19
Diabetic Neuropathy: A Position Statement by the American Diabetes Association Diabetes Care 2017;40:136154 | DOI: 10.2337/dc16-2042 Diabetic neuropathies are the most prevalent chronic complications of diabetes. This heterogeneous group of conditions affects different parts of the nervous system and presents with diverse clinical manifestations. The early recognition and appropriate man- agement of neuropathy in the patient with diabetes is important for a number of reasons: 1. Diabetic neuropathy is a diagnosis of exclusion. Nondiabetic neuropathies may be present in patients with diabetes and may be treatable by specic measures. 2. A number of treatment options exist for symptomatic diabetic neuropathy. 3. Up to 50% of diabetic peripheral neuropathies may be asymptomatic. If not recognized and if preventive foot care is not implemented, patients are at risk for injuries to their insensate feet. 4. Recognition and treatment of autonomic neuropathy may improve symptoms, reduce sequelae, and improve quality of life. Among the various forms of diabetic neuropathy, distal symmetric polyneurop- athy (DSPN) and diabetic autonomic neuropathies, particularly cardiovascular au- tonomic neuropathy (CAN), are by far the most studied (14). There are several atypical forms of diabetic neuropathy as well (14). Patients with prediabetes may also develop neuropathies that are similar to diabetic neuropathies (510). Table 1 provides a comprehensive classication scheme for the diabetic neuropathies. Due to a lack of treatments that target the underlying nerve damage, prevention is the key component of diabetes care. Screening for symptoms and signs of diabetic neuropathy is also critical in clinical practice, as it may detect the earliest stages of neuropathy, enabling early intervention. Although screening for rarer atypical forms of diabetic neuropathy may be warranted, DSPN and autonomic neuropathy are the most common forms encountered in practice. The strongest available evidence regarding treatment pertains to these forms. This Position Statement is based on several recent technical reviews, to which the reader is referred for detailed discussion and relevant references to the literature (3,4,1116). PREVENTION Prevention of diabetic neuropathies focuses on glucose control and lifestyle modica- tions. Available evidence pertains only to DSPN and CAN, and most of the large trials that have evaluated the effect of glucose control on the risk of complications have included DSPN and CAN as secondary outcomes or as post hoc analyses rather than as primary outcomes. In addition, in some of these trials, the outcome measures used to evaluate neuropathy may have limited ability to detect a benet, if present. Recommendations c Optimize glucose control as early as possible to prevent or delay the devel- opment of distal symmetric polyneuropathy and cardiovascular autonomic neuropathy in people with type 1 diabetes. A c Optimize glucose control to prevent or slow the progression of distal symmet- ric polyneuropathy in people with type 2 diabetes. B c Consider a multifactorial approach targeting glycemia among other risk factors to prevent cardiovascular autonomic neuropathy in people with type 2 diabetes. C 1 Division of Metabolism, Endocrinology & Diabe- tes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 2 University of Manchester and the Royal Inrmary, Manchester, U.K. 3 Department of Neurology, University of Michigan, Ann Arbor, MI 4 Department of Neurology, University of Toronto, Toronto, Ontario, Canada 5 Department of Neurology, Harvard Medical School, Boston, MA 6 Department of Medicine, Weill Cornell Medicine- Qatar, Doha, Qatar and New York, NY 7 Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL 8 German Diabetes Center D¨ usseldorf, Leibniz Center for Diabetes Research at Heinrich Heine University, and Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, D¨ usseldorf, Germany Corresponding author: Rodica Pop-Busui, rpbusui@ umich.edu. This position statement was reviewed and ap- proved by the American Diabetes Association Professional Practice Committee in September 2016 and ratied by the American Diabetes As- sociation Board of Directors in October 2016. R.P.-B. and A.J.M.B. served as co-chairs of the writing committee. © 2017 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for prot, and the work is not altered. More infor- mation is available at http://www.diabetesjournals .org/content/license. Rodica Pop-Busui, 1 Andrew J.M. Boulton, 2 Eva L. Feldman, 3 Vera Bril, 4 Roy Freeman, 5 Rayaz A. Malik, 6 Jay M. Sosenko, 7 and Dan Ziegler 8 136 Diabetes Care Volume 40, January 2017 POSITION STATEMENT

Diabetic Neuropathy: A Position Statement by the …care.diabetesjournals.org/content/diacare/40/1/136.full.pdf · Diabetic Neuropathy: A Position Statement by the American Diabetes

Embed Size (px)

Citation preview

Diabetic Neuropathy: A PositionStatement by the AmericanDiabetes AssociationDiabetes Care 2017;40:136–154 | DOI: 10.2337/dc16-2042

Diabetic neuropathies are the most prevalent chronic complications of diabetes. Thisheterogeneous group of conditions affects different parts of the nervous system andpresents with diverse clinical manifestations. The early recognition and appropriateman-agement of neuropathy in thepatientwith diabetes is important for a number of reasons:

1. Diabetic neuropathy is a diagnosis of exclusion. Nondiabetic neuropathies maybe present in patients with diabetes and may be treatable by specific measures.

2. A number of treatment options exist for symptomatic diabetic neuropathy.3. Up to 50% of diabetic peripheral neuropathies may be asymptomatic. If not

recognized and if preventive foot care is not implemented, patients are at riskfor injuries to their insensate feet.

4. Recognition and treatment of autonomic neuropathy may improve symptoms,reduce sequelae, and improve quality of life.

Among the various forms of diabetic neuropathy, distal symmetric polyneurop-athy (DSPN) and diabetic autonomic neuropathies, particularly cardiovascular au-tonomic neuropathy (CAN), are by far the most studied (1–4). There are severalatypical forms of diabetic neuropathy as well (1–4). Patients with prediabetes mayalso develop neuropathies that are similar to diabetic neuropathies (5–10). Table 1provides a comprehensive classification scheme for the diabetic neuropathies.Due to a lack of treatments that target the underlying nerve damage, prevention

is the key component of diabetes care. Screening for symptoms and signs of diabeticneuropathy is also critical in clinical practice, as it may detect the earliest stages ofneuropathy, enabling early intervention. Although screening for rarer atypical formsof diabetic neuropathy may be warranted, DSPN and autonomic neuropathy are themost common forms encountered in practice. The strongest available evidenceregarding treatment pertains to these forms.This Position Statement is based on several recent technical reviews, towhich the reader

is referred for detailed discussion and relevant references to the literature (3,4,11–16).

PREVENTION

Prevention of diabetic neuropathies focuses on glucose control and lifestyle modifica-tions. Available evidence pertains only to DSPN and CAN, and most of the large trialsthat have evaluated the effect of glucose control on the risk of complications haveincluded DSPN and CAN as secondary outcomes or as post hoc analyses rather than asprimary outcomes. In addition, in some of these trials, the outcome measures used toevaluate neuropathy may have limited ability to detect a benefit, if present.

Recommendations

c Optimize glucose control as early as possible to prevent or delay the devel-opment of distal symmetric polyneuropathy and cardiovascular autonomicneuropathy in people with type 1 diabetes. A

c Optimize glucose control to prevent or slow the progression of distal symmet-ric polyneuropathy in people with type 2 diabetes. B

c Consider amultifactorial approach targeting glycemia among other risk factors toprevent cardiovascular autonomic neuropathy in people with type 2 diabetes. C

1Division ofMetabolism, Endocrinology & Diabe-tes, Department of Internal Medicine, Universityof Michigan, Ann Arbor, MI2University of Manchester and the Royal Infirmary,Manchester, U.K.3Department of Neurology, University of Michigan,Ann Arbor, MI4Department of Neurology, University of Toronto,Toronto, Ontario, Canada5Department of Neurology, Harvard MedicalSchool, Boston, MA6Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar and New York, NY7Division of Endocrinology, University of MiamiMiller School of Medicine, Miami, FL8German Diabetes Center Dusseldorf, LeibnizCenter for Diabetes Research at Heinrich HeineUniversity, and Department of Endocrinologyand Diabetology, Medical Faculty, HeinrichHeine University, Dusseldorf, Germany

Corresponding author: Rodica Pop-Busui, [email protected].

This position statement was reviewed and ap-proved by the American Diabetes AssociationProfessional Practice Committee in September2016 and ratified by the American Diabetes As-sociation Board of Directors in October 2016.

R.P.-B. and A.J.M.B. served as co-chairs of thewriting committee.

© 2017 by the American Diabetes Association.Readers may use this article as long as the workis properly cited, the use is educational and notfor profit, and the work is not altered. More infor-mation is available at http://www.diabetesjournals.org/content/license.

Rodica Pop-Busui,1 Andrew J.M. Boulton,2

Eva L. Feldman,3 Vera Bril,4 Roy Freeman,5

Rayaz A. Malik,6 Jay M. Sosenko,7 and

Dan Ziegler8

136 Diabetes Care Volume 40, January 2017

POSITION

STATEMEN

T

Glucose ControlEnhanced glucose control in people withtype 1 diabetes dramatically reduces theincidence of DSPN (78% relative risk re-duction) (17–19). In contrast, enhancedglucose control in people with type 2 di-abetes reduces the risk of developingDSPN modestly (5%–9% relative risk re-duction) (20,21). In a small trial of Japa-nese patients with early type 2 diabetes,intensive insulin treatment was associ-ated with improvement in selectedDSPN measures (22), and the Action toControl Cardiovascular Risk in Diabetes(ACCORD) trial reported a modest butsignificant DSPN risk reduction withthe glycemia intervention in individualswith type 2 diabetes after 5 years offollow-up (21). Yet, no effects are ob-served in other large trials (20,23–25).This discrepancy highlights the differ-ences between type 1 and type 2 diabe-tes and emphasizes the point that many

people with type 2 diabetes developDSPN despite adequate glucose control(20,25). The presence of multiple comor-bidities, polypharmacy, hypoglycemia,and weight gain might have attenuatedthe effects of glucose control in thesetrials and contributed to inconsistentfindings (25). Specific glucose-loweringstrategies may also contribute to the dis-crepancy. For example, participants, par-ticularly men, in the Bypass AngioplastyRevascularization Investigation in Type 2Diabetes (BARI 2D) study treated with in-sulin sensitizers had a lower incidence ofDSPN over 4 years than those treatedwith insulin/sulfonylurea (26). This out-come may be a result of less weight gainand less hypoglycemia (26). Last, the factthat many patients have had asymptom-atic hyperglycemia for many years priorto the diagnosis of type 2 diabetes mayalso explain the limited benefit in thesepatients.

Similar to the findings in DSPN, themost robust evidence for CAN preven-tion was reported in type 1 diabetes.Intensive glucose control designed toachieve near-normal glycemia reducedthe risk of incident CAN during the Di-abetes Control and Complications Trial(DCCT) by 45% andby 31% in its follow-upstudy, the Epidemiology of Diabetes In-terventions and Complications (EDIC)study (27). The highly reproducible andsensitive testing protocol, the robustdefinitions used for CAN, and the largesample size in DCCT/EDIC enhance thevalidity of the results and support therationale for implementing and main-taining tight glucose control as early aspossible in the course of type 1 diabetes.In contrast, glycemic control in type 2diabetes has not consistently loweredthe risk of CAN (25). However, a multi-factorial intervention, including a life-style component, targeting glucose andcardiovascular disease risk factors re-duced the risk of CAN by 60% in peoplewith type 2 diabetes (28).

Lifestyle ModificationsThe best models to date regarding pa-rameters for an evidence-based, inten-sive lifestyle intervention come from theDiabetes Prevention Program (DPP)(29), the Steno-2 Study (28), the Italiansupervised treadmill study (30), and theUniversity of Utah type 2 diabetes study(31). The latter study recently reportednerve fiber regeneration in patients withtype 2 diabetes engaged in an exerciseprogram compared with loss of nerve fi-bers in those who only followed standardof care. Overall, such an approach focuseson either exercise alone (supervised aer-obic and/or resistance training) (30,31) orcombined dietary modification and exer-cise. There is no consensus regarding di-etary regimens, and although the DPPused a low-calorie, low-fat diet, othershave championed a Mediterranean dietthat is moderately lower in carbohydrate(45%) and higher in fat (35%–40%), withless than 10% of saturated fat.

Although theDPP (32) and the ImpairedGlucose Tolerance Neuropathy (IGTN)study (33) reported benefits of lifestyleinterventions on measures of CAN andDSPN, respectively, these trials did not in-clude subjects with established diabetes.In addition, in the DPP, indices of CANimproved with the lifestyle interventionand did not change in the other arms (32).

Table 1—Classification for diabetic neuropathies

Diabetic neuropathiesA. Diffuse neuropathyDSPN

c Primarily small-fiber neuropathyc Primarily large-fiber neuropathyc Mixed small- and large-fiber neuropathy (most common)

AutonomicCardiovascular

c Reduced HRVc Resting tachycardiac Orthostatic hypotensionc Sudden death (malignant arrhythmia)

Gastrointestinalc Diabetic gastroparesis (gastropathy)c Diabetic enteropathy (diarrhea)c Colonic hypomotility (constipation)

Urogenitalc Diabetic cystopathy (neurogenic bladder)c Erectile dysfunctionc Female sexual dysfunction

Sudomotor dysfunctionc Distal hypohydrosis/anhidrosis,c Gustatory sweating

Hypoglycemia unawarenessAbnormal pupillary function

B. Mononeuropathy (mononeuritis multiplex) (atypical forms)Isolated cranial or peripheral nerve (e.g., CN III, ulnar, median, femoral, peroneal)Mononeuritis multiplex (if confluent may resemble polyneuropathy)

C. Radiculopathy or polyradiculopathy (atypical forms)Radiculoplexus neuropathy (a.k.a. lumbosacral polyradiculopathy, proximal motor

amyotrophy)Thoracic radiculopathy

Nondiabetic neuropathies common in diabetesPressure palsiesChronic inflammatory demyelinating polyneuropathyRadiculoplexus neuropathyAcute painful small-fiber neuropathies (treatment-induced)

care.diabetesjournals.org Pop-Busui and Associates 137

DSPN

Most common among diabetic neurop-athies is chronic DSPN, accounting forabout 75% of the diabetic neuropathies(1,3). A simple definition of DSPN forclinical practice is the presence of symp-toms and/or signs of peripheral nervedysfunction in people with diabetes af-ter the exclusion of other causes.Experimental studies suggest a mul-

tifactorial pathogenesis of DSPN (Fig. 1),but the causes remain unknown(34–37). A prevailing view of the patho-genesis is that oxidative and inflamma-tory stress may, in the context ofmetabolic dysfunction, damage nervecells (34–37).Estimates of the incidence and prev-

alence of DSPN vary greatly (25,38–40),but evidence from several large obser-vational cohorts (41,42) and the DCCT/EDIC (27,43) suggests that DSPN occursin at least 20% of people with type 1 di-abetes after 20 years of disease dura-tion. DSPN may be present in at least10%–15% of newly diagnosed patientswith type 2 diabetes (44,45), with ratesincreasing to 50% after 10 years of diseaseduration (25,26). Rates in youth withtype 1 and type 2 diabetes approach those

observed in adult populations (46).DSPN has been associated with glyce-mia (14,33–35), height (47) (perhapsas a proxy for nerve length), smoking(48), blood pressure, weight, and lipidmeasures (49,50).

