Next Article in Journal
Diagnosis of Cervical Cancer and Pre-Cancerous Lesions by Artificial Intelligence: A Systematic Review
Previous Article in Journal
Placenta Accreta Spectrum (PAS) Disorder: Ultrasound versus Magnetic Resonance Imaging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

D-dimer Testing in Pulmonary Embolism with a Focus on Potential Pitfalls: A Narrative Review

1
Department of Laboratory Medicine, Clinique St-Luc Bouge, 5004 Namur, Belgium
2
Department of Pharmacy, Namur Research Institute for Life Sciences, University of Namur, 5000 Namur, Belgium
3
Namur Thrombosis and Hemostasis Center (NTHC), CHU UCL Namur, Université Catholique de Louvain, 5530 Yvoir, Belgium
4
Department of Pharmacy, Namur Thrombosis and Hemostasis Center (NTHC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
5
QUALIblood s.a., 5000 Namur, Belgium
6
Department of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
7
Department of Emergency Medicine, CHU Angers, Institut MITOVASC, Equipe CARME, UMR CNRS 6015-INSERM 1083, UNIV Angers, F-CRIN INNOVTE, 49000 Angers, France
8
Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam UMC Location Universiteit van Amsterdam, 1105 Amsterdam, The Netherlands
9
Department of Vascular Medicine, Amsterdam UMC Location Universiteit van Amsterdam, 1105 Amsterdam, The Netherlands
10
Clinicial Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
11
Department of Internal Medicine and Thrombosis Expertise Center, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
12
Center for Thrombosis and Hemostasis, Gutenberg University Medical Center, 55131 Mainz, Germany
13
Department of Nuclear Medicine, CHU UCL Namur, 5530 Yvoir, Belgium
14
Institute of Experimental and Clinical Research, UCLouvain, 1348 Louvain-la-Neuve, Belgium
15
Namur Research Institute for Life Sciences, University of Namur, 5000 Namur, Belgium
16
Department of Radiology, CHU UCL Namur, 5530 Yvoir, Belgium
17
Division of Vascular Medicine, CHRU de Nancy, Université de Lorraine, 54000 Nancy, France
18
Section of Clinical Biochemistry, University Hospital of Verona, 37134 Verona, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2022, 12(11), 2770; https://doi.org/10.3390/diagnostics12112770
Submission received: 27 September 2022 / Revised: 8 November 2022 / Accepted: 9 November 2022 / Published: 12 November 2022
(This article belongs to the Section Pathology and Molecular Diagnostics)

Abstract

:
D-dimer is a multifaceted biomarker of concomitant activation of coagulation and fibrinolysis, which is routinely used for ruling out pulmonary embolism (PE) and/or deep vein thrombosis (DVT) combined with a clinical pretest probability assessment. The intended use of the tests depends largely on the assay used, and local guidance should be applied. D-dimer testing may suffer from diagnostic errors occurring throughout the pre-analytical, analytical, and post-analytical phases of the testing process. This review aims to provide an overview of D-dimer testing and its value in diagnosing PE and discusses the variables that may impact the quality of its laboratory assessment.

1. Introduction

What is universally referred to as “D-dimer” is actually a mixture of fibrin degradation products (FnDP) that contain a “D-dimer” motif, obtained from cross-linked fibrin digestion by plasmin [1,2,3,4]. Thrombin, activated factor XIII (factor XIIIa), and plasmin are the three successive enzymes responsible for D-dimer formation. Fibrinogen contains a central E-domain connected to two outer D-domains and consists of three pairs of polypeptide chains (Aα-, Bβ-, and γ-) [5,6]. Thrombin releases fibrinopeptides A and B, generating fibrin monomers and unraveling two cryptic polymerization sites located on the E domain. Self-assembling of fibrin monomers leads to the formation of a network, which becomes insoluble (fibrin deposits) after reaching a critical size [1]. Thrombin activates FXIII into FXIIIa in a reaction that is enhanced by fibrin [7]. FXIIIa in turn promotes the generation of stable fibrin clots, the lysis of which is slowed down [1,4]. The generation of plasmin from plasminogen catalyzed by tissue plasminogen activator and urokinase-type plasminogen activator leads to the degradation of fibrin by cleavages at several specific sites [4]. Fibrinolysis generates products with a wide range of molecular masses, referred to as FnDP. The smallest final digestion product of fibrin clots is the D-dimer/fragment E (DD/E) complex, which presents two adjacent covalently bound D-domains from two fibrin monomers cross-linked by factor XIIIa [4].
Initially, the term “D-dimer fragment” only referred to the DD/E complex [1,8]. Nonetheless, the range of species recognized by antibodies in D-dimer assays is wider, including species with masses ranging from approximately 200 to over 10,000 kDa [1,9]. In healthy subjects, D-dimer is detectable in small amounts because of the physiological conversion of fibrinogen to fibrin [4,10]. The half-life of D-dimer in vivo is estimated to be approximately 8 h [11].
D-dimer immunoassays are designed to detect a specific epitope on degradation products of factor XIIIa-cross-linked fibrin and should therefore not recognize X, Y, D, or E fragments but only the D-dimer motif in the smallest DD/E fragment and in larger FnDP [3].

2. Interest in D-dimer Testing in PE

Pulmonary embolism (PE) is defined as the obstruction of blood flow in a pulmonary artery by an embolus that has usually originated from a thrombus in the lower limb veins. Its incidence is approximately 60 to 120 per 100,000 people/year. The clinical picture mainly includes acute chest pain, shortness of breath (i.e., dyspnea), or syncope, and diagnosis requires chest imaging [12]. D-dimer testing is widely used as a gold standard for ruling out venous thromboembolism (VTE) in patients classified as having a low-intermediate pre-test probability based on a clinical decision rule (CDR) or implicit assessment [1,4,13,14,15,16]. In addition to almost all cases of VTE, increased D-dimer levels may also be observed in many conditions, such as infection, pregnancy, trauma, cancer, and aging as well as in hematomas or interstitial hemorrhages [2]. Interpretation of D-dimer assays should be performed following local regulations that may apply, as some uses may fall outside the requirements of intended use approved by the local regulatory agency [17,18,19]. The use of tests outside those directives (e.g., age-adjusted cut-offs) may constitute a laboratory-developed test. Other indications for D-dimer testing include assessing the risk of recurrent thrombosis and guiding anticoagulant therapy and diagnosing and monitoring disseminated intravascular coagulation although the use of D-dimer in those settings is controversial or less well-established [1,4,20]. Table 1 summarizes the characteristics of some relevant central laboratory and point-of-care (POC) assays, including the method type and the “intended use” issued by the U.S. Food and Drugs Administration (FDA) [18]. Regarding test performance, the recommendations from the FDA are ≥95% (with a lower limit of confidence interval (CI) ≥90%) sensitivity and ≥97% (with a lower limit of CI ≥95%) negative predictive value (NPV) [21], while the Clinical and Laboratory Standards Institute (CLSI, former NCCLS) suggests specifications of ≥97% (with a lower limit of CI ≥90%) sensitivity and ≥98% (with a lower limit of CI ≥95%) NPV to rule out VTE. Quantitative assays have higher sensitivity than qualitative or semi-quantitative assays and should hence be preferred [3]. Of note, accurate estimation of the performance of D-dimer immunoassays is pivotal and required by the In Vitro Diagnostics Regulation (IVDR) [22]. Regarding POC assays, whole-blood D-dimer assays may display higher specificity for PE than laboratory assays (i.e., 69%) but at the expense of lower sensitivity (i.e., 87%) [23,24].
The use of validated CDR in combination with D-dimer testing is recommended to identify patients with suspected VTE in whom the diagnosis can be ruled out without imaging [2,4,13,25,26,27,28,29]. A score is assigned to an individual based on signs, symptoms, and risk factors, which can be used to classify patients into risk categories for VTE (e.g., low, intermediate, or high probability). Straightforward two-level scores are preferred since they perform as well as three-level scores and are easier to use [25,26]. Numerous CDR for suspected PE are available, among which the Wells’ PE and revised Geneva scores are the most widely used and display comparable diagnostic performances [1,16,30,31,32,33]. By using an approach that combines a CDR with D-dimer testing to rule out PE, the risk of withholding imaging in patients with false-negative D-dimer (failure rate) is very low [1,2,4,13,34]. The use of a combination of CDR and D-dimer testing may be safely applied when the incidence of thromboembolic events is below 1–2% after 3 months in patients in whom VTE was excluded without imaging testing [35,36]. Imaging testing is recommended, and D-dimer testing is unnecessary in patients with “high” or “likely” probability [2,36,37].
The use of D-dimer testing alone was evaluated with the YEARS algorithm that, in essence, excluded PE in all patients with a high-sensitive D-dimer < 500 µg/L fibrinogen-equivalent units (FEU) [33]. In the YEARS study, the overall failure rate (1 minus NPV) of this diagnostic algorithm was well below the accepted threshold of 2%. The recently introduced PEGeD algorithm, which also uses varying D-dimer thresholds dependent on the clinical pretest probability, maintains the approach of not using D-dimer alone to rule out PE in patients with a high pretest probability based on the Wells’ score [38].
An age-adjusted cut-off for D-dimer is defined as age times 10 µg/L in those aged 51 years or older, while the traditional threshold of 500 µg/L FEU is used in those 50 years or younger. Age-adjusted cut-offs combined with results of a clinical score (i.e., Wells’ or revised Geneva score) were demonstrated by the ADJUST-PE study to enable safe PE exclusion without additional computed tomography pulmonary angiography (CTPA) [27,39]. The use of such cut-offs is now one of the recommended diagnostic approaches for patients with suspected PE in order to enhance the clinical specificity while maintaining a clinically usable NPV [13,30,40,41,42,43]. For example, the European Society of Cardiology recently endorsed the use of age-adjusted cut-offs for ruling out PE [28].
Interestingly, D-dimer may also be used to rule out VTE in specific clinical circumstances such as pregnancy, cancer, or kidney disease [11].

3. Pre-Analytical Step

Among all the various steps of the total testing process, the pre-analytical phase is currently considered as the main source of diagnostic errors, accounting for 60–70% of all laboratory mistakes [44]. The International Organization for Standardization 15189:2012 standard for laboratory accreditation is defined as “processes that start, in chronological order, from the clinician’s request and include the examination request, preparation and identification of the patient, collection of the primary sample(s), the transport to and within the laboratory, and end when the analytical examination begins” [45]. The pre-analytical requirements for D-dimer analysis are often wrongly assimilated to those for other coagulation tests [46,47]. This misperception may come from the fact that, although testing methods are diametrically different, D-dimer and other hemostasis tests are generally performed using the same citrated tube.
The first variable that affects the D-dimer measurement is sample collection. To perform venipuncture, it is recommended that the tourniquet should not obstruct the arterial blood flow, and venous blood flow should be freed as soon as the needle enters the vein or when the first tube starts to fill [48,49]. If applied longer than 1 to 2 min, the obstruction of blood flow may induce both hemoconcentration and thrombin generation, therefore interfering with accurate hemostasis testing [48,49,50]. In a study on the effect of tourniquet application on D-dimer, 1 min of venous stasis already increased D-dimer values on average by 7.9% using the Vidas DD assay (BioMérieux, Marcy l’Etoile, France) [50].
The act of collection should be performed by ordinary straight needles with a caliber between 19–22 gauge (G) and confer as little trauma as possible [48]. Butterfly devices, even with small-sized needles, constitute an appropriate alternative approach to standard straight needles when needed [50,51] although a mandatory discard tube must be drawn prior to sample collection to ensure removing any air present within the tubing. The presence of air may be responsible for inadequate volume in the tube and subsequent disturbances or interferences [52]. Straight-needle venipunctures are to be preferred when other hemostasis analyses are requested [13]. In this case, the use of a discard tube does not have a significant impact on D-dimer results and is therefore unnecessary when D-dimer is the only test requested [53].
The tube material as well as the additive used for anticoagulation may also be sources of variation. Different tube materials were studied (i.e., glass or polyethylene terephthalate plastic collection tubes (Vacutainer, Becton Dickinson (BD), Franklin Lakes, NJ, USA) as well as glass citrated collection tubes (Vacuette, Grenier bio-one, Germany)) and showed no significant differences in D-dimer values [54,55,56]. Altogether, the choice of silicone-coated glass or polypropylene plastic tubes does not substantially impact the quality of D-dimer measurement. The CLSI and the World Health Organization recommend the use of collection tubes containing 3.2% (105–109 mmol/L) buffered sodium citrate anticoagulant [49,52]. Alternative sample matrices should be locally validated before being used in routine clinical practice. Few D-dimer assays (including POC tests) also allow using heparinized or ethylenediamine tetraacetic acid plasma, whilst only citrate anticoagulated blood is recommended for others [57]. Reports in the literature do not agree on the influence of heparin tubes, stating that a modest bias could appear, which could be corrected by a correction factor to adjust for the dilution induced by the presence of the anticoagulant [58,59,60]. Importantly, a blood-to-anticoagulant ratio of 9:1 must be respected once the specimen is collected to prevent underestimation of D-dimer due to excessive sample dilution [4,49,61]. Sample collection should ensure free flow into the tube and be mixed within 30 s after completing the phlebotomy (i.e., three to six complete inversions), thus ensuring homogenization of blood and anticoagulant [49].
Samples delivery should be performed as fast as possible (i.e., usually <1 h) after collection, in a vertical position and at ambient temperature (15–22 °C) [48,49,52]. Owing to accessibility and rapidity, a reduction in turnaround time (TAT) may be obtained using a pneumatic tube system (PTS). Although a low impact of PTS on D-dimer test results has been noted [58,62,63], PTS should be assessed and validated locally since these systems are rather heterogeneous in terms of length, materials, internal diameter, acceleration (or deceleration) forces, and speed [62,64].
Sample processing is another important pre-analytical variable. Once received in the laboratory and prior to analysis, the specimens should be carefully examined to identify those that do not comply with pre-analytical requirements (e.g., inappropriate additive, identification errors, insufficient volume, presence of clots). To ensure patient safety, noncompliant samples should be rejected, or test results should be otherwise suppressed [48]. Automation (i.e., pre-analytical modules) may help identify pre-analytical issues [65]. Then, prior to performing most D-dimer analysis, plasma needs to be separated from cellular components, usually through centrifugation at 1500× g for at least 15 min at ambient temperature [52]. Shorter centrifugation schemes may also be used, especially for stat testing, and are of major interest to ensure shorter TAT [66,67]. The impact of the temperature of centrifugation was also shown to be non-significant [34,68].
Finally, the stability of D-dimer in collecting tubes prior to analysis is of the utmost importance to ensure appropriate results. Various real-life studies suggested that sample stability might be sufficient to allow longer delays than stated by current recommendations (i.e., no more than 4 h room temperature (15–25 °C) prior to D-dimer testing [48]). D-dimer is stable for at least 24 h (in plasma/whole blood) at room temperature or at 2–8 °C with many different D-dimer immunoassays before performing D-dimer testing. Several freezing-thawing cycles did not induce significant changes in D-dimer values [58,69,70,71]. Due to the important clinical decisions that could be taken (e.g., ruling out acute episodes of PE), rapid D-dimer testing is typically required, and delayed analysis (e.g., frozen samples) would prove most useful in research settings [13,69,72,73,74,75].
Importantly, like all immunoassays, D-dimer testing may be affected by interferences that significantly impact test results (i.e., 0.4 to 4.0% of all analyses) [65]. With a prevalence ranging between 30–70% of all unsuitable specimens, in vitro hemolysis is the most frequent interference in hemostasis testing and is caused by various conditions (e.g., hemolytic anemia, metabolic disorders), traumatic venipuncture, sample transportation, processing (e.g., delay before centrifugation), and storage (e.g., unsuitable conditions of temperature and duration) [76,77,78,79,80]. Notably, it has been observed that the majority of hemolyzed samples (±95%) in clinical laboratories are only “mildly” hemolytic (e.g., cell-free hemoglobin concentration between 0.3–0.6 g/L, giving a faint reddish hue to the sample) [76,78,81]. D-dimer values might still be reliable when the concentration of cell-free hemoglobin remains within a non-interference limit (i.e., <3 g/L) and could hence be safely reported to the clinicians [78]. Greater hemolysis might, however, lead to biased results, which would need to be suppressed in order to safeguard patient safety [77,78,82,83]. The impact of other frequent sources of interference, such as lipemia, icterus, proteinemia (including monoclonal gammopathy) and heterophilic antibodies, is less-documented [77,84,85,86]. Some studies observed no impact of lipemia and icterus [87,88,89,90,91,92,93]. Spurious D-dimer results were only observed with the STA reagents (Diagnostica Stago, Asnières sur Seine, France) when adding high concentrations of free bilirubin at 80 mg/dL and above [94]. Moreover, the HIL check was able to detect lipemic samples (triglyceride levels > 400 mg/dL) on the CS-5100 D-dimer assay (Sysmex, Kobe, Japan) in three out of four samples [95]. Recently, patient pools with low D-dimer concentrations (<750 µg/L FEU) were shown to be significantly affected by lipemia (HemosIL HS500, Werfen, San Diego, CA, USA) [96]. Interestingly, excessive lipemia might be decreased using high-speed centrifugation (e.g., at 10,000 g) [97]. Multiple case reports suggested or illustrated an interference of monoclonal gammopathy in D-dimer assays [85,98,99,100]. Few reports on heterophilic antibodies are available on interference with D-dimer assays [84,101,102,103,104]. Recently, such cases were also reported in COVID-19 patients [105]. Such interferences may be detected and prevented using commercially available heterophilic blocking agents [84,85] or by using a second (and unaffected) assay [65,85]. Research and identification of interferences may be performed based on a practical algorithm [65].

