HSHD
HSHD
Edited by
Gregory Cheng
DEEP VEIN THROMBOSIS
Edited by Gregory Cheng
Deep Vein Thrombosis
Edited by Gregory Cheng
Published by InTech
Janeza Trdine 9, 51000 Rijeka, Croatia
As for readers, this license allows users to download, copy and build upon published
chapters even for commercial purposes, as long as the author and publisher are properly
credited, which ensures maximum dissemination and a wider impact of our publications.
Notice
Statements and opinions expressed in the chapters are these of the individual contributors
and not necessarily those of the editors or publisher. No responsibility is accepted for the
accuracy of information contained in the published chapters. The publisher assumes no
responsibility for any damage or injury to persons or property arising out of the use of any
materials, instructions, methods or ideas contained in the book.
Preface IX
1. Introduction
Deep vein thrombosis is a clinical challenge for doctors of all disciplines. It can complicate
the course of a disease but might also be encountered in the absence of precipitating
disorders. Thrombosis can take place in any section of the venous system, but arises most
frequently in the deep veins of the leg. Long-term morbidity due to post-thrombotic
syndrome is common and can be substantial. The major concern, however, is embolisation
of the thrombus to the lung, which can be fatal. Deep vein thrombosis is highly prevalent
and poses a burden on health economy. The disorder and its sequelae are also among the
best examples of preventable diseases. Relevant data for the frequency of deep vein
thrombosis derive from large community-based studies because they mainly reflect
symptomatic rather than asymptomatic disease. In a systematic review, the incidence of first
deep vein thrombosis in the general population was 0·5 per 1000 person-years.1 The
disorder is rare in children younger than 15 years,2,3 but its frequency increases with age,
with incidence per 1000 person-years of 1·8 at age 65–69 years and 3·1 at age 85–89 years.4
Two-thirds of first-time episodes of deep vein thrombosis are caused by risk factors,
including surgery, cancer, immobilisation, or admission for other reasons.5,6 Risk for first
deep vein thrombosis seems to be slightly higher in men than in women.6,9 In a population-
based cohort study, the age-adjusted incidence of first venous thromboembolism was 1·3
per 1000 person-years in men and 1·1 per 1000 person-years in women.2 It is noteworthy
that the risk for recurrence of this disorder is higher in men than in women.6,10
Antiphospholiped syndrome
Dyslipoproteinaemia
Nephrotic syndrome
Paroxysmal nocturnal haemoglobinuria
Myeloproliferative diseases
Behçet’s syndrome
Varicose veins
Superficial vein thrombosis
Congenital venous malformation
Long-distance travel
Prolonged bed rest
İmmobilisation
Limb paresis
Chronic care facility stay
Pregnancy/Puerperium
Oral contraceptives
Hormone replacement theraphy
Heparin-induced thrombocitopenia
Other drugs
Chemotheraphy
Tamoxifen
Thalidomide
Antipsychotics
Central Venous catheter
Vena cava filter
İntravenous drug abuse
Rudolph Virchow is recognized as the first person to link the development of VTE to the
presence of at least 1 of 3 conditions: venous stasis, vascular injury, and/or
hypercoagulability. 11 Each of these factors can alter the delicate hemostatic balance toward
hypercoagulability and development of thrombosis. Several aspects of surgery can be linked
to Virchow’s triad. Coleridge-Smith et al12 reported in 1990 that venous stasis occurs during
general surgery, with veins dilating 22% to 28% in patients undergoing general anesthesia
and surgery and up to 57% in those who also received an infusion of 1 L of saline during
surgery. The investigators suggested that it is this intraoperative venous distension that
underlies the risk for DVT in patients undergoing surgery. They suggested that the venous
distension is the result of loss of muscle tone that is caused by the muscle relaxants used
during surgery. Muscle paralysis resulting from regional anesthesia also can lead to venous
dilatation. These effects can be modified to some extent by the use of graduated
compression stockings during surgery.13 In a study of 40 patients undergoing surgery of the
abdomen or neck, the median vein diameter in the extremity studied was 2.6 mm at the
beginning of surgery in both the control and intervention groups (control group, n = 20;
median vein diameter, 2.6 mm; interquartile range [IQR], 2.1–3.3 mm; stocking group, n =20;
median vein diameter, 2.6 mm; IQR, 2.1–3.7 mm). This decreased to a median vein diameter
of 1.6 mm (IQR, 1.3–2.8 mm) after application of a stocking, whereas vein diameter
Risk Factors of Deep Vein Thrombosis 3
increased from 2.6 to 2.9 mm (IQR, 2.3– 4.0 mm) in the control group.13 Comerota et al14
found that in patients undergoing total hip replacement surgery, handling of soft tissue
(muscle) during surgery leads to venodilation, whereas bone manipulation leads to
venoconstriction. The venous dilatation that occurs during surgery causes cracks in the
endothelium, which provides a nidus for thrombosis as the blood coagulation system is
activated. The researchers also showed that pharmacologic control of venodilation during
surgery reduced postoperative DVT.14 Microscopic vessel wall damage, 15 such as that
demonstrated in patients undergoing hip and knee replacement surgeries, also contributes
to the development of VTE. 16,17 Tissue factor released from the blood vessel wall after
injury drives thrombus formation,18 which may help explain the increased risk of VTE in
patients undergoing surgery. The third factor in Virchow’s triad, hypercoagulability, is
linked to a number of factors, including certain genetic traits. Deficiencies of antithrombin,
protein C, or protein S, or mutations of factor V Leiden or factor II (prothrombin) G20210A
genes lead to hypercoagulable states.11 Although these genetic factors account for only a
small percentage of the total cases of VTE, more than half of all patients with juvenile or
idiopathic VTE have been identified with an inherited thrombophilic condition.11. Given
that VTE is the leading preventable cause of in-hospital deaths,19 every patient should be
screened before other lesser screens are performed (bedsores, risk of falls, nutritional
evaluation, and so forth). Stated another way— every patient deserves a proper history and
physical to uncover any possible factors that might increase their risk of a VTE.
In 1992, the Thromboembolic Risk Factors (THRIFT) Consensus Group identified acquired
risk factors for VTE.20 Sixteen years later, the most recent update of the American College of
Chest Physicians (ACCP) guidelines for VTE prophylaxis reveals essentially the same risk
factors for VTE as those identified by THRIFT, with the addition of a few new ones,
including acute medical illness, and the removal of smoking as a separate risk factor (Table
4 Deep Vein Thrombosis
1). 19 The incidence of VTE increases dramatically in tandem with the number of risk factors
identified in patients.11,21 Most hospitalized patients have at least one risk factor for VTE,
and the most recent ACCP review of VTE estimated that approximately 40% have 3 or more
risk factors.19 These include fracture (hip or leg), hip or knee replacement, major general
surgery, major trauma, and spinal cord injury,11 as well as a history of VTE,11
thrombophilia, 11 inflammatory bowel disease,22 postoperative infection, 19 and cancer.23
Bed rest for more than 72 hours,11,24 use of hormones,11 and impaired mobility11 are
additional risk factors. Many of these factors are not simple binary (ie, yes/no) risks. For
example, age is a significant risk factor, with the risk approximately doubling with each
decade beyond age 40.11,25 It is not sufficient to use a single age cut-off level to define high
or low risk.11 Similarly, the incidence of VTE increases with length of surgery.26,27 In
addition, Sugerman et al28 found higher rates of VTE in obese patients (mean body mass
index, 61) who also had venous stasis syndrome; a simple cut-off level based on a definition
of obesity would not capture this increased risk. In fact, Anderson and Spencer11 suggest
that the association of risk of VTE and weight alone is a weak one. As noted earlier,
hospitalized patients usually have at least 1 risk factor for VTE, and more than a third of
hospitalized patients have 3 risk factors or more.19 Risk factor weighting can be used to
calculate the risk for an individual patient, and the results may be used to determine several
aspects of prophylaxis, such as the length of prophylaxis (including out-of-hospital
prophylaxis), selection of prophylactic agent, timing of first dose, and the need for combined
use of physical and pharmacologic methods.
Risk assessment typically has taken 1 of 2 approaches, group risk assessment or individual
risk assessment. The group risk assessment approach assigns patients to one of a few broad
risk categories, whereas individual risk assessment seeks to define risk more accurately by
using individualized risk scores.19 The system recommended by the 2001 ACCP guidelines
used a group risk assessment in which the type of surgery (“major” vs “minor”), age
bracket, and presence of additional risk factors were used to assign patients to 1 of 4 risk
groups29; however, this was based on older studies, arbitrary age cut-off levels, and inexact
definitions.19 The ACCP has refined this recommendation with a newer one in which
patients are assigned 1 of 3 VTE risk levels based on type of surgery, patient mobility,
overall risk of bleeding, and moderate/high risk of VTE based on the presence of additional
risk factors 19 As the investigators note, this group risk assessment approach ignores the
substantial variability in patient-specific risk factors, but it does take into account what they
view as the principal risk factor (surgery vs acute medical illness). This approach is most
appropriate for patients who fit the criteria of the randomized clinical trials that were used
to develop the model; the investigators include a disclaimer for patient groups that have not
been included in clinical trials or for types of patients who have not been tested.19 However,
the group risk assessment approach recommended by the ACCP may not be appropriate for
all individual patients.30 Out-of-hospital prophylaxis is not addressed except for a few very
high risk groups (major cancer surgery, total joint replacement).19 It may be more
appropriate to use the individual risk assessment approach to identify and evaluate all
possible risk factors to determine the true extent of risk for a patient.30 The ACCP
guidelines, in fact, point out that “specific knowledge about each patient’s risk factors for
VTE” is an essential component of the decision-making process when prescribing
thromboprophylaxis. 19 Also, if many risk factors are present and a planned procedure is
Risk Factors of Deep Vein Thrombosis 5
based on a quality-of-life decision rather than a critical medical need, the patient may come
to a different decision about whether to proceed.30 A common misconception among
physicians is that individual risk assessment takes longer and is more cumbersome than
group risk assessment. However, individual assessment can be accomplished with, for
example, a simple assessment form that merely captures information from the history and
physical examination of the patient.
Among all patients with PE in the PIOPED II trial 94% had 1 or more of the following
assessed risk factors: bed rest within the last month of 3 days or more, travel within the last
month of 4 hours or more, surgery within 3 months, malignancy, past history of DVT or PE,
trauma of lower extremities or pelvis, central venous instrumentation within 3 months,
stroke, paresis or paralysis, heart failure or chronic obstructive pulmonary disease
(COPD).31 Immobilization of only 1 or 2 days may predispose to PE, and 65% of those who
were immobilized were immobilized for 2 weeks or less.32
confounding- associated risk factors, which make the risk of obesity inapparent. Previous
investigators used several indices of obesity including a BMI greater than 35 kg/m2 as well
as BMI 30 to 35 kg/m2,46 BMI 29 kg/m2 or greater,36 weight more than 20% of median
recommended weight for height,13 and for men, waist circumference 100 cm or greater.40 It
is likely that all patients diagnosed with obesity in the National Hospital Discharge Survey
database were obese, irrespective of the criteria used. However, some obese patients may
not have had a listed discharge diagnosis of obesity, and they would have been included in
the nonobese group. This situation would have tended to reduce the relative risk of obesity
in VTE. Various abnormalities of hemostasis have been described in obesity, in particular
increased plasminogen activator inhibitor-1 (PAI-1).47,48 Other abnormalities of coagulation
have been reported as well,48 including increased platelet activation,39 increased levels of
plasma fibrinogen, factor VII, factor VIII, and von Willebrand factor.49 Fibrinogen, factor
VIIc, and PAI-1 correlated with BMI.50 Regarding height, in the study of Swedish men,
those taller than 179 cm (5’ 10”) had a 1.5 times higher risk of VTE than men shorter than 172
cm.51 The Physicians’ Health Study of male physicians also showed that taller men had a
significantly increased risk of VTE.52
3. Air travel
The possibility of VTE after travel is not unique to air travel.53,54 Prolonged periods in
cramped quarters, irrespective of travel, can lead to PE.55 The term economy class
syndrome was introduced in 1988,56 but has since been replaced with flight-related DVT in
recognition that all travelers are at risk, irrespective of the class of travel57 Rates of
development of PE with air travel lasting 12 to 18 hours have been calculated as 2.6
PE/million travelers.58 With air travel of 8 hours or longer, 1.65/million passengers had
acute PE on arrival.59 With 6 to 8 hours of air travel the rate of acute PE on arrival was
0.25/million and among those who traveled for 6 hours or less none developed acute PE on
arrival.59 The trend showing increasing rates of PE with duration of travel is compelling,
but the incidence of DVT was about 3000 times higher in a prospective investigation.60 In a
prospective investigation of travelers who traveled for 10 hours or longer, 4 of 878 (0.5%)
developed PE and 5 of 878 (0.6%) developed DVT.60
4. Varicose veins
Varicose veins were found by some to be an agedependent risk factor for VTE.43 Among
patients aged 45 years the odds ratio for VTE was 4.2.43 In patients aged 60 years the odds
ratio was 1.9 and at aged 75 years, varicose veins were not associated with an increased risk
of VTE.43 However, others did not find varicose veins to be a risk factor for DVT61 or PE
found at autopsy.21
5. Oral contraceptives
Although the risk of VTE is higher among users of oral estrogen-containing contraceptives
than nonusers, 62,63 the absolute risk is low.64 An absolute risk of VTE of less than 1/10,000
patients/y increased to only 3 to 4/10,000 patients/y during the time oral contraceptives were
used.64 The relative risk for VTE in women using oral contraceptives containing 50 mg of
estrogen, compared with users of oral contraceptives that contained less than 50 mg was 1.5.65
The relative risk for VTE in women using oral contraceptives containing more than 50 mg of
Risk Factors of Deep Vein Thrombosis 7
estrogen, compared with users of oral contraceptives that contained less than 50 mg was 1.7.65
No difference in the risk of VTE was found with various levels of low doses of 20, 30, 40, and
50 mg/d.66 With doses of estrogen of 50 mg/d, the rate of VTE was 7.0/ 10,000 contraceptive
users/y and with more than 50 mg/d, the rate of VTE was 10.0/10,000 oral contraceptive
users/y.65 However, some found no appreciable difference in the relative risk of VTE in
relation to low or higher estrogen doses.67 Reports of the risk of VTE in relation to the
duration of use of oral contraceptives are inconsistent. Some showed relative risks increased as
the duration of use of estrogen-containing oral contraceptives increased.68 The relative risks
were 0.7 in women who used oral contraceptives for less than 1 year, 1.4 for those who used
oral contraceptives for 1 to 4 years and 1.8 in those who used it for 5 years or longer.68 Others
showed the opposite effect, with a decreasing relative risk with duration of use.66 The relative
risk for DVT or PE was 5.1 with use for less than 1 year, 2.5 with use for 1 to 5 years, and 2.1
with use for longer than 5 years.66 Some showed the risk to be unaffected by the duration of
use.67 A synergistic effect of oral contraceptives with obesity has been shown.69,71 The odds
ratio of DVT in obese women (BMI _30 kg/m2) who were users of oral contraceptives ranged
from 5.2 to 7.8 compared with obese women who did not use oral contraceptives37,69,71 and
among women with a BMI 35 kg/m2 or higher, the odds ratio was 3.1 compared with
similarly obese nonusers of oral contraceptives.71
6. Tamoxifen
Tamoxifen is a selective estrogen-receptor modulator used for treatment of breast cancer
and for prevention of breast cancer in high-risk patients.72,74 Among women with breast
cancer currently being treated with tamoxifen, compared with previous users or those who
never used it, the odds ratio was 7.1.74 Others found a lower odds ratio of 2.7.43 The odds
ratio for VTE in women at high risk of breast cancer who received tamoxifen to prevent
breast cancer was 2.1.73 Others found a hazard ratio of 1.63.72
8.5 Hyperhomocysteinemia
Homocysteine is an amino acid formed during the metabolism of methionine and may be
elevated secondary to inherited defects in two enzymes that are part of the conversion of
homocysteine to cysteine. The two enzymes involved are N5,N10–methylene
tetrahydrofolate reductase (MTHFR) or cystathionine beta-synthase.
Hyperhomocysteinemia has been shown to increase the risk of atherosclerosis,
atherothrombosis, and venous thrombosis. Elevated plasma homocysteine levels cause
various dysfunctions of endothelial cells leading to a prothrombotic state.
Hypercoagulable syndromes include inherited and acquired thrombophilias. The former is
discussed in detail in the article by Weitz in this issue. The latter includes the
antiphospholipid syndrome, heparin-induced thrombocytopenia, acquired
dysfibrinogenemia, myeloproliferative disorders, and malignancy. Myeloproliferative
disorders and malignancy are described elsewhere in this article. Regarding the
antiphospholipid syndrome, antiphospholipid antibodies are associated with both arterial
and venous thrombosis.89 The most commonly detected subgroups of antiphospholipid
antibodies are lupus anticoagulant antibodies, anticardiolipin antibodies and anti-b2-
glycoprotein I antibodies.90 DVT, the most common manifestation of the antiphospholipid
syndrome, occurs in 29% to 55% of patients with the syndrome, and about half of these
patients have pulmonary emboli.91,92 The risk of heparin-associated thrombocytopenia is
more duration related than dose related. Heparin-associated thrombocytopenia occurs more
frequently with unfractionated heparin when used for an extended duration than with
LMWH used for an extended duration.93 When used for prophylaxis, there was a higher
prevalence of heparin-associated thrombocytopenia inthose receiving unfractionated
heparin (1.6%, 57 of 3463) than in those receiving LMWH (0.6%, 23 of 3714).93 However,
treatment resulted in only a small difference in the prevalence of heparinassociated
thrombocytopenia comparing unfractionated heparin (0.9%, 22 of 2321) with LMWH (0.6%,
18 of 3126).93 Acquired dysfibrinogenemia occurs most often in patients with severe liver
disease.94 The impairment of the fibrinogen is a structural defect caused by an increased
carbohydrate content impairing the polymerization of the fibrin, depending on the degree of
abnormality of the fibrinogen molecule.94
9. Heart failure
Congestive heart failure (CHF) is considered amajor risk factor for VTE.13,41,61,95,96 Among
patients with established CHF, those with lower ejection fractions had a higher risk of
thromboembolic event.97,98 However, some investigators did not evaluate CHF among the
risk factors for VTE.99 The reported frequency of PE in patients with heart failure has ranged
widely from 0.9% to 39% of patients. 13,97,98,100,101 The reported frequency of DVT in patients
with CHF also ranged widely from 10% to 59%.13,41,61 The largest investigation was from the
National Hospital Discharge Survey.102 Among 58,873,000 patients hospitalized with heart
failure in short-stay hospitals from 1979 to 2003, 1.63% had VTE (relative risk 5 1.47).102 The
relative risk for VTE was highest in patients less than 40 years old (relative risk 5 6.91). Some
showed the lower the ejection fraction, the greater the risk of VTE.103 Among 755,807 adults
older than 20 years with heart failure who died from 1980 to 1998, PE was listed as the cause
of death in 20,387 (2.7%).104 Assuming that the accuracy of death certificates was only
26.7%,105 the rate of death from PE in these patients may have been as high as 10.1%.
10 Deep Vein Thrombosis
Therefore, the estimated death rate from PE in patients who died with heart failure was 3%
to 10%. CHF seems to be a stronger risk factor in women. Dries and colleagues97 reported a
higher proportion of PE in women (24%) compared with men (14%). We too showed a
higher relative risk of PE and of DVT in women with CHF than in men.102 Although these
data seemcompelling, multivariate logistic analysis failed to identify CHF as an independent
risk factor for DVT or PE.43 However, it was a risk factor for postmortem VTE that was not a
cause of death.43 In one study of pediatric patients with dilated cardiomyopathy awaiting
transplant the incidence of pulmonary embolism was 13.9% 106.
Heart failure is the second most common risk factor for VTE in hospitalized patients, as
shown in ENDORSE.107
10. COPD
Hospitalized patients with exacerbations of COPD, when routinely evaluated, showed PE in
25% to 29%.108,109From 1979 to 2003, 58,392,000 adults older than 20 years were hospitalized
with COPD in short-stay hospitals in the United States.110 PE was diagnosed in 381,000
(0.65%) and DVT in 632,000 (1.08%).110 The relative risk for PE in adults hospitalized with
COPD was 1.92 and for DVT it was 1.30. Among those aged 20 to 39 years with COPD, the
relative risk for PE was 5.34. Among patients with COPD aged 40 to 59 years, the relative risk
for PE decreased to 2.02, and among patients aged 60 to 79 years the relative risk for PE was
1.23.110 The relative risk for DVT was also higher in patients with COPD aged 20 to 39 years
(relative risk 5 2.58) than in patients aged 40 years or older (relative risk 0.92-1.17, depending
on age).110 In young adults, other risk factors in combination with COPD are uncommon, so
the contribution of COPD to the risk of PE becomes more apparent than in older patients.
Although these data strongly suggest that COPD is a risk factor for PE and DVT, multivariate
logistic analysis did not identify it as an independent risk factor.43 Others, with univariate
analysis, did not identify COPD as a risk factor.61
Neuhaus et al. 111 found pulmonary emboli in 27% of 66 autopsies performed in patients
who had respiratory failure (not only as a decompensation of COPD) and died after
admission to a Respiratory Intensive Care Unit.
The largest study was conducted by Schonhofer and Kohler 112 on a population of 196
patients admitted to a respiratory intensive care unit. The authors found a DVT rate of
10.7% as assessed by US. The majority (86%) of cases were asymptomatic and, interestingly,
almost all major clinical variables (such as age, weight, severity of dyspnea, lung function,
situation of blood gases) failed to predict patients who were more likely to develop DVT.
11. Stroke
There is considerable evidence that in spinal cord injury patients interruption of neurologic
impulses and the ensuing paralysis cause profound metabolic changes in blood vessels
accountable for venous thrombosis.
Vascular adaptations to inactivity and muscle atrophy, rather than the effect of a
nonworking leg-muscle pump and sympathetic denervation, cause thrombosis, indicating
that thrombosis established through venous incompetence cannot be reversed by
anticoagulation alone.
Risk Factors of Deep Vein Thrombosis 11
Spinal cord injuries with paralysis result in an immobile state with retardation of the blood
flow caused by the relaxation of muscle and the atony of blood vessels. It is not surprising
that spinal cord injuries are frequently complicated by the development of venous
thrombosis, which is inevitably linked to hospitalization, immobilization, vein wall damage,
stasis, and hypercoagulability. Deep vein thrombosis and pulmonary emboli remain the
major complications in spinal cord injuries below the C2 through T12 vertebrae associated
with motor complete or motor nonfunctional paralysis. 113,114,115,116,117,118,119 Two surprising
findings set spinal cord injury apart from other risk factors for venous thrombosis: incidence
of leg DVT and pulmonary embolism in spinal cord injury is three times higher than in the
general population.
Patients with stroke are at particular risk of developing DVT and PE because of limb
paralysis, prolonged bed rest, and increased prothrombotic activity.120 Among 14,109,000
patients with ischemic stroke hospitalized in short-stay hospitals from 1979 to 2003, VTE
was diagnosed in 165,000 (1.17%).121 Among 1,606,000 patients with hemorrhagic stroke,
the incidence of VTE was higher (1.93%).
Among patients with ischemic stroke who died from 1980 to 1998, PE was the listed cause of
death in 11,101 of 2,000,963 (0.55%).122 Based on an assumed sensitivity of death certificates
for fatal PE of 26.7% to 37.2%,105,123 the corrected rate of fatal PE was 1.5% to 2.1%. Death
rates from PE among patients with ischemic stroke decreased from 1980 to 1998, suggesting
effective use of antithrombotic prophylaxis.
12. Cancer
Cancer is a major risk factor of venous thromboembolism (VTE) 124,125 as defined by deep-
vein thrombosis (DVT) – including central venous catheter (CVC) related thrombosis – or
pulmonaryembolism (PE), which occur in 4 to 20% of cancer patients 126,127.
12.2 Histology
In certain types of cancer, higher rates of VTE are found in some histological subtypes
compared with others. For example, in patients with non-small-cell lung cancer, 9.9% of
those with adenocarcinoma subtype develop VTE in the first 6 months after diagnosis
compared with 7.7% with squamous cell carcinoma (HR 1.9, CI 1.7–2.1) [Link] and
colon cancer patients, the type of histology does not predict for the incidence of cancer-
associated VTE, but VTE-associated mortality rates are higher in patients with certain
histological subtypes 141,143.
12.4 Chemotherapy
Chemotherapy is one of the most important factors in VTE risk stratification of cancer
patients. Large population-based studies in groups of pooled cancer patients have
demonstrated a significantly increased risk in patients receiving chemotherapy. Heit et al
used a population-based study of patients with a new diagnosis of VTE, 23% of which had a
diagnosis of active malignancy, to demonstrate a significantly increased risk of VTE in those
on chemotherapy (OR 6.5, CI 2.11–20) 146.
Studies in specific types of cancer and with specific antineoplastic agents have also
supported the role of chemotherapy in predicting the risk of cancer-associated VTE. Two
prospective studies of breast cancer patients demonstrated that the risk of VTE in patients
receiving chemotherapy in addition to tamoxifen or surgery increased two- to seven-fold
147,148. A recent meta-analysis of breast cancer patients revealed that use of adjuvant
hormonal therapy was associated with a 1.5–7-fold increased risk of VTE 149.
Risk Factors of Deep Vein Thrombosis 13
12.5 Surgery
Surgery is a well-known risk factor for development of VTE in patients without cancer. The
incidence of DVT in cancer patients undergoing general surgery is estimated at 37%
compared with 20% in patients without cancer 150. Factors related to immobility, tissue
destruction and venous stasis are likely to be related to the increased risk of VTE after
surgery.
carcinoma of the pancreas (4.3%) and the lowest incidences were in patients with carcinoma
of the bladder and carcinoma of the lip, oral cavity, or pharynx (<0.6% to 1.0%). Incidences
with cancer were not age dependent.157 Myeloproliferative diseaseand lymphoma were
associated with relative risks for VTE of 2.9 and 2.5, respectively157 Leukemia was
associated with a lower relative risk (1.7). Based on death certificates from 1980 to 1998
among patients who died with cancer, PE was the listed cause of death in 0.21%.158
Adjustment of the data for the frailty of the diagnosis of fatal PE based on death certificates
indicated a likely range of 0.31% to 1.97%.158
13. Pregnancy
Pregnancy-associated DVT based on data from the National Hospital Discharge Survey was
diagnosed in 93,000 of 80,798,000 women (0.12%) from 1979 to 1999.151 The rate of
pregnancyassociated DVT (vaginal delivery and cesarean section) increased from 1982 to
1999, although the rate of nonpregnancy-associated DVT decreased for most of this period.
Some showed the rate of pregnancy-associated DVT was twice the rate of nonpregnancy-
associated DVT.159 A 6-fold increase in the rate of thromboembolism during pregnancy and
the puerperium compared with nonpregnant women has been reported by others.160
Although the rate of pregnancy-associated DVT was higher than the rate of
nonpregnancyassociated DVT, the rate of pregnancyassociated PE was lower than
Pathophysiology of venous thromboembolism during Pregnancy:
Increased venous distensibility and capacity, with a resultant reduction in the velocity of blood
flow in the lower limbs, are demonstrable from the first trimester of pregnancy162,163. These
changes are compounded by a 20–25% increase in the overall circulatory volume during
pregnancy164. Obstruction of the inferior vena cava by the enlarging gravid uterus may also
result in increased stasis165. Compression of the left iliac vein by the right iliac artery as they
cross 166 may explain the preponderance of left leg DVT during pregnancy 161,167.
Altered levels of coagulation factors have been described both during pregnancy and
postpartum. Hypercoagulability is thought to be promoted by increases in coagulation
factors such as fibrinogen, von Willebrand factor, and factor VIII:C 168,169–171, as well as by
decreases in natural inhibitors of coagulation such as protein S 172 and the development of
an acquired resistance to the endogenous anticoagulant, activated protein C 173. In addition,
a reduction in global fibrinolytic activity has been described during pregnancy 174, perhaps
as a consequence of increases in the levels of plasminogen activator inhibitor 1 (PAI 1) and
plasminogen activator inhibitor 2 (PAI 2) 174–176, the latter being produced by the placenta.
Exogenous risk factors also appear to determine the thrombotic risk associated with
pregnancy. In a retrospective cohort study of unselected consecutive patients with
confirmed pregnancy-related venous thromboembolism, approximately two-thirds of
patients had an identifiable acquired risk factor (for example, age over 35 years, intercurrent
illness, immobility, increased parity or caesarean section) 177.
The reason for this difference is unknown and could reflect difference of the natural history
of DVT in pregnancy. It also could reflect a reluctance to expose pregnant women to
ionizing radiation associated with imaging for PE, resulting in a decreased frequency of
diagnosis of PE. The rate of pregnancy-associated DVT was higher among women aged 35
to 44 years than in younger women. The rate of pregnancyassociated DVT among black
Risk Factors of Deep Vein Thrombosis 15
women was higher than among white women.159,178,179 DVT was more frequent among
women who underwent cesarean section (104/100,000/y) than those who underwent
vaginal delivery (47/ 100,000/y).159 VTE in pregnancy is discussed in detail in the article by
Marik elsewhere in this issue.
mutation, and increased levels of factor VIII.204 Based on data from the National Hospital
Discharge Survey, among 4,927,000 hospitalized patients with chronic alcoholic liver disease
from 1979 to 2006, the prevalence of VTE was 0.6% and among 4,565,000 hospitalized
patients with chronic nonalcoholic liver disease it was 0.9%.201 The prevalence of VTE was
higher in those with chronic alcoholic liver disease than with nonalcoholic liver disease, but
the difference was small and of no clinical consequence.201
Both showed a lower prevalence of VTE than in hospitalized patients with most other
medical diseases. It may be that both chronic alcoholic liver disease and chronic
nonalcoholic liver disease have protective antithrombotic mechanisms although the
mechanisms differ.
16.3 Hypothyroidism
Among 19,519,000 hospitalized patients with a diagnosis of hypothyroidism from 1979 to
2005, 119,000 (0.61%) had PE (relative risk 5 1.64).205 DVT was diagnosed in 1.36% of
hypothyroid patients (relative risk 5 1.62).205 The relative risk for PE in patients with
hypothyroidism was highest in patients younger than 40 years (relative risk 5 3.99) and the
relative risk for DVT was also highest in patients younger than 40 years (relative risk 5 2.25).
Hyperthyroidism was not associated with an increased risk for VTE (relative risk 5 0.98).
17. Sepsis
Initiation of coagulation takes place when TF is exposed, such as by fibroblasts, when there
is tissue damage or by cytokine-stimulated monocytes and endothelial cells216, as in sepsis.
While TF is the major initiator of coagulation, endotoxin, foreign bodies, and negatively
charged particles may initiate coagulation via contact system activation. TF binds to factor
18 Deep Vein Thrombosis
VIIa, and this complex (TF:VIIa) may then activate factor X and factor IX217. Factor Xa,
associated with factor Va, forms the prothrombinase complex, which subsequently turns
prothrombin into thrombin.
The relationship between coagulation and inflammation is complex and, as yet, not
completely understood. It is known that blood clotting not only leads to fibrin deposition
and platelet activation, but it also results in vascular cell activation, which contributes to
leukocyte activation218. On the other hand, inflammation can induce TF expression in
monocytes, via nuclear factor kappa-B (NF-kB) activation, thus initiating coagulation216.
Examples of this interaction are readily seen. First, leukocytes are found at relatively high
concentrations in venous thrombi, and leukocytes and activated platelets can form rosettes
mediated by P-selectin expression on the surface of the activated platelet 219,220.
These microscopic observations are probably elicited from the actions of thrombin, which
can activate platelets and endothelium, increasing the surface expression of P-selectin221,222.
P- electin is the primary initial mediator of leukocyte-endothelial cell rolling and is critical
for leukocyte adhesion. Second, TF:VIIa and factor Xa have been shown to activate cells and
generate responses similar to those mediated by thrombin218. Third, GAG and TM
expression on cell surfaces are inhibited by inflammatory cytokines 223,224,225,226 and
lipopolysaccharide (LPS)227, thus blocking the augmentation of AT action by GAG, and APC
formation by TM.
18. References
[1] Fowkes FJI, Price JF, Fowkes FGR. Incidence of diagnosed deep vein thrombosis in the
general population: systematic review. Eur J Vasc Endovasc Surg 2003; 25: 1–5.
[2] Heit JA, Silverstein MD, Mohr DN, et al. The epidemiology of venous thromboembolism
in the community. Thromb Haemost 2001; 86: 452–63.
[3] Stein PD, Patel KC, Kalra NK, et al. Deep venous thrombosis in a general hospital. Chest
2002; 122: 960–62.
[4] Kniffin WD, Baron JA, Barrett J, Birkmeyer JD, Anderson FA Jr. The epidemiology of
diagnosed pulmonary embolism and deep venous thrombosis in the elderly. Arch
Intern Med 1994; 154: 861–66.
[5] Heit JA, O’Fallon WM, Petterson TM, et al. Relative impact of risk factors for deep vein
thrombosis and pulmonary embolism: a population-based study. Arch Intern Med
2002; 162: 1245–48.
[6] Cushman M, Tsai AW, White RH, et al. Deep vein thrombosis and pulmonary embolism
in two cohorts: the longitudinal investigation of thromboembolism etiology. Am J
Med 2004; 117: 19–25.
[7] Patel RK, Lambie J, Bonner L, Arya R. Venous thromboembolism in the black
population. Arch Intern Med 2004; 164: 1348–49.
[8] Klatsky AL, Armstrong MA, Poggi J. Risk of pulmonary embolism and/or deep venous
thrombosis in Asian-Americans. Am J Cardiol 2000; 85: 1334–37.
[9] Anderson FA, Wheeler HB, Goldberg RJ, et al. A population-based perspective of the
hospital incidence and case-fatality rates of deep vein thrombosis and pulmonary
embolism: the Worcester DVT Study. Arch Intern Med 1991; 151: 933–38.
Risk Factors of Deep Vein Thrombosis 19
[10] Kyrle PA, Minar E, Bialonczyk C, Hirschl M, Weltermann A, Eichinger S. The risk of
recurrent venous thromboembolism in men and women. N Engl J Med 2004; 350:
2558–63.
[11] Anderson FA Jr, Spencer FA. Risk factors for venous thromboembolism. Circulation
2003;107:I9 –I16.
[12] Coleridge-Smith PD, Hasty JH, Scurr JH. Venous stasis and vein lumen changes during
surgery. Br J Surg 1990;77:1055–9.
[13] Coleridge Smith PD, Hasty JH, Scurr JH. Deep vein thrombosis: effect of graduated
compression stockings on distension of the deep veins of the calf. Br J Surg
1991;78:724–6.
[14] Comerota AJ, Stewart GJ, Alburger PD, et al. Operative venodilation: a previously
unsuspected factor in the cause of postoperative deep vein thrombosis. Surgery
1989;106:301–9.
[15] Comerota AJ, Stewart GJ, White JV. Combined dihydroergotamine and heparin
prophylaxis of postoperative deep vein thrombosis: proposed mechanism of action.
Am J Surg 1985;150:39–44.
[16] Stamatakis JD, Kakkar VV, Sagar S, et al. Femoral vein thrombosis and total hip
replacement. BMJ 1977;2:223–5.
[17] Mammen EF. Pathogenesis of venous thrombosis. Chest 1992;102:640S–4S.
[18] Mackman N. Role of tissue factor in hemostasis, thrombosis, and vascular development.
Arterioscler Thromb Vasc Biol 2004;24:1015–22.
[19] Geerts WH, Bergqvist D, Pineo GF, et al. Prevention of venous thromboembolism:
American College of Chest Physicians Evidence-Based Clinical Practice Guidelines.
Chest 2008;133:381S– 453S.
[20] Thromboembolic Risk Factors (THRIFT) Consensus Group. Risk of and prophylaxis for
venous thromboembolism in hospital patients. BMJ 1992;305:567–74.
[21] Wheeler HB. Diagnosis of deep vein thrombosis. Review of clinical evaluation and
impedance plethysmography. Am J Surg 1985;150:7–13.
[22] Nguyen GC, Sam J. Rising prevalence of venous thromboembolism and its impact on
mortality among hospitalized inflammatory bowel disease patients. Am J
Gastroenterol 2008;103:2272– 80.
[23] Kröger K, Weiland D, Ose C, et al. Risk factors for venous thromboembolic events in
cancer patients. Ann Oncol 2006;17:297–303.
[24] Kovacevich GJ, Gaich SA, Lavin JP, et al. The prevalence of thromboembolic events
among women with extended bed rest prescribed as part of the treatment for
premature labor or preterm premature rupture of membranes. Am J Obstet
Gynecol 2000;182:1089 –92.
[25] Anderson FA Jr, Wheeler HB, Goldberg RJ, et al. A population-based perspective of the
hospital incidence and case-fatality rates of deep vein thrombosis and pulmonary
embolism. The Worcester DVT Study. Arch Intern Med 1991;151:933– 8.
[26] Borow M, Goldson H. Postoperative venous thrombosis. Evaluation of five methods of
treatment. Am J Surg 1981;141:245–51.
[27] Borow M, Goldson HJ. Prevention of postoperative deep venous thrombosis and
pulmonary emboli with combined modalities. Am Surg 1983;49:599–605.
20 Deep Vein Thrombosis
[28] Sugerman HJ, Sugerman EL, Wolfe L, et al. Risks and benefits of gastric bypass in
morbidly obese patients with severe venous stasis disease. Ann Surg 2001;234:41–
6.
[29] Geerts WH, Heit JA, Clagett GP, et al. Prevention of venous thromboembolism. Chest
2001;119:132S–75S.
[30] Caprini JA. Thrombosis risk assessment as a guide to quality patient care. Dis Mon
2005;51:70–8.
[31] Stein PD, Beemath A, Matta F, et al. Clinical characteristics of patient with acute
pulmonary embolism: data from PIOPED II. Am J Med 2007;120:871e9.
[32] Stein PD, Terrin ML, Hales CA, et al. Clinical, laboratory, roentgenographic and
electrocardiographic findings in patients with acute pulmonary embolism and no
pre-existing cardiac or pulmonary disease. Chest 1991;100:598e603
[33] Heit JA, Silverstein MD, Mohr DN, et al. The epidemiology of venous
thromboembolism in the community. Thromb Haemost 2001;86:452e63.
[34] Anderson FA Jr, Wheeler HB, Goldberg RJ, et al. The prevalence of risk factors for
venous thromboembolism among hospital patients. Arch Intern Med
1992;152:1660e4.
[35] Hedley AA, Ogden CL, Johnson CL, et al. Prevalence of overweight and obesity among
US children, adolescents, and adults, 1999e2002. JAMA 2004;291:2847e50.
[36] Goldhaber SZ, Grodstein F, Stampfer MJ, et al. A prospective study of risk factors for
pulmonary embolism in women. JAMA 1997;277:642e5.
[37] Abdollahi M, Cushman M, Rosendaal FR. Obesity: risk of venous thrombosis and the
interaction with coagulation factor levels and oral contraceptive use. Thromb
Haemost 2003;89:493e8.
[38] Coon WW, Coller FA. Some epidemiologic considerations of thromboembolism. Surg
Gynecol Obstet 1959;109:487e501.
[39] Basili S, Pacini G, Guagnano MT, et al. Insulin resistance as a determinant of platelet
activation in obese women. J Am Coll Cardiol 2006;48:2531e8.
[40] Hansson PO, Eriksson H, Welin L, et al. Smoking and abdominal obesity: risk factors
for venous thromboembolism among middle-aged men: “the study of men born in
1913”. Arch Intern Med 1999;159:1886e90.
[41] Samama MM. An epidemiologic study of risk factors for deep vein thrombosis in
medical outpatients: the Sirius study. Arch Intern Med 2000;160: 3415e20.
