161
UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) UvA-DARE (Digital Academic Repository) Cancer, thrombosis and low-molecular-weight heparins Piccioli, A. Publication date 2015 Document Version Final published version Link to publication Citation for published version (APA): Piccioli, A. (2015). Cancer, thrombosis and low-molecular-weight heparins. 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. Download date:14 Jun 2021

UvA-DARE (Digital Academic Repository) Cancer, thrombosis and … · Piccioli, A. Link to publication Citation for published version (APA): Piccioli, A. (2015). Cancer, thrombosis

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

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

    UvA-DARE (Digital Academic Repository)

    Cancer, thrombosis and low-molecular-weight heparins

    Piccioli, A.

    Publication date2015Document VersionFinal published version

    Link to publication

    Citation for published version (APA):Piccioli, A. (2015). Cancer, thrombosis and low-molecular-weight heparins.

    General rightsIt 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 opencontent license (like Creative Commons).

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

    Download date:14 Jun 2021

    https://dare.uva.nl/personal/pure/en/publications/cancer-thrombosis-and-lowmolecularweight-heparins(1860fc0b-cb7d-46ab-9c9e-0e2327b181b7).html

  • CANCER, THROMBOSIS AND

    LOW-MOLECULAR-WEIGHT HEPARINS

    ANDREA PICCIOLI

    AN

    DR

    EA P

    ICC

    IOLI

    C

    AN

    CER

    , TH

    RO

    MBO

    SIS

    AN

    D L

    OW

    -MO

    LEC

    ULA

    R-W

    EIG

    HT

    HEP

    AR

    INS

  • CANCER,THROMBOSIS AND LOW-MOLECULAR-WEIGHT HEPARINS

    Andrea Piccioli

  • Cover: watercolor of Vicenza, Italy. Reproduced with permission of Gilberto Padovan Editore

  • CANCER, THROMBOSIS AND LOW-MOLECULAR-WEIGHT HEPARINS

    ACADEMISCH PROEFSCHRIFT

    ter verkrijging van de graad van doctor

    aan de Universiteit van Amsterdam op gezag van de Rector Magnificus

    prof.dr. D.C. van den Boom

    ten overstaan van een door het college voor promoties ingestelde

    commissie, in het openbaar te verdedigen in de Agnietenkapel

    op vrijdag 20 maart 2015, te 10.00 uur

    door

    Andrea Piccioli

    geboren te Vicenza, Italië

  • Promotiecommissie:

    Promotor: Prof. dr. H.R. Büller Co-promotor: Prof. dr. P. Prandoni Overige leden: Prof. dr. D.P.M. Brandjes Dr. S.C. Cannegieter Prof. dr. M.M. Levi Prof. dr. S. Middeldorp Dr. H.M. Otten Prof. dr. M.H. Prins

    Faculteit der Geneeskunde

  • Labor omnia vicit /

    Improbus et duris urgens in rebus egestas

    Verg., Georg. I, 145-146

  • CONTENTS

    Chapter 1. Piccioli A.

    Introduction and outline of the thesis...................................................................................Page 01

    Part A. Venous thromboembolism in cancer patient

    Chapter 2. Cancer and venous thromboembolism.

    Published in past in: Amer Heart J 1996;132:1010-4, Lancet Oncol 2005; 6:401, Expert Opin

    Pharmacother. 2014 Apr; 15 (6): 799-807 ..........................................................................Page 11

    Chapter 3. Recurrent venous thromboembolism and bleeding complications during anticoagulant

    treatment in patients with cancer and venous thrombosis.

    Blood 2002;100:3484 -8 ……..…………………………………………………………………..Page 31

    Chapter 4. Predicting recurrences or major bleeding in cancer patients with venous

    thromboembolism. Findings from the RIETE Registry.

    Thromb Haemost 2008;100:435-9. ....................................................................................Page 41

    Chapter 5. Approach to venous thromboembolism in the cancer patient.

    Curr Treat Options Cardiovasc Med 2011;13 (2): 159-68...................................................Page 49

    Part B. Venous thromboembolism and the risk of cancer

    Chapter 6. Venous thromboembolism as first manifestation of cancer.

    Acta Haematol 2001;106:13-7. ...........................................................................................Page 61

    Chapter 7. Screening for occult cancer in patients with idiopathic venous thromboembolism: Yes.

    J Thromb Haemost 2003;1: 2271-2....................................................................................Page 69

    Chapter 8. Extensive screening for occult malignant disease in idiopathic venous

    thromboembolism: a prospective randomized clinical trial.

    J Thromb Haemost 2004;2:884-9........................................................................................Page 75

    Chapter 9. Decision analysis for cancer screening in idiopathic venous thromboembolism.

    J Thromb Haemost 2005;3:2391-6......................................................................................Page 85

    Chapter 10. The value of CT scanning for detection of occult cancer in patients with idiopathic

    VTE. D’Acquapendente Study. Submitted 2014.................................................................Page 95

  • Chapter 11. The long-term risk of cancer in patients with a first episode of venous

    thromboembolism. J Thromb Haemost 2009;7:546-51. ………………………………..…..Page 107

    Chapter 12. The risk of cancer in patients with venous thromboembolism does not exceed

    that expected in the general population after the first 6 months.

    J Thromb Haemost 2010; 8 (5): 1126-7. ............................................................................Page 117

    Part C. Effect of low-molecular-weight heparins on survival in cancer patients

    Chapter 13. Anticoagulants and cancer survival.

    Semin Thromb Haemost 2006;32:810-3. ...........................................................................Page 123

    Chapter 14. The effect of low-molecular-weight heparin on survival in patients with advanced

    malignancy. J Clin Oncol 2005;23:2130-5.........................................................................Page 129

    Summary. ...........................................................................................................................Page 139

    Samenvatting. ....................................................................................................................Page 143

  • CHAPTER 1

    INTRODUCTION

    Andrea Piccioli

    University Hospital of Padua, Italy

  • 2

    BACKGROUND

    Since Trousseau’s time, the clinical association between cancer and venous thromboembolism

    (VTE) has been observed and documented (1).

    The relationship between cancer and VTE insist on a two way clinical correlation. In fact it has

    been clearly established that cancer patients exhibit a higher risk of developing a thrombotic

    event when compared to non-cancer patients, especially in the presence of the well-known risk

    factors for thrombosis, such as prolonged immobilization, surgery and chemo-radio-hormonal

    therapy. It has also been clearly noticed that a first episode of idiopathic VTE may represent the

    first manifestation of a yet undisclosed cancer, offering chances for an earlier diagnosis of the

    pathology (2,3,4).

    A substantial number of cancer patients receive anticoagulants either for prophylaxis, due to the

    high risk of developing thrombosis, or for treatment of an already established thrombotic event.

    The initial treatment of VTE in cancer patients should not differ from that of non cancer patients.

    In fact they will receive UFH or LMWH at therapeutic dosages. They begin also with warfarin.

    Once the INR has reached the therapeutic target, heparin or LMWH can be stopped and

    warfarin is continued for the remainder of the treatment period (4).

    Cancer patients are at high risk of recurrent VTE when anticoagulation is stopped: studies

    conducted during the past decade, assessing the clinical course of patients with thrombosis,

    have documented that the risk of recurrence increases progressively in the course of time,

    being as high as 30% after 10 years from the thrombotic event. If risk factors for recurrence are

    considered, malignancy is the most common, being more frequent than thrombophilia. For

    these reasons, in patients with cancer and thrombosis, anticoagulation is recommended for as

    long as cancer is active (2,5).

    Moreover cancer patients are at higher risk of both recurrence and major bleeding

    complications during anticoagulation, even if they are within the therapeutic range. Several

    published clinical trial have examined the administration of long-term LMWH as an alternative to

    warfarin therapy in cancer patients with VTE. There is strong evidence that LMWH is more

    efficacious than warfarin for preventing symptomatic recurrent VTE in cancer patients (4,6).

    A complex relation exists between the coagulation system and cancer. Hemostasisand

    malignancy share linking mechanisms so that it has been noticed that the inhibition of

    hemostasis activation may impact on outcomes from malignancy. In particular LMWH may have

    potential antitumor effects. Further evaluations is warranted to assess if anticoagulation has the

    potential to prolong survival in cancer patients (7,8).

    The incidence of newly diagnosed cancer during the follow-up of patients with VTE is higher

    than in the general population. VTE, especially in its idiopathic presentation, may represent the

    first manifestation of a yet undisclosed cancer, offering a possible chance for early diagnosis

    and treatment. Newly diagnosed malignancies are not confined to certain subtypes, but involve

    virtually all body systems. Some of these malignancies can be identified by routine assessment

    at the time of the diagnosis of the thrombotic event. Moreover, in patients with idiopathic VTE,

    who are apparently cancer free at baseline, there remains an approximate 7-10% incidence of

    clinically overt malignant disease during the follow-up period after the thrombotic event (1,2,9).

  • 3

    OUTLINE OF THE THESIS

    Part A “Venous thromboembolism in cancer patients”

    In chapter 2 we have analysedthe two way clinical correlation between cancer and venous

    thromboembolism presenting data from three review articles which have

    examinedepidemiology, risk factors, prophylaxis and treatment of VTE in cancer patients

    (1,2,3).

    The risk of recurrent VTE and bleeding complications during anticoagulant treatment in cancer

    patients is presented in chapter 3. In this prospective follow-up study we sought to determine

    whether in patients with established thrombosis those with cancer are at higher risk for recurrent

    venous thromboembolism or bleeding complications during anticoagulant treatment than those

    without cancer. It has been demonstrated that cancer patients with venous thrombosis are more

    likely to develop recurrent thromboembolic complications and major bleeding during

    anticoagulant treatment than those without malignancy. These risks correlate with the extent of

    cancer (5).

