• No results found

Coagulation, angiogenesis and cancer - Chapter 1: General Introduction

N/A
N/A
Protected

Academic year: 2021

Share "Coagulation, angiogenesis and cancer - Chapter 1: General Introduction"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Coagulation, angiogenesis and cancer

Niers, T.M.H.

Publication date

2008

Link to publication

Citation for published version (APA):

Niers, T. M. H. (2008). Coagulation, angiogenesis and cancer.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)
(3)
(4)
(5)

11 General introduction

Coagulation, angiogenesis and cancer

The relationship between cancer and thrombosis is twofold. First, cancer patients have an

increased risk to develop venous thromboembolism (VTE). Second, the coagulation system

affects cancer progression and metastasis. Furthermore, various coagulation factors, like

tissue factor (TF) and thrombin are involved in angiogenesis, new vessel formation, which

facilitates tumor growth and metastasis

1,2

. This thesis is focused on three major cancer

progression-stimulating factors: platelets, TF/thrombin and angiogenesis -and the

two-way interactions between cancer and coagulation.

Increased risk of thrombosis in cancer patients

Cancer patients have an increased risk to develop VTE. This was first described by

Bouillaud (1823)

3,4

and forty years later (1865) interpreted and published by Trousseau

5

. An

unprovoked VTE can be the first sign of occult cancer, which was described for the first time

in 1935

6

. According to Virchow, the hallmarks of VTE are pathological changes in blood

flow, coagulability and the condition of the vessel wall. All three phenomena may occur

in cancer patients. The proposed mechanisms to explain hypercoagulation associated

with cancer include a reaction of the patients body to the tumor such as abnormal protein

synthesis, angiogenesis and necrosis and more specific processes related to

tumor-mediated haemostatic activities (cancer cells interacting with platelets, endothelial cells,

monocytes and with the coagulation and fibrinolytic systems)

7,8

. Furthermore, cancer

treatment -irradiation, chemotherapy and surgery- may further upset the balance between

procoagulant and anticoagulant factors

9

.

The incidence of VTE is also associated with the use of central venous catheters (CVC).

Cancer patients frequently have to use CVCs for chemotherapy, stem cell infusion, blood

supply, medication, parenteral hyperalimentation and blood sampling. Risk factors for

CVC-related thrombosis include the type of malignancy, chemotherapy and CVC and insertion

sites of the catheter tip

10

. Many studies have addressed the incidence and associated risk

factors of CVC-related infections and VTE in patients with solid tumors but only few data

are available on haemato-oncological patients. These patients may differ from patients

with solid tumors, because of the more severe and prolonged thrombocytopenia and

leukopenia. Therefore, an important matter of debate is whether haematological cancer

patients should receive thrombosis prophylaxis or not. This issue is dealt with in chapter 9.

The coagulation system affects cancer progression and metastasis

Until the mid nineties of the last century, the initial treatment of VTE consisted of a brief

course of unfractionated heparin (UFH) followed by a course of vitamin K antagonists

for several months. In 1992, Prandoni et al compared the relative safety and efficacy of

(6)

12 Chapter 1

low molecular weight heparin (LMWH) and UFH for the treatment of VTE and concluded

that fixed-dose subcutaneous LMWH is at least as effective and safe as UFH for the initial

treatment of VTE

11

. This has been confirmed by others

12-14

. Unexpectedly, LWMH showed

a favourable effect on the survival of cancer patients. At 3 months, 44% (8 of 18) of the

cancer patients died in the UFH group vs. 7% (1 of 15) in the LMWH group (p=0.021)

11

.

These findings were confirmed in meta-analyses of 9 studies that compared LMWH with

UFH in the treatment of VTE

15,16

. These studies initiated clinical trials evaluating the effect

of anticoagulants on survival of cancer patients without thrombosis

17-20

. Thus, cancer

favours thrombosis and the coagulation system promotes cancer as suggested by the

marked survival advantage of patients using anticoagulants.

