7

Platelets in Hematogenous Breast Cancer Metastasis: Partners in Crime

Savannah R. Free Kermit L. Carraway III

Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, CA, USA

Abstract: Distant metastasis is the primary driver of breast cancer-associated mortality, and research into the mechanisms underlying hematogenous tumor cell dissemination could give rise to the development of novel and more effective therapeutic agents and strategies. Platelets are activated directly by tumor cell interaction and indirectly by tumor-secreted factors to trigger platelet aggregation, degranulation, and the subsequent release of pro-tumorigenic factors. Platelet presence within the primary tumor, bloodstream, and metastatic sites allows for continuous exposure of breast cancer cells to these factors, making platelets a powerful partner in tumor cell dissemination. Platelet-tumor cell crosstalk contributes to hematogenous breast cancer metastasis by providing physical and biochemical support to metastasizing cells via mechanisms including protection from shear forces, anoikis, and immune attack, and enhancement of angiogenesis, migration, and pro-tumorigenic inflammation. Here, we review platelets and their many benefits to metastatic breast cancer, their role in facilitating paraneoplastic thrombosis, and current research regarding their potential as a breast cancer therapeutic target.

Keywords: antiplatelet therapy; breast cancer; hematogenous metastasis; paraneoplastic thrombosis; platelets in breast cancer

Author for correspondence: Kermit Carraway III, Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, CA, USA. Email: klcarraway@ucdavis.edu

Cite this chapter as: Free SR, Carraway KL III. Platelets in Hematogenous Breast Cancer Metastasis: Partners in Crime. In: Mayrovitz HN. editor. Breast Cancer. Brisbane (AU): Exon Publications. Online first 10 Jun 2022.

Doi: https://doi.org/10.36255/exon-publications-breast-cancer-platelets

In: Mayrovitz HN, editor. Breast Cancer. Brisbane (AU): Exon Publications. ISBN: 978-0-6453320-3-2. Doi: https://doi.org/10.36255/exon-publications-breast-cancer

Copyright: The Authors.

License: This open access article is licenced under Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/

INTRODUCTION

One hundred and fifty years ago, Armand Trousseau first drew the insightful connection between platelets and cancer when he noted an increased incidence of thrombosis in patients with visceral cancer (1). Ninety years later, Gabriel and Tatiana Gasic observed that neuraminidase, a potent inducer of thrombocytopenia, reduced pulmonary metastasis in mouse models of cancer (24). Since then, the contribution of platelets to the progression and dissemination of cancer has been established across cancer types and tissues of origin. In breast cancer (BC) specifically, where patients with advanced, distantly metastatic disease face a 5-year survival rate of only 30% (5), the ability of platelets to support tumor cell survival and aggressiveness throughout the metastatic cascade is an important area of research and therapeutic consideration. Furthermore, high co-incidence of coagulopathies and cancer—acknowledged by Trousseau—poses an additional risk to patients and may be mitigated by platelet-directed therapies (6, 7). Herein, we review the role of platelets in promoting growth and invasion in primary breast tumors, enhancing circulating tumor cell (CTC) survival and extravasation from the bloodstream, enabling the seeding and growth of distant metastatic lesions, and fostering BC-associated coagulopathy.

PLATELETS

With their tremendous abundance (1.5–3.5 × 108/mL of human blood) and unique ability to rapidly aggregate and release hundreds of factors in response to activation, platelets are the first responders to arrive on the scene of vascular injury and begin the process of clot formation and healing (8). Though small (~2 μm in diameter) and anucleate, platelets contain ribosomes, mitochondria, a cytoskeletal network, in addition to distinct α-granules, lysosomal granules, and dense-granules, which store biologically active factors essential to injury response and hemostatic maintenance (9). These factors can be fully synthesized within platelet precursor megakaryocytes, acquired by receptor-mediated endocytosis or pinocytosis, or even synthesized within the platelets themselves using pre-mRNA generated in megakaryocytes; this allows for flexibility and diversity in platelet storage contents (9, 10). Release of granular contents is mediated by platelet activation and degranulation, which can be triggered by a variety of agonists (e.g., adhesive proteins such as Von Willebrand Factor (VWF) in the vascular wall, soluble factors such as thrombin, adenosine diphosphate (ADP), or thromboxane A2 (TXA2)) (8).

Unfortunately, platelets are unable to discern between vascular injury and cancer, which has been described as the “wound that does not heal” (11). Cancer cells secrete several platelet agonists, including ADP and TXA2, which induce constitutive reciprocal signaling with platelets that not only supports cancer growth and metastasis, but also induces a hypercoagulative state throughout the body (12). Cancer-specific signals stimulate changes in nearby platelet storage contents, creating “tumor-educated platelets” that may be useful as surrogate biomarkers for identifying tumor type, location, and even mutational profile (13, 14).

PLATELETS AND THE PRIMARY TUMOR

Primary breast tumor cells are exposed to platelets and platelet signals, as demonstrated by robust platelet marker CD42b staining of patient BC samples at the tumor’s leading edge (15, Figure 1A). As a result, platelets secrete factors within the primary tumor compartment which contribute to tumor cell survival, proliferation, and aggressiveness (discussed below). Accordingly, retrospective analysis of tumor biopsies from BC patients who subsequently received treatment and neo-adjuvant chemotherapy revealed that patients with CD42b-rich tumors were significantly less likely to achieve pathological complete response to chemotherapy (15).

Fig 1

Figure 1. Platelets aid BC cells throughout the metastatic cascade. A, Platelets help primary tumor cells migrate and intravasate by secreting factors that promote epithelial-to-mesenchymal transition and remodeling of the extracellular matrix (ECM; (20, 23)). B, Platelets protect BC cells from shear-induced apoptosis and encourage resistance to anoikis caused by loss of ECM attachment (55, 58, 59). C, Platelets contribute to BC immune evasion by directly inhibiting local immune cell activation, masking immune ligands on tumor cells, and conferring “pseudo-expression” of immune-inactivating molecules to tumor cells (26, 31, 34, 6064). D, Platelets support arrest and trans-endothelial migration of BC cells and help establish the pre-metastatic niche by recruiting granulocytes to the area (7275). Figure created with biorender.com.

