Abstract
IMPORTANCE
Small-fiber polyneuropathy involves preferential damage to the thinly myelinated A-delta fibers, unmyelinated C sensory fibers, or autonomic or trophic fibers. Although this condition is common, most patients still remain undiagnosed and untreated because of lagging medical and public awareness of research advances. Chronic bilateral neuropathic pain, fatigue, and nausea are cardinal symptoms that can cause disability and dependence, including pain medication dependence.
OBSERVATIONS
Biomarker confirmation is recommended, given the nonspecificity of symptoms. The standard test involves measuring epidermal neurite density within a 3-mm protein gene product 9.5 (PGP9.5)–immunolabeled lower-leg skin biopsy. Biopsies and autonomic function testing confirm that small-fiber neuropathy not uncommonly affects otherwise healthy children and young adults, in whom it is often associated with inflammation or dysimmunity. A recent meta-analysis concluded that small-fiber neuropathy underlies 49% of illnesses labeled as fibromyalgia. Initially, patients with idiopathic small-fiber disorders should be screened by medical history and blood tests for potentially treatable causes, which are identifiable in one-third to one-half of patients. Then, secondary genetic testing is particularly important for familial and childhood cases. Treatable genetic causes include Fabry disease, transthyretin and primary systemic amyloidosis, hereditary sensory autonomic neuropathy-1, and ion-channel mutations. Immunohistopathologic evidence suggests that small-fiber dysfunction and denervation, especially of blood vessels, contributes to diverse symptoms, including postexertional malaise, postural orthostatic tachycardia, and functional gastrointestinal distress. Preliminary evidence implicates acute or chronic autoreactivity in some cases, particularly in female patients and otherwise healthy children and young adults. Different temporal patterns akin to Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy have been described; here, corticosteroids and immunoglobulins, which are often efficacious for inflammatory neuropathic conditions, are increasingly considered.
CONCLUSIONS AND RELEVANCE
Because small fibers normally grow throughout life, improving contributory conditions may permit regrowth, slow progression, and prevent permanent damage. The prognosis is often hopeful for improving quality of life and sometimes for abatement or resolution, particularly in the young and otherwise healthy individuals. Examples include diabetic, infectious, toxic, genetic, and inflammatory causes. The current standard of care requires prompt diagnosis and treatment, particularly in children and young adults, to restore life trajectory. Consensus diagnostic and tracking metrics should be established to facilitate treatment trials.
Polyneuropathy may be the most common neurologic illness; it affects 14.8% of American solder than 40 years.1 This does not even include small-fiber polyneuropathy (SFN), the most common neuropathy presentation, because most patients currently remain undiagnosed. Small-fiber polyneuropathy refers to widespread preferential damage to the small-diameter somatic and autonomic unmyelinated C-fibers and/or thinly myelinated A-delta fibers. Neither surface nerve conduction studies nor electromyography testing, the universal diagnostic tests for large-fiber polyneuropathy, detect SFN. The only population-based estimate available, to our knowledge, is 52.95 affected individuals per 100 000 population in the Netherlands,2 which yields a global SFN prevalence of 4 077 150 individuals. But because ascertainment required the identification of 2 or more symptoms by specialists plus confirmatory skin biopsies or thermal sensory thresholds and normal nerve conduction study results,2 this represents the minimum prevalence. Some have estimated that only 10% of patients are diagnosed. Thus, global prevalence could exceed 10 million. Additionally, meta-analysis generated a 49% (95% CI, 38%−60%) prevalence of SFN in patients with fibromyalgia.3 With fibromyalgia reportedly affecting 2% to 5% of people globally,4 SFN could conceivably affect far more than 10 million people. Hence, there is urgency to improve awareness, diagnosis, treatment, and research.
In addition to pure SFN, mixed polyneuropathies (eg, diabetes) often start as SFN, and SFN often coexists with large fiber–predominant neuropathy.5,6 Small fibers’ high surface-to-volume ratios, which is perhaps the largest among human cells, make their axons most likely to degenerate. Most small fibers are unmyelinated, precluding energy-conserving saltatory conduction and requiring panaxonal ion-channel deployment. Small fibers’ enormous axonal surfaces are supported by miniscule cytoplasmic volumes that struggle to transport supplies along axons sometimes more than a meter long. Insights and treatments for SFN will likely apply to other neuropathic conditions as well.
Pathophysiology and Clinical Presentation
Small-diameter somatic and autonomic unmyelinated C-fibers and/or thinly myelinated A-delta fibers evolved to protect us from harm. They monitor for external dangers and send pain and itch signals centrally to trigger conscious and involuntary evasive maneuvers and monitor the internal physical environment to maintain homeostasis and marshal responses to injury and illness. The central nervous system integrates these exteroceptive and interoceptive inputs to improve the odds of survival. Sensory small fibers’ outward signaling capacities blur classic somatosensory, motor, and autonomic distinctions. Their efferent paracrine and trophic functions (Figure 1) include regulating immunocytes and remodeling bone.7 These diverse functions help explain why SFN can cause multiple symptoms (Figure 2). If individual patients see different specialists for each symptom, the unifying SFN diagnosis can be missed. Traditional descriptions of SFN emphasize spontaneous and stimulus-evoked distal skin pain and sensory loss, but deep aching, fatigue, postexertional malaise, and neuropathic itch (Figure 2E) are also common.8 Postural orthostatic tachycardia syndrome (POTS), gastrointestinal complaints, and sweating complaints reflect damage to post-ganglionic unmyelinated autonomic small fibers.9,10
Figure 1.
Multiple Functions of Normal Cutaneous Small Fibers Reduced in a Patient With Early-Onset Small-Fiber Polyneuropathy
Multilabel fluorescent immunohistochemistry with confocal microscopy. Axons are green (PGP 9.5), basement membrane and blood vessels are red (col IV), and epidermis and endothelia are blue (Ulex europaeus agglutinin). A, C, E, and G are from the skin biopsy of a normal control (a boy aged 10 years); B, D, F, and G are from a boy with Fabry disease (aged 13 years). Images are from the standard lower-leg site, except C and D, which are from a glabrous fingertip. A and B, Epidermal neurites; B shows morphological signs of degeneration (axon thinning and beading) and regeneration (clusters within tracts of epidermal denervation) and a disordered subepidermal neural plexus. C and D, Normal abundant innervation of arteriovenous anastomoses by vasomotor fibers to maintain tonic closure, which is severely reduced in D. E and F, Sudomotor innervation of sweat glands; F shows severe axonal denervation and derangement with resultant atrophy of the sweat gland. G and H, Pilomotor innervation of arrector pili muscles; the density is moderately reduced in H.
Figure 2.
Neurological Examination Findings
A, Classic stocking-and-glove erythromelalgia presentation in the distal extremities, with burning pain and neuropathic microvasculopathy causing skin redness and underlying edema. Severely affected patients often cool the affected skin to reduce C-fiber firing and pain. B, Non–length-dependent anhidrosis in a patient with small-fiber neuropathy from transthyretin amyloidosis. Thermoregulatory sweat testing results reveal residual sweating (dark staining) only in the axilla. C, Episodic hand redness, pain, and hyperhidrosis in woman with early-onset idiopathic small-fiber polyneuropathy. D and E, An adolescent with early-onset small-fiber polyneuropathy causing profound nausea, vomiting, and cachexia improved with a feeding tube (D); the same individual displays Adie pupils (E). F, A boy with biopsy-confirmed SFN attributable to SCN9A mutation cries from chronic neuropathic pain and demonstrates his lower-leg skin thickening and early painless foot ulcer from scratching neuropathic itch. G, A man with hereditary sensory and autonomic neuropathy type 2B from a RAB7 mutation has severe painless foot ulcers.
The classic erythromelalgia phenotype that Silas Weir Mitchell, MD, characterized in 1878 as swollen, red, burning feet soothed by cooling (Figure 2A)11 represents spontaneous activity of damaged sensory C-fibers that transmit unprovoked pain signals centrally while releasing vasoactive substance P and calcitonin gene-related peptide distally to cause neurogenic inflammation.12 Some SFN symptoms are caused by neuropathic microvasculopathy as tissues become unable to increase perfusion during peak demand. For instance, chronic fatigue and reduced exertional tolerance in SFN and fibromyalgia are explicated by studies showing that in both conditions, denervated dilated arteriovenous anastomoses (Figure 1D) in skeletal muscle shunt arterial blood directly into venules, bypassing capillaries, depriving exercising myocytes, and triggering local hypoxemia.13,14 This pathophysiologic profile reportedly identifies SFN with 90% sensitivity and 91% specificity.14 In addition, impaired venous contractility in the legs reduces cardiac return and thus cardiac output, which worsens perfusion throughout the body. Among 229 patients with chronic fatigue attributed to preload failure, 31% had skin biopsies consistent with SFN.15 Gastrointestinal specialists are increasingly considering neuropathic dysregulation of enteric vasculature (gastrointestinal angina) as an additional contributor, along with enteric small-fiber loss,16 to previously unexplained postprandial nausea, vomiting, and lower dysmotility syndromes. It is possible that neuropathic dysregulation also contributes to brain symptoms of SFN, such as chronic daily headache and cognitive dysfunction.9,17,18
Small-fiber polyneuropathy has documented effects on the brain. The central axons of small fibers synapse in the spinal cord to trigger ascending signals or directly ascend the dorsal columns to the brainstem. Central axons also degenerate (central-peripheral distal axonopathy) with trans synaptic and network effects.18 The C and A small-fiber nociceptors control the long-term potentiation–like pain amplification that can secondarily change in SFN (central sensitization).19 Inactivity, pain, fatigue, deconditioning, depression, and pain medications also affect the brain.17 Neuropsychological evaluations in patients with SFN associated with Sjögren syndrome yielded 100% with cognitive complaints, including 80% with problems in the executive domain and 70% with abnormal Wisconsin Card-Sorting Test results.20
Non–Length-Dependent and Patchy Presentations
One-quarter of patients with SFN do not present with classic symmetric stocking-and-glove presentations21 (Figure 2B and D). Patchy or proximal distributions suggest targeting of sensory or autonomic cell bodies (ganglionitis or neuronitis) rather than axonopathy. Cranial ganglionitis causes SFN symptoms, including pain and erythromelalgia, in the head and face. Most ganglionitis is inflammatory, particularly in female patients and individuals with rapid-onset cases. Small-fiber ganglia become vulnerable because they leave the protection of the central nervous system during embryo-genesis to become sentinels. Their fenestrated capillaries that facilitate sampling the internal environment further expose them to infection, immunity, and toxins (Table 1).29 Cerebrospinal fluid can reveal inflammatory markers, and mononuclear infiltrates and nodules of Nageotte are pathological hallmarks of cell-body degeneration.30 Because large-fiber cell bodies are often also attacked, patients can have ataxia or reduced proprioception, hypo-reflexia and abnormal nerve conduction study results, or somato-sensory-evoked potentials. Magnetic resonance imaging may become useful.31
Table 1.
