Paraneoplastic Autonomic Neuropathy

Back

Background

Autoimmune paraneoplastic autonomic neuropathy is a rare paraneoplastic neurological syndrome (PNS), which manifests as disturbance in sympathetic and/or parasympathetic nervous system function. Most often, autonomic problems in cancer patients are attributable to prolonged bed rest, neurotoxic chemotherapy, high-dose analgesics, and malnutrition. However, paraneoplastic autonomic neuropathy should be considered in all cancer patients who present with signs or symptoms of autonomic nervous system dysfunction. Patients may develop autonomic disturbances at any time relative to the diagnosis of cancer. The autonomic problems can precede the cancer diagnosis, and a high level of suspicion is then required to identify the underlying neoplasm.

An international expert group established diagnostic criteria in 2004 that divided patients with a suspected paraneoplastic neurological syndrome into "definite" and "probable" categories.[1] These criteria are based on the presence or absence of cancer, the presence of well-characterized paraneoplastic (onconeural) antibodies, and the type of clinical syndrome.

The clinical syndromes are divided into classical and non-classical categories.[1]

Classical paraneoplastic neurological syndromes include:

The most common non-classical paraneoplastic neurological syndromes include:

Patients with a definite PNS include those with the following:

Patients with a possible PNS include those with the following:

The main paraneoplastic syndromes associated with autonomic dysfunction include paraneoplastic autoimmune autonomic gangliopathy (AAG), paraneoplastic sensory neuropathies and neuronopathies, paraneoplastic encephalomyeloneuropathies, and Lambert-Eaton myasthenic syndrome (LEMS). Each one of these disorders may have other distinct symptoms and findings in addition to autonomic disturbances.

Pathophysiology

It has long been known that patients with small-cell lung cancer[2] develop neurological signs and symptoms with increased frequency[3] ; however, many cancers including other lung tumors[4] , thymoma, Hodgkin disease[5] , other lymphomas[6] , testicular, ovarian, and breast carcinoma can also cause paraneoplastic neurological syndromes.[7]

Exactly how cancers result in paraneoplastic neurological symptoms is incompletely understood. The development of these syndromes are not due to direct tumor invasion. Expression of onconeural antigens by the cancer cells resulting in autoimmunity appears to be the mechanism. The classical paraneoplastic antibodies are directed at intracellular antigens[8] and may not be directly pathogenic. Passive transfer experiments have generally been unsuccessful. However, antibodies directed against exposed antigens, namely ganglionic acetylcholine receptors and voltage-gated calcium and potassium channels, appear more directly pathogenic. With these antibody types, several passive transfer experiments have been positive, confirming the antibodies direct pathogenic effect.

Autoantibodies to these extracellularly exposed membrane proteins often occur without an underlying malignancy. A search for a malignancy should still be undertaken, but these conditions may occur through an idiopathic autoimmune process.

Many antineuronal antibodies have been described to date, and new antibodies are described each year. Sometimes, more than 1 type of antibody is found in a single patient, and the same type of antibody may be associated with very different syndromes in other patients.[9] It is also entirely possible that a patient who lacks identifiable autoantibodies but has a tumor known to express epitopes similar to neuronal structures (eg, small-cell lung cancer), may suffer from paraneoplastic autonomic dysfunction due to an antibody that has not been identified.

Autonomic dysfunction can occur when this autoimmune process causes sufficient damage to the autonomic nervous system. Limited data are available regarding the immune attack on preganglionic neurons or central autonomic pathways. Typical pathological changes include lymphocytic infiltrates[10] and vascular cuffing; as is shown in the image below from a case of anti-Hu encephalomyeloneuropathy. Similar attacks on autonomic postganglionic and myenteric neurons can occur with antineuronal antibodies.



View Image

Paraneoplastic autonomic neuropathy. Central nervous system sections from a patient with autonomic failure, oat-cell carcinoma of the lung, and positi....

The best understood syndromes involving paraneoplastic autonomic dysfunction are paraneoplastic autoimmune autonomic gangliopathy (AAG), paraneoplastic sensory neuropathy and/or neuronopathy, paraneoplastic encephalomyeloneuropathy, and Lambert-Eaton myasthenic syndrome.

Anti-ganglionic acetylcholine receptor antibodies

These antibodies directed at nicotinic acetylcholine in receptors containing the α3 subunit expressed on sympathetic and parasympathetic ganglia cause paraneoplastic AAG.[11] Ganglionic acetylcholine receptor antibodies do not bind to muscle acetylcholine receptors and are not known to cause myasthenia gravis (MG). These anti-ganglionic acetylcholine receptor antibodies are likely directly pathogenic and have an associated clinical picture that is usually characterized by autonomic failure. Only about 20% of cases with AAG appear to be paraneoplastic in origin[12] , relating to small-cell lung carcinoma, thymoma, bladder carcinoma, and rectal carcinoma[13] ; many of the remaining cases appear to be postinfectious analogues to Gullian-Barre syndrome. A case of paraneoplastic AAG coexisting with antibodies against muscle acetylcholine receptors has been reported.[14]

Anti-Hu

Anti-Hu antibodies (which are also called antineuronal nuclear antibody type 1 [ANNA-1])[2] are the second most pertinent to autonomic dysfunction; and are often seen in the setting of small-cell lung cancer. These antibodies can also be seen in non–small-cell lung cancer, neuroblastoma, and malignant disorders of the gastrointestinal tract, prostate, breast, bladder, kidney, pancreas, testicle, and ovary.[15, 13, 16] The autoimmune response is directed to the Hu antigen, an AU-rich 3′-untranslated m-RNA sequence that is expressed by many small-cell lung cancer cells and by all neurons.

