• No results found

Coagulation, angiogenesis and cancer - Chapter 2: Mechanism of heparin induced anti-cancer activity in experimental cancer models

N/A
N/A
Protected

Academic year: 2021

Share "Coagulation, angiogenesis and cancer - Chapter 2: Mechanism of heparin induced anti-cancer activity in experimental cancer models"

Copied!
29
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)

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)

2

(3)

induced anticancer activity in

experimental cancer models

Tatjana M.H. Niers, Clara P.W. Klerk, Marcello DiNisio, Cornelis J.F. Van Noorden, Harry R. Büller, Pieter H. Reitsma, Dick J. Richel

(4)

Abstract

Background

Retrospective analyses of clinical trials and prospective clinical studies have suggested that heparins may have an effect on cancer survival. This putative anticancer activity of heparins is supported by data from studies in animal tumor models.

Objective

To clarify the various potential mechanisms of heparin anticancer activity we evaluated the data from pre-clinical studies in which heparins have been tested as anticancer therapy.

Methods

Pre-clinical studies, published between 1960 and 2005 were assessed. Data was collected on the type and dose of heparin used, duration of exposure to heparin, interval between heparin administration and cancer cell inoculation, and the animal tumor model used. In addition, a distinction was made in the analysis between heparin effects on the primary tumor or on established metastases and effects on the metastatic potential of infused cells.

Results

Heparins seemed to affect the formation of metastasis rather than the growth of primary tumors. Chemically modified heparins with no or limited anticoagulant activity also showed anti-metastatic properties. Possible mechanisms to explain the effects on the process of metastases include inhibition of blood coagulation, inhibition of cancer cell-platelet and -endothelial interactions by selectin inhibition and inhibition of cell invasion and angiogenesis.

Conclusions

The anticancer activity of heparins depends more on inhibition of metastasis formation than on the effects on primary tumor growth. These effects are probably related to both coagulation and non-coagulation dependent factors. For a definitive proof of the anticancer activity of heparins in the clinic, prospective randomized trials especially in patients with early metastatic disease or in the adjuvant setting are urgently needed.

(5)

Introduction

Although the association of thrombosis and cancer is a well-established phenomenon, the role of activation of the coagulation cascade in cancer prognosis is not well defined. A critical role for activated coagulation factors in cancer biology has been suggested for years, and is supported by clinical studies with heparins and in preclinical studies in vivo and in vitro tumor models.

In a subgroup analysis of a study on venous thromboembolism (VTE) therapy, Prandoni et al observed a significant reduction in the three months mortality rate in the subgroup of cancer patients who received low molecular weight heparin (LMWH), nadroparin1. Forty-four percent (8 of 18) of the cancer patients who were treated with

unfractionated heparin (UFH) died during the study period as compared with only 7% (1 of 15) of those patients in the LMWH group (p=0.021).

The LMWH effect on survival was confirmed in a subsequent systematic review of nine studies on VTE-treatment2, which showed a 49% reduction of the 3 months mortality

(hazard ratio, HR 0.61:95%Cl, 0.40-0.93) with LMWH relative to UFH.

The reduction in mortality was observed for various types of cancer, it was not the result of differences in fatal VTE or bleeding events and was unaltered after adjustment for other prognostic variables.

Prospective randomized trials have evaluated the survival effects of UFH or LMWH in patients with cancer. Tempelhoff et al3 studied the survival effect of prophylactic-dose

LMWH versus UFH given for seven days postoperatively. In a group of 324 patients undergoing surgery for breast or pelvic cancer, the mortality rate in LMWH treated patients was 5.7% compared to 15.6% in UFH recipients (p=0.005).

In a trial of Lee et al4 676 cancer patients with VTE were randomized to receive 6 months

of dalteparin or Vitamin K antagonists (VKA). No difference in the one year mortality was detected. However, a post-hoc subgroup analysis5 suggested that in patients

without metastatic disease the 1 year mortality was significantly reduced by dalteparin group (20%) relative to the VKA group (35%) (HR 0.50; 95% CI 0.27-0.95) (p=0.03). This difference was apparently not attributed to a difference in fatal thrombosis.

Two recent studies (FAMOUS and MALT) have evaluated the effects of LMWH on survival in cancer patients without VTE 6,7. In the FAMOUS study 7, 385 cancer patients

without thrombosis were randomized between low-dose dalteparin (5000 IU/day) or placebo for 1 year. No significant difference in survival between the two study groups was detected. In the subgroup of patients with a relatively good prognosis at entrance in the study, dalteparin was associated with a better median survival (24 months versus 43 months; p=0.03). In the MALT study6, 302 cancer patients without thrombosis were

randomized to 6 weeks of nadroparin (2 weeks full therapeutic followed by 4 weeks half this dose) or placebo. The median survival was 6.5 months in the placebo group and 8.0 months in the nadroparin group (HR for mortality 0.75; 95% CI; 0.59-0.96). In

(6)

the a priory specified subgroup of patients with a life expectancy of at least 6 months, the median survival increased from 9.4 to 15.4 months (HR for mortality 0.64; 95% CI; 0.45-0.90).

Two trials studied the effect of UFH8 or LMWH9 in patients with small cell lung cancer.

In both these studies a statistically significant survival advantage was observed in patients randomized to chemotherapy plus UFH or LMWH relative to patients who received chemotherapy alone.

Although several clinical studies strongly support an anticancer activity of heparins, and especially LMWH, many questions remain unresolved. The precise mechanism(s) by which LMWHs exerts their anticancer activity even after a short-term LMWH administration2,3,6 for example, remains unknown. It is also unclear, whether the

observed anticancer effect of LMWHs is solely accounted for by their anticoagulant activity or if an influence on other biological processes is also involved. In this study we systematically reviewed the available evidence on the relationship between heparins and cancer from experimental studies in animal models.

Heparins

Heparins and low molecular weight heparins

Heparin is a highly sulphated member of the glycosaminoglycan (GAG) family. These molecules are long and unbranched disaccharide repeats and are located primarily in the cell membrane and in the extracellular matrix (ECM). Other members of the family of GAGs with known physiological significance are hyaluronic acid, dermatan sulphate, chondroitin sulphate and keratan sulphate. Heparin is abundant in granules of mast cells that line blood vessels and are present in mucosal tissue. The release of heparin from these granules in response to injury and its subsequent entry into the bloodstream leads to inhibition of blood clotting.

UFH, used in the clinic, are naturally-occurring glycosaminogycans from porcine or bovine origin with molecular weights in the range of 12.000-14.000 Daltons. LMWHs are derived from these UFH by chemical or enzymatic depolymerization, yielding fragments of approximately one third of the size of heparin (MW approximately 5000). The different types of LMWHs are prepared by different methods of degradation, and although they differ from each other to some extent in pharmacokinetic properties and anticoagulant profile, they are recognized to be quite comparable in clinical activity10.

The antithrombotic activity of heparin is mediated by interaction with antithrombin (AT). Heparin molecules bind AT via a unique pentasaccharide motif that induces a conformational change in AT that enhances its antithrombotic activity manifold11,12.

The heparin-AT complex binds activated factor X via direct binding of AT thereby inactivating factor Xa. This anti Xa activity is equivalent between UFH and LMWHs. The difference between UFH and LMWH is in their relative inhibitory activity against

(7)

factor Xa and factor ))a13. In contrast, the inactivation of factor ))a is mediated by

the formation of a ternary complex consisting of heparin, AT, and factor ))a14. An

18-saccharide sequence in the heparin molecule is required to form such a trimolecular complex. The majority of LMWH chains are too short to form this complex. Thus LMWH have relatively little anti-))a activity compared to UFH15.

Besides binding to antithrombin, UFH and LMWH can bind to a wide range of other proteins and molecules via electrostatic interactions with the polyanionic groups of the glycosaminoglycan chains16. These interactions are mediated by physicochemical

properties of heparin polymers such as sequence composition, sulfation pattern, charge distribution, overall charge density and molecular size. As a consequence, UFH and LMWH have a wide variety of biological activities other than their anticoagulant effects.