Various experimental studies showed the inhibitory effects of anticoagulants on

cancer progression and metastasis. The results are reviewed in chapter 2. However, the

mechanisms by which anticoagulants may interfere with tumor growth and metastasis

are diverse, remain poorly defined and seem to be dependent on the type of cancer and

individual anticoagulant

21-27

. In this thesis, experimental studies are described that focus

in particular on the phase of haematogenous dissemination when cancer cells are present

in the circulation. These studies are described in chapters 4, 5, 6 and 7. Chapter 3 reviews

validation of noninvasive bioluminescence imaging (BLI) for quantitative assessment of

tumor load in time in small animals, a technique we used in two of our studies.

Platelets

Platelet aggregation on cancer cells takes place rapidly when cancer cells have entered

the circulation. Cancer cells are thus masked for the immune system, protected against

shear stress in the vasculature and adhesion to vessel walls is facilitated. The process starts

with the interaction of activated platelets with cancer cells that express P-selectin ligands,

such as glycoprotein ligand-1, CD24, heparan sulphate proteoglycan (HSPG) and

sialyl-Lewis a/x

28,29

. The interactions of platelets and cancer cells may also involve ß3-containing

integrins binding von Willibrand factor (vWF), thrombomodulin and fibrinogen to form

molecular bridges

30,31

. Then, activation of procoagulant proteins such as TF can occur as is

described below

32,33

. These interactions enable rolling of cancer cells or cancer cell-platelet

complexes along vessel walls where endothelial cells constitutively express low amounts

of P-selectin

34

that facilitates adhesion of cancer cells to the wall and transmigration into

the subendothelium

35

or protect cancer cells within vessels against mechanical stress and

the immune system

36-40

.

P-selectin also occurs in a soluble form in blood plasma. Ferroni et al demonstrated that

soluble P-selectin (sP-selectin) plays a pivotal role in the pathogenesis of metastasis by

formation of cancer cell-platelet complexes. sP-selectin is considered to be a marker for

platelet activation and sP-selectin correlated inversely with prognosis in patients with

cancer

41

. It has been suggested that anticoagulants inhibit metastasis by blocking

(7)

13 General introduction

can be explained by its interference in P-selectin-mediated interactions between platelets

and cancer cells. This is described in chapter 8.

Thrombin

When TF is expressed on the plasma membrane of cancer cells, it activates circulating

liver-derived coagulation factors VII, V and X that leads to the generation of thrombin

from prothrombin (Figure 1). Thrombin has distinct effects on cells. Intracellular effects

of thrombin are mediated by protease activated receptors (PARs), members of the family

of G-coupled receptors

43

. Four PARs have been described: PAR-1, -2, -3 and -4. Thrombin

seems to be the major physiological activator of PAR-1

44,45

and PAR-4

46

, but it can also

activate PAR-3

47

, that functions as a cofactor of PAR-4. PAR-2 is not directly activated by

thrombin but via trypsin

48

, coagulation factor VIIa and factor X

49,50

. TF and PARs play an

important role in cancer progression

51,52

.

PAR signalling upregulates adhesion molecules on endothelial surfaces and triggers

production of chemokines by activating neutrophils and monocytes. This leads to binding,

rolling and attachment of platelets and leukocytes on the surface of endothelium. High

Figure 1: Coagulation cascade. HWMK=High molecular weight kinogen; PK=Prekallikrein; TFPI=Tissue factor pa-thway inhibitor. Grey arrows, functions of thrombin. Mediators studied or discussed in this thesis (TF, Factor Xa, thrombin, Factor V, Factor VIII, Fibrin and activated protein C).

(8)

14 Chapter 1

concentrations of locally-produced thrombin may lead to direct release of P-selectin

stored in Weibel bodies in endothelial cells through PAR-4-dependent signalling, resulting

in increased platelet aggregation and cancer cell-platelet binding. Local aggregates of

cells expressing TF

53

along with procoagulant activity of platelets

54

may trigger further

thrombin formation

1

and increased permeability of the endothelium

2

. Thrombin also

activates platelets to release growth factors that may sustain tumor development and may

aid angiogenesis by production of platelet derived growth factor (PDGF), basic fibroblast

growth factor (bFGF) and vascular endothelial growth factor (VEGF)

9,57-59

. Furthermore,

thrombin converts fibrinogen into fibrin, the end product of the coagulation cascade.