    Chapter 4 deals with the predictors of recurrent VTE or major bleedings in cancer patients with

    VTE, examining some variables available at entry among patients from the RIETE International

    Registry. We tried to identify which cancer patients are at a higher risk for recurrent pulmonary

    embolism (PE), deep vein thrombosis (DVT) or major bleeding. On multivariate analysis,

    patients aged

  • 4

    In chapter 7 evidence has been provided in favourof extensive screening procedures for cancer identification to be performed among patients with a first episode of idiopathic VTE in whom a routine initial screening for cancer identification is negative. Arguments originate from available literature on this topic (10). In chapter 8 we presented the SOMIT study, a prospective evaluation in which apparently

    cancer-free patients with acute idiopathic venous thromboembolism were randomized to either

    the strategy of extensive screening for occult cancer or to no further testing. Patients had a 2-

    year follow-up period. Although early detection of occult cancers may be associated with

    improved treatment possibilities, it is uncertain whether this improves the prognosis (12).

    In chapter 9, a decision analysis from the data of the SOMIT-trial is presented. The screening

    tests were divided in several possible strategies. The number of detected cancers and the

    number of patients who underwent further investigations eventually ending in a benign

    condition, were calculated for each strategy and the total costs were determined. Based on

    tumor type, stage, age and gender of the individual cancer patient, the difference in life-years

    gained (LYG) was calculated between the two study groups. It has been found that the

    screening for cancer including adbomino/pelvic CT with or without mammography and/or

    sputum cytology appears potentially cost effective in patients with idiopathic VTE (13).

    Chapter 10 presents the D’Acquapendente Study, a randomized open label trial among

    patients with a first episode of idiopathic VTE and a normal baseline routine screening, which

    compared an extensive compound strategy based of CT scan of thorax, abdomen and pelvis

    plus haemoccult to a common clinical practice strategy based on the attending physician

    preference (but excluding CT as a first line test) for the identification of hidden cance r(14).

    Chapter 11 and chapter 12 deals with the long-term risk of cancer in patients with VTE. In

    patients with VTE, 15-20% will have overt cancer when VTE is diagnosed but little is known

    about such patients' long-term risk, time course and predictors of new cancer. In patients with a

    first VTE and without clinically evident cancer, the risk for new cancer is about 1-2% per year,

    appears to be uniform over time, and is higher in patients with unprovoked VTE and those with

    advanced age. Moreover the risk of cancer in patients with VTE does not exceed that expected

    in the general population after the first 6 months (15,16).

    Part C. Effect of low-molecular-weight heparins on survival

    in cancer patients

    The association between cancer and activation of blood coagulation has been described since

    Trousseau's time (1). The hypercoagulable state often encountered in cancer patients not only

    acts as an important risk factor for thrombosis, but also may play a role in tumour progression

    and metastatization. An anti-neoplastic effect of anticoagulants in this setting has often been

    hypothesized. The results of recently conducted clinical trials suggest that cancer patients could

    benefit from the administration of low molecular weight heparins, particularly those with

  • 5

    nonadvanced disease. Additional clinical trials are needed to provide further insight into this

    challenging setting. Studies in cancer patients with venous thromboembolism suggested that

    low molecular weight heparin may prolong survival. Chapter 13 is an overview of the current

    knowledge in this field (17) and chapter 14 presents the results of a trial among patients with

    metastasized or locally advanced solid tumours, who were randomly assigned to receive a 6-

    week course of subcutaneous nadroparin or placebo. The primary efficacy analysis was based

    on time from random assignment to death. The primary safety outcome was major bleeding

    (18).

  • 6

    References

    1. Trousseau A Phlegmasia alba dolens. Lectures on clinical medicine. The New Sydenham Society, London 1868; 5:281-331

    2. Piccioli A, Prandoni P, Goldhaber SZ. Epidemiologic characteristics, management, and outcome of deep venous thrombosis in a tertiary care hospital: The Brigham and Women's Hospital DVT Registry. Amer Heart J 1996;132:1010-4

    3. Prandoni P, Falanga A, Piccioli A. Cancer and venous thromboembolism. Lancet Oncol 2005;6:401- 410

    4. Prandoni P, Piovella C, Filippi L, Vedovetto V, Dalla Valle F & Piccioli A. What are the pharmacotherapy options for treating venous thromboembolism in cancer patients? Expert Opin Pharmacother 2014 Apr;15(6):799-807

    5. Prandoni P, Lensing AWA, Piccioli A, Bernardi E, Simioni P, Girolami B, Marchiori A, Sabbion P, Prins MH, Noventa F, Girolami A. Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood 2002;100:3484-8

    6. Piccioli A, Prandoni P. Approach to venous thromboembolism in the cancer patient. Curr Treat Options Cardiovasc Med 2011; 13 (2): 159-68

    7. Piccioli A,Falanga A, Prandoni P. Anticoagulants and cancer survival. SeminThromb Haemost 2006;32:810-3

    8. Klerk CP, Smorenburg SM, Otten HM, Lensing AW, Prins MH, Piovella F, Prandoni P, Bos MM, Richel DJ, van Tienhoven G, Buller HR, for the MALT Investigators (including Piccioli A). The Effect of Low Molecular Weight Heparin on Survival in Patients With Advanced Malignancy. J ClinOncol 2005;23:2130-5

    9. Piccioli A, Prandoni P. Venous thromboembolism as first manifestation of cancer. Acta Haematol 2001;106:13-7

    10. Piccioli A.Prandoni P. Screening for occult cancer in patients with idiopathic venous thromboembolism: Yes. J ThrombHaemost 2003;1: 2271-2

    11. Tryjillo-Santos J, Nieto JA, Tiberio G, Piccioli A, Di Micco P, Prandoni P, Monreal M; RIETE Registry.Predicting recurrences or major bleeding in cancer patients with venous thromboembolism. Findings from the RIETE Registry. Thromb Haemost 2008;100:435-9

    12. Piccioli A, Lensing AW, Prins MH, Falanga A, Scannapieco GL, Ieran M, Cigolini M, Ambrosio GB, Monreal M, Girolami A, Prandoni P, for the SOMIT Investigators Group. Extensive screening for occult malignant disease in idiopathic venous thromboembolism: a prospective randomized clinical trial. J Thromb Haemost 2004;2:884-9

    13. Di Nisio M, Otten HM, Piccioli A, Lensing AWA, Prandoni P, Büller HR, Prins MH. Decision analysis for cancer screening in idiopathic venous thromboembolism. J Thromb Haemost 2005;3:2391-6

  • 7

    14. Piccioli A, Bernardi E, Dalla Valle F, Visonà A, Tropeano PF, BovaC,Bucherini E , Barbar S, Falanga A, Prandoni P. The value of CT scanning for detection of occult cancer in patients with idiopathic VTE: D’Acquapendente Study. Submitted 2014

    15. Douketis JD, Gu C, Piccioli A, Ghirarduzzi A, Pengo V, Prandoni P.The long-term risk of cancer in patients with a first episode of venous thromboembolism. J Thromb Haemost 2009;7:546-51

    16. Prandoni P, Casiglia A., Piccioli A,Ghirarduzzi A, Pengo V, Gu C, and Douketis JD. The risk of cancer in patients with venous thromboembolism does not exceed that expected in the general population after the first 6 months. J Thromb Haemost 2010;8 (5): 1126-7

    17. Piccioli A, Falanga A, Prandoni P. Anticoagulants and cancer survival. Semin Thromb Haemost 2006;32:810-3

    18. Klerk CP, Smorenburg SM, Otten HM, Lensing AW, Prins MH, Piovella F, Prandoni P, Bos MM, Richel DJ, van Tienhoven G, Buller HR, for the MALT Investigators (including Piccioli A). The Effect of Low Molecular Weight Heparin on Survival in Patients With Advanced Malignancy. J ClinOncol 2005;23:2130-5

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=

  • 8

  • 9

    A. VENOUS THROMBOEMBOLISM IN CANCER PATIENTS

  • 10

  • 11

    CHAPTER 2

    Cancer and venous thromboembolism

    Piccioli A, Prandoni P

    Published in part in: Andrea Piccioli MD, Paolo Prandoni, MD, Bruce M Ewenstein MD, and Samuel Z. Goldhaber MD. Padua, Italy and Boston, Mass. Cancer and venous thromboembolism, Amer Heart J 1996;132:1010-4. Paolo Prandoni, Anna Falanga, Andrea Piccioli. Cancer and venous thromboembolism. Lancet Oncol 2005;6:401-410. Piccioli A, Falanga A, Baccaglini U, Marchetti M, Prandoni P Cancer and venous Thromboembolism. Semin Thromb Hemost 2006; 32(7);694-9. Paolo Prandoni, Chiara Piovella, Lucia Filippi, Valentina Vedovetto, Fabio Dalla Valle & Andrea Piccioli What are the pharmacotherapy options for treating venous thromboembolism in cancer patients? Expert Opin Pharmacother 2014 Apr;15(6):799-80.7

