer Treatment. Front. Immunol. 2021,12, 807600. [CrossRef] [PubMed]
Cancers 2023,15, 1282 37 of 37
377.
Hughes, C.E.; Navarro-Núñez, L.; Finney, B.A.; Mourão-Sá, D.; Pollitt, A.Y.; Watson, S.P. CLEC-2 is not required for platelet
aggregation at arteriolar shear. J. Thromb. Haemost. 2010,8, 2328–2332. [CrossRef] [PubMed]
378.
Le, X.; Nilsson, M.; Goldman, J.; Reck, M.; Nakagawa, K.; Kato, T.; Ares, L.P.; Frimodt-Moller, B.; Wolff, K.; Visseren-Grul, C.; et al.
Dual EGFR-VEGF Pathway Inhibition: A Promising Strategy for Patients With EGFR-Mutant NSCLC. J. Thorac. Oncol. 2020,16,
205–215. [CrossRef]
379.
Subbiah, V.; Dumbrava, E.I.; Jiang, Y.; Thein, K.Z.; Naing, A.; Hong, D.S.; Fu, S.; Piha-Paul, S.A.; Tsimberidou, A.M.; Janku, F.;
et al. Dual EGFR blockade with cetuximab and erlotinib combined with anti-VEGF antibody bevacizumab in advanced solid
tumors: A phase 1 dose escalation triplet combination trial. Exp. Hematol. Oncol. 2020,9, 7–8. [CrossRef]
380.
Hu, Q.; Hada, A.; Han, L. Platelet count as a biomarker for monitoring treatment response and disease recurrence in recurrent
epithelial ovarian cancer. J. Ovarian Res. 2020,13, 78. [CrossRef]
381.
Soff, G.A.; Ray-Coquard, I.; Rivera, L.J.M.; Fryzek, J.; Mullins, M.; Bylsma, L.C.; Park, J.K. Systematic literature review
and meta-analysis on use of Thrombopoietic agents for chemotherapy-induced thrombocytopenia. PLoS ONE
2022
,17,
e0257673. [CrossRef] [PubMed]
382.
Wang, J.; Zhou, P.; Han, Y.; Zhang, H. Platelet transfusion for cancer secondary thrombocytopenia: Platelet and cancer cell
interaction. Transl. Oncol. 2021,14, 101022. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Several studies have found that tumor tissue factor expression in ovarian carcinoma cell-derived extracellular vesicles promoted VTE in both preclinical [9] and clinical scenarios [10,11]. Platelet-derived C-type lectin-like receptor-2 (CLEC-2) binding to podoplanin on CAFs and cancer cells promotes tumor development and venous thrombosis in HGSOC patients [12]. As a result, GATA3 is a promising option for multitarget strategy of immune cells such as TAMs, stem cells, CAFs, and platelets inside the TME of HGSOC patients, as well as lowering the risk of venous thrombosis (Fig. 2). ...
Article
Multitarget strategy of GATA3 and high-grade serous ovarian carcinoma: Where are we now?
... Differential expression and prognostic signi cance of CYB5D2 and TGF-β in LIHC TGF-β often acts as a tumor suppressor in early-stage HCC(16). By curtailing cell proliferation and promoting apoptosis, TGF-β forestalls the initiation and progression of this malignancy (17). First, the expression levels of CYB5D2 in both normal and TCGA-LIHC samples were determined using the Wilcoxon test in our investigation. ...
Preprint
Full-text available
Aim Hepatocellular carcinoma (HCC) is a common liver malignancy. In this study, our goal was to investigate how TGF-β and CYB5D2 function in the etiology of HCC and their potential as prognostic biomarkers. Methods Gene co-expression network and prognostic analysis were executed on the GSE101685 dataset, and CYB5D2 was determined to be a hub gene. Then the expression of CYB5D2 and TGF-β in HCC and their correlation were detected. In vitro experiments analyzed the effects of CYB5D2 and TGF-β on the progression of HCC. Tumor xenograft experiments were performed to detect the regulation of CYB5D2 overexpression on tumor growth. Results Immunohistochemistry (IHC) and expression analysis results showed that CYB5D2 can serve as a tumor suppressor in HCC. In contrast, TGF-β, which is inversely correlated with CYB5D2, was overexpressed in liver hepatocellular carcinoma (LIHC) and linked to poor patient prognosis. In vitro experiments confirmed that CYB5D2 expression was upregulated in HCC cell lines, while TGF-β expression was upregulated, and results from the Human Protein Atlas (HPA) database confirmed these findings. Functional analysis showed that CYB5D2 overexpression inhibited the proliferation, migration, and invasion of HCC cells and induced G1 phase arrest. Furthermore, TGF-β treatment counteracted CYB5D2-mediated epithelial-mesenchymal transition (EMT) marker expression and tumor progression. Finally, in vivo studies showed that CYB5D2 overexpression significantly reduced tumor growth, suggesting its potential anticancer activity against HCC. Conclusion Overall, the tumor suppressor function of CYB5D2 in HCC and its interaction with TGF-β offer fresh information on the molecular pathophysiology of HCC and possible treatment avenues.