Antibodies to the Hu onconeuronal antigen can affect almost any portion of the central nervous system (CNS) or peripheral nervous system.[17] The anti-Hu antibody is most often associated with a paraneoplastic sensory neuronopathy, which involves destruction of primary sensory neurons.[18] When anti-Hu is present, about 25% of patients have some form of dysautonomia, most frequently gastrointestinal manifestations.[19] This antibody is diagnostically useful, but the exact role of humoral immunity in causing neural degeneration remains uncertain.

Anti-NMDA receptor

A recently discovered antibody to the NMDA receptor has been found to be associated with psychosis, seizures, and autonomic instability. In a case series, it was found to be the cause of 4% of encephalitis cases.[20] The antibody is found in cerebrospinal fluid (CSF) in 94% of cases.[21] In one case series, 55% of patients were found to have a teratoma containing neural tissue with NMDA receptors expressed.[22] Typically, the tumor is an ovarian teratoma; however, testicular germ cell tumors, neuroblastoma, and Hodgkin lymphoma have also been reported.[21] There is some evidence that NMDA-receptor antibody encephalitis may result in centrally mediated sinus node dysregulation, including cardiac arrest.[23, 24]

Anti-VGCC

In Lambert-Eaton myasthenic syndrome (LEMS), antibodies against P/Q type voltage-gated calcium channels (VGCC) are present. These antibodies lead to impaired presynaptic calcium release at the neuromuscular junction, resulting in predominantly proximal muscle weakness. These antibodies not only block the voltage-gated calcium channels at the neuromuscular junction but also block them at parasympathetic and sympathetic nerve terminals, thus creating autonomic insufficiency and autonomic symptoms. This syndrome is associated with malignancy 50-60% of the time, typically small-cell lung cancer, but also non–small-cell lung cancer, malignant thymoma, lymphoma, leukemia, and carcinomas of the breast, prostate, larynx, and gallbladder.[15, 25, 19] Autonomic dysfunction in LEMS is normally mild.

Other paraneoplastic antibodies

Collapsin response-mediator protein (CRMP-5), also known as CV-2, is another paraneoplastic antibody that has associated autonomic dysfunction in 33% of cases.[19] CRMP-5 is most often seen with small-cell lung cancer. Purkinje cell antibody-2 (PCA-2) frequently causes cerebellar degeneration but can also be associated with autonomic failure.[26] Antibodies against synaptophysin may be associated with lung cancer and gastrointestinal disturbances from the myenteric plexus.[27] Voltage-gated potassium channel (VGKC) antibodies[28] have been reported to cause autonomic failure in 33% of cases. VGKC antibodies are associated with neuroendocrine tumors including small-cell lung carcinoma, retinoblastoma, oligodendroglioma, melanoma, leiomyosarcoma, and hematologic malignancies.[29] . There is a VGKC antibody-complex associated with contractin associate protein 2 (CASPR2) that has been observed with autonomic dysfunction with thymoma or small cell lung cancer.[30]

Gastric dysmotility paraneoplastic disorders can often be seen with antibodies such anti-Yo, anti-Hu, anti-ganglionic acetylcholine receptor, CRMP-5, anti-VGCC, anti-VGKC, GAD65, islet cell antigen 512 (IA-2), gastric parietal cell, muscle striational, thyroid peroxidase, or thyroglobulin autoantibodies.[31] . One case report documents a patient with functional colonic obstruction ultimately resulting from paraneoplastic ocular myasthenia associated with a thymoma.[32]

Stiff person syndrome, when caused by a paraneoplastic disorder (about 5% of cases), has been reported to associate with anti-GAD65, amphiphysin antibodies, anti-Ri (ANNA-2 antibodies), anti-gephyrin, anti ICA 105, and anti-17-B-hydroxysteroid dehydrogenase type 4. Associated tumors are found in breast, thymus, lung, and kidney cancers as well as multiple myeloma.[33]

Progressive encephalomyelitis with rigidity and myoclonus (PERM) has been found to have a paraneoplastic cause in 20% of patients associated with anti-Ri (ANNA-2), anti-amphiphysin, and anti-GAD65. Autonomic symptoms such as pyrexia and diaphoresis are exhibited in 66% of patients with PERM.[33]

Anti-Yo, also known as Purkinje cell antibody 1 (PCA-1) is associated with breast, ovarian, fallopian tube, or uterine cancers.[9] It targets an intracellular antigen called CDR2. The typical presentation is gastric dysmotility.[15]

Many other autoantibodies may also play a role in paraneoplastic dysautonomia, with more being found every year. Recent examples include synaptophysin, SOX, ZIC, anti-AMPA, and anti-GABA, and antibodies to inositol 1,4,5-trisphosphate receptor type 1 (ITRP1) and dipeptidyl-peptidase-like protein-6 (DPP6, also known as DPPX).[34, 35, 36, 37]

The autoimmunity in paraneoplastic neurological syndromes does appear to confer some degree of antitumor effect. Inflammation similar to what is seen in the nervous system also affects the tumor.

There have been several reported cases in which Hu antibody has occurred with classical paraneoplastic symptoms that spontaneously resolved without an underlying tumor being identified.[38] This could be due to a spontaneous cure of the underlying cancer, possibly due to an anti-tumor effect of the paraneoplastic antibodies.

Epidemiology

Frequency

United States

In the United States, the precise incidence of paraneoplastic autonomic disorders in patients with cancer is not known, but the current best estimate is that less than 1% of patients with solid tumors develop any kind of PNS.

International

In the United Kingdom, a national screening program found 63 patients with paraneoplastic neurological symptoms (not including LEMS) from 2000-2001.[7]

Mortality/Morbidity

Morbidity and death can result from severe failure of autonomic function. However, milder autonomic disturbances may be obscured by prominent symptoms of systemic cancer, anticancer therapy, or other peripheral nervous system and CNS damage.

Race

No data suggests differences in frequency or outcomes based on race.

Sex

Conflicting evidence exists with many case series suggesting higher frequency in women[7, 39] but others suggesting higher frequency in men.

Age

PNS can occur at any age, many case series have recorded median age of onset in the sixth[7] or seventh decade. Anti-NMDA receptor antibodies tend to affect much younger patients with a median age in the early 20s.[22]

History

Patients may present with autonomic neuropathy prior to diagnosis of cancer, at the time of cancer diagnosis, or after the treatment of cancer. Autonomic neuropathy often presents as orthostatic hypotension. This may be profound, keeping patients bedridden in spite of aggressive therapy to maintain blood pressure. However, careful evaluation may reveal more widespread disturbances. Abnormal gastrointestinal motility may cause a spectrum of problems from mild constipation and nausea to intestinal pseudo-obstruction[40] due to autoimmune attack on myenteric neurons. Urinary incontinence, erectile dysfunction, cardiac arrhythemias, dry eyes, dry mouth, pupillary changes (sluggish or fixed pupils), and abnormalities of sweating are also common.

Concomitant somatic neuropathy is common and may cause pain and sensory loss, often in a length-dependent "stocking and glove" pattern but occasionally in a patchy distribution. Pain may be lightning like or burning. When motor nerves are affected, patients may report weakness.

If the paraneoplastic process involves the CNS, symptoms can include reduced level of consciousness, seizures, impaired memory or cognitive problems, personality change (ie, limbic encephalitis), ataxia, or even focal signs such as aphasia. CNS involvement may occur early or late; it often is responsible for profound morbidity and death.

One of the minor symptoms seen in LEMS is an unpleasant metallic taste. In the setting of weakness this symptom is suggestive of LEMS, but it may not be reported by the patient and often it needs to be solicited.

A patient may manifest a paraneoplastic syndrome with any combination of autonomic, peripheral, or CNS involvement.[15]

As with any paraneoplastic neurological degeneration, autonomic dysfunction has been described in many types of cancer. These include small-cell lung cancer and other lung tumors, thymoma, and ovarian and breast carcinoma. In some cases, paraneoplastic autonomic dysfunction occurs in the apparent absence of cancer. In patients without known cancer, any clinical history suggesting an underlying tumor (e.g., unexplained weight loss) or high risk for particular cancers (eg, heavy smoking, personal or family history of cancer) can help suggest a link between autonomic symptoms and an underlying malignancy. Finding the primary tumor can prove very difficult in some patients.

The clinical course is usually subacutely progressive over weeks to months, leading to a bedridden condition in severe cases if untreated, and often in spite of treatment.

Physical

Physical findings in patients with paraneoplastic autonomic failure resemble those of any patient with autonomic dysfunction, and include the following:

Peripheral sensory neuronopathy often is evident as patchy superficial sensory loss and asymmetrically abnormal stretch reflexes.

Patchy asymmetric weakness and dyscoordination, or abnormal mental status, may occur in patients with CNS involvement.

Proximal muscle weakness is seen in LEMS.

It is important to note that some physical signs may be due directly to the oncological process or from treatment of the malignancy. For instance, chemotherapy with vincristine typically causes areflexia that is diffuse and symmetric. Cisplatin can cause a sensory neuropathy and hearing loss, both of which are typically symmetric. Carcinomatous meningitis can closely mimic the presentation of paraneoplastic encephalomyeloneuropathy.

Causes

Paraneoplastic autonomic dysfunction is a secondary effect of cancer. Small-cell lung cancer is particularly likely to cause paraneoplastic syndromes, but many types of malignancy can cause these types of syndromes, including teratoma, neuroblastoma, retinoblastoma, oligodendroglioma, melanoma, leiomyosarcoma, hematological tumors, and cancers of the thymus, gastrointestinal tract, rectum, prostate, breast, fallopian tubes, uterus, bladder, kidney, pancreas, testicles, malignancies and ovary.[9, 15, 41, 22, 16, 29]

Laboratory Studies

Serum analysis for the presence of onconeural autoantibodies using immunohistochemistry and immunoblotting is the key to diagnosis. Since there is an overlap between the clinical picture of individual paraneoplastic autoantibodies, screening for them using a panel of autoantibodies is appropriate. Lumbar puncture is often indicated, with ample volume of cerebrospinal fluid (CSF) sent for cytologic analysis in addition to the other usual tests. The spinal fluid often reveals a mononuclear pleocytosis, elevated protein, oligoclonal bands, and paraneoplastic onconeural antibodies.

Evidence of another process (eg, diabetes, scleroderma, amyloidosis, thyroid disease) should be considered and excluded by appropriate serologic tests and/or biopsies.

Serological markers of malignancies (eg, CEA, PSA, NSE, CA19-9, AFP, β-HCG, and ProGRP, if available) can help to identify a cancer that is not readily apparent by imaging.

Imaging Studies

If the patient is not known to harbor a malignancy, then chest imaging (eg, high resolution CT scan) is indicated, since small-cell lung cancer is most often the associated tumor. If any abnormalities are seen, then bronchoscopic or transthoracic needle biopsy is usually required. If no tumor is found, fluorodeoxyglucose-positron emission tomography (FDG-PET) should be considered as it markedly increases the detection rate of malignancy.[42] Imaging of the abdomen, pelvis, and brain is normally indicated to look for a primary tumor or metastasis. Mammography and gynecological ultrasonographic examinations can be of value in women, and testicular ultrasonography in men. If no malignancy is found at presentation, repeating the work-up every 3-6 months is advisable[41] for the first 4 years (or the first 2 years in the case of LEMS).[42]

Other Tests

Electromyography (EMG)/nerve conduction velocity (NCV) studies can identify a neuropathy or defect of neuromuscular transmission if present. Heart rate variability and autonomic skin responses can be performed in most laboratories. More extensive autonomic testing may be helpful to prove autonomic dysfunction in milder cases, although it is usually not necessary.

Histologic Findings

The images below show typical histologic findings from a patient with small-cell lung cancer and autoimmune paraneoplastic autonomic failure. Loss of neurons is evident in autonomic ganglia and dorsal root ganglia, as well as within the CNS. Inflammatory infiltrates are also typical in the vicinity of neuron loss. Similar findings have been reported for autoimmune-mediated paraneoplastic autonomic dysfunction caused by other cancers.



View Image

Paraneoplastic autonomic neuropathy. Histopathology of peripheral nerve and sympathetic ganglion from a patient with autonomic failure, oat-cell carci....



View Image

Paraneoplastic autonomic neuropathy. Hematoxylin and eosin (H&E)–stained sections from dorsal root ganglion showing the hallmark histopathology of ant....



View Image

Paraneoplastic autonomic neuropathy. Central nervous system sections from a patient with autonomic failure, oat-cell carcinoma of the lung, and positi....

The underlying tumor is often heavily infiltrated by inflammatory cells.

Medical Care

Care of patients with paraneoplastic autonomic neuropathy depends on severity of autonomic dysfunction and status of associated malignancy.

Surgical Care

Temporary pacing of unstable cardiac rhythms has been found to be helpful in certain cases of NMDA receptor antibody encephalitis.[23, 24]

Consultations

If no known underlying cancer is present, consider seeking consultation from a pulmonary medicine specialist. A pulmonologist can help in finding an obscure small-cell lung cancer or other lung tumor. A medical oncology specialist can be of diagnostic help, in addition to assisting in the treatment of the associated malignancy.

Gastroenterology and urology consultation may be needed if intestinal dysmotility and urinary incontinence are part of the clinical manifestations.

Diet

High fluid and salt intake is of potential value for orthostatic hypotension. A fiber-rich diet can help some gastric dysmotility symptoms.

Medication Summary

No specific drug treatment is of proven value; however, cytotoxic chemotherapy for the associated malignancy is rational. For symptomatic management of autonomic failure, see article on Idiopathic Orthostatic Hypotension and Other Autonomic Failure Syndromes for details.

In the case of LEMS, drugs that increase available acetylcholine (such as 3,4-diaminopyridine, which increases acetylcholine release, or cholinesterase inhibitors like pyridostigmine that prevent acetylcholine destruction) have been helpful for symptom control.[13, 16]

Further Outpatient Care

Custodial nursing home care may be needed if the autonomic failure is severe, and patients are bedridden and completely disabled.

Further Inpatient Care

Typically, the disorder is progressive, although stabilization with antineoplastic therapy has been reported. Subsequent follow-up is devoted to assessing adequacy of blood pressure support and bowel and bladder management.

Inpatient & Outpatient Medications

Medications are the same as those used in inpatient care, except doses are adjusted with time and disease progression. See article Idiopathic Orthostatic Hypotension and Other Autonomic Failure Syndromes for specific suggestions.

Deterrence/Prevention

No known method is effective in deterring or preventing occurrence of autonomic paraneoplastic failure, except to prevent exposure to known carcinogens such as tobacco smoke.

Complications

Sudden death, often due to cardiac causes, can occur. Intestinal pseudo-obstruction and urinary tract infections from incomplete bladder emptying are possible. Overheating due to reduced sweat function can occur in hot temperatures or with physical exertion.

Prognosis

Prognosis is generally poor. Prompt and effective treatment of the underlying malignancy may arrest progression of autonomic dysfunction, but in many cases, no improvement occurs. Survival is dependent on underlying cancer, patient age, and extent of nervous system involvement. The median survival of all patients with paraneoplastic syndromes has been estimated to be 1 to 3 years.[7] The two major exceptions are LEMS and NMDA receptor antibody disease. In Lambert-Eaton myasthenic syndrome, effective treatment often results in remission of the mild autonomic symptoms and better than expected survival.[45, 46, 47] In NMDA receptor antibody disease, treatment results in recovery 75% of the time; however, this disease is still poorly characterized and likely underdiagnosed, so these numbers may change as more data become available.[22]

What is autoimmune paraneoplastic autonomic neuropathy?What are the diagnostic criteria for paraneoplastic neurological syndromes (PNSs)?What is the pathophysiology of paraneoplastic neurological syndromes (PNSs)?What is the role of anti-ganglionic acetylcholine receptor antibodies in the pathophysiology of paraneoplastic neurological syndromes (PNSs)?What is the role of anti-Hu antibodies in the pathophysiology of paraneoplastic neurological syndromes (PNSs)?What is the role of anti-NMDA receptor antibodies in the pathophysiology of paraneoplastic neurological syndromes (PNSs)?What is the role of anti-voltage-gated calcium channels (VGCC) antibodies in the pathophysiology of paraneoplastic neurological syndromes (PNSs)?What is the role of paraneoplastic antibodies in the pathophysiology of paraneoplastic neurological syndromes (PNSs)?What is the prevalence of paraneoplastic autonomic neuropathy in the US?What is the global prevalence of paraneoplastic autonomic neuropathy?What is the morbidity and mortality associated with paraneoplastic autonomic neuropathy?What are the racial predilections of paraneoplastic autonomic neuropathy?What are the sexual predilections of paraneoplastic autonomic neuropathy?Which age group has the highest prevalence of paraneoplastic autonomic neuropathy?Which clinical history findings are characteristic of paraneoplastic autonomic neuropathy?Which physical findings are characteristic of paraneoplastic autonomic neuropathy?What causes paraneoplastic autonomic neuropathy?What are the differential diagnoses for Paraneoplastic Autonomic Neuropathy?What is the role of lab testing in the workup of paraneoplastic autonomic neuropathy?What is the role of imaging studies in the workup of paraneoplastic autonomic neuropathy?What is the role of EMG/NCV studies in the workup of paraneoplastic autonomic neuropathy?Which histologic findings are characteristic of paraneoplastic autonomic neuropathy?How is malignancy treated in patients with paraneoplastic autonomic neuropathy?How are autoimmune processes treated in patients with paraneoplastic autonomic neuropathy?How is autonomic failure treated in patients with paraneoplastic autonomic neuropathy?How is paraneoplastic autonomic neuropathy treated?What is the role of surgery in the treatment of paraneoplastic autonomic neuropathy?Which specialist consultations are beneficial to patients with paraneoplastic autonomic neuropathy?Which dietary modifications are used in the treatment of paraneoplastic autonomic neuropathy?What is the role of medications in the treatment of paraneoplastic autonomic neuropathy?What is included in the long-term monitoring of paraneoplastic autonomic neuropathy?When is inpatient care indicated for the treatment of paraneoplastic autonomic neuropathy?How is paraneoplastic autonomic neuropathy prevented?What are the possible complications of paraneoplastic autonomic neuropathy?What is the prognosis of paraneoplastic autonomic neuropathy?

Author

Stacy E Dixon, MD, PhD, Assistant Professor of Neurology, University of Colorado School of Medicine

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Biogen; Genetech.

Coauthor(s)

Dianna Quan, MD, Professor of Neurology, Director of Electromyography Laboratory, University of Colorado School of Medicine

Disclosure: Received research grant from: Alnylam; Pfizer; Cytokinetics; Momenta.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Jorge C Kattah, MD, Head, Associate Program Director, Professor, Department of Neurology, University of Illinois College of Medicine at Peoria

Disclosure: Nothing to disclose.

Chief Editor

Tarakad S Ramachandran, MBBS, MBA, MPH, FAAN, FACP, FAHA, FRCP, FRCPC, FRS, LRCP, MRCP, MRCS, Professor Emeritus of Neurology and Psychiatry, Clinical Professor of Medicine, Clinical Professor of Family Medicine, Clinical Professor of Neurosurgery, State University of New York Upstate Medical University; Neuroscience Director, Department of Neurology, Crouse Irving Memorial Hospital

Disclosure: Nothing to disclose.

Additional Contributors

Bjorn E Oskarsson, MD, Assistant Professor, Department of Neurology, University of California, Davis, School of Medicine

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Flex Pharma<br/>Serve(d) as a speaker or a member of a speakers bureau for: Grifols<br/>Received research grant from: Neuraltus, Glaxo, Eisai, Cytokinetics, Genentech,.

Daniel Mordechai Goldenholz, MD, PhD, Resident Physician, Department of Neurology, University of California Davis Medical Center

Disclosure: Nothing to disclose.

Ronald G Wiley, MD, PhD, Professor of Neurology and Pharmacology, Vanderbilt University, Chief of the Neurology Service, Veterans Affairs Tennessee Valley Healthcare System

Disclosure: Nothing to disclose.

References

  1. Graus F, Delattre JY, Antoine JC, et al. Recommended diagnostic criteria for paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry. 2004 Aug. 75(8):1135-40. [View Abstract]
  2. Anderson NE, Rosenblum MK, Graus F. Autoantibodies in paraneoplastic syndromes associated with small-cell lung cancer. Neurology. 1988 Sep. 38(9):1391-8. [View Abstract]
  3. Elrington GM, Murray NM, Spiro SG. Neurological paraneoplastic syndromes in patients with small cell lung cancer. A prospective survey of 150 patients. J Neurol Neurosurg Psychiatry. 1991 Sep. 54(9):764-7. [View Abstract]
  4. Veilleux M, Bernier JP, Lamarche JB. Paraneoplastic encephalomyelitis and subacute dysautonomia due to an occult atypical carcinoid tumour of the lung. Can J Neurol Sci. 1990 Aug. 17(3):324-8. [View Abstract]
  5. Blaes F, Strittmatter M, Schwamborn J, et al. Antineuronal antibody-associated paraneoplastic neuropathy in Hodgkin's disease. Eur J Neurol. 1998 Jan. 5(1):109-112. [View Abstract]
  6. Ficker DM, Hammack JE. Primary central nervous system lymphoma presenting as autonomic dysfunction. J Neurooncol. 1995. 23(3):245-8. [View Abstract]
  7. Candler PM, Hart PE, Barnett M, et al. A follow up study of patients with paraneoplastic neurological disease in the United Kingdom. J Neurol Neurosurg Psychiatry. 2004 Oct. 75(10):1411-5. [View Abstract]
  8. Altermatt HJ, Rodriguez M, Scheithauer BW, Lennon VA. Paraneoplastic anti-Purkinje and type I anti-neuronal nuclear autoantibodies bind selectively to central, peripheral, and autonomic nervous system cells. Lab Invest. 1991 Oct. 65(4):412-20. [View Abstract]
  9. Pittock SJ, Kryzer TJ, Lennon VA. Paraneoplastic antibodies coexist and predict cancer, not neurological syndrome. Ann Neurol. 2004 Nov. 56(5):715-9. [View Abstract]
  10. Panegyres PK, Reading MC, Esiri MM. The inflammatory reaction of paraneoplastic ganglionitis and encephalitis: an immunohistochemical study. J Neurol. 1993 Feb. 240(2):93-7. [View Abstract]
  11. Vernino S, Adamski J, Kryzer TJ. Neuronal nicotinic ACh receptor antibody in subacute autonomic neuropathy and cancer-related syndromes. Neurology. 1998 Jun. 50(6):1806-13. [View Abstract]
  12. Vernino S, Low PA, Fealey RD. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med. 2000 Sep 21. 343(12):847-55. [View Abstract]
  13. Freeman R. Autonomic peripheral neuropathy. Lancet. 2005 Apr 2-8. 365(9466):1259-70. [View Abstract]
  14. Peltier AC, Black BK, Raj SR, Donofrio P, Robertson D, Biaggioni I. Coexistent autoimmune autonomic ganglionopathy and myasthenia gravis associated with non-small-cell lung cancer. Muscle Nerve. 2010 Mar. 41(3):416-9. [View Abstract]
  15. Lorusso L, Hart IK, Ferrari D, Ngonga GK, Gasparetto C, Ricevuti G. Autonomic paraneoplastic neurological syndromes. Autoimmun Rev. 2007 Jan. 6(3):162-8. [View Abstract]
  16. de Beukelaar JW, Sillevis Smitt PA. Managing paraneoplastic neurological disorders. Oncologist. 2006 Mar. 11(3):292-305. [View Abstract]
  17. Lucchinetti CF, Kimmel DW, Lennon VA. Paraneoplastic and oncologic profiles of patients seropositive for type 1 antineuronal nuclear autoantibodies. Neurology. 1998 Mar. 50(3):652-7. [View Abstract]
  18. Camdessanche JP, Antoine JC, Honnorat J. Paraneoplastic peripheral neuropathy associated with anti-Hu antibodies. A clinical and electrophysiological study of 20 patients. Brain. 2002 Jan. 125(Pt 1):166-75. [View Abstract]
  19. Vernino S. Antibody testing as a diagnostic tool in autonomic disorders. Clin Auton Res. 2009 Feb. 19(1):13-9. [View Abstract]
  20. Granerod J, Ambrose HE, Davies NW, Clewley JP, Walsh AL, Morgan D. Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study. Lancet Infect Dis. 2010 Dec. 10(12):835-44. [View Abstract]
  21. Lancaster E, Martinez-Hernandez E, Dalmau J. Encephalitis and antibodies to synaptic and neuronal cell surface proteins. Neurology. 2011 Jul 12. 77(2):179-89. [View Abstract]
  22. Dalmau J, Gleichman AJ, Hughes EG, Rossi JE, Peng X, Lai M. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol. 2008 Dec. 7(12):1091-8. [View Abstract]
  23. Nazif TM, Vázquez J, Honig LS, Dizon JM. Anti-N-methyl-D-aspartate receptor encephalitis: an emerging cause of centrally mediated sinus node dysfunction. Europace. 2012 Feb 15. [View Abstract]
  24. Millichap JJ, Goldstein JL, Laux LC, Nordli DR Jr, Stack CV, Wainwright MS. Ictal asystole and anti-N-methyl-D-aspartate receptor antibody encephalitis. Pediatrics. 2011 Mar. 127(3):e781-6. [View Abstract]
