Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

The interaction of platelets with colorectal cancer cells inhibits tumor

growth but promotes metastasis

Léa Plantureux1, Diane Mège1-2, Lydie Crescence1, Estelle Carminita1, Stéphane Robert1,

Sylvie Cointe1,3, Nicolas Brouilly4, Walid Ezzedine1-2, Françoise Dignat-George1,3,

Christophe Dubois1*, Laurence Panicot-Dubois1.

1 Aix Marseille Univ, INSERM UMR1263, INRA 1260, C2VN, Marseille, France.

2 Department of Digestive Surgery, Timone University Hospital, Marseille, France.

3 Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille,

France.

4 Aix Marseille Univ, CNRS UMR 7288, Institut de Biologie du Développement de Marseille

(IBDM), Marseille, France.

* To whom correspondence should be addressed:

Christophe Dubois

Aix Marseille Univ, C2VN, Faculty of Pharmacy, 27 Boulevard Jean Moulin, 13385,

Marseille, France.

Tel: +33(0) 491 835 56

[email protected]

Running title: Paradoxical roles of platelets in cancer

Conflict of interest Statement: The authors declare no competing interests.

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

Platelets promote metastasis, however, their role in tumor growth remains controversial. Here, we investigated the effect of platelet interactions with colorectal tumor cells. Platelets extravasated into the tumor microenvironment and interacted with tumor cells in a -6- dependent manner. The interaction induced platelet spreading, release of their granule content and the generation of three types of microparticles (iMPs) that expressed platelet markers, tumor markers, or both. The presence of iMPs was confirmed in colorectal cancer tissue specimens. Platelets significantly reduced tumor growth and increased intratumoral macrophages. This was mediated by iMP recruitment of macrophages via the chemoattractants RANTES, MIF, CCL2 and CXCL12 and activation of their tumor cell killing capacity through IFN-gamma and IL-4 which led to cell cycle arrest of tumor cells in a p21-dependent manner. In contrast, in the bloodstream, iMPs activated endothelial cells and platelets and induced epithelial-to-mesenchymal transition of tumor cells, promoting metastasis. Altogether, these results indicate that depending on the environment, local or bloodstream, the consequences of the interactions between platelets and a tumor may promote or prevent cancer progression.

Statement of significance:

Tumor cell interaction with platelets produces chimeric extracellular vesicles that suppress primary tumor growth by activating tumor-eliminating macrophages, while promoting metastasis through EMT and endothelial activation.

2

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

Platelets were first described as the major effectors of hemostasis and thrombosis. However, platelets may also actively participate in the progression of malignancies(1). Since pioneering studies performed in the early 1970s, increasing evidence has shown that the release of platelet agonists and the expression of procoagulant , such as tissue factor (TF)

(2),(3),(4), by tumor cells induce platelet activation and aggregation in the bloodstream and actively participate in the process of metastasis. Subsequently, numerous studies, including ours, have demonstrated that the inhibition of platelet activation by clopidogrel, ticagrelor, or daily aspirin impairs tumor metastasis in different animal models and in humans (5),(6),(7). In addition to their role in the development of metastasis, platelets have also been reported to participate in tumor growth, although the latter is still subject to controversy. On the one hand, studies have demonstrated that platelet-released factors promote tumor cell proliferation and prosurvival signaling (8),(9). On the other hand, other studies have shown that platelets and platelet-derived microparticles exert antiproliferative and cytotoxic effects on many tumor cells (10),(11),(12).

The presence of extravasated intratumoral platelets was recently suggested in mouse models of ovarian cancer (13), melanomas and breast carcinomas and was associated with tumor vessel structure and metastasis (14). The mechanisms by which platelets extravasate appear to be dependent on P- (15) and on the platelet Focal Adhesion Kinase (FAK) in unstable and leaking tumor vessels (16). The role of intratumoral platelets in tumor growth is currently unknown. In this study, we investigated the presence of platelets within the colorectal tumor microenvironment and their roles in tumor growth and metastasis. Our findings demonstrated that platelets extravasate into the microenvironment of colorectal

3

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

tumors and interact directly with tumor cells without aggregation. Using a blocking antibody and shRNA strategy, we showed that these interactions are dependent on cadherin-6 and lead to decreased tumor growth in vivo. In contrast, in a circulatory and metastatic context, platelet-tumor cell interactions induce epithelial-to-mesenchymal transition (EMT) and increase the adhesive ability of colorectal tumor cells to interact with the endothelium.

4

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Methods

Cell culture

A stable clone of murine colorectal cancer cells expressing GFP and luciferase named

CT26GFP/Luci was obtained by infection of a CT26 WT cell line (ATCC® CRL-2638™, used at passage 7) with lentiviral particles (Amsbio LVP436-PBS) in the presence of polybrene (Life

Technologies, 10 µg). A CT26GFP/Luci clone was selected and cultured in RPMI 1640 supplemented by 10% of decomplemented FCS, 100 U/mL of penicillin, 100 mg/mL of streptomycin, 2 mM of glutamine, 0.1% fungizone and 0.02 mg/mL of blasticidin. Human colorectal cancer cell line HT-29 (ATCC® HTB-38™) was cultured in McCoy’s 5A medium supplemented by 10% decomplemented FCS, 100 U/mL of penicillin, 100 mg/mL of streptomycin, 2 mM glutamine and 0.1% fungizone. The cells were tested for mycoplasma

and were grown, mycoplasma free, at 37°C in a humidified atmosphere with 5% CO2. All cells were used with passages under 20.

Interactions between colorectal tumor cells and platelets

The kinetics of the interaction were measured at several end points (10, 30, 60, 120, 180 and

360 minutes). Briefly, 1.9×106 HT29 or 1.7×106 CT26GFP/Luci cells were seeded into 6-well plates (Thermo-Nunc, Thermo Fisher Scientific, France) for 18 hours. Washed human or murine platelets were added to tumor cells (50:1 ratio of platelets to tumor cells) in an appropriate prewarmed FCS-free medium. The interactions were stopped by extensive washing, and platelets interacting with tumor cells were fixed in 2% PFA or harvested to obtain total protein lysates. The interaction supernatants (called iSN) were collected, cellular debris was removed by centrifugation, and finally, the samples were snap frozen and stored at

-80°C. In the spreading experiments, nonadherent platelets were removed after 60 minutes of interaction by extensive washing, and then the appropriate prewarmed FCS-free medium was

5

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

added. Alternatively, interactions were performed in blocking conditions: platelets were incubated with cadherin-6 blocking antibody (20 or 40 µg/mL), RGDV (100 µM) or both for

20 minutes before contact with cancer cells.

Purification and characterization of the iMPs contained in the iSN and in the plasma of patients with colorectal cancer iSN were centrifuged at 1000 ×g for 15 minutes, then centrifuged at 20 000 ×g for one hour, and the iMP pellet was resuspended in FCS-free McCoy’s medium. The iSN or the purified iMPs were incubated with FITC- V (Tau technology, Ozyme, France), PE anti-human

Epcam and APC anti-human CD41. The flow cytometry instrument settings and microparticle gating were performed with Megamix beads.

A total of 13 patients who underwent surgery for colorectal cancer in the Digestive Surgical

Department of Timone Hospital (Marseille, France) were included in the study, and the samples were issued from our local collection database (DC2013-1815). Platelet-poor plasma containing MPs was obtained as previously described (17). The circulating iMPs contained in the PPP samples were analyzed using a Cytoflex flow cytometry instrument. The iMPs were detected with a combination of FITC-conjugated Annexin V, a PC7-conjugated CD41 antibody and a PE-Dazzle-conjugated Epcam antibody. All analyses were performed with

Kaluza software.

Interactions between human colorectal tumor cells, platelets and monocytes

Interactions between HT29 and monocytes were examined in the presence or absence of platelets. Briefly, purified monocytes and platelets were added simultaneously to HT29 cells at a ratio of 1:100 (monocytes: platelets) in FCS-free prewarmed McCoy’s 5A medium for 6 hours. Alternatively, in some experiments, the monocytes were added to the HT29 tumor cells after 6 hours of platelet-cancer cell interactions. The interactions were stopped after 6 hours by extensive washing, and cancer cells were harvested for protein lysates.

6

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Transwell assay iSN, iMPs (15 million total iMPs), and control supernatants were added to the lower chambers, and 50 000 calcein-green-labeled purified human monocytes were applied to the upper chamber. After 4 hours of incubation, the number of transmigrating monocytes was assessed by a tile scan of each well using a Leica DMi8 fluorescence microscope.

Dynamic interactions between HT29 cells and human umbilical vein endothelial cells

(HUVECs)

HUVECs, used at passages between 2 and 3, were seeded onto tissue-culture-treated microslides (Ibidi, Biovalley, France) and allowed to adhere for 18 hours under flow.

HUVECs were treated with or without iSN, iMPs, or controlSN for 3 hours. Calcein-green- labeled HT29 cells were primed with or without platelets for 1 hour. Then, 0.5×106 primed

HT29 cells were added to the perfusion set and allowed to interact with the endothelium monolayer at 4 dynes/cm2 for 1 hour. The noninteracting tumor cells were removed by extensive washing, and the endothelial cells were fixed using PFA 2%. HT29 cells interacting with HUVECs were analyzed and counted using a Leica DMi8 fluorescence microscope 20X objective on 20 fields per condition and per experiment.

Human cytokine array

Human cytokine arrays were performed according to the manufacturer’s instructions (R and D

System) with iSN and iMPs with or without permeabilizing conditions (1% Triton X-100).

Ectopic syngeneic colorectal cancer mouse model

Wild-type Balb/cByJRj mice were obtained from Janvier Elevage (Janvier Labs, France) and housed under standard conditions. All animal care and experimental procedures were performed as recommended by the European Community guidelines and were approved by local ethical committee number 14 (number 2018032713142114). Six- to eight-week-old mice were anesthetized with 2% isoflurane gas. CT26GFP/Luci or CT26 shRNA cdh-6 clones

7

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

were incubated with platelets or Tyrode buffer for one hour. Colorectal cancer cells were washed extensively before injection into the right flank of the mice (0.5×106 cells). Tumors were allowed to grow for 30 days and measured twice a week with an electronic caliper.

Statistics

For in vitro experiments and tumor volume measurement, significance was determined using a two-tailed Student’s t-test. The differences were significant at p<0.05.

Data sharing statement

For original data, please contact [email protected]

8

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Results

Platelets were present in the tumor microenvironment and interacted with tumors through the expression of Cadherin 6.

To decipher the role played by platelets in the tumor microenvironment, we first determined if platelets were present apart from blood vessels, namely, within a colorectal tumor. We developed an ectopic colorectal cancer model using CT26 GFP/Luci cancer cells.

Thirty days after the injection of the cells, when the tumors reached a volume of approximately 1500 mm3, some platelets were detected apart from the tumor blood vessels

(Figure 1A) and directly interacting with the cancer cells (Figure 1B). Intratumoral platelets were mostly detected at the periphery and not at the center of the tumors (Figures 1C and

1D) and represented approximately 20% of the total platelets within the tumor (Figure 1E).

The presence of intratumoral platelets was confirmed in poorly differentiated colorectal human adenocarcinoma (Figure 1F). We next studied the interactions of platelets with colorectal cancer cells in vitro, in the absence of plasma, to reproduce the intratumoral conditions. Interestingly, the colorectal cancer cell line HT29 did not induce aggregation in the presence of washed platelets (Supplemental Figure 1), indicating that the intratumoral interactions of platelets with cancer cells are independent of the classical IIß3-fibrinogen platelet aggregation pathway. This finding was confirmed by studying the kinetics of the interactions between the two cellular partners; single platelets, not aggregates, adhered to the cancer cells, and the number of adherent platelets increased over time to reach 200 platelets per field after 60 minutes (Figures 2A and 2B). This result was confirmed under dynamic conditions at 6 dynes/cm2. This interaction was specific for cancer cells: under the same conditions, platelets did not adhere to HUVECs (Figures 2C and 2D).

Dune and collaborators demonstrated that platelets express cadherin 6 (18), which is a type II cadherin that possesses an RGD motif, allowing it to interact in a homophilic manner with

9

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

other cadherin-6 proteins and in a heterophilic manner with through the RGD motif.

To date, the functional roles of in platelet-cancer cell interactions have not yet been investigated. We hypothesize that cadherin 6 could be involved in the interactions of platelets with colorectal cancer cells. Flow cytometry analysis confirmed that platelets and HT29 cancer cells expressed cadherin-6 (Figure 2E). The expression of cadherin-6 by HT29 was further confirmed by immunofluorescence (supplemental Figure 2A), RT-PCR (Figure 2F) and sequencing (supplemental Figures 2B and 2C). When using a cadherin 6 blocking antibody or the RGDV peptide, the interactions between platelets and cancer cells in the static

(supplemental Figures 3A and 3B) and dynamic conditions were reduced by 46% and 21%, respectively. When both the homophilic and heterophilic cadherin 6 interactions were blocked, the interactions between platelets and cancer cells were reduced by 76% in comparison to the control (Figure 2G). Taken together, these data indicate that the interactions of colorectal cancer cells with single platelets are mainly cadherin-6 dependent.

The interaction of platelets with cancer cells induced the generation of microparticles detected in patients with colorectal cancer.

During the first 30 minutes following the interactions of single platelets with colorectal cancer cells, we did not observe any changes in the platelet morphology. However, starting at 60 minutes postinteraction, nondiscoid platelets with an area greater than 10 µm2, corresponding to spread platelets, were detected on the surface of cancer cells (Figure 3A-B).

The interaction of platelets with cancer cells also led to the generation of microparticles

(MPs). Platelet-derived microparticles (PMPs) were observed in the supernatant after 60 minutes of interaction between platelets and cancer cells (iSN) and was significantly increased at 360 minutes to 2000 MPs/µL. This generation of PMPs was not due to the apoptosis of platelets since, in the absence of cancer cells, the control platelets generated

10

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

fewer than 100 MPs/µl at 360 minutes (Figure 3C, left panel). Tumor-derived microparticles

(TMPs) were also detected at a concentration of 420 MPs/µL after 360 minutes of interaction.

In this case, however, the number of MPs was not significantly different from the number of

MPs detected in the absence of platelets (Figure 3C, middle panel), mainly because the tumor cells constantly generated MPs (19). Interestingly, the interaction of platelets with tumor cells also led to the production of MPs expressing both CD41 and Epcam at their surface (T+PMPs) with a concentration of 200 MPs/µl at 360 minutes (Figure 3C, right panel). We excluded the possibility that T+PMPs were MP aggregates by performing serial dilutions of the sample. We indeed observed a direct correlation between the dilution of the sample performed and the quantity of T+PMPs detected (r2 = 0,9288). Thus, the interaction of platelets with cancer cells induced the production of 3 different types of MPs: 76% PMPs,

16% TMPs and 8% T+PMPs. We named these MPs (PMPs, TMPs and T+PMPs) iMPs for platelet- and tumor cell-interacting microparticles.

We next determined whether the T+PMPs could be detected at the surface of cancer cells interacting with platelets by confocal microscopy. As illustrated in Figure 3D, vesicles expressing Annexin V, CD41 and Epcam were identified on the surface of cancer cells interacting with platelets. Spread platelets, by secreting their granule contents into the cancer cells, might have induced a membrane redistribution that was involved in the production of

T+PMPs. To test this hypothesis, we injected fluorescent fibrinogen into recipient mice. After

1 day, platelets isolated from these mice were found to express fluorescent fibrinogen in their granules (Figure 3E). After interaction with tumor cells, a signal corresponding to fluorescent fibrinogen was detected in the cancer cells apart from the platelets (Figure 3F), which indicated that platelet granule contents were indeed released into the cancer cells and, consequently, that the platelet and cancer cell membranes were fused.

11

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

We next determined whether T+PMPs are indeed generated in patients with colorectal cancer. MPs expressing both Epcam and CD41 were detected in the plasma of all 13 patients studied, with concentrations ranging from 20 to 257 and a median of 70 MPs/µL of plasma

(Figure 3G). These T+PMPs were also detected in the tumor microenvironment of human colorectal tumors (Figure 3H).

Altogether, these results indicated that upon interacting with cancer cells, the platelets spread, released their granules into the cancer cells and secreted MPs. The fusion of platelet and cancer cell membranes led to the production of iMPs, including MPs expressing markers of both platelets and cancer cells, which were detected in patients with colorectal cancer.

To characterize the sizes of the iMPs generated in greater detail, the different populations of iMPs produced after 6 h of interactions between platelets and cancer cells were isolated using a cell sorter (Figures 4A and 4B) and analyzed by electron microcopy (Figures 4C and 4D).

More than 50% of the iMPs had a size between 100 and 300 nm. None of them was greater in size than 700 nm. All three types of iMPs had similar size distributions; however, 34% of

T+PMPs had a size between 200 and 300 nm, versus 24 and 27% for the PMPs and TMPs, respectively. Additionally, 12% of TMPs had a size between 500 and 600 nm versus 4% for

PMPs and T+PMPs.

The generated iMPs recruited monocytes and contained two cytokines (IFN-gamma and

IL-4) involved in activating the tumoricidal functions of macrophages.

To determine the role of iMPs, we primed mouse colorectal cancer cells

(CT26GFP/Luci) with platelets before injecting them into the right flank of wild-type mice.

When we compared the size of the tumors in the presence and absence of platelets, we observed, starting at day 18, that both the tumor volumes and the bioluminescent signal at day

12

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

28 were significantly lower in the group of mice that received CT26GFP/Luci cells primed with platelets than in the control group (Figure 5A). When Cadherin 6 was blocked from cancer cells using shRNA (Figures 5B and 5C), the priming of the cancer cells with platelets no longer affected the growth of the tumor (Figure 5A), indicating that cadherin 6-dependent interactions of platelets with cancer cells were involved in the decreased tumor size.

Interestingly, the shutdown of Cadherin 6 in cancer cells by shRNA almost completely blocked the inhibitory role of platelets in colon cancer cell progression in vivo. However,

Cadherin 6 knockdown inhibited platelet adhesion only slightly in vitro (supplemental

Figures 3A and 3B). To understand this apparent discrepancy, we next examined the generation of iMPs following the interaction of platelets with cancer cells not expressing

Cadherin 6. We observed that the shutdown of Cadherin 6 by shRNA significantly diminished the concentration of iMPs generated following interaction with platelets and mostly abolished the production of PMPs (Figure 5D), suggesting that cadherin 6 expressed by cancer cells is involved in the activation of platelets leading to the generation of PMPs. We concluded that

Cadherin 6 expressed by cancer cells plays a key role in the generation of iMPs.

When comparing the cellular composition of the primary tumor in the presence and absence of platelets, no difference was observed in the presence of intratumoral neutrophils in the two groups of mice. However, the F4/80 fluorescent signal corresponding to macrophages was significantly higher in the tumor microenvironment of mice injected with CT26GFP/Luci cells primed with platelets than in the control mice (Figure 5E). The presence of platelets significantly increased the number of intratumoral macrophages (Figure 5E).

13

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Using a Transwell assay, we observed that monocytes transmigrated from the upper to the lower chamber when the iSN or the purified iMPs were present in the lower chamber. In contrast, the supernatants from platelets alone or cancer cells alone containing PMPs and

TMPs, respectively, were significantly less effective than iMPs in attracting monocytes

(Figure 5F). Taken together, these results indicated that the iMPs produced following the cadherin 6-dependent interaction of platelets with cancer cells could attract and activate monocytes, whereas PMPs or TMPs alone could not. These results strongly suggested that the

T+PMPs were directly involved in this process.

To determine how iMPs attract macrophages at the site of a primary tumor, we next performed a cytokine array on the supernatant of platelets alone and of cancer cells alone, on the iSN and on purified iMPs (Figure 5G). RANTES and MIF, two cytokines known to be involved in the recruitment of leukocytes, were detected in the iSN and in the iMPs. Of note, only one cytokine, MIF, was detected in the supernatant of cancer cells. Interestingly, various cytokines were present in iMPs only when the iSN and the iMPs were permeabilized. These cytokines included CXCL12 and CCL2 (involved in the attraction of leukocytes), IFN- and

IL-4 (known to activate macrophages and to be involved in macrophage tumoricidal activity)(20). These results indicated that cytokines in iMPs can attract macrophages (CCL2,

CXCL12) and activate their tumoricidal capacity (IFN- and IL-4). Furthermore, based on the expression of iNOS and Arg1, we found that 89% (+/- 34) of the macrophages were polarized into a M1 phenotype in the tumor primed with platelets versus 38% (+/-32) in the control mice (Figure 5H).

We next determined whether platelets and macrophages could indeed affect cancer cell proliferation or apoptosis. The interaction of platelets alone (Figure 6A-C) or monocytes alone (Figure 6B) with cancer cells did not induce cancer cell apoptosis. The expression of

P21 (Waf1/Cip1) was unchanged in cancer cells after platelet interaction (Figures 6D and

14

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

6E). However, the interaction of monocytes alone or monocytes plus platelets with cancer cells induced the expression of the cyclin-dependent kinase inhibitor P21 (Waf1/Cip1) in a time-dependent manner (Figures 6F and 6G). When platelets and monocytes were sequentially added to cancer cells, the expression of Rb phosphorylated at positions 807 and

811 in HT29 cancer cells was significantly decreased (Figure 6H). Taken together, these results indicated that in the tumor microenvironment, tumoricidal macrophages were recruited and activated by iMPs.

Platelets and iMPs facilitated the interaction of cancer cells with the endothelium.

Our results indicated that in the tumor microenvironment, platelets play an important role in reducing tumor growth through the generation of iMPs and the recruitment of tumoricidal macrophages. However, different studies, including ours, have demonstrated that activated platelets may participate in the process of metastasis. To understand this apparent discrepancy, we next studied the role of platelets in cancer cell-endothelium interactions, a key step involved in metastasis. We first investigated the adhesive properties of HT29 colorectal cancer cells to the endothelium in the presence of absence of platelet priming in dynamic conditions (4 dynes/cm2). As illustrated in Figure 7A, compared to control levels, a tenfold increase in the adhesion of cancer cells to the endothelium was observed when cancer cells were primed with platelets. Interestingly, during priming, platelets released and transferred subunit ß3-positive (CD61) vesicles to the cancer cells (Figures 7B), and these vesicles might have participated in the interactions with the endothelium. As an alternative to this mechanism, a few studies have demonstrated that platelet-cancer cell interactions increase the migratory properties of cancer cells by inducing EMT. To determine whether platelet interaction with HT29 colorectal cancer cells induced EMT, we assessed the expression of E-cadherin and Snail1, two known markers involved in EMT. The interaction of

15

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

platelets with cancer cells decreased the expression of E-cadherin and increased the expression of snail1 in a time-dependent manner (Figure 7C). These results were confirmed in vivo using a model of experimental metastasis (21) (supplemental Figure 3C). The priming of cancer cells with platelets significantly increased the number of metastatic modules detected in the lungs.

To better understand the transfer of biological material from platelets to cancer cells, we next compared the expression of more than 200 mRNA involved in inflammation and metastasis in platelets alone, cancer cells alone and cancer cells primed with platelets (Figure

7D). Interestingly, some mRNAs that were absent in cancer cells alone were overexpressed when cancer cells were primed with platelets. For example, consistent with a previous publication (22), the mRNA PTGS2, coding for COX2, is overexpressed following the interaction of platelets with cancer cells. Other mRNAs are shut down following the interaction of cancer cells with platelets, including the mRNAs coding for SerpinB5, also known as Maspin, and NF-2, coding for two proteins reported to function as tumor suppressors, that suppress the ability of cancer cells to invade and metastasize to other tissues and are thus described as playing important roles in metastasis. We also confirmed that platelet interactions with cancer cells play an important role in EMT since the expression of

E-cadherin was diminished, whereas the mRNA CTNNA1, coding for beta-catenin, MMP10 and MMP2 (SNAIL et ZEB family) and fibronectin (SNAIL et TWIST family) were overexpressed in cancer cells primed with platelets.

We next investigated the ability of iMPs to prime the endothelium and facilitate the adhesion of cancer cells at the site of metastasis. Purified iMPs activated HUVECs, as attested by the induction of the expression of ICAM-1 at the surface of the endothelial cells (Figure

7E). In a dynamic assay, at 4 dynes/cm2, purified iMPs, as well as the interaction supernatant,

16

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

significantly increased the percentage of HT29 in interaction with HUVECs in comparison to the control (Figure 7F). Altogether, these results indicate that platelets and iMPs may contribute to metastasis by favoring interaction between cancer cells and the endothelium.

Discussion

The role of platelets and platelet activation in tumor growth has been subject to controversy in the literature. Some studies have demonstrated that the use of drugs that inhibit platelet activation, such as ticagrelor and clopidogrel, reduces tumor growth and metastasis in vivo in pancreatic cancer (1)(23). Other studies performed on breast, colorectal and prostate cancer did not observe any effect of clopidogrel or prasugrel alone on the tumor growth

(24,25). The activation of platelets might play different roles in tumor behavior and metastasis in different cancers. We previously reported that in pancreatic cancer, inhibition of platelets decreases tumor growth and metastasis (5). In this case, cancer cells strongly activate platelets, leading to TCIPA, which is mainly dependent on IIbß3 interactions and not cadherin-6. Here, in a colorectal model, we observed a paradoxical role of platelets

(Supplemental Figure 4). Platelets mainly interacted with the cancer cells through cadherin-

6, independent of TCIPA. Although our results indicate that Cadherin 6 plays a crucial role in generating iMPs, other receptors expressed by platelets, including GPVI or P-selectin, may also be involved in the interaction of platelets with cancer cells (22). A deep characterization of the role played by platelets depending on the type of cancer and its stage is needed before using antiplatelet drugs in the management of cancer and cancer-associated thrombosis.

The kinetics of platelet activation and spread on colorectal cancer cells was different from what is usually described, with the activation of platelets starting as late as 60 minutes following their interaction with cancer cells. Steele et al. demonstrated that the canonical

17

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Wnt/ß-catenin signaling components are present in platelets and negatively regulate platelet functions (including platelet αIIbß3 activation, platelet adhesion and spread, and dense granule secretion) by regulating small GTPase activity (26,27). Later, Maguire et al. demonstrated that platelet ß-catenin is strongly associated with cadherin adherents junctions including cadherin-6 (28). It is possible that the cadherin-6 dependent interactions between platelets and tumor cells are responsible for the absence of TCIPA and the observed kinetics of platelet spreading through activation of the Wnt/ß-catenin signaling pathway. Of note, consistent with previous publications (29)(30), the tumor size in the Cdh6 knock-down group was smaller than that in the control group, indicating that Cadherin 6 expressed by cancer cells is involved in the proliferation of colorectal cancer cells. Since we observed that the shutdown of

Cadherin 6 by shRNA strongly affected the generation of iMPs, it is possible that the role played by Cadherin 6 expressed by cancer cells in tumor growth is mainly mediated through its interactions with platelets and the generation of iMPs.

For a few years, platelets have been considered effectors of inflammation and immunity, mainly based on the fact that platelet -granules contain a plethora of chemokines, including RANTES, MIF, CCL-3, CXCL5 and CXCL12, which are all known to attract leukocytes in different pathological conditions such as bacterial infections and atherosclerosis

(31,32). One study also suggests that platelets alone can exert directly cytotoxic effects on tumor cells (12). This cytotoxicity is dependent on the type of cancer cells used and requires an interaction between platelets and cancer cells when the platelets are not artificially preactivated (33). Our results indicate that the cytotoxic effect of platelets on cancer cells is mainly due to the production of microparticles involved in recruiting and activating macrophages that facilitate cell cycle arrest. Further studies will determine if this pathway is

18

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

specific to colorectal cancer or is also activated following the interaction of platelets with other types of cancers.

Numerous studies underline the effects of platelet microparticles (PMPs) on the recruitment and activation of monocytes (34)(35). This process classically induces oxidative burst activation of monocytes and the release of proinflammatory cytokines such as TNF-

(36,37). In accordance with these studies, we showed that iMPs (containing RANTES, MIF,

CXCL-12, and IFN-) that were released following platelet-tumor cell interactions increased the attraction of monocytes and that monocyte – platelet – tumor cell interactions induced cell cycle arrest in vitro and decreased tumor growth in vivo. Proinflammatory cytokines, such as

IFN- and TNF-, are known to induce M1 macrophage polarization with strong tumoricidal activity (38). The tumoricidal activity of macrophages may be due to cytokine‐mediated induction of cell death and phagocytosis such as TRAIL, CCL2 and IL‐8 (39). Alternatively, macrophage‐mediated phagocytosis could be an important part of the antitumoral immune response. However, the exact mechanisms supporting the tumoricidal activity of the macrophages in vivo remained to be determined.

Recently, the notion of tumor-educated platelets has been highlighted and provides an interesting tool for cancer diagnostics (40–42). The main way a tumor influences platelet

RNA profiles is through the uptake of tumor-derived microvesicles (41). Here, we demonstrated that platelets can also educate tumor cells via (i) the release of their alpha- granule contents into the tumor cells (ii) the microvesicle-dependent transfer of adhesive proteins, such as the ß3-integrin subunit (iii) the fact that platelet transfer could induce the expression or repression of different RNAs in cancer cells. Based on our results, we propose that platelet-educated cancer cells also exist and play an important role in tumor behavior and metastasis formation. Here, we focused on and highlighted the role played by iMPs in these

19

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

processes. However, the overexpression/repression of mRNA could also be an essential step in the regulation of the tumor and the formation of metastases. These points need to be clarified.

These mechanisms lead to the induction of EMT and are responsible for the enhanced ability to interact with the endothelium. Moreover, the iMPs generated during platelet-tumor cell interactions prime the endothelium and enhance the adhesion of tumor cells to the endothelium. These results are in accordance with the findings of Pasquier et al., who demonstrated a MP-mediated cross-talk between endothelial cells and tumor cells involved in the activation of endothelial cells and the formation of a prometastatic vascular niche (43).

Janowska et al. demonstrated that PMPs can transfer platelet-derived CD41 in six lung cancer cell lines, which is known to enhance the metastatic potential of cancer cells with an increased ability to adhere to endothelial cells (44,45). Additionally, it has been recently shown that

PMPs can infiltrate solid tumors and suppress tumor growth mainly via the delivery of miR-

24 (10). Thus, platelets (as well as iMPs) that are released in the bloodstream during cancer progression may contribute to the metastatic process by facilitating the interactions between tumor cells and the endothelium.

Altogether, our results indicate that iMPs, by being present in the tumor microenvironment and by disseminating information (via cytokines and integrins) in the bloodstream, play an important role in the behavior of a tumor and may represent an interesting target to successfully treat colorectal cancer as well as a pertinent biomarker of cancer.

20

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Acknowledgements

We would like to thank Dr Pierre-André Jarrot and Pascal Weber for their technical assistance. This work was supported by a grant from the GFTC (Groupe Francophone

Thrombose et Cancer).

21

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References

1. Mezouar S, Frère C, Darbousset R, Mege D, Crescence L, Dignat-George F, et al. Role of platelets in cancer and cancer-associated thrombosis: Experimental and clinical evidences. Thrombosis Research. 2016;139:65–76. 2. Liu Y, Jiang P, Capkova K, Xue D, Ye L, Sinha SC, et al. Tissue factor-activated coagulation cascade in the tumor microenvironment is critical for tumor progression and an effective target for therapy. Cancer Res. 2011;71:6492–502. 3. Rickles FR, Falanga A. Molecular basis for the relationship between thrombosis and cancer. Thromb Res. 2001;102:V215-224. 4. Ruf W, Yokota N, Schaffner F. Tissue factor in cancer progression and angiogenesis. Thrombosis Research. 2010;125:S36–8. 5. 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;136:462–75. 6. Gebremeskel S, LeVatte T, Liwski RS, Johnston B, Bezuhly M. The reversible P2Y12 inhibitor ticagrelor inhibits metastasis and improves survival in mouse models of cancer. Int J Cancer. 2015;136:234–40. 7. Rothwell PM, Wilson M, Price JF, Belch JFF, Meade TW, Mehta Z. Effect of daily aspirin on risk of cancer metastasis: a study of incident cancers during randomised controlled trials. Lancet. 2012;379:1591–601. 8. 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;20:576–90. 9. Egan K, Crowley D, Smyth P, O’Toole S, Spillane C, Martin C, et al. Platelet adhesion and degranulation induce pro-survival and pro-angiogenic signalling in ovarian cancer cells. PLoS ONE. 2011;6:e26125. 10. 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;130:567–80. 11. Wang Y, Zhang H. Platelet-induced inhibition of tumor cell growth. Thromb Res. 2008;123:324–30. 12. Ibele GM, Kay NE, Johnson GJ, Jacob HS. Human platelets exert cytotoxic effects on

22

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

tumor cells. Blood. 1985;65:1252–5. 13. 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;366:610–8. 14. 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. 15. Qi C, Wei B, Zhou W, Yang Y, Li B, Guo S, et al. P-selectin-mediated platelet adhesion promotes tumor growth. Oncotarget. 2015;6:6584–96. 16. Haemmerle M, Bottsford-Miller J, Pradeep S, Taylor ML, Choi H-J, Hansen JM, et al. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal. The Journal of Clinical Investigation. 2016;126:1885. 17. Mege D, Panicot-Dubois L, Ouaissi M, Robert S, Sielezneff I, Sastre B, et al. The origin and concentration of circulating microparticles differ according to cancer type and evolution: A prospective single-center study. Int J Cancer. 2016;138:939–48. 18. Dunne E, Spring CM, Reheman A, Jin W, Berndt MC, Newman DK, et al. Cadherin 6 has a functional role in platelet aggregation and thrombus formation. Arterioscler Thromb Vasc Biol. 2012;32:1724–31. 19. Plantureux L, Mège D, Crescence L, Dignat-George F, Dubois C, Panicot-Dubois L. Impacts of Cancer on Platelet Production, Activation and Education and Mechanisms of Cancer-Associated Thrombosis. Cancers (Basel). 2018;10. 20. Suk K, Somers SD, Erickson KL. Regulation of murine macrophage function by IL-4: IL-4 and IFN-gamma differentially regulate macrophage tumoricidal activation. Immunology. 1993;80:617–24. 21. 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;105:178–85. 22. Dovizio M, Maier TJ, Alberti S, Di Francesco L, Marcantoni E, Münch 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;84:25–40. 23. Elaskalani O, Falasca M, Moran N, Berndt MC, Metharom P. The Role of Platelet- Derived ADP and ATP in Promoting Pancreatic Cancer Cell Survival and Gemcitabine Resistance. Cancers (Basel). 2017;9. 24. Denslow A, Świtalska M, Jarosz J, Papiernik D, Porshneva K, Nowak M, et al. Clopidogrel in a combined therapy with anticancer drugs—effect on tumor growth,

23

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

metastasis, and treatment toxicity: Studies in animal models. PLOS ONE. 2017;12:e0188740. 25. Serebruany VL. Platelet Inhibition with Prasugrel and Increased Cancer Risks: Potential Causes and Implications. The American Journal of Medicine. 2009;122:407–8. 26. Steele BM, Harper MT, Macaulay IC, Morrell CN, Perez-Tamayo A, Foy M, et al. Canonical Wnt signaling negatively regulates platelet function. Proc Natl Acad Sci USA. 2009;106:19836–41. 27. Steele BM, Harper MT, Smolenski AP, Alkazemi N, Poole AW, Fitzgerald DJ, et al. WNT-3a modulates platelet function by regulating small GTPase activity. FEBS Lett. 2012;586:2267–72. 28. Maguire PB, Donlon T, Parsons M, Wynne K, Dillon E, Áinle FN, et al. Proteomic Analysis Reveals a Strong Association of β‐Catenin with Cadherin Adherens Junctions in Resting Human Platelets. PROTEOMICS [Internet]. 2018 [cited 2018 Apr 5]; Available from: https://www.readcube.com/articles/10.1002/pmic.201700419 29. Bartolomé RA, Peláez-García A, Gomez I, Torres S, Fernandez-Aceñero MJ, Escudero-Paniagua B, et al. An RGD motif present in cadherin 17 induces integrin activation and tumor growth. J Biol Chem. 2014;289:34801–14. 30. Casal JI, Bartolomé RA. RGD cadherins and α2β1 integrin in cancer metastasis: A dangerous liaison. Biochim Biophys Acta Rev Cancer. 2018;1869:321–32. 31. Schrottmaier WC, Kral JB, Badrnya S, Assinger A. Aspirin and P2Y12 Inhibitors in platelet-mediated activation of neutrophils and monocytes. Thromb Haemost. 2015;114:478– 89. 32. Wirtz TH, Tillmann S, Strüßmann T, Kraemer S, Heemskerk JWM, Grottke O, et al. Platelet-derived MIF: a novel platelet chemokine with distinct recruitment properties. Atherosclerosis. 2015;239:1–10. 33. Sagawa T, Tominaga A, Kodama T, Okada M. Cytotoxicity of unstimulated and thrombin-activated platelets to human tumour cells. Immunology. 1993;78:650–6. 34. Hosseinkhani B, Kuypers S, van den Akker NMS, Molin DGM, Michiels L. Extracellular Vesicles Work as a Functional Inflammatory Mediator Between Vascular Endothelial Cells and Immune Cells. Front Immunol. 2018;9:1789. 35. Bei J-J, Liu C, Peng S, Liu C-H, Zhao W-B, Qu X-L, et al. Staphylococcal SSL5- induced platelet microparticles provoke proinflammatory responses via the CD40/TRAF6/NFκB signalling pathway in monocytes. Thromb Haemost. 2016;115:632–45. 36. Mause SF, von Hundelshausen P, Zernecke A, Koenen RR, Weber C. Platelet microparticles: a transcellular delivery system for RANTES promoting monocyte recruitment

24

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

on endothelium. Arterioscler Thromb Vasc Biol. 2005;25:1512–8. 37. Vasina EM, Cauwenberghs S, Staudt M, Feijge MA, Weber C, Koenen RR, et al. Aging- and activation-induced platelet microparticles suppress apoptosis in monocytic cells and differentially signal to proinflammatory mediator release. Am J Blood Res. 2013;3:107– 23. 38. Bekisz J, Sato Y, Johnson C, Husain SR, Puri RK, Zoon KC. Immunomodulatory effects of interferons in malignancies. J Interferon Cytokine Res. 2013;33:154–61. 39. Olingy CE, Dinh HQ, Hedrick CC. Monocyte heterogeneity and functions in cancer. J Leukoc Biol. 2019;106:309–22. 40. Calverley DC, Phang TL, Choudhury QG, Gao B, Oton AB, Weyant MJ, et al. Significant downregulation of platelet in metastatic lung cancer. Clin Transl Sci. 2010;3:227–32. 41. Nilsson RJA, Balaj L, Hulleman E, van Rijn S, Pegtel DM, Walraven M, et al. Blood platelets contain tumor-derived RNA biomarkers. Blood. 2011;118:3680–3. 42. Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, et al. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell. 2015;28:666–76. 43. Pasquier J, Thawadi HA, Ghiabi P, Abu-Kaoud N, Maleki M, Guerrouahen BS, et al. Microparticles mediated cross-talk between tumoral and endothelial cells promote the constitution of a pro-metastatic vascular niche through Arf6 up regulation. Cancer Microenvironment. 2014;7:41–59. 44. Janowska-Wieczorek A, Marquez-Curtis LA, Wysoczynski M, Ratajczak MZ. Enhancing effect of platelet-derived microvesicles on the invasive potential of breast cancer cells. Transfusion. 2006;46:1199–209. 45. Janowska-Wieczorek A, Wysoczynski M, Kijowski J, Marquez-Curtis L, Machalinski B, Ratajczak J, et al. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int J Cancer. 2005;113:752–60.

25

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure legends

Figure 1. Platelets are present apart from blood vessels in the tumor microenvironment of mouse and human colorectal tumors.