Article
Full-text available
In conventional T cells, OX40 has been identi!ed as a major costimulating receptor augmenting survival and clonal expansion of effector and memory T cell populations. In regulatory T cells, (Treg) OX40 signaling suppresses cellular activity and differentiation. However, clinical trials investigating OX40 agonists to enhance anti-tumor immunity, showed only limited success so far. Here we show that platelets from breast cancer patients express relevant levels of OX40L and platelet OX40L (pOX40L) inversely correlates with platelet-expressed immune checkpoint molecules GITRL (pGITRL) and TACI (pTACI). While high expression of pOX40L correlates with T and NK cell activation, elevated pOX40L levels identify patients with higher tumor grades, the occurrence of metastases, and shorter recurrence-free survival (RFS). Of note, OX40 mRNA levels in breast cancer correlate with enhanced expression of anti-apoptotic, immune- suppressive, and tumor-promoting mRNA gene signatures. Our data suggest that OX40L on platelets might play counteracting roles in cancer and anti-tumor immunity. Since pOX40L re"ects disease relapse better than the routinely used predictive markers CA15-3, CEA, and LDH, it could serve as a novel biomarker for refractory disease in breast cancer.
Article
Full-text available
Importance: Appropriate regimens of antithrombotic therapy for patients with atrial fibrillation (AF) and coronary artery disease (CAD) have not yet been established. Objective: To compare the total number of thrombotic and/or bleeding events between rivaroxaban monotherapy and combined rivaroxaban and antiplatelet therapy in such patients. Design, setting, and participants: This was a post hoc secondary analysis of the Atrial Fibrillation and Ischemic Events With Rivaroxaban in Patients With Stable Coronary Artery Disease (AFIRE) open-label, randomized clinical trial. This multicenter analysis was conducted from February 23, 2015, to July 31, 2018. Patients with AF and stable CAD who had undergone percutaneous coronary intervention or coronary artery bypass grafting 1 or more years earlier or who had angiographically confirmed CAD not requiring revascularization were enrolled. Data were analyzed from September 1, 2020, to March 26, 2021. Interventions: Rivaroxaban monotherapy or combined rivaroxaban and antiplatelet therapy. Main outcomes and measures: The total incidence of thrombotic, bleeding, and fatal events was compared between the groups. Cox regression analyses were used to estimate the risk of subsequent events in the 2 groups, with the status of thrombotic or bleeding events that had occurred by the time of death used as a time-dependent variable. Results: A total of 2215 patients (mean [SD] age, 74 [8.2] years; 1751 men [79.1%]) were included in the modified intention-to-treat analysis. The total event rates for the rivaroxaban monotherapy group (1107 [50.0%]) and the combination-therapy group (1108 [50.0%]) were 12.2% (135 of 1107) and 19.2% (213 of 1108), respectively, during a median follow-up of 24.1 (IQR, 17.3-31.5) months. The mortality rate was 3.7% (41 of 1107) in the monotherapy group and 6.6% (73 of 1108) in the combination-therapy group. Rivaroxaban monotherapy was associated with a lower risk of total events compared with combination therapy (hazard ratio, 0.62; 95% CI, 0.48-0.80; P < .001). Monotherapy was an independent factor associated with a lower risk of subsequent events compared with combination therapy. The mortality risk after a bleeding event (monotherapy, 75% [6 of 8]; combination therapy, 62.1% [18 of 29]) was higher than that after a thrombotic event (monotherapy, 25% [2 of 8]; combination therapy, 37.9% [11 of 29]). Conclusions and relevance: Rivaroxaban monotherapy was associated with lower risks of total thrombotic and/or bleeding events than combination therapy in patients with AF and stable CAD. Tapered antithrombotic therapy with a sole anticoagulant should be considered in these patients. Trial registration: ClinicalTrials.gov Identifier: NCT02642419.