Modified heparins

Several chemically modified heparins with a decreased anticoagulant activity have been synthesized to minimise the anticoagulant activity and to enhance the inhibitory effects of heparin on the tumor growth and metastasis. Periodate-oxidised (IO4- heparin)

and periodate-oxidised, alkaline-degraded LMWH (IO4- LMWH)17, for instance, do not

have a specific pentasaccharide structure to interact with antithrombin III and show a much lower anti-coagulant activity than UFH18. Other heparin derivatives have been

produced which reduced the anticoagulant activity, like; N-acetylated, N-desulfated, O-desulfated or carboxyl-reduced heparin19,20. Also treatment of heparin with sodium

borohybride markedly reduced the anticoagulant activity of heparin21. Other modified

heparins such as N-succinylated heparins (Succ 100 H) and N-succinylated LMWH (Succ 100-LMW-H) and very low molecular weight heparin (VLMWH) have been synthesised and showed to have also a low anticoagulant effect22. Same as the anticoagulant

activity of a series of semisynthetic sulfaminoheparosan sulfates (SAHSs) with different degrees and distributions of 2-O or 3-O sulfation at glucosamine residues obtained by chemical modification of the E coli K4 polysaccharide23.

Effects of heparins on experimental primary tumor growth and

metastasis

The effects of heparins on cancer have been studied in several animal models. Important variation in study design and type of tumor makes interpretation and comparison difficult. Variations include the type of heparin, heparin dose and duration of exposure, interval between heparin administration and cancer cell inoculation and the animal model used. In this review we discriminated between effects on the primary tumor or established metastases and effects on the metastatic potential of infused cells.

(8)

Effect of heparins on the outgrowth of primary tumors or established metastases

Seventeen studies have evaluated the effect of heparins on subcutaneous implanted or chemically induced tumors in animals (Table 1). In only 4 of these studies heparin treatment induced a significant reduction of the primary tumor volume. The effect of heparins was not affected by the different methods of tumor induction in the various animal models. In a similar way, the interval between the start of heparin treatment and tumor induction was not a determinant factor for tumor growth inhibition. Other potential factors influencing the effect of heparins were the dose, type of heparins used and the duration of treatment. High doses of heparin (100 U 3 times for 15 days)24,25 seemed to be effective, although

results have not been always concordant26. Low doses of heparins24,25 or less than 15 days

of administration24 did not affect tumor growth. Moreover, a very high dose of heparin

(200 U) given only three times in a week was not apparently capable to affect the primary tumor growth27. Therefore it appears that long-term heparin administration at high

concentration levels is necessary for an effect on the primary tumor growth.

In 2 studies a tumor growth inhibition was observed with chemically modified heparins (2,3-O-desulphated heparin, IO4-LMWH and NAC-HCPS) with low anticoagulant activity28,29. That this is not the case for all modified heparins was demonstrated in the

study of Sciumbata were Succ 100-LMWH and VLMH had no effect on tumor growth22.

In summary, the available data suggests that only high dosages of heparin and some chemically modified heparins may inhibit the primary tumor growth in various tumor models. In this regard the importance of a high dose of heparin is supported by studies in intra-peritoneal tumor models (Table 2). Heparin administration into the peritoneal cavity reduced the peritoneal tumor deposits in 6 out of 7 studies. This would suggest that high heparin concentrations around the tumor cells may be necessary. Because modified heparins with limited or no anticoagulant activity demonstrate anti-tumor activity suggests that the anti-tumor is not only dependent on the inhibition of coagulation.

Effects of heparins on the development of metastases

Metastases, rather than primary tumors, are responsible for most cancer deaths. Therefore it is worth speculating that heparins derive part or all of its effect on prognosis of cancer patients by interfering with haematogenous metastasis formation. This issue has been studied using both primary and secondary metastasis models. In the primary models, tumor cells are directly injected into the bloodstream and the resulting metastasis are recorded. In the secondary models, tumor cells are placed either subcutaneously or intraperitoneally followed by evaluation of the number and/or size of metastases developing in the lung. Table 3 summarizes data from 17 studies (1985-2005) investigating the effects of heparin on primary metastases (visceral metastases following intravenous cancer cell administration) and secondary metastases (visceral metastases following subcutaneous cancer cell inoculation). In most of these studies, 14 out of these17 studies, heparin induced a significant reduction in the number of metastases.

(9)

Table 1. Eff

ec

t of heparins on the out

g

ro

wth of primar

y tumors and established metastases

Ref e renc e C anc er t ype Animal C anc er dev elopment Sacrific ed (da y s) In te rv al Doses schedule Results Drugs [30] Sar coma T-241 C57BL6JN mic e L eg muscle 18 −2 h 1 dd sc No eff ec t Heparin 50–200 U Sar coma DBA49 DBA mic e B

earing tumor in leg

for sev eral y ears 21 −2 h 1 dd sc No eff ec t Heparin 50–200 U [31] Sar coma MC G1-SS CBA mic e s.c . 1 0 +48 h 6 da y s 3 dd s .c . No eff ec t

Heparin not fur

ther specified [32] M elanoma B16 C57BL/6J mic e s.c . 5–16 +48 h 6 da y s 3 dd s .c . No eff ec t

Heparin not fur

ther specified Sar coma MC G1-AS CBA mic e s.c . 5–14 +48 h 6 da y s 3 dd s .c . No eff ec t

Heparin not fur

ther specified [24] Sar coma MSLS Holtzman rats s. c. 8–15 +24 h 15 da y s 3 dd i.p . Tumor g ro wth reduc ed b y 44% Heparin 100 U [27] PA -III c ells 1 × 105 c ells R a ts Injec ted s .c . 42 +72 h 3 da y s/w eek i.v . No eff ec t Heparin 200 U [26] W alker 256 car cino sar coma Sprague –Da wley rat Injec ted s .c . 50 −48 h; 0; +48 h 7 da y s i.v .; 14 da y s i.v .; c o ntinuous No eff ec t Heparin 600 U [33] N b -P r-A I-II R a ts s. c. 21 T herap y tumor 90 mm3 1 dd i.v . No eff ec t Heparin 15 U; Heparin 150 U [34] N b -P r-A I-II R a ts s. c. 30 T herap y tumor 90 mm3 1 dd i.p . No eff ec t Heparin 15 U N b -P r-A I-III R a ts s. c. 30 0 4 da y s/3 w eeks i.p . No eff ec t Heparin 150 U [35] Sar coma NFSA 6 × 105 cells C3 H f/k am mic e Injec ted s .c . 23 +7 da y s 17 da y s 2 dd s .c . or orally No eff ec t

Heparin 15.7 U; Heparin not fur

ther specified M ammar y car cinoma MCA-K 6 × 105 c ells C3 H f/k am mic e Injec ted s .c . 24 +10 da y s 14 da y s 2 dd s .c . or orally No eff ec t

Heparin 15.7 U; Heparin not fur

ther specified [36] M ammar y car cinoma 3 × 104 c ells BR 6/Icr f mic e Injec ted s .c . 28 −24 h, 1 h 6 times/w eek i.p . No eff ec t Heparin 40 U [25] S quamous car cinoma Ddy mic e Induc ed 63 T herap y tumor 5 mm3 2 dd i.p . Tumor g ro wth reduc ed Heparin 12.5 U

(10)

Table 1. Eff

ec

t of heparins on the out

g

ro

wth of primar

y tumors and established metastases

Ref e renc e C anc er type Animal C anc er dev elopment Sacrific ed (da y s) In te rv al Doses schedule Results Drugs [37] C

olon adeno car

cinoma SW480 3 × 106 c ells Balb -c mic e Injec ted s .c . 84 −24 h 2 dd s .c . No eff ec t Heparin 3 U [22] B16-BL melanoma c ells 2 × 105 c ells C57BL/6 mic e Injec ted s .c . 20 −10 min 2 times/w eek s .c . No eff ec t

Chemically modified heparin

[28] C a P an-2 3 × 106 c ells Balb/c mic e Injec ted s .c . 35 +24 h 1 dd s .c . Tumor g ro wth reduc ed

Chemically modified heparin

[38]

C

olon adeno car

cinoma C C351s 1 × 105 c ells W A G-R ij rats Injec ted i.v . por tal v ein 24 −1 h 1 dd i.p . No eff ec t LMWH; not fur ther specified [29] L

ewis lung canc

er , 3LL 1 × 107 c ells C57BL/6 mic e Injec ted s .c . 20 Tumor 100–200 mm3 (Da y 14) Da y s 14–20 1 dd s .c . Tumor g ro wth reduc ed

Chemically modified heparin

B16 melanoma 1 × 107 cells C57BL/6 mic e Injec ted s .c . 20 Tumor 100–200 mm3 (Da y 14) Da y s 14–20 1 dd s .c . Tumor g ro wth reduc ed

Chemically modified heparin

[39]

L

ewis lung canc

er , 1 × 106 c ells C57/BL6 Injec ted s .c . 22 +6 da y s Da y s 6–22 ev er y 3 da y s s .c . No eff ec t Heparin 5 U; LMWH 5.7 U anti Xa In te rv al r e fers t o the int e rv al bet w een canc er c

ell administration and star

t of heparin tr eatment; MC G1-SS, S yngeneic 20MC (met h ylcholanthr ene) induc ed rhabdom y osar coma; MC G1-AS, rhabdom y osar

coma, a solid ascit

es f o rm of MC G1-SS; MSLS, Murph y -Sturm L ymphosar coma; P A -III c ells , R at pr ostat e adenocar cinoma cells; 3LL, L

ewis lung canc

er c

ells; C

a

P

an-2, human pancr

eatic adenocar cinoma; N b -P r-A.I-II, N b rat pr ostatic adenocar cinoma; N FSA, a fibr osar coma; MCA-K , mammar y car cinoma.

(11)

Table 2. Eff ec t on intraperit oneal tumor g ro wth af ter tr

eatment with heparins in a lapar

osc opic model Ref e renc e C anc er t ype Animal Sacrific ed (da y s) In te rv al Doses schedule Results Drugs [40] MB T-2 c ells 1 × 105 c ells C3H mic e 1 6 0 Single i.p . R educ ed tumor w eight Heparin 10 U [41] DHD/K12/TR b 1 × 104 c ells BD IX rats 28 0 Single i.p . R educ ed tumor w eight Heparin 20 U [42] DHD/K12/TR b 1 × 104 c ells DB IX rats 28 0 Single i.p . R educ ed tumor w eight Heparin 20 U [43] M ammar y adenocar cinoma 2 × 107 c ells Dark A goutti rats 7 +4 min Single i.p . R educ ed tumor w eighta Heparin 200 U [44] DHD/K12?TR b 1 × 106 c ells BD IX rats 28 0 Single i.p . No eff ec t Heparin 5 U [45] C olon car cinoma C C531 5 × 106 c ells/ml W A G rats 21 0 Single i.p . 1 dd s .c .; single i.p ./1 dd s .c . Reduc ed tumor w eight LMWH; not fur ther specified [46] C C 531 C olon car cinoma 5 × 106 c ells/ml W A G rats 21 0 Single i.p . 1 dd s .c .; single i.p ./1 dd s .c . Reduc ed tumor w eight LMWH; not fur ther specified MB

T-2, murine bladder tumor c

ells; DHD/K12/TR b (ECA C C ), c olonic adenocar

cinoma of the rat.

a A sig

nificant decr

ease in tumor w

eight was obser

v ed in a g roup of rats that r e ceiv ed heparin t

ogether with the g

roup that r

e

ce

iv

ed 2 ml fr

esh blood plus 200 U heparin

compar

ed with c

o

ntr

(12)

Table 3. Eff

ec

ts of heparins on experimentally induc

ed lung or liv er metastases af ter intra v enous injec tion of canc er c ells Ref e renc e C anc er t ype Animal Sit e of tastases Sacrific e (da y s) In te rv al Doses schedule Results Drugs [34] N b -P r-AI-II s .c . R a ts L ung 30 T herap y star t tumor 30 mm3 30 da y s 1 dd i.p . R educ ed spontaneous metastasis Heparin 17.5 U N b -P r-AI-III s .c . R a ts L ung 30 0 4 times/w eek during 3 w eeks i.p . Reduc ed spontaneous metastasis Heparin 175 U [35] NFSA 2 × 105 c ells i.v . C3 H f/K am mic e L ung 13 +4 da y s 9 da y s 2 dd s .c . Reduc ed metastasis Heparin 15.6 U MCA-K 3 × 105 c ells i.v . C3 H f/K am mic e L ung 27 +7 da y s 14 da y s 2 dd s .c . Reduc ed metastasis Heparin 15.6 U [19] B16-BL6 melanoma c ells 5 × 104 c ells i.v . C57BL/6 mic e L ung 20 0 C anc er c ells incubat ed with heparins f or 2 h Reduc ed metastasis

Heparin not fur

ther specified;

chemically modified heparins

[50] Sar coma L -1 5 × 104 cells i.v . Balb/c mic e L ung 14 −1 h 3 da y s 2 dd s .c . M oderat ely r educ ed metastasis Heparin 21 U Sar coma L -1 1 × 106 cells i.v . Balb/c mic e L ung 14 −1 h 3 da y s 2 dd s .c . M oderat ely r educ ed metastasis Heparin 21 U [51] BR 6 mammar y 3 × 104 cells i.v . BR 6/Icr f mic e L ung 18 −24 h and −1 h Single i.p . R educ ed metastasis Heparin 40 U BR 6 mammar y 3 × 104 cells s .c . BR 6/Icr f mic e L ung 26 −24 h and −1 h 4 da y s 3 dd i.p . R educ ed spontaneous metastasis Heparin 40 U [36] M ammar y car cinoma 3 × 104 c ells i.v . BR 6/Icr f mic e L ung 28 −24 h and −1 h Single i.p . R educ ed metastasis Heparin 40 U M ammar y car cinoma 3 × 104 c ells s .c . BR 6/Icr f mic e L ung 21–28 0 6 times/w eek i.p . R educ ed spontaneous metastasis Heparin 40 U [48]

DHD/K12/PRO 2 × 107 cells i.v

. BDIX rats Liv er 90 0 7 da y s i.v . C o ntinuous Reduc ed metastasis NS Heparin 100 U [22] B16-BL6 melanoma 1 × 105 c ells i.v . C57BL/6 mic e L ung 21 −10 min Single s .c . Reduc ed metastasis

Chemically modified heparin

[52] B16-BL6 melanoma 1 × 105 c ells i.v . C57/BL6 mic e L ung 15 −20 min Single i.p . R educ ed metastases LMWH 13 U 13762 M A T M ammar y 2 × 105 c ells i.v . F isher 344 rats L ung 15 −20 min Single i.p . R educ ed metastasis LMWH 468 U

(13)

Table 3. Eff

ec

ts of heparins on experimentally induc

ed lung or liv er metastases af ter intra v enous injec tion of canc er c ells Ref e renc e C anc er t ype Animal Sit e of tastases Sacrific e (da y s) In te rv al Doses schedule Results Drugs [38] C C531 C olon car cinoma 1 × 105 c ells i.v . W A G-R ij rats Liv er 8 − 1 h LMWH 8 da y s 1 dd i.p .; UFH 8 da y s 2 dd i.p . No eff ec t LMWH/heparin 2.0 mg/kg C C531 C olon car cinoma 1 × 105 c ells i.v . W A G-R ij rats Liv er 24 −1 h 24 da y s 1 dd i.p . No eff ec t LMWH/heparin 2.5 mg/kg [53] LS180 C olon car cinoma 3–4 × 105 c ells i.v . Litt ermat e RA G2 mic e L ung 42 −30 min Single i.v . R educ ed metastasis Heparin 100 U [29] L ewis lung 3 × 105 c ells C57BL/6 mic e L ung 14 ? 7 da y s 1 dd i.v . R educ ed metastasis

Chemically modified heparin

B16 melanoma 3 × 105 cells i.v

. C57BL/6 mic e L ung 14 ? 7 da y s 1 dd i.v . R educ ed metastasis

Chemically modified heparin

[54] B16-BL6 melanoma 1 × 105 c ells i.v . C57/BL6 mic e L ung 14 0 Single i.v . R educ ed metastasis

Chemically modified heparin

B16-BL6 melanoma 1 × 105 c ells i.v . C57/BL6 mic e L ung 14 −1 h Single i.v . R educ ed metastasis

Chemically modified heparin

[47]

B16 melanoma 2 × 105 cells i.v

. C57/BL6 mic e L ung 15 −4 h 14 da y s 1 dd s .c . Reduc ed metastasis LMWH 10 mg/kg

B16 melanoma 2 × 105 cells i.v

. C57/BL6 mic e L ung 15 −4 h Single s .c . Reduc ed metastasis LMWH 10 mg/kg [21] H11 c ells 1 × 106 c ells i.v .C57/BL6 mic e L ung 14 −10 min Single i.v . R educ ed metastasis 0.5–1.0–2.0 mg/mouse H11 c ells 1 × 106 c ells i.v .C57/BL6 mic e L ung 14 −1 h −10 min 0 +1 h Single i.v . R educ ed metastasis No eff ec t −1/+1 h

Chemically modified heparin

H11 c ells 1 × 106 c ells i.v .C57/BL6 mic e L ung 14 −10 min Single i.v .; single i.p .; single s .c . Reduc ed metastasis