  • 12

    Abstract

    Venous thromboembolism occurs commonly in patients with cancer. The pathogenetic mechanisms of thrombosis involve a complex interaction between tumour cells, the haemostatic system, and characteristics of the patient. Among risk factors for thromboembolism the most important are long-term immobilisation, especially in hospital, surgery, and chemotherapy with or without adjuvant hormone therapy. Although prophylaxis and treatment of thromboembolism in patients with cancer draw on the agents that are commonly used in those without cancer, there are many special features of patients with cancer that make use of these drugs more challenging. Low-molecular weight heparins are the cornerstone of prophylaxis and treatment of venous thromboembolism in patients with cancer. These drugs have the potential to increase survival, at least in patients with more favourable outlook. Introduction Thromboembolism is a well-recognised complication of malignant disease. Clinical manifestations vary from venous thromboembolism to disseminated intravascular coagulation and arterial embolism. Disseminated intravascular coagulation is most commonly observed in patients with haematological malignant disorders and those with widespread metastatic cancer, whereas arterial embolism is most commonly observed in patients undergoing chemotherapy and in those with non-bacterial thrombotic endocarditis. This review focuses on the relation between cancer and venous thromboembolism. Pathogenesis of thrombosis in cancer Cancer growth is associated with the development of a hypercoaguable state. Patients with malignant disorders but no thrombosis commonly present with abnormalities in laboratory coagulation tests, which suggest a continuous process of fibrin formation and removal at different rates in these patients (1). Fibrin and other clot components have roles not only in thrombogenesis but also in tumour adhesion,spread,and metastasis (2). Many histopathological studies have shown the presence of fibrin or platelet plugs in and around many types of tumours (3) which suggests local activation of blood coagulation and an involvement of clotting mechanisms in the growth of malignant tissues. Prothrombotic mechanisms The activation of blood coagulation in patients with cancer is complex and multifactorial (4). General prothromboticmechanisms are related to the host response to cancer and include the acute-phase reaction, paraprotein production, inflammation,necrosis, and hemodynamic disorders. Procoagulant effects are also exerted by anticancer chemotherapy and radiotherapy. However, a prominent part is played by tumour-specific clot-promoting mechanisms resulting from the prothrombotic properties expressed by tumour cells themselves. These properties are unique to the malignant state. Malignant cells can activate blood coagulation in several ways: by producing procoagulant, fibrinolytic, and proaggregating activities; by releasing pro-inflammatory and proangiogenic cytokines; and by interacting directly with host vascular and blood cells, such as endothelial cells, leucocytes, and platelets, by means of adhesion molecules. Procoagulant,fibrinolytic, and proaggregating activities Tumour cells produce procoagulant factors, among which the most studied are tissue factor and cancer procoagulant (5). Tissue factor, the primary activator of healthy blood coagulation, forms a complex with factor VII to activate factors X and IX by proteolysis. In healthy vascular cells, expression of tissue factor is tightly controlled; the factor is normally not expressed but is induced by inflammatory stimuli such as the cytokines interleukin 1 and tumour necrosis factor

  • 13

    (TNF) as well as bacterial lipopolysaccharides. In malignant cells, however, tissue factor is constitutively expressed. By contrast, cancer procoagulant is a cysteine proteinase that directly activates factor X independently of factor VII; it has been found in tumour cells and in tissues of the amnion and chorion but not in normal differentiated cells. Tissue factor and cancer procoagulant have been identified in several human and animal tumour tissues (6). The finding that severe coagulopathy in acute promyelocytic leukaemia resolves in parallel with the loss of blast-cell procoagulant activities from the patient’s bone marrow strongly supports the role of tumour procoagulants in promoting clotting complications in malignant disorders (7). Tumour cells can express all proteins that regulate the fibrinolytic system, including the urokinase-type and tissue-type plasminogen activators, plasminogen-activator inhibitors 1 and 2, and plasminogen-activator receptor (8). The increase in plasma concentrations of plasminogen-activator inhibitors and impairment in plasma fibrinolytic activity in patients with solid tumours indicate another tumour-associated prothrombotic mechanism. Tumour cells induce platelet activation and aggregation by direct cell–cell contact or by releasing soluble factors, such as ADP, thrombin, and other proteases (9). Circulating activated platelets expose on their surface the activation-dependent antigens P-selectin and CD 63. On aggregation they release their granule contents. Activation of platelets increases their capacity to interact by specific adhesive mechanisms with endothelial cells, leucocytes, and tumour cells. Tumour-cell-derived cytokines Tumour cells produce and release various cytokines, including TNF, interleukin 1, and vascular endothelial growth factor (VEGF), which can be involved in the development of thrombotic disorders in patients with cancer (10). The major targets of tumour-derived cytokines are the vascular endothelium and leucocytes. TNFa and interleukin 1 induce the expression of endothelial procoagulant activity (tissue factor) and simultaneously down regulate the expression of thrombomodulin, the endothelial surface high affinity receptor for thrombin, which complexes thrombin to activate the potent anticoagulant protein-C system. Together, up-regulation of tissue factor and down-regulation of thrombomodulin lead to a prothrombotic condition in the vascular wall. The same cytokines strongly stimulate the production of the fibrinolysis inhibitor plasminogen-activator inhibitor 1, thus impairing the endothelial antithrombotic response. Tumour-derived VEGF also induces expression of tissue factor by endothelial cells, which implies involvement of tissue factor in tumour neovascularisation (11). Finally, cytokines induce changes in expression of endothelial-cell adhesion molecule, increasing the capacity of the vessel wall to attract leucocytes and platelets and promoting localised clotting activation and fibrin formation. In a similar way to endothelial cells, monocytes are activated by tumour cells, their products, or both to express tissue factor on their surfaces (12). Tumour-associated macrophages obtained from experimental and human tumours express substantially more tissue factor than do control cells, and circulating monocytes from patients with various types of cancer show increased tissue-factor activity. Tumour cytokines also attract and activate polymorphonuclear leucocytes, which release reactive oxygen species and intracellular proteases that have several activities on endothelial cells and platelets, modifying the haemostatic balance towards a prothrombotic state (13). Cell–cell interactions The presence of cell-adhesion molecules on the surface of tumour cells allows the possibility of direct interaction with healthy cells. During haematogenous spread, this interaction occurs with endothelial cells, platelets, and leucocytes. The capacity of tumour cells to adhere to both resting and cytokine-stimulated endothelium is well known, and adhesion-molecule pathways specific to different tumour-cell types have been identified (2,4). Malignant cells attached to the vessel wall promote localised clotting activation and thrombus formation and promote the adhesion and arrest of leucocytes and platelets by releasing cytokines. Cancer cells also directly activate platelets, adhere and migrate through the vessel wall, and are assisted by polymorphonuclear leucocytes in their interaction with endothelial cells.

  • 14

    Prothrombotic mechanisms and tumour progression Tumour-specific prothrombotic properties contribute to the process of tumour growth and dissemination. The formation of thrombin, the final effector enzyme of the clotting cascade, and production of fibrin, the final product of the activation of blood coagulation, are coagulation-dependent mechanisms of tumour progression. In addition, tumour prothrombotic properties can interfere with the malignant process by coagulation-independent mechanisms . Relevant in this setting is the emerging role of the non-coagulant activities of tissue factor (2), particularly its capacity to modulate VEGF expression by malignant cells and normal vascular cells. This property regulates tumour neovascularisation and provides an important link in patients with cancer between activation of coagulation, inflammation, thrombosis, and cancer growth and metastasis (14). Epidemiology and risk factors for thromboembolism in patients with cancer Since the initial observation by Trousseau in 1865, many studies have addressed the relation between cancer and venous thromboembolism. This disorder is a common complication in patients with cancer. In some cases it is the first manifestation of cancer, so offering opportunities for diagnosis and treatment (15). In patients with malignant disorders, venous thromboembolism is an important cause of morbidity and mortality. Of every seven patients with cancer who die in hospital, one dies of pulmonary embolism (16). Of these patients, 60% have localised cancer or limited metastatic disease, and they would have survived for longer in the absence of pulmonary embolism. According to the Medicare Provider Analysis and Review Record database that records the primary discharge diagnosis and an additional four discharge diagnoses in the USA, initial or recurrent thromboembolism in patients with cancer exceeds by far that recorded in those without malignant disorders; thromboembolism complicates the course of cancers of virtually all body systems with similar frequency (17). The true frequency of venous thromboembolism in patients with cancer is not known, because of the surprising lack of information in almost all studies dealing with the natural course of malignant diseases. Most thrombotic episodes occur spontaneously, in the absence of triggering factors commonly accounting for thromboembolic complications in people without cancer (18,19). Patients with cancer have a highly increased risk of venous thromboembolism in the first few months after diagnosis and in the presence of distant metastases (19). The risk is further increased in the presence of inherited thrombophilic abnormalities (19). The most common situations that increase the risk of venous thromboembolism in patients with cancer include immobilisation, surgery, chemotherapy with or without hormone therapy, and the insertion of central venous catheters (20). Immobilisation One of the most important triggering factors for venous thromboembolism is long-term immobilisation, especially during a hospital stay. This pattern was clearly confirmed by Shenand Pollack, (16) who reported that up to 14% of patients with cancer admitted to hospital died of autopsy-confirmed pulmonary embolism, compared with 8% of those without cancer (16). Surgery In the absence of adequate prophylaxis, patients with active cancer face a very high risk of developing venous thromboembolism postoperatively. In the absence of thromboprophylaxis, the overall incidence of postoperative deep-vein thrombosis is about two times higher in patients with cancer than in patients without malignant disease (15). Many factors contribute to this high frequency, including advanced age, long and complicated surgical procedures, and late mobilisation with long postoperative course owing to the patient’s poor condition. If

  • 15

    thromboprophylaxis is not extended beyond the hospital stay, patients with cancer remain at risk of developing late venous thromboembolism (21, 22). Chemotherapy, radiotherapy, and adjuvant hormone therapy Patients with cancer are also at high risk of developing both venous and arterial thrombosis when they receive chemotherapy (23). In patients with high-grade glioma undergoing chemotherapy, the frequency of thromboembolic complications was as high as 16% (24). In a retrospective study in patients who had been given chemotherapy, the rate of thromboembolic complications arising within the first (3) months was unexpectedly high, giving an annual rate of 11% (25). The most reliable estimate of thromboembolic complications in patients undergoing chemotherapy comes from those with breast cancer. The frequency of chemotherapy-induced thromboembolic complications in women with stage II breast cancer undergoing chemotherapy was on average 7% in available studies assessing this risk (23). Among patients with stage IV breast cancer the risk was even higher (26). Hormone therapy combined with chemotherapy further increases the risk of thromboembolic complications in women with breast cancer (27). Even when given alone, tamoxifen to prevent recurrence or for prevention of breast cancer in women at high risk, slightly increases the rate of venous thromboembolism (28,29). By comparison with tamoxifen, third-generation oral aromatase inhibitors, such as the irreversible steroid inactivator exemestane, have the potential to be associated with a lower rate of thromboembolic events (30). Although radiotherapy is widely believed to be a risk factor for venous thromboembolism in patients with cancer, no study has as yet adequately investigated its role. Central venous catheters Long-dwelling central venous catheters have greatly improved the management of patients with cancer. However, their use has been associated with the occurrence of deep-vein thrombosis in the arms, especially in patients undergoing chemotherapy (31). The true frequency of deep-vein thrombosis in patients with central venous lines is difficult to estimate, because published data are somewhat conflicting. In the absence of thromboprophylaxis, Bern and colleagues (32) found that the rate of deep-vein thrombosis, as shown by phlebography, was 37%. Monreal and co-workers (33) found an even higher rate. By contrast, in case series in which ultrasonography or other non-invasive methods were used to detect arm deep-vein thromboses, a much lower rate has been reported (31,34). Along with the lower sensitivity of objective non-invasive methods in comparison with phlebography, the availability of new textures and coating of catheters, and the introduction of new procedures to reduce their invasiveness is likely to account for discrepancies between older and more recent studies. Thromboprophylaxis Although many patients with active cancer develop thrombotic complications spontaneously, in the absence of other risk factors, there is probably little point in providing thromboprophylaxis to all patients with cancer who are not undergoing surgical or medical therapy. However, a history of thromboembolism puts patients with cancer at such a high risk of recurrence that the systematic use of mechanical or pharmacological prophylaxis should be considered even in the absence of the common risk factors for thrombosis. Prevention of venous thromboembolism in cancer is an important challenge, because patients experiencing a thrombotic episode have a poor outcome with greater probability of death. Surgical interventions According to widely accepted guidelines, low-molecular-weight heparin (LMWH) in low doses, low-dose unfractionated heparin, or physical measures should be used in patients with cancer who face long-term immobilisation or low-risk surgical procedures (35). Patients with cancer

  • 16

    undergoing extensive surgery are at extremely high risk of postoperative venous thromboembolism. Accordingly, more intensive prophylactic regimens are needed, such as doses of LMWH about twice as high as suggested for low riskprocedures, adjusted-dose heparin, or oral anticoagulants (35). Once-daily injections of LMWH are at least as effective and safe as several injections of unfractionated heparin for prevention of postoperative venous thromboembolism in patients with cancer (36–38). In this setting, fondaparinux (a short-acting pentasaccharide) shows promise. In a trial to address the value of fondaparinux (2,5 mg once daily) for prevention of postoperative venous thromboembolism in patients undergoing major abdominal surgery, fondaparinux was more effective than enoxaparin in the subgroup of patients with cancer without increasing the risk of haemorrhage (39). Recent trials (22,40) have suggested that use of LMWH until 4 weeks after surgical intervention provides an additional thromboprophylactic effect without increasing the risk of haemorrhage.In patients who have had bleeding episodes or who are at high risk of bleeding, physical measures such as graduated compression stockings or external pneumatic compression should be used instead of pharmacological prophylaxis (41). According to the results of two randomised trials (42,43) in patients with cancer undergoing elective neurosurgery, the combination of LMWH (starting within 24 h of surgery) and graduated compression stockings is more effective than, and as safe as, elastic stockings alone for prevention of postoperative venous thromboembolism. Chemotherapy and radiotherapy In the only available study,(44) fixed low-dose warfarin (1 mg/day) for 6 weeks, followed by doses that maintained the international normalised ratio (INR) at 1·3–1·9was effective and safe for prevention of chemotherapy-induced thromboembolism in women with metastatic breast cancer. Whether this strategy or approaches that involve LMWH are effective and safe in other oncological settings remains to be shown.No adequate study has yet assessed the preventive value of antithrombotic strategies in patients undergoing radiotherapy. Central venous catheters Two randomised controlled studies (32,45) documented the benefit of fixed low-dose warfarin (1 mg once daily) in decreasingthe frequency of arm venous thrombosis related to indwelling central venous catheters. Subcutaneous dalteparin (2500 IU once daily for 90days) was also highly beneficial for prevention of arm thrombosis in patients with cancer who had venous access devices (33). However, three other clinical trials found no benefit from 1 mg warfarin daily (46,47) or 40 mg enoxaparin once daily (48) compared with no prophylaxis. Thus, neither low-dose warfarin nor prophylactic LMWH can be recommended as routine prophylaxis forpatients with cancer who have indwelling central venouslines (35). Treatment of venous thromboembolism Initial treatment Those patients who present with clinically unstable life threatening PEand who do not have contraindications tothrombolysis(such as ongoing or recent bleeding and brain metastasis) should promptly be administered drugs that have the potential to rapidly restore the patency of obstructed pulmonaryarteries (49-51). Among the drugs that have been shownto achieve a rapid and substantial lysis of fresh pulmonary emboli are urokinase, streptokinase and tissue-type plasminogenactivator (t-PA). The use of the last should been encouraged, because the administration of a loading dose of10 mg followed by the intravenous infusion of 90 mg produces in only 2 h the result that can be obtained by 12 - 24 h of infusion of urokinase or streptokinase (52,53). As compared to heparin alone, the administration of t-PA relieves patients’symptoms and improves prognosis to a greater extent (54-56). During the administration of t-PA or soon after its discontinuation heparin treatment should be implemented

  • 17

    (1). As far as the role of thrombolytic agents for acute deep-vein thrombosis (DVT) in cancer patients is concerned, available evidence is against their use except for very selected patients with massive ilio-femoral thrombosis who are at risk of limb gangrene and for whom a rapid venous decompression and flow restoration may be desirable (49-51). In patients with contraindications to catheter-directed thrombolysis, surgical thrombectomy can be considered (49-51). Anticoagulant therapy Except for selected patients requiring aggressive treatments, the large majority of cancer patients should be treated with therapeutic doses of low-molecular-weight heparin (LMWH), unfractionated heparin or fondaparinux (UFH) (49-51). Except for patients with severe renal failure, in whom UFH still represents the treatment of choice, in all other patients the VTE episode should be managed with LMWHs, as they represent the standard of long-term treatment (49-51). In the absenceof contraindications, LMWHs should be administered as soon as there is a high probability that venous thrombosis exists, even before the diagnostic algorithm is completed. LMWHs present a number of potential advantages overUFH, including a longer plasma half-life, an improved subcutaneous bioavailabilityand less variability in response to fixed doses (49). As a result of these pharmacokinetic properties, a stable and sustained anticoagulant effect is achieved when these drugs are administered subcutaneously in doses adjusted to bodyweight, once or twice daily, without laboratory monitoring(49).These compounds have the potential to greatly simplify the initial treatment of DVT and of selected low-risk patients with PE (9), making the treatment of suitable patients feasible in an outpatient setting (58-60). Treatment on home basis appears feasible and safe, which is particularly attractive for cancer patients, in whom prevention or reduction of hospital stay haste potential to improve the quality of life (61-62). According to the results of worldwide surveys, LMWHs are by far the most commonly used drugs for the initial treatment of VTE in cancer patients(63,64). Based on the results of many comparativetrials between UFH and LMWH for the initial treatment of patients with DVT that were conducted in the 1990s, LMWHs appear to be at least as effective and safe as UFH both in patients with and in those without cancer (65,66). It should be noted, however, that in these clinical trials cancer patients represented only 10 - 15% of the total population, as the majority of them were excluded because of their poor performance status. Of interest, the use of LMWH was associated with a significantly lower mortality, which was essentially dependent on the reduction of cancer-related mortality (65,66). It should not be forgotten that patients undergoing LMWH treatment require close monitoringof platelet count, as the risk of heparin-induced thrombocytopeniain medical patients treated with LMWHmay not be negligible (67). Although in clinical practice UFH has virtually been replaced by LMWHs, we think that several indications still remain for UFH, especially in cancer patients. The short half-life of intravenous UFH indeed allows for rapid reversal of anticoagulation in patients who begin to bleed or will require an invasive procedure. Also, the presence of severe renal insufficiency (i.e., creatinine clearance lower than 30 ml/min)makes it attractive to use as a short-acting drug that in addition can be timely monitored and possesses a specific antidote (the protamine sulfate). UFH is generally administered intravenously, whereas the use of nomograms assures that most patients will achieve the therapeutic range for the activated partial thromboplastin time (APTT), the most commonly recommended test for its monitoring (68).Subcutaneous heparin treatment has been suggested as an alternative to intravenous standard heparin, provided that the APTT is performed in order to achieve a full therapeutic effect(69). This modality of heparin administration, which is particularly desirable in those cancer patients who have difficult vein access, has been shown to be as effective and safe as LMWH for treatment of patients with acute VTE, including > 20% of cancer patients (70). In addition, fixed-dose unmonitored UFH isreasonableoption for out-of-hospital management in patients with severe renal impairment (71). Fondaparinux is the first drug of a new class of synthetic antithrombotic agents designed specifically for a single physiological target in the coagulation cascade and acts by indirect inhibition of factor Xa. This compound does not bind to platelet factor-4, which makes the development of immunethrombocytopenia extremely unlikely. In two large Phase III multicenter