Article
Full-text available
Background Currently, there are no approved options to prevent or treat chemotherapy-induced thrombocytopenia (CIT). We performed a systematic literature review and meta-analysis on use of thrombopoietic agents for CIT. Patients and methods We searched Cochrane Central Register of Controlled Trials, Cochrane Database of Systematic Reviews, PubMed, EMBASE, ClinicalTrials.gov, and health technology assessments from January 1995 to March 2021 for studies evaluating thrombopoietic agents for CIT, including recombinant human thrombopoietin (rhTPO), megakaryocyte growth and development factor (MGDF), romiplostim, and eltrombopag. Random effects meta-analyses were conducted for efficacy and safety endpoints. Results We screened 1503 titles/abstracts, assessed 138 articles, and abstracted data from 39 publications (14 recombinant human thrombopoietin, 7 megakaryocyte growth and development factor, 9 romiplostim, 8 eltrombopag, and 1 romiplostim/eltrombopag). Random effects meta-analyses of data from multiple studies comparing thrombopoietic agents versus control (comparator, placebo, or no treatment) showed that thrombopoietic agents did not significantly improve chemotherapy dose delays and/or reductions (21.1% vs 40.4%, P = 0.364), grade 3/4 thrombocytopenia (39.3% vs 34.8%; P = 0.789), platelet transfusions (16.7% vs 31.7%, P = 0.111), grade ≥ 2 bleeding (6.7% vs 16.5%; P = 0.250), or thrombosis (7.6% vs 12.5%; P = 0.131). However, among individual studies comparing thrombopoietic agents with placebo or no treatment, thrombopoietic agents positively improved outcomes in some studies, including significantly increasing mean peak platelet counts (186 x 10 ⁹ /L with rhTPO vs 122 x 10 ⁹ /L with no treatment; P < 0.05) in one study and significantly increasing platelet count at nadir (56 x 10 ⁹ /L with rhTPO vs 28 x 10 ⁹ /L with not treatment; P < 0.05) in another study. Safety findings included thrombosis ( n = 23 studies) and bleeding ( n = 11), with no evidence of increased thrombosis risk with thrombopoietic agents. Conclusion Our analyses generate the hypothesis that thrombopoietic agents may benefit patients with CIT. Further studies with well-characterized bleeding and platelet thresholds are warranted to explore the possible benefits of thrombopoietic agents for CIT.
Article
Full-text available
Simple Summary One-third of patients with ovarian cancer have elevated platelet counts associated with a poor prognosis. We found that platelets increase the expression of immune checkpoint (PD-L1) in ovarian cancer in mice and patients. Reducing platelet counts or inhibiting platelet function reduced the expression of PD-L1 in tumors. We investigated the mechanism of platelet-induced PD-L1 and showed that platelets increase PD-L1 on cancer cells both directly (contact-dependent through NF-κB signaling) and indirectly (contact-independent via TFGβ released from platelets through TFGβR1/Smad signaling). Our results show that platelets dampen the antitumor immune response in the tumor microenvironment. Based on our preclinical results, we speculate that platelet counts might be a predictive biomarker for immunotherapy, i.e., patients with thrombocytosis respond better to anti-PD1/PDL-1 therapy. On the other hand, the use of aspirin or other antiplatelet reagents may impact the effectiveness of immunotherapy. These speculations need to be examined in clinical trials. Abstract The interactions between platelets and cancer cells activate platelets and enhance tumor growth. Platelets increase proliferation and epithelial–mesenchymal transition in cancer cells, inhibit anoikis, enhance the extravasation of cancer cells, and protect circulating tumor cells against natural killer cells. Here, we have identified another mechanism by which platelets dampen the immune attack on cancer cells. We found that platelets can blunt the antitumor immune response by increasing the expression of inhibitory immune checkpoint (PD-L1) on ovarian cancer cells in vitro and in vivo. Platelets increased PD-L1 in cancer cells via contact-dependent (through NF-κB signaling) and contact-independent (through TFGβR1/Smad signaling) pathways. Inhibition of NF-κB or TGFβR1 signaling in ovarian cancer cells abrogated platelet-induced PD-L1 expression. Reducing platelet counts or inhibiting platelet functions reduced the expression of PD-L1 in ovarian cancer. On the other hand, an increase in platelet counts increased the expression of PD-L1 in tumor-bearing mice.