Chemically modified heparin

H11 c ells 1 × 106 c ells i.v .C57/BL6 mic e L ung 14 −10 min Single i.v . R educ ed metastasis

Chemically modified heparin

3LL; C olon26; B16F0; FBJ; 1 × 106 c ells i.v . C57/BL6 mic e L ung 14 −10 min Single i.v . R educ ed metastasis

Chemically modified heparin

[55] MC38GFP 1 × 105 c ells C57/BL 6J mic e L ung 27 −30 min Single s .c . Reduc ed metastasis

UFH 6.56 U/19.68 U Diff

er

ent LMWH 7.32 IU/21.96

(14)

Table 3. Eff

ec

ts of heparins on experimentally induc

ed lung or liv er metastases af ter intra v enous injec tion of canc er c ells Ref e renc e C anc er t ype Animal Sit e of tastases Sacrific e (da y s) In te rv al Doses schedule Results Drugs B16F1 1 × 105 c ells C57/BL 6J mic e L ung 17 −30 min Single s .c . Reduc ed metastasis

UFH 6.56 U/19.68 U Diff

er ent LMWH 7.32 IU/21.96 IU [56] HT168-M1 5 × 106 c ells

BALB/c SCID mic

e L ung 49 –? h 3 da y s 1 dd i.p . R educ ed metastasis UFH 0.5–5 IU LMWH 0.5–5 IU NFSA, fibr osar coma; MCA-K , mammar y car cinoma; N b -P

r-A I-II, rat pr

ostat e adenocar cinoma andr ogen insensitiv e tumor t ype II; N b -P

r-A I-III, rat pr

ostat e adenocar cinoma andr ogen insensitiv e tumor t ype III; H11 c

ell line established fr

om L ewis lung 3LL -deriv ed c ell line; LL, L ewis L ung car cinoma c ell line; C olon26, c olon car cinoma c ell line; B16F0, M elanoma c ell line; FBJ , Ost eosar coma c ell line; MC38GFP , mouse c olon car cinoma c

ell line stable tr

ansf

ec

ted with enhanc

ed g reen fluor esc ent pr ot

ein; HT168-M1, human melanoma c

ell line

(15)

The timing of heparin administration in relation to the intravenous inoculation of cancer cells is likely to be relevant, because intravascular exposure of cancer cells to heparins or heparin effects may affect the capacity of survival and endothelial adhesion of cancer cells, events necessary for successful metastases. Antimetastatic activity of heparins was showed in 10 studies in which heparin was administrated just before intravenously tumor cell inoculation. In 1 study, showing a inhibitory effect of heparin35, heparin administration

started at 4 or 7 days after cancer cell inoculation. In the study of Amirkhosravi et al the interval was 4 hours. In this study the LMWH, tinzaparin, resulted in an APTT of 200 seconds, 4 hours after administration, reflecting the presence of a significant anticoagulant effect at the time of intravenous cancer cell inoculation47.

Three studies investigated the anti-tumor effects of heparins with reduced anticoagulant activity. These chemically-modified heparins, NAH-HCPS29,LAC heparin21 and ITF 116422

were also able to inhibit tumor metastases, suggesting that the anti-metastatic effect was not dependent on anticoagulation. These chemically modified heparins had a pleiotropic action including anti-adhesion, anti ECM degradation, anti-platelet cancer complex formation and anti-angiogenesis activity, indicating the multipotency of these compounds in inhibition of almost all steps involved in the later phases of haematogenous metastasis development.

In 2 of the 3 studies which found no antimetastatic capacity of heparin38,48 the cancer

cells were directly injected into the portal vein to induce liver metastases. The inoculation model may not be the ideal experimental set-up to investigate the anti-metastatic effects of heparins. The formation of metastases in the liver after administration of cancer cells may have involved mechanical entrapment of the cancer cells in the sinusoids due to their large size and their limited plasticity as was demonstrated by Mook et al using the same animal model and the same cancer cells. Moreover Mook et al demonstrated that these interactions between cancer cells and endothelial cells did not occur because endothelial cells retracted rapidly and the interactions occurred between cancer cells and hepatocyts49. Therefore, in the model of Smorenburg et al and Nagawa et al, the molecular

interactions that are sensitive to heparin may not have occurred.

In the third negative study50 development of metastases was decreased in the heparin

group, but this effect was not significant. Also in this study the heparin dose was rather low.

Mechanisms of heparin induced anticancer activity

The reviewed studies in experimental models indicate that heparins are able to inhibit cancer metastasis more than tumor growth. Haematogenous metastasis is a complex and highly regulated process in which cancer cells detach from the primary tumor, migrate across blood vessel walls into the bloodstream, disperse throughout the body, adhere to endothelial cells and penetrate into surrounding tissues. During the

(16)

intravascular phase, cancer cells are extremely vulnerable to a variety of host defense mechanisms and only a few of the cancer cells that enter the bloodstream result in manifest metastases leading to fatal outcome.

However, most of the animal studies on experimental metastasis involve the intravenous injection of tumor cell in the tail vein, resulting in pulmonary tumor foci. Over 90% of the tumor cells become entrapped in the vasculature of the lungs immediately after tail vein injection57.This entrapment occurs regardless of anticoagulation or

fibrinogen deficiency and is immediately followed by tumor cell induced coagulation and intravascular fibrin deposition. Tissue factor (TF), often highly expressed on tumor cells, plays an important role in the local activation of the coagulation cascade and the formation of the tumor-fibrin clot. The rapid decline of platelet counts following intravascular tumor cell inoculation is probably the result of platelet sequestration in the tumor-fibrin clot58.Tumor cell induced coagulation and platelet activation

may prolong the survival of tumor cells in the lung vessels. In these fibrin platelet complexes tumor cells may growth or die intravascular, or adhere to the endothelial wall followed by invasion into the lung parenchyma. The last process is also facilitated by the local generation of thrombin, thrombin induced platelet activation, and the local release from platelets of angiogenesis (vascular endothelial growth factor, VEGF) and proliferation (platelet factor 4, PF-4) inducing factors. Thrombin induced platelet activation results in activation of the platelet integrin receptor GP IIb/IIIa59, allowing

binding of platelets to fibrinogen, and induction of P-selectin from A granules, a process that is dependent on protease-activated receptor-4 (PAR-4) signaling. Increased levels of P-selectin on platelets and endothelial cells, contributes to the adherence of platelets and endothelial cells to aberrantly expressed carbohydrates on cancer cells60 (see next chapter). These data demonstrate that platelet activation

and fibrin formation are both important mechanisms by which tumor cells promote haematogenous metastasis. Several experiments in animal models have demonstrated that a low platelet count or inhibition of platelet activation result in a decrease of the metastatic potential of tumor cells. In PAR-4 deficient mice and in Nf-E2 mice, mice lacking circulating platelets, haematogenous metastasis are significantly reduced61. In

addition, antagonists of GPIIb/IIIa, Abciximab and XV45, are strong inhibitors of tumor cell platelet activation and the development of metastasis62-64. The most challenging

problem of the use of antiplatelet drugs in cancer is the lack of selectivity. Indeed, the currently available anti platelet drugs affect both haemostasis and cancer-induced platelet activation. The next part of this paper describes the potential mechanisms of heparins as anticancer drugs. Potential effects of heparins on the process of metastasis are; inhibitory effects of heparins on blood coagulation, inhibition of cancer cell-platelet and endothelium interactions during intravascular dissemination, inhibition of heparanase, and competition with growth factor binding to heparan sulphate proteoglycans (HSPGs).

(17)

Coagulation as the target

The tumor associated activation of the coagulation cascade has been emended in both the formation of tumor stroma and the promotion of metastasis65. Most solid tumors

contain considerable amounts of fibrinogen-derived products suggesting that fibrin is important in tumor stroma formation65,66. Although inhibition of fibrin formation

by heparins is an attractive hypothesis for their anticancer effects, the evidence that heparins directly affect tumor growth is limited. In addition to the role of coagulation-induced fibrin in tumor stroma formation, coagulation factors may be involved in the process of metastasis as well. Studies in fibrinogen-deficient mice demonstrated that fibrin(ogen) plays an important role in determining the metastatic potential of cancer cells. Fibrin(ogen) appears to facilitate metastasis by enhancing the sustained survival of individual cell emboli in the vasculature67. In the fibrinogen deficient animal model the

development of lung metastasis, upon intravenous cancer cell inoculation, was strongly diminished compared to normal mice. In the same model the specific thrombin inhibitor hirudin further diminished the metastatic potential of circulating cancer cells, suggesting that thrombin facilitates metastasis partly in a fibrinogen-independent way. Heparins are able to interfere with these coagulation dependent mechanisms of metastases. Heparins have been shown to shorten the retention time of tumor cells in the lungs and reduce the development of lung metastasis in this way. It also inhibits thrombin and - fibrin formation and by virtue of preventing thrombin generation and activity, can thus inhibit platelet activation.