[42] Kabrhel C, Varraso R, Goldhaber SZ, et al. Prospective study of BMI and the risk of
pulmonary embolism in women. Obesity (Silver Spring) 2009; 17:2040e6.
[43] Heit JA, Silverstein MD, Mohr DN, et al. Risk factors for deep vein thrombosis and
pulmonary embolism: a population-based case-control study. Arch Intern Med
2000;160:809e15.
[44] US Department of Health and Human Services. Public Health Service, National Center
for Health Statistics National Hospital Discharge Survey 1979-2006 Multi-year
Public-Use Data File Documentation. Available at:
[Link] Accessed April 28,
2010.
[45] Stein PD, Beemath A, Olson RE. Obesity as a risk factor in venous thromboembolism.
Am J Med 2005;118:978e80.
Risk Factors of Deep Vein Thrombosis 21
[46] Farmer RD, Lawrenson RA, Todd JC, et al. A comparison of the risks of venous
thromboembolic disease in association with different combined oral contraceptives.
Br J Clin Pharmacol 2000;49:580e90.
[47] Pannaciulli N, De Mitrio V, Marino R, et al. Effect of glucose tolerance status on PAI-1
plasma levels in overweight and obese subjects. Obes Res 2002; 10:717e25.
[48] De Pergola G, Pannacciulli N. Coagulation and fibrinolysis abnormalities in obesity. J
Endocrinol Invest 2002;25:899e904.
[49] Mertens I, Van Gaal LF. Obesity, haemostasis and the fibrinolytic system. Obes Rev
2002;3:85e101.
[50] Bara L, Nicaud V, Tiret L, et al. Expression of a paternal history of premature
myocardial infarction on fibrinogen, factor VIIC and PAI-1 in European
offspringethe EARS study. European Atherosclerosis Research Study Group.
Thromb Haemost 1994;71:434e40.
[51] Rosengren A, Frede´n M, Hansson PO, et al. Psychosocial factors and venous
thromboembolism: a long-term follow-up study of Swedish men. J Thromb
Haemost 2008;6:558e64.
[52] Glynn RJ, Rosner B. Comparison of risk factors for the competing risks of coronary
heart disease, stroke, and venous thromboembolism. Am J Epidemiol
2005;162:975e82.
[53] Homans J. Thrombosis of the deep leg veins due to prolonged sitting. N Engl J Med
1954;250:148e9.
[54] Tardy B, Page Y, Zeni F, et al. Phlebitis following travel. Presse Med 1993;22:811e4.
[55] Simpson K. Shelter deaths from pulmonary embolism. Lancet 1940;2:744.
[56] Cruickshank JM, Gorlin R, Jennett B. Air travel and thrombotic episodes: the economy
class syndrome. Lancet 1988;2:497e8.
[57] Collins J. Thromboembolic disease related to air travel: what you need to know. Semin
Roentgenol 2005;40:1e2.
[58] Hertzberg SR, Roy S, Hollis G, et al. Acute symptomatic pulmonary embolism
associated with long haul air travel to Sydney. Vasc Med 2003;8:21e3.
[59] Perez-Rodriguez E, Jimenez D, Diaz G, et al. Incidence of air travel-related pulmonary
embolism at the Madrid-Barajas airport. Arch Intern Med 2003; 163:2766e70.
[60] Hughes RJ, Hopkins RJ, Hill S, et al. Frequency of venous thromboembolism in low to
moderate risk long distance air travellers: the New Zealand Air Traveller’s
Thrombosis (NZATT) study. Lancet 2003;362:2039e44.
[61] Cogo A, Bernardi E, Prandoni P, et al. Acquired risk factors for deep-vein thrombosis in
symptomatic outpatients. Arch Intern Med 1994;154:164e8.
[62] Lewis MA. The epidemiology of oral contraceptive use: a critical review of the studies
on oral contraceptives and the health of young women. Am J Obstet Gynecol
1998;179:1086e97.
[63] Realini JP, Goldzieher JW. Oral contraceptives and cardiovascular disease: a critique of
the epidemiologic studies. Am J Obstet Gynecol 1985;152:729e98.
[64] Vandenbroucke JP, Rosing J, BloemenkampK W, et al. Oral contraceptives and the risk
of venous thrombosis. N Engl J Med 2001;344:1527e35.
[65] Gerstman BB, Piper JM, Tomita DK, et al. Oral contraceptive estrogen dose and the risk
of deep venous thromboembolic disease. Am J Epidemiol 1991;133:32e7.
22 Deep Vein Thrombosis
[85] Nicolaes GAF, Dahlback B. Activated protein C resistance (FVLeiden) and thrombosis:
Factor V mutations causing hypercoagulable states, Hematol Oncol Clin N Am.
2003. 17: 37–61.
[86] Koppelman SJ, Hackeng TM, Sixma JJ et al. Inhibition of the intrinsic factor X activating
complex by protein S: Evidence for specifi c binding of protein S to factor VIII,
Blood. 1995. 86:1062–1071.
[87] Allaart CF, Poort SR, Rosendaal FR et al. Increased risk of venous thrombosis in carriers
of hereditary protein C defi ciency defect, Lancet. 1993. 341: 134–138.
[88] Silver D, Vouyouka A. The caput medusae of hypercoagulability, J Vasc Surg. 2000. 31:
396–495.
[89] Greaves M. Antiphospholipid antibodies and thrombosis. Lancet 1999;353:1348e53.
[90] Levine JS, Branch DW, Rauch J. The antiphospholipid syndrome. N Engl J Med
2002;346: 752e63.
[91] Asherson RA, Khamashta MA, Ordi-Ros J, et [Link] “primary” antiphospholipid
syndrome: major clinical and serological features. Medicine (Baltimore)
1989;68:366e74.
[92] Vianna JL, Khamashta MA, Ordi-Ros J, et al. Comparison of the primary and secondary
antiphospholipid syndrome: a European Multicenter Study of 114 patients. Am J
Med 1994;96:3e9.
[93] Stein PD, Hull RD, Matta F, et al. Incidence of thrombocytopenia in hospitalized
patients with venous thromboembolism. Am J Med 2009;122: 919e30.
[94] Brick W, Burgess R, Faguet GB. Dysfibrinogenemia. WebMD. Available at:
[104] Beemath A, Skaf E, Stein PD. Pulmonary embolism as a cause of death in adults who
died with heart failure. Am J Cardiol 2006;98:1073e5.
[105] Attems J, Arbes S, Bohm G, et al. The clinical diagnostic accuracy rate regarding the
immediate cause of death in a hospitalized geriatric population; an autopsy study
of 1594 patients. Wien Med Wochenschr 2004;154:159e62.
[106] Hsu DT, Addonizio LJ, Hordof AJ, Gersony WM. Acute pulmonary embolism in
pediatric patients awaiting heart transplantation. J Am Coll Cardiol 1991;17:1621–5.
[107] Cohen AT, Tapson VF, Bergmann JF, et al. Venous thromboembolism risk and
prophylaxis in the acute hospital care setting (ENDORSE study): a multinational
cross-sectional study. Lancet 2008;371: 387-94.
[108] Mispelaere D, Glerant JC, Audebert M, et al. Pulmonary embolism and sibilant types
of chronic obstructive pulmonary disease decompensations. Rev Mal Respir
2002;19:415e23.
[109] Tillie-Leblond I, Marquette CH, Perez T, et al. Pulmonary embolism in patients with
unexplained exacerbation of chronic obstructive pulmonary disease: prevalence
and risk factors. Ann Intern Med 2006;144:390e6.
[110] Stein PD, Beemath A, Meyers FA, et al. Pulmonary embolism and deep venous
thrombosis in patients hospitalized with chronic obstructive pulmonary disease. J
Cardiovasc Med 2007;8:253e7.
[111] Neuhaus A, Bentz RR, Weg JG. Pulmonary embolism in respiratory failure. Chest
1978;73(4):460 –5.
[112] Schonhofer B, Kohler D. Prevalence of deep-vein thrombosis of the leg in patients with
acute exacerbation of Chronic Obstructive Pulmonary Disease. Respiration
1998;65:173– 7.
[113] Gunduz S, Ogur E, Mohur H, et al: Deep vein thrombosis in spinal cord injured
patients. Paraplegia 31:606410, 1993
[114] Hull R Venous thromboembolism in spinal cord injury patients Chest 102:658-662,
1992
[115] Merli GJ: Management of deep vein thrombosis in spinal cord injury. Chest 102652-
657, 1992
[116] Myllynen P, Kammonen M, Rokkanen P, et al: Deep venous thrombosis and
pulmonary embolism in patients with acute spinal cord injury: A comparison with
non-paralyzed patients immobilized due to spinal fractures. J Trauma 25:541-543,
1985
[117] Prasad DK, Banerjee AK, Howard H Incidence of deep vein thrombosis and the effect
of the pneumatic compression if the calf in elderly hemiplegics. Age Aging 11:424,
1982
[118] Waring WP, Karunas RS: Acute spinal cord injuries and the incidence of clinically
occurring thromboembolic disease. Paraplegia 29:8-16, 1991
[119] Yao JST Deep vein thrombosis in spinal cord-injured patients. Evaluation and
assessment. Chest 102:645-648, 1992
[120] Harvey RL. Prevention of venous thromboembolism after stroke. Topics Stroke Rehab
2003;10:61e9.
[121] Skaf E, Stein PD, Beemath A, et al. Venous thromboembolism in patients with ischemic
and hemorrhagic stroke. Am J Cardiol 2005;96:1731e3.
Risk Factors of Deep Vein Thrombosis 25
[122] Skaf E, Stein PD, Beemath A, et al. Fatal pulmonary embolism and stroke. Am J
Cardiol 2006;97:1776e7.
[123] Dismuke SE, VanderZwaag R. Accuracy and epidemiological implications of the death
certificate diagnosis of pulmonary embolism. J Chronic Dis 1984;37:67e73.
[124] Heit JA, Silverstein MD, Mohr DN, Petterson TM, O’Fallon WM, Melton 3rd LJ. Risk
factors for deep vein thrombosis and pulmonary embolism: a populationbased
case-control study. Arch Intern Med 2000;160(6):809–15.
[125] Spencer FA, Lessard D, Emery C, Reed G, Goldberg RJ. Venous thromboembolism in
the outpatient setting. Arch Intern Med 2007;167(14):1471–5.
[126] Levitan N, Dowlati A, Remick SC, Tahsildar HI, Sivinski LD, Beyth R, et al. Rates
ofinitial and recurrent thromboembolic disease among patients with malignancy
versus those without malignancy. Risk analysis using Medicare claims data.
Medicine (Baltimore) 1999;78(5):285–91.
[127] Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH. Frequency, risk
factors, and trends for venous thromboembolism among hospitalized cancer
patients. Cancer 2007;110(10):2339–46.
[128] Stein PD, Beemath A, Meyers FA, et al. Incidence of venous thromboembolism in
patients hospitalized with cancer. Am J Med 2006;119:60–8.
[129] Khorana AA, Francis CW, Culakova E, et al. Thromboembolism in hospitalized
neutropenic cancer patients. J Clin Oncol 2006;24:484–90.
[130] Sallah S, Wan JY, Nguyen NP. Venous thrombosis in patients with solid tumors:
determination of frequency and characteristics. Thromb Haemost 2002;87:575–9.
[131] 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.
[132] Stein PD, Beemath A, Meyers FA, et al. Incidence of venous thromboembolism in
patients hospitalized with cancer. Am J Med 2006;119:60–8.
[133] Chew HK, Wun T, Harvey D, et al. Incidence of venous thromboembolism and its
effect on survival among patients with common cancers. Arch Intern Med
2006;166:458–64.
[134] Numico G, Garrone O, Dongiovanni V, et al. Prospective evaluation of major vascular
events in patients with nonsmall cell lung carcinoma treated with cisplatin and
gemcitabine. Cancer 2005;103:994–9.
[135] Weijl NI, Rutten MF, Zwinderman AH, et al. Thromboembolic events during
chemotherapy for germ cell cancer: a cohort study and review of the literature. J
Clin Oncol 2000;18:2169–78.
[136] White RH, Chew HK, Zhou H, et al. Incidence of venous thromboembolism in the year
before the diagnosis of cancer in 528,693 adults. Arch Intern Med 2005;165:1782–7.
[137] Blom JW, Doggen CJ, Osanto S, et al. Malignancies, prothrombotic mutations, and the
risk of venous thrombosis. JAMA 2005;293:715–22.
[138] Sallah S, Wan JY, Nguyen NP. Venous thrombosis in patients with solid tumors:
determination of frequency and characteristics. Thromb Haemost 2002;87:575–9.
[139] Khorana AA, Francis CW, Culakova E, et al. Thromboembolism in hospitalized
neutropenic cancer patients. J Clin Oncol 2006;24:484–90.
[140] Rodriguez AO, Wun T, Chew H, et al. Venous thromboembolism in ovarian cancer.
Gynecol Oncol 2007;105:784–90.
26 Deep Vein Thrombosis
[141] Tateo S, Mereu L, Salamano S, et al. Ovarian cancer and venous thromboembolic risk.
Gynecol Oncol 2005;99:119–25.
[142] Chew HK, Wun T, Harvey DJ, et al. Incidence of venous thromboembolism and the
impact on survival in breast cancer patients. J Clin Oncol 2007;25:70–6.
[143] Chew HK, Davies AM,Wun T, et al. The incidence of venous thromboembolism
among patients with primary lung cancer. J Thromb Haemost 2008;6:601–8.
[144] Alcalay A, Wun T, Khatri V, et al. Venous thromboembolism in patients with
colorectal cancer: incidence and effect on survival. J Clin Oncol 2006;24:1112–8.
[145] Mandala M, Reni M, Cascinu S, et al. Venous thromboembolism predicts poor
prognosis in irresectable pancreatic cancer patients. Ann Oncol 2007;18:1660–5.
[146] Chew HK, Wun T, Harvey D, et al. Incidence of venous thromboembolism and its
effect on survival among patients with common cancers. Arch Intern Med
2006;166:458–64.
[147] Heit JA, Silverstein MD, Mohr DN, et al. Risk factors for deep vein thrombosis and
pulmonary embolism: a populationbased case-control study. Arch Intern Med
2000;160:809–15.
[148] Pritchard KI, Paterson AH, Paul NA, et al. Increased thromboembolic complications
with concurrent tamoxifen and chemotherapy in a randomized trial of adjuvant
therapy for women with breast cancer. National Cancer Institute of Canada Clinical
Trials Group Breast Cancer Site Group. J Clin Oncol 1996;14:2731–7.
[149] Fisher B, Dignam J, Wolmark N, et al. Tamoxifen and chemotherapy for lymph node-
negative, estrogen receptor-positive breast cancer. J Natl Cancer Inst 1997;89:1673–
82.
[150] Deitcher SR, Gomes MP. The risk of venous thromboembolic disease associated with
adjuvant hormone therapy for breast carcinoma: a systematic review. Cancer
2004;101:439–49.
[151] Agnelli G, Caprini JA. The prophylaxis of venous thrombosis in patients with cancer
undergoing major abdominal surgery: emerging options. J Surg Oncol 2007;96:265–
72.
[152] 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.
[153] Khorana AA, Francis CW, Culakova E, et al. Risk factors for chemotherapy-associated
venous thromboembolism in a prospective observational study. Cancer
2005;104:2822–9.
[154] Edgington TS, Mackman N, Brand K, et al. The structural biology of expression and
function of tissue factor. Thromb Haemost 1991;66:67–79.
[155] Nemerson Y. Tissue factor and hemostasis. Blood 1988;71:1–8.
[156] Nemerson Y. Tissue factor: then and now. Thromb Haemost 1995;74:180–4.
[157] Khorana AA, Ahrendt SA, Ryan CK, et al. Tissue factor expression, angiogenesis, and
thrombosis in pancreatic cancer. Clin Cancer Res 2007;13:2870–5.
[158] Stein PD, Beemath A, Meyers FA, et al. Incidence of venous thromboembolism in
patients hospitalized with cancer. Am J Med 2006;119:60e8.
[159] Stein PD, Beemath A, Meyers FA, et al. Pulmonary embolism as a cause of death in
patients who died with cancer. Am J Med 2006;119:163e5.
[160] Stein PD, Hull RD, Kayali F, et al. Venous thromboembolism in pregnancy: 21 year
trends. Am J Med 2004;117:121e5.
Risk Factors of Deep Vein Thrombosis 27
[161] Anonymous. Oral contraception and thromboembolic disease. J R Coll Gen Pract
1967;13: 267e79.
[162] Ginsberg JS, Brill-Edwards P, Burrows RF, Bona R, Prandoni P, Buller HR, et al.
Venous thrombosis during pregnancy: leg and trimester of presentation. Thromb
Haemost 1992;67:519– 20
[163] Ikard RW, Ueland K, Folse R. Lower limb venous dynamics in pregnant women. Surg
Gynecol Obstet 1971;132:483– 8.
[164] Macklon NC, Greer IA, Bowman AW. An ultrasound study of gestational and postural
changes in the deep venous system of the leg in pregnancy. Br J Obstet Gynaecol
1997;104:191–7.
[165] Metcalfe J, Ueland K. Maternal cardiovascular adjustments to pregnancy. Prog
Cardiovasc Dis 1974;16:363–74.
[166] Kerr MG, Scott DB, Samuel E. Studies of the inferior vena cava in late pregnancy. Br
Med J 1964;1:532– 3.
[167] Cockett FB, Thomas ML. The iliac compression syndrome. Br J Surg 1965;52:816– 21.
[168] Hull RD, Raskob GE, Carter CJ. Serial impedance plethysmography in pregnant
patients with clinically suspected deep vein thrombosis. Ann Intern Med
1990;112:663– 7.
[169] Rutherford SE, Phelan JP. Thromboembolic disease in pregnancy. Clin Perinatol
1986;13:719– 39.
[170] Stirling Y, Woolf L, North WR, Seghatchian MJ, Meade TW. Haemostasis in normal
pregnancy. Thromb Haemost 1984;52:176–82.
[171] Woodhams BJ, Candotti G, Shaw R, Kernoff PB. Changes in coagulation and
fibrinolysis during pregnancy: evidence of activation of coagulation preceding
spontaneous abortion. Thromb Res 1989;55:99– 107.
[172] Bonnar J. Blood coagulation and fibrinolysis in obstetrics. Clin Hematol 1973;12:58 –
63.
[173] Comp PC, Thurnau GR, Welsh J, Esmon CT. Functional and immunologic protein S
levels are decreased during pregnancy. Blood 1986;68:881– 5.
[174] Clark P, Brennand J, Conkie JA, McCall F, Greer IA, Walker ID. Activated protein C
sensitivity, protein C, protein S, and coagulation in normal pregnancy. Thromb
Haemost 1998;79:1166– 70.
[175] Wright JG, Cooper P, Astedt B, Lecander I, Wilde JT, Preston FE, et al. Fibrinolysis
during normal human pregnancy: complex interrelationships between plasma
levels of tissue plasminogen activator and inhibitors and the euglobulin clot lysis
time. Br J Haematol 1988; 69:253– 8.
[176] Bremme K, Ostlund E, Almqvist I, Heinonen K, Blomback M. Enhanced thrombin
generation and fibrinolytic activity in normal pregnancy and the puerperium.
Obstet Gynecol 1992;80:132– 7.
[177] Kruithof EK, Tran-Thang C, Gudinchet A, Hauert J, Nicoloso G, Genton C, et al.
Fibrinolysis in pregnancy: a study of plasminogen activator inhibitors. Blood
1987;69:460–6.
[178] McColl MD, Ramsay JI, Tait RD, Walker ID, McCall F, Conkie JA, et al. Risk factors for
pregnancy associated venous thromboembolism. Thromb Haemost 1997;78:1183–8.
28 Deep Vein Thrombosis
[179] Geerts WH, Bergqvist D, Pineo GF, et al. Preventive of venous thromboembolism.
American College of Chest physicians evidence-based clinical practice guidelines
(8th edition). Chest 2008;133: 381Se453S.
[180] Kroger K, Schelo C, Gocke C, et al. Colour Doppler sonographic diagnosis of upper
limb venous thromboses. Clin Sci 1998;94:657e61.
[181] Dollery CM, Sullivan ID, Bauraind O, et al. Thrombosis and embolism in long-term
central venous access for parenteral nutrition. Lancet 1994;344: 1043e5.
[182] Mustafa S, Stein PD, Patel KC, et al. Upper extremity deep venous thrombosis. Chest
2003; 163:1213e9.
[183] Hingorani A, Ascher E, Lorenson E, et al. Upper extremity deep venous thrombosis
and its impact on morbidity and mortality rates in a hospitalbased population. J
Vasc Surg 1997;26:853e60.
[184] Monreal M, Lafoz E, Ruiz J, et al. Upperextremity deep venous thrombosis and
pulmonary embolism. A prospective study. Chest 1991;99:280e3.
[185] Horattas MC, Wright DJ, Fenton AH, et al. Changing concepts of deep venous
thrombosis of the upper extremityereport of a series and review of the literature.
Surgery 1988;104:561e7.
[186] Joffe HV, Kucher N, Tapson VF, et al. Upper-extremity deep vein thrombosis: a
prospective registry of 592 patients. Circulation 2004;110:1605e11.
[187] Broviac JW, Cole JJ, Scribner BH. A silicone rubber atrial catheter for prolonged
parenteral alimentation. Surg Gynecol Obstet 1973; 136:602– 606.
[188] Hickman RO, Buckner CD, Clift RA, Sanders JE, Stewart P, Thomas ED. A modified
right atrial catheter for access to the venous system in marrow trans-plant
recipients. Surg Gynecol Obstet 1979; 148:871– 875.
[189] Foley MJ. Radiologic placement of long-term central venous peripheral access system
ports (PAS Port): results in 150 patients. J Vasc Interv Radiol1995; 6:255–262.
[190] Rubenstein EB, Fender A, Rolston KV, et al. Vascular access by physician as-sistants:
evaluation of an implantable peripheral port system in cancer patients. J Clin Oncol
1995; 13:1513–1519.
[191] Shetty PC, Mody MK, Kastan DJ, et al. Outcome of 350 implanted chest ports placed
by interventional radiologists. J Vasc Interv Radiol 1997; 8:991–995.
[192] Schwarz RE, Groeger JS, Coit DG. Subcutaneously implanted central venous access
devices in cancer patients: a prospective analysis. Cancer 1997; 79: 1635–1640.
[193] Struk DW, Bennett JD, Kozak RI. Insertion of subcutaneous central venous infusion
ports by interventional radiologists. Can Assoc Radiol J 1995; 46:32–36.
[194] Hata Y, Morita S, Morita Y, et al. Peripheral insertion of a central venous access device
under fluoroscopic guidance using a peripherally accessed system (PAS) port in the
forearm. Cardiovasc Intervent Radiol 1998; 21:230 –233.
[195] Deppe G, Kahn ML, Malviya VK, Malone JM, Christensen CW. Experience with the
P.A.S.-Port venous access device in patients with gynecologic malignancies.
Gynecol Oncol 1996; 62:340 –343.
[196] Monreal M, Lafoz E, Ruiz J, Valls R, Alastrue A. Upper extremity deep venous
thrombosis and pulmonary embolism; a prospective study. Chest 1991;99:280–3.
[197] Saleh T, Matta F, Yaekoub AY, et al. Risk of venous thromboembolism with
inflammatory bowel disease. Clin Appl Thromb Hemost 2010. [Epub ahead of
print].
Risk Factors of Deep Vein Thrombosis 29
[198] Saleh T, Matta F, Alali F, et al. Liver disease and risk of venous thromboembolism.
Submitted for publication.
[199] Søgaard KK, Horva´th-Puho´ E, Grønbaek H, et al. Risk of venous thromboembolism
in patients with liver disease: a nationwide population-based case-control study.
Am J Gastroenterol 2009;104: 96e101.
[200] Northup PG, McMahon MM, Ruhl AP, et al. Coagulopathy does not fully protect
hospitalized cirrhosis patients from peripheral venous thromboembolism. Am J
Gastroenterol 2006;101:1524e8.
[201] Tripodi A, Primignani M, Chantarangkul V, et al. An imbalance of pro- vs anti-
coagulation factors in plasma from patients with cirrhosis. Gastroenterology
2009;137:2105e11.
[202] Danescu L, Badshah A, Danescu SC, et al. Venous thromboembolism in patients
hospitalized with thyroid dysfunction. Clin Appl Thromb Hemost 2009;15:676e80.
[203] Seriolo B, Accardo S, Garnero A, et al. Anticardiolipin antibodies, free protein S levels
and thrombosis: a survey in a selected population of rheumatoid arthritis patients.
Rheumatology 1999; 38:675e8.
[204] McEntegart A, Capell HA, Creran D, et al. Cardiovascular risk factors, including
thrombotic variables, in a population with rheumatoid arthritis. Rheumatology
2001;40:640e4.
[205] Matta F, Singala R, Yaekoub AY, et al. Risk of venous thromboembolism with
rheumatoid arthritis. Thromb Haemost 2009;101:134e8.
[206] Stein PD, Goldman J, Matta F, et al. Diabetes mellitus and risk of venous
thromboembolism. Am J Med Sic 2009;337:259e64.
[207] Matta F, Yaekoub AY, Stein PD. Human immunodeficiency virus infection and risk of
venous thromboembolism. Am J Med Sci 2008;336:402e6.
[208] Kayali F, Najjar R, Aswad F, et al. Venous thromboembolism in patients hospitalized
with nephrotic syndrome. Am J Med 2008;121:226e30.
[209] Stein PD, Beemath A, Meyers FA, et al. Deep venous thrombosis and pulmonary
embolism in patients hospitalized with sickle cell disease. Am J Med
2006;119:897e901.
[210] Navarro S, Ricart JM, Medina P, et al. Activated protein C levels in Behc¸ et’s disease
and risk of venous thrombosis. Br J Haematol 2004;126:550e6.
[211] Ray JG, Burows RF, Ginsberg JS, et al. Paroxysmal nocturnal hemoglobinuria and the
risk of venous thrombosis: review and recommendations for management of the
pregnant and nonpregnant patient. Haemostasis 2000;30:103e17.
[212] Fischer MD, Hopewell PC. Recurrent pulmonary emboli and Buerger’s disease. West J
Med 1981; 135:238e41.
[213] Osterud B (1998) Tissue factor expression by monocytes: regulation and
pathophysiological roles. Blood Coagul Fibrinolysis 9[Suppl 1]:S9–14
[214] Osterud B, Rapaport SI (1977) Activation of factor IX by the reaction product of tissue
factor and factor VII: additional pathway for initiating blood coagulation. Proc Natl
Acad Sci USA 74:5260–5264
[215] Esmon CT (2001) Role of coagulation inhibitors in inflammation. Thromb Haemost
86:51–56
30 Deep Vein Thrombosis
[216] Schaub RG, Simmons CA, Koets MH, Romano PJ, Stewart GJ (1984) Early events in the
formation of a venous thrombus following local trauma and stasis. Lab Invest
51:218–224
[217] Yang J, Furie BC, Furie B (1999) The biology of P-selectin glycoprotein ligand-1: its role
as a selectin counterreceptor in leukocyte-endothelial and leukocyte-platelet
interaction. Thromb Haemost 81:1–7
[218] Coughlan AF, Hau H, Dunlop LC, Berndt MC, Hancock WW (1994) P-selectin and
platelet-activating factor mediate initial endotoxin- induced neutropenia. J Exp
Med 179:329–334
[219] Lim YC, Snapp K, Kansas GS, Camphausen R, Ding H, Luscinskas FW (1998)
Important contributions of P-selectin glycoprotein ligand-1-mediated secondary
capture to human monocyte adhesion to P-selectin, E-selectin, and TNF-alpha-
activated endothelium under flow in vitro. J Immunol 161:2501–2508
[220] Ramasamy S, Lipke DW, McClain CJ, Hennig B (1995) Tumor necrosis factor reduces
proteoglycan synthesis in cultured endothelial cells. J Cell Physiol 162:119–126
[221] Klein NJ, Shennan GI, Heyderman RS, Levin M (1992) Alteration in
glycosaminoglycan metabolism and surface charge on human umbilical vein
endothelial cells induced by cytokines, endotoxin and neutrophils. J Cell Sci 102[Pt
4]:821–832
[222] Conway EM, Rosenberg RD (1988) Tumor necrosis factor suppresses transcription of
the thrombomodulin gene in endothelial cells. Mol Cell Biol 8:5588–5592
[223] Murugesan G, Rani MR, Ransohoff RM, Marchant RE, Kottke-Marchant K (2000)
Endothelial cell expression of monocyte chemotactic protein-1, tissue factor, and
thrombomodulin on hydrophilic plasma polymers. J Biomed Mater Res 49:396–408
[224] Moore KL, Andreoli SP, Esmon NL, Esmon CT, Bang NU (1987) Endotoxin enhances
tissue factor and suppresses thrombomodulin expression of human vascular
endothelium in vitro. J Clin Invest 79:124–130
2
1. Introduction
Laparoscopic surgery – is one of the most progressive minimal invasive surgery branches.
About 25–40% of all abdominal operations are performed laparoscopicaly in our days and
this rating is going in ascending order. Laparoscopic operations (cholecystectomy,
fundoplication, appendectomy, bypass due to morbid obesity et at.) have rapidly become
the operations of choice in abdominal surgery. Several authors reported that deep vein
thrombosis (DVT) in the legs developed in 30% of postoperative patients and pulmonary
embolism (PE) in 10% of these patients.
Many studies explored the frequency of deep leg vein thrombosis after various open
abdominal surgery operations. Some studies (Geerts and al.,1994) determined that deep leg
vein thrombosis develops in 55% of polytrauma patients. Clagett &Reisch, 1988; found 25%
rate of DVT after open abdominal surgery. Literature data on the incidence of DVT after
laparoscopic operations is limited. Patel MI and al., 1996; carried out the prospective clinical
study, studying the frequency of DVT after laparoscopic cholecystectomy. The rate of DVT,
diagnosed by ultrasound Doppler, was 55%. The incidence of DVT and PE after laparoscopic
fundoplications was 1.8% in our prospective randomized study. Lord RV and al., 1998;
performed the prospective clinical study and compared the incidence of DVT after
laparoscopic or microlaparotomic (open) cholecystectomy. The incidence of DVT was 1.7%
after laparoscopic and 2.4% after open cholecystectomy. Nevertheless, many authors states,
that the incidence of DVT should be less after laparoscopic surgery when comparing with
open one. Laparoscopic operations, in comparison with open ones, have few basic differences:
1. Laparoscopic operation involves a specific manipulation called abdominal insufflation
in addition to the routine procedure of general anesthesia. The increased intra-
abdominal pressure associated with pneumoperitoneum (12-14 mm Hg) during
laparoscopic upper gastrointestinal surgery has the potential to compound any lower–
limb venous stasis already present due to general anesthesia by compressing the
retroperitoneal vena cava and iliac veins.
2. Most of laparoscopic operations often last more than 1.5 hours and often are performed
with patient in the reverse Trendelenburg position. These differences also have the
potential for an increased risk of significant venous stasis.
32 Deep Vein Thrombosis
Fig. 1. Ultrasonography of the common femoral vein before the general anesthesia. Figure
on the left side shows blood velocity in the femoral vein using doppler ultrasound; the right
side shows longitudinal section of the femoral vein.
Fig. 2. Ultrasonography of the common femoral vein at the 12 mm Hg insufflation when the
patient was placed in the reverse Trendelenburg position (angle 45°). Figure on the left side
shows blood velocity in the femoral vein using doppler ultrasound; the right side shows
longitudinal section of the femoral vein.
34 Deep Vein Thrombosis
Fig. 3. Ultrasonography of the common femoral vein before the general anesthesia. Figure
shows the cross-sectional area of the femoral vein.
Fig. 4. Ultrasonography of the common femoral vein at the 12 mm Hg insufflation when the
patient was placed in the reverse Trendelenburg position (angle 45°). Figure shows the
cross-sectional area of the femoral vein.
Venous Stasis and Deep Vein Thrombosis Prevention in Laparoscopic Surgery 35
The decrease in the blood velocity of the femoral vein and increase of the cross-sectional
area differed significantly between 5-mm Hg insufflation, 10-mm Hg insufflation and 12-
mm Hg insufflation. Futhermore, the blood velocity of the femoral vein decreased
significantly and the femoral vein cross-sectional area increased significantly when the
patient was placed in the reverse Trendelenburg position with the presence of 12 mm Hg
pneumoperitoneum. These findings suggest that venous stasis, caused by abdominal
insufflation during laparoscopic operations, can be reduced by using lower pressures.
Postural changes during laparoscopic operation also greatly affect venous stasis. The large
increase in femoral venous blood flow and large decrease in femoral vein cross-sectional
area observed after release of the pneumoperitoneum in our study confirmed that venous
stasis is present through all laparoscopic operation.
Several other scientists (Ido et al.,1995; Jorgensen et al., 1994; Beebe et al., 1993) also
investigated femoral vein blood flow velocities during and after abdominal insufflation in
patients, who underwent laparoscopic cholecystectomy, using color Doppler
ultrasonography. They also found, that abdominal insufflation reduced the blood velocity in
the femoral vein and suggested that abdominal insufflation during laparoscopic operation
can cause femoral vein stasis. The femoral vein stasis, which appears in laparoscopic
operations, can be minimized by reducing the intraabdominal pressure during operation,
and avoiding reverse Trendelenburg position as much as possible.
The creation of pulsatile venous blood flow is thought to be crucial for the function of
mechanical antistasis devices. This pulsatile blood flow episodically flushes activated
clotting factors from stagnant soleal sinuses, thereby preventing thrombosis. Both IPC and
IECS were able to achieve pulsatile blood flow with a pneumoperitoneum (figure 5 and 6).
The maximum blood velocity generated by the IPC when a pneumoperitoneum (12 mm Hg)
was present and the patient was placed in the reverse Trendelenburg position was
significantly greater than the maximum blood velocity generated by the IECS. The femoral
vein cross-sectional area decreased 25 % when IPC was acting on the legs, when
pneumoperitoneum (12 mmHg) was present and the patient was placed in the reverse
Trendelenburg position, while the femoral vein cross-sectional area decreased only 3 %
when IECS was acting on the legs during laparoscopic operation. The femoral vein cross-
sectional area changes received by IPC were significantly greater than changes received by
IECS when the pneumoperitoneum (12 mm Hg) was present. These findings show that IPC
is more effective than IECS in reducing venous stasis induced by the pneumoperitoneum
and the reverse Trendelenburg position. Graded compression leg bandages is totally
ineffective in patients, undergoing laparoscopic operations (figure 7).
BA - Before the general anesthesia, * - the patient placed in the reverse Trendelenburg position (angle
45°), ** - the patient placed in the reverse Trendelenburg position, when the mechanical antistasis
devices is acting on the legs, *** - the patient placed in the reverse Trendelenburg position, when the
mechanical antistasis devices is acting on the legs and 1 h after the beginning of the operation
Fig. 7. Mean blood flow velocity changes in the relationship with pneumoperitoneum,
reverse Trendelenburg position and antistasis devices.
38 Deep Vein Thrombosis
With a pneumoperitoneum in place, neither device is able to return the depressed blood
flow velocity to the values recorded without a pneumoperitoneum. The incidence of DVT
and PE after laparoscopic fundoplications was 1.8% in our study.
1 h after
Before operation 10 min after
Variable introduction of
(Baseline) extubation
laparoscope
IPC group
1.07 (0.89-1.23) 1.0 (0.73-1.26) 1.85 (1.31-5.36)ab
(n = 10)
IPC + LMWH group
1.11 (0.83-1.94) 1.01 (0.77-1.93) 1.44 (0.89-2.17)
(n = 10)
Values are expressed as median (range)
a p < 0.0001 vs baseline
b p < 0.0001 vs 1h after introduction of laparoscope
Table 1. Changes of prothrombin fragment F1+2 plasma levels (nmol/L) in the IPC and IPC
+ LMWH groups.
Venous Stasis and Deep Vein Thrombosis Prevention in Laparoscopic Surgery 39
Table 2. Changes of thrombin – antithrombin complex plasma levels (µg/L) in the IPC and
IPC + LMWH groups.
Coagulation is regulated at several levels. Key inhibitors include tissue factor pathway
inhibitor, antithrombin, and the protein C pathway. The inhibition of the factor VIIa/tissue
factor complex (extrinsic coagulation pathway) is effected by TFPI. TFPI acts in a two-step
manner. In the first step, TFPI complexes and inactivates factor Xa to form a TFPI/factor Xa
complex. The TFPI within this complex then inactivates tissue factor-bound VIIa as the
second step. Because the formation of the TFPI/factor Xa complex is a prerequisite for the
efficient inactivation of factor VIIa, the system ensures that some factor Xa generation occurs
before factor VIIa-mediated initiation of the coagulation system is shut down. In this study
plasma free tissue factor pathway inhibitor as marker of hypocoagulation effect was used.
Our study results demonstrated that a combination of LMWH and IPC generates
hypocoagulation effect and are more effective than IPC alone to prevent deep-vein
thrombosis after laparoscopic fundoplication (table 3).
Table 3. Changes of free tissue pathway factor inhibitor plasma levels (ng/ml) in the IPC
and IPC + LMWH groups.
The antithrombotic effect of IPC is thought to be the result of increased venous velocity and
stimulation of endogenous fibrinolysis. However, the results of several studies on the
enhancement of hypocoagulation effect by an IPC have been controversial. Cahan et al.,
2000; showed that external pneumatic compression devices did not enhance systemic
fibrinolysis or prevent postoperative shutdown either by decreasing plasminogen activator
40 Deep Vein Thrombosis
inhibitor-1 activity or by increasing tissue plasminogen activator activity. Their data suggest
that external pneumatic compression devices do not prevent deep venous thrombosis by
fibrinolytic enhancement; effective prophylaxis is achieved only when the devices are used
in a manner that reduces lower extremity venous stasis. Jacobs et al., 1996; reported that
sequential gradient intermittent pneumatic compression induces prompt, but short-lived,
alterations in both fibrinolytic function, and the values quickly reverted to baseline on
termination of compression. Okuda et al., 2002; reported that intermittent compression boot
did not prevent increased intravascular thrombogenesis and platelet activation through
significant increases of plasma D-dimmer and β-thromboglobulin after laparoscopic
cholecystectomy. Killewich et al., 2002; also reported that enhanced regional fibrinolysis in
the lower extremities could not be detected with the use of external pneumatic compression
devices, as measured with tissue plasminogen activator and plasminogen activator
inhibitor-1 activity in common femoral venous blood samples in patients undergoing
abdominal surgery. On the other hand, Comerota et al., 1997; reported that external
pneumatic compression devices induced a significant decrease in plasminogen activator
inhibitor-1 activity in normal volunteers.
In our study, the IPC used alone during laparoscopic fundoplication, did not prevent
increased intravascular thrombogenesis through significant increases of plasma F1+2 and
TAT during and after laparoscopic fundoplication.
Giddings et al.,1999; reported that IPC led to highly significant falls in factor VIIa, associated
with increased levels of tissue factor pathway inhibitor in non-smoking volunteers.
Chouhan et al., 1999; investigated the effect of IPC on the tissue factor pathway in 6 normal
subjects and 6 patients with postthrombotic venous disease. Their study results
demonstrated that IPC results in an increase in plasma TFPI and decline in FVIIa in both
groups. Authors speculated that inhibition of tissue factor pathway, the initiating
mechanism of blood coagulation, is a possible mechanism for the antithrombotic effect of
IPC. Our study results demonstrate that IPC used alone did not increase TFPI in plasma and
didn‘t produce hypocoagulation effect during laparoscopic fundoplication.
Most circulating TFPI is bound to lipoproteins. TFPI is also found in platelet α-granules and
on the endothelium cell surface. TFPI bound to the endothelium is released with therapeutic
doses of heparin or low molecular weight heparin, suggesting that TFPI binds to
endogenous glycosaminoglycans on the endothelium wall surface.
Our clinical data suggest that LMWH, administered 1 h before operation, together with IPC
induce more favorable hypocoagulation profile compared with LMWH alone. However,
clinical data, comparing the rate of DVT between these two prophylactic methods are still
lacking. On the other hand, alone LMWHs have been evaluated in a large number of
randomized clinical trials and have been shown to be safe and effective for the prevention
and treatment of venous thrombosis in laparoscopic or in open surgery.
Our recommendation is LMWH, administered 1 h before operation, together with IPC
against postoperative venous tromboembolism in laparoscopic operations. Of course, this
recommendation has to be proved in future prospective randomized clinical trials,
comparing the incidence of DVT between these two prophylactic methods.
Venous Stasis and Deep Vein Thrombosis Prevention in Laparoscopic Surgery 41
3. Conclusions
1. Venous stasis, which appears in laparoscopic operations, can be minimized by reducing
the intraabdominal pressure during operation and avoiding reverse Trendelenburg
possition as much as possible.
2. IPC is more effective than IECS in reducing venous stasis induced by the
pneumoperitoneum and the reverse Trendelenburg position.
3. Graded compression leg bandages is ineffective in patients, undergoing laparoscopic
operations with pneumoperitoneum.
4. With a pneumoperitoneum in place, neither mechanical device is able to return the
depressed blood flow velocity to the values recorded without a pneumoperitoneum.
5. Hypercoagulable state is present during and after laparoscopic fundoplications when
using IPC alone for deep-vein thrombosis prevention: the IPC, used alone, did not
prevent increased intravascular thrombogenesis through significant increases of plasma
F1+2 and TAT during operation.
6. A combination of LMWH and IPC generates hypocoagulation effect and can be more
effective than IPC alone to prevent deep-vein thrombosis after laparoscopic operations.
7. Our recommendation is LMWH, administered 1 h before operation, together with IPC
against postoperative venous tromboembolism in laparoscopic operations.
4. References
Allan, A.; Williams, JT & Bolton J.P. (1983). The use of graduated compression stockings in
the prevention of postoperative deep vein thrombosis. Br J Surg 70:172-4.
Beebe, D.S.; Mc Nevin, M.P.; Crain, J.M. & al. (1993). Evidence of venous stasis after
abdominal insufflation for laparoscopic cholecystectomy. Surg Gynec Obstet
176:443-7.
Borow, M., & Goldson, H.J. (1981). Postoperative venous thrombosis: Evaluation of five
methods of prophylaxis. Am J Surg 141:245-51.
Broze, G.J.J. (1995). Tissue factor pathway inhibitor. Thromb Haemost 95:90-3.
Browse, N.L. & Negus, D. (1970). Prevention of postoperative leg vein thrombosis by
electrical muscle stimulation. An evaluation with I-labeled fibrinogen. Br Med J
3:615-8.
Cahan, M.A.; Hanna, D.J.; Wiley, L.A.; Cox , D.K. & Killewich, L.A. (2000). External
pneumatic compression and fibrinolysis in abdominal surgery. J Vasc Surg
32(3):537-43.
Caprini, J.A.; Arcelus, J.I.; Laubach, M.; Size, G.; Hoffman, K.N. & Coats, R.W. (1995).
Postoperative hypercoagulability and deep-vein thrombosis after laparoscopic
cholecystectomy. Surg Endosc 9(3):304-9.
Chouhan, V.D.; Comerota, A.J.; Sun, L.; Harada, R.; Gaughan, J.P. & Rao, A.K. (1999).
Inhibition of tissue factor pathway during intermittent pneumatic compression: A
possible mechanism for antithrombotic effect. Arterioscler Thromb Vasc Biol
19(11):2812-7.
Comerota ,A.J.; Gwendolyn, J. & Stewart, J. (1990). Operative venodilatation: a previously
unsuspected factor in the cause of postoperative deep vein thrombosis. Surgery
106:301-9.
42 Deep Vein Thrombosis
Comerota, A.J.; Chouhan, V.; Harada, R.N.; Sun, L.; Hosking, J. & Veermansunemi R. (1997).
The fibrinolytic effects of intermittent pneumatic compression: mechanism of
enhanced fibrinolysis. Ann Surg 226:306-13.
Dexter, S.P.; Griffith, J.P.; Grant, P.J. & McMahon, M.J. (1996). Activation of coagulation and
fibrinolysis in open and laparoscopic cholecystectomy. Surg Endosc 10(11):1069-1074.
Di, V.G.; Frazzetta, M.; Sciume, C.; Lauria, L.G.; Patti, R. & Leo P. (2000). Changes in the
hemostatic system after laparoscopic cholecystectomy. G Chir 21(5):213-8.
Ido, K. ; Suzuki, T. & Taniguchi Y. (1995). Femoral vein stasis during laparoscopic
cholecystectomy: effects of graded elastic compression leg bandages in preventing
thrombus formation. Gastrointestinal Endoscopy 42:151-5.
Ido, K.; Suzuki, T. & Kimura K. (1995). Lower-extremity venous stasis during laparoscopic
cholecystectomy as assessed using color Doppler ultrasound. Surg Endosc 9:310-3.
Jacobs, D.G.; Piotrowski, J.J.; Hoppensteadt, D.A.; Salvator, A.E. & Fareed, J. (1996).
Hemodinamic and fibrinolytic consequences of intermittent pneumatic
compression: preliminary results. J Trauma 40:710-7.
Jorgensen, J.O.; Lalak, N.J. & North L. (1994). Venous stasis during laparoscopic
cholecystectomy. Surgical laparoscopy and Endoscopy 4:128-33.
Killewich, L.A.; Cahan, M.A.; Hanna, D.J.; Murakami, M.; Uchida, T. & Wiley, L.A. (2002).
The effect of external pneumatic compression on regional fibrinolysis in a
prospective randomized trial. J Vasc Surg 36(5):953-8.
Kiudelis, M.; Endzinas, Z.; Mickevicius, A. & Pundzius, J. (2002). Venous stasis and deep
vein thrombosis prophylaxis during laparoscopic fundoplication. Zentrallbl Chir
(127):944-9.
Lindberg, F.; Rasmussen, I.; Siegbahn, A. & Bergqvist, D. (2000). Coagulation activation after
laparoscopic cholecystectomy in spite of thromboembolism prophylaxis. Surg
Endosc 14(9):858-61.
Lord, R.V.; Ling, J.J.; Hugh, T.B.; Coleman, M.J.; Doust, B.D. & Nivison-Smith, I. (1998).
Incidence of deep vein thrombosis after laparoscopic vs minilaparotomy
cholecystectomy. Arch Surg 133(9):967-73.
Okuda, Y.; Kitajima, T.; Egawa, H.; Hamaguchi, S.; Yamaguchi, S. & Yamazaki, H. (2002). A
combination of heparin and an intermittent pneumatic compression device may be
more effective to prevent deep-vein thrombosis in the lower extremities after
laparoscopic cholecystectomy. Surg Endosc 16:781-4.
Patel, M.I.; Hardman, D.T.; Nicholls, D.; Fisher; C.M. & Appleberg, M. (1996). The incidence
of deep venous thrombosis after laparoscopic cholecystectomy. Med J Aust
164(11):652-4, 656.
Risberg, B. (1988). Pathophysiological mechanisms of thromboembolism. Acta Chir Scand
Suppl (550):104-14.
Rosengarten, D.S.; Laird, J. & Jeyasingh K. (1970). The failure of compression stockings
(Tubigrip) to prevent deep venous thrombosis after operation. Br J Surg 57:296-9.
Schaub, R.G.; Lynch, P.R. & Stewart, G.J. (1978). The response of canine veins to three
abdominal surgery; a scanning and transmission electron microscopic study.
Surgery 83:411-24.
Vecchio, R.; Cacciola, E.; Martino, M.; Cacciola, R.R. & MacFadyen, B.V. (2003).
Modifications of coagulation and fibrinolytic parameters in laparoscopic
cholecystectomy. Surg Endosc 17(3):428-433.
3
1. Introduction
Deep venous thrombosis (DVT) is an illness of clinical interest, due to the associated
morbidity and mortality and its social and health care consequences. The etiology in young
patients has shown it frequently associated with congenital coagulation abnormalities and
acquired/inherited risk factors (table I) (a,b).
Inherited
Common
G169A mutation in the factor V gene (factor V Leiden)
G20219A mutation in the protrombin (factor II) gene
Homozygous C677T mutation in the methylenetetrahydrofolate reductase gene
Rare
Antitrombin deficiency
Protein C deficiency
Protein S deficiency
Very rare
Dysfibrinogenemia
Homozygous homocystinuria
Probably inherited
Increased levels of factor VII, IX, XI or fibrinogen
Acquired
Surgery and trauma
Prolonged immobilization
Older age
Cancer
Myeloproliferative disorders
Previous thrombosis
Pregnancy and the puerperium
Use of contraceptives or hormone-replacement therapy
44 Deep Vein Thrombosis
Fig 1) develop ventromedial to the posterior cardinal veins and ventrolateral to the aorta.
The intersubcardinal anastomosis forms between the paired subcardinal veins, anterior to
the aorta, and caudal to the superior mesenteric artery.
Fig. 1. Conceptual framework for development of the IVC. Composite schematic shows the
relative positions and interrelationships of the three paired embryonic vessels that
contribute to development of the IVC. The pictured veins are not all present simultaneously.
card= cardinal, post= posterior, SMA= superior mesenteric artery, v= vein, 1=
intersubcardinal anastomosis, 2 = intersupracardinal anastomosis.
Anastomosis between the posterior cardinal and subcardinal veins (light violet in Fig 1)
develop on each side at approximately the level of the intersubcardinal anastomosis. At the
same time, union occurs between the right subcardinal vein and the hepatic segment of the
IVC, which forms from the vitelline vein. As the cranial portions of the posterior cardinal
veins begin to atrophy, blood return from the lower extremities is shunted through the
postsubcardinal anastomosis, then through the subcardinal-hepatic anastomosis to the
hepatic segment of the IVC. This process establishes the pre-renal division of the IVC. The
next major development is the appearance of the paired supra-cardinal veins (goldenrod in
Fig 1), which lie dorso-medial to the posterior cardinal veins and dorso-lateral to the aorta.
Initially, multiple anastomosis form between the posterior and supracardinal veins. On each
side, a suprasubcardinal anastomosis (yellow in Fig 1) develops from union of the
postsupracardinal and the postsubcardinal anastomosis. In addition, intersupracardinal
anastomosis develop dorsal to the aorta. The supracardinal veins then separate into cranial
(azygos) and caudal (lumbar) ends. Meanwhile, inferiorly, anastomosis develop between the
two posterior cardinal veins and between the posterior and lumbar supracardinal veins.
With further atrophy of the posterior cardinal veins, blood return from the lower extremities
is shunted through the supracardinal system to the suprasubcardinal anastomosis, then to
the pre-renal division of the IVC. In addition, blood return from the left side of the body is
shunted to the right across the intersupracardinal and interpostcardinal anastomosis.
46 Deep Vein Thrombosis
Finally, the left supracardinal vein is one of the last veins to disappear, although Huntington
and McLure(j) state that the vessel does not so much atrophy as become incorporated into
the right supracardinal vein by coalescence of the multiple anastomosis. In summary, the
normal IVC is composed of four segments: hepatic, suprarenal, renal, and infrarenal. The
hepatic segment is derived from the vitelline vein. The right subcardinal vein develops into
the suprarenal segment by formation of the subcardinal-hepatic anastomosis. The renal
segment develops from the right suprasubcardinal and postsubcardinal anastomosis. It is
generally accepted that the infra-renal segment derives from the right supracardinal vein,
although this idea is somewhat controversial(i). In the thoracic region, the supracardinal
veins give rise to the azygos and hemiazygos veins. In the abdomen, the postcardinal veins
are progressively replaced by the subcardinal and supracardinal veins but persist in the
pelvis as the common iliac veins.
(a)
Fig. 2. Partial malrotation and left IVC in a 49-year-old man. (a) Schematic shows a left IVC
terminating at the left renal vein. (b-e) CT scans presented from caudal to cranial show the
anomaly. (b) Note the left IVC (arrow) inferior to the renal veins. (c) The left IVC joins the
left renal vein (arrow). (d) The left renal vein (arrow) crosses anterior to the aorta in the
normal fashion. (e) A normal right-sided prerenal IVC is formed from the confluence of the
left (straight arrow) and right (curved arrow) renal veins. Note the increased attenuation of
the right renal vein relative to that of the left due to absence of dilution from relatively
unenhanced lower-extremity venous return. The major clinical significance of this anomaly
is the potential for misdiagnosis as left-sided paraaortic adenopathy(k).
48 Deep Vein Thrombosis
(a)
Fig. 3. Double IVC in a 53-year-old woman with lymphoma. (a) Schematic shows left and
right infrarenal IVCs. The left IVC terminates at the left renal vein. (b) CT scan obtained
inferior to the renal veins shows left (straight arrow) and right (curved arrow) IVCs. (c-e) CT
scans show the left IVC ending at the confluence with the left renal vein (arrow in c), which
crosses anterior to the aorta in the normal fashion (arrow in d) to join a normal pre-renal
IVC (arrow in e). There may be morphological variation and asymmetry of the left and right
veins. Double IVC should be suspected in cases of recurrent pulmonary embolism following
placement of an IVC filter(i).
Vena Cava Malformations as an
Emerging Etiologic Factor for Deep Vein Thrombosis in Young Patients 49
(a)
Fig. 4. CT images of azygos continuation of the IVC in a 48-year-old man. (a) Schematic
shows lack of contiguity between the pre-renal segment of the IVC (arrow) and the hepatic
segment. The vessel parallel to the aorta under the crus is the azygos vein. (b, c) CT scans
obtained at the level of the diaphragmatic crus (b) and the level of the azygos vein arch (c)
show the enlarged azygos vein (straight arrow) draining into the superior vena cava (curved
arrow in c).
The azygos vein joins the superior vena cava at the normal location in the right para-tracheal
space. The hepatic segment (often termed the post-hepatic segment) is ordinarily not truly
absent; rather, it drains directly into the right atrium. Since the post-sub-cardinal
anastomosis does not contribute to formation of the IVC, each gonadal vein drains to the
ipsi-lateral renal vein(j). Formerly thought to be predominantly associated with severe
congenital heart disease and a-splenia or poly-splenia syndromes, azygos continuation of
the IVC has become increasingly recognized in otherwise asymptomatic patients since the
advent of cross-sectional imaging. It is important to recognize the enlarged azygos vein at
the confluence with the superior vena cava and in the retrocrural space to avoid
misdiagnosis as a right-sided para-tracheal mass or retro-crural adenopathy(k,l). Preoperative
knowledge of the anatomy may be important in planning cardiopulmonary bypass and to
avoid difficulties in catheterizing the heart(m).
50 Deep Vein Thrombosis
(a)
Fig. 5. Circumaortic left renal vein in a 73-year-old woman. (a) Schematic shows two left
renal veins, with the inferior vein crossing posterior to the aorta. (b-e) Contiguous 5-mm-
thick CT sections presented from cranial to caudal show the anomaly. (b) The superior left
renal vein (arrow) crosses anterior to the aorta. (c-e) The inferior vein (curved arrow)
descends approximately 2 cm and receives the left gonadal vein (straight arrow in d) before
crossing posterior to the aorta. The major clinical significance is in preoperative planning
prior to nephrectomy and in renal vein catheterization for venous sampling. Misdiagnosis as
retroperitoneal adenopathy should be avoided.
Vena Cava Malformations as an
Emerging Etiologic Factor for Deep Vein Thrombosis in Young Patients 51
(a)
Fig. 6. Retroaortic left renal vein in a 27-year-old man. (a) Schematic shows a single left renal
vein, which crosses posterior to the aorta. (b, c) CT scans show the left renal vein (arrow)
descending to cross posterior to the aorta.
52 Deep Vein Thrombosis
(a)
Fig. 7. Spinal dysraphism and double IVC with hemiazygos continuation in a 2-year-old
boy. (a) Schematic shows failed development of the right pre-renal IVC and hemi-azygos
continuation of the left IVC. (b-e) MR images presented from caudal to cranial show the
anomaly. (b) Note the right (straight arrow) and left (curved arrow) IVCs. (c) The right renal
vein (arrowhead) descends to receive the right IVC and crosses posterior to the aorta
(arrow) to join the left IVC. (d) The left IVC continues cephalad left of the aorta under the
diaphragmatic crus as the hemi-azygos vein (arrow). (e) In the thorax, the hemi-azygos vein
(straight arrow) crosses posterior to the aorta (arrowhead) to join a rudimentary azygos vein
(curved arrow) approximately 1-2 cm below the carina.
Vena Cava Malformations as an
Emerging Etiologic Factor for Deep Vein Thrombosis in Young Patients 53
2.6 Double IVC with retro-aortic right renal vein and hemi-azygos continuation of the
IVC
More than one anomaly can coexist in a patient. In the case of a double IVC with a retro-aortic
right renal vein and hemi-azygos continuation of the IVC, the embryologic basis is persistence
of the left lumbar and thoracic supra-cardinal vein and the left supra-sub-cardinal
anastomosis, together with failure of formation of the right sub-cardinal–hepatic anastomosis.
In addition, the right renal vein and right IVC meet and cross posterior to the aorta to join the
left IVC and continue cephalad as the hemi-azygos vein (Fig 7). Thus, there is also persistence
of the dorsal limb of the renal collar and regression of the ventral limb. In the thorax, the hemi-
azygos vein crosses posterior to the aorta at approximately T8 or T9 to join the rudimentary
azygos vein. Alternate collateral pathways for the hemi-azygos vein include cephalad
continuation to join the coronary vein of the heart via a persistent left superior vena cava and
an accessory hemi-azygos continuation to the left brachio-cephalic vein(n).
2.7 Double IVC with retro-aortic left renal vein and hemiazygos continuation of the
IVCA
Double IVC with a retro-aortic left renal vein and azygos continuation of the IVC is an
interesting combination. It results from persistence of the left supracardinal vein and the
dorsal limb of the renal collar with regression of the ventral limb. In addition, the sub-
cardinal-hepatic anastomosis fails to form (Fig 7). A recent study demonstrated that azygos
continuation of the IVC can be predicted with ultrasonography by identifying the right renal
artery crossing abnormally anterior to the IVC (15).
(a)
Fig. 8. Circumcaval ureter in a 65-year-old man. (a) Schematic shows the right ureter
encircling the IVC. (b-d) CT scans presented from cranial to caudal show the anomaly. (b)
The right ureter (arrow) is positioned posterior to the IVC. (c) The ureter (arrow) then
courses to the left of the IVC. (d) Finally, the ureter (arrow) crosses anterior to the IVC.
(Courtesy of Akira Kawashima, MD, Lyndon B. Johnson General Hospital, Houston, Tex.)
Vena Cava Malformations as an
Emerging Etiologic Factor for Deep Vein Thrombosis in Young Patients 55
(a)
Fig. 9. Absent infra-renal IVC. (a) Schematic shows absence of the IVC below the renal veins.
Collateral flow from the lower extremities reaches the azygos vein via para-vertebral
collateral veins. (b) CT scan obtained inferior to the aortic bifurcation shows absence of the
common iliac veins. Enlarged ascending lumbar veins are present (black arrow). Note the
iliac arteries (white arrow). (c) CT scan obtained inferior to the kidneys shows absence of the
IVC (white arrow). Enlarged ascending lumbar veins are present (black arrow). (d) CT scan
obtained at the level of the renal veins shows a normal pre-renal IVC formed at the
confluence of the renal veins (arrow). (e) CT scan obtained at the level of the pre-renal IVC
(white arrow) shows prominent para-vertebral collateral veins (black arrow), which lead to
a prominent azygos vein (arrowhead). (f) Coronal T1-weighted MR image shows the
enlarged ascending lumbar veins (arrow). (g) Lateral maximum-intensity projection
reconstruction of two-dimensional time-of-flight MR images shows formation of enlarged
ascending lumbar veins at the confluence of the internal and external iliac veins (solid
straight arrow). Note the anastomosis between the ascending lumbar veins and the azygos
vein (open straight arrow) via prominent anterior para-vertebral veins (white curved
arrow). Also note the pre-renal IVC (black arrowhead) posterior to the portal vein (black
curved arrow), as well as prominent anterior abdominal wall collateral veins (white
arrowheads). (Figs 10b, 10c, and 10g reprinted, with permission, from references).
56 Deep Vein Thrombosis
and vitamin-K antagonists(u). The diagnosis of anomalies in the inferior vena cava influences
the strategy for prevention the pulmonary embolism and long them maintenance treatment.
The use of mechanical device as caval filter is clearly limited by the anomalous anatomy of
the inferior vena cava and, generally, is-not indicated. On the other hand the use of oral anti-
coagulant (commonly warfarin) should be adjusted to maintain a target international
normalized ratio of 2.5 (range 2-3) and extended indefinitely in absence of main contra-
indications(d).
At present the introduction of new drugs as the factor Xa antagonists (rivarixaban,
apixaban, edoxaban, ect) and the direct thrombin inhibitors as dabigatran etexilate could
improve the therapeutic options. The promising results of the recent clinical studies in terms
of efficacy and safety, suggest that these new drugs may allow a reduction of the length of
hospital stay after an acute DVT, and a better adherence to guidelines in the long term
treatment. The principal advantages of these drugs are the absence of the need of a routine
coagulation monitoring and a therapeutic activity not influenced by dietary regimen and by
drugs as NSADIs and statins(u). Potential limitations are the lack of specific antidotes
(however the hal-life of these drugs is relative short) and the absence of a simple assay for
quantification of activity or plasma level.
In conclusion these interesting pharmacological characteristics could improve the benefit-
risk balance of long-term anti-coagulant therapy and the overall clinical outcome.
4. Conclusions
The complexity of the ontogeny of the IVC, with numerous anastomosis formed between the
three primitive paired veins, can lead to a wide array of variations in the basic plan of
venous return from the abdomen and lower extremity. Some of these anomalies have
significant clinical implications. Although vascular structures can usually be readily
identified on contrast-enhanced CT scans, identification of unusual venous arrangements
may be difficult in those cases in which intravenous contrast material is contraindicated. In
such patients, MR imaging may be used to distinguish aberrant vessels from masses by
demonstrating flow voids or flow-related enhancement. The echo-scanning may suggest the
presence of venous anomalies but usually it insufficient for a detailed diagnosis. A
knowledge of IVC and renal vein anomalies is essential to avoid diagnostic pitfalls.
5. References
[1] Rosendaal F. Thrombosis in the young: epidemiology and risk factors. A focus on
venous thrombosis. Thromb Haemost. 1997;78:1-6.
[2] García-Fuster MJ, Fernández C, Forner MJ, & Vayá A. Estudio prospectivo de los
factores de riesgo y las características clíni- cas de la enfermedad tromboembólica
en pacientes jóvenes. Med Clin (Barc). 2004;123:217-9.
[3] Ruggeri M, Tosseto A, Castaman G & Rodeghiero F. Congenital absence of the inferior
vena cava: a rare risk factor for idiopathic deep vein thrombosis. Lancet.
2001;357:441.
[4] M. Bianchi, D. Giannini, A. Balbarini, M.G. & Castiglioni. Congenital hypoplasia of
inferior vena cava and inherited thrombophilia: rare associated risk factor for
Vena Cava Malformations as an
Emerging Etiologic Factor for Deep Vein Thrombosis in Young Patients 57
[21] Mavrakanas T, Bounameaux H. The potential role of new oral anticoagulants in the
prevention and treatment of thromboembolism. Pharmacology & Therapeutics 130
(2011) 46–58
4
1. Introduction
Deep venous thrombosis of the lower limb is a common disease with an incidence of 80 per
10000 (Patel et al 2011) and has potential fatal consequences in the form of pulmonary
embolism.
It is usually seen in patients undergoing major surgery particularly orthopaedic surgery,
trauma, prolonged immobilisation or hypercoagulable states (such as in the context of
malignancy). There are associations with drugs such as the oral contraceptive pill and
hormone replacement therapy (tamoxifen) that predispose to hypercoagulability.
that early intervention in the setting of acute DVT is associated with lower recurrence rates,
lower incidence of post thrombotic syndrome and lower rates of fatal PE.
Pending the results of these trials however, clinicians should assess the need for invasive
measures on a case by case basis, based on the timing of the DVT, associated risk factors for
development of DVT, risk factors for thrombolysis related bleeding, age and prognosis of
the patient.
3. Endovascular techniques
The goals of endovascular treatment of acute DVT include: prevention of PE, early symptom
relief, and prevention of post thrombotic syndrome (Vendatham 2006).
The endovascular techniques available to achieve these goals include catheter directed
thrombolysis, mechanical/rheolytic thrombectomy and stent placement. IVC filters can
theoretically be used in conjunction with these techniques to reduce the rate of PE during
therapy, although this is controversial.
These techniques require knowledge of, and skills in, ultrasound guided needle punctures,
wire and catheter manipulation, thrombolysis drug administration, placement of
endovascular stents, and familiarity with the use of various mechanical/rheolytic
thrombectomy devices. The specific equipment will vary according to their availability and
preference.
passage of an infusion catheter), and a diagnostic venogram performed either via the sheath
or via a catheter. This may or may not be sufficient to visualise the extent of thrombosis.
In either case, this is followed by a wire and angiographic catheter (usually 0.035in
system, such as glidewire (Terumo, Somerset NJ) and a 5Fr angled tapered glidecatheter),
which traverses the occluded segment. A venogram past the level of the occlusion is
performed, usually in the IVC, to confirm intraluminal position, and absence of more
centrally located clot.
The thrombolytic agent is usually injected at this point, and a number of different
thrombolytic strategies have been described. At our institution, we would lace the
length of the thrombosed segment using 200,000IU of Urokinase as the diagnostic
catheter is being retracted. An infusion catheter with an infusion length that covers the
occluded segment is then selected, and is inserted over a wire. The active infusion
segment of the catheter is placed over the thrombosed segment of vein, which allows
direct delivery of thrombolytic agent throughout the length of the thrombus. An
infusion of Urokinase would then be commenced, at a rate of between 100,000-150,000
IU per hour. A Heparin infusion through the vascular sheath side arm is also
commenced. The infusion is continued overnight, and patient nursed in a High
Dependency Unit or Intensive Care Unit with one to one nursing. If the case arrives
early in the morning, the infusion is left running until the mid afternoon. The patient
will then return to the angiography suite for a venogram to reassess the degree of
thrombosis and treat any underlying lesions.
If significant thrombus remains after the initial infusion, the infusion can be continued if
it is felt that the clot will continue to disintegrate. However, this increases the dose and
the duration of therapy with the associated increased risks of thrombolysis. It also
increases the length of hospital stay and potentially increases the costs of treatment.
Currently, CDT combined with mechanical thrombectomy is the preferred treatment
(Sharaffudin 2003).
between proximal and distal balloons for control and to prevent PE. Rotational devices have
direct contact with the endothelium and subsequently have the potential for endothelial
damage. However there have been no studies to analyse their efficacy compared with
rheolytic devices.
Rheolytic devices include the Angiojet (Possis, Minneapolis,MN). The device uses high-
pressure saline jets to fragment the thrombus. The jets also create a negative pressure zone
which draws the fragmented thrombus toward the catheter where it is aspirated and
removed. A possible advantage of the Angiojet device is that there is no contact of the
maceration component of the device with the vessel wall. However, its use of high-pressure
saline jets carries a theoretical risk of haemolysis and the release of adenosine and
potassium. (Zhu, 2008) This has been linked to the incidence of bradyarrhythmia in cardiac
applications of the device (Lee et al, 2005) or haemoglobinuria.
Ultrasound enhanced devices include the EKOS Endowave (EKOS Corporation, Bothell,
WA, USA) and Omniwave (Omnisonics Medical Technologies, Wilmington, MA, USA).
These are catheters that contain multiple ultrasound transducers, which radially emit
high-frequency, low-energy ultrasound energy. The ultrasonic energy expands and thins
the fibrin component of thrombus, exposing plasminogen receptor sites, and the
ultrasound forces thrombolytic into the clot and keeps it there (Francis et al, 1995). This
technique may be associated with fewer haemolytic effects than rheolytic thrombectomy
(Lang et al, 2008) and has a lower potential for endothelial damage than rotational
thrombectomy devices.
We prefer the use of the Angiojet system at our institution, as an adjunctive modality to
CDT. The timing of its use is dependent on the case, and preference of the interventionist.
However, the method is the same in either situation. The Angiojet catheters come in a range
of sizes and lengths, we prefer the 5 and 6Fr systems. The device is passed several times
across the thrombosed segment over a wire and under fluoroscopic visualisation. In order to
minimise the risk of haemolysis, each pass is limited to 30 seconds with 10 second rests in
between.
The potential added benefit of the Angiojet system is the ability of the catheters to ‘pulse
spray’ thrombolytic agent using high pressure jets into the thrombus itself, improving
delivery.
4. Results
4.1 Catheter directed thrombolysis
To date, the largest DVT thrombolytic database is the venous registry (Mewissen 1999),
which is a prospective registry of patients with a DVT who underwent CDT with urokinase.
473 patients were enrolled with 287 patients followed up at 1 year. 83% of patients had
thrombolysis >50%. There were also a strong relationship between early thrombus removal
and 1- year patency (primary patency rate of 60%). Major bleeding complications occurred
in 11%, most often at the puncture site. 1% of patients developed a PE. Two patients (<1%)
died (one from PE and one from intracranial haemorrhage).
64 Deep Vein Thrombosis
Grunwald and Hofmann (2004) retrospectively analysed 74 patients who underwent CDT
for DVT and compared Urokinase, Alteplase and Reteplase. They found that there was no
statistical difference between infusion times, success rates and complication rates between
the three agents. However, they did find that the new recombinant agents are significantly
less expensive than Urokinase in the United States.
No RCTs have been published looking at CDT in acute DVT. However, currently the
TORPEDO trial is underway which is a large scale RCT looking at the efficacy of CDT vs
anticoagulation in treatment of DVT. Mid term results show that CDT is superior to
anticoagulation therapy alone in the prevention of recurrence of DVT, reduction in PTS, and
reduction of hospital stays.
Similarly the ATTRACT Trial is currently underway looking at the efficacy of CDT.
5. Adjunctive procedures
DVT, particularly in the iliocaval system, can be associated with chronic venous obstruction,
which can lead to valvular insufficiency and consequently venous hypertension. This in turn
is associated with a higher incidence of post thrombotic syndrome (PTS). There are multiple
other causes of venous obstruction, which include May Thurner syndrome, external
compression (e.g. cancer, lymphocoeles) and retroperitoneal fibrosis. Very frequently,
thrombolysis and thrombectomy can uncover the underlying lesion which precipitated the
venous thrombosis. Failure to identify and treat these lesions, despite successful
thrombolysis, can result in higher rates of recurrence, and the development of PTS.
The advantage of CDT as compared with anticoagulation therapy alone in the treatment of
acute DVT, is that it allows the opportunity to treat the underlying lesion and restore flow in
most cases. Obviously, non mechanical underlying issues must also be addressed, such as
underlying prothrombotic syndromes.
The objective is to restore flow and the measures employed usually involve angioplasty
and stenting of the lesion. Angioplasty and stenting in the setting of obstruction has been
shown to improve quality of life and improve symptoms (Hartung et al 2005, Neglen P et
al 2005, Raju S et al 2002;). The lesions treated usually lie within the IVC, iliac and
femoral veins. It has not been shown that angioplasty and stenting of lesions below this
Endovascular Therapies in Acute DVT 65
level is of any benefit. However, chronic lesions do benefit as well as acute obstructions
(Titus 2011).
The procedure is similar to angioplasty of the arterial system. Once access is achieved,
heparin is given if anticoagulation has not been already instituted. A wire is passed across
the lesion, followed by a catheter. Any wire can be used however 0.035in wires are
preferred. This is usually not too difficult in an acute thrombus, which is soft, and has not
had time to organise. Contrast is injected beyond the lesion to ensure intraluminal
position. The catheter is then exchanged for a balloon which is usually sized
approximately 20% greater than the expected calibre of the vein. Angioplasty of the
venous system is different from the arterial system, in that the balloons can be oversized
to a greater extent than in the arteries. There is also a greater propensity for veins to have
elastic recoil, such that even with aggressive angioplasty using high pressure balloons, the
veins collapse back to their obstructed state. In other cases there is persistent stenosis in
the vein post angioplasty. When this is the case, stenting is performed. These are also
oversized in relation to the vein.
Fig. 1. 20 y.o. girl with acute DVT and swollen, dusky leg. Popliteal access has been achieved
and venogram demonstrates thrombus in the femoral vein.
Endovascular Therapies in Acute DVT 67
Fig. 4. Same patient. IVC filter placed prior to commencement of procedure. Infusion
catheter placed through the thrombus and Urokinase infusion commenced.
70 Deep Vein Thrombosis
Fig. 10. Stent deployed in the left iliac vein with good flow through the vessel.
76 Deep Vein Thrombosis
7. Conclusion
Endovascular techniques are important therapeutic options in the prevention of limb loss,
recurrence and post thrombotic syndrome related to acute DVT, and have been shown to be
superior to anticoagulation therapy alone. It also is advantageous in uncovering and
treating underlying lesions that contribute to the DVT.
No guidelines are available currently in terms of patient selection, techniques and the use of
IVC filters and at present these decisions are made on a case by case basis at the discretion of
the interventionist. Large randomized controlled trials underway currently will hopefully
be able to shed more insight on these issues.
8. References
Arko FR, Davis CM, Murphy EH, et al. 2007. Aggressive Percutaneous Mechanical
Thrombectomy of Deep Venous Thrombosis. Archives of Surgery. 142
(6):513-519
Comerota AJ & Aldridge SC. 1993. Thrombolytic therapy for deep venous thrombosis: a
clinical review. Can J Surg. 36(4): 359-64.
Francis CW, Blinc A, Lee S, Cox C. 1995. Ultrasound accelerates transport of recombinant
tissue plasminogen activator into clots. Ultrasound Med Biol;21(3):419e24
Gallus AS, 1998. Thrombolytic therapy for venous thrombosis and pulmonary embolism.
Clin Haematol. 11(3):663-73.
Grosman C & McPherson S. 1999. Safety and efficacy of catheter directed thrombolysis for
iliofemoral venous thrombosis. Am J Roentgenol. 172: 667-672.
Grunwald MR and Hofmann LV. 2004. Comparison of UK, Alteplase, and reteplase for CDT
of DVT. Journal Vasc Interv Radiol; 15:347-352
Hartung O, Otero A, Boufi M, et al. 2005. Mid term results of endovascular treatment of
symptomatic chronic non-malignant iliocaval venous occlusive disease. J Vasc Surg.
42(6):1138-44.
Karthikesalingam, EL Young, RJ Hinchliffe, et al. 2011. A systematic review of percutaneous
Mechanical thrombectomy in the treatment of DVT. Eur J Vasc endovasc Surgery, 41:
554-565
Lang EV, Kulis AM, Villani M, et al. 2008. Hemolysis comparison between the OmniSonics
OmniWave endovascular system and the Possis AngioJet in a porcine model. J Vasc
Interv Radiol;19(8):1215e21
Lee MS, Makkar R, Singh V et al. 2005. Pre-procedural administration of aminophylline does
not prevent AngioJet rheolytic thrombectomy-induced bradyarrhythmias. J Invasive
Cardiol;17(1):19e22
Leizorovicz A, Simonneau G, Decousus H et al. 1994. Comparison of efficacy and safety of
low molecular weight heparins and unfractionated heparin in initial treatment of
deep venous thrombosis: a meta analysis. BMJ. 309 (6950): 299-304.
Lensing AW, Prins MH, Davidson BL, et al. 1995. Treatment of deep venous thrombosis
with low-molecular-weight heparins: a meta-analysis. Arch Intern Med;
155:601–607.
Endovascular Therapies in Acute DVT 77
Lorch H, Welger D, Wagner V, et al. 2000. Current practice of temporary vena cava filter
insertion: a multicenter registry. J Vasc Interv Radiol. 11(1): 83-8.
Mewissen. 1999. CDT for lower extremity Deep venous thrombosis: report of a national
multicenter registry, Radiolog; 211:39-49
Molina JE, Hunter DW & Yedlicka JW. Thrombolytic therapy for iliofemoral venous
thrombosis. 1992. Vasc Surg. 26:630-637.
Neglen P, Raju S. 2005. Endovascular treatment of chronic occlusions of the iliac veins and the
inferior vena cava. Rutherford RB, editor. Vascular Surgery. 6th ed. Philadelphia:
Elsevier Saunders;. pp 2321-32.
Patel K, Basson MD, Borsa JJ, et al, May 2011. Deep Venous Thrombosis. In: Emedicine.
Available from:
[Link]
Piccioli A, Prandoni P Goldhaber S. Epidemiologic characteristics, management
and outcome of deep venous thrombosis in a tertiary-care hospital: The
Brigham and Women’s Hospital DVT registry. 1996. American Heart Journal.
132(5).
Prandoni P, Lensing A, Cogo A, et al. 1996. The Long-Term clinical course of Acute Deep
Venous Thrombosis. Annals of Internal Medicine. 125:1-7.
Raju S, Fountain T & McPherson SH. 1998. Catheter directed thrombolysis for deep venous
thrombosis. J Miss State Med Assoc. 39(3):81-4.
Raju S, Owen S Jr, Neglen P. 2002. The clinical impact of iliac venous stents in the
management of chronic venous insufficiency. J Vasc Surg; 35:8-15.
Schweizer J, Kirch W, Koch R, et al. 1998. Short and long term results after thrombolytic
treatment of deep venous thrombosis. J Am Coll Cardiol. 36:1336-1343.
Sharafuddin MJ, Sun S, Hoballah JJ, et al. 2003. Endovascular management of venous
thrombotic and occlusive diseases of the lower extremities. J Vasc Interv Radiol.
14(4)4:405-23.
Sirgusa S, Cosmi B, Piovella F, et al. 1996. Low molecular weight heparins and
unfractionated heparin in the treatment of patients with acute venous
thromboembolism: results of a meta analysis. The American Journal of Medicine.
100(3): 269-277.
Tarry WC, Makhoul RG, Tisnado J, et al. 1994. Catheter directed thrombolysis following
ven cava filtration for severe deep venous thrombosis. Ann Vasc Surg.
8(6):583-590.
Titus JM, Moise MA, Bena J, et al. 2011. Ilioifemoral stenting for venous occlusive disease. J
Vasc Surg. 53(3): 706-712.
Vedantham S, Millward S, Cardella J, et al. 2006. Society of Interventional Radiology
Position Statement: Treatment of Acute Iliofemoral deep Vein Thrombosis with use
of Adjunctive Catheter directed Intrathrombus Thrombolysis. J Vasc Interv Radiol.
17:613-616.
Wells PS & Forster AJ. Thrombolysis in deep vein thrombosis: is there still an indication?
2001 Thromb Haemost. 86 (1):499-508.
78 Deep Vein Thrombosis
1. Introduction
Radiological imaging plays a central role in the diagnosis, and treatment, of deep venous
thrombosis (DVT) in the upper and lower limb. The intention of this chapter is not to
distract the reader with a detailed account of the physics behind generating ultrasound (US),
computed tomography (CT) and MR (magnetic resonance) vascular imaging. This would
demand a chapter in its own right, and this information can be readily found in textbooks.
Instead, emphasis will be placed on the clinical indications for requesting imaging in the
diagnosis of DVT, as well as the potential limitations of these modalities. This will be
supplemented with a review of current evidence and guidelines, and examples of the
common image findings. The latest advances in venous MR imaging will be discussed, as
will the role of interventional radiology in the treatment of DVT. Finally, considering that it
is now universally accepted that DVT and pulmonary embolus (PE) are essentially
manifestations of the same disease – namely, venous thromboembolism (Moser et al., 1994)
– the imaging and radiological management of PE will also be addressed.
An additional problem is the small, but recognised, risk of actually causing thrombosis
through the irritant effects of iodinated contrast medium on the vascular endothelium.
Even allowing for these limitations, the continued use of conventional angiography is not
sustainable in modern clinical practice considering it is a relatively time consuming and
hence expensive procedure, and there is a growing demand on hospital Radiology
departments to diagnose an increasingly prevalent disorder, affecting 200 per 100,000 of
those aged 70- 79 years. The argument for providing a robust and efficient means of
diagnosis is augmented by evidence that the initial clinical evaluation of DVT is often
ineffective (Barnes et al., 1975; Haeger, 1969, as cited in Fraser & Anderson, 1999). Other
conditions including lymphoedema, cellulitis, superficial phlebitis, muscle sprain and
ruptured baker’s cyst can be indistinguishable from DVT. Indeed, seventy five percent of
patients who present with signs and symptoms of DVT do not have the disease (Heijboer et
al., 1993; Wells et al., 1995).
Fig. 1. Lower limb venogram showing a normal superficial femoral vein. Note the normal
valves (arrow).
2.2 Ultrasound
Ultrasound successfully addresses many of the above requirements, and has clearly become
the first line imaging modality in suspected DVT (Cronan, 1993; Dorfman & Cronan, 1992).
Unlike other modalities, it can also be a portable technique, allowing assessment of critically
ill patients at the bedside. Cronan et al. gathered data from multiple studies to show a
sensitivity of 95% and a specificity of 98% in detecting lower limb disease. The performance
of venous ultrasound in the upper limb has been less studied, mainly because of the lower
incidence of upper limb thrombus. However, the frequency of upper limb venous
thrombosis is increasing considering that the two major risk factors are malignancy and
central venous catheter placement (Allen et al., 2000; Baron et al., 1998). The performance of
upper limb venous sonography should be high as ultrasound provides the highest spatial
Radiological Imaging and Intervention in Venous Thrombosis 81
resolution of any current imaging modality where veins are sonographically visible (Roditi
& Fink, 2009). A relatively large study of upper limb venous sonography including over one
hundred patients reported a sensitivity and specificity of 82% (Baarslag et al., 2002).
The most accurate ultrasound tool for diagnosing DVT is compressibility of the vein in the
transverse plane; a normally patent vein simply disappears when compressed by the
ultrasound transducer (fig. 2). The maximum pressure required to obliterate a vein is much
less than that required to deform the adjacent artery. Fortuitously, the entire deep venous
system of the lower limb consists of arteries that parallel veins. Compression should not be
performed in the longitudinal plane because the transducer may slide off the vessel with
compression resulting in a false – positive finding.
(a) (b)
Fig. 2. (a) Transverse ultrasound of a normal left common femoral vein (V) and common
femoral artery (A). (b) Transducer compression obliterates the vein, leaving the
accompanying artery unaffected (A).
Technique: In suspected lower limb DVT the veins are examined from the inguinal ligament
(junction of the great saphenous vein of the superficial system with the common femoral
vein of the deep system), to the popliteal vein within the popliteal fossa. There is varying
opinion on the usefulness of assessing the distal calf veins, and they are not routinely
scanned by the authors in whose institution’s protocol employs a repeat interval scan for
those with high pre-test probability (see later). There is currently no consensus on what, if
any, treatment is indicated in below knee thrombus (Righini, 2007; Schellong, 2007), and the
reliability of compression US in excluding calf DVT has been questioned (Dauzat et al., 1997,
as cited in Johnson et al., 2010; Kearon et al., 1998). A meta – analysis reported the sensitivity
for detecting isolated calf DVT to be 73% (Kearon et al., 1998). Anticoagulation of calf DVT
(that might spontaneously resolve) may unnecessarily place patients at increased risk of
potential side effects of such medication, with an estimated 1.1% risk of major bleeding
(Krakow & Ortel, 2005, as cited in Johnson et al., 2010). This particularly applies to frailer
patients vulnerable to intra - cerebral haemorrhage from even innocuous trauma.
Furthermore, the value of adding distal (calf) US to proximal US of the lower limbs for
diagnosis of PE was investigated in a sub-analysis of a large, randomised trial. A total of 855
patients with suspected PE underwent investigation by pre – test probability assessment, D-
82 Deep Vein Thrombosis
Fig. 3. Duplex US. Normal phasic variation in Doppler waveform within the superficial
femoral vein during deep respiration (arrow).
Other indicators of thrombus include distension of the vein in acute thrombosis (typically
long established clot does not expand the lumen), and visualising clot within the affected
vein (fig. 4). Unfortunately, a significant number of acute clots are isoechoic i.e. of the same
Radiological Imaging and Intervention in Venous Thrombosis 83
ultrasound density to flowing blood, rendering them invisible to the naked eye unless
colour mapping is used. Differentiating acute from chronic DVT is a challenge with all
imaging modalities, not just ultrasound. The maturation of thrombus from anechoic i.e. less
dense than blood, through to hyperechoic i.e. more dense is very variable, and exact age
determination is not possible. Six months following a DVT, 50% of patients will have
persisting abnormality on US (Dougherty RS & Brant WE, 2007), making the distinction
between acute–on–chronic versus chronic changes very difficult. In addition to thrombus
appearance, studies have assessed change in thrombus diameter (Kearon et al., 1998),
change in thrombus length (Linkins et al., 2004), and Doppler analysis of flow (Prandoni et
al., 2002) in an attempt to differentiate acute from chronic changes, but there remains no
consensus on which ultrasound measurement can be relied upon to solve this potentially
important dilemma. A sensible approach is to obtain a baseline scan at the time of
discontinuing anticoagulation to allow for comparison in the event of the patient re–
presenting with recurring symptoms.
(a) (b)
Fig. 4. (a) Transverse image showing non – compressible DVT within the right common
femoral vein on US. The lumen of the vein contains echogenic clot implying that it is
relatively chronic (arrow). (b) Longitudinal Duplex image highlighting non – occlusive clot
within the common femoral vein (arrow) with blood flowing around the thrombus.
Venous Thrombi are dynamic structures, especially within the first 1 to 2 weeks after their
onset (O’shaughnessy & Fitzgerald, 2000a, 2000b). Up to 25% of calf DVTs may propagate
into the proximal veins (Johnson et al., 2010). Therefore, it is routine practice to repeat a
negative scan after 5 to 7 days to assess for propagation into the proximal vasculature,
particularly in patients with high pre–test probability scores.
Importantly there are of course limitations to US. As discussed, the calf veins are not readily
identified, especially in the swollen oedematous leg, often necessitating a repeat
examination to exclude proximal clot propagation. In addition the iliac veins are not readily
assessable, and the adductor canal (at the junction of the superficial femoral vein [SFV] and
popliteal vein) is notoriously difficult to visualise even in thin patients. The saphenous vein
or collaterals can be mistaken for the SFV. In addition, the SFV is duplicated in
approximately 20% of patients, potentially leading to diagnostic error, particularly if one
system is occluded and the other patent. Obesity and oedema can render examinations
84 Deep Vein Thrombosis
inconclusive. Interestingly, studies have shown that whilst US is sensitive and specific for
symptomatic lower limb DVT, it has rather poor sensitivity for asymptomatic DVT
compared to conventional venography, with sensitivity between 29 and 38% (Davidson et
al., 1992; Turkstra et al., 1997, as cited in Roditi & Fink, 2009). US has been investigated as a
potential screening test in asymptomatic patients deemed to be at high risk of DVT
following surgical procedures. However, the sensitivity and specificity appear to be reduced
in this setting, a randomised – controlled trial discovering no added benefit of screening
patients for DVT after lower limb arthroplasty surgery (Robinson et al., 1997, as cited in
Fraser & Anderson, 1999).
A final potential pit – fall is worth clarifying, especially for the referring clinician acting
upon the radiological report. The superficial femoral vein is actually part of the deep venous
system, and thrombus involving this vein could easily be interpreted as only a superficial
phlebitis by the unaware clinician. The term should either be avoided, and replaced with the
deep femoral vein (the practice of the authors), or the conclusion of the report should clearly
indicate that there is DVT.
vessel wall. Clot forming an obtuse angle implies chronic thromboembolic disease, but this
can also be seen in the acute setting. Secondary signs on CT reflect the non-specific
abnormalities frequently seen on chest radiography. Pleural based wedge shaped
consolidation indicates peripheral haemorrhage or infarction. Peripheral oligaemia (paucity
of blood vessels distal to the occluded artery), pleural effusions and linear atelectasis (partial
collapse) can also be observed.
Detailed depiction of the lung parenchyma offers additional information not provided by V/Q
scans (fig. 6b). In the context of a negative test for PE, an alternative explanation for the
patient’s symptoms may be highlighted. A study found that as many as two – thirds of
patients with an initial suspicion of PE received another diagnosis following CTPA (Hull et al.,
1994, as cited in Schoepf & Costello, 2004). In another study, CTPA identified pleural or
parenchymal abnormalities that explained indeterminate defects on V/Q scans in 57% of
patients (van Rossum et al., 1996, as cited in Kanne et al., 2004). Although a normal V/Q scan
essentially excludes PE, a high probability scan has a sensitivity of 88%, compared to 94 – 96%
for CTPA (Kanne et al., 2004). Patients with intermediate or indeterminate probability scans
(because of background lung or pleural abnormalities) still have a 30 – 40% incidence of PE
(Klein JS, 2007). V/Q scans, however, should always be considered in young patients with low
pre-test probability and normal chest radiographs in view of its lower radiation dose.
Fig. 5. CTPA in the axial (transverse) plane demonstrating bilateral filling defects within the
contrast opacified pulmonary arteries diagnostic of PE (arrows).
The main cause of death within the first 30 days after a PE is right ventricular failure
(Schoepf et al., 2004). Right ventricular enlargement on CTPA has been shown to correlate
with right ventricular dysfunction on echocardiography, and to predict early death from
acute PE. In patients with confirmed PE, evidence of right heart strain / dysfunction should
always be sought as this can influence patient management with regards reperfusion
therapy. To accurately, and reproducibly, measure ventricular size the original CT data is
manipulated to allow reformatting in the 4 chamber orientation. This is simply performed at
the reporting workstation. The ventricle is measured at its maximum size at a level 1 cm
86 Deep Vein Thrombosis
ahead of the corresponding atrioventricular valve. A right ventricle : left ventricle ratio of >
0.9 is indicative of right ventricular enlargement (fig 6).
CT venography (CTV) can be combined with CTPA to evaluate both PE and DVT in a single
CT study (fig. 7). The lower limb veins are scanned at intervals 3 or 4 minutes following
completion of the pulmonary angiogram. The sensitivity and specificity of CTV has been
reported between 89 – 100% and 94 – 100% respectively (Begemann et al., 2003; Loud et al.,
2001, as sited in Kanne et al., 2004). The combined study also allows evaluation of the iliac
system, not afforded by US. However, a major concern is the additional radiation exposure.
A study found the addition of CTV to increase the gonadal dose by a factor of 500 in women
and 2000 in men (the dose is higher in men since the testes are external to the body cavity).
This translates to increased likelihood of birth defects and radiation – related death, albeit at
a very low added risk (Rademaker et al., 2001). Combined CTV also requires substantial
contrast medium dose for adequate venous opacification, significantly greater amounts than
the relatively small quantities (50 - 75 ml) required for CTPA on modern multidetector
scanners. The value of this combined study is therefore debatable, and CTV is not included
in the CTPA protocol in most European institutions, including our own.
(a) (b)
Fig. 6. Coronal oblique reformatted CTPA at the level of the atrioventricular valves
demonstrating right ventricular enlargement. (a) Maximum size of the left ventricle (line).
LA = left atrium. (b) Maximum size of the right ventricle (line). RA = right atrium. The
corresponding right ventricle : left ventricle ratio is > 0.9. Note the clot within the right
pulmonary artery (arrowhead); and a lung tumour (arrow) which was not clinically suspected.
The value of adding lower limb US in the evaluation of patients undergoing CTPA has been
evaluated in a large, randomised trial of 1819 patients with clinically suspected PE.
Following pre – test probability assessment, 909 patients were randomised to investigation
by D-dimer measurement and CTPA, and 916 patients were randomised to D-dimer
measurement, CTPA and venous US of the leg. The primary outcome was 3 month
thromboembolic risk in patients who were left untreated on the basis of exclusion of PE by
the investigation strategy. The prevalence of PE and the 3 month risk of thromboembolism
Radiological Imaging and Intervention in Venous Thrombosis 87
was the same in both investigation groups. Thus, venous US of the leg does not improve
diagnosis of 3 month thromboembolic risk when added to investigation by D-dimer analysis
and CTPA (Righini et al., 2008). Therefore, it can be argued that for patients who have
undergone CTPA for the investigation of PE, US of the leg is a redundant investigation.
The diagnostic power of current CT has provoked another interesting debate with the ability
to potentially identify clot down to sixth order branches with multi – detector row scanners
(MDCT). Older single detector row scanners (SDCT) have limited ability in detecting
isolated subsegmental PE. Whilst the treatment of embolus detected within third and even
fourth order subsegmental arteries is undisputed, the clinical relevance of detecting clot
within smaller, more peripheral branches is questionable (Kanne et al., 2004), and could be
unnecessarily subjecting patients to the side effects of anticoagulation. A review of 20
prospective cohort studies and 2 randomised controlled trials was done to evaluate the
importance of single and multiple detector row CTPA in the diagnosis of subsegmental PE.
This meta-analysis showed that the diagnosis rate of sub-segmental PE was 4.7% with SDCT
and 9.4% with MDCT. However, the 3 month risk of thromboembolic events in patients with
suspected PE who were left untreated based upon a diagnostic algorithm that included a
negative CTPA was 0.9% for SDCT and 1.1% for MDCT. Therefore, although MDCT
increases the proportion of patients diagnosed with PE compared with SDCT, it does not
substantially reduce the 3-month risk of thromboembolism. The authors suggest that
isolated sub-segmental PE may not be clinically relevant (Carrier et al., 2006). Small
peripheral emboli are believed to form even in healthy individuals (although this has never
been substantiated); and it is a function of the lung to prevent these small clots from
entering the arterial bed (Tetalman et al., 1973, as cited in Schoepf & Costello, 2004).
Fig. 7. CTV (combined with CTPA) demonstrating clot within the left superficial femoral
vein (arrow).
undergoing the greatest evolution in terms of venous imaging. Studies have already shown
the sensitivity and specificity of MR venography (MRV) to be comparable to conventional
venography in diagnosing femeropopliteal DVT (Cantwell et al., 2006; Fraser et al., 2002, as
cited in Cantwell et al., 2006). Conventional venography is poor by comparison in
opacifying the pelvic vessels (Cantwell et al., 2006; Spritzer, 2009). To reiterate, US also
performs poorly in assessing the iliac vessels. Where MRV has perhaps until now performed
less well than venography is in assessing the calf veins (Cantwell et al., 2006). With contrast
enhanced MRV this was largely because of difficulties in predicting the arrival of contrast in
the more distal veins to optimally time the acquisition of the images. This is confounded by
the very short transit time of standard extracellular contrast agents within the vascular bed
as they rapidly redistribute into the extracellular fluid space.
Recent advancements in the physical properties of contrast agents have overcome the
aforementioned difficulties in imaging the calf vessels. The “blood pool” contrast agent
gadofosveset trisodium (Vasovist, Bayer Schering Pharma, Berlin, Germany) binds to
plasma albumin extending the blood pool residence time. Not only does this eliminate time
constraints in acquiring satisfactory images, but allows very high spatial resolution imaging
of both the deep and superficial venous system (fig. 8). As previously mentioned, a potential
pitfall of venography and US is the not uncommon occurrence of duplicated veins. A study
investigating the effects of these anatomical variants in DVT suggested that DVT was twice
as likely to be missed (Liu et al., 1986, as cited in Cantwell et al., 2006). Fig. 9 shows
duplication of the SFV readily identified by high resolution MRI.
(a) (b)
Fig. 8. High resolution MRI using “blood pool” contrast allows excellent visualisation of both
veins and their accompanying arteries. (a) Coronal plane. The normal anatomy of the calf
arteries with their accompanying paired veins is clearly demonstrated with the anterior tibial
artery and veins (arrowhead) and the peroneal artery and veins (straight arrow). The superficial
(deep) femoral vein and artery are also shown (curved arrow). (b). Sagittal oblique view
showing clot within the common femoral vein (arrow) and great saphenous vein (arrowhead).
Radiological Imaging and Intervention in Venous Thrombosis 89
The lack of radiation makes MRI a more attractive option than CT, particularly when there
are concerns regarding pelvic DVT in younger patients as the reproductive organs are
within the scanning field. For this reason, MRI should always be considered in excluding
DVT in pregnancy. US is often equivocal especially in the latter stages due to technical
difficulties. A further venous complication of pregnancy is ovarian vein thrombosis, or
puerperal ovarian vein thrombophlebitis. Presentation is usually on the 2nd or 3rd day
postpartum with lower abdominal pain and fever. The major complications are septicaemia
and PE, which is reported to occur in up to 25%. MR is considered to be more sensitive than
CT or US in making the diagnosis (Kubik-Huch RA et al., 1999, as cited in Spritzer, 2009).
Several studies have evaluated the performance of pulmonary contrast enhanced MRA for the
diagnosis of PE. One of the larger studies (Oudkerk et al., 2002, as cited in Roditi & Fink, 2009)
assessed MRA in 141 patients with an abnormal perfusion lung scintigraphy and compared
the findings with those of pulmonary DSA. Sensitivity was 77%, and the demonstration of
emboli in two patients with a normal angiogram resulted in a specificity of 98%. The major
advantage of MR is the lack of radiation exposure. A study has shown that the radiation from
a single CTPA may cause an additional attributable lifetime risk of cancer of almost 1% in
young women (Einstein et al., 2007, as cited in Roditi & Fink, 2009), mainly because breast
tissue is relatively radiosensitive. With the introduction of “blood pool” contrast agents a
comprehensive examination can be performed for PE and DVT using a single low dose
contrast injection, without the associated radiation concerns that hamper CT.
Fig. 9. High resolution MRI in the coronal plane showing duplication of the superficial
femoral vein (arrow). Note that the superficial femoral artery is not visible because it was
occluded at the groin. The patient had known claudication.
90 Deep Vein Thrombosis
The limitations of MR are the relative expense limiting availability, and for some patients
the claustrophobic environment preventing completion of the examination. At present, MRI
should be considered when there is a strong clinical suspicion of pelvic DVT, and in young
women requiring investigation for PE with abnormal chest X – ray precluding a V/Q scan.
used for this purpose. Venography at the time of the procedure can help assess the clot
burden, plan adjunctive treatments (venoplasty, pharmaco-mechanical disruption) and also
help define a suitable end point.
There is however a lack of prospective randomised data assessing the benefits of
thrombolysis as compared to anticoagulant therapy (Pianta & Thomson, 2011). This, in
combination with haemorrhagic complications and lack of awareness among physicians has
limited acceptance of this procedure.
(a) (b)
Fig. 10. Catheter thrombectomy. (a) Catheter tip within the right main pulmonary artery,
adjacent to embolus (arrow). (b) Post – treatment shows disruption of the clot. Note the
striking difference in contrast opacification of the pulmonary arterial tree pre – and post –
treatment.
92 Deep Vein Thrombosis
Complications include vessel wall and valve injury and kidney failure due to haemolysis.
Although patients can experience transient shortness of breath presumably due to
pulmonary microemboli, experience gained from thrombectomy of clotted fistulas has
shown that concomitant use of a plasminogen activator significantly reduced the risk of
symptomatic pulmonary embolism from the procedure (O’Sullivan, 2010).
Pharmacomechanical thrombolysis involves the combined use of a thrombectomy device
in combination with catheter directed infusion of a thrombolytic agent. The advantages of
this combination include better permeation of the thrombolytic agent and a smaller
duration of treatment. Although some devices can lower the systemic dose of the drug,
others do not do so.
A retrospective study of 93 patients showed that pharmacomechanical thrombolysis was an
effective treatment modality in patients with significant DVT and compared to catheter
directed thrombolysis alone, it provided similar treatment success, reduced length of
intensive care and hospital stay, and reduced hospital costs (Lin et al., 2006). However,
another study has shown that that use of the Trerotola device alone constituted effective
treatment of acute ilio-femoral DVT independent of adjunct pharmacological thrombolysis
(Lee et al., 2006).
There is, however, a relative lack of randomised data on the use of these devices and further
randomised studies are necessary.
Unlike arteries, veins have a high elastic recoil and lower rates of flow which leads to less
satisfactory results with long term stent patency, in the iliac veins, with a greater than 50%
re-stenosis in up to 15% of patients (Hood & Alexander, 2004). These figures are much
worse for patients who are hypercoagulable, have longer stent lengths and need infra-
inguinal stents.
Stenting an underlying lesion has, however, shown to help prevent or prolong the interval
to recurrence and can result in 50% increased patency rate than thrombolysis alone (Hood &
Alexander, 2004) and lower recurrence rates of ilio-femoral DVT (up to 73% lower) in
patients with May-Thurner syndrome (Oguzkurt et al., 2004). Most experience has been
gained with Wallstents (Boston Scientific, Hemel Hempstead, Herts, UK) which are self
expanding stents with a good radial strength. Studies have also shown the efficacy and
durability of stents in the IVC (Ing et al., 2001; Razavi et al., 2000).
Radiological Imaging and Intervention in Venous Thrombosis 93
IVC filters are classified as temporary or permanent (Streiff, 2000) and there are various
devices available that are approved for use. To cite a few examples, the Bird`s Nest Filter
and the Trapease filter are permanent whereas the Gunther Tulip and the Cook Celect Filter
are retrievable. Timely removal of retrievable filters is important to reduce the long term
94 Deep Vein Thrombosis
risk of filter deployment. In terms of safety and efficacy, there is no significant difference
between the two types of devices (Nazir et al., 2009). Complications associated with the
device include those encountered at the time of insertion such as access site haematoma,
pneumothorax, inadvertent arterial puncture and misplacement. Delayed complications
include IVC thrombosis, occlusion, venous insufficiency and pulmonary embolism.
4. Conclusion
The incidence of deep venous thrombosis is increasing, not just in the lower limb but also
within the deep veins of the upper limb, where malignancy and central venous catheter
placement are the major precipitating factors. Ultrasound provides a rapid and readily
available assessment, and can be portably used at the bedside in critically ill patients. There
is however limitations to ultrasound, particularly the poor visualisation of below knee clot.
In high risk patients, a short interval repeat scan is indicated to exclude the 25% of such clots
which can propagate above the knee.
The iliac veins within the pelvis are also inaccessible to ultrasound in almost every patient.
If DVT is strongly suspected within the pelvis, MRI should be considered. This modality has
seen the greatest advancements in recent times, with current protocols able to visualise the
venous system in very high spatial resolution. CT angiography of the limbs, whilst sensitive
and easily incorporated into routine CT pulmonary angiograph in suspected PE, should be
avoided in view of the radiation burden. The major advantage of MRI is the lack of radiation
exposure. MRI will almost certainly feature more commonly in DVT evaluation in the near
future with new “blood pool” contrast agents allowing a comprehensive examination for PE
and DVT in the same scan. One specific application is in relatively young patients with
abnormal CXR precluding a V/Q scan. However, CT is currently the “gold standard” for
diagnosing PE.
There are a number of endovascular treatment options in DVT which aim to achieve
thrombus removal, restoring patency and potentially limiting the acute complications
associated with DVT. It is important to appreciate there are limitations to these
treatments, with a relative lack of randomised controlled trials evaluating their true
efficacy. They should however be given consideration in selected patients as outlined
above.
5. Acknowledgements
We would like to thank Dr Iain Robertson & Dr Richard Edwards, consultant interventional
radiologists, Gartnavel hospital, Glasgow for kindly providing the images of catheter
thrombectomy.
6. References
Allen AW, Megargell JL, Brown DB, Lynch FC, Singh H & Singh Y. Venous thrombosis
associated with the placement of peripherally inserted central catheters. J Vasc
Interv Radiol. 2000; 11: 1309-1314.
Radiological Imaging and Intervention in Venous Thrombosis 95
Baarslag HJ, van Beek, EJR, Koopman MMW & Reekers JA. Prospective study of color
duplex ultrasonography in patients suspected of having deep venous thrombosis of
the upper extremities. Annals of Int Med. 2002; 136(12): 865-872.
Baron JA, Gridley G, weiderpass E, Nyren O & Linet M. Venous thromboembolism and
cancer. Lancet. 1989; 351: 1077-1088.
Blachere H, Latrabe V, Montaudon M, valli N, Couffinhal T, Raherisson C, Leccia F &
Laurent F. Pulmonary embolism revealed on helical CT angiography: comparison
with ventilation- perfusion radionuclide lung scanning. Am J Roentgenol. 2000;
174: 1041-1047.
British Thoracic Society guidelines for the management of suspected acute pulmonary
embolism. Thorax. 2003; 58(6): 470-483.
Cantwell CP, Cradock A, Bruzzi J, Fitzpatrick P, Eustace S & Murray JG. MR venography
with true fast imaging with steady- state precession for suspected lower- limb deep
vein thrombosis. J Vasc Interv Rad 2006; 17: 1763-1769.
Carrier M, Righini M, Wells PS, Perrier A, Anderson DR, Rodger MA, Pleasance S & Le Gal
G. Pulmonary embolism diagnoses on computer tomography: incidental and
clinical implications. A systematic review and meta- analysis of the management
outcome studies. J Thromb Haemost. 2010; 8(8): 1716-1722.
Comerota AJ & Paolini D. Treatment of acute ileofemoral deep venous thrombosis: a
strategy of thrombus removal. Eur J Vasc Endovasc Surg. 2007; 33(3): 351-360.
Cronan JJ. Venous thromboembolic disease: the role of US. Radiology. 1993; 186: 619-630.
De Valois JC, van Schaik CC, Verzijibergen F, van Ramshorst B, Eikelboom BC &
Meuwissen OJ. Contrast venography. Eur J Radiol. 1990; 11(2): 131-137.
Dorfman GS & Cronan JJ. Venous ultrasonography. Radiol Clin North Am. 1992; 30: 879-
893.
Dougherty RS & Brant WE. 2007. vascular ultrasound, In: Fundamentals of diagnostic
radiology, Brant WE & Helms CA., pp 1019-1060. Lipincott Williams & Wilkins,
ISBN- 13: 978-0-7817-6518-3, Philadelphia, USA.
Ferris EJ, Lim WN, Smith PL & casali R. May-Thurner syndrome. Radiology. 1983; 147: 29-
31.
Fraser JD & Anderson DR. Deep venous thrombosis: recent advances and optimal
investigation with US. Radiology. 1999; 211: 9-24.
Heijboer H, Buller HR, lensing AW, Turpie AG, Colly LP & Ten Cate JW. A comparison of
real-time compression ultrasonography with impedance plethysmography for the
diagnosis of deep-vein thrombosis in symptomatic outpatients. N Engl J Med. 1993;
329: 1365-9.
Hood DB & Alexander JQ. Endovascular management of iliofemoral venous occlusive
disease. Surg Clin N Am. 2004; 84: 1381-1396.
Hyers TM, Agnelli G & Hull RD. 1998. Antithrombotic therapy for venous thromboembolic
disease. In, Fifth American College of Chest Physicians(ACCP) consensus conference on
antithrombotic therapy, Dalen JE, Hirsh J(eds). Chest. 114(suppl):561-579.
Ing FF, Fagan TE, Grifka RG et al. Reconstruction of stenotic or occluded ilio-femoral
veins and inferior vena cava using intravascular stents: re-establishing access for
96 Deep Vein Thrombosis
future cardiac catheterisation and cardiac surgery. J Am Coll Cardiol. 2001; 37:
251-257.
Johnson MS. Current strategies for the diagnosis of pulmonary embolus. J Vasc Interv
Radiology. 2002;13: 13-23.
Johnson SA, Stevens SM, Woller SC, Lake, E, Donadini M, Cheng J, Labarere J & Douketis J.
Risk of deep venous thrombosis following a single negative whole leg compression
ultrasound. 2010; 303(5); 438-445.
Kanne JP & Lalani TA. Role of computed tomography and magnetic resonance imaging for
deep vein thrombosis and pulmonary embolism. Circulation. 2004; 109: 15-21.
Kaufman JA, Kinney TB, Streiff MB, Sing R, Proctor M, Mark DB, A Cipolle, S Cornerota,
Millward F, Frederick B, Rogers D, Sacks A & Venbrux C. Guidelines for the use of
retrievable and convertible vena cava filters: report from the Society of
Interventional Radiology multidisciplinary consensus conference. J Vasc Interv
Radiol. 2006; 17: 449-459.
Kearon C, Julian JA, Newman TE & Ginsberg JS. Non- invasive diagnosis of deep venous
thrombosis. Ann Int Med. 1998; 128(8): 663-677.
Kearon C, Kahn SR, Agnelli G, Goldhaber S, Raskob GE & Comerota AJ. Antithrombotic
therapy for venous thromboembolic disease: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines (8th Edition). Chest. 2008; 133(6
Suppl):454S-545S.
Klein JS. 2007. Pulmonary vascular disease, In: Fundamentals of diagnostic radiology, Brant WE
& Helms CA., pp 417-432. Lipincott Williams & Wilkins, ISBN- 13: 978-0-7817-6518-
3, Philadelphia, USA.
Lee KH, Han H, Lee KJ, Yoon CS, Kim SH, Won JY & Lee DY. Mechanical thrombectomy of
acute iliofemoral deep vein thrombosis with the use of an Arrow Trerotola
percutaneous thrombectomy device. J Vas Interv Radiol. 2006; 17(3): 487-495.
Lin PH, Zhou W, Dardik, Mussa F, Kougias P, Hedayata N, Naoum JJ, Sayed HE, Peden EK
& Huynh TT. Catheter-direct thrombolysis versus pharmacomechanical
thrombectomy for treatment of symptomatic lower extremity deep venous
thrombosis. Am J Surg. 2006; 192(6): 782-788.
Linkins LA, Pasquale P, Paterson S & Kearon C. Change in thrombus length on venous
ultrasound and recurrent dep vein thrombosis. Arch Intern Med 2004; 164: 1973-
1796.
Moser KM, Fedullo PF, Littlejohn JK & Crawford R. Frequent asymptomatic pulmonary
embolism in patients with deep venous thrombosis. JAMA. 1994; 271(3): 223-225.
Nazir SA, Ganeshan A, Nazir S & Uberoi R. Endovascular treatment options in the
management of lower limb deep venous thrombosis. Cardiovasc Intervent Radiol.
2009; 32: 861-876.
Oguzkurt L, Ozkan U, Ulusan S, Koc Z & Tercan F. Compression of left common iliac vein
in asymptomatic subjects and patients with left iliofemoral deep vein thrombosis. J
Vasc Interv Radiol. 2008; 19: 366-371.
O’Shaugnessy AM & Fitzgerald DE. Determining the stage of organisation and natural
history of venous thrombosis using computer analysis. Int Angiol. 2000a; 19(3): 220-
227.
Radiological Imaging and Intervention in Venous Thrombosis 97
O’Shaugnessy AM & Fitzgerald DE. The value of computer analysis in predicting the long
term outcome of deep venous thrombosis. Int Angiol. 2000b; 19(4): 308-313.
O’Sullivan GJ. The role of interventional radiology in the management of deep venous
thrombosis: advanced therapy. Cardiovasc Intervent Radiol. 2011 ;34(3):445-61.
Pianta MJ & Thomson KR. Catheter-directed thrombolysis of lower limb thrombosis.
Cardiovasc Intervent Radiol. 2011; 34(1):25-36.
Prandoni P, Lensing AWA & Bernardi E. The diagnostic value of compression
ultrasonograpy in patients with suspected recurrent deep vein thrombosis. Thromb
Haemost. 2002; 88: 402-406.
Rademaker J, griesshaber V, Hidajat N, Oestmann JW & Felix R. Combined CT pulmonary
angiography and venography for diagnosis of pulmonary embolism and deep vein
thrombosis: radiation dose. Journal Thoracic Imaging. 2001; 16: 297-299.
Razavi MK,Hansch EC, Kee ST, Sze DY, Semba CP & Dake MD. Chronically occluded
inferior vena cavae: endovascular treatment. Radiology. 2000; 214: 133-138.
Remi- Jardin, Remy J, Baghaie F, Fribourg M, Artoud D & Duhamel A. Clinical value of thin
collimation in the diagnostic work up of pulmonary embolism. Am J Roentgenol.
2000; 175: 407-411.
Righini M. Is it worth diagnosing and treating distal deep venous thrombosis? No. J Thromb
Haemost. 2007; 5(suppl 1): 55-59.
Righini M, Le gal G, Aujesky D, roy PM, Sanchez O, Verschuren F, Rutschmann O, Nonent
M, Cornuz J, Thys F, Le Manach CP, Revel MP, Polleti PA, Meyer G, Mottier G,
Perneger T, Bounameaux H & Perrier A. Diagnosis of pulmonary embolism by
multidetector CT alone or combined with venous ultrasonography of the leg: a
randomised non- inferiority trial. Lancet. 2008; 371(9621): 1343-1352.
Righini M, Le Gal G, Aujesky D, Roy PM, Sanchez O & Verschuren F. Complete venous
ultrasound in outpatients with suspected pulmonary embolism. J Thromb
Haemost. 2009; 7(3): 406- 412.
Roditi G & Fink C. Venous MR imaging with blood pool agents. Eur Rad 2009; 18(suppl 5):
3-12.
Schellong SM. Distal DVT: worth diagnosing. Yes. J Thromb Haemost. 2007; 5(suppl 1): 51-
54.
Schoepf UJ & Costello P. CT angiography for diagnosis of pulmonary embolism: state of the
art. Radiology. 2004; 230(2): 329-337.
Schoepf UJ, Kucher N, Kipfmueller F, Quiroz R, Costello P & Goldhaber SZ. Circulation.
2004; 110: 3276- 3280.
Sevitt S & Gallagher N. Venous thrombosis and pulmonary embolism. A clinico-
pathological study in injured and burned patients. British Journal Surgery. 1961; 48:
475-489.
Spritzer CE. Progress in MR imaging of the venous system. Perspectives in vascular surgery
and endovascular therapy. 2009; 21(2): 105-116.
Streiff MB. Vena caval filters: a comprehensive review. Blood. 2000; 95: 3669-3677.
Vedantham S, Millward SF, Cardella JF, Hofmann LV, Razavi MK, Grassi CJ, Sacks D &
Kinney TB. Society of Interventional Radiology position statement: treatment of
98 Deep Vein Thrombosis
acute iliofemoral deep vein thrombosis with use of adjunctive catheter directed
intrathrombus thrombolysis. J vasc Interven Rad. 2004; 20 (Suppl 7):332-335.
Wells PS, Hirsh J, Anderson DR, Lensing AW, Foster G, Kearon C, Weitz J, D’Ovidio R,
Cogo A, Prandoni P, Girolami A & Jinsberg A. Accuracy of clinical assessment of
deep venous thrombosis. Lancet. 1995; 345: 1315-1380.
6
1. Introduction
This chapter addresses a new and emerging aspect of health in developing countries—one
that poses a serious and growing burden on individuals, health systems, and economies of
poor countries but is largely preventable. Deep Vein thrombosis (DVT) is a major medical,
social and economic problem in developed countries, but in developing countries scanty
information is available. Blood clots such as thrombus in a deep vein in the lower limb is the
most serious unexpected killer of hospitalized patients in developed countries and over the
years this has led to elaboration of numerous strategies directed towards reducing the risks
of formation of such thrombi and treating them when they occur. This area has been
covered extensively in the literature emerging from developed countries, and little is known
about the pattern and scale of problem in developing countries.
Another area that will be covered in this chapter relates to hypercoagulation in chronic liver
disease which is poorly understood till recently. Because of the relatively uncommon
occurrence of overt clinical thrombosis in patients with liver disease, and the complexity of
the haemostatic mechanism, in addition to the fact that clinicians often perceive that these
patients are at a reduced risk for venous thromboembolism, DVT in liver disease is an
understudied problem. In this chapter, we aim to discuss DVT from two aspects; DVT in
liver disease, and DVT in developing countries.
Initiation
TF
Fibroblast VIIa
IXa
Xa
Prothrombin Propagation
Thrombin
Thrombin
Platelet
Activated platelets
Amplification
Fig. 1. Cell-based model of the mechanism of blood coagulation
Emerging Issues in Deep Vein Thrombosis; (DVT) in Liver Disease and in Developing Countries 101
The classical cascade model of the coagulation cascade is being replaced by the new, cell-
based model of coagulation (Roberts et al.,2006 ) (Fig. 1), which emphasizes the interaction
of coagulation proteins with cell surfaces of platelets, subendothelial cells and the
endothelium. According to this model the coagulation is initiated (The Initiation Phase) by
the formation of a complex between tissue factor (TF) exposed on the surface of fibroblasts
as a result of a vessel wall injury, and activated factor VII (FVIIa), normally present in the
circulating blood. The TF-FVIIa complexes convert FX to FXa on the TF bearing fibroblasts.
FXa then activates prothrombin (FII) to thrombin (FIIa). The next phase is the Amplification
Phase in which this limited amount of thrombin activates FVIII, FV, FXI and platelets, on the
surface of blood platelets. Thrombin-activated platelets change shape, and as a result will
expose negatively charged membrane phospholipids, which form the perfect template for
the assembly of various clotting factors and full thrombin generation involving FVIIIa and
FIXa (The Propagation Phase). According to this cell-based model the tissue factor (TF)
extrinsic pathway is the principal cellular initiator of normal blood coagulation in vivo
(Mackman et al. 2007 ), and the major regulator of haemostasis and thrombogenesis, with
the intrinsic pathway, playing an amplification role.
mesenteric systems (Mammen et al., 1992), hepatic veins (Singh et al., 2000), and
peripherally in the extremities with associated pulmonary emboli (Northup et al., 2006). The
prothrombotic state may be involved in other sequelae of chronic liver disease, including
hepatic parenchymal extinction, fibrosis and portopulmonary hypertension. Thus, a
prolonged prothrombin time does not adequately portray the levels of other clotting factors,
particularly factors VIII, X and II that can be more than adequate to promote clot formation
(Violi et al., 1995). As well, it is known that the coagulation disorders associated with falling
liver can induce further hepatic damage, namely, parenchymal extinction. Wanless et al
(Wanless et al., 1995) have clearly demonstrated the histopathologic evidence of the
secondary hepatic damage caused by circulatory disturbances due to thrombotic occlusion
of intrahepatic blood vessels (microvascular thrombosis).
Given this to be the situation in the developed countries, the the magnitude of the problem
would be much lower in the developing countries. Indeed many population studies that are
carried in Western developed countries documented the lower incidence of VTE in Asians
and Hispanics compared to Caucasians (Kearon 2001, White et al 2009).
Although there is strong evidence that the prevalence of venous thrombo-embolism (VTE)
varies significantly among different ethnic/racial groups, the genetic, physiologic and/or
clinical basis for these differences remain largely undefined (White et al ., 2009).
Identifying the scale of DVT in developing countries is difficult due to scanty and conflicting
available published literature on the scale of the problem, the diagnostic tools, management
and treatment challenges facing these countries. Most published information on the DVT
was generated from small hospital-based studies that documented DVT as a significant
complication of orthopedic surgery particularly total knee arthroplasty (Chung et al 2010,
Ko et al. 2003, Leizorovicz et al 2005, Sen et al 2011, Sen et al 2011), and general hospital
patients (Ogeng'o et al 2001, Angchaisuksiri et al 2007, Sakon et al. 2006, Lee et al. 2009).
Essentially all these and other similar studies advocated the importance of
thrombohphylaxis to avoid the risk of VTE.
As to population studies very few could be identified and almost all from Asian Far Eastern
countries particularly China and Korea. In one study from Korea the incidence of VTE, DVT
and PE per 100,000 individuals was found to be 8.83, 3.91 and 3.74 in 2004 and increased to
13.8, 5.31 and 7.01 in 2008 (Jang et al 2001). Another recent study from Hong Kong
documented an annual incidence of of VTE at 16.6 events per 100,000 populations (Lui et al
2002). Another Chinese study reported the incidence of DVT and PE of 17.1 and 3.9 per
100,000 populations (Cheuk et al 2004). The incidence of DVT in all three studies is almost
one tenth that reported from developed counties; yet the problem of DVT remains to be a
health problem that clinicians should be aware of.
3.2.2 Registries
In reviewing the available evidence on the epidemiology of deep vein thrombosis (DVT) in
the developing countries, it is quite clear that there are few on-going registries that track
data on patients with DVT. Most of those registries are hospital-based rather than national.
For example in Saudi Arabia there is the Saudi Thrombosis and Familial Thrombophilia (S-
TAFT) Registry (Saour et al., 2009), which is considered the only registry in Gulf Region and
Emerging Issues in Deep Vein Thrombosis; (DVT) in Liver Disease and in Developing Countries 105
perhaps the Middle East. In developing countries there is very scanty and non-conclusive
data on the prevalence, incidence, risk factors, genetic predisposition, distribution of DVT
occurrences among different age groups and gender, and the burden of DVT on different
patient groups (e.g. post-surgical, pregnancy etc…). Most importantly, how physicians
manage DVT is also unknown and no cost-effective analysis is available on the current
treatment regimens deployed in these countries. Such registry for DVT should include
demographic data and extensive medical history (past and present). Detailed information on
environmental, lifestyle and occupational factors could help identifying certain groups who
are at increased risk of developing DVT or its complications. There is also need to
accumulate laboratory data which should include blood group, factor VIII, inherited
thrombophilic defects (such as factor V Leiden and prothrombin mutations), fibrinogen
level, as well as routine laboratory investigations. Screening for inherited thrombophilia and
other genetic diseases that predispose to DVT is crucial and has gained popularity
worldwide. The available data on the prevalence of thrombophilic risk factors for VTE,
particularly factor V Leidin, prothrombin G20210A, mutations C677T methylenetetra-
hydrfolate reductase and hyperhomocysteinaemia) in developing countries is scanty but
agree on their rarity and much lower prevalence than in developed countries (Jun et al 2006,
et al 2002, Lim et al 2004, Omar et al 2007).
3.2.3 Epidemiology
The burden of DVT in the developing world is unknown due to lack of documentation and
large-scale research projects aiming at identifying the different epidemiology aspects. Some of
the developing countries (Saudi Arabia, United Arab of Emirates and the rest of the Arab Gulf
countries), have the financial resources to setup such registries. However, setting up registries
requires substantial training to the current and future personnel who are working fulltime in
maintaining them. Policymakers, represented by the governments, academic medical centers
and, most importantly, local and regional funding agencies, must work together in order to
consider emphasizing DVT as a public health problem so that the appropriate increasing
proportion of public health resources is reallocated to address DVT and its related issues.
4. Conclusion
In conclusion, we believe that addressing DVT as a regional public health problem in the
developing countries should take a multi-dimensional approach targeting the epidemiology
of DVT and implementation of cost-effective preventive and therapeutic programs.
Emerging Issues in Deep Vein Thrombosis; (DVT) in Liver Disease and in Developing Countries 107
5. References
Abdulaziz Aldawood, Yaseen Arabi, Abdulrahman Aljumah, Alawi Alsaadi, Asgar Rishu,
Hasan Aldorzi, Saad Alqahtani, Mohammad Alsultan, and Afaf Felemban. The
incidence of venous thromboembolism and practice of deep venous thrombosis
prophylaxis in hospitalized cirrhotic patients. Thromb J. 2011; 9: 1
Abeer [Link]-Ghumlas AK, Gader AMA, Al Faleh FZ. Haemostatic abnormalities in liver
disease: could some haemostatic tests be useful as liver function tests? Blood Coag
Fibrinol. 2005;16:329-335
Al Sayegh F, Almahmeed W, Al Humood S, Marashi M, Bahr A, Al Mahdi H, Bakir S, Al
Farhan M. Global Risk Profile Verification in Patients with Venous
Thromboembolism (GRIP VTE) in 5 Gulf countries. Clin Appl Thromb Hemost.
2009 May-Jun;15(3):289-96.
Anderson FA Jr, Wheeler HB, Goldberg RJ, Hosmer DW, Patwardhan NA, Jovanovic B,
Forcier A, Dalen JE. A population-based perspective of the hospital incidence and
case-fatality rates of deep vein thrombosis and pulmonary embolism. The
Worcester DVT Study. Arch Intern Med 1991; 151: 933–8.
Angchaisuksiri P, Atichartakarn V, Aryurachai K, Archararit N, Rachakom B, Atamasirikul
K, Tiraganjana A. Risk factors of venous thromboembolism in thai patients. Int J
Hematol. 2007 Dec;86(5):397-402.
Angchaisuksiri P, Pingsuthiwong S, Sura T, Aryuchai K, Busabaratana M, Atichartakarn V.
Prevalence of the C677T methylenetetra- hydrofolate reductase mutation in Thai
patients with deep vein thrombosis. Acta Haematol. 2000;103(4):191-6.
Bechmann Lars P. , Matthias Sichau, Marc Wichert, Guido Gerken1, Knut Kro¨ ger, and
Philip Hilgard. Low-molecular-weight heparin in patientswith advanced cirrhosis.
Liver International 2010 :ISSN 1478-3223
Ben Ari Z, Panagou M, Patch D, Bates S, Osman E, Pasi J et al. (1997) Hypercoagulability in
patients with primary biliary cirrhosis and primarysclerosing cholangitis evaluated
by thrombelastography. J Hepato l26:554–559.
Cushman M, Tsai AW, White RH, Heckbert SR, Rosamond WD, Enright P, Folsom AR.
Deep vein thrombosis and pulmonary embolism in two cohorts: the longitudinal
investigation of thromboembolism etiology. Am J Med 2004; 117:19–25.
Chung LH, Chen WM, Chen CF, Chen Th, Liu CL. Deep Vein Thrombosis after total knee
arthoplasty in asian patients without prophylactic [Link].
2011 Jan 3;34(1):15.
Cheuk BL, Cheung GC, Cheng SW. Epidemiology of Venous Thromboembolism in a
Chinese Population. Br J Surg. 2004 Apr;91(4):424-8.
De Caterina M, Tarantino G, Farina C, Arena A, Di Maro G, Esposito P, Scopacasa F.
Haemostasis unbalance in Pugh-scored liver cirrhosis: characteristic changes of
plasma levels of protein C versus protein S. Haemostasis, 1993; 23: 229–35.
Dhillon KS, Askander A, Doraismay S. Postoperative Deep-Vein Thrombosis in Asian
patients is not a rarity: a prospective study of 88 patients with no prophylaxis. J
Bone Joint Surg Br. 1996 May;78(3):427-30.
Gader AA, Haggaz AE, Adam I. Epidemiology of Deep Venous Thrombosis during
pregnancy and puerperium in Sudanese Women. Vasc Health Risk Manag.
2009;5(1):85-7
108 Deep Vein Thrombosis
Geerts WH, Bergqvist D, Pineo GF, Heit JA, Samama CM, Lassen MRet al. (2008) Prevention
of venous thromboembolism: American Collegeof Chest Physicians Evidence-
Based Clinical Practice Guidelines (8thedition). Chest 133 (6 Suppl.):381S–453S.
Gunawan B, Runyon B. The efficacy and safety of epsilon-aminocaproic acid treatment in
patients with cirrhosis and hyperfibrinolysis. Aliment Pharmacol Ther 2006; 23:
115–20.
Hawkins D. Pharmacoeconomics of thrombosis management. Pharmacotherapy. 2004;24(7
pt 2):95S-99S.
Jang MJ, Bang SM, Oh D. Incidence of venous thromboembolism in Korea: from the Health
Insurance Review and Assessment Service database. J Thromb Haemost. 2011
Jan;9(1):85-91.
Jun ZJ, Ping T, Lei Y, Li L, Ming SY, Jing W. Prevalence of factor V Leiden and prothrombin
G20210A mutations in Chinese patients with deep venous thrombosis and
pulmonary embolism. Clin Lab Haematol. 2006 Apr;28(2):111-6.
Kearon C. Epidemiology of venous thromboembolism. Semin Vasc Med. 2001;1(1):7-26.
Kelly J, Hunt BJ., The utility of pretest probability assessment in patients with clinically
suspected venous thromboembolism. J Thromb Haemost. 2003 Sep;1(9):1888-96.
Kochanek KD, Murphy SL, Anderson RN, Scott C. Deaths: final data for 2002. Natl Vital Stat
Rep. 2004; 53 ( 5 ): 1 - 115
Ko PS, Chan WF, Siu TH, Khoo J, Wu WC, Lam JJ. Deep Venous Thrombosis after total hip
or knee arthroplasty in a “low-risk” Chinese population. Arthroplasty. 2003
Feb;18(2)174-9.
Kozak LJ , Owings MF , Hall MJ . National Hospital Discharge Survey: 2002 annual
summary with detailed diagnosis and procedure data . Vital Health Stat 13 . 2005 ;
158 : 1 - 199 .
Lee AD, Stephen E, Agarwal S, Premkumar P. Venous Thromboembolism in India. Eur J
Vasc Endovasc Surg. 2009 Apr;37(4):482-5. Epub 2009 Feb 8.
Leizorovicz A, Turpie AG, Cohen AT, Wong L, Yoo MC, Dans A; SMART Study Group.
Epidemiology of venous thromboembolism in Asian patients undergoing major
orthopedic surgery without thromboprophylaxis. The SMART study. J Thromb
Haemost. 2005 Jan;3(1):28-34.
Lim YW, Chong KC, Chong I, Low CO, See HF, Lam KS. Deep vein thrombosis following
hip fracture and prevalence of hyperhomocysteinaemia in the elderly. Ann Acad
Med Singapore. 2004 Mar;33(2):235-8.
Liu HS, Kho BC, Chan JC, Cheung FM, Lau KY, Choi FP, Wu WC, Yau TK. Venous
thromboembolism in the Chinese population--experience in a regional hospital in
Hong Kong. Hong Kong Med J. 2002 Dec;8(6):400-5.
Mackman N, Tilly RE, Key NS. The role of the extrinisic pathway of blood coagulation in
hemostasis and thrombosis. Arterioscler Thromb Vasc Biol. 2007;27:1687-1693
Mammen EF. Coagulation abnormalities in liver disease. Hematol Oncol Clin North Am
1992; 6: 1247–57.
Marco Senzolo, Maria Teresa Sartori , Ton [Link] we give thromboprophylaxis to
patients with liver cirrhosis and coagulopathy?.HPB 2009, 11, 459–464.
Misumi K, Ogawa H, Yasue H, Soejima H, Suefuji H, Nishiyama K, Takazoe K, Kugiyama
K, Tsuji I, Kumeda K, Nakamura S. Comparison of plasma tissue factor levels in
unstable and stable angina pectoris. Am J Cardiol. 1998;81:22–26
Emerging Issues in Deep Vein Thrombosis; (DVT) in Liver Disease and in Developing Countries 109
Shet AS, Aras O, Gupta K, Hass MJ, Rausch DJ, Saba N, Koopmeiners L, Key NS, Hebbel
RP. Sickle blood contains tissue factor-positive microparticles derived from
endothelial cells and monocytes. Blood. 2003;102:2678 –2683
Singh V, Sinha SK, Nain CK, Bambery P, Kaur U, Verna S, Chawla YK, Singh K, Budd-Chiari
syndrome: our experience of 71 patients. J GastroenrolHepatol 2000;15: 550-4.
Stein PD, Patel KC, Kalra NK, Petrina M, Savarapu P, Furlong JW Jr, Steele RD Jr, Check FE.
Estimated incidence of acute pulmonary embolism in a community/teaching
general hospital. Chest 2002; 121: 802–5.
Tesselaar ME, Romijn FP, van dL, I, Prins FA, Bertina RM, Osanto S Microparticle-associated
tissue factor activity: a link between cancer and thrombosis J Thromb Haemost.
2007 Mar;5(3):520-7.
Violi F, Ferro D, Basili S, Cimminiello C, Saliola M, Vezza E, Cordova C. Prognostic value of
clotting and fibrinolytic systems in a follow-up of 165 liver cirrhotic patients. CALC
Group. Hepatology 1995; 22: 96–100.
Violi F, Ferro D, Basili S, D’Angelo A, Mazzola G, Quintarelli C, Cordova C. Relation
between lupus anticoagulant and splanchnic venous thrombosis in cirrhosis of the
liver. BMJ 1994; 309: 239–40.
Vukovich T, Teufelsbauer H, Fritzer M, Kreuzer S, Knoflach P. Hemostasis activation in
patients with liver cirrhosis. Thromb Res1995; 77: 271–8.
Walker FJ. Protein C deficiency in liver disease. Ann Clin Lab Sci 1990; 20: 106–12.
Wanless IR, Wong F, Blendis LM, Greig P, Heathcote EJ, Levy G. Hepatic and portal vein
thrombosis in cirrhosis: possible role in development of parenchymal extinction
and portal hypertension. Hepatology 1995; 21: 1238–47.
Wang CJ, Wang JW, Chen LM, Chen HS, Yang BY, Cheng SM. Deep Vein Thrombosis after
total knee athroplasty. J Formos Med Assoc. 2000 Nov;99(11):848-53
Wells PS, Anderson DR, Bormanis J, Guy F, Mitchell M, Gray L, et al. Value of assessment of
pretest probability of deep-vein thrombosis in clinical management. Lancet
1997;350:1796.
White RH, Keenan CR. Effects of race and ethnicity on the incidence of venous
thromboembolism. Thromb Res. 2009;123 Suppl 4:S11-7).
World Health Organization The world health report 2006: Working Together For Health.
ISBN 92 4 156317 6 (NLM classification: WA 530.1)
Zenzolo M, Sartori MT, Lisman T. Should we give thromboprophylaxis to patients with liver
cirrhosis and coagulopathy? HPB (Oxford). 2009 Sp;11(6):459-65.
Zurborn KH, Kirch W, Bruhn HD. Immunological and functional determination of the
protease inhibitors, protein C and antithrombin III, in liver cirrhosis and in
neoplasia. Thromb Res 1988; 52: 325–36.
7
Venous Thromboembolism
Prophylaxis in Cancer Patients
Hikmat Abdel-Razeq
King Hussein Cancer Center, Amman,
Jordan
1. Introduction
Venous Thromboembolism (VTE) is a common disease, comprising the life-threatening
pulmonary embolism (PE) and its precursor deep vein thrombosis (DVT). In view of the
clinically silent nature of VTE; the incidence, prevalence and mortality rates are probably
under estimated (Kniffin et al., 1994). Although VTE is a common disease, fortunately it is
preventable; identifying high risk patients and the application of suitable prophylactic
measures is the best way to decrease the incidence of VTE and its associated complications.
Using unfractionated heparin (UFH), the rate of radiologically detected DVT was reduced
by 67% without significant bleeding complications (Belch et al., 1981).
Although most patients survive DVT, they often suffer serious and costly long-term
complications. Venous stasis syndrome (postphlebitic syndrome) with painful swelling and
recurrent ulcers is well known complication following DVT (Prandoni et al., 1996).
Additionally, PE is associated with substantial morbidity and mortality both tend to be
higher among cancer patients and those who survive such event may develop chronic
complications like pulmonary hypertension (Carson et al., 1992; Pengo et al., 2004). In a
large study, Sørensen et al. examined the survival of patients with cancer and VTE
compared to those without VTE matched for many factors including the type and duration
of cancer diagnosis; the one year survival rate for cancer patients with VTE was 12%
compared to 36% in the control group (P<0.001). Furthermore, the risk of VTE recurrence
was higher in cancer patients compared to those without (Sørensen et al., 2000).
stomach, ovary, lung, prostate, and kidney (Chew et al., 2006; Gerber DE, et al., 2006; Marras
et al., 2000; Sallah et al., 2002; Thodiyil& Kakkar, 2002), but lower in sites like skin and breast
(Andtbacka et al., 2006; Chew HK et al., 2007). In addition to primary tumor type, other
cancer-related factors play important role in VTE rates; the risk of VTE is highest during the
first 3–6 months after the initial diagnosis of cancer (Blom et al., 2005). Such risk also varies
with the stage of the disease; much higher with advanced stage compared to early stage
disease (Blom JW et al., 2005) and among cancer patients on active treatment with
chemotherapy or radiotherapy (Haddad & Greeno, 2006).
Certain anti-cancer therapies are known to increase the risk of VTE in cancer patients. The
rate of VTE increases by two to five folds in women with breast cancer treated with
tamoxifen, a selective estrogen receptor modulator (SERM), and this risk was even higher
when tamoxifen was combined with chemotherapy, a practice that was abandoned many
years ago (Fisher et al., 2005; Pritchard et al., 1996). Aromatase inhibitors (AI), however, like
anastrozole, letrozole and exemestane are less thrombogenic (Breast International Group
(BIG) 1–98 Collaborative Group, 2005; ATAC (Arimidex Tamoxifen Alone or in
Combination Trialists’ Group), 2002).
Cancer type:
High: Brain, Ovary, Pancreas, Colon, Stomach, Lung, Prostate, Kidney
Low: Skin, Thyroid, Breast
Duration since cancer diagnosis:
High: First 6 months
Low: After 12 months
Stage of disease:
High: Locally-advanced and metastatic disease
Low: Early-stage
Anticancer therapy:
High: Chemotherapy, radiotherapy, surgery
Low: No active treatment
Hormonal therapy:
High: Tamoxifen
Low: Aromatase inhibitors like letrozole, anastrozole and exemestene
Antiangiogenesis and immune modulators:
Thalidomide
Lenalidomide
Bevacizumab
Sorafenib
Sunitinib
Table 1. Cancer-related risk factors for thrombosis
thrombotic events (Kabbinavar et al., 2007; Shah et al, 2005). Lower rates, however,
wereobserved when different combination chemotherapy were used in the treatment of
non-small cell lung cancer (Johnson et al., 2004). In addition to thrombosis, bevacizumab
was associated with significant increase risk of bleeding; a fact that complicates decision
making (Kabbinavar et al., 2003) . However, the highest incidence of VTE was observed in
multiple myeloma patients treated with thalidomide, dexamethasone and doxorubicin-
containing chemotherapy [Zangari et al., 2002). Higher VTE rates were also observed with
thalidomide derivatives; lenalidomide and pomalidomide when used in combination with
dexamethasone (Zonder et al., 2006 & Dimopoulos et al., 2007). Cancer-related risk factors
are summarized in table-1.
Different antithrombotics including low molecular weight heparin (LMWH), low-dose
warfarin, full-dose warfarin with target international normalized ratio (INR) of 2 to 3, and
aspirin (ASA) were all tried to reduce the risk of VTE in cancer patients undergoing such
therapy (Baz et al., 2005; Cavo M et al., 2004; Minnema et al., 2004). Specific
recommendations in these clinical settings are beyond the scope of this review. However, in
a recent trial that included a total of 667 patients with previously untreated multiple
myeloma who received thalidomide-containing regimens and had no clinical indication or
contraindication for a specific antiplatelet or anticoagulant therapy were randomly assigned
to receive ASA (100 mg/d), fixed-dose warfarin (1.25 mg/d), or LMWH (enoxaparin 40
mg/d). A composite primary end point included serious thromboembolic events, acute
cardiovascular events, or sudden deaths during the first 6 months of treatment; of 659
analyzed patients, 43 (6.5%) had serious thromboembolic events, acute cardiovascular
events, or sudden death during the first 6 months (6.4% in the ASA group, 8.2% in the
warfarin group, and 5.0% in the LMWH group). Compared with LMWH, the absolute
differences were +1.3% (95% CI, - 3.0% to 5.7%; P =.544) in the ASA group and +3.2% (95%
CI, - 1.5% to 7.8%; P = .183) in the warfarin group (Palumbo et al., 2011).
In addition to chemotherapy agents, drugs that are commonly used to support cancer
patient while on active treatment may increase the risk of VTE. Erythropoiesis-stimulating
agents (ESA); erythropoietin and darbepoietin are both associated with higher VTE rates. A
meta-analysis of 35 trials in 6,769 cancer patients concluded that such treatment increased
the risk of thromboembolic events by 67% compared with patients not receiving this therapy
(Bohlius et al., 2006).
(Kahn et al., 2007). Similar findings were also reported in the IMPROVE study in which only
45% of cancer patients who either met the ACCP criteria for requiring prophylaxis or were
eligible for enrollment in randomized clinical trials that have shown the benefits of
pharmacologic prophylaxis actually received prophylaxis [Tapson et al., 2007]. In another
study conducted by our group, two hundred cancer patients with established diagnosis of
VTE were identified; majority (91.8%) had advanced-stage cancer at time of VTE diagnosis.
In addition to cancer, many patients had multiple coexisting risk factors for VTE with 137
(68.5%) patients had at least three, while 71 (35.5%) had four or more. Overall, 111(55.5%)
patients developed lower-extremity DVT while 52 (26%) patients developed PE, other sites
accounted for 18%. Almost three quarters of the patients (73.5%) had not received any
antecedent prophylaxis. Prophylaxis rate was 23% among patients with >3 risk factors and
50% among the highest risk group with >5 risk factors (Abdel-Razeq et al., 2011).
Compared to surgical patients, decisions on when to offer prophylaxis in cancer patients
admitted to medical units is difficult to make (Monreal et al., 2004); medical patients
typically have many risk factors, the interaction of which is difficult to quantify. In a recent
survey, The Fundamental Research in Oncology and Thrombosis (FRONTLINE), marked
differences were seen in the use of thromboprophylaxis for surgical and medical cancer
patients, with over 50% of surgeons reporting that they initiated thromboprophylaxis
routinely, while most medical oncologists reported using thromboprophylaxis in less than
5% of medical cancer patients (Kakkar et al., 2003). These studies and many others (Chopard
et al., 2005; Ageno et al., 2002), demonstrate that VTE prophylaxis in cancer patients is still
underutilized.
Many factors may contribute to the low VTE prophylaxis rate in cancer patients. Obviously,
concerns about bleeding especially in patients undergoing active treatment with
chemotherapy that can lead to low blood counts is one of these reasons; this issue was
evident in our study patients where 113 (18.6%) had prolonged PT and or PTT and another
92 (15.2%) had platelet counts < 100 K (Abdel-Razeq et al., 2001). While these may not
represent absolute or even relative contraindications for using anticoagulants for VTE
prophylaxis, nevertheless, such factors may prevent physicians from prescribing
anticoagulant prophylaxis for cancer patients. Other reasons may include concerns about
higher bleeding risks from tumor metastasis in vital structures like the brain. Such patients
can be offered mechanical methods if anticoagulants deemed contraindicated. However, the
absence of a suitable risk assessment model may also contribute to such low prophylaxis
rate; such risk assessment model should take into account the additive or even the
synergistic effect of the many other additional risk factors that cancer patients are typically
admitted with.
Caprini et al. had established a risk assessment model to help health professionals in
making the decision on when and how to prescribe VTE prophylaxis (Caprini et al., 2001 ;
Motykie et al., 2000). Though we found it useful, we faced several limitations when we tried
to apply such model in cancer patients. All cancer patients were given the same risk score;
while in fact type of cancer, stage, nature of anti-cancer therapy and time since cancer
diagnosis are, as discussed above, important factors that affect VTE rate in cancer patients
(Abdel-Razeq et al., 2010).
Venous Thromboembolism Prophylaxis in Cancer Patients 115
4. Published guidelines
Several clinical and scientific groups including the ACCP (Geerts et al., 2008), the American
Society of Clinical Oncology (ASCO) (Lyman et al., 2007) and the National Comprehensive
Cancer Network (NCCN) (Wagman et al., 2008) have established guidelines for VTE
prophylaxis in cancer patients. All have different and somewhat conflicting
recommendations but all lack a risk assessment model. While the ACCP guidelines were
very conservative and advised prophylaxis for cancer patients who are bedridden with an
acute medical illness, the NCCN, on the other hand, lowered their threshold for VTE
prophylaxis; their most recent updated guidelines stated: ‘‘The panel recommends
prophylactic anticoagulation therapy for all inpatients with a diagnosis of active cancer (or
for whom clinical suspicion of cancer exists) who do not have a contraindication to such
therapy (category 1).’’ Their recommendation was based on an assumption that ambulation
in hospitalized cancer patients is inadequate to reduce VTE risk (Wagman et al., 2008). The
ASCO guidelines published in 2007 have taken a more neutral position by stating in their
summary conclusions: “Hospitalized patients with cancer should be considered candidates
for VTE prophylaxis with anticoagulants in the absence of bleeding or other
contraindications to anticoagulation” (Lyman et al., 2007).
The concept of VTE prophylaxis for ambulatory cancer patients was tested in a recent
double-blind study; patients with metastatic or locally advanced cancer of lung, colo-rectal,
stomach, ovary, pancreas, or bladder who are initiating a new chemotherapy course, were
randomized to receive subcutaneous semuloparin ( a new ultra low molecular weight
heparin) or placebo. The drug was given at a dose of 20 mg subcutaneously and continued
until change of chemotherapy. Twenty of the 1,608 patients treated with semuloparin (1.2%)
and 55 of the 1,604 patients treated with placebo (3.4%) had a thromboembolic event,
representing a 64% risk reduction in such event rate (hazard ratio [HR] = 0.36, 95%
confidence interval [CI] 0.21–0.60, p<0.0001, intent-to-treat analysis). Nineteen of 1,589
patients (1.2%) in the semuloparin and 18 of the 1,583 patients (1.1%) in the placebo group
had a major bleeding (HR=1.05, 95%CI 0.55 to 1.99) (Agnelli et al., 2011).
More work is needed before taking findings of these studies to clinical practice; as such
ambulatory cancer patients on active chemotherapy may be considered for VTE prophylaxis
based on risk level and clinical judgment.
confidence interval 7-44). Bleeding events were not increased with prolonged compared
with short-term thromboprophylaxis (Rasmussen et al., 2006).
A recent meta-analysis of eligible clinical studies compared safety and efficacy of extended
use of LMWH (for three to four weeks after surgery) versus conventional in-hospital
prophylaxis among patients undergoing major abdominal surgeries. The indication for
surgery was neoplastic disease in 70.6% (780/1104) of patients. The administration of
extended LMWH prophylaxis significantly reduced the incidence of VTE, 5.93% versus
13.6%, RR 0.44 (CI 95% 0.28 - 0.7); DVT 5.93% versus 12.9%, RR 0.46 (CI 95% 0.29 - 0.74);
proximal DVT 1% versus 4.72%, RR 0.24 (CI 95% 0.09 - 0.67). These superior efficacy results
were obtained with no significant difference in major or minor bleeding between the two
groups: 3.85% in the extended thrombo-prophylaxis group versus 3.48% in the conventional
prophylaxis group; RR 1.12 (CI 95% 0.61 - 2.06) (Bottaro et al., 2008). Given the results of
these studies, one can conclude that extended thromboprophylaxis with LMWH should be
considered as a safe and useful strategy to prevent VTE in high-risk major abdominal and
pelvic surgeries especially in cancer patients. Similar conclusions were reached in a more
recent Cochrane database analysis (Rasmussen et al., 2009). Results of these studies are
summarized in Figure-1.
Fig. 1. Extended out-of-hospital VTE prophylaxis for cancer patients undergoing major
surgery
118 Deep Vein Thrombosis
Several clinical trials have addressed the issue of VTE prophylaxis in such patients. One
study showed a benefit in reducing VTE events when low fixed-dose warfarin (1mg/day)
was used for prophylaxis (Bern et al., 1990). However, two subsequent clinical trials failed to
show any benefit [Heaton et al., 2002: Couban et al., 2005).
Low molecular weight heparin was also tried in two large, double-blind clinical trials (Verso
et al., 2005; Karthaus et al., 2006). The first trial failed to show beneficial effect of enoxaparin
when used at a dose of 40 mg once daily versus placebo in a group of 385 cancer patients
with CVC (Verso et al., 2005). In the second trial, dalteparin at 5,000 units once daily was
tested against placebo in 439 cancer patients who were receiving chemotherapy through
such catheters; clinically relevant VTE occurred in 3.7% and 3.4% in the dalteparin and
placebo recipients, respectively (Karthaus et al., 2006). Nadroparin, another LMWH, showed
no advantage when tested against low fixed dose of warfarin (1 mg/day) in a small
randomized trial that involved 45 evaluable patients (Mismetti et al., 2003).
Given the results of these studies, thromboprophylaxis with anticoagulants for patients with
central venous catheters is not recommended.
as compared with 21 (11.6%) patients assigned to the no-filter group, had recurrent DVT
(odds ratio, 1.87; 95% CI, 1.10 to 3.20) ( Decousus et al., 1998). This study was updated 8 years
later; patients with IVC filters experienced a greater cumulative incidence of symptomatic
DVT (35.7%versus 27.5%; HR 1.52, CI 1.02 to 2.27; P = 0.042), but significantly fewer
symptomatic pulmonary emboli (6.2%versus 15.1%; HR 0.37, CI 0.17 to 0.79; P = 0.008) (The
PREPIC Study Group, 2005). The conclusion from this long-term follow-up was similar to the
original report; that is, with an IVC filter there is an equivalent trade-off of fewer PE at the cost
of more DVTs. There was no difference in long-term morbidity or mortality in both groups.
Main Indications:
Failure of anticoagulation: Recurrent VTE despite anticoagulation
Contraindications and/or severe complications of anticoagulation:
High risk for bleeding
Real bleeding (GI,GU,GYN, CNS)
Thrombocytopenia (Depends on count and etiology)
Immediate post-operative VTE
Large CNS Tumor: Primary or metastatic
Other indications:
Large, free-floating iliocaval thrombus
Limited cardiopulmonary reserve (Cor Pulmonale)
Poor compliance with medications
Patients at risk for falls while on anticoagulation therapy
IVC: Inferior Vena Cava, GI: Gastrointestinal, GU: Genitourinary, GYN: Gynecological, CNS: Central
Nervous System, VTE: Venous Thromboembolism
Given the lack of long term benefits of IVC filters; temporary, retrievable filters had gained
increasing interest. Many different retrievable filters had recently received approval for
temporary insertion. Recent data suggest that the use of these filters may be associated with
low rates of PE and insertion complications (Imberti & Prisco, 2008). Nevertheless; no
randomized clinical trials have been performed. In one large retrospective study that
included 252 evaluable patients who had retrievable filter placed for different indications;
only 47 filters were successfully retrieved yielding a retrieval rate of 18.7% ( Dabbagh et al.,
2010). Similar or higher retrieval rates were reported by others (Mismetti et al., 2007).
Regardless of the type of the filter placed, the most recent American Colleague of Chest
Physicians (ACCP) guidelines recommend systemic anticoagulation, when possible, even
with the filter in place (Kearon et al., 2008).
Cancer itself, or its treatment, might result in certain clinical complications that make
systemic anticoagulation very risky (Abdel-Razeq et al., 2011). Venous thromboembolic
disease is a frequent complication in patients with intracranial malignancies. Many of the
primary brain tumors like gliomas or secondary metastatic tumors to the brain are either
bulky or very vascular thus increasing the risk of bleeding with or without systemic
anticoagulation (Ruff & Posner, 1983). Brain metastases from melanoma, choriocarcinoma,
thyroid carcinoma, and renal cell carcinoma have particularly high propensities for
120 Deep Vein Thrombosis
spontaneous hemorrhage while metastatic tumors from sites like lung and breast are less
likely to bleed spontaneously (Mandybur, 1993). However, not all patients with intracranial
malignancies are at higher risk of bleeding with anticoagulation. Complication rate of IVC
filters in patients with brain tumors is higher than commonly perceived and may outweigh
the risk of anticoagulation. Researchers at Brigham and Women's Hospital in Boston
reviewed the records of 49 patients with intracranial malignancies and venous
thromboembolic disease to determine the effectiveness and complications resulting from
systemic anticoagulation or IVC filter placement. Of the 42 patients received IVC filters, a
strikingly high percentage (62%) developed one or more complications; 12% developed
recurrent PE, while 57% developed filter thrombosis, recurrent DVT, or post-phlebitic
syndrome. These complications severely reduced the quality of life of affected patients. Only
15 (31%) patients were treated with anticoagulation, and seven of these received it because
of continued thromboembolic disease. None of these 15 patients had proven hemorrhagic
complications (Levin et al., 1993).
Many recent studies questioned the need to insert IVC filters in cancer patients particularly
those with advanced-stage disease whose survival is short and prevention of PE may be of
little clinical benefit and could be a poor utilization of resources. In one retrospective study
performed to determine the clinical benefit of IVC filter placement in patients with
malignancy, 116 patients who had such filters inserted were included. Ninety one (78%)
patients had stage IV disease, 42 (46%) of them died of cancer within 6 weeks and only16
(14%) were alive at one year (Jarrett et al., 2002).
The benefits of IVC filter placement on overall survival, as measured from the time of VTE
was addressed in a recent retrospective study that examined 206 consecutive cancer patients
with VTE. Patients were classified into 3 treatment groups: anticoagulation-only (n= 62),
IVC filter-only (n=77), or combination of both IVC filter and anticoagulation (n=67). Median
overall survival was significantly greater in patients treated with anticoagulation (13
months) compared with those treated with IVC filters (2 months) or combination of both
(3.25 months; P < 0.0002). IVC patients were at 1.9 times more risk of death than
anticoagulation only (hazard ratio=0.528; 95% confidence interval=0.374 to .745).
Multivariate analysis revealed that performance status and type of thrombus were not
confounders and had no effect on overall survival (Barginear et al., 2009).
In another study, the survival benefit of IVC filters in patients with late-stage malignancy
was evaluated in a group of 5,970 patients who were treated with a primary diagnosis of
malignancy at a tertiary care facility. Retrospective analysis identified 55 consecutive
patients with stage III or IV malignant disease and VTE who received IVC filters. In a case
control study, 16 patients with VTE but without IVC filter were matched for age, sex, type of
malignancy, and stage of disease. Filter placement prevented PE in 52 (94.5%) patients,
however, four (7.3%) of patients had complications related to the procedure; 13 (23.6%)
patients with late-stage cancer survived less than 30 days following placement of the filter;
another 13 (23.6%) patients of this group, however, survived more than one year.
Ambulatory status differed significantly (P = 0.01) between these two subgroups. Authors
concluded that IVC filter placement conferred no survival benefit compared to the control
group and that the survival of such patients with advanced-stage cancer was limited
Venous Thromboembolism Prophylaxis in Cancer Patients 121
primarily by the malignant process (Schunn et al., 2006). Researchers at M.D. Anderson
Cancer center concluded, in a study that included 308 cancer patients with VTE and IVC
filters, that such filters are safe and effective in preventing PE-related deaths in selected
patients with cancer. However, patients with a history of DVT and bleeding or advanced
disease had the lowest survival after IVC filter placement (Wallace et al., 2004).
10. Acknowledgments
The authors would like to thank Ms. Haifa Al-Ahmad and Mrs. Alice Haddadin for their
help in preparing this manuscript.
11. References
Abdel-Razeq H. (2010). Venous thromboembolism prophylaxis for hospitalized medical
patients, current status and strategies to improve. Ann Thorac Med. 5:195-200.
Abdel-Razeq, HN.; Hijjawi, SB.; Jallad, SG.; Ababneh, BA. (2010). Venous
thromboembolism risk stratification in medically-ill hospitalized cancer patients.
A comprehensive cancer center experience. J Thromb Thrombolysis. 30: 286-93.
Abdel-Razeq, H.; Mansour, A.; Ismael, Y.; Abdulelah, H. (2011) Inferior vena cava filters
in cancer patients: to filter or not to filter. Ther Clin Risk Manag. 7:99-102.
Abdel-Razeq, H.; Albadainah, F.; Hijjawi, S.; Mansour, A.; Treish, I. (2011). Venous
Thromboembolism (VTE) in Hospitalized Cancer Patients: Prophylaxis Failure or
Failure to Prophylax. J Thromb Thrombolysis. 3: 107-12.
Ageno, W.; Squizzato, A.; Ambrosini, F. et al. (2002). Thrombosis prophylaxis in medical
patients: a retrospective review of clinical practice patterns. Haematologica 87:746–
50.
Agnelli, G.; Bolis, G.; Capussotti, L. et al. (2006). A clinical outcome based prospective
study on venous thromboembolism after cancer surgery: the ARISTOS project.
Ann Surg 243:89–95.
Agnelli, G.; George, D.; Fisher, K.; Kakkar, AK. et al. (2011). The ultra-low molecular
weight heparin (ULMWH) semuloparin for prevention of venous
thromboembolism (VTE) in patients with cancer receiving chemotherapy: SAVE
ONCO study. J Clin Oncol 29: (suppl; abstr LBA9014)
Andtbacka, RH.; Babiera, G.; Singletary, SE. et al. (2006). Incidence and prevention of
venous thromboembolism in patients undergoing breast cancer surgery and
treated according to clinical pathways. Ann Surg 243:96–101.
ATAC (Arimidex Tamoxifen Alone or in Combination) Trialists’ Group. (2002).
Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for
adjuvant treatment of postmenopausal women with early breast cancer: first
results of the ATAC randomised trial. Lancet 359: 2131-2139
Ay, C.; Simanek, R.; Vormittag, R. et al. (2008). High plasma levels of soluble P-selectin are
predictive of venous thromboembolism in cancer patients: results from the
Vienna Cancer and Thrombosis Study (CATS). Blood 112:2703–8.
Barginear, MF.; Lesser, M.; Akerman, ML. et al. (2009). Need for inferior vena cava filters
in cancer patients: a surrogate marker for poor outcome. Clin Appl Thromb
Hemost. 15:263-269.
Baz, R.; Li, L.; Kottke-Marchant, K. et al. (2005). The role of aspirin in the prevention of
thrombotic complications of thalidomide and anthracycline-based chemotherapy
for multiple myeloma. Mayo Clin Proc 80:1568-74.
Belch, JJ.; Lowe, GDO.; Ward, AG.; Forbes, CD.; Prentice, CRM. (1981). Prevention of deep
vein thrombosis in medical patients by low-dose heparin. Scott Med J 26:115–7.
Venous Thromboembolism Prophylaxis in Cancer Patients 123
Bergqvist, D.; Agnelli, G.; Cohen, AT. et al. (2002). Duration of prophylaxis against venous
thromboembolism with enoxaparin after surgery for cancer. N Engl J Med
346:975– 80.
Bern, MM.; Lokich, JJ.; Wallach, SR. et al. (1990). Very low doses of warfarin can prevent
thrombosis in central venous catheters: a randomized prospective trial. Ann
Intern Med 112,423-8.
Blom, JW.; Doggen, CJ.; Osanto, S.; Rosendaal, FR. (2005). Malignancies, prothrombotic
mutations, and risk of venous thrombosis. JAMA 293:715–22.
Bohlius, J.; Wilson, J.; Seidenfeld, J. et al. (2006). Recombinant human erythropoietins and
cancer patients: updated meta-analysis of 57 studies including 9353 patients. J
Natl Cancer Inst 98:708–14.
Bona, RD. (1999). Thrombotic complications of central venous catheters in cancer patients.
Semin Thromb Haemost 25,147-55.
Bottaro, FJ.; Elizondo, MC.; Doti, C. et al. (2008). Efficacy of extended thrombo-
prophylaxis in major abdominal surgery: what does the evidence show? A meta-
analysis. Thromb Haemost. 99:1104-11.
Breast International Group (BIG) 1–98 Collaborative Group. (2005). A comparison of
letrozole and tamoxifen in postmenopausal women with early breast cancer. N
Engl J Med 353, 2747-57.
Buller, HR.; Davidson, BL.; Decousus, H. et al. (2004). Fondaparinux or enoxaparin for the
initial treatment of symptomatic deep venous thrombosis: a randomized trial.
Ann Intern Med. 140: 867-873.
Caprini, J.; Arcelus, J.; Reyna, J. (2001). Effective risk stratification of surgical and
nonsurgical patients for venous thromboembolic disease. Semin Hematol 38:12–19.
Carson, JL.; Kelley, MA.; Duff, A. et al. (1992). The clinical course of pulmonary embolism.
N Engl J Med 326:1240–5.
Cavo, M., Zamagni, E., Tosi, P., et al. (2004). First-line therapy with thalidomide and
dexamethasone in preparation for autologous stem cell transplantation for
multiple myeloma. Haematologica 89:826-31.
Chen, M., Geng, JG., P-selectin. (2006). mediates adhesion of leukocytes, platelets, and
cancer cells in inflammation, thrombosis, and cancer growth and metastasis. Arch
Immunol Ther Exp 54:75-84.
Chew, HK.; Wun, T.; Harvey, D.; Zhou, H.; White, RH. (2006). Incidence of venous
thromboembolism and its effect on survival among patients with common
cancers. Arch Intern Med 166:458–64.
Chew, HK.; Wun, T.; Harvey, DJ.; Zhou, H.; White, RH. (2007). Incidence of venous
thromboembolism and the impact on survival in breast cancer patients. J Clin
Oncol 25:70–6.
Chopard, P.; Dörffler-Melly, J.; Hess, U. et al. (2005). Venous thromboembolism
prophylaxis in acutely ill medical patients: definite need for improvement. J
Intern Med 257:352–7.
Clark-Pearson, DL.; DeLong, E.; Synan, IS.; Soper, JT.; Creasman, WT.; Coleman, RE.
(1990). A controlled trial of two low-dose heparin regimens for the prevention of
postoperative deep vein thrombosis. Obstet Gynecol 75:684–9.
124 Deep Vein Thrombosis
Couban, S.; Goodyear, M.; Burnell, M. et al. (2005). Randomized placebo-controlled study
of low-dose warfarin for the prevention of central venous catheter-associated
thrombosis in patients with cancer. J Clin Oncol 23, 4063-9.
Dabbagh, O.; Nagam, N.; Chitima-Matsiga, R.; Bearelly, S.; Bearelly, D. (2010). Retrievable
inferior vena cava filters are not getting retrieved: where is the gap? Thromb Res.
126:493-497.
Decousus, H.; Leizorovicz, A.; Parent, F. et al. (1998). A clinical trial of vena caval filters in
the prevention of pulmonary embolism in patients with proximal deep-vein
thrombosis: Prevention duRisque d’Embolie Pulmonaire par Interruption Cave
Study Group. N Engl J Med. 338:409–415.
Dimopoulos, M.; Spencer, A.; Attal, M. et al. (2007). Lenalidomide plus dexamethasone for
relapsed or refractory multiple myeloma. N Engl J Med 357:2123-32.
Donati, MB. (1995). Cancer and thrombosis: from Phlegmasia alba dolens to transgenic
mice. Thromb Haemost 74:278.
Fisher, B.; Costantino, JP.; Wickerham, DL. et al. (2005). Tamoxifen for the prevention of
breast cancer: current status of the National Surgical Adjuvant Breast and Bowel
Project P-1 study. J Natl Cancer Inst 97: 1652-62.
Gallus, AS. (1997). Prevention of post-operative deep leg vein thrombosis in patients with
cancer. Thromb Haemost 78:126–32.
Geerts, WH.; Bergqvist, D.; Pineo, GF. et al. (2008). Prevention of venous
thromboembolism: American College of Chest Physicians evidence- based
clinical practice guidelines (8th edition). Chest 133:381–453.
Gerber, DE.; Grossman, SA.; Streiff, MB. (2006). Management of venous
thromboembolism in patients with primary and metastatic brain tumors. J Clin
Oncol 24:1310–8.
Haddad, TF.; Greeno, EW. (2006). Chemotherapy-induced thrombosis. Thromb Res
118:547–666
Heaton, DC.; Han, DY.; Inder, A. (2002). Minidose (1 mg) warfarin as prophylaxis for
central vein catheter thrombosis. Intern Med J 32: 84-8.
Heit, JA.; Silverstein, MD.; Mohr, DN.; Petterson, TM.; O'Fallon, WM.; Melton, LJ .; 3rd.
(2000). Risk factors for deep vein thrombosis and pulmonary embolism: a
population based case-control study. Arch Intern Med 160:809–15.
Imberti, D.; Prisco, D. (2008). Retrievable vena cava filters: key considerations. Thromb Res.
122:442-449.
Jarrett, BP.; Dougherty, MJ.; Calligaro, KD. (2002). Inferior vena cava filters in malignant
disease. J Vasc Surg. 36:704-707.
Johnson, DH.; Fehrenbacher, L.; Novotny, WF. et al. (2004). Randomized phase II trial
comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and
paclitaxel alone in previously untreated locally advanced or metastatic non-
small-cell lung cancer. J Clin Oncol 22:2184-91.
Kabbinavar, F.; Hurwitz, HI.; Fehrenbacher, L. et al. (2003). Phase II, randomized trial
comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV
alone in patients with metastatic colorectal cancer. J Clin Oncol 21:60-5.
Venous Thromboembolism Prophylaxis in Cancer Patients 125
Kahn, SR.; Panju, A.; Geerts, With. et al. (2007). Multicenter evaluation of the use of
venous thromboembolism prophylaxis in acutely ill medical patients in Canada.
Thromb Res 119:145–55.
Kakkar, AK.; Levine, M.; Pinedo, HM.; Wolff, R.; Wong, J. (2003). Venous thrombosis in
cancer patients: insights from the FRONTLINE survey. Oncologist 8:381-8.
Kakkar, AK.; Haas, S.; Wolf, H.; Encke, A. (2005). Evaluation of perioperative fatal
pulmonary embolism and death in cancer surgical patients: the MC-4 cancer
substudy. Thromb Haemost 94:867–71.
Karthaus, M.; Kretzschmar, A.; Kröning, H. et al. (2006). Dalteparin for prevention of
catheter-related complications in cancer patients with central venous catheters:
final results of a double-blind, placebo-controlled phase III trial. Ann Oncol 17:
289-96.
Kearon, C.; Kahn, SR.; Agnelli, G.; Goldhaber, S.; Raskob, GE.; Comerota, AJ.
(2008).American College of Chest Physicians. Antithrombotic therapy for venous
thromboembolic disease: American College of Chest Physicians Evidence-Based
Clinical Practice Guidelines (8th Edition). Chest. 133(6 Suppl):454S-545S.
Khorana, AA.; Kuderer, NM.; Culakova, E.; Lyman, GH.; Francis, CW. (2008).
Development and validation of a predictive model for chemotherapy-associated
thrombosis. Blood 111:4902–7.
Kniffin, WD.; Baron, JA.; Barret, J.; Bikmeyer, JD.; Anderson, FA. (1994). The
epidemiology of pulmonary embolism and deep venous thrombosis in the
elderly. Arch Intern Med 154:861–6.
Kyrle, PA.; Hron, G.; Eichinger, S.; Wagner, O. (2007). Circulating P-selectin and the risk
of recurrent venous thromboembolism. Thromb Haemost 97:880-883
Levin, JM.; Schiff, D.; Loeffler, JS.; Fine, HA.; Black, PM.; Wen, PY. (1993). Complications
of therapy for venous thromboembolic disease in patients with brain tumors.
Neurology. 43:1111-1114.
Lyman, GH.; Khorana, AA.; Falanga, A. et al. (2007). American Society of Clinical
Oncology Guideline: Recommendations for venous thromboembolism
prophylaxis and treatment in patients with cancer. J Clin Oncol 25:5490–5505.
Mandybur, TI. (1977). Intracranial hemorrhage caused by metastatic tumors. Neurology.
27:650-655.
Marras, LC.; Geerts, WH.; Perry, JR. (2000). The risk of venous thromboembolism is
increased throughout the course of malignant glioma: an evidence-based review.
Cancer 89:640–6.
Maynard,G.; Stein, J. (2010). Designing and implementing effective venous
thromboembolism prevention protocols: Lessons from collaborative efforts. J
Thromb Thrombolysis 29:159-66
Minnema, MC.; Breitkreutz, I.; Auwerda, JJ. et al. (2004). Prevention of venous
thromboembolism with low molecular-weight heparin in patients with multiple
myeloma treated with thalidomide and chemotherapy. Leukemia 18:2044-6.
Mismetti, P.; Mille, D.; Laporte, S. et al. (2003). Low-molecular-weight heparin
(nadroparin) and very low doses of warfarin in the prevention of upper
126 Deep Vein Thrombosis
Sallah. S.; Wan, JY.; Nguyen, NP. (2002). Venous thrombosis in patients with solid tumors:
determination of frequency and characteristics. Thromb Haemost 87:575–9.
Saour, J.; Al Harthi, A.; El Sherif, M.; Bakhsh, E.; Mammo, L. (2009). Inferior vena caval
filters: 5 years of experience in a tertiary care center. Ann Saudi Med. 29:446-449.
Schunn, C.; Schunn, GB.; Hobbs, G.; Vona-Davis, LC.; Waheed ,U. (2006). Inferior vena
cava filter placement in late-stage cancer. Vasc Endovascular Surg. 40:287-294.
Schwarz, RE.; Marrero, AM.; Conlon, KC.; Burt, M. (1996). Inferior vena cava filters in
cancer patients: indications and outcome. J Clin Oncol. 14:652-657.
Shah, MA.; Ilson, D.; Kelsen, DP. (2005). Thromboembolic events in gastric cancer: High
incidence in patients receiving irinotecan- and bevacizumabbased therapy. J Clin
Oncol 23:2574-6.
Sørensen, HT.; Mellemkjaer, L.; Olsen, JH.; Baron, JA. (2000). Prognosis of cancers
associated with venous thromboembolism. N Engl J Med 343:1846–50.
Spencer, FA.; Bates, SM.; Goldberg, RJ. et al. (2010). A population-based study of inferior
vena cava filters in patients with acute venous thromboembolism. Arch Intern
Med. 170:1456-1462.
Tapson, VF.; Decousus, H.; Pini, M. et al. (2007). Venous thromboembolism prophylaxis in
acutely ill hospitalized medical patients: findings from the international medical
prevention registry on venous thromboembolism. Chest 132:936–45.
The Joint Commission Manual for Performance Improvement Measures. (2011). Available
from: [Link] [last cited
on, Apr 06, 2011].
The PREPIC Study Group. (2005). Eight-year follow-up of patients with permanent vena
cava filters in the prevention of pulmonary embolism: the PREPIC (Pre´vention
du Risque d’Embolie Pulmonaire par Interruption Cave) randomized study.
Circulation. 112:416–422.
Thodiyil, PA.; Kakkar, AK. (2002). Variation in relative risk of venous thromboembolism
in different cancers. Thromb Haemost 87:1076–7.
Verso, M.; Agnelli, G.; Bertoglio, S. et al. (2005). 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 23, 4057-62.
Wagman, LD.; Baird, MF.; Bennett, CL. et al. (2008). Venous thromboembolic disease.
NCCN. Clinical practice guidelines in oncology. J Natl Compr Canc Netw 6:716–53.
Wallace, MJ.; Jean, JL.; Gupta, S. et al. (2004). Use of inferior vena caval filters and survival
in patients with malignancy. Cancer. 101:1902-1907.
White, RH.; Zhou, H.; Romano, PS. (2003). Incidence of symptomatic venous
thromboembolism after different elective or urgent surgical procedures. Thromb
Haemost 90:446–55.
Zangari, M.; Siegel, E.; Barlogie, B. et al. (2002). Thrombogenic activity of doxorubicin in
myeloma patients receiving thalidomide: implications for therapy. Blood
100:1168-71.
Zangari, M.; Fink, LM.; Elice, F.; Zhan, F.; Adcock, DM.; Tricot, GJ. (2009). Thrombotic
events in patients with cancer receiving antiangiogenesis agents. J Clin Oncol 27:
4865-73
128 Deep Vein Thrombosis
Zonder, JA.; Barlogie, B.; Durie, BG.; McCoy, J.; Crowley, J.; Hussein, MA. (2006).
Thrombotic complications in patients with newly diagnosed multiple myeloma
treated with lenalidomide and dexamethasone: Benefit of aspirin prophylaxis.
Blood 108:403.
8
1. Introduction
Deep vein thrombosis is often regarded as a disease limited of the veins of the lower
extremities, which may sometimes – in more severe cases - extend to the pelvic veins.
Although this holds true for over 90% of all thromboses, clinically relevant thromboses may
be found in virtually every vein system of the body. Of these uncommon localisations of
thromboses, deep vein thrombosis of the arms is one of the most frequent entities,
accounting for about 5% of all thromboses (Munoz et al., 2008; Isma et al., 2010). Most cases
of deep arm vein thrombosis develop secondary in patients with indwelling central venous
catheters, pacemakers, malignant disease, or after surgery. Conversely, primary upper
extremity deep venous thrombosis is observed in patients after strenuous arm exercise
(“thrombosis par effort”), in thoracic outlet syndrome and inherited or acquired
thrombophilia (Bernardi et al., 2006). Acute and long-time complications of upper extremity
thrombosis may be significant and include pulmonary embolism, post-thrombotic
syndrome and recurrent thromboembolism. In this chapter, the clinical presentation,
diagnostic procedures, treatment and prevention of thromboses of the upper extremity will
be reviewed. It is not unusual to find thromboses of proximal arm veins and deep veins of
the neck region at the same time. Therefore, thromboses of the internal jugular vein, which
are also most often observed in the presence of indwelling central venous catheters, will also
be discussed. In this review, special emphasis will be given to the practical aspects of the
disease, like risk factors, clinical presentation, diagnosis, and treatment of arm vein
thrombosis. For a detailed, comprehensive overview of pathophysiological mechanisms, the
reader will be referred to other, excellent reviews within this field.
2. Epidemiology
The frequency of deep arm vein thrombosis relative to all deep thromboses has been
reported to be between 1 and 14% (Hill & Berry, 1990; Joffe et al., 2004; Spencer et al.,
2007). Recently, the prospective RIETE registry and the population based Malmö
thrombophilia study reported both very similar rates of upper extremity deep vein
thrombosis (4.4% and 5% of all thrombosis, respectively (Munoz et al., 2008; Isma et al.,
2010). Therefore, it can be assumed that about 5% of all thrombosis will involve the deep
arm veins, which corresponds to an annual incidence of approximately 3 per 100.000
patients per year (Bernardi et al., 2006). Less than 50% of these arm vein thromboses can
be expected to extend into the internal jugular vein (Gbaguidi et al., 2011). About one
130 Deep Vein Thrombosis
third of patients with deep arm vein thrombosis will have primary thrombosis, i.e.
idiopathic and effort-related thrombosis (Paget-von Schroetter syndrome). The remaining
two thirds of patients will have secondary upper extremity thrombosis with exogenous
(e.g. central venous catheters) or endogenous (e.g. cancer) risk factors. In intensive care
patients as well as in patients suffering from malignant disease with central venous
catheters, rates of asymptomatic thrombosis as high as 30% to over 60% have been
reported (Timsit et al., 1998; Van Rooden et al., 2005). There appears to be an increase in
upper extremity deep vein thrombosis in the last decades, which may reflect the
increasing use of central venous catheters (S. Mustafa et al., 2003; Czihal & Hoffmann,
2011), improved diagnostic methods, or both.
Subclavian vein
Cephalic vein
Axillar vein
Brachial vein
distinct clinical entities, namely after recent oropharyngeal infections with anaerobic bacteria
(fusobacterium necrophorum) and in the ovarian hyperstimulation syndrome (Gbaguidi et al.,
2011). For the latter syndrome, the increased risk of thrombosis has been explained by the
drainage of excessive estrogen concentrations in the peritoneal fluid via the thoracic and right
lymphatic duct into the confluence region of the large neck veins (Bauersachs et al., 2007).
Odds 95 % Confidence
Risk factor Reference
ratio interval
1136 153 - 8448 Blom et al. 2005
Central venous catheter
9.7 7.8 – 12.2 Joffe et al. 2004
Active cancer 18.1 9.4 – 35.1 Blom et al. 2005
Arm surgery 13.1 2.1 – 80.6 Blom et al. 2005
Plaster cast 7.0 1.7 – 29.5 Blom et al. 2005
Factor V Leiden* 6.2 2.5 – 15.7 Martinelli et al. 2004
Prothrombin G20210A* 5.0 2.0 – 12.2 Martinelli et al. 2004
Protein C, Protein S 4.9 1.1 – 22.0 Martinelli et al. 2004
Family history of VTE 2.8 1.6 – 4.9 Blom et al. 2005
Arm injury 2.1 0.7 – 6.2 Blom et al. 2005
Oral contraceptives 2.0 1.1 -3.8 Blom et al. 2005
Unusual arm exercise 1.5 1.0 – 2.1 Blom et al. 2005
*heterozygous mutation. VTE venous thromboembolism
Table 1. Major risk factors for arm vein thrombosis with odds ratios adjusted for age and sex.
5. Clinical presentation
The presence of typical symptoms (swelling of the upper extremity, localized pain, and
superficial collaterals) will raise the suspicion of the clinician to consider deep vein
thrombosis of the arms. The most frequent symptom is edema of the upper extremity, which
has been reported in 80% of cases by Joffe and coworkers . In addition, about 40% of patients
report localized pain or aching discomfort in the involved extremity (Joffe et al., 2004). Other
symptoms include reddish-blue discoloration, a sensation of heat or heaviness in the
respective arm, or dilated superficial collateral veins on the upper arm, shoulder girdle,
neck, and anterior chest wall (Prandoni et al., 1997a; Kommareddy et al., 2002; Bernardi et
al., 2006). In at least 5% of cases, deep vein thrombosis of the arms will be completely
asymptomatic (Mai & Hunt, 2011). In some cases, patients will become symptomatic only
after the occurrence of complications, like dyspnea in the case of pulmonary embolism, or
rarely venous gangrene (Kaufman et al., 1998). In dialysis patients, arm vein thrombosis
may become symptomatic only by catheter dysfunction, like the inability to draw blood,
increased dialysis pressure or arm swelling after dialysis (Hernandez et al., 1998).
In addition, classical syndromes have been described which share some but not all of their
symptoms with the clinical picture described above. Superior vena cava syndrome has been
described as a complication especially of catheter related arm vein thrombosis, but may also
be caused by other mechanisms, e.g. venous compression by chest tumors (Lepper et al.,
2011). It comprises usually bilateral edema of the face, neck, and upper extremities, together
with cyanosis, plethora, and dilated subcutaneous vessels. Paget von Schroetter´s syndrome,
which was first described at the end of the 19th century, is defined as a primary thrombosis
Deep Vein Thrombosis of the Arms 133
of the subclavian vein at the costoclavicular junction. This syndrome is usually precipitated
by musculoskeletal compression and / or repetitive microtrauma by strenuous arm exercise
(thrombosis par effort) (Constans et al., 2008; Illig & Doyle, 2010). In some cases, a
compression of the subclavian vein by muscular hypertophy (anterior scalenus muscle,
subclavian muscle) or by the clavicle and the first rib with extreme arm movements
(hyperabduction and elevation) may be responsible (thoracic outlet syndrome). Likewise,
the syndrome has been linked to certain sport activities, as e.g. weight lifters, baseball
pitchers or tennis players (Sheeran et al., 1997; van Stralen et al., 2005). It is observed more
frequently in young patients, men (2:1), and in the dominant extremity (Illig & Doyle, 2010).
The typical clinical presentation of this syndrome is a sudden onset of the typical symptoms
of deep vein thrombosis described above (Illig & Doyle, 2010). Isolated thrombosis of the
internal jugular vein is a rare disease which may be due to local (central venous catheters,
recent oropharyngeal infections with anaerobic bacteria like fusobacterium necrophorum) or
systemic factors (cancer, thrombophilia, ovarian hyperstimulation syndrome) (Sheikh et al.,
2002; Gbaguidi et al., 2011). Lemierre´s syndrome, first described in 1910, is a severe
condition of septic thrombosis following oropharyngeal infections, mostly due to
fuosbacterium necrophorum. This syndrome is also known as human necrobacillosis or
post-anginal septicaemia. Besides local symptoms (cervical edema, localized pain, dilated
superficial collateral veins, erythrocyanosis, indurated vein), severe systemic complications
like septicaemia and septic pulmonary embolism may occur (Vargiami & Zafeiriou, 2010;
Gbaguidi et al., 2011).
When evaluating patients for clinical signs of deep arm vein thrombosis, it is important to
bear in mind possible alternative diagnoses. These include e.g. superficial thrombophebitis,
paravasates after peripheral infusions, lymphedema, cellulitis, haematoma, venous
compression and traumatic injuries (Kommareddy et al., 2002; Bernardi et al., 2006).
6. Diagnostic procedures
As in thrombosis of the lower extremities, there are no reliable clinical symptoms to
diagnose deep arm vein thrombosis. Therefore, diagnostic test are necessary to diagnose or
rule out upper extremity thrombosis. Contrast venography is the gold standard diagnostic
method, allowing an unparalleled overview of all arm veins with high resolution (Fig 2).
However, venography is invasive, inconvenient for the patient and suffers from moderate
inter-observer agreement rates (between 71 and 83%) in upper extremity deep vein
thrombosis (Baarslag et al., 2003). Furthermore, possible complications as contrast agent
mediated kidney damage, allergic reactions and even venography-induced thrombosis can
occur (Bernardi et al., 2006).
Ultrasound has several advantages compared to invasive methods and has become the
diagnostic method of choice in lower extremity deep vein thrombosis (Kearon et al., 1998;
Goodacre et al., 2006). The main strengths of ultrasound are its non-invasive nature, general
availability, as well as the lack of radiation and contrast material. However, ultrasound
suffers generally from some disadvantages, including observer variability and usually lack
of standardized documentation. In arm vein thrombosis, an additional obstacle is the
portion of the subclavian vein behind the clavicle, impeding compression manoeuvres in
this clinically important region. In addition, the brachiocephalic veins and the superior vena
134 Deep Vein Thrombosis
Fig. 2. Venogram of a chronic occlusion of the left subclavian vein with extensive
collateralization (Department of Radiology, Innsbruck Medical University).
Since there is no imaging method combining optimal accuracy and minimal burden for the
patient, alternative methods have been searched for. An interesting clinical prediction score
for arm vein thrombosis, reminiscent of the Wells score for lower extremity thrombosis
(Wells et al., 1997), has been published by Constans and coworkers (Constans et al., 2008).
This simple score assigns one point each for a central venous catheter or pacemaker lead,
localized pain and unilateral edema. One point is subtracted in the case that an alternative
diagnosis would seem at least as likely as deep arm vein thrombosis. D-dimer testing has
also been evaluated in a cohort of patients with suspected deep arm vein thrombosis
(Merminod et al., 2006). Although nearly 100% sensitive, D-dimer suffers from a low
specificity.
Deep Vein Thrombosis of the Arms 135
A B
A B
Fig. 4. Indirect sonographic diagnosis of a non-recent thrombosis of the left sublcavian vein
with partial recanalisation. Normal venous flow with cardiac and respiratory modulation in
the right subclavian vein (A) compared to linear flow in the left subclavian vein (B).
136 Deep Vein Thrombosis
8. Treatment
Due to the lack of adequately powered, randomized clinical trials the current guidelines for
the treatment of arm vein thrombosis are mainly based on small cohort studies, expert
opinion or extrapolation of data derived from larger studies performed in patients with
lower extremity deep vein thrombosis. Nevertheless, the eight edition of the American
college of chest physicians’ (ACCP) guidelines cover several important aspects of the
treatment of patients with upper extremity deep vein thrombosis (Kearon et al., 2008). For
the initial treatment, therapeutic doses of low molecular weight heparin, unfractionated
heparin or fondaparinux are recommended. Overlapping with this initial treatment, long-
term anticoagulation with a vitamin K antagonist should be started and continued for a
minimum of 3 months. No studies are available that have addressed the ideal duration of
Deep Vein Thrombosis of the Arms 137
Another point of debate is the question whether central venous catheters should be removed
when a diagnosis of deep vein thrombosis has been confirmed in the respective vessel. Most
experts opt against catheter removal, if the catheter is still needed and still functional. In a
cohort study of 74 cancer patients with acute upper extremity thrombosis, the catheters were
not removed and patients were treated for 3 months with standard anticoagulation without
recurrent episodes of venous thromboembolism (Kovacs et al., 2007). If the catheter is
removed, the ACCP guidelines recommend not to shorten the anticoagulation period below
3 months (Kearon et al., 2008).
138 Deep Vein Thrombosis
9. Prevention
Patients with central venous catheters carry a high risk of deep arm vein thrombosis, which
may exceed 60 % in certain patient groups (ICU patients, oncological and hematological
patients) Van Rooden et al., 2005. Therefore, approaches to prevent catheter-related
thrombosis by means of pharmacological prophylaxis e.g. in cancer patients, appear
attractive. However, despite an early study showing benefit of low dose warfarin in this
context (Bern et al., 1990), subsequent studies with warfarin and heparins could not confirm
this protective effect. A recent meta-analysis did show a trend, but no significant reduction
of symptomatic deep vein thrombosis with any form of thromboprophylaxis (Akl et al.,
2008). In accordance with these data, the current guidelines for the prevention of venous
thromboembolism do not recommend routine use of thromboprophyaxis in cancer patients
with indwelling central venous catheters (Geerts et al., 2008).
The placement of superior vena cava filters has been reported in case reports and small case
series. Although effective in preventing pulmonary embolism from thrombi in the upper
extremities, these filters may cause severe complications, like cardiac tamponade and aortic
perforation (Owens et al., 2010) and do not protect from thrombi in the lower extremities.
Therefore, the placement of these filters should be limited to special situations (Kucher, 2011).
11. References
Akl, E.A.; Kamath, G.; Yosuico, V.; Kim, S.Y.; Barba, M.; Sperati, F.; Cook, D.J. & Schunemann,
H.J. (2008) Thromboprophylaxis for patients with cancer and central venous
catheters. Cancer 112, 2483-2492.
Baarslag, H.J.; Van Beek, E.J.R.; Tijssen, J.G.P.; van Delden, O.M.; Bakker, A.J. & Reekers, J.A.
(2003) Deep vein thrombosis of the upper extremity: intra- and interobserver study of
digital subtraction venography. European Radiology 13, 251-255.
Deep Vein Thrombosis of the Arms 139
Baarslag, H.J.; Van Beek, E.J.R. & Reekers, J.A. (2004) Magnetic resonance venography in
consecutive patients with suspected deep vein thrombosis of the upper extremity:
Initial experience. Acta Radiologica 45, 38-43.
Bauersachs, R.M.; Manolopoulos, K.; Hoppe, I.; Arin, M.J. & Schleussner, E. (2007) More on:
the 'ART' behind the clot: solving the mystery. Journal of Thrombosis and Haemostasis 5,
438-439.
Beathard, G.A. (2001) Catheter thrombosis. Seminars in Dialysis 14, 441-445.
Becker, G.J.; Holden, R.W.; Rabe, F.E.; Castanedazuniga, W.R.; Sears, N.; Dilley, R.S. & Glover,
J.L. (1983) Local thrombolytic therapy for subclavian and axillary vein-thrombosis -
treatment of the thoracic inlet syndrome. Radiology 149, 419-423.
Bern, M.M.; Lokich, J.J.; Wallach, S.R.; Bothe, A.; Benotti, P.N.; Arkin, C.F.; Greco, F.A.;
Huberman, M. & Moore, C. (1990) Very low-doses of warfarin can prevent
thrombosis in central venous catheters - a randomized prospective trial. Annals of
Internal Medicine 112, 423-428.
Bernardi, E.; Pesavento, R. & Prandoni, P. (2006) Upper extremity deep venous thrombosis.
[Link]. 32, 729-736.
Blom, J.W.; Doggen, C.J.; Osanto, S. & Rosendaal, F.R. (2005) Old and new risk factors for
upper extremity deep venous thrombosis. [Link]. 3, 2471-2478.
Brismar, B; Hardstedt, C & Jacobson, S. (1981). Diagnosis of thrombosis by catheter
phlebography after prolonged central venous catheterization. Annals of Surgery
194,779-783.
Chen, S.C.; Chang, J.M.; Wang, C.S.; Wu, H.C. & Chen, H.C. (2009) Upper limb deep vein
thrombosis following calcium gluconate injection. Nephrology 14, 621.
Constans, J.; Salmi, L.R.; Sevestre-Pietri, M.A.; Perusat, S.; Nguon, M.; Degeilh, M.; Labarere, J.;
Gattolliat, O.; Boulon, C.; Laroche, J.P.; Le Roux, P.; Pichot, O.; Quere, I.; Conri, C. &
Bosson, J.L. (2008) A clinical prediction score for upper extremity deep venous
thrombosis. [Link]. 99, 202-207.
Czihal, M. & Hoffmann, U. (2011) Upper extremity deep venous thrombosis. Vascular Medicine
16, 191-202.
Di Nisio, M.; van Sluis, G.L.; Bossuyt, P.M.M.; Buller, H.R.; Porreca, E. & Rutjes, A.W.S. (2010)
Accuracy of diagnostic tests for clinically suspected upper extremity deep vein
thrombosis: a systematic review. Journal of Thrombosis and Haemostasis 8, 684-692.
Gbaguidi, X.; Janvresse, A.; Benichou, J.; Cailleux, N.; Levesque, H. & Marie, I. (2011) Internal
jugular vein thrombosis: outcome and risk factors. Qjm-An International Journal of
Medicine 104, 209-219.
Geerts, W.H.; Bergqvist, D.; Pineo, G.F.; Heit, J.A.; Samama, C.M.; Lassen, M.R. & Colwell,
C.W. (2008) Prevention of venous thromboembolism. Chest 133, 381S-453S.
Goltz, J.P.; Scholl, A.; Ritter, C.O.; Wittenberg, G.; Hahn, D. & Kickuth, R. (2010) Peripherally
placed totally implantable venous-access port systems of the forearm: clinical
experience in 763 consecutive patients. Cardiovascular and Interventional Radiology 33,
1159-1167.
Goodacre, S.; Sampson, F.; Stevenson, M.; Wailoo, A.; Sutton, A.; Thomas, S.; Locker, T. & Ryan,
A. (2006) Measurement of the clinical and cost-effectiveness of non-invasive diagnostic
testing strategies for deep vein thrombosis. Health Technology Assessment 10, 1-168.
Hernandez, D.; Diaz, F.; Rufino, M.; Lorenzo, V.; Perez, T.; Rodriguez, A.; De Bonis, E.; Losada,
M.; Gonzalez-Posada, J.M. & Torres, A. (1998) Subclavian vascular access stenosis in
dialysis patients: Natural history and risk factors. Journal of the American Society of
Nephrology 9, 1507-1510.
140 Deep Vein Thrombosis
Hill, S.L. & Berry, R.E. (1990) Subclavian vein thrombosis: a continuing challenge. Surgery 108,
1-9.
Illig, K.A. & Doyle, A.J. (2010) A comprehensive review of Paget-Schroetter syndrome. Journal
of Vascular Surgery 51, 1538-1547.
Isma, N.; Svensson, P.J.; Gottsater, A. & Lindblad, B. (2010) Upper extremity deep venous
thrombosis in the population-based Malmo thrombophilia study (MATS).
Epidemiology, risk factors, recurrence risk, and mortality. Thrombosis Research 125,
E335-E338.
Joffe, H.V.; Kucher, N.; Tapson, V.F. & Goldhaber, S.Z. (2004) Upper-extremity deep vein
thrombosis: a prospective registry of 592 patients. Circulation 110, 1605-1611.
Kahn, S.R.; Elman, E.; Bornais, C.; Blostein, M. & Wells, P.S. (2005) Post-thrombotic syndrome,
functional disability and quality of life after upper extremity deep venous thrombosis
in adults. Thrombosis and Haemostasis 93, 499-502.
Kaufman, B.R.; Zoldos, J.; Bentz, M. & Nystrom, N.A. (1998) Venous gangrene of the upper
extremity. Annals of Plastic Surgery 40, 370-377.
Kearon, C.; Ginsberg, J.S. & Hirsh, J. (1998) The role of venous ultrasonography in the
diagnosis of suspected deep venous thrombosis and pulmonary embolism. Annals of
Internal Medicine 129, 1044-1049.
Kearon, C.; Kahn, S.R.; Agnelli, G.; Goldhaber, S.; Raskob, G.E. & Comerota, A.J. (2008)
Antithrombotic therapy for venous thromboembolic disease: American College of
Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 133,
454S-545S.
Kim, H.C.; Chung, J.W.; Park, J.H.; Yin, Y.H.; Park, S.H.; Yoon, C.J. & Choi, Y.H. (2003) Role of
CT venography in the diagnosis and treatment of benign thoracic central venous
obstruction. Korean Journal of Radiology 4, 146-152.
Koksoy, C.; Kuzu, A.; Erden, I. & Akkaya, A. (1995) The risk-factors in central venous catheter-
related thrombosis. Australian and New Zealand Journal of Surgery 65, 796-798.
Kommareddy, A.; Zaroukian, M.H. & Hassouna, H.I. (2002) Upper extremity deep venous
thrombosis. [Link]. 28, 89-99.
Kovacs, M.J.; Kahn, S.R.; Rodger, M.; Anderson, D.R.; Andreou, R.; Mangel, J.E.; Morrow, B.;
Clement, A.M. & Wells, P.S. (2007). A pilot study of central venous catheter
survival in cancer patients using low-molecular-weight heparin (dalteparin) and
warfarin without catheter removal for the treatment of upper extremity deep vein
thrombosis (the catheter study). Journal of Thrombosis and Haemostasis 5:1650-1653
Kucher, N. (2011) Deep-vein thrombosis of the upper extremities. New England Journal of
Medicine 364, 861-869.
Lechner, D.; Wiener, C.; Weltermann, A.; Eischer, L.; Eichinger, S. & Kyrle, P.A. (2008)
Comparison between idiopathic deep vein thrombosis of the upper and lower
extremity regarding risk factors and recurrence. Journal of Thrombosis and Haemostasis
6, 1269-1274.
Lepper, P.M.; Ott, S.R.; Hoppe, H.; Schumann, C.; Stammberger, U.; Bugalho, A.; Frese, S.;
Schmucking, M.; Blumstein, N.M.; Diehm, N.; Bals, R. & Hamacher, J. (2011) Superior
vena cava syndrome in thoracic malignancies. Respiratory Care 56, 653-666.
Luciani, A.; Clement, O.; Halimi, P.; Goudot, D.; Portier, F.; Bassot, N.; Luciani, J.A.; Avan, P.;
Frija, G. & Bonfils, P. (2001) Catheter-related upper extremity deep venous
thrombosis in cancer patients: A prospective study based on Doppler US. Radiology
220, 655-660.
Deep Vein Thrombosis of the Arms 141
Machleder, H.I. (1993) Evaluation of a new treatment strategy for Paget-Schroetter syndrome -
spontaneous thrombosis of the axillary-subclavian vein. Journal of Vascular Surgery 17,
305-317.
Mai, C. & Hunt, D. (2011) Upper extremity deep venous thrombosis: a review. American Journal
of Medicine 124, 402-407.
Marschang, P.; Niederwanger, A.; Gasser, R.W.; Daniaux, M. & Sturm, W. (2008) Symptomatic
upper extremity deep vein thrombosis as a complication of ambulatory blood
pressure monitoring. [Link]. 100, 711-712.
Martin, C.; Viviand, X.; Saux, P. & Gouin, F. (1999) Upper-extremity deep vein thrombosis after
central venous catheterization via the axillary vein. Critical Care Medicine 27, 2626-2629.
Martinelli, I.; Battaglioli, T.; Bucciarelli, P.; Passamonti, S.M. & Mannucci, P.M. (2004) Risk
factors and recurrence rate of primary deep vein thrombosis of the upper extremities.
Circulation 110, 566-570.
Martinelli, I.; Bucciarelli, P. & Mannucci, P.M. (2010) Thrombotic risk factors: Basic
pathophysiology. Critical Care Medicine 38, S3-S9.
Merminod, T.; Pellicciotta, S. & Bounameaux, H. (2006) Limited usefulness of D-dimer in
suspected deep vein thrombosis of the upper extremities. Blood Coagulation &
Fibrinolysis 17, 225-226.
Monreal, M.; Raventos, A.; Lerma, R.; Ruiz, J.; Lafoz, E.; Alastrue, A. & Llamazares, J.F. (1994)
Pulmonary embolism in patients with upper extremity DVT associated to venous
central lines - a prospective-study. Thrombosis and Haemostasis 72, 548-550.
Munoz, F.J.; Mismetti, P.; Poggio, R.; Valle, R.; Barron, M.; Guil, M. & Monreal, M. (2008)
Clinical outcome of patients with upper-extremity deep vein thrombosis - Results
from the RIETE registry. Chest 133, 143-148.
Mustafa, B.O.; Rathbun, S.W.; Whitsett, T.L. & Raskob, G.E. (2002) Sensitivity and specificity of
ultrasonography in the diagnosis of upper extremity deep vein thrombosis - A
systematic review. Archives of Internal Medicine 162, 401-404.
Mustafa, S.; Stein, P.D.; Patel, K.C.; Otten, T.R.; Holmes, R. & Silbergleit, A. (2003) Upper
extremity deep venous thrombosis. Chest 123, 1953-1956.
Owens, C.A.; Bui, J.T.; Knuttinen, M.G.; Gaba, R.C. & Carrillo, T.C. (2010) Pulmonary
embolism from upper extremity deep vein thrombosis and the role of superior vena
cava filters: A review of the literature. Journal of Vascular and Interventional Radiology
21, 779-787.
Palamari, B.; Breen, J.F. & Wysokinski, W.E. (2010) Lipoma causing upper extremity deep vein
thrombosis: A case report. Journal of Thrombosis and Thrombolysis 30, 109-111.
Phipps, C. & Joo, H. (2008) Upper limb deep vein thrombosis and portable computer games.
American Journal of Medicine 121, E3.
Prandoni, P.; Polistena, P.; Bernardi, E.; Cogo, A.; Casara, D.; Verlato, F.; Angelini, F.; Simioni,
P.; Signorini, G.P.; Benedetti, L. & Girolami, A. (1997a.) Upper-extremity deep vein
thrombosis - Risk factors, diagnosis, and complication. Archives of Internal Medicine
157, 57-62.
Prandoni, P.; Villalta, S.; Bagatella, P.; Rossi, L.; Marchiori, A.; Piccioli, A.; Bernardi, E.;
Girolami, B.; Simioni, P. & Girolami, A. (1997b.) The clinical course of deep-vein
thrombosis. Prospective long-term follow-up of 528 symptomatic patients.
Haematologica 82, 423-428.
Prandoni, P.; Bernardi, E.; Marchiori, A.; Lensing, A.W.; Prins, M.H.; Villalta, S.; Bagatella, P.;
Sartor, D.; Piccioli, A.; Simioni, P.; Pagnan, A. & Girolami, A. (2004) The long term
142 Deep Vein Thrombosis
clinical course of acute deep vein thrombosis of the arm: prospective cohort study.
BMJ 329, 484-485.
Schoen N.; Netzsch, C. & Kröger K. (2007) Subclavian vein thrombosis and backpacking.
Clinical Research in Cardiology 96, 42-44.
Sheeran, S.R.; Hallisey, M.J.; Murphy, T.P.; Faberman, R.S. & Sherman, S. (1997) Local
thrombolytic therapy as part of a multidisciplinary approach to acute
axillosubclavian vein thrombosis (Paget-Schroetter syndrome). Journal of Vascular and
Interventional Radiology 8, 253-260.
Sheikh, M.A.; Topoulos, A.P. & Deitcher, S.R. (2002) Isolated internal jugular vein thrombosis:
risk factors and natural history. Vascular Medicine 7, 177-179.
Shivakumar, S.P.; Anderson, D.R. & Couban, S. (2009) Catheter-associated thrombosis in
patients with malignancy. Journal of Clinical Oncology 27, 4858-4864.
Sood, S.L. (2009) Cancer-associated thrombosis. Current Opinion in Hematology 16, 378-385.
Spencer, F.A.; Emery, C.; Lessard, D. & Goldberg, R.J. (2007) Upper extremity deep vein
thrombosis: A community-based perspective. American Journal of Medicine 120, 678-684.
Thomas, I.H. & Zierler B.K. (2005) An integrative review of outcomes in patients with acute
primary upper extremity deep venous thrombosis following no treatment or
treatment with anticoagulation, thrombolysis, or surgical algorithms. Vascular and
Endovascular Surgery 39, 163-174.
Timsit, J.F.; Farkas, J.C.; Boyer, J.M.; Martin, J.B.; Misset, B.; Renaud, B. & Carlet, J. (1998)
Central vein catheter-related thrombosis in intensive care patients - Incidence, risks
factors, and relationship with catheter-related sepsis. Chest 114, 207-213.
Urschel, H.C. & Razzuk, M.A. (1998) Neurovascular compression in the thoracic outlet -
Changing management over 50 years. Annals of Surgery 228, 609-615.
Van Rooden, C.J.; Molhoek, S.G.; Rosendaal, F.R.; Schalij, M.J.; Meinders, A.E. & Huisman,
M.V. (2004) Incidence and risk factors of early venous thrombosis associated with
permanent pacemaker leads. Journal of Cardiovascular Electrophysiology 15, 1258-1262.
Van Rooden, C.J.; Tesselaar, M.E.T.; Osanto, S.; Rosendaal, F.R. & Huisman, M.V. (2005) Deep
vein thrombosis associated with central venous catheters - a review. Journal of
Thrombosis and Haemostasis 3, 2409-2419.
Van Stralen, K.J.; Blom, J.W.; Doggen, C.J.M. & Rosendaal, F.R. (2005) Strenuous sport
activities involving the upper extremities increase the risk of venous thrombosis of
the arm. Journal of Thrombosis and Haemostasis 3, 2110-2111.
Vargiami, E.G. & Zafeiriou, D.I. (2010) Eponym The Lemierre syndrome. European Journal of
Pediatrics 169, 411-414.
Verso, M.; Agnelli, G.; Kamphuisen, P.W.; Ageno, W.; Bazzan, M.; Lazzaro, A.; Paoletti, F.;
Paciaroni, M.; Mosca, S. & Bertoglio, S. (2008) Risk factors for upper limb deep vein
thrombosis associated with the use of central vein catheter in cancer patients. Internal
and Emergency Medicine 3, 117-122.
Vik, A.; Holme, P.A.; Singh, K.; Dorenberg, E.; Nordhus, K.C.; Kumar, S. & Hansen, J.B. (2009)
Catheter-directed thrombolysis for treatment of deep venous thrombosis in the upper
extremities. Cardiovascular and Interventional Radiology 32, 980-987.
Wells, P.S.; Anderson, D.R.; Bormanis, J.; Guy, F.; Mitchell, M.; Gray, L.; Clement, C.;
Robinson, K.S. & Lewandowski, B. (1997) Value of assessment of pretest probability
of deep-vein thrombosis in clinical management. Lancet 350, 1795-1798.
Zimmermann, R.; Morl, H.; Harenberg, J.; Gerhardt, P.; Kuhn, H.M. & Wahl, P. (1981)
urokinase therapy of subclavian-axillary vein-thrombosis. Klinische Wochenschrift 59,
851-856. Supported by grant P 20825-B05 of the Austrian Science Funds (FWF).
9
1. Introduction
Despite appropriate anticoagulant therapy, at least 1 of every 2-3 patients with deep-vein
thrombosis (DVT) of the lower extremities will develop post-thrombotic sequelae. These vary
from minor signs (i.e., stasis pigmentation, venous ectasia, slight pain and swelling) to severe
manifestations such as chronic pain, intractable edema and leg ulcers (1). The established post-
thrombotic syndrome (PTS) remains a significant cause of chronic illness, with considerable
socio-economic consequences for both the patient and the health care services (2,3).
The precise incidence of the PTS following confirmed venous thrombosis is still
controversial, as the rate of post-thrombotic sequelae reported in published studies has
varied between 20% and 100%. In earlier studies, a surprisingly high rate of severe PTS
complications was reported (50 to 100% of the patients within 4 to 10 years after the
qualifying thrombotic episode) (4-6). This rate sharply decreased in studies performed in the
last 25 years (7-39), which could be due to improved diagnostic and therapeutic approaches
to patients with DVT. However, owing to large differences among studies in terms of study
design, definition of PTS, sample size, length of follow-up, and use of compression elastic
stockings, the reported incidence of both overall and severe PTS still shows considerable
variability. In the absence of elastic stockings, PTS is expected to develop in approximately
50% of patients suffering an episode of DVT, and is severe in one fifth of patients (1). Of
interest, PTS can develop, although to a lower extent, also after an asymptomatic episode of
postoperative DVT (40,41).
According to the results of the most recent studies, most patients who develop post-
thrombotic manifestations become symptomatic within two years from the acute episode of
DVT (1,18-20,29-32,35-37,39). These findings challenge the general view that the PTS
requires many years to become manifest.
tissues of the medial lower limb, a condition that has been termed "lipodermatosclerosis".
Pruritus and eczematous skin changes are frequently present, and a proportion of patients
develops secondary superficial varicose veins as the syndrome evolves. Ulceration, often
precipitated by minor trauma, arises in a considerable number of patients and is
characteristically chronic and indolent with a high recurrence rate, once healing has been
achieved. Uncommonly, patients with persistent obstruction may experience venous
claudication, a bursting pain in the leg during exercise, which, in some respects, mimics
arterial claudication (42).
The clinical picture of the PTS is non-specific, as clinical conditions other than DVT may
result in a similar set of symptoms and signs in the lower extremity, including superficial
venous insufficiency, increased body mass index, and trauma (43-45).
The diagnosis of the PTS is made based on the development of the above mentioned clinical
manifestations in patients with a history of DVT, irrespective of the presence of venous
abnormalities as shown by invasive or non-invasive diagnostic procedures. In the absence of
characteristic signs and symptoms, the demonstration of venous abnormalities (such as
venous reflux, persistent venous obstruction, or both) does not, in itself, allow a patient with
a history of DVT to be defined as having PTS.
Although the clinical picture of the PTS is classical, there is large variation among published
studies as to its clinical classification. Among the suggested scoring systems, the Villalta
scale and the CEAP classification are the most widely adopted (46). The former, based on
clinical findings alone (Table 1), has high interobserver agreement (47,48), and good ability
to discriminate patients with versus those without PTS and patients with mild versus those
with severe PTS (1,47,49). In addition, this scale correlates well with the patient's perception
of the interference of leg complaints with daily life (31,47,49). The Villalta scale has recently
been recommended as a standard to define PTS for use in clinical investigations by the
Scientific and Standardization Committee of the International Society on Thrombosis and
Haemostasis (50). The latter, known as CEAP (Clinical, Etiologic, Anatomic,
Pathophysiologic) classification, was developed as a result of the cooperative work of a
panel of experts in the field of vascular disease, and combines clinical and objective findings
into a sophisticated scoring system (Table 2) (51).
The Post Thrombotic Syndrome 145
that inflammation at the time of, or consequent to the episode of acute DVT may play a role
in the pathophysiology of PTS, a hypothesis that is being further explored in a large
prospective study (64).
system (1,2). The management of this condition is demanding and oftentimes frustrating.
Several treatment strategies, both conservative and surgical, have been tested, especially
aimed at ulcer healing.
In 1997, the results of a prospective randomized Dutch study on the prevention of the PTS
became available (19). In this trial, 194 consecutive patients with confirmed proximal DVT
were allocated to wear or to not wear elastic compression stockings. A predefined scoring
system was used to classify patients into three categories: no, mild-to-moderate, and severe
PTS. After a median follow-up of 76 months, mild-to-moderate PTS occurred in 19 (20%)
and severe PTS in 11 (11.5%) of the 96 patients with stockings, while these occurred in 46
(47%) and 23 (23.5%) of the 98 patients without stockings, respectively (p<0.001).
These results were recently confirmed by a prospective, controlled, randomized study
performed in Italy (29), in which 180 consecutive patients with a first episode of
symptomatic proximal DVT who were planned to receive conventional anticoagulant
treatment were randomized to wear or to not wear below-knee compression (30-40 mm Hg
at the ankle) elastic stockings for two years. Follow-up was performed for up to 5 years.
Post-thrombotic sequelae, as assessed with the Villalta scale, developed in 44 of the 90
control patients (severe in 10), and in 23 of the 90 patients who were randomized to wear
elastic stockings (severe in 3). After adjustment for baseline characteristics, the hazard ratio
for the PTS in the stockings group as compared to the control group was 0.5 (0.3 to 0.8). A
large, multicenter randomized trial is currently underway in North America to compare
active versus placebo stockings to prevent PTS after proximal DVT (64).
Although the results of an investigation conducted in Canada (101) were not consistent with
those of the above described studies (19,29,37), a recent meta-analytic review emphasized the
role of graduated compression stockings for preventing long-term post-thrombotic sequelae
(102). Accordingly, the latest edition of the American College of Chest Physicians has recently
recommended elastic stockings beside conventional anticoagulation in all patients with acute
symptomatic DVT, if feasible (99). While the effectiveness of compression stockings to prevent
PTS after distal DVT has not been studied, it would be reasonable to offer compression
stockings to patients with severe symptoms related to distal DVT.
Knee-length and thigh-length compression elastic stockings have similar physiologic effects
in decreasing venous stasis of the lower limb, but the former are easier to apply and are
more comfortable (103). A recent systematic review of knee versus thigh length graduated
compression stockings for the prevention of DVT concluded that knee length were as
effective as thigh length stockings and offer advantages in terms of patient compliance and
cost (104). In order to directly compare the effectiveness and tolerability of below-knee
versus thigh length stockings at the time of acute DVT to prevent PTS a randomized clinical
trial has been conducted at our institution, whose results will be available soon (105).
The optimal duration of the treatment with elastic stockings has received little attention. In a
recent trial, 169 patients with a first or recurrent proximal DVT who had received 6 months
of standard compression treatment were randomized to wear or to not wear graduated
elastic stockings for an additional 18 months (37). Overall, after 6 years of follow-up,
prolongation of compression therapy failed to confer an additional advantage - according to
the CEAP classification – over and above the initial 6-month period. However, when the
analysis was confined to women, there was a statistically significant advantage to
prolonging treatment with compression stockings. In a prospective cohort management
study, the discontinuation of elastic stockings in patients free from PTS complaints who had
been offered at least six months of compression therapy did not increase the rate of PTS
development over patients in whom stockings had been used for at least two years
The Post Thrombotic Syndrome 151
7. Prognosis
It is commonly believed that patients with established PTS have a poor prognosis, and that
the majority will have sustained disability. In recent years, a few reports have suggested that
prognosis of the PTS might not be as poor as previously reported (69-71). Indeed, when
provided with elastic compression stockings and regularly supervised, more than 50% of
patients either remain stable or improve during long-term follow-up, irrespective of the
initial degree of PTS (69-71). Clinical presentation helps predict the prognosis, as the
outcome of patients who have initially severe manifestations appears to be more favorable
than that of patients whose symptoms progressively deteriorate over time (71). However, at
present there is no way to reliably predict the course of PTS in individual patients.
8. Conclusion
PTS is a frequent, burdensome and costly complication of DVT. Currently, there are few
effective treatments for PTS. Until such treatments are identified, prevention of PTS will
have the greatest impact on reducing the overall burden of PTS on patients and society.
Preventing DVT recurrence is likely to reduce the risk of PTS. Daily use of graduated ECS
after DVT appears to reduce the risk of PTS. As of yet, there is no established role for
thrombolysis in preventing PTS, but trials are underway to address this important question.
Research is also underway to identify biologic markers that may predict the risk of PTS in
individual patients. Finally, a few emerging antithrombotic compounds may have the
potential to reduce the risk of PTS, however this requires further study.
152 Deep Vein Thrombosis
9. Acknowledgements
Dr Kahn is a recipient of a Senior Clinical Research Scientist Award from the Fonds de la
Recherche en Santé du Québec and received research funding from the Canadian Institutes
of Health Research and the Heart and Stroke Foundation of Canada.
10. References
[1] Kahn SR, Ginsberg JS. Relationship between deep venous thrombosis and the
postthrombotic syndrome. Arch Intern Med 2004; 164: 17-26.
[2] Bergqvist D, Jendteg S, Johansen L, Persson U, Ödegaard K. Cost of long term
complications of deep venous thrombosis of the lower extremities: an analysis of a
defined patient population in Sweden Ann Intern Med 1997; 126: 454-457.
[3] Kahn SR, M'Lan CE, Lamping DL, Kurz X, Bérard A, Abenhaim L. The influence of venous
thromboembolism on quality of life and severity of chronic venous disease. J Thromb
Haemost 2004; 2: 2146-2151.
[4] Bauer G. Roentgenological and clinical study of the sequelae of thrombosis Acta Chir
Scand 1942; 86 (suppl 74): 1-110.
[5] Gjores JE. The incidence of venous thrombosis and its sequelae in certain districts of
Sweden. Acta Chir Scand 1956; 206 (suppl 1): 1-88.
[6] O'Donnell TF, Browse NL, Burnand KG, Lea Thomas M. The socioeconomic effects of an
ilio-femoral venous thrombosis. J Surg Res 1977; 22: 483-488.
[7] Strandness DE, Langlois Y, Cramer M, Randlett A, Thiele BL. Long-term sequelae of acute
venous thrombosis. JAMA 1983; 250: 1289-1292.
[8] Widmer LK, Zemp E, Widmer T, Schmitt HE, Brandenberg E, Voelin R, Biland L, da Silva
A, Magos M. Late results in deep vein thrombosis of the lower extremity. Vasa 1985;
14: 264-268.
[9] Lindner DJ, Edwards JM, Phinney ES, Taylor LM, Porter JM. Long-term hemodinamic and
clinical sequelae of lower extremity deep vein thrombosis. J Vasc Surg 1986; 4: 436-
442.
[10] Heldal M, Seem E, Snadset PM, Abildgaard U. Deep vein thrombosis: a 7-year follow-up
study. J Intern Med 1993; 234: 71-75.
[11] Lagerstedt C, Olsson CG, Fagher B, Norgren L, Tengborn L. Recurrence and late sequelae
after first-time deep vein thrombosis Relationship to initial signs. Phlebology 1993; 8:
62-67.
[12] Monreal M, Martorell A, Callejas JM, Valls R, Llamazares J,F Lafoz E, Arias A.
Venographic assessment of deep vein thrombosis and risk of developing post-
thrombotic syndrome: a prospective study. J Intern Med 1993; 233: 854-859.
[13] Eichlisberger R, Frauchiger B, Widmer MT, Widmer LK, Jager K. Late sequelae of deep
venous thrombosis: a 13-year follow-up of 223 patients. Vasa 1994; 23: 234-243.
[14] Beyth RJ, Cohen AM, Landefeld CS. Long-term outcomes of deep-vein thrombosis. Arch
Intern Med 1995; 155: 1031-1037.
[15] Johnson BF, Manzo RA, Bergelin RO, Strandness DE. Relationship between changes in the
deep venous system and the development of the postthrombotic syndrome after an
acute episode of lower limb deep vein thrombosis: a one- to six-year follow-up. J Vasc
Surg 1995; 21: 307-313.
[16] Saarinen J, Sisto T, Laurikka J, Salenius JP, Tarkka M. Late sequelae of acute deep venous
thrombosis: evaluation five and ten years after. Phlebology 1995; 10: 106-109.
The Post Thrombotic Syndrome 153
[17] Franzeck UK, Schalch I, Jäger KA, Schneider E, Grimm J, Bollinger A. Prospective 12-year
follow-up study of clinical and haemodynamic sequelae after deep vein thrombosis
in low-risk patients (Zürich study). Circulation 1996; 93: 74-79.
[18] Prandoni P, Lensing AWA, Cogo A, Cuppini S, Villalta S, Carta M, Cattelan AM,
Polistena P, Bernardi E, Prins MH. The long-term clinical course of acute deep venous
thrombosis. Ann Intern Med 1996; 125: 1-7.
[19] Brandijes DPM, Büller HR, Heijboer H, Huisman MV, de Rijk M, Jagt H. Randomised trial
of effect of compression stockings in patients with symptomatic proximal-vein
thrombosis. Lancet 1997; 349: 759-762.
[20] Prandoni P, Villalta S, Bagatella P, Rossi L, Marchiori A, Piccioli A, Bernardi E, Girolami
B, Simioni P, Girolami A. The clinical course of deep-vein thrombosis Prospective
long-term follow-up of 528 symptomatic patients. Haematologica 1997; 2: 423-428.
[21] Biguzzi E, Mozzi E, Alatri A, Taioli E, Moia M, Mannucci PM. The post-thrombotic
syndrome in young women: retrospective evaluation of prognostic factors. Thromb
Haemost 1998; 80: 575-577.
[22] Masuda EM, Kessler DM, Kistner RL, Eklof B, Sato DT. The natural history of calf vein
thrombosis: lysis of thrombi and development of reflux. J Vasc Surg 1998; 8: 67-74.
[23] McLafferty RB, Moneta GL, Passmann MA, Brant BM, Taylor LM, Porter JM. Late clinical
and hemodynamic sequelae of isolated calf vein thrombosis. J Vasc Surg 1998; 27: 50-
57.
[24] Haenen JH, Janssen MCH, van Langen H, van Asten WNJC, Wollersheim H, van't Hof
MA, Skotnicki SH, Thien T. The postthrombotic syndrome in relation to venous
hemodynamics as measured by means of duplex scanning and strain-gauge
plethysmography. J Vasc Surg 1999; 29: 1071-1076.
[25] Holmström M, Åberg W, Lockner C, Paul C. Long term clinical follow-up in 256 patients
with deep-vein thrombosis initially treated with either unfractionated heparin or
dalteparin: a retrospective analysis. Thromb Haemost 1999; 82: 1222-1226.
[26] Saarinen J, Kallio T, Lehto M, Hiltunen S, Sisto T. The occurrence of the post-thrombotic
changes after an acute deep venous thrombosis. A prospective two-year follow-up
study J Cardiovasc Surg 2000; 41: 441-446.
[27] Mohr DN, Silverstein MD, Heit JA, Petterson TM, O'Fallon M, Melton LJ. The venous
stasis syndrome after deep venous thrombosis or pulmonary embolism: a
population-based study. Mayo Clin Proc 2000; 75: 1249-1256.
[28] Ziegler S, Schillinger M, Maca TH, Minar E. Post-thrombotic syndrome after primary
event of deep venous thrombosis 10 to 20 years ago. Thromb Res 2001; 101: 23-33.
[29] Prandoni P, Lensing AWA, Prins MH, Frulla M, Marchiori A, Bernardi E, Tormene D,
Mosena L, Pagnan A, Girolami A. Below-knee elastic compression stockings to
prevent the post-thrombotic syndrome A randomized controlled trial. Ann Intern
Med 2004; 141: 249-256.
[30] Gabriel F, Labios M, Portoles O, Guillen M, Corella D, Frances F, Martinez M, Gil J, Saiz C.
Incidence of post-thrombotic syndrome and its association with various risk factors in
a cohort of Spanish patients after one year of follow-up following acute deep venous
thrombosis. Thromb Haemost 2004; 92: 328-336.
[31] Kahn SR, Kearon C, Julian JA, Mackinnon B, Kovacs MJ, Wells P, Crowther MA,
Anderson DR, Van Nguyen P, Demers C, Solymoss S, Kassis J, Geerts W, Rodger M,
Hambleton J, Ginsberg JS. Predictors of the post-thrombotic syndrome during long-
154 Deep Vein Thrombosis
term treatment of proximal deep vein thrombosis. J Thromb Haemost 2005; 3: 718-
723.
[32] Roumen-Klappe EM, den Heijer M, Janssen MCH, van der Vleuten C, Thien T,
Wollersheim H. The post-thrombotic syndrome: incidence and prognostic value of
non-invasive venous examinations in a six-year follow-up study. Thromb Haemost
2005; 94: 825-830.
[33] Stain M, Schönauer V, Minar E, Bialonczyk C, Hirschl M, Weltermann A, Kyrle PA,
Eichinger S. The post-thrombotic syndrome: risk factors and impact on the course of
thrombotic disease. J Thromb Haemost 2005; 3: 2671-2676.
[34] Schulman S, Lindmarker P, Holmstrom M, Larfars G, Carlsson A, Nicol P, Svensson E,
Ljungberg B, Viering S, Nordlander S, Leijd B, Jahed K, Hjorth M, Linder O, Beckman
M. Post-thrombotic syndrome recurrence and death 10 years after the first episode of
venous thromboembolism treated with warfarin for 6 weeks or 6 months. J Thromb
Haemost 2005; 4: 734-742.
[35] González-Fajardo JA, Martin-Pedrosa M, Castrodeza J, Tamames S, Vaquero-Puerta C.
Effect of the anticoagulant therapy in the incidence of post-thrombotic syndrome and
recurrent thromboembolism: Comparative study of enoxaparin versus coumarin. J
Vasc Surg 2008; 48: 953-959.
[36] Roumen-Klappe EM, den Heijer M, van Rossum J, Wollersheim H, van der Vleuten C,
Thien T, Janssen MC. Multilayer compression bandaging in the acute phase of deep-
vein thrombosis has no effect on the development of the post-thrombotic syndrome. J
Thromb Thrombolysis 2009; 27: 400-405.
[37] Aschwanden M, Jeanneret C, Koller MT, Thalhammer C, Bucher HC, Jaeger KA. Effect of
prolonged treatment with compression stockings to prevent post-thrombotic
sequelae: a randomized controlled trial. J Vasc Surg 2008; 47: 1015-1021.
[38] Tick LW, Kramer MH, Rosendaal FR, Faber WR, Doggen CJJ. Risk factors for post-
thrombotic syndrome in patients with a first deep venous thrombosis. J Thromb
Haemost 2008; 6: 2075-2081.
[39] Kahn SR, Shrier I, Julian JA, Ducruet T, Arsenault L, Miron MJ, Roussin A, Desmarais S,
Joyal F, Kassis J, Solymoss S, Desjardins L, Lamping DL, Johri M, Ginsberg JS.
Determinants and time course of the postthrombotic syndrome after acute deep
venous thrombosis. Ann Intern Med 2008; 149: 698-707.
[40] Wille-Jorgensen P, Jorgensen LN, Crawford M. Asymptomatic postoperative deep vein
thrombosis and the development of postthrombotic syndrome A systematic review
and meta-analysis. Thromb Haemost 2005; 93: 236-241.
[41] Lonner JH, Frank J, McGuire K, Lotke PA. Postthrombotic syndrome after asymptomatic
deep vein thrombosis following total knee and hip arthroplasty. Am J Orthop 2006;
35: 469-472.
[42] Immelman EJ, Jeffrey PC. The postphlebitic syndrome Pathophysiology prevention and
management. Clin Chest Med 1984; 5: 537-550.
[43] Browse NL, Clemenson G, Lea Thomas M. Is the postphlebitic leg always postphlebitic?
Relation between phlebographic appearances of deep-vein thrombosis and late
sequelae. Br Med 1980; 281: 1167-1170.
[44] Raju S. Venous insufficiency of the lower limbs and stasis ulceration. Ann Surg 1983; 197:
688-697.
[45] Scott TE, LaMorte WW, Gorin DR, Menzoian JO. Risk factors for chronic venous
insufficiency: a dual case-control study. J Vasc Surg 1995; 22: 622-628.
The Post Thrombotic Syndrome 155
[46] Kolbach DN, Neumann HA, Prins MH. Definition of the post-thrombotic syndrome
differences between existing classifications. Eur J Vasc Endovasc Surg 2005; 30: 404-
414.
[47] Villalta S, Bagatella P, Piccioli A, Lensing AWA, Prins MH, Prandoni P. Assessment of
validity and reproducibility of a clinical scale for the post-thrombotic syndrome.
Haemostasis 1994; 24 (Suppl 1): 57a.
[48] Rodger MA, Kahn SR, Le Gal G, Solymoss S, Chagnon I,
[49] Anderson DR, Wells PS, Kovacs MJ. Inter-observer reliability of measures to assess the
post-thrombotic syndrome. Thromb Haemost 2008; 100: 164-166.
[50] Kahn SR, Hirsch A, Shrier I. Effect of post-thrombotic syndrome on health-related quality
of life after deep venous thrombosis. Arch Intern Med 2002; 162: 1144-1148.
[51] Kahn SR, Partsch H, Vedantham S, Prandoni P, Kearon C. Definition of post-thrombotic
syndrome of the leg for use in clinical investigations: A recommendation for
standardization. J Thromb Haemost 2009; 7: 879-883.
[52] Porter JM, Moneta GL. Reporting standards in venous disease: an update International
Consensus Commitee on Chronic Venous Disease. J Vasc Surg 1995; 21: 635-645.
[53] Bettmann MA, Paulin, S. Leg phlebography: the incidence, nature, and modification of
undesirable side effects Radiology 1977; 122: 101-104.
[54] Villalta S, Prandoni P, Cogo A, Bagatella P, Piccioli A, Bernardi E, Simioni P, Scarano L,
Girolami A. The utility of non-invasive tests for detection of previous proximal-vein
thrombosis. Thromb Haemost 1995; 73: 592-596.
[55] Coleridge Smith PD, Thomas P, Scurr JH, Dormandy JA. Causes of venous ulceration: a
new hypothesis. Br Med J 1988; 296: 1726-1727.
[56] Shami SK, Shields DA, Scurr JH, Coleridge Smith PD. Leg ulceration in venous disease.
Postgrad Med J 1992; 68: 779-785.
[57] Lindhagen A, Bergqvist D, Hallböök T, Efsing HO. Venous function five to eight years
after clinically suspected deep venous thrombosis. Acta Med Scand 1985; 217: 389-
395.
[58] Markel A, Manzo RA, Bergelin RO, Strandness DE. Valvular reflux after deep vein
thrombosis: incidence and time of occurrence. J Vasc Surg 1992; 15: 377-384.
[59] Franzeck UK, Schalch I, Bollinger A. On the relationship between changes in the deep
veins evaluated by Duplex sonography and the postthrombotic syndrome 12 years
after deep vein thrombosis. Thromb Haemost 1997; 77: 1109-1112.
[60] Haenen JH, Janssen MC, Wollersheim H, Van't Hof MA, de Rooij
[61] MJ, van Langen H, Skotnicki SH, Thien T. The development of postthrombotic syndrome
in relationship to venous reflux and calf muscle pump dysfunction at 2 years after the
onset of deep venous thrombosis. J Vasc Surg 2002; 35: 1184-1189.
[62] Asbeutah AM, Riha AZ, Cameron JD, McGrath BP. Five-year outcome study of deep vein
thrombosis in the lower limbs. J Vasc Surg 2004; 40: 1184-1189.
[63] Singh H, Masuda EM. Comparing short-term outcomes of femoral-popliteal and
iliofemoral deep venous thrombosis: early lysis and development of reflux. Ann Vasc
Surg 2005; 19: 74-79.
[64] Prandoni P, Frulla M, Sartor D, Concolato A, Girolami A. Vein abnormalities and the
post-thrombotic syndrome. J Thromb Haemost 2005; 3: 401-402.
[65] Shbaklo H, Holcroft CA, Kahn SR. Levels of inflammatory markers and the development
of the post thrombotic syndrome. Thromb Haemost 2009; 101: 505-12.
156 Deep Vein Thrombosis
[66] Kahn SR, Shbaklo H, Shapiro S, Wells PS, Kovacs MJ, Rodger MA, Anderson DR,
Ginsberg JS, Johri M, Tagalakis V. Effectiveness of compression stockings to prevent
the post-thrombotic syndrome (the SOX Trial and Bio-SOX biomarker substudy): a
randomized controlled trial. BMC Cardiovasc Disord 2007: 7: 21.
[67] Van Dongen CJ, Prandoni P, Frulla M, Marchiori A, Prins MH, Hutten BA. Relation
between quality of anticoagulant treatment and the development of the
postthrombotic syndrome. J Thromb Haemost 2005; 3: 939-942.
[68] Ageno W, Piantanida E, Dentali F, Steidl L, Mera V, Squizzato A, Marchesi C, Venco A.
Body mass index is associated with the development of the post-thrombotic
syndrome. Thromb Haemost 2003; 89: 305-309.
[69] Hafner J, Bounameaux H, Burg G, Brunner U. Management of venous leg ulcers. Vasa
1996; 25: 161-167.
[70] Evers EJ, Wuppermann T. Effect of different compression therapies on the reflux in deep
veins with a post-thrombotic syndrome. Vasa 1999; 28: 19-23.
[71] Erickson CA, Lanza DJ, Karp DL, Edwards JW, Seabrook GR, Cambria RA, Freishlag JA,
Towne JB. Healing of venous ulcers in an ambulatory care program: the roles of
chronic venous insufficiency and patients compliance. J Vasc Surg 1995; 22: 629-636.
[72] Milne AA, Ruckley CV. The clinical course of patients following extensive deep venous
thrombosis. Eur J Vasc Surg 1994; 8: 56-9.
[73] Prandoni P, Lensing AWA, Prins MH, Bagatella P, Scudeller A, Girolami A. Which is the
outcome of the post-thrombotic syndrome? Thromb Haemost 1999; 82: 1196-1197.
[74] Kahn SR, Elman E, Rodger MA, Wells PS. Use of elastic compression stockings after deep
venous thrombosis: a comparison of practices and perceptions of thrombosis
physicians and patients. J Thomb Haemost 2003; 1: 500-506.
[75] Kahn SR, Azoulay L, Hirsch A, Haber M, Strulovitch C, Shrier I. Effect of graduated
elastic compression stockings on leg symptoms and signs during exercise in patients
with deep venous thrombosis: a randomized cross-over trial. J Thomb Haemost 2003;
1: 494-499.
[76] Ginsberg JS, Magier D, Mackinnon B, Gent M, Hirsh J. Intermittent compression units for
severe post-phlebitic syndrome: a randomised crossover study. CMAJ 1999; 160:
1303-1306.
[77] O'Donnell MJ, McRae S, Kahn SR, Julian JA, Kearon C, Mackinnon B, Magier D,
Strulovich C, Lyons T, Robinson S, Hirsh J, Ginsberg JS. Evaluation of a venous-
return assist device to treat severe post-thrombotic syndrome (VENOPTS) A
randomized controlled trial. Thromb Haemost 2008; 99: 463-464.
[78] Kahn SR, Shrier I, Shapiro S, Houweling AH, Hirsch AM, Reid RD, Kearon C, Rabhi K,
Rodger MA, Kovacs MJ, Anderson DR, Wells PS. Six-month exercise training
program to treat post-thrombotic syndrome: a randomized controlled two-centre
trial. CMAJ 2011; 183: 37-44.
[79] Colgan MP, Dormandy JA, Jones PW, Schraibman IG, Shanik G, Young RA.
Oxpentifylline treatment of venous ulcers of the leg. Br Med J 1990; 300: 972-975.
[80] Layton AM, Ibbotson SH, Davies JA, Goodfield MJ. Randomized trial of oral aspirin for
chronic venous leg ulcer. Lancet 1994; 344: 164-165.
[81] Rudofsky G. Intravenous prostaglandin E1 in the treatment of venous ulcers – a double-
blind placebo-controlled trial. Vasa 1989; 28 (Suppl): 39-43.
[82] Salim AS. Role of sulphydril-containing agents in the management of venous (varicose)
ulceration A new approach. Clin Exp Dermatol 1992; 17: 427-432.
The Post Thrombotic Syndrome 157
[83] Salim AS. The role of oxygen-derived free radicals in the management of venous
(varicose) ulceration. A new approach World J Surg 1991; 15: 264-269.
[84] Coccheri S, Scandotto G, Agnelli G, Aloisi D, Palazzini E, Zamboni V. Randomised double
blind multicentre placebo controlled study of sulodexide in the treatment of venous
leg ulcers. Thromb Haemost 2002; 87: 947-952.
[85] Burnand K, Clemenson G, Morland M, Jarret PE, Browse NL. Venous
lipodermatosclerosis: treatment by fibrinolytic enhancement and elastic compression.
Br Med J 1980; 280: 7-11.
[86] de Jongste AB, Jonker JJC, Huisman MV, ten Cate JW, Azar AJ. A double blind three
center clinical trial on the short-term efficacy of 0-(-hydroxyethyl)-rutosides in
patients with post-thrombotic syndrome. Thromb Haemost 1989; 62: 826-829.
[87] Frulla M, Marchiori A, Sartor D, Mosena L, Tormene D, Concolato A, Hartmann L,
Prandoni P. Elastic stockings hydroxyethylrutosides or both for the treatment of post-
thrombotic syndrome. Thromb Haemost 2005; 93: 183-185.
[88] Baste JC, Midy F. Surgery for post-thrombotic syndrome of the lower limbs Rev Prat 1994;
44: 781-785.
[89] Gloviczki P, Bergan JJ, Rhodes JM, Canton LG, Harmsen S, Ilstrup DM. Mid-term results
of endoscopic perforator vein interruption for chronic venous insufficiency: lessons
learned from the north American subfascial endoscopic perforator surgery registry.
The north American study group J Vasc Surg 1999; 29: 489-502.
[90] Perrin M, Hiltbrand B, Bayon JM. Results of valvuloplasty in patients presenting deep
venous insufficiency and recurring ulceration. Ann Vasc Surg 1999; 13: 524-532.
[91] Goldhaber SZ, Buring JE, Lipnick RJ, Hennekens CH. Pooled analyses of randomized
trials of streptokinase and heparin in phlebographically documented acute deep
venous thrombosis. Am J Med 1984; 76: 393-397.
[92] Sidorov J. Streptokinase vs heparin for deep venous thrombosis Can lytic therapy be
justified? Arch Intern Med 1989; 149: 1841-1845.
[93] Rogers LQ, Lutcher CL. Streptokinase therapy for deep vein thrombosis: a comprehensive
review of the English literature. Am J Med 1990; 88: 389-395.
[94] Alesh I, Kayali F, Stein PD. Catheter-directed thrombolysis (intrathrombus injection) in
treatment of deep venous thrombosis: a systematic review. Catheter Cardiovasc
Interv 2007; 70: 143-148.
[95] Comerota AJ, Aldridge SC, Cohen G, Ball DS, Pliskin M, White JV. A strategy of
aggressive regional therapy for acute iliofemoral venous thrombosis with
contemporary venous thrombectomy or catheter-directed Thrombolysis. J Vasc Surg
1994; 20: 244-254.
[96] Bjarnason H, Kruse JR, Asinger DA, Nazarian GK, Dietz CA Jr, Caldwell MD, Key NS,
Hirsch AT, Hunter DW. Iliofemoral deep venous thrombosis: safety and efficacy
outcome during 5 years of catheter-directed thrombolytic therapy. JVIR 1997; 8: 405-
418.
[97] Comerota AJ, Paolini D. Treatment of acute iliofemoral deep venous thrombosis: a
strategy of thrombus removal. Eur J Vasc Endovasc Surg 2007; 33: 351-360.
[98] Manninen H, Juutilainen A, Kaukanen E, Lehto S. Catheter-directed thrombolysis of
proximal lower extremity deep vein thrombosis: A prospective trial with venographic
and clinical follow-up. Eur J Radiol 2011; epub ahead of print
[99] Park YJ, Choi JY, Min SK, Lee T, Jung IM, Chung JK, Chung JW, Park JH, Kim SJ, Ha J.
Restoration of patency in iliofemoral deep vein thrombosis with catheter-directed
158 Deep Vein Thrombosis
thrombolysis does not always prevent post-thrombotic damage. Eur J Vasc Endovasc
Surg 2008; 36: 725-730.
[100] Ghanima W, Kleven IW, Enden T, Rosales A, Wik HS, Pederstad L, Holme PA, Sandset
PM. Recurrent venous thrombosis, post-thrombotic syndrome and quality of life after
catheter-directed thrombolysis in severe proximal deep vein thrombosis. J Thromb
Haemost. 2011; epub ahead of print.
[101] Kearon C, Kahn SR, Agnelli G, Goldhaber S, Raskob GE,
[102] Comerota AJ. Antithrombotic therapy for venous thromboembolic disease: American
College of Chest Physicians evidence-based clinical practice guidelines (8th edition).
Chest 2008: 133 (6 Suppl): 454S-545S.
[103] Enden T, Sandvik L, Klow NE, Hafsahl G, Holme PA, Holmen LO, Ghanima W,
Njaastad AM, Sandbaek G, Slagsvold CE, Sandset PM. Catheter-directed Venous
Thrombolysis in acute iliofemoral vein thrombosis-the CaVenT Study: rationale and
design of a multicenter randomized controlled clinical trial. Am Heart J 2007; 154:
808-814.
[104] Ginsberg JS, Hirsh J, Julian J, Vander Laande Vries M, Magier D, MacKinnon B, Gent M.
Prevention and treatment of postphlebitic syndrome Results of a 3-part study. Arch
Intern Med 2001; 161: 2105-2109.
[105] Kakkos SK, Daskalopoulou SS, Daskalopoulos ME, Nicolaides AN, Geroulakos G.
Review on the value of graduated elastic compression stockings after deep vein
thrombosis. Thromb Haemost 2006; 96: 441-445.
[106] BenkoT, Cooke EA, McNally MA, Mollan RA. Graduated compression stockings: knee
length or thigh length Clin Orthop Relat Res 2001; 383: 197-203.
[107] Sajid MS, Tai NR, Goli G, Morris RW, Baker DM, Hamilton G. Knee versus thigh length
graduated compression stockings for prevention of deep venous thrombosis: a
systematic review. Eur J Vasc Endovasc Surg 2006; 32: 730-736.
[108] Full-leg vs below-knee elastic stockings for prevention of the post-thrombotic syndrome.
[Link] identifier: NCT00426075
[109] Ten Cate-Hoek AJ, Ten Cate H, Tordoir J, Hamulyák K, Prins MH. Individually tailored
duration of elastic compression therapy in relation to incidence of the postthrombotic
syndrome. J Vasc Surg 2010; 52: 132-138.
[110] Nelson EA, Harper DR, Prescott RJ, Gibson B, Brown D, Ruckley CV. Prevention of
recurrence of venous ulceration: randomized controlled trial of class 2 and class 3
elastic compression. J Vasc Surg 2006; 44: 803–808.
[111] Partsch H, Kaulich M, Mayer W. Immediate mobilisation in acute vein thrombosis
reduces post-thrombotic syndrome. Int Angiol 2004; 3: 206-212.
[112] Hull RD, Pineo GF, Brant R, Liang J, Cook R, Solymoss S, Poon MC, Raskob G. Home
therapy of venous thrombosis with long-term LMWH versus usual care: patient
satisfaction and post-thrombotic syndrome. Am J Med 2009; 122: 762-769.
[113] Schulman S, Kearon C, Kakkar AK, Mismetti P, Schellong S, Eriksson H, Baanstra D,
Schnee J, Goldhaber SZ. Dabigatran versus warfarin in the treatment of acute venous
thromboembolism. N Engl J Med 2009; 361: 2342-52.
[114] The Einstein Investigators. Oral rivaroxaban for symptomatic venous thromboembolism.
N Engl J Med 2010; 363: 2499-510.
10
Venous Thromboembolism
in Orthopaedic Surgery
Justin R. Knight and Michael H. Huo
Department of Orthopaedic Surgery
University of Texas Southwestern Medical Center,
Dallas, Texas
USA
1. Introduction
Venous Thromboembolism (VTE) is a common complication following orthopaedic
procedures. It is discussed most commonly as it relates to total hip arthroplasty (THA) and
total knee arthroplasty (TKA), though this disease process can be seen after any orthopaedic
surgery. It is associated with significant morbity and costs (Caprini et al., 2003). This chapter
will provide an overview of the epidemiology, pathophysiology, and management of
thromboembolic disease. This will include preventative strategies, evidence-based
guidelines and a focus on newer drug agents currently being developed.
2. Epidemiology
Total joint arthroplasties remain some of the most common orthopaedic procedures
performed worldwide. It is estimated that by 2015, over 500,000 total hip arthroplasties
and 1.3 million total knee arthroplasties will be done in the United States alone (Kim,
2008). The aggregate costs in 2007 totaled over $15 billion (US Agency for Healthcare
Research and Quality, 2007). Geerts et al. reported that VTE would occur in 40%-60% of
the patients undergoing total joint arthroplasty if no prophylaxis was administered
(Geerts et al., 2008). Despite appropriate chemophrophylaxis, one study noted
asymptomatic proximal DVT found on ultrasound in 6.7% of THA and TKA patients at
the time of transfer to a rehabilitation center (Schellong et al., 2005). As many as 80% of all
clinical VTE events associated with arthroplasty patients occur within 3 months after
surgery (Oster et al., 2004).
The costs of VTE are significant. Approximately 10% of the patients who develop VTE
following THAs or TKAs require re-admission to the hospital within 3 months after their
index surgery (Oster et al., 2004). The clinical sequelae are often significant and can include leg
swelling, venous stasis ulcers, pulmonary hypertension, post-thrombotic syndrome, and
recurrence (Heit, 2006). The one-year mortality following deep vein thrombosis (DVT) has
been reported as high as 14.6%. Pulmonary embolism (PE) is associated with even higher
mortality rate. Heit et al. reported as high as 52.3% in a recent cohort study (Heit et al., 1999).
160 Deep Vein Thrombosis
3. Pathophysiology
The coagulation cascade is a complex system in which multiple components are activated to
produce fibrin. An overview of the system along with the targets of various therapeutic
interventions is shown in Figure 1. The coagulation pathway is separated into the intrinsic
and the extrinsic pathways. The latter is activated in response to specific tissue injury. Both
lead to the eventual formation of thrombin. Thrombin causes the conversion of fibrinogen to
fibrin. Additionally, it activates factor XIII which stabilizes the fibrin. An endogenous
fibrinolytic system balances this system. It consists of antithrombins, proteins C and S, and
the plasmin-plasminogen system.
vWF
VIIIa VIII Platelet Activated Platelet
II
X
XI Va
Cell VIIIa
IIa IXa IIa
Xa Inhibitors V
XIa
Heparin/LMWH Va Tissue Factor VIIa
Xa
Va
Warfarin IX
VII
Direct Thrombin II Xa X
VIIa
Inhibitor
The primary pathophysiology factors that predispose any patient to VTE are the Virchow’s
Triad: endothelial injury, venous stasis (or turbulent blood flow), and hypercoagulability.
Endothelial injury can occur due to manipulation, and retractor placement during surgery.
Venous stasis can occur due to positioning and the use of a tourniquet. Hypercoagulability
can occur as a result of depletion or dilution of endogenous anticoagulants. It is also
associated with several pro-coagulant disease processes such as factor V Leiden deficiency,
protein C and S deficiency, and others.
4. Prevention
Clinical VTEs occur due to many different causes, but one significant factor is inadequate
prophylaxis (Amin et al., 2010). Several barriers exist for inadequate prophylaxis. These
include: expense, bleeding concerns, availability of agents, and conflicting
recommendations. The American College of Chest Physicians (ACCP) and the American
Venous Thromboembolism in Orthopaedic Surgery 161
ACCP AAOS
Recommendation Risk Reccommendation
PE Bleeding
Aspirin
Standard Standard LMWH
Fondaparinux
LMWH
Elevated Standard
Fondaparinux Warfarin
Warfarin Aspirin
LMWH Standard Elevated Warfarin
Fondaparinux
Aspirin
Elevated Elevated Warfarin
None
Table 1. Summary of ACCP and AAOS recommendations for pharmacologic
thromboprophylaxis in patients undergoing elective hip or knee surgery. (Reference: Huo
M. VTE prophylaxis after total joint arthroplasty: current challenges-potential solutions.
Current Orthopaedic Practice 2011;22:193-197.)
4.3.1 Mechanical
Mechanical prophylaxis using sequential compressive devices (SCDs) or foot pumps can be
used as a sole means of VTE prophylaxis. Their clinical efficacy and safety have been
documented in multiple studies. This is particularly useful in a patient that is perceived to
have an elevated bleeding risk (Geerts et al., 2008). In many practices, mechanical devices
are often used in conjunction with pharmacological prophylaxis. Newer devices may be
used in the outpatient setting upon hospital discharge. The clinical efficacy, safety, and
compliance have been documented in a few studies. It is necessary to continue to follow
larger cohorts of patients using outpatient mechanical prophylaxis alone to fully determine
the efficacy and compliance.
4.3.2 Warfarin
Warfarin has been used as VTE chemoprophylaxis in high-risk orthopedic patients for
decades. It is an efficacious agent. However, it requires close monitoring. It can be both
difficult and costly in the outpatient setting (Eikelboom & Weitz, 2007). It also has numerous
drug-drug and drug-food interactions. These interactions can be particularly challenging
considering the issue of poly-pharmacy in the elderly joint arthroplasty patient population.
It also has a delayed onset of action, which may require bridging with a shorter acting
anticoagulant such as LMWHs or unfractionated heparin. A recent paper by Caprini et al.
noted that physicians often used inadequate bridging protocols in the postoperative period.
This can have important clinical implications. They found that the 30-day mortality rate was
found to be 6% for DVT and 12% for PE in this cohort (Caprini et al., 2005).
4.3.3 Aspirin
The ACCP guidelines do not recommend using aspirin alone in any of the high-risk
orthopedic patient populations. The AAOS guidelines do sanction its use in patients with
standard risk profile for pulmonary embolism prevention (Geerts et al., 2008).
unfractionated heparin alone in total joint arthroplasty or hip fracture patients due to
inadequate evidence-based data to support its efficacy in these patient populations (Geerts
et al., 2008).
5. Newer agents
There are several new oral anticoagulants in various stages of clinical development. These
new classes target the inhibition of either thrombin or factor Xa. Most of the clinical trial
data have demonstrated equal or even superior efficacy in comparison to LMWH. However,
bleeding complications remain the primary concern. There are several other potential
complications that have been reported.
Rivaroxaban and apixabab are both inhibitors of factor Xa. Their mechanism involves the
inhibition of circulating factor Xa as well as bound factor Xa within the prothrombinase
complex (Weitz, 2006). There have been four phase III clinical trials comparing rivaroxaban
to enoxaparin (Eriksson et al., 2008). It also is approved in the European Union and Canada
for VTE prophylaxis in patients undergoing THAs and TKAs. It has recently been approved
in the United States.
Apixaban has been evaluated in several phase III clinical trials as well. It has not been
approved for use anywhere (Lassen et al., 2010a). It was found to be more efficacious than
once-daily dosing of enoxaparin, but failed to demonstrate non-inferiority to twice daily
dosing of enoxaparin (Lassen et al., 2009; Lassen et al 2010b).
RE-MODEL Dab 150-, 220- 8.1% Dab 150-QD; 6.8% Dab 150-QD;
(Eriksson, 2076 Knee 6-10 QD; Enox 40- 7.4% Dab 220-QD; N/A 5.9% Dab 220-QD;
2008b) QD 6.6% Enox 5.3% Enox
RE-
Dab 150-, 220- 3.1% Dab 150-QD; 2.5% Dab 150-QD;
MOBILIZE
2596 Knee 12-15 QD; Enox 30- 3.3% Dab 220-QD; N/A 2.7% Dab 220-QD;
(Ginsberg,
BID 3.8% Enox 2.4% Enox
2009)
ODIXa- Riv 2.5-, 5-,
KNEE 10-, 20-, 30- 2.9% Riv 5-BID; 0% Riv 5-BID; 2.9% Riv 5-BID;
613 Knee 5-9
(Turpie, BID; Enox 30- 4.8% Enox 1.9% Enox 2.9% Enox
2005) BID
ODIXa-OD- Riv 5-, 10-,
HIP 20-, 30-, 40- 2.8% Riv 10-QD; 2.1% Riv 10-QD;
845 Hip 5-9 N/A
(Eriksson, QD; Enox 40- 5,1% Enox 3.2% Enox
2006a) QD
Riv 2.5-, 5-,
ODIXa-HIP
10-, 20-, 30- 8.1% Riv 5-BID; 2.2% Riv 5-BID; 5.9% Riv 5-BID;
(Eriksson, 704 Hip 5-9
BID; Enox 40 1.5% Enox 0.8% Enox 0% Enox
2006b)
QD
Dose-
Riv 2.5-, 5-,
escalation
10-, 20-, 30- 3.8% Riv 5-BID; 2.5% Riv 5-BID; 1.3% Riv 5-BID;
study 625 Hip 5-9
BID; 30-QD; 1.9% Enox 0% Enox 1.9% Enox
(Eriksson,
Enox 40 QD
2007c)
RECORD1
Rivaroxaban Riv 10-QD; 3.2% Riv; 2.5% 2.9% Riv; 2.4%
(Eriksson, 4433 Hip 31-39 N/A
(Riv) Enox 40-QD Enox Enox
2008)
RECORD2 31-39
Riv 10-QD; 3.4% Riv; 2.8% 3.3% Riv; 2.7%
(Kakkar, 2457 Hip Riv; 10- N/A
Enox 40-QD Enox Enox
2008) 14 Enox
RECORD3
Riv 10-QD; 3.3% Riv; 2.8% 2.7% Riv; 2.3%
(Lassen, 2459 Knee 10-14 N/A
Enox 40-QD Enox Enox
2008)
RECORD4
Riv 10-QD; 3.0% Riv; 2.3% 2.6% Riv; 2.0%
(Turpie, 3034 Knee 10-14 N/A
Enox 30-BID Enox Enox
2009)
RECORD1-3
Riv 10-QD; 3.3% Riv; 2.7% 3.0% Riv; 2.5%
(Eriksson, 9349 Hip and Knee 10-39 N/A
Enox 40-QD Enox Enox
2009)
Table 3. Major bleeding rates in VTE prophylaxis clinical trails in THA and TKA. (Reference:
Huo, M. New oral anticoagulants in venous thromboembolism prophylaxis in orthopaedic
patients: Are they really better? Thromb Haemost 2011;106:45-57.
Venous Thromboembolism in Orthopaedic Surgery 167
6. Conclusion
VTE remains a challenging problem that complicates many orthopaedic procedures. The
incidence has been found to be particularly high following TKA and THA. Governmental
and consumer governing bodies are beginning to recognize it as a “never-event” indicating
that increased emphasis will be placed on prophylaxis in the years to come.
Recommendations have been released by both the ACCP and the AAOS and there remains
some disagreement as to the optimal management of VTE. Warfarin and LMWH remain the
standard of care in many practices, but newer agents show increasing promise.
The authors have several recommendations regarding the duration and type of therapy.
Patients should be anticoagulated for 25-30 days postoperatively following total hip
arthroplasty and for 14 days following a total knee arthroplasty. Certain patients with high
risk of VTE (obese, low mobility, prior VTE, family history of VTE, or protein C/S
deficiency) should be treated for 25-30 days as well following hip or knee replacement. At
our institution, we generally use enoxaparin for postoperative anticoagulation. For
inpatients, either 30mg twice daily or 40mg daily may be used following total hip
arthroplasty. The FDA has approved only the twice daily dosing after total knee
arthroplasty. For outpatients, enoxaparin 40mg daily is our regimen of choice.
7. References
Agnelli G, Eriksson BI, Cohen AT, et al. Safety assessment of new antithrombotic agents:
lessons from the EXTEND study on ximelagatran. Thromb Res 2009; 123:488–497.
AHRQ. Diagnosis and treatment of deep venous thrombosis and pulmonary embolism; US
Agency for Healthcare Research and Quality. 2007.
American Academy of Orthopaedic Surgeons. American Academy of Orthopaedic Surgeons
clinical guideline on prevention of symptomatic pulmonary embolism in patients
undergoing total hip or knee arthroplasty.
[Link]/Research/guidelines/PE_guideline pdf 2007.
Amin A, Spyropoulos AC, Dobesh P et al. Are hospitals delivering appropriate VTE
prevention? The venous thromboembolism study to assess the rate of
thromboprophylaxis (VTE start). J Thromb Thrombolysis 2010;29:326–339.
Anderson FA, Lieberman J, Pellegrini VD, et al. Practices in prevention of venous
thromboembolism in primary hip and knee arthroplasty vary with surgeon
operative volume: findings from a survey of US orthopedic surgeons. J Thromb
Haemost 2009; 7 (Suppl 2): Abstract PP-MO-248.
Aszalos A. Drug-drug interactions affected by the transporter protein, P-glycoprotein
(ABCB1, MDR1) II. Clinical aspects. Drug Discov Today 2007;12:838–843.
Bergqvist D, Agnelli G, Cohen AT, et al., for the ENOXACAN II Investigators. Duration of
prophylaxis against venous thromboembolism with enoxaparin after surgery for
cancer. N Engl J Med. 2002;346:975–980.
Burnett RS, Clohisy JC, Wright RW, et al. Failure of the American College of Chest
Physicians-1A protocol for Lovenox in clinical outcomes for thromboembolic
prophylaxis. J Arthroplasty 2007;22:317–324.
168 Deep Vein Thrombosis
Caprini JA, Botteman MF, Stephens JM et al. Economic burden of long-term complications of
deep vein thrombosis after total hip replacement surgery in the United States. Value
Health 2003;6:59–74.
Caprini JA, Tapson VF, Hyers TM et al.; NABOR Steering Committee. Treatment of venous
thromboembolism: adherence to guidelines and impact of physician knowledge,
attitudes, and beliefs. J Vasc Surg 2005;42:726–733.
Comp PC, Spiro TE, Friedman RJ, et al., for the Enoxaparin Clinical Trial Group. Prolonged
enoxaparin therapy to prevent venous thromboembolism after primary hip or knee
replacement. J Bone Joint Surg Am. 2001;83-A:336–345.
Conduah, AH; Lieberman JR (2007). Thromboembolism and Pulmonary Distress in the
Setting of Orthopaedic Surgery. In TA Einhorn, RJ O’Keefe, JA Buckwalter (Eds.),
Orthopaedic Basic Science: Foundations of Clinical Practice (3rd edition, pp. 105-113).
Rosemont, IL: AAOS.
Crowther MA, Berry LR, Monagle PT, et al. Mechanisms responsible for the failure of
protamine to inactivate low-molecular-weight heparin. Br J Haematol 2002;116:178–
186.
Eikelboom JW, Karthikeyan G, Fagel N et al. American Association of Orthopedic Surgeons
and American College of Chest Physicians guidelines for venous thromboembolism
prevention in hip and knee arthroplasty differ: what are the implications for
clinicians and patients? Chest 2009;135:513–520.
Eikelboom JW, Weitz JI. A replacement for warfarin: the search continues. Circulation
2007;116:131–133.
Eriksson BI, Agnelli G, Cohen AT, et al. Direct thrombin inhibitor melagatran followed by
oral ximelagatran compared with enoxaparin for prevention of venous
thromboembolism after total hip or knee replacement. Thromb Haemost 2003;89:
288–296.
Eriksson BI, Dahl OE, Ahnfelt L, et al. Dose escalating safety study of a new oral direct
thrombin inhibitor, dabigatran etexilate, in patients undergoing total hip
replacement: BISTRO I. J Thromb Haemost 2004; 2: 1573–1580.
Eriksson BI, Dahl OE, Buller HR, et al. A new oral direct thrombin inhibitor, dabigatran
etexilate, compared with enoxaparin for prevention of thromboembolic events
following total hip or knee replacement: the BISTRO II randomized trial. J Thromb
Haemost 2005; 3:103–111.
Eriksson BI, Borris LC, Dahl OE, et al. A once-daily, oral, direct Factor Xa inhibitor,
rivaroxaban (BAY 59–7939), for thromboprophylaxis after total hip re- placement.
Circulation 2006; 114: 2374–2381.
Eriksson BI, Borris L, Dahl OE, et al. Oral, direct Factor Xa inhibition with BAY 59–7939 for
the prevention of venous thromboembolism after total hip replacement. J Thromb
Haemost 2006; 4: 121–128.
Eriksson BI, Dahl OE, Rosencher N, et al. Dabigatran etexilate versus enoxaparin for
prevention of venous thromboembolism after total hip replacement: a randomised,
double-blind, non- inferiority trial. Lancet. 2007; 370:949--956.
Venous Thromboembolism in Orthopaedic Surgery 169
Eriksson BI, Dahl OE, Rosencher N, et al. Oral dabigatran etexilate versus subcutaneous
enoxaparin for the prevention of venous thromboembolism after total knee
replacement: the RE-MODEL randomized trial. J Thromb Haemost. 2007; 5: 2178--2185.
Eriksson BI, Borris LC, Dahl OE, et al. Dose-escalation study of rivaroxaban (BAY 59–7939)--
an oral, direct Factor Xa inhibitor--for the prevention of venous thromboembolism
in patients undergoing total hip replacement. Thromb Res 2007; 120: 685–693.
Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for
thromboprophylaxis after hip arthroplasty. N Engl J Med 2008; 358: 2765–2775. 30.
Eriksson BI, Kakkar AK, Turpie AG, et al. Oral rivaroxaban for the prevention of
symptomatic venous thromboembolism after elective hip and knee replacement. J
Bone Joint Surg Br 2009; 91: 636–644.
Freedman KB, Brookenthal KR, Fitzgerald RH Jr, et al. A meta-analysis of thromboembolic
prophylaxis following elective total hip arthroplasty. J Bone Joint Surg Am 2000; 82-
A: 929–938.
Geerts WH, Bergqvist D, Pineo GF et al. Prevention of venous thromboembolism: American
College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th ed).
Chest 2008;133(6 suppl):381S–453S.
Ginsberg JS, Davidson BL, et al. RE-MOBILIZE Writing Committee. Oral thrombin inhibitor
dabigatran etexilate vs North American enoxaparin regimen for prevention of venous
thromboembolism after knee arthroplasty surgery. J Arthroplasty 2009; 24: 1–9.
Heit JA. The epidemiology of venous thromboembolism in the community: implications for
prevention and management. J Thromb Thrombolysis 2006;21:23–29.
Heit JA, Silverstein MD, Mohr DN, Petterson TM, O’Fallon WM, Melton LJ III. Predictors of
survival after deep vein thrombosis and pulmonary embolism: a population-based,
cohort study. Arch Intern Med 1999;159:445–453.
Hirsh J, Guyatt G, Lewis SZ. Reflecting on eight editions of the American College of Chest
Physicians antithrombotic guidelines. Chest 2008;133:1293–1295.
Hoffman M, Dougald M. The action of high-dose factor VIIa in a cell-based model of
hemostasis. Disease a Month 2003; 49: 14-21
Huo, M. Prevalence and Economic Burden of Venous Thromboembolism Following Total
Joint Arthroplasty. Current Orthopaedic Practice [Link]–197.
Huo M. New oral anticoagulants in venous thromboembolism prophylaxis in orthopaedic
patients: are they really better? Thromb Haemost 2011; 106:45-57.
Imperiale TF, Speroff T. A meta-analysis of methods to prevent venous thromboembolism
following total hip replacement. J Am Med Assoc 1994; 271: 1780–1785.
Kakkar AK, Brenner B, Dahl OE, et al. Extended duration rivaroxaban versus short-term
enoxaparin for the prevention of venous thromboembolism after total hip
arthroplasty: a double-blind, randomised controlled trial. Lancet 2008;372: 31–39.
Kim S. Changes in surgical loads and economic burden of hip and knee replacements in the
US: 1997–2004. Arthritis Rheum 2008;59:481–488.
Kolb G, Bodemer I, Galster H et al. Reduction of venous thromboembolism following
prolonged prophylaxis with the low molecular weight heparin Certoparin after
endoprosthetic joint replacement or osteosynthesis of the lower limb in elderly
patients. Thromb Haemost 2003;90:1100–1105
170 Deep Vein Thrombosis
Lassen MR, Borris LC, Anderson BS, et al. Efficacy and safety of prolonged
thromboprophylaxis with a low molecular weight heparin (dalteparin) after total
hip arthroplasty—the Danish Prolonged Prophylaxis (DaPP) Study. Thromb Res.
1998;89:281–287.
Lassen MR, Davidson BL, Gallus A, et al. A phase II randomized, double-blind five-arm
parallel-group, dose-response study of a new oral directly-acting factor Xa
inhibitor, razaxaban, for the prevention of deep vein thrombosis in knee re-
placement surgery. Blood 2003; 102: Abstract 41.
Lassen MR, Davidson BL, Gallus A, et al. The efficacy and safety of apixaban, an oral, direct
factor Xa inhibitor, as thromboprophylaxis in patients following total knee
replacement. J Thromb Haemost 2007; 5: 2368–2375.
Lassen MR, Ageno W, Borris LC, et al. Rivaroxaban versus enoxaparin for
thromboprophylaxis after total knee arthroplasty. N Engl J Med 2008; 358: 2776–
2786.
Lassen MR, Raskob GE, Gallus A, et al. Apixaban or enoxaparin for thromboprophylaxis
after knee replacement. N Engl J Med 2009;361:594–604.
Lassen MR, Gallus A, Raskob GE, Pineo G, Chen D, Ramirez LM; ADVANCE-3
Investigators. Apixaban versus enoxaparin for thromboprophylaxis after hip re-
placement. N Engl J Med 2010; 363: 2487–2498.
Lassen MR, Raskob GE, Gallus A, Pineo G, Chen D, Hornick P; ADVANCE-2 investigators.
Apixaban versus enoxaparin for thromboprophylaxis after knee replacement
(ADVANCE-2): a randomised double-blind trial. Lancet 2010; 375: 807–815.
Lee WM. Drug-induced hepatotoxicity. N Engl J Med 2003;349:474–485.
Leizorovicz A, Haugh MC, Chapuis FR, et al. Low molecular weight heparin in prevention
of perioperative thrombosis. Br Med J 1992; 305: 913–920.
Markel DC, York S, Liston MJ Jr, Flynn JC, Barnes CL, Davis CM 3rd; AAHKS Research
Committee. Venous thromboembolism: management by American Association of
Hip and Knee Surgeons. J Arthroplasty 2010;25:3–9.
Mega JL, Braunwald E, Mohanavelu S, et al. Rivaroxaban versus placebo in patients with
acute coronary syndromes (ATLAS ACS-TIMI 46): a randomised, double-blind,
phase II trial. Lancet 2009; 374: 29–38.
Moyer TP, O’Kane DJ, Baudhuin LM, et al. Warfarin sensitivity genotying: a review of the
literature and summary of patient experience. Mayo Clin Proc [Link]–1094.
Ng HJ, Crowther MA. New anti-thrombotic agents: emphasis on hemorrhagic complications
and their management. Semin Hematol 2006; 43 (1 Suppl 1): S77–83.
Noble SI, Finlay IG. Is long-term low-molecular-weight heparin acceptable to palliative care
patients in the treatment of cancer related venous thromboembolism? A qualitative
study. Palliat Med. 2005;19:197–201.
Nurmohamed MT, Rosendaal FR, Büller HR, et al. Low-molecular-weight heparin versus
standard heparin in general and orthopaedic surgery: a meta-analysis. Lancet 1992;
340: 152–156.
Oster G, Ollendorf DA, Vera-Llonch M, Hagiwara M, Berger A, Edelsberg J. Economic
consequences of venous thromboembolism following major orthopedic surgery.
Ann Pharmacother 2004;38:377–382.
Venous Thromboembolism in Orthopaedic Surgery 171
Wolowacz SE, Roskell NS, Plumb JM, Caprini JA, Eriksson BI. Efficacy and safety of
dabigatran etexilate for the prevention of venous thromboembolism following total
hip or knee arthroplasty. A meta-analysis. Thromb Haemost 2009; 101: 77–85.
Xarelto product monograph, Bayer Inc., Canada, September 10, 2008. Available at:
[Link]/files/XARELTO-PM-ENG-10SEP2008–[Link]. Accessed
September 29, 2010.
11
1. Introduction
Deep venous thrombosis or DVT is a blood clot formation in one or more of the deep veins.
The blood clot does not break down and therefore, it can become larger and occlude the
blood flow within the affected vein. The most frequent sites are the leg veins (femoral and
popliteal) and the deep pelvic veins. Rarely, the arm veins are affected (Paget-Schrötter
disease). Pulmonary embolism (PE) is the most dangerous complication of DVT. PE occurs
when the clot breaks into small pieces (emboli) and travel to the lung. The embolus may
travel to other vital organs and cause life-threatening complications such as stroke or heart
attack.
The etiology of thrombosis is exactly unknown, however, the Virchow’s triad of slow
circulation (stasis), increased blood coagulability and vessel wall intimal injury is the alleged
mechanism.
DVT and PE developing after trauma and pelvic surgery are of a major concern to surgeons
of all subspecialties. Therefore, proper assessment of the patient risk to develop DVT is of
paramount importance. The risk of DVT can be decreased significantly by adopting some
appropriate prophylactic procedures.
Although adopting anti-DVT prophylactic measures is not debatable, the use of these
measures has not yet been a universal issue, even in patients having no contraindications to
their use.
In this chapter, the term “radical pelvic surgeries” mean all types of major surgeries
performed to treat malignancies developing in the pelvis, such as radical cystectomy,
salvage cystectomy, radical prostatectomy, radical or pan-hysterectomy, radical surgery for
colo-rectal cancer and excision of a local tumor recurrence after primary radical surgery or
after definitive radiotherapy
2. Incidence
DVT constitutes a major health problem, especially among the elderly. In comparison with
previous era, the incidence of DVT remains the same among men and possibly increasing in
elderly females (Silverstein et al., 1998). On the other hand, the incidence of PE is decreasing
174 Deep Vein Thrombosis
over years (Silverstein et al., 1998). However, the incidence of DVT and PE may be
underestimated because of the missed diagnosis, absence of pertinent symptoms or the
absence of laws to permit routine autopsies in sudden post-operative mortalities in most
centers (Dalen & Alpert, 1975; Clagett, 1994). Furthermore, unexplained DVT may be the
first presentation in some malignancies, such as prostate, colorectal and bladder (Monreal &
Prandoni, 1999).
In a series of 2373 patients, the incidence of DVT was 0.87% after urologic surgeries for
prostate and bladder tumors, 2.8% in general surgery and 2% in gynecological surgeries
(Scarpa etal., 2007).
The incidence of DVT may be as low 2% after radical cystectomy (Ali-El-Dein et al., 2008;
Ghoneim et al., 2008), or as high as 40% following prolonged gynecological or obstetrical
surgery (Walsh et al., 1974; Clarke-Pearson et al., 1983). Patients undergoing large bowel
surgery also have a considerable risk of DVT and pulmonary embolism. The incidence of
DVT following radical cystectomy in our hospital is 2% to 2.6% and PE following DVT or
without prior DVT has long been a leading cause of post-operative death (Ali-El-Dein et al.,
2008; Ghoneim et al., 2008). In patients undergoing surgery or radiotherapy for treatment of
localized prostate cancer the incidence of DVT was 2% for pelvic lymphadenectomy alone
and 1.9% following prostatectomy, while fatal PE occurred in 2 patients (3.7%) after
prostatectomy (Bratt et al., 1994).
The incidence of DVT after gynecologic and obstetrical surgeries varies according to the
presence or absence of the known risk factors among patients and according to the methods
of diagnosis. It has been reported that this incidence is 14% after benign gynecological
surgeries (Walsh et al., 1974), while the rate has been higher (38%) for patients undergoing
surgery for gynecological tumors (Crandon & Knotts, 1983). In addition, among all causes of
death following gynecologic surgeries, PE has been a leading cause of postoperative
mortality in high risk women with gynecologic malignancy (Clarke-Pearson et al., 1983).
Following laparoscopic radical hysterectomy for cervical carcinoma the incidence of DVT
has been 3% (Chen et al., 2008).
In the study of yang et al. on 3645 patients undergoing surgery for colorectal cancer, 31
(0.85%) developed symptomatic venous thromboembolism or VTE (Yang et al., 2011).
Stasis: -Immobilization.
-Pelvic masses.
- A gravid uterus
- Surgically induced hematomas.
- lymphocysts also can lead to venous stasis
Endothelial injury of the vessel wall may be induced by surgical dissection in various
radical pelvic surgeries (e.g. radical cystectomy) or from infiltration of the vessel wall by the
tumor. In addition, catheters placed distally or proximally in the venous system are among
the risk factors (Evans et al., 2010). However, in this situation, the risk of DVT/PE is
determined by multiple factors including catheter size (Evans et al., 2010), degree of vein
trauma during catheter insertion and dwell and hypercoagulability of the patient’ blood.
Hypercoagulability or thrombophilia or prothrombotic state is a blood coagulation disorder
with a subsequent increase in the incidence of thrombosis (Heit, 2007). There are multiple
genetic and acquired risk factors that influence thrombophilia. The presence of these
inherited risk factors alone usually does not cause thrombosis unless an additional risk
factor is present (Heit, 2007; Kyrle et al., 2010).
Antithrombin deficiency, which is the first major form of thrombophilia, was identified in
1965, while the most common defects, such as factor V Leiden mutation and prothrombin
gene mutation G20210A were described in the 1990s (Dahlbäck, 2008; Rosendaal & Reitsma,
2009). The risk of developing DVT/PE increases significantly if one of these abnormalities is
present in patients undergoing radical pelvic surgery.
There are various possibilities, which can induce a hypercoagulable state during major
radical pelvic surgeries. These possibilities include decreased fibrinolytic activity associated
with surgery (Egan et al., 1974), increased level of coagulation factors I, V, VIII, IX, X, and
XI, the presence of activated intermediate coagulation products such as thrombin-
antithrombin III complexes and abnormalities of the platelets (Piccioli et al., 1996). In
addition, the malignant cells may secrete a substance promoting coagulation, such as tissue
factor and cancer procoagulant or factors that influence vascular endothelial permeability
such as vascular endothelial growth factor and subsequently stimulate fibrin deposition
(Goad & Gralnick, 1996).
In the prospective study of Duke University Medical Center 411 patients undergoing major
abdominal and pelvic gynecologic surgery were evaluated for DVT and the related possible
risk factors (Clarke-Pearson et al., 1987). In this study, the important factors, which
maintained statistical significance in stepwise logistic regression model were age, edema of
the ankle, type of surgery, nonwhite race, presence of varicose veins, history of radiation
preoperatively, past DVT and duration of surgery.
It has been found that the risk factors for distal DVT are different from those of proximal
DVT. In the national (France) multicenter prospective OPTIMEV study, out of 6141 patients
with symptoms suggestive of DVT, diagnosis was objectively confirmed in only 1643 and
isolated distal DVT was more common than proximal one (Galanaud et al., 2009). In this
study, acute or transient risk factors, such as recent surgery, recent plaster immobilization
and recent travel, were more frequently discovered in distal DVT. On the other hand, in
proximal DVT chronic risk factors such as active cancer, congestive heart failure or
respiratory insufficiency and age above 75 years were more frequent.
Active cancer and related chemotherapy can increase the incidence of DVT by multiple
mechanisms. In chronic lymphocytic leukemia patients, studies showed a link between
lenalidomide associated DVTs and inflammation, upregulation of TNFα and endothelial cell
dysfunction (Aue etal., 2011).
Deep Venous Thrombosis After Radical Pelvic Surgery 177
4. Diagnosis of DVT/PE
The majority of cases of DVT/PE have one or more risk factor. Many cases of DVT/PE are
asymptomatic. Suspected pulmonary embolism is a medical emergency and can be fatal. In
symptomatic DVT cases, the patient may present with lower limb pain, unilateral leg swelling,
redness and sometimes prominent superficial veins. A tender calf, especially with dorsiflexion
(Homan’s sign) and rarely a palpable venous cord are among the possible physical signs.
However, the presence of these manifestations is nonspecific, because in more than 50% of the
cases presenting with these symptoms, DVT is absent (Dainty et al., 2004). Therefore, diagnosis
of DVT based on symptoms only is problematic and proper hospital assessment and further
diagnostic tools are needed for accurate diagnosis. Similarly, most of the symptoms and signs
of PE are nonspecific and simulate post-surgery pulmonary complications. However,
physicians should maintain a high degree of suspicion if the patient is complaining of pleuritic
chest pain, hemoptysis, dyspnea, tachycardia and tachypnea.
(Rabinov & Paulin, 1972). Currently, contrast venography is rarely indicated nowadays and
has been replaced by the noninvasive measures. It is sometimes performed to confirm the
diagnosis of a clinically suspected DVT. However, if noninvasive imaging is normal or
inconclusive and still DVT is clinically suspected, venography is done to confirm the
diagnosis. In the cases of clinical suspicion of DVT, a negative contrast venography rules out
the need for anticoagulant treatment (Hull et al., 1981). The test has certain limitations and
complications.
Magnetic resonance venography (MRV) is an accurate noninvasive venographic technique for
the detection of DVT. It has a sensitivity and specificity comparable to contrast venography
(Carpenter et al., 1993). Furthermore, it can detect thrombi places not seen by the conventional
venography, such as pelvic, ovarian veins or vena cava. Two major limitations for MRV are
present, namely, the expensive cost and prolonged time necessary (Carpenter et al., 1993).
Scintigraphy has been described as a diagnostic tool for DVT (Knight, 1993). However, the
data of its clinical efficacy compared to the standard methods are still lacking.
4.3 Imaging in PE
The diagnosis of PE may be made by a variety of imaging techniques, including chest X-ray,
ventilation-perfusion scan, computed tomography (CT) of the chest vessels and pulmonary
angiography.
On clinical suspicion of PE, the initial evaluation is made using chest X-ray,
Electrocardiography (ECG) and ABG. Further evaluation is made by ventilation-perfusion
scan, CT of the chest vessels (Gulsun Akpinar & Goodman, 2008).
Currently, CT venography combined with pulmonary CT angiography for the detection of
PE is increasingly used to confirm the diagnosis of suspected PE and the results have been
extremely promising (Krishan et al., 2011).
5. Prophylaxis of DVT/PE
The incidence of DVT and subsequent PE can be decreased by adopting certain prophylactic
mechanical and/ or pharmacologic measures, which have been proved to be safe and
effective in most types of major surgeries (Martino et al., 2007; Geerts et al., 2008).
Mechanical methods act by reducing stasis of venous blood and may stimulate endogenous
fibrinolysis, while pharmacologic agents act by clot prevention through the various steps of
the clotting cascade (Martino et al., 2007; Geerts et al., 2008).
(LMWH) are the drugs of choice in patients undergoing radical pelvic operations in the
fileds of general, vascular, major urologic and gynecologic surgeries (Agnelli, 2004). In
urologic patients judged as low-risk, early postoperative mobilization is the only measure
needed. On the other hand, higher-risk patients should receive vitamin K antagonists,
LMWH and/ or fondaparinux (Agnelli, 2004).
Some investigators recommended a double prophylaxis of mechanical measures as well as
pharmacologic measures using pre- and post-operative anticoagulation, usually in the form
of LMWH (Whitworthet al., 2011). They found that the use of preoperative anticoagulation
seems to significantly decrease the risk of DVT in high-risk patients undergoing major
gynecologic surgeries. In addition, there was no significant change in the rates of
complications secondary to this protocol.
6. Treatment of DVT/PE
The goals of treatment of patients with DVT and PE are to prevent local growth of the
thrombus, prevent the thrombus from breaking down into small pieces (emboli) and
traveling to other places, prevent complications of DVT, prevent recurrence of the thrombus
and in some clinical situations accelerate fibrinolysis (Hirsh & Hoak, 1996).
DVT is treated by immediate institution of anticoagulant therapy. Treatment is given as
either unfractionated heparin or low molecular weight heparins, followed by few weeks to 6
months of oral anticoagulant therapy (Clarke-Pearson & Abaid, 2008). However, life-long
anticoagulation has been recommended in some patients with active cancers after partial
improvement or failure of treatment, because they remain at very high risk to recurrent DVT
(Clarke-Pearson & Abaid, 2008).Low concentrations of heparin can inhibit the early stages of
blood coagulation. However, higher concentrations are needed to inhibit the much higher
concentrations of thrombin that are formed if the DVT process is not modulated (Hirsh &
Hoak, 1996).
180 Deep Vein Thrombosis
7. Conclusion
Deep venous thrombosis and pulmonary embolism are among the major post-operative
complications that develop after radical pelvic surgeries. Pulmonary embolism is one of the
leading causes of post-operative mortality in these patients. Most of the cases are
asymptomatic and in the majority of patients dying from pulmonary embolism the
embolism is diagnosed at autopsy. Treatment is essentially prophylactic and the primary
treatment objectives are to prevent PE, decrease morbidity and to prevent the risk of
developing the post-thrombotic syndrome (PTS). High-risk patients may be subject for dual
mechanical and pharmacologic prophylaxis with good results. Anticoagulation provides the
main stay of treatment. Thrombolytic therapy is currently used for massive pulmonary
embolism and some selected cases of deep venous thrombosis. Surgical (thrombectomy or
embolectomy) or endovascular techniques have been tried with promising results.
8. References
Ali-El-Dein, B., Shaaban, A.A., Abu-Eideh, R.H., El-Azab, M., Ashamallah, A. & Ghoneim,
M.A. (2008). Surgical complications following radical cystectomy and orthotopic
neobladders in women. J Urol,180,1,206-10.
Agnelli, G. (2004). Prevention of venous thromboembolism in surgical patients. Circulation,
110, (24 Suppl 1), IV4-12.
Aue, G., Nelson Lozier, J., Tian, X., Marie Cullinane, A., Soto, S., Samsel, L., McCoy, P. &
Wiestner, A. (2011). Inflammation, TNFα and endothelial dysfunction link
lenalidomide to venous thrombosis in chronic lymphocytic leukemia. Am J Hematol,
Jun 27. doi: 10.1002/ajh.22114. [Epub ahead of
print][[Link]
Bratt, O., Elfving, P., Flodgren, P. & Lundgren, R. (1994). Morbidity of pelvic
lymphadenectomy, radical retropubic prostatectomy and external radiotherapy in
patients with localised prostatic cancer. Scand J Urol Nephrol, 28,3,265-71.
Carpenter, J.P., Holland, G.A., Baum, R.A., Owen, R.S., Carpenter, J.T. & Cope, C. (1993).
Magnetic resonance venography for the detection of deep venous thrombosis:
comparison with contrast venography and duplex Doppler ultrasonography. J Vasc
Surg, 18,5, 734-41.
Chen, Y., Xu, H., Li, Y., Wang, D., Li, J., Yuan, J. & Liang, Z. (2008). The outcome of
laparoscopic radical hysterectomy and lymphadenectomy for cervical cancer: a
prospective analysis of 295 patients. Ann Surg Oncol, 15,10,2847-55.
Chung, J. & Owen, R.J.T. (2008). Using inferior vena cava filters to prevent pulmonary
embolism. Can Fam Physician, 54,1, 49 – 55 .
Clagett, G.P. (1994) Prevention of postoperative venous thromboembolism: An update. Am J
Surg, 168,6, 515-22.
Clark, C. & Cotton, L.T. (1968). Blood-flow in deep veins of leg: Recording technique and
evaluation of methods to increase flow during operation. Br J Surg,55,3, 211-4.
Clarke-Pearson, D.L., Jelovsek, F.R. & Creasman, W.T. (1983). Thromboembolism
complicating surgery for cervical and uterine malignancy: Incidence, risk factors
and prophylaxis. Obstet Gynecol, 61,1, 87-94.
Clarke-Pearson, D.L., DeLong, E.R., Synan, I.S., Coleman, R.E. & Creasman, W.T. (1987).
Variables associated with postoperative deep venous thrombosis: a prospective
182 Deep Vein Thrombosis