  • 18

    clinical trials, involving the treatment of almost 4500 patients with clinically symptomatic DVT or PE (~10% with cancer), the once-daily subcutaneous administration of 7.5 mg of fondaparinux (5 mg in individuals weighing < 50 kg, 10 mg in those weighing > 100 kg) overlapped with and followed by vitamin K antagonists (VKA) was found to be at least as effective and safe as UFH or LMWH for the treatment of DVT or PE (72,73). However, when the analysis is confined to the only cancer patients randomized to the Matisse DVT study, recurrent VTE was found to be significantly more frequent in patients who had received an initial treatment with fondaparinux than in those allocated to the enoxaparin arm (74). In any case, fondaparinux is rarely employed for the initial treatment of VTE in cancer patients, because unlike LMWHs it is not (yet) registered for the long-termtreatment of thromboembolic disorders nor can it be followed by VKAs, whose efficacy is definitely lower than that of LMWHs (75). Alternative options: intracaval filters On average, patients with cancer present with major-often permanent-contraindications to anticoagulant treatment much more frequently than patients free from malignancy. In these patients, the only therapeutic option is the insertion of a (either retrievable or permanent) vena caval filter (49-51), which should be done without hesitation, as it has the potential to prevent (recurrent) PE events in patients with acute VTE. Indeed, prolonging life and/or improving its quality are invaluable goals to be achieved even in patients with poor condition. Our view is supported by findings from a Spanish registry. In a large number of patients with acute VTE who were managed without the insertion of a vena caval filter after a recent episode of major bleeding, the incidence of fatal bleeding and that of fatal PE in patients with cancer was 10 times as high as that observed in those without malignancy (76). As soon as the bleeding resolves, anticoagulation should be resumed; accordingly, the filter should be removed (77). Long-term anticoagulation While on VKA treatment, cancer patients with venous thrombosis have a risk of recurrent VTE and major bleeding that is higher than that reported in patients free from malignancies. The best evidence comes from a retrospective analysis of data from two large randomized clinical trials and two prospective cohort studies (78-80). Hutten et al. extractedthe rates of recurrent VTE and major bleedings in more than1300 patients receiving at least 3 months of oral anticoagulant therapy for an acute episode of DVT (78). The overall incidence of recurrent thrombosis in patients with cancer was 27.1/100 patient-years, versus 9.0/100 patient-years in those without cancer. The risk of major bleeding was 13.3/100 patient-years and 2.1/100 patients-years, respectively.Palaretiet al. comparedthe outcome of anticoagulation courses in 95 cancer patients and 733 patients without malignancy (79). Based on 744 patient-years of treatment and follow up, there was a trend toward a higher rate of thrombotic complications in cancer patients (6.8 vs 2.5%; relative risk = 2.5). The rate of major bleeding was significantly higher in cancer patients (5.4%) than in those without malignancy (0.9%; relative risk = 6.0). We conducted a prospective cohort study in 842 consecutive patients with DVT who were administered conventional anticoagulation, of whom 181 had cancer(80). The 12-month cumulative incidence of recurrent thromboembolism in cancer patients was 20.7 versus 6.8% in patients without cancer, for an age-adjustedhazard ratio of 3.2 (95% CI: 1.9 - 5.4). The 12-month cumulative incidence of major bleeding was 12.4% in patients with cancer and 4.9% in patients without cancer, for an age-adjusted hazard ratio of 2.2 (95% CI:1.2 - 4.1). In summary, cancer patients have a three- to four-fold higher risk of recurrent VTE during anticoagulant therapy than cancer-free patients, very likely as a consequence of the release of cancer procoagulants that are not inhibited by conventional anticoagulation. This risk correlates with the extent and the type of cancer (80). Recently, a stratification score has been developed and validated that has the potential to help clinicians predict the VTE recurrence risk and thus tailor treatment, improving clinical outcomes while minimizing costs (81). According to the results of three randomized clinical trials, LMWHs in full doses for the first month followed by a dose ranging from 50 to

  • 19

    100% of the initial regimen have the potential to provide a more effective antithrombotic regimen in cancer patients with venous thrombosis than the conventional treatment and are not associated with an increasedhemorrhagic risk (75,82,83), even in patients with disseminated cancer such as those with liver or brain metastases(84). In addition, LMWHs provide an anticoagulation that is easier to administer, more convenient and flexible and not influenced by nutrition problems or liver impairment (1). Thus, the long-term administration of LMWH should now be considered the treatment of choice in patients with metastatic disease and in those with conditions limiting the use of oral anticoagulants (49-51). On average, after discontinuation of antithrombotic treatment cancer patients with venous thrombosis present a risk for recurrences that is almost twice as high as that observed in patients free from malignancies (85-87). However, the risk of recurrence after stopping anticoagulation depends on the setting. For example, a patient who develops VTE after surgery for cancer has a low risk of recurrence if the cancer was completely resected. Like wise, a patient who develops VTE while receiving neo-adjuvant chemotherapy for potential micrometastatic cancer has a considerably lower risk of recurrence if anticoagulation is stopped after the chemotherapy is completed than a patient with metastatic disease who continues to receive combination chemotherapy. In principle, prolongation of anticoagulation should be considered for as long as the malignant disorder is active provided that it is not contraindicated. This decision should be frequently reassessed during patients’ follow up. Treatment of challenging situations The treatment of challenging situations has recently been addressed by the Subcommittee of the International Society of Thrombosis and Haemostasis (77). Management of recurrent VTE despite anticoagulation Recurrent VTE despite appropriate anticoagulation is common among cancer patients (88,89). Cancer patients with symptomatic recurrent VTE despite therapeutic anticoagulation with VKA should be switched to therapeutic weight adjusted doses of LMWH. Cancer patients with symptomatic recurrent VTE despite anticoagulation with LMWH should continue with LMWH at a higher dose, starting at an increase of ~ 25% of the current dose or increasing it back up to the therapeutic weight-adjusted dose if they have been receiving on-therapeutic dosing. All cancer patients with recurrent VTE despite anticoagulation should be reassessed 5-7 days after a dose escalation of their anticoagulant therapy. Patients with symptomatic improvement should continue the same dose of LMWH and resume their usual follow up. In patients without symptomatic improvement, the peak anti-Xa level can be used to estimate the dose of further escalation (77). Management of cancer-associated VTE in patients with thrombocytopenia Thrombosis is commonly diagnosed in patients with malignancy and thrombocytopenia. Full therapeutic doses of anticoagulation without platelet transfusion should be given in patients with platelet count ≥ 50 109/l. In patients with platelet count < 50 109/l, the recommended strategy diverges in patients with acute (< 1 month) from those withsubacute (1-3 months) or chronic (> 3 months) VTE. In the former group, full therapeutic doses of anticoagulation with platelet transfusion should be given to maintain a platelet count ≥ 50 109/l. If platelet transfusion is not possible or contraindicated, the insertion of a retrievable filter is suggested, as well as its removal when platelet count recovers and anticoagulation can be resumed. In the remaining groups, subtherapeutic orprophylactic doses of LMWH should be used in patients with platelet count of 25 - 50 109/l, whereas anticoagulation should be discontinued in patients with platelet count< 25 109/l (77).

  • 20

    Management of cancer-associated VTE patients who are bleeding A careful and thorough assessment of each bleeding episode, including identification of the source, its severity or impact, and reversibility should be done, as well as the usual supportive care with transfusion and surgical intervention to correct the bleeding source, whenever indicated and possible. Withholding anticoagulation in patients having a major or life-threateningbleeding episode is mandatory. Insertion of a caval filter is suggested for patients with acute or subacute VTE who are having a major or life-threatening bleeding episode, whereas it is discouraged in patients with chronic VTE. Once the bleeding resolves, anticoagulation should be initiated or resumed, and the retrievable caval filter (if inserted)should be removed (77). Potential of the novel direct oral anticoagulants As new categories of drugs have emerged that have the potential to replace conventional treatment for the initial and long-term treatment of VTE, major improvements are expected for the management of cancer patients with venous thrombosis. They include direct inhibitors of factor Xa (such as rivaroxaban, apixaban and edoxaban) and direct inhibitors of factorIIa (such as dabigatran etexilate). They possess several advantages over conventional drugs, including the inhibition of fibrin-bound Xa or thrombin, respectively, a dose-response that is more predictable because there is no binding to plasma proteins, and a lack of potential to produce immune thrombocytopenia. As a consequence of their pharmacokinetic and pharmacodynamic properties, they can be administered orally, in fixed doses, without laboratory monitoring. Based on available information coming from well-designed and conducted Phase III randomized clinical trials, they possess a morefavourable benefit-to-risk profile than the old compounds, make it possible to implement the treatment from the beginning and cover the whole spectrum of clinical presentations, including severe PE (90-95). However, for the time being their use in patients with cancer requires caution. Indeed, only a small minority of patients with cancer (consistently around 5%) were included in theabovementioned studies. More importantly, in these studies the comparator was warfarin, and not a LMWH, which represents the standard of treatment in cancer patients with thrombosis (49). Severe liver and renal dysfunctions, which contraindicate the use of all new oral compounds, are quite common in patients with cancer. There is uncertainty about the proper management of patients requiring emergency procedures and in those with thrombocytopenia. Finally, for the time being these drugs still lack proper antidotes. The novel anticoagulants should be investigated more carefully before routine usage in cancer patients. There are lessons to be learned from the studies conducted long time ago withfondaparinux, a parenteral potent inhibitor of factor Xa.Indeed, when the analysis of the Matisse DVT study, addressing the treatment of patients with DVT, was confined to theonlysubgroup of cancer patients, recurrent VTE was found to be significantly more frequent inpatients who had received an initial treatment with fondaparinux than in those allocated toenoxaparin (74). Management of incidentally detected isolated VTE Asymptomatic PE is a common finding in medical oncology dueto the routine use of modern computed tomography (CT) scanners for cancer staging. Although the clinical relevance of these incidental findings is unknown, based on the results of a few investigations conducted in recent years, they are likely to impact on both the incidence of recurrent VTE and on the overall prognosis to the same degree as the symptomatic findings (96-102). Accordingly, the most recent international guidelines recommend the same initial and long-term anticoagulation as for comparable patients with symptomatic PE (49). However, there is still uncertainty about the optimal management of patients with the incidental detection of isolated(i.e., not associated with DVT) sub-segmental PE. Indeed, whether the outcome of these patients is comparable to that of patients with symptomatic involvement of sub-segmental arterial vessels (103) is unknown, as is the accuracy of detecting on CT scans that were not specifically ordered to diagnose PE.

