Abstract

Paraneoplastic neurological syndrome (PNS) comprises a group of neurological disorders that result from a misguided immune response to the nervous system triggered by a distant tumor. These disorders frequently manifest before the diagnosis of the underlying neoplasm. Since the first reported case in 1888 by Oppenheim, the knowledge in this area has evolved rapidly. Several classic PNS have been described, such as limbic encephalitis, paraneoplastic cerebellar degeneration, encephalomyelitis, opsoclonus-myoclonus, sensory neuronopathy, Lambert-Eaton Myasthenic syndrome, and chronic gastrointestinal dysmotility. It is now recognized that PNS can have varied nonclassical manifestations that extend beyond the traditional syndromic descriptions. Multiple onconeural antibodies with high specificity for certain tumor types and neurological phenotypes have been discovered over the past 3 decades. Increasing use of immune checkpoint inhibitors (ICIs) has led to increased recognition of neurologic ICI-related adverse events. Some of these resemble PNS. In this article, we review the clinical, oncologic, and immunopathogenic associations of PNS.

Paraneoplastic neurological syndromes (PNS) comprise a group of neurological disorders that result from a misguided immune response to the nervous system triggered by a distant tumor.1,2 In 1888, Oppenheim reported empiric association of peripheral neuropathy and lung cancer highlighting the remote or indirect effect of cancer on the nervous system.3 This was followed by another description of association between cancer and neuropathy in 1890 by Auchè.4 Subsequently, Dr. Derek Denny-Brown, described 2 cases of sensory neuronopathy with myopathy (neuromyopathy) associated with bronchogenic pulmonary carcinoma.5 At that time, it was presumed this phenomenon may be related to a metabolic disorder associated with tumor. Later in the 1960s, Dr. Wilkinson and Dr. Zeromski investigated sera from patients with neuromyopathy associated bronchial carcinoma and found circulating antibodies against brain extracts6 as well as central nervous system neurons.7 In the 1980s, this perinuclear neuronal staining pattern was further characterized as anti-Hu (a.k.a. anti-neuronal nuclear antibody type 1 [ANNA1]) and details about neurological syndromes and oncologic associations were published by researchers from Sloan Kettering Cancer Center.8–10 In 1976, Dr. Trotter and colleagues identified an antibody against cerebellar Purkinje cells in serum of a patient with Hodgkin’s disease.11 Subsequently, anti-cerebellar Purkinje cells antibody was described in two patients with ovarian carcinoma and paraneoplastic cerebellar degeneration (PCD) by Dr. Greenlee and Dr. Brashear,12 and this was confirmed and further characterized by independent groups.13–16 Furthermore, in 1999, investigators from Sloan Kettering Cancer Center described the first serological biomarker of paraneoplastic encephalitis associated with testicular germ cell tumor, anti-Ma2-IgG.17 During this time, there were significant contributions from Mayo Clinic Neuroimmunology group, including discovery of P/Q-type voltage-gated calcium channel (VGCC) antibodies among patients with Lambert-Eaton Myasthenic syndrome. Another onconeural antibody anti-CV2 IgGwas initially described by Honnorat et al. in 1999,18 and was confirmed to be collapsin response-mediated protein 5 (CRMP5) in 2001.19 These early contributions and biomarker discoveries laid the foundation for the field of paraneoplastic neurological disorders. In the last two decades, many novel biomarkers have been discovered (Figure 1) and varied neurological syndromes have been characterized in association with paraneoplastic autoimmunity.

Autoantibody discovery timeline. Antibodies with high neoplastic association are in yellow. Abbreviations: AGNA/SOX1, anti-glial nuclear antibody against SRY (sex determining region Y)-box 1; AK5, adenylate kinase 5; AMPAR, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; ANNA1 (anti-Hu), anti-neuronal nuclear antibody type 1; ANNA2 (anti-Ri), anti-neuronal nuclear antibody type 2; ANNA3, anti-neuronal nuclear antibody type 3; AP3B2, adapter protein 3, subunit B2; AQP4, aquaporin4; CASPR2, contactin-associated protein-like 2; CRMP5, collapsinresponse mediator protein 5; DPPX, dipeptidyl peptidase-like protein 6; GABAAR, gamma-aminobutyric acid type A receptor; GABABR, gamma-aminobutyric acid type B receptor; GAD65, glutamic acid decarboxylase 65; GFAP, glial fibrillary acidic protein; GlyR, glycine receptor; GRAF, GTPase regulator associated with focal adhesion kinase; IgLON5, immunoglobulin-like cell adhesion molecule 5; ITPR1, inositol 1,4,5-trisphophate receptor type 1; KLHL11, kelch-like protein 11; LGI-1, leucine-rich, glioma-inactivated 1; mAChR, muscarinic acetylcholine receptor; mGluR1, metabotropic glutamate receptor 1; mGluR5, metabotropic glutamate receptor 5; MuSK, muscle-specific tyrosine kinase; nAChR, nicotinic acetylcholine receptor; NfL, neurofilament; NMDA-R, N-methyl-d-aspartate receptor; PCA1 (anti-Yo) , Purkinje cell cytoplasmic antibody type 1; PCA2, Purkinje cell cytoplasmic antibody type 2; PCA-Tr/DNER, Purkinje cell cytoplasmic antibody type Tr or Delta/Notch-like epidermal growth factor-related receptor; PDE10A, phosphodiesterase 10A; Trim9/67, tripartite motif-containing protein 9 and 67; VGCC, voltage-gated calcium channel; VGKC, voltage-gated potassium channel. *Amphiphysin and septin-5 antibodies can have transient cell-surface expression.
Figure 1.

Autoantibody discovery timeline. Antibodies with high neoplastic association are in yellow. Abbreviations: AGNA/SOX1, anti-glial nuclear antibody against SRY (sex determining region Y)-box 1; AK5, adenylate kinase 5; AMPAR, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; ANNA1 (anti-Hu), anti-neuronal nuclear antibody type 1; ANNA2 (anti-Ri), anti-neuronal nuclear antibody type 2; ANNA3, anti-neuronal nuclear antibody type 3; AP3B2, adapter protein 3, subunit B2; AQP4, aquaporin4; CASPR2, contactin-associated protein-like 2; CRMP5, collapsinresponse mediator protein 5; DPPX, dipeptidyl peptidase-like protein 6; GABAAR, gamma-aminobutyric acid type A receptor; GABABR, gamma-aminobutyric acid type B receptor; GAD65, glutamic acid decarboxylase 65; GFAP, glial fibrillary acidic protein; GlyR, glycine receptor; GRAF, GTPase regulator associated with focal adhesion kinase; IgLON5, immunoglobulin-like cell adhesion molecule 5; ITPR1, inositol 1,4,5-trisphophate receptor type 1; KLHL11, kelch-like protein 11; LGI-1, leucine-rich, glioma-inactivated 1; mAChR, muscarinic acetylcholine receptor; mGluR1, metabotropic glutamate receptor 1; mGluR5, metabotropic glutamate receptor 5; MuSK, muscle-specific tyrosine kinase; nAChR, nicotinic acetylcholine receptor; NfL, neurofilament; NMDA-R, N-methyl-d-aspartate receptor; PCA1 (anti-Yo) , Purkinje cell cytoplasmic antibody type 1; PCA2, Purkinje cell cytoplasmic antibody type 2; PCA-Tr/DNER, Purkinje cell cytoplasmic antibody type Tr or Delta/Notch-like epidermal growth factor-related receptor; PDE10A, phosphodiesterase 10A; Trim9/67, tripartite motif-containing protein 9 and 67; VGCC, voltage-gated calcium channel; VGKC, voltage-gated potassium channel. *Amphiphysin and septin-5 antibodies can have transient cell-surface expression.

A diagnostic criterion of PNS was recommended by a panel of neurologist with expertise in the diagnosis and management of these disorders in 2004 (Euronetwork PNS criteria).20 Based on these criteria, a definite PNS diagnosis required a classical syndrome, and identification of neoplasm within 5 years of PNS onset and/or the presence of well-characterized onconeural antibodies. For a patient presenting with a nonclassical syndrome the diagnosis of a definite PNS could be made if a well-characterized onconeural antibody was detected, or an onconeural antibody not considered well characterized was detected and cancer developed within 5 years of PNS onset, or PNS significantly improved after cancer treatment without concomitant immunotherapy. A recent population-based Italian study utilized these Euronetwork PNS diagnostic criteria and reported incidence of PNS was 0.89 per 100,000 person-years with a prevalence of 4.37 per 100,000.21

Herein, we review the pathogenesis, clinical syndromes, antibody and oncological associations, and management of PNS. Furthermore, we discuss the neurological complications of immune checkpoint inhibitors (ICIs) and their management.

Pathophysiology of Paraneoplastic Syndrome

The cellular or humoral immune response against antigens shared by the tumor and neural tissues is hypothesized to contribute to PNS pathogenesis. The majority of well-characterized onconeural antibodies target intracellular antigens. These neural antibodies targeting intracellular antigens are generally considered to be surrogate markers for a cytotoxic T-cell response rather than being directly pathogenic.22 On the other hand, the onconeural antibodies targeting cell-surface antigens are potentially directly pathogenic.

Upon cell death, cell-surface and intracellular proteins are phagocytosed and processed by the dendritic cells (DCs).23 Subsequently, the antigens are presented to the lymphocytes in the regional lymph nodes. Intracellular antigens lead to cell-mediated adaptive immune response. The antigens presented by the DCs activate autoantigen-specific CD8+ cytotoxic T-lymphocytes.24 The activated neural antigen-specific CD8+ T-lymphocytes are hypothesized to recognize and bind to the neural cells and, release proinflammatory cytokines and cytotoxic granules leading to cell death.25,26 Some studies have also demonstrated that antibodies targeting intracellular antigens such as ANNA1 (anti-Hu) or Purkinje cell cytoplasmic antibody type 1 (PCA1, a.k.a. anti-Yo) IgG can be taken by neurons and may be directly responsible for neuronal damage.27–30 However, direct neuronal cytotoxic effect of antibodies targeting intracellular antigens has not been consistently demonstrated in all in vitro studies.31,32 Intraventricular administration of anti-Hu antibodies in animal models did not lead to development of PNS.33 Immunohistopathological studies of patients with PNS have also demonstrated CD3+ and CD8+ T-lymphocyte predominant infiltrates in brain or dorsal root ganglia which supports cytotoxic T-cell-mediated neuronal loss, rather than antibody-mediated dysfunction.24,34–37

In contrast, the immune response against cell-surface protein is predominantly humoral- or B-cell-mediated. The complex interaction between B- and CD4+ T-lymphocytes leads to B-cell proliferation and differentiation to plasma cells and memory B cells. In some instances, B-lymphocytes can enter CNS where they undergo re-stimulation, maturation, and differentiation to plasma cells leading to intrathecal antibody synthesis.38,39 Unlike studies of intracellular antigen, various in vitro studies suggested that antibodies against cell-surface antigen can bind to cell-surface proteins on live neurons,38,40,41 and postmortem tissue examination demonstrated anti-IgG deposition around the neurons without substantial cell death.42 In animal studies, injection of N-methyl-d-aspartic acid receptor (NMDA-R) IgG, an antibody against cell-surface protein, led to alteration of glutamatergic transmission and enhance cortical excitability in Wistar rats.43,44 These findings demonstrated the pathogenic roles of antibodies against cell-surface protein.

Central Nervous System Syndromes

Paraneoplastic Limbic Encephalitis

Limbic system is involved in a variety of functions including memory, emotional behavior, mood processing, and olfaction. It consists of the hippocampus, amygdala, cingulate gyrus, hypothalamus, and the limbic cortex. Inflammation of these structures termed limbic encephalitis may result from autoimmune, inflammatory, or infectious etiologies. Paraneoplastic limbic encephalitis generally has a subacute onset. Common presenting features include anterograde amnesia, behavioral disturbances, and at times seizures.45 Cerebrospinal fluid (CSF) lymphocytic pleocytosis is common among patients with paraneoplastic limbic encephalitis.45 The MRI hallmark of this syndrome is increased T2 signal in medial temporal lobes involving the hippocampus and amygdala.46

Among patients presenting with limbic encephalitis, underlying onconeural antibody specificities may guide targeted neoplasm search. Neuropsychiatric dysfunction, sleep disorder and seizures, and metabotropic glutamate receptor 5 (mGluR5) antibody seropositivity are suggestive of underlying Hodgkin’s lymphoma.47 This syndrome was initially described by Ian Carr in his 15-year-old daughter in 1982.48 He named it “Ophelia syndrome” after a character from Shakespeare’s play “Hamlet.” Similarly, limbic encephalitis in association with Ma2-IgG or kelch-like protein 11 (KLHL11) IgG is suggestive of testicular or extra-testicular germ cell tumor.17,24,49,50 Various paraneoplastic antibodies associated with limbic encephalitis are listed in Table 1. Early diagnosis, treatment of the underlying tumor, and early initiation of immunosuppressive therapy have been associated with favorable outcomes.45,51

Table 1.

Paraneoplastic Neurological Syndromes, Clinical Features, and Associated Antibodies

Paraneoplastic Syndromes Clinical FeatureAssociated Antibodies
Limbic encephalitis Memory impairment, seizures, behavioral change, neuropsychiatric symptoms, mood and sleep disturbancesANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), mGluR5, ANNA2 (anti-Ri), PCA2, KLHL11, GABAB receptor, AMPAR, LGI1, CASPR2, ANNA3
EncephalomyelitisEncephalopathy, myelitis, and peripheral nerve or DRG involvementANNA1 (anti-Hu), CRMP5 (anti-CV2), amphiphysin, GFAP
Cerebellar degenerationAtaxia (usually subacute onset), dysarthria, diplopiaPCA1 (anti-Yo), PCA2, PCA-Tr (DNER), mGluR1, ANNA1,2, Zic 4, VGCC (P/Q type), amphiphysin, Ma2, NfL, KLHL11
Sensory neuronopathyAsymmetric numbness, paresthesia, sensory ataxia, neuropathic painANNA1 (anti-Hu), amphiphysin, PCA2, CRMP5 (CV2)
Opsoclonus myoclonus syndromeMyoclonus and opsoclonus eye movementsANNA2 (anti-Ri), ANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), NMDA-R, GluD2
Gastrointestinal dysmotility Gastroparesis, constipation, pseudo-obstructionANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2
Lambert-Eaton myasthenic syndromeMuscle weakness, fatigue, and autonomic dysfunction and absent deep tendon reflexesP/Q-type VGCC
Brainstem encephalitisAtaxia, vertigo, diplopia, dysphagia, dysarthriaANNA2 (anti-Ri), KLHL11, ANNA1 (anti-Hu), Ma2, PCA2, CRMP5 (CV2), amphiphysin, Zic 4
Paraneoplastic myelopathySubacute myelopathy, usually tract-specific involvementANNA2 (anti-Ri), ANNA1 (anti-Hu), ANNA3, CRMP5 (anti-CV2), Amphiphysin, NfL
Paraneoplastic stiff person syndromeMuscle spasms and exaggerated startle responseAmphiphysin, DPPX, GAD65, glycine receptor
Paraneoplastic choreaChoreic movements with more extensive nervous system involvement CRMP5 (anti-CV2), PDE10A, ANNA1
PoyradiculoneuropathyPainful asymmetric weakness and numbness, neuropathic painCRMP5 (anti-CV2), ANNA1 (anti-Hu), PCA2, and amphiphysin
Pan dysautonomia Orthostatic hypotension, dry mouth, incontinence, gastroparesis, and cardiac arrhythmiasANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2, ganglionic AChR antibodies
Peripheral nerve hyperexcitabilityCramps, myokymia, fasciculationCASPR2, LGI1, CRMP5 (anti-CV2), Nectrin-1
Myasthenia gravisOculobulbar weakness, dysarthria, dysphagia, fatigable proximal limb weaknessAChR binding antibody
Immune-mediated necrotizing myopathyProximal weakness with rare respiratory or cardiac involvementHMGCR, SRP54
Dermatomyositis Proximal weakness, skin changesSAE1, TIF1-γ, NXP2
Paraneoplastic Syndromes Clinical FeatureAssociated Antibodies
Limbic encephalitis Memory impairment, seizures, behavioral change, neuropsychiatric symptoms, mood and sleep disturbancesANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), mGluR5, ANNA2 (anti-Ri), PCA2, KLHL11, GABAB receptor, AMPAR, LGI1, CASPR2, ANNA3
EncephalomyelitisEncephalopathy, myelitis, and peripheral nerve or DRG involvementANNA1 (anti-Hu), CRMP5 (anti-CV2), amphiphysin, GFAP
Cerebellar degenerationAtaxia (usually subacute onset), dysarthria, diplopiaPCA1 (anti-Yo), PCA2, PCA-Tr (DNER), mGluR1, ANNA1,2, Zic 4, VGCC (P/Q type), amphiphysin, Ma2, NfL, KLHL11
Sensory neuronopathyAsymmetric numbness, paresthesia, sensory ataxia, neuropathic painANNA1 (anti-Hu), amphiphysin, PCA2, CRMP5 (CV2)
Opsoclonus myoclonus syndromeMyoclonus and opsoclonus eye movementsANNA2 (anti-Ri), ANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), NMDA-R, GluD2
Gastrointestinal dysmotility Gastroparesis, constipation, pseudo-obstructionANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2
Lambert-Eaton myasthenic syndromeMuscle weakness, fatigue, and autonomic dysfunction and absent deep tendon reflexesP/Q-type VGCC
Brainstem encephalitisAtaxia, vertigo, diplopia, dysphagia, dysarthriaANNA2 (anti-Ri), KLHL11, ANNA1 (anti-Hu), Ma2, PCA2, CRMP5 (CV2), amphiphysin, Zic 4
Paraneoplastic myelopathySubacute myelopathy, usually tract-specific involvementANNA2 (anti-Ri), ANNA1 (anti-Hu), ANNA3, CRMP5 (anti-CV2), Amphiphysin, NfL
Paraneoplastic stiff person syndromeMuscle spasms and exaggerated startle responseAmphiphysin, DPPX, GAD65, glycine receptor
Paraneoplastic choreaChoreic movements with more extensive nervous system involvement CRMP5 (anti-CV2), PDE10A, ANNA1
PoyradiculoneuropathyPainful asymmetric weakness and numbness, neuropathic painCRMP5 (anti-CV2), ANNA1 (anti-Hu), PCA2, and amphiphysin
Pan dysautonomia Orthostatic hypotension, dry mouth, incontinence, gastroparesis, and cardiac arrhythmiasANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2, ganglionic AChR antibodies
Peripheral nerve hyperexcitabilityCramps, myokymia, fasciculationCASPR2, LGI1, CRMP5 (anti-CV2), Nectrin-1
Myasthenia gravisOculobulbar weakness, dysarthria, dysphagia, fatigable proximal limb weaknessAChR binding antibody
Immune-mediated necrotizing myopathyProximal weakness with rare respiratory or cardiac involvementHMGCR, SRP54
Dermatomyositis Proximal weakness, skin changesSAE1, TIF1-γ, NXP2

Abbreviations: AMPAR, α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor; AChR, acetylcholine receptor; ANNA, anti-neuronal nuclear antibody; AP3B2, adaptor protein 3, subunit B2; CASPR2, contactin-associated protein 2; CLL, chronic lymphoid leukemia; CRMP5, collapsin response-mediated protein 5; DRG, dorsal root ganglia; DPPX, dipeptidyl peptidase-like protein-6; GABA, gamma-aminobutyric acid; GAD65, glutamic acid decarboxylase, 65 kDa isoform; GFAP, glial fibrillary acidic protein; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; IgG, immunoglobulin; GluD2, glutamate receptor δ2; KLHL11, kelch-like protein 11; LGI1, leucine-rich glioma-inactivated 1; mGluR, metabotropic glutamate receptor; NfL, neuronal intermediate filament light chain; NMDA-R, N-methyl-d-aspartic acid receptor; POEMS, polyneuropathy organomegaly endocrinopathy M-protein and skin changes; PCA, Purkinje cell cytoplasmic antibody; SCLC, small cell lung carcinoma; VGCC, voltage-gated calcium channel; SRP 54, signal recognition particle 54; SAE1, small ubiquitin-like modifier activating enzyme heterodimer 1; TIF1-γ, transcription intermediary factor 1-γ; NXP2, nuclear matrix protein 2.

Table 1.

Paraneoplastic Neurological Syndromes, Clinical Features, and Associated Antibodies

Paraneoplastic Syndromes Clinical FeatureAssociated Antibodies
Limbic encephalitis Memory impairment, seizures, behavioral change, neuropsychiatric symptoms, mood and sleep disturbancesANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), mGluR5, ANNA2 (anti-Ri), PCA2, KLHL11, GABAB receptor, AMPAR, LGI1, CASPR2, ANNA3
EncephalomyelitisEncephalopathy, myelitis, and peripheral nerve or DRG involvementANNA1 (anti-Hu), CRMP5 (anti-CV2), amphiphysin, GFAP
Cerebellar degenerationAtaxia (usually subacute onset), dysarthria, diplopiaPCA1 (anti-Yo), PCA2, PCA-Tr (DNER), mGluR1, ANNA1,2, Zic 4, VGCC (P/Q type), amphiphysin, Ma2, NfL, KLHL11
Sensory neuronopathyAsymmetric numbness, paresthesia, sensory ataxia, neuropathic painANNA1 (anti-Hu), amphiphysin, PCA2, CRMP5 (CV2)
Opsoclonus myoclonus syndromeMyoclonus and opsoclonus eye movementsANNA2 (anti-Ri), ANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), NMDA-R, GluD2
Gastrointestinal dysmotility Gastroparesis, constipation, pseudo-obstructionANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2
Lambert-Eaton myasthenic syndromeMuscle weakness, fatigue, and autonomic dysfunction and absent deep tendon reflexesP/Q-type VGCC
Brainstem encephalitisAtaxia, vertigo, diplopia, dysphagia, dysarthriaANNA2 (anti-Ri), KLHL11, ANNA1 (anti-Hu), Ma2, PCA2, CRMP5 (CV2), amphiphysin, Zic 4
Paraneoplastic myelopathySubacute myelopathy, usually tract-specific involvementANNA2 (anti-Ri), ANNA1 (anti-Hu), ANNA3, CRMP5 (anti-CV2), Amphiphysin, NfL
Paraneoplastic stiff person syndromeMuscle spasms and exaggerated startle responseAmphiphysin, DPPX, GAD65, glycine receptor
Paraneoplastic choreaChoreic movements with more extensive nervous system involvement CRMP5 (anti-CV2), PDE10A, ANNA1
PoyradiculoneuropathyPainful asymmetric weakness and numbness, neuropathic painCRMP5 (anti-CV2), ANNA1 (anti-Hu), PCA2, and amphiphysin
Pan dysautonomia Orthostatic hypotension, dry mouth, incontinence, gastroparesis, and cardiac arrhythmiasANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2, ganglionic AChR antibodies
Peripheral nerve hyperexcitabilityCramps, myokymia, fasciculationCASPR2, LGI1, CRMP5 (anti-CV2), Nectrin-1
Myasthenia gravisOculobulbar weakness, dysarthria, dysphagia, fatigable proximal limb weaknessAChR binding antibody
Immune-mediated necrotizing myopathyProximal weakness with rare respiratory or cardiac involvementHMGCR, SRP54
Dermatomyositis Proximal weakness, skin changesSAE1, TIF1-γ, NXP2
Paraneoplastic Syndromes Clinical FeatureAssociated Antibodies
Limbic encephalitis Memory impairment, seizures, behavioral change, neuropsychiatric symptoms, mood and sleep disturbancesANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), mGluR5, ANNA2 (anti-Ri), PCA2, KLHL11, GABAB receptor, AMPAR, LGI1, CASPR2, ANNA3
EncephalomyelitisEncephalopathy, myelitis, and peripheral nerve or DRG involvementANNA1 (anti-Hu), CRMP5 (anti-CV2), amphiphysin, GFAP
Cerebellar degenerationAtaxia (usually subacute onset), dysarthria, diplopiaPCA1 (anti-Yo), PCA2, PCA-Tr (DNER), mGluR1, ANNA1,2, Zic 4, VGCC (P/Q type), amphiphysin, Ma2, NfL, KLHL11
Sensory neuronopathyAsymmetric numbness, paresthesia, sensory ataxia, neuropathic painANNA1 (anti-Hu), amphiphysin, PCA2, CRMP5 (CV2)
Opsoclonus myoclonus syndromeMyoclonus and opsoclonus eye movementsANNA2 (anti-Ri), ANNA1 (anti-Hu), Ma2, CRMP5 (anti-CV2), NMDA-R, GluD2
Gastrointestinal dysmotility Gastroparesis, constipation, pseudo-obstructionANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2
Lambert-Eaton myasthenic syndromeMuscle weakness, fatigue, and autonomic dysfunction and absent deep tendon reflexesP/Q-type VGCC
Brainstem encephalitisAtaxia, vertigo, diplopia, dysphagia, dysarthriaANNA2 (anti-Ri), KLHL11, ANNA1 (anti-Hu), Ma2, PCA2, CRMP5 (CV2), amphiphysin, Zic 4
Paraneoplastic myelopathySubacute myelopathy, usually tract-specific involvementANNA2 (anti-Ri), ANNA1 (anti-Hu), ANNA3, CRMP5 (anti-CV2), Amphiphysin, NfL
Paraneoplastic stiff person syndromeMuscle spasms and exaggerated startle responseAmphiphysin, DPPX, GAD65, glycine receptor
Paraneoplastic choreaChoreic movements with more extensive nervous system involvement CRMP5 (anti-CV2), PDE10A, ANNA1
PoyradiculoneuropathyPainful asymmetric weakness and numbness, neuropathic painCRMP5 (anti-CV2), ANNA1 (anti-Hu), PCA2, and amphiphysin
Pan dysautonomia Orthostatic hypotension, dry mouth, incontinence, gastroparesis, and cardiac arrhythmiasANNA1 (anti-Hu), CRMP5 (anti-CV2), PCA2, ganglionic AChR antibodies
Peripheral nerve hyperexcitabilityCramps, myokymia, fasciculationCASPR2, LGI1, CRMP5 (anti-CV2), Nectrin-1
Myasthenia gravisOculobulbar weakness, dysarthria, dysphagia, fatigable proximal limb weaknessAChR binding antibody
Immune-mediated necrotizing myopathyProximal weakness with rare respiratory or cardiac involvementHMGCR, SRP54
Dermatomyositis Proximal weakness, skin changesSAE1, TIF1-γ, NXP2

Abbreviations: AMPAR, α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor; AChR, acetylcholine receptor; ANNA, anti-neuronal nuclear antibody; AP3B2, adaptor protein 3, subunit B2; CASPR2, contactin-associated protein 2; CLL, chronic lymphoid leukemia; CRMP5, collapsin response-mediated protein 5; DRG, dorsal root ganglia; DPPX, dipeptidyl peptidase-like protein-6; GABA, gamma-aminobutyric acid; GAD65, glutamic acid decarboxylase, 65 kDa isoform; GFAP, glial fibrillary acidic protein; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; IgG, immunoglobulin; GluD2, glutamate receptor δ2; KLHL11, kelch-like protein 11; LGI1, leucine-rich glioma-inactivated 1; mGluR, metabotropic glutamate receptor; NfL, neuronal intermediate filament light chain; NMDA-R, N-methyl-d-aspartic acid receptor; POEMS, polyneuropathy organomegaly endocrinopathy M-protein and skin changes; PCA, Purkinje cell cytoplasmic antibody; SCLC, small cell lung carcinoma; VGCC, voltage-gated calcium channel; SRP 54, signal recognition particle 54; SAE1, small ubiquitin-like modifier activating enzyme heterodimer 1; TIF1-γ, transcription intermediary factor 1-γ; NXP2, nuclear matrix protein 2.

Paraneoplastic Encephalomyelitis

Paraneoplastic encephalomyelitis is a multifocal neurological syndrome, with multi-level involvement of neuroaxis.20 These patients usually have an involvement of peripheral nerves or myenteric plexus, suggesting encephalomyelitis is not exclusively limited to the central nervous system. Furthermore, multifocal neurological syndromes without encephalitis, such as paraneoplastic myeloneuropathy or paraneoplastic subacute combined degeneration have also been described.52 Patients presenting with a clinical syndrome predominantly involving one part of neuroaxis, should be described according to the focal syndrome rather than “encephalomyelitis.” Onconeural antibody biomarkers associated with encephalomyelitis include ANNA1 (anti-Hu), ANNA2 (ant-Ri), amphiphysin, and CRMP5 (CV2) (Table 2).2,10,53–57 This is a refractory syndrome with relatively poor long-term prognosis. In a series of 200 ANNA1 (anti-Hu)-IgG-associated encephalomyelitis cases, more than 50% of cases were severely disabled at the time of diagnosis.58 Higher mortality among these cases is associated with worse functional status at the time of diagnosis, involvement of more areas of the nervous system, age >60 years, and lack of treatment.58 Early treatment of underlying malignancy was found to be an independent predictor of at least stabilization of the neurological disability for 6 months or more. Because of the presumed role of cellular immunity and reported refractoriness to first-line immunotherapies, more aggressive immunosuppression in the form of early initiation of second-line agents such as cyclophosphamide should be considered.

Table 2.

Antibodies With High Cancer Association, Associated Antigens, and Oncological Associations

AntibodiesAssociated AntigensOncological Associations
ANNA-1 (anti-Hu)nELAVL (Hu)SCLC, neuroblastoma in children
ANNA-2 (anti-Ri)NOVA 1, 2 (Ri)Lung and breast adenocarcinoma
ANNA-3UnknownSCLC
AGNA-1 SOX1SCLC
PCA-1 (anti-Yo)CDR2 or CDR2LOvarian, fallopian, endometrial, and breast adenocarcinoma
PCA-2MAP1BSCLC
PCA-TrDNERHodgkin lymphoma
AmphiphysinAmphiphysinBreast adenocarcinoma, SCLC
CRMP5 (anti-CV2)CRMP5SCLC, thymoma
Ma2PNMA2Testicular germ cell tumor, non-small cell lung cancer
KLHL11-IgGKLHL11Testicular germ cell tumor
mGluR5mGluR5Hodgkin lymphoma
PDE10APDE10AAdenocarcinomas (lung, renal and pancreatic)
NfLNeuronal intermediate filament light chainNeuroendocrine tumor
AntibodiesAssociated AntigensOncological Associations
ANNA-1 (anti-Hu)nELAVL (Hu)SCLC, neuroblastoma in children
ANNA-2 (anti-Ri)NOVA 1, 2 (Ri)Lung and breast adenocarcinoma
ANNA-3UnknownSCLC
AGNA-1 SOX1SCLC
PCA-1 (anti-Yo)CDR2 or CDR2LOvarian, fallopian, endometrial, and breast adenocarcinoma
PCA-2MAP1BSCLC
PCA-TrDNERHodgkin lymphoma
AmphiphysinAmphiphysinBreast adenocarcinoma, SCLC
CRMP5 (anti-CV2)CRMP5SCLC, thymoma
Ma2PNMA2Testicular germ cell tumor, non-small cell lung cancer
KLHL11-IgGKLHL11Testicular germ cell tumor
mGluR5mGluR5Hodgkin lymphoma
PDE10APDE10AAdenocarcinomas (lung, renal and pancreatic)
NfLNeuronal intermediate filament light chainNeuroendocrine tumor

Abbreviations: AGNA, anti-glial nuclear antibody; ANNA, anti-neuronal nuclear antibody; CLL, chronic lymphoid leukemia; CRMP5, collapsin response-mediated protein 5; DNER, Delta/Notch-like growth factor-related receptor; nELAVL, neuronal embryonic lethal abnormal vision-like; GABA, gamma-aminobutyric acid; IgG, immunoglobulin; KLHL11, kelch-like protein 11; mGluR, metabotropic glutamate receptor; PCA, Purkinje cell cytoplasmic antibody; PDE10A, phosphodiesterase 10A; PERM, progressive encephalomyelitis with rigidity and myoclonus; SCLC, small cell lung carcinoma; SOX1; SRY-box transcription factor 1.

Table 2.

Antibodies With High Cancer Association, Associated Antigens, and Oncological Associations

AntibodiesAssociated AntigensOncological Associations
ANNA-1 (anti-Hu)nELAVL (Hu)SCLC, neuroblastoma in children
ANNA-2 (anti-Ri)NOVA 1, 2 (Ri)Lung and breast adenocarcinoma
ANNA-3UnknownSCLC
AGNA-1 SOX1SCLC
PCA-1 (anti-Yo)CDR2 or CDR2LOvarian, fallopian, endometrial, and breast adenocarcinoma
PCA-2MAP1BSCLC
PCA-TrDNERHodgkin lymphoma
AmphiphysinAmphiphysinBreast adenocarcinoma, SCLC
CRMP5 (anti-CV2)CRMP5SCLC, thymoma
Ma2PNMA2Testicular germ cell tumor, non-small cell lung cancer
KLHL11-IgGKLHL11Testicular germ cell tumor
mGluR5mGluR5Hodgkin lymphoma
PDE10APDE10AAdenocarcinomas (lung, renal and pancreatic)
NfLNeuronal intermediate filament light chainNeuroendocrine tumor
AntibodiesAssociated AntigensOncological Associations
ANNA-1 (anti-Hu)nELAVL (Hu)SCLC, neuroblastoma in children
ANNA-2 (anti-Ri)NOVA 1, 2 (Ri)Lung and breast adenocarcinoma
ANNA-3UnknownSCLC
AGNA-1 SOX1SCLC
PCA-1 (anti-Yo)CDR2 or CDR2LOvarian, fallopian, endometrial, and breast adenocarcinoma
PCA-2MAP1BSCLC
PCA-TrDNERHodgkin lymphoma
AmphiphysinAmphiphysinBreast adenocarcinoma, SCLC
CRMP5 (anti-CV2)CRMP5SCLC, thymoma
Ma2PNMA2Testicular germ cell tumor, non-small cell lung cancer
KLHL11-IgGKLHL11Testicular germ cell tumor
mGluR5mGluR5Hodgkin lymphoma
PDE10APDE10AAdenocarcinomas (lung, renal and pancreatic)
NfLNeuronal intermediate filament light chainNeuroendocrine tumor

Abbreviations: AGNA, anti-glial nuclear antibody; ANNA, anti-neuronal nuclear antibody; CLL, chronic lymphoid leukemia; CRMP5, collapsin response-mediated protein 5; DNER, Delta/Notch-like growth factor-related receptor; nELAVL, neuronal embryonic lethal abnormal vision-like; GABA, gamma-aminobutyric acid; IgG, immunoglobulin; KLHL11, kelch-like protein 11; mGluR, metabotropic glutamate receptor; PCA, Purkinje cell cytoplasmic antibody; PDE10A, phosphodiesterase 10A; PERM, progressive encephalomyelitis with rigidity and myoclonus; SCLC, small cell lung carcinoma; SOX1; SRY-box transcription factor 1.

Paraneoplastic Cerebellar Degeneration

PCD usually presents with subacute progressive cerebellar ataxia.59 MRI brain may be normal in the early course, but most cases have cerebellar atrophy later in the disease. Onconeural antibodies typically associated with PCD include PCA1 (anti-Yo), Purkinje cell cytoplasmic antibody type Tr (PCA-Tr), and mGlur-1. PCA-Tr targets Delta/Notch-like epidermal growth factor-related receptor (DNER), a transmembrane protein that is expressed in the dendrites of Purkinje cells. The mGlur-1 receptor is also located in dendrites and plays a role in rapid calcium signaling.60 Whereas, PCA1 (anti-Yo) antibody targets intracellular CDR2 and/or CDR2-L protein.1,61,62 The majority of PCD associated with PCA1 (anti-Yo) have relatively poor outcomes. Interestingly, some studies have reported better prognosis of PCA1 (anti-Yo) patients with breast cancer compared to those with gynecologic cancer.63

Paraneoplastic Brainstem Encephalitis

Paraneoplastic brainstem encephalitis manifests as diplopia, ataxia, vertigo, cranial neuropathy, dysarthria, dysphagia, and at times impaired level of consciousness. A majority of paraneoplastic brainstem encephalitis cases have inflammatory CSF. MRI brain in early disease course may be unremarkable or may show T2 hyper-intensity. However, as disease progresses patients’ brainstem atrophy becomes more apparent.

Onconeural antibodies associated with brainstem encephalitis are ANNA2 (anti-Ri), KLHL11, Ma2, CRMP5 (CV2), ANNA1 (anti-Hu), and amphiphysin. Laryngospasm and/or jaw dystonia are relatively distinguishable neurological presentations reported in 25% of ANNA2 (anti-Ri) paraneoplastic brainstem encephalitis cases.64 Whereas sensory neural hearing loss and tinnitus are early presenting features of KLHL11-IgG-associated neurological syndrome.50

A considerable proportion of patients with brainstem encephalitis continue to have progressive decline, despite treatment of underlying cancer and aggressive immunotherapy. However, among both Ma2 and KLHL11-IgG seropositive cases detection and treatment of testicular germ cell tumor has been associated with favorable clinical prognosis.49,65

Paraneoplastic Opsoclonus-Myoclonus Syndrome

Opsoclonus-myoclonus syndrome (OMS) also known as “dancing eye syndrome,” consists of spontaneous, arrhythmic, conjugate high-frequency oscillations of the eyes with horizontal, vertical, and torsional saccades that are associated with myoclonus of the head, trunk, or extremities.66 Opsoclonus is diagnosed clinically based on back-to-back multidirectional conjugate saccades without an inter-saccadic interval. In children, OMS age of onset ranges from 12 to 36 months.66,67 Approximately, 50% of pediatric OMS cases are associated with neuroblastoma.66 Glutamate receptor δ2 IgG has been identified as a serological biomarker in a subset of pediatric OMS cases.68 Among adult patients, paraneoplastic OMS has been associated with small cell lung carcinoma, breast adenocarcinoma, ovarian teratoma, and testicular germ cell tumor.69,70 Onconeural antibodies associated with adult paraneoplastic OMS include ANNA2 (anti-Ri), ANNA1 (anti-Hu), Ma2, CRMP5 (CV2), and NMDA-R antibodies.70 Treatment includes identification and management underlying of neoplasm, and early initiation of immunotherapy. Symptomatic treatments include clonazepam, levetiracetam, baclofen, and gabapentin.69 In comparison with idiopathic OMS, paraneoplastic OMS has a more refractory course. However, complete or partial neurological recovery after removal of underlying tumor has been reported.71

Paraneoplastic Myelopathies

Most of the cases have subacute or insidious onset myelopathic signs and symptoms, with a progressive course.72,73 MRI spine usually demonstrates longitudinally extensive (>3 vertebral segments) tract or gray matter-specific T2 hyperintense signal abnormalities.72,73 Lateral and dorsal columns and central gray matter are commonly involved. Gadolinium enhancement is seen in approximately 50% of the cases.72 The majority of cases have lymphocyte-predominant pleocytosis and elevated protein in the CSF. Supernumerary oligoclonal band have been reported in 30%-75% of patients.72,73 Onconeural antibodies associated with paraneoplastic myelopathies are amphiphysin, CRMP5 (CV2), ANNA1 (anti-Hu), ANNA2 (anti-Ri), ANNA3, and PCA2.72–74 Furthermore, paraneoplastic AQP4-IgG myelitis in association with non-small cell lung and breast cancers have also been reported.75

Paraneoplastic myelopathies are usually associated with poor outcome. The majority (52%) of cases reported in a large series from our institution required gait aid for ambulation.72 The median time from symptom onset to wheelchair dependence was 9 months (range 1-21 months).

Paraneoplastic Stiff Person Syndrome

“Stiff man” syndrome was first described in a series of male patients in 1956.76 With description of female cases the disease nomenclature was changed to gender-neutral “Stiff person syndrome” (SPS).77 SPS is characterized by fluctuating muscle stiffness, superimposed spasms, and exaggerated startle. The muscle spasms principally involve the trunk and lower limbs, typically symmetric, more proximal in distribution, and associated with an increased sensitivity to external stimuli.78 SPS spectrum disorders (SPSD) comprise of classic SPS, focal or segmental SPS such as stiff limb syndrome, progressive encephalomyelitis with rigidity and myoclonus (PERM), and overlapping syndromes.79 Glutamic acid decarboxylase (GAD) 65 IgG, glycine receptor (GlyR) IgG, amphiphysin IgG, and dipeptidyl peptidase-like protein 6 (DPPX) IgG are antibodies associated with SPSD.80,81 GAD65 and glycine receptor antibodies are usually associated with non-paraneoplastic SPSD. A minority of DPPX-IgG cases have underlying hematological malignancies. Amphiphysin IgG has a strong paraneoplastic association with breast cancer and small cell lung cancer.82,83 Treatment of paraneoplastic SPS is focused on symptomatic treatment, immunotherapy, and treatment of underlying tumor.84 Symptomatic treatment includes use of muscle relaxants such as benzodiazepines, baclofen, tizanidine, and botulinum toxin injections to help with the muscle spasms.78,84

Peripheral Nervous System Syndromes

Paraneoplastic Sensory Neuronopathy

Sensory neuronopathy is caused by damage to the neurons in dorsal root ganglia.85 Patients usually present with burning pain and numbness in upper and/or lower limbs, usually in an asymmetric pattern. Symptoms progress rapidly over weeks. Involvement of the face, abdomen, or trunk often provides a diagnostic clue. On examination, there is a loss of sensation to all modalities with marked impairment of proprioception, sensory ataxia, and deep tendon reflexes are diminished or absent. The electrophysiological evaluation shows absent or reduced sensory responses globally with normal to minimal abnormalities of the motor responses.86 CSF analysis commonly demonstrates elevated protein, pleocytosis, and the presence of oligoclonal bands. Small cell lung cancer is the most common cancer associated with this phenotype. Common onconeural antibody associations include ANNA1 (anti-Hu), amphiphysin, CRMP5 (CV2), and PCA2 antibodies.87 Other common cancer associations include thymoma and breast cancer.58 Even though majority of cases have a refractory course, combined therapeutic approach focused management of underlying cancer and early immunosuppressive therapy initiation have demonstrated most significant clinical benefit.88

Paraneoplastic Polyradiculoneuropathy

Polyradiculoneuropathy is characterized by clinical and electrophysiological involvement of nerve roots and proximal nerves.89 In some cases, MRI spine and/or lumbar plexus demonstrates T2 hyper-intensity and/or post-contrast gadolinium enhancement of nerve roots and plexus. CRMP5 (CV2), PCA2, ANNA1 (anti-Hu), and amphiphysin are the onconeural antibodies commonly associated with paraneoplastic polyradiculoneuropathy.89–91 CRMP5 CV2 IgG-associated neuropathy comprises a distinct clinical phenotype of subacute painful asymmetric polyradiculoneuropathy (both proximal and distal limb weakness and sensory involvement) with perivascular inflammation on nerve biopsy. It is commonly associated with small cell lung cancer and thymoma.89 Neuropathy has been described as the most frequent neurological presentation with the PCA2-IgG seropositivity. The majority of PCA2 neuropathy cases had polyradiculoneuropathy phenotype followed by sensory neuronopathy.91 A considerable proportion of amphiphysin IgG seropositive patients with breast cancer also present with symmetric axonal polyradiculoneuropathy with or without co-existing SPS.90

Autonomic Neuropathy

Autonomic neuropathy as a paraneoplastic syndrome can present in isolation or may co-exist with other central/peripheral nervous system manifestations. Chronic gastrointestinal dysmotility syndrome and acute pandysautonomia are well-characterized paraneoplastic phenotypes.92 Orthostatic hypotension, dry mouth, incontinence, and cardiac arrhythmias are other presenting symptoms/signs. Small cell lung cancer and ANNA1 (anti-Hu) and/or CRMP5 (CV2) antibodies are common oncological and serological associations, respectively. Autoimmune autonomic ganglionopathy associated with ganglionic AChR antibodies has also been rarely described in association with cancer.93,94

Peripheral Nerve Hyperexcitability Syndromes

Autoimmune peripheral nerve hyperexcitability syndromes can be of paraneoplastic origin, the two most commonly described phenotypes are Isaac syndrome and Morvan’s syndrome. Isaac syndrome is clinically characterized by generalized muscle stiffness (present even during sleep) and cramps, myokymia, fasciculation, and often associated with autonomic dysfunction-like sialorrhea and hyperhidrosis.95 Electromyography (EMG) characteristically shows myokymia and/or neuromyotonia. Patients with Morvan’s syndrome have CNS involvement (encephalopathy, sleep dysfunction) in addition to features of peripheral nerve hyperexcitability. Contactin-associated protein-like 2 (CASPR2) and/or Leucine-rich glioma-inactivated 1 (LGI1) IgG are neural-specific antibodies associated with this phenotype, while thymoma is the commonly associated tumor. Antibodies against Netrin-1 receptor have also been described in association with neuromyotonia and myasthenia gravis (MG) in patients with thymoma.96

Neuromuscular Junction Disorders

MG is an autoimmune disorder with antibodies against acetylcholine receptors (AChR) and other related proteins, attacking the post-synaptic membrane in the neuromuscular junction.97 Patients usually present with predominant oculobulbar weakness in the form of double vision, droopy eyelids, and often swallowing difficulty along with fatigable proximal limb weakness. In about 10%-15% of cases, MG occurs as a paraneoplastic phenomenon in association with thymoma and rarely lung cancer.97,98

Lambert-Eaton myasthenic syndrome is another rare neuromuscular junction disorder associated with presynaptic membrane P/Q-type VGCC with very distinctive clinical and electrophysiological features.99,100 Muscle weakness, fatigue, and autonomic dysfunction and absent deep tendon reflexes are the most common findings. More than 50% of cases are associated with cancer, most commonly small cell lung cancer. Diagnosis requires a high level of awareness and can initiate prompt screening for cancer.101

Paraneoplastic Myopathies

Immune-mediated necrotizing myopathy (IMNM) is a relatively rare autoimmune myopathy often associated with signal recognition particle 54 (SRP54) and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) autoantibodies. Patients usually present with acute or subacute onset progressive proximal weakness with rare respiratory or cardiac involvement. Histopathologically, there tends to be minimal or no inflammation on muscle biopsy as compared to other immune myopathies.102 While statin use is the most-recognized risk factor for HMGCR-IgG-mediated syndrome, a minority of IMNM also have paraneoplastic associations.103 Malignancy tends to occur more frequently in seronegative cases.104

Dermatomyositis is another neurological presentation historically considered to have a paraneoplastic association.105 Certain myositis antibodies like anti-small ubiquitin-like modifier activating enzyme heterodimer 1 (anti-SAE1), anti-transcription intermediary factor 1-γ (anti-TIF1-γ), and anti-nuclear matrix protein 2 (anti-NXP2) antibodies are associated with an increased risk of cancer.106 Cancer is usually recognized within 3 years of myositis diagnosis, with most diagnosed within 12 months.

Neurological Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitors

In current clinical practice, three classes of ICIs are being utilized including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death receptor 1 (PD-1), and programmed cell death ligand 1 (PDL-1) inhibitors.107 ICIs are being utilized for a variety of cancer indications including melanoma, non-small cell lung carcinoma, renal cell carcinoma, Hodgkin’s lymphoma, urothelial cancer, Merkel cell carcinoma, and extensive-stage small cell lung cancer.108

ICIs have been associated with a variety of immune-related adverse events (irAEs). All organ systems can be affected. The commonly involved organ systems are the lungs, gastrointestinal tract, liver, endocrine, and kidney. The involvement of the nervous system is less common.109 The neurologic iRAEs may resemble PNS phenotype, or preexisting PNS may be worsened by ICI administration. However, neurologic irAEs (even those resembling classic PNS phenotype) differ from PNS, as the former is caused by drug-induced disruption of immunologic self-tolerance and/or potentiation of autoimmunity. The median onset of neurologic irAEs is typically 2-16 weeks after the ICI initiation.110 The risk of irAEs appears to 3-fold higher during the first 4 weeks after ICI treatment compared to later timepoints.111

Pathophysiology

Physiologically, CTLA-4, PD-1, and PDL-1 are the immune checkpoint proteins that inhibit autoreactive T-lymphocytes preventing the occurrence of autoimmune disorder. In many cancers, the tumor cells enhance this inhibitory signal to escape the anti-tumor immune response. Therefore, blocking these molecules reactivates T-lymphocytes to destroy tumor cells. However, inhibition of the immune checkpoint proteins may lead to the development of immune-mediated complications as well as potentiation of the paraneoplastic disorders.112,113

The majority of the n-irAEs are presumed to be secondary to three potential mechanisms: (1) immune response against common autoantigens expressed by the cancer and neural tissue, (2) exacerbation of preexisting autoimmunity, and (3) direct toxic effect of therapeutic monoclonal antibody.114

Cancer antigen-driven paraneoplastic mechanism is supported by a PCD mouse model of subcutaneously implanted breast cancer treated CTLA-4 inhibitor.115 Additionally, autopsies of patients who developed fatal myocarditis after receiving CTLA-4 and PD-1 inhibitors have demonstrated robust T-cell and macrophage infiltration in myocardium, and selective clonal T-cell population infiltrating myocardium have been found to be identical to those present in tumor cells.116

Reports of MS exacerbation following administration of ICIs are good examples of n-irAEs secondary to preexisting autoimmunity.117

ICI-related hypophysitis in association with CTLA-4 inhibitors is a good example of potential role of these monoclonal antibodies in directly causing irAEs.114 However, expression of these checkpoint proteins does not seem to be restricted to pituitary gland and their expression in other neural tissues have also been described.

Central Nervous System irAEs

Various CNS disorders have been reported in association with ICIs, including aseptic meningitis, encephalitis, myelitis, cerebellar ataxia, sarcoidosis, and Vogt-Konyagi-Harada-like syndrome.2,113,118–120 Moreover, exacerbation as well as de novo multiple sclerosis have also been reported in association with ICI use, particularly ipilimumab (CTLA-4 inhibitor).121,122 CNS irAEs resembling classic PNS phenotypes include limbic encephalitis,123–125 cerebellar ataxia,126 OMS,127 and encephalomyelitis.128 Limbic encephalitis is one of the most common n-irAEs resembling PNS phenotype.2 The onconeural antibodies which were reported in association with ICI use include autoantibodies against ANNA1 (anti-Hu), Ma2, SOX1, and CRMP5 (CV2).2,112,118

Peripheral Nervous System irAEs

ICIs are also associated with peripheral nervous system involvements including myopathies, neuromuscular junction disorders as well as neuropathies.118,129 IMNM, and myositis have been reported in association with CTLA-4, PD-1, PDL-1 inhibitors or combination therapies.129 Of note, a considerable proportion of ICI-related myopathies (irMyopathies) may have concomitant myocarditis.129 Patients with ICI-related MG (irMG) may have co-existing features of myositis on histopathological assessments.118 Lambert-Eaton myasthenic syndrome in association with P/Q-type VGCC antibodies has also been reported among patients with squamous cell and small cell lung cancer.130,131

A wide range of neuropathies have been reported as a complication of ICI, including Guillain-Barre syndrome, sensory ganglionopathy, Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), sensorimotor polyneuropathy, axonal polyradiculoneuropathy, mononeuritis multiplex, cranial neuropathy, and phrenic nerve palsy.118,129 Elevated CSF protein has been reported in most of the cases, and more than 50% of cases have CSF pleocytosis.

Management of Paraneoplastic Syndromes

Tumor Detection

Detection and treatment of tumor are critical in the management of the PNS. Evaluation of a patient with suspected neurological paraneoplastic disorder requires a detailed history regarding risk factors of cancer (eg, smoking, family history of cancers), constitutional symptoms (eg, weight loss, prolonged fever), enlarged lymph nodes, hemoptysis. Computed tomography of the chest, abdomen, and pelvis, mammography, and ovarian and testicular ultrasonography should be considered if a PNS is suspected. Fluorodeoxyglucose positron emission tomography (FDG-PET) should be performed if the initial tests are unrevealing. FDG-PET considerably improves the sensitivity for tumor detection in the setting of paraneoplastic autoimmunity.132–134 If initial cancer screenings are negative, surveillance imaging at 6-month to yearly intervals should be considered to look for the emergence of tumor especially in patients with a positive onconeural antibody.2

Immunotherapy

There have been no data from placebo-controlled trials for immunotherapy in PNS. Discussion here will focus on our approach to patients with PNS. The major principles for PNS management are treatment of the underlying neoplasm and immunosuppression.135

In our practice, intravenous methylprednisolone (IVMP), intravenous immunoglobulin (IVIG), and plasma exchange (PLEX) are commonly utilized as acute immunotherapies either as single therapies or in combinations such as IVMP and PLEX or IVMP and IVIG.136,137 In most PNS cases, in the inpatient setting, we utilize IVMP 1 g daily over 5 days, unless there is a contraindication. We monitor clinical response for 3-5 days after the completion of IVMP before considering IVIG (0.4 g/kg/day for 5 days) or PLEX (5 sessions over 7-14 days). In the outpatient setting, we commonly utilize a 6-week or 12-week IVMP or IVIG regimens (Table 3). Ideally, we re-evaluate within a week of the last infusion. In addition to patient reported change in symptoms, objective measurements such as neurological examination, formal neurocognitive test, neuroimaging (MRI, PET brain), and electrodiagnostic studies, are used for evaluation of the treatment response. In most PNS cases, disease stabilization is considered as a favorable response.138,139

Table 3.

Immunotherapies for Management of Paraneoplastic Syndromes According to Mayo Clinic Experience (Non-Evidence-Based)

Treatment Route, Dose, and Duration Mechanism of Action Common Side Effects
Acute treatments
MethylprednisoloneIVMP 1000 mg/day for 3-5 days
6-week trial: IVMP 1000 mg/day for 3 days
then weekly for 6 weeks
12-week trial: IVMP 1000 mg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Bind to glucocorticoid receptor in cytoplasm, then process through non-genomics and genomics results in reducing chemokine, cytokine, and leukocyte migration to the target tissueHyperglycemia, insomnia, psychosis, cushingoid appearance, osteoporosis, increased risk of infection
Plasma exchange 1.5× plasma volume, 5-7 sessions alternate days (over 10-14 days)Removal of pathological antibodies, immune complex, complements, and cytokinesComplication from catheter insertion, hemodynamic instability, infection
Intravenous immunoglobulin IV 0.4 g/kg/day for 5 days
6-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 6 weeks
12-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Neutralize pathogenic antibodies and block Fc receptors on macrophages preventing phagocytosisHeadache, aseptic meningitis, thromboembolic events, acute renal failure, and anaphylaxis due to IgA deficiency
Maintenance therapy
Cyclophosphamide IV
(0.6-1.0 g/m2 monthly for 6 months)
Oral
(Typical dosing: 2 mg/kg, dosing based on GFR)
Alkylating agent that predominantly depletes T cellsInfection, malignancy (lymphoma, skin cancers, and others), hemorrhagic cystitis, infertility, alopecia, and nausea and vomiting
Rituximab Initial loading dose: 1000 mg once, followed by another 1000 mg dose 2 weeks later. Maintenance dosing: 1000 mg every 6 monthsDepletes CD20+ B cellsAllergic reaction, opportunistic infection, reactivation of tuberculosis infection or hepatitis B infection
AzathioprineStart at 1.5 mg/kg/day. If tolerated, increase to 2 mg/kg/ day. Further increases in dose depend on the MCV or monitoring results. Typical goal dose: 2-3 mg/kg/d.Inhibiting purine synthesis through metabolite 6-mercaptopurine leading to decreased B- and T-cell proliferationHypersensitivity reactions, Infection, nausea, anemia, rash, transaminitis, pancreatitis
Mycophenolate mofetilStart at 500 mg twice per day. If tolerated, then increase to 1000 mg twice per day. Typical goal dose: 2000 mg per dayInhibiting of inosine monophosphate dehydrogenase leading to decreased purine synthesis and DNA formation causing decreased B- and T-cell proliferationInfection, increased risk of malignancy (lymphoma, skin cancers, etc.), diarrhea, hypertension, hepatitis, and renal failure
Treatment Route, Dose, and Duration Mechanism of Action Common Side Effects
Acute treatments
MethylprednisoloneIVMP 1000 mg/day for 3-5 days
6-week trial: IVMP 1000 mg/day for 3 days
then weekly for 6 weeks
12-week trial: IVMP 1000 mg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Bind to glucocorticoid receptor in cytoplasm, then process through non-genomics and genomics results in reducing chemokine, cytokine, and leukocyte migration to the target tissueHyperglycemia, insomnia, psychosis, cushingoid appearance, osteoporosis, increased risk of infection
Plasma exchange 1.5× plasma volume, 5-7 sessions alternate days (over 10-14 days)Removal of pathological antibodies, immune complex, complements, and cytokinesComplication from catheter insertion, hemodynamic instability, infection
Intravenous immunoglobulin IV 0.4 g/kg/day for 5 days
6-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 6 weeks
12-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Neutralize pathogenic antibodies and block Fc receptors on macrophages preventing phagocytosisHeadache, aseptic meningitis, thromboembolic events, acute renal failure, and anaphylaxis due to IgA deficiency
Maintenance therapy
Cyclophosphamide IV
(0.6-1.0 g/m2 monthly for 6 months)
Oral
(Typical dosing: 2 mg/kg, dosing based on GFR)
Alkylating agent that predominantly depletes T cellsInfection, malignancy (lymphoma, skin cancers, and others), hemorrhagic cystitis, infertility, alopecia, and nausea and vomiting
Rituximab Initial loading dose: 1000 mg once, followed by another 1000 mg dose 2 weeks later. Maintenance dosing: 1000 mg every 6 monthsDepletes CD20+ B cellsAllergic reaction, opportunistic infection, reactivation of tuberculosis infection or hepatitis B infection
AzathioprineStart at 1.5 mg/kg/day. If tolerated, increase to 2 mg/kg/ day. Further increases in dose depend on the MCV or monitoring results. Typical goal dose: 2-3 mg/kg/d.Inhibiting purine synthesis through metabolite 6-mercaptopurine leading to decreased B- and T-cell proliferationHypersensitivity reactions, Infection, nausea, anemia, rash, transaminitis, pancreatitis
Mycophenolate mofetilStart at 500 mg twice per day. If tolerated, then increase to 1000 mg twice per day. Typical goal dose: 2000 mg per dayInhibiting of inosine monophosphate dehydrogenase leading to decreased purine synthesis and DNA formation causing decreased B- and T-cell proliferationInfection, increased risk of malignancy (lymphoma, skin cancers, etc.), diarrhea, hypertension, hepatitis, and renal failure

Abbreviations: IVMP, intravenous methylprednisone; IV, intravenous; IVIG, intravenous immunoglobulin; MCV, mean corpuscular volume.

Table 3.

Immunotherapies for Management of Paraneoplastic Syndromes According to Mayo Clinic Experience (Non-Evidence-Based)

Treatment Route, Dose, and Duration Mechanism of Action Common Side Effects
Acute treatments
MethylprednisoloneIVMP 1000 mg/day for 3-5 days
6-week trial: IVMP 1000 mg/day for 3 days
then weekly for 6 weeks
12-week trial: IVMP 1000 mg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Bind to glucocorticoid receptor in cytoplasm, then process through non-genomics and genomics results in reducing chemokine, cytokine, and leukocyte migration to the target tissueHyperglycemia, insomnia, psychosis, cushingoid appearance, osteoporosis, increased risk of infection
Plasma exchange 1.5× plasma volume, 5-7 sessions alternate days (over 10-14 days)Removal of pathological antibodies, immune complex, complements, and cytokinesComplication from catheter insertion, hemodynamic instability, infection
Intravenous immunoglobulin IV 0.4 g/kg/day for 5 days
6-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 6 weeks
12-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Neutralize pathogenic antibodies and block Fc receptors on macrophages preventing phagocytosisHeadache, aseptic meningitis, thromboembolic events, acute renal failure, and anaphylaxis due to IgA deficiency
Maintenance therapy
Cyclophosphamide IV
(0.6-1.0 g/m2 monthly for 6 months)
Oral
(Typical dosing: 2 mg/kg, dosing based on GFR)
Alkylating agent that predominantly depletes T cellsInfection, malignancy (lymphoma, skin cancers, and others), hemorrhagic cystitis, infertility, alopecia, and nausea and vomiting
Rituximab Initial loading dose: 1000 mg once, followed by another 1000 mg dose 2 weeks later. Maintenance dosing: 1000 mg every 6 monthsDepletes CD20+ B cellsAllergic reaction, opportunistic infection, reactivation of tuberculosis infection or hepatitis B infection
AzathioprineStart at 1.5 mg/kg/day. If tolerated, increase to 2 mg/kg/ day. Further increases in dose depend on the MCV or monitoring results. Typical goal dose: 2-3 mg/kg/d.Inhibiting purine synthesis through metabolite 6-mercaptopurine leading to decreased B- and T-cell proliferationHypersensitivity reactions, Infection, nausea, anemia, rash, transaminitis, pancreatitis
Mycophenolate mofetilStart at 500 mg twice per day. If tolerated, then increase to 1000 mg twice per day. Typical goal dose: 2000 mg per dayInhibiting of inosine monophosphate dehydrogenase leading to decreased purine synthesis and DNA formation causing decreased B- and T-cell proliferationInfection, increased risk of malignancy (lymphoma, skin cancers, etc.), diarrhea, hypertension, hepatitis, and renal failure
Treatment Route, Dose, and Duration Mechanism of Action Common Side Effects
Acute treatments
MethylprednisoloneIVMP 1000 mg/day for 3-5 days
6-week trial: IVMP 1000 mg/day for 3 days
then weekly for 6 weeks
12-week trial: IVMP 1000 mg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Bind to glucocorticoid receptor in cytoplasm, then process through non-genomics and genomics results in reducing chemokine, cytokine, and leukocyte migration to the target tissueHyperglycemia, insomnia, psychosis, cushingoid appearance, osteoporosis, increased risk of infection
Plasma exchange 1.5× plasma volume, 5-7 sessions alternate days (over 10-14 days)Removal of pathological antibodies, immune complex, complements, and cytokinesComplication from catheter insertion, hemodynamic instability, infection
Intravenous immunoglobulin IV 0.4 g/kg/day for 5 days
6-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 6 weeks
12-week trial: IVIG 0.4 g/kg/day for 3 days
then weekly for 5 weeks, then every 2 weeks for 6 weeks
Neutralize pathogenic antibodies and block Fc receptors on macrophages preventing phagocytosisHeadache, aseptic meningitis, thromboembolic events, acute renal failure, and anaphylaxis due to IgA deficiency
Maintenance therapy
Cyclophosphamide IV
(0.6-1.0 g/m2 monthly for 6 months)
Oral
(Typical dosing: 2 mg/kg, dosing based on GFR)
Alkylating agent that predominantly depletes T cellsInfection, malignancy (lymphoma, skin cancers, and others), hemorrhagic cystitis, infertility, alopecia, and nausea and vomiting
Rituximab Initial loading dose: 1000 mg once, followed by another 1000 mg dose 2 weeks later. Maintenance dosing: 1000 mg every 6 monthsDepletes CD20+ B cellsAllergic reaction, opportunistic infection, reactivation of tuberculosis infection or hepatitis B infection
AzathioprineStart at 1.5 mg/kg/day. If tolerated, increase to 2 mg/kg/ day. Further increases in dose depend on the MCV or monitoring results. Typical goal dose: 2-3 mg/kg/d.Inhibiting purine synthesis through metabolite 6-mercaptopurine leading to decreased B- and T-cell proliferationHypersensitivity reactions, Infection, nausea, anemia, rash, transaminitis, pancreatitis
Mycophenolate mofetilStart at 500 mg twice per day. If tolerated, then increase to 1000 mg twice per day. Typical goal dose: 2000 mg per dayInhibiting of inosine monophosphate dehydrogenase leading to decreased purine synthesis and DNA formation causing decreased B- and T-cell proliferationInfection, increased risk of malignancy (lymphoma, skin cancers, etc.), diarrhea, hypertension, hepatitis, and renal failure

Abbreviations: IVMP, intravenous methylprednisone; IV, intravenous; IVIG, intravenous immunoglobulin; MCV, mean corpuscular volume.

The decision for long-term immunotherapy can be challenging. In patients with classic PNS and typical cancer-onconeural antibody associations, who present within 2 years from the onset of symptoms, we consider a long-term immunotherapy in the early disease course. For patients who presented late and are not having active disease progression, we usually re-evaluate the clinical response after treatment of underlying tumor and 12-week course of acute therapy, before considering the further escalation of immunological treatment.

Among patients with onconeural antibodies targeting an intracellular epitope, our preferred options for chronic immunosuppression are azathioprine, mycophenolate, and cyclophosphamide which target both T- and B-lymphocytes because the pathogenesis of these PNS is presumed to involve cellular immune response. Severity and progression are the factors that we use to determine the agent of choice. In aggressive cases, oral or IV cyclophosphamide is the preferred option. A prospective study demonstrated that twice as many patients who were treated with a combination of PLEX and cyclophosphamide demonstrated an improvement in disability compared to PLEX alone; however, severe leukopenia was seen in 4 of 10 patients who received cyclophosphamide.139 Among PNS with cell-surface antibodies, such as GABA-B-R IgG or mGlur5, we prefer rituximab as chronic immunotherapy because these antibodies are thought to be directly pathogenic. Therefore, suppression of antibody production by targeting B cells is potentially beneficial (Table 3).

Management of n-irAEs

The American Society of Clinical Oncology and the Society for Immunotherapy of Cancer recommend holding ICI in all CTCAE grade 3-4 toxicity and initiation of corticosteroids.140,141 Most n-irAE with PNS phenotype are classified as grade 3 or 4.2 In our practice, all severe n-irAE patients are treated with IVMP 1 g/day for 3-5 days or oral prednisone 60-80 mg/day depending on the n-irAE phenotype.107 In the patients with favorable response to corticosteroids, 4- to 6-week course of prednisone taper is considered. In the refractory cases, early initiation of IVIG, plasmapheresis, and other immunosuppression medications such as rituximab should be considered.107,141 The majority of the n-irAE patients with classic paraneoplastic phenotype and onconeural antibody seropositivity usually have a refractory course and require more aggressive immunosuppression.142 In few of these cases, we have utilized cyclophosphamide with limited success.112

Retreating with ICI potentially increases the risk of n-irAE relapse.107 We try to avoid re-treatment among patients with n-irAEs resembling classic paraneoplastic syndrome due to risk of irreversible neurological deterioration.142 If an ICI has to be resumed, we treat the patients with corticosteroids until the symptom resolution or stabilization and observe for 2-8 weeks before restarting an ICI.

Conclusion

In the past century, the number of serological biomarkers and associated phenotypes has considerably expanded. Furthermore, as cancer indications of ICIs grow, we are likely to encounter more patients with clinical phenotypes resembling PNS in our neurologic practice. Recognition of various clinical phenotypes, onconeural antibodies, and their neoplastic association may help clinicians diagnose these rare but refractory conditions early and treat them appropriately.

Funding

None declared.

Conflict of interest statement. Dr. J.J., Dr. P.P., Dr. S.O.M., and Dr. S.T. have no disclosures. Dr. S.J.P. is a named inventor on filed patents that relate to functional AQP4/NMO-IgG assays and NMO-IgG as a cancer marker. He has a patent pending for Septin 5, MAP1B, and KLHL11-IgGs as markers of neurological autoimmunity and paraneoplastic disorders. He has consulted for Alexion and Medimmune. He has received research support from Grifols, Medimmune, and Alexion. All compensation for consulting activities is paid directly to Mayo Clinic. D.D. has received research support from Center of Multiple Sclerosis and Autoimmune Neurology, Center for Clinical and Translational Science, and Grifols Pharmaceuticals. He has consulted for UCB and Astellas Pharmaceuticals. All compensation for consulting activities is paid directly to Mayo Clinic. Dr. D.D. has a patent pending for KLHL11-IgG as a marker of neurological autoimmunity.

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.

References

1.

Darnell
RB
,
Posner
JB
.
Paraneoplastic syndromes involving the nervous system
.
N Engl J Med.
2003
;
349
(
16
):
1543
1554
.

2.

Graus
F
,
Dalmau
J
.
Paraneoplastic neurological syndromes in the era of immune-checkpoint inhibitors
.
Nat Rev Clin Oncol.
2019
;
16
(
9
):
535
548
.

3.

Oppenheim
H
.
Uber hirnsymptome gei carcinomatose ohne nachweibare veranderungen um gehirm
.
Charite-Ann Berlin
.
1888
;
13
:
335
344
.

4.

Auchè
M
.
Des nevrites peripheriques chez les cancereux
.
Rev Med
.
1890
;
10
:
785
807
.

5.

Denny-Brown
D
.
Primary sensory neuropathy with muscular changes associated with carcinoma
.
J Neurol Neurosurg Psychiatry.
1948
;
11
(
2
):
73
87
.

6.

Wilkinson
PC
.
Serological findings in carcinomatous neuromyopathy
.
Lancet.
1964
;
1
(
7346
):
1301
1303
.

7.

Wilkinson
PC
,
Zeromski
J
.
Immunofluorescent detection of antibodies against neurones in sensory carcinomatous neuropathy
.
Brain.
1965
;
88
(
3
):
529
583
.

8.

Graus
F
,
Cordon-Cardo
C
,
Posner
JB
.
Neuronal antinuclear antibody in sensory neuronopathy from lung cancer
.
Neurology.
1985
;
35
(
4
):
538
543
.

9.

Graus
F
,
Elkon
KB
,
Cordon-Cardo
C
, et al.
Sensory neuronopathy and small cell lung cancer. Antineuronal antibody that also reacts with the tumor
.
Am J Med.
1986
;
80
(
1
):
45
52
.

10.

Dalmau
J
,
Graus
F
,
Rosenblum
MK
, et al.
Anti-Hu-associated paraneoplastic encephalomyelitis/sensory neuronopathy. A clinical study of 71 patients
.
Medicine (Baltimore).
1992
;
71
(
2
):
59
72
.

11.

Trotter
JL
,
Hendin
BA
,
Osterland
CK
.
Cerebellar degeneration with Hodgkin disease. An immunological study
.
Arch Neurol.
1976
;
33
(
9
):
660
661
.

12.

Greenlee
JE
,
Brashear
HR
.
Antibodies to cerebellar Purkinje cells in patients with paraneoplastic cerebellar degeneration and ovarian carcinoma
.
Ann Neurol.
1983
;
14
(
6
):
609
613
.

13.

Jaeckle
KA
,
Graus
F
,
Houghton
A
, et al.
Autoimmune response of patients with paraneoplastic cerebellar degeneration to a Purkinje cell cytoplasmic protein antigen
.
Ann Neurol.
1985
;
18
(
5
):
592
600
.

14.

Anderson
NE
,
Rosenblum
MK
,
Posner
JB
.
Paraneoplastic cerebellar degeneration: clinical-immunological correlations
.
Ann Neurol.
1988
;
24
(
4
):
559
567
.

15.

Rodriguez
M
,
Truh
LI
,
O’Neill
BP
, et al.
Autoimmune paraneoplastic cerebellar degeneration: ultrastructural localization of antibody-binding sites in Purkinje cells
.
Neurology.
1988
;
38
(
9
):
1380
1386
.

16.

Cunningham
J
,
Graus
F
,
Anderson
N
, et al.
Partial characterization of the Purkinje cell antigens in paraneoplastic cerebellar degeneration
.
Neurology.
1986
;
36
(
9
):
1163
1168
.

17.

Voltz
R
,
Gultekin
SH
,
Rosenfeld
MR
, et al.
A serologic marker of paraneoplastic limbic and brain-stem encephalitis in patients with testicular cancer
.
N Engl J Med.
1999
;
340
(
23
):
1788
1795
.

18.

Honnorat
J
,
Byk
T
,
Kusters
I
, et al.
Ulip/CRMP proteins are recognized by autoantibodies in paraneoplastic neurological syndromes
.
Eur J Neurosci.
1999
;
11
(
12
):
4226
4232
.

19.

Yu
Z
,
Kryzer
TJ
,
Griesmann
GE
, et al.
CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity
.
Ann Neurol.
2001
;
49
(
2
):
146
154
.

20.

Graus
F
,
Delattre
JY
,
Antoine
JC
, et al.
Recommended diagnostic criteria for paraneoplastic neurological syndromes
.
J Neurol Neurosurg Psychiatry.
2004
;
75
(
8
):
1135
1140
.

21.

Vogrig
A
,
Gigli
GL
,
Segatti
S
, et al.
Epidemiology of paraneoplastic neurological syndromes: a population-based study
.
J Neurol.
2020
;
267
(
1
):
26
35
.

22.

Dalmau
J
,
Gultekin
HS
,
Posner
JB
.
Paraneoplastic neurologic syndromes: pathogenesis and physiopathology
.
Brain Pathol.
1999
;
9
(
2
):
275
284
.

23.

Finn
OJ
.
Cancer immunology
.
N Engl J Med.
2008
;
358
(
25
):
2704
2715
.

24.

Dubey
D
,
Wilson
MR
,
Clarkson
B
, et al.
Expanded clinical phenotype, oncological associations, and immunopathologic insights of paraneoplastic kelch-like protein-11 encephalitis
.
JAMA Neurol
. 2020;77(11):1420–1429.

25.

Altman
AJ
,
Baehner
RL
.
Favorable prognosis for survival in children with coincident opso-myoclonus and neuroblastoma
.
Cancer.
1976
;
37
(
2
):
846
852
.

26.

Maddison
P
,
Newsom-Davis
J
,
Mills
KR
, et al.
Favourable prognosis in Lambert-Eaton myasthenic syndrome and small-cell lung carcinoma
.
Lancet.
1999
;
353
(
9147
):
117
118
.

27.

Greenlee
JE
,
Clawson
SA
,
Hill
KE
, et al.
Purkinje cell death after uptake of anti-Yo antibodies in cerebellar slice cultures
.
J Neuropathol Exp Neurol.
2010
;
69
(
10
):
997
1007
.

28.

Greenlee
JE
,
Clawson
SA
,
Hill
KE
, et al.
Neuronal uptake of anti-Hu antibody, but not anti-Ri antibody, leads to cell death in brain slice cultures
.
J Neuroinflammation.
2014
;
11
:
160
.

29.

Greenlee
JE
,
Parks
TN
,
Jaeckle
KA
.
Type IIa (‘anti-Hu’) antineuronal antibodies produce destruction of rat cerebellar granule neurons in vitro
.
Neurology.
1993
;
43
(
10
):
2049
2054
.

30.

Greenlee
JE
,
Burns
JB
,
Rose
JW
, et al.
Uptake of systemically administered human anticerebellar antibody by rat Purkinje cells following blood-brain barrier disruption
.
Acta Neuropathol.
1995
;
89
(
4
):
341
345
.

31.

Hormigo
A
,
Lieberman
F
.
Nuclear localization of anti-Hu antibody is not associated with in vitro cytotoxicity
.
J Neuroimmunol.
1994
;
55
(
2
):
205
212
.

32.

Tanaka
K
,
Ding
X
,
Tanaka
M
.
Effects of antineuronal antibodies from patients with paraneoplastic neurological syndrome on primary-cultured neurons
.
J Neurol Sci.
2004
;
217
(
1
):
25
30
.

33.

Graus
F
,
Illa
I
,
Agusti
M
, et al.
Effect of intraventricular injection of an anti-Purkinje cell antibody (anti-Yo) in a guinea pig model
.
J Neurol Sci.
1991
;
106
(
1
):
82
87
.

34.

Hormigo
A
,
Dalmau
J
,
Rosenblum
MK
, et al.
Immunological and pathological study of anti-Ri-associated encephalopathy
.
Ann Neurol.
1994
;
36
(
6
):
896
902
.

35.

Wanschitz
J
,
Hainfellner
JA
,
Kristoferitsch
W
, et al.
Ganglionitis in paraneoplastic subacute sensory neuronopathy: a morphologic study
.
Neurology.
1997
;
49
(
4
):
1156
1159
.

36.

Aye
MM
,
Kasai
T
,
Tashiro
Y
, et al.
CD8 positive T-cell infiltration in the dentate nucleus of paraneoplastic cerebellar degeneration
.
J Neuroimmunol.
2009
;
208
(
1–2
):
136
140
.

37.

Graus
F
,
Ribalta
T
,
Campo
E
, et al.
Immunohistochemical analysis of the immune reaction in the nervous system in paraneoplastic encephalomyelitis
.
Neurology.
1990
;
40
(
2
):
219
222
.

38.

Hughes
EG
,
Peng
X
,
Gleichman
AJ
, et al.
Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis
.
J Neurosci.
2010
;
30
(
17
):
5866
5875
.

39.

Martinez-Hernandez
E
,
Horvath
J
,
Shiloh-Malawsky
Y
, et al.
Analysis of complement and plasma cells in the brain of patients with anti-NMDAR encephalitis
.
Neurology.
2011
;
77
(
6
):
589
593
.

40.

Dalmau
J
,
Gleichman
AJ
,
Hughes
EG
, et al.
Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies
.
Lancet Neurol.
2008
;
7
(
12
):
1091
1098
.

41.

Ramberger
M
,
Berretta
A
,
Tan
JMM
, et al.
Distinctive binding properties of human monoclonal LGI1 autoantibodies determine pathogenic mechanisms
.
Brain.
2020
;
143
(
6
):
1731
1745
.

42.

Tüzün
E
,
Zhou
L
,
Baehring
JM
, et al.
Evidence for antibody-mediated pathogenesis in anti-NMDAR encephalitis associated with ovarian teratoma
.
Acta Neuropathol.
2009
;
118
(
6
):
737
743
.

43.

Manto
M
,
Dalmau
J
,
Didelot
A
, et al.
In vivo effects of antibodies from patients with anti-NMDA receptor encephalitis: further evidence of synaptic glutamatergic dysfunction
.
Orphanet J Rare Dis.
2010
;
5
:
31
.

44.

Manto
M
,
Dalmau
J
,
Didelot
A
, et al.
Afferent facilitation of corticomotor responses is increased by IgGs of patients with NMDA-receptor antibodies
.
J Neurol.
2011
;
258
(
1
):
27
33
.

45.

Gultekin
SH
,
Rosenfeld
MR
,
Voltz
R
, et al.
Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumour association in 50 patients
.
Brain.
2000
;
123
(
Pt 7
):
1481
1494
.

46.

Graus
F
,
Titulaer
MJ
,
Balu
R
, et al.
A clinical approach to diagnosis of autoimmune encephalitis
.
Lancet Neurol.
2016
;
15
(
4
):
391
404
.

47.

Lancaster
E
,
Martinez-Hernandez
E
,
Titulaer
MJ
, et al.
Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome
.
Neurology.
2011
;
77
(
18
):
1698
1701
.

48.

Carr
I
.
The Ophelia syndrome: memory loss in Hodgkin’s disease
.
Lancet.
1982
;
1
(
8276
):
844
845
.

49.

Dalmau
J
,
Graus
F
,
Villarejo
A
, et al.
Clinical analysis of anti-Ma2-associated encephalitis
.
Brain.
2004
;
127
(
Pt 8
):
1831
1844
.

50.

Mandel-Brehm
C
,
Dubey
D
,
Kryzer
TJ
, et al.
Kelch-like protein 11 antibodies in seminoma-associated paraneoplastic encephalitis
.
N Engl J Med.
2019
;
381
(
1
):
47
54
.

51.

Grisold
W
,
Giometto
B
,
Vitaliani
R
, et al.
Current approaches to the treatment of paraneoplastic encephalitis
.
Ther Adv Neurol Disord.
2011
;
4
(
4
):
237
248
.

52.

Shah
S
,
Do Campo
RV
,
Kumar
N
, et al.
Paraneoplastic myeloneuropathies: clinical, oncologic, and serologic accompaniments
.
Neurology
. 2021;96(4):e632–9.

53.

Antoine
JC
,
Absi
L
,
Honnorat
J
, et al.
Antiamphiphysin antibodies are associated with various paraneoplastic neurological syndromes and tumors
.
Arch Neurol.
1999
;
56
(
2
):
172
177
.

54.

Dubey
D
,
Hinson
SR
,
Jolliffe
EA
, et al.
Autoimmune GFAP astrocytopathy: prospective evaluation of 90 patients in 1 year
.
J Neuroimmunol.
2018
;
321
:
157
163
.

55.

Flanagan
EP
,
Hinson
SR
,
Lennon
VA
, et al.
Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: analysis of 102 patients
.
Ann Neurol.
2017
;
81
(
2
):
298
309
.

56.

Long
Y
,
Liang
J
,
Xu
H
, et al.
Autoimmune glial fibrillary acidic protein astrocytopathy in Chinese patients: a retrospective study
.
Eur J Neurol.
2018
;
25
(
3
):
477
483
.

57.

Keime-Guibert
F
,
Graus
F
,
Broët
P
, et al.
Clinical outcome of patients with anti-Hu-associated encephalomyelitis after treatment of the tumor
.
Neurology.
1999
;
53
(
8
):
1719
1723
.

58.

Graus
F
,
Keime-Guibert
F
,
Reñe
R
, et al.
Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients
.
Brain.
2001
;
124
(
Pt 6
):
1138
1148
.

59.

Giometto
B
,
Grisold
W
,
Vitaliani
R
, et al.
Paraneoplastic neurologic syndrome in the PNS Euronetwork database: a European study from 20 centers
.
Arch Neurol.
2010
;
67
(
3
):
330
335
.

60.

Hartmann
J
,
Henning
HA
,
Konnerth
A
.
mGluR1/TRPC3-mediated synaptic transmission and calcium signaling in mammalian central neurons
.
Cold Spring Harb Perspect Biol
.
2011
;
3
(
4
):
a006726
.

61.

Schubert
M
,
Panja
D
,
Haugen
M
, et al.
Paraneoplastic CDR2 and CDR2L antibodies affect Purkinje cell calcium homeostasis
.
Acta Neuropathol.
2014
;
128
(
6
):
835
852
.

62.

Albert
ML
,
Darnell
JC
,
Bender
A
, et al.
Tumor-specific killer cells in paraneoplastic cerebellar degeneration
.
Nat Med.
1998
;
4
(
11
):
1321
1324
.

63.

Rojas
I
,
Graus
F
,
Keime-Guibert
F
, et al.
Long-term clinical outcome of paraneoplastic cerebellar degeneration and anti-Yo antibodies
.
Neurology.
2000
;
55
(
5
):
713
715
.

64.

Pittock
SJ
,
Lucchinetti
CF
,
Lennon
VA
.
Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments
.
Ann Neurol.
2003
;
53
(
5
):
580
587
.

65.

Ortega Suero
G
,
Sola-Valls
N
,
Escudero
D
, et al.
Anti-Ma and anti-Ma2-associated paraneoplastic neurological syndromes
.
Neurologia.
2018
;
33
(
1
):
18
27
.

66.

Gorman
MP
.
Update on diagnosis, treatment, and prognosis in opsoclonus-myoclonus-ataxia syndrome
.
Curr Opin Pediatr.
2010
;
22
(
6
):
745
750
.

67.

Tate
ED
,
Allison
TJ
,
Pranzatelli
MR
, et al.
Neuroepidemiologic trends in 105 US cases of pediatric opsoclonus-myoclonus syndrome
.
J Pediatr Oncol Nurs.
2005
;
22
(
1
):
8
19
.

68.

Berridge
G
,
Menassa
DA
,
Moloney
T
, et al.
Glutamate receptor δ2 serum antibodies in pediatric opsoclonus myoclonus ataxia syndrome
.
Neurology.
2018
;
91
(
8
):
e714
e723
.

69.

Klaas
JP
,
Ahlskog
JE
,
Pittock
SJ
, et al.
Adult-onset opsoclonus-myoclonus syndrome
.
Arch Neurol.
2012
;
69
(
12
):
1598
1607
.

70.

Oh
SY
,
Kim
JS
,
Dieterich
M
.
Update on opsoclonus-myoclonus syndrome in adults
.
J Neurol.
2019
;
266
(
6
):
1541
1548
.

71.

Bataller
L
,
Graus
F
,
Saiz
A
, et al. ;
Spanish Opsoclonus-Myoclonus Study Group
.
Clinical outcome in adult onset idiopathic or paraneoplastic opsoclonus-myoclonus
.
Brain.
2001
;
124
(
Pt 2
):
437
443
.

72.

Flanagan
EP
,
McKeon
A
,
Lennon
VA
, et al.
Paraneoplastic isolated myelopathy: clinical course and neuroimaging clues
.
Neurology.
2011
;
76
(
24
):
2089
2095
.

73.

Liu
Z
,
Jiao
L
,
Qiu
Z
, et al.
Clinical characteristics of patients with paraneoplastic myelopathy
.
J Neuroimmunol.
2019
;
330
:
136
142
.

74.

Flanagan
EP
,
Keegan
BM
.
Paraneoplastic myelopathy
.
Neurol Clin.
2013
;
31
(
1
):
307
318
.

75.

Sepúlveda
M
,
Sola-Valls
N
,
Escudero
D
, et al.
Clinical profile of patients with paraneoplastic neuromyelitis optica spectrum disorder and aquaporin-4 antibodies
.
Mult Scler.
2018
;
24
(
13
):
1753
1759
.

76.

Moersch
FP
,
Woltman
HW
.
Progressive fluctuating muscular rigidity and spasm (“stiff-man” syndrome); report of a case and some observations in 13 other cases
.
Proc Staff Meet Mayo Clin.
1956
;
31
(
15
):
421
427
.

77.

Asher
R
.
A woman with the stiff-man syndrome
.
Br Med J
.
1958
;
1
(
5065
):
265
266
.

78.

Blum
P
,
Jankovic
J
.
Stiff-person syndrome: an autoimmune disease
.
Mov Disord.
1991
;
6
(
1
):
12
20
.

79.

Sarva
H
,
Deik
A
,
Ullah
A
, et al.
Clinical spectrum of stiff person syndrome: a review of recent reports
.
Tremor Other Hyperkinet Mov (N Y).
2016
;
6
:
340
.

80.

Mckeon
A
,
Vincent
A
.
Autoimmune movement disorders
.
Handb Clin Neurol.
2016
;
133
:
301
315
.

81.

McKeon
A
,
Martinez-Hernandez
E
,
Lancaster
E
, et al.
Glycine receptor autoimmune spectrum with stiff-man syndrome phenotype
.
JAMA Neurol.
2013
;
70
(
1
):
44
50
.

82.

McKeon
A
,
Pittock
SJ
,
Lennon
VA
.
Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease
.
Neurology.
2009
;
73
(
24
):
2132
;
author reply 2133
.

83.

Tobin
WO
,
Lennon
VA
,
Komorowski
L
, et al.
DPPX potassium channel antibody: frequency, clinical accompaniments, and outcomes in 20 patients
.
Neurology.
2014
;
83
(
20
):
1797
1803
.

84.

Balint
B
,
Meinck
HM
.
Pragmatic treatment of stiff person spectrum disorders
.
Mov Disord Clin Pract.
2018
;
5
(
4
):
394
401
.

85.

Kuntzer
T
,
Antoine
JC
,
Steck
AJ
.
Clinical features and pathophysiological basis of sensory neuronopathies (ganglionopathies)
.
Muscle Nerve.
2004
;
30
(
3
):
255
268
.

86.

Camdessanché
JP
,
Jousserand
G
,
Ferraud
K
, et al.
The pattern and diagnostic criteria of sensory neuronopathy: a case-control study
.
Brain.
2009
;
132
(
Pt 7
):
1723
1733
.

87.

Antoine
JC
,
Camdessanché
JP
.
Paraneoplastic neuropathies
.
Curr Opin Neurol.
2017
;
30
(
5
):
513
520
.

88.

Grisold
W
,
Grisold
A
,
Löscher
WN
.
Neuromuscular complications in cancer
.
J Neurol Sci.
2016
;
367
:
184
202
.

89.

Dubey
D
,
Lennon
VA
,
Gadoth
A
, et al.
Autoimmune CRMP5 neuropathy phenotype and outcome defined from 105 cases
.
Neurology.
2018
;
90
(
2
):
e103
e110
.

90.

Dubey
D
,
Jitprapaikulsan
J
,
Bi
H
, et al.
Amphiphysin-IgG autoimmune neuropathy: a recognizable clinicopathologic syndrome
.
Neurology.
2019
;
93
(
20
):
e1873
e1880
.

91.

Jitprapaikulsan
J
,
Klein
CJ
,
Pittock
SJ
, et al.
Phenotypic presentations of paraneoplastic neuropathies associated with MAP1B-IgG
.
J Neurol Neurosurg Psychiatry.
2020
;
91
(
3
):
328
330
.

92.

Lennon
VA
,
Sas
DF
,
Busk
MF
, et al.
Enteric neuronal autoantibodies in pseudoobstruction with small-cell lung carcinoma
.
Gastroenterology.
1991
;
100
(
1
):
137
142
.

93.

Li
Y
,
Jammoul
A
,
Mente
K
, et al.
Clinical experience of seropositive ganglionic acetylcholine receptor antibody in a tertiary neurology referral center
.
Muscle Nerve.
2015
;
52
(
3
):
386
391
.

94.

McKeon
A
,
Lennon
VA
,
Lachance
DH
, et al.
Ganglionic acetylcholine receptor autoantibody: oncological, neurological, and serological accompaniments
.
Arch Neurol.
2009
;
66
(
6
):
735
741
.

95.

Huang
K
,
Luo
YB
,
Yang
H
.
Autoimmune channelopathies at neuromuscular junction
.
Front Neurol.
2019
;
10
:
516
.

96.

Torres-Vega
E
,
Mancheño
N
,
Cebrián-Silla
A
, et al.
Netrin-1 receptor antibodies in thymoma-associated neuromyotonia with myasthenia gravis
.
Neurology.
2017
;
88
(
13
):
1235
1242
.

97.

Gilhus
NE
,
Tzartos
S
,
Evoli
A
, et al.
Myasthenia gravis
.
Nat Rev Dis Primers.
2019
;
5
(
1
):
30
.

98.

Yamasaki
M
,
Funaishi
K
,
Saito
N
, et al.
Acetylcholine receptor antibody-positive myasthenia gravis associated with small-cell lung cancer: a case report
.
Medicine (Baltimore).
2018
;
97
(
17
):
e0541
.

99.

Lennon
VA
,
Lambert
EH
,
Whittingham
S
, et al.
Autoimmunity in the Lambert-Eaton myasthenic syndrome
.
Muscle Nerve.
1982
;
5
(
9S
):
S21
S25
.

100.

Lennon
VA
,
Kryzer
TJ
,
Griesmann
GE
, et al.
Calcium-channel antibodies in the Lambert-Eaton syndrome and other paraneoplastic syndromes
.
N Engl J Med.
1995
;
332
(
22
):
1467
1474
.

101.

Ivanovski
T
,
Miralles
F
.
Lambert-Eaton Myasthenic syndrome: early diagnosis is key
.
Degener Neurol Neuromuscul Dis.
2019
;
9
:
27
37
.

102.

Milone
M
.
Diagnosis and management of immune-mediated myopathies
.
Mayo Clin Proc.
2017
;
92
(
5
):
826
837
.

103.

Kassardjian
CD
,
Lennon
VA
,
Alfugham
NB
, et al.
Clinical features and treatment outcomes of necrotizing autoimmune myopathy
.
JAMA Neurol.
2015
;
72
(
9
):
996
1003
.

104.

Allenbach
Y
,
Keraen
J
,
Bouvier
AM
, et al.
High risk of cancer in autoimmune necrotizing myopathies: usefulness of myositis specific antibody
.
Brain.
2016
;
139
(
Pt 8
):
2131
2135
.

105.

Yang
Z
,
Lin
F
,
Qin
B
, et al.
Polymyositis/dermatomyositis and malignancy risk: a metaanalysis study
.
J Rheumatol.
2015
;
42
(
2
):
282
291
.

106.

Yang
H
,
Peng
Q
,
Yin
L
, et al.
Identification of multiple cancer-associated myositis-specific autoantibodies in idiopathic inflammatory myopathies: a large longitudinal cohort study
.
Arthritis Res Ther.
2017
;
19
(
1
):
259
.

107.

Dubey
D
,
David
WS
,
Reynolds
KL
, et al.
Severe neurological toxicity of immune checkpoint inhibitors: growing spectrum
.
Ann Neurol.
2020
;
87
(
5
):
659
669
.

108.

Park
YJ
,
Kuen
DS
,
Chung
Y
.
Future prospects of immune checkpoint blockade in cancer: from response prediction to overcoming resistance
.
Exp Mol Med.
2018
;
50
(
8
):
109
.

109.

Martins
F
,
Sofiya
L
,
Sykiotis
GP
, et al.
Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance
.
Nat Rev Clin Oncol.
2019
;
16
(
9
):
563
580
.

110.

Ramos-Casals
M
,
Brahmer
JR
,
Callahan
MK
, et al.
Immune-related adverse events of checkpoint inhibitors
.
Nat Rev Dis Primers.
2020
;
6
(
1
):
38
.

111.

Kanjanapan
Y
,
Day
D
,
Butler
MO
, et al.
Delayed immune-related adverse events in assessment for dose-limiting toxicity in early phase immunotherapy trials
.
Eur J Cancer.
2019
;
107
:
1
7
.

112.

Kunchok
A
,
Zekeridou
A
,
Pittock
S
.
CRMP5-IgG-Associated paraneoplastic myelopathy with PD-L1 inhibitor therapy
.
JAMA Neurol
.
2020;77(2):255–256
.

113.

Postow
MA
,
Sidlow
R
,
Hellmann
MD
.
Immune-related adverse events associated with immune checkpoint blockade
.
N Engl J Med.
2018
;
378
(
2
):
158
168
.

114.

Yshii
LM
,
Hohlfeld
R
,
Liblau
RS
.
Inflammatory CNS disease caused by immune checkpoint inhibitors: status and perspectives
.
Nat Rev Neurol.
2017
;
13
(
12
):
755
763
.

115.

Yshii
LM
,
Gebauer
CM
,
Pignolet
B
, et al.
CTLA4 blockade elicits paraneoplastic neurological disease in a mouse model
.
Brain.
2016
;
139
(
11
):
2923
2934
.

116.

Johnson
DB
,
Balko
JM
,
Compton
ML
, et al.
Fulminant myocarditis with combination immune checkpoint blockade
.
N Engl J Med.
2016
;
375
(
18
):
1749
1755
.

117.

Johnson
DB
,
McDonnell
WJ
,
Gonzalez-Ericsson
PI
, et al.
A case report of clonal EBV-like memory CD4+ T cell activation in fatal checkpoint inhibitor-induced encephalitis
.
Nat Med.
2019
;
25
(
8
):
1243
1250
.

118.

Dalakas
MC
.
Neurological complications of immune checkpoint inhibitors: what happens when you ‘take the brakes off’ the immune system
.
Ther Adv Neurol Disord.
2018
;
11
:
1756286418799864
.

119.

Crosson
JN
,
Laird
PW
,
Debiec
M
, et al.
Vogt-Koyanagi-Harada-like syndrome after CTLA-4 inhibition with ipilimumab for metastatic melanoma
.
J Immunother.
2015
;
38
(
2
):
80
84
.

120.

Gkiozos
I
,
Kopitopoulou
A
,
Kalkanis
A
, et al.
Sarcoidosis-like reactions induced by checkpoint inhibitors
.
J Thorac Oncol.
2018
;
13
(
8
):
1076
1082
.

121.

Gerdes
LA
,
Held
K
,
Beltrán
E
, et al.
CTLA4 as Immunological checkpoint in the development of multiple sclerosis
.
Ann Neurol.
2016
;
80
(
2
):
294
300
.

122.

Gettings
EJ
,
Hackett
CT
,
Scott
TF
.
Severe relapse in a multiple sclerosis patient associated with ipilimumab treatment of melanoma
.
Mult Scler.
2015
;
21
(
5
):
670
.

123.

Fellner
A
,
Makranz
C
,
Lotem
M
, et al.
Neurologic complications of immune checkpoint inhibitors
.
J Neurooncol.
2018
;
137
(
3
):
601
609
.

124.

Papadopoulos
KP
,
Romero
RS
,
Gonzalez
G
, et al.
Anti-Hu-Associated autoimmune limbic encephalitis in a patient with PD-1 inhibitor-responsive myxoid chondrosarcoma
.
Oncologist.
2018
;
23
(
1
):
118
120
.

125.

Williams
TJ
,
Benavides
DR
,
Patrice
KA
, et al.
Association of autoimmune encephalitis with combined immune checkpoint inhibitor treatment for metastatic cancer
.
JAMA Neurol.
2016
;
73
(
8
):
928
933
.

126.

Kawamura
R
,
Nagata
E
,
Mukai
M
, et al.
Acute cerebellar ataxia induced by nivolumab
.
Intern Med.
2017
;
56
(
24
):
3357
3359
.

127.

Maller
B
,
Peguero
E
,
Tanvetyanon
T
.
Ipilimumab/Nivolumab-related opsoclonus-myoclonus-ataxia syndrome variant in a patient with malignant pleural mesothelioma
.
J Immunother.
2018
;
41
(
9
):
411
412
.

128.

Matsuoka
H
,
Kimura
H
,
Koba
H
, et al.
Nivolumab-induced limbic encephalitis with anti-hu antibody in a patient with advanced pleomorphic carcinoma of the lung
.
Clin Lung Cancer.
2018
;
19
(
5
):
e597
e599
.

129.

Puwanant
A
,
Isfort
M
,
Lacomis
D
, et al.
Clinical spectrum of neuromuscular complications after immune checkpoint inhibition
.
Neuromuscul Disord.
2019
;
29
(
2
):
127
133
.

130.

Agrawal
K
,
Agrawal
N
.
Lambert-Eaton myasthenic syndrome secondary to nivolumab and ipilimumab in a patient with small-cell lung cancer
.
Case Rep Neurol Med.
2019
;
2019
:
5353202
.

131.

Nakatani
Y
,
Tanaka
N
,
Enami
T
, et al.
Lambert-Eaton myasthenic syndrome caused by nivolumab in a patient with squamous cell lung cancer
.
Case Rep Neurol.
2018
;
10
(
3
):
346
352
.

132.

Hadjivassiliou
M
,
Alder
SJ
,
Van Beek
EJ
, et al.
PET scan in clinically suspected paraneoplastic neurological syndromes: a 6-year prospective study in a regional neuroscience unit
.
Acta Neurol Scand.
2009
;
119
(
3
):
186
193
.

133.

McKeon
A
,
Apiwattanakul
M
,
Lachance
DH
, et al.
Positron emission tomography-computed tomography in paraneoplastic neurologic disorders: systematic analysis and review
.
Arch Neurol.
2010
;
67
(
3
):
322
329
.

134.

Rees
JH
,
Hain
SF
,
Johnson
MR
, et al.
The role of [18F]fluoro-2-deoxyglucose-PET scanning in the diagnosis of paraneoplastic neurological disorders
.
Brain.
2001
;
124
(
Pt 11
):
2223
2231
.

135.

Shams’ili
S
,
Grefkens
J
,
de Leeuw
B
, et al.
Paraneoplastic cerebellar degeneration associated with antineuronal antibodies: analysis of 50 patients
.
Brain.
2003
;
126
(
Pt 6
):
1409
1418
.

136.

Lancaster
E
.
Paraneoplastic disorders
.
Continuum (Minneap Minn).
2017
;
23
(
6, Neuro-oncology
):
1653
1679
.

137.

McKeon
A
,
Lennon
VA
,
Pittock
SJ
.
Immunotherapy-responsive dementias and encephalopathies
.
Continuum (Minneap Minn).
2010
;
16
(
2 Dementia
):
80
101
.

138.

Keime-Guibert
F
,
Graus
F
,
Fleury
A
, et al.
Treatment of paraneoplastic neurological syndromes with antineuronal antibodies (anti-Hu, anti-Yo) with a combination of immunoglobulins, cyclophosphamide, and methylprednisolone
.
J Neurol Neurosurg Psychiatry.
2000
;
68
(
4
):
479
482
.

139.

Vernino
S
,
O’Neill
BP
,
Marks
RS
, et al.
Immunomodulatory treatment trial for paraneoplastic neurological disorders
.
Neuro Oncol.
2004
;
6
(
1
):
55
62
.

140.

Brahmer
JR
,
Lacchetti
C
,
Schneider
BJ
, et al.
Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline
.
J Clin Oncol.
2018
;
36
(
17
):
1714
1768
.

141.

Puzanov
I
,
Diab
A
,
Abdallah
K
, et al.
Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group
.
J Immunother Cancer.
2017
;
5
(
1
):
95
.

142.

Sechi
E
,
Markovic
SN
,
McKeon
A
, et al.
Neurologic autoimmunity and immune checkpoint inhibitors: autoantibody profiles and outcomes
.
Neurology.
2020
;
95
(
17
):
e2442
e2452
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)