Medical Contributors to Initially Idiopathic Small-Fiber Neuropathy: Screening by History and Examinationa,b
Cost-Effective Screening Blood Tests in the United States | Definition of Abnormal Result | Medical Condition Tested | Prevalence of Abnormal Test Result in Sample, No. (%) | % Population Prevalence of Abnormal Test Result22 | Population or Geographic Location Studied22 |
---|---|---|---|---|---|
Immune-mediated disorders | |||||
Erythrocyte sedimentation rate | High | Inflammation or infection9,23 | 157 (28.0) | 5.0 | Norway |
Antinuclear antibodies | ≥1:160 | Lupus or rheumatic disease9 | 153 (27.5) | 8.9 | Brazil |
Complement C4 | Low | Inflammation and vasculitis9 | 115 (15.7) | 10.4 | WHS |
Complement C3 | Low | Autoimmunity and vasculitis9 | 118 (11.0) | 2.7 | WHS |
C-reactive protein | High | Injury or inflammation | 95 (12.6) | 7.1 | WHS |
Anti-Ro/Anti-Sjögren syndrome-associated antigen A | High | Sjögren syndrome24 | 98 (9.2) | 0.7 and 3.9 | WHS and NHANES, respectively |
IgA tissue transglutaminase antibodies | High | Celiac disease25 | 109 (3.5) | 0.5–1.0 | United States (estimate) |
Metabolic or endocrine disordersc | |||||
Fasting glucose or oral glucose tolerance test | High | Diabetes | 921 (7.7) | 6.126 | The Netherlands |
Thyrotropin | High | Hyperthyroidism | 145 (4.1) | 0.5 | NHANES |
Thyrotropin | Low | Hypothyroidism27 | 144 (2.1) | 0.3 | NHANES |
Folate | Low | Folate deficiency28 | 49 (2.0) | <0.1 | US residents >50 years old |
Hematologic disordersd | |||||
Serum protein electrophoresis or immunofixation | Variable | Monoclonal gammopathy | 128 (3.9) | 3.2 | US residents >50 years old |
Abbreviations: NHANES, National Health and Nutrition Examination Survey; WHS, World Health Survey.
Additional causes of small-fiber polyneuropathy include temporally appropriate neurotoxic exposures (cancer chemotherapy [eg, with vinca, taxanes], antiretroviral HIV drugs, colchicine, vitamin B6, metronidazole, nitrofurantoin, fluoroquinolones, and arsenic), and genetic disorders (Charcot-Marie-Tooth disease, transthyretin amyloidosis, Fabry disease, hereditary sensory and autonomic neuropathies, Ehlers-Danlos syndrome, and ion channelopathies).
Infectious causes of small-fiber polyneuropathy for which no tests are included in this Table: HIV, hepatitis B and C, Lyme disease, and leprosy.
Metabolic causes of small-fiber polyneuropathy for which screening blood tests are not recommended in the United States: alcohol abuse and glucose intolerance.
Hematologic causes of small-fiber polyneuropathy include Waldenström macroglobulinemia and multiple myeloma.
Sjögren syndrome and proximal diabetic radiculopathy can cause chronicnon–length-dependent SFN symptoms.32,33 Restricted ganglionitis can present without serologic markers, including in 60% of patients with Sjögren syndrome–associated SFN.24 Ophthalmologic evaluation and lip biopsy for salivary glands help confirm suspected cases and direct therapy. Paraneoplastic painful ganglionitis is most often associated with anti-Hu amphiphysin and CV2 autoantibodies; the lung is the most common site of a malignant condition, followed by hematological and gastrointestinal tumors.34 Thus, non–length-dependent SFN requires urgent evaluation and often disease-targeted treatment.29
Early Onset in Children and Healthy Young Adults
Childhood SFN is generally attributed to rare, deterministic, mendelian mutations (Table 2). However, affected families reveal more complex effects with wider implications. Presentation age varies from preschool, if affected neurons never form (hereditary sensory and autonomic neuropathies 3–5)35–37 to the second and third decade, if neurons form but degenerate quickly.38,40 Rare patients remain a symptomatic or sub clinical into adulthood, unless or until axons are additionally stressed (for instance, by aging, diabetes, chemo-therapy, or injury), representing variable penetrance.38,44 Mutations are often conceptualized as loss-of-function mutations, where neural degeneration permits painless injuries, infections, and (rarely) death (Figure 2E and F) vs gain-of-function mutations, where hyperexcitable small-fiber firing triggers pain, itch, and autonomic lability. However, excess action potentials trigger deleterious ion and fluid entry and add energy demands. Together, these can cause axon degeneration and loss of function, blurring the distinction. Single-nucleotide polymorphisms (SNPs) affecting voltage-gated sodium channels (Nav) preferentially expressed by small fibers have been linked to loss-of-function and gain-of-function SFN symptoms, but the most frequent abnormal test finding is a variant of uncertain significance (VUS). Evaluations of pathogenic significance of a VUS require integrating data on variant population prevalence, anticipated out comes, and reported phenotypes. Clinical significance may vary in different populations and environments. A Dutch study of patients with pure SFN–associated erythromelalgia symptoms with Nav SNPs,42 whereas a US study of patients with mixed neuropathy reported no correlations between the presence of Nav variants and pain status.45 The prevalence of rare Nav SNPs also far exceeded population prevalences in the Dutch study of patients with pure SFN.42,45 Excess Nav firing is the electrophysiological sub-strate of neuropathic pain. In knock-in mice with Fabry disease, globotriaosylceramide accumulations caused gain of function of Nav1.7 and potassium-sodium hyperpolarization–activated cyclic nucleotide-gated ion-channel 2 (HCN-2) ion channels expressed in small fibers and blood vessels, culminating in painful axonopathy. Also, Nav variants may influence susceptibility to painful diabetic neuropathy.41,44 Some VUS interact with other components or modulators of ion-channel complexes, and sequencing only channel exons will not detect them. Some genetic variants affect multiple fiber types. Hereditary sensory neuropathy type 1 presents in youth with distal pain, itch, and sensory loss, but weakness and muscle atrophy develop rapidly.38 Transient receptor potential ankyrin 1 (TRPA1), sodium voltage–gated channel alpha subunit 9 (SCN9A), collagen type VI alpha 5 chain (COL6A5), transient receptor potential cation channel subfamily V member 4 (TRPV4), and Piezo-type mechanosensitive ion channel component 2 (Piezo2) variants have been associated with neuropathic itch.8,43 An immune-system attack on channel complexes can mimic a genetic channelopathy.46 Sodium-channel blockers, including carbamaze-pine, oxcarbazepine, and mexiletine, are attractive for treating channelopathy-associated SFN and perhaps other types as well; trials are needed.
Table 2.
