Skip to main content

REVIEW article

Front. Oncol., 03 March 2023
Sec. Molecular and Cellular Oncology

Platelets in the tumor microenvironment and their biological effects on cancer hallmarks

Lilan Chen&#x;Lilan Chen1†Chunyan Zhu&#x;Chunyan Zhu1†Fan PanFan Pan1Ying ChenYing Chen2Lei XiongLei Xiong3Yan Li*Yan Li4*Xiaoyuan Chu*Xiaoyuan Chu1*Guichun Huang*Guichun Huang1*
  • 1Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
  • 2Division of Immunology, Medical School of Nanjing University, Nanjing, Jiangsu, China
  • 3Department of Cardio-Thoracic Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
  • 4Department of Respiratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China

The interplay between platelets and tumors has long been studied. It has been widely accepted that platelets could promote tumor metastasis. However, the precise interactions between platelets and tumor cells have not been thoroughly investigated. Although platelets may play complex roles in multiple steps of tumor development, most studies focus on the platelets in the circulation of tumor patients. Platelets in the primary tumor microenvironment, in addition to platelets in the circulation during tumor cell dissemination, have recently been studied. Their effects on tumor biology are gradually figured out. According to updated cancer hallmarks, we reviewed the biological effects of platelets on tumors, including regulating tumor proliferation and growth, promoting cancer invasion and metastasis, inducing vasculature, avoiding immune destruction, and mediating tumor metabolism and inflammation.

1 Introduction

As early as 1865, an association between cancer and thrombosis, closely linked to platelets, was observed by Trousseau (1). Clinical evidence suggested that thrombocytosis (elevated platelet counts) was correlated with increased cancer risk (2, 3), and high platelet-to-lymphocyte ratio (PLR) or increased platelet count was revealed as an adverse prognostic factor (4, 5). Technological advancement has made it possible to analyze proteins and RNAs of platelets comprehensively, challenging the old understanding of platelet transcriptome (6, 7). As a result, numerous studies have revealed that the proteins and RNAs within platelets vary among individuals with or without cancer, as well as between different cancer types. Tumor-educated platelets (TEPs), which are platelets isolated from the circulation of cancer patients and have distinct RNA and protein profiles, have emerged as potential indicators in liquid biopsies (8).

Platelets play a crucial role in invasive potential, intravasation, and survival in the circulation, arrest, adhesion, and extravasation into secondary sites during the hematogenous spread of tumor cells (9, 10). Platelets probably affect other aspects of cancer other than hematogenous metastasis. The hallmarks of cancer have evolved in 2022 as a result of further understanding of cancer (11). The initial six hallmarks of cancer include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing/accessing vasculature, and activating invasion and metastasis (12). Two more “emerging hallmarks” comprise reprogramming cellular metabolism and avoiding immune destruction (13). Unlike the aforementioned acquired capabilities, genome instability and tumor-promoting inflammation were defined as “enabling characteristics”. With increasing recognition of the tumor microenvironment (TME) in cancer development and the rapid progress of both the breadth and depth of cancer research, four new concepts comprising unlocking phenotypic plasticity, non-mutational epigenetic reprogramming, polymorphic microbiomes, and senescent cells were put forward (11). Platelets might play complex roles in multiple steps of tumor development and affects several hallmarks of cancer.

The tumor microenvironment is a complicated system composed of various non-cancerous cells (e.g., endothelial cells, fibroblasts, adipocytes, and immune cells) and non-cellular components (e.g., extracellular matrix, cytokines, growth factors, and extracellular vesicles). Platelets were engaged in tumor development along with other components of the tumor microenvironment through abundant tumor neovascularization (14). Evidence showed that platelets probably existed in the TME, but the underlying molecular mechanisms remain to be explored (15). We speculated that except the direct contact with tumor cells in the circulation, platelets could also adhere to highly permeable blood vessels and extravasate into tumor stroma through vascular leakage or other molecular pathways.

As a particular cellular component, the role of platelets in the TME has not been fully revealed, and the specific contributions of platelets to cancer require constant exploration. Thus, in conjunction with advances in cancer concepts, this review provides a synopsis of the contributions of platelets to cancer progression to provide a clear map for future research (Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1 Platelets in the primary tumor microenvironment and hematogenous metastasis. Platelets and tumors interact during the hematogenous dissemination of tumor cells. Furthermore, platelets extravasate into the tumor microenvironment via neovascularization leakage. Platelet-tumor cell interactions can affect the biological behavior of tumor cells (cancer hallmarks) through platelet activation, surface receptors, and released factors.

2 Platelets in the tumor microenvironment

Recent studies associated with cancer biology no longer focused solely on tumor cells but were based on the network of TME. To better understand the biological significance of platelets in cancer progression, the precise locations of platelet–tumor interactions need to be figured out. Research found that platelets infiltrated into the tumor stroma and had the predictive value in patients with pancreatic and colorectal cancer (1618). Platelets exist in the TME of ovarian, melanoma, lung, and colorectal cancers (15, 1922). Through the injection of platelets labeled with yellow fluorescence protein (YFP), extravascular platelets were found in the tumor bed, and the platelets outside blood vessels were tumor-specific compared with those in the peritonitis (20, 22). Therefore, like tumor-associated fibroblasts, platelets in the primary tumor microenvironment could be termed tumor-associated platelets (TAPs).

Not only platelet secretion but also the platelet itself can regulate the network of TME through vascular leakage (14). Platelet depletion by anti-platelet antibodies can significantly reduce microvessel density (20). Moreover, platelets in the primary TME can modulate the structure of tumor vessels by inducing vascular permeability (21). Platelets participate in vascular maturation and change the extracellular matrix (ECM), which is connected to the capacity of tumor cells to extravasate from the original site, indicating that platelet strongly affects the initial invasion of tumor cells. Meanwhile, the process of platelet extravasation has been explored. Platelet focal adhesion kinase (FAK) protein regulates their migration into TME (22).

Platelets play multiple roles in cancer biology through platelet-related molecules in the tumor microenvironment. The endocytic mechanism of platelets can ingest and store proteins derived from a tumor, thereby modulating the tumor microenvironment (23). Otherwise, platelet-derived microparticles (PMPs) infiltrating solid tumors can transfer RNAs to tumor cells (24). Micro-RNAs (miRNAs) and microparticles derived from platelets have emerged as novel research targets. Additionally, the bone and bone marrow microenvironment, which are connected with cancer bone metastasis, can facilitate the communication between cancer cells and platelets (25, 26).

2.1 Effects of TAPs on tumor proliferation and growth

Tumor cells have the fundamental hallmarks of sustaining proliferative signaling, evading growth suppressors, and resisting cell death. As has been demonstrated, platelets promote tumor growth and proliferation in ovarian, breast, lung, glioma, and hepatocellular carcinoma, as well as osteosarcoma (2732). Activation of platelets was reported to contribute to tumor growth in vivo in pancreatic cancer (33). After the anti-angiogenic therapy withdrawal, the platelet infiltration in TME increases, and the tumor growth accelerates (22). However, another study observed that the interactions between platelets and tumor cells in colorectal cancer led to the release of different types of microparticles which, although inducing epithelial-to-mesenchymal transition of tumor cells, incurred the intratumoral macrophages and reduced tumor growth (15).

Additionally, the glycoprotein (GP)VI on the platelet surface aided in the growth of primary tumors in Lewis lung carcinoma or melanoma (34). The latest study using a mouse model of hepatocellular carcinoma (HCC) with non-alcoholic fatty liver disease (NAFLD) also found an unexpected result that platelets suppressed liver cancer growth via activating CD8+T cells (35). Although most studies have suggested that platelets facilitate tumor growth, the precise mechanisms behind these phenomena remain unknown. It appears that different tumor types and different platelet-derived receptors and secretions had distinct effects on tumor proliferation and growth.

2.2 Effects of TAPs on cancer invasion and metastasis

The effects of tumor cells-TAPs interaction on invasion-metastasis cascades have been confirmed, including promoting epithelial-mesenchymal transition (EMT) of cancer cells, protecting circulating tumor cells (CTCs) survival in the bloodstream, enhancing adhesion to endothelial cells, and further extravasating into distant sites (15). Growing evidence suggests that anti-platelet agents are effective in hindering tumor metastasis. Upon tumor cells detaching from the primary tumor, invading the surrounding site, and subsequently arriving in the bloodstream, platelets are the first cells to contact with. Tumor cells initiate platelet activation through various mechanisms for stable adhesion, which allows platelets to protect CTCs from high shear stress and immune surveillance (36). Coating with platelets, tumor cells have increased adhesion to the vessel wall and be trapped within capillaries. The secretion of bioactive molecules from platelets assists tumor cells in vascular permeability and trans-endothelial migration. Several molecular mechanisms have been reported in this process, including Fcγ receptor IIa (FcγRIIa), C-type lectin-like receptor 2 (CLEC-2), glycoprotein VI (GPVI), transforming growth factor-β (TGF-β), heat shock protein 47 (HSP47), integrin α2β1& α6β1, adenosine triphosphate (ATP), and lysophosphatidic acid (LPA).

The immunoreceptor tyrosine-based activation motif (ITAM) containing receptors on platelets were rarely studied on the cross-talk between platelets and tumor cells. FcγRIIa, CLEC-2, and GPVI have been investigated as key members of these receptors for mechanisms concerning thrombosis and metastasis. FcγRIIa is required for prostate tumor cell-induced platelet activation (TCIPA), which is dependent on adenosine 5′-diphosphate (ADP) secretion from dense platelet granules, followed by platelet aggregation (37). Downstream signals of the ITAM cascade are potential mediators of tumor cell-induced platelet secretion (TCIPS), such as Syk kinase, phospholipase C, and protein kinase C. Pharmacologic antagonists of these signals as well as inhibitors of FcγRIIa have a critical impact on TCIPS and TCIPA. It was assumed that when platelets are exposed to prostate cancer cells, integrin αIIbβ3-FcγRIIa-P2Y12 cross-talk transduces the signal to release ADP whose receptor P2Y12 amplifies the response, thereby promoting platelet aggregation. Previously, the collaborative signaling of integrin αIIbβ3 and FcγRIIa was established during thrombus formation (38). Furthermore, immunoglobulin G (IgG) derived from cancer cells acts on platelet FcγRIIa, leading to platelet activation (39). Taken together, integrin/ITAM pair, cancer cell derived-IgG, FcγRIIa on platelets, and ADP secretion may be potential targets to cut off signaling during cancer cell induced-platelet activation.

CLEC-2, primarily expressed on platelets, is a receptor for platelet-activating snake venom and contains a hemi-ITAM (40). Podoplanin (PDPN), also known as aggrus, encodes a glycoprotein associated with cell migration and adhesion and is over-expressed in a variety of tumor cells (41). As the key endogenous ligand for CLEC-2, PDPN plays crucial roles in inducing platelet activation and aggregation (42). The research showed that thrombus formation was suppressed and experimental hematogenous metastasis in lungs was decreased in CLEC-2-depleted mice (43). Moreover, recombinant rhodocytin was generated by binding to CLEC-2 to inhibit platelet aggregation and lung metastasis (44). Mutant rhodocytin and anti-podoplanin antibodies also reversed platelets promoting lung colonization in osteosarcoma (45). These results suggest that the interactions between CLEC-2 and PDPN are essential to TCIPA which is the protection of tumor cells in hematogenous metastasis (46). CLEC-2-PDPN axis becomes promising for antiplatelet and antitumor drugs. By blocking the CLEC-2-PDPN interaction through a polysaccharide-containing fraction from Artemisia argyi or cobalt hematoporphyrin (Co-HP), TCIPA and, subsequently, tumor metastasis can be inhibited (47, 48). Moreover, it has been speculated that CLEC-2 depletion did not show severe bleeding tendency, so that to some extent targeting platelet CLEC-2 is a safe treatment strategy (43, 46, 48). In addition, PDPN is also expressed in tumor stroma, including cancer-associated fibroblasts (CAFs) (49). PDPN-positive CAFs exhibit poor clinical outcomes in cancers of the lung (50), breast (51, 52), pancreas (53), and esophagus (54). PDPN-expressing CAFs are tumor-promoting by constructing immunosuppressive TME, which may be modulated by TGF-β production and CD204+ tumor-associated microphages infiltration (55, 56). PDPN-positive CAFs generate tracks through Rho-ROCK pathway to assist with cancer cell invasion in the extracellular matrix (57). Taken together, platelets and CAFs are likely to mediate tumor cell activities in tumor stroma jointly through CLEC-2 and PDPN, but the underlying mechanisms have not yet been studied.

Glycoprotein VI (GPVI), an immunoglobulin superfamily receptor, exerts multiple functions of platelets, particularly collagen-induced platelet activation. GPVI is demonstrated to promote colon and breast cancer cell metastasis by enhancing vascular permeability in response to its counter receptor galectin-3 on tumor cells. GPVI blockade using JAQ1 F(ab′)2 impairs platelet–tumor cell interactions and metastasis with only minor hemostatic side effects (58). Moreover, because GPVI is a critical regulator of vascular integrity in growing tumors, it becomes a promising target for antimetastatic therapies. Functional inhibition of platelet GPVI induces intra-tumoral bleeding and increases the efficacy of chemotherapeutic drugs (59). Although the involvement of GPVI in metastasis has rarely been studied (6062), a few studies have reported that GPVI promotes experimental metastasis in Lewis lung carcinoma and melanoma models (34), and that GPVI inhibitor (revacept) and galectin-3 inhibitors prevent colon cancer metastasis in animal models (63). Very recently, an organ-on-chip platform called ovarian tumor microenvironment chip (OTME-Chip) was developed to verify how platelets extravasate through the endothelium into the tumor microenvironment, promoting tumor cell proliferation, metastasis, and chemoresistance. Again, the GPVI-galectin-3 interaction was identified to play a critical regulatory role in platelet–tumor interactions (64). Collectively, GPVI is exclusively expressed in platelets and megakaryocytes and hardly affects hemostasis as an antithrombotic target (65). Hence, targeting GPVI is a safe and effective antimetastatic strategy.

Reversible phenotypic changes in epithelial-mesenchymal transition of cancer cells involve loss of intercellular adhesion and enhanced motility and invasiveness. Platelets aid in the EMT process by enabling tumor cells to detach from the surrounding tissue of the primary tumor and intravasate into the circulation (66). TGF-β is one of the most important promoters in the EMT of tumor cells. Mesenchymal CTC clusters attached to platelets show high TGF-β signatures (67). It is pointed out that platelet-derived TGF-β could synergistically activate the TGF-β/Smad signaling and NF-κB pathways in tumor cells, and that abrogation of TGF-β signaling was sufficient to inhibit metastasis and EMT (68). Likewise, podoplanin-mediated EMT could be suppressed by a TGF-β neutralizing antibody (69). As an essential signaling molecule, TGF-β secreted from activated platelets is involved in multiple steps of metastasis cascades consisting of immunosurveillance, EMT, and invasiveness.

Interestingly, cancer cell-platelet interaction can be enhanced by the EMT process. Heat shock protein 47 (HSP47), a collagen-binding protein, has been found to be overexpressed in breast cancer and glioblastoma multiforme (GBM). Expression of HSP47 is correlated with cancer metastasis and tumor grade (70, 71). HSP47 is exposed on the surface of the platelet. It contributes to thrombosis and hemostasis, and its inhibitor, Col003, prevents the contact between platelet and collagen by inhibiting GPVI and mitogen-activated protein kinase (MAPK) signaling (72, 73). Furthermore, HSP47 expression in glioma vessels promotes glioma angiogenesis via HIF1α-VEGFR2 signaling (74). More importantly, the HSP47/collagen axis, which is crucial for cancer cell-platelet interaction, has been shown to promote cancer colonization and metastasis (75). During EMT, increased expression of HSP47 and collagen induces platelet recruitment and subsequently enhances CTCs clustering and extravasation. In the experiment, knockdown and silencing of HSP47, as well as the antibodies of collagen receptors in platelets (e.g., integrin α2β1 antibody and GPVI antibody (jAQ1)) are used to inhibit collagen-platelet interaction. However, targeting the interaction between HSP47 and collagen may depend on the small-molecule compound Col003 (76). Another study suggests that HSP47 could induce tumor cell stemness through the TGF-β pathway, implying a positive feedback loop between cancer cell–platelet interaction and EMT induction.

Among the three β1 integrin family members expressed on platelets, α2β1 and α6β1 are involved in direct interaction between tumor cells and platelets. According to recent research, integrin α2β1 is involved in platelet contact with the human breast cancer cell line MCF-7 and promotes EMT of tumor cells by activating the Wnt-β-catenin pathway. The activated Wnt-β-catenin pathway promotes the secretion of TGF-β1 in tumor cells and, in conjunction with TGF-β1/pSmad3 pathways, enhances the transcription of Snail and Slug, which are correlated with EMT (77). Both knockout of integrin α6 in the megakaryocytic lineage and pharmacological blocking of integrin α6 by GoH3 diminished experimental lung metastasis in mice. The study also identified ADAM9, which is expressed on tumor cells, as the counter receptor of α6β1. Their direct binding benefits platelet recruitment to CTCs and their subsequent activation and granule secretion. The interplay between platelet α6β1 and tumor ADAM9 facilitates tumor cell intravasation and extravasation, as evaluated by the trans-endothelial migration in vitro and in vivo (78). The potential role of the platelet β1 integrin family in tumor metastasis needs more experimental evidence and clinical studies (79).

The establishment of metastasis is inseparable from tumor cell arrest, adhesion, and extravasation in a distant site. ATP plays a vital role in the disruption of endothelial junctions. ATP secreted by tumor cell-activated platelet dense granules was identified to act on P2Y2 receptor on endothelial cells and induce tumor cell extravasation and metastasis, especially tumor cell endothelial transmigration (80). Later studies also showed that ATP from the tumor microenvironment was involved in cell invasion or metastasis in breast, prostate, and gastric cancer via the P2Y2 receptor expressed in tumor cells (8183).

Numerous studies have shown that platelet-derived lysophosphatidic acid (LPA) supports metastasis in breast cancer, ovarian cancer, osteosarcoma, and glioblastoma (8486). The mechanism by which platelets contribute to cancer metastasis through LPA depends on autotaxin (ATX) which is overexpressed in multiple types of cancers and stored in α-granules of resting platelets. When tumor cells initiate platelet activation, ATX is released and mediates the production of LPA through its lysophospholipase D activity (8789). Platelets are major sources of LPA, which regulates a variety of pleiotropic activities, such as proliferation, survival, motility, and autophagy (90). LPA receptor 1 (LPAR1) expressed in cancer cells is considered as an important response to LPA, and the absence of LPAR1 may affect vascular leak (84, 86, 88, 91). Tumor CD97, an adhesion G protein-coupled receptor (GPCR), stimulates platelet activation and mediates CD97-LPAR signaling (92). LPA is regarded as a significant bioactive molecule in tumor cell proliferation, invasion, and migration, especially trans-endothelial migration through vascular permeability. Therefore, the ATX-LPA signaling pathways give a new prospect, and LPAR1 and CD97 become promising therapeutic targets in the fight against cancer metastasis (93, 94). Interestingly, LPA and sphingosine 1-phosphate (S1P) also take part in angiogenesis by acting on endothelial cells (95).

2.3 Effects of TAPs on inducing or accessing the vasculature

The contribution of platelets to tumor angiogenesis has long been recognized. Multiple molecules secreted from platelet α-granules exhibit pro- and anti-angiogenic properties. However, the precise mechanisms of platelet granule secretion by different stimulations have not been fully understood. Pro-angiogenic factors contain vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF, also known as FGF2), epidermal growth factor (EGF), metalloproteinases (MMPs), etc. Anti-angiogenic factors include thrombospondin-1 (TSP-1), sphingosine 1 -phosphate (S1P), endostatin, platelet factor 4 (PF4/CXCL4), and so on. Previous studies showed that pro- and anti-angiogenic factors in separate platelet α-granules are released differently. Proteinase-activated receptors (PARs) counter-regulate the release of pro- and anti-angiogenic factors. To be specific, PAR1-activating peptide (PAR1-AP), ADP (via P2Y1/P2Y12), and GPVI-targeting collagen-related peptide induce the expressions of stromal cell-derived factor-1α (SDF-1α/CXCL12) and VEGF, but not endostatin. In contrast, PAR4 activation stimulates endostatin and platelet factor 4 release but suppresses the release of VEGF and SDF-1α (9698). Similarly, adenosine diphosphate (ADP) and thromboxane A2 (TXA2) also counter-regulate platelet release and have the opposite effect on angiogenesis. ADP stimulates the release of VEGF and promotes the migration and formation of capillary structures. Conversely, the release of endostatin stimulated by TXA2 has an inhibitory effect (99).

Pro-angiogenic platelets, which are the drivers for the tumor’s angiogenic switch to break dormancy, can mediate the primary tumor’s effect on the systemic macroenvironment (26). Platelet α-granules are required for bone marrow-derived cell (BMDC) recruitment which is also important for the angiogenic process (100). Platelet-derived growth factor B (PDGFB) in platelet maintains tumor vessel integrity in the TME, which is dependent on the recruitment of pericytes (101). Chemotherapy targeting tumor vasculature also utilizes platelet biomimetic technology to achieve intra-tumoral vascular destruction (102).

2.4 Effects of TAPs on avoiding immune destruction

The most essential aspect of tumor immunosurveillance has been the engagement of natural killer (NK) cells through cytotoxicity and IFN-γ production (103106). Platelets may directly protect tumor cells from NK tumor-lytic activity. Apart from surface shielding by platelet aggregates, numerous investigations showed that the interaction between platelet and fibrin (or fibrinogen) plays a crucial role in immune evasion (107, 108). Salih and colleagues explored various mechanisms, including the down-regulation of NKG2D on NK cells by platelet-derived TGF-β, platelet-derived MHC class I transfer onto the tumor cell surface, and forward signals from platelet-expressed glucocorticoid-induced TNF-related ligand (GITRL) to GITR on NK cells that result in the impaired anti-tumor reactivity of NK cells (109111). These findings shed light on how platelets impact tumor-NK cell interactions. Overexpression of a hypoxia-inducible factor (HIF)-target gene in renal cancer cells may also enhance platelet binding, protecting cancer cells from NK cell-mediated cytotoxicity (112).

Platelets have also been proven to impact adaptive immunity. TGF-β and lactate are major immunomodulators of T cell activity from platelet releases. Platelet-intrinsic glycoprotein A repetitions predominant (GARP) that dominantly contributes to TGF-β activation subsequently suppresses CD4+ and CD8+ T cells. Platelet-specific deletion of GARP by gp96, a molecular chaperone of GARP and GPIb-IX-V, leads to enhanced tumor-specific T-cell immunity (113). The authors then discovered that platelet GARP cleavage, which is required for TGF-β maturation, is thrombin dependent. Thrombin inhibition reduces activated TGF-β and hence avoids immune tolerance (114). Therefore, the GARP-TGF-β axis may be the molecular mechanism of combination therapy of immunotherapy and anti-platelet agents in cancer (113, 115).

2.5 Effects of TAPs on tumor-promoting inflammation

Platelets participate in the inflammatory process, including interactions with leukocytes and endothelial cells, in addition to their vital functions in hemostasis and thrombosis. Platelets recruit and activate various immune cells and promote the secretion of cytokines through surface proteins and the release of pro-inflammatory or regulatory inflammatory factors (10). Granulocytes can be recruited to form early metastatic niches through CXCL5/7 chemokines derived from platelets (116). Immune responses to tumors, like infections, are large and complex, involving various cellular components and molecular pathways. The interactions between platelets and host immune cells potentially influence tumor-promoting inflammation (66).

Neutrophils, the main component of the innate immune system, respond to infection and are involved in tumors. Platelets–neutrophils cross-talk in infection and tumor is inseparable from neutrophil extracellular traps (NETs), a web-like structure composed of extracellular DNA decorated by histones and granular proteases. Platelets induce NET formation and bind to them, while NETs act as a scaffold for platelet adhesion and aggregation and facilitate platelet activation (117119). A recent study demonstrated that surgical stress-activated platelets facilitated NETs-mediated capture of CTCs and synergized with the enhanced aggregation of platelet–tumor cells, contributing to distant metastasis via the Toll-like receptor 4 (TLR4)-ERK5-integrin GPIIb/IIIa axis (120). The high affinity between NETs and platelet-coated-CTCs created favorable conditions for tumor cell dissemination after surgical inflammation.

Furthermore, TLR4 is a mediator in microbial and sterile inflammation, as well as an important component of the lipopolysaccharide (LPS) receptor signaling complex expressed on platelets (121). The TLR4 pathway can activate platelets in response to high-mobility group box1 (HMGB1) released from dying tumor cells destroyed by NK cells and shear stress (122). These studies indicated that TLR4 on platelets is a potential target for reversing tumor-promoting inflammation and restrain tumor dissemination.

2.6 Effects of TAPs on deregulating cellular metabolism

Metabolic changes affect tumor growth, survival, and metastasis. Platelet metabolism is dependent on mitochondrial functions for energy requirements and even the lifespan of platelets (123). Intratumoral hypoxia triggers the release of chemokines and growth factors in platelet granules, while oxidative stress alters mitochondrial function of platelets (124). Platelets are metabolically active and can utilize glycolysis instead of oxidative phosphorylation (OXPHOS). Although no studies have shown that platelets can affect tumor cell mitochondria functions, platelets are thought to be biomarkers of mitochondrial dysfunction in cancers (125). Platelets can release mitochondria into circulation and transfer mitochondria to other cells, such as mesenchymal stem cells, under certain circumstances including wound healing, inflammation, and cancer, thereby building bridges for cell communications and promoting specific properties (126129). In addition, tumor cells can concentrate mitochondria in the most active areas of the platelet–tumor cell process (130). As a result, we can speculate that platelet–tumor cell interplays may affect the mitochondria location and function of tumor cells. Platelets promote the aggressive phenotype of tumor cells, and tumor cells undergo metabolic reprogramming during EMT (131). The metabolic advantages of CTCs that platelets may provide in circulation include increased glucose uptake and lactate production (132).

3 Perspectives

Given the recognition of the importance of TME, the roles of non-tumor cell components in tumor initiation, growth, and metastasis are increasingly studied. The roles of platelets in tumor biology are gradually recognized, and significant progress has been made in revealing the effects of platelets on cancer development. Herein, we reviewed the essential molecular mechanisms underlying tumor–platelet dynamic interactions from the perspectives of cancer hallmarks (Figure 2). Several platelet molecules can be potential anti-platelet targets and have been verified by experiments in animal models. However, platelet-targeted anti-tumor therapy has not formed a prospective treatment regimen. Profound knowledge of tumor–platelet interactions is critical for future research and novel therapeutic interventions.