An important aspect in coagulation activation is the expression of tissue factor (TF) on cancer cells.

The expression of TF is inhibited in a fXa-dependent manner by tissue factor pathway inhibitor (TFPI)68. TFPI is mainly synthesized in and localized to vascular endothelial cells69

and is able to inhibit metastatic tumor growth in mice also suggesting a role for thrombin in the metastatic process. Because heparins can also cause the release of tissue factor pathway inhibitor (TFPI), which reduces the pro-coagulant activity of cancer cells70, it is

tempting to speculate that TFPI is one of the targets involved in the anticancer activity of heparins.

Because several studies demonstrated that also non-anticoagulant heparins have the potential to inhibit metastasis19,21,22,29,71 other, non anti-coagulant, effects of heparins may

be important as well.

For example, non-anticoagulants can potentially cause the release of TFPI from the vascular endothelium and TFPI may contribute to their anti metastatic effect partially independent of the role in the coagulation cascade but as an inhibitor of angiogenesis72.

Selectins as the target

Ample evidence from pre-clinical studies supports the concept that intravascular survival and arrest of cancer cells on the endothelial surface is facilitated by interactions between

(18)

cancer cells and platelets73-75. The formation of cancer cell-platelet complexes provides

a shield that protects them from immune competent cells and favours cancer cell adhesion to vascular endothelium53. This interaction between cancer cells and platelets

and endothelial cells is facilitated by binding of glycoproteins in the plasma membrane of cancer cells to selectins on platelets and endothelium. Altered cell-surface glycosylation of glycans (mucins), which are high-molecular-weight molecules containing a protein core substituted with a large number of O-linked carbohydrates, is a prominent feature of cancer progression. Relatively few types of glycan alterations are associated with epithelial cancer. Sialyl Lewisx and Lewisa (sLex and sLea) epitopes are two of them. These

sLex/a epitopes on carcinoma mucins are frequently associated with an advanced and

further metastatic progression.

Selectins are adhesion receptors that recognize these altered carbohydrate structures. Their physiological function in mediating cell adhesion has been shown during inflammation, immune responses and wound repair76.77. Selectins are primary responsible for the first

steps in cell adhesion and in the absence of selectins, all subsequent steps mediated by integrins and other adhesion molecules are either substantially delayed or do not occur. Selectins are expressed on leukocytes (L-selectin), platelets (P-selectin) and the vascular endothelium (E- and P-selectin) .Whereas L-selectin is constitutivelyexpressed on neutrophils, monocytes, and naive lymphocytes, P-selectin is stored in secretory granules of resting plateletsand endothelium, is rapidly translocated to the cell surface upon activation by thrombin and histamine. E-selectin is newly synthesized in endothelial cells via transcriptional activation initiated by various proinflammatoryagonists such as IL-1, TNF-alpha and endotoxins78. All three selectins can bind sialylated, fucosylated, or

in some cases, sulfated glycans on glycoproteins, glycolipids, and proteoglycans. The tetrasaccharides sLex and sLea have been identified as the minimal ligands for all three

types of selectins.

Heparin and heparin-like oligosaccharides can inhibit L-selectin and P-selectin binding to SLex-related compounds79,80. Inhibition of L-selectin and P-selectin binding results in

inhibition of the inflammatory response81 and may attenuate tumor metastasis82.

Heparanase as target

A critical event in the process of cancer invasion and metastasis is the degradation of various components of the ECM, including collagen, laminin, fibronectin and HSPGs. Cancer cells are able to accomplish this task through the concerted actions of enzymes such as matrix metalloproteases, serine proteases, cysteine proteases and endoglycosidases83,84. Among

the endoglycosidases, heparanase, secreted by cancer cells, can destroy various ECM components favouring tumor invasion.

Heparanase expression is rare in normal tissue, but is evident in many human tumors where it significantly increases both the angiogenic and metastatic potential of cancer cells85. Elevated heparanase expression in humans has been correlated with advanced

(19)

disease and metastasis of tumors of the breast86, colon87, ovary88, bladder89, pancreas90,

acute myeloid leukemia91, non-small cell lung cancer92 and myelomas93.

Heparanase is important for the cleavage of heparan sulphate (HS) groups from HSPGs. These HS groups are growth factor-bearing structures linked to the protein core of HSPGs and localized in the ECM and in plasma membranes of cells. These structures are involved in storage of growth factors, such as bFGF and VEGF94,95 and function as their co-factors

in activation of plasma membrane receptors and downstream signaling94-96. Heparanase

does not completely digest HS chains but it cleaves the glycosidic bonds of HS chains at only a few sites, producing fragments which seems even more powerful than the native HS in potentiating the activity of bound growth factors91,97-99.

Several reports showed that heparin and some chemically modified species of heparin as well, inhibit tumor cell heparanase activity19-22,29,83,100-102 and that the inhibition of

heparanase activity correlates with a lower metastatic potential19,20,52,83,100.

Heparan sulphate proteoglycans as the target

Soluble heparins compete with HS groups on HSPGs for the binding of growth factors and other proteins and may cause release of these proteins from the ECM103. Both UFH

and LMWH have been shown to inhibit bFGF-induced angiogenesis in a human in vitro angiogenesis model104 by disrupting the bFGF-HS interactions. In man, therapeutic doses

of UFH can indeed cause an increase in plasma levels of growth factors, such as bFGF105,106.

In contrast, Soker et al showed that LMWH but not UFH can inhibit the binding of growth factors to their high affinity receptors as a result of its smaller size. In vitro, heparin fragments of less than 18 saccharide residues reduce the activity of VEGF107 and fragments

of less than 10 saccharide residues inhibits the activity of bFGF89,90,108.

Small heparin fractions have also been shown to inhibit VEGF and bFGF-mediated angiogenesis in vivo, in contrast to UFH109. The relevance of HSPGs and growth factors as

target for the heparin is very complicated and remains uncertain.

Concluding remarks

The process of tumor metastasis is a highly complicated, involving coagulation dependent and independent mechanism. Tumor cell induced platelet aggregation and activation plays an important role in the tumor cell protection and successful adherence to and invasion through the endothelial wall. Although some anti-platelet drugs exhibit potent anti-metastatic activity, this review focuses especially on the activities of heparins. Many experimental studies, reviewed here, support the hypothesis that heparin mainly affects the metastatic cascade rather than the primary tumor growth. In most experimental metastasis models tumor cells are injected intravenously in the tail vein resulting in pulmonary metastasis. During this process intravascular entrapment of tumor cells in the first capillary network (lungs) is followed by activation of coagulation via TF on the tumor

(20)

cells, platelet aggregation and activation. In this process thrombin plays an essential role, partially coagulation independent. Heparins are able to interfere at several sites in this sequence of events.

Because chemically modified heparins with no or limited anticoagulant activity also showed anti-metastatic properties in animal models, coagulation independent mechanisms may contribute to the anticancer activity of heparins as well21, 22, 54, 110. Heparin induced inhibition

of selectin-mediated cell-cell interactions, heparanase and angiogenesis inhibition and stimulation of TFPI release could at least partly explain the non anticoagulant heparin anti-metastatic activity.

How to explain the activity of agents that mainly affect metastasis when clinical anti cancer effects of heparins have been demonstrated in patients who already have a metastatic disease? Increasing evidence emerges that the ongoing process of metastasis in patients with cancer contributes to cancer progression and further dissemination. Several studies demonstrated that circulating cancer cells are detectable in the circulation of most patients with cancer111 and that the number of circulating cancer cells is correlated with

prognosis112. These data suggest that in patients with metastatic disease interruption of

the metastatic cascade with heparins may affect cancer outcome. This may also explain the phenomenon that heparins exert their anticancer activity mainly in patients with limited metastatic disease, whereas no effect is seen in patients with end stage disease6,7.

In contrast to cytotoxic chemotherapy, where the target is the cancer cell itself, heparins mainly affect targets outside the cancer cell. The process of metastasis is probably not cancer-type specific and more attributable to the contribution of common extra cellular mechanisms. This explains that the favorable impact of heparins on cancer outcome is a general phenomenon and probably not restricted to specific cancer types.

It can be concluded that heparins have an anticancer effect, but still many issues remain to be unanswered: what type of heparin, what dose and what duration of administration are optimizing anticancer effects. However the main issue is a definite proof that heparins contribute to a better cancer outcome. This answer is urgently needed and necessitates the design of prospective randomized trials especially in patients with early metastatic disease or in the adjuvant setting.

(21)

References

1. Prandoni P, Lensing AW, Buller HR, Carta M, Cogo A, Vigo M et al. Comparison of subcutaneous low-molecular-weight heparin with intravenous standard heparin in proximal deep-vein thrombosis. Lancet 1992; 339: 441-445.

2. 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.

3. von Tempelhoff GF, Heilmann L. Antithrombotic therapy in gynecologic surgery and gynecologic oncology. Hematol.Oncol.Clin.North Am. 2000; 14: 1151-1169.

4. Lee AY, Levine MN, Baker RI, Bowden C, Kakkar AK, Prins M et al. 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.

5. Lee AY, Rickles FR, Julian JA, Gent M, Baker RI, Bowden C et al. Randomized comparison of low mo-lecular weight heparin and coumarin derivatives on the survival of patients with cancer and venous thromboembolism. J.Clin.Oncol. 2005; 23: 2123-2129.

6. Klerk CP, Smorenburg SM, Otten HM, Lensing AW, Prins MH, Piovella F et al. The Effect of Low Mo-lecular Weight Heparin on Survival in Patients With Advanced Malignancy. J.Clin.Oncol. 2005; 23: 2130-2135.

7. Kakkar AK, Levine MN, Kadziola Z, Lemoine NR, Low V, Patel HK et al. Low molecular weight heparin, therapy with dalteparin, and survival in advanced cancer: the fragmin advanced malignancy out-come study (FAMOUS). J.Clin.Oncol. 2004; 22: 1944-1948.

8. Lebeau B, Chastang C, Brechot JM, Capron F, Dautzenberg B, Delaisements C et al. Subcutaneous heparin treatment increases survival in small cell lung cancer. “Petites Cellules” Group. Cancer 1994; 74: 38-45.

9. Altinbas M, Coskun HS, Er O, Ozkan M, Eser B, Unal A et al. 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.

10. Fareed J, Hoppensteadt D, Schultz C, Ma Q, Kujawski MF, Neville B et al. Biochemical and pharmaco-logic heterogeneity in low molecular weight heparins. Impact on the therapeutic profile. Curr.Pharm. Des. 2004; 10: 983-999.

11. Bauer KA, Rosenberg RD. Role of antithrombin III as a regulator of in vivo coagulation. Semin.Hema-tol. 1991; 28: 10-18.

12. Harenberg J. Pharmacology of low molecular weight heparins. Semin.Thromb.Hemost. 1990; 16 Suppl: 12-18.

13. Bauer KA, Rosenberg RD. Activation markers of coagulation. Baillieres Clin.Haematol. 1994; 7: 523-540.

14. Danielsson A, Raub E, Lindahl U, Bjork I. Role of ternary complexes, in which heparin binds both an-tithrombin and proteinase, in the acceleration of the reactions between anan-tithrombin and thrombin or factor Xa. J.Biol.Chem. 1986; 261: 15467-15473.

(22)

15. Boneu B, Caranobe C, Cadroy Y, Dol F, Gabaig AM, Dupouy D et al. Pharmacokinetic studies of standard unfractionated heparin, and low molecular weight heparins in the rabbit. Semin.Thromb. Hemost. 1988; 14: 18-27.

16. Hirsh J, Warkentin TE, Shaughnessy SG, Anand SS, Halperin JL, Raschke R et al. Heparin and low-mo-lecular-weight heparin: mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 2001; 119: 64S-94S.

17. Fransson LA, Carlstedt I. Alkaline and smith degradation of oxidized dermatan sulphate-chondroitin sulphate copolymers. Carbohydr.Res. 1974; 36: 349-358.

18. Conrad HE, Guo Y. Structural analysis of periodate-oxidized heparin. Adv.Exp.Med.Biol. 1992; 313: 31-36.

19. Irimura T, Nakajima M, Nicolson GL. Chemically modified heparins as inhibitors of heparan sulfate specific endo-beta-glucuronidase (heparanase) of metastatic melanoma cells. Biochemistry 1986; 25: 5322-5328.

20. Vlodavsky I, Mohsen M, Lider O, Svahn CM, Ekre HP, Vigoda M et al. Inhibition of tumor metastasis by heparanase inhibiting species of heparin. Invas.Metast. 1994; 14: 290-302.

21. Yoshitomi Y, Nakanishi H, Kusano Y, Munesue S, Oguri K, Tatematsu M et al. Inhibition of experimen-tal lung metastases of Lewis lung carcinoma cells by chemically modified heparin with reduced anticoagulant activity. Cancer Lett. 2004; 207: 165-174.

22. Sciumbata T, Caretto P, Pirovano P, Pozzi P, Cremonesi P, Galimberti G et al. Treatment with modi-fied heparins inhibits experimental metastasis formation and leads, in some animals, to long-term survival. Invas.Metast. 1996; 16: 132-143.

23. Poggi A, Rossi C, Casella N, Bruno C, Sturiale L, Dossi C et al. Inhibition of B16-BL6 melanoma lung colonies by semisynthetic sulfaminoheparosan sulfates from E. coli K5 polysaccharide. Semin. Thromb.Hemost. 2002; 28: 383-392.

24. Back N, Steger R. Effect of aprotinin, EACA and heparin on growth and vasopeptide system of Mur-phy-Sturm lymphosarcoma. Eur.J.Pharmacol. 1976; 38: 313-319.

25. Ohkoshi M, Akagawa T, Nakajima M. Effects of serine protease inhibitor FOY-305 and heparin on the growth of squamous cell carcinoma. Anticancer Res. 1993; 13: 963-966.

26. Owen CA, Jr. Anticoagulant treatment of rats with Walker 256 carcinosarcoma. J.Cancer Res.Clin. Oncol. 1982; 104: 191-193.

27. Chan SY, Pollard M. Metastasis-enhancing effect of heparin and its relationship to a lipoprotein fac-tor. J.Natl.Cancer Inst. 1980; 64: 1121-1125.

28. Lapierre F, Holme K, Lam L, Tressler RJ, Storm N, Wee J et al. Chemical modifications of heparin that diminish its anticoagulant but preserve its heparanase-inhibitory, angiostatic, tumor and anti-metastatic properties. Glycobiology 1996; 6: 355-366.

29. Ono K, Ishihara M, Ishikawa K, Ozeki Y, Deguchi H, Sato M et al. Periodate-treated, non-anticoagulant heparin-carrying polystyrene (NAC-HCPS) affects angiogenesis and inhibits subcutaneous induced tumor growth and metastasis to the lung. Br.J.Cancer 2002; 86: 1803-1812.

(23)

30. Retik AB, Arons MS, Ketcham AS, Mantel N. The effect of heparin on primary tumors and metastases. J.Surg.Res. 1962; 2: 49-53.

31. Hagmar B. Effect of heparin, epsilon-aminocaproic acid and coumarin on tumor growth and sponta-neous metastasis formation. Pathol.Eur. 1968; 3: 622-630.

32. Hagmar B. Tumor growth and spontaneous metastasis spread in two syngeneic systems. Acta Pathol. Microbiol.Scand.1970; 78: 131-142.

33. 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.

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

35. 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.

36. 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.

37. Antachopoulos CT, Gagos S, Iliopoulos DC, Karayannacos PE, Tseleni-Balafouta S, Alevras P et al. Low-dose heparin treatment does not inhibit SW480 human colon cancer growth and metastasis in vivo. In Vivo 1996; 10: 527-531.

38. Smorenburg SM, Vink R, te Lintelo M, Tigchelaar W, Maas A, Buller HR et al. In vivo treatment of rats with unfractionated heparin (UFH) or low molecular weight heparin (LMWH) does not affect experi-mentally induced colon carcinoma metastasis. Clin.Exp.Metast. 1999; 17: 451-456.

39. Bobek V, Boubelik M, Fiserova A, L’uptovcova M, Vannucci L, Kacprzak G et al. Anticoagulant drugs increase natural killer cell activity in lung cancer. Lung Cancer 2005; 47: 215-223.

40. Goldstein DS, Lu ML, Hattori T, Ratliff TL, Loughlin KR, Kavoussi LR. Inhibition of peritoneal tumor-cell implantation: model for laparoscopic cancer surgery. J.Endourol. 1993; 7: 237-241.

41. Jacobi CA, Ordemann J, Bohm B, Zieren HU, Sabat R, Muller JM. Inhibition of peritoneal tumor cell growth and implantation in laparoscopic surgery in a rat model. Am.J.Surg. 1997; 174: 359-363. 42. Jacobi CA, Peter FJ, Wenger FA, Ordemann J, Muller JM. New therapeutic strategies to avoid intra-

and extraperitoneal metastases during laparoscopy: results of a tumor model in the rat. Dig.Surg. 1999; 16: 393-399.

43. Neuhaus SJ, Ellis T, Jamieson GG, Watson DI. Experimental study of the effect of intraperitoneal hepa-rin on tumor implantation following laparoscopy. Br.J.Surg. 1999; 86: 400-404.

44. Eshraghi N, Swanstrom LL, Bax T, Jobe B, Horvath K, Sheppard B et al. Topical treatments of laparo-scopic port sites can decrease the incidence of incision metastasis. Surg.Endosc. 1999; 13: 1121-1124. 45. Pross M, Lippert H, Misselwitz F, Nestler G, Kruger S, Langer H et al. Low-molecular-weight heparin

(reviparin) diminishes tumor cell adhesion and invasion in vitro, and decreases intraperitoneal growth of colonadeno-carcinoma cells in rats after laparoscopy. Thromb.Res. 2003; 110: 215-220.

(24)

46. Pross M, Lippert H, Nestler G, Kuhn R, Langer H, Mantke R et al. Effect of low molecular weight hepa-rin on intra-abdominal metastasis in a laparoscopic experimental study. Int.J.Colorectal Dis. 2004; 19: 143-146.

47. Amirkhosravi A, Mousa SA, Amaya M, Francis JL. Antimetastatic effect of tinzaparin, a low-molecular-weight heparin. J.Thromb.Haemost. 2003; 1: 1972-1976.

48. Nagawa H, Paris P, Chauffert B, Martin F. Treatment of experimental liver metastases in the rat by continuous intraportal infusion of 5-fluorouracil and heparin: a pilot study. Anticancer Drugs 1990; 1: 149-156.

49. 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.

50. Beuth J, Ko HL, Uhlenbruck G, Pulverer G. Combined immunostimulation (Propionibacterium avi-dum KP 40) and anticoagulation (heparin) prevents metastatic lung and liver colonization in mice. J.Cancer Res.Clin.Oncol. 1987; 113: 359-362.

51. 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. 52. Miao HQ, Elkin M, Aingorn E, Ishai-Michaeli R, Stein CA, Vlodavsky I. Inhibition of heparanase activity

and tumor metastasis by laminarin sulfate and synthetic phosphorothioate oligodeoxynucleotides. Int.J.Cancer 1999; 83: 424-431.

53. 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.

54. Poggi A, Rossi C, Casella N, Bruno C, Sturiale L, Dossi C et al. Inhibition of B16-BL6 melanoma lung colonies by semisynthetic sulfaminoheparosan sulfates from E. coli K5 polysaccharide. Semin. Thromb.Hemost. 2002; 28: 383-392.

55. Stevenson JL, Choi SH, Varki A. Differential metastasis inhibition by clinically relevant levels of heparins--correlation with selectin inhibition, not antithrombotic activity. Clin.Cancer Res. 2005; 11: 7003-7011.

56. Bereczky B, Gilly R, Raso E, Vago A, Timar J, Tovari J. Selective antimetastatic effect of heparins in preclinical human melanoma models is based on inhibition of migration and microvascular arrest. Clin.Exp.Metast. 2005; 22: 69-76.

57. Im JH, Fu W, Wang H, Bhatia SK, Hammer DA, Kowalska MA et al. Coagulation facilitates tumor cell spreading in the pulmonary vasculature during early metastatic colony formation. Cancer Res. 2004; 64: 8613-8619.

58. Wang X, Wang M, Amarzguioui M, Liu F, Fodstad O, Prydz H. Downregulation of tissue factor by RNA interference in human melanoma LOX-L cells reduces pulmonary metastasis in nude mice. Int. J.Cancer 2004; 112: 994-1002.

59. Jurasz P, Alonso-Escolano D, Radomski MW. Platelet--cancer interactions: mechanisms and pharma-cology of tumor cell-induced platelet aggregation. Br.J.Pharmacol. 2004; 143: 819-826.

(25)

60. 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.

61. Camerer E, Qazi AA, Duong DN, Cornelissen I, Advincula R, Coughlin SR. Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood 2004; 104: 397-401.

62. Karpatkin S, Pearlstein E, Ambrogio C, Coller BS. Role of adhesive proteins in platelet tumor interac-tion in vitro and metastasis formainterac-tion in vivo. J.Clin.Invest 1988; 81: 1012-1019.

63. Cohen SA, Trikha M, Mascelli MA. Potential future clinical applications for the GPIIb/IIIa antagonist, abciximab in thrombosis, vascular and oncological indications. Pathol.Oncol.Res. 2000; 6: 163-174. 64. Amirkhosravi A, Mousa SA, Amaya M, Blaydes S, Desai H, Meyer T et al. Inhibition of tumor

cell-in-duced platelet aggregation and lung metastasis by the oral GpIIb/IIIa antagonist XV454. Thromb. Haemost. 2003; 90: 549-554.

65. Dvorak HF, Nagy JA, Berse B, Brown LF, Yeo KT, Yeo TK et al. Vascular permeability factor, fibrin, and the pathogenesis of tumor stroma formation. Ann.N.Y.Acad.Sci. 1992; 667: 101-111.

66. Costantini V, Zacharski LR. Fibrin and cancer. Thromb.Haemost. 1993; 69: 406-414.

67. Palumbo JS, Kombrinck KW, Drew AF, Grimes TS, Kiser JH, Degen JL et al. Fibrinogen is an important determinant of the metastatic potential of circulating tumor cells. Blood 2000; 96: 3302-3309. 68. Broze GJ, Jr. Tissue factor pathway inhibitor and the revised theory of coagulation. Annu.Rev.Med.

1995; 46: 103-112.

69. Sandset PM, Abildgaard U. Extrinsic pathway inhibitor--the key to feedback control of blood coagu-lation initiated by tissue thromboplastin. Haemostasis 1991; 21: 219-239.

70. Mousa SA, Mohamed S. Inhibition of endothelial cell tube formation by the low molecular weight heparin, tinzaparin, is mediated by tissue factor pathway inhibitor. Thromb.Haemost. 2004; 92: 627-633.

71. Poggi A, Rossi C, Casella N, Bruno C, Sturiale L, Dossi C et al. Inhibition of B16-BL6 melanoma lung colonies by semisynthetic sulfaminoheparosan sulfates from E. coli K5 polysaccharide. Semin. Thromb.Hemost. 2002; 28: 383-392.

72. Hembrough TA, Swartz GM, Papathanassiu A, Vlasuk GP, Rote WE, Green SJ et al. Tissue factor/factor VIIa inhibitors block angiogenesis and tumor growth through a nonhemostatic mechanism. Cancer Res. 2003; 63: 2997-3000.

73. Gasic GJ. Role of plasma, platelets, and endothelial cells in tumor metastasis. Cancer Metast.Rev. 1984; 3: 99-114.

74. Honn KV, Tang DG, Crissman JD. Platelets and cancer metastasis: a causal relationship? Cancer Metast.Rev. 1992; 11: 325-351.

75. Nieswandt B, Hafner M, Echtenacher B, Mannel DN. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999; 59: 1295-1300.

76. Subramaniam M, Saffaripour S, van de WL, Frenette PS, Mayadas TN, Hynes RO et al. Role of endothe-lial selectins in wound repair. Am.J.Pathol. 1997; 150: 1701-1709.

(26)

77. Talbott GA, Sharar SR, Harlan JM, Winn RK. Leukocyte-endothelial interactions and organ injury: the role of adhesion molecules. New Horiz. 1994; 2: 545-554.

78. Cummings RD, Smith DF. The selectin family of carbohydrate-binding proteins: structure and impor-tance of carbohydrate ligands for cell adhesion. Bioessays 1992; 14: 849-856.

79. Norgard-Sumnicht KE, Varki NM, Varki A. Calcium-dependent heparin-like ligands for L-selectin in nonlymphoid endothelial cells. Science 1993; 261: 480-483.