Genetic Small-Fiber Polyneuropathies
Name Inheritance | Inheritance | Pathogenesis | Pathogenic Gene(s) | Clinical Features | Age at Onset | Precision Treatments |
---|---|---|---|---|---|---|
Hereditary sensory and autonomic neuropathy IV | Autosomal recessive35 | Lack of development | NTRK1 | Insensitivity to pain and anhidrosis | Congenital | None |
Hereditary sensory and autonomic neuropathy V | Autosomal recessive | Lack of development | NGF-β | Insensitivity to pain | Congenital | None |
Hereditary sensory and autonomic neuropathy III (familial dysautonomia, Riley-Day Syndrome) | Autosomal recessive; almost exclusive to patients of Eastern-European Jewish ancestry36 | Lack of development | IKBKAP | Tearless crying, dry mouth, diarrhea/constipation, labile blood pressure, reduced taste, pain, and temperature | Early childhood | None |
Cold-induced sweating syndrome | Autosomal recessive37 | Abnormal maturation | CRLF1 | Cold-induced sweating | Congenital | None |
Hereditary sensory and autonomic neuropathy I | Autosomal dominant38 | Reduced survival | SPTLC1 and SPTLC2 | Reduced sensitivity to touch, pain, and thermal stimuli; muscle weakness; atrophy; and distal ulcers | Subtle onset in children, teens, worsens during life | Oral L-serine overloading reduces neurotoxic 1-deoxysphingolipids and modestly improves adults who have been long affected.39 Much earlier diagnosis and treatment consideration recommended. |
Hereditary sensory and autonomic neuropathy IIA, B, and C | Autosomal recessive40 | Reduced survival | ATL1, ATL3, DNMT1, RAB7, ATSV, WNK1, and FAM134B | Pan sensory loss and distal mutilation | Birth or early childhood | None |
Fabry disease | X-linked recessive41 | Lysosomal storage of globotriaosylceramide in small-fibers and endothelial cells | GLA | Pain and hypohydrosis | Late childhood to early adult | Enzyme replacement agalsidase alfa (Replagal) and agalsidase beta (Fabrazyme); only Fabrazyme has been cleared by the US Food and Drug Administration. |
Transthyretin amyloidosis | Autosomal dominant | Aggregation of insoluble, potentially toxic, fibrils that precipitate in tissues | Transthyretin | Pain, autonomic symptoms, carpal tunnel syndrome, and restrictive cardiomyopathy | Adults (20–80 y) | Patisiran (small, interfering RNA) and Inotersen (antisense oligonucleotide) lower transthyretin formation. Transthyretin stabilizers Tafamidis, Difluinsal, and Tolcapone inhibit release of monomers that form amyloid. |
Cation channelopathy | Autosomal dominant42,43 | Channelopathies; initial gain of function, with possible early or late loss of function | Nav 1.6–1.9 (SCN 8–11), HCN2, TRPA1, TRPV4, and Piezo2 | Episodic or chronic pain, itch, sensory loss, and vasomotor and autonomic symptoms | Usually prepubertal | Trials of subtype-specific sodium-channel antagonists (Nav1.7 and Nav 1.8) are in progress (ClinicalTrials.gov: NCT03339336). |
Abbreviations: ATL1, atlastin guanosine triphosphatase 1; ATL3, atlastin guanosine triphosphatase 3; ATSV, axonal transporter of synaptic vesicles; CRLF1, cytokine receptor-like factor 1; DNMT1, DNA methyltransferase 1; FAM134B, family with sequence similarity 134 member B; GLA, α-galactosidase A gene; HCN2, potassium-sodium hyperpolarization-activated cyclic nucleotide-gated ion channel 2; IKBKAP, inhibitor of κ light-polypeptide gene enhancer in B-cells, kinase complex–associated protein; Nav, voltage-gated sodium channel; NGF-β, nerve growth factor–β; NTRK1, neurotrophic receptor tyrosine kinase 1; Piezo2, Piezo-type mechanosensitive ion channel component 2; RAB7, Ras-related protein Rab-7a; SCN9A, sodium voltage-gated channel subunits 8 through 11; SPTLC1, serine palmitoyltransferase long-chain base subunit 1; SPTLC2, serine palmitoyltransferase long-chain base subunit 2; TRPA1, transient receptor potential ankyrin 1; TRPV4, transient receptor potential cation channel vanilloid subfamily member 4; WNK1, lysine-deficient protein kinase 1.
Most early-onset SFN is not genetic but rather appears inflammatory, involving autoreactive B cells. Auto antibodies have been associated with dysautonomic SFN symptoms, including POTS,47,48 and corticosteroid immune therapy has effectively treated young patients with rapid-onset painful SFN.23,49–51 The series that, to our knowledge, is largest, consisted of 41 patients with unexplained widespread pain beginning before age 21 years; objective testing identified definite SFN in 25 of 41 patients (59%) and probable SFN in 7 of 41 patients (17%).9 Inflammatory causality was proposed when comprehensive evaluations revealed neither familial, diabetic, nor toxic causes but rather histories of other autoimmune illnesses in 14 of 41 patients (33%) and inflammatory blood-test markers in 32 of 36 (89%). Corticosteroids benefitted 67% (10 of 15 patients) and intravenous immune globulins (IVIg) benefited 5 of 8 patients (63%). Overall, 80% benefited from immunotherapy.9 Reports of early-onset SFN after infectious exposures, particularly to human papil-lomavirus vaccination, suggest potential molecular mimicry.52,53 Autoreactive SFN also affects adults but is easiest to diagnose in children and otherwise healthy young people without other risk factors.
Testing children for SFN requires age-appropriate norms. For autonomic testing, quantitative sudomotor axon reflex test norms are only established for individuals 10 years or older, and short stature blunts the sensitivity of tilt-table testing (and reduces the prevalence of POTS).54 Skin biopsies are the best option for children currently, and preapplying local-anesthetic cream renders them painless. Biopsies are taken proportionately closer to the lateral malleolus, and a few laboratories accept 2-mm punches. Age-matched norms and statistical modeling are essential for reducing the very high rates of false-negative interpretations with adult norms. Children have 3 to 4 times more epidermal neurites, which are pruned during adolescence.55 Children and otherwise healthy young adults are excellent participants in research because most have recent, precisely documented onsets, and their parents are motivated to advance diagnosis and treatment. Regarding treatment, because younger patients have the greatest risk of life-derailing disease trajectories, plus greatest potential for recovery,9,56 lengthy observation or mere symptom palliation may be insufficient. Immuno-therapy and/or genetic testing should be more rapidly considered. Top priorities include improving pediatrician awareness of SFN and developing age-appropriate diagnostic and tracking metrics.
Assessment and Diagnosis
Recognition is straightforward with the classic painful-feet presentation, but experts estimate that most patients are currently undiagnosed. The lack of case definition and patient-reported and clinician-reported assessment tools hinders research as well as care. There are standardized patient-reported symptom surveys for single-cause mixed neuropathic conditions from diabetes,57–59 sarcoidosis,60 and specific chemotherapies,61 and validated general questionnaires for pain and dysautonomia.62,63 The SFN-specific Rasch-built overall disability scale and Small-Fiber Symptom Survey are comprehensive SFN symptom surveys validated for all causes.64–66 Regarding examination findings, the Utah Early Neuropathy Scale was developed and validated for sensory-predominant diabetic polyneuropathy.67 Insofar as we know, only the Massachusetts General Neuropathy Exam Tool (MAGNET) is validated for SFN independent of cause.68 A recent international Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) meeting is expected to generate the first consensus research case definition of SFN and research diagnostic criteria. These will incorporate symptoms, abnormalities on neurological examination, and objective confirmation by lower-leg skin biopsy or combinations of secondary tests. Research diagnoses prioritize specificity over sensitivity, so failure to fully meet them does not preclude SFN nor should research criteria be used to justify withholding treatment or reimbursement. The impression of expert clinicians remains the clinical diagnostic standard.
Confirming Diagnosis
When symptoms are nonspecific and examination findings are muted or subjective, objective confirmation is a critical step toward effective treatment and reimbursement. However, neither standard nerve conduction study nor electromyographic test assesses small fibers. They are invisible with conventional light microscopy, and biopsying nerves for an electron microscopic examination is not often feasible. The development of minimally invasive skin biopsies revolutionized SFN diagnosis and fuels discovery of new causes and treatments, with nerve biopsies now mostly reserved for vasculitis. The Peripheral Nerve Society and American and European Academies of Neurology endorse removing 3-mm punches from 10 cm above the lateral malleolus using intradermal anesthesia. Biopsy sections are immunolabeled against panneuronal marker protein gene product (PGP9.5), which reveals small-fiber axons with light or fluorescence microscopy.69,70 Biopsies can be performed locally and mailed to accredited laboratories for processing, and epidermal neurite density measured according to consensus standards for statistical comparison with epidermal neurite density in biopsies from healthy, demographically matched volunteers (Figure 3).70,72–74 Measured epidermal neurite densities less than the fifth percentile of the anticipated normal distribution confirm SFN in patients in whom this condition is suspected. However, many laboratories use nonrepresentative or published norms, which leads to inaccurate interpretations because of variability between laboratory methods and local reference populations.72,75,76 Insensitivity (false-negative results) is the biggest concern, not only as a result of poor norms (particularly for young patients and female patients), but also from sampling error and the fact that axons typically degenerate later than symptoms appear. Second biopsies (eg, from the thigh for non–length-dependent neuropathic conditions,75,77–79 or from the foot80) add sensitivity but also cost, and there are insufficient norms to recommend routine use. Immunofluorescence (Figure 1) visualizes twice as many fibers as conventional bright-field microscopy (Figure 3) and thus requires separate norms.81 Details about longitudinal stability and treatment responsiveness are needed for trial use. Researchers are exploring other biopsy sites and measures, such as density of sweat glands or arrector pili innervation (Figure 3), but these need more validation for clinical use.
Figure 3.
