Quantcast
Channel: Pediatric Neurosurgery – Surgical Neurology International
Viewing all 227 articles
Browse latest View live

Treatment options for pediatric craniopharyngioma

$
0
0
Gaddum D. Reddy, Daniel Hansen, Achal Patel, Yimo Lin, Andrew Jea, Sandi Lam
  1. Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA

Correspondence Address:
Sandi Lam
Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA

DOI:10.4103/2152-7806.178570

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Reddy GD, Hansen D, Patel A, Lin Y, Jea A, Lam S. Treatment options for pediatric craniopharyngioma. Surg Neurol Int 11-Mar-2016;7:

How to cite this URL: Reddy GD, Hansen D, Patel A, Lin Y, Jea A, Lam S. Treatment options for pediatric craniopharyngioma. Surg Neurol Int 11-Mar-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/treatment-options-for-pediatric-craniopharyngioma/

Date of Submission
24-Sep-2015

Date of Acceptance
26-Nov-2015

Date of Web Publication
11-Mar-2016

Abstract

Keywords: Adamantinomatous, brain tumor, craniopharyngioma, pediatric

ILLUSTRATIVE CASES

Case 1

A 13-year-old female with intermittent headaches evaluated by an ophthalmologist was noted to have a retinal abnormality, prompting a magnetic resonance imaging (MRI) scan and referral to the neurosurgery service. On initial exam, the patient was neurologically intact and without headache. Imaging revealed a complex heterogeneous cystic mass, arising from a suprasellar location, invading into the third ventricle, and closely apposed to the hypothalamus bilaterally. There was mild contrast enhancement peripherally and inferiorly. Of note, the initial MRI and clinical presentation [ Figure 1 ] showed no hydrocephalus. Likewise, initial endocrine evaluation was normal. On follow-up imaging, however, the ventricular system was noted to be enlarging. After extensive discussion of the risks, benefits, and alternatives to surgery, a right frontal endoscopic transventricular resection was planned with a goal of gross total resection (GTR). Intraoperatively, the tumor was found to be densely adherent to the walls of the third ventricle. Approximately 50% of the tumor could be safely debulked. Postoperatively, she was noted to have hypopituitarism and required hormonal replacement with desmopressin, hydrocortisone, and levothyroxine. The patient was discharged in stable condition but returned soon after with symptoms and imaging consistent with a trapped right ventricle for which she underwent a septostomy and eventual ventriculoperitoneal shunt placement. She subsequently completed proton radiation for the residual tumor, which has remained stable over 4-year follow-up. After radiation, she developed hypothalamic obesity and suffered a gradual decline in her vision bilaterally. At last follow-up, she could count fingers on the right and could only detect motion on the left. Since completing her treatments, she has required 24-h care. The patient has had multiple emergency room visits and hospital admissions for sodium fluctuations. She has also suffered multiple bone fractures secondary to osteoporosis from chronic steroid use.


Figure 1

Preoperative (left) and postoperative (right) T1-weighted magnetic resonance imaging with contrast images from the patient in Case 1. Coronal (top) and sagittal (bottom) images

 

Case 2

A 13-year-old female presented with headache, fatigue, and nausea. On exam, she was neurologically intact though with severe headache. Imaging revealed a large, complex, heterogeneous cystic mass, arising from a suprasellar location, invading into the third ventricle, and closely apposed to the hypothalamus bilaterally. She had significant hydrocephalus resulting from obstruction of the third ventricle. An external ventricular drain was placed, followed by endoscopic fenestration of the cyst, biopsy of the mass, and placement of a ventriculoperitoneal shunt. Pathology from the biopsy confirmed the suspected diagnosis of craniopharyngioma. The following year, an Ommaya reservoir was placed into an enlarging suprasellar cyst for intermittent as-needed drainage of accumulating fluid. Postoperative imaging showed a decompressed suprasellar cyst; she continues with expectant management of an inferior prepontine cyst [ Figure 2 ], which was clinically asymptomatic. Over the next 2 years, she developed mild hypopituitarism, requiring thyroid, steroid, and estrogen replacement, but she has had no issues with sodium balance. Neurologically, she remains intact and without visual decline. To date, the patient has not received any radiation treatment or chemotherapy infusions through the Ommaya. She continues with excellent academic performance and normal activities of daily living at 2-year follow-up.


Figure 2

Preoperative (left) and postoperative (right) T1-weighted magnetic resonance imaging with contrast images from the patient in Case 2. Coronal (top) and sagittal (bottom) images

 

EPIDEMIOLOGY AND CLINICAL PRESENTATION

Craniopharyngiomas are rare tumors, with an estimated incidence of 2–3/1 million. They are most common in children (age 5–15 years old) and older adults (60–70 years of age).[ 11 16 30 ] They are the most common nonglial tumor in the pediatric population, representing 6–9% of all brain tumors in this age range. No clear racial or gender predilection exists.[ 36 ]

These tumors are typically located in or above the sella turcica and produce symptoms by compression of adjacent neural structures. Slow growth and insidious onset of symptoms often delay arriving at a diagnosis. Potential symptoms are wide-ranging. They include visual deficits from compression of the optic apparatus, endocrine deficiencies, such as diabetes insipidus (DI) or pan-hypopituitarism from compression of the pituitary gland or stalk, hypothalamic compression and dysfunction resulting in abnormalities in sleep, appetite, or thermal regulation, or symptoms of hydrocephalus such as headache or vomiting from obstruction of cerebral spinal fluid pathways.[ 12 25 35 ] At the time of diagnosis, 20–50% of children are noted to have hormonal insufficiencies, making endocrine testing mandatory.[ 36 ]

TUMOR BIOLOGY

Craniopharyngiomas occur in two histological subtypes: An adamantinomatous form that is the most common pediatric variant and a papillary form that is found almost exclusively in adults. The pediatric form is thought to arise from epithelial remnants of the craniophayngeal duct or Rathke's pouch, an embryologic structure that develops into the anterior pituitary. These remnants are thought to enlarge during the development of the pituitary gland and thus present early in life. Grossly, these tumors typically have both solid and cystic components and are often calcified on imaging. The cyst fluid is dark, oily, and rich in lipids with birefringent cholesterol crystals.[ 3 15 16 29 ] Papillary craniopharyngiomas, or adult craniopharyngiomas, on the other hand, are theorized to arise from metaplasia of existing squamous cell rests and thus present later in life.

Recent genetic analysis has also shown differences between these two subtypes. Mutations in B-catenin (CTNNB1), a downstream effector of the Wnt pathway that is, involved in cellular growth and development, has been described in 60–96% of adamantinomatous craniopharyngiomas.[ 4 5 22 33 ] By contrast, papillary craniopharyngiomas recently have been discovered to frequently harbor V600E mutations of the BRAF gene,[ 4 ] which is a key player in the mitogen-activated protein kinase pathway.

MEDICAL WORKUP AND MANAGEMENT

A complete workup for craniopharyngioma should include MRI with and without gadolinium contrast to characterize the tumor and its relationship with critical nearby neural structures. As demonstrated in Case 1, these tumors may microscopically invade these structures even when this is not apparent on preoperative imaging. MR angiography can also be helpful in delineating the location of nearby vessels at the skull base. A noncontrasted computed tomography scan can also reveal complex calcifications and expansion of the sella, which is helpful in narrowing the differential. In addition, a workup for endocrinopathies should also be performed with measurements of growth hormone, thyroid stimulating hormone, follicular stimulating hormone/luteinizing hormone, prolactin, cortisol, and serum electrolytes. Any abnormalities ideally should be corrected before surgery is performed. Finally, formal visual acuity and visual field assessment are important to characterize any deficits that exist preoperatively.

TREATMENT AND OUTCOMES

Although histologically benign, these tumors frequently recur after treatment, and their close association with critical neurologic structures can lead to a much more malignant course. Surgical treatment options range from GTR to more conservative surgery (i.e., subtotal resection [STR] or biopsy only) followed by postoperative radiotherapy (RT), or other less invasive procedures such as endoscopic cyst fenestration or placement of an Ommaya reservoir into the tumor cyst for delivery of antineoplastic agents.[ 31 ] Nonsurgical options include stereotactic radiosurgery (SRS) or systemic chemotherapy. Over the past several decades, a paradigm shift has occurred in treatment from maximal resection to more scaled back interventions, in an attempt to balance tumor control and quality of life.

Historically, open cranial surgery with the goal of achieving GTR has been the treatment of choice, as it allows rapid decompression, provides a histological diagnosis, and is thought to minimize recurrence. The results of GTR have been influenced largely by surgeon experience, and the tendency of these tumors to invade nearby critical neuromuscular structures often leads to significant morbidity.[ 8 14 ] Multiple institutions have published their historical data in regard to this tumor, and we can extract from them several trends.[ 9 13 32 36 37 ] Reported reoccurrence rates after GTR range from 7% to 34%. Reported death rates were as high as 20%. Postoperative need for permanent hormone replacement was 80–86%, permanent DI from 75% to 90% and worsening of vision between 10% and 33%. Larger tumors and greater hypothalamic invasion were associated with worse outcomes. These results would lend to the conclusion that GTR is very often associated with a high surgical morbidity. Other factors that have also been shown to be associated with higher morbidity include a diagnosis before the age of 10 and the presence of intracranial hypertension on initial presentation.[ 17 ]

The lack of acceptable outcomes with GTR has led to groups approaching these tumors with a more conservative surgical plan, including STR or biopsy, followed in some cases with RT.[ 6 21 24 ] Groups have found no significant difference in progression-free survival at 5 years between GTR and STR + RT;[ 8 18 ] however, STR without RT has significantly increased recurrence rates.[ 8 ] When looking at mean quality-adjusted life years (QALY) as the outcome at 5-year follow-up, biopsy + RT was associated with the most mean QALY (3.9, standard deviation [SD] 0.2), followed by endoscopic surgery (3.7, SD 0.2) and, more distantly, by STR + RT (2.9, SD 0.2) and GTR (2.7, SD 0.1).[ 3 ]

For the surgeon experienced in endoscopy, an endoscopic endonasal approach, as compared to an open approach, has been shown to provide higher rates for GTR (66–69% vs. 48%), with lower reoccurrence (18% vs. 28%), with lower rates of permanent DI (27–32% vs. 48%), and less visual deterioration (1.7% vs. 11%).[ 24 ] This surgical corridor largely avoids traversing critical neurovascular structures and allows for better visualization of the subchiasmatic space and intrasellar portions of the tumor, which is a commonly missed and a frequent cause of recurrence after open surgery.[ 14 ] However, this approach does not obviate the risks of stretch or manipulation injury due to tumor adherence to surrounding structures.

Perhaps the least invasive treatment option is the insertion of an Ommaya reservoir into the cystic aspect of the tumor followed by drainage with or without subsequent instillation of antineoplastic agents. Several reports have shown follow-up out to 7 years with good cyst size control and 43–73% of patients needing no additional treatment.[ 28 34 ] It is possible that a longer follow-up period would influence these results, but this may be a method that allows for delaying RT in young children. Historically, bleomycin was used for intracystic infusion, but concerns for central nervous system toxicity have led to the use of interferon-a (INFa) in its place.[ 7 34 ] In addition to intracystic chemotherapy, intracystic irradiation with yttrium-90 has also demonstrated long-term results in reducing the size of recurrent craniopharyngioma cysts when used as part of a multimodal treatment regimen though this treatment strategy has not been widely replicated or implemented.[ 20 ]

Radiation therapy, either as a first-line treatment or as an adjuvant to surgical resection, has become a frequent care option. SRS can deliver radiation with a steeper dose gradient between tumor and adjacent brain structures. It is believed to lead to lower rates of neurotoxic side effects in comparison to traditional fractionated RT. The potential side effects of radiation are similar to that of open surgery and include the following: Panhypopituitarism, DI, hypothalamic dysfunction, vasculopathy, cognitive dysfunction, optic neuropathy, and secondary malignancies.[ 2 10 26 ] Published control rates for STR + SRS are good at 60–90%, depending on tumor type, with low rates of reported endocrine and visual deterioration in only 6% of patients using a lower marginal dose.[ 23 27 ] Because of high documented rates of optic neuropathy, SRS traditionally has been limited to smaller tumors (<3 cm) that were 3–5 mm away from the optic apparatus;[ 27 ] however, multisession SRS may allow for treatment of tumors closer to the chiasm.[ 1 ]

Finally, systemic chemotherapy treatment with INF-alpha-2b has been performed by the Pediatric Brain Tumor Consortium (PBTC-039) phase 2 trial in pediatric patients with recurrent craniopharyngioma. Three of the 12 patients tested experienced a response to the drug, and none developed any permanent side effects.[ 19 ] The same group recently published another study using the pegylated form of the drug in a cohort of five patients with recurrent disease in which four demonstrated response on imaging.[ 38 ]

CHOICE OF THERAPY

The treatment paradigm for craniopharyngioma has evolved over time, as our treatment options have expanded and our understanding of the long-term consequences of radical resection has grown. Originally, aggressive resection was the only hope of controlling this tumor; in spite of the often severe morbidity of such an approach, it remained the mainstay of treatment. But as the natural history of this tumor is recurrence, it may be considered a chronic disease, with the goal of maximizing control but minimizing patient morbidity. Our desire as surgeons to obtain tumor-free postoperative imaging may overlook the impact on our patient. Weighing risks, benefits, and alternatives to surgical goals and approaches are crucial in the treatment of this challenging tumor.

To date, no class I recommendations exist for the best treatment of these tumors. Management should be by a multidisciplinary team (neurosurgery, endocrinology, ophthalmology, psychology, oncology, and radiation oncology) and be individualized for each patient. There are some practical surgical considerations to keep in mind. In cases where total resection can be obtained without significant morbidity (i.e., cases where the tumor is not invading or adherent to the hypothalamus), GTR remains the treatment of choice. In cases where the tumor is small and the solid portions are primary intrasellar, without significant extension laterally in the suprasellar space or without encasement of vessels, an endoscopic approach may provide good outcomes. In cases where the tumor is densely involved in critical structures and has a significant cystic component causing mass effect, an Ommaya placement with or without chemotherapy infusion may represent a less invasive way to decompress neural structures and control tumor progression. Finally, in cases of STR or recurrent disease, particularly with a favorable margin between the tumor and the optic chiasm, adjuvant RT or SRS is likely to improve progression-free survival. Promising treatments from PBTC trials may offer hope for future therapies with lower side effect profiles. Ultimately, this tumor remains one of the most difficult pediatric neurosurgical problems, and recommendations will continue to evolve.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Adler JR, Gibbs IC, Puataweepong P, Chang SD. Visual field preservation after multisession cyberknife radiosurgery for perioptic lesions. Neurosurgery. 2006. 59: 244-54

2. Agha A, Sherlock M, Brennan S, O’Connor SA, O’sullivan E, Rogers B. Hypothalamic-pituitary dysfunction after irradiation of nonpituitary brain tumors in adults. J Clin Endocrinol Metab. 2005. 90: 6355-60

3. Ali ZS, Bailey RL, Daniels LB, Vakhshori V, Lewis DJ, Hossain AT. Comparative effectiveness of treatment options for pediatric craniopharyngiomas. J Neurosurg Pediatr. 2014. 13: 178-88

4. Brastianos PK, Taylor-Weiner A, Manley PE, Jones RT, Dias-Santagata D, Thorner AR. Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas. Nat Genet. 2014. 46: 161-5

5. Buslei R, Nolde M, Hofmann B, Meissner S, Eyupoglu IY, Siebzehnrübl F. Common mutations of beta-catenin in adamantinomatous craniopharyngiomas but not in other tumours originating from the sellar region. Acta Neuropathol. 2005. 109: 589-97

6. Caldarelli M, Massimi L, Tamburrini G, Cappa M, Di Rocco C. Long-term results of the surgical treatment of craniopharyngioma: The experience at the Policlinico Gemelli, Catholic University, Rome. Childs Nerv Syst. 2005. 21: 747-57

7. Cavalheiro S, Di Rocco C, Valenzuela S, Dastoli PA, Tamburrini G, Massimi L. Craniopharyngiomas: Intratumoral chemotherapy with interferon-alpha: A multicenter preliminary study with 60 cases. Neurosurg Focus. 2010. 28: E12-

8. Clark AJ, Cage TA, Aranda D, Parsa AT, Sun PP, Auguste KI. A systematic review of the results of surgery and radiotherapy on tumor control for pediatric craniopharyngioma. Childs Nerv Syst. 2013. 29: 231-8

9. Cohen M, Bartels U, Branson H, Kulkarni AV, Hamilton J. Trends in treatment and outcomes of pediatric craniopharyngioma, 1975-2011. Neuro Oncol. 2013. 15: 767-74

10. Darzy KH, Shalet SM. Hypopituitarism after cranial irradiation. J Endocrinol Invest. 2005. 28: 78-87

11. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro Oncol. 2012. 14: v1-49

12. Duff J, Meyer FB, Ilstrup DM, Laws ER, Schleck CD, Scheithauer BW. Long-term outcomes for surgically resected craniopharyngiomas. Neurosurgery. 2000. 46: 291-302

13. Elliott RE, Hsieh K, Hochm T, Belitskaya-Levy I, Wisoff J, Wisoff JH. Efficacy and safety of radical resection of primary and recurrent craniopharyngiomas in 86 children. J Neurosurg Pediatr. 2010. 5: 30-48

14. Fahlbusch R, Honegger J, Paulus W, Huk W, Buchfelder M. Surgical treatment of craniopharyngiomas: Experience with 168 patients. J Neurosurg. 1999. 90: 237-50

15. Garnett MR, Puget S, Grill J, Sainte-Rose C. Craniopharyngioma. Orphanet J Rare Dis. 2007. 2: 18-

16. Garrè ML, Cama A. Craniopharyngioma: Modern concepts in pathogenesis and treatment. Curr Opin Pediatr. 2007. 19: 471-9

17. Gautier A, Godbout A, Grosheny C, Tejedor I, Coudert M, Courtillot C. Markers of recurrence and long-term morbidity in craniopharyngioma: A systematic analysis of 171 patients. J Clin Endocrinol Metab. 2012. 97: 1258-67

18. Habrand JL, Ganry O, Couanet D, Rouxel V, Levy-Piedbois C, Pierre-Kahn A. The role of radiation therapy in the management of craniopharyngioma: A 25-year experience and review of the literature. Int J Radiat Oncol Biol Phys. 1999. 44: 255-63

19. Jakacki RI, Cohen BH, Jamison C, Mathews VP, Arenson E, Longee DC. Phase II evaluation of interferon-alpha-2a for progressive or recurrent craniopharyngiomas. J Neurosurg. 2000. 92: 255-60

20. Julow J, Backlund EO, Lányi F, Hajda M, Bálint K, Nyáry I. Long-term results and late complications after intracavitary yttrium-90 colloid irradiation of recurrent cystic craniopharyngiomas. Neurosurgery. 2007. 61: 288-95

21. Karavitaki N, Brufani C, Warner JT, Adams CB, Richards P, Ansorge O. Craniopharyngiomas in children and adults: Systematic analysis of 121 cases with long-term follow-up. Clin Endocrinol (Oxf). 2005. 62: 397-409

22. Kato K, Nakatani Y, Kanno H, Inayama Y, Ijiri R, Nagahara N. Possible linkage between specific histological structures and aberrant reactivation of the Wnt pathway in adamantinomatous craniopharyngioma. J Pathol. 2004. 203: 814-21

23. Kobayashi T, Kida Y, Mori Y, Hasegawa T. Long-term results of gamma knife surgery for the treatment of craniopharyngioma in 98 consecutive cases. J Neurosurg. 2005. 103: 482-8

24. Komotar RJ, Starke RM, Raper DM, Anand VK, Schwartz TH. Endoscopic endonasal compared with microscopic transsphenoidal and open transcranial resection of craniopharyngiomas. World Neurosurg. 2012. 77: 329-41

25. Manley PE, McKendrick K, McGillicudy M, Chi SN, Kieran MW, Cohen LE. Sleep dysfunction in long term survivors of craniopharyngioma. J Neurooncol. 2012. 108: 543-9

26. McCord MW, Buatti JM, Fennell EM, Mendenhall WM, Marcus RB, Rhoton AL. Radiotherapy for pituitary adenoma: Long-term outcome and sequelae. Int J Radiat Oncol Biol Phys. 1997. 39: 437-44

27. Minniti G, Esposito V, Amichetti M, Enrici RM. The role of fractionated radiotherapy and radiosurgery in the management of patients with craniopharyngioma. Neurosurg Rev. 2009. 32: 125-32

28. Moussa AH, Kerasha AA, Mahmoud ME. Surprising outcome of ommaya reservoir in treating cystic craniopharyngioma: A retrospective study. Br J Neurosurg. 2013. 27: 370-3

29. Müller HL. Childhood craniopharyngioma. Pituitary. 2013. 16: 56-67

30. Müller HL. Craniopharyngioma - A childhood and adult disease with challenging characteristics. Front Endocrinol (Lausanne). 2012. 3: 80-

31. Samii M, Tatagiba M. Surgical management of craniopharyngiomas: A review. Neurol Med Chir (Tokyo). 1997. 37: 141-9

32. Schoenfeld A, Pekmezci M, Barnes MJ, Tihan T, Gupta N, Lamborn KR. The superiority of conservative resection and adjuvant radiation for craniopharyngiomas. J Neurooncol. 2012. 108: 133-9

33. Sekine S, Shibata T, Kokubu A, Morishita Y, Noguchi M, Nakanishi Y. Craniopharyngiomas of adamantinomatous type harbor beta-catenin gene mutations. Am J Pathol. 2002. 161: 1997-2001

34. Steinbok P, Hukin J. Intracystic treatments for craniopharyngioma. Neurosurg Focus. 2010. 28: E13-

35. Weiner HL, Wisoff JH, Rosenberg ME, Kupersmith MJ, Cohen H, Zagzag D. Craniopharyngiomas: A clinicopathological analysis of factors predictive of recurrence and functional outcome. Neurosurgery. 1994. 35: 1001-10

36. Wisoff J, Donahue B, Albright AL, Pollack IF, Adelson PD.editors. Craniopharyngiomas. Principles and Practice of Pediatric Neurosurgery. New York: Thieme; 2007. p. 560-78

37. Yasargil MG, Curcic M, Kis M, Siegenthaler G, Teddy PJ, Roth P. Total removal of craniopharyngiomas. Approaches and long-term results in 144 patients. J Neurosurg. 1990. 73: 3-11

38. Yeung JT, Pollack IF, Panigrahy A, Jakacki RI. Pegylated interferon-a-2b for children with recurrent craniopharyngioma. J Neurosurg Pediatr. 2012. 10: 498-503


Malignant cerebellar peduncle lesions – rapid progression and poor outcome

$
0
0
Navneet Singla, Ankur Kapoor, Amey Savardekar, B. D. Radotra, Debjyoti Chatterjee, Sunil K. Gupta
  1. Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
  2. Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
  3. Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India

Correspondence Address:
Ankur Kapoor
Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India

DOI:10.4103/2152-7806.178569

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Singla N, Kapoor A, Savardekar A, Radotra BD, Chatterjee D, Gupta SK. Malignant cerebellar peduncle lesions - rapid progression and poor outcome. Surg Neurol Int 11-Mar-2016;7:

How to cite this URL: Singla N, Kapoor A, Savardekar A, Radotra BD, Chatterjee D, Gupta SK. Malignant cerebellar peduncle lesions - rapid progression and poor outcome. Surg Neurol Int 11-Mar-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/malignant-cerebellar-peduncle-lesions-%e2%80%91-rapid-progression-and-poor-outcome/

Date of Submission
01-Sep-2015

Date of Acceptance
20-Jan-2016

Date of Web Publication
11-Mar-2016

Abstract

Background:Tumors arising from cerebellar peduncle are extremely rare and behave aggressively. The inclusion of these into either cerebellar or brainstem gliomas is contentious.

Case Description:We performed clinicopathological review of three patients treated at our institute and surveyed the literature for previous such reported cases. Mean duration of symptoms in our patients was 2 weeks. Subtotal tumor resection was performed in two patients while the third underwent stereotactic biopsy followed by chemoradiotherapy. Histopathology revealed glioblastoma in initial two patients and medulloblastoma Grade IV in the third. The two patients who underwent surgical excision succumbed to the illness within 2 days and a month, respectively.

Conclusion:Malignant cerebellar peduncular lesions have poor overall survival despite surgical debulking. It is not confirmed whether these tumors should be considered as cerebellar lesions or brainstem gliomas due to aggressive clinical behavior, and so the ideal line of management is not yet known.

Keywords: Glioblastoma cerebellar peduncle, malignant peduncular lesions, outcome cerebellar lesions

INTRODUCTION

Primary cerebellar peduncle lesions are defined as the ones that arise directly from the peduncle and spread to involve the neighboring cerebellum and brainstem vital areas. Malignant lesions at this location are rare. It is a dilemma whether they behave as cerebellar tumors or as brainstem tumors. This ignites controversy of whether radical surgery is beneficial or histological diagnosis followed by radiotherapy should be attempted. There is scarce literature on the same and prognosis remains dismal despite surgery and radiotherapy. These tumors are known for their low overall survival (OS) and early recurrence.[ 7 ] We report a series of three such patients and discuss the clinicopathological features of the same.

CASE REPORTS

Case 1

A 10-year-old male presented with complaints of gait disturbance and vomiting for 2 weeks. On examination, he was drowsy and had left sided cerebellar signs and a weak gag reflex on left side. Magnetic resonance imaging (MRI) [ Figure 1 ] revealed solid-cystic lesion in left anterior cerebellum and cerebellar peduncle that was hypointense on T1-weighted (T1W), hyperintense on T2-weighted (T2W), had heterogenous contrast enhancement with surrounding edema. Midline suboccipital craniotomy was performed for tumor excision. Histology was suggestive of low-grade glioma. Thereafter, patient was lost to follow-up and presented 3 months later in unconscious state with a large recurrence and hydrocephalus. A critical review of previous biopsy showed few elements of atypical cells with high mitotic activity. The patient underwent a ventriculoperitoneal shunt followed by reexploration. Intraoperatively the tumor was vascular, infiltrating surrounding cerebellum with areas of necrosis. Histopathology revealed glioblastoma (World Health Organization [WHO] Grade IV) with highly cellular tumor showing nuclear pleomorphism and endocapillary proliferation. The patient did not gain consciousness after surgery. Computed tomography (CT) was suggestive of hypodensity and edema in brainstem. Further neurological deterioration was noticed next day with episodes of tachycardia and apnea. The patient finally succumbed to illness due to brainstem edema after 2 days.


Figure 1

(a) Noncontrast computed tomography (axial) showing hypodense lesion at left cerebellar peduncle. (b) T1-weighted contrast axial image showing nonenhancing lesion before first surgery. (c) Axial T1-weighted contrast image at recurrence showing solid-cystic lesion at peduncle. (d) Photomicrograph showing tumor cells with nuclear pleomorphism, high mitotic activity (H and E, ×40). (e) Ki-67 immunostain showing very high proliferation index (IP, ×40)

 

Case 2

A 9-year-old female presented with complaint of diplopia on far vision and facial deviation for 1 week. Examination revealed gait ataxia, left gaze, facial paresis, and subtle left cerebellar signs. MRI [ Figure 2 ] revealed a left peduncular lesion which was hypointense on T1W, hyperintense on T2W and had heterogenous enhancement on contrast. Two days later, she became unconscious, when a repeat CT showed intratumoral bleed again with mass effect. She underwent midline subocciptal craniotomy and had a friable, vascular tumor with necrosis. Histopathology revealed small cell glioblastoma (WHO Grade IV) with highly cellular tumor showing palisading necrosis, nuclear pleomorphism. Tumor cells stained positive for glial fibrillary acidic protein. Despite adequate decompression, the patient did not show clinical improvement. CT was suggestive of brainstem edema. There were scarce spontaneous respiratory efforts, pupil reaction was absent, and stimulus yielded minimal flexion response. She finally succumbed to illness after 1 month due to brainstem edema.


Figure 2

(a) Noncontrast computed tomography (axial) showing hemorrhagic lesion at left cerebellar peduncle. (b-d) Axial T1-weighted, axial T2-weighted, and sagittal T1-weighted contrast magnetic resonance imaging showing lesion hypointense on T1, heterogeneously hyperintense on T2 and dense contrast enhancing with edema and infiltrating pons. (e) Photomicrograph showing highly cellular glial tumor with high nuclear pleomorphism (H and E, ×40). (f) Tumor cell shows glial fibrillary acidic protein positive cytoplasmic processes (IP, ×40)

 

Case 3

A 30-year-old male patient presented with complaint of gait ataxia and visual blurring for 3 weeks. Examination revealed left cerebellar signs and a brun's nystagmus with coarse component to left. CT head demonstrated a hyperdense lesion in left anterior cerebellum spreading to middle cerebellar peduncle. MRI [ Figure 3 ] demonstrated lesion that was hypointense on T1W, iso- to hyper-intense on T2W with necrotic foci at inferior end. Taking into consideration the close approximation to vital brainstem area, he was taken up for stereotactic biopsy. Histopathology revealed medulloblastoma (Grade IV) with high mitotic activity, high nuclear: Cytoplasmic ratio, and round cells. The tumor stained positive for synaptophysin and neuron-specific enolase. He received adjuvant radiotherapy and concurrent chemotherapy with cisplatin and etoposide. After ten weeks, he presented with headache and vomiting. Repeat MRI revealed slight increase in edema and hydrocephalus for which a ventriculoperitoneal shunt was performed. At follow-up of 6 months, he is conscious and independent for daily routine activities.


Figure 3

(a-c) T1-weighted, T2-weighted, and T1-weighted contrast axial images showing well-defined lesion at left middle cerebellar peduncle hypointense on T1, isointense on T2, predominantly nonenhancing on contrast with small posterior nodule showing ring enhancement and necrosis. (d) Photomicrograph showing cellular tumor composed of round, blue cells with high nuclear: Cytoplasmic ratio and high mitotic activity and rosette formation (H and E, ×40).(e) Tumor cells show faint cytoplasmic synaptophysin positivity (IP, ×40)

 

DISCUSSION

Glioblastomas in cerebellum account for <5% of all cerebellar astrocytomas.[ 2 7 9 ] No apparent explanation accounts for the rarity of the infratentorial compartments as a site of origin for glioblastomas. Peduncular involvement in cerebellar lesions has been scarcely mentioned in the literature. The mean duration of symptoms in glioblastomas involving cerebellum is 4–5 weeks and OS of 9–15 months[ 13 ] while it is only slightly better for the anaplastic gliomas.[ 6 11 12 ] Symptomatology consists of headache, nausea and vomiting, diplopia, facial deviation, hearing disturbance, and gait ataxia. Brainstem signs are frequently present in these lesions.[ 3 ] Compared to hemispheric tumors, the ones arising from cerebellar peduncle behave more aggressively, are invading in nature, and recur early.

Tumor frequently extends beyond the enhancing part seen on imaging. Dissemination is believed to occur along ventricular wall and subarachnoid space[ 8 9 10 12 ] with reported rates of leptomeningeal spread ranging from 17% to 66%.[ 7 ] While an individual can tolerate removal of an entire cerebellar hemisphere, its proximity to brainstem via the cerebellar peduncles makes it easy for tumor to spread to regions that would be considered unresectable. The poor outcome despite surgery and chemoradiotherapy in primary cerebellar lesions that recur at peduncle depicts the effect of involvement vital brainstem regions.[ 4 ] The management of such aggressive lesions is a matter of debate as surgery may not provide a long-term survival.[ 10 ] Considering the high leptomeningeal spread, role of craniospinal irradiation is well established.[ 7 ] Peduncular involvement in medulloblastoma, as in case 3 in our study, has been reported earlier. The spread is believed to occur from flocculus that projects into cerebellopontine angle or residual of lateral medullary velum at this location.[ 5 ] Migratory process of germinal cells in a lateral direction explains involvement of cerebellopontine angle in adult medulloblastoma.[ 1 ] Although glioblastoma and medulloblastoma are entirely different pathologies, they may behave similarly as seen in our series where all lesions were high-grade (WHO Grade IV) with rapid dissemination.[ 11 ]

The rapidity of onset of symptoms in these patients and previously reported patients harboring cerebellar high-grade lesions is alarming,[ 10 ] with a mean duration of only 2 weeks. Two of three patients who underwent surgery died with a mean survival time of 2 weeks only. The third, after stereotactic biopsy, is under follow-up at 6 months. The overall scenario seems worse than that of supratentorial glioblastoma and brainstem gliomas, exact reason for which is not known. We believe that lesions which appear high grade and arise from cerebellar peduncle may have a natural history similar to aggressive brainstem gliomas.[ 3 ] Extraneural spread similar to that seen in medulloblastoma may be present in cerebellar glioblastoma.[ 11 ]

It is possible that the natural behavior and, therefore, the mode of failure of therapy, of cerebellar high-grade lesions invading cerebellar peduncle differ from hemispheric lesions. The truly aggressive nature of these lesions and the dismal survival in the presence of limited overall reported cases so far raises question of appropriate line of management.[ 10 ] It is a controversy whether these lesions should be considered merely cerebellar pathologies or brainstem lesions with poor overall outcome.

Our experience with such high-grade lesions at cerebellar peduncle has made us believe that proper case selection is imperative as aggressive surgical approach may be hazardous. With limited OS and associated risk of working in vital brainstem regions, biopsy followed by chemoradiotherapy may be a better option in few selected patients.

CONCLUSION

Malignant cerebellar peduncle lesions present with short history, progress rapidly, and may have a dismal outcome despite surgery. The small number of overall reported cases in literature leaves much to be sorted out in coming years. Although the number of patients in our study is small, certainly the focus of this controversy is the poorly defined natural history of high-grade lesions at cerebellar peduncle. We share our experience of managing such patients and together with a thorough literature review, put forth a rarely described entity, the ideal management of which is controversial.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Chung EJ, Jeun SS. Extra-axial medulloblastoma in the cerebellar hemisphere. J Korean Neurosurg Soc. 2014. 55: 362-4

2. Dohrmann GJ, Farwell JR, Flannery JT. Glioblastoma multiforme in children. J Neurosurg. 1976. 44: 442-8

3. Donaldson SS, Laningham F, Fisher PG. Advances toward an understanding of brainstem gliomas. J Clin Oncol. 2006. 24: 1266-72

4. Endo H, Kumabe T, Kon H, Yoshimoto T, Nakasato Y. A case of primary cerebellar glioblastoma in childhood. No Shinkei Geka. 2002. 30: 1325-9

5. Jaiswal AK, Mahapatra AK, Sharma MC. Cerebellopointine angle medulloblastoma. J Clin Neurosci. 2004. 11: 42-5

6. Kopelson G, Linggood R. Cerebellar glioblastoma. Cancer. 1982. 50: 308-11

7. Kulkarni AV, Becker LE, Jay V, Armstrong DC, Drake JM. Primary cerebellar glioblastomas multiforme in children. Report of four cases. J Neurosurg. 1999. 90: 546-50

8. Lapras C, Patet JD, Lapras C, Mottolese C. Cerebellar astrocytomas in childhood. Childs Nerv Syst. 1986. 2: 55-9

9. Pang D, Ashmead JW. Extraneural metastasis of cerebellar glioblastoma multiforme. Neurosurgery. 1982. 10: 252-7

10. Reddy GD, Sen AN, Patel AJ, Bollo RJ, Jea A. Glioblastoma of the cerebellum in children: Report of five cases and review of the literature. Childs Nerv Syst. 2013. 29: 821-32

11. Salazar OM. Primary malignant cerebellar astrocytomas in children: A signal for postoperative craniospinal irradiation. Int J Radiat Oncol Biol Phys. 1981. 7: 1661-5

12. Shinoda J, Yamada H, Sakai N, Ando T, Hirata T, Hirayama H. Malignant cerebellar astrocytic tumours in children. Acta Neurochir (Wien). 1989. 98: 1-8

13. Steinberg GK, Shuer LM, Conley FK, Hanbery JW. Evolution and outcome in malignant astroglial neoplasms of the cerebellum. J Neurosurg. 1985. 62: 9-17

McCune–Albright syndrome with craniofacial dysplasia: Clinical review and surgical management

$
0
0
Telmo Augusto Barba Belsuzarri, João Flavio Mattos Araujo, Carlos Alberto Morassi Melro, Maick Willen Fernandes Neves, Juliano Nery Navarro, Leandro Gomes Brito, Luis Otavio Carneiro Pontelli, Luis Gustavo de Abreu Mattos, Tiago Fernandes Gonçales, Wolnei Marques Zeviani
  1. Department of Neurosurgery, Hospital e Maternidade Celso Pierro, PUC-Campinas, São Paulo, Brazil

Correspondence Address:
Telmo Augusto Barba Belsuzarri
Department of Neurosurgery, Hospital e Maternidade Celso Pierro, PUC-Campinas, São Paulo, Brazil

DOI:10.4103/2152-7806.178567

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Barba Belsuzarri TA, João Flavio Mattos Araujo, Morassi Melro CA, Fernandes Neves MW, Navarro JN, Brito LG, Carneiro Pontelli LO, Abreu Mattos LG d, Tiago Fernandes Gonçales, Zeviani WM. McCune–Albright syndrome with craniofacial dysplasia: Clinical review and surgical management. Surg Neurol Int 11-Mar-2016;7:

How to cite this URL: Barba Belsuzarri TA, João Flavio Mattos Araujo, Morassi Melro CA, Fernandes Neves MW, Navarro JN, Brito LG, Carneiro Pontelli LO, Abreu Mattos LG d, Tiago Fernandes Gonçales, Zeviani WM. McCune–Albright syndrome with craniofacial dysplasia: Clinical review and surgical management. Surg Neurol Int 11-Mar-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/mccune-albright-syndrome-with-craniofacial-dysplasia-clinical-review-and-surgical-management/

Date of Submission
08-Oct-2015

Date of Acceptance
20-Nov-2015

Date of Web Publication
11-Mar-2016

Abstract

Background:Fibrous dysplasia (FD) is a benign fibro-osseous lesion related to an abnormal bone development and replacement by fibrous tissue. FD has three clinical patterns namely monostotic, polyostotic, and the McCune–Albright syndrome (MAS). MAS is a rare genetic disorder (about 3% of all FD's) that comprises a triad of polyostotic FD, café-au-lait skin macules, and precocious puberty. MAS can involve the orbit region and cause stenosis in the optic canal, leading the patient to a progressive visual loss.

Methods:We reported a case of craniofacial FD in MAS in a 9-year-old male with progressive visual loss, submitted to optic nerve decompression by fronto-orbito-zygomatic approach, with total recovery. A research was made at Bireme, PubMed, Cochrane, LILACS, and MEDLINE with the keywords: FD/craniofacial/McCune–Albright/Optic compression for the clinical review.

Results:A clinical review of the disease was made, the multiple, clinical, and surgical management options were presented, and the case report was reported.

Conclusion:MAS is a rare disease with a progressive polyostotic FD. Whenever it affects the orbit region, the optic canal, and it is associated with a progressive visual loss, the urgent optic nerve decompression is mandatory, either manually or with a rapid drill. It is known that aggressive approach is associated with less recurrence; it is also associated with worsening of the visual loss in optic nerve decompression. In MAS cases, multiple and less aggressive surgeries seem to be more suitable.

Keywords: Craniofacial dysplasia, fibrous dysplasia, McCune–Albright syndrome, orbit fibrous dysplasia, visual loss

INTRODUCTION

Fibrous dysplasia (FD) is a benign intramedullary fibro-osseous lesion related to an abnormal bone development and replacement of bone and marrow bone by fibrous tissue. FD has three clinical patterns namely monostotic, polyostotic, and the McCune–Albright Syndrome (MAS). MAS is a rare sporadic genetic disorder (about 3% of all FD's) that comprises a triad of polyostotic FD, café-au-lait skin macules, precocious puberty, and it is a subtype of the general FD.

CASE REPORT

Male, 9 years old, had a femur bone fracture while running; an orthopedic surgery was performed and referred to the Endocrinology Department [ Figure 1 ]. After 6 months of fracture, the patient started to present a progressive holocranial headache. After 1 month, he started to have a progressive temporal visual loss and proptosis on the right side [ Figure 2 ]. During the clinical investigation, the patient had café-au-lait spots [ Figure 3 ] and precocious puberty, with an increase of the testosterone levels and somatomedin. Moreover, the computed tomography (CT) and magnetic resonance imaging (MRI) scans showed that the lateral/roof of the orbital cavity was increased with optic nerve compression [Figures 4 and 5 ]. The patient was diagnosed with MAS, FD of the orbit bone, and optic nerve compression. Decompression surgery was performed with a fronto-orbito-zygomatic approach using a small sphenoid drill and optic canal decompression with manual instruments. The reconstruction of the lateral orbit was performed; the patient had a good recovery of the visual capacity after surgery and did not have new symptoms after 1 year of follow-up. The pathology confirmed the fibrous osseous tissue and FD [ Figure 6 ].


Figure 1

Scar from the surgical approach of the femur fracture

 

Figure 2

Proptosis of the right eye

 

Figure 3

Café-au-lait macule

 

Figure 4

Computed tomography scan shows fibrous dysplasia of the skull base and orbit with narrowing of the optic canal

 

Figure 5

Magnetic resonance imaging: High sign at T1 and low sign at T2 sequences show the fibrous dysplastic pattern

 

Figure 6

Substitution of the bone marrow by a soft fibroblastic tissue with irregular trabeculae

 

DISCUSSION

FD is a benign lesion wherein normal bone is replaced by fibrous tissue and immature bone due to a defect on osteoblastic differentiation and maturation.

There are three subtypes of FD: (1) Monostotic, with one bone involvement and the most common subtype. (2) Polyostotic, with multiple bones involvement. (3) MAS which combines polyostotic FD, endocrinopathy, and café-au-lait spots.[ 3 ] The clinical diagnosis of this uncommon syndrome can be made with two of these criteria. It occurs in children and adults, in both sex,[ 22 ] but the precocious puberty is more common in females (85%) and less common in males (10–15%).[ 16 ]

The genetic mutation is located in the chromosome 20q13 in the GNAS locus. The substitution of arginine for cysteine or histidine leads to a hyperactivation of the GSα−cyclic AMP. The overproduction and increased concentration of cAMP in bones over activate the proliferation and abnormal differentiation.[ 11 20 ] The elevated cAMP levels lead to an inflammatory cytokines response, in special the interleukin-6, and lead to this osteoclast differentiation.[ 25 ] This mutation can be also seen in hypersecretive thyroid tumors, Leydig cell tumors, and more than 40% of the secreting pituitary adenomas. The GNAS locus is a complex imprinted gene that generates multiple products, and the GSα is encoded by multiple exons (1–13) of the GNAS locus, most of them biallelic.[ 23 ] However, heterozygous mutations in maternal allele can lead to MAS with resistance for parathyroid hormone, thyroid-stimulating hormone, and gonadotropin, whereas mutation in paternal allele leads to MAS alone.[ 14 ] Genetic studies are important due to the differential diagnosis of the FD and other FD-like diseases, such as low-grade central osteosarcoma, which presents a low-incidence of GNAS mutation.[ 14 18 23 ]

The macroscopic white and brown aspect is typical, and microscopically there is a replace of marrow space by soft fibrous tissue composed of bland spindle cells and abnormal osseous component with irregular trabeculae.

Radiological findings are typical but not pathognomonic and can be divided into three groups:

Pagetoid pattern: The rate of bone-fibrous matrix is equal

Sclerotic pattern: The bone structure is in foreground

Radiolucent pattern: Fibrous matrix in the foreground, with cystic degeneration (simple or aneurysmal).[ 4 12 ]

During investigation, three differential diagnosis can be considered in skull lesions: Osteosarcoma, en plaque meningioma, and osteitis.[ 4 ]

When limited to the craniofacial region, it is considered to be the monostotic form, even if more than one bone is affected, because it is one focus of the disease.[ 19 ] The facial FD occurs more frequently in the unilateral form than the bilateral form, in both gender and equal side percentage.[ 24 ] Chen and Noordhoff[ 7 ] proposed a surgical classification by zones of the deformity, and indication of intervention based on functional or reconstructive criteria.[ 6 7 ] The most common symptoms are facial asymmetry (85%), orbital/facial mass (60%), blurred vision (24%), and eyelid position disturbance (10%).[ 15 ] Pain complaint is less frequent in children and more in adults, and female patients experience an increased pain level during pregnancy and during the menstrual cycle because of the estrogen receptors found in FD.[ 10 15 ] The pain management can be clinical for the persistent, severe pain; endovenous treatment with bisphosphonates is indicated. Oral bisphosphonates and alendronate have been shown to be ineffective for treatment of bone pain.[ 5 ]

Malignant transformation in FD is very rare (about 0.4–4%) and more common in the polyostotic disease and the histological types such as osteosarcoma, fibrosarcoma, and chondrosarcoma.[ 13 ]

Recent studies show that the narrowing of the optic canal is not directly correlated to visual loss,[ 14 19 ] even if the optic nerve is 100% evolved by fibrous bone.[ 21 ] Rahman et al. showed that most of the patients with optic canal stenosis did not progress to optic neuropathy, and about 40% will have a chronic visual loss in a long-term follow-up; also, just a few of them will have optic neuropathy progression. This data show that patients with FD without signs of aneurysmal bone cyst or mucoceles have a stable progression. On the other hand, in patients with excessive hormone drive, it tends to be more aggressive and recurrent, such as in MAS and other growth hormone-producing tumors.

Visual loss tends to be chronic and acute visual loss is related to aneurysmal bone cysts and mucoceles. Despite the low plasticity of the nerve, urgent decompression of the optic nerve can reverse it.[ 21 ] The real mechanism of the lesion is unclear and multifactorial, related to neuronal changes and blood flow.[ 1 2 ] In addition, it is known that the initial compression of the optic nerve can lead to a partially reversible demyelination, however, it could also lead to an irreversible conduction blockage and a Wallerian degeneration due to direct compression or traction.[ 8 17 ] This theory is congruent to justify the postoperative surgical complication of visual loss worsening. This important surgical complication might be considered for the decision of surgery, specially with the patients that could undergo to a prophylactic measure.[ 9 ]

The operative management of craniofacial dysplasia in MAS is not well-established because it is a rare syndrome with few case reports in long-term follow-up, with variable outcomes and timing of intervention.[ 6 7 14 19 ]

Surgical technique of decompression can be divided into two subtypes: Conservative decompression and radical surgery. The conservative surgery consists in shaping the dysplastic bone, and it is often repeated over time with the objective of postponing the radical surgery, if needed. The radical approach consists in the radical removal of dysplastic bone and reconstruction with autologous bone graft.[ 19 ] Moreover, the prophylactic surgery for the optic nerve is reserved for the lesions that could lead to acute visual loss such as the ones related to aneurysmal bone cysts and mucoceles.[ 9 ]

Postoperative complications occur in 50% of patients and include infections, binocular diplopia, cranial nerve palsy, pain, epistaxis, and ectropion.[ 19 ] Prophylactic decompression is controversial, and the removal of the dysplastic bone increases the risk of surgical visual loss complication.

CONCLUSION

MAS is a rare subtype of FD with endocrinopathy. The FD of the orbital region can lead to optic nerve compression and possibly to visual loss. Nowadays, there are no evidences that support the benefits of prophylactic surgery in children with normal visual fields and optic canal narrowing, shown by the CT or MRI, and there is no method to predict which child will stabilize or deteriorate the visual loss. The cystic degenerations can lead to sudden visual loss and is the only possible indication for prophylactic surgery, but the risk of nerve damage should be considered and well-explained. In all other cases of normal visual fields and CT/MRI optic canal narrowing, prophylactic surgery is not indicated, and follow-up should be done. On the other hand, early decompression of symptomatic children is a great standard for a better chance of visual loss reverse.