  25. Mahadeva B, Phillips LH 2nd, Juel VC. Autoimmune disorders of neuromuscular transmission. Semin Neurol. 2008 Apr. 28(2):212-27. [View Abstract]
  26. Vernino S, Lennon VA. New Purkinje cell antibody (PCA-2): marker of lung cancer-related neurological autoimmunity. Ann Neurol. 2000 Mar. 47(3):297-305. [View Abstract]
  27. Tschernatsch M, Klotz M, Probst C, Hosch J, Valtorta F, Diener M, et al. Synaptophysin is an autoantigen in paraneoplastic neuropathy. J Neuroimmunol. 2008 Jun 15. 197(1):81-6. [View Abstract]
  28. Tan KM, Lennon VA, Klein CJ, Boeve BF, Pittock SJ. Clinical spectrum of voltage-gated potassium channel autoimmunity. Neurology. 2008 May 13. 70(20):1883-90. [View Abstract]
  29. Tan KM, Lennon VA, Klein CJ, Boeve BF, Pittock SJ. Clinical spectrum of voltage-gated potassium channel autoimmunity. Neurology. 2008 May 13. 70(20):1883-90. [View Abstract]
  30. Vincent A, Bien CG, Irani SR, Waters P. Autoantibodies associated with diseases of the CNS: new developments and future challenges. Lancet Neurol. 2011 Aug. 10(8):759-72. [View Abstract]
  31. Dhamija R, Tan KM, Pittock SJ, Foxx-Orenstein A, Benarroch E, Lennon VA. Serologic profiles aiding the diagnosis of autoimmune gastrointestinal dysmotility. Clin Gastroenterol Hepatol. 2008 Sep. 6(9):988-92. [View Abstract]
  32. Malhotra K, Georgiades IB, Davies J. Functional colonic obstruction in an adult as a presenting feature of thymoma. Ann R Coll Surg Engl. 2011 Sep. 93(6):e61-3. [View Abstract]
  33. Duddy ME, Baker MR. Stiff person syndrome. Front Neurol Neurosci. 2009. 26:147-65. [View Abstract]
  34. Graus F, Saiz A, Dalmau J. Antibodies and neuronal autoimmune disorders of the CNS. J Neurol. 2010 Apr. 257(4):509-17. [View Abstract]
  35. Tschernatsch M, Klotz M, Probst C, Hosch J, Valtorta F, Diener M. Synaptophysin is an autoantigen in paraneoplastic neuropathy. J Neuroimmunol. 2008 Jun 15. 197(1):81-6. [View Abstract]
  36. Sven Jarius, Marius Ringelstein, Jurgen Hass, Irina I. Serysheva, Lars Komorowski, Kai Fechner, et al. Inositol 1,4,5-trisphosphate receptor type 1 autoantibodies in paraneoplastic and non-paraneoplastic peripheral neuropathy. Journal of Neuroinflammation. Oct 24, 2016. 13:278. [View Abstract]
  37. William Oliver Tobin, Vanda A. Lennon, Lars Komorowski, Christian Probst, Stacey Lynn Clardy, Allen J. Aksamit, et al. DPPX potassium channel antibody; frequency, clinical accompaniments, and outcomes in 20 patients. Neurology. Nov. 11, 2014. 83:1797-1803. [View Abstract]
  38. Byrne T, Mason WP, Posner JB, et al. Spontaneous neurological improvement in anti-Hu associated encephalomyelitis. J Neurol Neurosurg Psychiatry. 1997 Mar. 62(3):276-8. [View Abstract]
  39. Chalk CH, Windebank AJ, Kimmel DW, McManis PG. The distinctive clinical features of paraneoplastic sensory neuronopathy. Can J Neurol Sci. 1992 Aug. 19(3):346-51. [View Abstract]
  40. Chinn JS, Schuffler MD. Paraneoplastic visceral neuropathy as a cause of severe gastrointestinal motor dysfunction. Gastroenterology. 1988 Nov. 95(5):1279-86. [View Abstract]
  41. Dalmau J, Graus F, Rosenblum MK. Anti-Hu--associated paraneoplastic encephalomyelitis/sensory neuronopathy. A clinical study of 71 patients. Medicine (Baltimore). 1992 Mar. 71(2):59-72. [View Abstract]
  42. Titulaer MJ, Soffietti R, Dalmau J, Gilhus NE, Giometto B, Graus F, et al. Screening for tumours in paraneoplastic syndromes: report of an EFNS Task Force. Eur J Neurol. 2010 Sep 29. [View Abstract]
  43. Oh SJ, Dropcho EJ, Claussen GC. Anti-Hu-associated paraneoplastic sensory neuropathy responding to early aggressive immunotherapy: report of two cases and review of literature. Muscle Nerve. 1997 Dec. 20(12):1576-82. [View Abstract]
  44. Dhamija R, Tan KM, Pittock SJ, Foxx-Orenstein A, Benarroch E, Lennon VA. Serologic profiles aiding the diagnosis of autoimmune gastrointestinal dysmotility. Clin Gastroenterol Hepatol. 2008 Sep. 6(9):988-92. [View Abstract]
  45. Khurana RK, Koski CL, Mayer RF. Autonomic dysfunction in Lambert-Eaton myasthenic syndrome. J Neurol Sci. 1988 May. 85(1):77-86. [View Abstract]
  46. Maddison P, Lang B. Paraneoplastic neurological autoimmunity and survival in small-cell lung cancer. J Neuroimmunol. 2008 Sep 15. 201-202:159-62. [View Abstract]
  47. Mamdani MB, Walsh RL, Rubino FA. Autonomic dysfunction and Eaton Lambert syndrome. J Auton Nerv Syst. 1985 Apr. 12(4):315-20. [View Abstract]
  48. Block MS, Vassallo R. Lack of FDG uptake in small cell carcinoma associated with ANNA-1 positive paraneoplastic autonomic neuropathy. J Thorac Oncol. 2008 May. 3(5):542-4. [View Abstract]

Paraneoplastic autonomic neuropathy. Central nervous system sections from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive titer of anti-HU antibodies stained with hematoxylin and eosin (H&E). (a) Inferior olive showing a cluster of mononuclear cells (arrowhead); (b) hippocampus showing perivascular mononuclear infiltrate (arrowhead); (c) midbrain section showing a vessel encased in a mononuclear infiltrate; (d) ventral horn of the thoracic spinal cord showing clusters of mononuclear cells around degenerating motor neurons (arrowheads). Magnification bars indicate 100 mm. The bar in b applies also to a and the bar in d also applies to c.

Paraneoplastic autonomic neuropathy. Histopathology of peripheral nerve and sympathetic ganglion from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive anti-HU antibody titer. (a) Peripheral nerve in longitudinal section stained with Luxol fast blue-periodic acid-Schiff (PAS) showing scattered wallerian degeneration (arrowheads). (b) Low-power view of a paravertebral sympathetic ganglion stained with hematoxylin and eosin (H&E). Arrowhead indicates perivascular mononuclear infiltrates. (c) High-power view of the same sympathetic ganglion showing degenerating neurons (single arrowheads) and mononuclear infiltrates (double arrowhead). Magnification bars in a and b indicate 100 mm; c is 50 mm.

Paraneoplastic autonomic neuropathy. Hematoxylin and eosin (H&E)–stained sections from dorsal root ganglion showing the hallmark histopathology of anti-HU disease; a and c are from a healthy patient; b and d are from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive titer of anti-HU antibodies. The arrowheads in b and d indicate degenerating sensory neurons. Also note the interstitial hypercellularity and decreased numbers of neurons in b and d. Magnification bar in b indicates 100 mm and applies also to a. Similarly, the magnification bar in d indicates 50 mm, which also applies to c.

Paraneoplastic autonomic neuropathy. Central nervous system sections from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive titer of anti-HU antibodies stained with hematoxylin and eosin (H&E). (a) Inferior olive showing a cluster of mononuclear cells (arrowhead); (b) hippocampus showing perivascular mononuclear infiltrate (arrowhead); (c) midbrain section showing a vessel encased in a mononuclear infiltrate; (d) ventral horn of the thoracic spinal cord showing clusters of mononuclear cells around degenerating motor neurons (arrowheads). Magnification bars indicate 100 mm. The bar in b applies also to a and the bar in d also applies to c.

Paraneoplastic autonomic neuropathy. Histopathology of peripheral nerve and sympathetic ganglion from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive anti-HU antibody titer. (a) Peripheral nerve in longitudinal section stained with Luxol fast blue-periodic acid-Schiff (PAS) showing scattered wallerian degeneration (arrowheads). (b) Low-power view of a paravertebral sympathetic ganglion stained with hematoxylin and eosin (H&E). Arrowhead indicates perivascular mononuclear infiltrates. (c) High-power view of the same sympathetic ganglion showing degenerating neurons (single arrowheads) and mononuclear infiltrates (double arrowhead). Magnification bars in a and b indicate 100 mm; c is 50 mm.

Paraneoplastic autonomic neuropathy. Hematoxylin and eosin (H&E)–stained sections from dorsal root ganglion showing the hallmark histopathology of anti-HU disease; a and c are from a healthy patient; b and d are from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive titer of anti-HU antibodies. The arrowheads in b and d indicate degenerating sensory neurons. Also note the interstitial hypercellularity and decreased numbers of neurons in b and d. Magnification bar in b indicates 100 mm and applies also to a. Similarly, the magnification bar in d indicates 50 mm, which also applies to c.

Paraneoplastic autonomic neuropathy. Central nervous system sections from a patient with autonomic failure, oat-cell carcinoma of the lung, and positive titer of anti-HU antibodies stained with hematoxylin and eosin (H&E). (a) Inferior olive showing a cluster of mononuclear cells (arrowhead); (b) hippocampus showing perivascular mononuclear infiltrate (arrowhead); (c) midbrain section showing a vessel encased in a mononuclear infiltrate; (d) ventral horn of the thoracic spinal cord showing clusters of mononuclear cells around degenerating motor neurons (arrowheads). Magnification bars indicate 100 mm. The bar in b applies also to a and the bar in d also applies to c.