Next Article in Journal
Interventional Radiotherapy (Brachytherapy) for Nasal Vestibule: Novel Strategies to Prevent Side Effects
Next Article in Special Issue
Prognostic Factors for Restenosis of Superficial Femoral Artery after Endovascular Treatment
Previous Article in Journal
Increased Prevalence of Autoimmune Gastritis in Patients with a Gastric Precancerous Lesion
Previous Article in Special Issue
Aging and Vascular Disease: A Multidisciplinary Overview
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

No More Venous Ulcers—What More Can We Do?

by
Agata Stanek
1,2,3,*,
Giovanni Mosti
3,4,
Temirov Surat Nematillaevich
3,5,
Eva Maria Valesky
3,6,
Tanja Planinšek Ručigaj
3,7,
Malika Boucelma
3,8,
George Marakomichelakis
2,3,9,
Aaron Liew
2,3,10,
Bahar Fazeli
2,11,
Mariella Catalano
2,3,12 and
Malay Patel
2,3,13
1
Department of Internal Medicine, Angiology and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland
2
Vascular Independent Research and Education, European Foundation, 20157 Milan, Italy
3
VAS-International Consortium—International No More Venous Ulcers Strategic Network, 20157 Milan, Italy
4
Angiology Department, MD Barbantini Clinic, Via del Calcio 2, 55100 Lucca, Italy
5
Department of Specialized Surgery, Central Hospital of Ministry of Internal Affairs, Chimboy St. 2 A, Almazar District, Tashkent 100095, Uzbekistan
6
Department of Dermatology, Venereology and Allergology, University Hospital, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
7
Dermatovenereological Clinic, University Medical Centre Ljubljana, Gradiskova 10 Street, 1000 Ljubljana, Slovenia
8
Department of Internal Medicine, University of Algiers, Bachir Mentouri Hospital, Algiers 16208, Algeria
9
4th Department of Internal Medicine, General Hospital of Evangelismos, 16676 Athens, Greece
10
Department of Medicine, Portiuncula University Hospital, University of Galway, H91 TK33 Galway, Ireland
11
Support Association of Patients of Buerger’s Disease, Buerger’s Disease NGO, Mashhad 9183785195, Iran
12
Department of Biomedical, Clinical Sciences L Sacco Hospital, Inter-University Research Center on Vascular Disease, University of Milan, 20157 Milan, Italy
13
Department of Vascular Surgery, Apollo CVHF, Heart Institute, Ahmedabad 380059, India
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(19), 6153; https://doi.org/10.3390/jcm12196153
Submission received: 17 August 2023 / Revised: 12 September 2023 / Accepted: 19 September 2023 / Published: 23 September 2023

Abstract

:
Venous leg ulcers (VLUs) are the most severe complication caused by the progression of chronic venous insufficiency. They account for approximately 70–90% of all chronic leg ulcers (CLUs). A total of 1% of the Western population will suffer at some time in their lives from a VLU. Furthermore, most CLUs are VLUs, defined as chronic leg wounds that show no tendency to heal after three months of appropriate treatment or are still not fully healed at 12 months. The essential feature of VLUs is their recurrence. VLUs also significantly impact quality of life and could cause social isolation and depression. They also have a significant avoidable economic burden. It is estimated that the treatment of venous ulceration accounts for around 3% of the total expenditure on healthcare. A VLU-free world is a highly desirable aim but could be challenging to achieve with the current knowledge of the pathophysiology and diagnostic and therapeutical protocols. To decrease the incidence of VLUs, the long-term goal must be to identify high-risk patients at an early stage of chronic venous disease and initiate appropriate preventive measures. This review discusses the epidemiology, socioeconomic burden, pathophysiology, diagnosis, modes of conservative and invasive treatment, and prevention of VLUs.

1. Introduction

1.1. Definition of Venous Leg Ulcer

According to the CEAP classification, revised in 2004, a venous leg ulcer (VLU) is defined as a full-thickness skin defect, most frequently in the lower leg and ankle region, that fails to heal spontaneously and is sustained by venous hypertension due to chronic venous disease [1]. VLUs usually occur at the malleolar part on the medial and lateral sides of the ankle. However, they also may appear on the supra-malleolar and infra-malleolar areas of the leg and foot [2]. Furthermore, most VLUs are chronic leg ulcers (CLUs), defined as chronic leg wounds that show no tendency to heal after three months of appropriate treatment or are still not fully healed at 12 months [3]. Moreover, it is estimated that between 40% and 50% remain active between 6 and 12 months, and that 10% remain active up to 5 years [4]. It also has been shown that a VLU that fails to decrease in size by 30% (percentage area reduction) of its initial size over the first four weeks of treatment has a 68% probability of failing to heal within 24 weeks [5].

1.2. Epidemiology and Socioeconomic Burden

VLUs are the most severe complication caused by the progression of chronic venous insufficiency (CVI) [1,6,7]. They account for approximately 70%–90% of all chronic leg ulcers [8]. Women suffer three times more than men, and the incidence increases with age [9], and 40% of patients with VLUs present with underlying deep venous disease (DVD) [10].
O’Meara et al. say that 1% of the Western population sometimes suffers from a VLU [11]. Other population-based studies have reported similar prevalence rates, ranging from 0.7% to 2.7% [12]. VLUs across the globe are expected to rise due to an aging and increasingly overweight population.
The essential feature of VLUs is their recurrence. In the study conducted by Finlayson et al., the average relapse time was 42 weeks, with incidences of 22% at three months, 39% at six months, 57% at 12 months, 73% at two years, and 78% within a 3-year follow-up [6]. Moreover, it has been shown that deep venous disease-related VLUs (DRVs) are thought to have higher recurrence rates and an even more significant economic burden. In the study conducted by Kollour et al., the total annual incidences of new or recurrent DRVs in Australia, France, Germany, Italy, Spain, the U.K., and the U.S. were estimated at 122,000, 263,000, 345,000, 253,000, 85,000, 230,000, and 643,000 events, respectively, in 2019 [10].
Additionally, VLUs are related to a significant cause of morbidity. They also have an enormous impact on the social burden. It has been reported that 4.6 million workdays per year are lost secondary to CVI, in which VLUs are the most severe complication [12]. VLUs also significantly decrease quality of life and are painful [13]. Moreover, they could even cause social isolation and depression [14].
Apart from the substantial social burden, VLUs also significantly impact the economic burden. It has been estimated that treating venous ulceration accounts for around 3% of the total expenditure on healthcare [15]. An analysis of Secure Anonymized Information Linkage Databank data from 2007 to 2017, including approximately 2.5 million people over 11 years, showed that the direct cost of managing patients with VLUs was GBP 7706 per patient per annum, which translated to an annual cost of over GBP 2 billion when extrapolated to the UK population [16]. In the U.S., in 2019, the annual direct medical costs for patients with DRVs managed conservatively were approximately USD 10.73 billion (USD) or USD 5527 per person per year [10]. In Singapore (2017), the estimated gross healthcare cost per patient for inpatient, specialist outpatient, and primary care for VLUs was USD 16,761 for one year [17]. In Australia (2014), the total direct costs of treating VLUs (in public and private hospitals and residential care settings) were estimated to be USD 802.55 million [18]. In Germany (2007), the mean total cost of the ulcer per year and patient was EUR 9569 [19]. Guest et al., in their study, noticed that the patient care cost of an unhealed wound was 135% more than that of a healed wound. The authors concluded that the clinical and economic benefits to patients and the NHS could accrue from three strategies focusing on wound prevention, accurate diagnosis, and improving wound-healing rates [20].
A large International Collaborative Network of Scientific Societies (International No More Venous Ulcers Strategic Network) identified common education and awareness strategies to help prevention, intending to join expertise and energies worldwide in concrete plans. Coordination between the existing action plans by different organizations would be an effective way to follow in the future.
This review aims to present the current knowledge on the pathophysiology and treatment of VLUs and, furthermore, to consider what more can be done to decrease their number.

2. Pathophysiology of Venous Ulcers

Patients with venous leg ulcers have venous hypertension and abnormally sustained venous pressure elevation upon ambulation (normal venous pressure decreases with walking), which results from vein reflux or outflow obstruction.
Venous outflow may also be impaired due to poor calf muscle pump function, which damages the venous system’s ability to overcome the venous blood return to the heart. The limitation of ankle movement seems to be an essential contributor to calf muscle pump failure and a risk factor for ulceration [21]. Several risk factors of VLU development have been identified, including, among others, an age older than 55 years, an increased body mass index (BMI), a family history of CVI, a history of venous thromboembolism disease and superficial thrombophlebitis, a sedentary lifestyle, skeletal or joint disease of the legs, multiparous women, and severe stages of chronic venous diseases (lipodermatosclerosis, active or healed venous ulcers in history) [22].
Although venous hypertension results in ulceration, the exact mechanism remains unclear. Several hypotheses have been proposed, such as abnormalities of the fibrinolytic system, peripapillary fibrin deposition causing decreased oxygen diffusion to tissues, the trapping of growth factors by extravasated macromolecules around the vessels and in the dermis, limiting their function, and leucocyte margination and activation with the subsequent local release of inflammatory mediators [23]. Consequent microcirculatory changes lead to venous hypertensive microangiopathy (enlarged permeable capillaries, abnormally increased skin flux, edema, altered microlymphatic circulation, decreased partial pressure of oxygen, and increased carbon dioxide) that results in ulceration [24].
The effect on the microcirculation begins with altered shear stress on the endothelial cells, causing them to release vasoactive agents and express selectins, inflammatory molecules, chemokines, and prothrombotic precursors [25,26]. Mechanical forces and low shear stress are sensed by the endothelial cells via intercellular adhesion molecules-1 (ICAM1, CD 54), vascular cell adhesion molecules1 (VCAM-1, CD-106), and endothelial leucocyte adhesion molecule1 (CD-62, E-selectin). CVD patients have increased expressions of ICAM-1 and VCAM-1. A key component of inflammation in VLUs is the increased expressions of matrix metalloproteinases and the production of cytokines (Transforming growth factor-β1, Tumor necrosis factor-α, Interleukin-1) [27].
Another area of research in the pathophysiology of LVUs is gap junctions. Gap junctions are proteins that play critical roles in the pathogenesis of chronic wounds, mainly involved in inflammation, edema, and fibrosis. Connexins (a component of gap junctions) are abnormally elevated in the wound margins of VLUs. Connexins seem to play an essential role in the inflammatory response and VLU healing [28].
The pathophysiology of venous ulcers is shown in Figure 1.

3. Diagnosis of Venous Ulcers

The diagnosis of leg ulcers is based on medical history, clinical assessments, functional/diagnostics testing, blood tests, biopsy, bacteriologic or mycologic swabs, histopathology, and direct immunofluorescence [29,30,31,32] (Table 1). The clinical diagnosis as the first step is available everywhere, allowing for the diagnosis of the majority of VLUs. In the next step, several complementary diagnostic procedures are used [27,29] (Table 2).
Ulcers on the lower extremities occur from various etiologies; it is essential to know the possible differential diagnoses of the various wounds on the lower leg. The clinical findings may be similar, but the etiologies can differ [30,32] (Table 2).
Regarding investigations, individual studies’ recommendations could be more robust [25,26] (Table 3).
VLUs are more common in patients with positive family histories of chronic venous insufficiency, in patients with higher body mass indexes, in patients with histories of pulmonary embolism or superficial-/deep-vein thrombosis, in diseases of the skeleton or joints of the lower extremities (due to poor mobility or immobility of the ankle), in women with multiple pregnancies or on hormone therapy, in those patients who have previously had ulcers, in patients who have lipodermatosclerosis and other signs of venous disease or venous insufficiency, and in patients who have typical evening swelling of the legs. Poor prognostic signs for healing are an ulcer duration longer than three months, an initial ulcer size of 10 cm or more at the start of treatment, and the presence of lower-extremity arterial disease. Patients with venous ulcers have ulcers that are usually shallow with well-defined edges and that are often located in the lower two-thirds of the lower leg. Signs of venous disease, such as varicose veins, edema, corona phlebectatica, atrophie blanche, and eczema due to stasis, may also be present. At the same time, patients should have palpable pedal pulses and a calculated brachial–ankle index above 0.85. On Duplex ultrasound, superficial and/or deep veins or perforating veins are insufficient. Severe complications include infection and malignant changes in the ulcer. All these symptoms and signs, as well as associated diseases and specifics in the anamnesis and clinical picture, as well as objections to investigations, help us diagnose venous ulcers and distinguish them from other possible causes of leg ulcers [33,34,35].

4. Current Treatment of Venous Ulcers

Achieving the goal of having no more VLUs would prevent their formation. Prevention by conservative methods would be primary or secondary. Unfortunately, we have very few negative data for primary prevention. Indeed, only one paper reported that medical compression stockings (MCSs) could not prevent ulcer formation in patients with previous deep-vein thrombosis [36]. However, more data confirm that MCSs can effectively prevent ulcer recurrence. It is also essential to have less VLUs, as ulcer recurrence is up to about 78% of patients with histories of previous VLUs [37] in the first few months after ulcer healing [38,39]. The compression pressure must be as high as possible, as it was shown that the higher the compression pressure, the lower the risk of VLU recurrence [40].
Nevertheless, patient compliance must be considered, as it appears to be even more critical than the MCS compression pressure. Patients wearing compression are more likely to avoid ulcer recurrence than patients not wearing compression independent of the MCS pressure [41]. Even if this chapter deals with conservative treatment, it is worthwhile highlighting that surgical ablation is more effective than compression alone in preventing ulcer recurrence [42].
While it is important to have fewer VLUs, trying to shorten the ulcer healing time is as meaningful too. Both surgical and conservative procedures are effective at shortening the healing time. It has been proven that invasive procedures are more effective than compression alone to shorten the VLU healing time [43].
The different modes of the prevention and treatment of VLUs are shown in Figure 2.

4.1. Conservative Treatment

Conservative treatment of VLU patients mainly includes compression therapy, lifestyle changes, physical rehabilitation, local treatment, and pharmacologic treatment.

4.1.1. Compression Therapy

VLUs are primarily due to a venous hemodynamic impairment characterized by ambulatory venous hypertension. Any VLU treatment aims to reduce ambulatory venous hypertension to favor VLU healing. Surgical or conservative procedures can achieve ambulatory venous hypertension reduction. Compression therapy is considered the first treatment option in the conservative management of VLUs. It is the only therapeutical procedure that achieved grade 1A in the most recent guidelines and consensus papers on VLU treatment [44,45,46,47]. Compression therapy can be exerted using inelastic materials (short-stretch or inextensible bandages, adjustable compression wraps (ACWs), and hybrid pumps) and elastic fabrics (stockings or bandages) [48].

Compression Materials and Their Hemodynamic Effects

  • Inelastic materials
Inelastic materials exert their effect by resisting the increase in the muscle volume during muscular contraction in a standing position and during physical activity (the leg will give way). When standing up from the lying-down position, inelastic material causes a significant compression pressure increase compared with supine pressure, and very high pressure peaks when walking (Figure 3). The main stiffness index, the Static Stiffness Index (SSI), which is the difference between the standing and supine pressure, is >10 mm Hg, which characterizes inelastic materials [49,50,51].
The strong standing pressure and the intermittently strong or very strong pressure peaks during exercise will overcome the intravenous pressure [52] intermittently occluding the vein, mimicking a kind of valve mechanism. Due to these effects, inelastic materials can significantly improve impaired venous hemodynamics by reducing venous reflux [53,54], increasing the calf pumping function [55,56], and decreasing the AVH [57].
Inelastic materials are challenging, and well-educated personnel are required when applying inelastic bandages. A series of papers demonstrated that only 10–60% of health personnel treating venous ulcers could apply the target pressure with different inelastic bandages [58,59,60,61,62,63]. This is different for adjustable compression wraps, which are easier to use and are self-applied, facilitating self-management by the patients [64,65,66].
2.
Elastic materials
The elastic materials (elastic stockings or elastic bandages with extensibilities higher than 100%) cannot achieve this intense pressure or pressure peaks during walking. Elastic material gives way to muscle expansion, which occurs during standing and physical activity, resulting in a very low pressure increase when moving from the supine to the standing position. The SSI is always < 10 mm Hg in the elastic range, and the walking pressure amplitude (WPA) and dynamic stiffness index (DSI) are very low too (Figure 4).
Elastic materials tend to return to their initial length when stretched, and this “return force” is directly related to the stretch applied to the material. Suppose that the elastic material is strongly stretched to exert intense pressure (Figure 5), or several elastic stockings are superimposed. In this case, the final compression will be very painful and not tolerated by the patient after a short time from application. Consequently, when adequately stretched, an elastic bandage will exert a supine pressure of 30–40 mm Hg. Elastic stockings exert a wide range of pressure, from 18 to >50 mm Hg, according to their classification (I–IV class). Due to the very light pressure increase during standing and walking, the final pressure will never approach the intravenous pressure, resulting in the inability to narrow or occlude the veins [55], exerting a poor hemodynamic effect, which is significantly lower compared with inelastic material [56,57,58,59,60]. The advantage of elastic materials is that they are easy to apply and self-apply, usually as a single component.
The hemodynamic effects of compression therapy are summarized in Table 4.
3.
What material for ulcer treatment?
A long-standing debate exists about the preferred material used when facing a VLU. As described above, inelastic material (bandages or ACWs) is significantly more effective than elastic material in counteracting venous hemodynamic impairment [53,54,55,56,57]. As venous leg ulcers are due to a venous hemodynamic impairment, inelastic material, the best option to improve the venous hemodynamic impairment, should be the best treatment to increase the VLU healing rate. We have evidence that the higher the compression pressure, the higher the healing rate [11,67], which is in favor of inelastic bandages, which exert a much higher standing pressure than elastic materials.
  • Inelastic bandages. When correctly applied to exert strong pressure, inelastic bandages can achieve an ulcer healing rate close to 100% in three months of treatment [68]. Therefore, preferring inelastic material in VLU treatment is the right option from a logical point of view, but we need ultimate proof that this is correct at present. In contrast, many papers claim the greater effectiveness of elastic stockings or bandages than inelastic material in achieving ulcer healing [69,70,71,72,73,74,75,76,77,78,79,80,81,82]. However, studies comparing elastic and inelastic devices have so many flaws that their conclusions are hard to believe [83]. Two significant deficiencies in papers on compression therapy must be highlighted: the lack of compression pressure measurement and stiffness assessment, and the lack of healthcare professionals’ expertise in applying inelastic bandages. The compression pressure, the dosage of the compression therapy, was seldom measured in the papers comparing different materials, even though compression pressure measurements are easy to perform [84]. Not measuring the compression pressure and not knowing the level of expertise of healthcare professionals [58] make it impossible to tell whether the bandages were correctly applied, making any conclusion challenging to trust. Inelastic bandages could also be so poorly applied that their compression pressure is lower than that exerted by an elastic kit [80]. In addition, not measuring the pressure or assessing the SSI produced a notable mistake in almost all studies comparing elastic and inelastic bandages. In these studies [69,70,71,72,73,74,75,76], the prototype of elastic material was the so-called four-layer bandage, which was dogmatically considered elastic, as it comprises four different elastic components. Nevertheless, measuring the supine and standing pressure of the final bandage and calculating its SSI showed that the SSI is in the inelastic range [85]. In conclusion, all these studies compared two different inelastic bandages, and the conclusions must be critically reviewed;
  • Adjustable compression wraps (ACWs). These devices are quite inelastic and offer the advantages of inelastic material, already reported, in terms of improving the venous hemodynamics [86,87]. At the same time, they are straightforward to use and can be applied and re-adjusted by the patients themselves after a very short wear and education time [64,65,66]. ACWs have been proven more effective at achieving healing in patients with VLUs when compared with Unna boot bandages [88], four-layer bandages [89], or two-layer bandages [90]. Even though more studies must confirm these results, their outcomes seem promising;
  • Elastic stockings. Is there any role for MCSs? As already reported [40], MCSs are very effective in preventing ulcer recurrence after ulcer healing, but they also have a role in the active VLU treatment, especially when using the so-called elastic kits of two superimposed stockings and exerting pressure at about 40 mm Hg. The elastic kits were effective in small ulcers of recent onset. They achieved ulcer healing in 36–96% of patients with these VLU characteristics [77,78,79,80,81]. As ACWs, elastic kits do not require expert personnel to be applied and allow self-management.
In conclusion, inelastic materials are the most effective treatment modality, and we must choose between inelastic composite bandages (difficult to apply) or Velcro® devices, which are easier to apply and allow self-management, depending on personal expertise. Elastic kits may offer an alternative solution in small and recent-onset ulcers.

4.1.2. Lifestyle and Physical Exercise

Patients suffering from venous ulcers are often elderly and not infrequently affected by comorbidities. The most frequent of these comorbidities are orthopedic impairment and obesity. Orthopedic impairment may prevent them from normal ambulation, leading to calf muscle pump inefficiency and reduced venous output from the lower leg. In addition, painful joints and obesity may favor long sitting positions. This condition results in sedentarism, the further deterioration of the venous stasis in the lower leg, and a vicious cycle sets in [91]. Encouraging an alternation of rest in a supine position with elevated legs, physical activity, weight loss, and compression garments is mandatory in these patients. Leg elevation aids venous drainage, reducing leg swelling and improving microcirculation. Leg elevation combined with wearing compression garments is so important that it increases the VLU healing speed and reduces the VLU recurrence rate [92,93]. Elevation was given an IA recommendation from the European Society of Vascular Surgery in 2015 [94].
Physical activity can be simple walking when the patients do not have a significant mobility impairment. Correct walking activates the calf muscle pump and increases the venous output from the lower leg. When the patients suffer from substantial joint impairment, an exercise program to improve mobility, and especially the range of motion of the ankle, knee, and hip joints, may favor VLU improvement and healing. At least these procedures may result in reduced venous ulcer recurrence rates [95]. In turn, it was shown that patients who kept on with orthostatism, became obese, or did not wear compression hosiery developed progressive venous symptoms that could expose them to an increased risk of VLUs [96].
Physical activity, including walking speed, duration, and ankle motion, is significantly reduced in patients with VLUs [97]. This is precisely contrary to what is necessary, as venous return from the lower leg is based on the venous pumping system requiring active calf muscle contraction and ankle mobility with a normal range of motion. Exercise programs focused on calf muscle activities and ankle rehabilitation are the most effective at increasing the VLU healing rate.
Some reports demonstrated that patients adhering to exercise programs, specifically resistance exercise combined with ankle and/or foot range-of-motion exercises, as an adjunctive treatment to standard care, have a higher chance of healing and better functional outcomes than those who do not adhere to exercise programs [98,99,100,101]. In addition, they are also more likely to avoid an ulcer recurrence [95].
Although the evidence is still relatively weak (small studies, short intervention periods, etc.), at least we have some proof of the importance of physical exercise in improving the healing rate and preventing recurrences.

4.1.3. Local Treatment

Most healthcare providers usually apply local dressings on the ulcer bed when treating patients with leg ulcers. They are claimed to create a moist environment on the ulcer that aids in faster healing. A moist environment makes removing debris or dead tissue from the ulcer bed easier. In addition, local dressings contribute to controlling exudate excess-retaining fluids from moving to peri-wound tissue to control inflammation and infections.
Nevertheless, their effectiveness at increasing the VLU healing rate is still debated. A previous Cochrane review denied any role of local dressing in ulcer healing. The conclusion was as follows: “The type of dressing applied beneath compression has not been shown to affect ulcer healing” [102]. Only recently, a less undesirable conclusion more open to the positive effectiveness of local dressings was published by Cochrane reviewers: “In the light of the rest of the network meta-analysis evidence, we cannot be very confident about any conclusion, and the network as a whole represents low-certainty evidence” [103]. Even if data on VLU healing are not conclusive, it is possible to state that local dressings are, at least, effective in pain, infection, and exudate control, large ulcer healing, and cost reduction [104,105,106,107,108]. These effects may improve the patient’s quality of life [109].

4.1.4. Pharmacologic Treatment

The effectiveness of systemic drugs at improving the VLU healing rate has been reported in several papers. Micronized purified flavonoid fraction (MPFF) is one of the most studied drugs in this field.
Flavonoids interact with several biological systems. They have an anti-inflammatory effect, inhibiting granulocyte and macrophage infiltration through the venous endothelium and reducing circulating growth factors and endothelial cell adhesion molecules. Finally, flavonoids prevent/reduce edema formation by decreasing capillary filtration [110,111,112]. The clinical beneficial effects of MPFF are due to these properties. Micronized purified flavonoid fraction as an adjunct to compression therapy increased the ulcer healing rate and shortened the healing time while improving venous symptoms in many randomized controlled trials (RCTs) [113,114,115,116].
Sulodexide, a drug combining low-molecular-weight heparin and dermatan sulfate with profibrinolytic and antithrombotic effects, was demonstrated to improve the VLU healing rate in a randomized multicentric placebo-controlled trial that enrolled 235 patients treated for three months [117]. More recent papers and meta-analyses confirm the effectiveness of sulodexide in VLU healing [118,119]. Also, a recent Cochrane review reports positive effects of sulodexide in VLU patients but also highlights that “the evidence is only low quality and the conclusion is likely to be affected by new research”, and that “Further rigorous, adequately powered RCTs examining the effects of sulodexide on healing, ulcer recurrence, quality of life and costs are necessary” [120].
Pentoxifylline inhibits neutrophil activation, cytokine-mediated white cell adhesion to the endothelium, and oxidative stress [121]. It was initially used in patients with peripheral arterial disease (PAD) [122] and in the off-label in VLUs. Pentoxifylline could improve VLU healing via some different mechanisms: it improves the microcirculatory blood flow by increasing the erythrocyte flexibility, induces prostacyclin synthesis, exerts a vasodilatory effect, inhibits platelet aggregation, and exerts a fibrinolytic effect [123]. The actual effectiveness of pentoxifylline at increasing the VLU healing rate is controversial. Some studies report good results for improving VLU healing compared with placebos [124], while others report conflicting results [125,126]. These contrasting data reflect different conclusions in review papers. While the Cochrane review states that “Pentoxifylline is an effective adjunct to compression bandaging for treating venous ulcers and may be effective in the absence of compression” [127], a more recent meta-analysis concludes that the “Currently available evidence suggests that pentoxifylline could help venous leg ulcers heal more quickly and effectively. However, the evidence is insufficient to prove the results due to moderate-certainty evidence. Large-scale, well-designed, randomized clinical trials are warranted” [128].

4.2. Invasive Treatment

4.2.1. Surgical and Endovenous Treatment of Superficial Venous Insufficiency

The history of the surgical treatment of lower-limb varicosities goes back thousands of years. The first medical document describing ligation surgery and the surgical excision of varicosities was written by the Roman physician Celsus. It is followed by a description of the technique, like that of the contemporary saphenous stripping written by the Greek physician Galen, in which, for his technique, he described that varicosities were directly irritated by a hooked tool wire, aiming to extract as much of the vein as possible [129].
These methods are based on modern varicose vein treatment methods, such as ligation and stripping, as well as phlebectomy of the veins. Today’s varicose vein surgery is developing by leaps and bounds thanks to the basis obtained in ancient times.
The contemporary era of the surgical treatment of varicose vein disease began at the end of the nineteenth century when Friedrich Trendelenburg [130] described the great saphenous vein (GSV) ligation.
Today, there are two theories about the evolution of varicose veins and, accordingly, two main directions for their surgical treatment. The first one is descending theory, which proposes that varicose veins and chronic venous insufficiency are due to a failure of the proximal components of the superficial venous system: the valves at the saphenofemoral junction (SFJ) or saphenopopliteal junction (SPJ). According to this theory, treatment should begin with removing the SFJ or SPJ, abolishing venous reflux. Surgical methods such as high ligation, stripping, the ligation of varicose veins, and phlebectomy rely on this theory.
Until the first decade of the 21st century, the fundamental approach in the surgical treatment of varicose veins with great saphenous vein incompetence was long stripping [131] and its modifications.
This method has several variants: proximal stripping with a probe—the classical Babcock method; proximal stripping with the invagination (inversion) of the vessel; distal stripping with and without invagination [132,133,134]. These procedures have been performed safely for decades, although not always without associated morbidity. Postoperative pain, paresthesia due to saphenous nerve injury, hematoma, and bruising are the most common complications [135,136,137], which provoked the development of other methods, such as perforation imagination (PIN) stripping and cryo-stripping, in the hope of reducing postoperative complications [137,138,139,140]. Despite theoretically justified methods, the recurrence rate of varicose veins after five years has been reported to vary from 20% to 80% [141].
In the past decade, alternative treatments, such as endoluminal procedures, have gained popularity. These procedures can be divided into two groups: thermal procedures, in which the ablation of the veins is achieved via heat, and non-thermal procedures, in which the veins are obliterated chemically after the mechanical injuring of the vein wall by the tip of the catheter.
The most widely used endoluminal procedures are radiofrequency ablation (RFA) and endovenous laser ablation (EVLA). The RFA and EVLA procedures were introduced at the end of the 20th century. In contrast to stripping operations, the great advantage of endoluminal methods is that they are minimally invasive. Treated patients can undertake normal physical activities the same or next day, often including sports, and, as a rule, with no impairment due to the previous endovenous operation [142]. In radiofrequency procedures, the veins are heated to 120 °C in sections by a radiofrequency probe inserted through the lumen of the catheter [143]; for laser procedures, the use of high water absorption wavelengths (1470–1940 nm) and radially emitting laser probes is preferred. The temperature reached by a laser with a wavelength of 1470 nm with a radial probe is about 120–140 °C (±20 °C) [144,145]; it is now possible to precisely occlude a saphenofemoral recurrence with a remaining saphenofemoral stump [146]
Other new endoluminal methods are the non-thermal procedures: mechanochemical ablation (MOCA) and the cyanoacrylate (adhesive) closure system.
The MOCA procedure is used to seal a vein via a combination of mechanical action (the tip of the catheter rotates or has flexible cutting elements, resulting in painless damage to the intimate layer of the vein wall) and chemical action (a sclerosing agent is injected into the vein below the tip of the catheter) [147,148]. In the adhesive system, the vein is sealed with a special vein-adhesive material (mono-cyanoacrylate) [149].
Usually, all open surgical and endoluminal methods are completed with the mini phlebectomy of varicosities introduced by Muller [150] and supplemented by Varady hooks [151].
Based on descending theory, all these methods either remove or destroy the great saphenous (GSV) or short saphenous (SSV) vein to eliminate reflux in the superficial venous system. In addition to these methods, alternative techniques exist, relying on the ascending theory of varicosity, which advocates the proximal progression of distal venous incompetence and is supported by the observation that superficial venous incompetence often occurs with a competent saphenous junction. CHIVA (Ambulatory Conservative Hemodynamic Treatment of Venous Insufficiency) and ASVAL (Ambulatory Selective Varices Ablation Under Local Anesthesia) are two techniques characterized by the targeted therapy of the venous system without saphenous trunk destruction or removal [152].
CHIVA is the hemostatic column of blood fragmentation, achieved via the selective ligation of the truncal vein, the preservation of communicating branches draining into the deep venous system, and the ligation of refluxing saphenofemoral connections [152].
According to a review published by Cochrane in 2013, CHIVA reduced recurrence rates, improved quality-of-life results, and reduced side effects compared to stripping and compression [153]. A comparison between CHIVA, stripping, and EVLA found that CHIVA and EVLA patients had improved cosmetic results and reduced pain scores compared to those undergoing open surgery [154].
ASVAL is a minimally invasive technique based on the ascending theory of varicosity development. ASVAL aims to remove varicose tributaries, considered to be the origin of incompetence, without treating the saphenous trunk. The removal of varicosities reduces the hemodynamic load on the saphenous vein [155].
A retrospective analysis comparing the results of ASVAL and saphenous vein sparing revealed a reduction in more than two-thirds of cases over a four-year follow-up period, while symptoms improved or disappeared in most cases. There was no recurrence in 95% of patients at one year nor in 88.5% of patients at four years [156].
The summary of the invasive treatment of superficial venous insufficiency is presented in Figure 6.
In conclusion, the surgery for varicose veins is still evolving. There has yet to be a consensus on which treatment modality is the gold standard, and large multicenter studies are needed to prove the benefit of one of these surgical modalities in treating chronic venous insufficiency.

4.2.2. Surgical and Endovenous Treatment of Deep-Vein Occlusion

The main venous outflow of the lower limb is the deep venous system [157], and the foremost cause of vein occlusion syndromes is deep venous thrombosis (DVT), where the DVT of the iliofemoral segment is more clinically significant, such as iliofemoral DVT, which is associated with a higher risk for recurrent venous thromboembolism (VTE) and a higher risk for the development of a post-thrombotic syndrome (PTS) [158]. Other causes of deep-vein occlusion syndromes are anatomical conditions, such as the compression of the left common iliac vein between the right common iliac artery and the fifth lumbar vertebra, also known as May–Thurner syndrome [159], which causes non-thrombotic deep-vein occlusion syndromes. Unfortunately, May–Thurner syndrome causes non-thrombotic deep-vein occlusion syndromes and is also the source of iliofemoral DVTs [160].
Usually, the surgery of deep venous systems is the most challenging part of the work of a vascular surgeon, such that the results of surgical procedures in the deep venous system are worse than those in the arterial system.
Surgical procedures in the deep venous system are divided into two directions depending on the cause of the venous insufficiency: interventions to correct venous reflux and interventions to correct venous obstructions; both are consequences of PTS (Figure 7).
In turn, venous reflux surgery depends on the condition of the valve. For example, performing a valvuloplasty and a valve transposition in cases of intact but incompetent valves is better. Still, in the case of destroyed valves, an axillary vein autotransplantation is the best choice [161].
Dr. Kistner performed the first open valvuloplasty in 1968 [162], and, today, there are a few types of valvuloplasty: internal valvuloplasty, external valvuloplasty, and transcommissural valvuloplasty [163].
The most famous surgical procedure for deep venous obstruction is the crossover bypass (Palma–Dale procedure) [164,165]. This procedure is performed when it is impossible to recanalize the occluded area or with extended lesions of the iliofemoral segment. In a crossover bypass, the great saphenous vein of a donor limb is exposed and implanted into the opposite saphenofemoral junction. If it is not possible to use the GSV, then a 10 mm PTFE (polytetrafluoroethylene) femoro-femoral bypass graft can be used. Another option is an in-line bypass procedure, which is indicated in the long femoro-iliocaval occlusive segment using an expanded PTFE graft, the most used material for bypass. An additional arteriovenous fistula is created distally to maintain inflow, and it is closed 4–8 weeks after surgery [166]. Unfortunately, life-long anticoagulation is usually needed. When it is not possible to perform crossover or in-line procedures, an endo-phlebectomy is the last hope, in which the post-thrombotic vein is longitudinally exposed at various segments, and the synechiae attached to the intimal layer is carefully removed with scissors.
The treatment for deep-vein occlusion syndromes changed in the 1990s with the introduction of endovascular techniques. In the past, open surgery, including a bypass for chronic venous occlusive disease and thrombectomy for acute thrombosis, were invasive procedures, often used unselectively without proper pre- and intra-operative imaging and frequently with disappointing results. These techniques have been widely replaced by endovascular interventions, such as percutaneous recanalization combined with stenting [167].
Over the past decade, insights into treating deep venous disorders via stents have increased. Venous stenting is recognized as a possible treatment to help assist patients with symptomatic vein occlusion syndromes as an alternative to conventional surgery (Figure 8).
Treating deep-vein occlusion syndromes via stents, including involvement in the inferior vena cava, has acceptable patency rates and reduced symptoms in most patients [168]. In a published study with over 1500 patients, iliac vein stenting for vein occlusion syndromes was considered safe, with high patency rates of up to five years. Improvements in pain, ulceration, and limb swelling were noted [169]. A follow-up meta-analysis of 37 studies demonstrates good technical success, with minimal periprocedural complications and symptom relief at the final follow-up for five years. Complication rates are less than 1% for major bleeding, pulmonary embolism, and mortality, with 1.0–6.8% for early stent thrombosis [170].
The endovascular treatment of the occlusion of the iliac vein is presented in Figure 8.
Most initial venous stents were not designed for use in the venous system. Initial stent development addressed these challenges by increasing the crush resistance needed in venous stents [171]. Recently, dedicated venous stents were introduced, which, in theory, with increased chronic outward force and radial resistive force, should have better results than non-dedicated stents; today, we have a few works confirming that dedicated stents have results comparable to those for non-dedicated stents in terms of clinical outcomes, QOL improvement, and stent patency [172]. What about vein balloon angioplasty alone? The research work published by Aurshina A et al. in 2019 shows that in 49% of procedures, balloon dilatation relieved a previously stenotic area, and there was a highly variable response to balloon venoplasty, with 22% of procedures demonstrating no change and 29% of procedures resulting in a worsening of the cross-sectional area. This decrease in the area could be explained by the elastic recoil or spasm of the vessel wall upon dilatation. Regardless of the etiology, these require routine balloon dilatation before stent placement [173].
In conclusion, endovascular treatment, especially the stenting of the iliofemoral segment, has become an essential treatment option for chronic (thrombotic or non-thrombotic) obstructive venous disease. All vein occlusion syndromes should be treated with stenting because of the unintended chance of recoil and spasm. Balloon dilatation alone will not eliminate the external compression or PTS occlusions responsible for these lesions.

4.2.3. Surgical Debridement and Skin Grafting

The direct surgical treatment of venous ulcers involves surgical debridement and skin grafting. Regular debridement needs to reduce the necrotic burden and achieve healthy granulation tissue [174]. The debridement can be performed with sharp surgical, enzymatic, mechanical, autolytic, and chemical techniques. Surgical debridement is usually performed in the operating room with systematic or regional anesthesia by removing slough and necrotic tissue [175]. Wounds close by two main mechanisms: epithelial migration and wound contraction. Both processes occur from the edges of the wound and aim to fill the lesion and shrink the wound edges. Unfortunately, these natural mechanisms are not always sufficient. Skin grafts or flaps can be considered alternative solutions to accelerate wound healing [176]. Early skin grafting significantly improves quality of life by reducing pain and accelerating wound healing.
There are two types of skin grafts: split-thickness skin grafts (STSGs) and full-thickness skin grafts (FTSGs). Split-thickness skin grafts consist of the epidermis and some layers of the dermis. Full-thickness skin grafts are composed of the epidermis, dermis, and various layers of subcutaneous tissue [176]. STSGs are characterized by a poor cosmetic outcome, but they require lesser revascularization after implantation, which is why these grafts can be used to treat wounds with damaged blood supplies, such as venous insufficiency ulcers [177]. If the most crucial goal is the aesthetic outcome, then FTSGs are the best choice.

4.3. Challenges in the Treatment of Mixed Arterial–Venous Leg Ulcers

The coexistence of both venous and arterial diseases has been reported in 15–26% of patients with lower-extremity ulceration. Also, arterial insufficiency (ABI < 0.8) is postulated to be present in up to 30% of the patients with VLUs [178,179,180,181].
Discovering arterial disease in patients with VLUs is crucial because compression therapy is the primary treatment for VLUs and, simultaneously, it would be challenging in patients with PAD due to the probability of worsening in critical limb ischemia cases [182]. Patients with mixed arterial–venous ulcers may complain of more leg pain than those with venous ulcers alone [183]. Furthermore, the inadequate presence of granulation tissue and rolled wound edges of VLUs could be the contemporary signs of arterial insufficiency [183,184]. However, ABI would be determinative of PAD diagnosis, especially in old patients who may not have claudication because of decreased mobility. In addition, because the existence of pulsation cannot rule out the possibility of PAD, ABI should be performed for all patients who complain of VLUs [182,183,184].
Until recently, the optimal treatment for mixed arterial–venous ulcers remained unclear. However, compression therapy with a pressure of 20–30 mmHg would be acceptable in cases with ABIs between 0.5 and 0.8 to avoid the deterioration of the tissue vitality [185,186]. Patients with ABIs less than 0.5 should not receive compression therapy before any revascularization procedures [187,188].

5. Focus on Prevention: What More Can We Do?

5.1. Primary Prevention and Risk Factors

The risk factors for developing a first-time VLU need to be better investigated, and primary prevention in patients with CVD needs to be more appreciated in general practice. A systematic review identified several risk factors in developing a first VLU: older age, deep-vein reflux, DVT, higher body mass index (BMI), low physical activity, arterial hypertension, and family VLUs [189].
During the clinical examination, the ulcer surroundings and skin changes, such as corona phlebectatica paraplantaris, varicose veins, or signs of chronic skin inflammation, should be documented [190,191,192]. Hemosiderosis, lipodermatosclerosis, stasis dermatitis, atrophie blanche, and corona phlebectatica paraplantaris are considered to have predictive value for the development of VLUs [193] and/or the recurrence rate [194]. Of particular clinical relevance is the corona phlebectatica paraplantaris, as this can be an early isolated clinical sign of severe CVD [195]. The venous plexus of the foot does not carry venous valves; therefore, venous ectasia manifests early here in venous hypertension. The CEAP classification of CVD, updated in 2020 and established internationally, includes this aspect by adapting the clinical severity score (C4c: corona phlebectatica paraplantaris) [196].
A higher BMI and low physical activity indicate that lifestyle factors are also involved in VLU development. The risk factor “obesity in VLU” comprises several aspects. In addition to the limited general and ankle mobility of some obese patients, obesity directly influences venous hemodynamics by raising the intra-abdominal pressure. In these situations, the underlying CVD has a greater impact, increasing the risk of VLU recurrence and decreasing the ulcer healing rate [192,197,198,199]. Scholl et al. published a case series of patients who developed VLUs exclusively due to obesity-associated CVD without any venous pathology [200]. Several studies have shown that obesity correlates with increased intra-abdominal pressure and a concomitant increase in pressure in the external iliac and femoral veins, resulting in a reduced flow velocity in the femoral vein [201,202,203,204]. The altered pressure and flow characteristics lead to venous hypertension in the lower extremities. Reduced venous flow velocity, in turn, triggers the CVD-associated inflammatory response.
Moreover, obesity significantly impacts the development of VLUs through adipose tissue mass dysfunction by disrupting the microcirculation caused by the increased secretion of pro-inflammatory cytokines [198,205]. This pathophysiological relationship probably explains the association of the severity of post-thrombotic syndrome with the degree of obesity [206].
Although, as mentioned above, physical exercise, by increasing the movement of the ankle joint and strengthening the muscle pump in the calf of the leg, may help prevent the worsening of the disease [95,96]. The Cochrane meta-analysis published in 2016 and updated in 2023 showed that there is currently insufficient evidence to assess the benefits and harms of physical exercise in non-ulcerated CVI subjects. Therefore, the authors concluded that further research into the effect of physical activity considering the types of exercise protocols (intensity, frequency, and time), sample size, blinding, and homogeneity according to the severity of the disease is required [207].
Therefore, regarding primary prevention, it is essential to consider the risk factors and include them in the treatment strategies.

5.2. Recurrence Prevention

People with VLUs often live with a lifelong ulceration, healing, and recurrence cycle. As over 50% of venous leg ulcers will recur within 12 months of healing, a comprehensive knowledge of the risk factors associated with recurrence is required by health professionals caring for persons with VLUs [208,209,210].
Several randomized trials have shown that invasive therapy of the underlying superficial-vein insufficiency accelerates VLU healing [43,211,212] and reduces the incidence of recurrence [42,213]. These interventions can be endovenous (“ablative”) or open surgery (“surgical”). Even in patients with simultaneous segmental reflux in the deep venous system, the recurrence rates for VLUs were significantly lower in the intervention group (24%) compared to the compression group (52%) [42]. With the increasing severity of deep-vein insufficiency, the influence of invasive interventions on the recurrence rate becomes less, and the effect of the intervention is no longer statistically significant. Nevertheless, pre-existing reflux in the deep venous system should not be considered a contraindication for the invasive therapy of underlying superficial venous reflux because the intervention may improve CVD-associated symptoms [214,215].
Case series on peri-ulcerous ultrasound-guided foam sclerotherapy [216,217] showed a low recurrence rate in addition to the accelerated healing of the ulcerations [218]. Randomized controlled study results still need to be carried out.
So far, data with long-term follow-up after recanalizing the deep venous system in post-thrombotic syndrome still need to be included. Venous stenting as a strategy for preventing VLUs may be useful in selected individual cases [45].
In addition to the early invasive treatment of the underlying pathology in the superficial and/or deep venous system, continued medical compression therapy prevents VLU recurrence. A prospective controlled randomized trial investigated the effect of compression hosiery (ankle pressure of 35–45 mmHg) in 153 patients. The rate of VLU recurrence was significantly reduced from 54% in the compression group compared to 24% in the placebo group [219]. However, three other randomized controlled trials comparing different compression classes of hosiery showed no significant differences [40,41,214]. When selecting the compression class, it is crucial to consider that a higher compression class may be associated with a decrease in adherence [41,215,219,220]. In turn, poor adherence to medical compression therapy causes a higher rate of VLU recurrence [40,214,221]. Studies on the wearing time in the prevention of recurrence are required. In clinical practice, the recommendation is to wear the compression hosiery as consistently as possible whenever the legs are not elevated.
Some of the comorbidities associated with CVD (mentioned above) are also risk factors for a more severe course of CVD and negatively affect the healing process and increase the risk of recurrence [189].
Other relevant risk factors for recurrence are a long history of VLUs, a large wound area, obesity, limited mobility in the upper ankle, lack of patient education, lack of self-management, and lack of treatment adherence. These risk factors should be minimized preventively [42,100,204,213,222,223,224]. The positive effect of education on healing in VLUs has been shown in several studies [100,224,225,226]. The main reason is that structured education increases adherence. Adherence is essential for therapeutic success, especially in compression therapy [224]. Studies investigating a correlation between education and recurrence prevention have yet to be made available. However, it can be postulated from the available data that education increases adherence, thereby improving healing and preventing recurrences. Patients with healed VLUs may benefit from educational actions that include information on risk factors and long-term prevention strategies.
Physical activity and exercises should be recommended to patients with healed VLUs to improve venous return, strengthen the calf muscle pump, and increase the range of motion in the ankle joint [227,228]. A specification of suitable exercises is not possible due to the limited data. Activities should be customized to the patient’s physical capabilities.
Data on whether weight reduction in obesity reduces the recurrence rate of VLU are few. However, because severe comorbidities coexist in these patients, the correlation with the worsening of pre-existing CVD due to the obesity-related increase in the intra-abdominal pressure and the secretion of proinflammatory cytokines, it seems reasonable to recommend weight normalization in patients with VLUs and concomitant obesity.
VLU recurrence prevention involves a combination of strategies, including early interventions addressing underlying venous insufficiency, compression therapy, lifestyle modification, education, and the monitoring of early signs of recurrence by patients and health professionals.

6. Conclusions

A VLU-free world is an ambitious aim, but it may be challenging to achieve with the current knowledge of the pathophysiology and diagnostic, and therapeutical protocols. Conservative and invasive procedures, when properly prescribed, will undoubtedly help. Compression therapy and lifestyle adaptations may play significant roles in conventional approaches, but local and general treatment must be considered.
To decrease the incidence of VLUs, the long-term goal must be to identify high-risk patients early and initiate appropriate preventive measures. Patients with varicose veins are at a significantly increased risk of ulceration, especially those with skin changes of chronic venous insufficiency and associated deep-vein incompetence. Additionally, CVI is often not diagnosed until VLUs develop. Its treatment can be complex and challenging at this stage, often leading to patients experiencing repeated ulceration, healing, and recurrence patterns.
In the meantime, a joint effort of all the active stakeholders to provide a standard and universally acceptable indication with coordinated initiatives to prevent VLUs everywhere is essential, incorporating suggestions that consider the feasibility linked to countries’ different geographies and socioeconomics worldwide.

Author Contributions

Conceptualization, A.S.; writing—original draft preparation, writing—review and editing, A.S., G.M. (Giovanni Mosti), T.S.N. and G.M. (George Marakomichelakis); writing—original draft preparation, E.M.V., T.P.R., M.B. and B.F.; visualization G.M. (George Marakomichelakis), T.S.N. and G.M. (Giovanni Mosti); supervision, G.M. (Giovanni Mosti), A.L., M.C. and M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

We used PubMed and Web of Science to screen articles for this narrative review. We did not report any data.

Conflicts of Interest

G.M. (Giovanni Mosti), T.S.N., E.V., T.P.R., M.B., G.M. (George Marakomichelakis), A.L., B.F., M.C. and M.P. declare no conflict of interest; A.S.—Honoraria for lectures at symposia: Alfasigma, Bayer, Phizer.

References

  1. Eklöf, B.; Rutherford, R.B.; Bergan, J.J.; Carpentier, P.H.; Gloviczki, P.; Kistner, R.L.; Meissner, M.H.; Moneta, G.L.; Myers, K.; Padberg, F.T.; et al. American Venous Forum International Ad Hoc Committee for Revision of the CEAP Classification. Revision of the CEAP classification for chronic venous disorders: Consensus statement. J. Vasc. Surg. 2004, 40, 1248–1252. [Google Scholar] [CrossRef]
  2. Raffetto, J.D. The definition of the venous ulcer. J. Vasc. Surg. 2010, 52 (Suppl. 5), 46S–49S. [Google Scholar] [CrossRef]
  3. Kahle, B.; Hermanns, H.J.; Gallenkemper, G. Evidence-based treatment of chronic leg ulcers. Dtsch. Arztebl. Int. 2011, 108, 231–237. [Google Scholar] [CrossRef]
  4. Wipke-Tevis, D.D.; Rantz, M.J.; Mehr, D.R.; Popejoy, L.; Petroski, G.; Madsen, R.; Conn, V.S.; Grando, V.T.; Porter, R.; Maas, M. Prevalence, incidence, management, and predictors of venous ulcers in the long-term-care population using the MDS. Adv. Skin Wound Care 2000, 13, 218–224. [Google Scholar] [PubMed]
  5. Kantor, J.; Margolis, D.J. A multicentre study of percentage change in venous leg ulcer area as a prognostic index of healing at 24 weeks. Br. J. Dermatol. 2000, 142, 960–964. [Google Scholar] [CrossRef]
  6. Finlayson, K.; Wu, M.L.; Edwards, H.E. Identifying risk factors and protective factors for venous leg ulcer recurrence using a theoretical approach: A longitudinal study. Int. J. Nurs. Stud. 2015, 52, 1042–1051. [Google Scholar] [CrossRef] [PubMed]
  7. Hess, C.T. Venous Ulcer Assessment and Management: Using the Updated CEAP Classification System. Adv. Skin Wound Care 2020, 33, 614–615. [Google Scholar] [CrossRef] [PubMed]
  8. Sen, C.K.; Gordillo, G.M.; Roy, S.; Kirsner, R.; Lambert, L.; Hunt, T.K.; Gottrup, F.; Gurtner, G.C.; Longaker, M.T. Human skin wounds: A major and snowballing threat to public health and the economy. Wound Repair Regen. 2009, 17, 763–771. [Google Scholar] [CrossRef]
  9. Hellström, A.; Nilsson, C.; Nilsson, A.; Fagerström, C. Leg ulcers in older people: A national study addressing variation in diagnosis, pain and sleep disturbance. BMC Geriatr. 2016, 16, 25. [Google Scholar] [CrossRef]
  10. Kolluri, R.; Lugli, M.; Villalba, L.; Varcoe, R.; Maleti, O.; Gallardo, F.; Black, S.; Forgues, F.; Lichtenberg, M.; Hinahara, J.; et al. An estimate of the economic burden of venous leg ulcers associated with deep venous disease. Vasc. Med. 2022, 27, 63–72. [Google Scholar] [CrossRef]
  11. O’Meara, S.; Cullum, N.; Nelson, E.A.; Dumville, J.C. Compression for venous leg ulcers. Cochrane Database Syst. Rev. 2012, 11, CD000265. [Google Scholar] [CrossRef]
  12. Lal, B.K. Venous ulcers of the lower extremity: Definition, epidemiology, and economic and social burdens. Semin. Vasc. Surg. 2015, 28, 3–5. [Google Scholar] [CrossRef] [PubMed]
  13. Berszakiewicz, A.; Kasperczyk, J.; Sieroń, A.; Krasiński, Z.; Cholewka, A.; Stanek, A. The effect of compression therapy on quality of life in patients with chronic venous disease: A comparative 6-month study. Postepy Dermatol. Alergol. 2021, 38, 389–395. [Google Scholar] [CrossRef]
  14. Jones, J.E.; Robinson, J.; Barr, W.; Carlisle, C. Impact of exudate and odour from chronic venous leg ulceration. Nurs. Stand. 2008, 22, 53–61. [Google Scholar] [CrossRef] [PubMed]
  15. Scottish Leg Ulcer Participants. Effect of National Community Intervention Program on healing rates of chronic leg ulcer: Randomised controlled trial. Phlebology 2002, 17, 47–53. [Google Scholar] [CrossRef]
  16. Phillips, C.J.; Humphreys, I.; Thayer, D.; Elmessary, M.; Collins, H.; Roberts, C.; Naik, G.; Harding, K. Cost of managing patients with venous leg ulcers. Int. Wound J. 2020, 17, 1074–1082. [Google Scholar] [CrossRef]
  17. Lo, Z.J.; Lim, X.; Eng, D.; Car, J.; Hong, Q.; Yong, E.; Zhang, L.; Chandrasekar, S.; Tan, G.W.L.; Chan, Y.M.; et al. Clinical and economic burden of wound care in the tropics: A 5-year institutional population health review. Int. Wound J. 2020, 17, 790–803. [Google Scholar] [CrossRef]
  18. Graves, N.; Zheng, H. Modelling the direct health care costs of chronic wounds in Australia. Wound Pract. Res. 2014, 22, 20–24, 26–33. [Google Scholar]
  19. Purwins, S.; Herberger, K.; Debus, E.S.; Rustenbach, S.J.; Pelzer, P.; Rabe, E.; Schäfer, E.; Stadler, R.; Augustin, M. Cost-of-illness of chronic leg ulcers in Germany. Int. Wound J. 2010, 7, 97–102. [Google Scholar] [CrossRef]
  20. Guest, J.F.; Ayoub, N.; McIlwraith, T.; Uchegbu, I.; Gerrish, A.; Weidlich, D.; Vowden, K.; Vowden, P. Health economic burden that different wound types impose on the UK’s National Health Service. Int. Wound J. 2017, 14, 322–330. [Google Scholar] [CrossRef]
  21. Yim, E.; Richmond, N.A.; Baquerizo, K.; Van Driessche, F.; Slade, H.B.; Pieper, B.; Kirsner, R.S. The effect of ankle range of motion on venous ulcer healing rates. Wound Repair Regen. 2014, 22, 492–496. [Google Scholar] [CrossRef]
  22. Vivas, A.; Lev-Tov, H.; Kirsner, R.S. Venous Leg Ulcers. Ann. Intern. Med. 2016, 165, ITC17–ITC32. [Google Scholar] [CrossRef] [PubMed]
  23. Browse, N.L.; Burnand, K.G. The cause of venous ulceration. Lancet 1982, 2, 243–245. [Google Scholar] [CrossRef]
  24. Thomas, P.R.; Nash, G.B.; Dormandy, J.A. White cell accumulation in dependent legs of patients with venous hypertension: A possible mechanism for trophic changes in the skin. Br. Med. J. (Clin. Res. Ed.) 1988, 296, 1693–1695. [Google Scholar] [CrossRef]
  25. Schmid-Schönbein, G.W.; Takase, S.; Bergan, J.J. New advances in the understanding of the pathophysiology of chronic venous insufficiency. Angiology 2001, 52 (Suppl. 1), S27–S34. [Google Scholar] [CrossRef] [PubMed]
  26. Raffetto, J.D. Inflammation in chronic venous ulcers. Phlebology 2013, 28 (Suppl. 1), 61–67. [Google Scholar] [CrossRef] [PubMed]
  27. Raffetto, J.D. Pathophysiology of Chronic Venous Disease and Venous Ulcers. Surg. Clin. N. Am. 2018, 98, 337–347. [Google Scholar] [CrossRef]
  28. Ghatnekar, G.S.; Grek, C.L.; Armstrong, D.G.; Desai, S.C.; Gourdie, R.G. The effect of a connexin43-based Peptide on the healing of chronic venous leg ulcers: A multicenter, randomized trial. J. Investig. Dermatol. 2015, 135, 289–298. [Google Scholar] [CrossRef]
  29. Spoljar, S. Osnovni dijagnosticki postupci kod bolesnika s venskim ulkusom [List of diagnostic tests and procedures in leg ulcer]. Acta Med. Croat. 2013, 67 (Suppl. 1), 21–28. [Google Scholar]
  30. Himanshu, V.; Ramesh, K.T. Venous ulcer. In Ulcer of the Lower Extremity; Khanna, A.K., Tiwary, S.K., Eds.; Springer: New Delhi, India, 2016; pp. 141–162. [Google Scholar] [CrossRef]
  31. Planinšek Ručigaj, T. Diseases of the veins and arteries (leg ulcers), chronic wounds, and their treatment. In Atlas of Dermatology, Dermatopathology and Venereology: Inflammatory Dermatoses; Smoller, B.R., Ed.; Springer International Publishing: Cham, Switzerland, 2022; pp. 1205–1331. ISBN 9783319538044. [Google Scholar]
  32. Morison, M.J.; Moffat, C.J. A framework for patient assessment and care planning. In Leg Ulcers: A Problem-Based Learning Approach; Morison, M.J., Moffat, C.J., Franks, P.J., Eds.; Mosby, Elsevier: Philadelphia, PA, USA, 2007; pp. 119–139. [Google Scholar]
  33. Bonkemeyer Millan, S.; Gan, R.; Townsend, P.E. Venous Ulcers: Diagnosis and Treatment. Am. Fam. Physician 2019, 100, 298–305. [Google Scholar]
  34. Kecelj, N.; Kozak, M.; Slana, A.; Šmuc Berger, K.; Šikovec, A.; Makovec, M.; Blinc, A.; Žuran, I.; Planinšek Ručigaj, T. Recommendations of the diagnosis and treatment of chronic venous disease. Zdr. Vestn. Glas. Slov. Zdr. Društva 2017, 86, 345–361. [Google Scholar]
  35. Srisuwan, T.; Inmutto, N.; Kattipathanapong, T.; Rerkasem, A.; Rerkasem, K. Ultrasound Use in Diagnosis and Management of Venous Leg Ulcer. Int. J. Low. Extrem. Wounds 2020, 19, 305–314. [Google Scholar] [CrossRef] [PubMed]
  36. Doliner, B.; Jaller, J.A.; Lopez, A.J.; Lev-Tov, H. Treatments to prevent primary venous ulceration after deep venous thrombosis. J. Vasc. Surg. Venous Lymphat. Disord. 2019, 7, 260–271. [Google Scholar] [CrossRef]
  37. Abbade, L.P.; Lastória, S. Venous ulcer: Epidemiology, physiopathology, diagnosis and treatment. Int. J. Dermatol. 2005, 44, 449–456. [Google Scholar] [CrossRef]
  38. Finlayson, K.; Edwards, H.; Courtney, M. Factors associated with recurrence of venous leg ulcers: A survey and retrospective chart review. Int. J. Nurs. Stud. 2009, 46, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  39. Moffatt, C.J.; Dorman, M.C. Recurrence of leg ulcers within a community ulcer service. J. Wound Care 1995, 4, 57–61. [Google Scholar] [CrossRef]
  40. Nelson, E.A.; Harper, D.R.; Prescott, R.J.; Gibson, B.; Brown, D.; Ruckley, C.V. Prevention of recurrence of venous ulceration: Randomized controlled trial of class 2 and class 3 elastic compression. J. Vasc. Surg. 2006, 44, 803–808. [Google Scholar] [CrossRef] [PubMed]
  41. Clarke-Moloney, M.; Keane, N.; O’Connor, V.; Ryan, M.A.; Meagher, H.; Grace, P.A.; Kavanagh, E.; Walsh, S.R.; Burke, P.E. Randomised controlled trial comparing European standard class 1 to class 2 compression stockings for ulcer recurrence and patient compliance. Int. Wound J. 2014, 11, 404–408. [Google Scholar] [CrossRef] [PubMed]
  42. Gohel, M.S.; Barwell, J.R.; Taylor, M.; Chant, T.; Foy, C.; Earnshaw, J.J.; Heather, B.P.; Mitchell, D.C.; Whyman, M.R.; Poskitt, K.R. Long term results of compression therapy alone versus compression plus surgery in chronic venous ulceration (ESCHAR): Randomised controlled trial. BMJ 2007, 335, 83. [Google Scholar] [CrossRef]
  43. Gohel, M.S.; Heatley, F.; Liu, X.; Bradbury, A.; Bulbulia, R.; Cullum, N.; Epstein, D.M.; Nyamekye, I.; Poskitt, K.R.; Renton, S.; et al. A Randomized Trial of Early Endovenous Ablation in Venous Ulceration. N. Engl. J. Med. 2018, 378, 2105–2114. [Google Scholar] [CrossRef]
  44. O’Donnell, T.F.; Passman, M.A.; Marston, W.A.; Ennis, W.J.; Dalsing, M.; Kistner, R.L.; Lurie, F.; Henke, P.K.; Gloviczki, M.L.; Eklöf, B.G.; et al. Management of venous leg ulcers: Clinical practice guidelines of the Society for Vascular Surgery® and the American Venous Forum. J. Vasc. Surg. 2014, 60 (Suppl. 2), 3S–59S. [Google Scholar] [CrossRef]
  45. De Maeseneer, M.G.; Kakkos, S.K.; Aherne, T.; Baekgaard, N.; Black, S.; Blomgren, L.; Giannoukas, A.; Gohel, M.; de Graaf, R.; Hamel-Desnos, C.; et al. Editor’s Choice—European Society for Vascular Surgery (ESVS) 2022 Clinical Practice Guidelines on the Management of Chronic Venous Disease of the Lower Limbs. Eur. J. Vasc. Endovasc. Surg. 2022, 63, 184–267. [Google Scholar] [CrossRef]
  46. Mosti, G.; De Maeseneer, M.; Cavezzi, A.; Parsi, K.; Morrison, N.; Nelzén, O.; Rabe, E.; Partsch, H.; Caggiati, A.; Simka, M.; et al. Society for Vascular Surgery and American Venous Forum Guidelines on the management of venous leg ulcers: The point of view of the International Union of Phlebology. Int. Angiol. 2015, 34, 202–218. [Google Scholar]
  47. Franks, P.J.; Barker, J.; Collier, M.; Gethin, G.; Haesler, E.; Jawien, A.; Laeuchli, S.; Mosti, G.; Probst, S.; Weller, C. Management of Patients With Venous Leg Ulcers: Challenges and Current Best Practice. J. Wound Care 2016, 25 (Suppl. 6), S1–S67. [Google Scholar] [CrossRef] [PubMed]
  48. Berszakiewicz, A.; Sieroń, A.; Krasiński, Z.; Cholewka, A.; Stanek, A. Compression therapy in venous diseases: Current forms of compression materials and techniques. Postepy Dermatol. Alergol. 2020, 37, 836–841. [Google Scholar] [CrossRef] [PubMed]
  49. Partsch, H. The static stiffness index: A simple method to assess the elastic property of compression material in vivo. Dermatol. Surg. 2005, 31, 625–630. [Google Scholar] [CrossRef]
  50. Partsch, H. The use of pressure change on standing as a surrogate measure of the stiffness of a compression bandage. Eur. J. Vasc. Endovasc. Surg. 2005, 30, 415–421. [Google Scholar] [CrossRef]
  51. Partsch, B.; Partsch, H. Calf compression pressure required to achieve venous closure from supine to standing positions. J. Vasc. Surg. 2005, 42, 734–738. [Google Scholar] [CrossRef]
  52. Partsch, H.; Mosti, G.; Mosti, F. Narrowing of leg veins under compression demonstrated by magnetic resonance imaging (MRI). Int. Angiol. 2010, 29, 408–410. [Google Scholar] [PubMed]
  53. Partsch, H.; Menzinger, G.; Mostbeck, A. Inelastic leg compression is more effective to reduce deep venous refluxes than elastic bandages. Dermatol. Surg. 1999, 25, 695–700. [Google Scholar] [CrossRef] [PubMed]
  54. Mosti, G.; Partsch, H. Duplex scanning to evaluate the effect of compression on venous reflux. Int. Angiol. 2010, 29, 416–420. [Google Scholar]
  55. Mosti, G.; Mattaliano, V.; Partsch, H. Inelastic compression increases venous ejection fraction more than elastic bandages in patients with superficial venous reflux. Phlebology 2008, 23, 287–294. [Google Scholar] [CrossRef] [PubMed]
  56. Mosti, G.; Partsch, H. Measuring venous pumping function by strain-gauge plethysmography. Int. Angiol. 2010, 29, 421–425. [Google Scholar]
  57. Partsch, B.; Mayer, W.; Partsch, H. Improvement of ambulatory venous hypertension by narrowing of the femoral vein in congenital absence of venous valves. Phlebology 1992, 7, 101–104. [Google Scholar] [CrossRef]
  58. Protz, K.; Heyer, K.; Dörler, M.; Stücker, M.; Hampel-Kalthoff, C.; Augustin, M. Compression therapy: Scientific background and practical applications. J. Dtsch. Dermatol. Ges. 2014, 12, 794–801. [Google Scholar] [CrossRef]
  59. Berszakiewicz, A.; Sieroń, A.; Krasiński, Z.; Cholewka, A.; Stanek, A. Compression therapy in venous diseases: Physical assumptions and clinical effects. Postepy Dermatol. Alergol. 2020, 37, 842–847. [Google Scholar] [CrossRef] [PubMed]
  60. Keller, A.; Müller, M.L.; Calow, T.; Kern, I.K.; Schumann, H. Bandage pressure measurement and training: Simple interventions to improve efficacy in compression bandaging. Int. Wound J. 2009, 6, 324–330. [Google Scholar] [CrossRef] [PubMed]
  61. Reynolds, S. The impact of a bandage training programme. J. Wound Care 1999, 8, 55–60. [Google Scholar] [CrossRef] [PubMed]
  62. Nelson, E.A.; Ruckley, C.V.; Barbenel, J.C. Improvements in bandaging technique following training. J. Wound Care 1995, 4, 181–184. [Google Scholar] [CrossRef]
  63. Zarchi, K.; Jemec, G.B. Delivery of compression therapy for venous leg ulcers. JAMA Dermatol. 2014, 150, 730–736. [Google Scholar] [CrossRef]
  64. Partsch, H. Reliable self-application of short stretch leg compression: Pressure measurements under self-applied, adjustable compression wraps. Phlebology 2019, 34, 208–213. [Google Scholar] [CrossRef]
  65. Mosti, G.; Partsch, H. Self-management by firm, non-elastic adjustable compression wrap device. Veins Lymphat. 2017, 6, 7003. [Google Scholar] [CrossRef]
  66. Damstra, R.J.; Partsch, H. Prospective, randomized, controlled trial comparing the effectiveness of adjustable compression Velcro wraps versus inelastic multicomponent compression bandages in the initial treatment of leg lymphedema. J. Vasc. Surg. Venous Lymphat. Disord. 2013, 1, 13–19. [Google Scholar] [CrossRef] [PubMed]
  67. Milic, D.J.; Zivic, S.S.; Bogdanovic, D.C.; Jovanovic, M.M.; Jankovic, R.J.; Milosevic, Z.D.; Stamenkovic, D.M.; Trenkic, M.S. The influence of different sub-bandage pressure values on venous leg ulcers healing when treated with compression therapy. J. Vasc. Surg. 2010, 51, 655–661. [Google Scholar] [CrossRef]
  68. Mosti, G.; Crespi, A.; Mattaliano, V. Comparison between a New, Two-component Compression System with Zinc Paste Bandages for Leg Ulcer Healing: A Prospective, Multicenter, Randomized, Controlled Trial Monitoring Sub-bandage Pressures. Wounds 2011, 23, 126–134. [Google Scholar] [PubMed]
  69. Franks, P.J.; Moody, M.; Moffatt, C.J.; Martin, R.; Blewett, R.; Seymour, E.; Hildreth, A.; Hourican, C.; Collins, J.; Heron, A.; et al. Randomized trial of cohesive short-stretch versus four-layer bandaging in the management of venous ulceration. Wound Repair Regen. 2004, 12, 157–162. [Google Scholar] [CrossRef] [PubMed]
  70. Moffatt, C.J.; McCullagh, L.; O’Connor, T.; Doherty, D.C.; Hourican, C.; Stevens, J.; Mole, T.; Franks, P.J. Randomized trial of four-layer and two-layer bandage systems in the management of chronic venous ulceration. Wound Repair Regen. 2003, 11, 166–171. [Google Scholar] [CrossRef]
  71. Fletcher, A.; Cullum, N.; Sheldon, T.A. A systematic review of compression treatment for venous leg ulcers. BMJ 1997, 315, 576–580. [Google Scholar] [CrossRef]
  72. Callam, M.J.; Harper, D.R.; Dale, J.J.; Brown, D.; Gibson, B.; Prescott, R.J.; Ruckley, C.V. Lothian and Forth Valley Leg Ulcer Healing Trial Part 1. Elastic versus non-elastic bandaging in the treatment of chronic leg ulceration. Phlebology 1992, 7, 136–141. [Google Scholar] [CrossRef]
  73. Duby, T.; Hoffman, D.; Cameron, J.; Doblhoff-Brown, D.; Cherry, G.; Ryan, T. A randomized trial in the treatment of venous leg ulcers comparing short stretch bandages, four-layer bandage system, and a long stretch-paste bandage system. Wounds 1993, 5, 276–279. [Google Scholar]
  74. Ukat, A.; Konig, M.; Vanscheidt, W.; Münter, K.C. Short-stretch versus multilayer compression for venous leg ulcers: A comparison of healing rates. J. Wound Care 2003, 12, 139–143. [Google Scholar] [CrossRef] [PubMed]
  75. Scriven, J.M.; Taylor, L.E.; Wood, A.J.; Bell, P.R.; Naylor, A.R.; London, N.J. A prospective randomised trial of four-layer versus short stretch compression bandages for the treatment of venous leg ulcers. Ann. R. Coll. Surg. Engl. 1998, 80, 215–220. [Google Scholar] [PubMed]
  76. Nelson, E.A.; Iglesias, C.P.; Cullum, N.; Torgerson, D.J.; VenUS I collaborators. Randomized clinical trial of four-layer and short-stretch compression bandages for venous leg ulcers (VenUS I). Br. J. Surg. 2004, 91, 1292–1299. [Google Scholar] [CrossRef] [PubMed]
  77. Mariani, F.; Mattaliano, V.; Mosti, G.; Gasbarro, V.; Bucalossi, M.; Blättler, W. The treatment of venous leg ulcers with a specifically designed compression stocking kit. Phlebologie 2008, 37, 191–197. [Google Scholar] [CrossRef]
  78. Junger, M.; Partsch, H.; Ramelet, A.A.; Zuccarelli, F. Efficacy of a ready-made tubular compression device versus short stretch bandages in the treatment of venous leg ulcers. Wounds 2004, 16, 313–320. [Google Scholar]
  79. Jünger, M.; Wollina, U.; Kohnen, R.; Rabe, E. Efficacy and tolerability of an ulcer compression stocking for therapy of chronic venous ulcer compared with a below-knee compression bandage: Results from a prospective, randomized, multicentre trial. Curr. Med. Res. Opin. 2004, 20, 1613–1623. [Google Scholar] [CrossRef]
  80. Horakova, M.A.; Partsch, H. Compression stockings in treatment of lower leg venous ulcer. Wien. Med. Wochenschr. 1994, 144, 242–249. [Google Scholar]
  81. Brizzio, E.; Amsler, F.; Lun, B.; Blättler, W. Comparison of low-strength compression stockings with bandages for the treatment of recalcitrant venous ulcers. J. Vasc. Surg. 2010, 51, 410–416. [Google Scholar] [CrossRef]
  82. Amsler, F.; Willenberg, T.; Blättler, W. In search of optimal compression therapy for venous leg ulcers: A meta-analysis of studies comparing diverse [corrected] bandages with specifically designed stockings. J. Vasc. Surg. 2009, 50, 668–674. [Google Scholar] [CrossRef]
  83. Mosti, G. Elastic stockings versus inelastic bandages for ulcer healing: A fair comparison? Phlebology 2012, 27, 1–4. [Google Scholar] [CrossRef] [PubMed]
  84. Partsch, H.; Mosti, G. Comparison of three portable instruments to measure compression pressure. Int. Angiol. 2010, 29, 426–430. [Google Scholar] [PubMed]
  85. Mosti, G.; Mattaliano, V.; Partsch, H. Influence of different materials in multicomponent bandages on pressure and stiffness of the final bandage. Dermatol. Surg. 2008, 34, 631–639. [Google Scholar] [CrossRef]
  86. Spence, R.K.; Cahall, E. Inelastic versus elastic leg compression in chronic venous insufficiency: A comparison of limb size and venous hemodynamics. J. Vasc. Surg. 1996, 24, 783–787. [Google Scholar] [CrossRef]
  87. Murthy, G.; Ballard, R.E.; Breit, G.A.; Watenpaugh, D.E.; Hargens, A.R. Intramuscular pressures beneath elastic and inelastic leggings. Ann. Vasc. Surg. 1994, 8, 543–548. [Google Scholar] [CrossRef]
  88. DePalma, R.G.; Kowallek, D.; Spence, R.K.; Caprini, J.A.; Nehler, M.R.; Jensen, J.; Goldman, M.P. Comparison of Costs and Healing Rates of Two Forms of Compression in Treating Venous Ulcers. Vasc. Surg. 1999, 33, 683–690. [Google Scholar] [CrossRef]
  89. Blecken, S.R.; Villavicencio, J.L.; Kao, T.C. Comparison of elastic versus nonelastic compression in bilateral venous ulcers: A randomized trial. J. Vasc. Surg. 2005, 42, 1150–1155. [Google Scholar] [CrossRef]
  90. Mosti, G.; Mancini, S.; Bruni, S.; Serantoni, S.; Gazzabin, L.; Bucalossi, M.; Polignano, R.; Mariani, F.; Luca, B.; Partsch, H.; et al. Adjustable compression wrap devices are cheaper and more effective than inelastic bandages for venous leg ulcer healing. A Multicentric Italian Randomized Clinical Experience. Phlebology 2020, 35, 124–133. [Google Scholar] [CrossRef]
  91. Jawien, A. The influence of environmental factors in chronic venous insufficiency. Angiology 2003, 54 (Suppl. 1), S19–S31. [Google Scholar] [CrossRef] [PubMed]
  92. Dix, F.P.; Reilly, B.; David, M.C.; Simon, D.; Dowding, E.; Ivers, L. Effect of leg elevation on healing, venous velocity, and ambulatory venous pressure in venous ulceration. Phlebology 2005, 20, 87–94. [Google Scholar] [CrossRef]
  93. Simon, D.A.; Dix, F.P.; McCollum, C.N. Management of venous leg ulcers. BMJ 2004, 328, 1358–1362. [Google Scholar] [CrossRef]
  94. Wittens, C.; Davies, A.H.; Bækgaard, N.; Broholm, R.; Cavezzi, A.; Chastanet, S.; de Wolf, M.; Eggen, C.; Giannoukas, A.; Gohel, M.; et al. Editor’s Choice—Management of Chronic Venous Disease: Clinical Practice Guidelines of the European Society for Vascular Surgery (ESVS). Eur. J. Vasc. Endovasc. Surg. 2015, 49, 678–737. [Google Scholar] [CrossRef]
  95. Brown, A. Life-style advice and self-care strategies for venous leg ulcer patients: What is the evidence? J. Wound Care 2012, 21, 342–350. [Google Scholar] [CrossRef]
  96. Kostas, T.I.; Ioannou, C.V.; Drygiannakis, I.; Georgakarakos, E.; Kounos, C.; Tsetis, D.; Katsamouris, A.N. Chronic venous disease progression and modification of predisposing factors. J. Vasc. Surg. 2010, 51, 900–907. [Google Scholar] [CrossRef]
  97. Roaldsen, K.S.; Rollman, O.; Torebjörk, E.; Olsson, E.; Stanghelle, J.K. Functional ability in female leg ulcer patients--a challenge for physiotherapy. Physiother. Res. Int. 2006, 11, 191–203. [Google Scholar] [CrossRef]
  98. Mutlak, O.; Aslam, M.; Standfield, N. The influence of exercise on ulcer healing in patients with chronic venous insufficiency. Int. Angiol. 2018, 37, 160–168. [Google Scholar] [CrossRef]
  99. Qiu, Y.; Osadnik, C.R.; Team, V.; Weller, C.D. Effects of physical activity as an adjunct treatment on healing outcomes and recurrence of venous leg ulcers: A scoping review. Wound Repair Regen. 2022, 30, 172–185. [Google Scholar] [CrossRef] [PubMed]
  100. O’Brien, J.; Finlayson, K.; Kerr, G.; Edwards, H. Evaluating the effectiveness of a self-management exercise intervention on wound healing, functional ability and health-related quality of life outcomes in adults with venous leg ulcers: A randomised controlled trial. Int. Wound J. 2017, 14, 130–137. [Google Scholar] [CrossRef]
  101. Zhang, Q.; Lu, L.; Song, J.L.; Wang, L. Effects of exercise in treating patients with venous leg ulcers: A systematic review and meta-analysis. Int. Wound J. 2023, 20, 1776–1783. [Google Scholar] [CrossRef]
  102. Palfreyman, S.J.; Nelson, E.A.; Lochiel, R.; Michaels, J.A. Dressings for healing venous leg ulcers. Cochrane Database Syst. Rev. 2006, 3, CD001103. [Google Scholar] [CrossRef]
  103. Norman, G.; Westby, M.J.; Rithalia, A.D.; Stubbs, N.; Soares, M.O.; Dumville, J.C. Dressings and topical agents for treating venous leg ulcers. Cochrane Database Syst. Rev. 2018, 6, CD012583. [Google Scholar] [CrossRef] [PubMed]
  104. Callam, M.J.; Harper, D.R.; Dale, J.J.; Brown, D.; Gibson, B.; Prescott, R.J.; Ruckley, C.V. Lothian and Forth valley leg ulcer healing trial: 2 Knitted viscose dressing versus a hydrocellular dressing in the treatment of chronic leg ulceration. Phlebology 1992, 7, 142–145. [Google Scholar] [CrossRef]
  105. Skog, E.; Arnesjö, B.; Troëng, T.; Gjöres, J.E.; Bergljung, L.; Gundersen, J.; Hallböök, T.; Hessman, Y.; Hillström, L.; Månsson, T.; et al. A randomized trial comparing cadexomer iodine and standard treatment in the out-patient management of chronic venous ulcers. Br. J. Dermatol. 1983, 109, 77–83. [Google Scholar] [CrossRef] [PubMed]
  106. Smith, J.M.; Dore, C.J.; Charlett, A.; Lewis, J.D. A randomized trial of biofilm dressing for venous leg ulcers. Phlebology 1992, 7, 108–113. [Google Scholar] [CrossRef]
  107. Lindholm, C. Results of cost-efficacy aspects in wound care trials. In Proceedings of the Fourth Annual Meeting of the European Tissue Repair Society Academic Centre—John Radcliffe Hospital, Oxford, UK, 25–28 August 1994; p. 199. [Google Scholar] [CrossRef]
  108. Meridith, K.; Gray, E. Dressed to heal. J. District Nurs. 1988, 7, 8–10. [Google Scholar]
  109. Nair, H.; Venkateshwaran, N.; Seetharaman, S.S.; Deng, W.; Uthaipaisanwong, A.; Galea, E. Benefits of sucrose octasulfate (TLC-NOSF) dressings in the treatment of chronic wounds: A systematic review. J. Wound Care 2021, 30 (Suppl. 4), S42–S52. [Google Scholar] [CrossRef] [PubMed]
  110. Nicolaides, A.N. The Benefits of Micronized Purified Flavonoid Fraction (MPFF) Throughout the Progression of Chronic Venous Disease. Adv. Ther. 2020, 37 (Suppl. 1), 1–5. [Google Scholar] [CrossRef]
  111. Mansilha, A.; Sousa, J. Pathophysiological Mechanisms of Chronic Venous Disease and Implications for Venoactive Drug Therapy. Int. J. Mol. Sci. 2018, 19, 1669. [Google Scholar] [CrossRef]
  112. Coleridge-Smith, P.; Lok, C.; Ramelet, A.A. Venous leg ulcer: A meta-analysis of adjunctive therapy with micronized purified flavonoid fraction. Eur. J. Vasc. Endovasc. Surg. 2005, 30, 198–208. [Google Scholar] [CrossRef] [PubMed]
  113. Guilhou, J.J.; Février, F.; Debure, C.; Dubeaux, D.; Gillet-Terver, M.N.; Guillot, B.; Levesque, H.; Marzin, L.; Mignot, J.; Ouvry, P.; et al. Benefit of a 2-month treatment with a micronized, purified flavonoidic fraction on venous ulcer healing. A randomized, double-blind, controlled versus placebo trial. Int. J. Microcirc. Clin. Exp. 1997, 17 (Suppl. 1), 21–26. [Google Scholar] [CrossRef]
  114. Glinski, W.; Chodynicka, B.; Roszkiewicz, J.; Bogdanowski, T.; Lecewicz-Torun, B.; Kaszuba, A.; Bowszyc, J.; Nowak, A.; Wnorowski, J.; Wasik, F.; et al. Efficacia della frazione flavonoica purificata micronizzata (FFPM) nell’aumentare la guarigione della ulcere agli arti inferiori. Studio multicentrico in aperto, controllato e randomizato [Effectiveness of a micronized purified flavonoid fraction (MPFF) in the healing process of lower limb ulcers. An open multicentre study, controlled and randomized]. Minerva Cardioangiol. 2001, 49, 107–114. [Google Scholar]
  115. Smith, P.C. Daflon 500 mg and venous leg ulcer: New results from a meta-analysis. Angiology 2005, 56 (Suppl. 1), S33–S39. [Google Scholar] [CrossRef] [PubMed]
  116. Guilhou, J.J.; Dereure, O.; Marzin, L.; Ouvry, P.; Zuccarelli, F.; Debure, C.; Van Landuyt, H.; Gillet-Terver, M.N.; Guillot, B.; Levesque, H.; et al. Efficacy of Daflon 500 mg in venous leg ulcer healing: A double-blind, randomized, controlled versus placebo trial in 107 patients. Angiology 1997, 48, 77–85. [Google Scholar] [CrossRef]
  117. Coccheri, S.; Scondotto, G.; Agnelli, G.; Aloisi, D.; Palazzini, E.; Zamboni, V. Venous arm of the SUAVIS (Sulodexide Arterial Venous Italian Study) Group. Randomised, double blind, multicentre, placebo controlled study of sulodexide in the treatment of venous leg ulcers. Thromb. Haemost. 2002, 87, 947–952. [Google Scholar] [PubMed]
  118. González Ochoa, A. Sulodexide and phlebotonics in the treatment of venous ulcer. Int. Angiol. 2017, 36, 82–87. [Google Scholar] [CrossRef]
  119. Pompilio, G.; Nicolaides, A.; Kakkos, S.K.; Integlia, D. Systematic literature review and network Meta-analysis of sulodexide and other drugs in chronic venous disease. Phlebology 2021, 36, 695–709. [Google Scholar] [CrossRef]
  120. Wu, B.; Lu, J.; Yang, M.; Xu, T. Sulodexide for treating venous leg ulcers. Cochrane Database Syst. Rev. 2016, 2016, CD010694. [Google Scholar] [CrossRef]
  121. Sullivan, G.W.; Carper, H.T.; Novick WJJr Mandell, G.L. Inhibition of the inflammatory action of interleukin-1 and tumor necrosis factor (alpha) on neutrophil function by pentoxifylline. Infect. Immun. 1988, 56, 1722–1729. [Google Scholar] [CrossRef]
  122. Broderick, C.; Forster, R.; Abdel-Hadi, M.; Salhiyyah, K. Pentoxifylline for intermittent claudication. Cochrane Database Syst. Rev. 2020, 10, CD005262. [Google Scholar] [CrossRef]
  123. Ahmadi, M.; Khalili, H. Potential benefits of pentoxifylline on wound healing. Expert. Rev. Clin. Pharmacol. 2016, 9, 129–142. [Google Scholar] [CrossRef] [PubMed]
  124. Belcaro, G.; Cesarone, M.R.; Nicolaides, A.N.; De Sanctis, M.T.; Incandela, L.; Geroulakos, G. Treatment of venous ulcers with pentoxifylline: A 6-month randomized, double-blind, placebo controlled trial. Angiology 2002, 53 (Suppl. 1), S45–S47. [Google Scholar]
  125. Falanga, V.; Fujitani, R.M.; Diaz, C.; Hunter, G.; Jorizzo, J.; Lawrence, P.F.; Lee, B.Y.; Menzoian, J.O.; Tretbar, L.L.; Holloway, G.A.; et al. Systemic treatment of venous leg ulcers with high doses of pentoxifylline: Efficacy in a randomized, placebo-controlled trial. Wound Repair Regen. 1999, 7, 208–213. [Google Scholar] [CrossRef]
  126. Nelson, E.A.; Prescott, R.J.; Harper, D.R.; Gibson, B.; Brown, D.; Ruckley, C.V. A factorial, randomized trial of pentoxifylline or placebo, four-layer or single-layer compression, and knitted viscose or hydrocolloid dressings for venous ulcers. J. Vasc. Surg. 2007, 45, 134–141. [Google Scholar] [CrossRef] [PubMed]
  127. Jull, A.B.; Arroll, B.; Parag, V.; Waters, J. Pentoxifylline for treating venous leg ulcers. Cochrane Database Syst. Rev. 2012, 12, CD001733. [Google Scholar] [CrossRef] [PubMed]
  128. Sun, S.Y.; Li, Y.; Gao, Y.Y.; Ran, X.W. Efficacy and Safety of Pentoxifylline for Venous Leg Ulcers: An Updated Meta-Analysis. Int. J. Low. Extrem. Wounds. 2021. [Google Scholar] [CrossRef]
  129. van den Bremer, J.; Moll, F.L. Historical overview of varicose vein surgery. Ann. Vasc. Surg. 2010, 24, 426–432. [Google Scholar] [CrossRef]
  130. Trendelenburg, F. Uber die unterbindung der vena faphena magna bei unterschendelzaricen. Berl. Klin. Chir. 1890, 7, 195. [Google Scholar]
  131. Winterborn, R.J.; Earnshaw, J.J. Crossectomy and great saphenous vein stripping. J. Cardiovasc. Surg. 2006, 47, 19–33. [Google Scholar]
  132. Scheltinga, M.R.; Wijburg, E.R.; Keulers, B.J.; de Kroon, K.E. Conventional versus invaginated stripping of the great saphenous vein: A randomized, double-blind, controlled clinical trial. World J. Surg. 2007, 31, 2236–2242. [Google Scholar] [CrossRef]
  133. Lacroix, H.; Nevelsteen, A.; Suy, R. Invaginating versus classic stripping of the long saphenous vein. A randomized prospective study. Acta Chir. Belg. 1999, 99, 22–25. [Google Scholar] [CrossRef]
  134. Goren, G.; Yellin, A.E. Minimally invasive surgery for primary varicose veins: Limited invaginal axial stripping and tributary (hook) stab avulsion. Ann. Vasc. Surg. 1995, 9, 401–414. [Google Scholar] [CrossRef]
  135. Sam, R.C.; Silverman, S.H.; Bradbury, A.W. Nerve injuries and varicose vein surgery. Eur. J. Vasc. Endovasc. Surg. 2004, 27, 113–120. [Google Scholar] [CrossRef]
  136. Subramonia, S.; Lees, T. Sensory abnormalities and bruising after great saphenous vein stripping: Impact on short-term quality of life. J. Vasc. Surg. 2005, 42, 510–514. [Google Scholar] [CrossRef]
  137. Morrison, C.; Dalsing, M.C. Signs and symptoms of saphenous nerve injury after greater saphenous vein stripping: Preva lence, severity, and relevance for modern practice. J. Vasc. Surg. 2003, 38, 886–890. [Google Scholar] [CrossRef]
  138. Oesch, A. PIN-stripping: A novel method of atraumatic stripping. Phlebology 1993, 8, 171–173. [Google Scholar] [CrossRef]
  139. Durkin, M.P.; Turton, E.P.; Scott, D.J.; Berridge, D.C. A prospective randomised trial of PIN versus conventional stripping in varicose vein surgery. Ann. R. Coll. Surg. Eng. 1999, 81, 171–174. [Google Scholar]
  140. Garde, C. Cryosurgery of varicose veins. J. Dermatol. Surg. Oncol. 1994, 20, 56–58. [Google Scholar] [CrossRef]
  141. Kostas, T.; Ioannou, C.; Touloupakis, E.; Daskalaki, E.; Giannoukas, A.; Tsetis, D.; Katsamouris, A. Recurrent varicose veins after surgery: A new appraisal of a common and complex problem in vascular surgery. Eur. J. Vasc. Endovasc. Surg. 2004, 27, 275–282. [Google Scholar] [CrossRef]
  142. Hartmann, K. Endovenous (minimally invasive) procedures for treatment of varicose veins: The gentle and effective alternative to high ligation and stripping operations. Hautarzt 2020, 71 (Suppl. 2), 67–73. [Google Scholar] [CrossRef] [PubMed]
  143. Almeida, J.I.; Kaufman, J.; Göckeritz, O.; Chopra, P.; Evans, M.T.; Hoheim, D.F.; Makhoul, R.G.; Richards, T.; Wenzel, C.; Raines, J.K. Radiofrequency endovenous ClosureFAST versus laser ablation for the treatment of great saphenous reflux: A multicenter, single-blinded, randomized study (RECOVERY study). J. Vasc. Interv. Radiol. 2009, 20, 752–759. [Google Scholar] [CrossRef] [PubMed]
  144. Sroka, R.; Weick, K.; Sadeghi-Azandaryani, M.; Steckmeier, B.; Schmedt, C.G. Endovenous laser therapy—application studies and latest investigations. J. Biophotonics 2010, 3, 269–276. [Google Scholar] [CrossRef]
  145. Sroka, R.; Schaur, P.; Rühm, A.; Pongratz, T.; Schmedt, C.G. Ex-Vivo Investigations of Innovative Fibres for Use in Endoluminal Vein Treatment Procedures. 57; DGP: Bamberg, Germany, 2015. [Google Scholar]
  146. Hartmann, K.; Stenger, D.; Hartmann, M.; Rafi-Stenger, L. Endochirurgie versus offene Chirugie der Varikose. Vers. Einer Wertung. Hautarzt 2017, 68, 603–613. [Google Scholar] [CrossRef] [PubMed]
  147. Mueller, R.L.; Raines, J.K. ClariVein mechanochemical ablation: Background and procedural details. Vasc. Endovasc. Surg. 2013, 47, 195–206. [Google Scholar] [CrossRef] [PubMed]
  148. Terlecki, P.; Boryga, M.; Kołodziej, P.; Gołacki, K.; Stropek, Z.; Janczak, D.; Antkiewicz, M.; Zubilewicz, T. Mechanical Characteristics of the Flebogrif System—The New System of Mechano-Chemical Endovenous Ablation. Materials 2022, 15, 2599. [Google Scholar] [CrossRef]
  149. Gibson, K.; Ferris, B. Cyanoacrylate closure of incompetent great, small and accessory saphenous veins without the use of post-procedure compression: Initial outcomes of a post-market evaluation of the VenaSeal System (the WAVES Study). Vascular 2017, 25, 149–156. [Google Scholar] [CrossRef]
  150. Muller, R. Treatment of varicose veins by ambulatory phlebectomy. Phlebologie 1966, 19, 277–279. [Google Scholar] [PubMed]
  151. Fratila, A.; Rabe, E.; Biltz, H.; Kreysel, H.W. Stellenwert der perkutanen mikrochirurgischen Phlebextraktion nach Varady in der Varizenchirurgie [Value of Varady percutaneous microsurgical phlebologic extraction of varicose veins]. Z. Hautkr. 1990, 65, 487–491. [Google Scholar]
  152. Juan, J.; Escribano, J.M.; Criado, E.; Fontcuberta, F. Haemodynamic surgery for varicose veins: Surgical strategy. Phlebology 2005, 20, 2–13. [Google Scholar] [CrossRef]
  153. Bellmunt-Montoya, S.; Escribano, J.M.; Pantoja Bustillos, P.E.; Tello-Díaz, C.; Martinez-Zapata, M.J. CHIVA method for the treatment of chronic venous insufficiency. Cochrane Database Syst. Rev. 2021, 9, CD009648. [Google Scholar] [CrossRef]
  154. Solıs, J.V.; Ribe’, L.; Portero, J.L.; Rio, J. Stripping saphenectomy, CHIVA and Laser ablation for the treatment of the saphenous vein insufficiency. Amb. Surg. 2009, 15, 11–14. [Google Scholar]
  155. Onida, S.; Davies, A.H. CHIVA, ASVAL and related techniques—Concepts and evidence. Phlebology 2015, 30 (Suppl. 2), 42–45. [Google Scholar] [CrossRef]
  156. Pittaluga, P.; Chastanet, S.; Rea, B.; Barbe, R. Midterm results of the surgical treatment of varices by phlebectomy with conservation of a refluxing saphenous vein. K. Vasc. Surg. 2009, 50, 107–118. [Google Scholar] [CrossRef]
  157. Mavor, G.E.; Galloway, J.M. Iliofemoral venous thrombosis. Pathological considerations and surgical management. Br. J. Surg. 1969, 56, 45–59. [Google Scholar] [CrossRef]
  158. Douketis, J.D.; Crowther, M.A.; Foster, G.A.; Ginsberg, J.S. Does the location of thrombosis determine the risk of disease recurrence in patients with proximal deep vein thrombosis? Am. J. Med. 2001, 110, 515–519. [Google Scholar] [CrossRef]
  159. May, R.; Thurner, J. The cause of the predominantly sinistral occurrence of thrombosis of the pelvic veins. Angiology 1957, 8, 419–427. [Google Scholar] [CrossRef]
  160. Thijs, W.; Rabe, K.F.; Rosendaal, F.R.; Middeldorp, S. Predominance of left-sided deep vein thrombosis and body weight. J. Thromb. Haemost. 2010, 8, 2083–2084. [Google Scholar] [CrossRef]
  161. Raju, S.; Hardy, J.D. Technical options in venous valve reconstruction. Am. J. Surg. 1997, 173, 301–307. [Google Scholar] [CrossRef]
  162. Kistner, R. Surgical repair of a venous valve. Straub. Clin. Proc. 1968, 34, 41–43. [Google Scholar]
  163. Ajay, K. Khanna.; Shivanshu, Singh. Postthrombotic Syndrome: Surgical Possibilities. Thrombosis 2012, 2012, 520604. [Google Scholar] [CrossRef]
  164. Palma, E.C.; Esperon, R. Vein transplants and grafts in the surgical treatment of post phlebitic syndrome. J. Cardiovasc. Surg. 1960, 1, 94–107. [Google Scholar]
  165. Dale, W.A.; Hams, J. Crossover grafts for iliac and femoral venous occlusion. Ann. Surg. 1968, 168, 319–329. [Google Scholar] [CrossRef]
  166. Edwards, W.S. A-V fistula after venous reconstruction. Ann. Surg. 1982, 196, 669–671. [Google Scholar] [CrossRef]
  167. Kölbel, T.; Gottsäter, A.; Kühme, T.; Lindh, M.; Ivancev, K. Endovascular Treatment of Venous Occlusive Disease. Ann. Vasc. Dis. 2008, 1, 91–101. [Google Scholar] [CrossRef] [PubMed]
  168. Neglén, P.; Hollis, K.C.; Olivier, J.; Raju, S. Stenting of the venous outflow in chronic venous disease: Long-term stent-related outcome, clinical, and hemodynamic result. J. Vasc. Surg. 2007, 46, 979–990. [Google Scholar] [CrossRef]
  169. Raju, S. Best management options for chronic iliac vein stenosis and occlusion. J. Vasc. Surg. 2013, 57, 1163–1169. [Google Scholar] [CrossRef]
  170. Razavi, M.K.; Jaff, M.R.; Miller, L.E. Safety and Effectiveness of Stent Placement for Iliofemoral Venous Outflow Obstruction: Systematic Review and Meta-Analysis. Circ. Cardiovasc. Interv. 2015, 8, e002772. [Google Scholar] [CrossRef] [PubMed]
  171. Marston, W.A.; Chinubhai, A.; Kao, S.; Kalbaugh, C.; Kouri, A. In vivo evaluation of safety and performance of a nitinol venous stent in an ovine iliac venous model. J. Vasc. Surg. Venous Lymphat. Disord. 2016, 4, 73–79. [Google Scholar] [CrossRef]
  172. Powell, T.; Raju, S.; Jayaraj, A. Comparison between a dedicated venous stent and standard composite Wallstent–Z stent approach to iliofemoral venous stenting: Intermediate-term outcomes. J. Vasc. Surg. Venous Lymphat. Disord. 2023, 11, 82–90. [Google Scholar] [CrossRef]
  173. Aurshina, A.; Chait, J.; Kibrik, P.; Ostrozhynskyy, Y.; Rajaee, S.; Marks, N.; Ascher, E. Efficacy of balloon venoplasty alone in the correction of nonthrombotic iliac vein lesions. J. Vasc. Surg. Venous Lymphat. Disord. 2019, 7, 665–669. [Google Scholar] [CrossRef]
  174. Schultz, G.S.; Sibbald, R.G.; Falanga, V.; Ayello, E.A.; Dowsett, C.; Harding, K.; Romanelli, M.; Stacey, M.C.; Teot, L.; Vanscheidt, W. Wound bed preparation: A systematic approach to wound management. Wound Repair Regen. 2003, 11, S1–S28. [Google Scholar] [CrossRef] [PubMed]
  175. Gethin, G.; Cowman, S.; Kolbach, D.N. Debridement for venous leg ulcers. Cochrane Database Syst. Rev. 2015, 2015, CD008599. [Google Scholar] [CrossRef]
  176. Serra, R.; Rizzuto, A.; Rossi, A.; Perri, P.; Barbetta, A.; Abdalla, K.; Caroleo, S.; Longo, C.; Amantea, B.; Sammarco, G.; et al. Skin grafting for the treatment of chronic leg ulcers—A systematic review in evidence-based medicine. Int. Wound J. 2017, 14, 149–157. [Google Scholar] [CrossRef]
  177. Kirsner, R.S.; Eaglstein, W.H.; Kerdel, F.A. Split-thickness skin grafting for lower extremity ulcerations. Dermatol. Surg. 1997, 23, 85–91. [Google Scholar] [CrossRef] [PubMed]
  178. Hedayati, N.; Carson, J.G.; Chi, Y.W.; Link, D. Management of mixed arterial venous lower extremity ulceration: A review. Vasc. Med. 2015, 20, 479–486. [Google Scholar] [CrossRef] [PubMed]
  179. Ammermann, F.; Meinel, F.G.; Beller, E.; Busse, A.; Streckenbach, F.; Teichert, C.; Weinrich, M.; Neumann, A.; Weber, M.A.; Heller, T. Concomitant chronic venous insufficiency in patients with peripheral artery disease: Insights from MR angiography. Eur. Radiol. 2020, 30, 3908–3914. [Google Scholar] [CrossRef] [PubMed]
  180. Körber, A.; Klode, J.; Al-Benna, S.; Wax, C.; Schadendorf, D.; Steinstraesser, L.; Dissemond, J. Etiology of chronic leg ulcers in 31,619 patients in Germany analyzed by an expert survey. J. Dtsch. Dermatol. Ges. 2011, 9, 116–121. [Google Scholar] [CrossRef] [PubMed]
  181. Grey, J.E.; Harding, K.G.; Enoch, S. Venous and arterial leg ulcers. BMJ 2006, 332, 347–350. [Google Scholar] [CrossRef] [PubMed]
  182. Nag, F.; De, A.; Hazra, A.; Chatterjee, G.; Ghosh, A.; Surana, T.V. Chronic venous ulceration of leg associated with peripheral arterial disease: An underappreciated entity in developing country. Int. Wound J. 2014, 11, 546–549. [Google Scholar] [CrossRef] [PubMed]
  183. Lazarides, M.K.; Giannoukas, A.D. The role of hemodynamic measurements in the management of venous and ischemic ulcers. Int. J. Low. Extrem. Wounds 2007, 6, 254–261. [Google Scholar] [CrossRef]
  184. Treiman, G.S.; Copland, S.; McNamara, R.M.; Yellin, A.E.; Schneider, P.A.; Treiman, R.L. Factors influencing ulcer healing in patients with combined arterial and venous insufficiency. J. Vasc. Surg. 2001, 33, 1158–1164. [Google Scholar] [CrossRef] [PubMed]
  185. Ghauri, A.S.; Nyamekye, I.; Grabs, A.J.; Farndon, J.R.; Poskitt, K.R. The diagnosis and management of mixed arterial/venous leg ulcers in community-based clinics. Eur. J. Vasc. Endovasc. Surg. 1998, 16, 350–355. [Google Scholar] [CrossRef]
  186. Humphreys, M.L.; Stewart, A.H.; Gohel, M.S.; Taylor, M.; Whyman, M.R.; Poskitt, K.R. Management of mixed arterial and venous leg ulcers. Br. J. Surg. 2007, 94, 1104–1107. [Google Scholar] [CrossRef]
  187. Georgopoulos, S.; Kouvelos, G.N.; Koutsoumpelis, A.; Bakoyiannis, C.; Lymperi, M.; Klonaris, C.; Tsigris, C. The effect of revascularization procedures on healing of mixed arterial and venous leg ulcers. Int. Angiol. 2013, 32, 368–374. [Google Scholar] [PubMed]
  188. Lantis, J.C., 2nd; Boone, D.; Lee, L.; Mendes, D.; Benvenisty, A.; Todd, G. The effect of percutaneous intervention on wound healing in patients with mixed arterial venous disease. Ann. Vasc. Surg. 2011, 25, 79–86. [Google Scholar] [CrossRef]
  189. Meulendijks, A.M.; de Vries, F.M.C.; van Dooren, A.A.; Schuurmans, M.J.; Neumann, H.A.M. A systematic review on risk factors in developing a first-time Venous Leg Ulcer. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1241–1248. [Google Scholar] [CrossRef] [PubMed]
  190. Dissemond, J.; Bültemann, A.; Gerber, V.; Jäger, B.; Münter, C.; Kröger, K. Definitionen für die Wundbehandlung. Hautarzt 2016, 67, 265–266. [Google Scholar] [CrossRef] [PubMed]
  191. Gloviczki, P.; Comerota, A.J.; Dalsing, M.C.; Eklof, B.G.; Gillespie, D.L.; Gloviczki, M.L.; Lohr, J.M.; McLafferty, R.B.; Meissner, M.H.; Murad, M.H.; et al. The care of patients with varicose veins and associated chronic venous diseases: Clinical practice guidelines of the Society for Vascular Surgery and the American Venous Forum. J. Vasc. Surg. 2011, 53 (Suppl. 5), 2S–48S. [Google Scholar] [CrossRef]
  192. Cronenwett, J.; Johnston, K.W. (Eds.) Rutherford’s Vascular Surgery, 7th ed.; Saunders Elsevier: London, UK, 2010; Volume 1–2, 2448p. [Google Scholar]
  193. Robertson, L.; Lee, A.J.; Gallagher, K.; Carmichael, S.J.; Evans, C.J.; McKinstry, B.H.; Fraser, S.C.; Allan, P.L.; Weller, D.; Ruckley, C.V.; et al. Risk factors for chronic ulceration in patients with varicose veins: A case control study. J. Vasc. Surg. 2009, 49, 1490–1498. [Google Scholar] [CrossRef]
  194. Johnston, S.; Finlayson, K.; Bui, U.; O’Donoghue, E.; Fletcher, B.; Parker, C.N. Risk factors for the recurrence of venous leg ulcers in adults: A systematic review protocol. J. Tissue Viability 2022, 31, 804–807. [Google Scholar] [CrossRef]
  195. Uhl, J.F.; Cornu-Thénard, A.; Carpentier, P.H.; Widmer, M.T.; Partsch, H.; Antignani, P.L. Clinical and hemodynamic significance of corona phlebectatica in chronic venous disorders. J. Vasc. Surg. 2005, 42, 1163–1168. [Google Scholar] [CrossRef]
  196. Lurie, F.; Passman, M.; Meisner, M.; Dalsing, M.; Masuda, E.; Welch, H.; Bush, R.L.; Blebea, J.; Carpentier, P.H.; De Maeseneer, M.; et al. The 2020 update of the CEAP classification system and reporting standards. J. Vasc. Surg. Venous Lymphat. Disord. 2020, 8, 342–352. [Google Scholar] [CrossRef]
  197. Willenberg, T.; Schumacher, A.; Amann-Vesti, B.; Jacomella, V.; Thalhammer, C.; Diehm, N.; Baumgartner, I.; Husmann, M. Impact of obesity on venous hemodynamics of the lower limbs. J. Vasc. Surg. 2010, 52, 664–668. [Google Scholar] [CrossRef] [PubMed]
  198. Meulendijks, A.M.; Franssen, W.M.A.; Schoonhoven, L.; Neumann, H.A.M. A scoping review on Chronic Venous Disease and the development of a Venous Leg Ulcer: The role of obesity and mobility. J. Tissue Viability 2020, 29, 190–196. [Google Scholar] [CrossRef]
  199. Finlayson, K.J.; Parker, C.N.; Miller, C.; Edwards, H.E.; Campbell, J. Decreased mobility, lack of social support, haemosiderosis and use of antidepressant medications may predict recurrent venous leg ulcers within 12 months of healing: A prospective longitudinal study. Phlebology 2022, 37, 206–215. [Google Scholar] [CrossRef] [PubMed]
  200. Scholl, L.; Dörler, M.; Stücker, M. Ulkus bei Adipositas-assoziierter chronischer Veneninsuffizienz. Hautarzt 2017, 68, 560–565. [Google Scholar] [CrossRef]
  201. Sugerman, H.; Windsor, A.; Bessos, M.; Wolfe, L. Intra-abdominal pressure, sagittal abdominal diameter and obesity comorbidity. J. Intern. Med. 1997, 241, 71–79. [Google Scholar] [CrossRef]
  202. Arfvidsson, B.; Eklof, B.; Balfour, J. Iliofemoral venous pressure correlates with intraabdominal pressure in morbidly obese patients. Vasc. Endovasc. Surg. 2005, 39, 505–509. [Google Scholar] [CrossRef]
  203. Willenberg, T.; Clemens, R.; Haegeli, L.M.; Amann-Vesti, B.; Baumgartner, I.; Husmann, M. The influence of abdominal pressure on lower extremity venous pressure and hemodynamics: A human in-vivo model simulating the effect of abdominal obesity. Eur. J. Vasc. Endovasc. Surg. 2011, 41, 849–855. [Google Scholar] [CrossRef]
  204. Wiewiora, M.; Piecuch, J.; Glück, M.; Slowinska-Lozynska, L.; Sosada, K. Impact of weight loss due to sleeve gastrectomy on shear stress of the femoral vein in morbid obesity. Obes. Surg. 2014, 24, 806–812. [Google Scholar] [CrossRef]
  205. Castro-Ferreira, R.; Cardoso, R.; Leite-Moreira, A.; Mansilha, A. The Role of Endothelial Dysfunction and Inflammation in Chronic Venous Disease. Ann. Vasc. Surg. 2018, 46, 380–393. [Google Scholar] [CrossRef] [PubMed]
  206. Rattazzi, M.; Callegari, E.; Sponchiado, A.; Galliazzo, S.; Pagliara, V.; Villalta, S.; Pauletto, P. Visceral obesity, but not metabolic syndrome, is associated with the presence of post-thrombotic syndrome. Thromb. Res. 2015, 136, 225–228. [Google Scholar] [CrossRef]
  207. Araujo, D.N.; Ribeiro, C.T.; Maciel, A.C.; Bruno, S.S.; Fregonezi, G.A.; Dias, F.A. Physical exercise for the treatment of non-ulcerated chronic venous insufficiency. Cochrane Database Syst. Rev. 2016, 12, CD010637, Update in Cochrane Database Syst. Rev. 2023, 6, CD010637. [Google Scholar] [CrossRef]
  208. Weller, C.D.; Buchbinder, R.; Johnston, R.V. Interventions for helping people adhere to compression treatments for venous leg ulceration. Cochrane Database Syst. Rev. 2016, 3, CD008378. [Google Scholar] [CrossRef]
  209. Moffatt, C.J.; Franks, P.J.; Doherty, D.C.; Martin, R.; Blewett, R.; Ross, F. Prevalence of leg ulceration in a London population. QJM 2004, 97, 431–437. [Google Scholar] [CrossRef] [PubMed]
  210. Abbade, L.P.; Lastória, S.; Rollo Hde, A. Venous ulcer: Clinical characteristics and risk factors. Int. J. Dermatol. 2011, 50, 405–411. [Google Scholar] [CrossRef] [PubMed]
  211. Van Gent, W.B.; Catarinella, F.S.; Lam, Y.L.; Nieman, F.H.; Toonder, I.M.; van der Ham, A.C.; Wittens, C.H. Conservative versus surgical treatment of venous leg ulcers: 10-year follow up of a randomized, multicenter trial. Phlebology 2015, 30 (Suppl. 1), 35–41. [Google Scholar] [CrossRef]
  212. Van Gent, W.B.; Hop, W.C.; van Praag, M.C.; Mackaay, A.J.; de Boer, E.M.; Wittens, C.H. Conservative versus surgical treatment of venous leg ulcers: A prospective, randomized, multicenter trial. J. Vasc. Surg. 2006, 44, 563–571. [Google Scholar] [CrossRef]
  213. Gohel, M.S.; Mora MSc, J.; Szigeti, M.; Epstein, D.M.; Heatley, F.; Bradbury, A.; Bulbulia, R.; Cullum, N.; Nyamekye, I.; Poskitt, K.R.; et al. Long-term Clinical and Cost-effectiveness of Early Endovenous Ablation in Venous Ulceration: A Randomized Clinical Trial. JAMA Surg. 2020, 155, 1113–1121. [Google Scholar] [CrossRef]
  214. Kapp, S.; Miller, C.; Donohue, L. The clinical effectiveness of two compression stocking treatments on venous leg ulcer recurrence: A randomized controlled trial. Int. J. Low. Extrem. Wounds 2013, 12, 189–198. [Google Scholar] [CrossRef] [PubMed]
  215. Kankam, H.K.N.; Lim, C.S.; Fiorentino, F.; Davies, A.H.; Gohel, M.S. A Summation Analysis of Compliance and Complications of Compression Hosiery for Patients with Chronic Venous Disease or Post-thrombotic Syndrome. Eur. J. Vasc. Endovasc. Surg. 2018, 55, 406–416. [Google Scholar] [CrossRef]
  216. Bush, R.G. New technique to heal venous ulcers: Terminal interruption of the reflux source (TIRS). Perspect. Vasc. Surg. Endovasc. Ther. 2010, 22, 194–199. [Google Scholar] [CrossRef]
  217. Bush, R.; Bush, P. Percutaneous foam sclerotherapy for venous leg ulcers. J. Wound Care 2013, 22 (Suppl. 10), S20–S522. [Google Scholar] [CrossRef]
  218. Kamhawy, A.H.; Elbarbary, A.H.; Elhenidy, M.A.; Elwagih, A.M.M. Periulcer Foam Sclerotherapy Injection in Chronic Venous Leg Ulcers Using Near-Infrared Laser for Vein Visualization. Int. J. Low Extrem. Wounds 2020, 19, 63–69. [Google Scholar] [CrossRef] [PubMed]
  219. Vandongen, Y.K.; Stacey, M.C. Graduated Compression Elastic Stockings Reduce Lipodermatosclerosis and Ulcer Recurrence. Phlebology 2000, 15, 33–37. [Google Scholar] [CrossRef]
  220. Kapp, S.; Miller, C. The experience of self-management following venous leg ulcer healing. J. Clin. Nurs. 2015, 24, 1300–1309. [Google Scholar] [CrossRef]
  221. Nelson, E.A.; Bell-Syer, S.E. Compression for preventing recurrence of venous ulcers. Cochrane Database Syst. Rev. 2014, 2014, CD002303. [Google Scholar] [CrossRef]
  222. Moffatt, C.; Kommala, D.; Dourdin, N.; Choe, Y. Venous leg ulcers: Patient concordance with compression therapy and its impact on healing and prevention of recurrence. Int. Wound J. 2009, 6, 386–393. [Google Scholar] [CrossRef] [PubMed]
  223. Finlayson, K.J.; Parker, C.N.; Miller, C.; Gibb, M.; Kapp, S.; Ogrin, R.; Anderson, J.; Coleman, K.; Smith, D.; Edwards, H.E. Predicting the likelihood of venous leg ulcer recurrence: The diagnostic accuracy of a newly developed risk assessment tool. Int. Wound J. 2018, 15, 686–694. [Google Scholar] [CrossRef]
  224. Bar, L.; Brandis, S.; Marks, D. Improving Adherence to Wearing Compression Stockings for Chronic Venous Insufficiency and Venous Leg Ulcers: A Scoping Review. Patient Prefer. Adherence 2021, 15, 2085–2102. [Google Scholar] [CrossRef]
  225. Van Hecke, A.; Grypdonck, M.; Beele, H.; Vanderwee, K.; Defloor, T. Adherence to leg ulcer lifestyle advice: Qualitative and quantitative outcomes associated with a nurse-led intervention. J. Clin. Nurs. 2011, 20, 429–443. [Google Scholar] [CrossRef]
  226. Probst, S.; Weller, C.D.; Bobbink, P.; Saini, C.; Pugliese, M.; Skinner, M.B.; Gethin, G. Prevalence and incidence of venous leg ulcers-a protocol for a systematic review. Syst. Rev. 2021, 10, 148. [Google Scholar] [CrossRef]
  227. Klonizakis, M.; Tew, G.A.; Gumber, A.; Crank, H.; King, B.; Middleton, G.; Michaels, J.A. Supervised exercise training as an adjunct therapy for venous leg ulcers: A randomized controlled feasibility trial. Br. J. Dermatol. 2018, 178, 1072–1082. [Google Scholar] [CrossRef] [PubMed]
  228. Finlayson, K.; Edwards, H.; Courtney, M. Relationships between preventive activities, psychosocial factors and recurrence of venous leg ulcers: A prospective study. J. Adv. Nurs. 2011, 67, 2180–2190. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathophysiology of venous ulcers.
Figure 1. Pathophysiology of venous ulcers.
Jcm 12 06153 g001
Figure 2. Different modes of prevention and treatment of venous leg ulcers (CEAP C4—skin damage due to chronic insufficiency; IB—inelastic bandage; ACW—adjustable compression wrap; MCS—medical compression stocking.
Figure 2. Different modes of prevention and treatment of venous leg ulcers (CEAP C4—skin damage due to chronic insufficiency; IB—inelastic bandage; ACW—adjustable compression wrap; MCS—medical compression stocking.
Jcm 12 06153 g002
Figure 3. The pressure curve of an inelastic bandage exerts a compression pressure of 50 mm Hg. This pressure overcomes the intravenous pressure of about 75 mm Hg (red line) when performing foot dorsiflexion in a standing position and during walking, restoring a kind of valve mechanism; DSI—dynamic stiffness index; SSI—Static Stiffness Index; WPA—walking pressure amplitude.
Figure 3. The pressure curve of an inelastic bandage exerts a compression pressure of 50 mm Hg. This pressure overcomes the intravenous pressure of about 75 mm Hg (red line) when performing foot dorsiflexion in a standing position and during walking, restoring a kind of valve mechanism; DSI—dynamic stiffness index; SSI—Static Stiffness Index; WPA—walking pressure amplitude.
Jcm 12 06153 g003
Figure 4. The pressure curve of an elastic stocking exerts a compression pressure of about 20 mm Hg. The pressure never approaches the intravenous pressure of about 75 mm Hg (red line) when performing foot dorsiflexion in a standing position during walking. Veins are not compressed. DSI—dynamic stiffness index; SSI—Static Stiffness Index; WPA—walking pressure amplitude.
Figure 4. The pressure curve of an elastic stocking exerts a compression pressure of about 20 mm Hg. The pressure never approaches the intravenous pressure of about 75 mm Hg (red line) when performing foot dorsiflexion in a standing position during walking. Veins are not compressed. DSI—dynamic stiffness index; SSI—Static Stiffness Index; WPA—walking pressure amplitude.
Jcm 12 06153 g004
Figure 5. The pressure curve of an elastic bandage applied with full stretch exerts a compression pressure of about 75 mm Hg. Compression pressure is always the same or higher than the intravenous pressure (red line). Veins are always compressed. The bandage becomes painful within a short time. DSI—dynamic stiffness index; SSI—Static Stiffness Index; WPA—walking pressure amplitude.
Figure 5. The pressure curve of an elastic bandage applied with full stretch exerts a compression pressure of about 75 mm Hg. Compression pressure is always the same or higher than the intravenous pressure (red line). Veins are always compressed. The bandage becomes painful within a short time. DSI—dynamic stiffness index; SSI—Static Stiffness Index; WPA—walking pressure amplitude.
Jcm 12 06153 g005
Figure 6. Invasive methods of superficial venous insufficiency treatment (CHIVA—Ambulatory Conservative Hemodynamic Treatment of Venous Insufficiency; ASVAL—Ambulatory Selective Varices Ablation under Local Anesthesia; PIN—perforation imagination; EVLA—endovenous laser ablation; RFA—radiofrequency ablation; MOCA—mechanochemical ablation).
Figure 6. Invasive methods of superficial venous insufficiency treatment (CHIVA—Ambulatory Conservative Hemodynamic Treatment of Venous Insufficiency; ASVAL—Ambulatory Selective Varices Ablation under Local Anesthesia; PIN—perforation imagination; EVLA—endovenous laser ablation; RFA—radiofrequency ablation; MOCA—mechanochemical ablation).
Jcm 12 06153 g006
Figure 7. Invasive treatment of deep venous insufficiency.
Figure 7. Invasive treatment of deep venous insufficiency.
Jcm 12 06153 g007
Figure 8. Endovascular treatment of the occlusion of the iliac veins. (a,b) Right iliac phlebography shows occlusion of right common iliac vein (CIV) (white arrow) and severe collaterals from right internal iliac vein to contralateral iliac veins (red arrow); (c) balloon dilatation after recanalization of occluded part of right CIV (red arrow shows strong resistance due to scarring in the vein, 15 atm pressure was given); (d) right CIV and external iliac vein (EIV) is stented; (e) phlebography after stenting—vein inflow from CIV restored, all collateral veins disappeared.
Figure 8. Endovascular treatment of the occlusion of the iliac veins. (a,b) Right iliac phlebography shows occlusion of right common iliac vein (CIV) (white arrow) and severe collaterals from right internal iliac vein to contralateral iliac veins (red arrow); (c) balloon dilatation after recanalization of occluded part of right CIV (red arrow shows strong resistance due to scarring in the vein, 15 atm pressure was given); (d) right CIV and external iliac vein (EIV) is stented; (e) phlebography after stenting—vein inflow from CIV restored, all collateral veins disappeared.
Jcm 12 06153 g008
Table 1. Diagnostics of leg ulcers.
Table 1. Diagnostics of leg ulcers.
Medical History
History of present symptoms and signsDuration and presence of symptoms:
Cramps, tired legs, swollen legs, heavy legs, restless legs,
venous claudication,
itching
Pain: distribution, intensity (VAS score 0–10), duration, intermittent, during night/day, pain during dressing changes
Duration and presence of signs:
Varicose veins: duration, uni-/bilateral, bleeding from the vein
Swelling: uni-/bilateral, region: around the ankle, whole leg, relation to standing/sitting a whole day
Active ulcer: spontaneous/post-traumatic, duration, dressings (type/frequency of changes), compression therapy
Past signsPreviously healed/recurrent ulcers: spontaneous/post-traumatic, duration, dressings, compression therapy
DVT/SVT/PE: time of occurrence, therapy
Previous leg fractures
Previous surgical therapy
ComorbiditiesDiabetes mellitus, hypertension, chronic renal insufficiency, heart failures, malignancy, rheumatoid arthritis, PAD, obesity, back problems
TreatmentTreatment of present and past varicose veins: laser, sclerotherapy, surgical treatment, endovenous ablation (non-thermal/thermal), compression therapy (short-/long-stretch bandages, stockings, Velcro® materials)
Treatment of present ulcer: dressings, therapy of surrounding skin, compression; where and by whom treatment is provided (patient/nurse/in hospital/in healthcare center);
medications (anticoagulants, contraceptives, hormone replacement therapy, antidiabetics antihypertensives, immunosuppressive therapy, other)
AllergiesContact/systemic drug reaction
PregnancyWhen, number, signs, and symptoms during pregnancy, therapy of signs/symptoms
Family historyPresence of varicose veins in relatives, ulcers, DVT
OccupationProlonged standing/sitting
Bad habitsSmoking, alcohol consumption, drug use
TraumaMechanical, chemical, radiotherapy, chemotherapy, etc.
Clinical Assessment
Inspection and palpationMobility, BMI
Presence of varicose veins, corona phlebectatica
Limb swelling: Stemmer’s sign, non-pitting/pitting, Bisgaard sign
Skin changes: hyperpigmentations/redness (whole leg/during the vein, eczema),
lipodermatosclerosis/atrophie blanche
Peripheral arterial pulses, capillarity refilling
Groin lymph nodes
Leg temperature (cold/warm)
Scars after previous surgical therapy, trauma
Trophic changes in nails
Leg ulcers: where, number, size, wound bed (necrosis, fibrin (ogen), granulation tissue, epithelial tissue, isles in wound bed), edges, surrounding skin, smell, presence of infection, wound exudate,
possibility of ankle movements
Functional/Diagnostics Testing
Venous systemCW Doppler: S–F junction reflux
Duplex US
Photoplethysmography
Arterial systemMeasurement of ABI (with CW Doppler; automatic)
Lymphatic systemLimb circumferences, perimetry, bioimpedance
Non-invasive/invasive testsMonofilament test
Capillaroscopy
Venography
IVUS
Angiography
Lymphoscintigraphy
CT
MR
MicrobiologicalSwab for pathogenic bacteria and fungi
Skin/ulcer biopsyPathohistological examination
Direct immunofluorescence
Blood testsComplete/differential blood count, C-reactive protein, erythrocyte sedimentation rate,
blood glucose, HBA1c, blood lipids
electrolytes, urea, creatinine, liver function tests
tests of coagulations
total proteins, circulating immune complex,
immunoglobulins,
cryoglobulins, APC resistance, protein C, S, homocysteine
ANAs, ENA, anti-DNA, ANCAs, antiphospholipid antibodies, lupus antibodies,
pemphigus and pemphigoid antibodies,
vitamins (B12, D3, folic acid, A), trace elements (Fe, Zn, Mg, Cu)
Serological tests (lues tests—TPHA, leprosis, tbc)
VAS—visual analog scale for pain; DVT—deep-vein thrombosis; SVT—superficial-vein thrombosis; PE—pulmonary embolism; PAD—peripheral arterial disease; BMI—body mass index; CW Doppler—continuous-wave Doppler; S–F junction—saphenofemoral junction; Duplex US—duplex ultrasound; IVUS—intravascular ultrasound; CT—computed tomography; MR—magnetic resonance; HBA1c—hemoglobin A1c, glycated hemoglobin; APC—activated protein C; ANA—antinuclear antibodies; ENA—extractable nuclear antigen; anti-DNA—anti-double-stranded DNA (anti-dsDNA); ANCAs—antineutrophil cytoplasmic antibodies; Fe—ferrum; Zn—zinc; Mg—magnesium; Cu—copper; TPHA—Treponema Pallidum Hemagglutination Assay; tbc—tuberculosis.
Table 2. Assessment of leg ulcer.
Table 2. Assessment of leg ulcer.
The Questions We Ask OurselvesThe Causes/Symptoms/SignsTests for Making the Diagnosis
What is the immediate cause of the wound?
-
Trauma
-
Too-tight compression
-
Spontaneous
Is there any underlying pathology?
-
Venous disease: vein symptoms: varicose veins; mild ankle swelling during the day; hyperpigmentation; corona phlebectatica; lipodermatosclerosis; stasis eczema; atrophie blanche; past DVT/SVT; varicose veins/ulcer in family; reflux at SF junctions; insufficient veins at DU investigations; venography
-
Arterial disease: intermittent claudication; rest pain; poor perfusion (loss of pedal pulses, cold foot, pale skin); atrophic skin; loss of hair on the leg; trophic changes or gangrene; necrosis on the wound bed
-
Mixed venous–arterial etiology
-
Diabetic leg ulcer: neuropathic: warm, sensory deficit, diminished sweating, dry skin, motor neuropathy with a high arch and claw toes, Charcot foot; ischemic: pale, cold, absence of pulses, intermittent claudication; neuro-ischemic foot
-
Vasculitis: pain ulcers; small ulcers with necrosis
-
Malignancy: usually no pain; hypertrophic ulcers
-
Infection: high body temperature, redness around the ulcer
-
Bullous dermatosis
-
Lymphoedema stage III: primary or secondary cause lymphoedema: papillomatosis, Stemmer’s sign, non-pitting/pitting edema
-
Reflux at SF junctions, insufficient veins at DU investigations, plethysmography, venography
-
Blood lipids, low ABI (below 0.85 or below 0.5), arteriography
-
Blood glucose, HBA1c, monofilament test, capillaroscopy, ABI
-
ANCA, biopsy (HPE, DIF)
-
Biopsy
-
Swab, CRP, ESR, complete/differential blood count
-
Pemphigus and pemphigoid antibodies, biopsy
-
Limb circumferences, perimetry, bioimpedance, lymphoscintigraphy
Does the patient have any (medical) conditions?Diabetes mellitus, malnutrition, cardiovascular disease, anemia, renal disease, rheumatoid arthritis, cerebrovascular disease, old age, reduction in sensory perception, increasing susceptibility to trauma, therapy with immunosuppression agents, malignancies and their treatment, smoking, alcohol consumption, drug use, prolonged standing/sitting, obesity, poor mobility
SF−saphenofemoral; DU—Doppler ultrasound; DVT/SVT−deep-vein thrombosis/superficial-vein thrombosis; ABI−ankle–brachial index; HBA1c—hemoglobin A1c, glycated hemoglobin; ANCAs−antineutrophil cytoplasmic antibodies; HPE−histopathological examination; DIF−direct immunofluorescence microscopy; CRP−C-reactive protein; ESR−erythrocyte sedimentation rate.
Table 3. Recommendations of investigations in patients with venous leg ulcers/level of evidence.
Table 3. Recommendations of investigations in patients with venous leg ulcers/level of evidence.
Palpation of lower-extremity arterial pulses and calculated ABI are recommended for all patients with suspected venous leg ulcersC
Duplex ultrasound sonography is recommended for patients with venous leg ulcers to assess venous reflux and/or obstructionC
Biopsy is recommended for patients with venous leg ulcers if healing stallsC
Biopsy is recommended for patients with ulcers if there is suspicion that the ulcer may be venous, but it has an atypical appearanceC
Referral to a subspecialist is recommended for patients with venous leg ulcers if healing stallsC
Referral to a subspecialist is recommended for patients with ulcers if there is suspicion that the ulcer is not venous, but it is of an atypical appearanceC
Screening of patients using a hand-held Doppler detector makes sense only in mild involvement when only telangiectasias and venectasias are presentC
X-ray contrast venography, magnetic resonance, or computed tomography venography are reasonable to perform only in a small number of selected patients who have anatomical venous anomalies, and in those patients in whom surgical intervention on the deep venous system is planned.C
ABI−ankle–brachial index; C−based on expert opinion and consensus guidelines in the absence of clinical trials.
Table 4. Hemodynamic effects of compression therapy.
Table 4. Hemodynamic effects of compression therapy.
Venous Reflux
(VR)
Calf Pumping Function (CPF)Ambulatory Venous Hypertension (AVH)
Elastic material~~~
Inelastic material+++++++++
~ Very low influence; +++ high influence.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stanek, A.; Mosti, G.; Nematillaevich, T.S.; Valesky, E.M.; Planinšek Ručigaj, T.; Boucelma, M.; Marakomichelakis, G.; Liew, A.; Fazeli, B.; Catalano, M.; et al. No More Venous Ulcers—What More Can We Do? J. Clin. Med. 2023, 12, 6153. https://doi.org/10.3390/jcm12196153

AMA Style

Stanek A, Mosti G, Nematillaevich TS, Valesky EM, Planinšek Ručigaj T, Boucelma M, Marakomichelakis G, Liew A, Fazeli B, Catalano M, et al. No More Venous Ulcers—What More Can We Do? Journal of Clinical Medicine. 2023; 12(19):6153. https://doi.org/10.3390/jcm12196153

Chicago/Turabian Style

Stanek, Agata, Giovanni Mosti, Temirov Surat Nematillaevich, Eva Maria Valesky, Tanja Planinšek Ručigaj, Malika Boucelma, George Marakomichelakis, Aaron Liew, Bahar Fazeli, Mariella Catalano, and et al. 2023. "No More Venous Ulcers—What More Can We Do?" Journal of Clinical Medicine 12, no. 19: 6153. https://doi.org/10.3390/jcm12196153

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