Tumor growth and dissemination are dependent on the acquisition of intratumor vasculature to provide oxygen and nutrients to rapidly dividing cancer cells. Angiogenesis, the process by which tumors create new or co-opt existing endothelial vasculature, is mediated by a complex balance of numerous pro- and anti-angiogenic factors (16). Platelets are an important source of these factors, which they store in α-granules until agonists trigger their release. When cancer cells secrete agonists such as ADP and thrombin, nearby platelets are activated and release pro-angiogenic factors including vascular endothelial growth factor (VEGF), angiopoietin-1, and tumor necrosis factor-alpha (TNF-α) (17). Platelet-depleted tumors show hyperpermeable vasculature and poor tumor perfusion due to increased angiopoeitin-2 levels and diminished pericyte recruitment to hemorrhagic blood vessels (18). On the other hand, tumor cell-activated platelets enhance endothelial tube formation in vitro, and increase tumor vascularization and metastatic burden in vivo (19, 20). Patients with BC display significantly increased levels of systemic platelet-sequestered angiogenic factors VEGF, platelet-derived growth factor (PDGF), and transforming growth factor-beta (TGF-β; Figure 2) compared to healthy controls; this upregulation is correlated with higher incidence of lymph node metastasis and advanced disease stage (21).

To disseminate, tumor cells must invade the surrounding tissue and extracellular matrix (ECM) and penetrate the vasculature (Figure 1A). In one form of metastatic progression, tumor cells undergo epithelial-to-mesenchymal transition (EMT), wherein single epithelial tumor cells acquire invasive mesenchymal morphology and behavior (22). Breast tumors showing greater platelet presence tend to show more EMT-like characteristics (e.g., loss of apical-basal polarity, loss of E-cadherin expression, gain of vimentin expression) than those with less platelet activity (15). This is mediated largely through increased concentrations of intratumor TGF-β, which is abundantly secreted via platelet α-granules and activates transcription of EMT-related genes across cancer types (20, 23, 24). Platelets also engage EMT through direct interaction with cancer cell α2β1 surface integrins, which activates the Wnt-β-catenin pathway and upregulates autocrine TGF-β production by cancer cells (25). Furthermore, platelets constitutively express the cell surface TGF-β-docking Glycoprotein A Repetitions Predominant (GARP) receptor, which is essential for the conversion of TGF-β from its latent to active form and increases the pool of active TGF-β within the primary tumor (Figure 2) (26).

Fig 2

Figure 2. Platelets increase the local pool of active TGF-β. A, Platelets are the single greatest source of TGF-β in blood plasma, secreting huge amounts of latent TGF-β (LTGF-β) complexes in α-granules upon activation, which are then processed and activated by factors found on platelets and other cell types as shown in B (24). B, Platelets constitutively express the “Glycoprotein A Repetitions Predominant” (GARP) receptor, which docks LTGF-β (derived from platelets or other cell types) for activation. This substantially increases the local reservoir of active TGF-β (26). C, Direct contact between platelets and tumor cells via the cancer cell surface integrin α2β1 stimulates the Wnt-β-Catenin signaling pathway. This upregulates downstream transcription of tgfb1, resulting in increased TGF-β secretion by tumor cells (25). Active TGF-β secreted by platelets and tumor cells is bound by the TGF-β receptor on tumor cells, driving SMAD phosphorylation and downstream signaling that increases transcription of genes related to EMT, ECM remodeling, and more. TGF-βR, TGF-β receptor; pSMADs, phosphorylated “Suppressor of Mothers against Decapentaplegic” factors. Figure created with biorender.com

Whether mediated by EMT or collective cell migration (CCM), in which cells retain epithelial characteristics and invade as a group (27), remodeling of the ECM in the primary tumor compartment is an essential step in metastasis and is stimulated by bidirectional signaling between platelets and tumor cells (20). Platelet-stimulated paracrine and autocrine TGF-β signaling upregulates BC cell expression of matrix metalloproteinases-2 and -9 (MMP-2 and -9) (18, 20, 25), plasminogen activator inhibitor-1 (PAI-1) (18), and various proteases (18, 20, 25), which encourage the degradation of the ECM to enable cellular migration and invasion. Conversely, sustained TGF-β activity enhances tumor cell production of pro-tumorigenic ECM molecules like fibrin, fibronectin, and collagen, which enable cellular communication and migration (20, 25, 28).

To survive and spread, BC cells must evade immune detection and destruction (29). Many cancers avoid immunosurveillance by acquiring expression of programmed death-ligand 1 (PD-L1), which negatively regulates CD8+ T cell function; as a result, PD-L1 checkpoint blockade treatments have become an attractive therapeutic approach in recent years (30). Zaslavsky et al. observed that platelets express PD-L1—particularly after activation by tumor cells—and can confer PD-L1 “pseudo-expression” to PD-L1-/- mouse tumors, resulting in decreased T cell-driven cytotoxicity and increased tumor burden (31). Likewise, patients with PD-L1-negative lung cancer who responded to PD-L1 checkpoint inhibition tended to have greater platelet infiltration in biopsied tumors, suggesting that platelet presence sensitizes PD-L1-deficient cancers to PD-L1-directed therapies, though these results need to be confirmed on a larger cohort of patient samples (31). Other groups have observed that platelets also suppress cytotoxic T cell function in the tumor microenvironment through platelet-GARP driven activation of TGF-β (Figure 2B) (26), which inhibits T cell proliferation and activity through a diverse array of mechanisms reviewed by Gorelik and Flavell (32) and Thomas and Massagué (33). Furthermore, platelets provide well-documented protection of tumor cells from natural killer (NK) cells (34). Though this phenomenon likely exists within the primary tumor, it has been most thoroughly documented in circulating tumor cells (CTCs) and will be described in more detail below.

