Open Access Mini Review

Childhood-Onset Myasthenia Gravis, a Troublesome Clinical Entity with Heterogeneity in China

Yayun Cao and Bitao Bu*

Department of Neurology, China

Corresponding Author

Received Date:Jaunary 03, 2018;  Published Date: January 11, 2019

Abstract

Myasthenia gravis (MG) is a well-known autoimmune disease involving neuromuscular transmission leading to fluctuating muscle weakness and fatigability. With the advent of immunotherapy, majority of MG sufferers will have a good outcome. However, the childhood onset MG (CMG) in China is quite different from that reported in the Western countries in light of onset age distribution, clinical types, responsiveness to immunotherapy and the long-term outcome. The mini-review focuses on the differences of Chinese CMG patients from European cases.

Keywords: myasthenia gravis; Child; Juvenile; Heterogeneity; Chinese; Dug-resistance

Introduction

Myasthenia gravis (MG) is a T cell dependent, antibodymediated neuromuscular junction disease with complement system activation [1], often presenting with muscle weakness and fatigability of affected muscles. The distribution of affected muscles varies significantly from case to case, and the same regarding the onset ages. Some studies reported the bimodal distribution of ages at onset in the Western countries, with a peak of onset 20-40 years old in females and 50-60 years old in males [2,3]. However in China, the disease tends to more frequently affect children than adults [4- 6]. In literatures, the term, juvenile MG (JMG) is frequently used to describe the disease happened before age of 19 years [7], which are subdivided into prepubertal and postpubertal patients, according to occurrence of the first symptoms before or after puberty [8]. Childhood onset myasthenia gravis (CMG) depicts the condition that MG patients who develop the disease before age of 14 years and accounted for more than half of MG population in China and harbored unique features different from the cases of JMG in the Western countries or the ones of adult onset myasthenia gravis (AMG). The following paragraphs focus on the research concerning the CMG.

Demographic Differences

The estimated incidence of MG ranges from 15 to 179/1,000,000/year in the Western countries [9,10], of which the incidence of JMG is between 1 and 5/1,000,000/year accounting for 10–15% of all MG population [10-14]. The onset age distribution in Western JMG population has one peak in postpubertal, aged from 12 to 18 years, accounting for 66-90% [12,15]. Unfortunately, the epidemiology of MG is generally lacking in China, and a survey in Yiyang county in Hunan Province in 1986 discovered 60 MG patients in 590,000 residents (incidence was 10/100,000), with CMG 48 cases (80%) [16]. The Chinese hospital-based reports stated that CMG patients were estimated to account for more than 50% of the MG population, with a peak of onset age before 10 years, mainly aged 2-5 years [4-6,17].

Distinct Features

In the Western countries, the prepubertal patients have distinctive clinical characteristics compared with the postpubertal cases, who shared more features with AMG, such as presentations, transformation to the generalized types, immunological profiles, thymic abnormalities and responses to therapy [8,18-22].

Presentations and transformation

In prepubertal MG children, similar to late-onset AMG (LOMG) cases, no sex predominance has been observed [23]. While the postpubertal MG children had a predilection for females, just like early-onset AMG (EOMG) patients [24,25]. As reported, the prepubertal children are more likely to present with isolated ocular symptoms compared with the overwhelming generalized presentations among the postpubertal children and AMG patients [18-20]. Out of those prepubertal cases with initial ocular presentations, 50-60% would develop into generalized muscle weakness within 2 years, similar to the patients of AMG [22,26]. But in a sharp contrast, the Chinese CMG children only present with ocular type in as high as 71-95% of the CMG population with a pretty low rate of transformation to generalized types [6,27]. The conversion rate of CMG was significantly lower than that of AMG in our retrospective studies over 10 years follow-up (12.4% vs. 32.5%) [6], and much lower than that of JMG or AMG in the Western countries (50-60% and 80%, respectively) [22,28]. Interestingly, a cohort of study in Thailand disclosed the similar results in the JMG patients [29]. Though the risk factors for transformation have not been clearly pulled out yet [6,30], the long disease duration and onset age older than 10 years were reported to be associated with the transformation [31].

Immunological profiles

50-71% of the prepubertal children were AChR antibodies positive when compared with 80-90% seropositivity among the postpubertal or adult patients [15,18,20]. Numerous studies have demonstrated a high percentage of seronegativity among CMG children, especially in the prepubertal or those with isolated ocular symptoms [7,18-20,32-34]. MuSK-positive cases were very rare in seronegative children [13,35], while 40-50% of AMG patients who were negative for AChR antibodies had elevated titers of antibodies against MuSK [36]. Other antibodies profiles were not routinely determined in the literatures, such as LRP4, Titin, etc, in CMG patients. The possibility of other neuromuscular diseases mimicking CMG had been thoroughly investigated and ruled out, including the congenital myasthenic syndromes, mitochondrial myopathies, congenital myopathies or pure Graves disease in the CMG children.

Thymus abnormalities and thymectomy

Thymus abnormalities in CMG and JMG around the world are similar, of which thymic hyperplasia is the most common one, accounting for 73-89%, while thymoma is particularly rare, accounting for 0-3.8% [4,6,7,15,19,37,38]. However, in AMG patients, thymic hyperplasia is common in EOMG (46.6-80%), and thymoma is frequently observed in LOMG (50-75%) [1,39-41]. Indications for thymectomy are less clear for prepubertal JMG, and for seronegative children and the ones with isolated ocular symptoms. There are concerns that thymectomy performed at a young age may result in premature immunosenescence [42]. Nonetheless, thymectomy constitutes an important option in children if the pharmaceutical treatment proves unsatisfactory. According to a wide consensus, thymectomy is indicated in seropositive postpubertal children with moderate to severe generalized symptoms [43]. Thymectomy is mandatory if thymoma is present, but this situation is rare in children. The benefit of thymectomy to the children remains controversial. Popperud et al reported the efficacy of thymecotomy in the prepubertal onset patients who presented severe generalized symptoms or even frequent myasthenic crisis [22]. Similarly, Heng et al considered the trans-sternal thymectomy was effective in improving the outcome of 24 severe generalized children with positive AchR-ab in the United Kingdom [44].

On the contrary, we have performed a trans-sternal thymectomy in a cohort of 34 CMG patients (27 ocular type cases and 7 generalized type cases) who failed to respond well to drug therapy and have found thymoma in 6 cases and hyperplasia in 28 patients. But unfortunately, the general outcome of the cohort did not improve significantly, only slight improvement in the 7 generalized CMG patients [6]. The results may hint at the difference of CMG with isolated ocular type from those with generalized MG types.

Refractory cases

At present, the definition of refractory MG is not definitely delineated [45]. Although related researches are few, we have found that about 20% of ocular CMG patients in our database were resistant to drug therapy [6]. The longer follow-up observation of the Chinese CMG children disclosed that the majority of the cases had a tendency to relapse (over 50%) after they had achieved the status of being symptom-free for months or years. Slowly, the relapsing-remising cases could become resistance to oral corticosteroids and pyridostigmine [6]. The resistant or refractory cases would have troublesome symptoms such as symmetric or asymmetric ptosis, strabismus and amblyopia, which profoundly compromise the sufferers’ social and personal activities and social-psychological behavior as well. It is urgent for physicians to improve their symptoms. All the treatment of CMG has largely been extrapolated from adults’ data, so for refractory children, there is no clear guidance yet. Thymectomy may be beneficial to the CMG cases with generalized types, but no significant efficacy was observed in the cases with isolated ocular symptoms [38,43,46]. FK506 was initially tried to significantly improve the symptoms unresponsive to oral corticosteroids and pyridostigmine and had achieved satisfactory improvement in majority of the CMG cases who had the troublesome ocular symptoms [47,48]. Rituximab was occasionally used to treat both the intractable children and adults with generalized types and showed efficacy [49-52], but the data in CMG patients with sole isolated ocular symptoms is lacking. Based on the pathogenesis of autoimmunity, seeking more accurate novel immunosuppressive agents and biomarkers which may actually represent the severity and activity of the disease are needed in the refractory children.

Response to therapy and prognosis

There have been no large-scale prospective studies on the longterm outcome of CMG population all over the world except for some cross-sectional studies or retrospective studies [6,22,53-57]. Thus, the disease course and prognosis of MG children remained poorly defined. Even though spontaneous remission was reported to be high in children (15-34.7%) [19,58], especially in the prepubertal sufferers [18], or sole administration of acetylcholineesterase inhibitors may achieve satisfactory prognosis [53], the nature course has not been clearly depicted. Corticosteroids are the firstline immunosuppression therapy in treating MG, and both the ocular and generalized MG usually respond well to steroids. In order to avoid the side effects of long-term use of corticosteroids or to further improve the symptoms, cytotoxic agents, such as azathioprine, cyclosporine, mycophenolate mofetil, tacrolimus, as steroid-sparing or alternative therapy would be probably added on and achieved improvement in some case [7,38,54,59].

Most studies suggested a high remission rate after thymectomy in JMG [18,44,60,61], especially in postpubertal cases (29-68%) [38,56,62,63], similar to AMG population. Whereas the data about thymectomy in prepubertal was scarce [21]. In China, CMG children showed a good response to acetylcholineesterase inhibitors and immunotherapy initially, but majority of the patients would develop acquired drug-resistance or take a remising-relapsing course. These CMG children may relapse after they had achieved a complete clinical stable remission or pharmaceutical remission for years [6]. After multiple relapses, the symptoms will fail to respond to the therapy again (acquired drug-resistance) with unknown pathogenesis. Thus, the relapses after remission and acquired resistance to corticosteroids constitute the major concern in Chinese CMG populations.

Multidisciplinary Management

The ocular symptoms, such as symmetric or asymmetric ptosis, strabismus, diplopia or eyeball fixation, etc., with poor therapeutic efficacy would persist in a considerable group of patients with or without fluctuation and immensely affect the sufferers’ facial appearance, social activity and psychological status [19]. What is more, the chronic side effects of steroids such as growth-stunting and weight gain, would superimpose the MG symptoms and aftereffects [19]. In dealing with the sufferers with the intractable MG symptoms, it should be emphasized that multidisciplinary cooperation including neurologists, pediatricians, psychotherapists, and ophthalmologists is needed to improve the outcome of the CMG patients [20].

The Possible Reasons for the Chinese CMG

Up to now, the reasons for the distinctive Chinese autoimmune CMG patients remain elusive, although the CMG-mimics have been ruled out. Genetic background probably constitutes the cardinal factors [31,64]. HLA DRw9 and DRw13 were strongly associated with Japanese MG [31], Bw46DR9 with Chinese [27,65], DQ8 and DR3 with Caucasian, and DR5 with African descents [66]. Besides, environmental factors may play as a trigger to activate the autoimmunity by way of molecular mimicry, supra-antigen or by-stander mechanism [17]. In all, CMG is a clinical entity with worldwide heterogeneity concerning the clinical presentations and the responses to therapy. The Chinese CMG presents unique predominant ocular symptoms which harbor the remisingrelapsing course and acquired resistance to corticotherapy. More attention should be paid to elucidate the reasons why the CMG is likely to affect the Chinese and to investigate more accurate therapeutic methods to improve the ocular symptoms.

Acknowledgemnet

None.

Conflict of Interest

No conflict of interest.

References

  1. Berrih-Aknin S, Le Panse R (2014) Myasthenia gravis: a comprehensive review of immune dysregulation and etiological mechanisms. J Autoimmun 52: 90-100.
  2. Poulas K, Tsibri E, Kokla A, Papanastasiou D, Tsouloufis T, et al. (2001) Epidemiology of seropositive myasthenia gravis in Greece. J Neurol Neurosurg Psychiatry 71(3): 352-356.
  3. Thanvi B, Lo T (2004) Update on myasthenia gravis. Postgrad Med J 80(950): 690-700.
  4. Huang X, Liu WB, Men LN, Feng HY, Li Y, et al. (2013) Clinical features of myasthenia gravis in southern China: a retrospective review of 2,154 cases over 22 years. Neurol Sci 34(6): 911-917.
  5. Zhang X, Yang M, Xu J, Zhang M, Lang B, et al. (2007) Clinical and serological study of myasthenia gravis in HuBei Province, China. J Neurol Neurosurg Psychiatry 78(4): 386-390.
  6. Gui M, Luo X, Lin J, Li Y, Zhang M, et al. (2015) Long-term outcome of 424 childhood-onset myasthenia gravis patients. J Neurol 262(4): 823-830.
  7. Andrews PI (2004) Autoimmune myasthenia gravis in childhood. Semin Neurol 24 (1): 101-110.
  8. Della MA, Trippe H, Lutz S, Schara U (2014) Juvenile myasthenia gravis: recommendations for diagnostic approaches and treatment. Neuropediatrics 45(2): 75-83.
  9. Aisling S Carr, Chris R Cardwell, Peter O McCarron, John McConville (2010) A systematic review of population based epidemiological studies in Myasthenia Gravis. BMC Neurol 10: 46.
  10. McGrogan A, Sneddon S, de Vries CS (2010) The incidence of myasthenia gravis: a systematic literature review. Neuroepidemiology 34(3): 171- 183.
  11. Phillips LH (2004) The epidemiology of myasthenia gravis. Semin Neurol 24 (01): 17-20.
  12. Phillips LN, Torner JC, Anderson MS, Cox GM (1992) The epidemiology of myasthenia gravis in central and western Virginia. Neurology 42(10): 1888-1893.
  13. VanderPluym J, Vajsar J, Jacob FD, Mah JK, Grenier D, et al. (2013) Clinical characteristics of pediatric myasthenia: a surveillance study. Pediatrics 132 (4): e939-944.
  14. Matuja WB, Aris EA, Gabone J, Mgaya EM (2001) Incidence and characteristics of Myasthenia gravis in Dar Es Salaam, Tanzania. East Afr Med J 78(9): 473-476.
  15. Evoli A, Batocchi AP, Bartoccioni E, Lino MM, Minisci C, et al. (1998) Juvenile myasthenia gravis with prepubertal onset. Neuromuscul Disord 8(8): 561-567.
  16. You DY, Xu DD (1988) Epidemiological Survey of Myasthenia Gravis in 590,000 Population. Chin J Nerv Ment Dis (4).
  17. He D, Zhang H, Xiao J, Zhang X, Xie M, et al. (2018) Molecular and clinical relationship between live-attenuated Japanese encephalitis vaccination and childhood onset myasthenia gravis. Ann Neurol 84(3): 386-400.
  18. Andrews PI, Massey JM, Howard JF, Sanders DB (1994) Race, sex, and puberty influence onset, severity, and outcome in juvenile myasthenia gravis. Neurology 44(7): 1208-1214.
  19. Chiang LM, Darras BT, Kang PB (2009) Juvenile myasthenia gravis. Muscle Nerve 39(4): 423-431.
  20. Finnis MF, Jayawant S (2011) Juvenile myasthenia gravis: a paediatric perspective. Autoimmune Diseases 2011: 404101.
  21. Ionita CM, Acsadi G (2013) Management of Juvenile Myasthenia Gravis. Pediatr Neurol 48(2): 95-104.
  22. Popperud TH, Boldingh MI, Rasmussen M, Kerty E (2017) Juvenile myasthenia gravis in Norway: Clinical characteristics, treatment, and long-term outcome in a nationwide population-based cohort. Eur J Paediatr Neurol 21(5): 707-714.
  23. Haliloglu G, Anlar B, Aysun S, Topcu M, Topaloglu H, et al. (2002) Gender prevalence in childhood multiple sclerosis and myasthenia gravis. J Child Neurol 17(5): 390-392.
  24. Evoli A (2010) Acquired myasthenia gravis in childhood. Curr Opin Neurol 23(5): 536-540.
  25. Chiu HC, Vincent A, Newsom Davis J, Hsieh KH, Hung T (1987) Myasthenia gravis: population differences in disease expression and acetylcholine receptor antibody titers between Chinese and Caucasians. Neurology 37(12): 1854-1857.
  26. Evoli A, Tonali P, Bartoccioni E, Lo MM (1988) Ocular myasthenia: diagnostic and therapeutic problems. Acta Neurol Scand 77(1): 31-35.
  27. Wong V, Hawkins BR, Yu YL (1992) Myasthenia gravis in Hong Kong Chinese. 2. Paediatric disease. Acta Neurol Scand 86(1): 68-72.
  28. Kalb B, Matell G, Pirskanen R, Lambe M (2002) Epidemiology of myasthenia gravis: a population-based study in Stockholm, Sweden. Neuroepidemiology 21(5): 221-225.
  29. Vanikieti K, Lowwongngam K, Padungkiatsagul T, Visudtibhan A, Poonyathalang A (2018) Juvenile Ocular Myasthenia Gravis: Presentation and Outcome of a Large Cohort. Pediatr Neurol 87: 36-41.
  30. Luchanok U, Kaminski HJ (2008) Ocular myasthenia: diagnostic and treatment recommendations and the evidence base. Curr Opin Neurol 21(1): 8-15.
  31. Shinomiya N, Nomura Y, Segawa M (2004) A variant of childhood-onset myasthenia gravis: HLA typing and clinical characteristics in Japan. Clin Immunol 110(2) :154-158.
  32. Andrews PI, Massey JM, Sanders DB (1993) Acetylcholine receptor antibodies in juvenile myasthenia gravis. Neurology 43(5): 977-982.
  33. McMillan HJ, Darras BT, Kang PB (2011) Autoimmune neuromuscular disorders in childhood. Curr Treat Options Neurol 13(6): 590-607.
  34. Parr JR, Jayawant S (2007) Childhood myasthenia: clinical subtypes and practical management. Dev Med Child Neurol 49(8): 629-635.
  35. Takahashi Y, Sugiyama M, Ueda Y, Itoh T, Yagyu K, et al. (2012) Childhoodonset anti-MuSK antibody positive myasthenia gravis demonstrates a distinct clinical course. Brain Dev 34(9): 784-786.
  36. Deymeer F, Gungor-Tuncer O, Yilmaz V, Parman Y, Serdaroglu P, et al. (2007) Clinical comparison of anti-MuSK- vs anti-AChR-positive and seronegative myasthenia gravis. Neurology 68(8): 609-611.
  37. Ghosh JB, Roy M, Peters T (2009) Thymoma associated with myasthenia gravis in infancy. Indian J Pediatr 76(10): 1057-1058.
  38. Lindner A, Schalke B, Toyka KV (1997) Outcome in juvenile-onset myasthenia gravis: a retrospective study with long-term follow-up of 79 patients. J Neurol 244(8): 515-520.
  39. Murai H, Yamashita N, Watanabe M, Nomura Y, Motomura M, et al. (2011) Characteristics of myasthenia gravis according to onset-age: Japanese nationwide survey. J Neurol Sci 305(1-2): 97-102.
  40. Berrih-Aknin S, Frenkian-Cuvelier M, Eymard B (2014) Diagnostic and clinical classification of autoimmune myasthenia gravis. J Autoimmun 48-49: 143-148.
  41. Meriggioli MN, Sanders DB (2009) Autoimmune myasthenia gravis: emerging clinical and biological heterogeneity. Lancet Neurol 8(5): 475- 490.
  42. Sauce D, Larsen M, Fastenackels S, Duperrier A, Keller M, et al. (2009) Evidence of premature immune aging in patients thymectomized during early childhood. J Clin Invest 119 (10): 3070-3078.
  43. Andrews PI (1998) A treatment algorithm for autoimmune myasthenia gravis in childhood. Ann N Y Acad Sci 841: 789-802.
  44. Heng HS, Lim M, Absoud M, Austin C, Clarke D, et al. (2014) Outcome of children with acetylcholine receptor (AChR) antibody positive juvenile myasthenia gravis following thymectomy. Neuromuscul Disord 24(1): 25-30.
  45. Silvestri NJ, Wolfe GI (2014) Treatment-refractory myasthenia gravis. J Clin Neuromuscul Dis 15(4): 167-178.
  46. Madenci AL, Li GZ, Weil BR, Zurakowski D, Kang PB, et al. (2017) The role of thymectomy in the treatment of juvenile myasthenia gravis: a systematic review. Pediatr Surg Int 33(6): 683-694.
  47. Ishigaki K, Shishikura K, Murakami T, Suzuki H, Hirayama Y, et al. (2009) Benefits of FK 506 for refractory eye symptoms in a young child with ocular myasthenia gravis. Brain Dev 31(8): 634-637.
  48. Liu C, Gui M, Cao Y, Lin J, Li Y, et al. (2017) Tacrolimus Improves Symptoms of Children With Myasthenia Gravis Refractory to Prednisone. Pediatr Neurol 77: 42-47.
  49. Iorio R, Damato V, Alboini PE, Evoli A (2015) Efficacy and safety of rituximab for myasthenia gravis: a systematic review and meta-analysis. J Neurol 262 (5): 1115-1119.
  50. Koul R, Al FA, Abdwani R (2012) Rituximab in severe seronegative juvenile myasthenia gravis: review of the literature. Pediatr Neurol 47(3): 209-212.
  51. Wylam ME, Anderson PM, Kuntz NL, Rodriguez V (2003) Successful treatment of refractory myasthenia gravis using rituximab: a pediatric case report. J Pediatr 143(5): 674-677.
  52. Nelson RJ, Pascuzzi RM, Kessler K, Walsh LE, Faught PP, et al. (2009) Rituximab for the treatment of thymoma-associated and de novo myasthenia gravis: 3 cases and review. J Clin Neuromuscul Dis 10(4): 170-177.
  53. Ortiz S, Borchert M (2008) Long-term outcomes of pediatric ocular myasthenia gravis. Ophthalmology 115(7): 1245-1248.e1241.
  54. Ashraf VV, Taly AB, Veerendrakumar M, Rao S (2006) Myasthenia gravis in children: a longitudinal study. Acta Neurol Scand 114(2): 119-123.
  55. Castro D, Derisavifard S, Anderson M, Greene M, Iannaccone S (2013) Juvenile myasthenia gravis: a twenty-year experience. J Clin Neuromuscul Dis 14(3): 95-102.
  56. Rodriguez M, Gomez MR, Howard FJ, Taylor WF (1983) Myasthenia gravis in children: long-term follow-up. Ann Neurol 13 (5): 504-510.
  57. Melbourne Chambers R, Forrester S, Gray R, Tapper J, Trotman H (2012) Myasthenia gravis in Jamaican children: a 12-year institutional review. Paediatr Int Child Health 32(1): 47-50.
  58. Dural K, Yildirim E, Han S, Ozisik K, Ulasan N, et al. (2003) The importance of the time interval between diagnosis and operation in myasthenia gravis patients. J Cardiovasc Surg (Torino) 44(1): 125-129.
  59. Kakisaka Y, Haginoya K, Yokoyama H, Ishitobi M, Wakusawa K, et al. (2006) Successful treatment of a 2-year-old girl with intractable myasthenia gravis using tacrolimus. Brain Dev 28(8): 534-536.
  60. Tracy MM, McRae W, Millichap JG (2009) Graded response to thymectomy in children with myasthenia gravis. J Child Neurol 24(4): 454-459.
  61. Hennessey IA, Long AM, Hughes I, Humphrey G (2011) Thymectomy for inducing remission in juvenile myasthenia gravis. Pediatr Surg Int 27(6): 591-594.
  62. Adams C, Theodorescu D, Murphy EG, Shandling B (1990) Thymectomy in juvenile myasthenia gravis. J Child Neurol 5(3): 215-218.
  63. Ryniewicz B, Badurska B (1977) Follow-up study of myasthenic children after thymectomy. J Neurol 217(2): 133-138.
  64. Hawkins BR, Yu YL, Wong V, Woo E, Ip MS, et al. (1989) Possible evidence for a variant of myasthenia gravis based on HLA and acetylcholine receptor antibody in Chinese patients. Q J Med 70(263): 235-241.
  65. Chen WH, Chiu HC, Hseih RP (1993) Association of HLA-Bw46DR9 combination with juvenile myasthenia gravis in Chinese. J Neurol Neurosurg Psychiatry 56(4): 382-385.
  66. Christiansen FT, Pollack MS, Garlepp MJ, Dawkins RL (1984) Myasthenia gravis and HLA antigens in American blacks and other races. J Neuroimmunol 7(2-3): 121-129.
Citation
Keywords
Signup for Newsletter
Scroll to Top