There is emerging evidence thatDSPN, especially the painful small-fiberneuropathy subtype, may be present in10%–30% of subjects with impaired glu-cose tolerance, also known as prediabe-tes (5–10) or metabolic syndrome (51).

DSPN is the most important cause offoot ulceration, and it is also a prereq-uisite in the development of Charcotneuroarthropathy (CN) (52). The readeris referred to several other reviews thatcover this topic (52,53). Foot ulcerationand CN are both recognized as late com-plications of DSPN (52,54). These latecomplications drive amputation riskand economic costs of diabetic neurop-athy and are also predictors of mortality.

DSPN is also a major contributor tofalls and fractures (55–57), throughmore advanced small- and large-fiberdysfunction, with loss of sensory, pro-prioception, temperature discrimina-tion, and pain, all ultimately leading tounsteadiness, recurrent minor injuries,

and an increased risk of falls. These re-current minor injuries may further con-tribute to the pathogenesis of CN (58).

Screening and Diagnosis

Recommendations

c All patients should be assessed fordistal symmetric polyneuropathystarting at diagnosis of type 2 di-abetes and 5 years after the diag-nosis of type 1 diabetes and atleast annually thereafter. B

c Consider screening patients withprediabetes who have symptomsof peripheral neuropathy. B

c Assessment should include a care-ful history and either temperatureor pinprick sensation (small-fiberfunction) and vibration sensationusinga128-Hz tuning fork (large-fiberfunction). All patients should have anannual 10-g monofilament testing toassess for feet at risk for ulcerationand amputation. B

c Electrophysiological testing or re-ferral to a neurologist is rarelyneeded for screening, except insituations where the clinical fea-tures are atypical, the diagnosis isunclear, or a different etiology issuspected. Atypical features in-clude motor greater than sensoryneuropathy, rapid onset, or asym-metrical presentation. B

Patients with type 1 diabetes for 5 ormore years and all patients with type 2diabetes should be assessed annuallyfor DSPN using medical history andsimple clinical tests. Up to 50% of pa-tients may experience symptoms ofDSPN (Table 2), whereas the rest areasymptomatic. Patients may not volun-teer symptoms but on inquiry may revealthat they are experiencing numbness orother positive symptoms of DSPN.

Symptoms vary according to the classof sensory fibers involved. The mostcommon early symptoms are inducedby the involvement of small fibers andinclude pain and dysesthesias (unpleas-ant sensations of burning) (1,4,59,60).Neuropathic pain may be the first symp-tom that prompts patients to seek med-ical care and is present in up to 25% ofindividuals with DSPN (61–63). Charac-teristically, the pain is burning, lancinating,tingling, or shooting (electric shock–like);occurs with paresthesias; presents in

Figure 1—Mechanisms of diabetic neuropathy. Factors linked to type 1 diabetes (yellow), type 2diabetes (blue), and both (green) cause DNA damage, endoplasmic reticulum stress, mitochon-drial dysfunction, cellular injury, and irreversible damage. The relative importance of the path-ways in this network will vary with cell type, disease profile, and time. ER, endoplasmicreticulum; FFA, free fatty acids; PI3-K, phosphatidylinositol-3 kinase; RNS, reactive nitrogenspecies; ROS, reactive oxygen species. Adapted and reprinted from Callaghan et al. (20), withpermission from Elsevier.

138 Position Statement Diabetes Care Volume 40, January 2017

varying combinations; and is typicallyworse at night. Neuropathic pain may beaccompanied by an exaggerated responseto painful stimuli (hyperalgesia) and painevoked by contact, e.g., with socks, shoes,and bedclothes (allodynia). Neuropathicpain can lead to interference with dailyactivities, disability, psychosocial impair-ment, and reduced health-related qualityof life (64–66). The direct and indirect eco-nomic burden associated with neuro-pathic pain is substantial (67–69).The involvement of large fibers may

cause numbness, tingling without pain,and loss of protective sensation. Loss ofprotective sensation indicates the pres-ence of DSPN and is a risk factor for di-abetic foot ulceration. Patients can alsoinitially present with an insensate, numbfoot due to the loss of largefibers. Patientsfrequently state that their feet feel likethey arewrapped inwool or they arewalk-ing on thick socks. It is the loss of the “giftof pain” that permits patients with plantarneuropathic ulcers to walk on the lesions,inducing chronicity, frequently complicatedby infection (70).The following clinical tests may be

used to assess small- and large-fiberfunction distal to proximal (Table 2):

1. Small-fiber function: pinprick andtemperature sensation

2. Large-fiber function: vibration per-ception, proprioception, 10-g mono-filament, and ankle reflexes

A 128-Hz tuning fork can be used forthe assessment of vibration perception.Assessment of light-touch perceptionusing a 10-g monofilament should includeevaluation on the dorsal aspect of thegreat toe bilaterally as previously validatedby Perkins et al. (71). The 10-g monofila-ment is a useful clinical tool mainly fordetecting more advanced neuropathyand identifying patients at increased riskof ulceration and amputation (72).

Assessments should follow the typicalDSPN pattern, starting distally (the dor-sal aspect of the hallux) on both sidesand move proximally until a sensorythreshold is identified (72). Combiningat least two examinations will increasethe sensitivity and specificity of detect-ing DSPN, as demonstrated in severalcohorts of patients with type 1 andtype 2 diabetes including children andadolescents (26,46,73–79).

The diagnosis of DSPN is principally aclinical one (Table 2). A combination of

typical symptomatology and symmetrical

distal sensory loss or typical signs in the

absence of symptoms in a patient with

diabetes is highly suggestive of DSPNandmay not require additional evaluationor referral. As up to half of the patientsmay be asymptomatic, a diagnosis mayonly be made on examination or, insome cases, when the patient presentswith a painless foot ulcer.

Clinicians should note that the 10-gmonofilament test included for theannual DSPN screening and diagnosisis different than the diagnosis of the“high-risk foot” for ulceration, a lateDSPN complication that requires thatfour sites (first, third, and fifth metatarsalheads and plantar surface of distal hallux)be tested on each foot (80).

Consider excluding neuropathy withcauses other than diabetes (Table 3) byundertaking a family andmedication historyand performing relevant investigations (e.g.,serum B12, folic acid, thyroid function, com-plete blood count, metabolic panel, and aserumprotein immunoelectrophoresis) (81).

Electrophysiological testing or referralto a neurologist is rarely needed for di-agnosis, except in situations where theclinical features are atypical, the diagnosisis unclear, or a different etiology is sus-pected (2,38,40,80). Atypical features thatwarrant referral includemotor greater thansensory neuropathy, asymmetry of symp-toms and signs, and rapid progression.

Foot ComplicationsThe simple yet comprehensive clinicalexam is principally designed to identifythose at risk for the late complicationswho need education on preventativefoot self-care and regular podiatricfoot care. Recently an even simpler

Table 2—Symptoms and signs of DSPN

Large myelinated nerve fibers Small myelinated nerve fibers

Function Pressure, balance Nociception, protective sensation

Symptoms§ Numbness, tingling, poor balance Pain: burning, electric shocks, stabbing

Examination(clinically diagnostic)**

Ankle reflexes: reduced/absentVibration perception: reduced/absent10-g monofilament: reduced/absentProprioception: reduced/absent

Thermal (cold/hot) discrimination: reduced/absent**Pinprick sensation: reduced/absent**

§To document the presence of symptoms for diagnosis; **Documented in symmetrical, distal to proximal pattern.

Table 3—Differential diagnosis ofdiabetic neuropathiesMetabolic diseaseThyroid disease (common)Renal disease

Systemic diseaseSystemic vasculitisNonsystemic vasculitisParaproteinemia (common)Amyloidosis

InfectiousHIVHepatitis BLyme

InflammatoryChronic inflammatory demyelinating

polyradiculoneuropathy

NutritionalB12*PostgastroplastyPyridoxineThiamineTocopherol

Industrial agents, drugs, and metalsIndustrial agents

AcrylamideOrganophosphorous agents

DrugsAlcoholAmiodaroneColchicineDapsoneVinka alkaloidsPlatinumTaxol

MetalsArsenicMercury

HereditaryHereditary motor, sensory, and

autonomic neuropathies

*B12 deficiency is more commonlyassociated with malabsorption rather thannutritional deficiency.

care.diabetesjournals.org Pop-Busui and Associates 139

foot exam, the “3-minute diabetic footexam,” has been proposed (82). This isintended not only for physicians but alsofor other health care professionals whomay only have 15 min for the entire dia-betes annual review; it requires no equip-ment and provides simple advice oneducation on preventative foot self-care.

Management

Recommendations

c Tight glucose control targetingnear-normal glycemia in patientswith type 1 diabetes dramaticallyreduces the incidence of distalsymmetric polyneuropathy and isrecommended for distal symmet-ric polyneuropathy prevention intype 1 diabetes. A

c In patients with type 2 diabeteswith more advanced disease andmultiple risk factors and comorbid-ities, intensive glucose controlalone is modestly effective in pre-venting distal symmetric polyneur-opathy and patient-centered goalsshould be targeted. B

c Lifestyle interventions are recom-mended for distal symmetric poly-neuropathy prevention in patientswith prediabetes/metabolic syn-drome and type 2 diabetes. B

Prevention

Please refer to PREVENTION on page 136.

Pathogenetic TherapiesDespite the recent major advances inelucidating the pathogenesis of diabeticneuropathy, there remains a lack oftreatment options that effectively tar-get the natural history of DSPN (83) orreverse DSPN once established. Severalpathogenetic pharmacotherapies havebeen investigated (36), but evidencefrom randomized clinical trials is verylimited (81,83,84). Advances in DSPNdisease modification need to be con-firmed with further robust evidencefrom clinical trials, together with a bet-ter understanding of the mechanisms ofaction of promising treatments (83).

Pain Management

Recommendations

c Consider either pregabalin orduloxetine as the initial approachin the symptomatic treatment forneuropathic pain in diabetes. A

c Gabapentin may also be used asan effective initial approach, takinginto account patients’ socioeco-nomic status, comorbidities, andpotential drug interactions. B

c Although not approved by the U.S.Food and Drug Administration, tri-cyclic antidepressants are also ef-fective for neuropathic pain indiabetes but should be used withcaution given the higher risk of se-rious side effects. B

c Given the high risks of addictionand other complications, the useof opioids, including tapentadolor tramadol, is not recommendedas first- or second-line agents fortreating the pain associated withDSPN. E

No compelling evidence exists in supportof glycemic control or lifestyle manage-ment as therapies for neuropathic pain indiabetes or prediabetes (33,85), whichleaves only pharmaceutical interventions.

At present, pregabalin and duloxetinehave received regulatory approval for thetreatment of neuropathic pain in diabetesby the U.S. Food and Drug Administration(FDA), Health Canada, and the EuropeanMedicines Agency. The opioid, tapentadol,has regulatory approval in the U.S. andCanada, but the evidence of its use isweaker (15).

A large evidence base supports phar-macological treatment of neuropathicpain in diabetic neuropathy using otheragents of different classes, as docu-mented by several recent guidelinesand systematic reviews (15,16,20,86,87).It is important to mention that only afew trials that targeted pain in peripheralneuropathic painwere carried out in DSPNalone. However, the results of studies per-formed on peripheral nondiabetic neuro-pathic pain ormixed neuropathic painmaybe applicable to patients with neuropathicpain due to DSPN.

Although there are broad generalagreements among the recommenda-tions, there are some inconsistencies thatare, in part, a consequence of whether theguidelines are specific for painful DSPN orwhether they address neuropathic paindue to all causes (15,16,20,86,87).

Below we summarize the available ev-idence on the most effective agents forDSPN pain starting with the currently ap-proved drugs and continuing with the

other agents based on mechanism of ac-tion and strength of evidence. Evidencelevels are assigned based on the strengthof the published clinical evidence for theefficacy and safety of the agents for thetreatment of DSPN pain, which should beconsidered in clinical decision making.However, a certain degree of publicationbias should be considered, given thatmany negative trials may not have beenpublished (15).

Additional information on dose titra-tion, adverse effects, number needed totreat, and safety is presented in Table 4.

Approved Medications

Pregabalin and duloxetine have re-ceived regulatory approval for the treat-ment of neuropathic pain in diabetes inthe U.S., Europe, and Canada.

Pregabalin, a calcium channel a2-dsubunit ligand, is an effective treatmentfor neuropathic pain associated withDSPN. It is the most extensively studieddrug by far in DSPN, with the majorityof studies being positive regardingthe proportion of responders with atleast 30%–50% improvement in pain(15,86,88–94). There is also some evi-dence suggesting a dose response, with aweaker effect with 300 vs. 600 mg/day(88). However, not all trials with pregaba-lin have been positive (15,86,95,96), espe-cially when treating advanced refractorypatients (93). Pregabalin, in contrast togabapentin (see below), has a linear,dose-proportional absorption in the thera-peutic dose range (150–600 mg/day)(88). In addition, pregabalin has a morerapid onset of action and more limiteddosage range that requiresminimal titra-tion. Adverse effects may be more se-vere in older patients (97) and may beattenuated by lower starting doses andmore gradual titration.