Protein Gene Product 9.5–Immunolabeled Lower-Leg Skin Biopsies: Importance of Accurate Norms for Diagnostic Interpretation
This bright-field photomicrograph illustrates standard clinical processing and morphometric evaluations used most often for clinical diagnostic confirmation of small-fiber neuropathy. Skin biopsies are from 10 cm above the lateral malleolus in adults and proportionally less in children. They are cryosectioned vertically into 50-μm sections and hand-immunolabeled using monoclonal antibody targeting protein gene product–9.5, a ubiquitin hydrolase present in all axons.71 Skilled morphometrists count the number of neurites that penetrate the dermal-epidermal junction and express epidermal neurite density per mm2 of skin-surface area. Each patient’s epidermal neurite density is statistically compared with the anticipated normal distribution calculated using within-laboratory measurements made from biopsies of screened healthy control participants. Patient with epidermal neurite densities at less than the fifth percentile of expected amounts have pathologically confirmed clinical diagnoses, and predegenerative swellings, fragmentation, or inflammatory infiltrates (not shown) can be supportive; × 40 magnification; scale bar = 100 μm. A, Screened normal, healthy, white male control patient, aged 16 years. His epidermal neurite density (356 per mm2 of skin-surface area) was at the 58.0 percentile of the anticipated normal distribution of his age, sex, and race and provides no pathological evidence of small-fiber polyneuropathy. B, A 15.1-year-old white male patient with gastrointestinal symptoms, headache, fatigue, labile blood pressure, and postural orthostatic tachycardia syndrome (POTS) starting at age 3 years. His epidermal neurite density of 164 per mm2 of skin-surface area is at the 1.3rd percentile of the anticipated norm by age, sex, and race, confirming the clinician’s diagnosis of small-fiber polyneuropathy. Autonomic function testing results were also abnormal, antinuclear antibody test results were positive at 1:160, and a paraneoplastic panel identified autoantibodies against a voltage-gated potassium channel. C, This screened normal, healthy control participant, a 66.3-year-old white woman had 158 epidermal neurites per mm2 of skin-surface area and was at the 37.6th percentile of anticipated normal distribution for her age, sex, and race, well within the normal range, although lower than the pathological epidermal neurite density in B.
More accessible, cheaper, and more sensitive tests would improve care. The best accepted is physiological: quantitative autonomic function testing developed at the Mayo Clinic. It includes 4 site comparisons of quantitative sudomotor axon reflex text sweating to anticipated norms,73 which raises sensitivity toward 82% and is similar to a skin biopsy.82–84 However, because few hospitals have the equipment, and patients must stop potentially interfering medications beforehand and travel to the laboratory, autonomic function testing is still not regularly used or validated for trials.73 Quantitative sensory testing records only patients’ subjective sensations and is not an objective diagnostic test.85 Newer measures of sweating that are insufficiently validated for routine clinical use include the dynamic sweat test that assesses sweat gland density, distribution, and stimulated sweat production.86,87 In vivo corneal confocal microscopy, which visualizes the exclusively C-fiber innervation of the cornea, is noninvasive and requires no preparation. It is repeatable for longitudinal tracking but not yet routine.88 Neurophysiologic tests used mostly for research include laser-evoked and heat-evoked sensory potentials.89
Medical Causes and Contributors
Patients who have initially idiopathic cases of SFN (iiSFN) are first screened for cause using patient and family histories and prior test results (Table 1). Subsequent prospective blood screening identifies potential causes in 30% to 50% of patients with iiSFN.22,26,90 Screening recommendations must integrate pretest probability of an abnormal result, test availability, cost, and diagnostic performance. Diabetes mellitus is overall the most common cause of neuropathy, but the value of screening for undiagnosed diabetes in iiSFN differs depending on its local prevalence. Prediabetes conveys far less risk and is not often causal.22,91 Among US adults with biopsy-confirmed iiSFN, 2% had unappreciated diabetes mellitus and 22% had prediabetes, both of which were less than population prevalences (12%−14% and 37%−38%, respectively).26 In the Netherlands, testing patients with iiSFN revealed that 71 of 921 patients (7.7%) had undiagnosed diabetes, which is of unclear significance compared with the 6% population prevalence.26
We discourage universal screening with low-value or futile tests, including those for heavy metals and sarcoidosis, where high serum angiotensin-converting levels had 0% positive predictive value.22 Regarding vitamins, high B6 levels are the major risk.28 Universal screening for folate and B12 deficiencies is becoming less cost-effective in Western countries. A low folate level causes large-fiber rather than small-fiber sensory axonopathy; additionally, this deficiency is vanishing after mandated supplementation.92 In the United States, B12 deficiencies are now also rare, and prevalence in patients with iiSFN does not exceed population prevalence.22,28,90 In New England, where Lyme disease is prevalent, the prevalence of Western blot–confirmed Lyme disease was not elevated in patients with iiSFN.22
Secondary genetic sequencing (Table 2) is simultaneously underused in patients with a high probability of genetic causes (eg, pediatric patients, those with family history, those with highly specific phenotypes) and overused in patients with low probability of genetic causes.93 We at least consider it in adult patients with SFN without a cause identified after primary testing and attempt it in all children whose cases are still idiopathic, even absent a family history. The relative values of sequencing single genes, gene panels, vs initial whole-exome sequencing or whole-genome sequencing change as costs drop. Serial sequential testing delays diagnosis, whole-exome sequencing does not detect noncoding SNPs and structural effects such as gene duplications or translocations, and unbiased sequencing generates most VU Sand potentially adds cost. With more genes implicated, costs dropping, and more precision treatments available (Table 2), sequencing will be performed more often, sooner, and more comprehensively, potentially enabling creation of polygenic risk scores. We and other clinicians detect Nav variants most often in iiSFN, although their relevance is often unclear.42,45
Sequencing results are not final. Laboratories have limited medical information available from which to select sequences to analyze, and depth and quality of their analyses and reference comparators vary. New pathogenic variants are identified, new phenotypic correlates emerge, and some variants are later found to be spurious. Understanding of the significance of VUS in SFN-pathogenic genes changes is also evolving. Some so-called benign variants may be pathogenic in individuals with other genetic or environmental factors (ie, risk modifiers), and multiple benign variants could conceivably have combinatorial effects. Plus, we are only now appreciating that mutations in noncoding DNA can alter RNA biophysical properties in ways that influence its interactions. Initially unhelpful sequencing results may become useful later, and periodic reanalysis is a free service of many laboratories. Among 1519 patients referred to the National Institutes of Health Undiagnosed Diseases Network for advanced testing, nearly half were referred for neurological symptoms.94 Among all diagnostic tools including imaging, whole-exome sequencing and whole-genome sequencing were most productive, providing molecular diagnoses in 74% of these patients.94 Molecular diagnoses usually curtail further testing, can change treatments from palliative to precision-targeted to improve efficacy and safety, sometimes reduce cost, and influence reproductive planning.94 Patients and physicians report benefit from genetic diagnoses, and the research benefits and help to other patients are incalculable.
Inflammatory and autoreactive conditions are increasingly linked to SFN, unsurprisingly, given small fibers’ immunerole. Their cell bodies leave the protection of the central nervous system protection during gestation to become sentinels and first responders. Sensory ganglia develop fenestrated capillaries and distal C-fiber terminal nearly contact the exterior in the superficial epidermis and mucosa. Small fibers have receptors for and release immune or inflammatory signals distally. They contact mast cells and microvessels. Neurogenic inflammation mediated by C-fibers is sometimes visible as flushing and swelling in focal or generalized small-fiber neuropathy (Figure 2A and C). Medical histories and blood test results in large studies of patients with iiSFN suggest associations with dysimmunity.22,26,56 Among 921 Dutch patients with iiSFN, 12.9% had unspecified immunological conditions, and 6.1% had abnormal blood test results (eg, antinuclear antibodies, antineutrophil cytoplasmic antibodies, monoclonal gammopathy of undetermined significance, interleukin-2 receptor antibodies to tissue transglutaminase, and extractable nuclear antigens).22 Among 195 US patients with biopsy-confirmed iiSFN, high erythrocyte sedimentation rate levels, high antinuclear antibodies titers, low complement C3 levels, and antibodies for Sjögren syndrome and celiac disease were each present at more than 300% of population prevalence.22 High erythrocyte sedimentation rate and antinuclear antibodies of 1:160 titer or higher were most common, each affecting 28% of patients. These serologic markers can become false-negative or false-positive during immunotherapy, so screening should precede treatment.22
Primary Sjögren syndrome, the leading systemic immune condition associated with SFN, often remains undiagnosed. One-fourth to one-half of patients with all Sjögren syndrome–associated neuropathy are primarily affected by SFN.24 The prevalence of Sjögren syndrome autoantibodies is 9% to 12% in patients with SFN,22,74 but with nearly 60% of patients with Sjögren syndrome SFN seronegative, there may be more than 20% total prevalence of this disorder in iiSFN.24 Patients with Sjögren syndrome and neuropathy are more likely to be seronegative than those without neuropathy.95 Patients with Sjögren syndrome and SFN can have muted Sjögren symptoms or develop them after onset of neuropathic pain.33 In France, more than 90% of such patients in a large study were female, and their most prevalent symptoms were burning pains (36 of 40 [90%]), numbness (35 of 40 [88%]), tingling (33 of 40 [83%]), pins and needles (29 of 40 [73%]), electric discharges (28 of 40 [70%]), and allodynia (22 of 40 [55%]). Sixty-six percent (25 of 38 patients) had vasomotor symptoms, and 47% (18 of 38 patients) sweated abnormally. Antibodies against muscarinic receptor type 3 and calponin have been reported.96,97 Systemic lupus erythematosus, celiac disease, and psoriatic arthritis are less common associates of SFN.25 Paraneoplastic autoimmunity can cause painful sensory ganglionitis and axonopathy, particularly anti-Hu, anti-CV2/collapsin response-mediator protein-5 (CRMP5), and multiple endocrine neoplasia type 2 (MEN-2).98
What about most patients with iiSFN, who do not have systemic inflammatory diagnoses?56 Some will later receive systemic auto immune diagnoses, and some may have ill-defined systemic pre-dispositions toward dysimmunity, including selective immunoglobulin deficiency (SIgD).9 In 1 study, 15 of 55 patients (28%) had IgG SIgD, 10 of 55 (18%) had IgG subclass deficiency, 8 of 55 (14%) had IgM SIgD, and 6 of 55 (11%) had IgA SIgD.56 We and others have proposed the existence of small fiber–targeting inflammatory SFN, with acute and chronic presentations temporally resembling Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy, and preliminary evidence of episodic relapsing-remitting courses.99 Unlike in Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy, inflammatory cells are not prominent in SFN biopsies or cerebrospinal fluid. Current evidence links autoreactive B cells using complement, with low C4 levels implicating the classic or lectin pathways.9 Direct evidence is emerging in addition to the indirect evidence from medical histories, blood test results, and the responsiveness of some patients to corticosteroids and IVIg. Mice that were injected with sera from 3 patients with acute postinfectious SFN causing distal pain and dysautonomia developed SFN pain behaviors and small-fiber pathologic evidence of neuropathy.100 Passive transfer of human autoantibodies to contactin-associated protein-like 2 (CASPR2) to mice causes pain hypersensitivity and enhanced dorsal-root ganglia cell excitability by reducing potassium-channel Kv1 function.46 Five of 8 children with isolated iiSFN had IgM autoantibodies against trisulfated disaccharide IdoA2S-GlcNS-6S, as did 5 in a study of 22 patients with fibromyalgia, with 86% of these patients (n = 19) having SFN-diagnostic skin biopsies.79,101 Following examples learned from autoimmune encephalitis may enable us to identify cellular and molecular mechanisms of apparently autoimmune SFN. To do so, it is necessary to improve SFN phenotyping and conduct larger serologic studies.