PLATELETS AND CIRCULATING TUMOR CELLS

BC cells metastasize to distant organs by directly intravasating into blood vessels or indirectly entering into the lymphatic system which drains into the bloodstream (35). Upon hematogenous entry, an array of threats create a survival bottleneck for metastatic cells, and very few CTCs survive and successfully seed metastases (36). To survive these dangers, CTCs rapidly recruit platelets to provide physical protection and biochemical enhancements (37). In fact, platelets are the first cells in circulation to adhere to CTCs, forming microthrombi in seconds which then recruit fibrin, neutrophils, monocytes, and macrophages for additional aid (38).

Under normal physiologic conditions, platelets are recruited to sites of injury upon disruption of the basement membrane, which exposes subendothelial pro-coagulation factors (e.g., tissue factor (TF), VWF, collagen) typically unavailable in circulation (8). CTCs co-opt platelet activity by expressing these same factors. For example, widespread TF overexpression by cancer cells is driven by signaling programs associated with oncogenic transformation and EMT (39, 40). As a receptor for key components of the coagulation cascade, TF expression by cancer cells encourages localized thrombus formation and is associated with increased metastatic success and worse patient outcomes (41, 42). Likewise, aberrant expression of the adhesive glycoprotein VWF has been observed in BC (43) and other cancer types (44, 45). Tumor cell VWF secretion increases overall plasma VWF concentrations, contributing to paraneoplastic hypercoagulopathy; furthermore, tumor cells can directly bind VWF via αvβ3 integrins (46) or glycoprotein Ib/V/IX complexes (47), allowing them to self-aggregate, adhere to platelets, and arrest under flow conditions (45, 46).

In addition to the indirect modes of platelet-CTC aggregation described above, where cells need not interact directly but aggregate through mutual binding of coagulation factors, direct platelet-CTC binding can occur through a variety of receptor-ligand interactions. Platelet P-selectin interaction with CTC mucins (4850), platelet glycoprotein VI (GPVI) interaction with CTC galectin-3 (51), and platelet receptor FcγRIIa interaction with CTC immunoglobulin G (IgG) (52) have all been described (53). While these unique receptor-ligand dynamics present enticing therapeutic targets for inhibition of platelet-CTC interactions, the sheer number of molecules which can redundantly promote CTC binding and platelet activation may thwart these attempts.

BC cells in circulation are susceptible to anoikis, a type of programmed cell death triggered by loss of attachment to the ECM (54). Secreted platelet proteins promote anoikis resistance by upregulating CTC expression of the GTPase RhoA, thereby activating the transcription co-activator YAP1 and inducing expression of downstream genes involved in proliferation and apoptotic resistance (55). It is also widely believed that platelets physically guard CTCs from the shear forces of the bloodstream, which can induce cell cycle arrest (56), apoptosis, and necrosis (57). Although limited experimental evidence exists to support platelet-mediated shear force protection (58), in silico modeling of CTC-platelet interactions under shear indicates that platelets may reduce the total shear force magnitude applied to any single region of a tumor cell, decreasing deformation and damage of the membrane and preventing apoptosis (59, Figure 1B).

Perhaps the most widely studied benefit conferred by platelets to tumor cells is NK cell evasion, which is achieved by manipulating the behavior of both tumor cells and the NK cells themselves. Kopp and colleagues observed that platelet-secreted TGF-β downregulates expression of the activating immunoreceptor NKG2D by NK cells, diminishing their cytotoxic effects (60). Platelets also impair NK cell production of interferon gamma (IFNγ), which is a key effector molecule through which NK cells induce an adaptive immune response (61, 62). In addition, platelets release sheddases ADAM10 and ADAM17 which cleave NK ligands from the surface of tumor cells and prevent them from being targeted (63). Platelets can also confer “pseudo-expression” of the major histocompatibility complex (MHC) class I to tumor cells, which allows tumor cells to avoid NK targeting without triggering T-cell surveillance (64, Figure 1C).

Soon after entry into the bloodstream, CTCs travel to distant sites where they arrest and begin to extravasate. Vascular arrest occurs through both passive means (i.e., size-restricted arrest, where CTCs are physically trapped within small capillaries) (65) and active mechanisms (i.e., adhesion to the endothelium or subendothelial matrix) (66, 67); platelets have been implicated in both processes, though their roles are controversial (68). While platelets have been observed in CTC clusters mechanically arrested by size-restriction, it is unclear whether platelets are necessary for this entrapment, or if they simply pile up around CTCs that are blocking their passage (69). Likewise, direct CTC binding to both the endothelium (70) and the subendothelial matrix (67) has been noted even in the absence of preliminary platelet thrombus formation, suggesting that platelets are not strictly necessary for initial arrest. However, platelets have been implicated in tumor cell tethering, rolling, and adhesion to the endothelium via P-selectin interactions (71), suggesting that they may provide stability to CTC attachment.

PLATELETS AND DISTANT METASTASIS

Regardless of their role in vascular arrest, platelets significantly bolster the process of extravasation. In addition to the migratory advantage platelet-induced TGF-β signaling confers to tumor cells (23), platelets potentiate tumor cell-induced endothelial retraction (72) and induce the loss of endothelial tight junctions (73). These changes support trans-endothelial migration by tumor cells, which is further evidenced by observations that platelet inhibition significantly reduces tumor cell extravasation (73, Figure 1D).

Because they are rare and transient in vivo and difficult to model in vitro, relatively little is known about the initial stages of BC metastatic colonization, and specifically how these events are impacted by platelets. It is likely that many of the same platelet-tumor cell signaling programs that occur during primary tumor growth and intravasation are useful to disseminated tumor cells (e.g., platelet-induced tumor cell MMP secretion for ECM degradation, platelet expression of pro-angiogenic factors, etc.), though few of these dynamics have been demonstrated specifically during early metastatic colonization. On the other hand, platelets have been specifically implicated as early founders of the “pre-metastatic niche”. Labelle et al. observed that granulocytes are recruited to the vicinity of CTC-platelet aggregates arrested in the vasculature, where they help prepare a microenvironment amenable to tumor cell seeding (74, Figure 1D). Granulocyte recruitment is dependent on chemokine signaling by activated platelets, and metastatic success is significantly attenuated by depletion of either cell type. Another study found that by inhibiting platelet activation, deposition of fibronectin in metastatic sites in the lung was significantly decreased, resulting in a less hospitable metastatic environment and fewer lesions (75). These results support the notion that while platelets may be dispensable for initial vascular arrest, they are necessary for successful vascular retention and metastatic outgrowth (68).

PLATELETS AND PARANEOPLASTIC COAGULOPATHY

BC cells express high levels of pro-coagulant factors like TF and phosphatidylserine on their surfaces, or secrete them in extracellular vesicles (76). BC is also associated with increased platelet counts overall, likely stimulated by increased cytokine-driven thrombopoiesis, creating a positive feedback loop of platelet hyperactivation and systemic hypercoagulation (77). Consequently, patients with BC are at significantly higher risk than healthy patients of venous and arterial thromboembolism, myocardial infarction, and ischemic stroke, particularly following chemotherapeutic treatment (7). In fact, thromboembolic complications are one of the leading causes of death in patients undergoing chemotherapy (78). To mitigate the thrombotic risk associated with cancer and cancer treatment, clinicians regularly prescribe prophylactic anticoagulants like heparin as a supplement to cancer treatment regimens (79).

PLATELETS AS A THERAPEUTIC TARGET IN BREAST CANCER

Given their well-established localization to and support of BC metastases, platelets are an attractive anti-cancer target. In one approach, platelets are directly inhibited through antagonism of platelet receptors (e.g., P-selectin, GPVI, P2Y12) via antibodies or small molecules (80). Numerous such drugs exist and several have been approved for the treatment of cardiovascular disease (81), though their use in BC treatment is largely limited to pre-clinical studies. However, administration of anti-platelet agents alone or in combination with standard-of-care therapies has shown promise in metastatic prevention across studies and cancer types, as reviewed by Xu et al. (80).

One anti-platelet drug that has been thoroughly studied in the context of cancer is aspirin. Aspirin is a cyclooxygenase inhibitor that acts by inhibiting synthesis of platelet prostanoids, which promote activation of other nearby platelets. Aspirin treatment prevents BC cell-induced platelet activation and subsequent release of pro-tumorigenic factors in vitro (82), and leads to reduced metastasis in an in vivo mouse model of metastatic BC by suppressing platelet-enhanced anoikis resistance (83). In the clinic, observational studies of BC patients prescribed aspirin after cancer diagnosis show mixed effects on patient outcomes, though meta-analyses indicate that aspirin use is associated with reduced risk of BC death (84). Strikingly, in randomized controlled trials of aspirin for the prevention of cardiovascular disease, allocation to the aspirin group reduced the incidence of distantly metastatic adenocarcinoma by nearly half during trial follow-ups (85). This translated to significantly higher patient survival, regardless of whether the trials tested high or low-dose aspirin. These results suggest that low-dose aspirin may be useful in the prevention of distant metastasis, though therapeutic success may depend on preventative rather than responsive administration.

In another therapeutic approach, platelet localization to the tumor is exploited to allow enhanced delivery of therapeutic molecules into the intratumoral space. Bahmani et al. found that by encapsulating the toll-like receptor agonist resiquimod in platelet membrane-coated nanoparticles (PNPs), they enhanced uptake and retention of the drug in the tumors of a 4T1 mouse BC model, increasing intratumor immunity and decreasing tumor growth better than the drug alone (86). PNPs have also been used in vitro to effectively deliver chemotherapeutic agents, siRNAs, and photosensitizers to primary tumors and CTCs (87). Though PNPs are not currently being used in the clinic, they present a promising new method to increase the efficacy of existing therapeutics.

CONCLUSION

Platelets are essential partners to BC cells throughout the course of hematogenous metastasis. Through a multitude of reciprocal signaling events, tumor cells activate platelets, encouraging their aggregation and degranulation. In return, platelets secrete factors which enhance tumor cell survival and aggressiveness. This crosstalk threatens patients not only by its ability to foster metastatic disease, but also in its induction of a systemwide hypercoagulative state. Therapies which target the platelet-tumor cell interaction have the potential to mitigate the danger of BC metastasis, while also preventing cancer-associated thromboembolism. Therapeutic options for the prevention and treatment of distantly metastatic BC will be increased as researchers work to better understand and target the unique mechanisms of cancer-platelet cooperation.

Acknowledgement: We thank Michelle Hu for her comments and insights on this chapter. This work was supported by NIH grants CA230742 and CA250211 (K.L.C.).

Conflict of Interest: The authors declare no potential conflict of interest with respect to research, authorship and/or publication of this chapter.

Copyright and Permission Statement: The authors confirm that the materials included in this chapter do not violate copyright laws. Where relevant, appropriate permissions have been obtained from the original copyright holder(s), and all original sources have been appropriately acknowledged or referenced.