4. Analytical Step

The first method developed for D-dimer testing was a microplate enzyme-linked immunosorbent assay (ELISA) [1]. The principle relies on the use of D-dimer-specific capture antibodies fixed on a plate, followed by the generation of a colorimetric detection when a labeled antibody is added and binds to the immobilized D-dimer antigen (sandwich assay) [1,2]. These techniques were labor-intensive and had poor analytical precision and reproducibility [2,106]. The following generations of assays were mainly based on qualitative agglutination of antibody-coated latex microparticles detected by visual inspection [1,3,4,107]. Latex-enhanced immunoturbidimetric assays today reach a sensitivity close to ELISAs but offer significantly shorter TAT [3].
The automated Vidas enzyme-linked immunofluorescence assays (ELFA) were shown many years ago to display comparable analytical performance to microplate ELISA [3,87]. The Vidas method was thoroughly clinically validated [34,57,108,109,110,111] and is often considered the reference quantitative immunoassay for D-dimer testing [110,112,113]. Finally, chemiluminescent enzyme immunometric assays (CLIA) use magnetic particles coated with D-dimer-specific monoclonal antibodies, while detection is enabled by ulterior fixation anti-D-dimer antibodies labeled with chemiluminescent substances that generate a detectable light signal [3]. The sensitivity of CLIA is similar to or even better than ELISAs, latex-enhanced immunoturbidimetric, and ELFA assays [3,59,112].
Notably, POC D-dimer assays using methods involving monoclonal antibodies may facilitate the triage of patients suspected of VTE [3,114,115,116]. Accessibility to general practitioners shortens TAT [23,114,117], and the use of whole-blood samples enables testing without centrifugation [3,114]. Method design may be qualitative or (semi-)quantitative [3,118]. Only POC tests validated in clinical trials and cleared by the FDA, the European Community, or other similar regulatory agencies should be used for excluding PE. Table 1 recapitulates relevant central laboratory and POC D-dimer assays and their characteristics and intended use as issued by the FDA.
D-dimer assays may also vary importantly between laboratories. In a study on 353 laboratories using the seven most frequently used D-dimer immunoassays, the authors observed that different assays could produce high variations in D-dimer concentrations between assays, causing misclassifications around the 500 ng/mL FEU cut-off [119]. An inter-method coefficient of variation of 42% was, furthermore, observed by the Coagulation Resource Committee of the College of American Pathologists in a survey involving 3800 laboratories [120].
Importantly, D-dimer units vary according to the type of calibration material [9,121,122]. For FEU calibrators preparation, plasmin degradation of purified fibrinogen clotted in the presence of factor XIII is used, whilst calibrators are composed of purified D-dimer for D-dimer units (DDU), respectively [4]. Based on their molecular mass, DDU (195 kD) could be converted to FEU (340 kD) by applying a factor of correction (i.e., approximately 2×) [2,3,4,9,13].
Given the coexistence of multiple sources of heterogeneity in D-dimer assays (e.g., specificity of proprietary monoclonal antibodies, the existence of various fragments obtained from the digestion of cross-linked fibrin from low-molecular-mass forms to high-molecular-mass forms, the lack of certified IQC or calibrators, different reporting units and clinical cut-offs), the development of an international reference for calibration and standardization of D-dimer immunoassays are still unmet needs unlikely to be achieved [9,120,123,124,125,126,127,128,129,130,131,132]. Nonetheless, efforts are underway to provide more comparable results through the use of mathematical models and conversion factors [119,121,122,127,133]. Although such efforts towards harmonization have been made, a single diagnostic cut-off does not seem attainable since false-negative test results would have a harmful clinical impact [9]. According to the Fibrinolysis and Disseminated Intravascular Coagulation Scientific Standardization Subcommittees of the International Society on Hemostasis and Thrombosis, a consensus reference for D-dimer immunoassays might be obtained from a stable freeze-dried reference material containing high concentrations of D-dimer of heterogeneous species [132]. A study among the latest attempted to produce such material but found major instability due to structural rearrangements and amyloid formation of FnDP [134]. Another approach would be to selectively target low- and middle-molecular-mass FnDP species and higher-molecular-mass forms, using adequate monoclonal antibodies [126].
Prospective studies were carried out to validate cut-offs with some reagents (e.g., Vidas, AxSYM, STA Liatest) [1,4,34,108,109,110,129,135] but not with others. When validation studies are missing, a comparison with validated assays must be performed. This will also be required under the new IVDR [22]. It is the role of manufacturers to stay abreast of the most recent literature regarding their assays and suggest revised cut-offs when needed [136].
Recommendations stated that a coefficient of variation <10% should be observed around the diagnostic decision cut-off, and linearity should extend from 50 to 5000 µg/L FEU [132]. The CLSI has proposed to aim at a precision ≤7.5% around the assay-specific diagnostic threshold [17]. Importantly, there should be no cross-reaction with fibrinogen or fibrinogen-degradation products and preferably not with fibrin and fibrinogen fragments released from proteolysis mediated by various enzymes other than plasmin [132].

5. Post-Analytical Step

The International Organization for Standardization 15189:2012 standard defines the post-analytical phase as “processes following the examination including systematic review, formatting and interpretation, authorization for release, reporting and transmission of the results, and storage of samples of the examinations” [45]. The post-analytical phase is still the cause of a large number of diagnostic errors (15–20%), which may jeopardize patient safety [125].
Adequate reporting of D-dimer results is crucial. Several different measure units (i.e., ng/mL, μg/L, mg/L, g/L, μg/mL, mg/mL, and g/dL) are used [3] to report results obtained with diverse assays [2,137,138]. The coexistence of FEU, DDU, and different measure units is a major issue that may cause confusion among clinicians and potentially lead to misinterpretation of test results [3,4,125]. A total of 14 combinations of units and calibrations may hence be used, and surveys reveal an important variability in D-dimer test reporting between laboratories [9,120,125,139]. It is now recommended to favor “µg/L” (or “ng/mL”), which is the unit that best approximates the International System [13,125]. Additionally, the possibility to use age-adjusted cut-offs (or clinical probability-adjusted cut-offs) is another source of complexity for reporting D-dimer test results [125].
Recently, the SARS-CoV-2 pandemic and associated thrombotic events led to a considerable increase in D-dimer measurements as well as in the number of publications on this subject, with many presenting D-dimer reporting issues, ultimately generating confusion and misinterpretations (e.g., absence of units, identification and type of the assay used, age-specific cut-offs) [140]. Overcoming these challenges in reporting D-dimer through standardization is crucial [120,128,132].
Finally, to ensure the clinical usefulness of D-dimer in urgent situations, TAT offered by clinical laboratories must be the shortest possible. An acceptable TAT < 1 h has been proposed, which seems appropriate given the urgent (clinical) nature of this test [13]. A shorter TAT may be obtained by selecting assays that guarantee a wide range of linearity (i.e., up to 5000 µg/L FEU), therefore avoiding additional dilutions, ensuring faster pre-analytical steps (e.g., use of PTS and high-speed centrifugation), and using bedside POC analyzers [3,13,58,62,63,66,117,129]. Current methods (e.g., ELFA, CLIA) require an analytical processing time ranging between 15–40 min. In a European study, 81% of participant laboratories responded that D-dimer testing was available 24/7 [14].

6. Conclusions

D-dimer is a multifaceted biomarker of activation of coagulation and fibrinolysis and is the main laboratory test for patients with suspected VTE (PE and/or DVT). It is currently recommended to use D-dimer assays with high sensitivity to maximize the NPV (i.e., ≥95% sensitivity and ≥97% NPV). In patients with a low or intermediate clinical pretest probability based on structured CDR, a normal D-dimer test result safely rules out acute PE and the need for additional imaging, whilst values above the cut-off should trigger further testing to confirm or refute the diagnosis. The use of age-adjusted cut-offs and, more recently, clinical probability-adjusted cut-offs have been introduced to increase the diagnostic specificity. Pre-analytical requirements for D-dimer differ from other hemostasis tests, as this biomarker appears to be less sensitive to some pre-analytical variables. D-dimer suffers from high inter-laboratory variation, and results obtained from a particular assay cannot be simply extrapolated to another. Reporting of D-dimer is complex, and clinicians should be aware of the possible use of different measuring units. Further studies are needed to promote harmonization of D-dimer measurement and reporting. Systematic reviews on narrower topics on the use of D-dimer in PE are also needed. Future regulations such as the coming IVDR in Europe will have a major impact on the use of D-dimer for PE exclusion and will bring a better definition of the intended use of each D-dimer assay.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Adam, S.S.; Key, N.S.; Greenberg, C.S. D-dimer antigen: Current concepts and future prospects. Blood 2009, 113, 2878–2887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Linkins, L.A.; Takach Lapner, S. Review of D-dimer testing: Good, Bad, and Ugly. Int. J. Lab. Hematol. 2017, 39 (Suppl. 1), 98–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Riley, R.S.; Gilbert, A.R.; Dalton, J.B.; Pai, S.; McPherson, R.A. Widely Used Types and Clinical Applications of D-Dimer Assay. Lab. Med. 2016, 47, 90–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Thachil, J.; Lippi, G.; Favaloro, E.J. D-Dimer Testing: Laboratory Aspects and Current Issues. Methods Mol. Biol. 2017, 1646, 91–104. [Google Scholar] [CrossRef] [PubMed]
  5. Blomback, B. Fibrinogen and fibrin--proteins with complex roles in hemostasis and thrombosis. Thromb. Res. 1996, 83, 1–75. [Google Scholar] [CrossRef]
  6. Sidelmann, J.J.; Gram, J.; Jespersen, J.; Kluft, C. Fibrin clot formation and lysis: Basic mechanisms. Semin. Thromb. Hemost. 2000, 26, 605–618. [Google Scholar] [CrossRef] [PubMed]
  7. Crawley, J.T.; Zanardelli, S.; Chion, C.K.; Lane, D.A. The central role of thrombin in hemostasis. J. Thromb. Haemost. 2007, 5, 95–101. [Google Scholar] [CrossRef]
  8. Gaffney, P.J.; Edgell, T.; Creighton-Kempsford, L.J.; Wheeler, S.; Tarelli, E. Fibrin degradation product (FnDP) assays: Analysis of standardization issues and target antigens in plasma. Br. J. Haematol. 1995, 90, 187–194. [Google Scholar] [CrossRef]
  9. Reber, G.; De Moerloose, P. Standardization of D-dimer testing. In Quality in Laboratory Hemostasis and Thrombosis, 2nd ed.; Kitchen, S., Olson, J.D., Preston, F.E., Eds.; John Wiley & Sons: Oxford, UK, 2013; pp. 136–146. [Google Scholar]
  10. Haase, C.; Joergensen, M.; Ellervik, C.; Joergensen, M.K.; Bathum, L. Age- and sex-dependent reference intervals for D-dimer: Evidence for a marked increase by age. Thromb. Res. 2013, 132, 676–680. [Google Scholar] [CrossRef]
  11. Favresse, J.; Lippi, G.; Roy, P.M.; Chatelain, B.; Jacqmin, H.; Ten Cate, H.; Mullier, F. D-dimer: Preanalytical, analytical, postanalytical variables, and clinical applications. Crit. Rev. Clin. Lab. Sci. 2018, 55, 548–577. [Google Scholar] [CrossRef]
  12. Freund, Y.; Cohen-Aubart, F.; Bloom, B. Acute Pulmonary Embolism: A Review. JAMA 2022, 328, 1336–1345. [Google Scholar] [CrossRef] [PubMed]
  13. Lippi, G.; Cervellin, G.; Casagranda, I.; Morelli, B.; Testa, S.; Tripodi, A. D-dimer testing for suspected venous thromboembolism in the emergency department. Consensus document of AcEMC, CISMEL, SIBioC, and SIMeL. Clin. Chem. Lab. Med. 2014, 52, 621–628. [Google Scholar] [CrossRef] [PubMed]
  14. Spannagl, M.; Haverkate, F.; Reinauer, H.; Meijer, P. The performance of quantitative D-dimer assays in laboratory routine. Blood Coagul. Fibrinolysis 2005, 16, 439–443. [Google Scholar] [CrossRef] [PubMed]
  15. Lippi, G.; Bonfanti, L.; Saccenti, C.; Cervellin, G. Causes of elevated D-dimer in patients admitted to a large urban emergency department. Eur. J. Intern. Med. 2014, 25, 45–48. [Google Scholar] [CrossRef] [PubMed]
  16. Penaloza, A.; Verschuren, F.; Meyer, G.; Quentin-Georget, S.; Soulie, C.; Thys, F.; Roy, P.M. Comparison of the unstructured clinician gestalt, the wells score, and the revised Geneva score to estimate pretest probability for suspected pulmonary embolism. Ann. Emerg. Med. 2013, 62, 117–124.e112. [Google Scholar] [CrossRef]
  17. Clinical and Laboratory Standards Institute. Quantitative D-dimer for the Exclusion of Venous Thromboembolic Disease; Approved Guideline; CLSI Document H59-A; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2011. [Google Scholar]
  18. US Food and Drugs Administration. 510(k) Premarket Notification. Available online: https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpmn/pmn.cfm (accessed on 29 July 2021).
  19. Lippi, G.; Mullier, F.; Favaloro, E.J. D-dimer: Old dogmas, new (COVID-19) tricks. Clin. Chem. Lab. Med. 2022. [Google Scholar] [CrossRef]
  20. Tripodi, A. D-dimer testing in laboratory practice. Clin. Chem. 2011, 57, 1256–1262. [Google Scholar] [CrossRef] [Green Version]
  21. Spitz, I.M.; Le Roith, D.; Hirsch, H.; Carayon, P.; Pekonen, F.; Liel, Y.; Sobel, R.; Chorer, Z.; Weintraub, B. Increased high-molecular-weight thyrotropin with impaired biologic activity in a euthyroid man. N. Engl. J. Med. 1981, 304, 278–282. [Google Scholar] [CrossRef]
  22. Testa, S.; Meijer, P.; Lasne, D.; Mullier, F. Implementation of the new EUR IVD regulation and relation with ISO15189 accreditation: Guidance is urgently required for haemostasis testing. Int. J. Lab. Hematol. 2022, 44 (Suppl. 1), 71–78. [Google Scholar] [CrossRef]
  23. Lucassen, W.A.; Erkens, P.M.; Geersing, G.J.; Buller, H.R.; Moons, K.G.; Stoffers, H.E.; van Weert, H.C. Qualitative point-of-care D-dimer testing compared with quantitative D-dimer testing in excluding pulmonary embolism in primary care. J. Thromb. Haemost. 2015, 13, 1004–1009. [Google Scholar] [CrossRef]
  24. Di Nisio, M.; Squizzato, A.; Rutjes, A.W.; Buller, H.R.; Zwinderman, A.H.; Bossuyt, P.M. Diagnostic accuracy of D-dimer test for exclusion of venous thromboembolism: A systematic review. J. Thromb. Haemost. 2007, 5, 296–304. [Google Scholar] [CrossRef] [PubMed]
  25. Gardiner, C.; Pennaneac’h, C.; Walford, C.; Machin, S.J.; Mackie, I.J. An evaluation of rapid D-dimer assays for the exclusion of deep vein thrombosis. Br. J. Haematol. 2005, 128, 842–848. [Google Scholar] [CrossRef] [PubMed]
  26. Mazzolai, L.; Aboyans, V.; Ageno, W.; Agnelli, G.; Alatri, A.; Bauersachs, R.; Brekelmans, M.P.; Buller, H.R.; Elias, A.; Farge, D.; et al. Diagnosis and management of acute deep vein thrombosis: A joint consensus document from the European society of cardiology working groups of aorta and peripheral circulation and pulmonary circulation and right ventricular function. Eur. Heart J. 2017, 39, 4208–4218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Righini, M.; Van Es, J.; Den Exter, P.L.; Roy, P.M.; Verschuren, F.; Ghuysen, A.; Rutschmann, O.T.; Sanchez, O.; Jaffrelot, M.; Trinh-Duc, A.; et al. Age-adjusted D-dimer cutoff levels to rule out pulmonary embolism: The ADJUST-PE study. JAMA 2014, 311, 1117–1124. [Google Scholar] [CrossRef] [PubMed]
  28. Konstantinides, S.V.; Meyer, G.; Becattini, C.; Bueno, H.; Geersing, G.J.; Harjola, V.P.; Huisman, M.V.; Humbert, M.; Jennings, C.S.; Jimenez, D.; et al. 2019 ESC Guidelines for the diagnosis and management of acute pulmonary embolism developed in collaboration with the European Respiratory Society (ERS). Eur. Heart J. 2020, 41, 543–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Stals, M.A.M.; Klok, F.A.; Huisman, M.V. Diagnostic management of acute pulmonary embolism in special populations. Expert Rev. Respir. Med. 2020, 14, 729–736. [Google Scholar] [CrossRef] [Green Version]
  30. Douma, R.A.; le Gal, G.; Sohne, M.; Righini, M.; Kamphuisen, P.W.; Perrier, A.; Kruip, M.J.; Bounameaux, H.; Buller, H.R.; Roy, P.M. Potential of an age adjusted D-dimer cut-off value to improve the exclusion of pulmonary embolism in older patients: A retrospective analysis of three large cohorts. BMJ 2010, 340, c1475. [Google Scholar] [CrossRef] [Green Version]
  31. Robert-Ebadi, H.; Mostaguir, K.; Hovens, M.M.; Kare, M.; Verschuren, F.; Girard, P.; Huisman, M.V.; Moustafa, F.; Kamphuisen, P.W.; Buller, H.R.; et al. Assessing clinical probability of pulmonary embolism: Prospective validation of the simplified Geneva score. J. Thromb. Haemost. 2017, 15, 1764–1769. [Google Scholar] [CrossRef] [Green Version]
  32. Spencer, F.A.; Goldberg, R.J.; Lessard, D.; Reed, G.; Emery, C.; Gore, J.M.; Pacifico, L.; Weitz, J.I. Factors associated with adverse outcomes in outpatients presenting with pulmonary embolism: The Worcester Venous Thromboembolism Study. Circ. Cardiovasc. Qual. Outcomes 2010, 3, 390–394. [Google Scholar] [CrossRef] [Green Version]
  33. van der Hulle, T.; Cheung, W.Y.; Kooij, S.; Beenen, L.F.M.; van Bemmel, T.; van Es, J.; Faber, L.M.; Hazelaar, G.M.; Heringhaus, C.; Hofstee, H.; et al. Simplified diagnostic management of suspected pulmonary embolism (the YEARS study): A prospective, multicentre, cohort study. Lancet 2017, 390, 289–297. [Google Scholar] [CrossRef]
  34. Elf, J.L.; Strandberg, K.; Svensson, P.J. Performance of two relatively new quantitative D-dimer assays (Innovance D-dimer and AxSYM D-dimer) for the exclusion of deep vein thrombosis. Thromb. Res. 2009, 124, 701–705. [Google Scholar] [CrossRef] [PubMed]
  35. Dronkers, C.E.A.; Van Der Hulle, T.; Le Gal, G.; Kyrle, P.A.; Huisman, M.V.; Cannegieter, S.C.; Klok, F.A. Towards a tailored diagnostic standard for future diagnostic studies in pulmonary embolism: Communication from the SSC of the ISTH. J. Thromb. Haemost. 2017, 15, 1040–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Le Gal, G.; Righini, M.; Wells, P.S. D-dimer for pulmonary embolism. JAMA 2015, 313, 1668–1669. [Google Scholar] [CrossRef] [PubMed]
  37. Linkins, L.A.; Bates, S.M.; Lang, E.; Kahn, S.R.; Douketis, J.D.; Julian, J.; Parpia, S.; Gross, P.; Weitz, J.I.; Spencer, F.A.; et al. Selective D-dimer testing for diagnosis of a first suspected episode of deep venous thrombosis: A randomized trial. Ann. Intern. Med. 2013, 158, 93–100. [Google Scholar] [CrossRef] [Green Version]
  38. Kearon, C.; de Wit, K.; Parpia, S.; Schulman, S.; Afilalo, M.; Hirsch, A.; Spencer, F.A.; Sharma, S.; D’Aragon, F.; Deshaies, J.F.; et al. Diagnosis of Pulmonary Embolism with d-Dimer Adjusted to Clinical Probability. N. Engl. J. Med. 2019, 381, 2125–2134. [Google Scholar] [CrossRef] [PubMed]
  39. Solberg, R.; Glass, G. Adjusting D-dimer cutoffs: Brief literature summary and issues in clinical use. Am. J. Emerg. Med. 2018, 36, 2105–2107. [Google Scholar] [CrossRef]
  40. Farm, M.; Siddiqui, A.; Onelöv, L.; Chaireti, R.; Holmström, M.; Antovic, J.P. Age-Adjusted D-Dimer Cutoffs for DVT and Pulmonary Embolism: A Comparison of Five Assays. Blood 2016, 128, 1419. [Google Scholar] [CrossRef]
  41. Penaloza, A.; Roy, P.M.; Kline, J.; Verschuren, F.; Le Gal, G.; Quentin-Georget, S.; Delvau, N.; Thys, F. Performance of age-adjusted D-dimer cut-off to rule out pulmonary embolism. J. Thromb. Haemost. 2012, 10, 1291–1296. [Google Scholar] [CrossRef]
  42. Prochaska, J.H.; Frank, B.; Nagler, M.; Lamparter, H.; Weisser, G.; Schulz, A.; Eggebrecht, L.; Gobel, S.; Arnold, N.; Panova-Noeva, M.; et al. Age-related diagnostic value of D-dimer testing and the role of inflammation in patients with suspected deep vein thrombosis. Sci. Rep. 2017, 7, 4591. [Google Scholar] [CrossRef] [Green Version]
  43. Raja, A.S.; Greenberg, J.O.; Qaseem, A.; Denberg, T.D.; Fitterman, N.; Schuur, J.D.; Clinical Guidelines Committee of the American College of Physicians. Evaluation of Patients With Suspected Acute Pulmonary Embolism: Best Practice Advice From the Clinical Guidelines Committee of the American College of Physicians. Ann. Intern. Med. 2015, 163, 701–711. [Google Scholar] [CrossRef]
  44. Lippi, G.; Favaloro, E. Causes of Errors in Medical Laboratories. In Quality in Laboratory Hemostasis and Thrombosis, 2nd ed.; Kitchen, S., Olson, J.D., Preston, F.E., Eds.; John Wiley & Sons: Oxford, UK, 2013; pp. 22–31. [Google Scholar]
  45. ISO 15189:2012; Medical Laboratories—Particular Requirements for Quality and Competence. International Organization for Standardization: Geneva, Switzerland, 2012.
  46. Kitchen, S.; Adcock, D.M.; Dauer, R.; Kristoffersen, A.H.; Lippi, G.; Mackie, I.; Marlar, R.A.; Nair, S. International Council for Standardization in Haematology (ICSH) recommendations for processing of blood samples for coagulation testing. Int. J. Lab. Hematol. 2021, 43, 1272–1283. [Google Scholar] [CrossRef]
  47. Gosselin, R.C.; Marlar, R.A. Preanalytical Variables in Coagulation Testing: Setting the Stage for Accurate Results. Semin. Thromb. Hemost. 2019, 45, 433–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Magnette, A.; Chatelain, M.; Chatelain, B.; Ten Cate, H.; Mullier, F. Pre-analytical issues in the haemostasis laboratory: Guidance for the clinical laboratories. Thromb. J. 2016, 14, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Adcock, D. Sample integrity and preanalytical variables. In Quality in Laboratory Hemostasis and Thrombosis., 2nd ed.; Kitchen, S., Olson, J.D., Preston, F.E., Eds.; John Wiley & Sons: Oxford, UK, 2013; pp. 45–56. [Google Scholar]
  50. Lippi, G.; Salvagno, G.L.; Guidi, G.C. No influence of a butterfly device on routine coagulation assays and D-dimer measurement. J. Thromb. Haemost. 2005, 3, 389–391. [Google Scholar] [CrossRef] [PubMed]
  51. Lippi, G.; Salvagno, G.L.; Montagnana, M.; Brocco, G.; Cesare Guidi, G. Influence of the needle bore size used for collecting venous blood samples on routine clinical chemistry testing. Clin. Chem. Lab. Med. 2006, 44, 1009–1014. [Google Scholar] [CrossRef]
  52. Clinical and Laboratory Standards Institute. Collection, Transport, and Processing of Blood Specimens for Testing Plasma-Based Coagulation Assays and Molecular Hemostasis Assays; Approved Guideline, 5th ed.; CLSI Document H21-A5; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2008. [Google Scholar]
  53. Smock, K.J.; Crist, R.A.; Hansen, S.J.; Rodgers, G.M.; Lehman, C.M. Discard tubes are not necessary when drawing samples for specialized coagulation testing. Blood Coagul. Fibrinolysis 2010, 21, 279–282. [Google Scholar] [CrossRef] [PubMed]
  54. Leroy-Matheron, C.; Gouault-Heilmann, M. Influence of conditions of blood sampling on coagulation activation markers (prothrombin fragment 1 + 2, thrombin-antithrombin complexes and D-dimers) measurements. Thromb. Res. 1994, 74, 399–407. [Google Scholar] [CrossRef]
  55. Gosselin, R.C.; Janatpour, K.; Larkin, E.C.; Lee, Y.P.; Owings, J.T. Comparison of samples obtained from 3.2% sodium citrate glass and two 3.2% sodium citrate plastic blood collection tubes used in coagulation testing. Am. J. Clin. Pathol. 2004, 122, 843–848. [Google Scholar] [CrossRef]
  56. Yavas, S.; Ayaz, S.; Kose, S.K.; Ulus, F.; Ulus, A.T. Influence of blood collection systems on coagulation tests. Turk. J. Haematol. 2012, 29, 367–375. [Google Scholar] [CrossRef] [PubMed]
  57. Oude Elferink, R.F.; Loot, A.E.; Van De Klashorst, C.G.; Hulsebos-Huygen, M.; Piersma-Wichers, M.; Oudega, R. Clinical evaluation of eight different D-dimer tests for the exclusion of deep venous thrombosis in primary care patients. Scand. J. Clin. Lab. Investig. 2015, 75, 230–238. [Google Scholar] [CrossRef]
  58. Schutgens, R.E.; Haas, F.J.; Ruven, H.J.; Spannagl, M.; Horn, K.; Biesma, D.H. No influence of heparin plasma and other (pre)analytic variables on D-dimer determinations. Clin. Chem. 2002, 48, 1611–1613. [Google Scholar] [CrossRef]
  59. Lippi, G.; Salvagno, G.L.; Rossi, L.; Montagnana, M.; Franchini, M.; Guidi, G.C. Analytical performances of the D-dimer assay for the Immulite 2000 automated immunoassay analyser. Int. J. Lab. Hematol. 2007, 29, 415–420. [Google Scholar] [CrossRef] [PubMed]
  60. Vukovich, T.C.; Hamwi, A.; Bieglmayer, C. D-dimer testing within the routine clinical chemistry profile. Clin. Chem. 1998, 44, 1557–1558. [Google Scholar] [CrossRef] [Green Version]
  61. Favaloro, E.J.; Funk, D.M.; Lippi, G. Pre-analytical Variables in Coagulation Testing Associated With Diagnostic Errors in Hemostasis. Lab. Med. 2012, 43, 1–10. [Google Scholar] [CrossRef] [Green Version]
  62. Le Quellec, S.; Paris, M.; Nougier, C.; Sobas, F.; Rugeri, L.; Girard, S.; Bordet, J.C.; Negrier, C.; Dargaud, Y. Pre-analytical effects of pneumatic tube system transport on routine haematology and coagulation tests, global coagulation assays and platelet function assays. Thromb. Res. 2017, 153, 7–13. [Google Scholar] [CrossRef] [PubMed]
  63. Wallin, O.; Soderberg, J.; Grankvist, K.; Jonsson, P.A.; Hultdin, J. Preanalytical effects of pneumatic tube transport on routine haematology, coagulation parameters, platelet function and global coagulation. Clin. Chem. Lab. Med. 2008, 46, 1443–1449. [Google Scholar] [CrossRef] [PubMed]
  64. Koessler, J.; Kobsar, A.L.; Brunner, K.; Stolz, H.; Dossler, B.; Walter, U.; Steigerwald, U. The preanalytical influence of two different mechanical transport systems on laboratory analysis. Clin. Chem. Lab. Med. 2011, 49, 1379–1382. [Google Scholar] [CrossRef]
  65. Wauthier, L.; Plebani, M.; Favresse, J. Interferences in immunoassays: Review and practical algorithm. Clin. Chem. Lab. Med. 2022, 60, 808–820. [Google Scholar] [CrossRef]
  66. Bernard, M.; Ben Ayed, S.; Tazi, A.; Vamous, S.; Godet, G.; Arock, M.; Foglietti, M.J. Vidas Emergency panel: Decrease of the turnaround time by fast centrifugation. Clin. Chem. Lab. Med. 2002, 40 (Suppl. S9), S350. [Google Scholar]
  67. Wolfensberger, N.; Georgiou, G.; Giabbani, E.; Reusser, M.; Njue, L.M.; Fiedler, M.; Leichtle, A.B.; Nagler, M. Rapid Centrifugation in the Routine Hemostasis Laboratory. Thromb. Haemost. 2019, 119, 2025–2033. [Google Scholar] [CrossRef] [Green Version]
  68. Yazar, H.; Ozdemir, F.; Kose, E. Effect of Centrifuge Temperature on Routine Coagulation Tests. Acta Haematol. 2018, 139, 158–163. [Google Scholar] [CrossRef] [PubMed]
  69. Bohm-Weigert, M.; Wissel, T.; Muth, H.; Kemkes-Matthes, B.; Peetz, D. Long- and short-term in vitro D-dimer stability measured with INNOVANCE D-Dimer. Thromb. Haemost. 2010, 103, 461–465. [Google Scholar] [CrossRef]
  70. Gosselin, R.C.; Dwyre, D.W. Determining the effect of freezing on coagulation testing: Comparison of results between fresh and once frozen-thawed plasma. Blood Coagul. Fibrinolysis 2015, 26, 69–74. [Google Scholar] [CrossRef] [PubMed]
  71. Zurcher, M.; Sulzer, I.; Barizzi, G.; Lammle, B.; Alberio, L. Stability of coagulation assays performed in plasma from citrated whole blood transported at ambient temperature. Thromb. Haemost. 2008, 99, 416–426. [Google Scholar] [CrossRef]
  72. Linskens, E.A.; Devreese, K.M.J. Pre-analytical stability of coagulation parameters in plasma stored at room temperature. Int. J. Lab. Hematol. 2018, 40, 292–303. [Google Scholar] [CrossRef] [PubMed]
  73. Betsou, F.; Roussel, B.; Guillaume, N.; Lefrere, J.J. Long-term stability of coagulation variables: Protein S as a biomarker for preanalytical storage-related variations in human plasma. Thromb. Haemost. 2009, 101, 1172–1175. [Google Scholar] [CrossRef] [PubMed]
  74. Foshat, M.; Bates, S.; Russo, W.; Huerta, A.; Albright, K.; Giddings, K.; Indrikovs, A.; Qian, Y.W. Effect of freezing plasma at -20 degrees C for 2 weeks on prothrombin time, activated partial thromboplastin time, dilute Russell viper venom time, activated protein C resistance, and D-dimer levels. Clin. Appl. Thromb. Hemost. 2015, 21, 41–47. [Google Scholar] [CrossRef] [PubMed]
  75. Woodhams, B.; Girardot, O.; Blanco, M.J.; Colesse, G.; Gourmelin, Y. Stability of coagulation proteins in frozen plasma. Blood Coagul Fibrinolysis 2001, 12, 229–236. [Google Scholar] [CrossRef]
  76. Lippi, G.; Blanckaert, N.; Bonini, P.; Green, S.; Kitchen, S.; Palicka, V.; Vassault, A.J.; Plebani, M. Haemolysis: An overview of the leading cause of unsuitable specimens in clinical laboratories. Clin. Chem. Lab. Med. 2008, 46, 764–772. [Google Scholar] [CrossRef]
  77. Lippi, G.; Plebani, M.; Favaloro, E.J. Interference in coagulation testing: Focus on spurious hemolysis, icterus, and lipemia. Semin. Thromb. Hemost. 2013, 39, 258–266. [Google Scholar] [CrossRef]
  78. Lippi, G.; Avanzini, P.; Zobbi, V.; Ippolito, L. Influence of mechanical hemolysis of blood on two D-dimer immunoassays. Blood Coagul. Fibrinolysis 2012, 23, 461–463. [Google Scholar] [CrossRef] [PubMed]
  79. Salvagno, G.L.; Lippi, G.; Bassi, A.; Poli, G.; Guidi, G.C. Prevalence and type of pre-analytical problems for inpatients samples in coagulation laboratory. J. Eval. Clin. Pract. 2008, 14, 351–353. [Google Scholar] [CrossRef] [PubMed]
  80. Lippi, G.; von Meyer, A.; Cadamuro, J.; Simundic, A.M. Blood sample quality. Diagnosis 2019, 6, 25–31. [Google Scholar] [CrossRef]
  81. Carraro, P.; Servidio, G.; Plebani, M. Hemolyzed specimens: A reason for rejection or a clinical challenge? Clin. Chem. 2000, 46, 306–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Lippi, G.; Montagnana, M.; Salvagno, G.L.; Guidi, G.C. Interference of blood cell lysis on routine coagulation testing. Arch. Pathol. Lab. Med. 2006, 130, 181–184. [Google Scholar] [CrossRef]
  83. Hedeland, Y.; Gustafsson, C.M.; Touza, Z.; Ridefelt, P. Hemolysis interference in 10 coagulation assays on an instrument with viscosity-based, chromogenic, and turbidimetric clot detection. Int. J. Lab. Hematol. 2020, 42, 341–349. [Google Scholar] [CrossRef] [Green Version]
  84. Lippi, G.; Ippolito, L.; Tondelli, M.T.; Favaloro, E.J. Interference from heterophilic antibodies in D-dimer assessment. A case report. Blood Coagul Fibrinolysis 2014, 25, 277–279. [Google Scholar] [CrossRef]
  85. Mugler, K.; Lefkowitz, J.B. False-positive D-dimer result in a patient with Castleman disease. Arch. Pathol. Lab. Med. 2004, 128, 328–331. [Google Scholar] [CrossRef]
  86. Ma, H.; Chen, C.H.; Li, Y.M.; Wang, J.J.; Hu, Z.D. Falsely elevated D-dimer partially caused by heterophilic antibodies: A case report. Clin. Chim. Acta. 2022, 535, 140–142. [Google Scholar] [CrossRef]
  87. Pittet, J.L.; de Moerloose, P.; Reber, G.; Durand, C.; Villard, C.; Piga, N.; Rolland, D.; Comby, S.; Dupuy, G. VIDAS D-dimer: Fast quantitative ELISA for measuring D-dimer in plasma. Clin. Chem. 1996, 42, 410–415. [Google Scholar] [CrossRef] [Green Version]
  88. La’ulu, S.L.; Dominguez, C.M.; Roberts, W.L. Performance characteristics of the AxSYM D-dimer assay. Clin. Chim. Acta 2008, 390, 148–151. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, L.; Chen, Y. Performance Evaluation of the Sysmex CS-5100 Automated Coagulation Analyzer. Clin. Lab. 2015, 61, 653–660. [Google Scholar] [CrossRef] [PubMed]
  90. Talon, L.; Fourneyron, V.; Trapani, A.; Pereira, B.; Sinegre, T.; Lebreton, A. Analytical performance of a new immunoturbidimetric D-dimer assay and comparison with available assays. Res. Pract. Thromb. Haemost. 2022, 6, e12660. [Google Scholar] [CrossRef]
  91. Mastella, A.K.; de Carvalho, J.A.M.; Stein, C.S.; Bochi, G.V.; Moresco, R.N. Interference of icterus on plasma D-dimer levels measured using immunoturbidimetric assays. Blood Coagul. Fibrinolysis 2021, 32, 162–163. [Google Scholar] [CrossRef] [PubMed]
  92. Montaruli, B.; Guiotto, C.; Cosseddu, D. Influence of hemolysis, icterus and lipemia on coagulation tests as performed on Cobas t511 new analyzer. Blood Coagul. Fibrinolysis 2020, 31, 48–54. [Google Scholar] [CrossRef] [PubMed]
  93. Negrini, D.; Bernardi, D.; Antonelli, G.; Plebani, M. Interference of lipemia in samples for routine coagulation testing using a Sysmex CS-5100 coagulometer. Int. J. Lab. Hematol. 2019, 41, 772–777. [Google Scholar] [CrossRef]
  94. Park, S.J.; Chi, H.S.; Chun, S.H.; Jang, S.; Park, C.J. Evaluation of performance including influence by interfering substances of the Innovance D-dimer assay on the Sysmex coagulation analyzer. Ann. Clin. Lab. Sci. 2011, 41, 20–24. [Google Scholar]
  95. Flieder, T.; Gripp, T.; Knabbe, C.; Birschmann, I. The Sysmex CS-5100 coagulation analyzer offers comparable analytical performance and excellent throughput capabilities. Pract. Lab. Med. 2016, 6, 38–47. [Google Scholar] [CrossRef] [Green Version]
  96. Jensen, A.K.; Christensen, G.L.; Dalgard, A.; Jorgensen, N.R. Estimation of lipemia interference with automated HIL-test on d-dimer ACL TOP 50 series analysis—Reveals a higher cut-off than manufacturer’s recommendations. Scand. J. Clin. Lab. Investig. 2020, 80, 168–171. [Google Scholar] [CrossRef]
  97. Gardiner, C.; Lane, P.; Tailor, H.; Mackie, I.J. A practical method for reducing the interference due to lipaemia in coagulation tests. Int. J. Lab. Hematol. 2020, 42, 140–144. [Google Scholar] [CrossRef] [Green Version]
  98. Huang, H.; Li, H.; Li, D. Effect of serum monoclonal protein concentration on haemostasis in patients with multiple myeloma. Blood Coagul. Fibrinolysis 2015, 26, 555–559. [Google Scholar] [CrossRef] [PubMed]
  99. Roller, R.E.; Lahousen, T.; Lipp, R.W.; Korninger, C.; Schnedl, W.J. Elevated D-dimer results in a healthy patient. Blood Coagul. Fibrinolysis 2001, 12, 501–502. [Google Scholar] [CrossRef] [PubMed]
  100. Wu, X.Y.; Yin, Y.F.; Teng, J.L.; Zhang, L.W.; Yang, C.D. IgMk paraprotein from gammopathy patient can bind to cardiolipin and interfere with coagulation assay: A case report. BMC Immunol. 2017, 18, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Gardiner, C.; Pennaneac’h, C.; Mackie, I.J.; Sheldrake, A.; Harrison, J.; Machin, S.J. Falsely elevated D-dimer results in a healthy patient on account of heterophiletul antibodies. Br. J. Haematol. 2003, 122, 871–873. [Google Scholar] [CrossRef]
  102. Rouviere, J.A.; Devignes, J.; de Maistre, E.; Kennel, A.; Chabot, F.; Lecompte, T. Discrepancy between two methods of D-dimers measurement: One case of human anti-mouse antibody interference. Ann. Biol. Clin. 2008, 66, 441–446. [Google Scholar] [CrossRef]
  103. Wu, Y.; Xiao, Y.X.; Huang, T.Y.; Zhang, X.Y.; Zhou, H.B.; Zhang, X.X.; Wu, Y.E. What makes D-dimer assays suspicious-heterophilic antibodies? J. Clin. Lab. Anal. 2019, 33, e22687. [Google Scholar] [CrossRef] [Green Version]
  104. Sun, H.X.; Ge, H.; Xu, Z.Q.; Sheng, H.M. Clinical laboratory investigation of a patient with an extremely high D-dimer level: A case report. World J. Clin. Cases 2020, 8, 3560–3566. [Google Scholar] [CrossRef]
  105. Ozbalci, D.; Doguc, D.K.; Yilmaz, G.; Ozturk, O.; Sirin, F.B.; Akcam, F.Z. Interference of D-dimer levels from heterophilic antibody in COVID-19: A serious concern in treatment and follow-up of patients. Int. J. Lab. Hematol. 2022, 44, e13–e16. [Google Scholar] [CrossRef]
  106. Lippi, G.; Lima-Oliveira, G.; Brocco, G.; Bassi, A.; Salvagno, G.L. Estimating the intra- and inter-individual imprecision of manual pipetting. Clin. Chem. Lab. Med. 2017, 55, 962–966. [Google Scholar] [CrossRef]
  107. Greenberg, C.S.; Devine, D.V.; McCrae, K.M. Measurement of plasma fibrin D-dimer levels with the use of a monoclonal antibody coupled to latex beads. Am. J. Clin. Pathol. 1987, 87, 94–100. [Google Scholar] [CrossRef] [Green Version]
  108. Mountain, D.; Jacobs, I.; Haig, A. The VIDAS D-dimer test for venous thromboembolism: A prospective surveillance study shows maintenance of sensitivity and specificity when used in normal clinical practice. Am. J. Emerg. Med. 2007, 25, 464–471. [Google Scholar] [CrossRef] [PubMed]
  109. Perrier, A.; Desmarais, S.; Miron, M.J.; de Moerloose, P.; Lepage, R.; Slosman, D.; Didier, D.; Unger, P.F.; Patenaude, J.V.; Bounameaux, H. Non-invasive diagnosis of venous thromboembolism in outpatients. Lancet 1999, 353, 190–195. [Google Scholar] [CrossRef]
  110. Salvagno, G.L.; Lippi, G.; Manzato, F.; Giavarina, D.; Montagnana, M.; Poli, G.; Guidi, G.C. Analytical comparison of AxSYM, HemosIL DD HS and Innovance D-dimer immunoassays with the Vidas D-dimer. Int. J. Lab. Hematol. 2009, 31, 475–477. [Google Scholar] [CrossRef] [PubMed]
  111. Sidelmann, J.J.; Gram, J.; Larsen, A.; Overgaard, K.; Jespersen, J. Analytical and clinical validation of a new point-of-care testing system for determination of D-Dimer in human blood. Thromb. Res. 2010, 126, 524–530. [Google Scholar] [CrossRef]
  112. Lippi, G.; Ippolito, L.; Russello, T.; Ponzo, V.; Salvagno, G.L.; Guidi, G.C. Analytical performance of the new ACL AcuStar HemosIL D-Dimer. Blood Coagul. Fibrinolysis 2012, 23, 164–167. [Google Scholar] [CrossRef]
  113. van der Graaf, F.; van den Borne, H.; van der Kolk, M.; de Wild, P.J.; Janssen, G.W.; van Uum, S.H. Exclusion of deep venous thrombosis with D-dimer testing--comparison of 13 D-dimer methods in 99 outpatients suspected of deep venous thrombosis using venography as reference standard. Thromb. Haemost. 2000, 83, 191–198. [Google Scholar] [CrossRef] [Green Version]
  114. Helmersson-Karlqvist, J.; Karlsson, B.; Fredriksson, A.; Larsson, A. Evaluation of the Alere D-dimer test for point of care testing. J. Thromb. Thrombolysis 2014, 38, 250–252. [Google Scholar] [CrossRef]
  115. Marquardt, U.; Apau, D. Point-of-care D-dimer testing in emergency departments. Emerg. Nurse 2015, 23, 29–35. [Google Scholar] [CrossRef]
  116. Rooney, K.D.; Schilling, U.M. Point-of-care testing in the overcrowded emergency department—Can it make a difference? Crit. Care 2014, 18, 692. [Google Scholar] [CrossRef] [Green Version]
  117. Sen, B.; Kesteven, P.; Avery, P. Comparison of D-dimer point of care test (POCT) against current laboratory test in patients with suspected venous thromboembolism (VTE) presenting to the emergency department (ED). J. Clin. Pathol. 2014, 67, 437–440. [Google Scholar] [CrossRef]
  118. Geersing, G.J.; Toll, D.B.; Janssen, K.J.; Oudega, R.; Blikman, M.J.; Wijland, R.; de Vooght, K.M.; Hoes, A.W.; Moons, K.G. Diagnostic accuracy and user-friendliness of 5 point-of-care D-dimer tests for the exclusion of deep vein thrombosis. Clin. Chem. 2010, 56, 1758–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Meijer, P.; Haverkate, F.; Kluft, C.; de Moerloose, P.; Verbruggen, B.; Spannagl, M. A model for the harmonisation of test results of different quantitative D-dimer methods. Thromb. Haemost. 2006, 95, 567–572. [Google Scholar] [CrossRef] [PubMed]
  120. Olson, J.D.; Cunningham, M.T.; Higgins, R.A.; Eby, C.S.; Brandt, J.T. D-dimer: Simple test, tough problems. Arch. Pathol. Lab. Med. 2013, 137, 1030–1038. [Google Scholar] [CrossRef] [PubMed]
  121. Dempfle, C.E.; Zips, S.; Ergul, H.; Heene, D.L.; group, F.S. The fibrin assay comparison trial (FACT): Correlation of soluble fibrin assays with D-dimer. Thromb. Haemost. 2001, 86, 1204–1209. [Google Scholar] [CrossRef]
  122. Jennings, I.; Woods, T.A.; Kitchen, D.P.; Kitchen, S.; Walker, I.D. Laboratory D-dimer measurement: Improved agreement between methods through calibration. Thromb. Haemost. 2007, 98, 1127–1135. [Google Scholar] [CrossRef] [PubMed]
  123. Dempfle, C.E. Validation, calibration, and specificity of quantitative D-dimer assays. Semin. Vasc. Med. 2005, 5, 315–320. [Google Scholar] [CrossRef]
  124. Dempfle, C.E. D-dimer assays: The current status and new assay technologies. Thromb. Res. 2006, 118, 569–571. [Google Scholar] [CrossRef]
  125. Lippi, G.; Tripodi, A.; Simundic, A.M.; Favaloro, E.J. International survey on D-dimer test reporting: A call for standardization. Semin. Thromb. Hemost. 2015, 41, 287–293. [Google Scholar] [CrossRef] [Green Version]
  126. Madoiwa, S.; Kitajima, I.; Ohmori, T.; Sakata, Y.; Mimuro, J. Distinct reactivity of the commercially available monoclonal antibodies of D-dimer and plasma FDP testing to the molecular variants of fibrin degradation products. Thromb. Res. 2013, 132, 457–464. [Google Scholar] [CrossRef]
  127. Nieuwenhuizen, W. A reference material for harmonisation of D-dimer assays. Fibrinogen Subcommittee of the Scientific and Standardization Committee of the International Society of Thrombosis and Haemostasis. Thromb. Haemost. 1997, 77, 1031–1033. [Google Scholar]
  128. Goodwin, A.J.; Higgins, R.A.; Moser, K.A.; Smock, K.J.; Chandler, W.L.; Kottke-Marchant, K.; Hartman, S.K.; Volod, O.; Brown, A.F.; Johari, V.P.; et al. Issues Surrounding Age-Adjusted d-Dimer Cutoffs That Practicing Physicians Need to Know When Evaluating Patients With Suspected Pulmonary Embolism. Ann. Intern. Med. 2017, 166, 361–363. [Google Scholar] [CrossRef]
  129. Giannitsis, E.; Mair, J.; Christersson, C.; Siegbahn, A.; Huber, K.; Jaffe, A.S.; Peacock, W.F.; Plebani, M.; Thygesen, K.; Mockel, M.; et al. How to use D-dimer in acute cardiovascular care. Eur. Heart J. Acute Cardiovasc. Care 2017, 6, 69–80. [Google Scholar] [CrossRef]
  130. Tripodi, A.; Chantarangkul, V. Performance of quantitative D-dimer methods: Results of the Italian external quality assessment scheme. J. Thromb. Haemost. 2007, 5, 184–185. [Google Scholar] [CrossRef]
  131. Johnson, E.D.; Schell, J.C.; Rodgers, G.M. The D-dimer assay. Am. J. Hematol. 2019, 94, 833–839. [Google Scholar] [CrossRef]
  132. Longstaff, C.; Adcock, D.; Olson, J.D.; Jennings, I.; Kitchen, S.; Mutch, N.; Meijer, P.; Favaloro, E.J.; Lippi, G.; Thachil, J. Harmonisation of D-dimer—A call for action. Thromb. Res. 2016, 137, 219–220. [Google Scholar] [CrossRef]
  133. Garcia de Guadiana-Romualdo, L.; Morell-Garcia, D.; Favaloro, E.J.; Vilchez, J.A.; Bauca, J.M.; Alcaide Martin, M.J.; Gutierrez Garcia, I.; de la Hera Cagigal, P.; Egea-Caparros, J.M.; Perez Sanmartin, S.; et al. Harmonized D-dimer levels upon admission for prognosis of COVID-19 severity: Results from a Spanish multicenter registry (BIOCOVID-Spain study). J. Thromb. Thrombolysis 2022, 53, 103–112. [Google Scholar] [CrossRef]
  134. Bevan, S.; Longstaff, C. Is it possible to make a common reference standard for D-dimer measurements? Communication from the ISTH SSC Subcommittee on Fibrinolysis. J. Thromb. Haemost. 2022, 20, 498–507. [Google Scholar] [CrossRef]
  135. Pernod, G.; Wu, H.; de Maistre, E.; Lazarchick, J.; Kassis, J.; Aguilar, C.; Vera, P.M.; Palareti, G.; D’Angelo, A.; Di, E.T.S.G. Validation of STA-Liatest D-Di assay for exclusion of pulmonary embolism according to the latest Clinical and Laboratory Standard Institute/Food and Drug Administration guideline. Results of a multicenter management study. Blood Coagul. Fibrinolysis 2017, 28, 254–260. [Google Scholar] [CrossRef] [Green Version]
  136. Boeer, K.; Siegmund, R.; Schmidt, D.; Deufel, T.; Kiehntopf, M. Comparison of six D-dimer assays for the detection of clinically suspected deep venous thrombosis of the lower extremities. Blood Coagul. Fibrinolysis 2009, 20, 141–145. [Google Scholar] [CrossRef]
  137. Coen Herak, D.; Milos, M.; Zadro, R. Evaluation of the Innovance D-DIMER analytical performance. Clin. Chem. Lab. Med. 2009, 47, 945–951. [Google Scholar] [CrossRef]
  138. de Moerloose, P.; Palareti, G.; Aguilar, C.; Legnani, C.; Reber, G.; Peetz, D. A multicenter evaluation of a new quantitative highly sensitive D-dimer assay for exclusion of venous thromboembolism. Thromb. Haemost. 2008, 100, 505–512. [Google Scholar]
  139. Lucassen, W.; Geersing, G.J.; Erkens, P.M.; Reitsma, J.B.; Moons, K.G.; Buller, H.; van Weert, H.C. Clinical decision rules for excluding pulmonary embolism: A meta-analysis. Ann. Intern. Med. 2011, 155, 448–460. [Google Scholar] [CrossRef]
  140. Thachil, J.; Longstaff, C.; Favaloro, E.J.; Lippi, G.; Urano, T.; Kim, P.Y.; Thrombosis, S.S.C. Subcommittee on Fibrinolysis of the International Society on Thrombosis Haemostasis. The need for accurate D-dimer reporting in COVID-19: Communication from the ISTH SSC on fibrinolysis. J. Thromb. Haemost. 2020, 18, 2408–2411. [Google Scholar] [CrossRef]
Table 1. Characteristics of common D-dimer assays. Relevant central laboratory and point-of-care assays are shown along with the manufacturer, method design, method principle, unit type, cut-off mentioned by the manufacturer, and the intended use issued by the FDA regarding PE. DDU, D-dimer units; FDA, U.S. Food and Drugs Administration; FEU, fibrinogen-equivalent units; NA, not applicable; PE, pulmonary embolism; POC, point-of-care.
Table 1. Characteristics of common D-dimer assays. Relevant central laboratory and point-of-care assays are shown along with the manufacturer, method design, method principle, unit type, cut-off mentioned by the manufacturer, and the intended use issued by the FDA regarding PE. DDU, D-dimer units; FDA, U.S. Food and Drugs Administration; FEU, fibrinogen-equivalent units; NA, not applicable; PE, pulmonary embolism; POC, point-of-care.
Assay TypeAssay NameManufacturerDesignPrincipleUnit TypeManufacturer’s Cut-OffFDA Intended Use (PE)
Central labAdvanced D-DimerSiemens Healthcare Diagnostics (previously Dade Berhing)QuantitativeLatex-enhanced turbidimetric immunoassayFEUBCS System: 1.6 mg/L
Sysmex CA-1500: 1.0 mg/L
Aid in diagnosis
Central labAxSYM D-dimerAbbott laboratoriesQuantitativeEnzyme-linked fluorescent assayFEU500 µg/LNA
Central labDiazyme D-DimerDiazyme LaboratoriesQuantitativeLatex-enhanced turbidimetric immunoassayFEU0.5 µg/mLAid in diagnosis
Central labHemosIL
AcuStar D-Dimer
Werfen (previously Instrumentation Laboratory)QuantitativeChemiluminescent immunoassayFEU500 µg/LAid in diagnosis
Central labHemosIL D-Dimer (±HS)Werfen (previously Instrumentation Laboratory)QuantitativeLatex-enhanced turbidimetric immunoassayDDU230 µg/LExclusion
Central labHemosIL D-Dimer HS 500Werfen (previously Instrumentation Laboratory)QuantitativeLatex-enhanced turbidimetric immunoassayFEU500 µg/LExclusion
Central labInnovance D-DimerSiemens Healthcare DiagnosticsQuantitativeLatex-enhanced turbidimetric immunoassayFEU0.5 mg/LExclusion
Central labSTA Liatest D-DiDiagnostica StagoQuantitativeLatex-enhanced turbidimetric immunoassayFEU0.5 µg/mLExclusion
Central labTina-Quant D-DimerRoche DiagnosticsQuantitativeLatex-enhanced turbidimetric immunoassayFEU0.5 µg/mLExclusion
Central lab (or POC)VIDAS D-DimerbioMérieuxQuantitativeEnzyme-linked fluorescent assayFEU500 µg/LExclusion
POCAQT90 FLEX D-dimerRadiometer Medical ApSQuantitativeTime-resolved fluorometryNA500 µg/LNA
POCClearview SimplifyAgen BiomedicalQualitativeSolid-phase immunochromatographyNeg/pos80 µg/LNA
POCPathfast D-DimerMitsubishi Kagaku
Iatron
QuantitativeChemiluminescent immunoassayFEU0.686 µg/mLNA
POCRoche Cardiac D-dimerRoche DiagnosticsQuantitativeSolid-phase immunochromatographyFEU0.5 µg/mLNA
POCStratus CS Acute Care D-dimerSiemens Healthcare DiagnosticsQuantitativeFluorescent immunoassayFEU450 µg/LExclusion
POCTriage D-dimerBiosite DiagnosticsQuantitativeFluorescent immunoassayDDU350 µg/LAid in diagnosis
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wauthier, L.; Favresse, J.; Hardy, M.; Douxfils, J.; Le Gal, G.; Roy, P.-M.; van Es, N.; Ay, C.; ten Cate, H.; Vander Borght, T.; et al. D-dimer Testing in Pulmonary Embolism with a Focus on Potential Pitfalls: A Narrative Review. Diagnostics 2022, 12, 2770. https://doi.org/10.3390/diagnostics12112770

AMA Style

Wauthier L, Favresse J, Hardy M, Douxfils J, Le Gal G, Roy P-M, van Es N, Ay C, ten Cate H, Vander Borght T, et al. D-dimer Testing in Pulmonary Embolism with a Focus on Potential Pitfalls: A Narrative Review. Diagnostics. 2022; 12(11):2770. https://doi.org/10.3390/diagnostics12112770

Chicago/Turabian Style

Wauthier, Loris, Julien Favresse, Michaël Hardy, Jonathan Douxfils, Grégoire Le Gal, Pierre-Marie Roy, Nick van Es, Cihan Ay, Hugo ten Cate, Thierry Vander Borght, and et al. 2022. "D-dimer Testing in Pulmonary Embolism with a Focus on Potential Pitfalls: A Narrative Review" Diagnostics 12, no. 11: 2770. https://doi.org/10.3390/diagnostics12112770

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop