Attina G, Maurizi P, Triarico S, Capozza M. A, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. Biomed Pharmacol J 2020;13(4).
Manuscript received on :23-May-2020
Manuscript accepted on :7-Dec-2020
Published online on: 29-12-2020
Plagiarism Check: Yes
Reviewed by: Dr. Ahmar Rauf  
Second Review by: Dr. Elisabetta Liverani
Final Approval by: Fai Poon

How to Cite    |   Publication History
Views Views: (Visited 1,975 times, 1 visits today)   Downloads PDF Downloads: 593

Giorgio Attina, Palma Maurizi, Silvia Triarico, Michele Antonio Capozza, Alberto Romano, Stefano Mastrangelo and Antonio Ruggiero

Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy.

Corresponding Author E-mail: antonio.ruggiero@unicatt.it

DOI : https://dx.doi.org/10.13005/bpj/2035

Abstract

Optic pathway gliomas (OPG)  are a common cancer in children with neurofibromatosis type 1. OPGs can cause clinical symptoms such as  reduction of visual acuity, alterations of the visual field, pallor of the optical papilla, strabismus, endocrinological alterations up to diencephalic syndrome.The current guidelines provide for wait and see as the main approach if the tumor is not causing visual deterioration and adopting treatment only in the event of significant impairment of the visual function. Therefore, it is essential to early detect the visual deterioration changes as well as  the identification of children eligible for treatment.

Keywords

Children; Neurofibromatosis Type 1; Optic Pathway Glioma

Download this article as: 
Copy the following to cite this article:

Attina G, Maurizi P, Triarico S, Capozza M. A, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. Biomed Pharmacol J 2020;13(4).

Copy the following to cite this URL:

Attina G, Maurizi P, Triarico S, Capozza M. A, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. Biomed Pharmacol J 2020;13(4). Available from: https://bit.ly/3hnGY45

Introduction

Neurofibromatosis type 1 (NF1) is a rare autosomal dominant disease with full penetration and variable expressivity, with an incidence of 1 in 3000.It is caused by mutations or very rarely by microdeletion in the tumor suppressor gene NF1 located on chromosome 17q11.2 which encodes for a protein (neurofibromine 1) regulating cell growth and differentiation; it is an oncosuppressive gene with an important biological role in a pathway of signal transduction that regulates cell proliferation. 1,2

It is a very heterogeneous disease from the clinical point of view, so the diagnosis is placed in the presence of two or more of the following criteria: more than 5 coffee-milk spots; 2 or more neurofibromas or a plexiform neurofibroma; glioma of the optical pathways; freckles; 2 or more Lish nodules; specific skeletal dysplasia; a family history.

One of the most relevant features of NF1 is the predisposition of affected patients to the development of neoplasms mainly of the central and peripheral nervous system or exceptionally of tumors outside the nervous system: in most cases they are benign tumors, very rarely malignant tumors. 1-3

Optic Pathway Glioma in NF1

Optic pathway gliomas are the most common cancers in children with NF1: 15-20% of children with NF1 develop before the age of 7 years (with a peak between 4 and 6 years) a  low grade glioma (LGG) along the optical pathways (Optic Pathway Glioma, OPG).4

Such tumors can be found along all optical pathways, with a higher incidence on optical nerves and optical chiasm, while more rarely they affect the post-chiasmatic optical pathway (optical traits and radiations).5 Gliomas that arise at the level of the optic nerve are usually monolateral, while in sporadic forms (non-NF1-related) the optic chiasm is more frequently involved.

The brain stem is the second most affected site (15% of NF1-related LGG), while neoplasms of the cerebellum, cerebral cortex and base nuclei rarely affect pediatric patients with NF1, involving mainly adolescents and adults. 6-8

These are infiltrating tumors with a low proliferative index: data in the literature highlight that the LGGs that arise in children with NF1 exhibit different biologicalbehaviour, although with great clinical variability, compared to sporadic tumors, with a particularly slow clinical course, up to the description of some cases of spontaneous regression.5,9-13

Despite this about half of children with NF1 and LGG develop clinical symptoms in relation to localization: reduction of visual acuity, alterations of the visual field, pallor of the optical papilla, strabismus, endocrinological alterations up to diencephalic syndrome.9,14-18

Monitoring and Diagnosis

A major problem in NF1 clinical management lies in the absence of validated methods that can predict the development of a LGG in these patients and, similarly, in patients where the tumor has developed and diagnosed, it is not possible to predict reliably which forms will be symptomatic (that is, cause visual deterioration) and therefore deserving of treatment. So, a careful clinical and radiological follow-up programme is essential to identify early onset of symptoms.

The eye evaluation must include the evaluation of visual acuity, fundus oculi and visual field: in order to obtain a more complete and exhaustive evaluation,the execution of visual Evoked Potentials (PEV)should be performed in order to acquire more accurate data, that guide to the execution and timing of surveillance examinations or the decision to start a treatment.19-22

The gold standard investigation to assess the presence and extent of the tumor is cerebral MRI (possibly requiring high resolution sequences of optic nerves and chiasm), also used to monitor radiological progression and response to therapy. Typically, sequences weighed in T1 with and without contrast and sequences weighed in T2 are used. Since contrast enhancement is often heterogeneous and variable, T2-weighted sequences are the most useful to define the tumor involvement in the optical pathway.23

However, even with this type of sequence, the definition of the neoplasm margins is sometimes problematic due to presence in NF1, up to 70% of NF1 patients, of the so-called Focal Areas of Signal Intensity (FASI) or Unknown Bright Objects (Ubos), isointense focal lesions in T1 and hyperintense lesions in T2/FLAIR representing areas of myelin vacuolization. The FASI mainly involve optical pathways, brain stem, basal ganglia and cerebellum and their appearance in regions where NF1 related gliomas can arise, may pose differential diagnostic problems.10,14 However, the FASI do not have mass effect nor contrast-enhancement and often disappear with advancing age; their increase in size or number after 10 years of age should give rise to suspicion of cancer.24,25

Children with NF1 (suspected or diagnosed) but without an associated OPG must undergo an annual examination by an experienced medical equipe and, up to 8 years of age, a complete eye examination. After the age of 8, since visual decline is less likely to occur, eye examinations can be carried out every other year.

For children diagnosed with NF1-OPG, the eye check should be performed every 3 months for the first year, every 6 months for the second year and then move on to an annual visit up to the age of 18 in the event of a stable illness. In these patients, the eye examination must be integrated with neuroimaging (i.e. with the execution of an MRI of the brain with or without contrast) every 3 months for the first year, every 6 months for the second year, every year up to the fifth year and then, less frequently (according to the clinicians judgement up to 18 years of age). It is not recommended to perform MRI as screening for OPG (baseline MRI) in patients with NF1, as it has been demonstrated that the forms diagnosed at the onset of symptoms do not differ substantially from those diagnosed incidentally by MRI in terms of clinical outcomes.10,11

Treatment

The current guidelines provide for wait and see as the main approach, using treatment only in the event of significant deterioration of the visual function and/or radiological progression of the disease.It is possible to distinguish, with reference to this aspect, between absolute indications of therapy, indications related to therapy and signs and symptoms of alarm.10

Absolute Indications for Therapy

Those that define a clinically significant worsening of the visual function are considered absolute indications for therapy, namely: a)worsening of visual acuity ≥0.2 logMAR; b) reduction of the field of view of new appearance.

Indications Relating to Therapy

The indications related to therapy are the same as those that indicate therapy, also considering other prognostic factors, such as the site of the neoplasia, the tumor progression, the reliability of the measurement of visual acuity. This includes the following: a) visual acuity <1 logMAR in an eye at <0.2 logMAR in the other eye; b) Visual acuity between 0.6 and 1 logMAR in one eye; c) Significant progression of disease documented by MRI, associated with suspected visual decline but with no reliable ability to test visual acuity.

Warning Signs and Symptoms

A number of clinical signs and symptoms are able to raise suspicion of disease progression in patients with OPG. The combined evaluation of these factors in an appropriate clinical setting may support the decision to subject the patient to chemotherapy, but in themselves they are intended as indications for closer surveillance and not necessarily treatment. Such warning signs and symptoms include:

Radiological progression of the OPGwithout alterations of visual acuity or visual field is not an indication for starting chemotherapy;

Other ophthalmological findings, such as worsening of the chromatic vision, pallor of the optical disc, swelling of the optic nerve, afferent pupil defect, strabismus, nystagmus required an increased close vigilance in order to evaluate their progression for starting chemotherapy.