A. Representative images of the presence of extravasated platelets in the microenvironment of mouse colorectal tumors. Four-micrometer colorectal tumor sections were stained with the indicated antibodies. The tumor microenvironment is depicted in green (CT26GFP/luci), with isolectin B4 in yellow and GPIb- in red. Confocal microscopy, 60X. B. Representative images showing the colocalization of platelets and cancer cells (white). C. Representative images showing differential platelet extravasation into the tumor periphery in comparison to the tumor center. Platelets are depicted in red, vessel are labeled with IB4 in white (upper panel). The tumor localization was confirmed by HIF-1 staining (red), representative of the degree of hypoxia (lower panel). Confocal microscopy, 60X. D. HIF-1positive staining area expressed in µm2 in the tumor periphery and in the tumor center. The data are presented as the mean value +/- SD (p-value: 0.0072; n=3) E. Percentage of the fluorescent signal corresponding to platelet extravasation into the tumor center versus the tumor periphery. The data are presented as the mean value +/- SEM (p-value<0.0001; n=3) F. Representative images of extravasated platelets in the microenvironment of human colorectal tumors. Frozen tissue slides containing 5 µm sections from a poorly differentiated colorectal human adenocarcinoma were stained with the indicated antibodies. CD41 is depicted in red, and

CD31 is shown in green. Confocal microscopy, 60X. All the experiments were performed at least three times. Quantifications were performed in 10 fields per experiment using SlideBook

6.0 software. Two-tailed Student’s t-test, * p-value<0.05; ** p-value<0.01*** p-value<0.001.

26

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2. Interactions of platelets with colorectal tumors are cadherin-6 dependent.

A. Representative images of platelet-HT29 cell interactions at 10, 30 and 60 minutes. DiD- stained human washed platelets (hWPs) were added to calcein-green-stained HT29 cells at a ratio of 50:1 (platelets: cancer cells) for 10, 30 and 60 minutes. HT29 cells are depicted in brightfield (left), and DiD-stained hWPs are shown in red (middle); merged image (right).

Bars – 50 µm. B. Graph representing the mean number (+/- SD) of DiD-positive platelets in interaction with HT29 cells / field, with 10 fields analyzed per experiment (p-value<0.05; n=3) C. Representative images showing the DiD-stained hWP and calcein-green-stained

HT29 cell interaction after 60 minutes of interaction under dynamic conditions (6 dynes/cm2)

(upper panel). Representative images showing the absence of an interaction between DiD- stained hWPs (red) and Hoechst 33342-stained endothelial cells (HUVEC-blue) after 60 minutes under dynamic conditions (6 dynes/cm2) (Lower panel). D. The graph depicts the mean number (+/-SD) of DiD-positive platelets/field in interaction with HT29 cells or

HUVECs. (p-value<0.001; n=3) E. Expression of cadherin-6 on HT29 cells and hWP assessed by flow cytometry using indirect immunostaining with an unconjugated anti-human cadherin-6 IgG antibody (10 µg/mL) and AF649-conjugated anti-rabbit IgG secondary antibody (5 µg/mL). Cadherin-6-specific staining (green); matched isotype control (red). F.

RT-PCR using SSVI retrotranscriptase and cadherin-6 forward and reverse primers on total extracted RNA from HT29 cells and Mo59K human malignant glioblastoma cells (ATCCⓇ

CRL-2365TM, positive control). G. Cadherin-6 inhibition decreases the interaction of hWPs with HT29 cells. The hWPs were incubated with blocking antibody against cadherin-6 (40

µg/mL) and/or RGDV (100 µM) or control IgG (40 µg/mL) for 20 minutes before their addition to the HT29 cells in dynamic conditions (6 dynes/cm2). The results are expressed as the mean percentage of the interaction between the hWPs and HT29 cells in blocking

27

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

conditions in comparison to the control after 60 minutes of interaction +/- SD. (p-value<0.05; n=4). Quantifications were performed on 10 fields per experiment using ImageJ software.

Two-tailed Student’s t-test, * p-value<0.05; ** p-value<0.01*** p-value<0.001. hWPs: human washed platelets; HUVECs: human umbilical vein endothelial cells.

Figure 3. The interaction of platelets with tumor cells induces the generation of microparticles.

A. Representative images of hWP spreading on HT29 cells. CD41 (red), CD29 (green).

Confocal microscopy, 100X. B. Quantification of platelet spreading. The results are presented as the mean percentage value of spread platelets relative to the total number of platelets +/-

SD (n=4). Spread platelets were defined by their form (nondiscoid platelets) and their area

(above 10 µm2). C. Quantification of the generation of microparticles (MPs) in the supernatant of the interaction (iSN), control platelets supernatant or control tumor cells supernatant. The results are expressed as the mean concentration of the MPs released in the supernatants in MPs/µL at different timepoints +/- SD (n=4); left panel: Annexin V+ CD41 + and Epcam- microparticles (Platelet microparticles, PMPs); medium panel: Annexin V+

CD41– and Epcam+ microparticles (Tumor cell derived MPs, TMPs) and right panel:

Annexin V+ CD41+ and Epcam+ microparticles (T+PMPs). D. Representative images showing the formation of iMPs at 6 hours. CD41 is shown in red, Epcam in blue, AnV in green, and colocalization in white. Confocal microscopy, 100X. E. Flow cytometry characterization (left) and immunofluorescence (right) of fibrinogen (FGN)-positive washed platelets 24 hours after the intravenous injection of AF488-conjugated FGN in recipient mice.

FGN is present in the platelet granules. CD41 is displayed in red. F. Representative images demonstrating the internalization of AF488 FGN in the tumor cells after 6 hours of interaction. CD29 is presented in blue and CD41 in red; the platelets were stained in vivo as previously described. Leica microscope DMi8, 63X oil immersion; 0.2 µm stack conditions

28

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and Huygens deconvolution processing. G. Quantification of iMPs (AnV+, CD41+ and

Epcam+ MPs) in plasma from patients with colorectal cancer. H. Representative images demonstrating the presence of iMPs in the microenvironment of human colorectal tumors.

Frozen tissue slides containing 5 µm sections from a poorly differentiated, human colorectal adenocarcinoma. Right – crops corresponding to the white rectangular region. CD41, red;

Epcam, green. Confocal microscopy, 60X. Two-tailed Student’s t-test, * p-value<0.05; ** p- value<0.01*** p-value<0.001. hWP: human washed platelets.

Figure 4. Isolation and characterization of the iMPs.

A. Astrios FSC an SSC parameters were adjusted with Megamix + FSC and SSC beads and

MP gate was defined. B. Supernatant of cocultured cells and platelets were first analyzed on the cell sorter. Sorting regions were defined using isotypic controls for EpCAM and CD41 antibodies and analyzed to evaluate sort purity. C. Electron microscope images of sorted microparticles: platelet-derived microparticles (PMPs, left), cancer cells-derived microparticles (TMPs, middle) and platelet + cancer cells-derived microparticles (T+PMPs, right). Microparticles were fixed in PFA 1%, left on formvar grids overnight at 4°C. After night sedimentation, grids were contrasted with uranyl acetate for 5 minutes. Magnification:

89K (up) and 175K (bottom). D. Size repartition of microparticles in percentage of MPs counted in function of the size. At least 117 microparticles were analyzed for each condition.

Figure 5. Platelet-tumor cell interactions decrease tumor growth in vivo via the attraction of tumoricidal macrophages.

A. Tumor growth curve of 60 minutes +/- platelet-primed colorectal tumors expressing (sh

RNA MOCK) or not (sh RNA Cdh6) cadherin 6. Tumor volumes are expressed in mm3 and are presented as the mean value +/- SD (p-value<0.05; n=11 per group). B. RT-PCR using

29

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

SSVI retrotranscriptase and cadherin-6 forward and reverse primers on total extracted RNA from the CT26 shRNA control and the CT26 shRNA cdh-6. C. Western blot analysis of the cadherin-6 protein expression on total protein lysates from the CT26 shRNA control and the

CT26 shRNA cdh-6. D. Percentage MPs generated (iMPs, TMPs, PMPs and T+PMPs) following 60 minutes interaction of platelets when cadherin 6 is shut down (shRNA Cdh6) from CT26 cancer cells in comparison with control (shRNA Mock). E. Tumor growth curve of 60 minutes platelet-primed CT26 shRNA cdh6 and control CT26 shRNA. (ns; n=6 per group). F. Representative images showing macrophage infiltration (F4/80-positive cells) in the microenvironment of platelet-primed colorectal tumors (CT26GFP/Luci + platelets) and control colorectal tumors (CT26GFP/Luci). Four-micrometer tumor sections were stained by

F4/80 antibody (red). Confocal microscopy, 60X (left panel). The quantification of the fluorescent signal is represented on the right panel (p-value<0.01; n=3). G. Representative graph of the monocyte transwell assay in the presence of free FCS McCoy’s 5A medium

(medium control), 6-hour platelet supernatant control (Plts SN control), 6-hour HT29 supernatant control (HT29 SN control), TNF-a (100 U/mL), iSN and purified iMPs. The results are expressed as the mean percentage of transmigrating monocytes in comparison with that in the medium control (p-value<0.05; n=3) H. Quantification of cytokine and chemokine levels in the iSN and in the purified iMPs under permeabilized conditions (0.1% Triton X100) or nonpermeabilized conditions. The graph depicts the normalized level of cytokines relative to reference spots using ImageJ software. I. Quantification of the fluorescent signal of

Arginase 1 (Arg1 - left) and inducible Nitric oxide synthase (iNOS- right) colocalizing with

F4/80 signal in platelet primed CT26 cells tumor microenvironement and in control CT26 cells tumor microenvironment. Two-tailed Student’s t-test, * p-value<0.05; ** p- value<0.01*** p-value<0.001. hWPs: human washed platelets; iSN: 6-hour hWP-HT29 interaction supernatant; iMPs: iSN-purified MPs.

30

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 6. Platelets and their MPs affect tumor cell proliferation in the presence of monocytes.

A. Western blot analysis of the cleavage of caspase 3: noncleaved caspase 3 (35 kDa) and cleaved caspase 3 (17 and 12 kDa). The positive control was a treatment of HT29 cells with

200 µM camptothecin (CPT) for 24 h. B. Western blot analysis of the cleavage of caspase 3 induced by monocytic interaction. C. Flow cytometry analysis of the apoptosis of HT29 cells.

The graph depicts the mean percentage of AnV-positive and 7-AAD-positive HT29 cells at different times of interaction relative to the total number of analyzed HT29 cells +/- SD (p- value<0.05; n=3, Kaluza software). D. Western blot analysis of the P21 expression in total protein lysates from HT29 cells that interacted with hWPs. E. Relative protein expression level of P21 in HT29 cells at different times of interaction with hWPs +/- SD. (ns; n=3) F.

Western blot analysis of the P21 expression in total protein lysates from HT29 cells that interacted with human purified monocytes, or human purified monocytes and hWPs. Kinetic measurements of the interaction between human HT29 cells and human purified monocytes in the presence or absence of hWPs from the 60-minute timepoint to the 360-minute timepoint were performed. G. The relative protein expression level of P21 in HT29 cells at different times of interaction with human purified monocytes in the presence or absence of hWPs +/-

SD. (p-value < 0.05; n=3) H. Western blot analysis of the PRb 807/811 expression in total protein lysates from HT29 cells that interacted with hWPs for 5 hours followed by 5 hours of interactions with purified human monocytes (top panel). The relative protein expression level is presented in the bottom panel (p-value<0.05; n=3, ImageJ software). Two-tailed Student’s t-test, * p-value<0.05; ** p-value<0.01*** p-value<0.001.

31

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 7. Platelets and iMPs facilitate the interaction of tumor cells with the endothelium. A. Left – Representative images of the HUVEC-HT29 tumor cell interaction in dynamic conditions (4 dynes/cm2). Calcein-green-stained HT29 cells (green), HUVEC monolayer (nuclei, blue). Right – The graph depicts the mean number of HT29 cells interacting with HUVECs/field +/- SD (n=3, 10 field/condition). B. Representative images of the 3 integrin subunit (depicted in green), platelet vesicles (white arrow) and HT29 cells (F- actin in red). Confocal microscopy, 100X. C. Western blot analysis of E-cadherin and Snail-1 expression in total lysate proteins from HT29 cells that interacted with hWPs. D. mRNA expression was compared in platelets (PLTs), HT29 cells (HT29) and HT29 cells preincubated with platelets (iHT29PLTs) using the Applied Biosystems TaqMan Human

Inflammation or Tumor Metastasis Array Plates. Results were analyzed with the manufacturer’s online software and are represented using a color code: black: no expression;

Green: low expression and red : strong expression of the mRNA. E. ICAM-1 expression on

HUVECs following 3 hours of control McCoy’s 5A medium, iSN, iMPs, platelet control supernatant (cSN(Plts)) and HT29 control supernatant (cSN(HT29)) treatments. TNF- (10 ng/mL) was used as a positive control for endothelial cell activation and degranulation.

Endothelial cells were stained with Hoechst 33342 (nuclei, blue) and PE-conjugated ICAM-1 mouse IgG antibody (green). F. The graph depicts the mean percentage of HT29 cells interacting with the treated HUVEC monolayer in dynamic conditions (4 dynes/cm2) relative to the control medium +/- SD. (p-value<0.01; n=3). Two-tailed Student’s t-test, * p- value<0.05; ** p-value<0.01*** p-value<0.001. hWPs: human washed platelets; HUVECs: human umbilical vein endothelial cells; iSN: 6-hour hWP-HT29 interaction supernatant; iMPs: iSN-purified MPs.

32

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 1

A GPIb-a IB4

B C Tumor periphery Tumor center

GPIb-a GPIb-a GPIb-a IB4 IB4

HIF1-a HIF1-a

D E F

CD41

s t

) CD31

15 e l

2 *

** e

t 40

m

a

l

l

µ

p

a

(

n

d

g

a

i e

t 30

s

e

a r

10 t

s

a

n

a

e

v

g c

a 20

s

n

r

i

t

e

l

l

r

x

o

e

e

u

l

b

o f

t 10

a 5

l

e

g

h

n

t

i

a

-

f

d

o 1

n 0

o

F

I

%

p

s

H e

0 r -10 r

r y o r y e r c r t e te e n h n h e p e p c ri c ri r e r e o p o p m r m r u o u o T m T m u u T T

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2

A HT29 hWP Merged B

d

l

250 *

e s

10 mn mn 10

i

t

f

/

e

l

)

e

s t

t 200

a

n

l

e

p

f

m o

150

e

r

l

e

e

b

s e

m 100

v

u

i

t

i n

30 mn mn 30

s

n o

a 50

p

e

D

M

I D

( 0 n n n m m m 0 0 0 1 3 6 60 mn mn 60

D

C HT29 hWP Merged

d

l

40

e s

i ***

t

f

/

e

l

)

e

s

t

t

a n

l 30

e

p

f

m

o

e

r

l

e e

20

b

s e

HUVECs hWP Merged m

v

u

i

t

i

n

s

n 10

o

a

p

e

D

M

I D

( 0 9 s 2 C T E H V U H E G

150

h

)

t l

HT29 hWP i

o

w

r

t *

n

n

o

o

i

t

c

c

o a

t 100

r

e

e

t

v

i

n

t

i

a

l

n

i

e

r

s

(

t

e

s

l

l

l

e t

e 50

c

a

l

9

p

f

2

o

T

H % F HT29 Mo59K 0 l o G ) V l tr g M D ro n I µ t o 6 0 G n C h 0 R o d 1 + c 793 bp C ( Cdh-6 G G V g Ig D I G 6 h R d C

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 3 A

30 mn 60 mn 120 mn 180 mn

CD41 CD29 B C T+PMPs PMPs TMPs 100 AnV+ CD41+/Epcam+

AnV+ CD41+/EPCAM- AnV+ CD41-/Epcam +

)

s t

e 3000 250

l 600 *

ns L L e 80 **

t *

s

L

µ µ

t *

/

a

/

µ

l

e

/

s

s

l

p

s t

200

t

P

P

e

l

t

n

t

P

n

a

M

a

t

M

a

n

l

a

t

f

M

t f

o 60 2000

a

p

t

f

a

t

o

a

400 o

o

d

a n

o 150

n

t

n

r

r

n

a

n

n

o

r

e

e

o

n

i

e

i

t

o

r

e

t

p

p

i

o

t

a

p

p

s

a

u

u r

i 40

a

t s

r 100

r

s

u

s

t

r

f

t

n

f s

1000

a

n

f

o

n

e f

o 200

p

e

o

c

e

o

c

m

%

c

n n

o 50

n

o c

20 o

o

C

n

C

C i ( 0 0 0 N N N N N N N N N 0 iS S iS S iS S iS S S N N N N N N N N N N N N N N N N N N s s s s S S S S S S S S S n lt n lt n lt n lt C iS S iS S iS S iS S S i i i i n n n n n m p m p m p m p T n s n s n s n s C n ts n ts n ts n ts C l l l l l lt lt lt lt l l l l T m m m m m 0 tr 0 tr 0 tr 0 tr ro m p m p m p m p l T m p m p m p m p l 0 0 0 0 0 3 c 6 c 8 c 6 c t 0 rl 0 rl 0 rl 0 rl o 0 rl 0 rl 0 rl 0 rl o 3 6 9 2 8 1 3 n 3 t 6 t 8 t 6 t tr 3 t 6 t 8 t 6 t tr 1 1 n n n n o c c 1 c 3 c n c c 1 c 3 c n m m m m C n n n n o n n n n o 0 0 0 0 n 3 6 8 6 m m m m C m m m m C 1 3 m 0 0 0 0 n 0 0 0 0 n 0 3 6 8 6 3 6 8 6 6 1 3 m 1 3 m 3 0 0 6 6 3 3 D E CD41 Ep-cam CD41 AnV FGN

F G

CD41 CD29 FGN H Ep-cam CD41

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 4

A

A B

B

C D E F

PE EpCAM

CD41 APC C D

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 5 A B C

Cdh-6 Cdh-6

Actin ß-actin

D E

1.5×1007

**

0

)

8

/

U

4

A

F

( f

1.0×1007

o

e

c

y

t

n

i

e

s

c

n

s

e

t e

r 06 n

i 5.0×10

o

u

l

m

f

u S

0.0 r o ts m le u te T la p ith w r o m u T

1.0 iSN 0.8

) Permeabilized iSN

F G y t * i Permeabilized iMPs

s 0.6

800 n Platelets SN

e d

0.4

t HT29 SN

o

p

g

s

o

s 0.15

t

n

i

e

e

t

n

c

n

i

a

n

o k

r 600

e

s

o

r

g

i

t

l

i

r

e

y f

o ns

c

a

r

m

e

t

f

r

p

s

o

n

n

n

l

m

o

a

e

a

c

o

e

r

v

t

c

e

m

l 0.10

f

/

400m

n

y

o

d

i

t

u

i

i

e

s

e

s

z

d

i

n

l

g

e

e

t

e

a

a

y

d

t

m

m

t

c

r

n

o

o

o

e

p

n

c N

200 s

r

o

n

e a

m 0.05

P

e

m

o

i

t

a r

0 ( l l l o N s a o o tr S P - tr tr n i M F n n o i N o o 0.00 c T c c m N N 1 1 S L 1 1 1 a a l4 E 8 F -1 u S S L P E 0 L 1 F m 1 I 7 1 I I i C C T 4 C L D lR IL M A d s 9 C D X C S m I I P e lt 2 /M N C X , a / 2 A C 2 -g 1 M P T L R C 1 E H L N n C IF i C C rp X e C S

H

150 ***

s

150 *** 1

s

S

e

g

r

e

g

O

g

A

a

N

a

f

i

h

f

h

o

p

o

p

o

e 100

r

o

e g

r 100

c

g

c

a

a

t

a

a

t

n

m

n

m

e

e

g

c

g

r

c

n

r

i

n

e

i s

e 50 p

s 50

s

p

s

n

e

e

n

r

r

a

a

p

p

e

x

e

x

M

e

e M 0 0 6 s 6 ts 2 lt 2 l T P T P C + C + 6 6 2 2 T T C C

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 6 A B

Ctrl 30 60 180 CPT Ctrl 60 180 360 CPT

35 kDa Pro -Caspase 3 Pro-Caspase 3

17 kDa Cleaved Cleaved 12 kDa Caspase 3 caspase 3 45 kDa ß-actin ß-actin

C D

s

l l

e 25 c

* Ctrl 30 60 180 360

+

D A

) 20

s

A l

l P21

7

e

/

c +

15

c

V

i

t

n

o

t

A

p

f 10

o o

ß-actin

p

e

a

g

( a

t 5

n

e c

r 0 e

P l n n n ro T t m m m P Tumor cells + Monocytes n 0 0 0 C o 3 6 8 C 1 E F Ctrl 60 180 360

P21 0.20

Platelets

)

n

n

o

i i

t ns

s

c s

a ß-actin -

e 0.15

r

ß

/

p

1

x

2

e

P 1

0.10 2

o Tumor cells + Platelets + Monocytes

i

t

P

a

e

R

v (

i Ctrl 60 180 360

t

l 0.05

a

e

l

v

e

e l R P21 0.00 l 0 0 0 0 o 3 6 8 6 tr 1 3 n o ß-actin C Time (minutes)

G H Tumor cells + Platelets 5h + Monocytes 5h Ctrl P+M 5/5

Monocytes Platelets & monocytes

l

2.0 e PRb

2.0

) v

n * *

e

n

o

l

i

i

t 807/811

s

)

c

n

s

n

a

o

i

i

- t e 1.5

r 1.5

s

c

ß

/

s

p

a

1

- x

e ß-actin

r

2

ß

e

/

p

P

1

1

1.0 x

2 2

o 1.0

e

i

t

P

P

1

a

2

o

e

i

R

t

v

P )

( Platelets & Monocytes

i

n t

a 1.5

n l

0.5 i

e

o a

0.5 t

i

e

R

l

v

c

(

i

s

v

t

e a

s *

e

-

a

l

R

e

l

ß

r

e

/

p

0.0 x R

0.0 1

e 1 l 0 0 0 l

o 0 0 0 8 1.0 o 1 tr 6 8 6 tr 6 8 6 / 1 3 1 3 1 n n 7

o 8

o 0

C C /

8

7

0

b

8

Time (minutes) Time (minutes) R

b

P

R 0.5

o

i

P

t

a

e

v

R

i

(

t

l

a

l

e

e

v

e R l 0.0 l o s tr te n cy o o C n o M + ts le te la P

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 7 A B HT29

s 80 ***

l

l s

e 360 mn

C 60 mn

c

E

9 V

2 60

U

T

H

H

f

h

t

o

i

r

w 40

e

g

b

n

i

m

t u

HT29 + Platelets c

n a

20

r

n

e

t

a

n

e

i

M 0 9 s 2 t T le H te la p + 9 2 T H C D

Ctrl - 30 60 180 PLTs HT29 iHT29PLTs PTGS2 E-cdh SerpinB5 NF2 Snail1 CTNNA1 FN1 MMP10 ß-actin MMP2

E F

cMed TNF-a iSN

**

g )

n 300

l

i

t

o

c

r

t

a

r

n

e

o

t

c

n

i

o

t

s

l l

e 200

e

v

i

c

t

a

9

l

2

e

r

T

(

H

s

f

C

o

E 100

e V

iMPs cSN(Plts) cSN(HT29) g

U

a

t

H

n

e

h

t

c

i

r w

e 0 P ) ) d N s s e S P lt 9 M i 2 c iM (P T N (H S N c S c

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 14, 2019; DOI: 10.1158/0008-5472.CAN-19-1181 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

The interaction of platelets with colorectal cancer cells inhibits tumor growth but promotes metastasis

Léa Plantureux, Diane Mège, Lydie Crescence, et al.

Cancer Res Published OnlineFirst November 14, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-19-1181

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2019/11/14/0008-5472.CAN-19-1181.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet Manuscript been edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2019/11/14/0008-5472.CAN-19-1181. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research.