Duloxetine is a selective norepineph-rine and serotonin reuptake inhibitor.Doses of 60 and 120 mg/day showed ef-ficacy in the treatment of pain associatedwith DSPN in multicenter randomized tri-als, although some of these had a ratherhigh drop-out rate (15,86,94,96,98–101).Duloxetine was also suggested to in-duce improvement in neuropathy-relatedquality of life (100). In longer-term stud-ies, a small increase in A1C was reportedin people with diabetes treated with du-loxetine compared with placebo (102).Adverse eventsmay again bemore severein older people but may be attenuated

140 Position Statement Diabetes Care Volume 40, January 2017

Table

4—Treatm

entforpain

associatedwithDSP

N(15,16,20,86,87)

Drugclass

Agent

Dose

NNTrange

30–50

%im

provemen

t**

Commonadverse

even

tsMajoradverse

even

tsInitial

Effective

Anticonvulsan

ts

Pregabalin*(15,86

,88–

94)

25–75

mg,1–

33/day

300–60

0mg/day

3.3–

8.3

cSomnolence

cAngioed

ema

cDizziness

cHep

atotoxicity

cPeripheraled

ema

cRhab

domyolysis

cHeadache

cSuicidalthoughtsandbeh

avior

cAtaxia

cSeizuresafterrapid

discontinuation

cFatigue

cThrombocytopen

ia

cXerostomia

cWeigh

tgain

Gabap

entin(15,86

,96,10

5–11

1)100–300mg,1–33

/day

900–3,60

0mg/day

3.3–

7.2

cSomnolence

cSteven

s-Johnsonsyndrome

cDizziness

cSuicidalthoughtsandbeh

avior

cAtaxia

cSeizuresafterrapid

discontinuation

cFatigue

Antidep

ressants

Serotonin-norepinephrine

reuptake

inhibitors

Duloxetine*

(15,86

,94,96

,98–10

1)20

–30

mg/day

60–12

0mg/day

3.8–

11cNausea

cSteven

s-Johnsonsyndrome

cSomnolence

cHep

atotoxicity

cDizziness

cHypertensive

crisis

cConstipation

cGastrointestinalhem

orrhage

cDyspep

sia

cDelirium

cDiarrhea

cMyocardialinfarction

cXerostomia

cCardiacarrhythmias

cAnorexia

cGlaucoma

cHeadache

cSuicidalthoughtsandbeh

avior

cDiaphoresis

cShiftto

maniain

patients

with

bipolardisorder

cInsomnia

cSeizures

cFatigue

cSevere

hyponatremia

cDecreased

libido

cFragility

bonefractures

cSerotonin

syndrome

cNeurolepticmalignant

syndrome

Ven

lafaxine(15,16

,20,86

,87,126

,127

)37

.5mg/day

75–22

5mg/day

5.2–

8.4

cNausea

cSameas

duloxetine

cSomnolence

cDizziness

cConstipation

cDyspep

sia

cDiarrhea

cXerostomia

cAnorexia

Con

tinuedon

p.14

2

care.diabetesjournals.org Pop-Busui and Associates 141

Table

4—Continued

Drugclass

Agent

Dose

NNTrange

30–50

%im

provemen

t**

Commonadverse

even

tsMajoradverse

even

tsInitial

Effective

cHeadache

cDiaphoresis

cInsomnia

cFatigue

cDecreased

libido

Tricyclic

antidep

ressants

Amitriptyline(16,11

0,11

2–11

6)10

–25

mg/day

25–10

0mg/day

2.1–

4.2

cXerostomia

cDelirium

cSomnolence

cCardiacarrhythmias

cFatigue

cConductionabnorm

alities

cHeadache

cMyocardialinfarction

cDizziness

cHeartfailure

exacerbation

cInsomnia

cStroke

cOrthostatichypotension

cSeizures

cAnorexia

cHep

atotoxicity

cNausea

cBonemarrowsuppression

cUrinaryretention

cSuicidalthoughtsandbeh

avior

cConstipation

cShiftto

maniain

bipolardisorder

cBlurred

vision

cNeu

rolepticmalignan

tsyndrome

cAccommodation

cSerotonin

syndrome

cDisturbance

cSevere

hyponatremia

cMydriasis

cFragility

bonefractures

cWeigh

tgain

Desipramine(113

,118

–12

1,12

2)cSameas

above

cSameas

above

Nortriptyline

(15,16

,86,87

,113

,114

,120

,121

,123

)cSameas

above

cSameas

above

Opioids

Tram

adol(15

,16,86

,87,10

9,13

0)50

mg,1–

23/day

210mg/day

3.1–

6.4

cSomnolence

cConfusion

cNausea

cSeizures

cVomiting

cCardiacarrhythmias

cConstipation

cHypertension

cLigh

t-headed

ness

cHypersensitivity

reactions

cDizziness

cSteven

s-Johnsonsyndrome

cHeadache

Tapen

tadol*(103

,104

,135

)Im

med

iate

release:

50–10

0mg,

4–63

/day

Immediate-release:d

ay1:

700mg;afterday1,

60mg/day

N/A

cSomnolence

cRespiratory

dep

ression

cNausea

cSerotonin

syndrome

Extended

release:

50mg,23

/day

Extended

release:

50mg,23

/day

cVomiting

cSeizures

cConstipation

cHypertension

cDizziness

cNeo

natalopioid

withdrawal

syndrome

NNT,number

needed

totreat.*FDAapproved.**FD

Aconsiders30

–50

%im

provemen

tto

besign

ificant.

142 Position Statement Diabetes Care Volume 40, January 2017

with lower doses and progressive titra-tions of duloxetine.Tapentadol extended release is a

novel centrally acting opioid analgesicthat exerts its analgesic effects throughboth m-opioid receptor agonism andnoradrenaline reuptake inhibition.Extended-release tapentadol was ap-proved by the FDA for the treatmentof neuropathic pain associated withdiabetes based on data from two multi-center randomized withdrawal, placebo-controlledphase3 trials (103,104).However,both used an enriched design and there-fore are not generalizable, and a recentsystematic review and meta-analysis bythe International Association for theStudy of Pain Special Interest Group onNeuropathic Pain (NeuPSIG) found theevidence of the effectiveness of tapenta-dol in reducing neuropathic pain incon-clusive (15). Therefore, given the highrisk for addiction and safety concernscompared with the relatively modestpain reduction, the use of tapentadolextended release is not recommendedas first- or second-line treatment.

Anticonvulsants

Gabapentin, like pregabalin, also bindsthe calcium channel a2-d subunit andhas shown efficacy in a number of clin-ical trials for treating the pain associatedwith DSPN (15,86,96,105–111). How-ever, not all painful DSPN studies,some of which are unpublished, havebeen positive (15,107).Given its pharmacokinetic profile,

gabapentin requires gradual titrationand doses up to 1,800–3,600 mg are gen-erally needed to be clinically effective(96,105–107). Adverse effects may bemore severe in older patients (97).

Monoamine Reuptake Inhibitors

The monoamine reuptake inhibitorsdtricyclic antidepressants, selectiveserotoninreuptake inhibitors, and norepinephrineand serotonin reuptake inhibitorsdincrease synaptic monoamine levels anddirectly influence the activity of the de-scending neurons.Amitriptyline, although not FDA ap-

proved, is the most used of the tricyclicagents. Many previous guidelines rec-ommend the medication as a first-linetreatment based on few randomized,blinded, placebo-controlled clinical tri-als that reported significant improvementin neuropathic pain (110,112–116). Theeffectiveness appeared unrelated to the

antidepressant effect (112). A recent Co-chrane Review questioned the quality ofevidence on amitriptyline by raising con-cerns for bias given the small sample sizein most and concluded that in fact thereis no clear evidence for a beneficial effectfor amitriptyline on DSPN pain, especiallywhen balanced against spectrum of sideeffects (117). However, there was nogood evidence of a lack of effect either(117).

The secondary amines, nortriptylineand desipramine, have a less trouble-some side effect profile than the tertiaryamines, amitriptyline and imipramine,although fewer randomized controlledtrials were performed with these agents,and the potential for bias was high giventhe small size (113,118–123). The use ofthese agents is preferable, particularly inolder and side effect–prone patients(113,118–121).

Several studies have suggested thatthere is an increased risk of myocardialischemia and arrhythmogenesis associ-atedwith tricyclic agents (124,125).Becauseof concerns of possible cardiotoxicity, tricy-clic antidepressants should be used withcaution inpatientswithknownor suspectedcardiac disease.

Venlafaxine, a selective norepineph-rine and serotonin reuptake inhibitor,in doses between 150 and 225 mg/dayhas shown some effectiveness in thetreatment of painful DSPN (126,127).Both venlafaxine and duloxetine (seeabove) inhibit the reuptake of serotoninand norepinephrine without the musca-rinic, histaminic, and adrenergic side ef-fects that accompany the use of thetricyclic agents (98–100,102). However,the level of evidence for pain reductionassociated with DSPN is higher with du-loxetine (see above). Venlafaxine maylower the seizure threshold, and gradualtapering is recommended to avoid theemergence of adverse events upon dis-continuation (126,127).

Opioid and Atypical Opioid Analgesics

Tramadol is a centrally acting analgesicwith pain relief mediated by a weakm-opioid receptor agonist activity andinhibition of norepinephrine and seroto-nin reuptake (128,129). It is an effectiveagent in the treatment of painful dia-betic peripheral neuropathy comparedwith placebo as demonstrated by twolarge multicenter trials (129,130), andit appears to have long-term effects

(131). Although tramadol has a lowerpotential for abuse compared with otheropioids, given these safety concerns, itis not recommended for use as first- orsecond-line agent.

Controlled-release oxycodone im-proved pain scores in two single-centertrials in patients with painful diabeticneuropathy, one of which had a smallsample size (132,133). It may provideadditional analgesia for patients on a2-dligand treatment (134). As with all opioids,it is not recommended for use as first-,second-, or third-line agent.

Warnings on All Opioids

Despite the demonstrated effectivenessof opioids in the treatment of neuro-pathic pain (15,132,134,135), there is ahigh risk of addiction, abuse, sedation,and other complications and psychoso-cial issues even with short-term opioiduse. For these reasons, opioids are notrecommended in the treatment of pain-ful DSPN before failure of other agentsthat do not have these associated con-cerns (136–138).

Although add-on therapy with strongopioids may be required in some pa-tients who do not respond to all othercombinations, referral to specialized painclinics is recommended in these cases toavoid risks.

Additional Considerations for Pain

Management

Combination therapy, including combi-nations with opioids, may provide effec-tive treatment for diabetic neuropathicpain at lower doses (94,139). A detailedapproach for pain management is amplycovered in other literature (15,109),and a simple algorithm for clinical prac-tice use is shown in Fig. 2.

Treatment of Foot ComplicationsDetailed treatment of foot ulcerationand CN is beyond the scope of this state-ment, and the reader is referred to arelevant review (54). Effective off-loadingthat prevents patients with plantar neu-ropathic ulcers to walk on the lesions isthe key to successful management(52,54). Off-loading, usually with casting,and careful follow-up and repeated in-vestigations are also key componentsfor the management of CN (52,54). On-going education and regular podiatry fol-low-up can reduce the incidence of footcomplications in those found to be at“high risk.” Early intervention for foot

care.diabetesjournals.org Pop-Busui and Associates 143

lesions and CN or suspected CN can slowor reverse progression.

Fall Prevention

Recommendation

c Tests assessing gait and balancemay be considered in people withdistal symmetric polyneuropathyto evaluate the risk of falls. E

DSPN may also compromise balance indaily activities (58). For instance, pro-gressive loss of proprioception (dimin-ished sensation) and later weakness,superimposed on age-related functionalimpairments, lead to imbalance and un-steadiness in gait, with increased like-lihood of a fall (55,58). A decline incognitive function, polypharmacy, andneuropathic pain may further contribute.In addition, treatment of neuropathicpain often requires dosages and drugcombinations that may further increasethe fall risk due to cognitive impair-ment, drowsiness, dizziness, blurred vi-sion, and gait disturbances (97,109).Older patients are the most susceptible(97,109). Therefore, tests assessing gaitand balance may be considered in

clinical practice to evaluate risk of fallsin patients at risk (55,58).

Psychosocial Factors

Recommendations

c Consider treatment with duloxe-tine, pregabalin, and gabapentin toimprove quality of life in patientswith neuropathic pain. C

c Assess the effects of distal symmet-ric polyneuropathy on quality of lifeto improve adherence and responseto neuropathic pain treatment. E

Assessing the effects of DSPN on a pa-tient’s quality of life is emerging as acomponent of patient care and mayplay an important part in the adher-ence and the response to therapiesin patients with neuropathic pain(140). Some studies report an improve-ment in quality of life in people withpainful DSPN treated with duloxetine(100), pregabalin (141), and gabapentin(106,142). A longitudinal study hasshown that DSPN is a risk factor for de-pression and the strongest symptomassociated with depression was un-steadiness. Pain with DSPN may also

give rise to symptoms of anxiety(143). Two research tools that can beused to assess quality of life that areneuropathy specific are the Neuro-QoL (Quality of Life in Neurological Dis-orders) (144) and QOL-DN (NorfolkQuality of Life-Diabetic Neuropathy)instruments (145).

DIABETIC AUTONOMICNEUROPATHIES

Autonomic neuropathies affect the auto-nomic neurons (parasympathetic, sympa-thetic, or both) and are associated with avariety of site-specific symptoms. Thesymptoms and signs of autonomic dys-function should be elicited carefully dur-ing the medical history and physicalexamination. Major clinical manifesta-tions of diabetic autonomic neuropathyinclude hypoglycemia unawareness, rest-ing tachycardia, orthostatic hypotension,gastroparesis, constipation, diarrhea, fe-cal incontinence, erectile dysfunction,neurogenic bladder, and sudomotor dys-function with either increased or de-creased sweating.

Although CAN is themost studied andclinically relevant of the diabetic auto-nomic neuropathies, gastrointestinal,genitourinary, and sudomotor dysfunc-tion should be considered in the optimalcare of patients with diabetes.

CANAlthough CAN prevalence is very low innewly diagnosed patients with type 1diabetes (146), CAN prevalence in-creases substantially with diabetes du-ration (13,25), and prevalence rates ofat least 30% were observed in theDCCT/EDIC cohort after 20 years of di-abetes duration (27,147). In type 2 di-abetes, the prevalence of CAN alsoincreases with diabetes duration andmay be present in up to 60% of patientswith type 2 diabetes after 15 years(13,148,149). CAN may affect youth, es-pecially young women and those withelevated A1C levels, with prevalencerates of at least 20% reported in youthwith type 1 or type 2 diabetes (150). Inaddition, CAN is present in patients withimpaired glucose tolerance, insulinresistance, or metabolic syndrome(10,32,151).

A timely diagnosis of CAN may haveimportant clinical implications, as CAN isan independent risk factor for cardio-vascular mortality, arrhythmia, silent

Figure 2—Algorithm for management of the patient with pain because of DSPN. AE, adverseevents.*Pregabalin is FDA approved for painful DSPN, whereas gabapentin is not. Pharmaco-kinetic profile, spectrum of AEs, drug interactions, comorbidities, and costs to be consideredin selecting the agent of choice. **Duloxetine is FDA approved for painful DSPN, whereasvenlafaxine is not. Pharmacokinetic profile, spectrum of AEs, drug interactions, comorbidities,and costs to be considered in selecting the agent of choice. #None is FDA approved for painfulDSPN. Spectrum of AEs, drug interactions, and comorbidities need be considered if selectingthese agents.

144 Position Statement Diabetes Care Volume 40, January 2017

ischemia, any major cardiovascularevent, and myocardial dysfunction(152–157). Data from two large cardio-vascular outcomes trials that included31,531 patients with stable heart dis-ease and/or diabetes followed for a me-dian of 5 years reported that heart rate,an indirect measure of CAN, analyzed aseither categorical (baseline heart rate.70 vs. #70 bpm) or across heart ratequintile, was independently associatedwith significant increases in cardiovas-cular disease (CVD) events and all-causedeath (158). CANmay also be associatedwith hemodynamic instability or cardio-respiratory arrest (159). CAN was thestrongest risk factor for mortality in alarge cohort of patients with type 1 di-abetes participating in the EURODIABProspective Cohort Study (160), and ameta-analysis of several trials reportedhigher mortality risk with worse mea-sures of CAN (152).Conclusive evidence that supports

CAN as an independent predictor ofmortality was confirmed in more than8,000 participants with type 2 diabetesin the ACCORD trial (154). Hazard ratiosfor all-cause and CVD mortality in thosewith CAN were as high as 2.14 after ad-justing for all traditional CVD risk factors

and many other risk factors, includinguse of various classes of medicationuse (154).

It was also suggested that intensifi-cation of glucose and blood pressuremanagement may increase the risk ofa cardiovascular event in people withsigns of CAN (161–165). Similarly, emerg-ing evidence demonstrates an associa-tion between CAN and glucose variability,especially in the hypoglycemic range(150,164).

In addition, CAN independently predictsthe progression of diabetic nephropathyand chronic kidney disease in diabetes(13,166–168).

Screening and Diagnosis

Recommendations

c Symptoms and signs of autonomicneuropathy should be assessed inpatients with microvascular andneuropathic complications. E

c In the presence of symptoms orsigns of cardiovascular autonomicneuropathy, tests excluding othercomorbidities or drug effects/interactions that could mimic car-diovascular autonomic neuropa-thy should be performed. E

c Consider assessing symptoms andsigns of cardiovascular autonomicneuropathy in patients with hypo-glycemia unawareness. C

The most common symptoms of CANoccur upon standing and include light-headedness, weakness, palpitations, faint-ness, and syncope (13,169,170) (Table 5).The patient should be asked about thesesymptoms when a medical history is takenin the office, although the correlation ofsymptoms with overall autonomic deficitsis weak (149,171). However, these symp-toms may occur quite late in the diseasecourse (25,27,147,149). Itmaybe appropri-ate to screen patients with hypoglycemiaunawareness, as this may be associatedwith CAN (81).

In its early stages, CAN may be com-pletely asymptomatic and detected onlyby decreased heart rate variability (HRV)with deep breathing (13,169,170). Test-ing HRV may be done in the office byeither 1) taking an electrocardiogram re-cording as a patient begins to rise from aseated position or 2) taking an electro-cardiogram recording during 1–2 min ofdeep breathing with calculation of HRV(11,81,170).

Table 5—Symptoms and signs associated with diabetic autonomic neuropathy

CAN Gastrointestinal Urogenital Sudomotor

Resting tachycardia Gastroparesis (Gastropathy) Bladder dysfunction Dry skinAbnormal blood pressure regulation

c Nondippingc Reverse dipping

c Nauseac Bloatingc Loss of appetitec Early satietyc Postprandial vomitingc Brittle diabetes

c Frequencyc Urgencyc Nocturiac Hesitancyc Weak streamc Dribblingc Urinary incontinencec Urinary retention

c Anhidrosisc Gustatorysweating

Orthostatic hypotension (all with standing) Esophageal dysfunction Male sexual dysfunctionc Light-headednesscWeaknessc Faintnessc Visual impairmentc Syncope

c Heartburnc Dysphagia for solids

c Erectile dysfunctionc Decreased libidoc Abnormal ejaculation

Orthostatic tachycardia or bradycardiaand chronotropic incompetence(all with standing)

Diabetic diarrhea Female sexual dysfunction

c Light-headednesscWeaknessc Faintnessc Dizzinessc Visual impairmentc Syncope

c Profuse and watery diarrheac Fecal incontinencec May alternate with constipation

c Decreased sexual desirec Increased pain during intercoursec Decreased sexual arousalc Inadequate lubrication

Exercise intolerance Constipationc May alternate with explosive diarrhea

care.diabetesjournals.org Pop-Busui and Associates 145

In more advanced cases, patients maypresent with resting tachycardia (.100bpm) and exercise intolerance (13,170).Advanced disease may also be associatedwith orthostatic hypotension (a fall in sys-tolic or diastolic blood pressure by.20 mmHg or .10 mmHg, respectively,upon standing without an appropriate in-crease in heart rate) (172). Orthostatichypotension is usually easy to documentin the office. In most cases of CAN, thereis no compensatory increase in the heartrate, despite hypotension (173).The diagnosis includes documentation

of symptoms (Table 5) and signs of CAN,which include impaired HRV, higher rest-ing heart rate, and presence of ortho-static hypotension. In a symptomaticpatient presenting with resting tachycar-dia, with a history of poor glucose control,

or when the diagnosis of CAN is likely,clinicians may not need to perform addi-tional tests given costs and burden.

Exclusion of other comorbidities ordrug effects/interactions thatmaypresentwith the symptoms or signs of CAN andthat mimic CAN may be needed (81,174)(Table 6). In addition, polypharmacy mayalso directly or indirectly impact CAN.

Treatment

Prevention. Please refer to PREVENTION onpage 136.

Recommendations

c Optimize glucose control as earlyas possible to prevent or delay thedevelopment of cardiovascularautonomic neuropathy in peoplewith type 1 diabetes. A

c Consider a multifactorial approachtargeting glycemia among otherrisk factors to prevent cardiovas-cular autonomic neuropathy inpeople with type 2 diabetes. C

c Consider lifestyle modifications toimprove cardiovascular autonomicneuropathy in patients with predia-betes. C

Aswith DSPN,multiple other therapies tar-geting various pathogenetic mechanismshave failed to reverse established CAN.CAN treatment is generally focusedonalle-viating symptoms and should be targetedto the specific clinical manifestation.Symptomatic Treatment of Orthostatic

Hypotension. Treatment for orthostatichypotension is challenging and usually

Table 6—Diagnostic algorithm for CAN

Symptoms Signs/diagnostic tests Differential workup

Resting tachycardia Palpitations Clinical exam: resting heartrate .100 bpm

c AnemiaCould be asymptomatic c Hypothyroidism

c Fever

c CVD (atrial fibrillation, flutter, other)

c Dehydration

c Adrenal insufficiency

c Medications

c Sympathomimetic agents (asthma)

c Over-the-counter cold agents containingephedrine or pseudoephedrine

c Dietary supplements (e.g., ephedra alkaloids)

c Smoking, alcohol, caffeine

c Recreational drugs (cocaine, amphetamines,methamphetamine, mephedrone)

Orthostatic hypotension Light-headedness Clinical exam: a reduction of.20 mmHg inthe systolic blood pressure or.10mmHgin diastolic blood pressure

c Adrenal insufficiencyWeakness c Intravascular volume depletionFaintness c Blood loss/acute anemiaVisual impairment c DehydrationSyncope c Pregnancy/postpartum

c CVDc Arrhythmiasc Heart failurec Myocarditisc Pericarditisc Valvular heart disease

c Alcoholc Medicationc Antiadrenergicsc Antianginalsc Antiarrhythmicsc Anticholinergicsc Diureticsc ACE inhibitors/angiotensin receptor blockerc Narcoticsc Neurolepticsc Sedatives

146 Position Statement Diabetes Care Volume 40, January 2017

involves both pharmacological and non-pharmacological interventions. Physicalactivity and exercise should be encour-aged to avoid deconditioning, which isknown to exacerbate orthostatic into-lerance. Volume repletion with fluidsand salt is central to the management oforthostatic hypotension. Low-dose flu-drocortisone may be beneficial in sup-plementing volume repletion in somepatients, although there are growing con-cerns on risk of supine hypertension.As neurogenic orthostatic hypoten-

sion is in large part a consequence ofthe failure of norepinephrine releasefrom sympathetic neurons, the adminis-tration of sympathomimetic medicationsis central to the care of patients whosesymptoms are not controlled with othermeasures (173). Midodrine, a peripheral,selective, direct a1-adrenoreceptor ago-nist, is an FDA-approved drug for the treat-ment of orthostatic hypotension (175).Midodrine should be titrated gradually toefficacy. It should be used only when pa-tients intend to be upright or seated tominimize supine hypertension (173). Re-cently, droxidopa was approved by theFDA for the treatment of neurogenic or-thostatic hypotension but not specificallyfor patients with orthostatic hypotensiondue to diabetes (176).

Gastrointestinal NeuropathiesGastrointestinal neuropathies may involveany portion of the gastrointestinal tractwith manifestations including esophagealdysmotility, gastroparesis (delayed gastricemptying), constipation, diarrhea, and fe-cal incontinence. The prevalence data ongastroparesis are limited, as most reportswere from selected case series rather thanlarger populations, and therewas inconsis-tency in the outcome measures used(177). In the only community-based study,the cumulative incidence of gastroparesisover 10 yearswas higher in type 1 diabetes(5%) than in type 2 diabetes (1%) and incontrol subjects (1%) (178).Gastroparesis may directly affect gly-

cemic management (e.g., insulin dose orother antidiabetes agents) and may be acause of glucose variability and unex-plained hypoglycemia due to the disso-ciation between food absorption andthe pharmacokinetic profiles of insulinand other agents (12,179–182). Gastro-paresis is mainly found in patients withlong-standing diabetes (183).

Screening and Diagnosis

Recommendations

c Evaluate for gastroparesis in peo-ple with diabetic neuropathy, ret-inopathy, and/or nephropathy byassessing for symptoms of unex-pected glycemic variability, earlysatiety, bloating, nausea, andvomiting. C

c Exclusion of other causes docu-mented to alter gastric emptying,such as use of opioids or glucagon-like peptide 1 receptor agonistsandorganic gastric outlet obstruction,is needed before performing special-ized testing for gastroparesis. C

c To test for gastroparesis, eithermea-sure gastric emptying with scintigra-phy of digestible solids at 15-minintervals for 4 h after food intakeor use a 13C-octanoic acid breathtest. B

Gastroparesis may manifest with abroad spectrum of symptoms and signs(12,177,179,181). As part of a medicalhistory, providers are encouraged todocument symptoms of gastroparesis,such as early satiety, fullness, bloating,nausea, vomiting, dyspepsia, and ab-dominal pain. However, gastroparesismay be clinically silent in the majorityof cases, and symptoms do not neces-sarily correspond with severity of gas-troparesis and are poorly associatedwith abnormal gastric emptying (184,185).Symptoms such as anorexia, nausea,vomiting, and dyspepsia are nonspecificand resemble many other conditions(186) and may just be associated withthe presence of diabetes (181). Impor-tantly, hyperglycemia, hypoglycemia,and acute changes in blood glucose arewell documented to alter gastric emptying(182,187,188), as are some medications,especially opioids, other pain manage-ment agents, and glucagon-like pep-tide 1 receptor agonists (189,190).Therefore, all these factors known toaffect gastric emptying should alwaysbe considered before a firm diagnosis isestablished.

Exclusion of organic causes of gastricoutlet obstruction or peptic ulcer dis-ease (with esophagogastroduodenoscopyor a barium study of the stomach) isneeded before considering specializedtesting for gastroparesis. The diagnostic

gold standard is the measurement ofgastric emptying with scintigraphy of di-gestible solids at 15-min intervals for 4 hafter food intake; the use of 13C-octanoicacid breath test is emerging as a viablealternative (12,179). Optimization ofglucose levels prior to scanning isneeded (182,186–188) to avoid false-positive results.

Treatment

Recommendation

c Consider short-term metoclopra-mide in the treatment of diabeticgastroparesis. E

Treatment for diabetic gastroparesismay be very challenging. Dietary changesmay be useful, such as eating multiplesmall meals and decreasing dietary fatand fiber intake. Withdrawing drugs witheffects on gastrointestinal motility, suchas opioids, anticholinergics, tricyclicantidepressants, glucagon-like peptide1 receptor agonists, pramlintide, andpossibly dipeptidyl peptidase 4 inhibi-tors, may also improve intestinal motility(180,191). In cases of severe gastropare-sis, pharmacological interventions areneeded. Only metoclopramide, a proki-netic agent, is approved by the FDA forthe treatment of gastroparesis. How-ever, the level of evidence regardingthe benefits of metoclopramide for themanagement of gastroparesis is weak,and given the risk for serious adverseeffects (extrapyramidal symptoms, suchas acute dystonic reactions; drug-induced parkinsonism; akathisia; andtardive dyskinesia), its use in the treat-ment of gastroparesis beyond 5 days isno longer recommended by the FDA andthe European Medicines Agency. Itshould be reserved for severe casesthat are unresponsive to other therapies(191).