  • 21

    Higher risk of false positive diagnosis compared to patients with suspected PE cannot be excluded. In a series of70 patients diagnosed with subsegmental PE, this diagnosis was confirmed in only 51% by a reviewing radiologist (104). PE may not be acute but chronic. In a series of 65 cases of untreatedsubsegmental PE reported so far, none of these patients developed recurrent VTE (105). Finally, there is uncertainty about the outcome of patients with incidental PE who receive anticoagulants. In a cohort study of 51 patients with incidental PE, 5 (9.8%) patients developed major bleeding of which 2 cases were fatal (100). Thus, a careful evaluation should be individually done rather than giving full-dose anticoagulation to all cancer patients with the occasional detection of isolated subsegmental PE. An exception can be made for patients with the involvement of multiple sub-segmental vessels, as these patients are unlikely to differ from those with the involvement of more proximal arteries. While patients with incidentally detected proximal DVTshould be managed not differently from those with clinical symptoms, in patients with incidentally detected below-knee DVT, as well as in those with incidental thrombosis in other sites, such as portal, splenic or mesenteric veins, there is no evidencefavouring anticoagulation (49). Treatment of catheter-related thrombosis Central venous catheters are extensively used in patients with cancer to secure delivery of chemotherapy and to facilitate phlebotomy. Unfortunately, considerable morbidity can result from early complications or late sequelae, ranging from arterial puncture, pneumothorax and bloodstream infections to catheter-related thrombosis. Contemporary studies have shown that the incidence of symptomatic catheter-related thrombosis is ~ 5%, whereas the incidence of the asymptomatic one is higher, at 14 -- 18% (106). The significance and mechanisms of catheter design, material, insertion location and technique, position of the catheter tip and other risk factors in contributing to the development of catheter-related thrombosis are not well understood. Efforts to reduce thrombotic complications, involving flushing the catheter with heparinized solutions, the use of heparin-bonded catheters and systemic anticoagulant prophylaxis, have been largely ineffective (106). As published data and clinical experience suggest that catheter-related thrombosis is associated with a low risk of thrombosis recurrence and post-thrombotic syndrome (107), conservative treatment is recommended. A sensible approach is to remove the catheter only if central venous access is no longer required, the device is non-functional or defective or line-related sepsis is suspected or documented. Unless contraindicated, therapeutic anticoagulation should be given using either LMWH alone or LMWH followed by warfarin therapy. A short period of anticoagulation (3 - 5 days of LMWH) may even salvage some thrombosed catheters and obviate the need to remove and replace the line. Anticoagulation is recommended for a minimum of 3 months and while the catheter remains in place (49). Impact of antithrombotic drugs on cancer evolution Anticoagulant treatment of cancer patients, particularly those with lung cancer, has been reported to improve survival (108). Since then, studies conducted in animal tumour models have demonstrated that both UFH and LMWH interfere with various processes involved in tumour growth and metastasis (109). These processes might include fibrin formation, binding of heparin to angiogenic growth factors such as basic fibroblast growthfactor and vascular endothelial growth factor, modulation of tissue factor and other mechanisms (109). The evidence of lowered cancer mortality in patients on LMWH in comparison with those treated with UFH (65,66) has stimulated renewed interest in these agents as antineoplastic drugs. Five randomized studies have compared the long-term survival of cancer patients receiving conventional treatment with that of patients receiving a supplementary dose of LMWH in therapeutic or prophylactic doses (110-114). Two of these studies showed a favourable impact of the tested heparin on patients’ survival, this result being particularly evident in those with better prognosis (112,113). In the other two studies, post-hoc analysis showed a better survival in subgroups of patients with less

  • 22

    advanced disease (110,111). The fifth virtually excluded any appreciable advantages (114). Although a meta-analysis gave some support to this hypothesis (115), current evidence that LMWH reduces mortality is weak.

  • 23

    References

    1. Falanga A, Barbui T, Rickles FR, Levine MN. Guidelines for clotting studies in cancer patients. Thromb Haemost 1993;70:343–50 2. Rickles FR, Falanga A. Molecular basis for the relationship between thrombosis and cancer. Thromb Res 2001;102:V215–24 3. Costantini V, Zacharski LR. Fibrin and cancer. Thromb Haemost 1993;69:406–14 4. Falanga A, Donati MB. Pathogenesis of thrombosis in patients with malignancy. Int J Hematol 2001;73:137–44 5. Gale AJ, Gordon SG. Update on tumor cell procoagulant factors. Acta Haematol 2001;106: 25–32 6. Falanga A, Marchetti M, Vignoli A, Balducci D. Clotting mechanism and cancer: implications in thrombus formation and tumor progression. Clin Adv Hematol Oncol 2003;1:673–78 7. Falanga A, Iacoviello L, Evangelista V, et al. Loss of blast cell procoagulant activity and improvement of hemostatic variables in patients with acute promyelocytic leukemia given all-trans-retinoic acid. Blood 1995;86:1072–84 8. Kwaan HC, Keer HN. Fibrinolysis and cancer. Semin Thromb Haemost 1990;16:230–35 9. Varon D, Brill A. Platelets cross-talk with tumor cells. Haemostasis 2001;31(suppl 1):64–66 10. Grignani G, Maiolo A. Cytokines and hemostasis. Haematologica 2000;85:967–72 11. Contrino J, Hair G, Kreutzer DL, Rickles FR. In situ detection of tissue factor in vascular endothelial cells: correlation with the malignant phenotype of human breast disease. Nat Med 1996;2:209–15 12. Semeraro N, Colucci M. Tissue factor in health and disease. Thromb Haemost 1997;78: 759–64 13. Falanga A, Marchetti M, Evangelista V, et al. Polymorphonuclear leukocyte activation and hemostasis in patients with essential thrombocythemia and polycythemia vera. Blood 2000;96: 4261–66 14. Shoji M, Hancock WW, Abe K, et al. Activation of coagulation and angiogenesis in cancer. Immunohistochemical localization in situ of clotting proteins and VEGF in human cancers. Am J Pathol 1998;152: 399–411 15. Prandoni P, Piccioli A, Girolami A. Cancer and venous thromboembolism: an overview. Haematologica 1999;84:437–45 16. Shen VS, Pollak EW. Fatal pulmonary embolism in cancer patients: is heparin prophylaxis justified? South Med J 1980;73:841–43 17. Levitan N, Dowlati A, Remick SC, et al. Rates of initial and recurrent thromboembolic disease among patients with malignancy versus those without malignancy: risk analysis using Medicare claims data. Medicine (Baltimore) 1999;78:285–91

  • 24

    18. Otten HM, Prins MH. Venous thromboembolism and occult malignancy. Thromb Res 2001; 102:V187–94 19. Blom JW, Doggen CJ, Osanto S, Rosendaal FR. Malignancies, prothrombotic mutations, and the risk of venous thrombosis. JAMA 2005;293:715–22 20. Piccioli A, Prandoni P, Ewenstein BM, Goldhaber SZ. Cancer and venous thromboembolism. Am Heart J 1996;132:850–55 21. Huber O, Bounameaux H, Borst F, Rohner A. Postoperative pulmonary embolism after hospital discharge: an underestimated risk. Arch Surg 1992;127:310–13 22. Bergqvist D, Agnelli G, Cohen AT, et al. Duration of prophylaxis against venous thromboembolism with enoxaparin after surgery for cancer. N Engl J Med 2002;346:975–80. 23. Levine MN. Prevention of thrombotic disorders in cancer patients undergoing chemotherapy. Thromb Haemost 1997;78:133–36. 24. Cheruku R, Tapazoglou E, Ensley J, et al. The incidence and significance of thromboembolic complications in patients with high-grade gliomas. Cancer 1991;68:2621–24 25. Otten HMMB, Mathijssen J, ten Cate H, et al. Symptomatic venous thromboembolism in cancer patients treated with chemotherapy: an underestimated phenomenon. Arch Intern Med 2004;164:190–94 26. Goodnough LT, Saito A, Manni A, et al. Increased incidence of thromboembolism in stage IV breast cancer patients treated with a five-drug chemotherapy regimen: a study of 159 patients. Cancer 1984;54:1264–68 27. Prichard KI, Paterson AHG, Paul NA, et al. Increased thromboembolic complication with concurrent tamoxifen and chemotherapy in a randomized trial of adjuvant therapy for women with breast cancer. J Clin Oncol 1996;14:2731–37 28. Deitcher SR, Gomes MPV. The risk of venous thromboembolic disease associated with adjuvant hormone therapy for breast carcinoma. Cancer 2004;101:439–49 29. Goldhaber SZ. Tamoxifen: preventing breast cancer and placing the risk of deep vein thrombosis in perspective. Circulation 2005;111:539–41 30. Coombes RC, Hall E, Gibson LJ, et al. A randomized trial of exemestane after two to three years of tamoxifen therapy in postmenopausal women with primary breast cancer. N Engl J Med 2004;350:1081–92 31. Verso M, Agnelli G. Venous thromboembolism associated with long-term use of central venous catheters in cancer patients. J Clin Oncol 2003;21:3665–75 32. Bern MM, Lokich JJ, Wallach SR, et al. Very low dose of warfarin can prevent thrombosis in central venous catheters: a prospective trial. Ann Intern Med 1990;112:423–28 33. Monreal M, Alastrue A, Rull M, et al. Upper extremity deep venous thrombosis in cancer patients with venous access devicesprophylaxis with a low molecular weight heparin (fragmin). Thromb Haemost 1996;75:251–53 34. Levine MN, Lee AY, Kakkar AK. From Trousseau to targeted therapy: new insights and innovations in thrombosis and cancer. J Thromb Haemost 2003;1:1456–63

  • 25

    35. Geerts WH, Pineo GF, Heit JA, et al. Prevention of venous thromboembolism: the seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy. Chest 2004;126:338–400S 36. Bergqvist D, Burmark US, Flordal PA, et al. Low molecular weight heparin started before surgery as prophylaxis against deep vein thrombosis: 2500 versus 5000 XaI units in 2070 patients. Br J Surg 1995;82:496–501 37. Enoxacan Study group. Efficacy and safety of enoxaparin versus unfractionated heparin for prevention of deep vein thrombosis in elective cancer surgery: a double-blind randomized multicentre trial with venographic assessment. Br J Surg 1997;84:1099–103 38. McLeod RS, Geerts WH, Sniderman KW, et al. Subcutaneous heparin versus low-molecular-weight heparin as thromboprophylaxis in patients undergoing colorectal DVT prophylaxis trial: a randomized, double-blind trial. Ann Surg 2001;233:438–44 39. Agnelli G, Bergqvist D, Cohen A, et al. A randomized doubleblind study to compare the efficacy and safety of fondaparinux with dalteparin in the prevention of venous thromboembolism after high-risk abdominal surgery: the Pegasus study. In: Programs and abstracts of the XIX Congress of the International Society on Thrombosis and Haemostasis, Birmingham, UK; July 12–18, 2003. Malden: Blackwell Publishing, 2003 (abstr) 40. Rasmussen MS, Jorgensen LN, Wille-Jorgensen JP, et al. Prolonged prophylaxis with low molecular weight heparin (dalteparin) after major abdominal surgery: the Fame study. In: Programs and abstracts of the XIX Congress of the International Society on Thrombosis and Haemostasis, Birmingham, UK; July 12–18, 2003. Malden: Blackwell Publishing, 2003 (abstr) 41. Wells PS, Lensing AWA, Hirsh J. Graduated compression stockings in the prevention of postoperative venous thromboembolism: a meta-analysis. Arch Intern Med 1994;154:67–72 42. Nurmohamed MT, van Riel AM, Henkens CM, et al. Low molecular weight heparin and compression stockings in the prevention of venous thromboembolism in neurosurgery. Thromb Haemost 1996;75:233–38 43. Agnelli G, Piovella F, Buoncristiani P, et al. Enoxaparin plus compression stockings compared with compression stockings alone in the prevention of venous thromboembolism after elective neurosurgery. N Engl J Med 1998;339:80–85 44. Levine MN, Hirsh J, Gent M, et al. Double-blind randomised trial of very-low-dose warfarin for prevention of thromboembolism in stage IV breast cancer. Lancet 1994;343:886–89. 45. Bern MM, Bothe A Jr, Bistrian B, et al. Prophylaxis against central vein thrombosis with low-dose warfarin. Surgery 1986;99:216–21 46. Couban S, Goodyear M, Burnell M, et al. Randomized, placebo-controlled study of low-dose warfarin for the prevention of central venous catheter-associated thrombosis in patients with cancer. J Clin Oncol 2005 (in press) 47. Heaton DC, Han DY, Inder A. Minidose warfarin as prophylaxis for central vein catheter thrombosis. Intern Med 2002;32:84–88 48. Verso M, Agnelli G, Bertoglio S, et al. Enoxaparin for the prevention of venous thromboembolism associated with central vein catheter: a double -blind, placebo-controlled, randomized study in cancer patients J Clin Oncol 2005 (in press)

  • 26

    49. Kearon C, Akl EA, Comerota AJ, et al. Antithrombotic therapy for VTE disease: antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest2012;141(2 Suppl):e419-94S 50. Lyman GH, Khorana AA, Kuderer NM, et al. Venous thromboembolism prophylaxis and treatment in patients with cancer: American Society of Clinical Oncology Clinical Practice GuidelineUpdate. J Clin Oncol 2013;31:2189-204 51. Farge D, Debourdeau P, Beckers M,et al. International clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer.J Thromb Haemost 2013;11:56-70 52. Goldhaber SZ, Kessler CM, Heit J, et al.Randomised controlled trial of recombinant tissue plasminogen activator versus urokinase in the treatment of acute pulmonary embolism. Lancet1988;2:293-8 53. Meyer G, Sors H, Charbonnier B, et al.Effects of intravenous urokinase versus alteplase on total pulmonary resistance in acute massive pulmonary embolism: a European multicenter double-blind trial. J Am Coll Cardiol 1992;19:239-45 54. Dalla Volta S, Palla A, Santolicandro A, et al. Alteplase combined with heparin versus heparin in the treatment of acutepulmonary embolism. J Am Coll Cardiol 1992;20:520-6 55. Goldhaber SZ, Haire WD,Feldstein ML, et al. Alteplase versus heparin in acute pulmonary embolism:randomised trial assessing right ventricular function and pulmonary perfusion. Lancet 1993;341:507-11 56. Konstantinides S, Gebel A, Heusel G,et al. Heparin plus alteplase compared with heparin alone in patients with submassive pulmonary embolism. N Engl J Med 2002;347:1143-50 57. Hirsh J, Raschke R. Heparin and low-molecular-weight heparin. Chest 2004;126:188-203S 58. Aujesky D, Roy PM, Verschuren F, et al.Outpatient versus inpatient treatment for patients with acute pulmonary embolism:an international, open-label, randomised, non-inferiority trial. Lancet 2011;378:41-8 59. Zondag W, Kooiman J, Klok F, et al. Outpatient versus inpatient treatment in patients with pulmonary embolism: a meta-analysis. Eur Respir J 2012;42:134-44 60. Wells PS, Kovacs MJ, Bormanis J, et al.Expanding eligibility for outpatient treatment of deep venous thrombosis and pulmonary embolism with low-molecular-weight heparin:a comparison of patient self-injection with homecare injection. Arch Intern Med 1998;158:1809-12 61. Ageno W, Grimwood R, Limbiati S,et al. Home-treatment of deep vein thrombosis in patients with cancer. Haematologica 2005;90:220-4 62. Siragusa S, Arcara C, Malato A, et al. Home therapy for deep vein thrombosis and pulmonary embolism in cancer patients. Ann Oncol 2005;16:136-9S 63. Kakkar AK, Levine M, Pinedo HM,et al. Venous thrombosis in cancer patients: insights from the Front-line survey. Oncologist 2003;8:381-8

  • 27

    64. Kleinjan A, van Doormaal FF, Prins MH, et al. Limitations of screening for occult cancer in patients with idiopathic venous thromboembolism. Neth J Med 2012;70:311-17 65. Gould MK, Dembitzer AD, Doyle RL,et al. Low-molecular-weight heparins compared with unfractionated heparin for treatment of acute deep venous thrombosis. A meta-analysis of randomized, controlled trials. Ann Intern Med 1999;130:800-9 66. Dolovich LR, Ginsberg JS, Douketis JD,et al. A meta-analysis comparing low-molecular-weight heparins with unfractionated heparin in the treatment of venous thromboembolism. Arch Intern Med 2000;160:181-8 67. Prandoni P, Siragusa S, Girolami B,Fabris F. The incidence of heparin-induced thrombocytopenia in medical patients treated with low-molecular-weight heparin. Blood 2005;106:3049-54 68. Cruickshank MK, Levine MN, Hirsh J,et al. A standard heparin nomogram for the management of heparin therapy. Arch Intern Med 1991;151:333-7 69. Hommes DW, Bura A, Mazzolai L, et al. Subcutaneous heparin compared with continuous intravenous heparin administration in the initial treatment ofdeep vein thrombosis. A meta-analysis. Ann Intern Med 1992;116:279-84 70. Prandoni P, Carnovali M, Marchiori A. Subcutaneous adjusted-dose unfractionated heparin vs fixed-dose low-molecular-weight heparin in the initial treatment of venous thromboembolism. Arch Intern Med 2004;164:1077-83 71. Kearon C, Ginsberg JS, Julian JA, et al.Comparison of fixed-dose weight-adjusted unfractionated heparin and low-molecular-weight heparin for acute treatment of venous thromboembolism. JAMA 2006;296:935-42 72. Buller HR, Davidson BL, Decousus H,et al. Fondaparinux or enoxaparin for the initial treatment of symptomatic deep venous thrombosis. Ann Intern Med2004;140:867-73 73. The Matisse Investigators. Subcutaneous fondaparinux versus intravenous unfractionated heparin in the initial treatment of pulmonary embolism. N Engl J Med 2003;349:1695-702 74. van Doormaal FF, Raskob GE,Davidson BL, et al. Treatment of venous thromboembolism in patients with cancer: subgroup analysis of the Matisse clinical trials. Thromb Haemost 2009;101:762-9 75. Lee AY, Levine MN, Baker RI, 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;349:146-53 76. Nieto JA, De Tuesta AD, Marchena PJ, et al. Clinical outcome of patients with venous thromboembolism and recent major bleeding: findings from a prospective registry (RIETE). J Thromb Haemost 2005;3:703-9 77. Carrier M, Khorana A, Zwicker J, et al.Management of challenging cases of patients with cancer-associated thrombosis including recurrent thrombosis and bleeding: guidance from the SSC of the ISTH. J Thromb Haemost 2013;11:1760-5 78. Hutten B, Prins M, Gent M, et al.Incidence of recurrent thromboembolic and bleeding complications among patients with venous thromboembolism in relation to both malignancy and achieved international normalized ratio: a retrospective analysis. J Clin Oncol 2000;18:3078-83

  • 28

    79. Palareti G, Legnani C, Agnes L, et al. A comparison of the safety and efficacy of oral anticoagulation for the treatment of venous thromboembolic disease in patients with or without malignancy.Thromb Haemost 2000;84:805-10 80. Prandoni P, Lensing AWA, Piccioli A, et al. Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood 2002;100:3484-8 81. Louzada ML, Carrier M,Lazo-Langner A, et al. Development of a clinical prediction rule for risk stratification of recurrent venous thromboembolism in patients with cancer-associated venous thromboembolism. Circulation 2012;126:448-54 82. Meyer G, Marjanovic Z, Valcke J, et al.Comparison of low-molecular-weight heparin and warfarin for the secondary prevention of venous thromboembolism in patients with cancer. Arch Intern Med 2002;162:1729-35 83. Hull RD, Pineo GF, Brant RF, et al.Long-term low-molecular-weight heparin versus usual care in proximal-vein thrombosis patients with cancer. Am J Med 2006;119:1062-72 84. Monreal M, Zacharski L, Jimenez JA, et al. Fixed-dose low-molecular-weight heparin for secondary prevention of venous thromboembolism in patients with disseminated cancer: a prospective cohort study. J Thromb Haemost 2004;2:1311-15 85. Prandoni P, Lensing AWA, Cogo A,et al. The long-term clinical course of acute deep venous thrombosis. Ann Intern Med 1996;125:1-7 86. Hansson PO, Sorbo J, Eriksson H. Recurrent venous thromboembolism after deep vein thrombosis. Incidence and risk factors. Arch Intern Med 2000;1260:769-74 87. Heit JA, Mohr DN, Silverstein MD,et al. Predictors of recurrence after deep vein thrombosis and pulmonary embolism. A population-based cohort study. Arch Intern Med 2000;160:761-8 88. Carrier M, Le Gal G, Cho R, et al. Doseescalation of low molecular weight heparin to manage recurrent venous thromboemblic events despite systemic anticoagulation in cancer patients.J Thromb Haemost 2009;7:760-5 89. Luk C, Wells PS, Anderson D, kovacs MJ. Extended outpatient therapy with low molecular weight heparin for the treatment of recurrent venous thromboembolism despite warfarin therapy. Am J Med 2001;111:270-3 90. Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med2009;361:2342-52 91. Schulman S, Kakkar AK, Goldhaber SZ, et al. Treatment of acute venous thromboembolism with dabigatran or warfarin and pooled analysis. Circulation 2013, doi:10.1161/CIRCULATIONAHA.113.004450 92. The Einstein Investigators. Oral rivaroxaban for symptomatic venous thromboembolism. N Engl J Med 2010;363:2499-510 93. The Einstein Investigators. Oral rivaroxaban for the treatment of symptomatic pulmonary embolism.N Engl J Med 2012;366:1287-97

  • 29

    94. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med 2013;368:699-708 95. The Hokusai-VTE Investigators. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism. N Engl J Med 2013;369:1406-15 96. Menapace LA, Peterson DR, Berry A,et al. Symptomatic and incidental thromboembolism are both associated with mortality in pancreatic cancer.Thromb Haemost 2011;106:371-8 97. Sahut D’Izarn M, Caumont Prim A,Planquette B, et al. Risk factors and clinical outcome of unsuspected pulmonary embolism in cancer patients: a case-control study. J Thromb Haemost 2012;10:2032-8 98. Agnelli G, Verso M, Mandala` M, et al. A prospective study on survival in cancer patients with and without venous thromboembolism. Intern Emerg Med 2013;doi: 10.1007/s11739-013-0985-z 99. Connolly GC, Menapace L, Safadjou S,et al. Prevalence and clinical significance of incidental and clinically suspected venous thromboembolism in lung cancer patients.Clin Lung Cancer 2013;14:713-18 100. den Exter PL, Hooijer J, Dekkers OM,Huisman MV. Risk of recurrent venous thromboembolism and mortality in patients with cancer incidentally diagnosed with pulmonary embolism: a comparison with symptomatic patients. J Clin Oncol 2011;29:2405-9 101. Heidrich H, Konau E, Hesse P. Asymptomatic venous thrombosis in cancer patients. A problem often overlooked. Results of a retrospective and prospective study. Vasa 2009;38:160-6 102. O’Connell C, Razavi P, Ghalichi M, et al. Unsuspected pulmonary emboli adversely impact survival in patients with cancer undergoing routine staging MDCTscanning. J Thromb Haemost 2010;9:305-11. 103. den Exter PL, van Es J, Klok FA, et al.Risk profile and clinical outcome of symptomatic subsegmental acute pulmonary embolism. Blood 2013;122:1144-9 104. Pena E, Kimpton M, Dennie C, et al.Difference in interpretation of computedtomography pulmonary angiography diagnosis of subsegmental thrombosis in patients with suspected pulmonaryembolism. J Thromb Haemost2012;10:496-8 105. Donato AA, Khoche S, Santora J,Wagner B. Clinical outcomes in patients with isolated subsegmental pulmonary emboli diagnosed by multidetector CTpulmonary angiography. Thromb Res 2010;126:e266-70 106. Lee AY, Kamphuisen PW. Epidemiologyand prevention of catheter-related thrombosis in patients with cancer.J Thromb Haemost 2012;10:1491-9 107. Frank DA, Meuse J, Hirsch D, et al.The treatment and outcome of cancer patients with thrombosis on central venous catheters. J Thromb Thrombolysis 2000;10:271-5 108. Zacharski LR, Henderson WG, Rickles FR, et al. Effect of warfarin anticoagulation on survival in carcinoma of the lung, colon, head and neck, and prostate. Cancer 1984;53:2046-52109

  • 30

    109.Mousa SA. Anticoagulants in thrombosis and cancer: the missing link. Expert Rev Anticancer Ther 2002;2:227-33 110. Klerk CP, Smorenburg SM, Otten HM,et al. The effect of low molecular weight heparin on survival in patients with advanced malignancy. J Clin Oncol 2005;23:2130-5 111. Kakkar AK, Levine MN, Kadziola Z,et al. Low molecular weight heparin therapy with dalteparin, and survival in advanced cancer: the Fragmin Advanced Malignancy Outcome Study. J Clin Oncol 2004;22:1944-8 112. Lee AY, Rickles FR, Julian JA, et al. Randomized comparison of low molecular weight heparin and coumarin derivatives on the survival of patients with cancer and venous thromboembolism. J Clin Oncol 2005;23:2123-9 113. Altinbas M, Coskun HS, Er O, et al.A randomized clinical trial of combination chemotherapy with andwithout low-molecular-weight heparin in small cell lung cancer.J Thromb Haemost 2004;2:1266-71 114. van Doormaal FF, Di Nisio M,Otten HM, et al. Randomized trial of theeffect of the low molecular weight heparin nadroparin on survival in patients with cancer. J Clin Oncol 2011;29:2071-6 115. Lazo-Langner A, Goss GD, Spaans JN, Rodger MA. The effect of low-molecular-weight heparin on cancer survival. A systematic review and meta-analysis of randomized trials. J Thromb Haemost 2007;5:729-37

  • 31

    CHAPTER 3

    Recurrent venous thromboembolism and bleeding

    complications during anticoagulant treatment in

    patients with cancer and venous thrombosis.

    ●Prandoni P, ■Lensing AWA, ●Piccioli A, ●Bernardi E, ●Simioni P

    ●From the Department of Medical and Surgical Sciences, Second Chair ofInternal Medicine, and the Department of Clinical and Experimental Medicine,Clinical Epidemiology Group, Fifth Chair of Internal Medicine, University Hospital of Padua, Italy; ■Center for Vascular Medicine and the Department of Clinical Epidemiology, Academic Medical Center, University of Amsterdam, The Netherlands.

    Published in:

  • 32

    Blood 2002;100:3484-8

    Abstract

    A small proportion of patients with deep-vein thrombosis develop recurrent complications orbleeding during anticoagulant treatment. These complications may occur more frequently if these patients have concomitantcancer. This prospective follow-up study sought to determine whether in thrombosis patients those with cancer have a higher risk for recurrent venousthromboembolism or bleeding during anticoagulant treatment than those without cancer. Of the 842 included patients, 181 had known cancer at entry. The 12-month cumulative incidence of recurrent thromboembolism cancer patients was 20.7%(95% CI, 15.6%-25.8%) versus 6.8% (95%CI, 3.9%- 9.7%) in patients without cancer, for a hazard ratio of 3.2 (95% CI, 1.9-5.4)The 12-month cumulative incidence of major bleeding was 12.4% (95% CI, 6.5%-18.2%) in patients with cancer and 4.9% (95% CI, 2.5%-7.4%) in patients without cancer, for a hazard ratio of 2.2 (95% CI,1.2-4.1). Recurrence and bleeding were bothrelated to cancer severity and occurred predominantly during the first month of anticoagulant therapy but could not be explained by sub- or overanticoagulation.Cancer patients with venous thrombosis are more likely to develop recurrent thromboembolic complications and major bleeding during anticoagulant treatment than those without malignancy. These risks correlate with the extent of cancer. Possibilities for improvement using the current paradigms of anticoagulation seem limited and new treatment strategies should be developed. (Blood. 2002;100:3484-3488).

    Introduction

    Based on studies performed in the 1970s, it was established that patients with deep vein thrombosis (DVT) should receive anticoagulants for period of 3 months to prevent recurrent venous thromboembolism (VTE) (1). Further studies demonstrated that the presence of active canceror thrombophilic conditions were persisting risk factors for recurrent DVT (2-5). Subsequently, longer durations of anticoagulation became gradually adopted in these patient groups (6,7). Despite effective treatment, still approximately 5% to 7% of patients with DVT have recurrentvenousthromboembolic complications during heparinization or the subsequent 3-month period of oral anticoagulant therapy (6,7). Patients with DVT who also have cancer seem to be at a higher risk for recurrent venous thromboembolic complications during anticoagulation (8-10).This risk may be related to release of procoagulants by tumor cells that could make patients with cancer resistant to the usual intensities of anticoagulant drugs (11-13). Also the risk of anticoagulant-induced bleeding seems to be enhanced in patients with cancer, (9,14-16) although this is not consistently found (17,18). Little is known about whether the perceived higher risks for recurrent VTE and bleeding during anticoagulation apply to all patients with cancer or to specific groups. We analyzed the data of our cohort of consecutive patients with first episode of symptomatic DVT with specific attention to the period of anticoagulation. The aim of this analysis was to assess the relative risk for recurrent VTE and major bl