FIGURE 2
www.frontiersin.org

Figure 2 Examples of interactions between platelets and tumor cells. Platelet surface receptors and released 880 granules provide favorable conditions for tumor progression. Platelets recruit granulocytes via released factors and induce NET formation to bind to CTCs. Platelet surface receptors, such as FcγRIIa and CLEC-2, bind to tumor cells and promote platelet activation and aggregation. LPA and ATP released by platelets as well as platelet GPVI and integrin α6β1 aid tumor cell trans-endothelial migration. Platelets rich in TGF sources promote EMT of tumor cells. Platelets suppress NK and T cell activity, allowing tumor cells to survive in the bloodstream.

Aspirin is widely used to prevent and treat cardiovascular and cerebrovascular diseases, and cyclooxygenase (COX) is an important target of aspirin. Clinical evidence has demonstrated that aspirin exerts the roles in chemoprevention of multiple cancers, such as colorectal, gastric, pancreatic, and breast cancers (133136). However, numerous cohort studies found no effect or even increased risks of several types of cancers (137). The efficacy of aspirin for cancer prevention still needs more clinical trials to verify; furthermore, whether the combination of aspirin and other agents may be more effective than aspirin alone; whether patients can benefit from aspirin in the presence of potential side-effects or apply novel aspirin derivatives; whether aspirin can have a preventive effect in different population types or specific cancer subtypes; and whether the efficiency of aspirin may be dose-dependent, particularly at low doses. Many questions must be answered so that more individuals can benefit from aspirin use (138). Furthermore, the efficacy of aspirin for adjuvant cancer therapy has been validated (139). Aspirin inhibits platelet activation via COX-1 and epithelia and tumor cells via COX-2. Although aspirin and its metabolites have a variety of targets, platelet COX-1 may be the key mechanisms for the anticancer effects of low-dose aspirin (140). The anti-cancer effects of aspirin, which involve multiple cancer hallmarks, including tumor growth (141), metastasis (142, 143), angiogenesis (144, 145), immune evasion (113), are consistent with the platelet biology.

Drug delivery systems using nanoparticles are being developed as an alternative therapeutic route for chemotherapeutic drug transport that can minimize the side effects of chemotherapy in cancer patients (146). Nanoparticles modified with P-selectin-targeting agents contain the antiplatelet agent ticagrelor and the anti-inflammatory agent celecoxib. This nanoplatform focuses on tumor inflammation and tumor–platelet crosstalk and thereby various steps of metastatic cascades can be influenced. Platelet-blocking nanoparticles reduce the platelet-derived TGF-β and downregulate CXCL5 which decreases granulocytes recruitment. Inflammation inhibiting nanoparticles downregulate the MMPs and reverse the inflammatory status. The combination has synergetic effects on inhibiting EMT and interfering with inflammatory microenvironment (147). Based on the essential role of TGF-β in immune suppression and EMT, blocking the major source of TGF-β, which are the platelets, is an essential approach to prevent tumor metastasis. For this reason, Paclitaxel and a nitric oxide (NO) donor-modified albumin shell is constructed to release NO, which can block platelet functions including aggregation, adhesion, and coagulation. Platelet-induced EMT which includes morphological changes and protein markers can be weakened. Obviously, the levels of TGF-β derived from platelets can be downregulated so that tumor immunosuppression can be reversed. In other words, NO-inhibited platelet can be restrained from contacting with tumor cells which involves platelet adhesion around CTC, EMT, and distant metastasis (148). Platelet membrane-coated nanoparticles are also used as a biomimetic delivery approach of immunomodulator agents to inhibit tumor-promoting immune signaling which can make efficient immunotherapy come true (149). These studies exactly take advantage of platelet biological characteristics and abundant connections with cells in the tumor microenvironment. An important question is whether modified platelets carrying cancer therapeutics can be used to target established tumor masses in addition to tumor cells in the circulation. Better understanding of the dominant role of platelets in tumor progression has no doubt to be of guiding significance. Whether directly targeting the interaction between platelets and cancer cells by antiplatelet drugs or using platelets themselves as anticancer drug carriers, these still need to be verified in future clinical practice.

Both the old drug aspirin and the new nanotherapeutics affect tumor development by inhibiting platelet activity. In conclusion, platelet receptors, their binding partners, signaling proteins, and soluble molecules are all potential targets for anti-cancer drugs that target platelet–tumor cell interactions. Extensive experimental evidence for antiplatelet drugs supports the importance of platelets in tumor progression. It is hoped that traditional anti-platelet drugs and novel nanotechnology can be used in cancer treatment as a result of a comprehensive understanding of platelet physiology and the complex mechanisms underlying platelets in cancer biology.

Author contributions

LC and FP wrote the original draft. LC and YC designed the figure. CZ collected information and revised this manuscript. LX reviewed this manuscript. YL, XC, and GH designed and revised this manuscript. All authors contributed to the article and approved the submitted version.

Funding

The work was supported by the National Natural Science Foundation of China (no. 82273326) and the Jiangsu Provincial Youth Medical Key Talents Project (QNRC2016887).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Koupenova M, Kehrel BE, Corkrey HA, Freedman JE. Thrombosis and platelets: an update. Eur Heart J (2017) 38(11):785–91. doi: 10.1093/eurheartj/ehw550

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Giannakeas V, Kotsopoulos J, Cheung MC, Rosella L, Brooks JD, Lipscombe L, et al. Analysis of platelet count and new cancer diagnosis over a 10-year period. JAMA Netw Open (2022) 5(1):e2141633. doi: 10.1001/jamanetworkopen.2021.41633

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Zhu Y, Wei Y, Zhang R, Dong X, Shen S, Zhao Y, et al. Elevated platelet count appears to be causally associated with increased risk of lung cancer: A mendelian randomization analysis. Cancer Epidemiol Biomarkers Prev (2019) 28(5):935–42. doi: 10.1158/1055-9965.EPI-18-0356

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Zhao QT, Yuan Z, Zhang H, Zhang XP, Wang HE, Wang ZK, et al. Prognostic role of platelet to lymphocyte ratio in non-small cell lung cancers: A meta-analysis including 3,720 patients. Int J Cancer. (2016) 139(1):164–70. doi: 10.1002/ijc.30060

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Pedersen LM, Milman N. Prognostic significance of thrombocytosis in patients with primary lung cancer. Eur Respir J (1996) 9(9):1826–30. doi: 10.1183/09031936.96.09091826

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Roweth HG, Battinelli EM. Lessons to learn from tumor-educated platelets. Blood (2021) 137(23):3174–80. doi: 10.1182/blood.2019003976

PubMed Abstract | CrossRef Full Text | Google Scholar

7. D'Ambrosi S, Nilsson RJ, Wurdinger T. Platelets and tumor-associated RNA transfer. Blood (2021) 137(23):3181–91. doi: 10.1182/blood.2019003978

PubMed Abstract | CrossRef Full Text | Google Scholar

8. In 't Veld S, Wurdinger T. Tumor-educated platelets. Blood (2019) 133(22):2359–64. doi: 10.1182/blood-2018-12-852830

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Labelle M, Hynes RO. The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discovery (2012) 2(12):1091–9. doi: 10.1158/2159-8290.CD-12-0329

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Gaertner F, Massberg S. Patrolling the vascular borders: platelets in immunity to infection and cancer. Nat Rev Immunol (2019) 19(12):747–60. doi: 10.1038/s41577-019-0202-z

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Hanahan D. Hallmarks of cancer: New dimensions. Cancer Discovery. (2022) 12(1):31–46. doi: 10.1158/2159-8290.CD-21-1059

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell (2000) 100(1):57–70. doi: 10.1016/S0092-8674(00)81683-9

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell (2011) 144(5):646–74. doi: 10.1016/j.cell.2011.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

14. De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. (2017) 17(8):457–74. doi: 10.1038/nrc.2017.51

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Plantureux L, Mege D, Crescence L, Carminita E, Robert S, Cointe S, et al. The interaction of platelets with colorectal cancer cells inhibits tumor growth but promotes metastasis. Cancer Res (2020) 80(2):291–303. doi: 10.1158/0008-5472.CAN-19-1181

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Miao Y, Xu Z, Feng W, Zheng M, Xu Z, Gao H, et al. Platelet infiltration predicts survival in postsurgical colorectal cancer patients. Int J Cancer. (2022) 150(3):509–20. doi: 10.1002/ijc.33816

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Zhang S-R, Yao L, Wang W-Q, Xu J-Z, Xu H-X, Jin W, et al. Tumor-infiltrating platelets predict postsurgical survival in patients with pancreatic ductal adenocarcinoma. Ann Surg Oncol (2018) 25(13):3984–93. doi: 10.1245/s10434-018-6727-8

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Xu S-S, Xu H-X, Wang W-Q, Li S, Li H, Li T-J, et al. Tumor-infiltrating platelets predict postoperative recurrence and survival in resectable pancreatic neuroendocrine tumor. World J Gastroenterology. (2019) 25(41):6248–57. doi: 10.3748/wjg.v25.i41.6248

CrossRef Full Text | Google Scholar

19. Pucci F, Rickelt S, Newton AP, Garris C, Nunes E, Evavold C, et al. PF4 promotes platelet production and lung cancer growth. Cell Rep (2016) 17(7):1764–72. doi: 10.1016/j.celrep.2016.10.031

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Stone RL, Nick AM, McNeish IA, Balkwill F, Han HD, Bottsford-Miller J, et al. Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med (2012) 366(7):610–8. doi: 10.1056/NEJMoa1110352

PubMed Abstract | CrossRef Full Text | Google Scholar

21. 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) 14:167. doi: 10.1186/1471-2407-14-167

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Haemmerle M, Bottsford-Miller J, Pradeep S, Taylor ML, Choi HJ, Hansen JM, et al. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal. J Clin Invest. (2016) 126(5):1885–96. doi: 10.1172/JCI85086

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Mendoza-Almanza G, Burciaga-Hernández L, Maldonado V, Melendez-Zajgla J, Olmos J. Role of platelets and breast cancer stem cells in metastasis. World J Stem Cells (2020) 12(11):1237–54. doi: 10.4252/wjsc.v12.i11.1237

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Michael JV, Wurtzel JGT, Mao GF, Rao AK, Kolpakov MA, Sabri A, et al. Platelet microparticles infiltrating solid tumors transfer miRNAs that suppress tumor growth. Blood (2017) 130(5):567–80. doi: 10.1182/blood-2016-11-751099

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Kerr BA, McCabe NP, Feng W, Byzova TV. Platelets govern pre-metastatic tumor communication to bone. Oncogene (2013) 32(36):4319–24. doi: 10.1038/onc.2012.447

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Kuznetsov HS, Marsh T, Markens BA, Castano Z, Greene-Colozzi A, Hay SA, et al. Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells. Cancer Discovery (2012) 2(12):1150–65. doi: 10.1158/2159-8290.CD-12-0216

PubMed Abstract | CrossRef Full Text | Google Scholar

27. 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) 117(5):695–703. doi: 10.1038/bjc.2017.214

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Cho MS, Noh K, Haemmerle M, Li D, Park H, Hu Q, et al. Role of ADP receptors on platelets in the growth of ovarian cancer. Blood (2017) 130(10):1235–42. doi: 10.1182/blood-2017-02-769893

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Takagi S, Sato S, Oh-hara T, Takami M, Koike S, Mishima Y, et al. Platelets promote tumor growth and metastasis via direct interaction between aggrus/podoplanin and CLEC-2. PloS One (2013) 8(8):e73609. doi: 10.1371/journal.pone.0073609

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Takagi S, Takemoto A, Takami M, Oh-Hara T, Fujita N. Platelets promote osteosarcoma cell growth through activation of the platelet-derived growth factor receptor-akt signaling axis. Cancer Sci (2014) 105(8):983–8. doi: 10.1111/cas.12464

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Morrone FB, Vargas P, Rockenbach L, Scheffel TB. P2Y12 purinergic receptor and brain tumors: Implications on glioma microenvironment. Molecules (2021) 26(20):6146. doi: 10.3390/molecules26206146

PubMed Abstract | CrossRef Full Text | Google Scholar

32. He AD, Xie W, Song W, Ma YY, Liu G, Liang ML, et al. Platelet releasates promote the proliferation of hepatocellular carcinoma cells by suppressing the expression of KLF6. Sci Rep (2017) 7(1):3989. doi: 10.1038/s41598-017-02801-1

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Mezouar S, Darbousset R, Dignat-George F, Panicot-Dubois L, Dubois C. Inhibition of platelet activation prevents the p-selectin and integrin-dependent accumulation of cancer cell microparticles and reduces tumor growth and metastasis in vivo. Int J Cancer (2015) 136(2):462–75. doi: 10.1002/ijc.28997

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Jain S, Russell S, Ware J. Platelet glycoprotein VI facilitates experimental lung metastasis in syngenic mouse models. J Thromb Haemost. (2009) 7(10):1713–7. doi: 10.1111/j.1538-7836.2009.03559.x

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Ma C, Fu Q, Diggs LP, McVey JC, McCallen J, Wabitsch S, et al. Platelets control liver tumor growth through P2Y12-dependent CD40L release in NAFLD. Cancer Cell (2022) 40(9):986–98.e5. doi: 10.1016/j.ccell.2022.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Egan K, Cooke N, Kenny D. Living in shear: platelets protect cancer cells from shear induced damage. Clin Exp Metastasis. (2014) 31(6):697–704. doi: 10.1007/s10585-014-9660-7

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Mitrugno A, Williams D, Kerrigan SW, Moran N. A novel and essential role for FcgammaRIIa in cancer cell-induced platelet activation. Blood (2014) 123(2):249–60. doi: 10.1182/blood-2013-03-492447

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Zhi H, Rauova L, Hayes V, Gao C, Boylan B, Newman DK, et al. Cooperative integrin/ITAM signaling in platelets enhances thrombus formation in vitro and in vivo. Blood (2013) 121(10):1858–67. doi: 10.1182/blood-2012-07-443325

PubMed Abstract | CrossRef Full Text | Google Scholar

39. 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 FcgammaRIIa. Cell Death Dis (2019) 10(2):87. doi: 10.1038/s41419-019-1367-x

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Suzuki-Inoue K, Kato Y, Inoue O, Kaneko MK, Mishima K, Yatomi Y, et al. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J Biol Chem (2007) 282(36):25993–6001. doi: 10.1074/jbc.M702327200

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Astarita JL, Acton SE, Turley SJ. Podoplanin: emerging functions in development, the immune system, and cancer. Front In Immunol (2012) 3:283. doi: 10.3389/fimmu.2012.00283

CrossRef Full Text | Google Scholar

42. Suzuki-Inoue K. Platelets and cancer-associated thrombosis: focusing on the platelet activation receptor CLEC-2 and podoplanin. Blood (2019) 134(22):1912–8. doi: 10.1182/blood.2019001388

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Shirai T, Inoue O, Tamura S, Tsukiji N, Sasaki T, Endo H, et al. C-type lectin-like receptor 2 promotes hematogenous tumor metastasis and prothrombotic state in tumor-bearing mice. J Thromb Haemostasis JTH. (2017) 15(3):513–25. doi: 10.1111/jth.13604

CrossRef Full Text | Google Scholar

44. Sasaki T, Shirai T, Tsukiji N, Otake S, Tamura S, Ichikawa J, et al. Functional characterization of recombinant snake venom rhodocytin: rhodocytin mutant blocks CLEC-2/podoplanin-dependent platelet aggregation and lung metastasis. J Thromb Haemostasis JTH. (2018) 16(5):960–72. doi: 10.1111/jth.13987

CrossRef Full Text | Google Scholar

45. Ichikawa J, Ando T, Kawasaki T, Sasaki T, Shirai T, Tsukiji N, et al. Role of platelet c-type lectin-like receptor 2 in promoting lung metastasis in osteosarcoma. J Bone Mineral Res Off J Am Soc For Bone Mineral Res (2020) 35(9):1738–50. doi: 10.1002/jbmr.4045

CrossRef Full Text | Google Scholar

46. Hwang BO, Park SY, Cho ES, Zhang X, Lee SK, Ahn HJ, et al. Platelet CLEC2-podoplanin axis as a promising target for oral cancer treatment. Front Immunol (2021) 12:807600. doi: 10.3389/fimmu.2021.807600

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Tseng CP, Huang YL, Chang YW, Liao HR, Chen YL, Hsieh PW. Polysaccharide-containing fraction from artemisia argyi inhibits tumor cell-induced platelet aggregation by blocking interaction of podoplanin with c-type lectin-like receptor 2. J Food Drug Anal (2020) 28(1):115–23. doi: 10.1016/j.jfda.2019.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Tsukiji N, Osada M, Sasaki T, Shirai T, Satoh K, Inoue O, et al. Cobalt hematoporphyrin inhibits CLEC-2-podoplanin interaction, tumor metastasis, and arterial/venous thrombosis in mice. Blood Adv (2018) 2(17):2214–25. doi: 10.1182/bloodadvances.2018016261

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Suzuki H, Kaneko MK, Kato Y. Roles of podoplanin in malignant progression of tumor. Cells (2022) 11(3):575. doi: 10.3390/cells11030575

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Kawase A, Ishii G, Nagai K, Ito T, Nagano T, Murata Y, et al. Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int J Cancer. (2008) 123(5):1053–9. doi: 10.1002/ijc.23611

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Schoppmann SF, Berghoff A, Dinhof C, Jakesz R, Gnant M, Dubsky P, et al. Podoplanin-expressing cancer-associated fibroblasts are associated with poor prognosis in invasive breast cancer. Breast Cancer Res Treat (2012) 134(1):237–44. doi: 10.1007/s10549-012-1984-x

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Pula B, Jethon A, Piotrowska A, Gomulkiewicz A, Owczarek T, Calik J, et al. Podoplanin expression by cancer-associated fibroblasts predicts poor outcome in invasive ductal breast carcinoma. Histopathology (2011) 59(6):1249–60. doi: 10.1111/j.1365-2559.2011.04060.x

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Shindo K, Aishima S, Ohuchida K, Fujiwara K, Fujino M, Mizuuchi Y, et al. Podoplanin expression in cancer-associated fibroblasts enhances tumor progression of invasive ductal carcinoma of the pancreas. Mol Cancer. (2013) 12(1):168. doi: 10.1186/1476-4598-12-168

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Schoppmann SF, Jesch B, Riegler MF, Maroske F, Schwameis K, Jomrich G, et al. Podoplanin expressing cancer associated fibroblasts are associated with unfavourable prognosis in adenocarcinoma of the esophagus. Clin Exp Metastasis. (2013) 30(4):441–6. doi: 10.1007/s10585-012-9549-2

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Sakai T, Aokage K, Neri S, Nakamura H, Nomura S, Tane K, et al. Link between tumor-promoting fibrous microenvironment and an immunosuppressive microenvironment in stage I lung adenocarcinoma. Lung Cancer. (2018) 126:64–71. doi: 10.1016/j.lungcan.2018.10.021

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Suzuki J, Aokage K, Neri S, Sakai T, Hashimoto H, Su Y, et al. Relationship between podoplanin-expressing cancer-associated fibroblasts and the immune microenvironment of early lung squamous cell carcinoma. Lung Cancer (Amsterdam Netherlands) (2021) 153:1–10. doi: 10.1016/j.lungcan.2020.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Neri S, Ishii G, Hashimoto H, Kuwata T, Nagai K, Date H, et al. Podoplanin-expressing cancer-associated fibroblasts lead and enhance the local invasion of cancer cells in lung adenocarcinoma. Int J Cancer. (2015) 137(4):784–96. doi: 10.1002/ijc.29464

PubMed Abstract | CrossRef Full Text | Google Scholar

58. 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) 135(14):1146–60. doi: 10.1182/blood.2019002649

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Volz J, Mammadova-Bach E, Gil-Pulido J, Nandigama R, Remer K, Sorokin L, et al. Inhibition of platelet GPVI induces intratumor hemorrhage and increases efficacy of chemotherapy in mice. Blood (2019) 133(25):2696–706. doi: 10.1182/blood.2018877043

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Schlesinger M. Role of platelets and platelet receptors in cancer metastasis. J Hematol Oncol (2018) 11(1):125. doi: 10.1186/s13045-018-0669-2

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Erpenbeck L, Schon MP. Deadly allies: the fatal interplay between platelets and metastasizing cancer cells. Blood (2010) 115(17):3427–36. doi: 10.1182/blood-2009-10-247296

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest. (2019) 129(1):12–23. doi: 10.1172/JCI122955

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Dovizio M, Maier TJ, Alberti S, Di Francesco L, Marcantoni E, Munch G, et al. Pharmacological inhibition of platelet-tumor cell cross-talk prevents platelet-induced overexpression of cyclooxygenase-2 in HT29 human colon carcinoma cells. Mol Pharmacol (2013) 84(1):25–40. doi: 10.1124/mol.113.084988

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Saha B, Mathur T, Tronolone JJ, Chokshi M, Lokhande GK, Selahi A, et al. Human tumor microenvironment chip evaluates the consequences of platelet extravasation and combinatorial antitumor-antiplatelet therapy in ovarian cancer. Sci Adv (2021) 7(30):eabg5283. doi: 10.1126/sciadv.abg5283

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Dutting S, Bender M, Nieswandt B. Platelet GPVI: a target for antithrombotic therapy? ! Trends Pharmacol Sci (2012) 33(11):583–90. doi: 10.1016/j.tips.2012.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Ward MP, Kane L E, Norris L A, Mohamed BM, Kelly T, Bates M, et al. Platelets, immune cells and the coagulation cascade; friend or foe of the circulating tumour cell? Mol Cancer (2021) 20(1):59. doi: 10.1186/s12943-021-01347-1

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science (2013) 339(6119):580–4. doi: 10.1126/science.1228522

PubMed Abstract | CrossRef Full Text | Google Scholar

68. 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) 20(5):576–90. doi: 10.1016/j.ccr.2011.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Takemoto A, Okitaka M, Takagi S, Takami M, Sato S, Nishio M, et al. A critical role of platelet TGF-beta release in podoplanin-mediated tumour invasion and metastasis. Sci Rep (2017) 7:42186. doi: 10.1038/srep42186

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Zhu J, Xiong G, Fu H, Evers BM, Zhou BP, Xu R. Chaperone Hsp47 drives malignant growth and invasion by modulating an ECM gene network. Cancer Res (2015) 75(8):1580–91. doi: 10.1158/0008-5472.CAN-14-1027

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Jiang X, Zhou T, Wang Z, Qi B, Xia H. HSP47 promotes glioblastoma stemlike cell survival by modulating tumor microenvironment extracellular matrix through TGF-beta pathway. ACS Chem Neurosci (2017) 8(1):128–34. doi: 10.1021/acschemneuro.6b00253

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Sasikumar P, AlOuda KS, Kaiser WJ, Holbrook LM, Kriek N, Unsworth AJ, et al. The chaperone protein HSP47: a platelet collagen binding protein that contributes to thrombosis and hemostasis. J Thromb Haemost. (2018) 16(5):946–59. doi: 10.1111/jth.13998

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Wu S, Liang C, Xie X, Huang H, Fu J, Wang C, et al. Hsp47 inhibitor Col003 attenuates collagen-induced platelet activation and cerebral ischemic-reperfusion injury in rats. Front Pharmacol (2021) 12:792263. doi: 10.3389/fphar.2021.792263

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Wu ZB, Cai L, Lin SJ, Leng ZG, Guo YH, Yang WL, et al. Heat shock protein 47 promotes glioma angiogenesis. Brain Pathol (2016) 26(1):31–42. doi: 10.1111/bpa.12256

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Xiong G, Chen J, Zhang G, Wang S, Kawasaki K, Zhu J, et al. Hsp47 promotes cancer metastasis by enhancing collagen-dependent cancer cell-platelet interaction. Proc Natl Acad Sci U S A. (2020) 117(7):3748–58. doi: 10.1073/pnas.1911951117

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Ito S, Ogawa K, Takeuchi K, Takagi M, Yoshida M, Hirokawa T, et al. A small-molecule compound inhibits a collagen-specific molecular chaperone and could represent a potential remedy for fibrosis. J Biol Chem (2017) 292(49):20076–85. doi: 10.1074/jbc.M117.815936

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Zuo XX, Yang Y, Zhang Y, Zhang ZG, Wang XF, Shi YG. Platelets promote breast cancer cell MCF-7 metastasis by direct interaction: surface integrin alpha2beta1-contacting-mediated activation of wnt-beta-catenin pathway. Cell Commun Signal (2019) 17(1):142. doi: 10.1186/s12964-019-0464-x

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Mammadova-Bach E, Zigrino P, Brucker C, Bourdon C, Freund M, De Arcangelis A, et al. Platelet integrin alpha6beta1 controls lung metastasis through direct binding to cancer cell-derived ADAM9. JCI Insight (2016) 1(14):e88245. doi: 10.1172/jci.insight.88245

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Lavergne M, Janus-Bell E, Schaff M, Gachet C, Mangin PH. Platelet integrins in tumor metastasis: Do they represent a therapeutic target? Cancers (Basel) (2017) 9(10):133. doi: 10.3390/cancers9100133

PubMed Abstract | CrossRef Full Text | Google Scholar

80. 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) 24(1):130–7. doi: 10.1016/j.ccr.2013.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Li WH, Qiu Y, Zhang HQ, Liu Y, You JF, Tian XX, et al. P2Y2 receptor promotes cell invasion and metastasis in prostate cancer cells. Br J Cancer. (2013) 109(6):1666–75. doi: 10.1038/bjc.2013.484

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Zhang JL, Liu Y, Yang H, Zhang HQ, Tian XX, Fang WG. ATP-P2Y2-beta-catenin axis promotes cell invasion in breast cancer cells. Cancer Sci (2017) 108(7):1318–27. doi: 10.1111/cas.13273

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Reyna-Jeldes M, de la Fuente-Ortega E, Cerda D, Velazquez-Miranda E, Pinto K, Vazquez-Cuevas FG, et al. Purinergic P2Y2 and P2X4 receptors are involved in the epithelial-mesenchymal transition and metastatic potential of gastric cancer derived cell lines. Pharmaceutics (2021) 13(8):1234. doi: 10.3390/pharmaceutics13081234

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Boucharaba A, Serre C-M, Grès S, Saulnier-Blache JS, Bordet J-C, Guglielmi J, et al. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J Clin Invest (2004) 114(12):1714–25. doi: 10.1172/JCI200422123

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Kishi Y, Okudaira S, Tanaka M, Hama K, Shida D, Kitayama J, et al. Autotaxin is overexpressed in glioblastoma multiforme and contributes to cell motility of glioblastoma by converting lysophosphatidylcholine to lysophosphatidic acid. J Biol Chem (2006) 281(25):17492–500. doi: 10.1074/jbc.M601803200

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Takagi S, Sasaki Y, Koike S, Takemoto A, Seto Y, Haraguchi M, et al. Platelet-derived lysophosphatidic acid mediated LPAR1 activation as a therapeutic target for osteosarcoma metastasis. Oncogene (2021) 40(36):5548–58. doi: 10.1038/s41388-021-01956-6

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Umezu-Goto M, Kishi Y, Taira A, Hama K, Dohmae N, Takio K, et al. Autotaxin has lysophospholipase d activity leading to tumor cell growth and motility by lysophosphatidic acid production. J Cell Biol (2002) 158(2):227–33. doi: 10.1083/jcb.200204026

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Hama K, Aoki J, Fukaya M, Kishi Y, Sakai T, Suzuki R, et al. Lysophosphatidic acid and autotaxin stimulate cell motility of neoplastic and non-neoplastic cells through LPA1. J Biol Chem (2004) 279(17):17634–9. doi: 10.1074/jbc.M313927200

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Leblanc R, Lee SC, David M, Bordet JC, Norman DD, Patil R, et al. Interaction of platelet-derived autotaxin with tumor integrin alphaVbeta3 controls metastasis of breast cancer cells to bone. Blood (2014) 124(20):3141–50. doi: 10.1182/blood-2014-04-568683

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Chang CL, Liao JJ, Huang WP, Lee H. Lysophosphatidic acid inhibits serum deprivation-induced autophagy in human prostate cancer PC-3 cells. Autophagy (2007) 3(3):268–70. doi: 10.4161/auto.3909

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M, Zhao Z, et al. The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nat Med (2008) 14(1):45–54. doi: 10.1038/nm1685

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Ward Y, Lake R, Faraji F, Sperger J, Martin P, Gilliard C, et al. Platelets promote metastasis via binding tumor CD97 leading to bidirectional signaling that coordinates transendothelial migration. Cell Rep (2018) 23(3):808–22. doi: 10.1016/j.celrep.2018.03.092

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Panupinthu N, Lee HY, Mills GB. Lysophosphatidic acid production and action: critical new players in breast cancer initiation and progression. Br J Cancer. (2010) 102(6):941–6. doi: 10.1038/sj.bjc.6605588

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Leblanc R, Houssin A, Peyruchaud O. Platelets, autotaxin and lysophosphatidic acid signalling: win-win factors for cancer metastasis. Br J Pharmacol (2018) 175(15):3100–10. doi: 10.1111/bph.14362

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Boucharaba A, Guillet B, Menaa F, Hneino M, van Wijnen AJ, Clezardin P, et al. Bioactive lipids lysophosphatidic acid and sphingosine 1-phosphate mediate breast cancer cell biological functions through distinct mechanisms. Oncol Res (2009) 18(4):173–84. doi: 10.3727/096504009790217399

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Italiano JE Jr., Richardson JL, Patel-Hett S, Battinelli E, Zaslavsky A, Short S, et al. Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released. Blood (2008) 111(3):1227–33. doi: 10.1182/blood-2007-09-113837

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Chatterjee M, Huang Z, Zhang W, Jiang L, Hultenby K, Zhu L, et al. Distinct platelet packaging, release, and surface expression of proangiogenic and antiangiogenic factors on different platelet stimuli. Blood (2011) 117(14):3907–11. doi: 10.1182/blood-2010-12-327007

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Ma L, Perini R, McKnight W, Dicay M, Klein A, Hollenberg MD, et al. Proteinase-activated receptors 1 and 4 counter-regulate endostatin and VEGF release from human platelets. Proc Natl Acad Sci U S A. (2005) 102(1):216–20. doi: 10.1073/pnas.0406682102

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Battinelli EM, Markens BA, Italiano JE Jr. Release of angiogenesis regulatory proteins from platelet alpha granules: modulation of physiologic and pathologic angiogenesis. Blood (2011) 118(5):1359–69. doi: 10.1182/blood-2011-02-334524

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Feng W, Madajka M, Kerr BA, Mahabeleshwar GH, Whiteheart SW, Byzova TV. A novel role for platelet secretion in angiogenesis: mediating bone marrow-derived cell mobilization and homing. Blood (2011) 117(14):3893–902. doi: 10.1182/blood-2010-08-304808

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Zhang Y, Cedervall J, Hamidi A, Herre M, Viitaniemi K, D'Amico G, et al. Platelet-specific PDGFB ablation impairs tumor vessel integrity and promotes metastasis. Cancer Res (2020) 80(16):3345–58. doi: 10.1158/0008-5472.CAN-19-3533

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Zhang M, Ye JJ, Xia Y, Wang ZY, Li CX, Wang XS, et al. Platelet-mimicking biotaxis targeting vasculature-disrupted tumors for cascade amplification of hypoxia-sensitive therapy. ACS Nano. (2019) 13(12):14230–40. doi: 10.1021/acsnano.9b07330

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Glasner A, Levi A, Enk J, Isaacson B, Viukov S, Orlanski S, et al. NKp46 receptor-mediated interferon-gamma production by natural killer cells increases fibronectin 1 to alter tumor architecture and control metastasis. Immunity (2018) 48(1):107–19.e4. doi: 10.1016/j.immuni.2017.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Lopez-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of metastasis by NK cells. Cancer Cell (2017) 32(2):135–54. doi: 10.1016/j.ccell.2017.06.009

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat Rev Drug Discovery (2020) 19(3):200–18. doi: 10.1038/s41573-019-0052-1

CrossRef Full Text | Google Scholar

106. Wu SY, Fu T, Jiang YZ, Shao ZM. Natural killer cells in cancer biology and therapy. Mol Cancer. (2020) 19(1):120. doi: 10.1158/1557-3125.HIPPO19-IA12

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, et al. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood (2005) 105(1):178–85. doi: 10.1182/blood-2004-06-2272

PubMed Abstract | CrossRef Full Text | Google Scholar

108. 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) 110(1):133–41. doi: 10.1182/blood-2007-01-065995

PubMed Abstract | CrossRef Full Text | Google Scholar

109. 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) 69(19):7775–83. doi: 10.1158/0008-5472.CAN-09-2123

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Placke T, Orgel 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) 72(2):440–8. doi: 10.1158/0008-5472.CAN-11-1872

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Placke T, Salih HR, Kopp HG. GITR ligand provided by thrombopoietic cells inhibits NK cell antitumor activity. J Immunol (2012) 189(1):154–60. doi: 10.4049/jimmunol.1103194

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Robinson CM, Poon BPK, Kano Y, Pluthero FG, Kahr WHA, Ohh M. A hypoxia-inducible HIF1-GAL3ST1-Sulfatide axis enhances ccRCC immune evasion via increased tumor cell-platelet binding. Mol Cancer Res (2019) 17(11):2306–17. doi: 10.1158/1541-7786.MCR-19-0461

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Rachidi S, Metelli A, Riesenberg B, Wu BX, Nelson MH, Wallace C, et al. Platelets subvert T cell immunity against cancer via GARP-TGFbeta axis. Sci Immunol (2017) 2(11):eaai7911. doi: 10.1126/sciimmunol.aai7911

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Metelli A, Wu BX, Riesenberg B, Guglietta S, Huck JD, Mills C, et al. Thrombin contributes to cancer immune evasion via proteolysis of platelet-bound GARP to activate LTGF-beta. Sci Transl Med (2020) 12(525):eaay4860. doi: 10.1126/scitranslmed.aay4860

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Metelli A, Salem M, Wallace CH, Wu BX, Li A, Li X, et al. Immunoregulatory functions and the therapeutic implications of GARP-TGF-beta in inflammation and cancer. J Hematol Oncol (2018) 11(1):24. doi: 10.1186/s13045-018-0570-z

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Labelle M, Begum S, Hynes RO. Platelets guide the formation of early metastatic niches. Proc Natl Acad Sci U S A. (2014) 111(30):E3053–61. doi: 10.1073/pnas.1411082111

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Cedervall J, Hamidi A, Olsson AK. Platelets, NETs and cancer. Thromb Res (2018) 164 Suppl 1:S148–S52. doi: 10.1016/j.thromres.2018.01.049

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Olsson A-K, Cedervall J. NETosis in cancer - platelet-neutrophil crosstalk promotes tumor-associated pathology. Front In Immunol (2016) 7:373. doi: 10.3389/fimmu.2016.00373

CrossRef Full Text | Google Scholar

119. Abdol Razak N, Elaskalani O, Metharom P. Pancreatic cancer-induced neutrophil extracellular traps: A potential contributor to cancer-associated thrombosis. Int J Mol Sci (2017) 18(3):487. doi: 10.3390/ijms18030487

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Ren J, He J, Zhang H, Xia Y, Hu Z, Loughran P, et al. Platelet TLR4-ERK5 axis facilitates NET-mediated capturing of circulating tumor cells and distant metastasis after surgical stress. Cancer Res (2021) 81(9):2373–85. doi: 10.1158/0008-5472.CAN-20-3222

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med (2007) 13(4):463–9. doi: 10.1038/nm1565

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Yu LX, Yan L, Yang W, Wu FQ, Ling Y, Chen SZ, et al. Platelets promote tumour metastasis via interaction between TLR4 and tumour cell-released high-mobility group box1 protein. Nat Commun (2014) 5:5256. doi: 10.1038/ncomms6256

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Gremmel T, Frelinger AL, Michelson AD. Platelet physiology. Semin Thromb Hemost. (2016) 42(3):191–204. doi: 10.1055/s-0035-1564835

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Palacka P, Gvozdjakova A, Rausova Z, Kucharska J, Slopovsky J, Obertova J, et al. Platelet mitochondrial bioenergetics reprogramming in patients with urothelial carcinoma. Int J Mol Sci (2021) 23(1):388. doi: 10.3390/ijms23010388

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Kramer PA, Ravi S, Chacko B, Johnson MS, Darley-Usmar VM. A review of the mitochondrial and glycolytic metabolism in human platelets and leukocytes: implications for their use as bioenergetic biomarkers. Redox Biol (2014) 2:206–10. doi: 10.1016/j.redox.2013.12.026

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Al Amir Dache Z, Otandault A, Tanos R, Pastor B, Meddeb R, Sanchez C, et al. Blood contains circulating cell-free respiratory competent mitochondria. FASEB J (2020) 34(3):3616–30. doi: 10.1096/fj.201901917RR

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Chen E, Chen Z, Chen L, Hu X. Platelet-derived respiratory-competent mitochondria transfer to mesenchymal stem cells to promote wound healing via metabolic reprogramming. Platelets (2022) 33(2):171–3. doi: 10.1080/09537104.2021.1961717

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Torralba D, Baixauli F, Sánchez-Madrid F. Mitochondria know no boundaries: Mechanisms and functions of intercellular mitochondrial transfer. Front In Cell Dev Biol (2016) 4:107. doi: 10.3389/fcell.2016.00107

CrossRef Full Text | Google Scholar

129. Levoux J, Prola A, Lafuste P, Gervais M, Chevallier N, Koumaiha Z, et al. Platelets facilitate the wound-healing capability of mesenchymal stem cells by mitochondrial transfer and metabolic reprogramming. Cell Metab (2021) 33(2):283–299.e9. doi: 10.1016/j.cmet.2020.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Menter DG, Hatfield JS, Harkins C, Sloane BF, Taylor JD, Crissman JD, 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. doi: 10.1007/BF00116627

PubMed Abstract | CrossRef Full Text | Google Scholar

131. El Hassouni B, Granchi C, Vallés-Martí A, Supadmanaba IGP, Bononi G, Tuccinardi T, et al. The dichotomous role of the glycolytic metabolism pathway in cancer metastasis: Interplay with the complex tumor microenvironment and novel therapeutic strategies. Semin Cancer Biol (2020) 60:238–48. doi: 10.1016/j.semcancer.2019.08.025

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Liu M, Quek L-E, Sultani G, Turner N. Epithelial-mesenchymal transition induction is associated with augmented glucose uptake and lactate production in pancreatic ductal adenocarcinoma. Cancer Metab (2016) 4:19. doi: 10.1186/s40170-016-0160-x

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Burn J, Sheth H, Elliott F, Reed L, Macrae F, Mecklin J-P, et al. Cancer prevention with aspirin in hereditary colorectal cancer (Lynch syndrome), 10-year follow-up and registry-based 20-year data in the CAPP2 study: a double-blind, randomised, placebo-controlled trial. Lancet (2020) 395(10240):1855–63. doi: 10.1016/S0140-6736(20)30366-4

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Cheung KS, Chan EW, Wong AYS, Chen L, Seto WK, Wong ICK, et al. Aspirin and risk of gastric cancer after helicobacter pylori eradication: A territory-wide study. J Natl Cancer Inst (2018) 110(7):743–9. doi: 10.1093/jnci/djx267

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Bosetti C, Santucci C, Gallus S, Martinetti M, La Vecchia C. Aspirin and the risk of colorectal and other digestive tract cancers: an updated meta-analysis through 2019. Ann Oncol (2020) 31(5):558–68. doi: 10.1016/j.annonc.2020.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Zhang SM, Cook NR, Manson JE, Lee IM, Buring JE. Low-dose aspirin and breast cancer risk: results by tumour characteristics from a randomised trial. Br J Cancer. (2008) 98(5):989–91. doi: 10.1038/sj.bjc.6604240

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Loomans-Kropp HA, Pinsky P, Umar A. Evaluation of aspirin use with cancer incidence and survival among older adults in the prostate, lung, colorectal, and ovarian cancer screening trial. JAMA Network Open (2021) 4(1):e2032072. doi: 10.1001/jamanetworkopen.2020.32072

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Hua H, Zhang H, Kong Q, Wang J, Jiang Y. Complex roles of the old drug aspirin in cancer chemoprevention and therapy. Med Res Rev (2019) 39(1):114–45. doi: 10.1002/med.21514

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Jankowski JAZ, de Caestecker J, Love SB, Reilly G, Watson P, Sanders S, et al. Esomeprazole and aspirin in barrett's oesophagus (AspECT): a randomised factorial trial. Lancet (2018) 392(10145):400–8. doi: 10.1016/S0140-6736(18)31388-6

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Lichtenberger LM, Vijayan KV. Are platelets the primary target of aspirin's remarkable anticancer activity? Cancer Res (2019) 79(15):3820–3. doi: 10.1158/0008-5472.CAN-19-0762

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Lichtenberger LM, Fang D, Bick RJ, Poindexter BJ, Phan T, Bergeron AL, et al. Unlocking aspirin's chemopreventive activity: Role of irreversibly inhibiting platelet cyclooxygenase-1. Cancer Prev Res (Phila). (2017) 10(2):142–52. doi: 10.1158/1940-6207.CAPR-16-0241

PubMed Abstract | CrossRef Full Text | Google Scholar

142. 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 Advances. (2019) 3(2):198–211. doi: 10.1182/bloodadvances.2018026161

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Lucotti S, Cerutti C, Soyer M, Gil-Bernabé AM, Gomes AL, Allen PD, et al. Aspirin blocks formation of metastatic intravascular niches by inhibiting platelet-derived COX-1/thromboxane A2. J Clin Invest (2019) 129(5):1845–62. doi: 10.1172/JCI121985

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Huang Y, Lichtenberger LM, Taylor M, Bottsford-Miller JN, Haemmerle M, Wagner MJ, et al. Antitumor and antiangiogenic effects of aspirin-PC in ovarian cancer. Mol Cancer Ther (2016) 15(12):2894–904. doi: 10.1158/1535-7163.MCT-16-0074

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Holmes CE, Jasielec J, Levis JE, Skelly J, Muss HB. Initiation of aspirin therapy modulates angiogenic protein levels in women with breast cancer receiving tamoxifen therapy. Clin Transl Sci (2013) 6(5):386–90. doi: 10.1111/cts.12070

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Dovizio M, Ballerini P, Fullone R, Tacconelli S, Contursi A, Patrignani P. Multifaceted functions of platelets in cancer: From tumorigenesis to liquid biopsy tool and drug delivery system. Int J Mol Sci (2020) 21(24):9585. doi: 10.3390/ijms21249585

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Li S, Li L, Lin X, Chen C, Luo C, Huang Y. Targeted inhibition of tumor inflammation and tumor-platelet crosstalk by nanoparticle-mediated drug delivery mitigates cancer metastasis. ACS Nano (2022) 16(1):50–67. doi: 10.1021/acsnano.1c06022

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Xu Y, Liu J, Liu Z, Ren H, Yong J, Li W, et al. Blockade of platelets using tumor-specific NO-releasing nanoparticles prevents tumor metastasis and reverses tumor immunosuppression. ACS Nano. (2020) 14(8):9780–95. doi: 10.1021/acsnano.0c01687

PubMed Abstract | CrossRef Full Text | Google Scholar

149. 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) 12(1):1999. doi: 10.1038/s41467-021-22311-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Glossary

www.frontiersin.org

Keywords: platelets, cancer biology, tumor microenvironment, tumor-associated platelets, tumor metastasis

Citation: Chen L, Zhu C, Pan F, Chen Y, Xiong L, Li Y, Chu X and Huang G (2023) Platelets in the tumor microenvironment and their biological effects on cancer hallmarks. Front. Oncol. 13:1121401. doi: 10.3389/fonc.2023.1121401

Received: 11 December 2022; Accepted: 17 February 2023;
Published: 03 March 2023.

Edited by:

Almudena Porras, Complutense University of Madrid (UCM), Spain

Reviewed by:

Laura Gutiérrez, University of Oviedo, Spain
Stefania Tacconelli, University of Studies G. d’Annunzio Chieti and Pescara, Italy
Sachio Fushida, NPO corporation Digestive Disease Support Organization (DDSO), Japan

Copyright © 2023 Chen, Zhu, Pan, Chen, Xiong, Li, Chu and Huang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Guichun Huang, huangguichun@nju.edu.cn; Yan Li, yanli_med@nju.edu.cn; Xiaoyuan Chu, chuxiaoyuan000@163.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.