Urogenital NeuropathiesDiabetic autonomic neuropathy mayalso cause genitourinary disturbances,including sexual dysfunction and blad-der dysfunction. In men, diabetic auto-nomic neuropathy may cause erectiledysfunction (ED) and/or retrogradeejaculation. ED is three timesmore com-mon in men with diabetes than thosewithout the disease (192–194). Sexualdysfunction is also more common inwomen with diabetes (195–199).

care.diabetesjournals.org Pop-Busui and Associates 147

Recommendations

c Consider screening men with otherforms of diabetic neuropathy annu-ally for erectile dysfunctionwith sim-ple questions about a patient’s libidoand ability to reach and maintain anerection. C

c Consider screening patients withother forms of diabetic neuropathyfor lower urinary tract symptomsand female sexual dysfunction inthe presence of recurrent urinarytract infections using targeted ques-tioning regarding symptoms, suchas nocturia, pain during intercourse,and others. E

ED

ED may be a consequence of autonomicneuropathy, as autonomic neurotrans-mission controls the cavernosal and de-trusor smooth muscle tone and function(200). The etiology, however, is multi-factorial, and clinicians should also eval-uate other vascular risk factors such ashypertension, hyperlipidemia, obesity,endothelial dysfunction, smoking, CVD,concomitant medication, and psycho-genic factors (12). There is evidence ofassociations between ED and other di-abetes complications, including CAN(201–203).A diagnosis should be made after es-

tablishing the signs and symptoms of EDand after excluding alternate causes. Cli-nicians should consider performing hor-monal evaluation (luteinizing hormone,testosterone, free testosterone, pro-lactin) to rule out hypogonadism. Inaddition, a variety of medications andorganic causes should be excluded (12).Glucose control was associated with a

lower incidence of erectile dysfunctionin men with type 1 diabetes (204,205).Evidence is less strong for type 2 diabe-tes. Control of other risk factors such ashypertension and hyperlipidemia mayalso improve the condition (12). Pharma-cological treatment includes phosphodi-esterase type 5 inhibitors as first-linetherapy and transurethral prostaglan-dins, intracavernosal injections, vacuumdevices, and penile prosthesis in moreadvanced cases.

Lower Urinary Tract Symptoms and Female

Sexual Dysfunction

Lower urinary tract symptoms manifestas urinary incontinence and bladder dys-function (nocturia, frequent urination,

urination urgency, weak urinary stream)and is linked to the presence of diabeticneuropathy in both men and women(12,206). Female sexual dysfunction oc-curs more frequently in women with di-abetes than in those without diabetes(196,207) and presents as decreasedsexual desire, increased pain during in-tercourse, decreased sexual arousal,and inadequate lubrication.

Evaluation of bladder function shouldbe performed for individuals with diabe-tes who have recurrent urinary tract in-fections, pyelonephritis, incontinence,or a palpable bladder. The medical his-tory should include simple questions tounveil symptoms of lower urinary tractsymptoms and female sexual dysfunc-tion (196,207,208).

Sudomotor DysfunctionSudomotor dysfunctionmaymanifest asdry skin, anhidrosis, or heat intolerance(209,210). A rare form of sudomotordysfunction is gustatory sweating thatcomprises excessive sweating limitedexclusively to the head and neck regiontriggered by food consumption or, insome cases, the smell of food. Originallydescribed as being solely due to auto-nomic neuropathy, gustatory sweatingis also described in patients with dia-betic nephropathy on dialysis (211).

On the basis of the available evi-dence, the routine screening for sudo-motor dysfunction in clinical practiceis not recommended at this time. Theefficacy of the topical antimuscarinicagent glycopyrrolate in the treatmentof gustatory sweating was confirmedin a randomized controlled trial, anddaily application attenuates this compli-cation in most patients for at least 24 h(212).

ATYPICAL NEUROPATHIES

MononeuropathiesMononeuropathies occur more com-monly in patients with diabetes thanin those without diabetes (1) and canoccur as a result of involvement of themedian, ulnar, radial, and common pe-roneal nerves (213). Cranial neuropa-thies present acutely and are rare;primarily involve cranial nerves III, IV,VI, and VII; and usually resolve sponta-neously over several months (213).Electrophysiological studies are mosthelpful in identifying nerve conductionslowing or conduction block at the site

of nerve entrapment. Nerve entrap-ments may require surgical decompres-sion. The improvement in symptomseverity and functional status score isno different between patients withand without diabetes (213).

Diabetic Radiculoplexus NeuropathyDiabetic radiculoplexus neuropathy,a.k.a. diabetic amyotrophy or diabeticpolyradiculoneuropathy, typically involvesthe lumbosacral plexus (214–216). Thecomplication occurs mostly in men withtype 2 diabetes. People with the condi-tion routinely present with extreme uni-lateral thigh pain and weight loss,followed by motor weakness. Electro-physiological assessment is required todocument the extent of disease andalternative etiologies, including degen-erative disc disease or neoplastic, infec-tious, and inflammatory spinal disease(215,216). The disorder is usually self-limiting, and patients improve overtime with medical management andphysical therapy (214,215). There ispresently no evidence from randomizedtrials to support any recommendationon the use of any immunotherapy treat-ment in this condition (217).

Treatment-Induced NeuropathyTreatment-induced neuropathy in diabe-tes (also referred to as insulin neuritis) isconsidered a rare iatrogenic small-fiberneuropathy caused by an abrupt im-provement in glycemic control in the set-ting of chronic hyperglycemia, especiallyin patientswith very poor glucose control(218). The prevalence and risk factors ofthis disorder are not known but are cur-rently under study.

NEUROPATHY END POINTS FORRESEARCH AND CLINICAL TRIALS

There are currently no approved disease-modifying therapies for DSPN, CAN, orother forms of diabetic neuropathy,and multiple clinical trials for theseconditions have failed. Important con-tributing factors include a lack of agree-ment and uniformity in the use of themost sensitive DSPN measures that cap-ture the natural history of the diseaseand detect repair in the specific nervefiber populations, as well as the inclusionof appropriate patient populations.Thus, a valid and careful diagnosis forDSPN in clinical research is critical forcorrectly identifying the appropriatepatient population targeted for either a

148 Position Statement Diabetes Care Volume 40, January 2017

specific intervention or for prognosticimplications.The use of validated clinical instru-

ments such as the Michigan NeuropathyScreening Instrument (MNSI) (mostwidely used in large cohorts of pa-tients with type 1 and type 2 diabetes)(21,26,27,46,74,75), the modified Tor-onto Clinical Neuropathy Scale (mTCNS)(73), the Utah Early Neuropathy Scale(UENS) (77), or the Neuropathy Disabil-ity Score (NDS) (44) are recommended.These may be combined with electro-physiology; measures of small-fiberdamage and repair, such as intraepider-mal nerve fiber density (219–221) orcorneal confocal microscopy (222); andobjective measures of patient functionin the design of DSPN trials.The recommended CAN measures for

clinical trials targeting either a specificintervention or for prognostic implica-tions include 1) standardized cardiovas-cular autonomic reflex tests that aresimple, sensitive, specific, reproducible,and assess the changes in the R-R inter-val on electrocardiogram recordings inresponse to simple clinical maneuvers(deep breathing, Valsalva, and standing)(13,81,191,223); 2) indices of HRV (seeabove) (11,151,169); and 3) restingheart rate and QTc (154,156,157). Othermethods such as baroreflex sensitivity,cardiac sympathetic imaging, and micro-neurography require sophisticated infra-structure and highly trained personnel andare quite expensive and time-consuming(11,13,224).

Duality of Interest. No potential conflicts ofinterest relevant to this article were reported.

References1. Albers JW, Pop-Busui R. Diabetic neuropa-thy: mechanisms, emerging treatments, andsubtypes. Curr Neurol Neurosci Rep 2014;14:4732. Callaghan BC, Kerber KA, Lisabeth LL, et al.Role of neurologists and diagnostic tests on themanagement of distal symmetric polyneurop-athy. JAMA Neurol 2014;71:1143–11493. Dyck PJ, Albers JW, Andersen H, et al.; Tor-onto Expert Panel on Diabetic Neuropathy. Di-abetic polyneuropathies: update on researchdefinition, diagnostic criteria and estimation ofseverity. Diabetes Metab Res Rev 2011;27:620–6284. Malik RA, Veves A, Tesfaye S, et al.; TorontoConsensus Panel on Diabetic Neuropathy. Smallfibre neuropathy: role in the diagnosis of dia-betic sensorimotor polyneuropathy. DiabetesMetab Res Rev 2011;27:678–684

5. Smith AG, Singleton JR. Diabetic neuropathy.Continuum (Minneap Minn) 2012;18:60–846. Singleton JR, Smith AG, Bromberg MB.Increased prevalence of impaired glucose toler-ance in patients with painful sensory neuropa-thy. Diabetes Care 2001;24:1448–14537. Asghar O, Petropoulos IN, Alam U, et al. Cor-neal confocal microscopy detects neuropathy insubjects with impaired glucose tolerance. Dia-betes Care 2014;37:2643–26468. Bongaerts BW, Rathmann W, Heier M, et al.Older subjects with diabetes and prediabetesare frequently unaware of having distal senso-rimotor polyneuropathy: the KORA F4 study.Diabetes Care 2013;36:1141–11469. Im S, Kim SR, Park JH, Kim YS, Park GY. As-sessment of themedial dorsal cutaneous, dorsalsural, and medial plantar nerves in impairedglucose tolerance and diabetic patients withnormal sural and superficial peroneal nerve re-sponses. Diabetes Care 2012;35:834–83910. Ziegler D, Rathmann W, Dickhaus T,Meisinger C, Mielck A; KORA Study Group. Prev-alence of polyneuropathy in pre-diabetes anddiabetes is associated with abdominal obesityand macroangiopathy: the MONICA/KORAAugsburg Surveys S2 and S3. Diabetes Care2008;31:464–46911. Bernardi L, Spallone V, Stevens M, et al.;Toronto Consensus Panel on Diabetic Neuropa-thy. Investigation methods for cardiac auto-nomic function in human research studies.Diabetes Metab Res Rev 2011;27:639–65312. Kempler P, Amarenco G, Freeman R, et al.;Toronto Consensus Panel on Diabetic Neuropa-thy. Gastrointestinal autonomic neuropathy,erectile-, bladder- and sudomotor dysfunctionin patients with diabetes mellitus: clinical im-pact, assessment, diagnosis, and management.Diabetes Metab Res Rev 2011;27:665–67713. Spallone V, Ziegler D, Freeman R, et al.; Tor-onto Consensus Panel on Diabetic Neuropathy.Cardiovascular autonomic neuropathy in diabe-tes: clinical impact, assessment, diagnosis, andmanagement. DiabetesMetab Res Rev 2011;27:639–65314. Tesfaye S, Boulton AJ, Dyck PJ, et al.; Tor-onto Diabetic Neuropathy Expert Group. Dia-betic neuropathies: update on definitions,diagnostic criteria, estimation of severity, andtreatments. Diabetes Care 2010;33:2285–229315. Finnerup NB, Attal N, Haroutounian S, et al.Pharmacotherapy for neuropathic pain inadults: a systematic review and meta-analysis.Lancet Neurol 2015;14:162–17316. Bril V, England JD, Franklin GM, et al.; Amer-ican Academy of Neurology; American Asocia-tion of Neuromuscular and ElectrodiagnosticMedicine; American Academy of PhysicalMedicineand Rehabilitation. Evidence-based guideline:treatment of painful diabetic neuropathy–reportof the American Association of Neuromuscularand Electrodiagnostic Medicine, the AmericanAcademyofNeurology, and theAmericanAcademyof Physical Medicine & Rehabilitation. MuscleNerve 2011;43:910–91717. Diabetes Control and Complications Re-search Group. The effect of intensive treatmentof diabetes on the development and progres-sion of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med1993;329:977–986

18. Diabetes Control and Complications Re-search Group Effect of intensive diabetes treat-ment on nerve conduction in the DiabetesControl and Complications Trial. Ann Neurol1995;38:869–88019. Linn T, Ortac K, Laube H, Federlin K. Inten-sive therapy in adult insulin-dependent diabe-tes mellitus is associated with improved insulinsensitivity and reserve: a randomized, con-trolled, prospective study over 5 years innewly diagnosed patients. Metabolism 1996;45:1508–151320. Callaghan BC, Cheng HT, Stables CL, SmithAL, Feldman EL. Diabetic neuropathy: clinicalmanifestations and current treatments. LancetNeurol 2012;11:521–53421. Ismail-Beigi F, Craven T, Banerji MA, et al.;ACCORD trial group. Effect of intensive treat-ment of hyperglycaemia on microvascular out-comes in type 2 diabetes: an analysis of theACCORD randomised trial. Lancet 2010;376:419–43022. Ohkubo Y, Kishikawa H, Araki E, et al. Inten-sive insulin therapy prevents the progression ofdiabetic microvascular complications in Japa-nese patients with non-insulin-dependent dia-betesmellitus: a randomized prospective 6-yearstudy. Diabetes Res Clin Pract 1995;28:103–11723. Charles M, Ejskjaer N, Witte DR, Borch-Johnsen K, Lauritzen T, Sandbaek A. Prevalenceof neuropathy and peripheral arterial diseaseand the impact of treatment in people withscreen-detected type 2 diabetes: the ADDITION-Denmark study. Diabetes Care 2011;34:2244–224924. Charles M, Fleischer J, Witte DR, et al.Impact of early detection and treatment ofdiabetes on the 6-year prevalence of cardiacautonomic neuropathy in people with screen-detected diabetes: ADDITION-Denmark, acluster-randomised study. Diabetologia 2013;56:101–10825. Ang L, Jaiswal M, Martin C, Pop-Busui R.Glucose control and diabetic neuropathy: les-sons from recent large clinical trials. Curr DiabRep 2014;14:52826. Pop-Busui R, Lu J, Brooks MM, et al.; BARI2D Study Group. Impact of glycemic controlstrategies on the progression of diabetic periph-eral neuropathy in the Bypass Angioplasty Re-vascularization Investigation 2 Diabetes (BARI2D) Cohort. Diabetes Care 2013;36:3208–321527. Martin CL, Albers JW, Pop-Busui R; DCCT/EDIC Research Group. Neuropathy and relatedfindings in the Diabetes Control and Complica-tions Trial/Epidemiology of Diabetes Interven-tions and Complications study. Diabetes Care2014;37:31–3828. Gaede P, Vedel P, Larsen N, Jensen GV,Parving HH, Pedersen O. Multifactorial inter-vention and cardiovascular disease in patientswith type 2 diabetes. N Engl J Med 2003;348:383–39329. Knowler WC, Barrett-Connor E, Fowler SE,et al.; Diabetes Prevention Program ResearchGroup. Reduction in the incidence of type 2 di-abetes with lifestyle intervention or metformin.N Engl J Med 2002;346:393–40330. Balducci S, Iacobellis G, Parisi L, et al. Exer-cise training can modify the natural history ofdiabetic peripheral neuropathy. J DiabetesComplications 2006;20:216–223

