Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3490 2023-03-09 16:43:17 |
2 layout & references Meta information modification 3490 2023-03-10 01:49:44 | |
3 acronym explained + 1 word(s) 3491 2023-03-13 09:51:47 |

Video Upload Options

We provide professional Video Production Services to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Oncul, S.; Cho, M.S. Interactions of Platelets with TME in Ovarian Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/42034 (accessed on 21 December 2024).
Oncul S, Cho MS. Interactions of Platelets with TME in Ovarian Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/42034. Accessed December 21, 2024.
Oncul, Selin, Min Soon Cho. "Interactions of Platelets with TME in Ovarian Cancer" Encyclopedia, https://encyclopedia.pub/entry/42034 (accessed December 21, 2024).
Oncul, S., & Cho, M.S. (2023, March 09). Interactions of Platelets with TME in Ovarian Cancer. In Encyclopedia. https://encyclopedia.pub/entry/42034
Oncul, Selin and Min Soon Cho. "Interactions of Platelets with TME in Ovarian Cancer." Encyclopedia. Web. 09 March, 2023.
Interactions of Platelets with TME in Ovarian Cancer
Edit

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 (TME) 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.

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, mucinous, 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]. Non-epithelial ovarian cancers (e.g., germ cell and sex cord–stromal) constitute approximately 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]. Additional 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 associated with high mortality because of late diagnosis due to its anatomical location and mostly indistinguishable symptoms, high recurrence rate [2], primary or acquired resistance 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 principal 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]. Membrane proteins on activated platelets, including P-selectin [16], GPIbα [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. Additionally, 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. The interactions between platelets and the TME components of ovarian cancer are illustrated in Figure 1.
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.
Healthy individuals have platelet counts of between 150,000 and 450,000 per microliter 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 chemotherapy [30]. Aside from contributing to the formation of venous thromboembolisms, platelets contribute to cancer progression via distinct mechanisms, including increasing proliferation [31], epithelial–mesenchymal transition (EMT) [32], and anoikis resistance in cancer cells [33]; promoting the formation of the premetastatic niche and metastasis [33]; enhancing angiogenesis [27] and the integrity of tumor vasculature [34]; inducing immune tolerance [35]; and reducing the impact of chemotherapy [30].
Table 1. The major components reside on activated platelets or are released from platelet granules.

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 proliferation 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 endothelial 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 vasculature, 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 leukocyte rolling in activated endothelial cells [91] and the generation of platelet—cancer cell complexes [92]. Adhesion molecules, including integrins, von Willebrand factor (vWF), fibrinogen, 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 factors 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 P2Y2 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 involvement 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 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.
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. Additionally, 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 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 lymphatic 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 lymphangiogenesis 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 association 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 microvessels [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 antiangiogenic agents such as bevacizumab may interfere with the incorporation of pericytes into new blood vessels [131].
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 presumably 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 facilitate 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 α11 is a CAF marker, and its expression is related to myofibroblast differentiation and ECM alteration. CAFs expressing integrin α11 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 tumor 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-1α)-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 progression 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, tumor growth, and ECM reorganization, mainly by mediating STAT3 phosphorylation [157]. 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 mesothelial 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 various 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.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2022, 72, 7–33.
  2. Jelovac, D.; Armstrong, D.K. Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J. Clin. 2011, 61, 183–203.
  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.
  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.
  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.
  6. Jin, M.-Z.; Jin, W.-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 166.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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 αIIbβ3 (GPIIb/IIIa) with ανβ3 Integrin Mediates Interaction of Melanoma Cells with Platelets. J. Biol. Chem. 2012, 287, 2168–2178.
  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.
  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.
  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.
  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.
  23. Pavlovic, N.; Rani, B.; Gerwins, P.; Heindryckx, F. Platelets as Key Factors in Hepatocellular Carcinoma. Cancers 2019, 11, 1022.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  32. Guo, Y.; Cui, W.; Pei, Y.; Xu, D. Platelets promote invasion and induce epithelial to mesenchymal transition in ovarian cancer cells by TGF-β signaling pathway. Gynecol. Oncol. 2019, 153, 639–650.
  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.
  34. Davis, A.N.; Afshar-Kharghan, V.; Sood, A.K. Platelet Effects on Ovarian Cancer. Semin. Oncol. 2014, 41, 378–384.
  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.
  36. Saboor, M.; Ayub, Q.; Ilyas, S. Moinuddin Platelet receptors; an instrumental of platelet physiology. Pak. J. Med. Sci. 2013, 29, 891–896.
  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.
  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.
  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.
  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.
  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.
  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.
  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 Ibα supports experimental lung metastasis. Proc. Natl. Acad. Sci. USA 2007, 104, 9024–9028.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  60. Fitch-Tewfik, J.L.; Flaumenhaft, R. Platelet Granule Exocytosis: A Comparison with Chromaffin Cells. Front. Endocrinol. 2013, 4, 77.
  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.
  62. Kim, S.; Garcia, A.; Jackson, S.P.; Kunapuli, S.P. Insulin-like growth factor-1 regulates platelet activation through PI3-Kα isoform. Blood 2007, 110, 4206–4213.
  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.
  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.
  65. Walsh, P.N. Platelets and coagulation proteins. Fed. Proc. 1981, 40, 2086–2091.
  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.
  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.
  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.
  69. Santos-Martínez, M.J.; Medina, C.; Jurasz, P.; Radomski, M.W. Role of metalloproteinases in platelet function. Thromb. Res. 2008, 121, 535–542.
  70. Lishko, V.K.; Yakubenko, V.P.; Ugarova, T.P.; Podolnikova, N.P. Leukocyte integrin Mac-1 (CD11b/CD18, αMβ2, CR3) acts as a functional receptor for platelet factor 4. J. Biol. Chem. 2018, 293, 6869–6882.
  71. Walz, A.; Dewald, B.; von Tscharner, V.; Baggiolini, M. Effects of the neutrophil-activating peptide NAP-2, platelet basic protein, connective tissue-activating peptide III and platelet factor 4 on human neutrophils. J. Exp. Med. 1989, 170, 1745–1750.
  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.
  73. Chen, Y.; Zhong, H.; Zhao, Y.; Luo, X.; Gao, W. Role of platelet biomarkers in inflammatory response. Biomark. Res. 2020, 8, 28.
  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.
  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.
  76. Kim, H.; Conway, E.M. Platelets and Complement Crosstalk in Early Atherogenesis. Front. Cardiovasc. Med. 2019, 6, 131.
  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.
  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.
  79. de Jong, J.S.S.G.; Dekker, L.R.C. Platelets and Cardiac Arrhythmia. Front. Physiol. 2010, 1, 166.
  80. Chesney, C.M.; Harper, E.; Colman, R.W. Human Platelet Collagenase. J. Clin. Investig. 1974, 53, 1647–1654.
  81. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770.
  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.
  83. Klein, D. The Tumor Vascular Endothelium as Decision Maker in Cancer Therapy. Front. Oncol. 2018, 8, 367.
  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.
  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.
  86. Dvorak, H.F.; Brown, L.F.; Detmar, M.; Dvorak, A.M. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am. J. Pathol. 1995, 146, 1029–1039.
  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.
  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 Epithelial Ovarian Cancer or Primary Peritoneal Cancer: A Gynecologic Oncology Group Study. J. Clin. Oncol. 2007, 25, 5165–5171.
  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.
  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.
  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.
  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.
  93. Sang, Y.; Roest, M.; de Laat, B.; de Groot, P.G.; Huskens, D. Interplay between platelets and coagulation. Blood Rev. 2020, 46, 100733.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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 GPIbα far exceeds that of its main ligand, von Willebrand factor, in arterial thrombosis. Proc. Natl. Acad. Sci. USA 2006, 103, 16900–16905.
  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.
  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.
  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.
  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.
  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.
  113. Da, M.-X.; Wu, Z.; Tian, H.-W. Tumor Lymphangiogenesis and Lymphangiogenic Growth Factors. Arch. Med. Res. 2008, 39, 365–372.
  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.
  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.
  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.
  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.
  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.
  120. Sweeney, M.; Ayyadurai, S.; Zlokovic, B.V. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat. Neurosci. 2016, 19, 771–783.
  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.
  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.
  123. Bergers, G.; Song, S. The role of pericytes in blood-vessel formation and maintenance. Neuro-Oncology 2005, 7, 452–464.
  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.
  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.
  126. Armulik, A.; Genové, G.; Betsholtz, C. Pericytes: Developmental, Physiological, and Pathological Perspectives, Problems, and Promises. Dev. Cell 2011, 21, 193–215.
  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.
  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.
  130. Heldin, C.-H.; Westermark, B. Mechanism of Action and In Vivo Role of Platelet-Derived Growth Factor. Physiol. Rev. 1999, 79, 1283–1316.
  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.
  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.
  133. Bagavandoss, P.; Wilks, J. Specific inhibition of endothelial cell proliferation by thrombospondin. Biochem. Biophys. Res. Commun. 1990, 170, 867–872.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  143. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598.
  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.
  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.
  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.
  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.
  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.
  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.
  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. α11β1 Integrin is Induced in a Subset of Cancer-Associated Fibroblasts in Desmoplastic Tumor Stroma and Mediates In Vitro Cell Migration. Cancers 2019, 11, 765.
  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.
  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.
  153. Rother, E.; Brandl, R.; Baker, D.L.; Goyal, P.; Gebhard, H.; Tigyi, G.; Siess, W. Subtype-Selective Antagonists of Lysophosphatidic Acid Receptors Inhibit Platelet Activation Triggered by the Lipid Core of Atherosclerotic Plaques. Circulation 2003, 108, 741–747.
  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-1α from human mesenchymal stem cells. Exp. Mol. Med. 2010, 42, 280–293.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  170. Ghoneum, A.; Afify, H.; Salih, Z.; Kelly, M.; Said, N. Role of tumor microenvironment in ovarian cancer pathobiology. Oncotarget 2018, 9, 22832–22849.
  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.
  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.
More
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 534
Revisions: 3 times (View History)
Update Date: 13 Mar 2023
1000/1000
Video Production Service