Article
Full-text available
Among the surrounding cells influencing tumor biology, platelets are recognized as novel players as they release microvesicles (MVs) that, once delivered to cancer cells, modulate signaling pathways related to cell growth and dissemination. We have previously shown that physiological delivery of platelet MVs enriched in miR-126 exerted anti-tumor effects in different breast cancer (BC) cell lines. Here, we seek further insight by identifying AKT2 kinase as a novel miR-126-3p direct target, as assessed by bioinformatic analysis and validated by luciferase assay. Both ectopic expression and platelet MV-mediated delivery of miR-126-3p downregulated AKT2 expression, thus suppressing proliferating and invading properties, in either triple negative (BT549 cells) or less aggressive Luminal A (MCF-7 cells) BC subtypes. Accordingly, as shown by bioinformatic analysis, both high miR-126 and low AKT2 levels were associated with favorable long-term prognosis in BC patients. Our results, together with the literature data, indicate that miR-126-3p exerts suppressor activity by specifically targeting components of the PIK3/AKT signaling cascade. Therefore, management of platelet-derived MV production and selective delivery of miR-126-3p to tumor cells may represent a useful tool in multimodal therapeutic approaches in BC patients.
Article
Full-text available
Tertiary lymphoid structures (TLSs) are transient ectopic lymphoid aggregates whose formation might be caused by chronic inflammation states, such as cancer. However, how TLSs are induced in the tumor microenvironment (TME) and how they affect patient survival are not well understood. We investigated TLS distribution in relation to tumor infiltrating lymphocytes (TILs) and related gene expression in high grade serous ovarian cancer (HGSC) specimens. CXCL13 gene expression correlated with TLS presence and the infiltration of T cells and B cells, and was a favorable prognostic factor for HGSC patients. Coexistence of CD8+ T cells and B-cell lineages in the TME significantly improved the prognosis of HGSC and was correlated with the presence of TLSs. CXCL13 expression was predominantly coincident with CD4+ T cells in TLSs and CD8+ T cells in TILs, and shifted from CD4+ T cells to CD21+ follicular dendritic cells as TLS matured. In a mouse ovarian cancer model, recombinant CXCL13 induced TLSs and enhanced survival by the infiltration of CD8+ T cells. These results suggest that TLS formation was associated with CXCL13-producing CD4+ T cells and that TLSs facilitated the coordinated antitumor response of cellular and humoral immunity in ovarian cancer.
Article
Full-text available
Simple Summary Metformin is a drug commonly used to treat diabetes but it may play a role in cancer treatment. It is not known exactly how metformin acts on cancer cells and cells in the tumor microenvironment (TME). Our previous work suggested that metformin may be altering specific cells in the tumor microenvironment called cancer-associated mesenchymal stem cells (CA-MSC). The aim of this study was to build on our previous work to understand the impact of metformin on the TME. We demonstrated that when individuals with epithelial ovarian cancer received metformin in addition to chemotherapy, their tumors were less likely to have CA-MSC and T regulatory cells, which are both known to promote tumor growth, when compared to tumors from individuals who received chemotherapy alone. Additional experiments with ovarian cancer cells and tumors grown in mice suggested that metformin might be best used to prevent tumor growth rather than treat advanced stage disease. Abstract Controversy persists regarding metformin’s role in cancer therapy. Our recent work suggested metformin acts by impacting the tumor microenvironment (TME), normalizing the epigenetic profile of cancer-associated mesenchymal stem cells (CA-MSC). As CA-MSC can negatively impact tumor immune infiltrates, we evaluated metformin’s impact on the human TME, focusing on the interplay of stroma and immune infiltrates. Tumor samples from (i) 38 patients treated with metformin and chemotherapy and (ii) 44 non-metformin matched controls were included in a tissue microarray (TMA). The TMA was used to compare the presence of CA-MSC, desmoplasia and immune infiltrates in the TME. In vitro and in vivo models examined metformin’s role in alteration of the CA-MSC phenotype. The average percentage of CA-MSC was significantly lower in metformin-treated than in chemotherapy alone-treated tumors (p = 0.006). There were fewer regulatory T-cells in metformin-treated tumors (p = 0.043). Consistent with CA-MSC’s role in excluding T-cells from tumor islets, the T-cells were primarily present within the tumor stroma. Evaluation of metformin’s impact in vitro suggested that metformin cannot reverse a CA-MSC phenotype; however, the in vivo model where metformin was introduced prior to the establishment of the CA-MSC phenotype supported that metformin can partially prevent the reprogramming of normal MSC into CA-MSC. Metformin treatment led to a decrease in both the presence of protumorigenic CA-MSC and in immune exclusion of T cells, leading to a more immune-permissive environment. This suggests clinical utility in prevention and in treatment for early-stage disease and putatively in immune therapy.
Article
Full-text available
Programmed death-ligand 1 (PD-L1) is an immune checkpoint protein that suppresses cytotoxic T lymphocytes and is often overexpressed in cancers. Due to favorable clinical trial results, immune checkpoint inhibition (ICI) is now part of Food and Drug Administration-approved immuno-oncology therapies; however, not all patients derive benefit from ICI therapy. High blood platelet-to-lymphocyte ratio has been associated with failure of ICI treatment, but whether platelets have a role in hindering ICI response is not clear. Here, we report that co-culturing platelets with breast and lung cancer cell lines increased protein and gene expression of tumor cell PD-L1. Platelet-induced tumor cell PD-L1 upregulation was reduced by anti-platelet agents, such as aspirin and ticagrelor. Platelet cytokine arrays revealed that the well-established cytokines, including interferon-gamma, were not the main regulators of platelet-mediated PD-L1 upregulation. Instead, the high molecular weight epidermal growth factor (HWM-EGF) is abundant in platelets, which caused an upregulation of tumor cell PD-L1. Both an EGF neutralizing antibody and the drug, Cetuximab (EGF-receptor neutralizing antibody) inhibited platelet-induced increases in tumor cell PD-L1, suggesting that platelets induce tumor cell PD-L1 in an EGFR-dependent manner. Our data reveal a novel mechanism for platelets in tumor immune escape and warrant further investigation to determine if targeting platelets improves ICI therapeutic responses.
Article
Full-text available
T cell-based immunotherapy, for example, with T cell- recruiting bispecific antibody (bsAb), has revolutionized oncological treatment. However, many patients do not respond to treatment, and long-term remissions are still rare. Several tumor immune evasion mechanisms have been reported to counteract efficiency of T cell- engaging therapeutics. Platelets largely affect cancer pathophysiology by mediating tumor invasion, metastasis, and immune evasion. On treatment of patients in a clinical trial with a PSMA×CD3 bsAb (NCT04104607), we observed profound treatment-associated platelet activation, mirrored by a decrease of total platelet count. On modeling the treatment setting, we found that platelet activation significantly reduced bsAb-mediated CD4+ and CD8+ T-cell reactivity as revealed by impaired T-cell degranulation, secretion of perforin, and ultimately, inhibition of target cell lysis. This effect occurred in a transforming growth factor beta (TGF-β)-dependent manner and was not restricted to PSMA×CD3 bsAb, but rather observed with various CD3-directed bispecific constructs, including the approved CD19×CD3 bsAb blinatumomab. BsAb-mediated T-cell reactivity could be restored by platelet inhibition and specifically by blocking the TGF-β axis. Together, our findings demonstrate that platelets undermine the efficacy of T cell-recruiting bsAb and identify modulation of platelet function as a means to reinforce the effectiveness of bsAb treatment.
Article
Platelets, the often-overlooked component of the immune system, have been shown to promote tumor growth. Non-alcoholic fatty liver disease (NAFLD) is a common disease in the Western world and rising risk for hepatocellular carcinoma (HCC). Unexpectedly, we observed that platelets can inhibit the growth of established HCC in NAFLD mice. Through pharmacological inhibition and genetic depletion of P2Y12 as well as in vivo transfusion of wild-type (WT) or CD40L−/− platelets, we demonstrate that the anti-tumor function of platelets is mediated through P2Y12-dependent CD40L release, which leads to CD8⁺ T cell activation by the CD40 receptor. Unlike P2Y12 inhibition, blocking platelets with aspirin does not prevent platelet CD40L release nor accelerate HCC in NAFLD mice. Similar findings were observed in liver metastasis models. All together, our study reveals a complex role of platelets in tumor regulation. Anti-platelet treatment without inhibiting CD40L release could be considered for liver cancer patients with NAFLD.