The surgical technique is not well-established and depends on the case and surgeon expertise, but the conservative approach seems to be more adequate and multiple interventions might be required, especially in McCune–Albright and polyostotic lesions.

Declaration of patient consent

The authors certify that they have obtained all appropriate patient consent forms. In the form the patient(s) has/have given his/her/their consent for his/her/their images and other clinical information to be reported in the journal. The patients understand that their names and initials will not be published and due efforts will be made to conceal their identity, but anonymity cannot be guaranteed.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Abe T, Sato K, Otsuka T, Kawamura N, Shimazu M, Izumiyama H. Optic nerve decompression for orbitofrontal fibrous dysplasia. Skull Base. 2002. 12: 145-52

2. Abe T, Satoh K, Wada A. Optic nerve decompression for orbitofrontal fibrous dysplasia: Recent development of surgical technique and equipment. Skull Base. 2006. 16: 145-55

3. Albright F, Butler A, Hampton A, Smith P. Syndrome characterized by osteitis fibrosa disseminata, areas of pigmentation and endocrine dysfunction, with precocious puberty in females: Report of five cases. N Engl J Med. 1937. 216: 727-46

4. Bousson V, Rey-Jouvin C, Laredo JD, Le Merrer M, Martin-Duverneuil N, Feydy A. Fibrous dysplasia and McCune–Albright syndrome: Imaging for positive and differential diagnoses, prognosis, and follow-up guidelines. Eur J Radiol. 2014. 83: 1828-42

5. Boyce AM, Kelly MH, Brillante BA, Kushner H, Wientroub S, Riminucci M. A randomized, double blind, placebo-controlled trial of alendronate treatment for fibrous dysplasia of bone. J Clin Endocrinol Metab. 2014. 99: 4133-40

6. Chen YR, Breidahl A, Chang CN, Kurimoto M, Endo S, Onizuka K. Extradural optic nerve decompression for fibrous dysplasia with a favourable visual outcome. Neurol Med Chir (Tokyo). 1996. 36: 102-5

7. Chen YR, Noordhoff MS. Treatment of craniomaxillofacial fibrous dysplasia: How early and how extensive?. Plast Reconstr Surg. 1990. 86: 835-42

8. Clifford-Jones RE, Landon DN, McDonald WI. Remyelination during optic nerve compression. J Neurol Sci. 1980. 46: 239-43

9. Cruz AA, Constanzi M, de Castro FA, dos Santos AC. Apical involvement with fibrous dysplasia: Implications for vision. Ophthal Plast Reconstr Surg. 2007. 23: 450-4

10. DiCaprio MR, Enneking WF. Fibrous dysplasia. Pathophysiology, evaluation, and treatment. J Bone Joint Surg Am. 2005. 87: 1848-64

11. Dumitrescu CE, Collins MT. McCune-Albright syndrome. Orphanet J Rare Dis. 2008. 3: 12-

12. Hanifi B, Samil KS, Yasar C, Cengiz C, Ercan A, Ramazan D. Craniofacial fibrous dysplasia. Clin Imaging. 2013. 37: 1109-15

13. Harris WH, Dudley HR, Barry RJ. The natural history of fibrous dysplasia, an orthopaedic, pathological and roentgenographic study. Am J Orthop. 1962. 44: 207-33

14. Lee JS, FitzGibbon E, Butman JA, Dufresne CR, Kushner H, Wientroub S. Normal vision despite narrowing of the optic canal in fibrous dysplasia. N Engl J Med. 2002. 347: 1670-6

15. McCarthy EF. Fibro-osseous lesions of the maxillofacial bones. Head Neck Pathol. 2013. 7: 5-10

16. McCune DJ. Osteita fibrosa cystica: The case of nine-year-old girl who also exhibits precocious puberty, multiple pigmentation of the skin and hyperthyroidism. Am J Dis Child. 1936. 52: 743-4

17. McDonald WI. The symptomatology of tumours of the anterior visual pathways. Can J Neurol Sci. 1982. 9: 381-90

18. Pollandt K, Engels C, Kaiser E, Werner M, Delling G. Gsalpha gene mutations in monostotic fibrous dysplasia of bone and fibrous dysplasia-like low-grade central osteosarcoma. Virchows Arch. 2001. 439: 170-5

19. Rahman AM, Madge SN, Billing K, Anderson PJ, Leibovitch I, Selva D. Craniofacial fibrous dysplasia: Clinical characteristics and long-term outcomes. Eye (Lond). 2009. 23: 2175-81

20. Riminucci M, Fisher LW, Shenker A, Spiegel AM, Bianco P, Gehron Robey P. Fibrous dysplasia of bone in the McCune-Albright syndrome: Abnormalities in bone formation. Am J Pathol. 1997. 151: 1587-600

21. Sammut SJ, Kandasamy J, Newman W, Sinha A, Ross J, Blair JC. Relief of severe retro-orbital pain and vision improvement after optic-nerve decompression in polyostotic fibrous dysplasia: Case report and review of the literature. Childs Nerv Syst. 2008. 24: 515-20

22. Siegal G, Dal Cin P, Araujo ES, Fletucher DM, Unni KK, Mertens F.editors. Fibrous dysplasia. WHO Classification of Tumors of Soft Tissue and Bone. Lyon: IARC Press; 2002. p. 341-2

23. Vasilev V, Daly AF, Thiry A, Petrossians P, Fina F, Rostomyan L. McCune-Albright syndrome: A detailed pathological and genetic analysis of disease effects in an adult patient. J Clin Endocrinol Metab. 2014. 99: E2029-38

24. Wu H, Yang L, Li S, Jin X, Xu J, Lu J. Clinical characteristics of craniomaxillofacial fibrous dysplasia. J Craniomaxillofac Surg. 2014. 42: 1450-5

25. Yamamoto T, Ozono K, Kasayama S, Yoh K, Hiroshima K, Takagi M. Increased IL-6-production by cells isolated from the fibrous bone dysplasia tissues in patients with McCune-Albright syndrome. J Clin Invest. 1996. 98: 30-5

Pediatric knowledge update: Approach to the management of vein of Galen aneurysmal malformations in neonates

$
0
0
Daniel Hansen, Peter T. Kan, Gaddum D. Reddy, Arvind Chintagumpala Mohan, Andrew Jea, Sandi Lam
  1. Department of Neurosurgery, Division of Pediatric Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA

Correspondence Address:
Sandi Lam
Department of Neurosurgery, Division of Pediatric Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA

DOI:10.4103/2152-7806.182415

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Hansen D, Kan PT, Reddy GD, Mohan AC, Jea A, Lam S. Pediatric knowledge update: Approach to the management of vein of Galen aneurysmal malformations in neonates. Surg Neurol Int 13-May-2016;7:

How to cite this URL: Hansen D, Kan PT, Reddy GD, Mohan AC, Jea A, Lam S. Pediatric knowledge update: Approach to the management of vein of Galen aneurysmal malformations in neonates. Surg Neurol Int 13-May-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/pediatric-knowledge-update-approach-to-the-management-of-vein-of-galen-aneurysmal-malformations-in-neonates/

Date of Submission
27-Nov-2015

Date of Acceptance
26-Mar-2016

Date of Web Publication
13-May-2016

Abstract

Keywords: Neonate, pediatric, vascular, vein of Galen aneurysmal malformation, vein of Galen malformation

CASE EXAMPLE

A newborn female born via cesarean section at 39 weeks gestation was transferred to our institution for severe respiratory distress after delivery. She was intubated and required 100% fraction of inspired oxygen to maintain adequate saturations. She was also started on vasopressor medications secondary to hypotension. An echocardiogram of the heart showed diastolic flow reversal in the transverse aortic arch and increased flow in the superior vena cava. A bruit was auscultated over the anterior fontanelle that was concerning for an intracranial vascular malformation. Bedside, head ultrasound confirmed the presence of a vein of Galen malformation. Her liver was enlarged without signs of liver failure. Her anuria resolved with improved renal function after her hypotension was treated. Electroencephalography was negative for seizure activity. Her Bicêtre score was calculated to be between 9 and 11. As per the Lasjaunias algorithm, the baby was a candidate for emergent endovascular embolization.

Cerebral angiogram revealed an extensive choroidal type vein of Galen malformation with severe arteriovenous shunting [Figures 1 and 2 ]. Partial embolization was achieved using a combination of platinum coils and the liquid embolic agent Onyx, with a significant reduction in posttreatment arteriovenous shunting. Posttreatment echocardiogram showed improvement in the left ventricular function. Repeated partial embolizations were performed 3 more times over the next 3 months [ Figure 3 ]. Ventriculomegaly remained stable on serial imaging. Her cardiac function as assessed by her brain natriuretic peptide normalized. Her pulmonary hypertension steadily improved, allowing her to be discharged to home on room air.


Figure 1

First embolization treatment. Right internal carotid injection. Pretreatment (top) and posttreatment (bottom). Blue arrows highlighting arterial feeders pre- and post-coil embolization. Note slightly decreased contrast filling of venous outflow after coil placement, highlighted by red arrows

 

Figure 2

Magnetic resonance imaging of the brain without contrast. Sagittal T1 (left) sequence and axial T2 (right) sequence highlighting dilated vessels (arrows) converging to the vein of Galen varix, dilatations of the draining straight sinus

 

Figure 3

Third and fourth embolization treatments. Left vertebral injection. Pretreatment (top) and posttreatment (bottom). Multiple coil constructs (blue arrows) are now visible with further reduction in contrast filling of the venous outflow (red arrows)

 

BACKGROUND

Neonatal vein of Galen aneurysmal malformations (VGAMs) are rare; the incidence is approximately 1 in 25,000.[ 13 17 22 ] The most common presentation of VGAM as a neonate is high-output congestive heart failure, which carries high rates of morbidity and mortality.[ 6 ] The age of symptomatic onset is often characterized by a distinct clinical presentation: Neonates usually present with high-output cardiac failure; infants present with increasing head circumference and hydrocephalus; toddlers present with developmental delay, hydrocephalus, and/or seizures; and older children can have subarachnoid hemorrhage, headache, and/or seizures.[ 1 21 ] Reviewing treatment outcomes by age shows that mortality affects 37% of neonates following endovascular treatment, and 6.5% and 3.2%, respectively, of infants and children following treatment.[ 1 10 15 ]

VGAM represents an embryonic arteriovenous shunt in the subarachnoid space and the choroidal fissure. Steinheil first described this entity in 1895, but it was Boldrey and Miller in 1949 who first documented a true form of VGAM by identifying multiple arteriovenous communications draining into the dilated vein of Galen.[ 1 ] In 1989, Rayboud traced the anatomy to a persistent embryological precursor to the vein of Galen: The median prosencephalic vein of Markowski (MProsV).[ 12 ] Lasjaunias classified VGAM into two anatomical forms, a mural form (direct fistula from MProsV) and a choroidal form (multiple choroidal arteries contributing to an intermediary network before drainage into the large venous pouch), the predominant form.[ 16 ]

The diagnosis can be made in utero by ultrasound and can be identified between the 6th and 11th week of gestation. Fetal magnetic resonance imaging can better evaluate cerebral atrophy, ventricular size, and cardiac insufficiency. Fetal distress presents as cardiac insufficiency and/or hydrops. Neonatal diagnosis is typically made after identifying severe cardiopulmonary distress during or acutely after birth, along with the associated pulmonary, neurological, hepatic, and renal dysfunction.[ 9 ]

Before the advent of endovascular interventions, mortality in neonatal VGAM was nearly 90%.[ 19 ] With the introduction of embolization techniques and advanced neonatal critical care for severe cardiopulmonary illness, mortality has decreased to around 50%.[ 19 ] Immediate multidisciplinary neonatal assessment and care are clearly imperative.

In 2006, Lasjaunias described his large experience with VGAM management. The primary and immediate goal is stabilization of the life-threatening congestive heart failure. The next step is to attempt treatment of the VGAM. He recommends a staged approach to better manage the risk that comes with the instant flow reversal with obliteration of the VGAM. Any presenting hydrocephalus is not treated until after treatment of the VGAM to avoid worsening flow through the VGAM, which can occur with lowering of intracranial pressure.[ 5 10 16 ]

EVALUATION AND TREATMENT

Initial consultation should include a thorough neurological examination, bedside electroencephalography, auscultation for cranial bruit, measurement of head circumference, and signs or symptoms of hydrocephalus. Although the immediate clinical concern is cardiac failure, VGAM has direct cerebral consequences including encephalopathy, hydrocephalus, seizures, and developmental delay.

One option for evaluation of the newborn with VGAM is based on the Bicκtre score to determine potential treatment options.[ 10 16 ] This 21-point scale gives points for the severity of signs and symptoms pertaining to the cardiac, pulmonary, neurological, hepatic, and renal systems [ Table 1 ]. Clinical and laboratory values are used to calculate the Bicêtre score for the neonate presenting with VGAM. A score of <8 out of 21 suggests a near fatal prognosis, and the infant is considered too unstable for emergent embolization. A score between 8 and 12 characterizes neonates who are most likely to benefit from emergent embolization. A score >12 suggests infants who are candidates for medical management of their cardiopulmonary insufficiency. Medical management is continued until around 5 months of age when their larger size decreases the risks of extended embolization. Figure 4 summarizes this treatment algorithm.


Table 1

Bicêtre score

 

Figure 4

Treatment algorithm

 

Cardiology assessment of cardiac function and superior vena cava flow should proceed right away. An echocardiogram should be performed to assess for the function of all the cardiac chambers, the presence of a pulmonary ductus arteriosus shunt, degree of pulmonary arterial pressure (measured indirectly with the velocity of flow through the insufficient tricuspid valve), and other cardiac anomalies such as atrial septal defect and aortic coarctation.

Potential medical treatments are aimed at controlling the heart's left-to-right shunting caused by the VGAM. Nitric oxide can aid with persistent pulmonary hypertension. Other pharmacologic agents include diuretics and beta agonists such as dopamine, inotropes, prostaglandins, or phosphodiesterase inhibitors.[ 8 ] No guidelines for medical treatment exist, as no clinical trials have been conducted to date. If the diagnosis is made antenatally, one study has suggested that dosing maternal digoxin may result in concurrent fetal dosing and possible treatment while in utero.[ 3 ]

Laboratory tests should evaluate liver and renal function, in addition to serving as secondary markers of cardiac and pulmonary status. Brain natriuretic protein levels act as a marker for cardiac injury due to pulmonary hypertension.[ 4 ] Serum tests for coagulopathy and ultrasound examinations of size assess liver function, while volume of urine output and blood urea nitrogen/serum creatinine levels aid in determining the systemic impact of the heart failure.

TREATMENT

Treatment options for VGAMs have historically included either open surgery, a direct transtorcular approach to ligation, transarterial embolization, or transvenous embolization. Reports show nearly 100% mortality with open surgical approaches; transtorcular approaches have, likewise, not been successful; and transvenous approaches are reported to have poor cognitive outcomes.[ 11 ] Thus, their use has been relegated to second-line options, for use when a transarterial approach cannot be achieved.

Transarterial embolization, in a staged fashion, for partial occlusion of the VGAM is the only current treatment that has been shown to result in a safe reduction of cardiac failure. Access in neonates is often found through the umbilical artery. Protecting this route of access during birth and the acute postnatal period is important. This artery has the benefit of providing adequate access for the needed endovascular catheters, while sparing trauma to the femoral artery in small neonates. It is essential to maintain the patency of this vessel and utilize other routes for serial blood work.[ 9 ]

Embolization aims to occlude, or partially occlude, the arterial side of the fistula. Currently, there are two primary materials used: (1) N-butyl-cyanoacrylate (NBCA) or (2) Onyx liquid, an ethylene vinyl alcohol copolymer that is delivered in dimethyl sulfoxide (DMSO) solvent. Once in contact with an ionic substance (blood), the DMSO quickly dissolves, leaving a firm spongy substance that rapidly occludes the vessel. It is admixed with tantalum powder to render it visible on fluoroscopy and computed tomography. This gives it a dark purple/black appearance in vivo. The use of Onyx has been largely described in the adult population, but there is a growing body of literature supporting its use as safe in pediatric patients when used by experienced practitioners.[ 2 23 24 ]

The extreme flow through the VGAM is a major concern during endovascular treatment. Injection of the embolic material is difficult to control at such high flow rates and can lead to the NBCA or Onyx passing through the fistula without successfully depositing in the VGAM. There are three general techniques to deal with this problem.First, coils can be placed distally in the arterial pedicle to catch the Onyx or NBCA.[ 18 ] Second, Onyx can be injected through a dual lumen balloon catheter, with the balloon inflated in the distal arterial pedicle to arrest flow, which allows the Onyx to form. However, Care must be taken to not dislodge the Onyx cast once the balloon is deflated. Despite this risk, the double-balloon technique has been shown to be an effective treatment option.[ 20 ] Finally, a mixture of 80–100% NBCA can be used so that it polymerizes immediately on contact with blood. However, it is more difficult to visualize fluoroscopically and can become glued to the catheter; the development of detachable catheter tips has helped to reduce this potential problem.

After treating the primary problem of the VGAM, there may still be associated hydrocephalus to be addressed. Diagnosis is made by a combination of anterior fontanelle evaluation, head circumference measurements, and ultrasound assessment of ventricular caliber; more advanced imaging modalities may be used as well. The purported causes for the development of hydrocephalus in this setting include ineffective cerebrospinal fluid (CSF) reabsorption due to venous stenosis outside the VGAM, underdeveloped arachnoid granulations, and aqueductal obstruction resulting from the mass effect of the VGAM. Treatment should not precede control of the cardiac failure and VGAM.[ 3 ] CSF shunting in the setting of a persistent VGAM exacerbates the low vascular resistance and can lead to intracerebral hemorrhage or worsening of flow through the lesion.[ 13 21 ] Any considered interventions should be performed secondarily if needed.

OUTCOMES

VGAM treated in neonates carries significant risks. The largest series from Lasjaunias at the Hospital Bicêtre determined that of the 216 patients treated with endovascular embolization, 23 (10.6%) died, 20 (10.5%) suffered severe intellectual disabilities, and 30 (15.6%) were moderately intellectually disabled. The remaining 143 (74%) were neurologically normal at follow-up.[ 16 ] Frawley et al. reported similar outcomes with nine patients with follow-up times from 6 months to 4 years.[ 6 ] Others have reported that congestive heart failure, perinatal presentation, and choroidal angioarchitecture showed the worst outcomes.[ 7 ] In another series of 13 patients with VGAM treated between 1987 and 2001, two of five patients with choroidal malformations died, and one had a significant impairment; three of seven children with mural lesions died, and four were neurologically normal after treatment.[ 14 ]

The management of VGAM is evolving, and the addition of transarterial embolization techniques is allowing increased treatment of these complex lesions with improving outcomes. Nevertheless, overall prognosis remains poor. Even with recent technological advances, a large population of patients remains untreatable or has poor long-term outcomes. Moving forward, there is a need for prospective studies, clinical registries, and ideally, a randomized controlled trial to determine the best medical and interventional treatments in these complex neonates.

At our institution, our collaborative multidisciplinary team follows the diagnostic and treatment algorithm outlined in Figure 4 . For an infant with VGAM, neonatology, pediatric neurosurgery, pediatric neurology, neurointerventional endovascular, and pediatric cardiology services all combine to provide a tailored and individualized response to this challenging problem.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Alvarez H, Garcia Monaco R, Rodesch G, Sachet M, Krings T, Lasjaunias P. Vein of Galen aneurysmal malformations. Neuroimaging Clin N Am. 2007. 17: 189-206

2. Ashour R, Aziz-Sultan MA, Soltanolkotabi M, Schoeneman SE, Alden TD, Hurley MC. Safety and efficacy of Onyx embolization for pediatric cranial and spinal vascular lesions and tumors. Neurosurgery. 2012. 71: 773-84

3. Berenstein A, Fifi JT, Niimi Y, Presti S, Ortiz R, Ghatan S. Vein of Galen malformations in neonates: New management paradigms for improving outcomes. Neurosurgery. 2012. 70: 1207-13

4. Chevret L, Durand P, Alvarez H, Lambert V, Caeymax L, Rodesch G. Severe cardiac failure in newborns with VGAM.Prognosis significance of hemodynamic parameters in neonates presenting with severe heart failure owing to vein of Galen arteriovenous malformation. Intensive Care Med. 2002. 28: 1126-30

5. Foran A, Donohue V, McParland P, Lynch B, Lasjaunias P, Rodesch G. Vein of Galen aneurysm malformation (VGAM): Closing the management loop. Ir Med J. 2004. 97: 8-10

6. Frawley GP, Dargaville PA, Mitchell PJ, Tress BM, Loughnan P. Clinical course and medical management of neonates with severe cardiac failure related to vein of Galen malformation. Arch Dis Child Fetal Neonatal Ed. 2002. 87: F144-9

7. Fullerton HJ, Aminoff AR, Ferriero DM, Gupta N, Dowd CF. Neurodevelopmental outcome after endovascular treatment of vein of Galen malformations. Neurology. 2003. 61: 1386-90

8. Gailloud P, O’riordan DP, Burger I, Lehmann CU. Confirmation of communication between deep venous drainage and the vein of Galen after treatment of a vein of Galen aneurysmal malformation in an infant presenting with severe pulmonary hypertension. AJNR Am J Neuroradiol. 2006. 27: 317-20

9. Gailloud P, O’Riordan DP, Burger I, Levrier O, Jallo G, Tamargo RJ. Diagnosis and management of vein of Galen aneurysmal malformations. J Perinatol. 2005. 25: 542-51

10. Gold A, Ransohoff J, Carter S. Vein of Galen malformation. Acta Neurol Scand Suppl. 1964. 40: S1-31

11. Gupta AK, Varma DR. Vein of Galen malformations: Review. Neurol India. 2004. 52: 43-53

12. Iizuka Y, Kakihara T, Suzuki M, Komura S, Azusawa H. Endovascular remodeling technique for vein of Galen aneurysmal malformations – Angiographic confirmation of a connection between the median prosencephalic vein and the deep venous system. J Neurosurg Pediatr. 2008. 1: 75-8

13. Jea A, Bradshaw TJ, Whitehead WE, Curry DJ, Dauser RC, Luerssen TG. The high risks of ventriculoperitoneal shunt procedures for hydrocephalus associated with vein of Galen malformations in childhood: Case report and literature review. Pediatr Neurosurg. 2010. 46: 141-5

14. Jones BV, Ball WS, Tomsick TA, Millard J, Crone KR. Vein of Galen aneurysmal malformation: Diagnosis and treatment of 13 children with extended clinical follow-up. AJNR Am J Neuroradiol. 2002. 23: 1717-24

15. Khullar D, Andeejani AM, Bulsara KR. Evolution of treatment options for vein of Galen malformations. J Neurosurg Pediatr. 2010. 6: 444-51

16. Lasjaunias PL, Chng SM, Sachet M, Alvarez H, Rodesch G, Garcia-Monaco R. The management of vein of Galen aneurysmal malformations. Neurosurgery. 2006. 59: S184-94

17. Li AH, Armstrong D, terBrugge KG. Endovascular treatment of vein of Galen aneurysmal malformation: Management strategy and 21-year experience in Toronto. J Neurosurg Pediatr. 2011. 7: 3-10

18. Li MH, Li WB, Fang C, Gao BL. Transarterial embolization with berenstein liquid coils and N-butyl cyanoacrylate in a vein of Galen aneurysmal malformation: A case report. Korean J Radiol. 2007. 8: 164-8

19. Malhotra RK, Florez L, White D, Papasozomenos S, Covinsky M, Bhattacharjee M. Vein of Galen aneurysmal malformation associated with high output cardiac failure in three neonates. J Neuropathol Exp Neurol. 2007. 66: 438-

20. Pop R, Manisor M, Wolff V, Kehrli P, Marescaux C, Beaujeux R. Flow control using Scepter™ balloons for Onyx embolization of a vein of Galen aneurysmal malformation. Childs Nerv Syst. 2015. 31: 135-40

21. Schneider SJ, Wisoff JS, Epstein FJ. Complications of ventriculoperitoneal shunt procedures or hydrocephalus associated with vein of Galen malformations in childhood. Neurosurgery. 1992. 30: 706-8

22. Stephan S, Rodesch G, Elolf E, Wiemann D, Jorch G. Vein of Galen aneurysmal malformations: An ultrasonographic incidental finding – A case report. Case Rep Pediatr 2012. 2012. p.

23. Thiex R, Williams A, Smith E, Scott RM, Orbach DB. The use of Onyx for embolization of central nervous system arteriovenous lesions in pediatric patients. AJNR Am J Neuroradiol. 2010. 31: 112-20

24. Zaidi HA, Kalani MY, Spetzler RF, McDougall CG, Albuquerque FC. Multimodal treatment strategies for complex pediatric cerebral arteriovenous fistulas: Contemporary case series at Barrow Neurological Institute. J Neurosurg Pediatr. 2015. 15: 615-24

Intracranial extra-axial hemangioma in a newborn: A case report and literature review

$
0
0
Marcos Dalsin, Rafael Sodré Silva, Jennyfer Paula Galdino Chaves, Francine Hehn Oliveira, Ápio Cláudio Martins Antunes, Leonardo Modesti Vedolin
  1. Department of Neurosurgery, Hospital de Clínicas de Porto Alegre, RS, Brazil
  2. Universidade Federal de Rio Grande, RS, Brazil
  3. Department of Pathology, Hospital de Clínicas de Porto Alegre, RS, Brazil
  4. Head of Neurosurgery Unit, Hospital de Clínicas de Porto Alegre, RS, Brazil
  5. Head os Radiology Unit, Hospital de Clínicas de Porto Alegre, RS, Brazil

Correspondence Address:
Marcos Dalsin
Head os Radiology Unit, Hospital de Clínicas de Porto Alegre, RS, Brazil

DOI:10.4103/2152-7806.182414

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Dalsin M, Rafael Sodré Silva, Galdino Chaves JP, Oliveira FH, Ápio Cláudio Martins Antunes, Vedolin LM. Intracranial extra-axial hemangioma in a newborn: A case report and literature review. Surg Neurol Int 13-May-2016;7:

How to cite this URL: Dalsin M, Rafael Sodré Silva, Galdino Chaves JP, Oliveira FH, Ápio Cláudio Martins Antunes, Vedolin LM. Intracranial extra-axial hemangioma in a newborn: A case report and literature review. Surg Neurol Int 13-May-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/intracranial-extra%e2%80%91axial-hemangioma-in-a-newborn-a-case-report-and-literature-review/

Date of Submission
02-Sep-2015

Date of Acceptance
06-Feb-2016

Date of Web Publication
13-May-2016

Abstract

Background:Congenital hemangiomas are benign vascular tumors, and the intracranial counterpart was described in very few cases.

Case Description:A newborn presented with an intracranial tumor associated with an arachnoid cyst, diagnosed by antenatal ultrasound at 37 weeks of gestation. Surgery was indicated due to increased head circumference and bulging fontanelle, and a complete resection of an extra-axial red–brown tumor was performed at the 3rd week of life. Microscopy revealed a hemangioma.

Conclusion:Hemangioma is a rare differential diagnosis that must be considered in extra-axial intracranial tumors affecting infants and neonates. The radiological features are not helpful in differentiating from other tumors, and surgery is indicated when the diagnosis is uncertain or whenever there are signs of increased intracranial pressure.

Keywords: Brain tumor, intracranial hemangioma, newborn

INTRODUCTION

Hemangioma is an uncommon benign vascular intracranial tumor, and the occurrence in newborns is extremely rare.[ 3 8 ] Despite being the most common tumor in childhood, hemangiomas rarely affect the central nervous system (CNS).[ 2 ] When it occurs, the clinical presentation is usually with increased head circumference and intracranial hypertension signals.[ 2 3 ] Although it can undergo spontaneous regression, surgical resection is the treatment of choice.[ 3 ]

CASE REPORT

A 30-year-old woman, with gestational diabetes and at second pregnancy, was admitted to the hospital at 38 weeks gestation, after diagnosis of a large ultrasound fetal brain tumor at 37 weeks gestation [ Figure 1 ]. She had been attending regular antenatal examination, including normal fetal ultrasound at 12, 22, 28, and 32 weeks of gestation, without a family history of brain tumor.


Figure 1

Fetal ultrasound at 37 weeks gestation. (a) Axial view showing a solid echogenic mass in the left hemisphere (18 mm). (b) Axial view showing a large cyst (51 mm × 38 mm × 63 mm) adjacent to the tumor

 

The patient, a full-term female baby, was delivered by cesarean section, weighing 4455 g with an Apgar score of 5–9. On physical examination at birth, there were no skin lesions, the head circumference was 37 cm (higher than 95th percentile), the anterior fontanelle was normotensive, and the neurologic examination was normal.

Magnetic resonance imaging (MRI) demonstrated a solid mass lesion with intense gadolinium enhancement in the left middle fossa, associated with a large cyst dislocating the left hemisphere and the brainstem [ Figure 2 ]. Due to progressive head enlargement and tense fontanelle, two transfontanel punctures on the 1st and 2nd weeks were performed.


Figure 2

Preoperative magnetic resonance imaging at 5 days of life: (a) Axial T2-weighted. (b) Coronal T2-weighted sequence demonstrating a lobulated heterogeneous solid mass (22 mm × 18 mm × 10 mm) associated with a large extra-axial cyst that compress and dislocate brainstem of the left hemisphere. (c) Axial T2/fluid attenuation inversion recovery-weighted sequence demonstrating hyperintense lesion. (d) Sagittal T1-weighted with gadolinium showing intense enhancement of the lesion after contrast. (e and f) Magnetic resonance angiography showing the intense relation of the tumor with the left middle cerebral artery and one branch inside the mass

 

A small temporal craniotomy was done at the 3rd week of life: An extra-axial red–brown tumor was completely removed after coagulation of a clear vascular pedicle and the cyst was communicated with the subarachnoid cisterns. Microscopy revealed a tumor showing juvenile forms of capillary hemangioma and canalization of most vessels [ Figure 3 ].


Figure 3

Low power view of tumor showing juvenile forms of capillary hemangioma and canalization of most vessels (left: H and E, ×4); high power view of tumor demonstrating plump endothelial that lines vascular spaces and mitotic figure (arrow) (right: H and E, ×40)

 

Postoperative MRI demonstrated complete resection of the tumor, but a residual extra-axial cyst [ Figure 4 ]. The patient had a satisfactory recovery without neurological deficits and was discharged at the 5th week of life with normal open fontanels, without signs of intracranial hypertension, and referred to outpatient follow-up.


Figure 4

Postoperative magnetic resonance imaging 7 days after surgery. Axial and coronal T2-weighted demonstrating complete resection of the lesion and persistence of large extra-axial cyst, although smaller than preoperatively

 

DISCUSSION

Epidemiology

In childhood, malignant brain tumors are the most common solid organ tumors, accounting approximately for 20–30% of the cancers in this age.[ 7 ] Large intracranial lesions in the 1st year of life are uncommon[ 2 ] and more likely to be primitive neuroectodermal tumors, desmoplastic infantile astrocytoma, teratomas, and choroid plexus papilloma.[ 2 7 ] Congenital brain tumors occurs in an incidence of 1.1 to 3.3 per 100.000 newborns and constitute <2% of all pediatric brain tumors.[ 8 ]

Hemangiomas are the most common tumors of infancy,[ 2 5 ] present congenitally or in early infancy,[ 6 ] and described as benign vascular tumor typically involving the skin of the face, scalp, neck, and chest.[ 2 5 6 ] Although a common skin incidence, hemangiomas involving the CNS are extremely rare,[ 2 6 8 ] with the prevalence of 0.37–0.57% in the overall pediatric population, even more rare in the 1st year of life.[ 8 ]

Clinical and radiological features

The most common clinical presentation in neonates is the enlargement of head circumference (62%) and/or signs of raised intracranial pressure such as vomiting and irritability (14%),[ 2 3 8 ] instead of cerebral cavernous malformations whose main symptoms are due to hemorrhage.[ 4 ] Seizures and other neurologic symptoms are less frequent.[ 8 ] During the perinatal period, early diagnosis is important and requires an obstetric ultrasound for monitoring.[ 8 ] After delivery, cranial ultrasound demonstrates a well-circumscribed echogenic solid mass,[ 3 ] and computed tomography and MRI are useful to demonstrate the brain tumor,[ 8 ] but are not helpful in differential diagnosis.[ 2 ] In the few reported cases, the lesions were well-defined, highly vascularized extra-axial lesions, with intense contrast enhancement, calcification areas,[ 2 8 ] and high-signal intensity on both T1-weighted and T2-weighted images in MRI.[ 8 ]

Anatomopathological considerations

Hemangioma is a CNS vascular neoplasm that may occur sporadically or exhibit autosomal dominant inheritance.[ 4 8 ] It is usually an extra-axial lesion, involving the dura mater, dural sinuses, ventricles, and tentorium,[ 2 3 ] often associated with cutaneous lesions,[ 8 ] and have the potential to grow and regress spontaneously over the time.[ 2 ] The differential diagnosis includes hemangioblastoma, hemangioendothelioma, and hemangiopericytoma.[ 6 ]

Macroscopically, the tumor is encapsulated with high-flow vascular arterial and venous pedicles. Histologically, hemangiomas are benign vascular neoplasms characterized by thin-walled endothelial-lined dilated spaces that can be filled with blood or thrombus.[ 2 8 ] The vascular nature can be confirmed by immunohistochemistry to CD31 and CD34.[ 3 6 ]

Prognosis and treatment

Because these lesions are extremely uncommon, the natural evolution is unclear.[ 2 ] Surgical resection is the treatment of choice[ 3 ] and should be indicated for large tumors and whenever signs of intracranial hypertension caused by tumor.[ 2 ] Spontaneous regression of extracranial hemangiomas during childhood is common,[ 1 ] so small intracranial tumors associated with cutaneous hemangiomas could be followed to watch for regression.[ 2 6 ] Tumor biopsy is unsafe because of the high vascularity of the tumor.[ 1 ]

CONCLUSION

Although rare, hemangioma is a differential diagnosis that must be considered in extra-axial intracranial tumors affecting infants. Because the radiological features are not helpful in differentiating other tumors, surgery is indicated when the diagnosis is uncertain or whenever there are signs of increasing mass effect and/or intracranial hypertension.

The termination of pregnancy in case of defects or serious diseases of the fetus is not permitted by Brazilian law. In this related case or similar cases, it would be a serious mistake because this lesion has a good prognosis.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Azam M, O’Donovan DJ. Intracranial cavernous hemangioma and seizures in a newborn infant. J Pediatr. 2009. 155: 298-

2. Jalloh I, Dean AF, O’Donovan DG, Cross J, Garnett MR, Santarius T. Giant intracranial hemangioma in a neonate. Acta Neurochir (Wien). 2014. 156: 1151-4

3. Karmazyn B, Michovitz S, Sirota L, Drozd T, Horev G. Intracranial cavernous hemangioma in a neonate. Pediatr Radiol. 2001. 31: 610-2

4. Mazza C, Scienza R, Beltramello A, Da Pian R. Cerebral cavernous malformations (cavernomas) in the pediatric age-group. Childs Nerv Syst. 1991. 7: 139-46

5. Restrepo R, Palani R, Cervantes LF, Duarte AM, Amjad I, Altman NR. Hemangiomas revisited: The useful, the unusual and the new. Part 1: Overview and clinical and imaging characteristics. Pediatr Radiol. 2011. 41: 895-904

6. Simon SL, Moonis G, Judkins AR, Scobie J, Burnett MG, Riina HA. Intracranial capillary hemangioma: Case report and review of the literature. Surg Neurol. 2005. 64: 154-9

7. Winn HR.editorsYoumans Neurological Surgery. Philadelphia: Elsevier-Saunders; 2011. 2: 2040-2

8. Yang CY, Hsu JF, Lin KL, Jung SM, Lien R, Chang YL. An extra-axial hemangioma mimicking a large prenatal brain tumor. Brain Dev. 2010. 32: 883-6

Hydrocephalus caused by unilateral foramen of Monro obstruction: A review on terminology

$
0
0
Flavio Nigri, Gabriel Neffa Gobbi, Pedro Henrique da Costa Ferreira Pinto, Elington Lannes Simões, Egas Moniz Caparelli-Daquer
  1. Department of Surgical Specialties, Neurosurgery Teaching and Assistance Unit, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
  2. Nervous System Electric Stimulation Laboratory (LabEEL) – Neurosurgery Teaching and Assistance Unit, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
  3. Physiological Sciences Department, Roberto Alcântara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil

Correspondence Address:
Flavio Nigri
Nervous System Electric Stimulation Laboratory (LabEEL) – Neurosurgery Teaching and Assistance Unit, Pedro Ernesto University Hospital, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
Physiological Sciences Department, Roberto Alcântara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil

DOI:10.4103/2152-7806.182392

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Nigri F, Gobbi GN, Gobbi GN, Elington Lannes Simões, Caparelli-Daquer EM. Hydrocephalus caused by unilateral foramen of Monro obstruction: A review on terminology. Surg Neurol Int 13-May-2016;7:

How to cite this URL: Nigri F, Gobbi GN, Gobbi GN, Elington Lannes Simões, Caparelli-Daquer EM. Hydrocephalus caused by unilateral foramen of Monro obstruction: A review on terminology. Surg Neurol Int 13-May-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/hydrocephalus-caused-by-unilateral-foramen-of-monro-obstruction-a-review-on-terminology/

Date of Submission
09-Dec-2015

Date of Acceptance
16-Feb-2016

Date of Web Publication
13-May-2016

Abstract

Background:Hydrocephalus caused by unilateral foramen of Monro (FM) obstruction has been referred to in literature by many different terminologies. Precise terminology describing hydrocephalus confined to just one lateral ventricle has a very important prognostic value and determines whether or not the patient can be shunt free after an endoscopic procedure.

Methods:Aiming to define the best term for unilateral FM obstruction, 19 terms were employed on PubMed database (http://www.ncbi.nlm.nih.gov/pubmed) as quoted phrases.

Results:A total of 194 articles were found. Four patterns of hydrocephalus were discriminated as a result of our research term query and were divided by types for didactic purpose. Type A - partial dilation of the lateral ventricle; Type B - pure unilateral obstruction of the FM; Type C - previously shunted patients with secondary obstruction of the FM; and Type D - asymmetric lateral ventricles with patent FM.

Conclusion:In unilateral FM obstruction hydrocephalus, an in-depth review on terminology application is critical to avoid mistakes that may compromise comparisons among different series. This terminology review suggests that Type B hydrocephalus, i.e., the hydrocephalus confined to just one lateral ventricle with no other sites of cerebrospinal fluid circulation blockage, are best described by the terms unilateral hydrocephalus (UH) and monoventricular hydrocephalus, the first being by far the most popular. Type A hydrocephalus is best represented in the literature by the terms uniloculated hydrocephalus and loculated ventricle; Type C hydrocephalus by the terms isolated lateral ventricle and isolated UH; and Type D hydrocephalus by the term asymmetric hydrocephalus.

Keywords: Foramen of Monro, isolated lateral ventricle, monoventricular hydrocephalus, unilateral hydrocephalus

INTRODUCTION

Unilateral ventricle dilation, unrelated to ex-vacuum hemisphere atrophy,[ 68 ] is caused by unilateral foramen of Monro (FM) obstruction.[ 22 80 ] It may be just an obstructed compartment within a more complex hydrocephalus or a unique compartment with hydrocephalus. Hydrocephalus caused by unilateral FM obstruction has been referred to in literature by many different terms. Precise terminology describing hydrocephalus confined to just one lateral ventricle has a very important prognostic value and determines whether or not the patient can be shunt free after endoscopic procedure.[ 32 36 ] Here, we discuss terminology for unilateral FM obstruction hydrocephalus based on literature review.

METHODS

The terms asymmetric hydrocephalus (AH), asymmetric lateral ventricle, asymmetric lateral ventricles, asymmetric ventricle, asymmetric ventricles, compartmentalized hydrocephalus, isolated lateral ventricle (ILV), isolated unilateral hydrocephalus (IUH), isolated ventricle, isolated ventricles, loculated hydrocephalus, loculated lateral ventricle, loculated ventricle (LV), monoventricular hydrocephalus (MH), trapped lateral ventricle, trapped ventricle, UH, unilateral ventriculomegaly, and uniloculated hydrocephalus (ULH) were employed on PubMed database (http://www.ncbi.nlm.nih.gov/pubmed) as quoted phrases. Only articles with clear described cases were selected. Review articles were excluded. Only human articles were considered. Bilateral FM obstruction articles were not included.

RESULTS

A total of 194 articles were found [ Table 1 ]. As depicted on Figure 1 , four general patterns of hydrocephalus were discriminated as a result of our research terms query and were divided by types for didactic purpose. Type A - partial dilation of the lateral ventricle, i.e., ventricular septa, ventricular cyst, or trigone obstruction; Type B - unilateral obstruction of the FM; Type C - previously shunted patients with secondary obstruction of the FM; and Type D - hydrocephalus with asymmetric lateral ventricles and patent FM.


Table 1

Literature review listing terminology as quoted phrases; number of articles found (n); ventricles involved in terminology description; if the term always described cases with hydrocephalus; and if the terms are used to describe heart ventricles

 

Figure 1

Schematic view of four different types of hydrocephalus described in the literature by the terms asymmetric hydrocephalus, isolated lateral ventricle, isolated unilateral hydrocephalus, loculated ventricle, monoventricular hydrocephalus, unilateral hydrocephalus, and uniloculated hydrocephalus. (a) Partial dilation of the lateral ventricle. (b) Exclusive unilateral obstruction of the foramen of Monro. (c) Secondary unilateral foramen of Monro obstruction in shunted patients. (d) Asymmetric lateral ventricles with patent foramen of Monro

 

DISCUSSION

The best term for pure unilateral FM obstruction shall be specific to describe hydrocephalus of the lateral ventricles at the foramina level and at the same time shall not be used to describe heart ventricles. The terms asymmetric lateral ventricle, asymmetric ventricle, loculated lateral ventricle, and trapped lateral ventricle disclosed no result [ Table 1 ]. The terms isolated ventricle and isolated ventricles are also used to describe heart ventricle disease [ Table 1 ].[ 55 86 ] The terms compartmentalized hydrocephalus,[ 61 ] loculated hydrocephalus,[ 6 ] and trapped ventricle[ 29 ] although specific to brain ventricles are not specific for lateral ventricle involvement [ Table 1 ]. The terms asymmetric lateral ventricles,[ 89 ] asymmetric ventricles,[ 9 ] and unilateral ventriculomegaly[ 68 ] not always describe hydrocephalus [ Table 1 ]. Then these terms did not seem appropriate for further discussion.

Obstruction of the FM causing dilation of ipsilateral ventricle admits several etiologies.[ 32 48 84 ] It may be the single component of the hydrocephalus, or it may also be associated with other sites of cerebrospinal fluid (CSF) circulation blockage. Among all terms applied, the terms AH, ILV, IUH, LV, MH, UH, and ULH have shown to describe hydrocephalus of the lateral ventricles and not heart ventricles [ Table 1 ]. For this reason, these terms were chosen for further discussion [ Table 2 ].


Table 2

Literature review listing terminology, type of hydrocephalus as depicted in Figure 1, and authors’ affiliation country

 

Asymmetric hydrocephalus

The term AH literally means the coexistence of hydrocephalus with asymmetric ventricles. It can occur in patients with hydrocephalus and different size ventricles caused by congenital or acquired unilateral brain atrophy.[ 1 ] Durfee et al., reserve the term AH for cases where asymmetrical dilation of the lateral ventricles has more than 2 mm, thus including patients sustaining bilateral and unilateral ventricular ectasia.[ 27 ] According to the literature [ Table 3 ], AH can mean any type of hydrocephalus depicted on Figure 1 . Although it can be used to describe hydrocephalus caused by FM obstruction, it does not exclude cases with patent FM.[ 10 46 ]


Table 3

Number of articles matching terminology (rows) with type of the hydrocephalus as depicted in Figure 1 (columns)

 

Isolated lateral ventricle

The term ILV literally means that the whole lateral ventricle is isolated from the rest of the ventricular system, including cases where patients also have other sites of CSF circulation obstruction. The literature review has shown that ILV has been employed to designate communicating hydrocephalus associated with postshunt FM obstruction,[ 7 39 73 76 ] pure lateral ventricle hydrocephalus[ 48 ] and also combining the two conditions.[ 36 ] Krucoff et al. used the term ILV as a UH synonym.[ 48 ]

Isolated unilateral hydrocephalus

The term IUH has been used to indicate that the hydrocephalus is confined to one brain side including complete or partial lateral ventricle dilation. In the literature, this term specifically refer to FM obstruction, but it is employed in situations when hydrocephalus is restricted[ 63 ] and also when not restricted[ 37 64 69 ] to the lateral ventricle [ Table 2 ].

Loculated ventricle

The term LV literally describes a compartment separated from the rest of the ventricular system. It has been applied on situations where there is a partial dilation of the lateral ventricle, like Type A depicted on Figure 1 .[ 64 70 ]

Monoventricular hydrocephalus

The term MH specifically defines a single lateral ventricle obstruction that by anatomical reasons can only indicate the involvement of the lateral ventricle, inasmuch as an obstruction of the third ventricle or fourth ventricles will necessarily cause biventricular and triventricular hydrocephalus respectively. Indeed, the term MH has been used to describe hydrocephalus restricted to one lateral ventricle, as shown in four articles [ Table 3 ]. However, Gangemi et al. included cases with FM obstruction associated with communicating hydrocephalus.[ 32 ] Furthermore, there is a logical preference for terms describing the number of ventricles involved, such as monoventricular, biventricular, triventricular, and tetraventricular hydrocephalus, as described in Mori and Raimondi's classification of hydrocephalus.[ 8 ] Cultural influence seems to play an important role in terminology choice. Eighty percent of articles using the term MH come from Latin language speaking countries [ Table 4 ].


Table 4

Distribution of terms appearing in articles dealing with foramen of Monro obstruction by country (from articles listed in Table 2)

 

Unilateral hydrocephalus

The term UH literally means that the hydrocephalus is confined to one brain side. It may indicate a complete or partial dilation of the lateral ventricle. Probably due to the universal use of English language [ Table 4 ] the term UH is by far the most commonly employed in Type B hydrocephalus [ Figure 1 ] as shown in fifty articles [ Table 3 ]. However, it has also been employed through literature in different types of hydrocephalus [ Table 3 ]. Brück et al.,[ 16 ] and Takeshita et al.,[ 78 ] used the term UH not describing FM obstruction, but in cases with partial dilation of the lateral ventricle. Cantini et al.,[ 19 ] and Lazareff and Sadowinski,[ 51 ] used the term UH for cases of hydrocephalus not restricted to the lateral ventricle. Nishizaki et al., described a case of biventricular hydrocephalus, which he designated left dominant UH.[ 60 ] Hageman et al., described an arthrogryposis newborn with left lateral ventricle dilation, midline shift, asymmetric head, and right hemiparesis.[ 35 ] Although it may be considered a case of UH, the abnormality was diagnosed as unilateral cerebral hypoplasia, so no treatment was indicated. Suzuki et al. described a case of unilateral hydranencephaly and named it UH.[ 77 ] The term bilateral hydrocephalus, albeit employing the same terminological criteria as UH, is not frequently employed.

Uniloculated hydrocephalus

The term ULH literally means that a ventricle is compartmentalized or is by itself a unique hydrocephalic compartment. El-Ghandour,[ 28 ] and Lewis et al.,[ 53 ] use the term ULH as a general term describing cases with partial dilation of the lateral ventricle, complete dilation of one lateral ventricle, and shunt complicated unilateral FM obstruction. Nowoslawska et al., use to describe a pure FM obstruction case [ Table 2 ].[ 61 ] Andresen and Juhler, proposed a very practical classification dividing hydrocephalus in loculations and its variants.[ 6 ] Loculated hydrocephalus is a condition in which discrete fluid-filled compartments form in or in relation to the ventricular system of the brain.[ 6 ] Applying Andresen and Juhler, classification in FM obstruction situations, pure FM obstruction [ Figure 1 , Type B] is termed “simple uniloculated hydrocephalus” and FM obstruction associated with communicating hydrocephalus [ Figure 1 , Type C] is classified as “complex uniloculated hydrocephalus.”[ 6 ]

Term meaning X application in the literature

Table 5 depicts the comparison of the literal meaning of a terminology and how this term is used in the literature. All the terms show a strong correlation between terminology meaning and application in the literature, but there is some misuse that may cause confusion.


Table 5

Comparison of the term meaning and its application in the literature

 

Endoscopic treatment of unilateral foramen of Monro obstruction hydrocephalus

Since 1994, endoscopy is the treatment of choice for Type A, Type B, and Type C hydrocephalus.[ 2 7 11 17 18 21 25 30 31 32 34 36 38 43 44 48 50 52 58 63 64 71 73 75 82 84 87 ] Many endoscopic techniques have been employed with high success rates such as septostomy,[ 2 7 17 18 25 30 31 32 34 36 38 48 50 52 58 64 71 73 75 82 84 87 ] open membranes,[ 11 34 58 ] lesion removal,[ 36 43 44 84 ] Monro foraminoplasty,[ 21 48 50 63 64 84 ] and cyst fenestration.[ 31 32 ] Some authors still prefer standards methods like shunt,[ 23 47 72 90 ] open craniotomy lesion removal and shunt,[ 83 ] and Rickham reservoir and shunt.[ 24 ] Endoscopic technique to communicate ventricular compartments is not indicate for Type D hydrocephalus where both FM are patent. On Type B hydrocephalus, patients are shunt free after endoscopic treatment of hydrocephalus.[ 2 11 17 18 21 30 31 32 34 38 43 44 48 52 58 75 82 84 87 ] On Type C hydrocephalus, the endoscopic technique is employed to avoid a second shunt implantation, and the patient remains shunt-dependent.[ 7 32 64 73 ]

CONCLUSION

This study indicates that, in unilateral FM obstruction hydrocephalus, a thorough review of the terminology application is critical to avoid mistakes that may compromise comparisons among different series. There are different terminologies meaning the same and also cases where the same terminology is applied for different clinical situations. This terminology review suggests that Type B hydrocephalus, i.e., the hydrocephalus confined to just one lateral ventricle with no other sites of CSF circulation blockage, are best described by the terms UH and MH hydrocephalus, the first being by far the most popular. Type A hydrocephalus is best represented in the literature by the terms ULH and LV, Type C hydrocephalus by the terms ILV and IUH; and Type D hydrocephalus by the term AH.

Financial support and sponsorship

Authors received FAPERJ financial support

Conflicts of interest

There are no conflicts of interest.

References

1. Abdel-Salam GM, Flores-Sarnat L, El-Ruby MO, Parboosingh J, Bridge P, Eid MM. Muenke syndrome with pigmentary disorder and probable hemimegalencephaly: An expansion of the phenotype. Am J Med Genet A. 2011. 155A: 207-14

2. Abderrahmen K, Aouidj ML, Kallel J, Zammel I, Khaldi MM. Hydrocephalus due to non tumoral stenosis of foramens of Monro: Report of four cases. Neurochirurgie. 2008. 54: 72-8

3. Alexander E, Botterell EH. Unilateral hydrocephalus resulting from occlusion of foramen of Monro; complication of radical removal of brain abscess. J Neurosurg. 1949. 6: 197-206

4. Alonso A, Taboada D, Alvarez JA, Paramo C, Vila M. Spontaneous ventriculostomy and ventricular diverticulum. Radiology. 1979. 133: 651-4

5. Anderson N, Malpas T, Davison M. Prenatal diagnosis of unilateral hydrocephalus. Pediatr Radiol. 1993. 23: 69-70

6. Andresen M, Juhler M. Multiloculated hydrocephalus: A review of current problems in classification and treatment. Childs Nerv Syst. 2012. 28: 357-62

7. Ang BT, Steinbok P, Cochrane DD. Etiological differences between the isolated lateral ventricle and the isolated fourth ventricle. Childs Nerv Syst. 2006. 22: 1080-5

8. Aronyk KE. The history and classification of hydrocephalus. Neurosurg Clin N Am. 1993. 4: 599-609

9. Atad-Rapoport M, Schweiger A, Lev D, Sadan-Strul S, Malinger G, Lerman-Sagie T. Neuropsychological follow-up at school age of children with asymmetric ventricles or unilateral ventriculomegaly identified in utero. BJOG. 2015. 122: 932-8

10. Atalay B, Yilmaz C, Cekinmez M, Altinors N, Caner H. Treatment of hydrocephalus with functionally isolated ventricles. Acta Neurochir (Wien). 2006. 148: 1293-6

11. Aydin K, Cokluk C, Gokce E, Diren B, Iyigun O, Rakunt C. Use of 3DFT-CISS sequences and virtual MR endoscopy for the neuroendoscopic treatment of unilateral hydrocephalus: Case illustration. Minim Invasive Neurosurg. 2007. 50: 239-42

12. Baumann B, Danon L, Weitz R, Blumensohn R, Schonfeld T, Nitzan M. Unilateral hydrocephalus due to obstruction of the foramen of Monro: Another complication of intrauterine mumps infection?. Eur J Pediatr. 1982. 139: 158-9

13. Bhagwati S. A case of unilateral hydrocephalus secondary to occlusion of one foramen of Monro. J Neurosurg. 1964. 21: 226-9

14. Bhattacharyya A, Boruah DK, Handique A, Singh V, Kalita J, Misra UK. Involvement of the choroid plexus in neurotuberculosis: MR findings in six cases. Neuroradiol J. 2010. 23: 590-5

15. Boyar B, Ildan F, Bagdatoglu H, Cetinalp E, Karadayi A. Unilateral hydrocephalus resulting from occlusion of foramen of Monro: A new procedure in the treatment: Stereotactic fenestration of the septum pellucidum. Surg Neurol. 1993. 39: 110-4

16. Brück W, Sander U, Blanckenberg P, Friede RL. Symptomatic xanthogranuloma of choroid plexus with unilateral hydrocephalus. Case report. J Neurosurg. 1991. 75: 324-7

17. Burtscher J, Sweeney R, Bale R, Eisner W, Twerdy K. Neuroendoscopy based on computer assisted adjustment of the endoscope holder in the laboratory. Minim Invasive Neurosurg. 2003. 46: 208-14

18. Cai Q, Song P, Chen Q, Chen Z, Huang S, Xu H. Neuroendoscopic fenestration of the septum pellucidum for monoventricular hydrocephalus. Clin Neurol Neurosurg. 2013. 115: 976-80

19. Cantini R, Lenzi B, Meozzi A. Ventriculo-atrial shunt (delayed unilateral hydrocephalus). J Neurosurg Sci. 1980. 24: 45-50

20. Chang Y, Horoupian DS, Lane B, Fross RD, Smyth LT, Seiling RJ. Inflammatory pseudotumor of the choroid plexus in Sjögren's disease. Neurosurgery. 1991. 29: 287-90

21. Chun HJ, Lee Y, Park HK, Kim YS. Neuroendoscopic fenestration of the foramen of Monro without septostomy for unilateral hydrocephalus following neonatal intraventricular hemorrhage. Childs Nerv Syst. 2011. 27: 473-8

22. Dandy WE. Experimental hydrocephalus. Ann Surg. 1919. 70: 129-42

23. Dastgir G, Awad A, Salam A, Attia M. Unilateral hydrocephalus due to foramen of Monro stenosis. Minim Invasive Neurosurg. 2006. 49: 184-6

24. de Vries LS, Groenendaal F, Gooskens R, Hanlo P. Unilateral posthaemorrhagic hydrocephalus in the neonatal period or later in infancy. Acta Paediatr. 2000. 89: 77-81

25. Decq P, Yepes C, Anno Y, Djindjian M, Nguyen JP, Kéravel Y. Neurosurgical endoscopy.Diagnostic and therapeutic indications. Neurochirurgie. 1994. 40: 313-21

26. Dorwling-Carter D, Scherpereel B, Baudrillart JC, Omez F, Lejeune JP, Rousseaux P. Unilateral non-tumor hydrocephalus in children. Atresia of the foramen of Monro?. Neurochirurgie. 1987. 33: 129-34

27. Durfee SM, Kim FM, Benson CB. Postnatal outcome of fetuses with the prenatal diagnosis of asymmetric hydrocephalus. J Ultrasound Med. 2001. 20: 263-8

28. El-Ghandour NM. Endoscopic cyst fenestration in the treatment of uniloculated hydrocephalus in children. J Neurosurg Pediatr. 2013. 11: 402-9

29. Ferreira M, Nahed BV, Babu MA, Walcott BP, Ellenbogen RG, Sekhar LN. Trapped fourth ventricle phenomenon following aneurysm rupture of the posterior circulation: Case reports. Neurosurgery. 2012. 70: E253-8

30. Fondop J, Lagmari M, Metellus P, Fuentes S, Ngah E, Djentcheu V. Unilateral hydrocephalus secondary to a brain temporal abscess treated by endoscopic septotomy: A case report. Neurochirurgie. 2010. 56: 337-9

31. Freppel S, Marchal JC, Joud A, Pinelli C, Klein O. Early surgical management of antenatal diagnosed cystic lesions of the foramen of Monro causing monoventricular hydrocephalus. Childs Nerv Syst. 2009. 25: 1131-5

32. Gangemi M, Maiuri F, Donati PA, Signorelli F, Basile D. Endoscopic surgery for monoventricular hydrocephalus. Surg Neurol. 1999. 52: 246-50

33. Gaston BM, Jones BE. Perinatal unilateral hydrocephalus.Atresia of the foramen of Monro. Pediatr Radiol. 1989. 19: 328-9

34. Greenlee JD, Teo C, Ghahreman A, Kwok B. Purely endoscopic resection of colloid cysts. Neurosurgery. 2008. 62: 51-5

35. Hageman G, Gooskens RH, Willemse J. A cerebral cause of arthrogryposis: Unilateral cerebral hypoplasia. Clin Neurol Neurosurg. 1985. 87: 119-22

36. Hamada H, Hayashi N, Kurimoto M, Umemura K, Hirashima Y, Endo S. Neuroendoscopic septostomy for isolated lateral ventricle. Neurol Med Chir (Tokyo). 2003. 43: 582-7

37. Hayashi T, Hashimoto T, Fukuda S, Anegawa S, Torigoe R. Clinical analysis of shunted hydrocephalic neonates and sucklings.Observation of postshunt complication due to overdrainage from intraventricular CSF. No To Shinkei. 1990. 42: 1167-71

38. Hongo K, Morota N, Watabe T, Isobe M, Nakagawa H. Giant basilar bifurcation aneurysm presenting as a third ventricular mass with unilateral obstructive hydrocephalus: Case report. J Clin Neurosci. 2001. 8: 51-4

39. Hubballah MY, Hoffman HJ. The isolated lateral ventricle.Experience at the hospital for sick children. Surg Neurol. 1987. 27: 220-2

40. Husag L, Wieser HG, Probst C. Unilateral hydrocephalus due to membranous occlusions of the foramen of Monro (author's transl). Acta Neurochir (Wien). 1976. 33: 183-212

41. Ismail EA, Shafik MH, Al-Mutairi G. A case of non-O:1 Vibrio cholerae septicemia with meningitis, cerebral abscess and unilateral hydrocephalus in a preterm baby. Eur J Clin Microbiol Infect Dis. 2001. 20: 598-600

42. Ito M, Ishikawa S, Ono Y, Akiyama I. Unilateral hydrocephalus associated with congenital hemihypertrophy. Neurol Med Chir (Tokyo). 1978. 18: 49-57

43. Jeon JH, Lee SW, Ko JK, Choi BG, Cha SH, Song GS. Neuroendoscopic removal of large choroid plexus cyst: A case report. J Korean Med Sci. 2005. 20: 335-9

44. Jivan K, Mochan A, Modi G. Intraventricular neurocysticercosis causing acute unilateral hydrocephalus. Afr J Psychiatry (Johannesbg). 2010. 13: 315-7

45. Kasantikul V, Shuangshoti S, Taecholarn C. Primary phycomycosis of the brain in heroin addicts. Surg Neurol. 1987. 28: 468-72

46. Kehler U, Gliemroth J, Arnold H. Asymmetric hydrocephalus: Safe endoscopic perforation of septum pellucidum: Technical note. Minim Invasive Neurosurg. 1997. 40: 101-2

47. Koga Y, Tahara Y, Kida T, Matumoto Y, Negishi H, Fujimoto S. Prenatal diagnosis of congenital unilateral hydrocephalus. Pediatr Radiol. 1997. 27: 319-20

48. Krucoff MO, Chinn M, Babington P, Litvack ZN. Controversial neuroendoscopic Monro foraminoplasty in the management of isolated lateral ventricle in an adult. Interdisciplinary Neurosurgery: Advanced Techniques and Case Management. 2015. 2: 108-10

49. Kumar R, Bhagat P. A severe and rapidly progressive case of proteus syndrome in a neonate who presented with unilateral hydrocephalus apart from other typical features of the proteus syndrome. J Clin Neonatol. 2012. 1: 152-4

50. Kumar R. Unilateral hydrocephalus in paediatric patients, a trial of endoscopic fenestration. Neurol India. 1999. 47: 282-5

51. Lazareff JA, Sadowinski S. The probable role of hydrocephalus in the development of intraventricular septa.An observation of one case. Childs Nerv Syst. 1992. 8: 139-41

52. Leonardo J, Grand W. Enlarged thalamostriate vein causing unilateral Monro foramen obstruction.Case report. J Neurosurg Pediatr. 2009. 3: 507-10

53. Lewis AI, Keiper GL, Crone KR. Endoscopic treatment of loculated hydrocephalus. J Neurosurg. 1995. 82: 780-5

54. Mampalam TJ, Harsh GR, Tien RD, Dillon WP, Wilson CB. Unilateral hydrocephalus in adults. Surg Neurol. 1991. 35: 14-9

55. Meadows J, Pigula F, Lock J, Marshall A. Transcatheter creation and enlargement of ventricular septal defects for relief of ventricular hypertension. J Thorac Cardiovasc Surg. 2007. 133: 912-8

56. Milhorat TH, Hammock MK, Breckbill DL. Acute unilateral hydrocephalus resulting from oedematous occlusion of foramen of Monro: Complication of intraventricular surgery. J Neurol Neurosurg Psychiatry. 1975. 38: 745-8

57. Miyahara N, Saito Y, Tabuchi S, Watanabe T, Maegaki Y, Ohno K. Unilateral hydrocephalus due to congenital stenosis of foramen of Monro - Observation of the slowly progressive ventricular dilatation during asymptomatic period. No To Hattatsu. 2008. 40: 489-91

58. Mohanty A, Das BS, Sastry Kolluri VR, Hedge T. Neuro-endoscopic fenestration of occluded foramen of Monro causing unilateral hydrocephalus. Pediatr Neurosurg. 1996. 25: 248-51

59. Nakamura S, Makiyama H, Miyagi A, Tsubokawa T, Ushinohama H. Congenital unilateral hydrocephalus. Childs Nerv Syst. 1989. 5: 367-70

60. Nishizaki T, Orita T, Abiko S, Aoki H, Ito H. Subependymal giant cell astrocytoma associated with tuberous sclerosis: With special reference to cell kinetic studies – Case report. Neurol Med Chir (Tokyo). 1990. 30: 695-7

61. Nowoslawska E, Polis L, Kaniewska D, Mikolajczyk W, Krawczyk J, Szymanski W. Effectiveness of neuroendoscopic procedures in the treatment of complex compartmentalized hydrocephalus in children. Childs Nerv Syst. 2003. 19: 659-65

62. Ohkawa S, Ohsumi Y, Tabuchi M, Yamadori A. Acute unilateral hydrocephalus caused by a small intracerebral hemorrhage obstructing the foramen of Monro. Stroke. 1993. 24: 1602-

63. Oi S, Enchev Y. Neuroendoscopic foraminal plasty of foramen of Monro. Childs Nerv Syst. 2008. 24: 933-42

64. Oi S, Hidaka M, Honda Y, Togo K, Shinoda M, Shimoda M. Neuroendoscopic surgery for specific forms of hydrocephalus. Childs Nerv Syst. 1999. 15: 56-68

65. Oi S, Yamada H, Sasaki K, Matsumoto S. Atresia of the foramen of Monro resulting in severe unilateral hydrocephalus with subfalcial herniation and infratentorial diverticulum. Neurosurgery. 1985. 16: 103-6

66. Patten RM, Mack LA, Finberg HJ. Unilateral hydrocephalus: Prenatal sonographic diagnosis. AJR Am J Roentgenol. 1991. 156: 359-63

67. Pfeiffer G, Friede RL. Unilateral hydrocephalus from early developmental occlusion of one foramen of Monro. Acta Neuropathol. 1984. 64: 75-7

68. Piro E, Piccione M, Marrone G, Giuffrè M, Corsello G. Dyke-Davidoff-Masson syndrome: Case report of fetal unilateral ventriculomegaly and hypoplastic left middle cerebral artery. Ital J Pediatr. 2013. 39: 32-

69. Salmon JH. Isolated unilateral hydrocephalus following ventriculoatrial shunt. J Neurosurg. 1970. 32: 219-26

70. Schlitt M, Duvall ER, Bonnin J, Morawetz RB. Neurosarcoidosis causing ventricular loculation, hydrocephalus, and death. Surg Neurol. 1986. 26: 67-71

71. Schroeder HW. Intraventricular tumors. World Neurosurg. 2013. 79: 15-9

72. Schulman H, Landau D, Schulman P, Hertzanu Y. Congenital unilateral hydrocephalus - CT findings. Eur J Radiol. 2000. 36: 161-4

73. Schulz M, Bührer C, Spors B, Haberl H, Thomale UW. Endoscopic neurosurgery in preterm and term newborn infants – A feasibility report. Childs Nerv Syst. 2013. 29: 771-9

74. Sharifi G, Rezaee O, Jahanbakhshi A. Unilateral hydrocephalus due to idiopathic anomaly of foramen of Monro, treated successfully with endoscopic technique.Report of three cases. Cent Eur Neurosurg. 2010. 71: 143-6

75. Singh DK, Rastogi M, Sharma A, Husain M. Unilateral hydrocephalus: Atypical presentation of intracranial tuberculoma. Turk Neurosurg. 2011. 21: 242-5

76. Steinbok P, Poskitt KJ, Cochrane DD, Kestle JR. Prevention of postshunting ventricular asymmetry by transseptal placement of ventricular catheters. A randomized study. Pediatr Neurosurg. 1994. 21: 59-64

77. Suzuki M, Seki H, Yoshimoto T. Unilateral hydrocephalus combined with occlusion of the ipsilateral internal carotid artery. Surg Neurol. 1985. 24: 27-30

78. Takeshita M, Miyazaki T, Kubo O, Kagawa M, Kitamura K. Case report of paraventricular cerebral cyst of infant (author's transl). No Shinkei Geka. 1980. 8: 73-8

79. Terrier A, Jourdan C, Remond J, Vighettho A, Peloux A, Naous H. Unilateral hydrocephalus caused by abscess of the choroid plexus. Rev Neurol (Paris). 1992. 148: 234-6

80. Thomas WS. Experimental hydrocephalus. J Exp Med. 1914. 19: 106-20

81. Tien R, Harsh GR, Dillon WP, Wilson CB. Unilateral hydrocephalus caused by an intraventricular venous malformation obstructing the foramen of Monro. Neurosurgery. 1990. 26: 664-6

82. Tillmann BU, Emons D, Bartmann P, Fahnenstich H. Posthemorrhagic unilateral hydrocephalus: Fenestration of septum pellucidum as an alternative to shunt implantation. J Pediatr. 2004. 144: 126-8

83. Vajramani GV, Devi BI, Hegde T, Santosh V, Khanna N, Vasudev MK. Intraventricular tuberculous abscess: A case report. Neurol India. 1999. 47: 327-9

84. Vaz-Guimarães Filho FA, Ramalho CO, Suriano ÍC, Zymberg ST, Cavalheiro S. Neuroendoscopic surgery for unilateral hydrocephalus due to inflammatory obstruction of the Monro foramen. Arq Neuropsiquiatr. 2011. 69: 227-31

85. Venkataramana NK, Kolluri VR, Swamy KS, Arya BY, Das BS, Reddy GN. Progressive unilateral hydrocephalus in adults. Neurosurgery. 1989. 24: 282-4

86. Volz HC, Laohachewin D, Seidel C, Lasitschka F, Keilbach K, Wienbrandt AR. S100A8/A9 aggravates post-ischemic heart failure through activation of RAGE-dependent NF-κB signaling. Basic Res Cardiol. 2012. 107: 250-

87. Whyte CA. Images from headache: Unilateral hydrocephalus. Headache. 2011. 51: 142-4

88. Wilberger JE, Vertosick FT, Vries JK. Unilateral hydrocephalus secondary to congenital atresia of the foramen of Monro. Case report. J Neurosurg. 1983. 59: 899-901

89. Winchester P, Brill PW, Cooper R, Krauss AN, Peterson HD. Prevalence of “compressed” and asymmetric lateral ventricles in healthy full-term neonates: Sonographic study. AJR Am J Roentgenol. 1986. 146: 471-5

90. Zoran MJ, Biljana SS, Ivana MP. Relocation of ventricular catheter trough ventriculostomy due to congenital unilateral hydrocephalus: Nine year follow-up. Surg Neurol Int. 2011. 2: 141-

A rare case of giant multiseptated thoracic myelomeningocele with segmental placode: Commentary

$
0
0
Pierre-Aurelien Beuriat, Alexandru Szathmari, Federico Di Rocco, Carmine Mottolese
  1. Department of Pediatric Neurosurgery, Neurological and Neurosurgical Hospital Pierre Wertheimer, Lyon, France

Correspondence Address:
Pierre-Aurelien Beuriat
Department of Pediatric Neurosurgery, Neurological and Neurosurgical Hospital Pierre Wertheimer, Lyon, France

DOI:10.4103/2152-7806.182390

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Beuriat P, Szathmari A, Rocco FD, Mottolese C. A rare case of giant multiseptated thoracic myelomeningocele with segmental placode: Commentary. Surg Neurol Int 13-May-2016;7:

How to cite this URL: Beuriat P, Szathmari A, Rocco FD, Mottolese C. A rare case of giant multiseptated thoracic myelomeningocele with segmental placode: Commentary. Surg Neurol Int 13-May-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/a-rare-case-of-giant-multiseptated-thoracic-myelomeningocele-with-segmental-placode-commentary/

Date of Submission
12-Jan-2016

Date of Acceptance
09-Mar-2016

Date of Web Publication
13-May-2016

Sir,

It is with great interest that we read the paper entitled, “A rare case of giant multiseptated thoracic myelomeningocele with segmental placode” by Patnaik and Mahapatra.[ 4 ] The authors described a rare presentation of a thoracic spinal dysraphism. The child they presented had no neurological and urologic deterioration, which is very uncommon in a thoracic myelomeningocele, and the magnetic resonance imaging (MRI) showed a multiloculated swelling with a fibroneural stalk that linked the skin malformation to the underlying spinal cord.

However, the complete description of the malformation and the fine MRI images and peroperative pictures provided by the authors may not be consistent with the diagnosis of a true myelomeningocele. We believed that it is a form of spinal dysraphism called “limited dorsal myeloschisis” (LDM). Such entity first described in 1993[ 1 ] has been nicely reviewed by Pang et al. with its complete description[ 2 ] and classification.[ 3 ] The case described by Patnaik and Mahapatra would correspond to a typical case of the saccular form. It has the skin-based sac which is filled with cerebrospinal fluid (CSF). The top of the skin lesion is recovered by desquamated tissue which is not normal skin. The neurologic condition is excellent in this case report as reported by Pang et al. in 40–50% of the thoracic and lumbar cases, where the urologic deterioration is only present in 15–20% of the cases.[ 2 ] In the present report, MRI findings are also consistent with Pang's description. The fibroneural stalk traverses the CSF-filled sac to reach the top of the skin anomaly. We can see on the axial view that there is a little depression at the junction between the top of the sac and the stalk that corresponds to the skin crater and presumably to the original site of the disjunction failure.[ 2 ]

There is one very important point brought by the authors: The presence of an evolutive hydrocephalus that required a shunt.[ 4 ] This is very interesting as Pang et al. did not report the presence of hydrocephalus in patients with thoracic LDM.[ 2 ]

This distinction between LDM and myelomeningocele has also an embryological implication: The difference lying in the degree of the neurulation stage.[ 2 ] In LDM, only the final stage of the primary neurulation is incomplete. Therefore, a limited disjunction between cutaneous and neural ectoderms is present. It prevents the complete midline skin closure (the top of the sac is covered by desquamated tissue which is not normal skin) and allows persistence of a physical link (fibroneural stalk) between the disjunction site and the dorsal neural tube. This is well documented by the peroperative pictures provided by the authors. The swelling was connected by this stalk to the dura trough a very narrow midline bone defect shown in Figure 3 from Patnaik and Mahapatra.[ 4 ]

The report from Patnaik and Mahapatra[ 4 ] underlines the complexity in the description and classification of spinal dysraphisms. Because these LDMs and myelomeningoceles have such great difference both in the natural history and final outcome, especially at the thoracic level, the proper distinction between these two entities is of paramount importance for a correct management of the child and a proper counseling to the parents.[ 5 ]

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Pang D, Dias MS. Cervical myelomeningoceles. Neurosurgery. 1993. 33: 363-72

2. Pang D, Zovickian J, Oviedo A, Moes GS. Limited dorsal myeloschisis: A distinctive clinicopathological entity. Neurosurgery. 2010. 67: 1555-79

3. Pang D, Zovickian J, Wong ST, Hou YJ, Moes GS. Limited dorsal myeloschisis: A not-so-rare form of primary neurulation defect. Childs Nerv Syst. 2013. 29: 1459-84

4. Patnaik A, Mahapatra AK. A rare case of giant multiseptated thoracic myelomeningocele with segmental placode. Surg Neurol Int. 2015. 6: 170-

5. Russell NE, Chalouhi GE, Dirocco F, Zerah M, Ville Y. Not all large neural tube defects have a poor prognosis: A case of prenatally diagnosed limited dorsal myeloschisis. Ultrasound Obstet Gynecol. 2013. 42: 238-9

Intracranial subdural empyema after surgery for lumbar lipomyelomeningocele: A rare complication

$
0
0
Ha Son Nguyen, Andrew Foy, Peter Havens
  1. Department of Neurosurgery, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, WI, USA
  2. Department of Infectious Diseases, Children's Hospital of Wisconsin, Milwaukee, WI, USA

Correspondence Address:
Ha Son Nguyen
Department of Infectious Diseases, Children's Hospital of Wisconsin, Milwaukee, WI, USA

DOI:10.4103/2152-7806.182388

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Nguyen HS, Foy A, Havens P. Intracranial subdural empyema after surgery for lumbar lipomyelomeningocele: A rare complication. Surg Neurol Int 13-May-2016;7:

How to cite this URL: Nguyen HS, Foy A, Havens P. Intracranial subdural empyema after surgery for lumbar lipomyelomeningocele: A rare complication. Surg Neurol Int 13-May-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/intracranial-subdural-empyema-after-surgery-for-lumbar-lipomyelomeningocele-a-rare-complication/

Date of Submission
21-Sep-2015

Date of Acceptance
07-Mar-2016

Date of Web Publication
13-May-2016

Abstract

Background:Surgery is routinely recommended for lumbar lipomyelomeningocele, especially in the setting of tethered cord syndrome. The most common complications are wound infections and cerebrospinal fluid (CSF) leak, which remain confined to the surgical site. To the best of our knowledge, there have been no prior reports relating an intracranial subdural empyema following detethering surgery. Prompt diagnosis is essential since subdural empyema is a neurosurgical emergency.

Case Description:The patient was an 11-month-old male who underwent detethering surgery for a lumbar lipomyelomeningocele. This was followed by wound drainage consistent with CSF leak, requiring revision. Cultures grew three aerobes (Escherichia coli, Enterococcus, and Klebsiella) and three anaerobes (Clostridium, Veillonella, and Bacteroides). He was started on cefepime, vancomycin, and flagyl. The patient required two more wound revisions and placement of an external ventricular drain (EVD) secondary to persistent wound leakage. A subsequent magnetic resonance imaging (MRI) brain was carried out due to protracted irritability, which revealed extensive left subdural empyema along the parietooccipital region and the inferior and anterior temporal lobe. He underwent evacuation of the subdural empyema where cultures exhibited no growth. Subsequently, he progressed well. His lumbar incision continued to heal. Serial MRI brains and inflammatory markers were reassuring. He weaned off his EVD and went home to complete a 6-week course of antibiotics. Upon completion of his antibiotics, he returned for a clinic visit; he exhibited no interim fevers or wound issues; cranial imaging documented no evidence of a residual or recurrent subdural empyema.

Conclusion:Intracranial subdural empyema may occur after wound complications from detethering surgery despite early initiation of broad-spectrum antibiotics. Possible etiology may be local wound infection that seeds the subdural space and travels to the cranium, leading to meningitis and subdural empyema. Such a scenario should prompt surveillance imaging of the head as undiagnosed subdural empyema may lead to devastating consequences.

Keywords: Lipomyelomeningocele, subdural empyema, tethered cord syndrome

INTRODUCTION

Lumbar lipomyelomeningocele is a form of occult spinal dysraphism that can cause tethered cord syndrome, leading to neurological, urological, and orthopedic dysfunction.[ 31 ] Surgery is typically recommended for tethered cord syndrome. The most common surgical complications are wound infections and cerebrospinal fluid (CSF) leak, which are generally confined to the surgical site.[ 4 12 13 15 17 19 20 23 25 31 ] To the best of our knowledge, there have been no prior reports relating an intracranial subdural empyema to detethering surgery. We report a child who received surgery for lipomyelomeningocele, developed wound complications and, unfortunately, sustained an intracranial subdural empyema. Moreover, we postulate the underlying mechanisms for this rare, but potentially devastating complication.

CASE PRESENTATION

The patient was a male who had been followed for an S-shaped gluteal crease since age 2 weeks. At age 3 months, a magnetic resonance imaging (MRI) L spine revealed a lumbar lipomyelomeningocele (2.7 cm rostrocaudal × 1.4 cm transverse × 1 cm anterior-posterior). There was absence of the posterior elements at S3, S4, and S5 vertebrae. The conus medullaris terminated at S2 [ Figure 1 ]. On examination, he had good motor function of his lower extremities. At age 7 months, a voiding cystourethrogram revealed a smooth walled bladder without trabeculation or reflux; urethra appeared normal during voiding. However, formal urodynamic testing demonstrated over-reactivity of the detrusor muscle, with concern for a neurogenic bladder. At age 11 months, he underwent a lumbosacral laminectomy and detethering of his spinal cord with debulking of the lumbar lipomyelomeningocele.


Figure 1

(a) Magnetic resonance imaging L spine T2 (arrow) and (b) T1 demonstrates lumbar lipomyelomeningocele (arrow)

 

Postoperatively, he exhibited his baseline neurological function. He was kept flat for 2 days, sedated with a dexmedetomidine drip in the Intensive Care Unit. Initial course was complicated by profuse diarrhea, emesis, and intermittent fevers up to 40.3°C, attributed to viral gastroenteritis. On 5 days later, the wound was draining CSF. Wound exploration revealed a CSF fistula at the lower third of the incision with no gross signs of infection. The child was started on vancomycin, cefepime, and flagyl. Cultures grew three aerobes (Escherichia coli, Enterococcus, and Klebsiella) and three anaerobes (Clostridium, Veillonella, and Bacteroides). Because of persistent fevers and CSF drainage 2 days later, a right frontal external ventricular drain (EVD) was placed, and the lumbar wound was reexplored. The leak emanated from the superior 1 cm part of the incision where devitalized tissue was evident; this was debrided to bleeding tissue and closed. EVD was left at 5 cm above his external auditory canal. He was kept flat bed rest, intubated and sedated. Unfortunately, the wound exhibited further wound drainage 4 days later and was again re-explored. Frank purulent discharge was immediately encountered as the wound fell open superiorly. The suprafascial space was cultured, which eventually grew E. coli. There was no pus extending from the epidural/subfascial space. He was kept sedated and paralyzed postoperatively for 3 days to allow for wound healing.

MRI brain performed because of persistent irritability showed extensive left-sided subdural empyema, greatest at the parietooccipital region and the inferior and anterior temporal lobe [ Figure 2 ]. MRI spine was negative for obvious signs of infection. Via two separate linear incisions, two different craniotomies were performed along the left temporal and left parietooccipital region to evacuate the subdural empyema collections. No significant membranes were encountered, but fairly extensive firm and fibrous debris was encountered at both sites and were removed with copious irrigation/gentle pituitary work/gentle aspiration; cultures remained negative for aerobes and anaerobes. Subsequently, patient progressed well with reduced irritability. His lumbar incision continued to heal. Serial MRI brain and inflammatory markers were reassuring. He weaned off his EVD; CSF was tested daily since its insertion and remained negative throughout the hospitalization. He continued ampicillin, meropenem, vancomycin, and flagyl; once operating room (OR) cultures from the craniotomy were negative, vancomycin was discontinued. Ampicillin remained for enteroccocal coverage, meropenem for Gram-negative coverage, and flagyl for anaerobic coverage. At discharge, he remained on intravenous meropenem and enteral metronidazole to complete a 6-week course. Upon completion of his antibiotics, patient returned for a follow-up clinic visit; he exhibited no interim fevers and his cranial imaging [ Figure 3 ] documented no evidence of a residual or recurrent subdural empyema. His lumbosacral wound was well healed.


Figure 2

(a) Magnetic resonance imaging brain T1 with contrast demonstrates extra-axial rim enhancement along left anterior temporal (arrow) and (b) left occipital (arrow). (c) Left occipital collection demonstrates diffusion-weighted restriction (arrow)

 

Figure 3

Magnetic resonance imaging brain T1 with contrast demonstrates resolution of subdural along left anterior temporal (a) and left occipital (b). Prior diffusion-weighted imaging signal has also resolved (c)

 

DISCUSSION

Intracranial subdural empyema may cause headaches, fevers, altered mental status, motor deficits, and seizures.[ 9 ] Devastating consequences include epilepsy, hemiparesis, and death. The infection may result from cranial procedures, meningitis, sinusitis, otogenic infection, or trauma.[ 5 7 9 14 18 27 28 ] Mortality rates can be as high as 4.4–24%.[ 7 21 24 30 ] Failure to initiate treatment within 1–2 days may lead to a rapid decline toward coma and death.[ 3 ] Pathogenesis is contingent on the etiology. A sinus infection may spread along valveless veins existing between extracranial and intracranial structures; direct extension can also occur from an infected sinus or mastoid.[ 3 ] Furthermore, a cranial neurosurgical procedure (ventricular shunting or subdural drain placement) may seed bacteria in the subdural space or prompt a secondary infection of a distant subdural effusion.[ 6 16 ]

The spread of infection from a spinal location to a cranial location is rare. Lumbar punctures, spinal anesthesia, and infected intrathecal baclofen pumps have been associated with meningitis.[ 2 11 ] To the best of our knowledge, however, the spread of infection from a spinal location to a cranial location has not been reported to develop into subdural empyema. Perhaps the most suitable analogy is congenital dermal sinus tracts, which connect the surface of the skin to the central nervous system.[ 1 ] The pathology has been associated with meningitis[ 10 22 26 ] and intraspinal abscess (epidural, subdural, and intramedullary).[ 22 26 ] Moreover, Mount[ 22 ] reported two instances of intracranial infection (a cerebellar abscess and an infected dermoid cyst) secondary to dermal sinus tracts tracking to the occipital protuberance. Radmanesh et al.[ 26 ] and Emami-Naeini et al.[ 8 ] each reported a case with intracranial abscess from a spinal dermal sinus tract.

Our patient had several neurosurgical procedures: A right frontal EVD placement and the initial detethering surgery followed by three wound revisions. It seems reasonable that the etiology of the subdural empyema was from a wound infection after the initial surgery (secondary to wound drainage coupled with profuse diarrhea/wound contamination); the wound infection may seed the CSF space and travel to the cranium to form an empyema from the beginning. On the other hand, the persistent CSF leakage from the wound, compounded by EVD drainage, may induce formation of a left subdural hygroma/effusion, which was subsequently seeded with infection from the lumbar wound. The patient was kept flat for a prolonged period, which may facilitate the spread of infection to the cranium. Nevertheless, his MRI spine rostral to the surgical site did not exhibit any signs of infection. The polymicrobial bacteria from the cultures were consistent with a gastrointestinal source. Another possible source was EVD though its location was contralateral to the subdural empyema; moreover, the CSF was tested daily and remained negative throughout the hospitalization course. In addition, there were no signs of infection along the ventricular catheter on the MRI brain. On the other hand, only about 9% of CSF cultures are positive in patients with subdural empyema.[ 29 ]

The patient was on prolonged sedation/paralysis to encourage wound healing, but such a scenario limits the ability to assess for intracranial complications based on a physical examination. Given this rare complication, we recommend routine surveillance cranial imaging to rule out subdural empyema, as prompt diagnosis is associated with better outcomes.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Ackerman LL, Menezes AH. Spinal congenital dermal sinuses: A 30-year experience. Pediatrics. 2003. 112: 641-7

2. Baer ET. Post-dural puncture bacterial meningitis. Anesthesiology. 2006. 105: 381-93

3. Bruner DI, Littlejohn L, Pritchard A. Subdural empyema presenting with seizure, confusion, and focal weakness. West J Emerg Med. 2012. 13: 509-11

4. Chern JJ, Tubbs RS, Patel AJ, Gordon AS, Bandt SK, Smyth MD. Preventing cerebrospinal fluid leak following transection of a tight filum terminale. J Neurosurg Pediatr. 2011. 8: 35-8

5. Dashti SR, Baharvahdat H, Spetzler RF, Sauvageau E, Chang SW, Stiefel MF. Operative intracranial infection following craniotomy. Neurosurg Focus. 2008. 24: E10-

6. Dickerman RD, Piatt JH, Hsu F, Frank EH. Subdural empyema complicating cerebrospinal fluid shunt infection. Pediatr Neurosurg. 1999. 30: 310-1

7. Dill SR, Cobbs CG, McDonald CK. Subdural empyema: Analysis of 32 cases and review. Clin Infect Dis. 1995. 20: 372-86

8. Emami-Naeini P, Mahdavi A, Ahmadi H, Baradaran N, Nejat F. Brain abscess as a manifestation of spinal dermal sinus. Ther Clin Risk Manag. 2008. 4: 1143-7

9. French H, Schaefer N, Keijzers G, Barison D, Olson S. Intracranial subdural empyema: A 10-year case series. Ochsner J. 2014. 14: 188-94

10. Givner LB, Baker CJ. Anaerobic meningitis associated with a dermal sinus tract. Pediatr Infect Dis. 1983. 2: 385-7

11. Haranhalli N, Anand D, Wisoff JH, Harter DH, Weiner HL, Blate M. Intrathecal baclofen therapy: Complication avoidance and management. Childs Nerv Syst. 2011. 27: 421-7

12. Hayashi T, Takemoto J, Ochiai T, Kimiwada T, Shirane R, Sakai K. Surgical indication and outcome in patients with postoperative retethered cord syndrome. J Neurosurg Pediatr. 2013. 11: 133-9

13. Hsieh MH, Perry V, Gupta N, Pearson C, Nguyen HT. The effects of detethering on the urodynamics profile in children with a tethered cord. J Neurosurg. 2006. 105: S391-5

14. Idowu OE, Adekoya VA, Adeyinka AP, Beredugo-Amadasun BK, Olubi OO. Demography, types, outcome and relationship of surgically treated intracranial suppuration complicating chronic suppurative otitis media and bacterial rhinosinusitis. J Neurosci Rural Pract. 2014. 5: S48-52

15. James HE, Williams J, Brock W, Kaplan GW, Hoi SU. Radical removal of lipomas of the conus and cauda equina with laser microneurosurgery. Neurosurgery. 1984. 15: 340-3

16. Kasliwal MK, Sinha S, Kumar R, Sharma BS. Giant hemicranial calcified subdural empyema – Unusual complication following ventriculoperitoneal shunt insertion. Indian J Pediatr. 2009. 76: 651-2

17. Kumar A, Mahapatra AK, Satyarthee GD. Congenital spinal lipomas: Role of prophylactic surgery. J Pediatr Neurosci. 2012. 7: 85-9

18. Le Roux PC, Wood M, Campbell RA. Subdural empyema caused by an unusual organism following intracranial haematoma. Childs Nerv Syst. 2007. 23: 825-7

19. Lee GY, Paradiso G, Tator CH, Gentili F, Massicotte EM, Fehlings MG. Surgical management of tethered cord syndrome in adults: Indications, techniques, and long-term outcomes in 60 patients. J Neurosurg Spine. 2006. 4: 123-31

20. Mehta VA, Bettegowda C, Ahmadi SA, Berenberg P, Thomale UW, Haberl EJ. Spinal cord tethering following myelomeningocele repair. J Neurosurg Pediatr. 2010. 6: 498-505

21. Miller ES, Dias PS, Uttley D. Management of subdural empyema: A series of 24 cases. J Neurol Neurosurg Psychiatry. 1987. 50: 1415-8

22. Mount LA. Congenital dermal sinuses as a cause of meningitis, intraspinal abscess and intracranial abscess. J Am Med Assoc. 1949. 139: 1263-8

23. Pang D, Zovickian J, Oviedo A. Long-term outcome of total and near-total resection of spinal cord lipomas and radical reconstruction of the neural placode: Part I-surgical technique. Neurosurgery. 2009. 65: 511-28

24. Pathak A, Sharma BS, Mathuriya SN, Khosla VK, Khandelwal N, Kak VK. Controversies in the management of subdural empyema. A study of 41 cases with review of literature. Acta Neurochir (Wien). 1990. 102: 25-32

25. Proctor MR, Scott RM. Long-term outcome for patients with split cord malformation. Neurosurg Focus. 2001. 10: e5-

26. Radmanesh F, Nejat F, El Khashab M. Dermal sinus tract of the spine. Childs Nerv Syst. 2010. 26: 349-57

27. Salomão JF, Cervante TP, Bellas AR, Boechat MC, Pone SM, Pone MV. Neurosurgical implications of Pott's puffy tumor in children and adolescents. Childs Nerv Syst. 2014. 30: 1527-34

28. Santarius T, Qureshi HU, Sivakumaran R, Kirkpatrick PJ, Kirollos RW, Hutchinson PJ. The role of external drains and peritoneal conduits in the treatment of recurrent chronic subdural hematoma. World Neurosurg. 2010. 73: 747-50

29. Scheld MW, Marra CM, Whitley RJ.editorsInfections of the Central Nervous System. Philadelphia, Pennsylvania: Wolters Kluwer Health; 2014. p.

30. Tewari MK, Sharma RR, Shiv VK, Lad SD. Spectrum of intracranial subdural empyemas in a series of 45 patients: Current surgical options and outcome. Neurol India. 2004. 52: 346-9

31. Thuy M, Chaseling R, Fowler A. Spinal cord detethering procedures in children: A 5 year retrospective cohort study of the early post-operative course. J Clin Neurosci. 2015. 22: 838-42


Extranodal right-optic nerve Rosai–Dorfman disease: A rare localization case report

$
0
0

Jakob Nemir, Ines Trninic, Kresimir S. Duric, Antonia Jakovcevic, Goran Mrak, Josip Paladino
  1. Department of Neurosurgery, University Hospital Center Zagreb, School of Medicine, Zagreb, Croatia
  2. Department of Pathology, University Hospital Center Zagreb, School of Medicine, Zagreb, Croatia
  3. Department of Neurosurgery, Dubrovnik General Hospital, Dubrovnik, Croatia

Correspondence Address:
Jakob Nemir
Department of Neurosurgery, University Hospital Center Zagreb, School of Medicine, Zagreb, Croatia

DOI:10.4103/2152-7806.196933

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Jakob Nemir, Ines Trninic, Kresimir S. Duric, Antonia Jakovcevic, Goran Mrak, Josip Paladino. Extranodal right-optic nerve Rosai–Dorfman disease: A rare localization case report. 28-Dec-2016;7:

How to cite this URL: Jakob Nemir, Ines Trninic, Kresimir S. Duric, Antonia Jakovcevic, Goran Mrak, Josip Paladino. Extranodal right-optic nerve Rosai–Dorfman disease: A rare localization case report. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/extranodal-right%e2%80%91optic-nerve-rosai-dorfman-disease-a-rare-localization-case-report/

Date of Submission
09-Feb-2016

Date of Acceptance
17-Jun-2016

Date of Web Publication
28-Dec-2016

Abstract

Background:Rosai–Dorfman is a rare disease that usually occurs in young adults. It is characterized with massive painless cervical lymphadenopathy and histiocyte proliferation. Isolated intracranial involvement is extremely rare. Our aim is to present a new rare case of extranodal Rosai–Dorfman disease that involved the right optic nerve in a 4-year-old boy.

Case Description:A 4-year-old boy with right-sided convergent strabismus and amblyopia lasting for 1 year was treated at the Department of pediatric ophthalmology. Initial optical fundus examination was normal. Examination repeated after 1 year noted the atrophy of the optic nerve papilla. Visual evoked potentials of the right eye showed normal findings of prechiasmatic visual pathway with severe dysfunction of the right optic nerve. Magnetic resonance imaging (MRI) of the brain and orbits showed expansive changed and elongated right optic nerve with contrast enhancement, and smaller lesion in the right temporal operculum region visible in T2 and fluid-attenuated inversion recovery sequence. Through small eyebrow “keyhole” osteoplastic frontoorbital craniotomy the fusiform enlarged (to 2 cm) right optic nerve was identified, resected between the eyeball and optic chiasm, and transferred for pathohistological analysis. Early postoperative course had no complications. Histological, immunohistochemical, and ultrastructural analyses revealed extranodal Rosai–Dorfman disease. Right periorbital edema was verified on the 7th postoperative day and regressed to supportive therapy. Control multi slice computed tomography (MSCT) and MRI of endocranium and orbits showed total tumor removal with no signs of complications.

Conclusion:Although rare, extranodular intracranial Rosai–Dorfman disease should be taken into account in the differential diagnosis of intracranial and intraorbital lesions, especially in the pediatric age group.

Keywords: Extranodal, optic nerve, pediatric tumor, Rosai–Dorfman disease

INTRODUCTION

Rosai–Dorfman disease (RDD), also known as sinus histiocytosis with massive lymphadenopathy (SHML) is an uncommon benign histiocytic proliferative disorder of unknown origin.[ 1 2 7 9 12 14 19 ] It predominantly affects the lymph nodes but can also be found extranodally in other organs, and usually presents with other constitutional symptoms such as fever, malaise, weight loss, and raised inflammatory markers.[ 1 2 7 9 12 14 19 ] Approximately, one-third of the patients have concurrent extranodal involvement, most commonly in the skin, salivary glands, and upper respiratory tract.[ 21 ] Nervous system involvement is rare and in most cases intracranial.[ 2 9 11 12 ] Only 18 cases of intracranial involvement have been described previously. In addition, only 4 cases described the lesion with involvement of the orbits. Our report documents the first case, to our knowledge, of optic nerve localization of RDD.