80. Koenig A, Norgard-Sumnicht K, Linhardt R, Varki A. Differential interactions of heparin and heparan sulfate glycosaminoglycans with the selectins. Implications for the use of unfractionated and low molecular weight heparins as therapeutic agents. J.Clin.Invest 1998; 101: 877-889.

81. Nelson RM, Cecconi O, Roberts WG, Aruffo A, Linhardt RJ, Bevilacqua MP. Heparin oligosaccharides bind L- and P-selectin and inhibit acute inflammation. Blood 1993; 82: 3253-3258.

82. Varki NM, Varki A. Heparin inhibition of selectin-mediated interactions during the hematogenous phase of carcinoma metastasis: rationale for clinical studies in humans. Semin.Thromb.Hemost. 2002; 28: 53-66.

83. Nakajima M, Irimura T, Nicolson GL. Heparanases and tumor metastasis. J.Cell Biochem. 1988; 36: 157-167.

84. Vlodavsky I, Korner G, Ishai-Michaeli R, Bashkin P, Bar-Shavit R, Fuks Z. Extracellular matrix-resident growth factors and enzymes: possible involvement in tumor metastasis and angiogenesis. Cancer Metast.Rev. 1990; 9: 203-226.

85. Vlodavsky I, Goldshmidt O, Zcharia E, Atzmon R, Rangini-Guatta Z, Elkin M et al. Mammalian hepa-ranase: involvement in cancer metastasis, angiogenesis and normal development. Semin.Cancer Biol. 2002; 12: 121-129.

86. Maxhimer JB, Quiros RM, Stewart R, Dowlatshahi K, Gattuso P, Fan M et al. Heparanase-1 expression is associated with the metastatic potential of breast cancer. Surgery 2002; 132: 326-333.

87. Friedmann Y, Vlodavsky I, Aingorn H, Aviv A, Peretz T, Pecker I et al. Expression of heparanase in normal, dysplastic, and neoplastic human colonic mucosa and stroma. Evidence for its role in colonic tumorigenesis. Am.J.Pathol. 2000; 157: 1167-1175.

88. Ginath S, Menczer J, Friedmann Y, Aingorn H, Aviv A, Tajima K et al. Expression of heparanase, Mdm2, and erbB2 in ovarian cancer. Int.J.Oncol. 2001; 18: 1133-1144.

89. Gohji K, Hirano H, Okamoto M, Kitazawa S, Toyoshima M, Dong J et al. Expression of three extracel-lular matrix degradative enzymes in bladder cancer. Int.J.Cancer 2001; 95: 295-301.

90. Koliopanos A, Friess H, Kleeff J, Shi X, Liao Q, Pecker I et al. Heparanase expression in primary and metastatic pancreatic cancer. Cancer Res. 2001; 61: 4655-4659.

91. Bitan M, Polliack A, Zecchina G, Nagler A, Friedmann Y, Nadav L et al. Heparanase expression in hu-man leukemias is restricted to acute myeloid leukemias. Exp.Hematol. 2002; 30: 34-41.

92. Takahashi H, Ebihara S, Okazaki T, Suzuki S, Asada M, Kubo H et al. Clinical significance of heparanase activity in primary resected non-small cell lung cancer. Lung Cancer 2004; 45: 207-214.

(27)

93. Yang Y, Macleod V, Bendre M, Huang Y, Theus AM, Miao HQ et al. Heparanase promotes the sponta-neous metastasis of myeloma cells to bone. Blood 2005; 105: 1303-1309.

94. Ishai-Michaeli R, Eldor A, Vlodavsky I. Heparanase activity expressed by platelets, neutrophils, and lymphoma cells releases active fibroblast growth factor from extracellular matrix. Cell Regul. 1990; 1: 833-842.

95. Whitelock JM, Murdoch AD, Iozzo RV, Underwood PA. The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases. J.Biol.Chem. 1996; 271: 10079-10086.

96. Sanderson RD, Yang Y, Suva LJ, Kelly T. Heparan sulfate proteoglycans and heparanase--partners in osteolytic tumor growth and metastasis. Matrix Biol. 2004; 23: 341-352.

97. Elkin M, Ilan N, Ishai-Michaeli R, Friedmann Y, Papo O, Pecker I et al. Heparanase as mediator of angio-genesis: mode of action. FASEB J. 2001; 15: 1661-1663.

98. Vlodavsky I, Friedmann Y. Molecular properties and involvement of heparanase in cancer metastasis and angiogenesis. J.Clin.Invest 2001; 108: 341-347.

99. Kato M, Wang H, Kainulainen V, Fitzgerald ML, Ledbetter S, Ornitz DM et al. Physiological degradation converts the soluble syndecan-1 ectodomain from an inhibitor to a potent activator of FGF-2. Nat. Med. 1998; 4: 691-697.

100. Parish CR, Coombe DR, Jakobsen KB, Bennett FA, Underwood PA. Evidence that sulphated polysac-charides inhibit tumor metastasis by blocking tumor-cell-derived heparanases. Int.J.Cancer 1987; 40: 511-518.

101. Vlodavsky I, Friedmann Y, Elkin M, Aingorn H, Atzmon R, Ishai-Michaeli R et al. Mammalian heparan-ase: gene cloning, expression and function in tumor progression and metastasis. Nat.Med. 1999; 5: 793-802.

102. Poggi A, Rossi C, Casella N, Bruno C, Sturiale L, Dossi C et al. Inhibition of B16-BL6 melanoma lung colonies by semisynthetic sulfaminoheparosan sulfates from E. coli K5 polysaccharide. Semin. Thromb.Hemost. 2002; 28: 383-392.

103. Colin S, Jeanny JC, Mascarelli F, Vienet R, Al Mahmood S, Courtois Y et al. In vivo involvement of heparan sulfate proteoglycan in the bioavailability, internalization, and catabolism of exogenous basic fibroblast growth factor. Mol.Pharmacol. 1999; 55: 74-82.

104. Collen A, Smorenburg SM, Peters E, Lupu F, Koolwijk P, Van Noorden C et al. Unfractionated and low molecular weight heparin affect fibrin structure and angiogenesis in vitro. Cancer Res. 2000; 60: 6196-6200.

105. D’Amore PA. Capillary growth: a two-cell system. Semin.Cancer Biol. 1992; 3: 49-56.

106. Folkman J, Weisz PB, Joullie MM, Li WW, Ewing WR. Control of angiogenesis with synthetic heparin substitutes. Science 1989; 243: 1490-1493.

107. Soker S, Goldstaub D, Svahn CM, Vlodavsky I, Levi BZ, Neufeld G. Variations in the size and sulfation of heparin modulate the effect of heparin on the binding of VEGF165 to its receptors. Biochem. Biophys.Res.Commun. 1994; 203: 1339-1347.

(28)

108. Lepri A, Benelli U, Bernardini N, Bianchi F, Lupetti M, Danesi R et al. Effect of low molecular weight heparan sulphate on angiogenesis in the rat cornea after chemical cauterization. J.Ocul.Pharmacol. 1994; 10: 273-280.

109. Jayson GC, Gallagher JT. Heparin oligosaccharides: inhibitors of the biological activity of bFGF on Caco-2 cells. Br.J.Cancer 1997; 75: 9-16.

110. Kragh M, Binderup L, Vig Hjarnaa PJ, Bramm E, Johansen KB, Frimundt PC. Non-anti-coagulant hepa-rin inhibits metastasis but not primary tumor growth. Oncol.Rep. 2005; 14 :99-104.

111. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N.Engl.J.Med. 2004; 351: 781-791.

112. Cristofanilli M, Hayes DF, Budd GT, Ellis MJ, Stopeck A, Reuben JM et al. Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer. J.Clin.Oncol. 2005; 23: 1420-1430.

(29)

Referenties

GERELATEERDE DOCUMENTEN

The most prominent misfit between laypeople and experts was found in the textual questions, for the definitions of river basin (10 log BF of 14.9), river (10 log BF of 11.9),

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

Hij gaat terug in de tijd op zoek naar de wortels van het 'Publiek Orgaan voor de Arbeidsallocatie' ( po a ), een door Van Bekkum geïntroduceerd begrip dat als

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

It was first reported that in ZS fibroblasts from com- plementation group 4 at KKI (group C at Gifu [PEX6 defect]) the PMPs were located in unusual empty membrane structures

We find that allowing for (asymmetric) stochastic volatility reduces option pricing errors and explains the implied volatility smile, and that allowing for correlation between