Precision Treatment
Others have reviewed treatments to palliate pain and other SFN symptoms,102,103 so we limit discussion to disease-modifying treatments that restore or preserve small-fiber function. As the benefits of eliminating or curtailing diabetes and neuropathic infectious diseases are established, we discuss emerging treatments, even though long-term benefits and cost-effectiveness are currently unknown. Most treatments for genetic SFN are very expensive, except those for hereditary sensory and autonomic neuropathy type 1. Although oral L-serine can be purchased without prescription or physician over-sight, purity, dosing, and guidelines are not established, particularly for the children who are most likely to benefit.5 Established consortia and charities that support costs of genetic testing and treatment of patients with motor-predominant genetic neuropathies (Charcot-Marie-Tooth disease) typically exclude patients with SFN, so parity for supporting patients with genetic SFN should be accelerated. Massachusetts General Hospital is developing secure internet metrics and data-sharing for such families (https://NeuropathyCommons.org). Large sets of longitudinal, real-world data would advance research and trials.
Potentially disease-modifying treatments are widely available but untrialed for inflammatory SFN, partly because of inadequate case definitions, metrics, and outcome measures. In the interim, trials for other inflammatory neuropathic conditions provide guidance. Treatment with IVIg, which is primary for Guillain-Barré syndrome, chronic inflammatory demyelinating polyneuropathy, and other autoimmune neurological conditions has the most preliminary support for off-label use in apparently immune SFN. In the first large un-controlled study, 55 children and adults received 1 g/kg or more of IVIg every 4 weeks for 3 months or longer, and pretreatment pain severity with a mean of 6.3 (on a scale of 0 to 10) dropped to 5.2 (P = .007).56 Three-quarters of patients and neurologists reported improvement, and 8 of 51 patients (16%) entered sustained remission, permitting IVIg withdrawal.56 Adverse events were all as expected; these were mostly typical infusion reactions.56 Objectively, the prevalence of autonomic function testing diagnostic for SFN dropped from 31 of 35 patients (89%) before treatment to 20 of 35 patients (57%).9 Treatment with IVIg is reportedly helpful for sarcoidosis-associated SFN,104 and a randomized clinical trial for idiopathic SFN is underway in the Netherlands (NCT02637700).105 Smaller reports offer preliminary evidence of efficacy and safety of corticosteroids for acute presentations of apparently inflammatory SFN, particularly in young patients at lower risk of complications.23,49 In a small case series of young patients, 10 of 15 patients (67%) receiving corticosteroid treatment had sustained improvement in symptoms and quantitative autonomic function testing with only 1 significant adverse event, which was cataract development.9 In addition to the absence of untreated control participants, potential confounders included nonspecific benefits for pain, activity, and mood. Oral corticosteroids are globally available, inexpensive, and often the only immunotherapy available, so trials with them should be prioritized.
Conclusions
Small-fiber neuropathy has unappreciated symptoms, and undiagnosed cases appear common, particularly in the fibromyalgia syndrome. Presentations can vary from entirely somatic to entirely autonomic, but most patients have mixed symptoms. Skin biopsy is currently the best established test for objective confirmation of diagnosis. Because small-fiber axons grow throughout life, definitive diagnosis and treatment can lay the foundation for axonal and functional recovery. The young, otherwise healthy, and recently ill should be rapidly diagnosed and optimally treated to preserve life trajectory. Careful evaluation and screening identifies medical causes for almost half of patients whose cases are initially unknown, including seronegative Sjögren syndrome and rare genetic contributors. Multiple factors convey risk of SFN, so in all patients, it is important to manage potential secondary contributors and encourage better axonal perfusion and nutrition. Otherwise healthy children and young adults can develop acute or chronic SFN, apparently often from B-cell autoreactivity and only rarely from genetic causes. These cases, along with non–length-dependent presentations that are typically autoimmune and rarely paraneoplastic, are increasingly treatable, so they require urgent evaluation. Developing and validating simpler diagnostic and tracking metrics is a much-needed precursor for clinical trials. Most importantly, the rapid pace of discovery requires better education of clinicians and the public to improve medical care today.
Funding/Support:
This study was supported by the National Institutes of Health (grant R01-NS093653 [Dr Oaklander] and UL1 TR001102 [Harvard Catalyst/REDCap]), the US Department of Defense (grant GW140169 [Dr Oaklander]), the Freeman fund (Dr Oaklander), and the Italian Ministry of Health (grant/award 5/1000-2014 [Dr Nolano]).
Role of the Funder/Sponsor:
The funders had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Additional Contributions:
The authors gratefully acknowledge the mentorship of visionaries who developed the field of small-fiber neuropathy: John W. Griffin, MD, The Johns Hopkins University School of Medicine, and William R. Kennedy, MD, University of Minnesota. They were not compensated for their contributions. We thank the families of the patients in Figure 2D and F for granting permission to publish this information.
Footnotes
Conflict of Interest Disclosures: None reported.
REFERENCES
- 1.Gregg EW, Sorlie P, Paulose-Ram R, et al. ; 1999–2000 National Health and Nutrition Examination Survey. Prevalence of lower-extremity disease in the US adult population ≥40 years of age with and without diabetes: 1999–2000 National Health and Nutrition Examination Survey. Diabetes Care. 2004;27(7):1591–1597. doi: 10.2337/diacare.27.7.1591 [DOI] [PubMed] [Google Scholar]
- 2.Peters MJ, Bakkers M, Merkies IS, Hoeijmakers JG, van Raak EP, Faber CG. Incidence and prevalence of small-fiber neuropathy: a survey in the Netherlands. Neurology. 2013;81(15):1356–1360. doi: 10.1212/WNL.0b013e3182a8236e [DOI] [PubMed] [Google Scholar]
- 3.Grayston R, Czanner G, Elhadd K, et al. A systematic review and meta-analysis of the prevalence of small fiber pathology in fibromyalgia: implications for a new paradigm in fibromyalgia etiopathogenesis. Semin Arthritis Rheum. 2019;48 (5):933–940. doi: 10.1016/j.semarthrit.2018.08.003 [DOI] [PubMed] [Google Scholar]
- 4.Branco JC, Bannwarth B, Failde I, et al. Prevalence of fibromyalgia: a survey in five European countries. Semin Arthritis Rheum. 2010; 39(6):448–453. doi: 10.1016/j.semarthrit.2008.12.003 [DOI] [PubMed] [Google Scholar]
- 5.Fridman V, Suriyanarayanan S, Novak P, et al. Randomized trial of l-serine in patients with hereditary sensory and autonomic neuropathy type 1. Neurology. 2019;92(4):e359–e370. doi: 10.1212/WNL.0000000000006811 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nolano M, Provitera V, Manganelli F, et al. Non-motor involvement in amyotrophic lateral sclerosis: new insight from nerve and vessel analysis in skin biopsy. Neuropathol Appl Neurobiol. 2017;43(2):119–132. doi: 10.1111/nan.12332 [DOI] [PubMed] [Google Scholar]
- 7.Fukuda T, Takeda S, Xu R, et al. Sema3A regulates bone-mass accrual through sensory innervations. Nature. 2013;497(7450):490–493. doi: 10.1038/nature12115 [DOI] [PubMed] [Google Scholar]
- 8.Steinhoff M, Schmelz M, Szabó IL, Oaklander AL. Clinical presentation, management, and pathophysiology of neuropathic itch. Lancet Neurol. 2018;17(8):709–720. doi: 10.1016/S1474-4422(18)30217-5 [DOI] [PubMed] [Google Scholar]
- 9.Oaklander AL, Klein MM. Evidence of small-fiber polyneuropathy in unexplained, juvenile-onset, widespread pain syndromes. Pediatrics. 2013;131(4):e1091–e1100. doi: 10.1542/peds.2012-2597 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Thieben MJ, Sandroni P, Sletten DM, et al. Postural orthostatic tachycardia syndrome: the Mayo clinic experience. Mayo Clin Proc. 2007;82(3):308–313. doi: 10.1016/S0025-6196(11)61027-6 [DOI] [PubMed] [Google Scholar]
- 11.Mitchell SW. On a rare vaso-motor neurosis of the extremities, and on the maladies with which it may be confounded. Am J Med Sci. 1878;151:17–36. doi: 10.1097/00000441-187815100-00001 [DOI] [Google Scholar]
- 12.Sousa-Valente J, Brain SD. A historical perspective on the role of sensory nerves in neurogenic inflammation. Semin Immunopathol. 2018;40(3):229–236. doi: 10.1007/s00281-018-0673-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Albrecht PJ, Hou Q, Argoff CE, Storey JR, Wymer JP, Rice FL. Excessive peptidergic sensory innervation of cutaneous arteriole-venule shunts (AVS) in the palmar glabrous skin of fibromyalgia patients: implications for widespread deep tissue pain and fatigue. Pain Med. 2013;14(6):895–915. doi: 10.1111/pme.12139 [DOI] [PubMed] [Google Scholar]
- 14.Dori A, Lopate G, Keeling R, Pestronk A. Myovascular innervation: axon loss in small-fiber neuropathies. Muscle Nerve. 2015;51(4):514–521. doi: 10.1002/mus.24356 [DOI] [PubMed] [Google Scholar]
- 15.Joseph P, Sanders J, Rodriguez C, et al. The pathophysiology of chronic fatigue syndrome: results from an invasive cardiopulmonary exercise laboratory. Paper presented at: American Thoracic Society; May 22, 2019; Dallas, Texas. [Google Scholar]
- 16.Selim MM, Wendelschafer-Crabb G, Redmon JB, et al. Gastric mucosal nerve density: a biomarker for diabetic autonomic neuropathy? Neurology. 2010;75(11):973–981. doi: 10.1212/WNL.0b013e3181f25f19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Poda R, Guaraldi P, Solieri L, et al. Standing worsens cognitive functions in patients with neurogenic orthostatic hypotension. Neurol Sci. 2012;33(2):469–473. doi: 10.1007/s10072-011-0746-6 [DOI] [PubMed] [Google Scholar]
- 18.Hsieh PC, Tseng MT, Chao CC, et al. Imaging signatures of altered brain responses in small-fiber neuropathy: reduced functional connectivity of the limbic system after peripheral nerve degeneration. Pain. 2015;156(5):904–916. doi: 10.1097/j.pain.0000000000000128 [DOI] [PubMed] [Google Scholar]
- 19.Henrich F, Magerl W, Klein T, Greffrath W, Treede RD. Capsaicin-sensitive C- and A-fibre nociceptors control long-term potentiation-like pain amplification in humans. Brain. 2015;138(pt 9):2505–2520. doi: 10.1093/brain/awv108 [DOI] [PubMed] [Google Scholar]
- 20.Indart S, Hugon J, Guillausseau PJ, et al. Impact of pain on cognitive functions in primary Sjögren syndrome with small fiber neuropathy: 10 cases and a literature review. Medicine (Baltimore). 2017;96(16):e6384. doi: 10.1097/MD.0000000000006384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gemignani F, Giovanelli M, Vitetta F, et al. Non-length dependent small fiber neuropathy: a prospective case series. J Peripher Nerv Syst. 2010;15(1):57–62. doi: 10.1111/j.1529-8027.2010.00252.x [DOI] [PubMed] [Google Scholar]
- 22.Lang M, Treister R, Oaklander AL. Diagnostic value of blood tests for occult causes of initially idiopathic small-fiber polyneuropathy. J Neurol. 2016;263(12):2515–2527. doi: 10.1007/s00415-016-8270-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dabby R, Gilad R, Sadeh M, Lampl Y, Watemberg N. Acute steroid responsive small-fiber sensory neuropathy: a new entity? J Peripher Nerv Syst. 2006;11(1):47–52. doi: 10.1111/j.1085-9489.2006.00062.x [DOI] [PubMed] [Google Scholar]
- 24.Lefaucheur JP, Sène D, Oaklander AL. Primary Sjögren’s syndrome. N Engl J Med. 2018;379(1):96. doi: 10.1056/NEJMc1804598 [DOI] [PubMed] [Google Scholar]
- 25.Brannagan TH III, Hays AP, Chin SS, et al. Small-fiber neuropathy/neuronopathy associated with celiac disease: skin biopsy findings. Arch Neurol. 2005;62(10):1574–1578. doi: 10.1001/archneur.62.10.1574 [DOI] [PubMed] [Google Scholar]
- 26.de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy—a large cohort study and review of the literature. Eur J Neurol. 2018;25(2):348–355. doi: 10.1111/ene.13508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ørstavik K, Norheim I, Jørum E. Pain and small-fiber neuropathy in patients with hypothyroidism. Neurology. 2006;67(5):786–791. doi: 10.1212/01.wnl.0000234035.13779.4a [DOI] [PubMed] [Google Scholar]
- 28.Latov N, Vo ML, Chin RL, Carey BT, Langsdorf JA, Feuer NT. Abnormal nutritional factors in patients evaluated at a neuropathy center. J Clin Neuromuscul Dis. 2016;17(4):212–214. doi: 10.1097/CND.0000000000000128 [DOI] [PubMed] [Google Scholar]
- 29.Hsu JL, Liao MF, Hsu HC, et al. A prospective, observational study of patients with uncommon distal symmetric painful small-fiber neuropathy. PLoS One. 2017;12(9):e0183948. doi: 10.1371/journal.pone.0183948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Griffin JW, Cornblath DR, Alexander E, et al. Ataxic sensory neuropathy and dorsal root ganglionitis associated with Sjögren’s syndrome. Ann Neurol. 1990;27(3):304–315. doi: 10.1002/ana.410270313 [DOI] [PubMed] [Google Scholar]
- 31.Birnbaum J, Lalji A, Piccione EA, Izbudak I. Magnetic resonance imaging of the spinal cord in the evaluation of 3 patients with sensory neuronopathies: diagnostic assessment, indications of treatment response, and impact of autoimmunity. a case report. Medicine (Baltimore). 2017;96(49):e8483. doi: 10.1097/MD.0000000000008483 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lauria G, McArthur JC, Hauer PE, Griffin JW, Cornblath DR. Neuropathological alterations in diabetic truncal neuropathy: evaluation by skin biopsy. J Neurol Neurosurg Psychiatry. 1998;65(5):762–766. doi: 10.1136/jnnp.65.5.762 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Birnbaum J, Lalji A, Saed A, Baer AN. Biopsy-proven small-fiber neuropathy in primary Sjogren’s syndrome: neuropathic pain characteristics, autoantibody findings, and histopathological features. Arthritis Care Res (Hoboken). 2019;71(7):936–948. doi: 10.1002/acr.23762 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zis P, Sarrigiannis PG, Rao DG, Hadjivassiliou M. Gluten neuropathy: prevalence of neuropathic pain and the role of gluten-free diet. J Neurol. 2018;265(10):2231–2236. doi: 10.1007/s00415-018-8978-5 [DOI] [PubMed] [Google Scholar]
- 35.Nolano M, Crisci C, Santoro L, et al. Absent innervation of skin and sweat glands in congenital insensitivity to pain with anhidrosis. Clin Neurophysiol. 2000;111(9):1596–1601. doi: 10.1016/S1388-2457(00)00351-5 [DOI] [PubMed] [Google Scholar]
- 36.Hilz MJ, Axelrod FB, Bickel A, et al. Assessing function and pathology in familial dysautonomia: assessment of temperature perception, sweating and cutaneous innervation. Brain. 2004;127(pt 9):2090–2098. doi: 10.1093/brain/awh235 [DOI] [PubMed] [Google Scholar]
- 37.Di Leo R, Nolano M, Boman H, et al. Central and peripheral autonomic failure in cold-induced sweating syndrome type 1. Neurology. 2010;75(17):1567–1569. doi: 10.1212/WNL.0b013e3181f96237 [DOI] [PubMed] [Google Scholar]
- 38.Fridman V, Oaklander AL, David WS, et al. Natural history and biomarkers in hereditary sensory neuropathy type 1. Muscle Nerve. 2015;51(4):489–495. doi: 10.1002/mus.24336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fridman V, Novak P, David W, et al. A randomized, double-blind, placebo-controlled, delayed-start trial to evaluate the safety and efficacy of L-serine in subjects with hereditary sensory and autonomic neuropathy type 1 (HSAN1). Ann Neurol. 2016;80:S243. [Google Scholar]
- 40.Manganelli F, Pisciotta C, Provitera V, et al. Autonomic nervous system involvement in a new CMT2B family. J Peripher Nerv Syst. 2012;17(3):361–364. doi: 10.1111/j.1529-8027.2012.00415.x [DOI] [PubMed] [Google Scholar]
- 41.Hofmann L, Hose D, Grießhammer A, et al. Characterization of small fiber pathology in a mouse model of Fabry disease. Elife. 2018;7:e39300. doi: 10.7554/eLife.39300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Eijkenboom I, Sopacua M, Hoeijmakers JGJ, et al. Yield of peripheral sodium channels gene screening in pure small fibre neuropathy. J Neurol Neurosurg Psychiatry. 2019;90(3):342–352. doi: 10.1136/jnnp-2018-319042 [DOI] [PubMed] [Google Scholar]
- 43.Salvatierra J, Diaz-Bustamante M, Meixiong J, Tierney E, Dong X, Bosmans F. A disease mutation reveals a role for NaV1.9 in acute itch. J Clin Invest. 2018;128(12):5434–5447. doi: 10.1172/JCI122481 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Blesneac I, Themistocleous AC, Fratter C, et al. Rare NaV1.7 variants associated with painful diabetic peripheral neuropathy. Pain. 2018;159(3):469–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wadhawan S, Pant S, Golhar R, et al. NaV channel variants in patients with painful and nonpainful peripheral neuropathy. Neurol Genet. 2017;3(6):e207. doi: 10.1212/NXG.0000000000000207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dawes JM, Weir GA, Middleton SJ, et al. Immune or genetic-mediated disruption of CASPR2 causes pain hypersensitivity due to enhanced primary afferent excitability. Neuron. 2018;97(4): 806–822.e10. doi: 10.1016/j.neuron.2018.01.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Suarez GA, Fealey RD, Camilleri M, Low PA. Idiopathic autonomic neuropathy: clinical, neurophysiologic, and follow-up studies on 27 patients. Neurology. 1994;44(9):1675–1682. doi: 10.1212/WNL.44.9.1675 [DOI] [PubMed] [Google Scholar]
- 48.Watari M, Nakane S, Mukaino A, et al. Autoimmune postural orthostatic tachycardia syndrome. Ann Clin Transl Neurol. 2018;5(4):486–492. doi: 10.1002/acn3.524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Paticoff J, Valovska A, Nedeljkovic SS, Oaklander AL. Defining a treatable cause of erythromelalgia: acute adolescent autoimmune small-fiber axonopathy. Anesth Analg. 2007;104(2): 438–441. doi: 10.1213/01.ane.0000252965.83347.25 [DOI] [PubMed] [Google Scholar]
- 50.Seneviratne U, Gunasekera S. Acute small fibre sensory neuropathy: another variant of Guillain-Barré syndrome? J Neurol Neurosurg Psychiatry. 2002;72(4):540–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Morales PS, Escobar RG, Lizama M, et al. Paediatric hypertension-associated erythromelalgia responds to corticosteroids and is not associated with SCN9A mutations. Rheumatology (Oxford). 2012;51(12):2295–2296. doi: 10.1093/rheumatology/kes098 [DOI] [PubMed] [Google Scholar]
- 52.Schofield JR, Hendrickson JE. Autoimmunity, autonomic neuropathy, and the HPV vaccination: a vulnerable subpopulation. Clin Pediatr (Phila). 2018;57(5):603–606. doi: 10.1177/0009922817728701 [DOI] [PubMed] [Google Scholar]
- 53.Kafaie J, Kim M, Krause E. Small fiber neuropathy following vaccination. J Clin Neuromuscul Dis. 2016;18(1):37–40. doi: 10.1097/CND.0000000000000130 [DOI] [PubMed] [Google Scholar]
- 54.Low PA, Denq JC, Opfer-Gehrking TL, Dyck PJ, O’Brien PC, Slezak JM. Effect of age and gender on sudomotor and cardiovagal function and blood pressure response to tilt in normal subjects. Muscle Nerve. 1997;20(12):1561–1568. doi: [DOI] [PubMed] [Google Scholar]
- 55.Klein MM, Downs H, O’Neil K, Oaklander AL. Skin biopsy of normal children demonstrates inverse correlation between age and epidermal nerve fiber density, meaning age-specific norms are needed. Ann Neurol. 2014;76(suppl 18):S69. [Google Scholar]
- 56.Liu X, Treister R, Lang M, Oaklander AL. IVIg for apparently autoimmune small-fiber polyneuropathy: first analysis of efficacy and safety Ther Adv Neurol Disord. 2018;11:1756285617744484. doi: 10.1177/1756285617744484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bastyr EJ III, Price KL, Bril V; MBBQ Study Group. Development and validity testing of the neuropathy total symptom score-6: questionnaire for the study of sensory symptoms of diabetic peripheral neuropathy. Clin Ther. 2005;27(8):1278–1294. doi: 10.1016/j.clinthera.2005.08.002 [DOI] [PubMed] [Google Scholar]
- 58.Zilliox L, Peltier AC, Wren PA, et al. Assessing autonomic dysfunction in early diabetic neuropathy: the Survey of Autonomic Symptoms. Neurology. 2011;76(12):1099–1105. doi: 10.1212/WNL.0b013e3182120147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Meijer JW, Smit AJ, Sonderen EV, Groothoff JW, Eisma WH, Links TP. Symptom scoring systems to diagnose distal polyneuropathy in diabetes: the Diabetic Neuropathy Symptom score. Diabet Med. 2002;19(11):962–965. doi: 10.1046/j.1464-5491.2002.00819.x [DOI] [PubMed] [Google Scholar]
- 60.Hoitsma E, De Vries J, Drent M. The small fiber neuropathy screening list: construction and cross-validation in sarcoidosis. Respir Med. 2011;105(1):95–100. doi: 10.1016/j.rmed.2010.09.014 [DOI] [PubMed] [Google Scholar]
- 61.Oldenburg J, Fosså SD, Dahl AA. Scale for chemotherapy-induced long-term neurotoxicity (SCIN): psychometrics, validation, and findings in a large sample of testicular cancer survivors. Qual Life Res. 2006;15(5):791–800. doi: 10.1007/s11136-005-5370-6 [DOI] [PubMed] [Google Scholar]
- 62.Treister R, O’Neil K, Downs HM, Oaklander AL. Validation of the composite autonomic symptom scale 31 (COMPASS-31) in patients with and without small fiber polyneuropathy. Eur J Neurol. 2015;22(7):1124–1130. doi: 10.1111/ene.12717 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dworkin RH, Turk DC, Revicki DA, et al. Development and initial validation of an expanded and revised version of the Short-Form McGill Pain Questionnaire (SF-MPQ-2). Pain. 2009;144(1–2):35–42. doi: 10.1016/j.pain.2009.02.007 [DOI] [PubMed] [Google Scholar]
- 64.Lodahl M, Treister R, Oaklander AL. Specific symptoms may discriminate between fibromyalgia patients with vs without objective test evidence of small-fiber polyneuropathy. Pain Rep. 2017;3(1):e633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Brouwer BA, Bakkers M, Hoeijmakers JG, Faber CG, Merkies IS. Improving assessment in small fiber neuropathy. J Peripher Nerv Syst. 2015;20(3):333–340. doi: 10.1111/jns.12128 [DOI] [PubMed] [Google Scholar]
- 66.Treister R, Lodahl M, Lang M, Tworoger SS, Sawilowsky S, Oaklander AL. Initial development and validation of a patient-reported symptom survey for small-fiber polyneuropathy. J Pain. 2017; 18(5):556–563. doi: 10.1016/j.jpain.2016.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Singleton JR, Bixby B, Russell JW, et al. The Utah Early Neuropathy scale: a sensitive clinical scale for early sensory predominant neuropathy. J Peripher Nerv Syst. 2008;13(3):218–227. doi: 10.1111/j.1529-8027.2008.00180.x [DOI] [PubMed] [Google Scholar]
- 68.Zirpoli G, Klein MM, Downs S, Farhad K, Oaklander AL. Validation of the Mass General Neuropathy Exam Tool (MAGNET) for initial diagnosis of length-dependent small-fiber polyneuropathy; 2018 Peripheral Nerve Society Annual Meeting, 21‐25 July, 2018: Baltimore, Maryland. J Peripher Nerv Syst. 2018;23(4):249–405. doi: 10.1111/jns.12290 [DOI] [Google Scholar]
- 69.Wilkinson KD, Lee KM, Deshpande S, Duerksen-Hughes P, Boss JM, Pohl J. The neuron-specific protein PGP 9.5 is a ubiquitin carboxyl-terminal hydrolase. Science. 1989;246 (4930):670–673. doi: 10.1126/science.2530630 [DOI] [PubMed] [Google Scholar]
- 70.Lauria G, Hsieh ST, Johansson O, et al. ; European Federation of Neurological Societies; Peripheral Nerve Society. European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy: report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. Eur J Neurol. 2010;17(7):903–912, e44–e49. doi: 10.1111/j.1468-1331.2010.03023.x [DOI] [PubMed] [Google Scholar]
- 71.Thompson RJ, Doran JF, Jackson P, Dhillon AP, Rode J. PGP 9.5–a new marker for vertebrate neurons and neuroendocrine cells. Brain Res. 1983; 278(1–2):224–228. doi: 10.1016/0006-8993(83)90241-X [DOI] [PubMed] [Google Scholar]
- 72.Provitera V, Gibbons CH, Wendelschafer-Crabb G, et al. A multi-center, multinational age- and gender-adjusted normative dataset for immunofluorescent intraepidermal nerve fiber density at the distal leg. Eur J Neurol. 2016;23(2): 333–338. doi: 10.1111/ene.12842 [DOI] [PubMed] [Google Scholar]
- 73.England JD, Gronseth GS, Franklin G, et al. ; American Academy of Neurology. Practice parameter: evaluation of distal symmetric polyneuropathy, role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review): report of the American Academy of Neurology, American Association of Neuromuscular and Electrodiagnostic Medicine, and American Academy of Physical Medicine and Rehabilitation. Neurology. 2009;72(2):177–184. doi: 10.1212/01.wnl.0000336345.70511.0f [DOI] [PubMed] [Google Scholar]
- 74.Devigili G, Tugnoli V, Penza P, et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain. 2008;131(pt 7):1912–1925. doi: 10.1093/brain/awn093 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.McArthur JC, Stocks EA, Hauer P, Cornblath DR, Griffin JW. Epidermal nerve fiber density: normative reference range and diagnostic efficiency. Arch Neurol. 1998;55(12):1513–1520. doi: 10.1001/archneur.55.12.1513 [DOI] [PubMed] [Google Scholar]
- 76.Jin P, Cheng L, Chen M, Zhou L. Low sensitivity of skin biopsy in diagnosing small fiber neuropathy in Chinese Americans. J Clin Neuromuscul Dis. 2018; 20(1):1–6. doi: 10.1097/CND.0000000000000223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Lauria G, Holland N, Hauer P, Cornblath DR, Griffin JW, McArthur JC. Epidermal innervation: changes with aging, topographic location, and in sensory neuropathy. J Neurol Sci. 1999;164(2):172–178. doi: 10.1016/S0022-510X(99)00063-5 [DOI] [PubMed] [Google Scholar]
- 78.Provitera V, Gibbons CH, Wendelschafer-Crabb G, et al. The role of skin biopsy in differentiating small-fiber neuropathy from ganglionopathy. Eur J Neurol. 2018;25(6):848–853. doi: 10.1111/ene.13608 [DOI] [PubMed] [Google Scholar]
- 79.Malik A, Lopate G, Hayat G, et al. Prevalence of axonal sensory neuropathy with IgM binding to trisulfated heparin disaccharide in patients with fibromyalgia. J Clin Neuromuscul Dis. 2019;20(3):103–110. doi: 10.1097/CND.0000000000000236 [DOI] [PubMed] [Google Scholar]
- 80.Walk D, Wendelschafer-Crabb G, Davey C, Kennedy WR. Concordance between epidermal nerve fiber density and sensory examination in patients with symptoms of idiopathic small fiber neuropathy. J Neurol Sci. 2007;255(1–2):23–26. doi: 10.1016/j.jns.2007.01.052 [DOI] [PubMed] [Google Scholar]
- 81.Nolano M, Biasiotta A, Lombardi R, et al. Epidermal innervation morphometry by immunofluorescence and bright-field microscopy. J Peripher Nerv Syst. 2015;20(4):387–391. doi: 10.1111/jns.12146 [DOI] [PubMed] [Google Scholar]
- 82.Low VA, Sandroni P, Fealey RD, Low PA. Detection of small-fiber neuropathy by sudomotor testing. Muscle Nerve. 2006;34(1):57–61. doi: 10.1002/mus.20551 [DOI] [PubMed] [Google Scholar]
- 83.Singer W, Spies JM, McArthur J, et al. Prospective evaluation of somatic and autonomic small fibers in selected autonomic neuropathies. Neurology. 2004;62(4):612–618. doi: 10.1212/01.WNL.0000110313.39239.82 [DOI] [PubMed] [Google Scholar]
- 84.Thaisetthawatkul P, Fernandes Filho JA, Herrmann DN. Contribution of QSART to the diagnosis of small fiber neuropathy. Muscle Nerve. 2013;48(6):883–888. doi: 10.1002/mus.23891 [DOI] [PubMed] [Google Scholar]
- 85.Freeman R, Chase KP, Risk MR. Quantitative sensory testing cannot differentiate simulated sensory loss from sensory neuropathy. Neurology. 2003;60(3):465–470. doi: 10.1212/WNL.60.3.465 [DOI] [PubMed] [Google Scholar]
- 86.Duchesne M, Richard L, Vallat JM, Magy L. Assessing sudomotor impairment in patients with peripheral neuropathy: comparison between electrochemical skin conductance and skin biopsy. Clin Neurophysiol. 2018;129(7):1341–1348. doi: 10.1016/j.clinph.2018.04.608 [DOI] [PubMed] [Google Scholar]
- 87.Provitera V, Nolano M, Caporaso G, Stancanelli A, Santoro L, Kennedy WR. Evaluation of sudomotor function in diabetes using the dynamic sweat test. Neurology. 2010;74(1):50–56. doi: 10.1212/WNL.0b013e3181c7da4b [DOI] [PubMed] [Google Scholar]
- 88.Azmi S, Ferdousi M, Petropoulos IN, et al. Corneal confocal microscopy shows an improvement in small-fiber neuropathy in subjects with type 1 diabetes on continuous subcutaneous insulin infusion compared with multiple daily injection. Diabetes Care. 2015;38(1):e3–e4. doi: 10.2337/dc14-1698 [DOI] [PubMed] [Google Scholar]
- 89.Lefaucheur JP, Wahab A, Planté-Bordeneuve V, et al. Diagnosis of small fiber neuropathy: a comparative study of five neurophysiological tests. Neurophysiol Clin. 2015;45(6):445–455. doi: 10.1016/j.neucli.2015.09.012 [DOI] [PubMed] [Google Scholar]
- 90.Farhad K, Traub R, Ruzhansky KM, Brannagan TH III. Causes of neuropathy in patients referred as “idiopathic neuropathy”. Muscle Nerve. 2016;53(6): 856–861. doi: 10.1002/mus.24969 [DOI] [PubMed] [Google Scholar]
- 91.Dyck PJ, Clark VM, Overland CJ, et al. Impaired glycemia and diabetic polyneuropathy: the OC IG Survey. Diabetes Care. 2012;35(3):584–591. doi: 10.2337/dc11-1421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Koike H, Takahashi M, Ohyama K, et al. Clinicopathologic features of folate-deficiency neuropathy. Neurology. 2015;84(10):1026–1033. doi: 10.1212/WNL.0000000000001343 [DOI] [PubMed] [Google Scholar]
- 93.de Greef BT, Hoeijmakers JG, Wolters EE, et al. No Fabry Disease in Patients Presenting with Isolated Small Fiber Neuropathy. PLoS One. 2016;11(2):e0148316. doi: 10.1371/journal.pone.0148316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Splinter K, Adams DR, Bacino CA, et al. ; Undiagnosed Diseases Network. Effect of genetic diagnosis on patients with previously undiagnosed disease. N Engl J Med. 2018;379(22):2131–2139. doi: 10.1056/NEJMoa1714458 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Sène D, Cacoub P, Authier FJ, et al. Sjögren syndrome-associated small fiber neuropathy: characterization from a prospective series of 40 cases. Medicine (Baltimore). 2013;92(5):e10–e18. doi: 10.1097/MD.0000000000000005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dawson L, Tobin A, Smith P, Gordon T. Antimuscarinic antibodies in Sjögren’s syndrome: where are we, and where are we going? Arthritis Rheum. 2005;52(10):2984–2995. doi: 10.1002/art.21347 [DOI] [PubMed] [Google Scholar]
- 97.Birnbaum J, Höke A. Anti-calponin-3 autoantibodies: a new specificity in patients with Sjögren’s syndrome. Arthritis Rheum. 2018;70(10):1610–1616. doi: 10.1002/art.40550 [DOI] [PubMed] [Google Scholar]
- 98.Zis P, Paladini A, Piroli A, McHugh PC, Varrassi G, Hadjivassiliou M. Pain as a first manifestation of paraneoplastic neuropathies: a systematic review and meta-analysis. Pain Ther. 2017;6(2):143–151. doi: 10.1007/s40122-017-0076-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Spoendlin J, Roschach B, Landolt M, Oaklander AL. Characterization of a relapsing/remitting disease course for corticosteroid-responsive small-fiber polyneuropathy. J Peripher Nerv Syst. in press. [Google Scholar]
- 100.Yuki N, Chan AC, Wong AHY, et al. Acute painful autoimmune neuropathy: a variant of Guillain-Barré syndrome. Muscle Nerve. 2018;57(2):320–324. doi: 10.1002/mus.25738 [DOI] [PubMed] [Google Scholar]
- 101.Kafaie J, Al Balushi A, Kim M, Pestronk A. Clinical and laboratory profiles of idiopathic small fiber neuropathy in children: case series. J Clin Neuromuscul Dis. 2017;19(1):31–37. doi: 10.1097/CND.0000000000000178 [DOI] [PubMed] [Google Scholar]
- 102.Finnerup NB. Nonnarcotic methods of pain management. N Engl J Med. 2019;380(25):2440–2448. doi: 10.1056/NEJMra1807061 [DOI] [PubMed] [Google Scholar]
- 103.Low PA, Tomalia VA. Orthostatic hypotension: mechanisms, causes, management. J Clin Neurol. 2015;11(3):220–226. doi: 10.3988/jcn.2015.11.3.220 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tavee JO, Karwa K, Ahmed Z, Thompson N, Parambil J, Culver DA. Sarcoidosis-associated small fiber neuropathy in a large cohort: Clinical aspects and response to IVIG and anti-TNF alpha treatment. Respir Med. 2017;126:135–138. doi: 10.1016/j.rmed.2017.03.011 [DOI] [PubMed] [Google Scholar]
- 105.de Greef BT, Geerts M, Hoeijmakers JG, Faber CG, Merkies IS. Intravenous immunoglobulin therapy for small fiber neuropathy: study protocol for a randomized controlled trial. Trials. 2016;17(1): 330. doi: 10.1186/s13063-016-1450-x [DOI] [PMC free article] [PubMed] [Google Scholar]