REFERENCES

  1. Metharom P, Falasca M, Berndt M. The History of Armand Trousseau and Cancer-Associated Thrombosis. Cancers. 2019;11(2):158. https://doi.org/10.3390/cancers11020158
  2. Gasic G, Gasic T. Removal of Sialic Acid from the cell coat in tumor cells and vascular endothelium, and its effects on metastasis. Proc Natl Acad Sci. 1962 Jul;48(7):1172–7. https://doi.org/10.1073/pnas.48.7.1172
  3. Gasic GJ, Gasic TB, Stewart CC. Antimetastatic effects associated with platelet reduction. Proc Natl Acad Sci U S A. 1968 Sep;61(1):46–52. https://doi.org/10.1073/pnas.61.1.46
  4. Gasic GJ, Gasic TB, Galanti N, Johnson T, Murphy S. Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease. Int J Cancer. 1973 May 15;11(3):704–18. https://doi.org/10.1002/ijc.2910110322
  5. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022 Jan;72(1):7–33. https://doi.org/10.3322/caac.21708
  6. Navi BB, Reiner AS, Kamel H, Iadecola C, Okin PM, Elkind MSV, et al. Risk of Arterial Thromboembolism in Patients With Cancer. J Am Coll Cardiol. 2017 Aug;70(8):926–38. https://doi.org/10.1016/j.jacc.2017.06.047
  7. Walker AJ, West J, Card TR, Crooks C, Kirwan CC, Grainge MJ. When are breast cancer patients at highest risk of venous thromboembolism? A cohort study using English health care data. Blood. 2016 Feb 18;127(7):849–57. https://doi.org/10.1182/blood-2015-01-625582
  8. Li Z, Delaney MK, O’Brien KA, Du X. Signaling During Platelet Adhesion and Activation. Arterioscler Thromb Vasc Biol. 2010 Dec;30(12):2341–9. https://doi.org/10.1161/ATVBAHA.110.207522
  9. Thon JN, Italiano JE. Platelets: Production, Morphology and Ultrastructure. In: Gresele P, Born GVR, Patrono C, Page CP, editors. Antiplatelet Agents [Internet]. Berlin, Heidelberg: Springer Berlin Heidelberg; 2012 [cited 2022 May 18]. p. 3–22. (Handbook of Experimental Pharmacology; vol. 210). Available from: http://link.springer.com/10.1007/978-3-642-29423-5_1 https://doi.org/10.1007/978-3-642-29423-5_1
  10. Burkhart JM, Vaudel M, Gambaryan S, Radau S, Walter U, Martens L, et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood. 2012 Oct 11;120(15):e73–82. https://doi.org/10.1182/blood-2012-04-416594
  11. Flier JS, Underhill LH, Dvorak HF. Tumors: Wounds That Do Not Heal. N Engl J Med. 1986 Dec 25;315(26):1650–9. https://doi.org/10.1056/NEJM198612253152606
  12. Chang MC, Jeng JH. Tumor Cell-Induced Platelet Aggregation. In: Schwab M, editor. Encyclopedia of Cancer [Internet]. Berlin, Heidelberg: Springer Berlin Heidelberg; 2011 [cited 2022 May 18]. p. 3793–5. Available from: http://link.springer.com/10.1007/978-3-642-16483-5_6023 https://doi.org/10.1007/978-3-642-16483-5_6023
  13. Best MG, Wesseling P, Wurdinger T. Tumor-Educated Platelets as a Noninvasive Biomarker Source for Cancer Detection and Progression Monitoring. Cancer Res. 2018 Jun 19;canres;0008-5472.CAN-18-0887v1. https://doi.org/10.1158/0008-5472.CAN-18-0887
  14. D’Ambrosi S, Nilsson RJ, Wurdinger T. Platelets and tumor-associated RNA transfer. Blood. 2021 Jun 10;137(23):3181–91. https://doi.org/10.1182/blood.2019003978
  15. Ishikawa S, Miyashita T, Inokuchi M, Hayashi H, Oyama K, Tajima H, et al. Platelets surrounding primary tumor cells are related to chemoresistance. Oncol Rep. 2016 Aug;36(2):787–94. https://doi.org/10.3892/or.2016.4898
  16. Yan M, Lesyk G, Radziwon-Balicka A, Jurasz P. Pharmacological Regulation of Platelet Factors That Influence Tumor Angiogenesis. Semin Oncol. 2014 Jun;41(3):370–7. https://doi.org/10.1053/j.seminoncol.2014.04.007
  17. Battinelli EM, Markens BA, Italiano JE. Release of angiogenesis regulatory proteins from platelet alpha granules: modulation of physiologic and pathologic angiogenesis. Blood. 2011 Aug 4;118(5):1359–69. https://doi.org/10.1182/blood-2011-02-334524
  18. Li R, Ren M, Chen N, Luo M, Deng X, Xia J, et al. Presence of intratumoral platelets is associated with tumor vessel structure and metastasis. BMC Cancer. 2014 Dec;14(1):167. https://doi.org/10.1186/1471-2407-14-167
  19. Jiang L, Luan Y, Miao X, Sun C, Li K, Huang Z, et al. Platelet releasate promotes breast cancer growth and angiogenesis via VEGF-integrin cooperative signalling. Br J Cancer. 2017 Aug;117(5):695–703. https://doi.org/10.1038/bjc.2017.214
  20. Andrade SS, Sumikawa JT, Castro ED, Batista FP, Paredes-Gamero E, Oliveira LC, et al. Interface between breast cancer cells and the tumor microenvironment using platelet-rich plasma to promote tumor angiogenesis - influence of platelets and fibrin bundles on the behavior of breast tumor cells. Oncotarget. 2017 Mar 7;8(10):16851–74. https://doi.org/10.18632/oncotarget.15170
  21. Han H, Cao FL, Wang BZ, Mu XR, Li GY, Wang XW. Expression of Angiogenesis Regulatory Proteins and Epithelial-Mesenchymal Transition Factors in Platelets of the Breast Cancer Patients. Sci World J. 2014;2014:1–7. https://doi.org/10.1155/2014/878209
  22. Ribatti D, Tamma R, Annese T. Epithelial-Mesenchymal Transition in Cancer: A Historical Overview. Transl Oncol. 2020 Jun;13(6):100773. https://doi.org/10.1016/j.tranon.2020.100773
  23. Labelle M, Begum S, Hynes RO. Direct Signaling between Platelets and Cancer Cells Induces an Epithelial-Mesenchymal-Like Transition and Promotes Metastasis. Cancer Cell. 2011 Nov;20(5):576–90. https://doi.org/10.1016/j.ccr.2011.09.009
  24. Karolczak K, Watala C. Blood Platelets as an Important but Underrated Circulating Source of TGF β. Int J Mol Sci. 2021 Apr 26;22(9):4492. https://doi.org/10.3390/ijms22094492
  25. Zuo X xiao, Yang Y, Zhang Y, Zhang Z gang, Wang X fei, Shi Y gang. Platelets promote breast cancer cell MCF-7 metastasis by direct interaction: surface integrin α2β1-contacting-mediated activation of Wnt-β-catenin pathway. Cell Commun Signal. 2019 Dec;17(1):142. https://doi.org/10.1186/s12964-019-0464-x
  26. Rachidi S, Metelli A, Riesenberg B, Wu BX, Nelson MH, Wallace C, et al. Platelets subvert T cell immunity against cancer via GARP-TGFβ axis. Sci Immunol. 2017 May 5;2(11):eaai7911. https://doi.org/10.1126/sciimmunol.aai7911
  27. VanderVorst K, Dreyer CA, Konopelski SE, Lee H, Ho HYH, Carraway KL. Wnt/PCP Signaling Contribution to Carcinoma Collective Cell Migration and Metastasis. Cancer Res. 2019 Apr 15;79(8):1719–29. https://doi.org/10.1158/0008-5472.CAN-18-2757
  28. Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 2020 Dec;11(1):5120. https://doi.org/10.1038/s41467-020-18794-x
  29. Hanahan D, Weinberg RA. Hallmarks of Cancer: The Next Generation. Cell. 2011 Mar;144(5):646–74. https://doi.org/10.1016/j.cell.2011.02.013
  30. Planes-Laine, Rochigneux, Bertucci, Chrétien, Viens, Sabatier, et al. PD-1/PD-L1 Targeting in Breast Cancer: The First Clinical Evidences Are Emerging. A Literature Review. Cancers. 2019 Jul 22;11(7):1033. https://doi.org/10.3390/cancers11071033
  31. Zaslavsky AB, Adams MP, Cao X, Maj T, Choi JE, Stangl-Kremser J, et al. Platelet PD-L1 suppresses anti-cancer immune cell activity in PD-L1 negative tumors. Sci Rep. 2020 Dec;10(1):19296. https://doi.org/10.1038/s41598-020-76351-4
  32. Gorelik L, Flavell RA. Transforming growth factor-β in T-cell biology. Nat Rev Immunol. 2002 Jan;2(1):46–53. https://doi.org/10.1038/nri704
  33. Thomas DA, Massagué J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005 Nov;8(5):369–80. https://doi.org/10.1016/j.ccr.2005.10.012
  34. Nieswandt B, Hafner M, Echtenacher B, Männel DN. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999 Mar 15;59(6):1295-300.
  35. Wong SY, Hynes RO. Lymphatic or Hematogenous Dissemination: How Does a Metastatic Tumor Cell Decide? Cell Cycle. 2006 Apr 15;5(8):812–7. https://doi.org/10.4161/cc.5.8.2646
  36. Fidler IJ. Metastasis: Quantitative Analysis of Distribution and Fate of Tumor Emboli Labeled With 125I-5-Iodo-2’-deoxyuridine23. JNCI J Natl Cancer Inst [Internet]. 1970 Oct [cited 2021 Feb 9]; Available from: https://academic.oup.com/jnci/article-lookup/doi/10.1093/jnci/45.4.773
  37. Labelle M, Hynes RO. The Initial Hours of Metastasis: The Importance of Cooperative Host-Tumor Cell Interactions during Hematogenous Dissemination. Cancer Discov. 2012 Dec 1;2(12):1091–9. https://doi.org/10.1158/2159-8290.CD-12-0329
  38. Gil-Bernabé AM, Ferjančič Š, Tlalka M, Zhao L, Allen PD, Im JH, 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 Mar 29;119(13):3164–75. https://doi.org/10.1182/blood-2011-08-376426
  39. van den Berg YW, Osanto S, Reitsma PH, Versteeg HH. The relationship between tissue factor and cancer progression: insights from bench and bedside. Blood. 2012 Jan 26;119(4):924–32. https://doi.org/10.1182/blood-2011-06-317685
  40. Bourcy M, Suarez-Carmona M, Lambert J, Francart ME, Schroeder H, Delierneux C, et al. Tissue Factor Induced by Epithelial-Mesenchymal Transition Triggers a Procoagulant State That Drives Metastasis of Circulating Tumor Cells. Cancer Res. 2016 Jul 15;76(14):4270–82. https://doi.org/10.1158/0008-5472.CAN-15-2263
  41. Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, et al. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood. 2007 Jul 1;110(1):133–41. https://doi.org/10.1182/blood-2007-01-065995
  42. Kato S, Pinto M, Carvajal A, Espinoza N, Monso C, Sadarangani A, et al. Progesterone Increases Tissue Factor Gene Expression, Procoagulant Activity, and Invasion in the Breast Cancer Cell Line ZR-75–1. J Clin Endocrinol Metab. 2005 Feb;90(2):1181–8. https://doi.org/10.1210/jc.2004-0857
  43. Lehrer S, Green S, Dembitzer FR, Rheinstein PH, Rosenzweig KE. Increased RNA Expression of von Willebrand Factor Gene Is Associated With Infiltrating Lobular Breast Cancer and Normal PAM50 Subtype. Cancer Genomics - Proteomics. 2019;16(3):147–53. https://doi.org/10.21873/cgp.20120
  44. Lehrer S, Rheinstein P, Rosenzweig K. Increased expression of von Willebrand factor gene is associated with poorer survival in primary lower grade glioma. Glioma. 2018;1(4):132. https://doi.org/10.4103/glioma.glioma_17_18
  45. Yang A jun, Wang M, Wang Y, Cai W, Li Q, Zhao T ting, et al. Cancer cell-derived von Willebrand factor enhanced metastasis of gastric adenocarcinoma. Oncogenesis. 2018 Jan;7(1):12. https://doi.org/10.1038/s41389-017-0023-5
  46. Pilch J, Habermann R, Felding-Habermann B. Unique Ability of Integrin αvβ3 to Support Tumor Cell Arrest under Dynamic Flow Conditions. J Biol Chem. 2002 Jun;277(24):21930–8. https://doi.org/10.1074/jbc.M201630200
  47. Suter CM, Hogg PJ, Price JT, Chong BH, Ward RL. Identification and Characterisation of a Platelet GPIb/V/IX-like Complex on Human Breast Cancers: Implications for the Metastatic Process. Jpn J Cancer Res. 2001 Oct;92(10):1082–92. https://doi.org/10.1111/j.1349-7006.2001.tb01063.x
  48. Borsig L, Wong R, Hynes RO, Varki NM, Varki A. Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metastasis. Proc Natl Acad Sci U S A. 2002 Feb 19;99(4):2193–8. https://doi.org/10.1073/pnas.261704098
  49. Kim YJ, Borsig L, Han HL, Varki NM, Varki A. Distinct Selectin Ligands on Colon Carcinoma Mucins Can Mediate Pathological Interactions among Platelets, Leukocytes, and Endothelium. Am J Pathol. 1999 Aug;155(2):461–72. https://doi.org/10.1016/S0002-9440(10)65142-5
  50. Rowson-Hodel AR, Wald JH, Hatakeyama J, O’Neal WK, Stonebraker JR, VanderVorst K, et al. Membrane Mucin Muc4 promotes blood cell association with tumor cells and mediates efficient metastasis in a mouse model of breast cancer. Oncogene. 2018 Jan;37(2):197–207. https://doi.org/10.1038/onc.2017.327
  51. Mammadova-Bach E, Gil-Pulido J, Sarukhanyan E, Burkard P, Shityakov S, Schonhart C, et al. Platelet glycoprotein VI promotes metastasis through interaction with cancer cell-derived Galectin-3. Blood. 2020 Feb 7;blood.2019002649. https://doi.org/10.1182/blood.2019002649
  52. Miao S, Shu D, Zhu Y, Lu M, Zhang Q, Pei Y, et al. Cancer cell-derived immunoglobulin G activates platelets by binding to platelet FcγRIIa. Cell Death Dis. 2019 Feb;10(2):87. https://doi.org/10.1038/s41419-019-1367-x
  53. Schlesinger M. Role of platelets and platelet receptors in cancer metastasis. J Hematol OncolJ Hematol Oncol. 2018 Dec;11(1):125. https://doi.org/10.1186/s13045-018-0669-2
  54. Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta BBA - Mol Cell Res. 2013 Dec;1833(12):3481–98. https://doi.org/10.1016/j.bbamcr.2013.06.026
  55. Haemmerle M, Taylor ML, Gutschner T, Pradeep S, Cho MS, Sheng J, et al. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat Commun. 2017 Dec;8(1):310. https://doi.org/10.1038/s41467-017-00411-z
  56. Chang SF, Chang CA, Lee DY, Lee PL, Yeh YM, Yeh CR, et al. Tumor cell cycle arrest induced by shear stress: Roles of integrins and Smad. Proc Natl Acad Sci. 2008 Mar 11;105(10):3927–32. https://doi.org/10.1073/pnas.0712353105
  57. Mitchell MJ, Denais C, Chan MF, Wang Z, Lammerding J, King MR. Lamin A/C deficiency reduces circulating tumor cell resistance to fluid shear stress. Am J Physiol-Cell Physiol. 2015 Dec 1;309(11):C736–46. https://doi.org/10.1152/ajpcell.00050.2015
  58. Egan K, Cooke N, Kenny D. Living in shear: platelets protect cancer cells from shear induced damage. Clin Exp Metastasis. 2014 Aug;31(6):697–704. https://doi.org/10.1007/s10585-014-9660-7
  59. Anvari S, Osei E, Maftoon N. Interactions of platelets with circulating tumor cells contribute to cancer metastasis. Sci Rep. 2021 Dec;11(1):15477. https://doi.org/10.1038/s41598-021-94735-y
  60. Kopp HG, Placke T, Salih HR. Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009 Oct 1;69(19):7775–83. https://doi.org/10.1158/0008-5472.CAN-09-2123
  61. Placke T, Salih HR, Kopp HG. GITR Ligand Provided by Thrombopoietic Cells Inhibits NK Cell Antitumor Activity. J Immunol. 2012 Jul 1;189(1):154–60. https://doi.org/10.4049/jimmunol.1103194
  62. Clar K, Hinterleitner C, Schneider P, Salih H, Maurer S. Inhibition of NK Reactivity Against Solid Tumors by Platelet-Derived RANKL. Cancers. 2019 Feb 26;11(3):277. https://doi.org/10.3390/cancers11030277
  63. Maurer S, Kropp KN, Klein G, Steinle A, Haen SP, Walz JS, et al. Platelet-mediated shedding of NKG2D ligands impairs NK cell immune-surveillance of tumor cells. OncoImmunology. 2018 Feb;7(2):e1364827. https://doi.org/10.1080/2162402X.2017.1364827
  64. Placke T, Örgel M, Schaller M, Jung G, Rammensee HG, Kopp HG, et al. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012 Jan 15;72(2):440–8. https://doi.org/10.1158/0008-5472.CAN-11-1872
  65. Chambers AF, MacDonald IC, Schmidt EE, Koop S, Morris VL, Khokha R, et al. Steps in tumor metastasis: new concepts from intravital videomicroscopy. Cancer Metastasis Rev. 1995 Dec;14(4):279–301. https://doi.org/10.1007/BF00690599
  66. Gassmann P, Kang ML, Mees ST, Haier J. In vivo tumor cell adhesion in the pulmonary microvasculature is exclusively mediated by tumor cell - endothelial cell interaction. BMC Cancer. 2010 Dec;10(1):177. https://doi.org/10.1186/1471-2407-10-177
  67. Wang H, Fu W, Im JH, Zhou Z, Santoro SA, Iyer V, et al. Tumor cell α3β1 integrin and vascular laminin-5 mediate pulmonary arrest and metastasis. J Cell Biol. 2004 Mar 15;164(6):935–41. https://doi.org/10.1083/jcb.200309112
  68. Foss A, Muñoz-Sagredo L, Sleeman J, Thiele W. The contribution of platelets to intravascular arrest, extravasation, and outgrowth of disseminated tumor cells. Clin Exp Metastasis. 2020 Feb;37(1):47–67. https://doi.org/10.1007/s10585-019-10009-y
  69. Menter DG, Hatfield JS, Harkins C, Sloane BF, Taylor JD, Crissman JC, et al. Tumor cell-platelet interactions in vitro and their relationship to in vivo arrest of hematogenously circulating tumor cells. Clin Exp Metastasis. 1987;5(1):65–78. https://doi.org/10.1007/BF00116627
  70. Crissman JD. Morphological Study of the Interaction of Intra vascular Tumor Cells with Endothelial Cells and Subendothelial Matrix. Cancer Res. 1988;
  71. McCarty OJT, Mousa SA, Bray PF, Konstantopoulos K. Immobilized platelets support human colon carcinoma cell tethering, rolling, and firm adhesion under dynamic flow conditions. Blood. 2000 Sep 1;96(5):1789–97. https://doi.org/10.1182/blood.V96.5.1789.h8001789_1789_1797
  72. Honn KV, Tang DG, Grossi IM, Renaud C, Duniec ZM, Johnson CR, et al. Enhanced Endothelial Cell Retraction Mediated by 12(S)-HETE: A Proposed Mechanism for the Role of Platelets in Tumor Cell Metastasis. Exp Cell Res. 1994 Jan;210(1):1–9. https://doi.org/10.1006/excr.1994.1001
  73. Schumacher D, Strilic B, Sivaraj KK, Wettschureck N, Offermanns S. Platelet-Derived Nucleotides Promote Tumor-Cell Transendothelial Migration and Metastasis via P2Y2 Receptor. Cancer Cell. 2013 Jul;24(1):130–7. https://doi.org/10.1016/j.ccr.2013.05.008
  74. Labelle M, Begum S, Hynes RO. Platelets guide the formation of early metastatic niches. Proc Natl Acad Sci. 2014 Jul 29;111(30):E3053–61. https://doi.org/10.1073/pnas.1411082111
  75. Wang Y, Sun Y, Li D, Zhang L, Wang K, Zuo Y, et al. Platelet P2Y12 Is Involved in Murine Pulmonary Metastasis. Kalinichenko VV, editor. PLoS ONE. 2013 Nov 13;8(11):e80780. https://doi.org/10.1371/journal.pone.0080780
  76. Rousseau A, Van Dreden P, Khaterchi A, Larsen AK, Elalamy I, Gerotziafas GT. Procoagulant microparticles derived from cancer cells have determinant role in the hypercoagulable state associated with cancer. Int J Oncol. 2017 Dec;51(6):1793–800. https://doi.org/10.3892/ijo.2017.4170
  77. Stravodimou A, Voutsadakis IA. Pretreatment Thrombocytosis as a Prognostic Factor in Metastatic Breast Cancer. Int J Breast Cancer. 2013;2013:1–6. https://doi.org/10.1155/2013/289563
  78. Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH. Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. J Thromb Haemost. 2007 Mar;5(3):632–4. https://doi.org/10.1111/j.1538-7836.2007.02374.x
  79. Pfrepper C. Paraneoplastic Thromboembolism and Thrombophilia: Significance in Visceral Medicine. Visc Med. 2020;36(4):280–7. https://doi.org/10.1159/000509150
  80. Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood. 2018 Apr 19;131(16):1777–89. https://doi.org/10.1182/blood-2017-05-743187
  81. Michelson AD. Antiplatelet therapies for the treatment of cardiovascular disease. Nat Rev Drug Discov. 2010 Feb;9(2):154–69. https://doi.org/10.1038/nrd2957
  82. Johnson KE, Ceglowski JR, Roweth HG, Forward JA, Tippy MD, El-Husayni S, et al. Aspirin inhibits platelets from reprogramming breast tumor cells and promoting metastasis. Blood Adv. 2019 Jan 22;3(2):198–211. https://doi.org/10.1182/bloodadvances.2018026161
  83. Xu R, Yan Y, Zheng X, Zhang H, Chen W, Li H, et al. Aspirin suppresses breast cancer metastasis to lung by targeting anoikis resistance. Carcinogenesis. 2022 Mar 24;43(2):104–14. https://doi.org/10.1093/carcin/bgab117
  84. Chen WY, Holmes MD. Role of Aspirin in Breast Cancer Survival. Curr Oncol Rep. 2017 Jul;19(7):48. https://doi.org/10.1007/s11912-017-0605-6
  85. Rothwell PM, Wilson M, Price JF, Belch JF, Meade TW, Mehta Z. Effect of daily aspirin on risk of cancer metastasis: a study of incident cancers during randomised controlled trials. The Lancet. 2012 Apr;379(9826):1591–601. https://doi.org/10.1016/S0140-6736(12)60209-8
  86. Bahmani B, Gong H, Luk BT, Haushalter KJ, DeTeresa E, Previti M, et al. Intratumoral immunotherapy using platelet-cloaked nanoparticles enhances antitumor immunity in solid tumors. Nat Commun. 2021 Dec;12(1):1999. https://doi.org/10.1038/s41467-021-22311-z
  87. Wang S, Duan Y, Zhang Q, Komarla A, Gong H, Gao W, et al. Drug Targeting via Platelet Membrane-Coated Nanoparticles. Small Struct. 2020 Oct;1(1):2000018. https://doi.org/10.1002/sstr.202000018