Fibrin depositions have been found in and around many types of tumors, providing a

scaffold for angiogenesis and possibly also protecting the cancer cells against the host

defence system

60,61

. Thus, possible mechanisms by which anticoagulants prolong survival

of cancer patients may also be reduction of thrombin and fibrin formation. The relationship

between thrombin and cancer is described in the chapters 5 and 7.

Angiogenesis

Several coagulation factors, such as TF and thrombin play a role in angiogenesis, that is

required for tumor growth and metastasis

1,2

. First, thrombin can activate angiogenesis

by reduction of endothelial cell attachment to lamina basalis proteins and activation of

matrix metalloproteinases

62

. Second, thrombin has chemotactic and apoptotic effects on

endothelial cells and upregulates expression of VEGF receptors (VEGFR). Third, thrombin

upregulates expression of αvβ3 integrin, the marker of the angiogenic phenotype of

endothelial cells

63

. Platelets may contribute to this process because they also release

angiogenic factors like VEGF upon activation by thrombin via PAR-1

64

.

VEGF is one of the most important angiogenic factors. It binds to specific tyrosine kinase

receptors on the surface of endothelial cells including VEGFR-1 (Flt-1) and VEGFR-2 (KDR/

flk-1) on vascular endothelium and VEGFR-3 (Flt-4) expressed on lymphatic endothelium,

resulting in cell migration, proliferation and survival

65,66

. Clinical research on angiogenesis

has two major directions in cancer patients. First, quantification of angiogenesis for

diagnosis, prognosis as well as for the monitoring of responses. Second, the inhibition of

angiogenesis to halt tumor growth

2

. However, serum, plasma and whole blood have been

indiscriminately used to determine VEGF levels in the body. Because coagulation results

in the release of VEGF from platelets, serum VEGF levels include plasma-derived VEGF and

platelet-derived VEGF

67

. Therefore, VEGF levels in serum do not reflect the true circulating

levels of VEGF. In citrate or EDTA plasma, where less platelet activation and subsequent

VEGF release occurs than in serum, VEGF levels were found to be higher in cancer patients

than in controls and this was interpreted as a reflection of the higher levels of VEGF in the

circulation of cancer patients

68

. However, the release of VEGF from platelets may contribute

to increased VEGF levels in plasma as well under these conditions. Therefore, the effects

of the different blood collection protocols on the measurement of circulating VEGF and

(9)

15 General introduction

their impact on VEGF release from platelets by in vitro platelet activation is described in

chapter 10.

VEGF expression is regulated by a number of factors. In renal cell carcinoma (RCC),

VEGF expression is a consequence of inactivation of the von Hippel-Lindau (VHL) tumor

suppressor gene, resulting in a remarkable overexpression of VEGF. Because of the

high levels of VEGF occurring in RCC, VEGF may be identified as a critical component of

angiogenesis in RCC and as a potential therapeutic target to treat RCC. The strategy to

inhibit the activity of VEGF includes binding of the VEGF protein and blockade of VEGFR.

Besides the classical prognostic markers for advanced RCC

69

, novel validated biomarkers

are needed to predict the outcome of targeted therapy and the development of drug

resistance. Circulating levels of VEGF, placenta growth factor (PlGF), sVEGFR-1 and -2 and

bFGF are potential candidates to predict outcome of the various therapies. We correlated

base line levels and changes in the levels during treatment of these potential markers

with disease outcome and the development of resistance during therapy in chapter 11.

(10)

16 Chapter 1

References

1. Folkman J. Tumor angiogenesis: therapeutic implications. N.Engl.J.Med. 1971; 285: 1182-1186. 2. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat.Med. 1995; 1: 27-31. 3. Bouillaud S. De L’Obliteration des veines et de son influence sur la formation des hydropisies

par-teilles: consideration sur la hydropisies passive er general. Arch.Gen.Med. 2007; 1: 188-204.

