This site is intended for healthcare professionals only

The Diabetic
Foot Journal

Issue:

Share this article

Screening developments for the foot in diabetes

Aditya Dutta, Ashu Rastogi, Edward B Jude
Foot complications in people with diabetes are often neglected, which leads to significant morbidity and even mortality. Screening of the foot at initial diagnosis of type 2 diabetes and periodically on subsequent clinic visits is helpful in early recognition of foot complications. Foot screening involves a thorough history pertaining to risk factors for foot complications and prior pedal ulcers; assessment for diabetic peripheral neuropathy, peripheral vascular disease and foot deformities. A simple tuning fork, monofilament sensation, palpation of pedal pulses and Ankle Brachial Index assessment provide necessary information for categorising the risk for future foot complications.

Foot complications in people with diabetes is an outcome of increased longevity (Bhansali and Rastogi, 2016). As people with diabetes live longer, they develop microvascular complications like neuropathy and macrovascular complexity of vasculopathy, both of which contribute to foot complications. Once people with diabetes develop foot complications then it contributes to excess economic burden, morbidity and even mortality (Boulton et al, 2005; Al-Rubeaan et al, 2017; Kerr et al, 2019). Unfortunately, most patients are referred late to healthcare professionals, which adds to the seriousness of the condition. Therefore, screening for foot complications and especially the ‘foot at risk’ in a given individual with diabetes takes a precedence during each visit to the healthcare facility. The following review provides an overview of screening procedures for the foot in diabetes and their pragmatic use in resource constraint settings.

What is the diabetic foot?
The ‘diabetic foot’ has been defined as infection, ulceration or destruction of tissues of the foot associated with neuropathy and/or arterial disease in the lower-extremity of a person with diabetes (van Netten et al, 2019). The risk factors for diabetic foot include presence of signs or symptoms of diabetic peripheral neuropathy (DPN), autonomic neuropathy, peripheral arterial disease (PAD), pre-ulcerative lesions (like callus), foot deformities (like hallux valgus), previous foot ulcer or amputation, oedema, smoking or nicotine use, male sex, duration of diabetes, complications of diabetes (especially end-stage renal disease and retinopathy) and post-transplant status (Boulton et al, 2018).
Numerous classification schemes are available to guide the risk stratification and follow-up frequency in a diabetic foot patient on a case-to-case basis (Boyko et al, 2006; Boulton et al, 2008; Scottish Intercollegiate Guidelines Network [SIGN], 2010; International Diabetes Federation, 2017; Bus et al, 2020). These classification systems point out five key risk factors related to diabetic foot ulcer (DFU) development. First, DPN itself can increase the risk of development of first foot ulcer by seven-fold by virtue of an insensate foot, decreasing proprioception and hampering the balance (Young et al, 1994). Second, PAD has a causal role in pathway to ulceration in up to 35% of cases (Reiber et al, 1999) and its prevalence in patients with DFU is nearly 50% (Prompers et al, 2007). Third, foot deformities worsen plantar pressures and result in DFU at areas of high pressure and recurrent stress (Singh et al, 2005; Chatwin et al, 2019). Fourth, 30–50% ulcers may recur in individuals with prior history of DFU and/or amputation (Armstrong et al, 2017). In addition to the above, end-stage renal disease (ESRD) and dialysisare independent risk factors for foot ulceration (Lavery et al, 2010; Ndip et al, 2010). The role of these risk factors have been assessed in a recent systematic review (Crawford et al, 2015).

Why to screen for the diabetic foot?
The annual incidence of diabetic foot ulcers (DFU) is 2% (Abbott et al, 2002). The lifetime risk of DFU in a person with diabetes is thought to be between 19–34% (Armstrong et al, 2017). Diabetes-related lower-extremity complications (LEC) rank within the top-10 leading causes of the global disability burden (Lazzarini et al, 2018). Mortality data are staggeringly high, exceeding 70% at 5 years for people with diabetes with some level of amputation (Lavery et al, 2010). A study conducted in 1983 found the incidence of foot examination performed in people with diabetes by physicians to be only 12.3% (Bailey et al, 1985). Nearly three decades later, this figure has not improved much (Jayaprakash et al, 2011). Many studies have shown that provision of foot-care services and preventive care can reduce amputations and financial burden in people with diabetes (McCabe et al, 1998; Sloan et al, 2010; Carls et al, 2011). In one such study, visiting both a podiatrist and a LEC specialist in the year before diagnosis of LEC, was protective of undergoing lower-extremity amputation (Sloan et al, 2010).