care.diabetesjournals.org Pop-Busui and Associates 149

31. Singleton JR, Marcus RL, Jackson JE,K Lessard M, Graham TE, Smith AG. Exerciseincreases cutaneous nerve density in diabeticpatients without neuropathy. Ann Clin TranslNeurol 2014;1:844–84932. Carnethon MR, Prineas RJ, Temprosa M,Zhang ZM, Uwaifo G, Molitch ME; DiabetesPrevention Program Research Group. The asso-ciation among autonomic nervous system func-tion, incident diabetes, and intervention arm inthe Diabetes Prevention Program. DiabetesCare 2006;29:914–91933. Smith AG, Russell J, Feldman EL, et al. Life-style intervention for pre-diabetic neuropathy.Diabetes Care 2006;29:1294–129934. Biessels GJ, Bril V, Calcutt NA, et al. Pheno-typing animal models of diabetic neuropathy: aconsensus statement of the Diabetic Neuropa-thy Study Group of the EASD (Neurodiab). J Pe-ripher Nerv Syst 2014;19:77–8735. O’Brien PD, Hinder LM, Sakowski SA,Feldman EL. ER stress in diabetic peripheral neu-ropathy: a new therapeutic target. Antioxid Re-dox Signal 2014;21:621–63336. Vincent AM, Callaghan BC, Smith AL,Feldman EL. Diabetic neuropathy: cellular mech-anisms as therapeutic targets. Nat Rev Neurol2011;7:573–58337. Zenker J, Ziegler D, Chrast R. Novel patho-genic pathways in diabetic neuropathy. TrendsNeurosci 2013;36:439–44938. Boulton A, Malik R. Diabetes mellitus: neu-ropathy. In Endocrinology: Adult and Pediatric.Jameson JL, De Groot LJ, Eds. Philadelphia, Sa-unders Elsevier, 2010, p. 984–99839. Boulton AJ, Valensi P, Tesfaye S.Interna-tional Neuropathy Workshop of 2009: introduc-tion to the final reports. Diabetes Metab ResRev 2011;27:617–61940. Boulton AJ, Vinik AI, Arezzo JC, et al.; Amer-ican Diabetes Association. Diabetic neuropa-thies: a statement by the American DiabetesAssociation. Diabetes Care 2005;28:956–96241. Maser RE, Steenkiste AR, Dorman JS, et al.Epidemiological correlates of diabetic neurop-athy. Report from Pittsburgh Epidemiology ofDiabetes Complications Study. Diabetes 1989;38:1456–146142. Tesfaye S, Stevens LK, Stephenson JM, et al.Prevalence of diabetic peripheral neuropathyand its relation to glycaemic control and poten-tial risk factors: the EURODIAB IDDM Complica-tions Study. Diabetologia 1996;39:1377–138443. Albers JW, Herman WH, Pop-Busui R, et al.;Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions andComplications Research Group. Effect of prior in-tensive insulin treatment during the DiabetesControl and Complications Trial (DCCT) on periph-eral neuropathy in type 1 diabetes during theEpidemiology of Diabetes Interventions and Com-plications (EDIC) Study. Diabetes Care 2010;33:1090–109644. Young MJ, Boulton AJ, MacLeod AF,Williams DR, Sonksen PH. A multicentre studyof the prevalence of diabetic peripheral neurop-athy in the United Kingdom hospital clinic pop-ulation. Diabetologia 1993;36:150–15445. UK Prospective Diabetes Study (UKPDS)Group. Intensive blood-glucose control with sul-phonylureas or insulin compared with conven-tional treatment and risk of complications in

patients with type 2 diabetes (UKPDS 33). Lan-cet 1998;352:837–85346. Jaiswal M, Lauer A, Martin CL, et al.;SEARCH for Diabetes in Youth Study Group. Pe-ripheral neuropathy in adolescents and youngadults with type 1 and type 2 diabetes from theSEARCH for Diabetes in Youth follow-up cohort:a pilot study. Diabetes Care 2013;36:3903–390847. Sosenko JM, Gadia MT, Fournier AM,O’Connell MT, Aguiar MC, Skyler JS. Body stat-ure as a risk factor for diabetic sensory neurop-athy. Am J Med 1986;80:1031–103448. Clair C, CohenMJ, Eichler F, Selby KJ, RigottiNA. The effect of cigarette smoking on diabeticperipheral neuropathy: a systematic review andmeta-analysis. J Gen Intern Med 2015;30:1193–120349. Tesfaye S, Chaturvedi N, Eaton SE, et al.;EURODIAB Prospective Complications StudyGroup. Vascular risk factors and diabetic neu-ropathy. N Engl J Med 2005;352:341–35050. Van Acker K, Bouhassira D, De Bacquer D,et al. Prevalence and impact on quality of life ofperipheral neuropathy with or without neuro-pathic pain in type 1 and type 2 diabetic patientsattending hospital outpatients clinics. DiabetesMetab 2009;35:206–21351. Callaghan BC, Xia R, Banerjee M, et al.;Health ABC Study. Metabolic syndrome compo-nents are associated with symptomatic poly-neuropathy independent of glycemic status.Diabetes Care 2016;39:801–80752. Boulton AJ. The pathway to foot ulcerationin diabetes. Med Clin North Am 2013;97:775–79053. Mayfield JA, Reiber GE, Sanders LJ, JanisseD, Pogach LM; American Diabetes Association.Preventive foot care in people with diabetes.Diabetes Care 2003;26(Suppl. 1):S78–S7954. Boulton AJ. Diabetic neuropathy and footcomplications. Handb Clin Neurol 2014;126:97–10755. Morrison S, Colberg SR, Parson HK, Vinik AI.Relation between risk of falling and posturalsway complexity in diabetes. Gait Posture2012;35:662–66856. Schwartz AV, Hillier TA, Sellmeyer DE, et al.Older womenwith diabetes have a higher risk offalls: a prospective study. Diabetes Care 2002;25:1749–175457. Wallace C, Reiber GE, LeMaster J, et al. In-cidence of falls, risk factors for falls, and fall-related fractures in individuals with diabetesand a prior foot ulcer. Diabetes Care 2002;25:1983–198658. Brown SJ, Handsaker JC, Bowling FL,Boulton AJ, Reeves ND. Diabetic peripheral neu-ropathy compromises balance during daily ac-tivities. Diabetes Care 2015;38:1116–112259. Baron R, Tolle TR, Gockel U, Brosz M,Freynhagen R. A cross-sectional cohort surveyin 2100 patients with painful diabetic neuropa-thy and postherpetic neuralgia: differences indemographic data and sensory symptoms.Pain 2009;146:34–4060. Freeman R, Baron R, Bouhassira D, CabreraJ, Emir B. Sensory profiles of patients with neu-ropathic pain based on the neuropathic painsymptoms and signs. Pain 2014;155:367–37661. Daousi C, MacFarlane IA, Woodward A,Nurmikko TJ, Bundred PE, Benbow SJ. Chronicpainful peripheral neuropathy in an urban

community: a controlled comparison of peoplewith and without diabetes. Diabet Med 2004;21:976–98262. Davies M, Brophy S, Williams R, Taylor A.The prevalence, severity, and impact of painfuldiabetic peripheral neuropathy in type 2 diabe-tes. Diabetes Care 2006;29:1518–152263. Ziegler D, Rathmann W, Dickhaus T,Meisinger C, Mielck A; KORA Study Group. Neu-ropathic pain in diabetes, prediabetes and nor-mal glucose tolerance: the MONICA/KORAAugsburg Surveys S2 and S3. Pain Med 2009;10:393–40064. Vileikyte L, Leventhal H, Gonzalez JS, et al.Diabetic peripheral neuropathy and depressivesymptoms: the association revisited. DiabetesCare 2005;28:2378–238365. Vileikyte L, Rubin RR, Leventhal H. Psycho-logical aspects of diabetic neuropathic foot com-plications: an overview. Diabetes Metab Res Rev2004;20(Suppl. 1):S13–S1866. Vinik E, Silva MP, Vinik AI. Measuring therelationship of quality of life and health status,including tumor burden, symptoms, and bio-chemical measures in patients with neuroendo-crine tumors. Endocrinol Metab Clin North Am2011;40:97–109, viii67. Hogan P, Dall T, Nikolov P; American Di-abetes Association. Economic costs of diabe-tes in the US in 2002. Diabetes Care 2003;26:917–93268. O’Brien JA, Patrick AR, Caro J. Estimates ofdirect medical costs for microvascular and mac-rovascular complications resulting from type 2diabetes mellitus in the United States in 2000.Clin Ther 2003;25:1017–103869. O’Connor AB. Neuropathic pain: quality-of-life impact, costs and cost effectiveness of ther-apy. Pharmacoeconomics 2009;27:95–11270. Boulton AJ, Malik RA, Arezzo JC, SosenkoJM. Diabetic somatic neuropathies. DiabetesCare 2004;27:1458–148671. Perkins BA, Olaleye D, Zinman B, Bril V. Sim-ple screening tests for peripheral neuropathy inthe diabetes clinic. Diabetes Care 2001;24:250–25672. Tan LS. The clinical use of the 10g mono-filament and its limitations: a review. DiabetesRes Clin Pract 2010;90:1–773. Bril V, Perkins BA. Validation of the TorontoClinical Scoring System for diabetic polyneurop-athy. Diabetes Care 2002;25:2048–205274. Herman WH, Pop-Busui R, Braffett BH,et al.; DCCT/EDIC Research Group. Use of theMichigan Neuropathy Screening Instrumentas a measure of distal symmetrical peripheralneuropathy in type 1 diabetes: results fromthe Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions andComplications. Diabet Med 2012;29:937–94475. Martin CL, Albers J, Herman WH, et al.;DCCT/EDIC Research Group. Neuropathy amongthe Diabetes Control and Complications Trialcohort 8 years after trial completion. DiabetesCare 2006;29:340–34476. Pop-Busui R, Lu J, Lopes N, Jones TL; BARI2D Investigators. Prevalence of diabetic periph-eral neuropathy and relation to glycemic controltherapies at baseline in the BARI 2D cohort.J Peripher Nerv Syst 2009;14:1–1377. Singleton JR, Bixby B, Russell JW, et al. TheUtah Early Neuropathy Scale: a sensitive clinical

150 Position Statement Diabetes Care Volume 40, January 2017

scale for early sensory predominant neuropa-thy. J Peripher Nerv Syst 2008;13:218–22778. Zilliox LA, Ruby SK, Singh S, Zhan M,Russell JW. Clinical neuropathy scales in neu-ropathy associated with impaired glucose tol-erance. J Diabetes Complications 2015;29:372–37779. Herder C, Bongaerts BW, Rathmann W,et al. Association of subclinical inflammationwith polyneuropathy in the older population:KORA F4 study. Diabetes Care 2013;36:3663–367080. Boulton AJ, Armstrong DG, Albert SF, et al.;American Diabetes Association; American Asso-ciation of Clinical Endocrinologists. Comprehen-sive foot examination and risk assessment: areport of the task force of the foot care interestgroup of the American Diabetes Association,with endorsement by the American Associationof Clinical Endocrinologists. Diabetes Care 2008;31:1679–168581. Ziegler D, Keller J, Maier C, Pannek J; Ger-man Diabetes Association. Diabetic neuropa-thy. Exp Clin Endocrinol Diabetes 2014;122:406–41582. Miller JD, Carter E, Shih J, et al. How to do a3-minute diabetic foot exam. J Fam Pract 2014;63:646–65683. Boulton AJ, Kempler P, Ametov A, Ziegler D.Whither pathogenetic treatments for diabeticpolyneuropathy? Diabetes Metab Res Rev 2013;29:327–33384. Ziegler D, Low PA, Litchy WJ, et al. Efficacyand safety of antioxidant treatmentwitha-lipoicacid over 4 years in diabetic polyneuropathy: theNATHAN 1 trial. Diabetes Care 2011;34:2054–206085. Oyibo SO, Prasad YD, Jackson NJ, Jude EB,Boulton AJ. The relationship between blood glu-cose excursions and painful diabetic peripheralneuropathy: a pilot study. Diabet Med 2002;19:870–87386. Griebeler ML, Morey-Vargas OL, Brito JP,et al. Pharmacologic interventions for painfuldiabetic neuropathy: An umbrella systematicreview and comparative effectiveness networkmeta-analysis. Ann Intern Med 2014;161:639–64987. Moulin D, Boulanger A, Clark AJ, et al.; Ca-nadian Pain Society. Pharmacological manage-ment of chronic neuropathic pain: revisedconsensus statement from the Canadian PainSociety. Pain Res Manag 2014;19:328–33588. Freeman R, Durso-Decruz E, Emir B. Effi-cacy, safety, and tolerability of pregabalintreatment for painful diabetic peripheral neurop-athy: findings from seven randomized, controlledtrials across a range of doses. Diabetes Care 2008;31:1448–145489. Freynhagen R, Strojek K, Griesing T, WhalenE, Balkenohl M. Efficacy of pregabalin in neuro-pathic pain evaluated in a 12-week, randomised,double-blind, multicentre, placebo-controlled tri-al of flexible- and fixed-dose regimens. Pain 2005;115:254–26390. Moore RA, Straube S, Wiffen PJ, Derry S,McQuay HJ. Pregabalin for acute and chronicpain in adults. Cochrane Database Syst Rev2009;3):CD00707691. Rosenstock J, Tuchman M, LaMoreaux L,Sharma U. Pregabalin for the treatment of pain-ful diabetic peripheral neuropathy: a double-

blind, placebo-controlled trial. Pain 2004;110:628–63892. RichterRW,PortenoyR,SharmaU,LamoreauxL,Bockbrader H, Knapp LE. Relief of painful dia-betic peripheral neuropathy with pregabalin: arandomized, placebo-controlled trial. J Pain2005;6:253–26093. Raskin P, Huffman C, Toth C, et al. Prega-balin in patients with inadequately treatedpainful diabetic peripheral neuropathy: a ran-domized withdrawal trial. Clin J Pain 2014;30:379–39094. Tesfaye S, Wilhelm S, Lledo A, et al. Dulox-etine and pregabalin: high-dose monotherapyor their combination? The “COMBO-DN study”–amultinational, randomized, double-blind, parallel-group study in patients with diabetic periph-eral neuropathic pain. Pain 2013;154:2616–262595. Ziegler D, Duan WR, An G, Thomas JW,Nothaft W. A randomized double-blind,placebo-, and active-controlled study of T-typecalcium channel blocker ABT-639 in patientswith diabetic peripheral neuropathic pain. Pain2015;156:2013–202096. Quilici S, Chancellor J, Lothgren M, et al.Meta-analysis of duloxetine vs. pregabalin andgabapentin in the treatment of diabetic periph-eral neuropathic pain. BMC Neurol 2009;9:697. Dworkin RH, Jensen MP, Gammaitoni AR,Olaleye DO, Galer BS. Symptom profiles differin patients with neuropathic versus non-neuro-pathic pain. J Pain 2007;8:118–12698. Goldstein DJ, Lu Y, Detke MJ, Hudson J,Iyengar S, Demitrack MA. Effects of duloxetineon painful physical symptoms associated withdepression. Psychosomatics 2004;45:17–2899. Goldstein DJ, Lu Y, Detke MJ, Lee TC,Iyengar S. Duloxetine vs. placebo in patientswith painful diabetic neuropathy. Pain 2005;116:109–118100. Wernicke JF, Pritchett YL, D’Souza DN,et al. A randomized controlled trial of duloxe-tine in diabetic peripheral neuropathic pain.Neurology 2006;67:1411–1420101. Raskin J, Wang F, Pritchett YL, GoldsteinDJ. Duloxetine for patients with diabetic periph-eral neuropathic pain: a 6-month open-labelsafety study. Pain Med 2006;7:373–385102. Hardy T, Sachson R, Shen S, ArmbrusterM,Boulton AJ. Does treatment with duloxetine forneuropathic pain impact glycemic control? Di-abetes Care 2007;30:21–26103. Schwartz S, Etropolski M, Shapiro DY, et al.Safety and efficacy of tapentadol ER in patientswith painful diabetic peripheral neuropathy:results of a randomized-withdrawal, placebo-controlled trial. Curr Med Res Opin 2011;27:151–162104. Vinik AI, Shapiro DY, Rauschkolb C, et al. Arandomized withdrawal, placebo-controlledstudy evaluating the efficacy and tolerabilityof tapentadol extended release in patientswith chronic painful diabetic peripheral neurop-athy. Diabetes Care 2014;37:2302–2309105. Adriaensen H, Plaghki L, Mathieu C,Joffroy A, Vissers K. Critical review of oral drugtreatments for diabetic neuropathic pain-clinicaloutcomes based on efficacy and safety data fromplacebo-controlled and direct comparative stud-ies. Diabetes Metab Res Rev 2005;21:231–240

106. BackonjaM, Beydoun A, Edwards KR, et al.Gabapentin for the symptomatic treatment ofpainful neuropathy in patients with diabetesmellitus: a randomized controlled trial. JAMA1998;280:1831–1836107. Backonja M, Glanzman RL. Gabapentindosing for neuropathic pain: evidence from ran-domized, placebo-controlled clinical trials. ClinTher 2003;25:81–104108. Dallocchio C, Buffa C, Mazzarello P, ChiroliS. Gabapentin vs. amitriptyline in painful dia-betic neuropathy: an open-label pilot study.J Pain Symptom Manage 2000;20:280–285109. Dworkin RH, O’Connor AB, Backonja M,et al. Pharmacologic management of neuro-pathic pain: evidence-based recommendations.Pain 2007;132:237–251110. Morello CM, Leckband SG, Stoner CP,Moorhouse DF, Sahagian GA. Randomizeddouble-blind study comparing the efficacy ofgabapentin with amitriptyline on diabetic pe-ripheral neuropathy pain. Arch Intern Med1999;159:1931–1937111. Simpson DA. Gabapentin and venlafaxinefor the treatment of painful diabetic neuropa-thy. J Clin Neuromuscul Dis 2001;3:53–62112. MaxMB, CulnaneM, Schafer SC, et al. Am-itriptyline relieves diabetic neuropathy pain inpatients with normal or depressed mood. Neu-rology 1987;37:589–596113. Max MB, Lynch SA, Muir J, Shoaf SE,Smoller B, Dubner R. Effects of desipramine,amitriptyline, and fluoxetine on pain in diabeticneuropathy. N Engl J Med 1992;326:1250–1256114. Saarto T, Wiffen PJ. Antidepressants forneuropathic pain. Cochrane Database Syst Rev2007;4:CD005454115. Biesbroeck R, Bril V, Hollander P, et al. Adouble-blind comparison of topical capsaicinand oral amitriptyline in painful diabetic neu-ropathy. Adv Ther 1995;12:111–120116. Boyle J, Eriksson ME, Gribble L, et al. Ran-domized, placebo-controlled comparison ofamitriptyline, duloxetine, and pregabalin in pa-tients with chronic diabetic peripheral neuro-pathic pain: impact on pain, polysomnographicsleep, daytime functioning, and quality of life.Diabetes Care 2012;35:2451–2458117. Moore RA, Derry S, Aldington D, Cole P,Wiffen PJ. Amitriptyline for neuropathic painin adults. Cochrane Database Syst Rev 2015;7:CD008242118. Max MB, Kishore-Kumar R, Schafer SC,et al. Efficacy of desipramine in painful diabeticneuropathy: a placebo-controlled trial. Pain1991;45:3–9; discussion 1–2119. Sindrup SH, Ejlertsen B, Frøland A, SindrupEH, Brøsen K, Gram LF. Imipramine treatment indiabetic neuropathy: relief of subjective symp-toms without changes in peripheral and auto-nomic nerve function. Eur J Clin Pharmacol 1989;37:151–153120. Sindrup SH, Jensen TS. Pharmacologictreatment of pain in polyneuropathy. Neurology2000;55:915–920121. Joss JD. Tricyclic antidepressant use in di-abetic neuropathy. Ann Pharmacother 1999;33:996–1000122. Hearn L, Moore RA, Derry S, Wiffen PJ,Phillips T. Desipramine for neuropathic pain inadults. Cochrane Database Syst Rev 2014;9:CD011003

care.diabetesjournals.org Pop-Busui and Associates 151

123. Derry S, Wiffen PJ, Aldington D,Moore RA.Nortriptyline for neuropathic pain in adults. Co-chrane Database Syst Rev 2015;1:CD011209124. Glassman AH, Roose SP, Bigger JT Jr. Thesafety of tricyclic antidepressants in cardiac pa-tients. Risk-benefit reconsidered. JAMA 1993;269:2673–2675125. Ray WA, Meredith S, Thapa PB, Hall K,Murray KT. Cyclic antidepressants and the riskof sudden cardiac death. Clin Pharmacol Ther2004;75:234–241126. Rowbotham MC, Goli V, Kunz NR, Lei D.Venlafaxine extended release in the treatmentof painful diabetic neuropathy: a double-blind,placebo-controlled study. Pain 2004;110:697–706127. Sindrup SH, Bach FW, Madsen C, Gram LF,Jensen TS. Venlafaxine versus imipramine inpainful polyneuropathy: a randomized, con-trolled trial. Neurology 2003;60:1284–1289128. Raffa RB, Friderichs E, Reimann W, ShankRP, Codd EE, Vaught JL. Opioid and nonopioidcomponents independently contribute to themechanism of action of tramadol, an ‘atypical’opioid analgesic. J Pharmacol Exp Ther 1992;260:275–285129. Freeman R, Raskin P, Hewitt DJ, et al.;CAPSS-237 Study Group. Randomized study oftramadol/acetaminophen versus placebo inpainful diabetic peripheral neuropathy. CurrMed Res Opin 2007;23:147–161130. Harati Y, Gooch C, SwensonM, et al. Double-blind randomized trial of tramadol for the treat-mentof thepainof diabeticneuropathy.Neurology1998;50:1842–1846131. Harati Y, Gooch C, SwensonM, et al. Main-tenance of the long-term effectiveness of tra-madol in treatment of the pain of diabeticneuropathy. J Diabetes Complications 2000;14:65–70132. Gimbel JS, Richards P, Portenoy RK.Controlled-release oxycodone for pain in di-abetic neuropathy: a randomized controlledtrial. Neurology 2003;60:927–934133. Watson CP, Moulin D, Watt-Watson J,Gordon A, Eisenhoffer J. Controlled-release oxy-codone relieves neuropathic pain: a randomizedcontrolled trial in painful diabetic neuropathy.Pain 2003;105:71–78134. HannaM, O’Brien C,WilsonMC. Prolonged-release oxycodone enhances the effects ofexisting gabapentin therapy in painful diabeticneuropathy patients. Eur J Pain 2008;12:804–813135. Sommer C, Welsch P, Klose P, Schaefert R,Petzke F, Hauser W. Opioids in chronic neuro-pathic pain. A systematic review and meta-analysis of efficacy, tolerability and safety inrandomized placebo-controlled studies of atleast 4 weeks duration [published correctionappears in Schmerz 2015;29:308]. Schmerz2015;29:35–46 [in German]136. Højsted J, Sjøgren P. Addiction to opioidsin chronic pain patients: a literature review. EurJ Pain 2007;11:490–518137. Katz NP, Adams EH, Benneyan JC, et al.Foundations of opioid risk management. Clin JPain 2007;23:103–118138. Martell BA, O’Connor PG, Kerns RD, et al.Systematic review: opioid treatment for chronicback pain: prevalence, efficacy, and associationwith addiction. Ann InternMed 2007;146:116–127

139. Chaparro LE, Wiffen PJ, Moore RA, Gilron I.Combination pharmacotherapy for the treatmentof neuropathic pain in adults. Cochrane DatabaseSyst Rev 2012;7:CD008943140. Vileikyte L, Gonzalez JS. Recognition andmanagement of psychosocial issues in diabeticneuropathy. Handb Clin Neurol 2014;126:195–209141. Vinik A, Emir B, Cheung R, Whalen E. Re-lationship between pain relief and improve-ments in patient function/quality of life inpatients with painful diabetic peripheral neu-ropathy or postherpetic neuralgia treated withpregabalin. Clin Ther 2013;35:612–623142. Gilron I, Bailey JM, Tu D, Holden RR,Weaver DF, Houlden RL. Morphine, gabapentin,or their combination for neuropathic pain.N Engl J Med 2005;352:1324–1334143. Vileikyte L, Peyrot M, Gonzalez JS, et al.Predictors of depressive symptoms in personswith diabetic peripheral neuropathy: a longitu-dinal study. Diabetologia 2009;52:1265–1273144. Vileikyte L, Peyrot M, Bundy C, et al. Thedevelopment and validation of a neuropathy-and foot ulcer-specific quality of life instrument.Diabetes Care 2003;26:2549–2555145. Vinik EJ, Hayes RP, Oglesby A, et al. Thedevelopment and validation of the NorfolkQOL-DN, a newmeasure of patients’ perceptionof the effects of diabetes and diabetic neurop-athy. Diabetes Technol Ther 2005;7:497–508146. DCCT. The effect of intensive diabetestherapy on measures of autonomic nervous sys-tem function in the Diabetes Control and Com-plications Trial (DCCT). Diabetologia 1998;41:416–423147. Pop-Busui R, Low PA, Waberski BH, et al.;DCCT/EDIC Research Group. Effects of prior in-tensive insulin therapy on cardiac autonomicnervous system function in type 1 diabetes mel-litus: the Diabetes Control and ComplicationsTrial/Epidemiology of Diabetes Interventionsand Complications study (DCCT/EDIC). Circula-tion 2009;119:2886–2893148. Low PA. Diabetic autonomic neuropathy.Semin Neurol 1996;16:143–151149. Low PA, Benrud-Larson LM, Sletten DM,et al. Autonomic symptoms and diabetic neu-ropathy: a population-based study. DiabetesCare 2004;27:2942–2947150. Jaiswal M, Divers J, Isom S, et al. Preva-lence and correlates of cardiovascular autonomicneuropathy in youth with type 1 diabetes:SEARCH for Diabetes in Youth Study [Abstract].Diabetes 2014;63 (Suppl. 1):A145151. Ziegler D, Voss A, Rathmann W, et al.;KORA Study Group. Increased prevalence of car-diac autonomic dysfunction at different degreesof glucose intolerance in the general popula-tion: the KORA S4 survey. Diabetologia 2015;58:1118–1128152. Maser RE, Mitchell BD, Vinik AI, Freeman R.The association between cardiovascular auto-nomic neuropathy and mortality in individualswith diabetes: a meta-analysis. Diabetes Care2003;26:1895–1901153. Pop-Busui R, Cleary PA, Braffett BH, et al.;DCCT/EDIC Research Group. Association be-tween cardiovascular autonomic neuropathyand left ventricular dysfunction: DCCT/EDICstudy (Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and

Complications). J Am Coll Cardiol 2013;61:447–454154. Pop-Busui R, Evans GW, Gerstein HC,et al.; Action to Control Cardiovascular Risk inDiabetes Study Group. Effects of cardiac auto-nomic dysfunction on mortality risk in the Ac-tion to Control Cardiovascular Risk in Diabetes(ACCORD) trial. Diabetes Care 2010;33:1578–1584155. Young LH, Wackers FJ, Chyun DA, et al.;DIAD Investigators. Cardiac outcomes afterscreening for asymptomatic coronary artery dis-ease in patients with type 2 diabetes: the DIADstudy: a randomized controlled trial. JAMA2009;301:1547–1555156. Ziegler D, Zentai CP, Perz S, et al.; KORAStudy Group. Prediction of mortality using mea-sures of cardiac autonomic dysfunction in thediabetic and nondiabetic population: theMONICA/KORA Augsburg Cohort Study. Diabetes Care2008;31:556–561157. Lykke JA, Tarnow L, Parving HH, Hilsted J. Acombined abnormality in heart rate variationand QT corrected interval is a strong predictorof cardiovascular death in type 1 diabetes.Scand J Clin Lab Invest 2008;68:654–659158. Lonn EM, Rambihar S, Gao P, et al. Heartrate is associated with increased risk of majorcardiovascular events, cardiovascular and all-cause death in patients with stable chronic car-diovascular disease: an analysis of ONTARGET/TRANSCEND. Clin Res Cardiol 2014;103:149–159159. Kadoi Y. Perioperative considerations indiabetic patients. Curr Diabetes Rev 2010;6:236–246160. Soedamah-Muthu SS, Chaturvedi N, WitteDR, Stevens LK, Porta M, Fuller JH; EURODIABProspective Complications Study Group. Rela-tionship between risk factors and mortality intype 1 diabetic patients in Europe: the EURODIABProspective Complications Study (PCS). DiabetesCare 2008;31:1360–1366161. Marques JL, George E, Peacey SR, et al.Altered ventricular repolarization during hypo-glycaemia in patients with diabetes. DiabetMed1997;14:648–654162. Robinson RT, Harris ND, Ireland RH,Macdonald IA, Heller SR. Changes in cardiac re-polarization during clinical episodes of noctur-nal hypoglycaemia in adults with type 1diabetes. Diabetologia 2004;47:312–315163. Koivikko ML, Salmela PI, Airaksinen KE,et al. Effects of sustained insulin-induced hypogly-cemia on cardiovascular autonomic regulation intype 1 diabetes. Diabetes 2005;54:744–750164. Koivikko ML, Tulppo MP, Kiviniemi AM,et al. Autonomic cardiac regulation during spon-taneous nocturnal hypoglycemia in patientswith type 1 diabetes. Diabetes Care 2012;35:1585–1590165. Jaiswal M, McKeon K, Comment N, et al.Association between impaired cardiovascularautonomic function and hypoglycemia in pa-tients with type 1 diabetes. Diabetes Care2014;37:2616–2621166. Astrup AS, Tarnow L, Rossing P, HansenBV, Hilsted J, Parving HH. Cardiac autonomicneuropathy predicts cardiovascular morbidityand mortality in type 1 diabetic patients withdiabetic nephropathy. Diabetes Care 2006;29:334–339

152 Position Statement Diabetes Care Volume 40, January 2017

167. Orlov S, Cherney DZ, Pop-Busui R, et al.Cardiac autonomic neuropathy and early pro-gressive renal decline in patients with nonma-croalbuminuric type 1 diabetes. Clin J Am SocNephrol 2015;10:1136–1144168. Wheelock KM, Jaiswal M, Martin CL, et al.Cardiovascular autonomic neuropathy asso-ciates with nephropathy lesions in AmericanIndians with type 2 diabetes. J Diabetes Compli-cations 2016;30:873–879169. Task Force of the European Society of Car-diology and the North American Society of Pac-ing and Electrophysiology. Heart rate variability:standards of measurement, physiological inter-pretation and clinical use. Circulation 1996;93:1043–1065170. Pop-Busui R. Cardiac autonomic neuropa-thy in diabetes: a clinical perspective. DiabetesCare 2010;33:434–441171. Suarez GA, Opfer-Gehrking TL, Offord KP,Atkinson EJ, O’Brien PC, Low PA. The AutonomicSymptom Profile: a new instrument to assessautonomic symptoms. Neurology 1999;52:523–528172. The Consensus Committee of the Ameri-can Autonomic Society and the American Acad-emy of Neurology. Consensus statement on thedefinition of orthostatic hypotension, pure au-tonomic failure, and multiple system atrophy.Neurology 1996;46:1470173. Freeman R. Clinical practice. Neurogenicorthostatic hypotension. N Engl J Med 2008;358:615–624174. Spallone V, Bellavere F, Scionti L, et al.;Diabetic Neuropathy Study Group of the ItalianSociety of Diabetology. Recommendations forthe use of cardiovascular tests in diagnosing di-abetic autonomic neuropathy. Nutr MetabCardiovasc Dis 2011;21:69–78175. Low PA, Gilden JL, Freeman R, Sheng KN,McElligott MA; Midodrine Study Group. Efficacyof midodrine vs placebo in neurogenic ortho-static hypotension. A randomized, double-blind multicenter study. JAMA 1997;277:1046–1051176. Low PA, Tomalia VA. Orthostatic hypoten-sion: mechanisms, causes, management. J ClinNeurol 2015;11:220–226177. Bharucha AE. Epidemiology and naturalhistory of gastroparesis. Gastroenterol ClinNorth Am 2015;44:9–19178. Choung RS, Locke GR 3rd, Schleck CD,Zinsmeister AR, Melton LJ 3rd, Talley NJ. Riskof gastroparesis in subjects with type 1 and 2 di-abetes in the general population. Am J Gastro-enterol 2012;107:82–88179. Camilleri M. Clinical practice. Diabetic gas-troparesis. N Engl J Med 2007;356:820–829180. Camilleri M, Parkman HP, Shafi MA, AbellTL, Gerson L; American College of Gastroenter-ology. Clinical guideline:management of gastro-paresis. Am J Gastroenterol 2013;108:18–37;quiz 38181. Bytzer P, Talley NJ, Leemon M, Young LJ,Jones MP, Horowitz M. Prevalence of gastroin-testinal symptoms associated with diabetesmellitus: a population-based survey of 15,000adults. Arch Intern Med 2001;161:1989–1996182. Schvarcz E, Palmer M, Aman J, Horowitz M,Stridsberg M, Berne C. Physiological hyper-glycemia slows gastric emptying in normal sub-jects and patients with insulin-dependent

diabetes mellitus. Gastroenterology 1997;113:60–66183. American Diabetes Association. Stan-dards of medical care in diabetes-2015: sum-mary of revisions. In Standards of Medical Carein Diabetesd2015. Diabetes Care 2015;38(Suppl. 1):S4184. Hasler WL, Parkman HP, Wilson LA, et al.;NIDDK Gastroparesis Clinical Research Consor-tium. Psychological dysfunction is associatedwith symptom severity but not disease etiologyor degree of gastric retention in patients withgastroparesis. Am J Gastroenterol 2010;105:2357–2367185. Samsom M, Vermeijden JR, Smout AJ,et al. Prevalence of delayed gastric emptyingin diabetic patients and relationship to dyspep-tic symptoms: a prospective study in unselecteddiabetic patients. Diabetes Care 2003;26:3116–3122186. Parkman HP, Camilleri M, Farrugia G, et al.Gastroparesis and functional dyspepsia: ex-cerpts from the AGA/ANMSmeeting. Neurogas-troenterol Motil 2010;22:113–133187. Fraser R, HorowitzM, Dent J. Hyperglycae-mia stimulates pyloric motility in normal sub-jects. Gut 1991;32:475–478188. Schvarcz E, Palmer M, Aman J, LindkvistB, Beckman KW. Hypoglycaemia increases thegastric emptying rate in patients with type 1diabetes mellitus. Diabet Med 1993;10:660–663189. Schirra J, Nicolaus M, Roggel R, et al.Endogenous glucagon-like peptide 1 controlsendocrine pancreatic secretion and antro-pyloro-duodenal motility in humans. Gut2006;55:243–251190. Murphy DB, Sutton JA, Prescott LF,Murphy MB. Opioid-induced delay in gastricemptying: a peripheral mechanism in humans.Anesthesiology 1997;87:765–770191. Pop-Busui R, Stevens M. Autonomic neu-ropathy in diabetes. In Therapy for DiabetesMellitus and Related Disorders. 6th ed. Umpier-rez GE, Ed. Alexandria, VA, American DiabetesAssociation, 2014, p. 834–863192. Feldman HA, Goldstein I, HatzichristouDG, Krane RJ, McKinlay JB. Impotence and itsmedical and psychosocial correlates: results ofthe Massachusetts Male Aging Study. J Urol1994;151:54–61193. Giugliano F, Maiorino M, Bellastella G,Gicchino M, Giugliano D, Esposito K. Determi-nants of erectile dysfunction in type 2 diabetes.Int J Impot Res 2010;22:204–209194. Saigal CS, Wessells H, Pace J, Schonlau M,Wilt TJ; Urologic Diseases in America Project.Predictors and prevalence of erectile dysfunc-tion in a racially diverse population. Arch InternMed 2006;166:207–212195. Enzlin P, Mathieu C, Van den Bruel A,Bosteels J, Vanderschueren D, Demyttenaere K.Sexual dysfunction in women with type 1 diabe-tes: a controlled study. Diabetes Care 2002;25:672–677196. Enzlin P, Rosen R, Wiegel M, et al.; DCCT/EDIC Research Group. Sexual dysfunction inwomen with type 1 diabetes: long-term findingsfrom the DCCT/EDIC study cohort. DiabetesCare 2009;32:780–785197. Esposito K, Maiorino MI, Bellastella G,Giugliano F, RomanoM, Giugliano D. Determinants

of female sexual dysfunction in type 2 diabetes. IntJ Impot Res 2010;22:179–184198. Mazzilli R, Imbrogno N, Elia J, et al. Sexualdysfunction in diabetic women: prevalence anddifferences in type 1 and type 2 diabetes mel-litus. Diabetes Metab Syndr Obes 2015;8:97–101199. Rutte A, van Splunter MM, van derHeijden AA, et al. Prevalence and correlates ofsexual dysfunction in men and women withtype 2 diabetes. J Sex Marital Ther 2015;41:680–690200. Pop-Busui R, Hotaling J, Braffett BH, et al.;DCCT/EDIC Research Group. Cardiovascularautonomic neuropathy, erectile dysfunctionand lower urinary tract symptoms in menwith type 1 diabetes: findings from the DCCT/EDIC. J Urol 2015;193:2045–2051201. Fedele D, Coscelli C, Santeusanio F, et al.Erectile dysfunction in diabetic subjects inItaly. Gruppo Italiano Studio Deficit Erettilenei Diabetici. Diabetes Care 1998;21:1973–1977202. Pop-Busui R, Braffett BH, Hotaling J, et al.Diabetic neuropathy and urologic complicationsin men with type 1 diabetes in the DiabetesControl and Complications Trial/Epidemiologyof Diabetes Intervention and ComplicationsStudy (DCCT/EDIC) [Abstract]. Diabetes 2014;63 (Suppl. 1):A149–A580203. Gazzaruso C, Giordanetti S, De Amici E,et al. Relationship between erectile dysfunctionand silent myocardial ischemia in apparentlyuncomplicated type 2 diabetic patients. Circula-tion 2004;110:22–26204. Van Den Eeden SK, Sarma AV, RutledgeBN, et al.; Diabetes Control and ComplicationsTrial/Epidemiology of Diabetes Research Group.Effect of intensive glycemic control and diabetescomplications on lower urinary tract symptomsin men with type 1 diabetes: Diabetes Con-trol and Complications Trial/Epidemiology ofDiabetes Interventions and Complications(DCCT/EDIC) study. Diabetes Care 2009;32:664–670205. Wessells H, Penson DF, Cleary P, et al.; Di-abetes Control and Complications Trial/Epide-miology of Diabetes Interventions andComplications Research Group. Effect of inten-sive glycemic therapy on erectile function inmenwith type 1 diabetes. J Urol 2011;185:1828–1834206. Ueda T, Yoshimura N, Yoshida O. Diabeticcystopathy: relationship to autonomic neurop-athy detected by sympathetic skin response.J Urol 1997;157:580–584207. Pontiroli AE, Cortelazzi D, Morabito A. Fe-male sexual dysfunction and diabetes: a system-atic review and meta-analysis. J Sex Med 2013;10:1044–1051208. Barry MJ, Fowler FJ Jr, O’Leary MP, et al.;The Measurement Committee of the AmericanUrological Association. The American UrologicalAssociation symptom index for benign prostatichyperplasia. J Urol 1992;148:1549–1557; dis-cussion 1564209. Smith AG, Lessard M, Reyna S, Doudova M,Singleton JR. The diagnostic utility of Sudo-scan for distal symmetric peripheral neuropa-thy. J Diabetes Complications 2014;28:511–516210. Casellini CM, Parson HK, Richardson MS,Nevoret ML, Vinik AI. Sudoscan, a noninvasive

care.diabetesjournals.org Pop-Busui and Associates 153

tool for detecting diabetic small fiber neuropa-thy and autonomic dysfunction. Diabetes Tech-nol Ther 2013;15:948–953211. Shaw JE, Parker R, Hollis S, Gokal R,Boulton AJ. Gustatory sweating in diabetes mel-litus. Diabet Med 1996;13:1033–1037212. Shaw JE, Abbott CA, TindleK, Hollis S, BoultonAJ. A randomised controlled trial of topical glyco-pyrrolate, the first specific treatment for diabeticgustatory sweating. Diabetologia 1997;40:299–301213. Smith BE. Focal and entrapment neuropa-thies. Handb Clin Neurol 2014;126:31–43214. Dyck PJ, Windebank AJ. Diabetic and non-diabetic lumbosacral radiculoplexus neuropa-thies: new insights into pathophysiology andtreatment. Muscle Nerve 2002;25:477–491215. Massie R, Mauermann ML, Staff NP, et al.Diabetic cervical radiculoplexus neuropathy: adistinct syndrome expanding the spectrum ofdiabetic radiculoplexus neuropathies. Brain2012;135:3074–3088

216. Laughlin RS, Dyck PJ. Electrodiagnostictesting in lumbosacral plexopathies. Phys MedRehabil Clin N Am 2013;24:93–105217. Chan YC, Lo YL, Chan ES. Immunotherapyfor diabetic amyotrophy. Cochrane DatabaseSyst Rev 2012;6:CD006521218. Gibbons CH, Freeman R. Treatment-inducedneuropathy of diabetes: an acute, iatrogenic com-plication of diabetes. Brain 2015;138:43–52219. Lauria G, Bakkers M, Schmitz C, et al. Intra-epidermal nerve fiber density at the distal leg: aworldwide normative reference study. J Pe-ripher Nerv Syst 2010;15:202–207220. Lauria G, Devigili G. Skin biopsy as a diag-nostic tool in peripheral neuropathy. Nat ClinPract Neurol 2007;3:546–557221. Lauria G, Hsieh ST, Johansson O, et al.; Eu-ropean Federation of Neurological Societies;Peripheral Nerve Society. European Federationof Neurological Societies/Peripheral Nerve Soci-ety guideline on the use of skin biopsy in the

diagnosis of small fiber neuropathy. Report of ajoint task force of the European Federation ofNeurological Societies and the Peripheral NerveSociety. Eur J Neurol 2010;17:903–912,e944–e909222. Chen X, Graham J, Dabbah MA, et al.Small nerve fiber quantification in the diagno-sis of diabetic sensorimotor polyneuropathy:comparing corneal confocal microscopy withintraepidermal nerve fiber density. DiabetesCare 2015;38:1138–1144223. Low PA, Denq JC, Opfer-Gehrking TL,Dyck PJ, O’Brien PC, Slezak JM. Effect of age andgender on sudomotor and cardiovagal func-tion and blood pressure response to tilt innormal subjects. Muscle Nerve 1997;20:1561–1568224. Pop-Busui R. What do we know and we donot know about cardiovascular autonomic neu-ropathy in diabetes. J Cardiovasc Transl Res2012;5:463–478

154 Position Statement Diabetes Care Volume 40, January 2017