4. Otten HM. Thrombosis and Cancer, Thesis. 2002. Academic Medical Center, University of Amsterdam. 5. Trousseau A. Phlegmasia alba dolens. Clinique medicinale de l’Hotel-Dieu de Paris 1865; Paris:

Bail-liere J.-B. et fils: 645-712.

6. Illtyd James T, Matheson N. Thromboplebitis in cancer. Practitioner 1935; 134: 683-684.

7. Donati MB. Cancer and thrombosis: from Phlegmasia alba dolens to transgenic mice. Thromb.Hae-most. 1995; 74: 278-281.

8. Mousa SA. Anticoagulants in thrombosis and cancer: the missing link. Semin Thromb.Hemost. 2002; 28: 45-52.

9. De Cicco M. The prothrombotic state in cancer: pathogenic mechanisms. Crit.Rev.Oncol.Hematol. 2004; 50: 187-196.

10. Boersma RS, Jie KS, Verbon A, van Pampus EC, Schouten HC. Thrombotic and infectious complica-tions of central venous catheters in patients with hematological malignancies. Ann.Oncol. 2008; 19: 433-442.

11. Prandoni P, Lensing AW, Buller HR, Carta M, Cogo A, Vigo M, Casara D, Ruol A, ten Cate JW. Compari-son of subcutaneous low-molecular-weight heparin with intravenous standard heparin in proximal deep-vein thrombosis. Lancet 1992; 339: 441-445.

12. Low-molecular-weight heparin in the treatment of patients with venous thromboembolism. The Columbus Investigators. N.Engl.J.Med. 1997; 337: 657-662.

13. Koopman MM, Prandoni P, Piovella F, Ockelford PA, Brandjes DP, van der MJ, Gallus AS, Simonneau G, Chesterman CH, Prins MH. Treatment of venous thrombosis with intravenous unfractionated heparin administered in the hospital as compared with subcutaneous low-molecular-weight heparin admin-istered at home. The Tasman Study Group. N.Engl.J.Med. 1996; 334: 682-687.

14. Levine M, Gent M, Hirsh J, Leclerc J, Anderson D, Weitz J, Ginsberg J, Turpie AG, Demers C, Kovacs M. A comparison of low-molecular-weight heparin administered primarily at home with unfractionated heparin administered in the hospital for proximal deep-vein thrombosis. N.Engl.J.Med. 1996; 334: 677-681.

15. Hettiarachchi RJ, Smorenburg SM, Ginsberg J, Levine M, Prins MH, Buller HR. Do heparins do more than just treat thrombosis? The influence of heparins on cancer spread. Thromb.Haemost. 1999; 82: 947-952.

16. Siragusa S, Cosmi B, Piovella F, Hirsh J, Ginsberg JS. Low-molecular-weight heparins and unfraction-ated heparin in the treatment of patients with acute venous thromboembolism: results of a meta-analysis. Am.J.Med. 1996; 100: 269-277.

(11)

17 General introduction 17. Altinbas M, Coskun HS, Er O, Ozkan M, Eser B, Unal A, Cetin M, Soyuer S. A randomized clinical trial

of combination chemotherapy with and without low-molecular-weight heparin in small cell lung cancer. J.Thromb.Haemost. 2004; 2: 1266-1271.

18. Kakkar AK, Levine MN, Kadziola Z, Lemoine NR, Low V, Patel HK, Rustin G, Thomas M, Quigley M, Williamson RC. Low molecular weight heparin, therapy with dalteparin, and survival in advanced cancer: the fragmin advanced malignancy outcome study (FAMOUS). J.Clin.Oncol. 2004; 22: 1944-1948.

19. Klerk CP, Smorenburg SM, Otten HM, Lensing AW, Prins MH, Piovella F, Prandoni P, Bos MM, Richel DJ, van Tienhoven G, Buller HR. The effect of low molecular weight heparin on survival in patients with advanced malignancy. J.Clin.Oncol. 2005; 23: 2130-2135.

20. Lee AY, Levine MN, Baker RI, Bowden C, Kakkar AK, Prins M, Rickles FR, Julian JA, Haley S, Kovacs MJ, Gent M. Low-molecular-weight heparin versus a coumarin for the prevention of recurrent venous thromboembolism in patients with cancer. N.Engl.J.Med. 2003; 349: 146-153.

