Venous thromboembolism and cancer: Guidelines of the Italian Association of Medical Oncology (AIOM)

https://doi.org/10.1016/j.critrevonc.2006.05.001Get rights and content

Abstract

Thromboembolic complications represent one of the most important cause of morbidity and mortality in cancer patients. Although several data have been published demonstrating the strong association between cancer and venous thromboembolism (VTE), there is poor perception, among oncologists, of the level of risk of thrombosis and of relevance of managing VTE in these patients. The Associazione Italiana di Oncologia Medica (AIOM) has provided some recommendations to direct clinical practice according to evidence-based data concerning cancer and VTE. In fact, we conducted an extensive literature review (1996–2005) to produce evidence-based recommendations to improve perceptions of the magnitude of this risk among Italian medical and surgical oncologists and alert on the new approaches to prophylaxis and treatment of VTE in cancer patients. Levels of evidence are given according to a five-point rating system, and similarly for each key recommendation a five-point rating system suggests if the evidence is strong and indicate that the benefits do, or do not, outweigh risks and burden.

Introduction

The strict two-way clinical relationship between cancer and VTE has been evaluated since Trousseau's time [1]. This author described the high incidence of venous thromboembolism (VTE) in a cohort of patients with gastrointestinal carcinoma. Thereafter it has been extensively recognised that VTE is a common complication of patients with malignancy [2].

Even though the association between cancer and hypercoagulability is well established, the pathogenesis of VTE in these patients is not entirely elucidated. While coagulation activation is encountered in up to 90% of cancer patients, only 4–15% of them develop deep venous thrombosis (DVT) and/or pulmonary embolism (PE) [3].

Thromboembolic disease can also act as an epiphenomenon of a hidden cancer. Several studies have demonstrated that patients with idiopathic thrombosis have a substantial likelihood to develop cancer [4]. It is noteworthy that local growth and metastatic dissemination of malignancies are influenced by the coagulation system [5], [6]. Tissue factor, thrombin, fibrin, platelets and other hemostatic components can all play a role in tumor progression. Chemotherapy and hormone therapy may further impair the hemostatic balance by causing alterations of blood vessel walls or regulatory proteins of the coagulation cascade [7], [8]. The incidence of VTE in solid tumors, and particularly for each histotype is difficult to establish because most clinical studies presented in current literature show a great variability based on the type of diagnostic procedure utilized to diagnose VTE, the most frequent tumor histotype, the medical or surgical procedures, and finally the presence or not of an indwelling central venous catheter, which in itself increases the risk of thrombosis of the axillary/subclavian vein. In addition, most data in the literature have been derived from retrospective and prospective studies not specifically designed to evaluate the issue. No studies have been able to definitively quantify the risk of developing a VTE for each histotype and/or site of the primary tumor.

Even in absence of symptomatic thrombosis most cancer patients present with abnormalities of coagulation tests, underlying a subclinical hypercoagulable state, including thrombin–antithrombin complex, prothrombin fragments 1 + 2, fibrinopeptide A, and D-dimer [9].

The developing of VTE is a multifactorial event involving several mechanisms. General mechanisms include inflammation due to necrosis or release of acute phase reactants and hemodynamic disorder such as stasis. Tumor-specific mechanisms include the capacity of tumor cells to activate the coagulation cascade by several ways. The tumor cells are able to interact with host blood cells, such as platelets, leukocytes, and endothelial cells, by releasing inflammatory cytokines (IL-1, TNF, VEGF) or by direct cell-to-cell interactions. As a final result, a downregulation of anticoagulant and an upregulation of procoagulant properties of these cells occur in the host, contributing to the general hypercoagulable condition of these subjects [9]. Cancer cells also produce a number of procoagulant substances including tissue factor [10], [11], [12] and cancer procoagulant [13], [14]. Therefore, tumor tissue can directly activate the clotting cascade, leading to thrombin generation and fibrin formation. It is now well demonstrated that tissue factor and other hemostatic proteins, including thrombin and fibrin, besides favouring thrombotic phenomenon, are also involved in the neoplastic dissemination and metastasis [10].

Albeit the pathophysiology of VTE in cancer has been extensively studied and VTE commonly occurs in patients with cancer, however most oncologists underestimate the prevalence of VTE and its negative impact on their patients. With this background in mind the Associazione Italiana di Oncologia Medica (AIOM) decided to draft recommendations for clinical practice in order to improve perceptions about the magnitude of VTE risk in patients with malignancy and to improve prophylaxis and treatment of VTE in cancer patients among Italian medical and surgical oncologists. For this purpose, we performed an extensive “Medline” and Cancerlit literature review (1996–2005) to produce evidence-based recommendations. Various combinations of search terms were used depending on the requirements of the database being searched. These terms included: “Thrombosis”, “Thromb*”, “Venous Thrombosis”, “Coagulation”, “Cancer”, “Tumor”, “prophylaxis*”, “therapy”, “Surgery”, “Chemotherapy”, “Hormonotherapy”, “Occult Cancer”, “Prognosis”, “Survival”, “Heparin”, “Coumarin”, “Warfarin*”. Relevant references in each article were scanned and we also did manual searches of abstracts from the annual meetings of the American Society of Hematology (1993–2004), American Society for Clinical Oncology (1993–2004), and European Haematology Association (1993–2004).

We focused on five distinct issues: (1) VTE and occult cancer; (2) prophylaxis of VTE in cancer surgery; (3) prophylaxis of VTE during chemotherapy or hormonal therapy; (4) prophylaxis of VTE and central venous catheters; (5) treatment of VTE in patients with cancer and VTE; (6) anticoagulation and prognosis of cancer patients.

For this purpose, the AIOM established a panel of experts in clinical oncology, clinical research, and clinical hematology. The panel included three academically affiliated and four community-based practicing hematology/oncology specialists. The panel used a systematic review of the evidence as its foundation for making recommendations. This process included a systematic weighting of the level of evidence and a systematic grading of the evidence for making a recommendation. In order to design a hierarchical grading system we gave greater weight to well-designed randomised controlled trials and meta-analyses and progressively less weight to studies with weaker internal validity. We considered with particular interest, and scored as high quality results those derived from studies with false positive rate ≤5% and false-negative rate ≤20%. Furthermore the concordant findings of controlled studies that have not been subject to meta-analysis were highly considered in order to score the levels of evidence. When evidence was lacking, the panel determined that it was appropriate to reach conclusions based on expert opinion. The strength of evidence and grade of recommendations are reported in Table 1, Table 2 and the scheme is identical to that in use by American Society of Clinical Oncology and European Society of Medical Oncology.

Section snippets

VTE and occult malignancy

Several studies have compared the incidence of cancer in patients with idiopathic VTE with those with VTE secondary to well-recognised risk factors [15], [16], [17], [18], [19], [20], [21], [22], [23], [24]. When considered overall, cancer was more commonly found in the follow-up of patients with idiopathic VTE or pulmonary embolism (PE) than in patients with known risk factors for such conditions. Prandoni et al. [18] published the results of a prospective study evaluating 154 patients with

Prophylaxis of VTE in surgical patients

A meta-analysis of perioperative prophylaxis demonstrated a reduced incidence of VTE in patients who received heparin prophylaxis (13.6%) compared to patients with no prophylaxis (30.6%) [28]. The therapeutic approaches used for the prevention of post-surgical VTE include compression stockings, low-dose unfractionated heparin (UFH) (5000 IU given daily every 8–12 h starting 1–2 h before the operation), and more recently the low-molecular weight heparins (LMWH) at a fixed single daily dose. The

Prophylaxis of VTE during chemotherapy or hormone therapy

Non-surgical cancer therapies also increase the risk of thromboembolic disease. The relation between VTE and chemotherapy has been most extensively investigated in patients with breast cancer. Levine et al. [43] demonstrated that chemotherapy contributes to thrombosis in patients with breast cancer. They performed a randomised trial comparing 12 weeks of chemo-hormone therapy (using cyclophosphamide, methotrexate, fluorouracil, vincristine, prednisone, doxorubicin, and tamoxifen) with 36 weeks

Prophylaxis of central venous catheters related VTE

In the last two decades two open label randomised clinical trials suggested a role of prophylaxis with warfarin or a LMWH in patients with a central venous catheter (CVC). Bern et al. [52] described the results obtained in 82 assessable patients at risk for thrombosis associated with chronic indwelling CVC who were prospectively and randomly assigned to receive or not to receive 1 mg fixed dose warfarin/day, beginning 3 days before catheter insertion and continuing for 90 days. Venograms were

Initial treatment

The standard initial treatment of an acute episode of VTE in cancer patients is similar to that adopted for patients without cancer. It consists in the administration of LMWH at a dose adjusted to body weight or UFH i.v. in continuous infusion. UFH is first administered as a bolus of 5000 IU, followed by continuous infusion, nearly 30,000 IU over 24 h, adjusted to achieve and maintain an activated partial thromboplastin time (aPTT) prolongation of 1.5–2.5 times the basal value.

Based on the results

Anticoagulation and prognosis of cancer

In the last 30 years basic research studies have indicated that: (i) tumor cells possess the capacity to activate blood coagulation by various mechanisms; (ii) this capacity parallels tumor cell malignant transformation; (iii) fibrin formation in tumor tissues, as a final product of the clotting cascade activation, is involved in tumor growth and dissemination. A number of experimental studies in vitro or in animal models the hypothesis that inhibiting blood coagulation may interfere with the

Reviewers

Guy Meyer, Doctor, Service de Pneumologie, soins intensifs, Hôpital Européen Georges-Pompidou 20, rue Leblanc, FR-75015 Paris, France.

Sylvia Haas, Professor, Institute for Experimental Oncology and Therapeutic Research, Technical University of Munich, Ismaninger Strasse 22, DE-81675 Munich, Germany.

Ajay K. Kakkar, Professor, Centre for Surgical Sciences, Thrombosis Research Institute, Emmanuel Kage Building, Manresa Road, London SW3 6LR, UK.

M. Mandalà received a MD from Rome Catholic University in 1995 and a fellowship in Oncology from Milan University in 2000. He is in training for a fellowship in Haematology at the Milan University. He is currently Clinical Assistant in the Division of Medical Oncology, Ospedali Riuniti Bergamo.

References (87)

  • A. Trousseau

    Phlegmasia alba dolens

    (1865)
  • A.K. Kakkar et al.

    Venous thrombosis in cancer patients: insight from the FRONTLINE survey

    Oncologist

    (2003)
  • A. Falanga et al.

    Deep vein thrombosis in cancer: the scale of the problem and approaches to management

    Ann Oncol

    (2005)
  • A. Piccioli et al.

    Risk of cancer in patients presenting with venous thromboembolism

  • J. Folkman

    Tumor angiogenesis and tissue factor

    Nat Med

    (1996)
  • C. Boccaccio et al.

    The MET oncogene drives a genetic programme linking cancer to haemostasis

    Nature

    (2005)
  • A.Y.Y. Lee et al.

    The thrombophilic state induced by therapeutic agents in the cancer patients

    Semin Thromb Hemost

    (1999)
  • H.C. Kwaan

    Cancer and hypercoagulability

  • A. Falanga et al.

    Pathophysiology of the thrombophilic state in cancer patients

    Semin Thromb Haemost

    (1999)
  • A.K. Kakkar et al.

    Tissue factor expression correlates histological grade in human pancreatic cancer

    Br J Surg

    (1995)
  • K. Hamada et al.

    Expression of tissue factor correlates with grade in human glioma

    Cancer

    (1996)
  • S.G. Gordon et al.

    A factor X-activating cysteine protease from malignant tissue

    J Clin Invest

    (1981)
  • A. Falanga et al.

    Isolation and characterization of cancer procoagulant: a cysteine protease from malignant tissue

    Biochemistry

    (1985)
  • H.T. Sorensen et al.

    The risk of a diagnosis of cancer after primary deep venous thrombosis or pulmonary embolism

    N Engl J Med

    (1998)
  • D. Aderka et al.

    Idiopathic deep vein thrombosis in an apparently healthy patient as a premonitory sign of occult cancer

    Cancer

    (1986)
  • M. Monreal et al.

    Occult cancer in patients with deep venous thrombosis. A systematic approach

    Cancer

    (1991)
  • P. Prandoni et al.

    Deep-vein thrombosis and the incidence of subsequent symptomatic cancer

    N Engl J Med

    (1992)
  • M. Monreal et al.

    Occult cancer in patients with venous thromboembolism: which patients, which cancers

    Thromb Haemost

    (1997)
  • M. Monreal et al.

    Occult cancer in patients with acute pulmonary embolism. A prospective study

    Chest

    (1993)
  • E.A. Bastounis et al.

    The incidence of occult cancer in patients with deep venous thrombosis: a prospective study

    J Intern Med

    (1996)
  • Z. Ahmed et al.

    Deep vein thrombosis as a predictor of cancer

    Angiology

    (1996)
  • R.J.K. Hettiarachchi et al.

    Undiagnosed malignancy in patients with deep vein thrombosis

    Cancer

    (1998)
  • S. Schulman et al.

    Incidence of cancer after prophylaxis with warfarin against recurrent venous thromboembolism

    N Engl J Med

    (2000)
  • J.T. Murchison et al.

    Excess risk of cancer in patients with primary venous thromboembolism: a national, population-based cohort study

    Br J Cancer

    (2004)
  • G.P. Clagett et al.

    Prevention of venous thromboembolism in general surgical patients. Results of meta-analysis

    Ann Surg

    (1988)
  • A.K. Kakkar et al.

    Prevention of venous thromboembolism in cancer patients

    Semin Thromb Hemost

    (1999)
  • D. Bergqvist et al.

    Low molecular weight heparin once daily compared with conventional low-dose heparin twice daily. A prospective double-blind multicentre trial on prevention of postoperative thrombosis

    Br J Surg

    (1986)
  • D. Bergqvist et al.

    Low molecular weight heparin given in the evening before surgery compared with conventional low-dose heparin in the prevention of thrombosis

    Br J Surg

    (1988)
  • M. Samama et al.

    Low molecular weight heparin compared with unfractionated heparin in the prevention of postoperative thrombosis

    Br J Surg

    (1988)
  • A. Leizorovicz et al.

    Prevention of postoperative deep vein thrombosis in general surgery: a multicenter double-blind study comparing two doses of logiparin and standard heparin

    Br J Surg

    (1991)
  • B. Boneu

    An international multicenter study: clivarin in the prevention of venous thromboembolism in patients undergoing general surgery

    Blood Coagul Fibrinolysis

    (1993)
  • 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)
  • Comparison of a low molecular weight heparin and unfractionated heparin for the prevention of deep vein thrombosis in patients undergoing abdominal surgery

    Br J Surg

    (1988)
  • Cited by (117)

    • Review: Inhibitory potential of low molecular weight Heparin in cell adhesion; emphasis on tumor metastasis

      2021, European Journal of Pharmacology
      Citation Excerpt :

      A vast range of heparin derivatives with various anticoagulant activities includes unfractionated Heparin (UFH), low molecular weight heparin (LMWH), and modified heparins are produced through Biochemical modifications of Heparin (Borsig, 2010b; Smorenburg and Van Noorden, 2001). As of today, for anticoagulation in cancer patients, low molecular weight heparin (LMWH) has remained a favored medication (Ay et al., 2017; Lyman et al., 2007; Mandala et al., 2006) The properties which cause an increase in usage of LMWH includes its antithrombotic effect, the prolonged half-life, and good bioavailability(Medicine, 1997; Prandoni et al., 1992). Evidence from the carryout research with experimental models in particular cell lines, tissues, and mice in several types of cancers have suggested that LMWHs possess antitumor and antimetastatic aside from their anticoagulant activity (Dimakakos et al., 2018; Mousa et al., 2009; Thomas et al., 2017).

    • Cancer-associated thrombosis

      2016, Disease-a-Month
    View all citing articles on Scopus

    M. Mandalà received a MD from Rome Catholic University in 1995 and a fellowship in Oncology from Milan University in 2000. He is in training for a fellowship in Haematology at the Milan University. He is currently Clinical Assistant in the Division of Medical Oncology, Ospedali Riuniti Bergamo.

    A. Falanga is Attending Hematologist in the Department of Hematology, Head of the Blood Coagulation Research Program at the Ospedali Riuniti in Bergamo, Italy, and appointed Professor at the School of Haematology, University “La Bicocca” Milan, Italy. She received her medical degree and board certification in internal medicine at the University of Naples, and her board certification in hematology at the University of Verona, Italy. Dr. Falanga worked as a Post-doctoral Fellow for 3 years in the Mario Negri Institute in Milan, Italy and subsequently at the University of Colorado School of Medicine, CO, USA for 2 years. From 2002, Dr. Falanga has been Elected Member of the Board of the Scientific and Standardization Committee of the International Society of Thrombosis and Hemostasis; she is currently Chairman of the Sub-committee on Hemostasis and Malignancy. She is involved in numerous professional societies, including the American Society of Hematology and the American Association for Cancer Research, and she is on the Council of the Italian Society of Thrombosis and Haemostasis.

    View full text