New visual field loss or change in visual acuity worse than 0.2 logMAR require prompt treatment.

When treatment is indicated for NF1-OPG, there is no indication to surgery as in most cases it is not possible to obtain a radical removal while preserving visual functionality. Therefore the choice necessarily falls on non-surgical therapeutic options.14

Radiation therapy plays a very limited role in the management of patients with NF1, as it has been shown that it involves a particularly high risk of developing secondary malignant tumors, neuro cognitive disorders, neuro endocrine disorders and radiation-induced vasculitis, such as moya-moya syndrome.26-30

Therefore, when it is necessary to treat a patient with NF1-OPG, chemotherapy is the main therapeutic option.The most widely used chemotherapy scheme involves the combination of Carboplatin and Vincristine: this schedule is usually well tolerated, although about 40% of patients may have hypersensitivity reactions to Carboplatin.31-34As an alternative therapy, the treatment with Vinblastineh as been proven to be effective.  Other proposed schemes include the PCV (Procarbazine, CCNU, Vincristine), the association between Cisplatin and Etoposide and Temozolomide. Etoposide or alkylating agents are generally avoided as their use carry a risk of secondary tumors as NF1 patients have predisposition of patients to the development of malignant neoplasms.35-38

Despite being the best therapeutic option currently available and despite its effectiveness in stopping the growth of the tumor ,39,40 traditional chemotherapy is notoriously burdened with short and long-term adverse effects and, above all, is not always effective in improving or preserving visual function.5,41-51

Although for a long time LGGs were considered tumours with few molecular alterations, numerous authors have reported  the spectrum of the biological characteristics of these tumors, paving the way for the discovery of personalized therapies.These include MEK inhibitors, Bevacizumab and especially Vemurafenib if the BRAF mutation is present.52-57

In order to improve the visual function in such patients, promising studies are underway on the use of Nerve growth factor (NGF) administered locally as eyedrops: in some patients an improvement in the field of vision and PEVs were observed following  this treatment.58-63

Conclusions

In children with NF1 the appearance of OPGs is relatively frequent: in such patients accurate clinical-radiological monitoring is essential in order to allow the early diagnosisof visual deterioration changes and therefore the identification of children for treatment.

The available treatments do not always allow improved visual outcome, hence it is important to identify new treatments for these patients.

Acknowledgments

This work was supported by “Sara un angelo con la bandana Onlus”

Conflict of interest

The authors declare that they have no conflict of interest.

Funding support

There are not funding sourse

References

  1. Upadhyaya M. Neurofibromatosis type 1: diagnosis and recent advances. Expert Opin Med Diagn. 2010; 4: 307-322.
    CrossRef
  2. Jett K, Friedman JM. Clinical and genetic aspects of neurofibromatosis 1. Genet Med. 2010;12:1-11.
    CrossRef
  3. Boyd KP, Korf BR, Theos A. Neurofibromatosis type 1. J Am AcadDermatol. 2009; 61: 1-14
    CrossRef
  4. Albers AC, Gutmann DH. Gliomas in patients with neurofibromatosis type 1. Expert Rev Neurother. 2009;9(4):535-539.
    CrossRef
  5. Listernick R, Ferner RE, Liu GT, Gutmann DH. Optic pathway gliomas in neurofibromatosis-1: controversies and recommendations. Ann Neurol. 2007; 6:189-198.
    CrossRef
  6. Mahdi J, Shah AC, Sato A, et al. A multi-institutional study of brainstem gliomas in children with neurofibromatosis type 1. 2017; 88: 1584-1589.
    CrossRef
  7. Gutmann DH, Rasmussen SA, Wolkenstein P, et al. Gliomas presenting after age 10 in individuals with neurofibromatosis type 1 (NF1). Neurology. 2002; 59: 759-761.
    CrossRef
  8. Vinchon M, Soto-Ares G, Ruchoux MM, et al. Cerebellar gliomas in children with NF1: pathology and surgery. 2000; 16: 417-420.
    CrossRef
  9. Melloni G, Eoli M, Cesaretti C, Bianchessi D, et al. Risk of Optic Pathway Glioma in Neurofibromatosis Type 1: No Evidence of Genotype-Phenotype Correlations in A Large Independent Cohort. Cancers (Basel). 2019 Nov 21;11(12).
    CrossRef
  10. de Blank PM, Fisher MJ, Liu GT, et al. Optic Pathway Gliomas in Neurofibromatosis Type 1: An Update: Surveillance, Treatment Indications, and Biomarkers of Vision. J Neuroophthalmol. 2017; 37 Suppl 1: S23-S32.
    CrossRef
  11. King A, Listernick R, Charrow J, et al. Optic pathway gliomas in neurofibromatosis type 1: the effect of presenting symptoms on outcome. Am J Med Genet A. 2003;122A(2):95-99.
    CrossRef
  12. Thiagalingam S, Flaherty M, Billson F, et al. Neurofibromatosis type 1 and optic pathway gliomas: follow-up of 54 patients. Ophthalmology. 2004; 111: 568-577.
    CrossRef
  13. Rubin J. Intersections at the crossroads: Neurofibromatosis type 1, cAMP, sex, and glioma risk. Molecular & Cellular Oncology. 2016; 3(3): e1069917.
    CrossRef
  14. Khatua S, Gutmann DH, Packer RJ. Neurofibromatosis type 1 and optic pathway glioma: molecular interplay and therapeutic insights. Pediatr Blood Cancer. 2018;65(3).doi: 10.1002/pbc.26838
    CrossRef
  15. HernáizDriever P, von Hornstein S, Pietsch T, et al. Natural history and management of low-grade glioma in NF-1 children. J Neurooncol. 2010; 100: 199-207.
    CrossRef
  16. Blanchard G, Lafforgue MP, Lion-François L, et al. Systematic MRI in NF1 children under six years of age for the diagnosis of optic pathway gliomas. Study and outcome of a French cohort. Eur J Paediatr Neurol. 2016; 20: 275-281.
    CrossRef
  17. Prada CE, Hufnagel RB, Hummel TR, et al. The use of magnetic resonance imaging screening for optic pathway gliomas in children with neurofibromatosis type 1. J Pediatr. 2015; 167: 851-856.
    CrossRef
  18. Fisher MJ, Avery RA, Allen JC, et al. Functional outcome measures for NF1-associated optic pathway glioma clinical trials. Neurology. 2013; 81(21 Suppl 1): S15 and S24.
    CrossRef
  19. Falsini B, Ziccardi L, Lazzareschi I, et al. Longitudinal assessment of childhood optic gliomas: relationship between flicker visual evoked potentials and magnetic resonance imaging findings. J Neurooncol. 2008; 88: 87-96.
    CrossRef
  20. Trisciuzzi MT, Riccardi R, Piccardi M, et al. A fast visual evoked potential method for functional assessment and follow-up of childhood optic gliomas. 2004; 115: 217-226
    CrossRef
  21. Jabbari B, Maitland CG, Morris LM, et al. The value of visual evoked potential as a screening test in neurofibromatosis. Arch Neurol. 1985; 42:1072-1074.
    CrossRef
  22. North K, Cochineas C, Tang E, et al. Optic gliomas in neurofibromatosis type 1: role of visual evoked potentials. Pediatr Neurol. 1994; 10:117-123.
    CrossRef
  23. Balcer LJ, Liu GT, Heller G, et al. Visual loss in children with neurofibromatosis type 1 and optic pathway gliomas: Relation to tumor location by magnetic resonance imaging. American Journal of Ophthalmology. 2001; 131(4), 442-445.
    CrossRef
  24. Lopes FerrazFilho JR, Munis MP, Soares Souza A, et al. Unidentified bright objects on brain MRI in children as a diagnostic criterion for neurofibromatosis type 1. 2008; 38: 305-310.
    CrossRef
  25. DeBella K, Poskitt K, Szudek J, et al. Use of “unidentified bright objects” on MRI for diagnosis of neurofibromatosis 1 in children. 2000; 54: 1646-1651.
    CrossRef
  26. Sharif S, Ferner R, Birch JM, et al. Second primary tumors in neurofibromatosis 1 patients treated for optic glioma: substantial risks after radiotherapy. J ClinOncol. 2006; 24: 2570-2575.
    CrossRef
  27. Armstrong GT, Conklin HM, Huang S, et al. Survival and long-term health and cognitive outcomes after low-grade glioma. Neuro Oncol. 2011; 13:223-234.
    CrossRef
  28. Shalitin S, Gal M, Goshen Y, et al. Endocrine outcome in long-term survivors of childhood brain tumors. Horm Res Paediatr. 2011; 76: 113-122.
    CrossRef
  29. Grill J, Couanet D, Cappelli C, et al. Radiation-induced cerebral vasculopathy in children with neurofibromatosis and optic pathway glioma. 1999; 45: 393, 396.
    CrossRef
  30. Iuvone L, Peruzzi L, Colosimo C, et al. Pretreatment neuropsychological deficits in children with brain tumors. 2011;13(5):517-524
    CrossRef
  31. Ruggiero A, Triarico S, Trombatore G, et al. Incidence, clinical features and management of hypersensitivity reactions to chemotherapeutic drugs in children with cancer. Eur J Clin Pharmacol. 2013;69(10):1739-1746
    CrossRef
  32. Lazzareschi I, Ruggiero A, Riccardi R, Attinà G, Colosimo C, Lasorella A. Hypersensitivityreactions to carboplatin in children. J Neurooncol. 2002; 58:33-37
    CrossRef
  33. Ruggiero A, Rizzo D, Mastrangelo S, Battaglia D, Attinà G, Riccardi R. Interactions between antiepileptic and chemotherapeutic drugs in children with brain tumors: is it time to change treatment?. Pediatr Blood Cancer. 2010;54(2):193-198
    CrossRef
  34. Ater JL, Xia C, Mazewski CM, et al. Nonrandomized comparison of neurofibromatosis type 1 and non-neurofibromatosis type 1 children who received carboplatin and vincristine for progressive low-grade glioma: a report from the Children’s Oncology Group. 2016; 122: 1928-1936.
    CrossRef
  35. Mahgoub N, Taylor BR, Le Beau MM, et al. Myeloid Malignancies Induced by Alkylating Agents in Nf1 Mice. Blood. 1999 93: 3617-3623.
    CrossRef
  36. Riccardi A, Mazzarella G, Cefalo G, et al. Pharmacokinetics of Temozolomide given three times a day in pediatric and adult patients. Cancer Chemother Pharmacol. 2003; 52: 459-464
    CrossRef
  37. Rizzo D, Scalzone M, Ruggiero A, et al. Temozolomide in the treatment of newly diagnosed diffuse brainstem glioma in children: a broken promise?. J Chemother. 2015;27(2):106-110
    CrossRef
  38. Ruggiero A, Rizzo D, Attinà G, et al. Phase I study of temozolomide combined with oral etoposide in children with malignant glial tumors. J Neurooncol.2013;113(3):513-518
    CrossRef
  39. Perilongo G. Considerations on the role of chemotherapy and modern radiotherapy in the treatment of childhood low grade glioma. J Neurooncol. 2005; 75(3): 301-307.
    CrossRef
  40. Packer RJ, Ater J, Allen J, et al. Carboplatin and vincristine chemotherapy for children with newly diagnosed progressive low-grade gliomas. J Neurosurg. 1997; 86: 747-754.
    CrossRef
  41. Fisher MJ, Loguidice M, Gutmann DH, et al. Visual outcomes in children with 35 neurofibromatosis type 1-associated optic pathway glioma following chemotherapy: a multicenter retrospective analysis. Neuro Oncol. 2012; 14: 790-797.
    CrossRef
  42. Moreno L, Bautista F, Ashley S, et al. Does chemotherapy affect the visual outcome in children with optic pathway glioma? A systematic review of the evidence. Eur J Cancer. 2010; 46: 2253-2259.
    CrossRef
  43. Dalla Via P, Opocher E, Pinello ML, et al. Visual outcome of a cohort of children with neurofibromatosis type 1 and optic pathway glioma followed by a pediatric neuro-oncology program. Neuro Oncol. 2007; 9: 430-437.
    CrossRef
  44. Dodgshun AJ, Elder JE, Hansford, JR, et al. Long-term visual outcome after chemotherapy for optic pathway glioma in children: Site and age are strongly predictive. Cancer. 2015; 121(23), 4190-4196.
    CrossRef
  45. Shofty B, Ben-Sira L, Freedman S, et al. Visual outcome following chemotherapy for progressive optic pathway gliomas. Pediatr Blood Cancer. 2011; 57(3), 481-485.
    CrossRef
  46. Kalin-Hajdu E, Décarie JC, Marzouki M, et al. Visual acuity of children treated with chemotherapy for optic pathway gliomas. Pediatr Blood Cancer. 2014;61(2):223-227.
    CrossRef
  47. Cefalo MG, Ruggiero A, Maurizi P, Attinà G, Arlotta A, Riccardi R. Pharmacological management of chemotherapy-induced nausea and vomiting in children with cancer. J Chemother. 2009;21(6):605-610
    CrossRef
  48. Ruggiero A, Ferrara P, Attinà G, Rizzo D, Riccardi R. Renal toxicity and chemotherapy in children with cancer. Br J Clin Pharmacol. 2017;83(12):2605-2614
    CrossRef
  49. Chiaretti A, Ruggiero A, Barbi E, et al. Comparison of propofol versus propofol-ketamine combination in pediatric oncologic procedures performed by non-anesthesiologists. Pediatr Blood Cancer. 2011;57(7):1163-1167
    CrossRef
  50. Chiaretti A, Ruggiero A, Barone G, et al.Propofol/alfentanil and propofol/ketamine procedural sedation in children with acute lymphoblastic leukaemia: safety, efficacy and their correlation with pain neuromediator expression. Eur J Cancer Care. (Engl) 2010;19(2):212-220
    CrossRef
  51. Timeus F, Crescenzio N, Longoni D, et al. Paroxysmal nocturnal hemoglobinuria clones in children with acquired aplastic anemia: a multicentre study. PLoS One.2014;9(7):e101948
    CrossRef
  52. Theeler BJ, Ellezam B, Yust-Katz Shlomit, et al. Prolonged survival in adult neurofibromatosis type 1 patients with recurrent high-grade gliomas treated with bevacizumab. J Neurol. 2014; 261(8): 1559-64.
    CrossRef
  53. Sikkema AH, de Bont ES, Molema G, et al. Vascular endothelial growth factor receptor 2 (VEGFR-2) signalling activity in paediatric pilocytic astrocytoma is restricted to tumour endothelial cells. 2011; 37: 538-548.
    CrossRef
  54. Rodriguez FJ, Ligon AH, Horkayne-Szakaly I, et al. BRAF duplications and MAPK pathway activation are frequent in gliomas of the optic nerve proper. J NeuropatholExp Neurol. 2012; 71:789- 794.
    CrossRef
  55. See WL, Tan IL, Mukherjee J, et al. Sensitivity of glioblastomas to clinically available MEK inhibitors is defined by neurofibromin 1 deficiency. Cancer Research. 2012; 72(13), 3350-3359.
    CrossRef
  56. Yalon M, Rood B, MacDonald TJ, et al. A feasibility and efficacy study of rapamycin and erlotinib for recurrent pediatric low-grade glioma (LGG). Pediatr Blood Cancer. 2013; 60:71- 76.
    CrossRef
  57. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res.2007;13(4):1253-1259.
    CrossRef
  58. Falsini B, Chiaretti A, Barone G, et al. Topical nerve growth factor as a visual rescue strategy in pediatric optic gliomas: a pilot study including electrophysiology. NeurorehabilNeuralRepair. 2011; 25: 512-520.
    CrossRef
  59. Falsini B, Chiaretti A, Rizzo D, et al. Nervegrowth factor improves visual loss in childhood optic gliomas: a randomized, double-blind, phase II clinical trial. Brain.  2016; 139(Pt 2): 404-414.
    CrossRef
  60. Falsini B, Iarossi G, Chiaretti A, et al. NGF eye-drops topical administration in patients with retinitis pigmentosa, a pilot study. J Transl Med.2016;14:8-14
    CrossRef
  61. Chiaretti A, Antonelli A, Piastra M, Genovese O, Polidori G, Aloe L. Expression of neurotrophic factors in cerebrospinal fluid and plasma of children with viral and bacterial meningoencephalitis. Acta Paediatr.2004;93(9):1178‐
    CrossRef
  62. Fantacci C, Capozzi D, Ferrara P, Chiaretti A. Neuroprotective role of nerve growth factor in hypoxic-ischemic brain injury. Brain Sci. 2013;3(3):1013‐
    CrossRef
  63. Chiaretti A, Aloe L, Antonelli A, et al. Neurotrophic factor expression in childhood low-grade astrocytomas and ependymomas. Childs Nerv Syst. 2004; 20:412-419
    CrossRef
Share Button
(Visited 1,975 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.