21. Drago JR, Weed P, Fralisch A. The evaluation of heparin in control of metastasis of Nb rat androgen-insensitive prostate carcinoma. Anticancer Res. 1984; 4: 171-172.

22. Drago JR, Lombard JS. Metastasis in the androgen-insensitive Nb rat prostatic carcinoma system. J.Surg.Oncol. 1985; 28: 252-256.

23. Lee AE, Rogers LA, Jeffery RE, Longcroft JM. Comparison of metastatic cell lines derived from a mu-rine mammary tumor, and reduction of metastasis by heparin. Clin.Exp.Metast. 1988; 6: 463-471. 24. Lee AE, Rogers LA, Longcroft JM, Jeffery RE. Reduction of metastasis in a murine mammary tumor

model by heparin and polyinosinic-polycytidylic acid. Clin.Exp.Metast. 1990; 8: 165-171. 25. Milas L, Hunter N, Basic I. Treatment with cortisone plus heparin or hexuronyl hexoaminoglycan

sulfates of murine tumors and their lung deposits. Clin.Exp.Metast. 1985; 3: 247-255.

26. Niers TM, Klerk CP, Dinisio M, van Noorden CJ, Buller HR, Reitsma PH, Richel DJ. Mechanisms of hepa-rin induced anti-cancer activity in experimental cancer models. Crit.Rev.Oncol.Hematol. 2007; 61: 195-207.

27. Smorenburg SM, van Noorden CJ. The complex effects of heparins on cancer progression and metas-tasis in experimental studies. Pharmacol.Rev. 2001; 53: 93-105.

28. Felding-Habermann B. Tumor cell-platelet interaction in metastatic disease. Haemostasis 2001; 31 Suppl 1: 55-58.

29. McCarty OJ, Mousa SA, Bray PF, Konstantopoulos K. Immobilized platelets support human colon carcinoma cell tethering, rolling, and firm adhesion under dynamic flow conditions. Blood 2000; 96: 1789-1797.

30. Chopra H, Timar J, Rong X, Grossi IM, Hatfield JS, Fligiel SE, Finch CA, Taylor JD, Honn KV. Is there a role for the tumor cell integrin alpha IIb beta 3 and cytoskeleton in tumor cell-platelet interaction? Clin.Exp.Metast. 1992; 10: 125-137.

31. Steinert BW, Tang DG, Grossi IM, Umbarger LA, Honn KV. Studies on the role of platelet eicosanoid metabolism and integrin alpha IIb beta 3 in tumor-cell-induced platelet aggregation. Int.J.Cancer 1993; 54: 92-101.

(12)

18 Chapter 1

32. Lip GY, Chin BS, Blann AD. Cancer and the prothrombotic state. Lancet Oncol. 2002; 3: 27-34. 33. Nash GF, Walsh DC, Kakkar AK. The role of the coagulation system in tumor angiogenesis. Lancet

Oncol. 2001; 2: 608-613.

34. Stone JP, Wagner DD. P-selectin mediates adhesion of platelets to neuroblastoma and small cell lung cancer. J.Clin.Invest. 1993; 92: 804-813.

35. Kim YJ, Borsig L, Varki NM, Varki A. P-selectin deficiency attenuates tumor growth and metastasis. Proc.Natl.Acad.Sci.U.S.A. 1998; 95: 9325-9330.

36. Mook OR, Van Marle J, Vreeling-Sindelarova H, Jonges R, Frederiks WM, van Noorden CJ. Visualization of early events in tumor formation of eGFP-transfected rat colon cancer cells in liver. Hepatology 2003; 38: 295-304.

37. Borsig L, Wong R, Feramisco J, Nadeau DR, Varki NM, Varki A. Heparin and cancer revisited: mechanis-tic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proc.Natl. Acad.Sci.U.S.A. 2001; 98: 3352-3357.

38. Borsig L, Wong R, Hynes RO, Varki NM, Varki A. Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metasta-sis. Proc.Natl.Acad.Sci.U.S.A. 2002; 99: 2193-2198.