In a recent survey, it has been estimated that one-third reduction in prevalence of DFU in England would result in an annual saving of £240m (Kerr et al, 2019). Unfortunately, these data are not corroborated with data from randomised control trials (RCT) (Dorresteijn and Valk, 2012). Rather, more data is available for secondary prevention, i.e., reducing the risk of foot ulcer recurrence. A systematic review of studies evaluating the role of integrated foot care, self-management, therapeutic footwear and foot surgery has shown a mean effect size ranging from 30.9% to 61.8% in reducing the risk of recurrent foot ulcer in the intervention groups (van Netten et al, 2020).
Patient targeted education by itself is insufficient in providing clinical benefit at the level of secondary preventiondue to inherent constant physical abnormalities in the diabetic foot (Lincoln et al, 2008). Thus, there is a compelling need for clinical screening of the diabetic foot in people with diabetes.

Whom and when to screen?
The microvascular complications can be observed at the onset of type 2 diabetes (T2D); hence, screening of the diabetic foot should start at the outset. The Diabetic Foot Risk Stratification and Triage System (Figure 1) outlines the screening frequency (the traffic light) and the subsequent intervention (Stang and Leese, 2016). Certain populations like patients who have end-stage renal disease (ESRD) or post-renal transplant should be screened more frequently (Bus et al, 2020). However, in young people with diabetes (especially type 1 diabetes) the screening protocol is not well defined. In the latter subgroup, we believe that screening for neuropathy (at least) should begin within 5 years after diagnosis, mirroring the retinopathy assessment (American Diabetes Association [ADA], 2020). For vasculopathy, the ADA suggests at least annual history and examination of pulses in a person with diabetes, and Ankle Brachial Index (ABI) in patients with symptoms or signs of PAD (ADA, 2020). There are multiple other recommendations for (Gerhard-Herman et al, 2017; National Institute for Health and Care Excellence [NICE], 2018; Bus et al, 2020) and against (US Preventive Services Task Force et al, 2018) the use of ABI for screening of PAD in asymptomatic but high risk individuals (like people with diabetes). In short, for screening of PAD in people with diabetes, annual clinical examination is a must and the use of ABI is at the discretion of the healthcare professional. Screening for cardiovascular risk and emphasis on smoking cessation can be integrated in the diabetic foot annual assessment (Stang and Leese, 2016).

How to screen?
History
A detailed history should be taken keeping in mind the following points:

  • Neuropathy symptoms (positive: burning or shooting pain, tingling sensations; negative: numbness, walking on cotton/air and loss of temperature sensation)
  • Musculoskeletal symptoms (feet too large for the shoe, slippage of slippers and foot drop)
  • Vascular symptoms (claudication, rest pain, discolouration, non-healing ulcer and fatigue)
  • Diabetes duration, complications of diabetes (retinopathy precludes foot-care and dialysis or post-transplant status)
  • Past history of DFU, gangrene, amputation, revascularisation, cardiovascular disorder and tobacco use.

Inspection of the foot
Examination of the foot should start withscrutiny of the skin, nails, interdigital areas, skin over the deformities, pre-ulcerative signs, oedema, prominent veins and erythema.

  • Pre-ulcerative signs (callus, maceration, blisters, fissures and bleeding in callus) serve as pointers for diabetic foot
  • Callus develops due to abnormal foot pressures at sites like deformities (claw toes and prominent metatarsal heads), dorsum of toes (cramped footwear) or midfoot (Charcot neuroarthropathy)
  • Presence of nail changes (ingrown nail, onychomycosis, onychogryphosis and onycholysis) and nail or interdigital infection (paronychia, intertrigo and dermatophytosis) should prompt a visit to the specialist
  • Lack of hair and skin/nail discolouration point to existence of PAD
  • Ill-fitting, worn-out or lack of footwear should also be recorded.

Musculoskeletal assessment
Common structural deformities in a diabetic foot include hammer toes, mallet toes, claw toes, hallux valgus (bunion), hallux rigidus, prominent metatarsal heads, pes cavus, pes planus and rocker-bottom foot (residual of Charcot neuroarthropathy). Dorsal and plantar flexion of the foot, guttering of the foot and gait (loss of proprioception) should also be checked.

Neurological assessment
Establishing the presence of DPN is fundamental to identify the diabetic foot. Diabetes is characterised by a ‘dying back’ axonopathy affecting C (small) and A (large) fibres. This causes impairment of sensory functions in the foot (e.g. loss of pain sensation, unsteadiness and dryness etc.) and predisposes to deformities and ulceration. The past decade has seen a trend to objectify the neurological testing in order to minimise the receiver-operator bias and make it easy to execute at the patients’ bedside. Several clinical examination methods, point-of-care (POC) devices, instruments and chemical indicators are now available for screening of neuropathy (Table 1).

Traditional screening methods
Current American Diabetes Association (ADA) recommendations include taking a detailed history, and assessment of either temperature or pinprick sensation (small fibre) and vibration perception threshold (VPT) using a 128-Hz tuning fork (large fibre) along with 10-g monofilament testing (ADA, 2020). DPN has been defined as presence of loss of protective sensation (LOPS) along with absence of either pin-prick, temperature sensation, vibration sensation or ankle reflex. The diverse options given by the ADA are based on regional practices and near-similar performance of tests against each other (Perkins et al, 2001; Jayaprakash et al, 2011).

10-g Semmes-Weinstein monofilament has been the most advocated test for foot examination due to ease in performing the same and widespread availability. Its outcome measure, loss of protective sensation (LOPS) is defined as inability to sense light pressure (10-g force). A recent meta-analysis of monofilament tests (using nerve conduction study as a reference) has shown pooled sensitivity and specificity of 0.53 and 0.88, respectively, with heterogeneous sensitivities (16.7%–95.8%; Wang et al, 2017). These results may reflect inconsistency in the technique (number and sites of testing), reference standards and wear and tear. To maintain the accuracy, the monofilament should be regularly replaced (6 monthly or if bent). Other monofilaments available in clinical practice include Bailey’s (retractable) 10-g monofilament and Owen Mumford’s Neuropen.

Vibration sensation testing by 128-Hz tuning fork is considered one of the best screening modalities for the foot in diabetes (Young et al, 1994; Jayaprakash et al, 2011). It is validated, inexpensive, durable and easy to perform with high sensitivity (>80%; Meijer et al, 2003; Martin et al, 2010). Grading of severity of DPN (mild, moderate and severe) is done with the use of the biothesiometer or neurothesiometer. Here, a vibration perception threshold (VPT) of ≥25 is considered as diagnostic for neuropathy. However, these take longer time to operate and are expensive.
Absence of ankle reflex is an easy bedside sign to demonstrate DPN. Studies evaluating ankle reflex alone or as part of neuropathy disability score (NDS) have found high sensitivity (>80%) but variable specificity (Shehab et al, 2012; Malik et al, 2013). It is unreliable as a single test due to high incidence of absent ankle reflex in general population and older adults (Bowditch et al, 1996).

By combining traditional methods (like ankle reflex and VPT) and the appearance of the foot during inspection, several scores like Michigan Neuropathy Screening Instrument (MNSI) and NDS have been developed to aid in quick out-patient screening.

Advances in neuropathy screening
The Ipswich Touch Test (IpTT) is a simple bedside test for neuropathy screening. It has been prospectively evaluated in a head-to-head trial with 10-g monofilament and was found to have good sensitivity and specificity (k=0.88; P<0.0001), and positive predictive value (89%) in detecting LOPS (Rayman et al, 2011). It has been validated in various studies (Sharma et al, 2014; Madanat et al, 2015) and is likely to supplant 10-g monofilament in diabetic foot examination. VibraTip is a small handheld battery-operated device. It has been studied prospectively against the neurothesiometer and NDS thresholds and has demonstrated good sensitivity (>80%) and specificity (>82%) (Bracewell et al, 2012; Papanas et al, 2020). Smartphones appear to have future potential for checking VPT, as well as temperature sensation testing as they are able to generate vibration of 25-Hz. This feature has been tested in a small trial of 21 patients with DPN and was found to fare better (accuracy — 0.88) than either the tuning fork or the 10-g monofilament (May and Morris, 2017).

NC-stat DPNCheck is a POC device that measures conduction velocity and action potential of sensory nerves in lateral thigh (sural nerve). It is free of patient bias and also identifies patients without symptoms of neuropathy (Poulose et al, 2015). It has been validated in people with diabetes with DPN (Binns-Hall et al, 2018) and seems to be a promising tool.

NeuroQuick is another handheld device emitting cold air at a standardised distance to the dorsum of the foot. With its 10 levels of fan velocity, one can grade the temperature sensation at which cold airflow is recognised. It has been studied in early DPN, and found to outperform traditional thermal testing and tuning fork test (Ziegler et al, 2005).

Neuropad indicator test to study the sudomotor function of the plantar skin is a good screening test to exclude DPN, with a high negative predictive value (98%) and reproducible results (Manes et al, 2014). It has been shown to predict the development of DPN in people with diabetes and pre-diabetes (Ziegler et al, 2012). Conversely, due to poor specificity, abnormal results require confirmation by additional testing (Manes et al, 2014). Neuropad automated continuous image analysis software has been tested which may improve the diagnostic yield of this test (Ponirakis et al, 2015).

Sudoscan is another non-invasive test for testing small fiber and autonomic neuropathy. It relies on the production an electric current from sodium chloride in the sweat. No discomfort is felt during the test and the results are reproducible (Gin et al, 2011). The test correlated well with both NDS and VPT in a prospective study for asymptomatic diabetic neuropathy (Mao et al, 2017). However, the test lacks consistent normative data on its outcome measure, namely the electrochemical skin conductance (ESC) (Rajan et al, 2019).

Vascular assessment
Historical points relevant to PAD assessment are mentioned in section 4.1. It is imperative to suspect PAD in a patient with current or prior history of non-healing DFU of >6 weeks duration (Hinchliffe et al, 2016). Examination for PAD should include:

  • Observing the feet for lack of hair and skin/nail discolouration
  • looking at calf muscle girth (for atrophy)
  • Checking pedal pulses (femoral, popliteal, posterior tibial, dorsalis pedis) bilaterally
  • Evaluating for bruit and slow venous filling time.

Regrettably, none of these clinical markers are accurate enough to detect PAD (Collins et al, 2006). Currently, the ADA suggests at least annual history and examination of pulses in a patient with diabetes, and ABI in patients with symptoms or signs of PAD (ADA, 2020). ABI represents the ratio of the systolic blood pressure (SBP) at the ankle divided by SBP at the arm. SBP of both the arms is noted and the higher value becomes the denominator. A value between 0.91–1.30 is considered as normal (Table 2). Depending on the device function, doppler waveforms can also be generated or printed. The test is easy to perform at the bedside, requires minimal training, is cost-effective, non-invasive and less time consuming. Sensitivity can further be improved by 6-minute treadmill walk test. In a systematic review, the sensitivity of ABI<0.9 in diagnosing PAD, ranged from 29 to 95% (median at 63%), and its specificity varied between 58% and 97% (median 93%) (US Preventive Services Task Force et al, 2018). Limitations include inconsistent inter- and intra-tester reliability (Casey et al, 2019), non-reliability in patients with medial arterial calcification (especially patients with ESRD) and operator bias (Tóth-Vajna et al, 2019). Despite these limitations, handheld ABI measurement is unlikely to lose its importance as a valuable tool in screening undiagnosed PAD. Automated oscillometric ABI devices have been developed to minimise operator bias. These have been found to be as reliable as colour doppler sonography in detecting PAD in people with diabetes (Ma et al, 2017). PAD in diabetes: difficulties in screening
PAD in diabetes has certain distinctive features. It is insidious, preferentially affects infra-popliteal arterial system, has diffuse involvement, has poor collateral formation and has faster progression. It is associated with a high risk for first foot ulcer, non-healing DFU, amputation, cardiovascular events and mortality. Thus, it seems appropriate to institute early screening for PAD in diabetes. However, three difficulties are commonly encountered. First, diabetic neuropathy may shield the symptoms of PAD and predispose to medial arterial calcification (Jeffcoate et al, 2009). Second, pedal pulses may remain palpable even when underlying stenosis is present, and is otherwise unreliable in a busy clinic (Lundin et al, 1999). Third, screening of asymptomatic population may have undue financial repercussions. Still, three small studies have yielded a high prevalence of undiagnosed PAD (26–57%) using handheld ABI Doppler in people with diabetes (Formosa et al, 2012; Ogbera et al, 2015; Tummala et al, 2018).

Screening in resource-constrained settings
The screening practices in resource constraint settings should be the ones that are cost-effective, accessible, less technically demanding, less time consuming and reliable. The 10-g monofilament, 128-Hz tuning fork, ankle reflex, IpTT, palpation of pedal pluses and the handheld ABI device have been used successfully in community-based studies in  developing nations (Viswanathan et al, 2005; Jayaprakash et al, 2011; Formosa et al, 2012; Malik et al, 2013; Madanat et al, 2015; Tummala et al, 2018). A plethora of both short and comprehensive examinations are available at our behest (Boulton et al, 2008; Miller et al, 2014). While a detailed examination entails assessment of dermatological, sensory, musculoskeletal and vascular systems, it is often not practical in resource constraint settings. The authors suggest the use of the 3-minute foot examination module to actively screen and triage people with diabetes for various risk factors (Miller et al, 2014). The foot examination should be followed by at least an annual risk stratification (traffic light system, ADA or IWGDF risk scores) to allow for timely intervention.

Emphasis by the healthcare professional on foot-care education, including daily foot inspection, avoiding walking barefoot, not to cut callosities with razors or knives at home, use of appropriate footwear in high-risk patients and early presentation to the hospital at the onset of a foot lesion, can serve to offload the burden of the foot in diabetes.

To access the CPD module, click here.

REFERENCES:

Abbott CA, Carrington AL, Ashe, H et al (2002) The North-West Diabetes Foot Care Study: incidence of, and risk factors for, new diabetic foot ulceration in a community-based patient cohort. Diabet Med 19(5): 377–84
Al-Rubeaan K, Almashouq MK, Youssef AM et al (2017) All-cause mortality among diabetic foot patients and related risk factors in Saudi Arabia. PloS One 12(11): e0188097 
Armstrong DG, Boulton AJM, Bus SA (2017) Diabetic foot ulcers and their recurrence. N Engl J Med 376(24): 2367–75
Bailey TS, Yu HM, Rayfield EJ (1985) Patterns of foot examination in a diabetes clinic. Am J Med 78(3): 371–4
Bhansali A, Rastogi A (2016) Diabetic foot: an outcome of increasing longevity. J Foot Ankle Surg 3(2): 65
Binns-Hall O, Selvarajah D, Sanger D et al (2018) One-stop microvascular screening service: an effective model for the early detection of diabetic peripheral neuropathy and the high-risk foot. Diabet Med 35(7): 887–94
Boulton AJ, Vileikyte L, Ragnarson-Tennvall G, Apelqvist J (2005) The global burden of diabetic foot disease. Lancet 366(9498): 1719–24
Bowditch MG, Sanderson P, Livesey JP (1996) The significance of an absent ankle reflex. J Bone Joint Surg Br 78(2): 276–9
Boyko EJ, Ahroni JH, Cohen V et al (2006) Prediction of diabetic foot ulcer occurrence using commonly available clinical information: The Seattle Diabetic Foot Study. Diabetes Care 29(6): 1202–7
Bracewell N, Game F, Jeffcoate W, Scammell BE (2012) Clinical evaluation of a new device in the assessment of peripheral sensory neuropathy in diabetes. Diabet Med 29(12): 1553–5
Bus SA, Lavery LA, Monteiro-Soares M et al (2020) Guidelines on the prevention of foot ulcers in persons with diabetes (IWGDF 2019 update). Diabetes Metab Res Rev 36(Suppl 1): e3269
Carls GS, Gibson TB, Driver VR et al (2011) The economic value of specialized lower-extremity medical care by podiatric physicians in the treatment of diabetic foot ulcers. J Am Podiatr Med Assoc 101(2): 93–115
Casey S, Lanting S, Oldmeadow C, Chuter V (2019) The reliability of the Ankle Brachial Index: a systematic review. J Foot Ankle Res 12: 39
Chatwin KE, Abbott CA, Boulton AJM (2019) The role of foot pressure measurement in the prediction and prevention of diabetic foot ulceration — a comprehensive review. Diabetes Metab Res Rev 36(4): e3258
Collins TC, Suarez-Almazor M, Peterson NJ (2006) An absent pulse is not sensitive for the early detection of peripheral arterial disease. Fam Med 38(1): 38–42
Crawford F, Cezard G, Chappell FM et al (2015) A systematic review and individual patient data meta-analysis of prognostic factors for foot ulceration in people with diabetes: the international research collaboration for the Prediction of Diabetic Foot Ulcerations (PODUS). Health Technol Assess 19(57): 1–210
Dorresteijn JAN, Valk GD (2012) Patient education for preventing diabetic foot ulceration. Diabetes Metab Res Rev 28(Suppl 1): 101–6
Formosa C, Gatt A, Chockalingam N (2012) Screening for peripheral vascular disease in patients with type 2 diabetes in Malta in a primary care setting. Qual Prim Care 20(6): 409–14
Gerhard-Herman MD, Gornik HL, Barrett C et al (2017) 2016 AHA/ACC guideline on the management of patients with Lower extremity peripheral artery disease: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines. Circulation 135(12): e686–e725
Gin H, Baudoin R, Raffaitin CH et al (2011) Non-invasive and quantitative assessment of sudomotor function for peripheral diabetic neuropathy evaluation. Diabetes Metab 37(6): 527–32
Hinchliffe RJ, Brownrigg JRW, Apelqvist J et al (2016) IWGDF guidance on the diagnosis, prognosis and management of peripheral artery disease in patients with foot ulcers in diabetes.Diabetes Metab Res Rev 32(Suppl 1): 37–44.
Jayaprakash P, Bhansali A, Bhansali S (2011) Validation of bedside methods in evaluation of diabetic peripheral neuropathy. Indian J Med Res 133(6): 645–9
Jeffcoate WJ, Rasmussen LM, Hofbauer LC, Game FL (2009) Medial arterial calcification in diabetes and its relationship to neuropathy. Diabetologia 52(12): 2478–88
Kerr M, Barron E, Chadwick P et al (2019) The cost of diabetic foot ulcers and amputations to the National Health Service in England. Diabet Med 36(8): 995–1002
Lavery LA, Hunt NA, Ndip A et al (2010) Impact of chronic kidney disease on survival after amputation in individuals with diabetes. Diabetes Care 33(11): 2365–9
Lazzarini PA, Pacella RE, Armstrong DG, van Netten JJ (2018) Diabetes-related lower-extremity complications are a leading cause of the global burden of disability. Diabet Med doi: 10.1111/dme.13680. [Online ahead of print]
Lincoln NB, Radford KA, Game FL, Jeffcoate WJ (2008) Education for secondary prevention of foot ulcers in people with diabetes: a randomised controlled trial. Diabetologia 51(11): 1954–61
Lundin M, Wiksten JP, Peräkylä T et al (1999) Distal pulse palpation: is it reliable? World J Surg 23(3): 252–5
Ma J, Liu M, Chen D et al (2017) The validity and reliability between automated oscillometric measurement of Ankle-Brachial Index and standard measurement by Eco-Doppler in diabetic patients with or without diabetic foot. Int J Endocrinol 2017: 2383651
Madanat A, Sheshah E, Badawy E-B et al (2015) Utilizing the Ipswich Touch Test to simplify screening methods for identifying the risk of foot ulceration among diabetics: the Saudi experience. Prim Care Diabetes 9(4): 304–6
Malik MM, Jindal S, Bansal S et al (2013) Relevance of ankle reflex as a screening test for diabetic peripheral neuropathy. Indian J Endocrinol Metab 17(7): 340
Manes C, Papanas N, Exiara T et al (2014) The indicator test Neuropad in the assessment of small and overall nerve fibre dysfunction in patients with type 2 diabetes: a large multicentre study. Exp Clin Endocrinol Diabetes 122(3): 195–9
Mao F, Liu S, Qiao X et al (2017) Sudoscan is an effective screening method for asymptomatic diabetic neuropathy in Chinese type 2 diabetes mellitus patients. J Diabetes Investig 8(3): 363–8
Martin CL, Waberski BH, Pop-Busui R et al (2010) Vibration perception threshold as a measure of distal symmetrical peripheral neuropathy in type 1 diabetes: results from the DCCT/EDIC study. Diabetes Care 33(12): 2635–41
May JD, Morris MWJ (2017) Mobile phone generated vibrations used to detect diabetic peripheral neuropathy. Foot Ankle Surg 23(4): 281–4.
McCabe CJ, Stevenson RC, Dolan AM (1998) Evaluation of a diabetic foot screening and protection programme. Diabet Med 15(1): 80–4
Meijer J-WG, Bosma E, Lefrandt JD et al (2003) Clinical diagnosis of diabetic polyneuropathy with the diabetic neuropathy symptom and diabetic neuropathy examination scores. Diabetes Care 26(3): 697–701
NICE (2012) Peripheral Arterial Disease: Diagnosis and Management. Clinical guideline [CG147]. Available at: https://bit.ly/303whgl (accessed 04.06.2020)
Ndip A, Rutter MK, Vileikyte L et al (2010) Dialysis treatment is an independent risk factor for foot ulceration in patients with diabetes and stage 4 or 5 chronic kidney disease. Diabetes Care 33(8): 1811–6
Ogbera AO, Adeleye O, Solagberu B, Azenabor A (2015) Screening for peripheral neuropathy and peripheral arterial disease in persons with diabetes mellitus in a Nigerian University Teaching Hospital. BMC Res Notes 8: 533
Papanas N, Pafili K, Demetriou M et al (2020) The diagnostic utility of VibraTip for distal symmetrical polyneuropathy in type 2 diabetes mellitus. Diabetes Ther 11(1): 341–6
Perkins BA, Olaleye D, Zinman B, Bril V (2001) Simple screening tests for peripheral neuropathy in the diabetes clinic. Diabetes Care 24(2): 250–6
Ponirakis G, Fadavi H, Petropoulos IN et al (2015) Automated quantification of Neuropad improves its diagnostic ability in patients with diabetic neuropathy. J Diabetes Res 2015: 847854
Poulose S, Cheriyan E, Poulose A et al (2015) Usefulness of the NC-stat DPNCheck nerve conduction test in a community pharmacy as an educational tool for patients with diabetes. Can Pharm J (Ott) 148(1): 17–20
Prompers L, Huijberts M, Apelqvist J et al (2007) High prevalence of ischaemia, infection and serious comorbidity in patients with diabetic foot disease in Europe. Baseline results from the Eurodiale study. Diabetologia 50(1): 18–25
Rajan S, Campagnolo M, Callaghan B, Gibbons CH (2019) Sudomotor function testing by electrochemical skin conductance: does it really measure sudomotor function? Clin Auton Res 29(1): 31–9
Rayman G, Vas PR, Baker N et al (2011) The Ipswich Touch Test: a simple and novel method to identify inpatients with diabetes at risk of foot ulceration. Diabetes Care 34(7): 1517–8
Reiber GE, Vileikyte L, Boyko EJ et al (1999) Causal pathways for incident lower-extremity ulcers in patients with diabetes from two settings. Diabetes Care 22(1): 157–62
Scottish Intercollegiate Guidelines Network (2010) 116 Management of Diabetes. A National Clinical Guideline. Edinburgh: SIGN. Available at: https://bit.ly/2YdQTQD (accessed 04.06.2020)
Sharma S, Kerry C, Atkins H, Rayman G (2014) The Ipswich Touch Test: a simple and novel method to screen patients with diabetes at home for increased risk of foot ulceration. Diabet Med 31(9): 1100–3
Shehab DK, Al-Jarallah KF, Abraham M et al (2012) Back to basics: ankle reflex in the evaluation of peripheral neuropathy in type 2 diabetes mellitus. QJM 105(4): 315–20
Singh N, Armstrong DG, Lipsky BA (2005) Preventing foot ulcers in patients with diabetes. JAMA 293(2): 217–28
Sloan FA, Feinglos MN, Grossman DS (2010) Receipt of care and reduction of lower extremity amputations in a nationally representative sample of U.S. elderly. Health Serv Res 45(6 Pt 1) 1740–62
Stang D, Leese GP (2016) The Scottish Diabetes Foot Action Group 2016 update of the Diabetic Foot Risk Stratification and Triage System. The Diabetic Foot Journal 19(4): 182–6
Tóth-Vajna Z, Tóth-Vajna G, Gombos Z et al (2019) Screening of peripheral arterial disease in primary health care. Vasc Health Risk Manag 15: 355–63
Tummala R, Banerjee K, Mahajan K et al (2018) Utility of ankle-brachial index in screening for peripheral arterial disease in rural India: a cross-sectional study and review of literature. Indian Heart J 70(2): 323–5
US Preventive Services Task Force, Curry SJ, Krist AH et al (2018) Screening for peripheral artery disease and cardiovascular disease risk assessment with the Ankle-Brachial Index: US Preventive Services Task Force Recommendation Statement. JAMA 320(2): 177–83
van Netten JJ, Raspovic A, Lavery LA et al (2020) Prevention of foot ulcers in the at-risk patient with diabetes: a systematic review. Diabetes Metab Res Rev 36(Suppl 1): e3270
Viswanathan V, Thomas N, Tandon N (2005) Profile of diabetic foot complications and its associated complications — a multicentric study from India. J Assoc Physicians India 53: 933–6
Wang F, Zhang J, Yu J et al (2017) Diagnostic accuracy of monofilament tests for detecting diabetic peripheral neuropathy: a systematic review and meta-analysis. J Diabetes Res 2017: 8787261
Young MJ, Breddy JL, Veves A, Boulton AJ (1994) The prediction of diabetic neuropathic foot ulceration using vibration perception thresholds. A prospective study. Diabetes Care 17(6): 557–60
Ziegler D, Papanas N, Rathmann W et al (2012) Evaluation of the Neuropad sudomotor function test as a screening tool for polyneuropathy in the elderly population with diabetes and pre-diabetes: the KORA F4 survey. Diabetes Metab Res Rev 28(8): 692–7
Ziegler D, Siekierka-Kleiser E, Meyer B, Schweers M (2005) Validation of a novel screening device (NeuroQuick) for quantitative assessment of small nerve fiber dysfunction as an early feature of diabetic polyneuropathy. Diabetes Care 28(5): 1169–74

Related content
Is artificial intelligence the key to better foot self-care in diabetes?
;
Free for all UK & Ireland healthcare professionals

Sign up to all DiabetesontheNet journals

 

By clicking ‘Subscribe’, you are agreeing that DiabetesontheNet.com are able to email you periodic newsletters. You may unsubscribe from these at any time. Your info is safe with us and we will never sell or trade your details. For information please review our Privacy Policy.

Are you a healthcare professional? This website is for healthcare professionals only. To continue, please confirm that you are a healthcare professional below.

We use cookies responsibly to ensure that we give you the best experience on our website. If you continue without changing your browser settings, we’ll assume that you are happy to receive all cookies on this website. Read about how we use cookies.