CASE DESCRIPTION

History and examination

A 4-year-old boy presented with right-sided convergent strabismus and amblyopia that lasted for 1 year and was admitted to the Department of Pediatric Ophthalmology. His medical history was unremarkable except for the thrombocytopenia treated at 2 years of age. Before admission, he had regular ophthalmological exams and was treated with corrective glasses. An eye examination was performed by an ophthalmologist. Visual acuity was normal on his left eye, but visual acuity on his right eye was poor, he did not have the sense of light. Both eye bulb motility was normal with good pupil function. Initial optical fundus examination was normal. The cover test was positive on his right eye with right-sided convergent strabismus. Visual field was not done because of the patient's age. On the last ophthalmologic control exam, due to the right-sided atrophy of the optic nerve papilla and amblyopia, performing visual evoked potentials (VEP) and magnetic resonance imaging (MRI) were recommended. VEP of the right eye showed normal findings of prechiasmatic visual pathway with severe dysfunction of the right optic nerve. MRI of the brain and orbits showed expansive changed and elongated right optic nerve with contrast enhancement; furthermore, a smaller lesion in the right temporal operculum region was visible in T2 and fluid-attenuated inversion recovery sequence, as well as a small oval lesion in the left cerebellar lobe [Figures 1 and 2 ]. Optic nerve glioma was considered to be the most likely radiological diagnosis.


Figure 1

(a, b) Preoperative T1 magnetic resonance (MR) images demonstrates expansive changed and elongated right optic nerve. (c) Preoperative coronal plane T1 gadolinium-enhanced MR image. (d) Preoperative sagittal plane T2 MR image

 

Figure 2

(a, b) Preoperative magnetic resonance imaging showing left cerebellar and right temporal intracranial lesions

 

Surgical technique

The position of the patient was supine, with the head turned to the left at 15°, leaving the eyebrow as the most prominent point. Skin was incised through the eyebrow, medially up to the supraorbital notch leaving the supraorbital nerve intact. With one small burr hole at the superior temporal line, small supraorbital bone flap was performed which was 3 cm in width and 2.5 cm in height using a craniotome, including linear extensions over the supraorbital arch. Periorbita was detached from the bone and the bony flap was pushed down toward the orbit until the orbital roof brakes. With slight elevation of the bone, dura was detached from the orbital roof and the whole bone flap including supraorbital arch was removed in one piece. Using a diamond drill, the whole orbital roof was drilled out, optic canal widely opened, and anterior clinoid removed extraduraly. Periorbit was incised longitudinally, including annulus of Zinni and the dura over the optic nerve and frontobasaly, meticulously dissecting orbital muscles and nerves before reaching the optic nerve. Almost the whole optic nerve was thickened, and was irregularly shaped 2 mm from the eyeball and up to the chiasm. The nerve with the infiltrating tumor was cut leaving the normal white tissue at the resected planes of the nerve. Macroscopically, the tumor was firm, avascular, and had a gray-yellow color infiltrating the whole width of the optic nerve. There was no bleeding at the resected planes of the nerve and dura and periorbit was partly sutured and sealed. Bone flap was attached using microscrews, and the wound was closed in the standard manner.

Postoperative course

Early postoperative course was uneventful, except right-sided periorbital hematoma that spontaneously regressed few days later. Later, the patient received corneal ulcer refractory to treatment with topical antibiotic drops and cream for 3 months, after which the amniotic membrane transplant was performed to heal the ulcer completely. After 3 years of follow-up, MRI showed complete tumor removal without any sign of recurrence [ Figure 3 ].


Figure 3

(a, b) Postoperative T2 images, axial and coronal plane. (c) Postoperative T1 axial plane image

 

Histopathological workup

Microscopic examination of the surgical sample revealed a mainly histiocytic lesion in a fibrous background admixed with lesser populations of lymphocytes and plasma cells. The histiocytic cells showed evidence of emperipolesis. Immunohistochemically, tumor cells were CD68 and S100 positive and negative for langerin and CD1a. The phenotype was consistent with RDD [Figures 4 and 5 ]. There were no microorganisms, necrosis, or granuloma formation. Because of the extremely rare diagnosis, especially on this localization, paraffin blocks of tumor tissue were sent to the Department of Pathology Brigham and Women's Hospital in Boston, USA, for a second opinion. They confirmed our diagnosis of RDD.


Figure 4

(a) Microscopic features; mainly histiocytes and a few chronic inflammatory cells in fibrous background (hematoxylin and eosin ×100). (b) Emperipolesis; an additional common histopathologic finding in Rosai–Dorfmann disease (hematoxylin and eosin ×400)

 

Figure 5

Immunohistochemistry: (a) Glial fibrillary acidic protein small fragments of brain tissue were positive whereas tumour was negative (×100). (b) CD68 positive histiocytic tumor cells (×400); (c) negative staining for CD1a (×100)

 

DISCUSSION

RDD is a rare histiocytic disorder initially described as a separate entity in 1969 by Rosai and Dorfman using the term SHML.[ 19 ] The causes of RDD are not fully understood, and treatment strategies can be different according to the severity of vital organ involvement. It is usually seen in young adult patients but may occur in any age group.[ 13 ] Patients presenting with isolated intracranial disease tend to be older.[ 6 ] Moreover, it is more common in men, with a possible predilection for African Americans, and the mean age at presentation is approximately 20 years.[ 7 9 19 ] The etiology is uncertain, although agents such as the Epstein-Barr or herpes viruses are important in the pathogenesis.[ 10 ] The neck lymph nodes are the most frequently involved, followed by inguinal, axillary, and mediastinal lymph nodes.[ 3 16 ] The most common extranodal sites are the skin, upper respiratory tract, and bones. Head and neck involvement—approximately 22% of extranodal disease—include involvement of the nasal cavity, the paranasal sinuses, the nasopharynx, submandibular glands, the parotid, the larynx, the temporal bone, the intratemporal fossa, the pterygoid fossa, the meninges, and the orbit.[ 3 18 ] The skin is also commonly affected. Half of the patients have another associated extranodal site. Orbit and ocular glove involvement have been reported, usually as a retroorbitary mass and proptosis.[ 8 ] To our knowledge, the present case is the first example of optic nerve localization caused by RDD. Intracranial RDD usually occurs without extracranial lymphadenopathy, and most intracranial lesions are attached to the dura with only few extending intraparynchemally. Central nervous system disease can present clinically and radiologically as meningioma, however, the presence of emperipolesis in the cerebrospinal fluid is usually diagnostic of RDD.[ 15 ] In our case, glioma was considered to be the most likely radiological diagnosis. Emperipolesis is not a unique phenomenon to RDD and has been seen in both normal and leukemic processes,[ 19 ] however, it appears to be a prerequisite for the diagnosis. The clinical course of RDD is unpredictable with episodes of exacerbation and remissions that could last many years. The disease is often self-limiting with a very good outcome, nevertheless 5–11% of patients die from their disease. In the present case, clinical presentation of RDD was right-sided convergent strabismus and amblyopia without painless cervical lymphadenopathy with fever, which has been seen in other patients. The presenting symptoms depended on the location of the lesions and were manifested by cranial nerve deficits or nonlocalizing symptoms of raised intracranial pressure or seizures. Laboratory features in RDD are often nonspecific. Leukocytosis, elevated sedimentation rate, and polyclonal hypergammaglobulinemia have been reported in most patients, but in our case there were no laboratory abnormalities.[ 7 20 ] The differential diagnosis of extranodal SHLM may be a challenge, and is based on the clinical and histological examination. Histology shows typical features, such as diffuse lymphoplasmatic infiltration, Russel bodies, foamy histiocytes, and histiocytes with phagocytosed lymphocytes within the cytoplasm (emperipolesis). Immunohistochemical features include positive S-100, alpha-antichymotrypsin and CD1a and CD68 antigens.[ 3 18 ] Imaging (computed tomography and MRI) may be used to assess disease extension. If there is cervical lymph node enlargement, fine needle aspiration biopsy or lymph node biopsies may be useful for the diagnosis.[ 10 ] The differential diagnosis is made with lymphoreticular malignancies such as lymphomas, Hodgkin's disease, malignant histiocytosis, and monocytic leukemia, all of which have similar histopathological features. Atypia in cytology and the aggressive clinical course establish the diagnosis in most case. Other histiocytoses, such as rhinoscleromas and Wegener's granulomatosis, may also be included in the differential diagnosis.[ 8 ] Treatment is controversial. In the majority of cases, RDD has a benign course and treatment is not necessary.[ 22 ] Therapy is required, however, for patients with extranodal RDD having vital organ involvement or those with nodal disease causing life-threatening complications.[ 18 ] The role of surgery is mostly in biopsies and to relieve obstruction.[ 10 ] Local recurrence is frequent following surgical resection. The role of radiotherapy is not well understood; some reports have described full resolution with this treatment, whereas others have shown no response.[ 4 5 ] Systemic corticosteroids are usually helpful in decreasing nodal size and symptoms, however, they can be immunosuppressive and recurrence of RDD lesions can occur after a short period of interruption.[ 21 ] Chemotherapy has resulted in controversial results. A possible efficacy of methotrexate and 6-mercaptopurine requires further investigation. Other reports have suggested using alpha-interferon, although its side effects have its limited use.[ 14 ] Furthermore, the efficacy of the anti-CD20 monoclonal antibody/rituximab has been described in one case.[ 17 ] We treated our patient with the surgical form of treatment, to which he responded well, without radiotherapy and others modalities. Because of the rarity of such cases, long-term clinical and radiological follow-up is mandatory. In summary, we described a new case of isolated optic nerve RDD causing optic nerve infiltration. It should be considered among the rare differential diagnosis of optic nerve infiltration disease.

CONCLUSION

Extranodular intracranial RDD should be taken into account in the differential diagnosis of intracranial and intraorbital lesions, especially in pediatric age group.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

Acknowledgements

We thank Prof. Christopher D. M. Fletcher, MD, FRCPath, from the Department of pathology Brigham and Women's Hospital Boston, USA, for his support in diagnosis.

References

1. Al-Saad K, Thorner P, Ngan BY, Gerstle JT, Kulkarni AV, Babyn P. Extranodal Rosai-Dorfman disease with multifocal bone and epidural involvement causing recurrent spinal cord compression. Pediatr Dev Pathol. 2005. 8: 593-8

2. Andriko JA, Morrison A, Colegial CH, Davis BJ, Jones RV. Rosai-Dorfman disease isolated to the central nervous system: A report of 11 cases. Mod Pathol. 2001. 14: 172-8

3. Carbone A, Passannante A, Gloghini A, Devaney KO, Rinaldo A, Ferlito A. Review of sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) of head and neck. Ann Otol Rhinol Laryngol. 1999. 108: 1095-104

4. Carpenter RJ, Banks PM, Mc Donald TJ, Sanderson DR. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): Report of a case with respiratory tract involvement. Laryngoscope. 1978. 88: 1963-9

5. Cooper SL, Chavis PS, Fortney JA, Watkins JM, Caplan MJ, Jenrette JM. A case of orbital Rosai-Dorfman disease responding to radiotherapy. J Pediatr Hematol Oncol. 2008. 30: 744-8

6. Deodhare SS, Ang LC, Bilbao JM. Isolated intracranial involvement in Rosai-Dorfman disease: A report of two cases and a review of the literature. Arch Pathol Lab Med. 1998. 122: 161-5

7. Foucar E, Rosai J, Dorfman R. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): Review of the entity. Semin Diagn Pathol. 1990. 7: 19-73

8. Goodnight JW, Wang MB, Sercarz JA, Fu YS. Extranodal Rosai-Dorfman disease of the head and neck. Laryngoscope. 1996. 106: 253-6

9. Hargett C, Bassett T. Atypical presentation of sinus histiocytosis with massive lymphadenopathy as an epidural spinal cord tumor: A case presentation and literature review. J Spinal Disord Tech. 2005. 18: 193-6

10. Hazarika P, Nayak DR, Balakrishnan R, Kundaje HG, Rao PL. Rosai-Dorfman disease of the subglottis. J Laryngol Otol. 2000. 114: 970-3

11. Hollowell JP, Wolfla CE, Shah NC, Mark LP, Whittaker MH. Rosai-Dorfman disease causing cervical myelopathy. Spine. 2000. 25: 1453-6

12. Huang YC, Than HY, Jung SM, Chuang WY, Chuang CC, Hsu PW. Spinal epidural Rosai-Dorfman disease proceeded by relapsing uveitis: A case report with literature review. Spinal Cord. 2007. 45: 641-4

13. Juskevicius R, Finlay JL. Rosai-Dorfman disease of the parotid gland, cytologic and histopathologic findings with immunohistochemical correlation. Arch Pathol Lab Med. 2001. 125: 1348-50

14. Kidd DP, Revesz T, Miller NR. Rosai-Dorfman disease presenting with widespread intracranial and spinal cord involvement. Neurology. 2006. 67: 1551-5

15. Krafet SK, Honig M, Krishnamurthy S. Emperipolesis in the cerebrospinal fluid from a patient with Rosai-Dorfman disease. Diagn Cytopathol. 2007. 36: 67-8

16. Lauwers GY, Perez-Atayed A, Dorfman RF, Rosai . The digestive system manifestations of Rosai-Dorfman disease (sinus histiocytosis with massive lymphadenopathy): Review of 11 cases. Hum Pathol. 2000. 31: 380-5

17. Petschner F, Walker UA, Scmitt-Graf A. Catastrophic systemic lupus erythematous with Rosai-Dorfman sinus histiocytosis. Successful treatment with anti-CD20/rituximab. Dtsch Med Wochenschr. 2001. 126: 998-1001

18. Pulsoni A, Anghel G, Falcucci P, Matera , R Pescarmona, Ribersani M. Treatment of sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): Report of a case and literature Review. Am J Hematol. 2000. 69: 61-71

19. Rosai J, Dorfman RF. Sinus histiocytosis with massive lymphadenopathy: A newly recognized benign clinicopathological entity. Arch Pathol. 1969. 87: 63-70

20. Sanchez R, Rosai J, Dorfman RF. Sinus histiocytosis with massive lymphadenopathy: An analysis of 113 cases with special emphasis on its extranodal manifestations. Lab Invest. 1977. 36: 349-50

21. Scheel MM, Rady PL, Tyring SK, Pandya AG. Sinus histiocytosis with massive lymphadenopathy: Presentation as giant granuloma annulare and detection of human herpesvirus 6. J Am Acad Dermatol. 1997. 37: 643-6

22. Tian Y, Wang J, Li M, Lin S, Wang G, Wu Z, Ge M, Pirotte BJ. Rosai-Dorfman disease involving the central nervous system: Seven cases from one institute. Acta Neurochir. 2015. 157: 1565-71

Volumetric growth analysis of an insular dysembryoplastic neuroepithelial tumor over a 10-year follow-up

$
0
0

Takehiro Uno, Masashi Kinoshita, Takuya Furuta, Katsuyoshi Miyashita, Hemragul Sabit, Mitsutoshi Nakada
  1. Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan

Correspondence Address:
Mitsutoshi Nakada
Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan

DOI:10.4103/2152-7806.196931

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Takehiro Uno, Masashi Kinoshita, Takuya Furuta, Katsuyoshi Miyashita, Hemragul Sabit, Mitsutoshi Nakada. Volumetric growth analysis of an insular dysembryoplastic neuroepithelial tumor over a 10-year follow-up. 28-Dec-2016;7:

How to cite this URL: Takehiro Uno, Masashi Kinoshita, Takuya Furuta, Katsuyoshi Miyashita, Hemragul Sabit, Mitsutoshi Nakada. Volumetric growth analysis of an insular dysembryoplastic neuroepithelial tumor over a 10-year follow-up. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/volumetric-growth-analysis-of-an-insular-dysembryoplastic-neuroepithelial-tumor-over-a-10%e2%80%91year-follow%e2%80%91up/

Date of Submission
31-Jan-2016

Date of Acceptance
23-Jul-2016

Date of Web Publication
28-Dec-2016

Abstract

Background:Dysembryoplastic neuroepithelial tumors (DNETs) are benign tumors characterized by a cortical location; they result in symptoms of drug-resistant partial seizures in children. The development of DNETs is poorly understood because most of them are resected immediately upon diagnosis without any observation period owing to the intractable seizures.

Case Description:We report the first DNET case with the growth rate analyzed in the natural course of development for a period of 10 years. The patient was a right-handed man who was initially referred to another hospital with mild head injury when he was 8 years old. A tumor located in the right insular cortex was incidentally detected on magnetic resonance imaging (MRI) and followed-up with annual MRI for 10 years.

Conclusion:In this case, the volume of the DNET increased in direct proportion to the length of time in its clinical course. The tumor doubling time was approximately 10 years. This case suggests DNET is a slow-growing but not stable tumor.

Keywords: Dysembryoplastic neuroepithelial tumor, insular, growth analysis

INTRODUCTION

Dysembryoplastic neuroepithelial tumors (DNETs) are benign, hamartomatous tumors thought to arise from the cortical gray matter. They are mixed neuronal-glial tumors, classified as grade I by the World Health Organization (WHO). Progression or post-surgical recurrence of DNETs is perceived to be extremely rare. DNETs typically cause intractable seizures in children, and are removed surgically without observation.[ 1 10 ] Therefore, the natural course and development of DNETs is poorly understood. The DNET case reported here was observed for 10 years without surgery because of the absence of symptoms. The lesion demonstrated gradual growth. We report an analysis of the DNET growth rate for the first time.

CASE REPORT

Our patient was initially referred to another hospital with mild head injury when he was 8 years old. An intra-axial tumor located in the right insular cortex was incidentally detected on MRI. Surgical resection was waived and followed-up for 10 years until the patient was 18 years old because of slowly growing tumor without symptoms. At the end of the observation period, the tumor size was measured to be one and a half times the diameter measured on the first MRI scan. After the end of the follow-up period, he visited our hospital for intensive examination and treatment.

The patient had no neurological deficit. Computed tomography (CT) imaging showed a low-density lesion with no calcification located in the right insular cortex [ Figure 1a ]. T1-weighted MRI demonstrated a hypointense lesion in the right insular cortex [ Figure 1b ]. T2-weighted MRI showed a hyperintense lesion that corresponded with the hypointensity on the T1-weighted image [ Figure 1c ]. T1-weighted MRI with gadolinium administration did not show any enhanced lesions [Figure 1d and e ]. Arterial spin labeling study suggested decreased blood flow at the lesion [ Figure 1f ].


Figure 1

Axial computed tomography (CT) (a) and magnetic resonance imaging (MRI) scans (b-d, f) and coronal MRI scan (e). (a) CT scan shows a low-density lesion in right insular cortex with no calcification. (b) T1-weighted MRI demonstrates a hypointense lesion. (c) T2-weighted MRI shows a hyperintense lesion that corresponds with the hypointensity on the T1-weighted MRI image. (d, e) T1-weighted MRI with gadolinium administration did not demonstrate any enhanced lesion. (f) Arterial spin labeling study shows decreased blood flow at the lesion

 

The lesion located in the right insular cortex demonstrated gradual growth for 10 years [Figure 2a - d ]. The change in lesion volume was assessed using polygonal tracing with fusion. Fluid-attenuated inversion recovery (FLAIR) signals were assessed using the DICOM image viewer OsiriX (®) (v. 7.0; Pixmeo SARL, Bernex, Switzerland) by slice-by-slice region of interest tracings. The growth rate of this lesion was found to be almost directly proportional to time [ Figure 2e ].


Figure 2

(a-d) T2-FLAIR MRI scans in the axial plane from age 8 to 18 years show gradual growth. An inserted picture in the corner of images is a three-dimensional reconstructed model of the tumor in each figure. (e) A dot graph with an almost straight line shows that the increase in tumor volume is directly proportional to time

 

In order to remove the lesion and obtain histopathological diagnosis, an awake craniotomy was performed using cortical and subcortical stimulation mapping with a bipolar direct electrical stimulator at 3.5 mA/60 Hz biphasic current to monitor motor and somatosensory response, speech or language difficulties, and other higher brain functions.[ 6 ] An anarthria was induced by stimulation of the ventral precentral gyrus [ Figure 3a ]. Tumor resection was performed via a transopercular approach. Intraoperatively, the nature of the tumor was gray, soft, and jelly-like tissue with clear boundaries. Fiber structures in the peripheral zone were relatively well-defined and we promoted excision of the tumor using an ultrasonic surgical aspirator. A postoperative MRI showed gross total resection of the tumor [ Figure 3b ]. Postoperative course was uneventful without neurological deficits. No recurrence was recognized postoperatively for 12 months.


Figure 3

Intraoperative (a) and postoperative (b) images. (a) Intraoperative view after tumor resection during awake surgery. Tag 1 suggests the area where anarthria was induced on the ventral precentral gyrus by direct electrical stimulation. An inserted picture is the three-dimensional cortical view. (b) Fluid-attenuated inversion recovery magnetic resonance imaging performed 3 months after the operation shows gross total resection. Arrows indicate the Sylvian fissure

 

Histological examination of the hypointense area on T1-weighted MRI showed multiple cystic structures with myxomatous background and proliferation of oligodendroglia-like cells with oval nuclei in the wall of the cystic spaces [ Figure 4a ]. Neuronal elements featuring “floating neurons” were observed, indicating a glioneuronal lesion within the cystic cavity [ Figure 4b ]. Immunohistochemical analysis revealed intense positive staining for Olig2, S-100 and synaptophysin, and less reactivity for IDH-1 [ Figure 4c ]. The Ki-67 staining index (SI) was 1% [ Figure 4d ]. A combined deletion of 1p and 19q chromosomes was absent. The histological diagnosis was WHO grade I DNET.


Figure 4

Photomicrographs of the surgical specimen showing the hypointense lesion on T1-weighted magnetic resonance imaging. (a) Multiple cystic structures are observed on a mucinous background. Hematoxylin and eosin (H and E) staining, original magnification ×100. (b) High-power view of a, showing floating neurons (arrows) scattered in the cystic structure. H and E staining, ×200. (c) Negative staining for IDH1 mutation, ×100. Inset: Positive control (d) The Ki-67 staining index was 1%, ×100

 

DISCUSSION

This rare case of DNET was followed for 10 years with annual MRI. Our statistical analysis of growth rate showed that the tumor volume gradually increased in direct proportion to time. According to this analysis, the tumor doubling time was determined to be approximately 3473 days. To date, only a few studies on DNET growth pattern have been reported.[ 3 4 ] We report the first case of DNET growth rate analysis in its natural course over a period of 10 years.

DNETs are usually stable tumors. However, malignant transformation in DNETs results in rapid growth rates.[ 3 10 ] It is still debated whether DNET is stable at birth or exhibits gradual growth in cases without malignant transformation. Jensen et al. reported a case of DNET that was stable for 15 years.[ 7 ] Conversely, Alexander et al. reported a case of DNET in which the occipital lobe grew from 5.2 cm to approximately 10.4 cm, accompanied by the appearance of enhanced tumor lesions on MRI for 10 years.[ 1 ] In our patient, the volume of DNET increased in direct proportion to the length of time without the appearance of enhanced lesions on MRI during its clinical course.

The lesion was located in the right insular cortex in our patient. DNETs typically occur in the temporal lobe in 62%, the frontal lobe in 31%, the parietal and/or occipital lobe in 7% of cases,[ 4 ] and rarely in the periventricular white matter, basal ganglia, thalamus, brainstem, and cerebellum[ 12 ] including the pons and third ventricle.[ 8 ] To the best of our knowledge, this is the first report of DNET located in the insular cortex.

DNET is generally positive for Olig2, S-100, and synaptophysin. The genetic background of DNETs has not been systemically investigated. Loss of heterozygosity at 1p/19q and TP53[ 5 ] or IDH1[ 2 ] mutations were not detected in DNETs. However, Maria et al. reported a case with 1p/19q chromosomal deletion and IDH1 mutation.[ 11 ] Most DNETs show very low proliferative activity and Ki-67 SIs lower than 1%.[ 9 ] Our results agreed with these findings.

The patient underwent an awake craniotomy. Gross total resection of the lesion was achieved. We expected an extremely low risk of tumor recurrence.[ 11 ] This case indicated that partial resection of DNETs might result in the regrowth of residual lesion in direct proportion to the length of time. Therefore, we suggest that patients with partial or subtotal resection of DNET be followed-up for longer time after surgery.

Despite being recognized for only less than three decades, DNETs are becoming an important part of epilepsy neurosurgical practice. However, their natural growth rate remains poorly understood. We present this case to increase the knowledge related to the growth pattern of these tumors, and to suggest that the growth rate of DNETs is directly proportional to time.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

Acknowledgments

We thank Erika Komura for assistance with preparation of the immunohistochemistry.

References

1. Alexander H, Tannenburg A, Walker DG, Coyne T. Progressive dysembryoplastic neuroepithelial tumour. J Clin Neurosci. 2015. 22: 221-4

2. Capper D, Reuss D, Schittenhelm J, Hartmann C, Bremer J, Sahm F. Mutation-specific IDH1 antibody differentiates oligodendrogliomas and oligoastrocytomas from other brain tumors with oligodendroglioma-like morphology. Acta Neuropathol. 2011. 121: 241-52

3. Daghistani R, Miller E, Kulkarni AV, Widjaja E. Atypical characteristics and behavior of dysembryoplastic neuroepithelial tumors. Neuroradiology. 2013. 55: 217-24

4. Daumas-Duport C, Scheithauer BW, Chodkiewicz JP, Laws ER, Vedrenne C. Dysembryoplastic neuroepithelial tumor: A surgically curable tumor of young patients with intractable partial seizures. Report of thirty-nine cases. Neurosurgery. 1988. 23: 545-56

5. Fujisawa H, Marukawa K, Hasegawa M, Tohma Y, Hayashi Y, Uchiyama N. Genetic differences between neurocytoma and dysembryoplastic neuroepithelial tumor and oligodendroglial tumors. J Neurosurg. 2002. 97: 1350-5

6. Herbet G, Lafargue G, Almairac F, Moritz-Gasser S, Bonnetblanc F, Duffau H. Disrupting the right pars opercularis with electrical stimulation frees the song: Case report. J Neurosurg. 2015. 123: 1401-4

7. Jensen RL, Caamano E, Jensen EM, Couldwell WT. Development of contrast enhancement after long-term observation of a dysembryoplastic neuroepithelial tumor. J Neurooncol. 2006. 78: 59-62

8. Leung SY, Gwi E, Ng HK, Fung CF, Yam KY. Dysembryoplastic neuroepithelial tumor. A tumor with small neuronal cells resembling oligodendroglioma. Am J Surg Pathol. 1994. 18: 604-14

9. Mano Y, Kumabe T, Shibahara I, Saito R, Sonoda Y, Watanabe M. Dynamic changes in magnetic resonance imaging appearance of dysembryoplastic neuroepithelial tumor with or without malignant transformation. J Neurosurg Pediatr. 2013. 11: 518-25

10. Moazzam AA, Wagle N, Shiroishi MS. Malignant transformation of DNETs: A case report and literature review. Neuroreport. 2014. 25: 894-9

11. Thom M, Toma A, An S, Martinian L, Hadjivassiliou G, Ratilal B. One hundred and one dysembryoplastic neuroepithelial tumors: An adult epilepsy series with immunohistochemical, molecular genetic, and clinical correlations and a review of the literature. J Neuropathol Exp Neurol. 2011. 70: 859-78

12. Yang AI, Khawaja AM, Ballester-Fuentes L, Pack SD, Abdullaev Z, Patronas NJ. Multifocal dysembryoplastic neuroepithelial tumours associated with refractory epilepsy. Epileptic Disord. 2014. 16: 328-32

Transanal presentation of a distal ventriculoperitoneal shunt catheter: Management of bowel perforation without laparotomy

$
0
0

James Bales, Ryan P. Morton, Nathan Airhart, David Flum, Anthony M. Avellino
  1. Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
  2. Department of Surgery, University of Washington School of Medicine, Seattle, Washington, USA

Correspondence Address:
James Bales
Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA

DOI:10.4103/2152-7806.196930

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: James Bales, Ryan P. Morton, Nathan Airhart, David Flum, Anthony M. Avellino. Transanal presentation of a distal ventriculoperitoneal shunt catheter: Management of bowel perforation without laparotomy. 28-Dec-2016;7:

How to cite this URL: James Bales, Ryan P. Morton, Nathan Airhart, David Flum, Anthony M. Avellino. Transanal presentation of a distal ventriculoperitoneal shunt catheter: Management of bowel perforation without laparotomy. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/transanal-presentation-of-a-distal-ventriculoperitoneal-shunt-catheter-management-of-bowel-perforation-without-laparotomy/

Date of Submission
06-Jan-2016

Date of Acceptance
20-Jun-2016

Date of Web Publication
28-Dec-2016

Abstract

Background:Bowel perforation is a serious but rare complication after a ventriculoperitoneal shunt (VPS) procedure. Prior studies have reported spontaneous bowel perforation after VPS placement in adults of up to 0.07%. Transanal catheter protrusion is a potential presentation of VPS bowel perforation and places a patient at risk for both peritonitis and ventriculitis/meningitis via retrograde migration of bacteria. This delayed complication can be fatal if unrecognized, with a 15% risk of mortality secondary to ventriculitis, peritonitis, or sepsis.

Case Description:We describe a unique case of a patient with distal VPS catheter protrusion from the anus whose bowel perforation did not cause clinical sequelae of infection. We were able to manage the patient without laparotomy.

Conclusions:A subset of patients can be managed without laparotomy and only with externalization of the ventricular shunt with antibiotics until the cerebrospinal fluid cultures finalize without growth.

Keywords: Bowel perforation, complications, ventriculoperitoneal shunt

INTRODUCTION

Peritoneal complications of ventricular shunt placement (VPS) are uncommon along with serious events.[ 1 7 9 14 ] Spontaneous bowel perforation after VPS has been reported with an incidence of 0.01–0.07% with a high mortality of up to 15%.[ 9 14 ] However, it still remains an underappreciated potential complication. There has been a shift toward the assistance of general surgery for the laparoscopic placement of the distal shunt tubing, and it is unclear if this affects the rate of bowel perforation.

Regardless of prompt recognition of this uncommon condition, important and delayed recognition can have significant consequences for patient care.

CASE REPORT

A 29-year-old male with shunted congenital hydrocephalus of unknown etiology with previous revisions in infancy and as a young child initially presented to the neurosurgery clinic with worsening headaches and complaints of blurred vision for more than 18 months. Computerized tomography (CT) of the head demonstrated a slight increase in his ventricular size; physical evaluation noted mild chronic papilledema and reduced visual acuity in his left eye, which was baseline following a car accident a few years earlier. He had a right parietal VPS in place; shunt series X-rays showed no shunt disconnections. In addition, abdominal X-rays demonstrated a retained peritoneal distal catheter from his previous shunt revision operations. Given the concerns for shunt failure, he underwent shunt exploration and revision for management. During the shunt revision surgery, the valve was found to be nonfunctional and was replaced; the retained peritoneal distal catheter was also removed laparoscopically by the general surgery team. His initial postoperative course was uncomplicated, and he was discharged on postoperative day 1 with decrease in his headaches and improvement in his subjective complaint of blurry vision.

He again presented 17 months later with continual headaches, decrease in vision, and increased ventricular size. This time he was noted to have acute papilledema and worsened visual acuity in his right eye. Given the concern regarding the age of the ventricular and distal catheter in his right parietal system, which had been placed at 4 months of age, it was determined that the placement of a new shunt system would be the best clinical option. He underwent another VPS revision with placement of a new right frontal VPS shunt and a new distal peritoneal catheter placed laparoscopically by the general surgery team [ Figure 1 ]. His initial postoperative course was uncomplicated, and the patient's headaches decreased, however, he did experience a lasting deficit in his visual acuity. Two months postoperatively, he presented to the emergency room with complaints of an object intermittently protruding from his rectum. At this initial emergency room evaluation, his rectal exam was unremarkable; on shunt series X-rays, the distal catheter was within the peritoneal cavity [ Figure 2 ]. The patient was subsequently discharged without neurosurgical consultation.


Figure 1

Postoperative abdominal X-ray film following the patient's second shunt revision demonstrating appropriate shunt placement. As noted, the peritoneal portion was placed laparoscopically by general surgery and was visualized to be within the peritoneal space

 

Figure 2

Initial evaluation in the emergency department demonstrating shunt catheter placement in the abdomen, which was originally interpreted as intraperitoneal

 

One month later, he presented with continued complaints of an object intermittently protruding from his rectum. During the emergency room evaluation, a neurosurgical consultation was obtained and the rectal exam revealed that the distal peritoneal catheter was protruding through his anus. X-rays corroborated the physical exam [ Figure 3a ]; CT imaging clearly revealed the distal peritoneal catheter within the large colon and rectum [ Figure 3b ]. On examination, the patient had no signs of peritonitis or meningitis, and he described no abdominal pain, feeding concerns, fevers, worsening headaches, or blurry vision.


Figure 3

(a) Abdominal shunt series X-ray showing the distal peritoneal catheter protruding through the rectum (arrow), and (b) abdominal computed tomography showing the distal peritoneal catheter within the bowel (arrow)

 

INTERVENTION PERFORMED

Because the patient was clinical asymptomatic except for the distal catheter protrusion through the anus, the general surgery team was consulted, and we decided to expose and externalize the distal catheter at the clavicle with simultaneous removal of the remaining distal catheter through the previous laparoscopic abdominal incision in a manner described previously.[ 1 ] Cerebrospinal fluid (CSF) taken at the time of the externalization of the shunt did not show any organisms on gram stain or subsequent bacterial growth on culture, with a normal glucose level of 72 mg/dL and a normal protein level of 23 mg/dL.

POSTOPERATIVE COURSE

The patient was placed on a triple antibiotic regiment of flagyl, vancomycin, and cefepime; daily CSF cultures from the externalized shunt revealed no bacterial growth. After 5 days of negative cultures, he underwent removal of all previous hardware and placement of a new right frontal ventriculoatrial shunt. His antibiotic regimen was continued for 1 day and he was discharged on postoperative day 2 without further antibiotics. At the patient's 6 month postoperative visit, he was doing well with no signs of infection and decreased headaches.

DISCUSSION

At our institution, it is common practice to enlist the assistance of general surgery to place the distal catheter laparoscopically. The reason for this is the theoretical advantage of visualizing the distal catheter within the peritoneal space, thus reducing the likelihood of incorrect placement; it also allows for a small incision and good wound healing at the distal site. However, as with any laparoscopic procedure, it is possible that a bowel injury was caused at the time of operation. The reported incidence of laparoscopic-induced bowel perforation is 0.22%, and most are recognized at the time of surgery.[ 12 ] There have been no reports of shunt bowel perforation with a laparoscopic approach to placement of the distal catheter. At this time, spontaneous bowel perforation appears to be the most likely cause, however, as laparoscopic approaches become more common, it will be important to pay close attention to the incidence of this uncommon complication.

The exact pathogenesis of spontaneous bowel perforation is unclear having been first reported by Wilson and Bertran[ 13 ] in two pediatric patients. Since the initial report, there have been approximately 90 documented cases in the literature regarding VPS-induced bowel perforation. In cases that have warranted surgical intervention, or by autopsy, the authors have described an encasing fibrotic scar anchoring the tubing to an area of the bowel and causing ulceration, and theoretically, eventual perforation.[ 3 ]

Clinical presentation may be straightforward with 44% of the patients having abdominal pain, vomiting, and fever, and 50% with clinical signs of meningitis.[ 4 6 14 ] Abdominal radiology can be diagnostic in a majority of these cases, and both X-ray and CT have been used with success.[ 4 5 11 ] Notably though, almost half of the patients with distal catheter bowel perforation may present without abdominal pain or signs of infection within the abdomen or shunt, which may hinder accurate diagnosis. Further complicating the clinical presentation of VPS-bowel perforation is the delayed nature of its presentation from the original surgery and the uncommon nature of this complication. Its presentation is so rare that colleagues outside the neurosurgical field may not be aware of this entity.[ 2 4 ] As with our patient, initial evaluation with abdominal shunt series X-rays 1 month prior to definitive diagnosis was interpreted as unremarkable and only on re-evaluation was the concern raised that the distal peritoneal catheter may be within the bowel based on the pattern of the distal catheter following the transverse and descending colon [ Figure 2 ].

Management of bowel perforation is highly individualized and dependent upon the presenting signs and symptoms of the patient. Immediate externalization is necessary to maintain shunt patency, as well as to limit the retrograde spread of bacteria along the shunt system which can cause ventriculitis or meningitis.[ 6 ] If there is a concern of abdominal abscess or peritonitis, laparotomy is the preferred treatment choice to manage the bacterial infection.[ 8 11 ] However, in cases where there is no evidence of peritoneal involvement and the patient's exam remains benign, it is believed that the fistulous opening should close spontaneously after removal of the catheter.[ 10 14 ] As we demonstrated here, this subset of patients can be managed without laparotomy and only with externalization of the ventricular shunt with antibiotics until the CSF cultures finalize without growth. Importantly, when re-shunting a patient, we highly recommend choosing a different terminus outside the abdominal cavity, as there remains the concern that the factors leading to bowel perforation are still present, such as the atrium (as in our case) or pleura.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Abu-Dalu K, Pode D, Hadani M, Safar A. Colonic complications of ventriculoperitoneal shunts. Neurosurgery. 1983. 13: 167-9

2. Akcora B, Serarslan Y, Sangun O. Bowel perforation and transanal protrusion of a ventriculoperitoneal shunt catheter. Pediatr Neurosurg. 2006. 42: 129-31

3. Brownlee JD, Brodley JS, Schaefer IK. Colonic perforation by ventriculoperitoneal shunt tubing: A case of suspected silicon allergy. Surg Neurol. 1998. 49: 21-4

4. Ferreira PR, Bizzi JJ, Amantea SL. Protrusion of ventriculoperitoneal shunt catheter through the anal orifice. A rare abdominal complication. J Pediatr Surg. 2005. 40: 1509-10

5. Hornig GW, Shillito J. Intestinal perforation by peritoneal shunt tubing: Report of two cases. Surg Neurol. 1990. 33: 288-90

6. Ibrahim AW. E-coli meningitis as an indicator of intestinal perforation by V-P shunt tube. Neurosurg Rev. 1998. 21: 194-7

7. Sathyanarayana S, Wylen EL, Baskaya MK, Nanda A. Spontaneous bowel perforation after ventriculoperitoneal shunt surgery: Case report and a review of 45 cases. Surg Neurol. 2000. 54: 388-96

8. Schulhof LA, Worth RM, Kalsbeck JE. Bowel perforation due to peritoneal shunt. A report of 7 cases and a review of the literature. Surg Neurol. 1975. 3: 265-9

9. Sells CJ, Loeser JD. Peritonitis following perforation of the bowel: A rare complication of a ventriculoperitoneal shunt. J Pediatr. 1973. 83: 823-4

10. Sharma A, Pandey AK, Radhakrishnan M, Kumbhani D, Das HS, Desai N. Endoscopic management of anal protrusion of ventriculo-peritoneal shunt. Indian J Gastroenterol. 2003. 22: 29-30

11. Snow RB, Lavyne MH, Fraser RA. Colonic perforation by ventriculoperitoneal shunts. Surg Neurol. 1986. 25: 173-7

12. van der Voort M, Heijnsdijk EA, Gouma DJ. Bowel injury as a complication of laparoscopy. Br J Surg. 2004. 91: 1253-8

13. Wilson CB, Bertan V. Perforation of the bowel complicating peritoneal shunt for hydrocephalus. Report of two cases. Am Surg. 1966. 32: 601-3

14. Yousfi MM, Jackson NS, Abbas M, Zimmerman RS, Fleischer DE. Bowel perforation complicating ventriculoperitoneal shunt: Case report and review. Gastrointest Endosc. 2003. 58: 144-8

Squamous cell carcinoma arising from neglected meningocele

$
0
0

Abrar A. Wani, Uday K. Raswan, Nayil K. Malik, Altaf U. Ramzan, Iqbal Lone
  1. Department of Neurosurgery, Sher-i- Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
  2. Department of Pathology, Sher-i- Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India

Correspondence Address:
Abrar A. Wani
Department of Pathology, Sher-i- Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India

DOI:10.4103/2152-7806.196928

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Abrar A. Wani, Uday K. Raswan, Nayil K. Malik, Altaf U. Ramzan, Iqbal Lone. Squamous cell carcinoma arising from neglected meningocele. 28-Dec-2016;7:

How to cite this URL: Abrar A. Wani, Uday K. Raswan, Nayil K. Malik, Altaf U. Ramzan, Iqbal Lone. Squamous cell carcinoma arising from neglected meningocele. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/squamous-cell-carcinoma-arising-from-neglected-meningocele/

Date of Submission
22-Jul-2016

Date of Acceptance
13-Sep-2016

Date of Web Publication
28-Dec-2016

Abstract

Background:A neural tube defect (NTD) is a common congenital anomaly with an incidence of 6.57–8.21 per 1000 live births. Patients usually present early because of obvious swelling or due to neurological deficit. However, neglecting the obvious cystic swelling on the back till its transformation into malignant tumor is rare.

Case Description:We describe a case of malignant transformation of meningocele in a 60-year-old man. Magnetic resonance imaging showed sacral meningocele. Neurological examination revealed intact motor and sensory examination with normal bladder and bowel function. There were no signs of meningitis and hydrocephalus. Excision was done and biopsy revealed it as squamous cell carcinoma.

Conclusion:Meningocele should be treated early and possibility of malignant change should be kept in mind in neglected cases presenting in adulthood.

Keywords: Adult, meningocele, neural tube defects, squamous cell carcinoma

INTRODUCTION

A neural tube defect (NTD) is a common congenital anomaly with an incidence of 6.57–8.21 per 1000 live births.[ 1 3 ] Patients usually present early because of obvious swelling or due to neurological deficit. However, neglecting the obvious cystic swelling on the back till adulthood is rare. To the best of our literature search, we could find only few such cases.[ 2 4 5 6 7 ]

CASE REPORT

A 60-year-old man presented with complaints of discharge from a swelling in the sacral area. At the time of birth he was noted to have a sacral meningocele for which he was advised surgery, however, his family had refused and the wound surface slowly became abraded and exudated repeatedly over a period of years. One month before the admission, the swelling started discharging foul smelling fluid and increased in size. Inspection showed a swelling in the sacral region of 5 cm in diameter, consisting of cauliflower-shaped swelling with yellowish slough [ Figure 1 ]. The area smelled foul and was constantly draining serosanguinous fluid. Neurological examination revealed intact motor and sensory examination with normal bladder and bowel function. There were no signs of meningitis and hydrocephalus. Magnetic resonance imaging (MRI) showed sacral meningocele with sinus tract [Figure 2a and b ]. The tumor was excised, dural attachment was removed, and dura was closed again [ Figure 3 ].


Figure 1

Adult sacral meningocele with yellowish slough over it

 

Figure 2

(a, b) Magnetic resonance imaging of the spine (T1, T2 sagittal view) showing sacral meningocele

 

Figure 3

Operative photograph showing the swelling being excised

 

Pathological finding

The tissue sections were stained in hematoxylin and eosin (H and E) stain, and the histopathology study revealed tumor cells arranged in sheets and nests with keratin pearl formation [ Figure 4 ], suggestive of well-differentiated squamous cell carcinoma. On high power examination, these tumor cells were large with high nuclei/cytoplasmic ratio and prominent nucleoli.


Figure 4

Photomicrograph shows tumor cells arranged in sheets and central keratin pearl (marked arrow), which is suggestive of it being squamous cell carcinoma (H and E stain, ×400)

 

Postoperative course

The postoperative recovery was uneventful and the wounds healed by primary intention. Further, the patient was sent to the oncology department for adjuvant therapy. Postoperatively, the patient has been on follow-up for a year without any recurrence.

DISCUSSION

A meningocele is a congenital anomaly of neural arch fusion in association with an open neural tube defect, and is characterized by protrusion of spinal meninges which contain cerebrospinal fluid without involvement of the neural tissue. Most meningoceles are surgically repaired during the new-born period or at least in childhood. The incidence of survival is low without intervention, and hence, adult meningoceles are rarely seen. Life expectancy at birth is shorter in myelomeningocele patients, although effective treatment for hydrocephalus and intermittent catheterization for the management of the neurogenic bladder can improve the quality of life for these patients. Posterior lumbosacral meningocele cases have rarely been reported. The long-term follow up results for adults with sacral myelomeningocele are not as good as in children because other neurological abnormalities such as hydromyelia, syringomyelia, tethered cord, Chiari malformations, and hydrocephalus accompany this lesion.[ 3 ]

A search of the literature revealed four cases similar to the one we have described. Saskun et al.[ 6 ] biopsied a neglected case of lumbosacral myelomeningocele who presented with fungating growth. Histology showed squamous cell carcinoma and radiotherapy was instituted. In the case reported by Thorp,[ 7 ] surgical treatment was chosen initially for a 26-year-old man with a carcinoma at the site of a lumbar meningomyelocele. Six months after resection, the tumor recurred and was treated with radiotherapy. Later, the tumor appeared again, necessitating further surgery. Three weeks postoperatively, the patient the patient died of septicemia. In the case described by Pope and Todorovl,[ 5 ] a squamous cell carcinoma developed at the site of a cervical meningomyelocele in a 37-year-old man. This lesion was easily excised, without complications because the defect was a meningocele, and hence did not contain neural elements or connect with the spinal canal. Hong- Zhou et al.[ 2 ] excised a cauliflower-shaped lumbosacral myelomeningocele in an 11-year-old boy, who had the swelling since birth. It turned out to be squamous cell carcinoma on histology. Our patient presented as a neglected case of sacral meningocele with a discharge from the swelling and normal neurological examination. Excision of the mass was done and biopsy revealed squamous cell carcinoma.

MRI is a good investigation choice for evaluating the meningocele sac in the sagittal plane, and to observe the spinal cord itself, as well as the possible congenital anomalies associated with it.[ 2 4 ]. In meningocele cases, neurological involvement are not seen as often as in myelomeningocele lesions, however, the local signs of sacral nerve involvement are seen as pain in both legs and bladder dysfunction. Taking this into consideration, somatosensory evoked potentials (SSEP) can be used in these patients as in myelomeningoceles.[ 4 ]

One must be aware of possible neoplastic change in untreated meningomyelocele after years of mechanical irritation and chronic bacterial infection. This seems to be a probable cause in our case. The defect, lacking a protective epithelial cover, must be viewed regularly with a high degree of suspicion, as with other chronic ulcers. Squamous cell carcinoma is a well-known complication of burn and chronic venous ulcer, pilonidal sinuses, longstanding bacterial and fungal infections, vaccination scars, and even tattoos.[ 6 ]

Once change is noted, biopsies should be done to establish a histological diagnosis. If malignancy is established, an intensive search for metastases, lymph node involvement, and local invasion must be made for proper staging and subsequent treatment.[ 6 ]

CONCLUSION

In conclusion, meningocele should be treated as soon as diagnosed and possibility of malignant change should be kept in mind in neglected cases presenting in adulthood.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Cherian A, Serena S, Bullock RK, Antony AC. Incidence Of Neural Tube Defects In The Least-Developed Area Of India: A Population-Based Study. Lancet. 2005. 366: 930-1

2. Duan HZ, Zhang Y, Zhang JY, Bao SD, Zhou CQ. A Case Report of Squamous Cell Carcinoma Arising In a Patient with Meningomyelocele. Beijing Da Xue Xue Bao. 2009. 41: 489-91

3. Laharwal MA, Sarmast AH, Ramzan AU, Wani AA, Malik NK, Arif SH. Epidemiology of the neural tube defects in Kashmir valley. Surg Neurol Int. 2016. 7: 35-

4. Ozdemir NG, Atci IB, Antar V, Yllmaz H, Bitirak G, Katar S. Lumbosacral Meningocele In Adulthood. Cukurova Med J. 2015. 40: 131-5

5. Pope M, Todorov AB. Cutaneous squamous cell carcinoma as a rare complication of cervical meningocele. Birth Defects. 1975. 11: 336-

6. Saskun JM, Fisher BK. Squamous Cell Carcinoma Arising In a Meningomyelocele. Can Med Assoc J. 1978. 119: 739-41

7. Thorp RH. Carcinoma Associated With Myelomeningocele. Case Report. J Neurosurg. 1967. 27: 446-8

Rare complication of ventriculoperitoneal shunt: Catheter protrusion to subcutaneous tissue – Case report

$
0
0

Luana Antunes Maranha Gatto, Roger Mathias, Rogério Tuma, Ricardo Abdalla, Paulo Henrique Pires de Aguiar
  1. Department of Neurosurgery and Interventional Neuroradiology, University Hospital Cajuru, Curitiba, PR, Brazil
  2. Department of Neurosurgery of Bragança University, Division of Neurosurgery, Sirio LIbanês Hospital, São Paulo, SP, Brazil
  3. Divisions of Neurology, Sirio LIbanês Hospital, São Paulo, SP, Brazil
  4. Divisions of Surgery, Sirio LIbanês Hospital, São Paulo, SP, Brazil
  5. Divisions of Neurosurgery, Sirio LIbanês Hospital, São Paulo, SP, Brazil

Correspondence Address:
Luana Antunes Maranha Gatto
Divisions of Neurosurgery, Sirio LIbanês Hospital, São Paulo, SP, Brazil

DOI:10.4103/2152-7806.196926

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Luana Antunes Maranha Gatto, Roger Mathias, Rogério Tuma, Ricardo Abdalla, Paulo Henrique Pires de Aguiar. Rare complication of ventriculoperitoneal shunt: Catheter protrusion to subcutaneous tissue – Case report. 28-Dec-2016;7:

How to cite this URL: Luana Antunes Maranha Gatto, Roger Mathias, Rogério Tuma, Ricardo Abdalla, Paulo Henrique Pires de Aguiar. Rare complication of ventriculoperitoneal shunt: Catheter protrusion to subcutaneous tissue – Case report. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/rare-complication-of-ventriculoperitoneal-shunt-catheter-protrusion-to-subcutaneous-tissue-case-report/

Date of Submission
18-Apr-2016

Date of Acceptance
30-Sep-2016

Date of Web Publication
28-Dec-2016

Abstract

Background:Ventriculoperitoneal (VP) shunt is a day-to-day procedure performed by a neurosurgeon. The most frequent associated complications are obstructive and infectious. Although rare, there are well-reported complications related to the poor positioning of the distal catheter, with perforation of organs and tissues. Still rarer are the complications related to the migration of this catheter.

Case Description:We describe an atypical case of VP shunt postoperative by normal pressure hydrocephalus. After well-documented proper positioning of the distal catheter into the intraperitoneal cavity, it protruded into the subcutaneous space. Even on a new documented satisfactory abdominal tomography, this catheter migrated back again to the subcutaneous tissue.

Conclusion:We did not find plausible explanation for this rare event.

Keywords: Catheters, cerebrospinal fluid shunts, normal pressure hydrocephalus, postoperative complications, surgically-created structures, ventriculoperitoneal shunt

INTRODUCTION

Placement of a ventriculoperitoneal (VP) shunt is the most common treatment for hydrocephalus.[ 6 ] It is a routine, common, and effective procedure in Neurosurgery.[ 2 ] Complications of VP shunts may occur anywhere along their course from the cerebral ventricle to the peritoneal cavity.[ 6 ] Valvular dysfunction secondary to the obstruction of proximal catheter is relatively frequent in the emergency room. However, non-infectious obstruction of distal catheter is exceptional.[ 9 ] Rare complications such as migration of the peritoneal catheter into the stomach, liver, gallbladder, vagina, scrotum, bladder, bowel, colon, pulmonary artery, diaphragm, cardiac ventricle, cervical area, umbilicus, rectum, anus, and mouth have been described in the literature. We report a case of a male patient with normal pressure hydrocephalus (NPH) submitted to VP shunt. The distal catheter protruded from the intraperitoneal cavity and lodged in the subcutaneous tissue, leading to collection of cerebrospinal fluid (CSF). The same thing occurred after another surgery for catheter repositioning. During both times, control computed tomography (CT) scans had recorded proper positioning of all shunt systems. This unusual complication is extremely uncommon. According to our records, there is no case cited in the past.

CASE REPORT

A male patient, 63-year-old, with no known comorbidities and a cocaine user, presented with over 1 year of classic triad of dementia, urinary incontinence, and ataxia march. The diagnosis of NPH was confirmed by CT scan, with Evans index of 0.62, as shown in Figure 1 . Therapeutic test (tap test) was positive for improvement of symptoms after 50 mL drain of CSF by lumbar puncture. He was submitted to ventriculoperitoneal shunt uneventfully. In our hospital, we routinely perform CT scans of the skull and abdomen as postoperative control, and both showed the entire system to be positioned properly [Figures 2 and 3 ]. However, on the second postoperative day, the patient presented with clinical worsening, with recurrence of early symptoms, and an abdominal collection was identified by superficial palpation. New abdomen CT identified the distal catheter protruding from the peritoneal cavity and housed beneath the subcutaneous tissue, producing a collection of CSF, as shown in Figure 4 . During further surgery to reposition the catheter, it was identified immediately below the skin after the incision, and no other pathological findings were noteworthy [Figures 5 and 6 ]. The shunt was otherwise functioning, however, there was a subcutaneous collection filled with a turbid CSF. New control CT showed the catheter well positioned in the peritoneal cavity [ Figure 7 ] and the patient symptoms improved. However, again 2 days after the last surgery, a superficial collection in the abdomen was identified, similar to the previous event. Another CT scan suggested the same event, with prolapsed and allocated catheter in abdominal subcutaneous and a collection of CSF, as illustrated in Figure 8 . New surgical repositioning of the entire DVP system was performed uneventfully, with appropriate control scans [Figures 9 and 10 ].


Figure 1

Computed tomography scan showing Evans index of 0.62, confirming the diagnosis of normal pressure hydrocephalus

 

Figure 2

Computed tomography scan after ventriculoperitoneal shunt with the catheter well positioned in the anterior horn of the right lateral ventricle

 

Figure 3

Abdomen tomography after ventriculoperitoneal shunt with the catheter well positioned in the peritoneal cavity

 

Figure 4

Abdomen tomography two days later, with the catheter lodged in the subcutaneous tissue, and cerebrospinal fluid collection in the same space

 

Figure 5

Aspect of coiling of the distal catheter after opening the abdominal incision

 

Figure 6

Distal catheter functioning wrapped in the subcutaneous space resulting in collection of cerebrospinal fluid

 

Figure 7

Abdomen tomography after repositioning of the distal catheter spread into the peritoneal cavity with no other complications

 

Figure 8

Abdominal tomography 2 days after the second surgery demonstrating new protrusion of the catheter from the peritoneal cavity into the subcutaneous space

 

Figure 9

Final control CT scan, with the ventricular catheter in the proper position

 

Figure 10

Final control abdomen tomography, with the peritoneal catheter in proper position

 

The patient was discharged on the fifth postoperative day of the second surgical shunt revision. No further problems were noted during regular follow-ups at the outpatient office, with significant clinical improvement in gait and bladder control, although with no marked cognitive improvement. CT scan showed reduction of brain ventricles globally. He had clean and dry operative wounds, functioning valve to palpation; and the abdomen flat, flaccid, and not painful on palpation.

DISCUSSION

Infectious and obstructive are the most frequent complications after shunt surgery.[ 1 8 ] Other types are less common, and eventually occur due to technical errors during brain ventricular puncture, opening the intraperitoneal cavity and the catheter tunneling between the two points.

There are several cases reports of distal catheter migration to cardiac ventricle.[ 2 ] The hypothesis is an incidental perforation of internal jugular vein during tunnelization, added to negative inspiratory pressure and orthograde, as well as slow blood flow that may draw the catheter proximally through vein and eventually to the heart or even to the pulmonary artery.[ 2 4 6 8 ] These could be resolved with a pericardial window by cardiac surgeons[ 4 ] by an endovascular approach, either guided by fluoroscopy through the internal jugular vein, or via femoral and inferior vena cava.[ 2 6 ]

Other documented complication of VP drainage is effusion of CSF through pleura (hydrothorax), peritoneum (ascite) and peritoneum-vaginal processus (hydrocele),[ 7 9 ] all for absorption issues and normally in children.

In a large series of 1585 VP shunts, only 0.7% developed large abdominal cysts, and the associated causes were infection, particularly by Staphylococcus epidermidis and multiple-shunt revisions. Simple parencentesis and replacement of the shunt usually are sufficient treatment for this complication, if infection is not present.[ 5 ]

Our case had a complication not as serious as the aforementioned because there was no perforation of the hollow viscus or penetration of involved organs, and there was no infection. Nevertheless, the involved mechanisms with twice protrusion of distal catheter are not easily explicated.

One revision in 2013 of 12 cases of upward VP catheter migration postulated the following hypothesis.[ 1 ] (1) Reabsorption of subgaleal fluid may generate negative pressure, dragging the distal tubing proximally; (2) “Windlass effect,” with some granulation tissue or valve placed below the scalp, which acts as an anchoring point, and the patient's repeated head motion allows the distal tubing to be pulled in a proximal direction; (3) Increased abdominal pressure during Valsalva maneuver may act as a pushing force for distal tubing to migrate upward; (4) The memory of devices placed in packaging allows the tubing to recoil, as explained by the “memory phenomenon;”[ 3 ] (5) Unishunt catheters with a spring coil mechanism and no interposed valves or flushing devices are more frequently involved in migration. Shunt migration depends on various factors such as the type of catheter and reservoir used (for example, shunt migration in children is more frequent). None of the migration hypotheses are clearly defined but many similar cases include the “windlass effect.”[ 1 ]

One of these cases was a child with proximal migration of the VP catheter and extrusion of the ventricular catheter. This resulted in the entire VP shunt along with the shunt chamber lying in a subgaleal pocket in the occipital region in a tightly coiled manner. This coiling was very similar in appearance to that of the pre-insertion shunt in the packaging when it was supplied; hence, it was postulated that the migration was secondary to retained “memory” of the shunt tubing.[ 3 ]

Regardless of what mechanism was involved in the case of our patient, we did not find any cases in the literature with the occurrence of two consecutive extrusions of distal catheter. Further, we did not find references about protrusion only for abdominal subcutaneous space. We noted that in both instances there were imaging studies documenting the good positioning of the entire system previously.

CONCLUSIONS

We emphasize the importance of careful and proper placement of the distal catheter during the tunneling procedure to prevent complications, as well as the importance of carrying out control tests to document the satisfactory placement of the entire shunt system.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Cho KR, Yeon JY, Shin HJ. Upward migration of a peritoneal catheter following ventriculoperitoneal shunt. J Korean Neurosurg Soc. 2013. 53: 383-5

2. Chong JY, Kim JM, Cho DC, Kim CH. Upward migration of distal ventriculoperitoneal shunt catheter into the heart: Case report. J Korean Neurosurg Soc. 2008. 44: 170-3

3. Dominguez CJ, Tyagi A, Hall G, Timothy J, Chumas PD. Sub-galeal coiling of the proximal and distal components of a ventriculo-peritoneal shunt. An unusual complication and proposed mechanism. Childs Nerv Syst. 2000. 16: 493-5

4. Frazier JL, Wang PP, Patel SH, Benson JE, Cameron DE, Hoon AH. Unusual migration of the distal catheter of a ventriculoperitoneal shunt into the heart: Case report. Neurosurgery. 2002. 51: 819-22

5. Gutierrez FA, Raimondi AJ. Peritoneal cysts: A complication of ventriculoperitoneal shunts. Surgery. 1976. 79: 188-92

6. Hermann EJ, Zimmermann M, Marquardt G. Ventriculoperitoneal shunt migration into the pulmonary artery. Acta Neurochir. 2009. 151: 647-52

7. Kim JH, Roberts DW, Bauer DF. CSF hydrothorax without intrathoracic catheter migration in children with ventriculoperitoneal shunt. Surg Neurol Int. 2015. 23(Suppl 11): S330-3

8. Manix M, Sin A, Nanda A. Distal ventriculoperitoneal shunt catheter migration to the right ventricle of the heart--A case report. J La State Med Soc. 2014. 166: 21-5

9. Rivero-Garvía M, Barbeito Gaído JL, Morcillo J, Márquez Rivas J. Shunt dysfunction secondary to peritoneal catheter migration to the scrotum. Arch Argent Pediatr. 2013. 111: e14-6

Commentary

Jorge Lazareff
  1. Center for World Health, Department of Neurosurgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA E-mail: jalazareff@mednet.ucla.edu

The natural history of a surgical complication has always been a puzzle. We surgeons tend to believe that the complication is the derailment of a neat set of events, and when it happens, we search for the culprit.

Following this logic, “Rare complication of ventriculo peritoneal shunt: catheter protrusion to subcutaneous tissue-Case report” can be read as an improper closure of the peritoneal wall, or an increase in abdominal pressure that perhaps was combined with a minimally imperfect stitching of the wound. If so, a heartfelt praise for the authors honesty would be enough.

However, I propose that complications reports should be read assuming, without reservations, that those directly involved in the surgical procedure performed it up to the more stringent standard. Hence, there is no smoking gun in anybody hands. And this seems to be the conclusion of the authors unless until they retract and state that careful and proper placement of the distal catheter during the tunneling procedure to prevent complications, as well as the importance of carrying out control tests to document the satisfactory placement of the entire shunt system. But, I ask, do we not do it all the time? Did they not do it the first time? Of course they did it, of course we all do it in every case. And still the events recur. To the authors of the paper it happened twice on the same patient. And I feel it is a disservice to what nature is trying to tell us to treat this report as the report of a complication. The epistemological question of the complication is, has the complication a different set of rules? Philosophy is the process of interrogating nature, with surgical complications we surgeons interrogate ourselves.

Descartes stated that a stopped watch had a different ontology than a working one. Perhaps we could borrow the analogy and apply it to the surgical complications, certainly not to those were an egregious mistake was committed (those are rarely if ever reported) but those as the one reported here that lead us to pause and ponder the phenomenon as an independent entity not necessarily a continuum from the original procedure.

Modern management of medulloblastoma: Molecular classification, outcomes, and the role of surgery

$
0
0

Visish M. Srinivasan, Michael G. Z. Ghali, Robert Y. North, Zain Boghani, Daniel Hansen, Sandi Lam
  1. Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA
  2. Department of Neurobiology, Drexel University College of Medicine, Philadelphia, USA

Correspondence Address:
Sandi Lam
Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA

DOI:10.4103/2152-7806.196922

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Visish M. Srinivasan, Michael G. Z. Ghali, Robert Y. North, Zain Boghani, Daniel Hansen, Sandi Lam. Modern management of medulloblastoma: Molecular classification, outcomes, and the role of surgery. 28-Dec-2016;7:

How to cite this URL: Visish M. Srinivasan, Michael G. Z. Ghali, Robert Y. North, Zain Boghani, Daniel Hansen, Sandi Lam. Modern management of medulloblastoma: Molecular classification, outcomes, and the role of surgery. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/modern-management-of-medulloblastoma-molecular-classification-outcomes-and-the-role-of-surgery/

Date of Submission
20-Aug-2016

Date of Acceptance
14-Oct-2016

Date of Web Publication
28-Dec-2016

Keywords: Medulloblastoma, pediatric, posterior fossa syndrome

ILLUSTRATIVE CASE

A 4-year-old boy presented with papilledema and nystagmus. He had developed eye deviation, clumsiness, and episodes of dizziness starting 6 months prior to presentation, which progressed in frequency. Following an episode of emesis, he was brought to our attention. One week prior to presentation, the patient had one episode of emesis in the morning. His development had been notable for speech delay, with expressive language limited to speaking in short phrases without full sentence formation; he otherwise had normal growth and motor development.

Physical examination was significant for sluggish and dilated pupils, agitation, crying, and attention difficulty. Computer tomography (CT) scan of the brain revealed a large posterior fossa mass arising from the vermis with multiple calcifications and associated obstructive hydrocephalus. Magnetic resonance imaging (MRI) with and without contrast of the brain showed a mass with patchy enhancement and associated metastatic lesions located throughout the cerebellar hemispheres [ Figure 1 ]. MRI of the spine demonstrated two focal enhancing nodules of spinal cord in the cervical and thoracic spine.


Figure 1

Preoperative MRI Brain, T1 post-gadolinium in sagittal (a) and axial (b) sections from patient in the example case. The large midline medulloblastoma arises from the cerebellar vermis and compresses the fourth ventricle, causing obstructive hydrocephalus. There is nodular leptomeningeal spread throughout the posterior fossa

 

Once in the operating room, a frontal external ventricular drain was placed prior to positioning the patient prone for a midline suboccipital craniotomy with transvermian approach splitting the inferior aspect of the vermis. The tumor was highly vascular and noted to be involving the floor of the fourth ventricle, bilateral foramina of Luschka, and left cerebellar peduncle, precluding a complete resection. Histopathology confirmed a medulloblastoma with subsequent molecular definition of a non-WNT(wingless)/non-SHH(sonic hedgehog) subgroup.

Postoperatively, the patient exhibited decreased responsiveness, mutism, fixed downward gaze, and inability to follow commands. Postoperative imaging did not demonstrate any hemorrhage [ Figure 2 ]. He was dependent on cerebrospinal fluid (CSF) drainage, requiring a ventriculoperitoneal shunt on postoperative day 14. He had an extended postoperative course complicated by shunt infection but eventually was able to undergo adjuvant radiation and chemotherapy. Following craniospinal proton radiotherapy, he improved in his mental status and purposeful interaction and regained three-word speech. However, significant disease burden still persisted on imaging [ Figure 3 ].


Figure 2

Postoperative MRI Brain, T1 post-gadolinium in sagittal (a) and axial (b) sections. The medial portion of the lesion has been largely resected, but the lesion remains in the bilateral cerebellar hemispheres, totaling >1.5 cm2

 

Figure 3

Post-radiotherapy MRI Brain, T1 post-gadolinium in sagittal (a) and axial (b) sections. There has been some interval improvement of leptomeningeal spread and nodular lesions. However, there has been recurrence of disease in the fourth ventricle

 

OVERVIEW OF MEDULLOBLASTOMA

Medulloblastoma (MB) is the most common posterior fossa tumor in children, presenting at a mean age of 9 years and, more commonly, in males (ratio 2:1).[ 2 ] It usually presents with obstructive hydrocephalus and resultant symptoms, which may also be accompanied by ataxia, cranial neuropathies, brainstem dysfunction, or nerve root/spinal cord compression from metastatic disease. Symptoms are often progressive over weeks to months, and it is not uncommon for patients to have an extended symptomatic period prior to initial diagnosis. Metastatic disease is commonly present at diagnosis (40%), and imaging of the entire craniospinal axis is an essential part of the initial diagnostic evaluation. Most cases of MB arise sporadically but are also linked to a number of syndromes, including Gorlin, Li-Fraumeni, and Turcot syndromes.

Molecular subgrouping

In addition to traditional histologic classification [classic, desmoplastic/nodular (DNMB), MB with extensive nodularity (MBEN), large cell/anaplastic (LCA)), MB has been classified into four distinct molecular subgroups according to transcriptional profiling studies.[ 41 ] The consensus four subgroups, which include WNT, SHH, Group 3, and Group 4, have been correlated to both clinical outcomes and histologic appearance.[ 2 41 ] These genetically defined molecular subgroups form a crucial aspect of the forthcoming World Health Organization 2016 guidelines, which divides MB into WNT, SHH-TP53 wild type, SHH-TP53 mutant, non-WNT/non-SHH, and MB-NOS.[ 31 ] Unfortunately, molecular studies, while available, remain isolated to large tertiary centers, even in developed countries.

Genetics

The WNT and SHH classifications identify the underlying oncopathogenic pathway, whereas Groups 3 and 4 retain generic designations pending elucidation of underlying cellular pathobiology. WNT tumors result from unregulated WNT signaling leading to increased beta-catenin-mediated increase in transcriptional activity and consequent tumorigenesis. They are associated with monosomy 6, germline APC mutations (Turcot syndrome), somatic CTNNB1 mutations, and nuclear positivity for β-catenin.[ 17 48 ] Oncogenesis of SHH MBs results from upregulation of sonic hedgehog signaling as a consequence of loss of function of the tumor suppressors suppressor of fused gene (SUFU) and patched-1 (PTCH1). Other events implicated in pathogenesis of SHH MBs include mutations of PTCH1, PTCH2, SUFU, and SMO, as well as amplification of GLI1 and GLI2.[ 7 8 26 27 28 37 39 40 ]

The precise oncopathogenic mechanisms for non-WNT/SHH tumors are still under investigation. Altered MYC and KDM6A signaling has been implicated in Groups 3 and 4 MBs. Whereas MYC is often overamplified in Group 3 MBs, Group 4 MBs are occasionally characterized by MYCN and CDK6 amplification. Isochromosome 17q is characteristic of Group 4 tumors but is also occasionally observed in Group 3 MBs, making it poorly specific. An alternative proposed marker for Group 4 lesions is KCNA1.[ 26 ]

Whereas molecular subgrouping of MB is based on differential gene transcriptional profiles, the different subgroups are recapitulated by subgroup-specific differential heterogeneity of cis-regulatory elements in the epigenome.[ 21 ] Based on these studies, it has been proposed that Group 4 MBs may arise from cells in the deep cerebellar nuclei of the cerebellar nuclear transitory zone or the upper rhombic lip.[ 21 ] Probing and further delineating epigenetic and gene regulatory differences may serve not only as better molecular subgrouping classification metrics but also the dual purpose of uncovering oncopathogenesis of different types of MB as well as of offering novel therapeutic targets.

Another compelling area of investigation in the genetics of MB is recurrent disease. Much of our understanding of the molecular and genetic profiles of MB is derived from experiments with treatment naïve and primary site disease. However, much like other malignancies, there is mounting evidence that suggests clonal selection and genetic divergence may play an important role in MB recurrence.[ 25 ] Although it has been demonstrated that molecular subgroups (WNT, SHH, Group 3, Group 4) are preserved in both metastatic and recurrent disease, for recurrence this may be insufficient to guide molecularly targeted therapies because significant genetic divergence, including increased mutational burden with both loss and gain of actionable molecular targets, has been demonstrated.[ 25 44 ]

Histologic correlation

There is an association between molecular subgroup and histologic type. For instance, 97% of WNT MBs are of the classic histologic variant.[ 19 ] However, this correlation is not absolute; in infants, children, and adults, 89%, 25%, and 100% of DNMBs were of the SHH molecular subgroup, respectively.[ 19 ] LCA tumors in infants are most commonly Group 3 lesions, however, in other ages they are evenly distributed across molecular subgroups.

Epidemiologic correlation

WNT, SHH, Group 3, and Group 4 MBs account for 10%, 30%, 25%, and 35% of MBs overall, respectively, with a 1:1 M:F ratio for WNT and SHH subgroups, and a 2:1 male predominance for non-SHH/WNT tumors. WNT tumors are typically seen in children and adults, whereas Group 3 tumors are more often seen in infants and children. SHH and Group 4 tumors are seen across all age groups, with the former exhibiting a bimodal age distribution, most typically occurring in patients <4 and >16 years of age.[ 10 ]

Surgical treatment

Extent of resection

The extent of resection indicated in MBs largely depends on the unique anatomy of the tumor and what can be done safely and without the incurrence of neurological deficit, as with all tumors in the eloquent areas of brain. Although no clinical trials have been designed to specifically evaluate the role of surgery for MB, there have been many studies supporting a relationship of extent of resection with event-free survival. The most influential is likely a retrospective analysis of 233 children involved in a randomized controlled trial of differing chemotherapy regimens by Albright et al. determining that a radiographically measured residual tumor less than 1.5 cm3 was associated with improvement in 5-year PFS of greater than 20% in patients with M0 disease and an 11% difference for all patients irrespective of age, M stage, or any other measured factors.[ 3 ] However, there have also been a number of studies that question a definitive association between the extent of resection and survival. Possibly, the most compelling of these studies was a recent retrospective analysis of 787 patients by Thompson et al. They demonstrated that the benefit of increased extent of resection is largely attenuated after taking into account molecular subtype and not significant when comparing STR (>1.5 cm3) versus NTR (<1.5 cm3) or gross total resection (GTR; no radiographic residual) versus NTR (<1.5 cm3).[ 43 ] In addition, aggressive resection of brainstem disease is not indicated owing to the high potential for morbidity incurred with this approach as well as the high sensitivity of the tumor to radiation and chemotherapy. For tumors involving the brainstem, investigators have found no difference in outcome between GTR and residual tumor <1.5 cc.[ 46 ]

Hydrocephalus

Following resection, between 10 and 40% of the patients have hydrocephalus requiring CSF diversion.[ 2 20 33 ] Riva-Cambrin et al. developed the Canadian Preoperative Prediction Rule for Hydrocephalus (CPPRH) for use in the preoperative prediction of shunt dependence, which can aid in surgical planning and patient counseling.[ 33 ] Our patient had a CPPRH score of 7/10 and, thus, a predicted risk of hydrocephalus of 79.9%. It is important to expedite shunt placement and not to delay adjuvant therapies, especially if leptomeningeal spread or metastases have occurred.

Adjuvant therapies

Risk stratification and radiotherapy

Traditionally, children older than 3 years of age are stratified into average and high-risk prognostic groups based on the presence of metastatic disease and post-resection residual less or greater than 1.5 cm3. “High-risk” MB is defined as having any one of the following characteristics: >1.5 cm3 postoperative residual, evidence of radiographic metastases, or presence of leptomeningeal disease/CSF seeding, with the remaining patients defined as “average-risk.”[ 2 ] Those less than 3 years of age constitute a unique group in which current standard of care is chemotherapy alone as first-line adjunctive therapy, with radiation therapy eschewed in order to avoid the very poor neurocognitive outcomes associated with typical doses of craniospinal irradiation (CSI) in such young patients.

Under the above scheme, “high-risk” patients undergo posterior fossa or surgical bed radiation (54–55.8 Gy) with high-dose CSI (36.0 Gy) followed by adjuvant chemotherapy, whereas “average-risk” patients undergo posterior fossa or surgical bed radiation (54–55.8 Gy) with reduced-dose CSI (23.4 Gy) followed by adjuvant chemotherapy.[ 2 ] However, molecular subgrouping is already being applied to clinical trials of varied adjuvant therapy schemes, and this classic scheme is likely to be modified or supplanted by a scheme reliant on molecular subgroups, as suggested in a recent consensus paper by Ramaswamy et al.[ 31 ]

Chemotherapy

Cytotoxic chemotherapy may be used in the initial treatment, maintenance therapy, or for recurrent disease. It may be radiation sparing, which would allow one to eschew the use of radiation in children <3 years of age and permit dose-reduction in older patients. Various regimens exist for initial treatment, with standard therapy being post-radiation cisplatin-based chemotherapy for 4–9 cycles.[ 42 ]

Many previous trials investigating chemotherapy and radiation regimens have compared outcomes based on the histologic type. However, the histologic types and molecular subgroups have not been completely congruent. Following the new stratification of MB by molecular subgroup, inroads have been made to tailor therapies to these pathways or predict response to traditional therapy.[ 47 ] In cases where tumor characteristics or metastases preclude adequate resection, biopsy followed by targeted chemotherapy may serve as a favorable alternative.[ 46 ]

Specifically targeted chemotherapies based on pathways believed to be involved in oncogenesis are a promising future application of molecular subgrouping. At present, molecularly targeted agents for each of the four molecular subgroups are being studied in either clinical and pre-clinical models—the most well-studied of these being smoothened (SMO) receptor antagonists, such as vismodigib, that have demonstrated some utility in both preclinical and clinical models.[ 9 ]

Alternative strategies include sensitization of MB tumor cells to chemotherapeutic treatment. For example, thiostrepton, an antagonist of FOXM1 (an oncogene shown to be upregulated in a variety of malignancies), was shown to sensitize MB cells to cisplatin in vitro.[ 22 ]

Options for recurrent disease are more limited. A regimen of ifosfamide, cisplatin, and etoposide has been investigated but may be limited by significant attendant toxicities, most notable for profound myelosuppression.[ 18 ] An alternative regimen combines bevacizumab and irinotecan with or without temozolomide, with an objective response rate of 55% at 6 months and may be better tolerated.[ 1 ] Further studies are required to identify an efficacious regimen with tolerable toxicity for recurrent medulloblastoma, perhaps targeted to molecular subgroup.

Prognosis and outcomes

The overall prognosis of MB is relatively good compared to other high-grade tumors, with a 5-year overall survival of approximately 70%.[ 38 ] Prognostic factors include age at diagnosis, post-resection residual, histologic type, presence of metastasis, and molecular subgroup. Positive prognostic markers include DNMB and MBEN histologic types, WNT subgroup tumors, and expression of beta-catenin in the nucleus[ 11 13 ] and TrkB.[ 16 35 ]

Histological phenotype has classically been very important in prognostication, especially in young children in whom it is highly predictive of outcome. Favorable results can be seen in DNMBs, which account for half of MB cases seen in children ≤3 years old.[ 46 ] In another study, DNMB and MBEN accounted for more than half of the cases in patients <3 years old and had a 5-year overall survival of ~53% compared to ~17% for the classic variant of MB.[ 23 ] DNMB and MBEN are also unique in that complete remission is a realistic and realizable goal with resection and postoperative chemotherapy.[ 14 15 34 36 ] In patients >3 years old, MB histology still retains prognostic significance, with significant differences in outcome for DNMB versus classic versus large cell/anaplastic types.

The WNT subgroup has a very favorable prognosis, with only a 10% probability of metastasis at diagnosis and a 5-year overall survival of 95–100%. SHH MBs carry a slightly greater risk of metastasis than WNT MBs but less than the Groups 3 and 4 subgroups. Prognosis of SHH tumors is inversely correlated with age, with 10-year overall survival of 77%, 51%, and 34% in infants, children, and adults, respectively.[ 32 ] The presence of TP53 mutations in SHH tumors, unlike its presence in the WNT subgroup MBs, carries an additional poor prognostic risk.

Non-WNT/SHH (Groups 3 and 4) MBs are more likely to be metastatic than WNT and SHH subgroups, with approximately 30% of the patients having metastasis at diagnosis. Group 4 tumors carry an intermediate prognosis with 5-year progression-free survival of 95%, with negative risk modifiers including MYCN amplification and presence of metastasis.[ 19 32 ] A categorically poor prognosis is carried by Group 3 MBs, with a 10-year overall survival of 39% and 50% in infants and children, respectively; these tumors are associated with large cell/anaplastic histology and MYC amplification.[ 29 ] These and other distinguishing features are summarized in Table 1 .


Table 1

Summary of characteristics and prognosis of the four recognized subgroups of medulloblastoma

 

A meta-analysis examining event-free survival/disease-free survival favored the inclusion of chemotherapy in treatment of pediatric MB when omitting, but not when including, disease progression as an event.[ 24 ] The authors could not discount a benefit for chemotherapy in the treatment of MB, but also could not make firm positive recommendations for the same. The decision regarding whether or not to include chemotherapy in the treatment of a specific patient and what intensity to use should be individualized based on patient risk factors and tumor characteristics (i.e., molecular subgroup, histology, presence of metastasis).

Posterior fossa syndrome

The posterior fossa syndrome (PFS), also known as cerebellar mutism syndrome, is a complication that occurs in 8–24% of infratentorial brain tumor resections.[ 12 ] PFS usually presents between 1 and 2 days postoperatively and is characterized as a triad of cerebellar mutism, ataxia or axial hypotonia, and affective symptoms such as irritability and emotional lability. These children are commonly inconsolable, apathetic, and/or hypokinetic. While the pathophysiology is poorly understood, there are some purported mechanisms, including disruption of the dentate-thalamo-cortical (DTC) pathway, which has been identified on diffusion tensor imaging of patients affected with PFS.[ 5 ] Specifically, Avula et al. have shown, across several imaging analyses, possible involvement of the proximal efferent cerebellar pathway (pECP), which interconnects the dentate nucleus, superior cerebellar peduncle, and midbrain tegmentum.[ 4 ] The DTC and pECP constitute a sufficiently large area that may account for the anatomic heterogeneity of lesions associated with PFS.

While some studies have suggested vermian injury as the cause of PFS,[ 30 ] equivocal data exist for this hypothesis.[ 45 ] While potentially associated with a variety of posterior fossa tumors and even hemorrhagic AVMs,[ 6 ] MB appears to be associated with the highest risk for development of PFS. While some recent progress has been made regarding the anatomical pathways associated with PFS, a wide variety of partially supported hypotheses still exist regarding the pathophysiology, including postoperative vasospasm, axonal injury, neuronal dysfunction, thermal injury, and postoperative edema.[ 5 ]

CONCLUSION

MB is one of the most well-studied and frequently encountered CNS malignancies with a relatively good response to current treatments. However, there remains a subset of patients with poor outcomes despite numerous studies trying to optimize chemotherapy and radiation regimens. The current understanding of MB biology has vastly outpaced breakthroughs in treatment over the past 5–10 years, and application of this knowledge holds promise for continued improvements in outcomes for the future.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Aguilera D, Mazewski C, Fangusaro J, MacDonald TJ, McNall-Knapp RY, Hayes LL. Response to bevacizumab, irinotecan, and temozolomide in children with relapsed medulloblastoma: A multi-institutional experience. Childs Nerv Syst. 2013. 29: 589-96

2. Albright AL, Pollack IF, Adelson PD.editorsPrinciples and practice of pediatric neurosurgery. New York: Thieme; 2014. p.

3. Albright AL, Wisoff JH, Zeltzer PM, Boyett JM, Rorke LB, Stanley P. Effects of medulloblastoma resections on outcome in children: A report from the Children's Cancer Group. Neurosurgery. 1996. 38: 265-71

4. Avula S, Kumar R, Pizer B, Pettorini B, Abernethy L, Garlick D. Diffusion abnormalities on intraoperative magnetic resonance imaging as an early predictor for the risk of posterior fossa syndrome. Neuro Oncol. 2015. 17: 614-22

5. Avula S, Mallucci C, Kumar R, Pizer B. Posterior fossa syndrome following brain tumour resection: Review of pathophysiology and a new hypothesis on its pathogenesis. Childs Nerv Syst. 2015. 31: 1859-67

6. Baillieux H, Weyns F, Paquier P, De Deyn PP, Marien P. Posterior fossa syndrome after a vermian stroke: A new case and review of the literature. Pediatr Neurosurg. 2007. 43: 386-95

7. Brechbiel J, Miller-Moslin K, Adjei AA. Crosstalk between hedgehog and other signaling pathways as a basis for combination therapies in cancer. Cancer Treat Rev. 2014. 40: 750-9

8. Brugieres L, Pierron G, Chompret A, Paillerets BB, Di Rocco F, Varlet P. Incomplete penetrance of the predisposition to medulloblastoma associated with germ-line SUFU mutations. J Med Genet. 2010. 47: 142-4

9. Coluccia D, Figuereido C, Isik S, Smith C, Rutka JT. Medulloblastoma: Tumor Biology and Relevance to Treatment and Prognosis Paradigm. Curr Neurol Neurosci Rep. 2016. 16: 43-

10. DeSouza RM, Jones BR, Lowis SP, Kurian KM. Pediatric medulloblastoma-Update on molecular classification driving targeted therapies. Front Oncol. 2014. 4: 176-

11. Ellison DW, Onilude OE, Lindsey JC, Lusher ME, Weston CL, Taylor RE. beta-Catenin status predicts a favorable outcome in childhood medulloblastoma: The United Kingdom Children's Cancer Study Group Brain Tumour Committee. J Clin Oncol. 2005. 23: 7951-7

12. Gadgil N, Hansen D, Barry J, Chang R, Lam S. Posterior fossa syndrome in children following tumor resection: Knowledge update. Surg Neurol Int. 2016. 7: S179-83

13. Gajjar A, Chintagumpala M, Ashley D, Kellie S, Kun LE, Merchant TE. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): Long-term results from a prospective, multicentre trial. Lancet Oncol. 2006. 7: 813-20

14. Giangaspero F, Perilongo G, Fondelli MP, Brisigotti M, Carollo C, Burnelli R. Medulloblastoma with extensive nodularity: A variant with favorable prognosis. J Neurosurg. 1999. 91: 971-7

15. Grill J, Sainte-Rose C, Jouvet A, Gentet JC, Lejars O, Frappaz D. Treatment of medulloblastoma with postoperative chemotherapy alone: An SFOP prospective trial in young children. Lancet Oncol. 2005. 6: 573-80

16. Grotzer MA, Janss AJ, Fung K, Biegel JA, Sutton LN, Rorke LB. TrkC expression predicts good clinical outcome in primitive neuroectodermal brain tumors. J Clin Oncol. 2000. 18: 1027-35

17. Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell SM. The molecular basis of Turcot's syndrome. N Engl J Med. 1995. 332: 839-47

18. Kanamori M, Kumabe T, Saito R, Yamashita Y, Sonoda Y, Tominaga T. The safety of combination chemotherapy with ifosfamide, cisplatin, and etoposide (ICE): Single-institution retrospective review of 108 cases. No Shinkei Geka. 2010. 38: 997-1005

19. Kool M, Korshunov A, Remke M, Jones DT, Schlanstein M, Northcott PA. Molecular subgroups of medulloblastoma: An international meta-analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, Group 3, and Group 4 medulloblastomas. Acta Neuropathol. 2012. 123: 473-84

20. Lam S, Reddy GD, Lin Y, Jea A. Management of hydrocephalus in children with posterior fossa tumors. Surg Neurol Int. 2015. 6: S346-8

21. Lin CY, Erkek S, Tong Y, Yin L, Federation AJ, Zapatka M. Active medulloblastoma enhancers reveal subgroup-specific cellular origins. Nature. 2016. 530: 57-62

22. Lin J, Zheng Y, Chen K, Huang Z, Wu X, Zhang N. Inhibition of FOXM1 by thiostrepton sensitizes medulloblastoma to the effects of chemotherapy. Oncol Rep. 2013. 30: 1739-44

23. McManamy CS, Pears J, Weston CL, Hanzely Z, Ironside JW, Taylor RE. Nodule formation and desmoplasia in medulloblastomas-defining the nodular/desmoplastic variant and its biological behavior. Brain Pathol. 2007. 17: 151-64

24. Michiels EM, Schouten-Van Meeteren AY, Doz F, Janssens GO, van Dalen EC. Chemotherapy for children with medulloblastoma. Cochrane Database Syst Rev. 2015. 1: CD006678-

25. Morrissy AS, Garzia L, Shih DJ, Zuyderduyn S, Huang X, Skowron P. Divergent clonal selection dominates medulloblastoma at recurrence. Nature. 2016. 529: 351-7

26. Northcott PA, Hielscher T, Dubuc A, Mack S, Shih D, Remke M. Pediatric and adult sonic hedgehog medulloblastomas are clinically and molecularly distinct. Acta Neuropathol. 2011. 122: 231-40

27. Northcott PA, Nakahara Y, Wu X, Feuk L, Ellison DW, Croul S. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet. 2009. 41: 465-72

28. Pastorino L, Ghiorzo P, Nasti S, Battistuzzi L, Cusano R, Marzocchi C. Identification of a SUFU germline mutation in a family with Gorlin syndrome. Am J Med Genet A. 2009. 149A: 1539-43

29. Pfister S, Remke M, Benner A, Mendrzyk F, Toedt G, Felsberg J. Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci. J Clin Oncol. 2009. 27: 1627-36

30. Puget S, Boddaert N, Viguier D, Kieffer V, Bulteau C, Garnett M. Injuries to inferior vermis and dentate nuclei predict poor neurological and neuropsychological outcome in children with malignant posterior fossa tumors. Cancer. 2009. 115: 1338-47

31. Ramaswamy V, Remke M, Bouffet E, Bailey S, Clifford SC, Doz F. Risk stratification of childhood medulloblastoma in the molecular era: The current consensus. Acta Neuropathol. 2016. 131: 821-31

32. Ramaswamy V, Remke M, Bouffet E, Faria CC, Perreault S, Cho YJ. Recurrence patterns across medulloblastoma subgroups: An integrated clinical and molecular analysis. Lancet Oncol. 2013. 14: 1200-7

33. Riva-Cambrin J, Detsky AS, Lamberti-Pasculli M, Sargent MA, Armstrong D, Moineddin R. Predicting postresection hydrocephalus in pediatric patients with posterior fossa tumors. J Neurosurg Pediatr. 2009. 3: 378-85

34. Rutkowski S, Bode U, Deinlein F, Ottensmeier H, Warmuth-Metz M, Soerensen N. Treatment of early childhood medulloblastoma by postoperative chemotherapy alone. N Engl J Med. 2005. 352: 978-86

35. Rutkowski S, von Bueren A, von Hoff K, Hartmann W, Shalaby T, Deinlein F. Prognostic relevance of clinical and biological risk factors in childhood medulloblastoma: Results of patients treated in the prospective multicenter trial HIT’91. Clin Cancer Res. 2007. 13: 2651-7

36. Saito R, Kumabe T, Sonoda Y, Kanamori M, Yamashita Y, Watanabe M. Combination chemotherapy with ifosfamide, cisplatin, and etoposide for medulloblastoma: Single-institute experience and differences in efficacy for subgroups of medulloblastoma. Childs Nerv Syst. 2011. 27: 1399-406

37. Slade I, Murray A, Hanks S, Kumar A, Walker L, Hargrave D. Heterogeneity of familial medulloblastoma and contribution of germline PTCH1 and SUFU mutations to sporadic medulloblastoma. Fam Cancer. 2011. 10: 337-42

38. Smoll NR. Relative survival of childhood and adult medulloblastomas and primitive neuroectodermal tumors (PNETs). Cancer. 2012. 118: 1313-22

39. Taylor MD, Liu L, Raffel C, Hui CC, Mainprize TG, Zhang X. Mutations in SUFU predispose to medulloblastoma. Nat Genet. 2002. 31: 306-10

40. Taylor MD, Mainprize TG, Rutka JT. Molecular insight into medulloblastoma and central nervous system primitive neuroectodermal tumor biology from hereditary syndromes: A review. Neurosurgery. 2000. 47: 888-901

41. Taylor MD, Northcott PA, Korshunov A, Remke M, Cho YJ, Clifford SC. Molecular subgroups of medulloblastoma: The current consensus. Acta Neuropathol. 2012. 123: 465-72

42. Taylor RE, Bailey CC, Robinson K, Weston CL, Ellison D, Ironside J. Results of a randomized study of preradiation chemotherapy versus radiotherapy alone for nonmetastatic medulloblastoma: The International Society of Paediatric Oncology/United Kingdom Children's Cancer Study Group PNET-3 Study. J Clin Oncol. 2003. 21: 1581-91

43. Thompson EM, Hielscher T, Bouffet E, Remke M, Luu B, Gururangan S. Prognostic value of medulloblastoma extent of resection after accounting for molecular subgroup: A retrospective integrated clinical and molecular analysis. Lancet Oncol. 2016. 17: 484-95

44. Wang X, Dubuc AM, Ramaswamy V, Mack S, Gendoo DM, Remke M. Medulloblastoma subgroups remain stable across primary and metastatic compartments. Acta Neuropathol. 2015. 129: 449-57

45. Wells EM, Khademian ZP, Walsh KS, Vezina G, Sposto R, Keating RF. Postoperative cerebellar mutism syndrome following treatment of medulloblastoma: Neuroradiographic features and origin. J Neurosurg Pediatr. 2010. 5: 329-34

46. Wong TT, Liu YL, Ho DM, Chang KP, Liang ML, Chen HH. Factors affecting survival of medulloblastoma in children: The changing concept of management. Childs Nerv Syst. 2015. 31: 1687-98

47. Zhang ZY, Xu J, Ren Y, Li KK, Ng HK, Mao Y. Medulloblastoma in China: Clinicopathologic analyses of SHH, WNT, and non-SHH/WNT molecular subgroups reveal different therapeutic responses to adjuvant chemotherapy. PLoS One. 2014. 9: e99490-

48. Zurawel RH, Chiappa SA, Allen C, Raffel C. Sporadic medulloblastomas contain oncogenic beta-catenin mutations. Cancer Res. 1998. 58: 896-9

Pediatric cerebral cavernous malformations: Genetics, pathogenesis, and management

$
0
0

Michael G. Z. Ghali, Visish M. Srinivasan, Arvind C. Mohan, Jeremy Y. Jones, Peter T. Kan, Sandi Lam
  1. Department of Neurobiology, Drexel University College of Medicine, Philadelphia, USA
  2. Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
  3. Department of Radiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA

Correspondence Address:
Sandi Lam
Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA

DOI:10.4103/2152-7806.196921

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Michael G. Z. Ghali, Visish M. Srinivasan, Arvind C. Mohan, Jeremy Y. Jones, Peter T. Kan, Sandi Lam. Pediatric cerebral cavernous malformations: Genetics, pathogenesis, and management. 28-Dec-2016;7:

How to cite this URL: Michael G. Z. Ghali, Visish M. Srinivasan, Arvind C. Mohan, Jeremy Y. Jones, Peter T. Kan, Sandi Lam. Pediatric cerebral cavernous malformations: Genetics, pathogenesis, and management. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/pediatric-cerebral-cavernous-malformations-genetics-pathogenesis-and-management/

Date of Submission
06-Apr-2016

Date of Acceptance
14-Jun-2016

Date of Web Publication
28-Dec-2016

Keywords: Cerebral cavernous malformation, cavernous angioma, cavernous hemangioma, vascular lesion, neurovascular, intracranial

CASE REPORTS

Case 1

A 2-year-old Hispanic girl presented with a 3-day history of vomiting followed by a sudden onset of right facial droop and right eye ptosis. She had achieved normal developmental milestones until then. In retrospect, she had several months of occasional left leg weakness causing falls. There was no history of seizures. She had a family history with two second-degree relatives with cerebral cavernous malformations (CCMs) of the cerebrum and brainstem. She had not received prior imaging or screening. Genetic testing revealed a heterozygous mutation in the KRIT1 gene.

Computerized tomography (CT) of the head demonstrated multiple hemorrhagic lesions of different ages, with the largest being a 3 × 2.5 cm lesion in the right thalamus with mesencephalic extension, along with 1 cm lesions in the left forceps minor and right atrium. There was obstructive hydrocephalus without intraventricular hemorrhage [ Figure 1a ].


Figure 1

Evolution of hemorrhage from familial cavernous malformation (Case 1). (a) Computerized tomography (CT) of head at presentation, showing 2.5 × 3 cm hemorrhagic CM in right thalamic-mesencephalic junction. (b) Magnetic resonance imaging (MRI) of the brain, axial gradient echo sequence showing the same. (c) Expansion of hemorrhage to 3.5 × 3 × 3 cm. (d) CT head, sagittal section, day 9, showing further extension in the craniocaudal dimension. (e) Preoperative MRI brain, axial gradient echo sequence, showing lesion growing to 3.3 cm anteroposterior × 3.7 cm transverse × 3.7 cm craniocaudal. (f) Postoperative CT head showing complete resection of lesion and hematoma

 

An external ventricular drain (EVD) was placed. Magnetic resonance imaging (MRI) of the brain characterized these lesions as CCMs [ Figure 1b ]. Due to the deep and eloquent location of the hemorrhage, the initial plan was directed toward supportive care and rehabilitation. Over 2 weeks of supportive care, the patient's neurologic status declined, and she required intubation. The lesions exhibited continued episodic hemorrhage with rupture into the ventricular system [Figure 1c and d ]. The hemorrhage extended deeper into the midbrain and pons, causing a complete third nerve palsy, quadriplegia, and obtundation. The hematoma eventually presented to the surface of the tectal plate, allowing a direct surgical corridor for access [ Figure 1e ]. Evacuation of the hematoma and right thalamic-mesencephalic CCM was performed via a midline supracerebellar–infratentorial approach [ Figure 1f ]. The patient recovered some right-sided motor function. Ventilator support and EVD were both weaned successfully. She continues with postoperative rehabilitation.

Case 2

A 7-year-old girl presented with diplopia, ataxia, and headaches. Imaging revealed a hemorrhagic right middle cerebellar peduncle lesion suggestive of a solitary CCM [ Figure 2a ]. Her presenting deficits resolved within a week, and she received a brief course of physical therapy with serial follow-up. In the following month, the patient had acute headache and ataxia. Repeat imaging showed extension of the hemorrhagic lesion past the ependymal surface into the lateral fourth ventricle without obstructive hydrocephalus [Figures 2b and c ]. Surgery was timed 4 weeks following the acute hemorrhage to allow for rehabilitation and evolution of the hematoma cavity. She underwent suboccipital craniotomy with complete resection of the CM [ Figure 2d ]. The patient had excellent postoperative recovery without deficits; she was more active and energetic than the year prior to surgery.


Figure 2

Evolution of hemorrhage in brainstem cavernous malformation (Case 2). (a) Computerized tomography (CT) of the head at presentation showing hemorrhage in right middle cerebellar peduncle that does not extend to the pial surface. (b) CT head 1 month later during recurrence of symptoms shows worsened perilesional edema and progression toward the ependymal surface of the fourth ventricle. (c) Magnetic resonance imaging (MRI) of the brain T1-weighted sequence with contrast at the time of second hemorrhage demonstrates surgical corridor made possible by hemorrhage extending to the right lateral recess of fourth ventricle. (d) Postoperative MRI demonstrates complete resection of lesion

 

REVIEW

Epidemiology

The epidemiology of CCMs, or cavernomas, is an area of extensive investigation in adults. However, less data exist regarding their incidence and prevalence in the pediatric age group. Moreover, a prior false assumption of an exclusively congenital etiology may have led to an underestimation of the overall hemorrhage risk. The development of CCMs appears to increase with age, reaching a plateau in late adolescence, as demonstrated by Al-Holou et al. in 2012.[ 3 ] They reported a prevalence of ~0.2% in infants and an overall prevalence of ~0.6% in children. Males and females are equally affected in the pediatric and adult populations.[ 3 34 35 ] According to studies by Gross et al. in 2011 and 2015, among children with CCMs, 10% of cases are familial and approximately 17% have multiple lesions.[ 34 35 ] Also evident is an apparent racial disparity in etiology, with the familial form accounting for half of the cases in Hispanic patients and only 10–20% of affected Caucasians.[ 22 63 ]

The overall incidence for the development of new CCMs in children is correlated with their pre-existing cavernoma burden. Gross et al. reported incidences of approximately 1.2% per patient per year and 2.5% per lesion per year; that is, the presence of multiple CMs confers a greater risk of de novo cavernoma-genesis compared with patients with solitary CMs (7.1% versus 0.6% per lesion per year).[ 34 ] Prior radiation also appears to confer a risk of de novo CCM formation, representing 9% of pediatric CCMs. Patients harboring CCMs who subsequently received radiation developed further CCMs at a rate of 2.6% per lesion per year,[ 34 ] usually with a latency period of 9 years until detection.[ 55 ]

Histopathology

Cavernous malformations (CMs) are hamartomatous, cystically-dilated vascular spaces composed of a single layer of endothelium with possible infrequent subendothelial cells, without elastic lamina or smooth muscle cells, embedded in a collagenous extracellular matrix.[ 9 17 64 ] The endothelial cells characteristically lack tight junctions. The rudimentary vessels coalesce to form a compact mulberry-like mass devoid of intervening neural parenchyma, distinguishing them from capillary telangiectasias and arteriovenous malformations (AVMs).[ 35 64 ] On gross pathology, they are well-demarcated vascular masses measuring up to several centimeters. CMs may exhibit reactive astrogliosis, calcification, and hemosiderin deposition, with the latter being a consequence of recurrent hemorrhages.

Clinical presentation and diagnosis

Approximately 85% of CMs are supratentorial in children (92% lobar, 8% deep). The remainder are located infratentorially (57% brainstem, 43% cerebellar), with rare occurrence in the spinal cord.[ 34 ] CCMs may remain asymptomatic for the lifetime of the patient in both children and adults. In the pediatric population, clinical presentation of CCM includes hemorrhage (62%), seizures (35%), and incidental radiographic finding (26%). A bleeding episode is followed by thrombosis of the lesion and subsequent recanalization, predisposing to recurrent hemorrhage.[ 72 80 ] Presentation by hemorrhage is more common among brainstem CMs (80%), which also have higher annual re-hemorrhaging rates (17% versus 11% for CMs overall).[ 34 48 ] Patients may or may not have neurological sequelae.

By definition, all CCMs exhibit varying degrees of microhemorrhage, as evidenced by hemosiderin deposition. When located in noneloquent areas of the brain, slightly larger degrees of hemorrhage may be tolerated. Concern arises when CCMs occur in critical brain regions, such as the brainstem, where even small hemorrhages may compromise vital functions.

The overall risk for CCM hemorrhage in the pediatric population is approximately 0.5% per lesion per year.[ 3 4 29 34 35 43 ] A history of prior hemorrhage is perhaps the most important risk factor, shown in one study to confer an overall annual hemorrhage risk of 11.3%.[ 4 6 9 29 34 35 43 ] Recurrent hemorrhages have a tendency to “cluster,” usually seen within 2–3 years of the index hemorrhage event.[ 6 34 ]

Imaging

CT scanning has poor sensitivity for detection of CCMs. T2-weighted MRI, especially gradient echo (GRE) or susceptibility weighted imaging (SWI) sequences, possesses the greatest sensitivity for detection of CCMs and reveals a mixed signal core and surrounding low signal rim, often with evidence of microhemorrhages. Brain MRI screening is indicated for first-degree relatives of patients with CMs with two or more affected family members.

Developmental venous anomalies (DVAs) are seen in up to 20% of patients with CMs,[ 35 54 59 ] and have been suggested to confer an increased risk of CCM hemorrhage risk in past studies.[ 61 ]

Genetics and etiopathogenesis

CCMs may occur sporadically or be transmitted in an autosomal dominant fashion with variable penetrance; the familial type has been reported to account for roughly half of the cases in Hispanics and up to one-fifth of the cases in Caucasians, and is also associated with a greater annual risk of symptomatic bleeding.[ 10 16 37 40 63 80 ] It has been shown that many cases thought to be sporadic do, in fact, harbor familial mutations.[ 23 47 ] Multiple lesions are present in 84% of the familial cases and up to one-third of the sporadic cases.

Three genetic loci have been associated with and account for ~80% of familial CCM: CCM1/Krit1, CCM2/MGC4607, and CCM3/PDCD10.[ 19 25 51 ] CCM1 was definitively localized and mapped to chromosome 7q21-q22,[ 66 67 ] CCM2 to 7p15-p13,[ 19 ] and CCM3 to 3q25.25-27.[ 19 ] Multilocus analysis reveals 40%, 20%, and 40% of familial CCM are linked to CCM1, CCM2, and CCM3, respectively.[ 19 ] Penetrance of CCM1, CCM2, and CCM3 mutations is approximated at 60–88%, 100%, and 63%, respectively.[ 19 21 ] Several different loss-of-function mutations have been identified in CCM1-3, suggesting that their products function as tumor suppressors. A fourth locus has been postulated to be present at 3q26.3-q27.2.[ 8 19 ] Other candidate genes include ZPLDC1, found mutated in a single patient with CCM, and those encoding ephrin-B2[ 74 ] and Notch[ 76 ] proteins.

Furthermore, single nucleotide polymorphisms (SNPs) of inflammatory and immune response genes have been associated with different features of CCM natural history, including disease burden and severity of risk of intracerebral hemorrhage; identified SNPs include IL-1RN, TGFBR2, CHUK, SELS, CD3G, IGH, and IGL. This information may have implications for risk stratification and treatment planning.

A Knudsonian two-hit hypothesis has been presented to account for all cases of CCM. This appears to hold true for the familial form, wherein an inherited germline CCM mutation and a single acquired somatic mutation in the homologue affects cavernoma-genesis,[ 42 ] which has been demonstrated for CCM1-3.[ 2 30 31 ] In the sporadic form of the disease, the Knudsonian two-hit hypothesis posits the acquisition of two somatic mutations in homologous genes, which has never been directly shown, and thus remains to be explained. One theory is that, in the setting of vascular insult, such as trauma or radiation shown to predispose to cavernoma-genesis,[ 34 55 ] context-dependent haploinsufficiency of a CCM gene may occur. In this model, a single functional copy of a CCM gene in the normal uninjured state would be sufficient to prevent cavernoma genesis by maintaining vascular integrity and blunting unmitigated cellular proliferation via actions of CCM protein products (see Molecular Pathogenesis). Injury may create a greater requirement for larger amounts of this protein product to keep rho-associated protein kinase (ROCK) signaling and other pathways in check when a single functional copy of a CCM gene becomes inadequate (haploinsufficiency).

Molecular pathogenesis

The molecular pathogenesis of CCMs is linked to the gene products of CCM1, CCM2, and CCM3, which are also known as Krev Interaction Trapped 1 (Krit1), malcavernin, and PDCD10, respectively. Table 1 summarizes the effects of these mutations.


Table 1

Summary of pathogenesis of mutations in the cerebral cavernous malformation (CCM) genes associated with familial cavernous malformations (CM)

 

CCM1 encodes Krit1, a microtubule-associated protein that also interacts with Rap1A, ICAP-1, and a variety of other proteins. Rap1A is a Ras-family GTPase involved in cellular differentiation and morphogenesis, as well as regulation of cellular polarity and cytoskeletal organization.[ 58 65 ] It is expressed in endothelial and vascular smooth muscle cells.[ 77 ] Among the proposed functions of Krit1 is localization of Rap1A to specific subcellular compartments. An abnormality of this nature in vascular smooth muscle cells may result in perturbation of endothelial cell organization[ 38 67 ] or affect the stability of endothelial adherens junctions.[ 26 81 83 84 ]

In addition, Krit1 may contribute to regulation of transmembrane β1-integrin-mediated signal transduction and cell-cell as well as cell-extracellular matrix (ECM) signaling, both of which are critical in the formation of microtubules and, in turn, endothelial structure/function and angiogenesis. An important intermediary protein in β1-integrin-mediated signal transduction is ICAP-1, which binds the cytoplasmic domain and links it to the cytoskeleton. Further, ICAP possesses binding sites for Krit1, which may play a regulatory function in the β1-integrin/ICAP-1 interaction.[ 81 83 ] CCM1 mutations prevent Krit1/ICAP-1 association and, thus, may effect cavernoma-genesis consequent to defective transmembrane protein-mediated signaling involved in the angiogenesis and organization of vascular endothelium.[ 83 ]

Krit1 also interacts with malcavernin, a scaffold protein that associates with mitogen-activated protein kinase (MAPK)-extracellular-regulated kinase (ERK) kinase 3 (MEKK3), a pathway critical in the regulation of endothelial proliferation and migration, adhesion, and cytoskeletal regulation.[ 52 81 82 ] The malcavernin/MEKK3 complex promotes the nuclear-to-cytoplasmic translocation of Krit1/ICAP-1, which associates to form a ternary complex that regulates p38 MAPK activity;[ 82 ] malcavernin mutations alter the phosphotyrosine binding domains by which Krit1 associates. P38 MAPK promotes endothelial proliferation via promotion of VEGF and COX-2 signaling, while also negatively modulating differentiation and apoptosis via promotion of Bad (an inhibitor of MEK1/2 and ERK1/2) and inhibition of Bcl-x (an inhibitor of the caspase cascade).[ 12 20 ]

Programmed cell death 10 gene (PDCD10) encodes for a protein involved in apoptosis and associates with both Krit1 and malcavernin.[ 8 ] PDCD10 also affects cellular proliferation and growth via interaction with MST4 kinase and the MAPK and ERK pathways.[ 50 ] PDCD10 has been proposed to function in the pruning of newly formed blood vessels. A perturbation in this mechanism may effect cavernoma-genesis.

CCM1-3 share their ability to negatively modulate RhoA/Rho kinase;[ 12 ] loss-of-function mutations result in upregulated ROCK signaling. ROCK increases the synthesis of microtubules and phosphorylates myosin light chains, leading to increased cell contractility, which promotes disruption of endothelial intercellular adhesion, leading to the formation of abnormally dilated leaky vessels.[ 11 33 71 76 79 ] Thus, CCM genes contribute to the maintenance of vascular integrity through negative modulation of ROCK [ Figure 3 ]. This may offer a promising therapeutic target for CCMs;[ 49 ] direct pharmacological inhibition of ROCK using Fasudil has been demonstrated in both in vitro and animal models.[ 71 ] Another target could be RhoA, whose hyperactivation could be inhibited by HMG-CoA reductase inhibitors such as statins.[ 49 ] Human trials appear to be the next step in the future; there are no open clinical trials as of May 2016. An increased risk of hemorrhage in zebrafish is reported, but not in murine or mammalian models.[ 27 ] Short hairpin RNA (shRNA) silencing of ROCK has been used in vitro[ 11 ] and, in theory, may also be targeted to locally treat CCMs.


Figure 3

Summary of main pathways of cavernoma-genesis and potential targeted therapies. The molecular pathogenesis of familial cavernomas centers around modulation of Rho Kinase (ROCK), which modulates microtubule synthesis. This, in turn, alters contractility of endothelial cells, intercellular adhesion, and vascular integrity. Loss of vascular integrity allows cavernoma-genesis

 

Treatment

Standard management options for CCMs have classically included observation and surgical removal.[ 41 ] Asymptomatic lesions are generally treated conservatively. Surgery is indicated for accessible symptomatic lesions. Complete resection eliminates the risk of hemorrhage from that particular lesion but may risk neurological morbidity, especially for CCMs located in eloquent cortex or brainstem.[ 24 75 ] Assessment of optimal surgical timing and approach is important to minimize treatment morbidity.

Especially for deep-seated CCMs in the pons or brainstem, surgeons typically prefer to wait for the CCM lesion to present to a surgically accessible surface without the need for direct surgical dissection through eloquent tracts.[ 1 ] In addition, the timing of microsurgery is optimal several weeks after hemorrhage to allow perilesional edema to subside, as well as hematoma contents to evolve and soften.[ 15 62 ]

Surgical resection of CMs in the setting of epilepsy is a topic of continuing investigation. In this scenario, the hemosiderin ring associated with CCMs is associated with epileptogenic potential[ 44 45 ] and is generally recommended to be resected along with the CCM lesion.[ 7 36 46 70 ] High rates of seizure control (73–85%) can be attained with complete resection of the hemosiderin ring.[ 14 28 69 ] Use of intraoperative electrocorticography (ECoG) has also been correlated with more favorable seizure outcomes, increasing seizure freedom from 77% without ECoG to 90% with ECOG-directed resection at 6 months postoperatively,[ 73 ] though in other studies the benefit has not reached statistical significance.[ 28 ] Englot et al. have postulated that this may be due to a selection bias of ECoG use in patients with more severe epilepsy or lesions in eloquent regions.[ 13 28 ] However, safety of full resection of hemosiderin-stained tissue may be limited in eloquent areas such as those subserving motor or language function. Hemosiderin resection is typically not performed in brainstem CCMs.[ 24 ]

Evolution of minimally invasive therapies

Stereotactic radiosurgery (SRS) has been reported by some groups as a potential treatment option.[ 5 60 85 ] However, any reported decrease in the risk of hemorrhage appears to occur at 2 years after SRS; thus, it is difficult to discern whether this reflects true therapeutic benefit versus the natural history of decreased hemorrhage rates after initial “clustering” of hemorrhage events.[ 5 39 57 60 ] Because arteriopathy is not an etiology of CCM, some experts contend that the use of SRS for CCMs does not have an explanation grounded in biological basis.[ 13 ]

Magnetic resonance-guided focused ultrasound (MRgFUS) has been reported as a novel minimally invasive procedure for the treatment of central nervous system pathologies, including CCMs.[ 68 ] This treatment focuses multiple intersecting ultrasonic waves to overcome the attenuation of wave energy by the skull and prevents overheating of nontargeted foci in a manner analogous to SRS.[ 32 ] Known mechanisms of therapeutic efficacy include local thermal rise, acoustic cavitation, and immunomodulation.[ 18 ] The treatment is guided by magnetic resonance thermal imaging, which confirms correct targeting and sufficient heating to effect ablation. In the treatment of vascular malformations, such as CCMs, MRgFUS may theoretically be used to activate the release of anti-angiogenic or endothelial-targeting nanoparticles carried on microbubbles. Limitations include difficulty in focusing ultrasonic waves in paraconvexity regions and long procedure duration due to the need for MRI acquisition or large lesions requiring multiple sonications. It must be stressed that, to date, this technology has had neither widespread application nor long-term follow-up.

Another minimally invasive treatment option for CCMs may include MR-guided laser interstitial thermal therapy (MRgLITT), also called MR-guided stereotactic laser ablation (MRgSLA). MRgSLA has been used in the treatment of tumors, epilepsy, and chronic pain syndromes. A non-negligible risk of focal neurologic deficits has been reported, especially for deep targets.[ 56 ] MRgSLA has been used successfully at Texas Children's Hospital to treat hypothalamic hamartomas, with high rates of control of gelastic seizures and a low complication rate.[ 78 ] Application of MRgSLA for ablation of CCMs has been demonstrated in a series of 5 adults with medically intractable epilepsy associated with CCMs with abolition of disabling seizures in 4 patients at 12 to 28 months’ postintervention.[ 53 ] CCM ablation was confirmed immediately following the procedure, and follow-up imaging (6–21 months) revealed perilesional necrotic encephalomalacia, consistent with the intent to effect extended lesionectomy. Because there are no long-term treatment outcomes and experience is limited, thus far, critical evaluation and further study are needed.

CONCLUSION

CCMs are a common cause of intracranial hemorrhage in the pediatric population. A significant proportion of CCMs identified in pediatric patients, especially those with a history of symptomatic hemorrhage, may be associated with a familial subtype with identifiable genetic mutations in genes CCM1, CCM2, or CCM3. Future research will further identify genetic pathophysiology, risk of rupture, and risk of CCM formation based on genotyping. Surgery remains the gold standard of treatment. Directions for future evaluation include minimally invasive procedures, as well as potential for an increased role of medical management using targeted molecular therapies.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Abla AA, Turner JD, Mitha AP, Lekovic G, Spetzler RF. Surgical approaches to brainstem cavernous malformations. Neurosurg Focus. 2010. 29: E8-

2. Akers AL, Johnson E, Steinberg GK, Zabramski JM, Marchuk DA. Biallelic somatic and germline mutations in cerebral cavernous malformations (CCMs): Evidence for a two-hit mechanism of CCM pathogenesis. Hum Mol Genet. 2009. 18: 919-30

3. Al-Holou WN, O’Lynnger TM, Pandey AS, Gemmete JJ, Thompson BG, Muraszko KM. Natural history and imaging prevalence of cavernous malformations in children and young adults. J Neurosurg Pediatr. 2012. 9: 198-205

4. Al-Shahi Salman R, Hall JM, Horne MA, Moultrie F, Josephson CB, Bhattacharya JJ. Untreated clinical course of cerebral cavernous malformations: A prospective, population-based cohort study. Lancet Neurol. 2012. 11: 217-24

5. Amin-Hanjani S, Ogilvy CS, Candia GJ, Lyons S, Chapman PH. Stereotactic radiosurgery for cavernous malformations: Kjellberg's experience with proton beam therapy in 98 cases at the Harvard Cyclotron. Neurosurgery. 1998. 42: 1229-36

6. Barker FG, Amin-Hanjani S, Butler WE, Lyons S, Ojemann RG, Chapman PH. Temporal clustering of hemorrhages from untreated cavernous malformations of the central nervous system. Neurosurgery. 2001. 49: 15-24

7. Baumann CR, Schuknecht B, Lo Russo G, Cossu M, Citterio A, Andermann F. Seizure outcome after resection of cavernous malformations is better when surrounding hemosiderin-stained brain also is removed. Epilepsia. 2006. 47: 563-6

8. Bergametti F, Denier C, Labauge P, Arnoult M, Boetto S, Clanet M. Mutations within the programmed cell death 10 gene cause cerebral cavernous malformations. Am J Hum Genet. 2005. 76: 42-51

9. Bertalanffy H, Benes L, Miyazawa T, Alberti O, Siegel AM, Sure U. Cerebral cavernomas in the adult. Review of the literature and analysis of 72 surgically treated patients. Neurosurgical Rev. 2002. 25: 1-53

10. Bicknell JM, Carlow TJ, Kornfeld M, Stovring J, Turner P. Familial cavernous angiomas. Arch Neurol. 1978. 35: 746-9

11. Borikova AL, Dibble CF, Sciaky N, Welch CM, Abell AN, Bencharit S. Rho kinase inhibition rescues the endothelial cell cerebral cavernous malformation phenotype. J Biol Chem. 2010. 285: 11760-4

12. Cavalcanti DD, Kalani MY, Martirosyan NL, Eales J, Spetzler RF, Preul MC. Cerebral cavernous malformations: From genes to proteins to disease. J Neurosurg. 2012. 116: 122-32

13. Chang EF, Gabriel RA, Potts MB, Berger MS, Lawton MT. Supratentorial cavernous malformations in eloquent and deep locations: Surgical approaches and outcomes. Clinical article. J Neurosurg. 2011. 114: 814-27

14. Chang EF, Gabriel RA, Potts MB, Garcia PA, Barbaro NM, Lawton MT. Seizure characteristics and control after microsurgical resection of supratentorial cerebral cavernous malformations. Neurosurgery. 2009. 65: 31-7

15. Chen L, Zhao Y, Zhou L, Zhu W, Pan Z, Mao Y. Surgical strategies in treating brainstem cavernous malformations. Neurosurgery. 2011. 68: 609-20

16. Clark JV. Familial occurrence of cavernous angiomata of the brain. J Neurol Neurosurg Psychiatry. 1970. 33: 871-6

17. Clatterbuck RE, Moriarity JL, Elmaci I, Lee RR, Breiter SN, Rigamonti D. Dynamic nature of cavernous malformations: A prospective magnetic resonance imaging study with volumetric analysis. J Neurosurg. 2000. 93: 981-6

18. Cohen-Inbar O, Xu Z, Sheehan JP. Focused ultrasound-aided immunomodulation in glioblastoma multiforme: A therapeutic concept. J Ther Ultrasound. 2016. 4: 2-

19. Craig HD, Gunel M, Cepeda O, Johnson EW, Ptacek L, Steinberg GK. Multilocus linkage identifies two new loci for a mendelian form of stroke, cerebral cavernous malformation, at 7p15-13 and 3q25.2-27. Hum Mol Genet. 1998. 7: 1851-8

20. Dashti SR, Hoffer A, Hu YC, Selman WR. Molecular genetics of familial cerebral cavernous malformations. Neurosurg Focus. 2006. 21: e2-

21. Davenport WJ, Siegel AM, Dichgans J, Drigo P, Mammi I, Pereda P. CCM1 gene mutations in families segregating cerebral cavernous malformations. Neurology. 2001. 56: 540-3

22. Denier C, Goutagny S, Labauge P, Krivosic V, Arnoult M, Cousin A. Mutations within the MGC4607 gene cause cerebral cavernous malformations. Am J Hum Genet. 2004. 74: 326-37

23. Denier C, Labauge P, Bergametti F, Marchelli F, Riant F, Arnoult M. Genotype-phenotype correlations in cerebral cavernous malformations patients. Ann Neurol. 2006. 60: 550-6

24. Duckworth EA. Modern management of brainstem cavernous malformations. Neurol Clin. 2010. 28: 887-98

25. Dupre N, Verlaan DJ, Hand CK, Laurent SB, Turecki G, Davenport WJ. Linkage to the CCM2 locus and genetic heterogeneity in familial cerebral cavernous malformation. Can J Neurol Sci. 2003. 30: 122-8

26. Eerola I, McIntyre B, Vikkula M. Identification of eight novel 5’- exons in cerebral capillary malformation gene-1 (CCM1) encoding KRIT1. Biochim Biophys Acta. 2001. 1517: 464-7

27. Eisa-Beygi S, Wen XY, Macdonald RL. A call for rigorous study of statins in resolution of cerebral cavernous malformation pathology. Stroke. 2014. 45: 1859-61

28. Englot DJ, Han SJ, Lawton MT, Chang EF. Predictors of seizure freedom in the surgical treatment of supratentorial cavernous malformations. J Neurosurg. 2011. 115: 1169-74

29. Flemming KD, Link MJ, Christianson TJ, Brown RD, Jr . Prospective hemorrhage risk of intracerebral cavernous malformations. Neurology. 2012. 78: 632-6

30. Gault J, Awad IA, Recksiek P, Shenkar R, Breeze R, Handler M. Cerebral cavernous malformations: Somatic mutations in vascular endothelial cells. Neurosurgery. 2009. 65: 138-44

31. Gault J, Shenkar R, Recksiek P, Awad IA. Biallelic somatic and germ line CCM1 truncating mutations in a cerebral cavernous malformation lesion. Stroke. 2005. 36: 872-4

32. Ghanouni P, Pauly KB, Elias WJ, Henderson J, Sheehan J, Monteith S. Transcranial MRI-Guided Focused Ultrasound: A Review of the Technologic and Neurologic Applications. AJR Am J Roentgenol. 2015. 205: 150-9

33. Glading AJ, Ginsberg MH. Rap1 and its effector KRIT1/CCM1 regulate beta-catenin signaling. Dis Model Mech. 2010. 3: 73-83

34. Gross BA, Du R, Orbach DB, Scott RM, Smith ER. The natural history of cerebral cavernous malformations in children. J Neurosurg Pediatr. 2015. p. 1-6

35. Gross BA, Lin N, Du R, Day AL. The natural history of intracranial cavernous malformations. Neurosurgical focus. 2011. 30: E24-

36. Hammen T, Romstock J, Dorfler A, Kerling F, Buchfelder M, Stefan H. Prediction of postoperative outcome with special respect to removal of hemosiderin fringe: A study in patients with cavernous haemangiomas associated with symptomatic epilepsy. Seizure. 2007. 16: 248-53

37. Hayman LA, Evans RA, Ferrell RE, Fahr LM, Ostrow P, Riccardi VM. Familial cavernous angiomas: Natural history and genetic study over a 5-year period. Am J Med Genet. 1982. 11: 147-60

38. Henkemeyer M, Rossi DJ, Holmyard DP, Puri MC, Mbamalu G, Harpal K. Vascular system defects and neuronal apoptosis in mice lacking ras GTPase-activating protein. Nature. 1995. 377: 695-701

39. Karlsson B, Kihlstrom L, Lindquist C, Ericson K, Steiner L. Radiosurgery for cavernous malformations. J Neurosurg. 1998. 88: 293-7

40. Kidd HA, Cumings JN. Cerebral angiomata in an Icelandic family. Lancet. 1947. 1: 747-

41. Kivelev J, Niemela M, Hernesniemi J. Treatment strategies in cavernomas of the brain and spine. J Clin Neurosci. 2012. 19: 491-7

42. Knudson AG. Mutation and cancer: Statistical study of retinoblastoma. Proc Natl Acad Sci U S A. 1971. 68: 820-3

43. Kondziolka D, Lunsford LD, Kestle JR. The natural history of cerebral cavernous malformations. J Neurosurg. 1995. 83: 820-4

44. Kotagal P, Luüders H.editorsThe Epilepsies: Etiologies and Prevention. San Diego: Academic Press; 1999. p.

45. Kraemer DL, Awad IA. Vascular malformations and epilepsy: Clinical considerations and basic mechanisms. Epilepsia. 1994. 35: S30-43

46. Kraemer DL, Griebel ML, Lee N, Friedman AH, Radtke RA. Surgical outcome in patients with epilepsy with occult vascular malformations treated with lesionectomy. Epilepsia. 1998. 39: 600-7

47. Labauge P, Laberge S, Brunereau L, Levy C, Tournier-Lasserve E. Hereditary cerebral cavernous angiomas: Clinical and genetic features in 57 French families. Societe Francaise de Neurochirurgie. Lancet. 1998. 352: 1892-7

48. Li D, Hao SY, Tang J, Xiao XR, Jia GJ, Wu Z. Clinical course of untreated pediatric brainstem cavernous malformations: Hemorrhage risk and functional recovery. J Neurosurg Pediatr. 2014. 13: 471-83

49. Li DY, Whitehead KJ. Evaluating strategies for the treatment of cerebral cavernous malformations. Stroke. 2010. 41: S92-4

50. Ma X, Zhao H, Shan J, Long F, Chen Y, Chen Y. PDCD10 interacts with Ste20-related kinase MST4 to promote cell growth and transformation via modulation of the ERK pathway. Mol Biol Cell. 2007. 18: 1965-78

51. Marchuk DA, Gallione CJ, Morrison LA, Clericuzio CL, Hart BL, Kosofsky BE. A locus for cerebral cavernous malformations maps to chromosome 7q in two families. Genomics. 1995. 28: 311-4

52. Matos P, Skaug J, Marques B, Beck S, Verissimo F, Gespach C. Small GTPase Rac1: Structure, localization, and expression of the human gene. Biochem Biophys Res Commun. 2000. 277: 741-51

53. McCracken DJ, Willie JT, Fernald BA, Saindane AM, Drane DL, Barrow DL. Magnetic Resonance Thermometry-Guided Stereotactic Laser Ablation of Cavernous Malformations in Drug-Resistant Epilepsy: Imaging and Clinical Results. Neurosurgery. 2015. p.

54. Moriarity JL, Wetzel M, Clatterbuck RE, Javedan S, Sheppard JM, Hoenig-Rigamonti K. The natural history of cavernous malformations: A prospective study of 68 patients. Neurosurgery. 1999. 44: 1166-71

55. Nimjee SM, Powers CJ, Bulsara KR. Review of the literature on de novo formation of cavernous malformations of the central nervous system after radiation therapy. Neurosurg Focus. 2006. 21: e4-

56. Patel P, Patel NV, Danish SF. Intracranial MR-guided laser-induced thermal therapy: Single-center experience with the Visualase thermal therapy system. J Neurosurg. 2016. p. 1-8

57. Pham M, Gross BA, Bendok BR, Awad IA, Batjer HH. Radiosurgery for angiographically occult vascular malformations. Neurosurg Focus. 2009. 26: E16-

58. Pizon V, Chardin P, Lerosey I, Olofsson B, Tavitian A. Human cDNAs rap1 and rap2 homologous to the Drosophila gene Dras3 encode proteins closely related to ras in the ‘effector’ region. Oncogene. 1988. 3: 201-4

59. Porter PJ, Willinsky RA, Harper W, Wallace MC. Cerebral cavernous malformations: Natural history and prognosis after clinical deterioration with or without hemorrhage. Journal of neurosurgery. 1997. 87: 190-7

60. Porter RW, Detwiler PW, Han PP, Spetzler RF. Stereotactic radiosurgery for cavernous malformations: Kjellberg's experience with proton beam therapy in 98 cases at the Harvard Cyclotron. Neurosurgery. 1999. 44: 424-5

61. Pozzati E, Acciarri N, Tognetti F, Marliani F, Giangaspero F. Growth, subsequent bleeding, and de novo appearance of cerebral cavernous angiomas. Neurosurgery. 1996. 38: 662-9

62. Ramina R, Mattei TA, de Aguiar PH, Meneses MS, Ferraz VR, Aires R. Surgical management of brainstem cavernous malformations. Neurol Sci. 2011. 32: 1013-28

63. Rigamonti D, Hadley MN, Drayer BP, Johnson PC, Hoenig-Rigamonti K, Knight JT. Cerebral cavernous malformations. Incidence and familial occurrence. N Eng J Med. 1988. 319: 343-7

64. Robinson JR, Awad IA, Little JR. Natural history of the cavernous angioma. J Neurosurg. 1991. 75: 709-14

65. Ruggieri R, Bender A, Matsui Y, Powers S, Takai Y, Pringle JR. RSR1, a ras-like gene homologous to Krev-1 (smg21A/rap1A): Role in the development of cell polarity and interactions with the Ras pathway in Saccharomyces cerevisiae. Mol Cell Biol. 1992. 12: 758-66

66. Sahoo T, Johnson EW, Thomas JW, Kuehl PM, Jones TL, Dokken CG. Mutations in the gene encoding KRIT1, a Krev-1/rap1a binding protein, cause cerebral cavernous malformations (CCM1). Hum Mol Genet. 1999. 8: 2325-33

67. Serebriiskii I, Estojak J, Sonoda G, Testa JR, Golemis EA. Association of Krev-1/rap1a with Krit1, a novel ankyrin repeat-containing protein encoded by a gene mapping to 7q21-22. Oncogene. 1997. 15: 1043-9

68. Sheehan J. Transcranial ultrasound for arteriovenous malformations: Something old is new again. World Neurosurg. 2012. 77: 269-70

69. Stavrou I, Baumgartner C, Frischer JM, Trattnig S, Knosp E. Long-term seizure control after resection of supratentorial cavernomas: A retrospective single-center study in 53 patients. Neurosurgery. 2008. 63: 888-96

70. Stefan H, Hammen T. Cavernous haemangiomas, epilepsy and treatment strategies. Acta Neurol Scand. 2004. 110: 393-7

71. Stockton RA, Shenkar R, Awad IA, Ginsberg MH. Cerebral cavernous malformations proteins inhibit Rho kinase to stabilize vascular integrity. J Exp Med. 2010. 207: 881-96

72. Tomlinson FH, Houser OW, Scheithauer BW, Sundt TM, Okazaki H, Parisi JE. Angiographically occult vascular malformations: A correlative study of features on magnetic resonance imaging and histological examination. Neurosurgery. 1994. 34: 792-9

73. Van Gompel JJ, Rubio J, Cascino GD, Worrell GA, Meyer FB. Electrocorticography-guided resection of temporal cavernoma: Is electrocorticography warranted and does it alter the surgical approach?. J Neurosurg. 2009. 110: 1179-85

74. Wang HU, Chen ZF, Anderson DJ. Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell. 1998. 93: 741-53

75. Wang Z, Qian C, Shi L, Wang L, Zhang J, Wang Y. Surgery Approaches to Brainstem Cavernous Malformations. J Craniofac Surg. 2015. 26: e577-80

76. Whitehead KJ, Plummer NW, Adams JA, Marchuk DA, Li DY. Ccm1 is required for arterial morphogenesis: Implications for the etiology of human cavernous malformations. Development. 2004. 131: 1437-48

77. Wienecke R, Maize JC, Reed JA, de Gunzburg J, Yeung RS, DeClue JE. Expression of the TSC2 product tuberin and its target Rap1 in normal human tissues. Am J Pathol. 1997. 150: 43-50

78. Wilfong AA, Curry DJ. Hypothalamic hamartomas: Optimal approach to clinical evaluation and diagnosis. Epilepsia. 2013. 54: 109-14

79. Yadla S, Jabbour PM, Shenkar R, Shi C, Campbell PG, Awad IA. Cerebral cavernous malformations as a disease of vascular permeability: From bench to bedside with caution. Neurosurg Focus. 2010. 29: E4-

80. Zabramski JM, Wascher TM, Spetzler RF, Johnson B, Golfinos J, Drayer BP. The natural history of familial cavernous malformations: Results of an ongoing study. J Neurosurg. 1994. 80: 422-32

81. Zawistowski JS, Serebriiskii IG, Lee MF, Golemis EA, Marchuk DA. KRIT1 association with the integrin-binding protein ICAP-1: A new direction in the elucidation of cerebral cavernous malformations (CCM1) pathogenesis. Hum Mol Genet. 2002. 11: 389-96

82. Zawistowski JS, Stalheim L, Uhlik MT, Abell AN, Ancrile BB, Johnson GL. CCM1 and CCM2 protein interactions in cell signaling: Implications for cerebral cavernous malformations pathogenesis. Hum Mol Genet. 2005. 14: 2521-31

83. Zhang J, Clatterbuck RE, Rigamonti D, Chang DD, Dietz HC. Interaction between krit1 and icap1alpha infers perturbation of integrin beta1-mediated angiogenesis in the pathogenesis of cerebral cavernous malformation. Hum Mol Genet. 2001. 10: 2953-60

84. Zhang J, Clatterbuck RE, Rigamonti D, Dietz HC. Cloning of the murine Krit1 cDNA reveals novel mammalian 5’ coding exons. Genomics. 2000. 70: 392-5

85. Zhang N, Pan L, Wang BJ, Wang EM, Dai JZ, Cai PW. Gamma knife radiosurgery for cavernous hemangiomas. J Neurosurg. 2000. 93: S74-7


Management of intracerebral hemorrhage in pediatric neurosurgery

$
0
0

Visish M. Srinivasan, Loyola V. Gressot, Bradley S. Daniels, Jeremy Y. Jones, Andrew Jea, Sandi Lam
  1. Department of Neurosurgery, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, USA

Correspondence Address:
Sandi Lam
Department of Neurosurgery, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, USA

DOI:10.4103/2152-7806.196919

Copyright: © 2016 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Visish M. Srinivasan, Loyola V. Gressot, Bradley S. Daniels, Jeremy Y. Jones, Andrew Jea, Sandi Lam. Management of intracerebral hemorrhage in pediatric neurosurgery. 28-Dec-2016;7:

How to cite this URL: Visish M. Srinivasan, Loyola V. Gressot, Bradley S. Daniels, Jeremy Y. Jones, Andrew Jea, Sandi Lam. Management of intracerebral hemorrhage in pediatric neurosurgery. 28-Dec-2016;7:. Available from: http://surgicalneurologyint.com/surgicalint_articles/management-of-intracerebral-hemorrhage-in-pediatric-neurosurgery/

Date of Submission
21-Mar-2016

Date of Acceptance
07-Apr-2016

Date of Web Publication
28-Dec-2016

Keywords: Arteriovenous malformation, cavernoma, cavernous malformation, intracerebral hemorrhage, intracranial hemorrhage, neurovascular

INTRODUCTION

Pediatric stroke is a relatively rare occurrence, with an annual incidence of 1.2–13 cases per 100,000. Hemorrhagic strokes account for half of these cases.[ 28 ] In adults, hemorrhagic strokes are predominantly hypertensive in etiology. However, in the pediatric population, they are frequently associated with vascular lesions such as AVMs (47%), arteriovenous fistulas, or CMs [ Table 1 ].[ 21 ] Other causes of ICH in adults, such as amyloid angiopathy or drug-related vascular damage, are rarely seen in the pediatric population. Workup of pediatric ICH should include vascular imaging consisting of either CTA or DSA. An MRI of the brain should be obtained to detect CMs, which are angiographically occult. MR angiography may lack the sensitivity to allow for visualization of the smaller vessels that may be involved with some of these lesions.[ 25 ] In a series of 137 patients with ICH described by Hino et al., 9% were found to have an “occult” vascular lesion that was not visualized on first angiogram. The clinical index of suspicion should guide the workup further if a causative lesion cannot be identified upon initial imaging. This may include repeat DSA, which is considered the gold standard for the assessment of vascular lesions. In the setting of clinically symptomatic hemorrhage, any vascular lesion, including AVM, CM, capillary telangiectasias, or developmental venous anomalies, may present in occult fashion, though AVM is most common.[ 7 ]


Table 1

Most common etiologies of spontaneous intracerebral hemorrhage in adults and children

 

ILLUSTRATIVE CASES

Case 1

A 7-year-old girl presented with acute onset headache, aphasia, and right hemiparesis. She was found to have a large left temporal intracerebral hemorrhage (ICH) and underwent emergent decompressive craniectomy without direct evacuation of hematoma [ Figure 1a ]. Postoperative imaging studies, including magnetic resonance imaging (MRI) and digital subtraction angiogram (DSA), failed to show a causative lesion [Figures 1b and c ]. She went to rehab and had considerable improvement of her aphasia and hemiparesis. Six weeks later, repeat imaging with DSA was performed after resolution of the initial hemorrhage, which also failed to show an associated vascular lesion. Because of the high index of clinical suspicion for arteriovenous malformation (AVM), a third angiogram was performed at 4 months post-hemorrhage. This showed a small left temporal AVM with a small nidus and early draining vein, which was the likely culprit of the patient's initial hemorrhage [ Figure 1d ]. Because of its small size, the lesion was not visualized on MRI or computed tomography (CT) angiogram; thus, radiosurgery was not a treatment option. Endovascular embolization was considered but deemed a poor option due to the high degree of difficulty in accessing the feeding vessel and concerns of the durability of treatment in this young patient. Because of the difficulty in localization, DynaCT was performed with the catheter in the feeding vessel from the inferior branch of the left MCA. This allowed preoperative study, planning, and intraoperative neuronavigation.[ 26 ] The lesion was successfully resected in total, and cranioplasty was performed in the same setting. Postoperative DSA demonstrated complete extirpation of the AVM, with tissue confirmation by pathology. The patient made an excellent recovery. She is ambulatory without focal motor deficit and has regained normal language function.


Figure 1

Left frontal hemorrhagic lesion from Case 1. (a) Computed tomography of the head at presentation showed a 9 × 4 × 4 cm hematoma with midline shift. (b) Magnetic resonance imaging brain, T2 sequence showed surrounding vessels but no definite arteriovenous malformation nidus around the large hematoma. (c) Left internal carotid artery digital subtraction angiogram at presentation, without evidence of vascular malformation. (d) Left internal carotid artery digital subtraction angiogram at 4 months post-hemorrhage reveals a left temporal Grade 1 arteriovenous malformation (arrow)

 

Case 2

A 9-year-old girl presented with acute onset of headache and obtundation. CT of the head demonstrated a large right cerebellar hemorrhage with tonsillar herniation, brainstem compression, and obstructive hydrocephalus [ Figure 2a ]. CT angiography (CTA) was suspicious for a vascular lesion. In emergent fashion, ventriculostomy was placed, followed by posterior fossa decompression. Because of the suspicion of a vascular lesion requiring further characterization, suboccipital decompression with craniectomy and dural decompression and minimal subtotal resection of the hematoma were performed. After surgical stabilization, the patient underwent DSA and MRI, which revealed an AVM, fed by branches of the right anterior and posterior inferior cerebellar arteries (AICA and PICA) [Figure 2b and c ]. She returned to the operating room the next day for definitive resection of the AVM. Postoperative DSA confirmed complete extirpation of the ruptured AVM and surrounding hematoma [ Figure 2d ]. Ventriculostomy was subsequently weaned, and the patient recuperated with rehabilitation. At one-year follow-up, the patient was independent with activities and had returned to school.


Figure 2

Right cerebellar hemorrhagic lesion from Case 2. (a) Computed tomography of the head at presentation showed a large hemorrhage of the right cerebellar hemisphere with displacement of the fourth ventricle. (b) Magnetic resonance imaging brain, T2 sequence showed perilesional edema and some associated vessels. (c) Right vertebral artery injection DynaCT, coronal view, showed an arteriovenous malformation with feeders from the right anterior and posterior cerebellar arteries. (d) Postoperative angiogram, AP projection, showed no residual malformation

 

Case 3

A 14-year-old girl presented with acute onset of headache, nausea, and vomiting for 1 day. She had similar but mild symptoms 2 weeks prior. She had a generalized seizure upon arrival to the hospital. CT of the head revealed a large 5 cm left frontal hemorrhage with mixed density and significant mass effect with exuberant perilesional edema [ Figure 3a ]. She was intubated for diminished sensorium and subsequently deteriorated as a result of uncal herniation. CT of the head with contrast demonstrated a lesion that appeared to be consistent with a tumor, though not definitive [ Figure 3b ]. She was taken for emergent decompressive craniectomy for stabilization. Subsequently, she returned to her prior neurological baseline. She proceeded to undergo MRI and MRA of the brain, which were suggestive of a cavernous malformation (CM) rather than a hemorrhagic tumor [ Figure 3c ]. The lesion was completely resected [ Figure 3d ], followed by replacement of her bone flap. Pathology confirmed hemorrhage and CM. The patient made an excellent functional recovery, with resolving mild abulia secondary to her dominant frontal lobe injury.


Figure 3

Left frontal hemorrhage from Case 3. (a) Computed tomography of the head showed a large left frontal intracerebral hemorrhage with subfalcine shift and perilesional edema. (b) Computed tomography with contrast showed partial enhancement of the lesion without any vascular feeders. (c) Magnetic resonance imaging brain, gradient echo sequence, showed a central lesion with surrounding hemorrhage, suggestive of cavernous malformation. (d) Postoperative magnetic resonance imaging brain with contrast showed complete resection of the lesion and hematoma, with resolution of mass effect

 

MANAGEMENT

Upon initial diagnosis of intracerebral hemorrhage on noncontrast CT, workup and treatment should be initiated without delay. The guidelines for the treatment of spontaneous ICH were last updated in 2010; these recommendations also apply to pediatric patients [ Table 2 ].[ 22 ] Based on institutional availability and clinical suspicion, workup should proceed with both vascular imaging (CTA or DSA) and MRI of the brain. Some authors have reported that MRI, MRA, and MRV are sufficient to diagnose vascular lesions in up to 66% of patients.[ 19 ] We suggest that MR vascular studies be supplemented with their CT-based counterparts if clinical suspicion is high for the improved resolution and sensitivity of CTA as MR imaging has a false-negative rate of 7%.[ 19 ] Intensive care and monitoring are indicated. Conservative management with supportive care may be appropriate for self-limited hemorrhage without progressive mass effect or elevated intracranial pressure (ICP).


Table 2

Summary of guidelines for management of spontaneous intracerebral hemorrhage

 

SURGICAL NUANCES

Several surgical options exist for managing acute ICH requiring intervention. First and foremost are the evaluation and management of “ABCs:” Airway, breathing, and circulation. If high intracranial pressure is present, it must be addressed with external ventricular drainage, evacuation of the hematoma, and/or decompressive craniectomy with expansile duraplasty depending on the clinical situation and the location of the hemorrhage. If there is suspicion of underlying AVM, limited evacuation of the hematoma is advised only if necessary in cases of mass effect because aggressive evacuation of the hematoma may precipitate AVM re-rupture. The thrombus cap over the rupture site can be tenuous.[ 16 ] In non-eloquent areas or in the posterior fossa where mass effect can be significant, evacuation of hematoma is typically undertaken with care, cognizant that an underlying AVM may be present.

In hemorrhages seemingly admixed with eloquent brain tissue, our experience supports a large craniectomy with expansile duraplasty in cases of significant cerebral edema, without surgical destruction of eloquent areas. This allows for the possibility of recovery of function. Direct removal of the hematoma and the vascular malformation at initial surgery may not be possible due to high ICP and incomplete workup due to clinical instability, and thus, incomplete appreciation for the anatomy and localization of the lesion, as described in Case 1. Craniectomy or minimal evacuation of hematoma, followed by a more lesion-tailored approach with the support of diagnostic imaging, may be prudent in these cases. In contrast to ruptured aneurysms that have a high short-term risk of re-rupture, thus warranting urgent securement, ruptured AVMs and cavernomas have relatively low acute re-rupture rates, which allow slightly more conservative management.[ 8 9 ] This permits the benefit of proceeding to surgery under optimal circumstances with adequate imaging and a medically optimized, stable patient. Multiple-modality imaging should be used, and in the case of equivocal findings, repeat imaging is indicated as obscuring hemorrhage resolves. Modern management can include the use of intraoperative indocyanine green videoangiography (ICG-VA) or intraoperative DSA in a hybrid OR-angiography suite.

ARTERIOVENOUS MALFORMATIONS

AVMs are the most common cause of ICH in children. They carry an annual risk of hemorrhage of approximately 3.2%, a 5–10% mortality rate, and a 50% risk of neurological morbidity.[ 4 23 ] The large majority of AVMS are clearly evident on DSA, although several authors dating back several decades have described angiographically “occult” AVMs that were obscured following rupture.[ 24 27 ] Hypotheses for this phenomenon include possible lesional thrombosis, compressive occlusion of lesion from hematoma, transient vasospasm, or small nidus.[ 24 ] The Lawton supplementary scale has been used in recent years as an adjunct to the well-established Spetzler–Martin grading scale for assessment of surgical risk in AVMs.[ 14 17 ] The Lawton supplementary scale validates the experience that pediatric patients and lesions with previous hemorrhage carry lower surgical morbidity. This helps to further guide the treatment decision-making in AVM-associated pediatric ICH. While SRS may play a role in lesions with unacceptably high surgical risk,[ 1 ] the risk of short-term hemorrhage and lesional persistence is concerning in pediatric patients with a higher cumulative lifetime risk of hemorrhage.[ 13 ] With a 20% failure rate of primary SRS therapy, radiosurgery is best used as an adjunct to a more definitive cure in the pediatric patient.[ 10 ] Endovascular embolization, frequently performed with Onyx or n-BCA at our center, usually plays an adjunct role to surgical resection but is not used as the primary treatment modality.

Postoperative surveillance of AVM

AVMs in children are fraught with a high recurrence rate (as high as 14%),[ 23 ] especially in lesions with a diffuse nidus[ 15 ] or deep venous drainage. There are no definitive guidelines for postoperative surveillance, however, undoubtedly, DSA has the highest resolution and sensitivity. Although surveillance patterns vary greatly, at our institution we recommend intraoperative/immediate postoperative DSA, followed by 1-year DSA, yearly CTA, and DSA every 3 years until age 18. Concerns over morbidity of the procedure are discussed, though the overall procedural complication rate is less than 1%.[ 18 ] A recent series suggests that MRA, although benign in the sense of radiation and contrast exposure, has reduced sensitivity.[ 23 ] CTA is considered as a noninvasive surveillance tool with higher sensitivity at recurrent AVM detection than MRA[ 23 ] and is used at our institution at yearly intervals between surveillance DSA performed at 3 years intervals.

CAVERNOUS MALFORMATIONS

CMs represent 20–25% of spontaneous ICH in children.[ 12 ] CM-associated ICH is generally smaller and has better outcomes compared to ICH from AVM. Rarely, CM-associated ICH can produce significant mass effect and may even be fatal.[ 2 ] Various factors have been associated with symptomatic hemorrhage risk. Chief among them are prior hemorrhage, brainstem location, and associated developmental venous anomalies (DVAs). The natural history of CMs includes temporal clustering of hemorrhages, often seen within the first 3 years of a herald bleed. This known grouping of hemorrhages should be an important consideration in management.[ 9 ] Unhemorrhaged lesions have an annual bleeding rate of <2%, whereas previously hemorrhaged lesions can have much higher rates (4.5–22.6%).[ 3 ] The appearance on MRI has been described in four classes by Zabramski et al.[ 30 ] While the classical “popcorn” appearance (Zabramski Type 2) is the most common, it is important to be familiar with the more atypical appearances as well. CMs are angiographically silent; thus, MRI is the only way to visualize them. Large ICH from CMs can obscure the lesion itself, making initial diagnosis challenging at times.[ 11 29 ] The initial hemorrhage can sometimes take months to fully resolve. In pediatric patients with ICH, it is imperative to maintain a high index of suspicion for causative vascular lesions and to continue aggressive diagnostic workup given the relatively high incidence in this population.[ 5 ] Therefore, MRI following the resolution of the acute hemorrhage is mandatory to rule out underlying vascular lesions. Advanced MRI sequences, such as gradient echo (GRE) and susceptibility weighted imaging (SWI), have a better sensitivity for blood and may better characterize the lesion[ 6 ] or identify other lesions in the setting of multiple CMs. Specifically, because there are no intrinsic 180-degree refocusing pulses in gradient echo sequences (as compared to a spin echo sequences), GRE images are more sensitive to the dephasing and signal losses that result from paramagnetic blood products. SWI further increases sensitivity to dephasing from these blood products by incorporating and mathematically amplifying this phase information in the image generation process. Cavernomas typically have hemosiderin rings on T2-weighted sequences.

CONCLUSION

Intracerebral hemorrhage in children requires special consideration in management compared to the same in adults. The rate of associated vascular malformations, including but not limited to AVMs and CMs, is high. These should be considered during initial evaluation, hematoma management, and surgical management of the offending lesion.[ 20 ]

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Abla AA, Rutledge WC, Seymour ZA, Guo D, Kim H, Gupta N. A treatment paradigm for high-grade brain arteriovenous malformations: Volume-staged radiosurgical downgrading followed by microsurgical resection. J Neurosurg. 2015. 122: 419-32

2. Al-Holou WN, O’Lynnger TM, Pandey AS, Gemmete JJ, Thompson BG, Muraszko KM. Natural history and imaging prevalence of cavernous malformations in children and young adults. J Neurosurg Pediatr. 2012. 9: 198-205

3. Bilginer B, Narin F, Hanalioglu S, Oguz KK, Soylemezoglu F, Akalan N. Cavernous malformations of the central nervous system (CNS) in children: Clinico-radiological features and management outcomes of 36 cases. Childs Nerv Syst. 2014. 30: 1355-66

4. Blauwblomme T, Bourgeois M, Meyer P, Puget S, Di Rocco F, Boddaert N. Long-term outcome of 106 consecutive pediatric ruptured brain arteriovenous malformations after combined treatment. Stroke. 2014. 45: 1664-71

5. Clatterbuck RE, Moriarity JL, Elmaci I, Lee RR, Breiter SN, Rigamonti D. Dynamic nature of cavernous malformations: A prospective magnetic resonance imaging study with volumetric analysis. J Neurosurg. 2000. 93: 981-6

6. de Champfleur NM, Langlois C, Ankenbrandt WJ, Le Bars E, Leroy MA, Duffau H. Magnetic resonance imaging evaluation of cerebral cavernous malformations with susceptibility-weighted imaging. Neurosurgery. 2011. 68: 641-

7. Greenberg MS.editorsHandbook of Neurosurgery. Tampa: Greenberg Graphics Inc; 2010. p.

8. Gross BA, Du R. Natural history of cerebral arteriovenous malformations: A meta-analysis. J Neurosurg. 2013. 118: 437-43

9. Gross BA, Du R, Orbach DB, Scott RM, Smith ER. The natural history of cerebral cavernous malformations in children. J Neurosurg Pediatr. 2015. p. 1-6

10. Hoh BL, Ogilvy CS, Butler WE, Loeffler JS, Putman CM, Chapman PH. Multimodality treatment of nongalenic arteriovenous malformations in pediatric patients. Neurosurgery. 2000. 47: 346-

11. Ide C, De Coene B, Baudrez V. MR features of cavernous angioma. JBR-BTR. 2000. 83: 320-

12. Jordan LC, Hillis AE. Hemorrhagic stroke in children. Pediatr Neurol. 2007. 36: 73-80

13. Kano H, Kondziolka D, Flickinger JC, Yang HC, Flannery TJ, Awan NR. Stereotactic radiosurgery for arteriovenous malformations, part 2: Management of pediatric patients. J Neurosurg Pediatr. 2012. 9: 1-10

14. Kim H, Abla AA, Nelson J, McCulloch CE, Bervini D, Morgan MK. Validation of the supplemented Spetzler-Martin grading system for brain arteriovenous malformations in a multicenter cohort of 1009 surgical patients. Neurosurgery. 2015. 76: 25-31

15. Klimo P, Rao G, Brockmeyer D. Pediatric arteriovenous malformations: A 15-year experience with an emphasis on residual and recurrent lesions. Childs Nerv Syst. 2007. 23: 31-7

16. Lawton MT.editorsSeven AVMs: Tenets and Techniques for Resection. New York: Thieme; 2014. p.

17. Lawton MT, Kim H, McCulloch CE, Mikhak B, Young WL. A supplementary grading scale for selecting patients with brain arteriovenous malformations for surgery. Neurosurgery. 2010. 66: 702-13

18. Lin N, Smith ER, Scott RM, Orbach DB. Safety of neuroangiography and embolization in children: Complication analysis of 697 consecutive procedures in 394 patients. J Neurosurg Pediatr. 2015. 16: 432-8

19. Liu AC, Segaren N, Cox TS, Hayward RD, Chong WK, Ganesan V. Is there a role for magnetic resonance imaging in the evaluation of non-traumatic intraparenchymal haemorrhage in children?. Pediatr Radiol. 2006. 36: 940-6

20. Meretoja A, Strbian D, Putaala J, Curtze S, Haapaniemi E, Mustanoja S. SMASH-U: A proposal for etiologic classification of intracerebral hemorrhage. Stroke. 2012. 43: 2592-7

21. Meyer-Heim AD, Boltshauser E. Spontaneous intracranial haemorrhage in children: Aetiology, presentation and outcome. Brain Dev. 2003. 25: 416-21

22. Morgenstern LB, Hemphill JC, Anderson C, Becker K, Broderick JP, Connolly ES. Guidelines for the management of spontaneous intracerebral hemorrhage: A guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2010. 41: 2108-29

23. Morgenstern PF, Hoffman CE, Kocharian G, Singh R, Stieg PE, Souweidane MM. Postoperative imaging for detection of recurrent arteriovenous malformations in children. J Neurosurg Pediatr. 2015. p. 1-7

24. Ogilvy CS, Heros RC, Ojemann RG, New PF. Angiographically occult arteriovenous malformations. J Neurosurg. 1988. 69: 350-5

25. Osborn AG.editorsOsborn's Brain: Imaging, Pathology, and Anatomy. Salt Lake City: Amirsys Publishing, Inc; 2013. p.

26. Srinivasan VM, Schafer S, Ghali MG, Arthur A, Duckworth EA. Cone-beam CT angiography (Dyna CT) for intraoperative localization of cerebral arteriovenous malformations. J Neurointerv Surg. 2016. 8: 69-74

27. Tsitsopoulos P, Andrew J, Harrison MJ. Occult cerebral arteriovenous malformations. J Neurol Neurosurg Psychiatry. 1987. 50: 218-20

28. Tsze DS, Valente JH. Pediatric stroke: A review. Emerg Med Int 2011. 2011. p. 734506-

29. Wardlaw JM, Keir SL, Seymour J, Lewis S, Sandercock PA, Dennis MS. What is the best imaging strategy for acute stroke?. Health Technol Assess. 2004. 8: iii-ix-x

30. Zabramski JM, Wascher TM, Spetzler RF, Johnson B, Golfinos J, Drayer BP. The natural history of familial cavernous malformations: Results of an ongoing study. J Neurosurg. 1994. 80: 422-32

Surgical management of lipomyelomeningocele in children: Challenges and considerations

$
0
0

Kathryn M. Wagner, Jeffrey S. Raskin, Daniel Hansen, Gaddum D. Reddy, Andrew Jea, Sandi Lam
  1. Department of Neurosurgery, Baylor College of Medicine/Section of Pediatric Neurosurgery, Texas Children's Hospital, Texas, USA

Correspondence Address:
Sandi Lam
Department of Neurosurgery, Baylor College of Medicine/Section of Pediatric Neurosurgery, Texas Children's Hospital, Texas, USA

DOI:10.4103/2152-7806.205268

Copyright: © 2017 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Kathryn M. Wagner, Jeffrey S. Raskin, Daniel Hansen, Gaddum D. Reddy, Andrew Jea, Sandi Lam. Surgical management of lipomyelomeningocele in children: Challenges and considerations. 26-Apr-2017;8:63

How to cite this URL: Kathryn M. Wagner, Jeffrey S. Raskin, Daniel Hansen, Gaddum D. Reddy, Andrew Jea, Sandi Lam. Surgical management of lipomyelomeningocele in children: Challenges and considerations. 26-Apr-2017;8:63. Available from: http://surgicalneurologyint.com/surgicalint-articles/surgical-management-of-lipomyelomeningocele-in-children-challenges-and-considerations/

Date of Submission
20-Oct-2016

Date of Acceptance
08-Dec-2016

Date of Web Publication
26-Apr-2017

Keywords: Spinal lipoma, lipomyelomeningocele, spinal dysraphism, pediatric

ILLUSTRATIVE CASES

Case 1

A 3-month-old boy was referred to neurosurgery when a subcutaneous bulge in the lower lumbar region was incidentally noted. Initial workup included an ultrasound of the region that was concerning for spinal dysraphism, including sacral agenesis and an associated intraspinal mass. These findings prompted a lumbosacral magnetic resonance image (MRI), which confirmed the diagnosis of lipomyelomeningocele. The patient was clinically asymptomatic, with normal strength in his lower extremities, no evidence of hydrocephalus, and normal bowel and bladder function. Because he was meeting developmental milestones, he was managed observantly with annual clinical exams, which remained normal. At 3 years of age, he underwent urodynamic studies, which were unremarkable, and he was able to successfully toilet train. At around this time, his parents reported transient morning stiffness in the back and lower legs, which would resolve by the afternoon. This prompted a repeat MRI [ Figure 1 ], which demonstrated the development of a syrinx in the lumbar region of the spinal cord. Surgical debulking and untethering of the lipomyelomeningocele was discussed with the parents, along with the associated risks and potential benefits, and, ultimately, the decision was made to continue expectant management. At 5 years of age, he remains clinically asymptomatic and continues to meet developmental milestones.


Figure 1

(a) T1 (left) and T2 (right) sagittal sections of an MRI showing the presence of a caudal lipomyelomeningocele with an associated lumbar syrinx (arrows) in a clinically asymptomatic patient. (b) Selected T2 axial sections as identified by the color coding on the sagittal image in A

 

Case 2

An 11-day-old girl with an uncomplicated birth was noted by her parents to have a lump on her lower back, prompting further workup by her pediatrician. A physical exam revealed a 4–5 cm soft, nontender mass in the lumbar spine with hyperpigmented changes [ Figure 2 ]. An ultrasound was performed, which showed the absence of sacral lamina and dorsal elements with an associated intraspinal mass and bilateral hydronephrosis. She subsequently underwent an MRI showing a spinal dysraphism of the sacrum associated with a large cystic intraspinal mass, concerning for a lipomyelomeningocele versus a terminal lipomyelocystocele. Her motor exam showed normal strength in both lower extremities but poor deep tendon reflexes and a slightly distended abdomen. Further urological workup revealed the presence of grade 4 vesicoureteral reflux for which she was started on a clean intermittent catheterization protocol. After a thorough discussion about the risks and benefits of operative intervention, she was taken to the operating room (OR) for debulking of the lipomatous mass and untethering. Electrical stimulation and neuromonitoring were used to monitor function. The dural tube was reconstructed using a synthetic patch duroplasty. No cyst was encountered during the debulking. There were no intraoperative complications, however, 1 week postoperatively she developed a cerebrospinal fluid (CSF) leak, which precipitated a Klebsiella wound infection. She returned to the OR for irrigation and debridement, placement of a lumbar drain, and revision of the dural closure. Postoperatively, she remained intubated and prone for 1 week, at which point the lumbar drain was removed and she was extubated. She had no further leakage from the wound and was discharged approximately 5 weeks after her first surgery. At 18-months’ postoperative follow-up, her wound was well healed, and she retained normal strength in her bilateral lower extremities. She progressed to grade 5 vesicoureteral reflux with moderate hydronephrosis without urinary tract infections, managed by clean intermittent catheterization every 3 hours.


Figure 2

(a) Photograph demonstrating a lumbar mass and associated hyperpigmentation. (b) T1 (left) and T2 (right) sagittal sections of an MRI showing the lumbosacral dysraphism and associated cystic lumbar mass. (c) Selected T2 axial sections as identified by color coding on the sagittal image in A

 

INTRODUCTION

Lipomyelomeningocele (LMMC) is a closed neural tube defect in which neural elements are incorporated into a spinal lipoma. This is an uncommon defect, occurring in 3–6 patients per 100,000 live births.[ 14 ] Clinical decision-making regarding treatment is complicated by the varied pathology and the spectrum of presentations. Herein, LMMC embryology, morphology, treatment options, and outcomes are reviewed.

Embryology

Central nervous system development consists of primary and secondary neurulation. During primary neurulation, the notochord induces folding of the neural plate to form the neural tube, which extends in both the rostral and caudal directions. The ectoderm overlying the neural tube separates to ultimately form the skin dorsal to the spine, a process known as dysjunction. Mesoderm around the neural tube differentiates into the posterior vertebral elements, fat, and paraspinal musculature. In premature dysjunction, mesoderm can migrate into the neural tube before it is fully closed, disrupting the neurulation process. This mesoderm then differentiates into fat and forms a border between the neural placode and the now entrapped lipoma. As development continues, meninges form around the neural tube except at the placode-lipoma interface, leaving a dorsal diaschisis traversed by a lipoma. This often results in a distinct transition point between normal planes and anatomy, anteriorly, and the lipoma, posteriorly.[ 13 ]

In secondary neurulation, a caudal mass of mesenchymal mesoderm cavitates and fuses with the primary neural tube, forming the spine below S2. After fusion of the primary and secondary neural tubes, mesoderm can migrate caudally and interfere with secondary neurulation in a mechanism similar to the disruption of primary neurulation. Secondary neurulation differs phylogenetically and is incompletely understood. Humans lack mature tail structure and have less complexity of secondary neurulation comparatively. In chick embryos, dynamic histology describes a coalescing of radially oriented tubules around a central lumen, ultimately cavitating within the caudal cell mass and joining the primary neurulated structure. Prevailing theories involve morphogenetic determinants, with candidate genes including sonic hedgehog and Pax transcription factors.[ 10 ] Environmental factors are posited to interfere with the enfolding mechanism of secondary neurulation including folate deficiency, viremia, and teratogens as examples; however, little incidence data supports this supposition.[ 8 ]

The morphology of spinal lipomas is thought to be determined by which of the two neurulation processes is affected. Regardless of the morphology, in LMMC the placode-lipoma junction lies outside the spinal canal with dorsal extension of the meninges through an accompanying bony defect, in contrast to residing inside the canal, as seen in a lipomyelocele.[ 29 ] After the lipoma exits the dural defect, it continues through a fascial defect to communicate with subcutaneous tissue. This tethers the spinal cord and restricts its ability to ascend normally, making it susceptible to stretch injury during spine growth or repetitive ischemic insults resulting from flexion/extension movements.[ 15 21 24 ]

Classification

Traditionally, spinal lipomas have been classified into three groups based on the location of the neural placode–lipoma junction: Dorsal, caudal, and transitional, known as the Chapman classification[ 3 5 ] [ Figure 3 ]. In dorsal spinal lipomas, the junction is on the dorsal aspect of the lumbar spinal cord and spares the conus medullaris. The dorsal root entry zone (DREZ) and neural elements are displaced lateral and ventrolateral to the placode–lipoma junction, respectively. The nerve roots emerge from the spinal cord tissue anterior to the junctional zone, where the lipoma, dura, and conus medullaris converge. In contrast, the conus is involved with caudal lipomas, and neural elements are located rostral to the junction. In caudal lipomas, the fatty tissue can extend from within the central canal caudally, where the fat is intermixed with nerve roots. The distal cord thus appears progressively larger in diameter toward the caudal aspect. Other findings may include a restrictive transverse fibrous band at the level of the last intact lamina. Transitional lipomas have characteristics of both dorsal and caudal types, with viable nerve roots passing through the lipoma tissue. These lesions tend to be asymmetric, with a rotational component on the spinal cord. The placode–lipoma junction is thus typically rotated. In addition, Pang et al. described a chaotic type, which has an irregular border between the placode and lipoma. Fat extends around the spinal cord and onto its ventral aspect, obscuring the DREZ. Most LMMCs are of the dorsal or transitional type.[ 30 ]


Figure 3

Classification of lipomas of the conus medullaris, as described by Chapman. Dorsal, caudal, and transitional types (right to left)

 

Premature dysjunction is necessary for all types of spinal lipomas, except the chaotic type. Primary and secondary neurulation is disrupted in the dorsal and caudal types, respectively, whereas both are affected in the transitional type. The chaotic type is thought to involve only secondary neurulation, where mesenchymal cells may become mixed with the caudal stem cell mass.

Presentation

Spinal lipomas and LMMCs are frequently associated with cutaneous and musculoskeletal abnormalities in addition to sensorimotor deficits and urological dysfunction.[ 29 ] Cutaneous lesions include subcutaneous lipomas, capillary hemangiomas, complex dimples, and hypertrichosis, whereas complex malformations, such as dermal appendages, are rare.[ 12 15 ] Musculoskeletal findings include scoliosis, unilateral or bilateral foot deformities, such as pes cavus, club feet, or abnormal rotation, or asymmetry of the foot or leg. Any of these findings should prompt consideration of an underlying embryomorphic etiology. Urological dysfunction, such as incontinence, frequency, urgency, and urinary tract infections, are also commonly associated. Neurological symptoms frequently correspond to those expected of a tethered cord syndrome, such as back or leg pain at rest that worsens with activity, in addition to weakness, sensory disturbances, or gait abnormalities.[ 9 ]

At birth, neurological symptoms may be absent in nearly half of the cases.[ 16 ] As the infant ages and axial growth occurs, the infant may experience progressive loss of neurological function.[ 19 ] Often, a change in the pattern of bladder and bowel function is the presenting symptom of LMMC.[ 9 ] As axial growth continues, lower limb, and sacral motor and sensory dysfunction, such as radicular pain, leg spasticity, foot deformities, and gait abnormalities, can develop.[ 17 ] Consequently, older children who escape early detection of LMMC are more likely to present with more pronounced urological and neurological complaints.[ 2 ] In addition to the symptomatic progression that correlates with axial growth, morphology of the defect also plays a large role in the presentation of LMMC patients. Symmetric malformations without a rotational component to the lipoma–placode interface tend to cause bilateral neurological or orthopedic abnormalities, which present at a later age. In contrast, asymmetric malformations tend to cause unilateral functional abnormalities and present earlier in life, usually on the side to which the neural placode was rotated.[ 2 ] Finally, LMMCs can become symptomatic from spinal stenosis secondary to mass effect as the lipomatous malformation increases mass over time.[ 30 ]

LMMC can be associated with additional pathologies, including Chiari malformation type 1 (13%), spina bifida (14.4%), split cord malformations (3.1%), associated dermal sinuses (3.1%), dermoid or epidermoid cysts (3.1%), diastematomyelia (3.1%), terminal hydromyelia (3.1%), anal stenosis (1.0%), and Down syndrome (1.0%).[ 18 23 33 ]

DIAGNOSTIC STUDIES

Ultrasound is an effective screening tool because it is low risk and widely available, however, it has limited use after the initial diagnosis or following surgical treatment and should not be relied upon as the sole preoperative assessment.[ 22 ] A detailed MRI is the definitive imaging evaluation for spinal-neural lipomas. The anatomical detail of the placode-lipoma junction can be shown in relation to the normal spinal cord. Plain radiographs or computed tomography (CT) imaging may be useful to assess for scoliosis and evaluate the spine's bony anatomy during preoperative planning.

MANAGEMENT

Historical studies have shown that surgical interventions may briefly stabilize or relieve neurological symptoms but ultimately fail to improve upon the natural history of LMMCs.[ 6 21 23 28 34 ] Symptoms are typically progressive and worsen with age. Kulkarni et al. shed light on the natural history, citing a 33% risk of symptom deterioration with conservative management versus 46% for surgical treatment at nine-year follow-up.[ 28 ] One downfall of this study was that the conservative group was prospectively followed, whereas the surgical cohort was treated in the 1970s and retrospectively analyzed. Nevertheless, this study has changed the outlook and management of asymptomatic patients previously thought to require surgical intervention. The argument against surgical intervention for asymptomatic spinal lipomas was reinforced again in a 2012 London study that found a 40% cumulative risk of deterioration at 10 years.[ 35 ]

Surgical intervention may provide temporary relief or lessening of symptoms by releasing tension on the spinal cord, however, there is a risk of retethering with subsequent return or progression of neurologic symptoms, with reported rates of 5–50%. In a study by Colak et al. of 94 patients who underwent initial repair of a LMMC, 20.2% required subsequent operations for symptomatic retethering, with an average follow-up of 52 months after surgery. Of these reoperated patients, 6.4% exhibited repetitive symptomatic tethering, which became more difficult to treat and with shorter times between return of symptoms. Colak concluded that even after an adequate initial operation, symptomatic retethering is a common problem and that no current duraplasty graft material entirely prevents this from occurring.[ 7 ] There has been a confusing array of studies showing progression of neurologic symptoms after surgery to be lower,[ 4 ] similar to,[ 3 6 ] or worse than[ 11 ] the natural history. Cochrane et al. suggested that the variance in these results may be due to differences in the symptoms assessed, the duration of follow-up, the type of malformation, and timing of surgery.[ 6 ] Many experts argue for conservative treatment, with close clinical evaluations and surgical intervention only as patients develop worsening symptoms, as the natural history is similar or marginally better than the long-term outcome of surgically-treated patients.

A recent retrospective review attempted to identify radiological correlates predicting neurological decline. Over 16 years, a 24-patient population with LMMC that underwent an observational management strategy at a single institution was dichotomized into those experiencing early (less than 18 months) and late (18–30 months) neurological deterioration. Nine patients experiencing early deterioration were more likely to have large intradural lipomatous masses, which grew within the first year to exert regional mass effect on neural structures and were associated with a large expanded syrinx. Early decliners were more likely to present with motor deficits, whereas 15 patients experiencing late neurological decline presented with mixed urologic and motor deficits.[ 32 ] This study supports prophylactic untethering in infants with large intradural lipomas with syrinx, which exert mass effect on neural structures.

Several factors are thought to affect the treatment outcome of LMMCs. Age, gender, morphology, the presence and severity of neurological symptoms, and absence or presence of an associated spinal cord syrinx are all taken into consideration. Of these, morphology is considered the most crucial factor affecting outcome. For example, transitional lipomas appear to have a higher rate of retethering after surgery than dorsal and caudal types.[ 7 ] It is unclear if this is related to a factor intrinsic to the embryology or related to lesional complexity precluding adequate untethering.[ 30 31 ] It is also hypothesized that these factors may affect the outcome through a common pathway described by Pang et al. as the postoperative cord-to-sac ratio, which his group noted to be directly correlated to the progression or return of neurologic symptoms.[ 30 ] They suggested that cord retethering is a result of too little space in the dural sac for the spinal cord. This forces into close proximity any remaining residual lipoma surface with the normal spinal cord, which can then adhere. If the lipoma is completely removed and space within the dural sac is increased, they posited there is less chance of the cord contacting the dura or the “sticky” lipoma surface and retethering.

The traditional surgical technique described in historical studies involves partial resection of the lipoma to avoid injury to the neural placode, followed by untethering of the cord, then apposition of the edges of the placode, and finally duraplasty.[ 1 ] This technique, however, does not dramatically affect the cord-to-sac ratio. Pang et al. suggested a more aggressive approach involving total or near-total resection of the lipoma, complete reconstruction of the neural placode, followed by expansile duraplasty, preferably using bovine pericardium.[ 30 ] Using their technique, they showed no neurologic deterioration in 88.1% of patients over 20 years of follow-up, compared to 34.6% risk of progression over 10 years in patients with only partial resection. This included lipomas of all types, including symptomatic, asymptomatic, unoperated, and redo subgroups. Progression free survival (PFS) for asymptomatic unoperated spinal lipomas with this aggressive resection method was 98.8% over 20 years. Multivariate analysis of their data showed cord-to-sac ratio to be the only independent factor predicting outcome. These results have not been reproduced in any other series to date, however, they are compelling numbers for those who hold that surgical intervention can improve upon the natural history of LMMCs, whether or not symptoms are present.

A radically different approach to the treatment of tethered cord comes in the form of vertebral column shortening, which offers an alternative method for relieving tension on the spinal cord without risking injury to the neural placode and possibly stabilizing or improving neurological outcome.[ 20 ] The three-column osteotomy typically involves T12 or L1, and the average reduction in height is approximately 20 mm. Potential surgical complications include pseudoarthrosis, neurological injury, and significant perioperative blood loss. Kokubun et al. used this surgical method to treat eight patients (ages 15–54 years), 3 of which had previous conventional untethering procedures.[ 26 ] Six patients remained stable, and 2 patients experienced neurological decline over an average follow-up of 6.2 years. For these patients with decline, it is unclear if additional height reduction is needed, or perhaps changes within the neural placode occur independent of tension produced by the tethering. Although thought to be a relatively safe treatment option when used by experienced surgeons, its use should be limited to symptomatic patients with recurrent tethering despite conventional surgical detetherings, until studies with larger patient numbers can demonstrate its safety and effectiveness as a primary treatment option.

Regardless of the type of surgical intervention, the use of operative microscope is recommended, and intraoperative neurophysiological monitoring should be performed. There are a number of various neurophysiologic measures that can be used, and institutional practice may dictate what is available. Whatever monitoring type is chosen, the goal remains to avoid unintended injury to intact nervous structures, which may be hidden by or attached to the LMMC.[ 25 27 ]

As mentioned earlier, management of symptomatic LMMCs may not always entail surgical untethering. Complex variants of the transitional or chaotic spinal lipomas associated with isolated urological symptoms or orthopedic deformities may be observed because these abnormalities are less likely to improve with surgical intervention. They also carry a higher risk of unsuccessful untethering and incomplete lipoma resection, which may lead to an increased risk of neurological deterioration after surgery.

After careful evaluation, “simple” LMMCs with symptoms or any LMMC with an associated sensorimotor deficit should be considered for possible surgical intervention. Caudal and dorsal spinal lipomas are typically more amenable to surgical treatment than the rest. Immediate postoperative complication rates range from 10–30% and include infection, CSF leak, or neurological deterioration.[ 1 23 ]

LMMCs associated with embryomorphic malformations of other systems may represent the more severe end of the spectrum of congenital defects. For example, OEIS (omphalocele, exstrophy, imperforate anus, spinal defects) and VATER (vertebral defects, anal atresia, tracheoesophageal fistula, renal abnormalities) represent associated multisystem abnormalities, which complicate management and are beyond the scope of this review.

CASES IN CONTEXT

Table 1 summarizes the salient details of Cases 1 and 2, along with the resultant clinical decision. Case 1 included incidental identification of an asymptomatic caudal LMMC with associated syrinx and without intradural mass effect. The patient had normal motor and urological exams, with only the development of leg tightening over 2 months. The clinical and radiographic data were combined into a clinical decision rule supported by the literature, and the patient has been stable through observational treatment. Case 2 is defined radiographically by caudal LMMC with large associated syrinx and mass effect on intradural neural structures. The patient also exhibited signs of tethering, including diminished deep tendon reflexes and abdominal distention, along with intercurrent embryomorphic malformations of the spine and renal system. In the presentation for Case 2, the literature supports untethering and debulking, which was performed. The patient had essentially static symptoms without significant worsening or improvement on follow-up at 18 months.


Table 1

Clinical decision-making for patients with lipomyelomeningocele

 

CONCLUSION

LMMC management remains a challenge. The selected cases demonstrate important factors integrated within a clinical decision rule. Although there is no high-quality clinical outcome data to provide guidance regarding the treatment options for LMMCs, conservative management of asymptomatic patients is appropriate. Clearly progressive symptomatic patients should be considered for surgical untethering with the goal of managing symptoms, with the patient and family prepared for an iterative process. Patients with static neurological deficits should be managed observationally. Prophylactic surgery may, theoretically, prevent the onset of neurological deterioration or stabilize and reverse early-onset symptoms at diagnosis, especially in infants with large intradural lipoma and associated syrinx, which compress neural structures, however, this has not been shown to offer immunity against further deterioration. When surgical management is elected, experts advocate aggressive resection of the lipoma, along with reconstruction of the placode and large expansile duraplasty, but the literature shows this is technically difficult and may not greatly improve upon the natural history of LMMCs.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Arai H, Sato K, Okuda O, Miyajima M, Hishii M, Nakanishi H. Surgical experience of 120 patients with lumbosacral lipomas. Acta Neurochir. 2001. 143: 857-64

2. Atala A, Bauer SB, Dyro FM, Shefner J, Shillito J, Sathi S. Bladder functional changes resulting from lipomyelomeningocele repair. J Urol. 1992. 148: 592-4

3. Blount JP, Elton S. Spinal lipomas. Neurosurg Focus. 2001. 10: e3-

4. Byrne RW, Hayes EA, George TM, McLone DG. Operative resection of 100 spinal lipomas in infants less than 1 year of age. Pediatr Neurosurg. 1995. 23: 182-6

5. Chapman PH. Congenital intraspinal lipomas: Anatomic considerations and surgical treatment. Childs Brain. 1982. 9: 37-47

6. Cochrane DD. Cord untethering for lipomyelomeningocele: Expectation after surgery. Neurosurg Focus. 2007. 23: E9-

7. Colak A, Pollack IF, Albright AL. Recurrent tethering: A common long-term problem after lipomyelomeningocele repair. Pediatr Neurosurg. 1998. 29: 184-90

8. Copp AJ, Stanier P, Greene ND. Neural tube defects: Recent advances, unsolved questions, and controversies. Lancet Neurol. 2013. 12: 799-810

9. Cornette L, Verpoorten C, Lagae L, Plets C, Van Calenbergh F, Casaer P. Closed spinal dysraphism: A review on diagnosis and treatment in infancy. Eur J Paediatr Neurol. 1998. 2: 179-85

10. Detrait ER, George TM, Etchevers HC, Gilbert JR, Vekemans M, Speer MC. Human neural tube defects: Developmental biology, epidemiology, and genetics. Neurotoxicol Teratol. 2005. 27: 515-24

11. Dorward NL, Scatliff JH, Hayward RD. Congenital lumbosacral lipomas: Pitfalls in analysing the results of prophylactic surgery. Childs Nerv Syst. 2002. 18: 326-32

12. Drolet B. Birthmarks to worry about. Cutaneous markers of dysraphism. Dermatol Clin. 1998. 16: 447-53

13. Finn MA, Walker ML. Spinal lipomas: Clinical spectrum, embryology, and treatment. Neurosurg Focus. 2007. 23: E10-

14. Forrester MB, Merz RD. Descriptive epidemiology of lipomyelomeningocele, Hawaii, 1986-2001. Birth Defects Res A Clin Mol Teratol. 2004. 70: 953-6

15. Guggisberg D, Hadj-Rabia S, Viney C, Bodemer C, Brunelle F, Zerah M. Skin markers of occult spinal dysraphism in children: A review of 54 cases. Arch Dermatol. 2004. 140: 1109-15

16. Hertzler DA, DePowell JJ, Stevenson CB, Mangano FT. Tethered cord syndrome: A review of the literature from embryology to adult presentation. Neurosurg Focus. 2010. 29: E1-

17. Hoffman HJ, Hendrick EB, Humphreys RP. The tethered spinal cord: Its protean manifestations, diagnosis and surgical correction. Childs Brain. 1976. 2: 145-55

18. Hoffman HJ, Taecholarn C, Hendrick EB, Humphreys RP. Management of lipomyelomeningoceles. Experience at the Hospital for Sick Children, Toronto. J Neurosurg. 1985. 62: 1-8

19. Hoving EW, Haitsma E, Oude Ophuis CM, Journee HL. The value of intraoperative neurophysiological monitoring in tethered cord surgery. Childs Nerv Syst. 2011. 27: 1445-52

20. Hsieh PC, Stapleton CJ, Moldavskiy P, Koski TR, Ondra SL, Gokaslan ZL. Posterior vertebral column subtraction osteotomy for the treatment of tethered cord syndrome: Review of the literature and clinical outcomes of all cases reported to date. Neurosurg Focus. 2010. 29: E6-

21. Huang SL, Shi W, Zhang LG. Surgical treatment for lipomyelomeningocele in children. World J Pediatr. 2010. 6: 361-5

22. . International Society of Ultrasound in O, Gynecology Education C. Sonographic examination of the fetal central nervous system: Guidelines for performing the ‘basic examination’ and the ‘fetal neurosonogram’. Ultrasound Obstet Gynecol. 2007. 29: 109-16

23. Kanev PM, Lemire RJ, Loeser JD, Berger MS. Management and long-term follow-up review of children with lipomyelomeningocele, 1952-1987. J Neurosurg. 1990. 73: 48-52

24. Kannu P, Furneaux C, Aftimos S. Familial lipomyelomeningocele: A further report. Am J Med Genet A. 2005. 132A: 90-2

25. Khealani B, Husain AM. Neurophysiologic intraoperative monitoring during surgery for tethered cord syndrome. J Clin Neurophysiol. 2009. 26: 76-81

26. Kokubun S, Ozawa H, Aizawa T, Ly NM, Tanaka Y. Spine-shortening osteotomy for patients with tethered cord syndrome caused by lipomyelomeningocele. J Neurosurg Spine. 2011. 15: 21-7

27. Kothbauer KF, Novak K. Intraoperative monitoring for tethered cord surgery: An update. Neurosurg Focus. 2004. 16: E8-

28. Kulkarni AV, Pierre-Kahn A, Zerah M. Conservative management of asymptomatic spinal lipomas of the conus. Neurosurgery. 2004. 54: 868-73

29. Muthukumar N. Congenital spinal lipomatous malformations: Part I--Classification. Acta Neurochir. 2009. 151: 179-88

30. Pang D, Zovickian J, Wong ST, Hou YJ, Moes GS. Surgical treatment of complex spinal cord lipomas. Childs Nerv Syst. 2013. 29: 1485-13

31. Pierre-Kahn A, Zerah M, Renier D, Cinalli G, Sainte-Rose C, Lellouch-Tubiana A. Congenital lumbosacral lipomas. Childs Nerv Syst. 1997. 13: 298-334

32. Tu A, Hengel AR, Cochrane DD. Radiographic predictors of deterioration in patients with lumbosacral lipomas. J Neurosurg Pediatr. 2016. 18: 171-6

33. Tubbs RS, Bui CJ, Rice WC, Loukas M, Naftel RP, Holcombe MP. Critical analysis of the Chiari malformation Type I found in children with lipomyelomeningocele. J Neurosurg. 2007. 106: 196-200

34. Wu HY, Kogan BA, Baskin LS, Edwards MS. Long-term benefits of early neurosurgery for lipomyelomeningocele. J Urol. 1998. 160: 511-4

35. Wykes V, Desai D, Thompson DN. Asymptomatic lumbosacral lipomas--A natural history study. Childs Nerv Syst. 2012. 28: 1731-9

Musculoskeletal neck pain in children and adolescents: Risk factors and complications

$
0
0

Jawad Fares, Mohamad Y. Fares, Youssef Fares
  1. Department of Neurosurgery, Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
  2. Faculty of Medicine, American University of Beirut, Beirut, Lebanon
  3. Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon

Correspondence Address:
Jawad Fares, Youssef Fares
Department of Neurosurgery, Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon

DOI:10.4103/sni.sni_445_16

Copyright: © 2017 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Jawad Fares, Mohamad Y. Fares, Youssef Fares. Musculoskeletal neck pain in children and adolescents: Risk factors and complications. 10-May-2017;8:72

How to cite this URL: Jawad Fares, Mohamad Y. Fares, Youssef Fares. Musculoskeletal neck pain in children and adolescents: Risk factors and complications. 10-May-2017;8:72. Available from: http://surgicalneurologyint.com/surgicalint-articles/musculoskeletal-neck-pain-in-children-and-adolescents-risk-factors-and-complications/

Date of Submission
10-Nov-2016

Date of Acceptance
09-Mar-2017

Date of Web Publication
10-May-2017

Abstract

Background:Neck pain is a major public health concern that has been extensively studied in adults but not in children and adolescents. Therefore, the purpose of this article is to explore musculoskeletal neck pain in children and adolescents, as well as to discuss its possible risk factors and complications.

Methods:Participants were patients under 18 years of age, who had presented to the clinic (Beirut, Lebanon) in 2015, with nonspecific neck pain. They were examined and asked to evaluate and localize the pain. Neck positioning during various activities along with other complications were explored. Patients reporting pain associated with congenital or systemic diseases and fractures were excluded.

Results:Two-hundred-and-seven children and adolescents presented with nonspecific neck pain. Musculoskeletal neck pain with spasm was diagnosed in 180 patients (N = 180). Participants did not show any findings on physical examination and radiological studies, and had no comorbidities. More females (57%) than males (43%) and more adolescents (60%) than children (40%) were affected. All the 180 participants (100%) reported flawed flexion of their back and neck while studying and/or using smartphones and tablets. Eye symptoms were reported in 21% of the cases, and parents of most participants (82%) reported a change in the psychological and social behavior of their children.

Conclusions:Musculoskeletal neck pain is an important disease in children and adolescents with numerous risk factors contributing to its development. Increased stresses regarding the cervical spine may lead to cervical degeneration along with other developmental, medical, psychological, and social complications.

Keywords: Adolescents, children, neck flexion, neck pain, smartphones, text neck

INTRODUCTION

Neck pain is a major public health problem in modern societies.[ 4 14 ] It can originate from any structure in the neck including intervertebral discs, ligaments, muscles, facet joints, dura, and nerve roots.[ 3 ] Potential causes can be tumors, infection, inflammatory diseases, and congenital disorders. In most cases, however, no systemic illness can be detected, and the complaint is labeled as musculoskeletal neck pain.[ 2 ]

Prevalence data have shown that, in a general population, the 1-year incidence of neck pain can be as high as 40%.[ 1 ] The World Health Organization (WHO) has ranked neck pain and other musculoskeletal diseases at 4th and 10th, respectively, among all health conditions for years lived with disability.[ 32 ] These conditions were also acknowledged as the key drivers of the increase in years lived with disability over the past 20 years.[ 32 ] In addition, data from the WHO Global Burden of Disease study showed that neck pain is the 8th ranked reason for most years lived with disability for 15–19 year olds of any health condition, which is higher than well-known adolescent public health problems such as asthma, alcohol use, drug use, and road injury.[ 21 ]

Although the epidemiology, burden, and treatment of musculoskeletal pain have been extensively explored in adults, the same is not true for children. The lack of clinical research pertinent to children and adolescents has been emphasized by multiple studies.[ 7 15 25 26 29 ] Emerging evidence shows that children, especially adolescents who report persistent pain, are at an increased risk of chronic pain as adults.[ 5 19 23 ] Moreover, many musculoskeletal illnesses follow a long-term pattern of recurring exacerbations and remissions, with the most consistent predictor of a new episode being the experience of a previous episode.[ 30 ]

As the surge in prevalence of musculoskeletal conditions occurs in childhood and adolescence, it may be necessary to investigate the condition at these stages of life.[ 24 ] Understanding aspects and risk factors surrounding the initial onset provides the best opportunity to develop efficacious treatments, and is central to any efforts at primary prevention.

Therefore, the purpose of this article is to explore musculoskeletal neck pain in children and adolescents under 18 years of age, and to discuss possible risk factors and complications related to this pain.

MATERIALS AND METHODS

This study explored cases that presented to our clinic in Beirut, Lebanon, in 2015 with nonspecific neck pain. Demographic information including age, sex, level of education, use of technology, and study habits were collected from individuals and their parents.

Interviewed individuals were under 18 years of age. Those whose ages ranged between 8 and 11 years were considered children, and those whose ages ranged between 12 and 17 years were considered adolescents. Patients were asked to localize the pain on themselves and on a model provided to them. Neurological assessment was conducted to test for sensory and motor deficits. Individuals were also asked about their pertinent daily habits, studying conditions, sitting positions, and sleeping positions. Further investigation explored the use of technology such as cell phones, tablets, computers, and television; participants were asked to show their usual neck position when engaged in these activities. All interviewees underwent radiological studies by radiography to rule out scoliosis or any other pathologies at the cervical spine level. In addition, psychological and social symptoms were investigated.

For the purposes of this study, we did not include patients reporting pain associated with congenital or systemic diseases, such as scoliosis. We also excluded patients reporting pain resulting from frank injuries, such as fractures, and pain following surgical interventions. Chronic neck pain was defined as continuous neck complaints for more than 6 months.

RESULTS

In total, 207 children and adolescents presented with nonspecific neck pain. All patients reported having cervical neck pain of more than 6 months’ duration that radiates dorsally down the back and to the shoulders. None had sensory or motor deficits. Twenty-seven patients were found to have dorsolumbar scoliosis and were excluded from the study. The remaining 180 patients did not show any finding on physical examination and radiological studies, and had no comorbidities. They were diagnosed with musculoskeletal neck pain with spasm, and were the focus of our study (N = 180). Ages ranged between 8 and 17 years, with a mean age of 14 years. Demographics of the children and adolescents participating in the study are presented in Table 1 .


Table 1

Demographics of the children and adolescents participating in the study (N=180)

 

All the 180 participants (100%) reported flawed flexion of their back and neck while studying. They also admitted to using smartphones and/or tablets. When asked to demonstrate how they used these devices, all participants (100%) showed strong flexion of the neck (≥45 degrees) when engaged in the activity. Children and adolescents in our sample spent an average of 5 and 7 hours a day, respectively, on their smartphones and handheld devices. Table 2 shows the pain sites, eye symptoms, and psychological and social effects that were found in the children and adolescents.


Table 2

Pain location, eye symptoms, and psychological and social effects in the children and adolescents participating in the study (N=180)

 

A change in behavior, defined by a change in daily habits and usual social interactions, as noticed by parents, was reported in 147 young participants (82%). They were described as having become more irritable and alienated. Parents of 115 participants (64%) reported that their grades in school have been declining.

DISCUSSION

Musculoskeletal neck pain in children and adolescents is very common. Almost 87% (180 of 207) of the children and adolescents presented in our study were diagnosed with musculoskeletal neck pain. Other studies and reviews have shown 1-year incidences of 28% and 40% for neck pain.[ 1 22 ]

All our participants reported flexing their back and neck while studying. Ariëns et al.[ 1 ] found a positive relationship between neck flexion and neck pain, suggesting an increased risk of neck pain for individuals studying with the neck at a minimum of 20° of flexion for more than 70% of the studying time. Moreover, all our participants showed strong flexion of the neck when using smartphones. “Text neck,” a 21st-century syndrome, is a term derived from the onset of cervical spinal degeneration resulting from the repeated stress of frequent forward head flexion while looking down at the screens of mobile devices and “texting” for long periods of time.[ 31 ] Text neck is becoming more common as more people, especially teens and adolescents, hunch over smartphones.[ 6 ] It is estimated that 75% of the world's population spends hours daily hunched over their handheld devices with their heads flexed forward.[ 19 ] In our sample, children and adolescents spent averages of 5 and 7 hours a day, respectively, with their heads tilted over reading and texting on their smartphones and handheld devices. Cumulatively, this is an average of 1825 and 2555 hours a year, respectively, of excess stresses seen in the cervical spine area. Hansraj adds that it is possible that a high school student may spend an extra 5000 hours in poor posture.[ 18 ]

The weight put on the spine dramatically increases when flexing the head forward at varying degrees [ Figure 1 ]. A full-grown head weighs 4.54 to 5.44 kg in the neutral position.[ 18 ] As the head tilts forward, the forces seen by the neck surges to 12.25 kg at 15°, 18.14 kg at 30°, 22.23 kg at 45°, and 27.22 kg at 60°.[ 18 ] The frequent forward flexion causes changes in the cervical spine, curvature, supporting ligaments, tendons, and musculature, as well as the bony segments, commonly causing postural change and pain felt in the neck and other associated areas.[ 31 ]


Figure 1

A chart depicting the stress and weight put on the neck and spine as a result of hunching over a smartphone and handheld devices at varying degrees. The neck flexion angle is the angle between the global vertical and the vector pointing from C7 to the occipitocervical joint. A full-grown head weighs 5 kg in the neutral position. As the head bends forward, the weight seen by the neck increases to 18 kg at 30° and 27 kg at 60°

 

The effects of forward flexion of the neck transcend pain to contribute to more associated complications. Often times, the effects of prolonged neck flexion can contribute to nearsightedness, eye strain, or dry eyes, as the eyes are forced to focus on an object placed nearby.[ 8 ] New research suggests a link between forward leaning postures that people use while texting, studying, surfing the web, emailing, and playing video games, and hyperkyphosis, which is associated with pulmonary disease and cardiovascular problems.[ 9 ] It is suggested that when someone drops their head and rounds their shoulders while looking at a smartphone or a tablet, it is harder for them to take a full breath because of the restriction to their muscles.[ 9 ] In addition, the ribs cannot move properly; thus, the heart and lungs cannot function to their full effectiveness.[ 9 ] Most times, children and adolescents do not know they could be doing serious long-term damage to their body because the short-term effects are not as noticeable. It is only in later life that the effects can seriously affect the quality of life. This increases fears that younger people, who are society's biggest users of smartphones and tablets, could be facing a future of pain and disability, or even taking years off of their life expectancy.

Emre et al.[ 10 ] indicate that computers, wireless internet, cell phones, and televisions emit an extremely low-frequency electromagnetic field. Electromagnetic radiation can cause difficulty sleeping, dizziness, headaches, tingling in the hands, ringing in the ears, eye pain, “unexplained” cardiac conditions, electrosensitivity, low immunity, attention deficit hyperactivity disorder, and autism.[ 20 ] When electrical properties are considered, the absorption of electromagnetic radiation through a child's head can be over two times greater, and absorption of the skull's bone marrow ten times greater, than in adults.[ 16 ]

Parents of most of our participants reported that their children have become more isolated and easily irritable. In addition, their grades in school have been negatively affected. Children who use more than the expert recommended 1–2 hours per day of technology, have a 60% increase in psychological disorders.[ 28 ] As mentioned previously, children and adolescents in our sample spent averages of 5 and 7 hours a day, respectively, on their smartphones and handheld devices. It is indicated that the more time students spend consuming media, and the more violent its contents are, the worse their grades in school, even when controlling for vital factors such as family, education, or immigrant background.[ 27 ] In addition, spending too many hours on handheld devices and smartphones will negatively affect children and adolescents’ communication skills, particularly with regard to face-to-face communication skills, bullying, and teasing.[ 17 ]

The lack of validated instruments to measure musculoskeletal neck pain and its consequences in children and adolescents is a limitation in similar studies. This is due to the fact that it becomes difficult to draw conclusions and compare between different samples, populations, and cultures. In addition, the method of administration is critical because self-administered instruments cannot be conducted among illiterate and young children, and the need for a parent to collect information may hinder the pain evaluation process.

Future studies should aim to find a unified instrument to measure musculoskeletal neck pain, and validation across cultures should occur. The pathways and mechanisms for the association of pain experienced in childhood and pain experienced in adulthood are unknown. It can be hypothesized that a physiological or behavioral trigger is set when a child has a particular painful experience, which predisposes them to pain as an adult. Genetic susceptibility may also underlie the experience of pain. Future studies can aim to explore these mechanisms. Moreover, there is a particular need for studies to investigate the nature of the relationships between neck pain and other adverse health risk factors.

CONCLUSIONS

Musculoskeletal neck pain is a common multifactorial disease in children and adolescents, implying that there are numerous risk factors contributing to its development. Bending the head, neck, and shoulders over cell phones and handheld devices, along with distorted neck positioning when sitting, studying, and watching television, can lead to incrementally increased stresses in the cervical spine area. These stresses may lead to early wear, tear, degeneration, and possibly surgeries. Other developmental, medical, psychological, and social complications are also of concern. While it is nearly impossible to avoid the habits and technologies that cause these issues, young individuals should make an effort to perform activities with a neutral spine and to avoid neck flexion for hours each day.[ 11 12 13 14 ] Cervical spine surgeons, pediatric neurologists, public health advocates, and social workers should run campaigns to raise awareness on musculoskeletal neck pain and its medical, psycholgical, and societal consequences, and methods of prevention and treatment.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Ariëns GAM, Borghouts JAJ, Koes BW, Crombie IK, Croft PR, Linton SJ, LeResche L, Korff M.editors. Neck pain. Epidemiology of pain. Seattle: IASP Press; 1999. p. 235-6

2. Bogduk N. Neck pain. Aust Fam Physician. 1984. 13: 26-30

3. Bogduk N. Neck pain: An update. Aust Fam Physician. 1988. 17: 75-

4. Bouter LM, van der Wal G. Psychosocial risk factors for neck pain: A systematic review. Am J Ind Med. 2011. 39: 180-93

5. Brattberg G. Do pain problems in young school children persist into early adulthood? A 13-year follow-up. Eur J Pain. 2004. 8: 187-99

6. Last accessed on 2016 Sep 01. https://health.clevelandclinic.org/2015/03/text-neck-is-smartphone-use-causing-your-neck-pain/.

7. Cohen E, Uleryk E, Jasuja M, Parkin PC. An absence of pediatric randomized controlled trials in general medical journals, 1985–2004. J Clin Epidemiol. 2007. 60: 118-23

8. Last accessed on 2016 Sep 01. http://www.correctvision.com/eye-health/effects-of-texting/.

9. Curtis SLast accessed on 2016 Sep 01. http://www.telegraph.co.uk/technology/mobile-phones/10721014/Texting-for-long-periods-could-lower-lifeexpectancy.html.

10. Emre M, Cetiner S, Zencir S, Unlukurt I, Kahraman I, Topcu Z. Oxidative stress and apoptosis in relation to exposure to magnetic field. Cell Biochem Biophys. 2011. 59: 71-7

11. Fares J, Al Tabosh H, Saadeddin Z, El Mouhayyar C, Aridi H. Stress, burnout and coping strategies in preclinical medical students. N Am J Med Sci. 2016. 8: 75-

12. Fares J, Fares Y. The role of yoga in relieving medical student anxiety and stress. N Am J Med Sci. 2016. 8: 202-

13. Fares J, Saadeddin Z, Al Tabosh H, Aridi H, El Mouhayyar C, Koleilat MK. Extracurricular activities associated with stress and burnout in preclinical medical students. J Epidemiol Glob Health. 2016. 6: 177-85

14. Fares Y. Conseils pour une Colonne Vertebrale Saine. Beirut. 2000. p.

15. Fares Y, Fares J. Neurosurgery in Lebanon: History, Development, and Future Challenges. World Neurosurg. 2017. p.

16. Gandhi OP, Morgan LL, de Salles AA, Han YY, Herberman RB, Davis DL. Exposure limits: The underestimation of absorbed cell phone radiation, especially in children. Electromagn Biol Med. 2012. 31: 34-51

17. Graham JB. Impacts of Text Messaging on Adolescents’ Communication Skills: School Social Workers’ Perceptions. Master of Social Work Clinical Research Papers, Paper 184, St. Catherine University, St. Paul and Minneapolis, Minnesota, US. 2013. p.

18. Hansraj KK. Assessment of stresses in the cervical spine caused by posture and position of the head. Surg Technol Int. 2014. 25: 277-9

19. Hestbaek L, Leboeuf-Yde C, Kyvik KO, Manniche C. The course of low back pain from adolescence to adulthood: Eight-year follow-up of 9600 twins. Spine. 2006. 31: 468-72

20. Hossmann KA, Hermann DM. Effects of electromagnetic radiation of mobile phones on the central nervous system. Bioelectromagnetics. 2003. 24: 49-62

21. Last accessed on 2016 Sep 01. http://vizhub.healthdata.org/gbd-compare/.

22. Jeffries LJ, Milanese SF, Grimmer-Somers KA. Epidemiology of adolescent spinal pain: A systematic overview of the research literature. Spine. 2007. 32: 2630-7

23. Jones GT, Silman AJ, Power C, Macfarlane GJ. Are common symptoms in childhood associated with chronic widespread body pain in adulthood?: Results from the 1958 british birth cohort study. Arthritis Rheum. 2007. 56: 1669-75

24. Kamper SJ, Henschke N, Hestbaek L, Dunn KM, Williams CM. Musculoskeletal pain in children and adolescents. Braz J Phys Ther. 2016. 20: 275-84

25. McBeth J, Jones K. Epidemiology of chronic musculoskeletal pain. Best Pract Res Clin Rheumatol. 2007. 21: 403-25

26. Michaleff ZA, Kamper SJ, Maher CG, Evans R, Broderick C, Henschke N. Low back pain in children and adolescents: A systematic review and meta-analysis evaluating the effectiveness of conservative interventions. Eur Spine J. 2014. 23: 2046-58

27. Mössle T, Kleimann M, Rehbein F, Pfeiffer C. Media use and school achievement–boys at risk?. Br J Dev Psychol. 2010. 28: 699-725

28. Page AS, Cooper AR, Griew P, Jago R. Children's screen viewing is related to psychological difficulties irrespective of physical activity. Pediatrics. 2010. 126: e1011-7

29. Rodgers A. Managing chronic pain in children and adolescents. BMJ. 2002. 324: 1570-6

30. Taylor JB, Goode AP, George SZ, Cook CE. Incidence and risk factors for first-time incident low back pain: A systematic review and meta-analysis. Spine J. 2014. 14: 2299-319

31. Last accessed on 2016 Aug 29. http://text-neck.com/.

32. Vos T, Barber RM, Bell B, Bertozzi-Villa A, Biryukov S, Bolliger I. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015. 386: 743-800

Chiari I malformation and syringomyelia in mucopolysaccharidosis type I (Hurler syndrome) treated with posterior fossa decompression: Case report and review of the literature

$
0
0

Vyacheslav Makler, Christina L. Goldstein, Daniel Hoernschemeyer, Tomoko Tanaka
  1. Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, Missouri, USA
  2. Department of Orthopedic Surgery, University of Missouri School of Medicine, Columbia, Missouri, USA

Correspondence Address:
Tomoko Tanaka
Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, Missouri, USA

DOI:10.4103/sni.sni_463_16

Copyright: © 2017 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Vyacheslav Makler, Christina L. Goldstein, Daniel Hoernschemeyer, Tomoko Tanaka. Chiari I malformation and syringomyelia in mucopolysaccharidosis type I (Hurler syndrome) treated with posterior fossa decompression: Case report and review of the literature. 26-May-2017;8:80

How to cite this URL: Vyacheslav Makler, Christina L. Goldstein, Daniel Hoernschemeyer, Tomoko Tanaka. Chiari I malformation and syringomyelia in mucopolysaccharidosis type I (Hurler syndrome) treated with posterior fossa decompression: Case report and review of the literature. 26-May-2017;8:80. Available from: http://surgicalneurologyint.com/surgicalint-articles/chiari-i-malformation-and-syringomyelia-in-mucopolysaccharidosis-type-i-hurler-syndrome-treated-with-posterior-fossa-decompression-case-report-and-review-of-the-literature/

Date of Submission
02-Dec-2016

Date of Acceptance
22-Feb-2017

Date of Web Publication
26-May-2017

Abstract

Background:Hurler Syndrome is the most severe phenotype of mucopolysaccharidosis type I. With bone marrow transplant and enzyme replacement therapy, the life expectancy of a child with Hurler syndrome has been extended, predisposing them to multiple musculoskeletal issues most commonly involving the spine.

Case Description:This is the case report of a 6-year-old male with Hurler syndrome who was diagnosed with Chiari I malformation and cervicothoracic syringomyelia on a preoperative magnetic resonance imaging (MRI) for his thoracolumbar kyphosis. This report details the successful management of a Chiari I malformation and syringomyelia with posterior fossa decompression in a child with Hurler syndrome.

Conclusion:Children born with MPS I can have complex spine issues that require surgical management. The most common orthopedic spinal condition for these patients, thoracolumbar kyphosis, requires evaluation with an MRI before performing surgery. This resulted in the diagnosis of a Chiari I malformation and syringomyelia in our patient with Hurler syndrome. This was successfully treated with decompression of the posterior fossa.

Keywords: Chiari I Malformation, Hurler syndrome, mucopolysaccharidosis type I, posterior fossa decompression, syrinx

INTRODUCTION

Mucopolysaccharidosis type I (MPSI) is an autosomal recessive lysosomal storage disease caused by deficient or absent activity of the α-L-iduronidase enzyme (IDUA), which catalyzes the degradation of the glycosaminoglycans (i.e., dermatan and heparan sulfates), the most severe form of which is Hurler syndrome (HS).[ 25 ] Without treatment, patients with HS suffer from multisystem manifestations including mental retardation, skeletal deterioration, severe cardiopulmonary disease, hepatosplenomegaly, visual impairment, and deafness, usually leading to death within the first decade of life.[ 8 ] The advent of allogeneic hematopoietic stem cell transplantation from bone marrow, peripheral blood, or unrelated umbilical cord has resulted in the reversal of organomegaly, preservation of neurocognitive development, and improved hearing, vision, and cardiopulmonary function in most transplanted patients.[ 1 8 9 13 18 20 21 ] With improvement in treatment, not only is the life expectancy of a child with HS increased, so is the risk of development of other medical conditions. To our knowledge, there have been only two papers published in the English medical literature describing syringomyelia in patients with mucopolysaccharidosis type II (Hunter syndrome)[ 14 ] and type VI (Maroteaux–lamy syndrome).[ 10 ] There have been no reports on the diagnosis and management of a Chiari I malformation (CM-I) and syringomyelia in a patient with HS. In this manuscript, we present a case of a 6-year-old child with HS who was diagnosed with CM-I and a cervicothoracic syrinx during a preoperative magnetic resonance imaging (MRI) for the surgical management of his thoracolumbar kyphosis.

CASE REPORT

The patient is a 6-year-old male who was referred to a pediatric clinic for an incidentally found CM-I and cervicothoracic syrinx [ Figure 1 ] identified during a preoperative workup prior to the surgical management of a progressive thoracolumbar kyphosis [ Figure 2 ]. The child has been experiencing daily headaches, but denied dysphagia, neck pain, or numbness or weakness in the upper extremities. His family also noted difficulty with hand coordination and strength when compared to his peers. Born via caesarian section at 41 weeks with a birth weight of 8 pounds and 12 ounces, he was diagnosed with HS and underwent a bone marrow transplant at the age of 14 months.


Figure 1

Preoperative MRI findings. (a) MRI brain, T1WI sagittal view. Demonstrates cerebellar ectopia, measuring approximately 5.9 mm with crowding within the foramen magnum. (b) MRI brain, T2WI coronal view. Demonstrates prominent retrocerebellar cystic space. The differential diagnosis includes major cisterna magna versus an arachnoid cyst. (c) MRI cervicothoracic spine, T2WI sagittal view. Demonstrates 8 mm syrinx from C5 to T1-2. (d) MRI cervicothoracic spine, T2WI axial view at the level of C6-7

 

Figure 2

Spinal Imaging. (a) MRI thoracolumbar spine, sagittal view. (b) CT thoracolumbar spine, sagittal view. Both demonstrate severe gibbus deformity centered at L1 causing moderate to severe bony spinal canal stenosis

 

On physical examination, the patient was noted to be alert and oriented with clear speech and normal cranial nerve function. Muscle strength was 5/5 in bilateral biceps, triceps, and deltoids, 4+/5 in hand grip and finger abduction. Deep tendon reflexes were grade 2/4 in the upper and lower extremities with a negative Hoffman's sign bilaterally, no ankle clonus, and a downgoing Babinski test bilaterally. The patient was unable to perform single-leg stance.

On August 4, 2015 the patient was taken to the operating room where he underwent a successful suboccipital craniectomy measuring 3 × 3 cm, C1 laminectomy, intradural exploration with coagulation of cerebellar tonsils, lysis of arachnoid adhesions, resection of thick posterior arachnoid membrane, and duraplasty measuring 3 × 3 cm utilizing Dura-Guard™ (Baxter Healthcare Corporation, Mountain Home, Arizona) and DuraSeal™ (Medtronic, Minneapolis, Minnesota). Intraoperatively, he was noted to have numerous arachnoid adhesions, an arachnoid cyst, and cerebellar tonsillar herniation to the level of C1 was confirmed. Postoperative course was unremarkable, and he was ultimately discharged home on postoperative day 3.

At his first 2-week postoperative follow-up, his mother reported more stability with his gait and improved balance. The patient returned to school. The patient's physical examination was unchanged from his preoperative exam except that he was now able to perform a single-leg stance. At his 3-month follow-up visit, his parents reported that he continued to improve. His daily headaches had resolved, and the MRI of the cervicothoracic spine revealed improvement in the size of the syrinx measuring 3.5 mm at its widest point [ Figure 3 ].


Figure 3

3-month postoperative MRI. (a) MRI cervicothoracic spine, T2 WI sagittal view. Demonstrates improvement in syrinx from C5 to T1-2. (b) MRI cervicothoracic spine, T2 WI axial view at the level of C6-7

 

Six months after suboccipital craniectomy, the patient underwent an uncomplicated anterior release and posterior spinal fusion with correction of his thoracolumbar kyphosis. During the most recent 3-month follow-up visit, the patient was found to have complete resolution of his gibbus deformity and no changes with his neurologic exam. The MRI of the cervical and thoracic spine revealed a stable syrinx. This case report was approved by the University of Missouri Health Sciences Institutional Review Board, and an informed consent was obtained from the patient's parents.

DISCUSSION

In 1891, Hans Chiari documented three cases of congenital defects of the rhombencephalon, classified as type I, II, and III.[ 6 ] The most common type is CM-1, which is present in 0.56–1% of the population.[ 16 ] The radiologic diagnosis of CM-I is best made on cranial midsagittal MRI studies, with cerebellar tonsil herniation of at least 3 mm below the basion-opisthion line suggesting the condition [ Table 1 ].[ 3 11 15 ] The symptoms of CM-I include head, neck, and back pain, cape pain (shoulders), nonradicular limb pain, weakness, paresthesias, vestibular symptoms, diplopia, tinnitus, hearing loss, syncope, slurred speech, dysphagia, urinary incontinence, and sleep disturbance.[ 16 ] A syrinx is present in 30–70% of cases of CM-I.[ 15 ]


Table 1

Diagnostic quick reference for Chiari I malformation and variants. This table is reproduced with the permission of the authors and the Neurologic Clinics

 

MPSI is an autosomal recessive lysosomal storage disease caused by mutation in the IDUA gene located on chromosome 4p16.3,[ 12 ] resulting in deficient or absent activity of the IDUA, which catalyzes the degradation of the glycosaminoglycans (i.e., dermatan and heparan sulfates).[ 25 ] These molecules can be found in free form in the extracellular matrix or as part of the structure of different types of proteoglycans, with important functions both in the structure of tissues and intercellular communication. Intralysosomal accumulation of these substrates results in pathological processes that produce a progressive dysfunction resulting in multiorgan deterioration that includes hepatosplenomegaly, dysostosis multiplex, short stature, coarse facial features, corneal clouding, joint contractures, umbilical hernias, failure to thrive, intellectual disability, and developmental delay.[ 5 12 ] The extensive storage of these glycosaminoglycans is also known to cause meningeal thickening.[ 4 ] Intraoperatively, our patient was noted to have thickened arachnoid membrane, which was biopsied and sent for pathological assessment. The sample did not reveal any intracellular storage of Luxol Fast Blue or Periodic Acid-Schiff positive contents, which neither supports nor refutes the clinical diagnosis of mucopolysaccharidosis.

Historically, MPSI has been divided into three clinical subtypes – Hurler (severe), Scheie (mild/attenuated), and Hurler–Scheie (intermediate).[ 1 ] This classification is based on clinical factors such as the age of onset, the rate of functional deterioration, and the range of affected organs (i.e., CNS involvement).[ 1 ] HS is the most severe phenotype in the spectrum of MPSI, with a prevalence of approximately 0.69 cases per 100,000 births.[ 2 ] Diagnosis of MPSI is based on IDUA enzyme analysis in leukocytes or dried blood spots followed by molecular confirmation of the IDUA gene mutations in individuals with low enzyme activity.[ 12 ]

HS progresses rapidly from 6 to 24 months resulting in significant multiorgan dysfunction.[ 5 ] Historically, the natural history of HS involved death before the age of 10 due to respiratory complications or cardiomyopathy.[ 17 ] Because new therapies such as allogeneic hematopoietic stem cell transplantation became a viable treatment option, reversal of organomegaly, preservation of neurocognitive development, and improved hearing, vision, and cardiopulmonary function in most transplanted patients have been observed.[ 1 8 9 13 18 20 21 ] To our knowledge, there have been only two papers published in the English language medical literature describing the diagnosis and treatment of syringomyelia in mucopolysaccharidosis patients. No one has reported this diagnosis in a patient with HS.

Manara et al.,[ 14 ] reported a case of CM-I and holocord syringomyelia in a patient who was diagnosed with Hunter syndrome at the age of 18 months. Patient was ultimately treated with enzyme replacement therapy with idursulfase. A brain MRI revealed enlarged cisterna magna along with cerebellar tonsils ectopia consistent with CM-I. MRI of the cervical spine failed to reveal a syringomyelia, whereas a repeat MRI at the age of 5 years revealed a focal thin syrinx. The child only complained of upper limbs numbness and loss of sphincter control without any other neurological deficits. A follow-up MRI 1 year later showed a holocord syrinx extending from the cervicomedullary junction to the conus medullaris. The child underwent posterior fossa decompression (PFD) for CM-I. At 7-months follow-up preoperative symptoms had resolved, whereass the MRI demonstrated significant decreased in size of the syrinx.

Hite et al.,[ 10 ] reported the case of a 6-month-old boy who initially presented for evaluation of hepatosplenomegaly and increased head circumference. He was diagnosed with mucopolysaccharidosis type VI (Maroteaux–lamy syndrome) and treated with allogeneic bone marrow transplantation at the age of 14 months. Pretransplant spinal MRI was essentially normal, however, by 4 years of age a follow-up MRI revealed a holocord syringomyelia from C2 to L1. The syrinx remained stable on serial MRIs and the patient continued to have no focal motor or sensory abnormalities. Thus, no neurosurgical intervention was performed.

In 2003, Zafeiriou et al.,[ 26 ] summarized typical brain and spine MRI findings in patients with mucopolysaccharidosis. It was noted that the most prominent brain features identified in almost all types of mucopolysaccharidosis were white and gray matter changes, ventriculomegaly and hydrocephalus, cortical atrophy, and enlargement of the perivascular spaces. Spinal MRIs usually revealed canal stenosis and cord compression with spinal cord signal changes [ Table 2 ].[ 26 ] Looking specifically at the posterior fossa, mega cisterna magna was the most common radiologic finding. Ultimately, the correlation between imaging findings and the disease severity remains unclear. Intraoperatively, in our patient, the posterior fossa thickened membrane versus arachnoid cyst appeared to be causing compression on the surrounding tissue, which may have played a role in the tonsillar ectopia.


Table 2

Overview of the most frequent brain and spinal MRI abnormalities identified in MPS according to disease type. This table is reproduced with the permission of the authors and the American Journal of Neuroradiology

 

Tandon et al.,[ 22 ] published a case series among 12 patients with HS with a mean of 4.5 years follow-up after undergoing bone marrow transplantation. High lumbar kyphosis was noted in 10 patients, which was associated with thoracic scoliosis in one, whereas isolated thoracic scoliosis was seen in another. One patient did not have any significant problems in the thoracic or lumbar spine but had odontoid hypoplasia, which was also seen in three other children. Four of the 8 patients in whom MRI of the cervical spine had been performed had abnormal soft tissue around the tip of the odontoid. Neurological problems were only seen in two patients. In one it was caused by cord compression in the lower dorsal spine 9.5 years after posterior spinal fusion for progressive kyphosis, and in the other by angular kyphosis with thecal indentation in the high thoracic spine associated with symptoms of spinal claudication.

PFD has long been performed to relieve compression and restore normal CSF pathways at the craniocervical junction.[ 24 ] A survey on surgical treatment of CM-I with syringomyelia conducted by the American Society of Pediatric Neurosurgeons showed that 85% of the respondents perform PFD as first-line treatment, whereas less than 3% offer syrinx drainage as first-line therapy.[ 7 19 ] In addition, routine practice consisted of bony decompression alone for 7%, decompression with duraplasty in 36%, and additional tonsil reduction for 27%.[ 7 ] Xie et al.[ 24 ] examined 87 patients aged 5–18 years who had undergone PFD. They noted 72.4% improvement of symptoms at final follow-up. In 90.8% of the cases, significant syrinx resolution was also noted. Wu et al.[ 23 ] reported that typically the syrinx resolved within 6 months after PFD. In their retrospective review of patients with CM-I who had undergone PFD, Chotai et al.[ 7 ] noted that 87% of the patients indicated they would choose to undergo the surgery again.

CONCLUSION

MPSI is an autosomal recessive lysosomal storage disorder causing a chronic, progressive multiorgan disease by deficient or absent activity of the α-L-iduronidase enzyme, which catalyzes the degradation of glycosaminoglycans.[ 25 ] Bone marrow transplant and enzyme replacement therapy has led to an increased life expectancy for this once fatal disease. Patients with HS are now at an increased risk of developing other medical conditions associated with their disease, including progressive TL kyphosis, symptomatic carpal tunnel syndrome, angular deformity of the lower limbs, and hip dysplasia that may require surgical treatment. Diagnosing and properly managing a CM-I and syrinx in this patient population is necessary to avoid neurologic complications and spinal cord injury that could occur from anesthesia and surgical management of HS patients. Though these three reports represent a small case series, given the fact that all three patients developed a syrinx on serial imaging, and two of the three demonstrating neurologic abnormalities, we would suggest that patients with mucopolysaccharidosis require routine follow-up for clinical assessments with spinal imaging as indicated by history and physical examination.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Aldenhoven M, Boelens JJ, de Koning TJ. The clinical outcome of Hurler syndrome after stem cell transplantation. Biol Blood Marrow Transplant. 2008. 14: 485-98

2. Baehner F, Schmiedeskamp C, Krummenauer F, Miebach E, Bajbouj M, Whybra C. Cumulative incidence rates of the mucopolysaccharidoses in Germany. J Inherit Metab Dis. 2005. 28: 1011-7

3. Barkovich AJ, Wippold FJ, Sherman JL, Citrin CM. Significance of cerebellar tonsillar position on MR. AJNR Am J Neuroradiol. 1986. 7: 795-9

4. Boor R, Miebach E, Bruhl K, Beck M. Abnormal somatosensory evoked potentials indicate compressive cervical myelopathy in mucopolysaccharidoses. Neuropediatrics. 2000. 31: 122-7

5. Campos D, Monaga M. Mucopolysaccharidosis type I: Current knowledge on its pathophysiological mechanisms. Metab Brain Dis. 2012. 27: 121-9

6. Chiari H. Concerning alterations in the cerebellum resulting from cerebral hydrocephalus. 1891. Pediatr Neurosci. 1987. 13: 3-8

7. Chotai S, Kshettry VR, Lamki T, Ammirati M. Surgical outcomes using wide suboccipital decompression for adult Chiari I malformation with and without syringomyelia. Clin Neurol Neurosurg. 2014. 120: 129-35

8. Coletti HY, Aldenhoven M, Yelin K, Poe MD, Kurtzberg J, Escolar ML. Long-term functional outcomes of children with hurler syndrome treated with unrelated umbilical cord blood transplantation. JIMD Rep. 2015. 20: 77-86

9. Guffon N, Souillet G, Maire I, Straczek J, Guibaud P. Follow-up of nine patients with Hurler syndrome after bone marrow transplantation. J Pediatr. 1998. 133: 119-25

10. Hite SH, Krivit W, Haines SJ, Whitley CB. Syringomyelia in mucopolysaccharidosis type VI (Maroteaux-Lamy syndrome): Imaging findings following bone marrow transplantation. Pediatr Radiol. 1997. 27: 736-8

11. Jayarao M, Sohl K, Tanaka T. Chiari malformation I and autism spectrum disorder: An underrecognized coexistence. J Neurosurg Pediatr. 2015. 15: 96-100

12. Johnson BA, Dajnoki A, Bodamer OA. Diagnosing lysosomal storage disorders: Mucopolysaccharidosis type i. Curr Protoc Hum Genet. 2015. 84: 11-8

13. Malm G, Gustafsson B, Berglund G, Lindstrom M, Naess K, Borgstrom B. Outcome in six children with mucopolysaccharidosis type IH, Hurler syndrome, after haematopoietic stem cell transplantation (HSCT). Acta Paediatr. 2008. 97: 1108-12

14. Manara R, Concolino D, Rampazzo A, Zanetti A, Tomanin R, Faggin R. Chiari 1 malformation and holocord syringomyelia in hunter syndrome. JIMD Rep. 2014. 12: 31-5

15. McVige JW, Leonardo J. Imaging of Chiari type I malformation and syringohydromyelia. Neurol Clin. 2014. 32: 95-126

16. McVige JW, Leonardo J. Neuroimaging and the clinical manifestations of Chiari Malformation Type I (CMI). Curr Pain Headache Rep. 2015. 19: 18-

17. Moore D, Connock MJ, Wraith E, Lavery C. The prevalence of and survival in Mucopolysaccharidosis I: Hurler, Hurler-Scheie and Scheie syndromes in the UK. Orphanet J Rare Dis. 2008. 3: 24-

18. Peters C, Balthazor M, Shapiro EG, King RJ, Kollman C, Hegland JD. Outcome of unrelated donor bone marrow transplantation in 40 children with Hurler syndrome. Blood. 1996. 87: 4894-902

19. Rocque BG, George TM, Kestle J, Iskandar BJ. Treatment practices for Chiari malformation type I with syringomyelia: Results of a survey of the American Society of Pediatric Neurosurgeons. J Neurosurg Pediatr. 2011. 8: 430-7

20. Shapiro EG, Lockman LA, Balthazor M, Krivit W. Neuropsychological outcomes of several storage diseases with and without bone marrow transplantation. J Inherit Metab Dis. 1995. 18: 413-29

21. Souillet G, Guffon N, Maire I, Pujol M, Taylor P, Sevin F. Outcome of 27 patients with Hurler's syndrome transplanted from either related or unrelated haematopoietic stem cell sources. Bone Marrow Transplant. 2003. 31: 1105-17

22. Tandon V, Williamson JB, Cowie RA, Wraith JE. Spinal problems in mucopolysaccharidosis I (Hurler syndrome). J Bone Joint Surg Br. 1996. 78: 938-44

23. Wu T, Zhu Z, Jiang J, Zheng X, Sun X, Qian B. Syrinx resolution after posterior fossa decompression in patients with scoliosis secondary to Chiari malformation type I. Eur Spine J. 2012. 21: 1143-50

24. Xie D, Qiu Y, Sha S, Liu Z, Jiang L, Yan H. Syrinx resolution is correlated with the upward shifting of cerebellar tonsil following posterior fossa decompression in pediatric patients with Chiari malformation type I. Eur Spine J. 2015. 24: 155-61

25. Yang JS, Min HK, Oh HJ, Woo HI, Lee SY, Kim JW. A simple and rapid method based on liquid chromatography-tandem mass spectrometry for the measurement of alpha-L-iduronidase activity in dried blood spots: An application to mucopolysaccharidosis I (Hurler) screening. Ann Lab Med. 2015. 35: 41-9

26. Zafeiriou DI, Batzios SP. Brain and spinal MR imaging findings in mucopolysaccharidoses: A review. AJNR Am J Neuroradiol. 2013. 34: 5-13

mTOR activation is increased in pilocytic astrocytomas from older adults compared with children

$
0
0

Mahlon D. Johnson, Mary O’Connell, Kevin Walter, Howard Silberstein
  1. Department of Pathology, Division of Neuropathology, University of Rochester School of Medicine, Rochester, New York, USA
  2. Department of Neurosurgery, University of Rochester School of Medicine, Rochester, New York, USA

Correspondence Address:
Mahlon D. Johnson
Department of Neurosurgery, University of Rochester School of Medicine, Rochester, New York, USA

DOI:10.4103/sni.sni_367_16

Copyright: © 2017 Surgical Neurology International This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Mahlon D. Johnson, Mary O’Connell, Kevin Walter, Howard Silberstein. mTOR activation is increased in pilocytic astrocytomas from older adults compared with children. 26-May-2017;8:85

How to cite this URL: Mahlon D. Johnson, Mary O’Connell, Kevin Walter, Howard Silberstein. mTOR activation is increased in pilocytic astrocytomas from older adults compared with children. 26-May-2017;8:85. Available from: http://surgicalneurologyint.com/surgicalint-articles/mtor-activation-is-increased-in-pilocytic-astrocytomas-from-older-adults-compared-with-children/

Date of Submission
14-Sep-2016

Date of Acceptance
20-Feb-2017

Date of Web Publication
26-May-2017

Abstract

Background:Recent studies suggest that the behavior and biology of WHO grade I pilocytic astrocytomas (PAs) in adults is different than that associated with grade I PAs in children.

Methods:We evaluated Ki-67 labeling, BRAF abnormalities, isocitrate dehydrogenase R132 immunoreactivity phosphorylation (activation) of p44/42 mitogen activated protein kinase (MAPK), and mammalian target of rapamycin (mTOR) in formalin-fixed tissue from 21 adult (18 years or older, mean age 37 years) and 10 children (mean age 9.4 years) WHO grade I PAs.

Results:The mean Ki-67 labeling was 4.8% in adults and 3.8% in children. There was no significant difference between Ki-67 labeling in children and adults or either subgroups of adults. No differences were found in phospho p44/42MAPK in adult subgroups (18–33 years and 34 and older) compared to children. Activation/phosphorylation of mTOR was biphasic in adults being significantly lower than children in young adults but significantly higher than children in older adults (age 34 and older).

Conclusions:Identifying mTOR phosphorylation/activation may represent a difference in biology and a new marker to guide chemotherapy with recently approved mTOR inhibitors.

Keywords: mTOR, pilocytic astrocytoma, p44/42MAPK

INTRODUCTION

A recent study suggests that the recurrence rate for pilocytic astrocytomas (PA) in adults may be significantly higher than that in children. Progression after gross total resection was found to be approximately 40% in adults compared to relatively infrequent in children.[ 22 ] The molecular differences promoting recurrence likely involve growth regulatory kinase cascades,[ 2 4 13 ] however, this has not been extensively evaluated in adult PAs.

Activation of the p44/42 mitogen activated protein kinase (MAPK) has been implicated in several neoplasias[ 5 13 14 ] and is thought to be important in the pathogenesis of PAs in children.[ 2 10 ] Activation of the MAPK pathway appears to be, in some cases, by activation of upstream BRAF by the BRAF tandem duplication KIAA 1549 fusion or a mutation at BRAF V600E.[ 2 10 11 ] These result in the activation of the MAPK kinase (Raf-1)–MAP kinase/ERK kinase (MEK-1)-p44/42 MAPK cascade. BRAF activation also results in the activation of mammalian target of rapamycin (mTOR) by the phosphoinositide 3 kinase (PI3K)-protein kinase PKB/Akt–mTOR pathways in part by activation through the p44/42MAPK kinase.[ 7 ] Activation of the RAF1-MEK1 p44/42MAPK and PI3K-Akt-mTOR and other kinase cascades that promote cell proliferation and inhibit apoptosis[ 15 16 20 21 ] have been studied along with many types of gliomas, but have not been extensively evaluated in adult PAs.

MATERIALS AND METHODS

Pilocytic astrocytoma tissue

Thirty-one formalin-fixed paraffin embedded World Health Organization (WHO) grade I PAs from 21 adults (mean age 37 years, adults range 18–70, 10 females and 11 males) and 10 children, mean age 9.4 years (9 females and 1 male), were identified in the University of Rochester Medical Center archives and consultations with Institutional Review Board approval from 2008 to 2015 and reviewed following WHO criteria and recent findings.[ 3 4 ] In adult cases, the distribution of cases based on age shows a distinct clustering of cases in young and those in older adults. Consequently, adult cases were grouped as those 18 to 33 and those older than 33. Characteristics are listed in Table 1 .


Table 1

Pilocytic astrocytomas characteristics and findings

 

Immunohistochemistry for phospho-p44/42MAPK and phospho- mTOR in pilocytic astrocytomas

Each case was analyzed with a polyclonal antibody to phospho p44/42 MAPK (Thr 202/Tyr 204, 1:400) and human phospho-mTOR (Ser 2448, 1:100 Cell Signaling, Beverly MA.) and MAC4 universal HRP-polymer (Biocare) with diaminobenzidene (DAB) chromagen and hematoxylin counterstain (Biocare), as described previously.[ 9 ] For antigen retrieval, tissue sections were incubated in a thermoresistant chamber with X Reveal Decloaker (Biocare Medical, Concord CA) at 120–123°C and pressure of 20–24 psi, according to manufacturer's specifications. Immunoreactivity in tumor cells (excluding blood vessels) was assessed by two of us as “0” for no distinct immunoreactivity, “1+” if 1–30% of cells were immunoreactive, “2+” if 30–50%, and “3+” if greater than 50% of cells were positive. Scores were analyzed by two-way t-tests comparing cumulative scores between young and adults, young and adult subgroups, and between 18 and 33 versus patients older than 33 years of age.

RESULTS

Comparison of pathological features of pilocytic astrocytomas in adults and children

The mean Ki-67 labeling index was 4.8% in adults and 3.8% in children. The Ki-67 labeling was not significantly different in either the 18–33 or 34 and older adults compared to children, and was not statistically different between the subgroups in adults. Neither adults nor children showed IDHR132 immunoreactivity. None of the 6 adults analyzed had a BRAFv600E mutation but one showed the BRAF-KIAA fusion. Two of the 4 tumors in children had BRAF mutations [ Table 1 ].

Phospho-p44/42MAPK and phospho-mTOR immunoreactivity in pilocytic astrocytomas

As summarized in Table 1 the mean immunoreactivity for phospho p44/42 MAPK was 1.1 ± 0.93 for children, 0.8 ± 1.03 SD for 18–33-year olds and 1.11 for 34–70-year olds. This was not statistically different comparing younger and older adults and children.

Phospho-mTOR immunoreactivity had a mean of 1.37 ± 1.18 SD in children (< 18 years of age, 0.33 ± 0.487 in young adults (18–33) and 1.36 ± 1.28 in adults 34 or greater years of age. Phospho-mTOR was biphasic in adults being significantly lower than children in young adults (P = 0.05) but significantly higher than children in older adults (age 34–70 years) (P = 0.05) [ Figure 1 ].


Figure 1

Phospho-MAPK 44/42 (a) phospho-mTOR (b), immunohistochemical control (c) in adult pilocytic astrocytoma (case 21) (Hematoxylin counterstain. Diaminobenzidine chromagen, original magnification ×400)

 

DISCUSSION

The BRAF v600E mutation was found in only 1 of the 6 cases analyzed, which is consistent with previous reports that BRAF v600E mutations are rare in adults.[ 2 4 ] Similarly, BRAF fusion appears less common in adult PAs.[ 4 6 ]

Two studies suggest that the MEK-1-p44/42 MAPK pathway is involved in the pathogenesis of childhood PAs.[ 2 10 ] Our findings suggest that p44/42 MAPK phosphorylation/activation occurs in many Pas, however, this is not significantly different between adult and childhood WHO grade I PAs. In contrast, in children, this activation appears associated with anaplastic progression.[ 18 ] In addition, activation of p44/42 MAPK also influences oncogene-induced senescence in Pas.[ 8 17 ] The role of this pathway in tumor behavior warrants additional review after longer follow-up.

The PI3K- PKB/Akt-mTOR pathway influences several functions central to neoplasia including cell proliferation and metabolism.[ 1 21 24 ] mTOR is a serine/threonine kinase present in two compositionally distinct complexes with different functions. The mTOR complex 1 (mTORC1) contains PRAS40, and is activated by numerous extracellular growth factors and cellular mitogens.[ 23 ] Activation of mTORC1 phosphorylates S6 kinase stimulating a number of growth-related functions including protein synthesis and cell proliferation. In contrast, mTORC2 is associated with cell metabolism and cytoskeletal organization. mTORC1 is strongly activated by rapamycin.[ 24 ] The effectiveness and side effect profiles of various mTOR inhibitors depends on their relative inhibition of the more growth regulatory mTORC1 compared to mTORC2.[ 1 21 ]

The PI3K- PKB/Akt-mTOR pathway is also activated in many WHO grade I adult PAs, and is significantly higher in older adults compared to children. This raises the possibility that activation of this pathway may participate in the pathogenesis of PAs in the older population. Activation of mTOR is also increased in anaplastic PAs.[ 18 ] Consequently, drugs inhibiting mTOR activation may represent a viable chemotherapy for recurrent PAs in this population. Nonetheless, identifying the optimal therapy may require considerable study because several recently developed rapamycin analogues show different activities for mTORC1 and mTORC2.[ 1 ] Rapamycin (sirolimus) is a highly effective inhibitor of mTORC1 and associated components of the PI3K-PKB/Akt-mTOR pathway. Nonetheless, it has limited effectiveness in tuberous sclerosis, except possibly in some angiomyolipomas. This may reflect secondary activation of mTORC2 and Akt.[ 1 ] Everolimus has been approved for the treatment of tuberous sclerosis subependymal giant cell astrocytomas, neuroendocrine carcinomas of the pancreas, and advanced renal carcinomas.[ 1 12 ] More recently developed orally available analogues such as temsirolimus is currently approved for other malignancies such as mantle cell lymphoma and renal carcinoma.[ 1 ] Because of the secondary activation of other pathways, use of combination therapy with other kinase inhibitors may be more effective.[ 1 ]

Phosphorylation of p44/42MAPK and mTOR have been found to be less stable with cold ischemia after surgical removal than some other phosphoproteins.[ 23 ] Nonetheless, our tissues are rapidly placed in formalin (typically in less than 30 minutes). Moreover, in our recent studies on meningiomas, phospho p44/42 MAPK was consistently found in almost 10% of the cases by immunohistochemistry and western blot.[ 9 ]

In summary, phosphorylation/activation of mTOR appears increased, particularly in PAs in adults 34 years of age and older. Identifying mTOR phosphorylation/activation likely represents a new marker to guide chemotherapy because several rapamycin analogues that inhibit mTOR activation are now in clinical trials or FDA approved.[ 1 12 19 ]

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

1. Benjamin D, Colombi M, Moroni C, Hall MN. Rapamycin passes the torch: A new generation of mTOR inhibitors. Nature. 2011. 10: 868-80

2. Brokinkel B, Peetz-Dienhart S, Ligges S, Brentrup A, Stummer W, Paulus W. A comparative analysis of MAPK pathway hallmark alterations in pilocytic astrocytomas: Age related and mutually exclusive. Neuropathol Appl Neurobiol. 2015. 41: 258-61

3. Collins VP, Tihan T, Tihan T, Vandenberg SR, Burger PC, Hawkins C, Louis DN, Ohgaki H, Wiestler OD, Cavenee WK.editors. Pilocytic astrocytoma. Tumours of the Nervous System. Geneva, Switzerland: WHO Press; 2016. p. 80-8

4. Cykowki MD, Allen RA, Kabaly AC, Fung KM, Marshall R, Perry A. The differential diagnosis of pilocytic astrocytoma with atypical features and malignant glioma: An analysis of 16 cases with emphasis on distinguishing molecular features. J Neurooncol. 2013. 115: 477-86

5. Enralp Y, Derin D, Ozluk Y, Yavuz E, Guney E, Saip P. MAPK overexpression is associated with anthracycline resistance and increased risk for recurrence in patients with triple-negative breast cancer. Ann Oncol. 2008. 19: 669-74

6. Hasselblatt M, Riesmeier B, Lechtape B, Brentrup A, Stummer W, Albert FK. BRAF-KIAA 1549 fusion transcripts are less frequent in pilocytic astrocytomas diagnosed in adults. Neuropathol Appl Neurbiol. 2011. 37: 803-6

7. Gutmann DH, Chen YH. The molecular and cell biology of pediatric low grade gliomas. Oncogene. 2014. 33: 2019-26

8. Jacob K, Quang-Khuong DA, Jones DT, Witt H, Lambert S, Albrecht S. Genetic alterations leading to MAPK pathway activation mediate oncogene-induced senescence in sporadic pilocytic astrocytomas. Clin Cancer Res. 2011. 17: 4650-60

9. Johnson MD, O’Connell M, Vito F, Bakos RS. Increased STAT-3 and synchronous activation of Raf-1-MEK-1-MAPK, and phosphatidylinositol 3-Kinase-Akt-mTOR pathways in atypical and anaplastic meningiomas. J Neuro-Oncol. 2009. 92: 129-35

10. Jones DTW, Gromuch J, Lichter P, Witt O, Pfister SM. MAPK pathway activation in pilocytic astrocytoma. Cell Mol Life Sci. 2012. 69: 799-81

11. Jones DTW, Hutter B, Jager N, Korshunov A, Kool M, Warnatz HJ. Reccurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet. 2013. 45: 927-32

12. Kieran M, Yao X, Macy M. A prospective multi-institutional Phase II study of everolimus (Rad001), an mTOR inhibitor in pediatric patients with recurrent or progressive low-grade gliomas. Ped Blood Cancer. 2013. 60: 300-10

13. McCubrey JA, Steelman LS, Chappell WH, Sun L, Davis NM, Abrams SL. Advances in targeting signal transduction pathways. Oncotarget. 2012. 3: 123456-1521

14. Nan Y, Yang S, Tian Y, Zhang W, Zhou B, Bu L, Huo S. Analysis of the expression protein profiles of lung squamous carcinoma cell using shot-gun proteomics strategy. Med Oncol. 2009. 26: 215-21

15. Nicholson KM, Anderson NG. The Akt/PKB signaling pathway in human malignancy. Cell Signal. 2002. 14: 381-95

16. Peti W, Page R. Molecular basis of MAP kinase regulation. Protein Sci. 2013. 12: 1698-710

17. Raabe EH, Lim KS, Kim JM, Meeker A, Mao XG, Nikkhah G. BRAF activation induces transformation and then senescence in human stem cells: A pilocytic astrocytoma model. Clin Cancer Res. 2011. 17: 3590-9

18. Rodriguez EF, Scheithauer BW, Giannini C, Rynearson A, Cen Ling, Hoesley B. PI3K/AKT pathway alterations are associated with clinically aggressive and histologically anaplastic subsets of pilocytic astrocytoma. Acta Neuropathol. 2011. 121: 407-20

19. Rodriguez FJ, Raabe EH. mTOR: A new therapeutic target for pediatric low-grade glioma?. CNS Oncol. 2014. 3: 89-91

20. Santarpia L, Lippman SL, El-Naggar A. Targeting the mitogen-activated protein kinase RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets. 2012. 16: 103-19

21. Sheppard K, Kinross KM, Soloman B, Pearson RB, Phillips WA. Targeting PI3 Kinase/AKT/mTOR signaling in cancer. Crit Rev Oncol. 2012. 17: 69-95

22. Theeler BJ, Ellezum B, Sadighi ZS, Mehta V, Tran MD, Adesina AM. Adult pilocytic astrocytomas: Clinical features and molecular analysis. Neuro-oncol. 2014. 16: 841-7

23. Vassilakopoulou M, Parisi F, Siddiqui S, England AM, Zarell ER, Anagnostou V. Preanalytic variables and phosphepitope expression in FFPE tissue: A quantitative epitope assessment after variable cold ischemic time. Lab Invest. 2015. 95: 334-41

24. Yu JSL, Cui W. Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination. Development. 2015. 143: 3050-60

Viewing all 227 articles
Browse latest View live