39. Borsig L. Selectins facilitate carcinoma metastasis and heparin can prevent them. News Physiol.Sci. 2004; 19: 16-21.

40. Borsig L. Antimetastatic activities of modified heparins: selectin inhibition by heparin attenuates metastasis. Semin.Thromb.Hemost. 2007; 33: 540-546.

41. Ferroni P, Roselli M, Martini F, D’Alessandro R, Mariotti S, Basili S, Spila A, Aloe S, Palmirotta R, Maggini A, Del Monte G, Mancini R, Graziano F, Cosimelli M, Guadagni F. Prognostic value of soluble P-selectin levels in colorectal cancer. Int.J.Cancer 2004; 111: 404-408.

42. Borsig L, Wong R, Feramisco J, Nadeau DR, Varki NM, Varki A. Heparin and cancer revisited: mechanis-tic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proc.Natl. Acad.Sci.U.S.A. 2001; 98: 3352-3357.

43. Coughlin SR. How the protease thrombin talks to cells. Proc.Natl.Acad.Sci.U.S.A. 1999; 96: 11023-11027.

44. Rasmussen UB, Vouret-Craviari V, Jallat S, Schlesinger Y, Pages G, Pavirani A, Lecocq JP, Pouyssegur J, Obberghen-Schilling E. cDNA cloning and expression of a hamster alpha-thrombin receptor coupled to Ca2+ mobilization. FEBS Lett. 1991; 288: 123-128.

45. Vu TK, Hung DT, Wheaton VI, Coughlin SR. Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation. Cell 1991; 64: 1057-1068.

46. Kahn ML, Zheng YW, Huang W, Bigornia V, Zeng D, Moff S, Farese RV, Jr., Tam C, Coughlin SR. A dual thrombin receptor system for platelet activation. Nature 1998; 394: 690-694.

47. Ishihara H, Connolly AJ, Zeng D, Kahn ML, Zheng YW, Timmons C, Tram T, Coughlin SR. Protease-acti-vated receptor 3 is a second thrombin receptor in humans. Nature 1997; 386: 502-506.

(13)

19 General introduction 48. Nystedt S, Emilsson K, Wahlestedt C, Sundelin J. Molecular cloning of a potential proteinase

acti-vated receptor. Proc.Natl.Acad.Sci.U.S.A. 1994; 91: 9208-9212.

49. Camerer E, Huang W, Coughlin SR. Tissue factor- and factor X-dependent activation of protease-acti-vated receptor 2 by factor VIIa. Proc.Natl.Acad.Sci.U.S.A. 2000; 97: 5255-5260.

50. Riewald M, Ruf W. Mechanistic coupling of protease signaling and initiation of coagulation by tissue factor. Proc.Natl.Acad.Sci.U.S.A. 2001; 98: 7742-7747.

51. Camerer E. Protease signaling in tumor progression. Thromb.Res 2007; 120 Suppl 2: S75-S81. 52. Ruf W. Tissue factor and PAR signaling in tumor progression. Thromb.Res 2007; 120 Suppl 2: S7-S12. 53. Osterud B. Tissue factor expression by monocytes: regulation and pathophysiological roles. Blood

Coagul. Fibrinolysis 1998; 9 Suppl 1: S9-14.

54. Sims PJ, Wiedmer T, Esmon CT, Weiss HJ, Shattil SJ. Assembly of the platelet prothrombinase complex is linked to vesiculation of the platelet plasma membrane. Studies in Scott syndrome: an isolated defect in platelet procoagulant activity. J.Biol.Chem. 1989; 264: 17049-17057.

55. Palabrica T, Lobb R, Furie BC, Aronovitz M, Benjamin C, Hsu YM, Sajer SA, Furie B. Leukocyte accumu-lation promoting fibrin deposition is mediated in vivo by P-selectin on adherent platelets. Nature 1992; 359: 848-851.

56. Vogel SM, Gao X, Mehta D, Ye RD, John TA, Andrade-Gordon P, Tiruppathi C, Malik AB. Abrogation of thrombin-induced increase in pulmonary microvascular permeability in PAR-1 knockout mice. Physiol.Genomics 2000; 4: 137-145.

57. Nash GF, Turner LF, Scully MF, Kakkar AK. Platelets and cancer. Lancet Oncol. 2002; 3: 425-430. 58. Wojtukiewicz MZ, Tang DG, Nelson KK, Walz DA, Diglio CA, Honn KV. Thrombin enhances tumor cell

adhesive and metastatic properties via increased alpha IIb beta 3 expression on the cell surface. Thromb.Res. 1992; 68: 233-245.

59. Wojtukiewicz MZ, Tang DG, Ciarelli JJ, Nelson KK, Walz DA, Diglio CA, Mammen EF, Honn KV. Throm-bin increases the metastatic potential of tumor cells. Int.J.Cancer 1993; 54: 793-806.

60. Dvorak HF, Senger DR, Dvorak AM. Fibrin as a component of the tumor stroma: origins and biological significance. Cancer Metast.Rev. 1983; 2: 41-73.

61. Klerk CPW. Cancer, Coagulation and Antithrombotics. 2005. Thesis, Academic Medical Center, Univer-sity of Amsterdam.

62. Mook OR, Frederiks WM, van Noorden CJ. The role of gelatinases in colorectal cancer progression and metastasis. Biochim.Biophys.Acta 2004; 1705: 69-89.

63. Tsopanoglou NE, Maragoudakis ME. Role of thrombin in angiogenesis and tumor progression. Semin.Thromb.Hemost. 2004; 30: 63-69.

64. Ma L, Perini R, McKnight W, Dicay M, Klein A, Hollenberg MD, Wallace JL. Proteinase-activated recep-tors 1 and 4 counter-regulate endostatin and VEGF release from human platelets. Proc.Natl.Acad.Sci. U.S.A. 2005; 102: 216-220.

(14)

20 Chapter 1

65. Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tu-mor angiogenesis and a potential target for diagnosis and therapy. J.Clin.Oncol. 2002; 20: 4368-4380. 66. Witmer AN, Vrensen GF, van Noorden CJ, Schlingemann RO. Vascular endothelial growth factors and

angiogenesis in eye disease. Prog.Retin.Eye Res. 2003; 22: 1-29.

67. Verheul HM, Hoekman K, Luykx-de Bakker S, Eekman CA, Folman CC, Broxterman HJ, Pinedo HM. Platelet: transporter of vascular endothelial growth factor. Clin.Cancer Res. 1997; 3: 2187-2190. 68. Hyodo I, Doi T, Endo H, Hosokawa Y, Nishikawa Y, Tanimizu M, Jinno K, Kotani Y. Clinical significance

of plasma vascular endothelial growth factor in gastrointestinal cancer. Eur.J.Cancer 1998; 34: 2041-2045.

69. Motzer RJ, Bacik J, Schwartz LH, Reuter V, Russo P, Marion S, Mazumdar M. Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma. J.Clin.Oncol. 2004; 22: 454-463.

(15)

Referenties

GERELATEERDE DOCUMENTEN

order to obtain a reasonable worth for each coalition in these games, the Nash bargaining model is employed and we show that there exists at most one rational Nash bargaining

Figure 4: Regression coefficients estimated through the explanatory model built by using the whole HMP spatio-temporal information across China. The covariates shown in this figure

It was mentioned before the tendency of scholars studying policy transfer on proposing terms that fit their respective case studies. With the purpose of understanding some

Dit houdt onder meer in: naast een vakopleiding leerlingen in het initieel on­ derwijs ook algemene kennis en vaardigheden (sleutelkwalificaties) bijbrengen,

Het tijdschrift stelt zijn kolommen open voor wetenschappelijke artike­ len die gekenmerkt worden door een zekere actuali­ teitswaarde e n /o f

De 'public employment service' in noord­ west Europa.. W esterlaken

Public expenditure on active and passive la­ bour market policy shows considerable variety in volume and composition across the north­ western member-states of the

Titles in the ILLC Dissertation Series:. ILLC DS-2001-01: