Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Cancer Letters 228 (2005) 275–282

www.elsevier.com/locate/canlet

Opsoclonus myoclonus syndrome in neuroblastoma a report from


a workshop on the dancing eyes syndrome at the advances
in neuroblastoma meeting in Genoa, Italy, 2004
Katherine K. Matthaya, Franz Blaesb, Barbara Heroc, Dominique Plantazd,
Pedro De Alarcone, Wendy G. Mitchellf, Michael Pikeg, Vito Pistoiah,*
a
Department of Pediatrics, University of California, San Francisco, CA, USA
b
Department of Neurology, Justus-Liebig-University, Giessen, Germany
c
Children’s Hospital, University of Cologne, Köln, Germany
d
Department of Pediatrics, CHU Grenoble, Grenoble, France
e
Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
f
Neurology Division, Children’s Hospital, Los Angeles, CA, USA
g
Department of Paediatrics, John Radcliffe Hospital, Headington, Oxford, UK
h
Laboratory of Oncology, Giannina Gaslini Children’s Hospital, Largo Gerolamo Gaslini 5, 16148 Genova, Italy
Received 27 December 2004; accepted 20 January 2005

Abstract
Opsoclonus-myoclonus syndrome (OMS) is a rare neurologic syndrome that, in children, associates with neuroblastoma in
more than half of the cases. The etiology of this condition is thought to be immune mediated, but, though immunosuppressive
therapies may ameliorate the acute symptoms, no effective treatment to prevent the common neuropsychologic sequelae has
been established. This paper summarizes the results obtained at the 2004 Advances in Neuroblastoma Research meeting,
providing status of the art information on immune pathogenesis, clinical features, acute and chronic neurologic manifestations,
current and novel therapeutic approaches. It is emphasized that, due to the rarity of OMS in general and neuroblastoma-
associated OMS in particular, international collaborations are needed to better define the pathogenesis and therapy of this
disease, propose common evaluation criteria and identify new treatment modalities.
q 2005 Elsevier Ireland Ltd. All rights reserved.

Keywords: Opsoclonus-myclonus syndrome; Neuroblastoma; Immunopathogenesis; Clinical features; Novel therapies

1. Definition

Opsoclonus-myoclonus syndrome (OMS) is a rare


paraneoplastic or possibly post-viral serious neuro-
* Corresponding author. Tel.: C39 010 56 36342; fax: C39 010 logic syndrome. In children, OMS is associated in
377 9820. more than half of cases with neuroblastoma [1,2].
E-mail address: vitopistoia@ospedale-gaslini.ge.it (V. Pistoia).

0304-3835/$ - see front matter q 2005 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.canlet.2005.01.051
276 K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282

Often called ‘dancing eye syndrome’, OMS is defined called anti-Ri can be found in the serum of the patients
by acute onset of rapid and chaotic eye movements, [14]. In contrast to adult OMS, the pediatric disease
myoclonic jerking of the limbs and trunk, ataxia, and has not been associated with anti-Ri antibodies. The
behavioral disturbance. While children with OMS and most frequently described autoimmune reactivities in
neuroblastoma most often display favorable prognos- pediatric OMS are directed against Purkinje cell
tic tumor characteristics and a high survival rate [3], cytoplasmic antigens [15]. Other authors failed to
the majority of children also experience develop- detect a common autoantigen [9], and in our own
mental delays and long-term nervous system dysfunc- investigations a reactivity against a 55 kD protein was
tion, which may be devastating [2,4–7]. the only common autoantibody in some OMS children
The etiology of this condition is thought to be (Blaes et al., unpublished results).
immune mediated, but the exact pathogenesis has not The lack of a universal autoantibody in these
yet been elucidated. Furthermore, though immuno- children led to the suspicion of a predominantly T-cell
suppressive therapies may ameliorate the acute driven autoimmune process in OMS. However, recent
symptoms, no effective treatment to prevent the work demonstrated marked B-cell activation in the
common neuropsychologic sequelae has been estab- CSF of OMS children [16]. Additionally, OMS
lished. The recent workshop at the 2004 Advances in children often show a favorable response to steroids
Neuroblastoma Research meeting was held to sum- and intravenous immunoglobulin [17], which makes
marize recent results and propose international an auto-antibody mediated disease more likely.
collaborations in order to better define the pathogen- Recently, we detected IgG surface-binding and pro-
esis and therapy of this disease, and propose common apoptotic effects of OMS-IgG to neuroblastoma cells
evaluation criteria to be used. in vitro (Blaes et al., unpublished results), which also
supports the idea of an auto-antibody-mediated
disease. Taken together, there is evidence for a
2. Immune pathogenesis of opsoclonus-myoclonus cross-reactive autoimmunity between neuroblastoma
syndrome and nervous system tissue in OMS, and the identifi-
cation of the underlying auto-antigen is an important
Since autoantibodies have been described in both goal for further research.
adult and pediatric OMS, an autoimmune hypothesis New clues to a better understanding of OMS
of the syndrome is suspected. Rare pathological immunopathogenesis have come from the histological
examinations of pediatric OMS suggest cerebellar and immunohistochemical investigation of neuroblas-
pathology; therefore, most investigators have toma tumors affecting children with associated OMS.
searched for autoantibodies against cerebellar struc- These tumors are usually ganglioneuroblastomas with
tures [8]. One cooperative study in the Children’s interstitial or perivascular lymphoid infiltrates, that
Cancer Group (CCG) compared sera from 16 patients often organize in aggregates closely resembling
with neuroblastoma-associated OMS to age and stage secondary lymphoid follicles. Although these features
matched control sera from 48 non-OMS patients with may be found also in ganglioneuroblastomas not
neuroblastoma, and showed a significantly higher associated with OMS, lymphoid infiltrates are more
proportion of children with anti-neuronal antibodies prominent in OMS-related tumors, and the number
in the OMS group (81 versus 25%, P!0.001) [9]. and size of follicles is definitely higher in the latter
The first autoantibody described was anti-neurofi- tumors [18,19]. Immunophenotyping of lymphoid
lament [10]. However, this antibody was not specifi- cells infiltrating OMS-associated ganglioneuroblasto-
cally associated with OMS and was observed even in mas showed that the follicles had typical germinal
healthy controls [11]. In some children with para- centers containing a mesh of CD21C follicular
neoplastic OMS, anti-Hu antibodies could be detected dendritic cells. CD20C B lymphocytes were the
in the serum [9,12]. The anti-Hu autoantibody is major component of the GC and the follicular mantle,
mainly associated with paraneoplastic encephalomye- where a minority of CD3C T cells were also detected.
litis and small cell lung cancer in adults [13]. In some The bulk of T lymphocytes localized around
adults with OMS and breast cancer, an autoantibody the follicles and displayed mainly a CD8C
K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282 277

immunophenotype, although also CD4C T cells were (for CXCL13) [22]. In preliminary experiments, we
observed. [18,19]. have investigated expression of these receptors in
The germinal center reaction is a T cell-dependent OMS-related tumors by immunohistochemistry.
phenomenon initiated by an antigen-activated naïve B CCR7, CXCR4 and CXCR5 were found to be
cell that migrates to a lymphoid follicle where it gives expressed on the majority of tumor cells, with higher
rise first to centroblasts and subsequently to centro- intensity on the more differentiated component.
cytes. Those centrocytes selected to survive differen- CXCR5, but not CXCR4 or CCR7, was detected on
tiate into memory B cells or plasma cells. Memory B infiltrating lymphoid cells (Raffaghello et al., unpub-
cells, in turn, can undergo multiple passages through lished results). Studies on expression of the ligands of
germinal centers to improve the affinity, and there- these receptors are in progress. These preliminary
fore, the quality, of antibody responses [20]. These results suggest that chemokine–chemokine receptor
rules apply not only to secondary follicles in lymphoid interactions are indeed involved in lymphoid recruit-
organs, but also to ectopic lymphoid follicles devel- ment into OMS-associated ganglioneuroblastomas.
oping in inflamed tissues in the course of autoimmune
disorders. Thus, detection of huge lymphoid follicles
in OMS-associated ganglioneuroblastoma may
suggest the occurrence of T cell dependent responses 3. Clinical features
to tumor-associated antigens with subsequent B cell
activation and antibody production, a hypothesis OMS of childhood is diagnosed primarily between
supported by the detection of plasma cells in the age one and 3 years (median age 18–22 months), not
interfollicular areas. significantly different than neuroblastoma as a whole
As discussed by Raffaghello and coworkers in this [2,5,23]. However, unlike neuroblastoma in general, it
supplement, one of the major mechanisms of immune is very rare before age 1. It occurs in 2–3% of all
escape used by neuroblastoma cells is the down- children diagnosed with neuroblastoma [1,2].
regulation of HLA class I molecules from the cell Although in most patients the syndrome itself leads
surface, that prevents presentation of peptides from to the diagnosis of neuroblastoma, it may rarely occur
processed tumor-associated antigens to specific after tumor resection or at relapse [24–26]. Literature
CD8C T cells. Nonetheless, since locally produced review suggests that in at least half of the children
interferon (IFN)-g might upregulate HLA class I affected, OMS is associated with underlying occult or
molecules on tumor cells, we investigated by clinically apparent neuroblastoma. In one author’s
immunohistochemistry whether neuroblasts from personal experience [WG Mitchell] of 30 patients,
patients with ganglioneuroblastoma and OMS however, more than 80% of children with OMS who
expressed these molecules. The results of these underwent extensive evaluation including thin-cut
experiments demonstrated that malignant cells were computerized tomography (CT), had neuroblastoma
completely HLA class I negative, as opposed to that in some cases had escaped detection with the
infiltrating lymphoid cells that displayed evident usual diagnostic procedures. Given this observation, it
staining [19]. Altogether, these results provide seems possible that a small undetectable neuroblastic
compelling evidence, although do not prove, that tumor or one that had already regressed completely
lymphoid infiltration is unrelated to an antigen may have occurred in the patients with ‘idiopathic’
specific T cell response directed against neuroblasts. OMS. Although an advantage of tumor resection for
An alternative hypothesis to explain tumor infiltra- ultimate neurological outcome is not proven, it often
tion by lymphoid cells is that malignant neuroblasts ameliorates the acute symptoms of OMS.
produce chemokines capable of attracting lympho- A thorough diagnostic evaluation for neuroblas-
cytes within the tumor mass [21]. Key players in toma at presentation is necessary in all patients with
lymphoid neogenesis are the chemokines CCL19, OMS, after exclusion of central nervous system
CCL21, CXCL12 and CXCL13, that interact with pathology, with brain MRI and lumbar puncture.
their cognate receptors, i.e. CCR7 (for CCL19 and This should include CT or magnetic resonance
CCL21, CXCR4 (for CXCL12) and CXCR5 imaging (MRI) with thin cuts through the adrenals if
278 K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282

appropriate, 123I-metaiodobenzylguanidine (MIBG) conjugate, non-phasic, fast, and multi-directional. It


scan [27,28], and urinary VMA, HVA, and dopamine. tends to occur in bursts and, when mild, can some-
The tumor is usually localized, very small and times be provoked by change in gaze fixation, usually
differentiated (ganglioneuroblastoma). Recent from far to near. If it is not prominent, identification of
reported series show that 90% of patients were the opsoclonus may require a period of close
without metastases at diagnosis, compared to 40– observation with elicited changes in fixation [30–32].
50% in non-OMS patients. Tumor biology, including The shaky movements of OMS are variable in
MYCN copy number and Shimada histopathologic quality ranging from tremulous polymyoclonia to
classification are usually favorable in OMS patients, coarse multi-focal jerks. They may involve all body
which correlates with the excellent survival rate found parts including extremities, trunk and face. Whilst they
in patients with OMS and neuroblastoma, even when may be persistent, they are more often exacerbated by
survival analysis is stratified by age at diagnosis and attempts at movement and by emotional distress.
stage [2,3,5]. Personality change and developmental regression
Although death from tumor progression is rare typically occur involving loss of speech and language
in this condition, late neurological deficits are in addition to the obvious motor deficits. Severe
frequent, seen in 70–80% of children affected irritability and sleep disturbance occur in almost all
by OMS, long after the acute opsoclonus and children and provide a useful distinguishing feature
myoclonus have resolved. Several detailed studies from other acquired ataxias. The typical child with
with follow-up times of 5–20 years have shown OMS appears extremely uncomfortable, often pre-
persistent neurologic deficits in more than 70% of senting in the office or hospital screaming incon-
patients in speech, coordination, and, occasionally, solably and being passed back and forth between
eye movements. More than half the patients displayed distraught parents who find that they cannot put the
deficits on detailed testing in cognition, adaptive child down without exacerbating the distress. Vomit-
behavior, and development, ranging from mild to ing occurs in some children, often associated with
severe [2,4–7,23,29]. changes in position [6,33,34]. Children with OMS in
whom irritability is less prominent may present with
apathy, loss of social interaction and unwillingness to
4. Acute neurologic findings of OMS engage in age-appropriate play.
Very occasionally occult neuroblastoma may
Children with OMS with or without neuroblastoma present with some, but not all, of the features
present with an acute or sub-acute onset of ataxia, described [6,35]. Nor is every case of childhood
losing the ability to walk and/ or sit over a period of OMS paraneoplastic in origin. Given this element of
one to several days - occasionally weeks. The variability diagnostic criteria have been proposed—
movement disorder is accompanied by severe irrit- the presence of at least 3 out of the following four
ability and, at some point, by opsoclonus. The first features: (1) opsoclonus, (2) myoclonus/ataxia,
neurological diagnosis proposed for most children (3) behavioral change and/or sleep disturbance,
who are ultimately found to have OMS is almost (4) neuroblastoma.
invariably post-infectious acute cerebellar ataxia of
childhood. In fact, until opsoclonus appears, the
conditions are clinically similar, and acute cerebellar 5. Chronic neurological findings
ataxia of childhood is far more common. While
opsoclonus is occasionally seen in the first day or two With or without treatment, opsoclonus usually
of OMS, it may appear as late as a few weeks after the resolves eventually. Residual opsoclonus may
onset of motor symptoms. reappear after apparent complete resolution with
Opsoclonus is a distinctive eye movement reduction in medication dose, or with intercurrent
rarely seen in other childhood conditions, and is illnesses. Minimal opsoclonus may be elicited by
dissimilar to nystagmus, which is more often seen in having the child repeatedly re-fixate from near to far, or
other acute cerebellar ataxias. Opsoclonus is by having them partially squeeze the eye-lids shut, then
K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282 279

allow them to be partly opened by the examiner. Even outcome in patients with OMS. Published results are
with resolution of opsoclonus, abnormalities of smooth sparse and report only on small patient numbers
pursuit eye movements and hypometric saccades are ranging from 10 to 29 patients per series [2,4–7,23,29,
commonly seen even years after treatment. 36]. Current evidence suggests that the disease is
Motor deficits generally remain significant without mediated by an immune response directed against the
immunosuppressive treatment (and may be proble- tumor and cross-reacting with central neuronal
matic in spite of treatment). With treatment, children tissues, particularly cerebellum. Therefore, treatment
with OMS usually walk independently, but even with of OMS has been with immunosuppressive therapy,
vigorous treatment they are usually not as coordinated primarily with either long term corticosteroids or
as their peers throughout childhood and adolescence. ACTH. Although acute symptoms may be relieved by
Speech and language are also significantly affected steroid treatment, exacerbations often occur with
by this condition. Most children who have had OMS weaning of the drugs or intercurrent viral illness.
continue to show expressive language and speech Intravenous application of immunoglobulin has been
deficits. Receptive language tends to be better than used in some children with success, either in
expressive language. When all OMS symptoms are combination with steroids or alone [38–40]. Other
controlled with immunosuppressive medication, some immunomodulating therapies, including cyclopho-
children will experience catch-up in their speech and sphamide, azathioprine and plasmapheresis [41,42],
language skills, but most will be left with residual have been reported to occasionally be effective in the
learning and language disabilities and will require treatment of acute symptoms of OMS. Resection
educational assistance and speech therapy well into their of the neuroblastoma often improves the acute
elementary school years, often longer [2,4–7,29,36,37]. symptoms, but only rarely prevents neurologic
As patients with OMS reach their teen years, sequelae [4,6,36].
cognitive deficits may become more evident with Despite immunosuppressive treatment for the
increasingly difficult academic tasks. Adequate pre- acute symptoms of OMS, many children have
vocational planning and support is essential for significant neurological and developmental deficits
progression into successful and independent function- [2,4–7,23,29]. Therefore, retrospective analyses have
ing in young adulthood. Behavior problems including been performed to see if any particular therapy helped
aggressive and impulsive behavior, mood disorders to decrease the incidence of these devastating late
and sleep disturbances may persist indefinitely, with sequelae. Although in most patients, surgical resec-
some OMS patients manifesting night terrors well past tion was the only anti-tumor treatment necessary due
the toddler years. Better overall control of OMS to the frequency of low stage tumors, some patients
symptoms with immunosuppressive medication received chemotherapy as part of their treatment.
sometimes produces improvement in behavioral Retrospective analysis of 29 patients with OMS in
symptoms as well as improving motor function. Pediatric Oncology Group (POG) trials showed a
However, psychotropic medications may be necessary significant benefit for resolution of symptoms in
in selected children who have OMS sequelae with patients who where treated with chemotherapy [5]. On
severe behavioral or sleep disturbances. There is some the other hand, neither the Children’s Cancer Group
evidence that even very late treatment with corticos- study of 19 evaluable patients nor a French study of
teroids, corticotropin or other immunosuppression, 16 evaluable patients was able to show a significant
years after onset, will improve motor function. decrease in late neurologic problems for those
However, cognitive deficits, particularly when long- children treated with chemotherapy [2,23]. However,
standing, do not generally reverse. the combination of the 64 patients from these
cooperative group series showed that only 12/25
patients (48%) treated with chemotherapy had
6. Treatment approaches neurologic sequelae, compared to 32/39 (82%) who
did not receive chemotherapy [2,5,23]. It remains
So far, no prospective trial has been carried out to unclear whether this benefit is due to a positive effect
address treatment and its correlation with long-term of chemotherapy or due to less severe activation of
280 K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282

the immune system in patients with more advanced behavior and cognition and the regular cognitive
stages of the tumor. Similarly, the role of intravenous assessment in the follow up of patients, so that the
immunoglobulin in the treatment of OMS is not clear. comparison of the long-term results of both studies
In the past immunoglobulin may have been used in will be feasible in the future.
patients with severe symptoms or with problems when
weaning of steroids, thus impeding interpretation of
retrospective results. 8. New approaches

The current response to steroid therapy, immune


7. Current approaches globulin therapy and chemotherapy as well as the
presence of anti neural tissue antibodies in patients
The poor neurologic outcome despite steroid with neuroblastoma associated OMS strongly support
treatment and the possible positive effect seen after investigation of other immunosuppressive approaches
chemotherapeutic intervention led to the current to therapy in patients who fail traditional therapy. A
hypothesis that prompt and strong immunosuppres- single patient with this syndrome with severe ataxia
sion is necessary in the treatment of OMS. Because of who had failed to respond to therapy with ACTH,
the rarity of the disease and the difficulties in intravenous immune globulin and azathioprine,
interpretation of retrospective analyses, prospective underwent plasmapheresis in combination with ster-
cooperative trials have been initiated, with inter- oid and azathioprine [41]. Within 1 week of therapy
national collaboration. the subject had improved and 6 months later was able
The COG has opened a Phase III study for patients to ambulate without support and attended school. In
with OMS and neuroblastoma. All patients will the series of patients from the POG, two patients were
receive prednisone and will be randomized to either treated with azathioprine without full resolution of
receive intravenous immunoglobulin or not. Patients their symptoms [5]. High dose steroids (30 mg/kg/day
with low risk neuroblastoma who would otherwise not or methylprednisolone for 10 days) have been used
receive chemotherapy will receive immunosuppres- successfully in a patient for whom therapy with
sion with cyclophosphamide, while patients with prednisone was ineffective [43]. Pranzatelli and co-
intermediate or high-risk neuroblastoma will receive workers have documented the presence of activated T
chemotherapy according the appropriate trial. Stan- and B lymphocytes in the cerebrospinal fluid of
dardized neurocognitive, developmental and beha- subjects with OMS, in both idiopathic or neuroblas-
vioral testing will be performed at specified intervals, toma-associated cases [16]. Rituximab is a humanized
and further investigation of the pathogenesis of the monoclonal antibody against CD20. It has been used
syndrome will be assessed with tumor and cerebrosp- for therapy of autoimmune disorders as well as CD20
inal fluid biology studies, anti-neuronal antibodies positive non-Hodgkin lymphoma [44,45]. Based on
and blood lymphocyte studies, and serial brain MRI the expansion of B cells reported in the CSF of some
scans to evaluate anatomic changes. children with OMS, rituximab is being tested in pilot
In the European countries a study is currently studies for children unresponsive to steroid therapy. It
planned for patients with OMS with or without the is important that these alternative therapies be utilized
diagnosis of neuroblastoma with stepwise intensifica- in a systematic and prospective study so that we can
tion of treatment, starting with dexamethasone pulse evaluate their role and not be left with anecdotal
therapy, adding first intravenous immunoglobulin and experience as it is now the case for much of the
second immunosuppression with cyclophosphamide therapy for OMS.
for patients with insufficient response. This study will
also incorporate similar biologic and neurocognitive
testing. Acknowledgements
Both studies plan a systematic registration of
symptoms with a common, newly elaborated scoring The idea of organizing a workshop devoted to the
system for neurologic exam, opsoclonus, myoclonus, Dancing Eye/Opsoclonus Myoclonus Syndrome in
K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282 281

the frame of the 11th Conference ‘Advances in [11] K. Stefansson, L.S. Marton, M.E. Dieperink, G.K. Molnar,
Neuroblastoma Research’ (Genova, Italy, June 16– W.W. Schlaepfer, C.M. Helgason, Circulating autoantibodies
to the 200,000-dalton protein of neurofilaments in the serum of
19, 2004) came from the Second Workshop on
healthy individuals, Science 228 (1985) 1117–1119.
Dancing Eye Syndrome, Clinical and Basic Science, [12] P.G. Fisher, D.S. Wechsler, H.S. Singer, Anti-Hu antibody in a
held in Abingdon, UK (February 6th–8th, 2003) and neuroblastoma-associated paraneoplastic syndrome, Pediatr.
sponsored by The Dancing Eye Syndrome Support Neurol. 10 (1994) 309–312.
Trust. The Authors are grateful to this charity that [13] F. Blaes, Immunotherapeutic approaches to paraneoplastic
undertook the brave effort of bringing together basic neurological disorders, Expert Opin. Biol. Ther. 2 (2002) 419–
430.
and clinical scientists in the field and providing them
[14] N.E. Anderson, C. Budde-Steffen, M.K. Rosenblum, F. Graus,
with an unique opportunity of informal and in depth D. Ford, B.J. Synek, J.B. Posner, Opsoclonus, myoclonus,
discussion. ataxia, and encephalopathy in adults with cancer: a distinct
paraneoplastic syndrome, Medicine (Baltimore) 67 (1988)
100–109.
[15] A.M. Connolly, A. Pestronk, S. Mehta, M.R. Pranzatelli III,
References M.J. Noetzel, Serum autoantibodies in childhood opsoclonus-
myoclonus syndrome: an analysis of antigenic targets in neural
[1] M.R. Pranzatelli, The neurobiology of the opsoclonus- tissues, J. Pediatr. 130 (1997) 878–884.
myoclonus syndrome, Clin. Neuropharmacol. 15 (1992) [16] M.R. Pranzatelli III, A.L. Travelstead, E.D. Tate, T.J. Allison,
186–228. E.J. Moticka, D.N. Franz, et al., B- and T-cell markers in
[2] E. Rudnick, Y. Khakoo, N.L. Antunes, R.C. Seeger, opsoclonus-myoclonus syndrome: immunophenotyping of
G.M. Brodeur, H. Shimada, et al., Opsoclonus-myoclonus- CSF lymphocytes, Neurology 62 (2004) 1526–1532.
ataxia syndrome in neuroblastoma: clinical outcome and [17] M.R. Pranzatelli III, The immunopharmacology of the
antineuronal antibodies-a report from the Children’s Cancer opsoclonus-myoclonus syndrome, Clin. Neuropharmacol. 19
Group Study, Med. Pediatr. Oncol. 36 (2001) 612–622. (1996) 1–47.
[3] A.J. Altman, R.L. Baehner, favorable prognosis for survival in [18] R. Cooper, Y. Khakoo, K.K. Matthay, J.N. Lukens,
children with coincident opso-myoclonus and neuroblastoma, R.C. Seeger, D.O. Stram, et al., Opsoclonus-myoclonus-ataxia
Cancer 37 (1976) 846–852. syndrome in neuroblastoma: histopathologic features-a report
[4] P.S. Koh, J.G. Raffensperger, S. Berry, M.B. Larsen, from the Children’s Cancer Group, Med. Pediatr. Oncol. 36
H.S. Johnstone, P. Chou, et al., Long-term outcome in (2001) 623–629.
children with opsoclonus-myoclonus and ataxia and coinci- [19] C. Gambini, M. Conte, G. Bernini, P. Angelini, A. Pession,
dent neuroblastoma, J. Pediatr. 125 (1994) 712–716. P. Paolucci, et al., Neuroblastic tumors associated with
[5] C. Russo, S.L. Cohn, M.J. Petruzzi, P.A. de Alarcon, Long- opsoclonus-myoclonus syndrome: histological, immunohisto-
term neurologic outcome in children with opsoclonus- chemical and molecular features of 15 Italian cases, Virchows
myoclonus associated with neuroblastoma: a report from the
Arch. 442 (2003) 555–562.
Pediatric Oncology Group, Med. Ped. Oncol. 28 (1997) 284–
[20] I.C. MacLennan, Germinal centers, Annu. Rev. Immunol. 12
288.
(1994) 117–139.
[6] W.G. Mitchell, Y. Davalos-Gonzalez, V.L. Brumm,
[21] L.S. Metelitsa, H.W. Wu, H. Wang, Y. Yang, Z. Warsi,
S.K. Aller, E. Burger, S.B. Turkel, et al., Opsoclonus-ataxia
S. Asgharzadeh, et al., Natural killer T cells infiltrate
caused by childhood neuroblastoma: developmental and
neuroblastomas expressing the chemokine CCL2, J. Exp.
neurologic sequelae, Pediatrics 109 (2002) 86–98.
Med. 199 (2004) 1213–1221.
[7] K. Hayward, R.J. Jeremy, S. Jenkins, A.J. Barkovich,
S.H. Gultekin, J. Kramer, et al., Long-term neurobehavioral [22] P. Hjelmstrom, Lymphoid neogenesis: de novo formation of
outcomes in children with neuroblastoma and opsoclonus- lymphoid tissue in chronic inflammation through expression of
myoclonus-ataxia syndrome: relationship to MRI findings and homing chemokines, J. Leukoc. Biol. 69 (2001) 331–339.
anti-neuronal antibodies,, J. Pediatr. 139 (2001) 552–559. [23] D. Plantaz, J. Michon, D. Valteau-Couanet, C. Coze,
[8] F.A. Ziter, P.F. Bray, P.A. Cancilla, Neuropathologic findings P. Chastagner, C. Bergeron, et al., Opsoclonus-myoclonus
in a patient with neuroblastoma and myoclonic encephalo- syndrome associated with non-metastatic neuroblastoma.
pathy, Arch. Neurol. 36 (1979) 51. Long-term survival. Study of the French Society of Pediatric
[9] N.L. Antunes, Y. Khakoo, K.K. Matthay, R.C. Seeger, Oncologists, Arch. Pediatr. 7 (2000) 621–628.
D.O. Stram, E. Gerstner, et al., Antineuronal antibodies in [24] C. Delalieux, C. Ebinger, R. Maurus, H. Sliwowski, Letter:
patients with neuroblastoma and paraneoplastic opsoclonus- myoclonic encephalopathy and neuroblastoma, N. Engl.
myoclonus, J. Pediatr. Hematol. Oncol. 22 (2000) 315–320. J. Med. 292 (1975) 46–47.
[10] M.J. Noetzel, L.P. Cawley, V.L. James, B.J. Minard, [25] L. Rupprecht, W. Mortier, Cerebellar motor disorder and
H.C. Agrawal, Anti-neurofilament protein antibodies in neuroblastoma, Monatsschr. Kinderheilkd. 123 (1975)
opsoclonus-myoclonus, J. Neuroimmunol. 15 (1987) 137–145. 392–393.
282 K.K. Matthay et al. / Cancer Letters 228 (2005) 275–282

[26] W.T. Basco Jr., M. Abboud, K.R. Holden, Opsoclonus- [36] M.S. Hammer, M.B. Larsen, C.V. Stack, Outcome of children
myoclonus and recurrent neuroblastoma, J. Pediatr. 126 with opsoclonus-myoclonus regardless of etiology, Pediatr.
(1995) 847–848. Neurol. 13 (1995) 21–24.
[27] J.F. Swart, J. de Kraker, N. van der Lely, Metaiodobenzyl- [37] K.R. Pohl, J. Pritchard, J. Wilson, Neurological sequelae of the
guanidine total-body scintigraphy required for revealing dancing eye syndrome, Eur. J. Pediatr. 155 (1996) 237–244.
occult neuroblastoma in opsoclonus-myoclonus syndrome, [38] M.J. Petruzzi, P.A. de Alarcon, Neuroblastoma-associated
Eur. J. Pediatr. 161 (2002) 255–258. opsoclonus-myoclonus treated with intravenously adminis-
[28] M.T. Parisi, R.S. Hattner, K.K. Matthay, B.O. Berg, tered immune globulin G, J. Pediatr. 127 (1995) 328–329.
E.D. Sandler, Optimized diagnostic strategy for neuroblastoma [39] C. Borgna-Pignatti, R. Balter, P. Marradi, V. Colamaria,
in opsoclonus-myoclonus, J. Nucl. Med. 34 (1993) 1922–1926. Treatment with intravenously administered immunoglobulins
[29] P.H. Papero, M.R. Pranzatelli, L.J. Margolis, E. Tate, of the neuroblastoma-associated opsoclonus-myoclonus,
L.A. Wilson, P. Glass, Neurobehavioral and psychosocial J. Pediatr. 129 (1996) 179–180.
functioning of children with opsoclonus-myoclonus syn-
[40] E. Veneselli, M. Conte, R. Biancheri, A. Acquaviva, B. De
drome, Dev. Med. Child. Neurol. 37 (1995) 915–932.
Bernardi, Effect of steroid and high-dose immunoglobulin
[30] H.E. Brissaud, P. Beauvais, Opsoclonus and neuroblastoma,
therapy on opsoclonus-myoclonus syndrome occurring in
N. Engl. J. Med. 280 (1969) 1242.
neuroblastoma, Med. Pediatr. Oncol. 30 (1998) 15–17.
[31] E. Boltshauser, T. Deonna, H.R. Hirt, Myoclonic encephalo-
[41] V.W. Yiu, T. Kovithavongs, L.F. McGonigle, P. Ferreira,
pathy of infants or ‘dancing eyes syndrome’. Report of 7 cases
Plasmapheresis as an effective treatment for opsoclonus-
with long-term follow-up and review of the literature (cases
with and without neuroblastoma), Helv. Paediatr. Acta 34 myoclonus syndrome, Pediatr. Neurol. 24 (2001) 72–74.
(1979) 119–133. [42] S.R. Sheela, P.J. Mani, Opsoclonus myoclonus syndrome:
[32] J. Blake, C. Fitzpatrick, Eye signs in neuroblastoma, Trans. response to plasmapheresis, Indian Pediatr. 41 (2004) 499–
Ophthalmol. Soc. UK 92 (1972) 825–833. 502.
[33] P.G. Moe, G. Nellhaus, Infantile polymyoclonia-opsoclonus [43] W.L. Yeung, C.K. Li, E.A. Nelson, K.W. Chik, G.M. Joynt,
syndrome and neural crest tumors, Neurology 20 (1970) 756– E. Yuen, C.K. Yeung, Unusual neurological presentation of
764. neuroblastoma, Hong Kong Med. J. 9 (2003) 142–144.
[34] W.G. Mitchell, S.R. Snodgrass, Opsoclonus-ataxia due to [44] B. Coiffier, New treatment strategies in lymphomas: aggres-
childhood neural crest tumors: a chronic neurologic syndrome, sive lymphomas, Ann. Hematol. 83 (Suppl. 1) (2004) S73–
J. Child Neurol. 5 (1990) 153–158. S74.
[35] K.B. Roberts, Cerebellar ataxia and ‘occult neuroblastoma’ [45] L. Virgolini, V. Marzocchi, Rituximab in autoimmune
without opsoclonus, Pediatrics 56 (1975) 464–465. diseases, Biomed. Pharmacother. 58 (2004) 299–309.

You might also like