Medulloblastoma

Earn CME/CE in your profession:


Continuing Education Activity

While leukemias are the most common type of malignancy affecting the pediatric population, brain tumors are the most common solid tumors in this population. Medulloblastoma is the most common malignant brain tumor in children, constituting nearly 20 percent of all pediatric brain tumors. It is categorized as an embryonal neuroepithelial tumor of the cerebellum. Surgical resection followed by radiation and chemotherapy are the mainstay of therapy, with five-year survival rates ranging between 20 and 100 percent. This wide range is multifactorial, owing in part to age at diagnosis, presence of metastases at diagnosis, subgroup variant of medulloblastoma, and concurrent cytogenetic abnormalities including copy-number variations. Regardless of long-term survival, treatment-related cognitive, neurologic, and endocrinologic effects remain a debilitating concern and an impetus for research for additional therapeutic modalities. This activity describes the evaluation and management of patients with medulloblastoma and highlights the role of the interprofessional team in improving care for affected children.

Objectives:

  • Describe the epidemiology of medulloblastoma.
  • Explain when the diagnosis of medulloblastoma should be considered.
  • Identify treatment considerations for patients with medulloblastoma.
  • Describe how enhanced coordination of the interprofessional team can lead to more rapid recognition of medulloblastoma and subsequently improve their evaluation, enhancing detection of pathology and allowing for treatment when indicated.

Introduction

While leukemias are the most common type of malignancy to afflict the pediatric population, brain tumors are the most common solid tumors in this age group.[1] Medulloblastoma is the most common malignant brain tumor in children constituting nearly 20% of all pediatric brain tumors.[2] It is categorized as an embryonal neuroepithelial tumor of the cerebellum.

This is a high-grade tumor that has a propensity to spread via the cerebrospinal fluid.

Within the first few years of diagnosis, mortality approximates 15%; however, cure rates can reach as high as 60% with current therapeutic modalities.[3][4] Surgical resection preceded and/or followed by radiation and chemotherapy is the mainstay of therapy, with five-year survival rates of between 50% to 90%. This wide range is multifactorial, owing in part to age at diagnosis, the presence of metastases at diagnosis, and a histologic variant of medulloblastoma.[1][5] Regardless of long-term survival, treatment-related cognitive, neurologic, and endocrinologic effects remain a debilitating concern and an impetus for the search for further therapeutic modalities.

Etiology

There is no clear etiology to medulloblastoma. Some studies have found a link between maternal diet and blood/immune disorders during pregnancy.[6] Others report an association with viral infections, for example, early John Cunningham (JC) viral infections or human cytomegalovirus (CMV) infections in childhood.[7][8]

Medulloblasotms may have a familial association and are also known to be associated with:

  • Gorlin syndrome
  • Fanconi anemia
  • Turcot syndrome
  • Li-Fraumeni syndrome

Epidemiology

Examining data from the Surveillance, Epidemiology, and End-Results (SEER) database from 1973 through 2007, the annual incidence for medulloblastoma was reported at six per million children, in other words, approximately 450 new pediatric cases per year. Children age 4 to 9 years old had the highest incidence at 44%, followed by adolescents (10 to 16 years old) at 23%, and only a 12% incidence in infants/toddlers (0 to 3 years old). Pediatric incidence was calculated to be ten-fold higher than adult incidence. Medulloblastoma affected males 1.5 times more than females.[9][10]

Pathophysiology

Thought to originate from the granule cell precursors in the external germinal layer (EGL) of the developing cerebellum, tumor growth starts in the fourth ventricle and can grow to occupy it completely. After that, the tumor spreads to the cerebellar vermis and the brainstem, seeding the craniospinal axis. Medulloblastoma is a highly malignant tumor with a propensity for local invasion and distant metastatic spread through the subarachnoid system (i.e., within the brain and along the spinal cord, also known as "drop mets").[11]

Extraneural metastases in pediatric medulloblastomas are an infrequent occurrence (approximately 7%). The most frequent sites of extraneural metastasis in children include bone (78%), lymph nodes (33%), liver (15%), and lungs (11%). The average time to develop after maximal surgical resection is approximately 20 months. Survival in these cases is dismal and in most cases, can be less than six months.[12]

Notably, the most common cytogenetic mutation encountered in medulloblastoma is isochromosome 17q, resulting from the loss of the short arm (p) with a resultant gain of genetic material from the long arm (q). Also, deletions in the short arm have also been frequently reported, leading to loss of heterozygosity of 17p, (i.e., 17pLOH).[13][14][15] Interestingly, the tumor suppressor gene, TP53, which is located on chromosome 17p, is rarely mutated in medulloblastoma.[16][17] Thus, the search for putative tumor suppressor genes on chromosome 17p in the context of medulloblastoma is ongoing.

Histopathology

The five histologic subtypes encountered in medulloblastoma include:

  1. classic
  2. desmoplastic-nodular (D/N)
  3. large-cell anaplastic (LC/A)
  4. melanotic
  5. medullomyoblastoma 

In classic medulloblastoma, sheets of small round cells, possessing a high nuclear-to-cytoplasmic ratio, are noted. They have a high invasive tendency and possess occasional neuroblastic differentiation. The classic type constitutes approximately 70% of medulloblastomas. Nodules of tumor cells displaying neurocytic differentiation on a collagen-rich matrix characterize the desmoplastic variant; these tumors are less aggressive than the classic variant and account for 15% of medulloblastomas. Large-cell anaplastic medulloblastomas, as the designation suggests, demonstrate features of anaplasia. These features are large tumor cells with abundant cytoplasm, pleomorphic nuclei, and prominent nucleoli. These tumors are typically located in the cerebellar vermis and are highly aggressive, demonstrating high mitotic and apoptotic activity with large areas of necrosis. Consequently, the prognosis is especially poor with short survival times after diagnosis. They constitute approximately 10% of medulloblastomas.[18] The last two variants are rare and make up the remaining 5% of medulloblastomas.[19]

History and Physical

Given its origin in the cerebellum with the propensity of locally spreading into the fourth ventricle, patients most often present with a combination of cerebellar signs like clumsiness, gait disturbances, and obstructive hydrocephalus, for example, early morning headaches, nausea/vomiting, double vision, or blurry vision.

Since the majority of medulloblastomas arse in the fourth ventricle, obstructive hydrocephalus is not uncommon. These patients may present with emesis, headache, and lethargy. Time from symptom onset to diagnosis is usually short, usually two to three months.[11][20]

Almost all patients have early signs of elevated intracranial pressure that is associated with headache early in the morning and relieved over the course of the day. Patients with Cushing triad usually have impending herniation and need immediate treatment.

The tumor in the cerebellum can also lead to ataxia and truncal unsteadiness. If the brain stem is involved, it may lead to cranial nerve palsies that may present as diplopia, hearing loss, facial weakness, and head tilt.

Fundoscopy may show optic pallor or papilledema. Paralysis of the abducens nerve may lead to an inability to abduct the eye. The sun setting sign may be seen in some infants. Measurement of the head circumference in infants may reveal macrocephaly.

Evaluation

Investigations should include routine laboratories, including thyroid function tests.

Computed tomogram (CT) of the brain may reveal a mass in the 4th ventricle. Most medulloblastomas are contrast-enhancing. Hydrocephalus is common in most patients.

Magnetic resonance imaging (MRI) is performed in all patients as it demonstrates the anatomy better than CT scan. MRI is often used to detect post-surgical residual disease. Spinal MRI is also useful for detecting metastatic spinal lesions.

Other tests include audiometry prior to starting cisplatin treatment. Echocardiogram and pulmonary function tests are obtained as a baseline prior to the start of chemotherapy.

A lumbar puncture may reveal leptomeningeal tumor spread. But despite a positive MRI, cerebrospinal fluid (CSF) may be normal. Prior to a lumbar puncture, fundoscopy is done. Following surgery, lumbar puncture is delayed for at least 2 weeks because the surgery may have caused tumor dissemination, leading to a false-positive result.

Treatment / Management

Current treatment modalities for medulloblastoma combine surgical resection with chemotherapy and radiation. By traditional risk stratification, cure rates in the average-risk group reached three-quarters of patients. However, post-surgical treatment-related neurologic, cognitive, and endocrinologic sequelae, including intellectual retardation and growth hormone deficiency, remain a source of morbidity in up to 80% of survivors. The high-risk group experienced up to 50% mortality due not only to the presence of extraneural metastases at diagnosis but also due to their young age at diagnosis, which poses significant limitations to their therapeutic options, namely lower doses of radiation and chemotherapeutic agents.[1]

Newer subgroup classification systems have facilitated the development of more targeted therapeutics aimed at disrupting signal transduction pathways critical to phenotypic transformation. These are currently under clinical investigation:

SHH Subgroup

The SHH pathway is activated by the binding of Sonic Hedgehog to its receptor Patched 1 (PTCH1), which activates downstream signaling via a key mediator Smoothened (SMO). The most widely studied targeted therapeutic agents today are SMO inhibitors, i.e., cyclopamine, HhAntag, vismodegib, saridegib, and sonidegib.[21][22][23][24] Sonidegib (NCT01708174) and vismodegib are currently in phase 2 clinical trials in patients with relapsed SHH subgroup medulloblastoma.[25]

WNT Subgroup

The key step in WNT signal transduction leading to malignant transformation is a lack of degradation of beta-catenin due to mutations at key amino acid residues that are normally destined for phosphorylation. Hence, new drugs have been developed to target steps in downstream signaling by beta-catenin. These include naturally-occurring protein phosphatase inhibitors, cantharidin, norcantharidin, and ginkgetin.[26][27][28][29] However, given the benign prognosis of patients in this subgroup (five-year overall survival > 90%), current efforts are being aimed at limiting cytotoxic therapies such as radiation dosing (NCT02724579).

Non-SHH/WNT Subgroup

Unfortunately, not much is known about the signaling pathways implicated in non-SHH/WNT subgroup medulloblastoma. As a result, targeted therapeutics have yet to be developed for this type of medulloblastoma. Promising strategies currently under investigation include myc inhibition, cell cycle checkpoint inhibitors, and histone deacetylase (HDAC) inhibitors.[30][31][32][33]

Differential Diagnosis

  • Choroid plexus carcinoma
  • Ependymoma
  • Teratoma

Surgical Oncology

Surgery aims to excise as much tumor as possible and to relieve the obstruction of the CSF. However, care should be taken during surgery as the tumor is friable and can quickly disseminate. Following surgery, more than 40% of patients have residual neurological deficits. Besides cerebellar dysfunction, others may develop mutism, cranial nerve palsies, and hemiparesis. Some symptoms do reverse with time, but extensive rehabilitation is required. At least 50% will have permanent deficits.

In most patients, a ventriculoperitoneal shunt is necessary for the relief of hydrocephalus. Some patients may obtain benefits from an endoscopic third ventriculostomy. Besides central venous access for medications, an Ommaya reservoir for chemotherapy is often used. However, all these procedures have risks, and technical complications are common.

Radiation Oncology

Radiation therapy is often used for local control and management of the micrometastatic disease. Radiation is administered to the craniospinal axis. To maximize the response, it should be given within four weeks after surgery. Reducing the dose of radiation to minimize the complications is associated with higher relapse rates and poor outcomes. For those who relapse, the prognosis is dismal. At this stage, additional radiation may not help.

Staging

Patients with medulloblastoma were traditionally stratified into average-risk or high-risk groups based on three primary characteristics: (1) age, (2) extent of residual tumor post-op, and (3) degree of dissemination at the time of diagnosis. Patients who were at least three years old at diagnosis, had, at most, 1.5 cm of postoperative residual disease by MRI, and those who had no extraneural metastases were categorized as average-risk. The rest were high-risk.[1][34]

This traditional risk stratification discounted the influence of tumor histology on tumor behavior and impact on patient prognosis. Recent transcriptional profiling studies derived from microarray data of large numbers of patients with medulloblastoma revealed clusters of patients with similar transcriptomes, proteomes, and cytogenetic signatures involving unique signal transduction pathways. This has led to the currently accepted classification system of medulloblastoma into four primary subgroups: (1) SHH, sonic hedgehog, (2) WNT, wingless; (3) non-SHH/WNT group 3; and (4) non-SHH/WNT group 4.[10][34] Moreover, this system has not only augmented prognostication but has also facilitated the development of novel therapeutic options. That said, the field of medulloblastoma is ever-evolving, and there may be as many as 12 subtypes of medulloblastoma that cluster with high fidelity.[35][36] 

Using a combination of molecular profiling and histology, the World Health Organization (WHO) presently divides medulloblastoma into five subtypes[37]:

  1. WNT-activated with classic histology
  2. SHH-activated (wild-type TP53), with classic, desmoplastic/nodular, or large-cell/anaplastic histology
  3. SHH-activated (mutant TP53), with classic or large-cell/anaplastic histology
  4. non-WNT/non-SHH, group 3, with classic or large-cell anaplastic histology
  5. non-WNT/non-SHH, group 4, with classic or large-cell/anaplastic histology

Prognosis

The outcomes depend on several factors, including age, stage, the extent of residual disease after surgery, and response to treatment. Children with WNT subtype have a good outcome, whereas those with MYCN or MYC amplification have poor outcomes. Even after successful treatment, many patients are left with permanent residual neurological and cognitive deficits. Children have difficulties with learning, and there is growth impairment. In addition, deficiency of gonadotrophin and thyroid hormones are common. The key reason for these complications is craniospinal radiation.

The five-year survivor rate for those with average risk disease is 85% as long as there is no significant residual disease and no evidence of spread.

In the high-risk group, the 5-year survival is less than 40%.

In children less than three years, survival varies from 30%-70%. Children with metastatic disease do poor, whereas those with SHH or WNT activated lesions have better survival.

Transcriptional and methylation profiling date coupled with cytogenetic aberrations and copy number variations and patient outcomes has helped subclassify medulloblastoma in as many as 12 subtypes, two WNT, four SHH, three group 3 and three group 4. The highest survival rates have consistently been seen for the WNT subgroup at 97-100% at 5 years. The SHH subgroup remains intermediate, with 5-year overall survival between 70%-88%. The non-SHH/WNT subgroups have been thought to have the worst prognosis though group 4 patients tend to do better. Group 4 MB has a 65%-85% 5-year overall survival, while group 3 MB has dismal rates between 40%-65% 5-year overall survival, with myc amplification portending the poorest outcomes.[36]

Complications

  • Headaches
  • Emesis
  • Ataxia
  • Posterior fossa syndrome
  • Shunt infection/malfunction
  • Bleeding
  • Hemiparesis
  • Neurocognitive impairment
  • Somnolence syndrome
  • Radiation necrosis
  • Ototoxicity
  • Secondary malignancies
  • Chemotherapy associated complications

Deterrence and Patient Education

The patients (or more often the parents of the patients) should be educated on the prognosis of the disease and proper consent should be taken including all the risks involved, given the critical location of the tumor, before proceeding with any type of surgical intervention.

Enhancing Healthcare Team Outcomes

The diagnosis and management of medulloblastoma are best done by an interprofessional team that consists of a pediatrician, neurologist, neurosurgeon, oncologist, radiation therapist, and specialty trained nurses. The majority of children first present with cerebellar symptoms to the primary provider, pediatrician, or nurse practitioner. Once diagnosed the treatment modalities for medulloblastoma combine surgical resection with chemotherapy and radiation. Oncology board-certified pharmacists can consult with the oncologist on the regimens for chemotherapy, and the alternatives based on patient response. The pharmacist should also educate the patient on the management of headache and emesis. The oncology nurse is vital for patient monitoring and educating the patient and family. The oncology nurse should educate the patient on the types of treatments, their benefits, and potential complications. Mental health professionals may be important is vital as many patients develop depression and anxiety. The role of speech, occupational, and physical therapy cannot be understated. The members of the team should communicate and hold weekly meetings to determine the steps in care and ensure that the patient is receiving the current standard of care.

By traditional risk stratification, cure rates in the average-risk group reached three-quarters of patients. However, post-surgical treatment-related neurologic, cognitive, and endocrinologic sequelae, including intellectual retardation and growth hormone deficiency, remain a source of morbidity in up to 80% of survivors. The high-risk patient may have mortality in excess of 50%-75%. For those who survive, complete functional recovery may take months or even years, but an interprofessional team will optimize outcomes.[38][39][40] [Level V]


Details

Updated:

6/26/2023 9:22:26 PM

References


[1]

Gilbertson RJ. Medulloblastoma: signalling a change in treatment. The Lancet. Oncology. 2004 Apr:5(4):209-18     [PubMed PMID: 15050952]


[2]

Rossi A, Caracciolo V, Russo G, Reiss K, Giordano A. Medulloblastoma: from molecular pathology to therapy. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008 Feb 15:14(4):971-6. doi: 10.1158/1078-0432.CCR-07-2072. Epub     [PubMed PMID: 18281528]


[3]

Pfister S, Remke M, Benner A, Mendrzyk F, Toedt G, Felsberg J, Wittmann A, Devens F, Gerber NU, Joos S, Kulozik A, Reifenberger G, Rutkowski S, Wiestler OD, Radlwimmer B, Scheurlen W, Lichter P, Korshunov A. Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009 Apr 1:27(10):1627-36. doi: 10.1200/JCO.2008.17.9432. Epub 2009 Mar 2     [PubMed PMID: 19255330]


[4]

Wu X, Northcott PA, Croul S, Taylor MD. Mouse models of medulloblastoma. Chinese journal of cancer. 2011 Jul:30(7):442-9. doi: 10.5732/cjc.011.10040. Epub     [PubMed PMID: 21718590]


[5]

Huse JT, Holland EC. Genetically engineered mouse models of brain cancer and the promise of preclinical testing. Brain pathology (Zurich, Switzerland). 2009 Jan:19(1):132-43. doi: 10.1111/j.1750-3639.2008.00234.x. Epub     [PubMed PMID: 19076778]


[6]

Bunin GR,Kushi LH,Gallagher PR,Rorke-Adams LB,McBride ML,Cnaan A, Maternal diet during pregnancy and its association with medulloblastoma in children: a children's oncology group study (United States). Cancer causes     [PubMed PMID: 16132798]


[7]

Baryawno N, Rahbar A, Wolmer-Solberg N, Taher C, Odeberg J, Darabi A, Khan Z, Sveinbjörnsson B, FuskevÅg OM, Segerström L, Nordenskjöld M, Siesjö P, Kogner P, Johnsen JI, Söderberg-Nauclér C. Detection of human cytomegalovirus in medulloblastomas reveals a potential therapeutic target. The Journal of clinical investigation. 2011 Oct:121(10):4043-55. doi: 10.1172/JCI57147. Epub 2011 Sep 26     [PubMed PMID: 21946257]


[8]

Krynska B, Del Valle L, Croul S, Gordon J, Katsetos CD, Carbone M, Giordano A, Khalili K. Detection of human neurotropic JC virus DNA sequence and expression of the viral oncogenic protein in pediatric medulloblastomas. Proceedings of the National Academy of Sciences of the United States of America. 1999 Sep 28:96(20):11519-24     [PubMed PMID: 10500209]


[9]

Smoll NR, Drummond KJ. The incidence of medulloblastomas and primitive neurectodermal tumours in adults and children. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia. 2012 Nov:19(11):1541-4. doi: 10.1016/j.jocn.2012.04.009. Epub 2012 Sep 13     [PubMed PMID: 22981874]


[10]

Taylor MD,Northcott PA,Korshunov A,Remke M,Cho YJ,Clifford SC,Eberhart CG,Parsons DW,Rutkowski S,Gajjar A,Ellison DW,Lichter P,Gilbertson RJ,Pomeroy SL,Kool M,Pfister SM, Molecular subgroups of medulloblastoma: the current consensus. Acta neuropathologica. 2012 Apr;     [PubMed PMID: 22134537]

Level 3 (low-level) evidence

[11]

Raffel C. Medulloblastoma: molecular genetics and animal models. Neoplasia (New York, N.Y.). 2004 Jul-Aug:6(4):310-22     [PubMed PMID: 15256053]

Level 3 (low-level) evidence

[12]

Rochkind S, Blatt I, Sadeh M, Goldhammer Y. Extracranial metastases of medulloblastoma in adults: literature review. Journal of neurology, neurosurgery, and psychiatry. 1991 Jan:54(1):80-6     [PubMed PMID: 2010766]


[13]

Biegel JA, Rorke LB, Packer RJ, Sutton LN, Schut L, Bonner K, Emanuel BS. Isochromosome 17q in primitive neuroectodermal tumors of the central nervous system. Genes, chromosomes & cancer. 1989 Nov:1(2):139-47     [PubMed PMID: 2487154]


[14]

Bigner SH, Mark J, Friedman HS, Biegel JA, Bigner DD. Structural chromosomal abnormalities in human medulloblastoma. Cancer genetics and cytogenetics. 1988 Jan:30(1):91-101     [PubMed PMID: 3422050]


[15]

Cogen PH, McDonald JD. Tumor suppressor genes and medulloblastoma. Journal of neuro-oncology. 1996 Jul:29(1):103-12     [PubMed PMID: 8817421]


[16]

Adesina AM, Nalbantoglu J, Cavenee WK. p53 gene mutation and mdm2 gene amplification are uncommon in medulloblastoma. Cancer research. 1994 Nov 1:54(21):5649-51     [PubMed PMID: 7923211]


[17]

Batra SK, McLendon RE, Koo JS, Castelino-Prabhu S, Fuchs HE, Krischer JP, Friedman HS, Bigner DD, Bigner SH. Prognostic implications of chromosome 17p deletions in human medulloblastomas. Journal of neuro-oncology. 1995:24(1):39-45     [PubMed PMID: 8523074]


[18]

Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC, Cavenee WK. The WHO classification of tumors of the nervous system. Journal of neuropathology and experimental neurology. 2002 Mar:61(3):215-25; discussion 226-9     [PubMed PMID: 11895036]


[19]

Kool M, Korshunov A, Remke M, Jones DT, Schlanstein M, Northcott PA, Cho YJ, Koster J, Schouten-van Meeteren A, van Vuurden D, Clifford SC, Pietsch T, von Bueren AO, Rutkowski S, McCabe M, Collins VP, Bäcklund ML, Haberler C, Bourdeaut F, Delattre O, Doz F, Ellison DW, Gilbertson RJ, Pomeroy SL, Taylor MD, Lichter P, Pfister SM. 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 neuropathologica. 2012 Apr:123(4):473-84. doi: 10.1007/s00401-012-0958-8. Epub 2012 Feb 23     [PubMed PMID: 22358457]

Level 1 (high-level) evidence

[20]

Ryzhova MV, Zheludkova OG, Kumirova ÉV, Shishkina LV, Panina TN, Gorelyshev SK, Khukhlaeva EA, Mazerkina NA, Matuev KB, Medvedeva OA, Tarasova EM, Kholodov BV, Kapitul'skaia OIu. Characteristics of medulloblastoma in children under age of three years. Zhurnal voprosy neirokhirurgii imeni N. N. Burdenko. 2013:77(1):3-10; discussion 11     [PubMed PMID: 23659115]


[21]

Lee MJ, Hatton BA, Villavicencio EH, Khanna PC, Friedman SD, Ditzler S, Pullar B, Robison K, White KF, Tunkey C, LeBlanc M, Randolph-Habecker J, Knoblaugh SE, Hansen S, Richards A, Wainwright BJ, McGovern K, Olson JM. Hedgehog pathway inhibitor saridegib (IPI-926) increases lifespan in a mouse medulloblastoma model. Proceedings of the National Academy of Sciences of the United States of America. 2012 May 15:109(20):7859-64. doi: 10.1073/pnas.1114718109. Epub 2012 May 1     [PubMed PMID: 22550175]


[22]

Romer JT, Kimura H, Magdaleno S, Sasai K, Fuller C, Baines H, Connelly M, Stewart CF, Gould S, Rubin LL, Curran T. Suppression of the Shh pathway using a small molecule inhibitor eliminates medulloblastoma in Ptc1(+/-)p53(-/-) mice. Cancer cell. 2004 Sep:6(3):229-40     [PubMed PMID: 15380514]


[23]

Rudin CM, Hann CL, Laterra J, Yauch RL, Callahan CA, Fu L, Holcomb T, Stinson J, Gould SE, Coleman B, LoRusso PM, Von Hoff DD, de Sauvage FJ, Low JA. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. The New England journal of medicine. 2009 Sep 17:361(12):1173-8. doi: 10.1056/NEJMoa0902903. Epub 2009 Sep 2     [PubMed PMID: 19726761]


[24]

Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, Tibes R, Weiss GJ, Borad MJ, Hann CL, Brahmer JR, Mackey HM, Lum BL, Darbonne WC, Marsters JC Jr, de Sauvage FJ, Low JA. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. The New England journal of medicine. 2009 Sep 17:361(12):1164-72. doi: 10.1056/NEJMoa0905360. Epub 2009 Sep 2     [PubMed PMID: 19726763]


[25]

Robinson GW, Orr BA, Wu G, Gururangan S, Lin T, Qaddoumi I, Packer RJ, Goldman S, Prados MD, Desjardins A, Chintagumpala M, Takebe N, Kaste SC, Rusch M, Allen SJ, Onar-Thomas A, Stewart CF, Fouladi M, Boyett JM, Gilbertson RJ, Curran T, Ellison DW, Gajjar A. Vismodegib Exerts Targeted Efficacy Against Recurrent Sonic Hedgehog-Subgroup Medulloblastoma: Results From Phase II Pediatric Brain Tumor Consortium Studies PBTC-025B and PBTC-032. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015 Aug 20:33(24):2646-54. doi: 10.1200/JCO.2014.60.1591. Epub 2015 Jul 13     [PubMed PMID: 26169613]


[26]

Yang J, Wu J, Tan C, Klein PS. PP2A:B56epsilon is required for Wnt/beta-catenin signaling during embryonic development. Development (Cambridge, England). 2003 Dec:130(23):5569-78     [PubMed PMID: 14522869]


[27]

Ye ZN, Yu MY, Kong LM, Wang WH, Yang YF, Liu JQ, Qiu MH, Li Y. Biflavone Ginkgetin, a Novel Wnt Inhibitor, Suppresses the Growth of Medulloblastoma. Natural products and bioprospecting. 2015 Mar 29:5(2):91-7     [PubMed PMID: 25821199]

Level 2 (mid-level) evidence

[28]

Baryawno N, Sveinbjörnsson B, Eksborg S, Chen CS, Kogner P, Johnsen JI. Small-molecule inhibitors of phosphatidylinositol 3-kinase/Akt signaling inhibit Wnt/beta-catenin pathway cross-talk and suppress medulloblastoma growth. Cancer research. 2010 Jan 1:70(1):266-76. doi: 10.1158/0008-5472.CAN-09-0578. Epub 2009 Dec 22     [PubMed PMID: 20028853]


[29]

Cimmino F, Scoppettuolo MN, Carotenuto M, De Antonellis P, Dato VD, De Vita G, Zollo M. Norcantharidin impairs medulloblastoma growth by inhibition of Wnt/β-catenin signaling. Journal of neuro-oncology. 2012 Jan:106(1):59-70. doi: 10.1007/s11060-011-0645-y. Epub 2011 Jul 7     [PubMed PMID: 21735115]


[30]

Milde T, Oehme I, Korshunov A, Kopp-Schneider A, Remke M, Northcott P, Deubzer HE, Lodrini M, Taylor MD, von Deimling A, Pfister S, Witt O. HDAC5 and HDAC9 in medulloblastoma: novel markers for risk stratification and role in tumor cell growth. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010 Jun 15:16(12):3240-52. doi: 10.1158/1078-0432.CCR-10-0395. Epub 2010 Apr 22     [PubMed PMID: 20413433]


[31]

Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, Chesi M, Schinzel AC, McKeown MR, Heffernan TP, Vakoc CR, Bergsagel PL, Ghobrial IM, Richardson PG, Young RA, Hahn WC, Anderson KC, Kung AL, Bradner JE, Mitsiades CS. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011 Sep 16:146(6):904-17. doi: 10.1016/j.cell.2011.08.017. Epub 2011 Sep 1     [PubMed PMID: 21889194]

Level 2 (mid-level) evidence

[32]

Faria CC, Agnihotri S, Mack SC, Golbourn BJ, Diaz RJ, Olsen S, Bryant M, Bebenek M, Wang X, Bertrand KC, Kushida M, Head R, Clark I, Dirks P, Smith CA, Taylor MD, Rutka JT. Identification of alsterpaullone as a novel small molecule inhibitor to target group 3 medulloblastoma. Oncotarget. 2015 Aug 28:6(25):21718-29     [PubMed PMID: 26061748]


[33]

Morfouace M, Shelat A, Jacus M, Freeman BB 3rd, Turner D, Robinson S, Zindy F, Wang YD, Finkelstein D, Ayrault O, Bihannic L, Puget S, Li XN, Olson JM, Robinson GW, Guy RK, Stewart CF, Gajjar A, Roussel MF. Pemetrexed and gemcitabine as combination therapy for the treatment of Group3 medulloblastoma. Cancer cell. 2014 Apr 14:25(4):516-29. doi: 10.1016/j.ccr.2014.02.009. Epub 2014 Mar 27     [PubMed PMID: 24684846]


[34]

Gottardo NG, Hansford JR, McGlade JP, Alvaro F, Ashley DM, Bailey S, Baker DL, Bourdeaut F, Cho YJ, Clay M, Clifford SC, Cohn RJ, Cole CH, Dallas PB, Downie P, Doz F, Ellison DW, Endersby R, Fisher PG, Hassall T, Heath JA, Hii HL, Jones DT, Junckerstorff R, Kellie S, Kool M, Kotecha RS, Lichter P, Laughton SJ, Lee S, McCowage G, Northcott PA, Olson JM, Packer RJ, Pfister SM, Pietsch T, Pizer B, Pomeroy SL, Remke M, Robinson GW, Rutkowski S, Schoep T, Shelat AA, Stewart CF, Sullivan M, Taylor MD, Wainwright B, Walwyn T, Weiss WA, Williamson D, Gajjar A. Medulloblastoma Down Under 2013: a report from the third annual meeting of the International Medulloblastoma Working Group. Acta neuropathologica. 2014 Feb:127(2):189-201. doi: 10.1007/s00401-013-1213-7. Epub 2013 Nov 22     [PubMed PMID: 24264598]


[35]

Bavle A, Parsons DW. From One to Many: Further Refinement of Medulloblastoma Subtypes Offers Promise for Personalized Therapy. Cancer cell. 2017 Jun 12:31(6):727-729. doi: 10.1016/j.ccell.2017.05.013. Epub     [PubMed PMID: 28609651]


[36]

Cavalli FMG, Remke M, Rampasek L, Peacock J, Shih DJH, Luu B, Garzia L, Torchia J, Nor C, Morrissy AS, Agnihotri S, Thompson YY, Kuzan-Fischer CM, Farooq H, Isaev K, Daniels C, Cho BK, Kim SK, Wang KC, Lee JY, Grajkowska WA, Perek-Polnik M, Vasiljevic A, Faure-Conter C, Jouvet A, Giannini C, Nageswara Rao AA, Li KKW, Ng HK, Eberhart CG, Pollack IF, Hamilton RL, Gillespie GY, Olson JM, Leary S, Weiss WA, Lach B, Chambless LB, Thompson RC, Cooper MK, Vibhakar R, Hauser P, van Veelen MC, Kros JM, French PJ, Ra YS, Kumabe T, López-Aguilar E, Zitterbart K, Sterba J, Finocchiaro G, Massimino M, Van Meir EG, Osuka S, Shofuda T, Klekner A, Zollo M, Leonard JR, Rubin JB, Jabado N, Albrecht S, Mora J, Van Meter TE, Jung S, Moore AS, Hallahan AR, Chan JA, Tirapelli DPC, Carlotti CG, Fouladi M, Pimentel J, Faria CC, Saad AG, Massimi L, Liau LM, Wheeler H, Nakamura H, Elbabaa SK, Perezpeña-Diazconti M, Chico Ponce de León F, Robinson S, Zapotocky M, Lassaletta A, Huang A, Hawkins CE, Tabori U, Bouffet E, Bartels U, Dirks PB, Rutka JT, Bader GD, Reimand J, Goldenberg A, Ramaswamy V, Taylor MD. Intertumoral Heterogeneity within Medulloblastoma Subgroups. Cancer cell. 2017 Jun 12:31(6):737-754.e6. doi: 10.1016/j.ccell.2017.05.005. Epub     [PubMed PMID: 28609654]


[37]

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta neuropathologica. 2016 Jun:131(6):803-20. doi: 10.1007/s00401-016-1545-1. Epub 2016 May 9     [PubMed PMID: 27157931]


[38]

Salloum R, Chen Y, Yasui Y, Packer R, Leisenring W, Wells E, King A, Howell R, Gibson TM, Krull KR, Robison LL, Oeffinger KC, Fouladi M, Armstrong GT. Late Morbidity and Mortality Among Medulloblastoma Survivors Diagnosed Across Three Decades: A Report From the Childhood Cancer Survivor Study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2019 Mar 20:37(9):731-740. doi: 10.1200/JCO.18.00969. Epub 2019 Feb 7     [PubMed PMID: 30730781]


[39]

Goschzik T, Schwalbe EC, Hicks D, Smith A, Zur Muehlen A, Figarella-Branger D, Doz F, Rutkowski S, Lannering B, Pietsch T, Clifford SC. Prognostic effect of whole chromosomal aberration signatures in standard-risk, non-WNT/non-SHH medulloblastoma: a retrospective, molecular analysis of the HIT-SIOP PNET 4 trial. The Lancet. Oncology. 2018 Dec:19(12):1602-1616. doi: 10.1016/S1470-2045(18)30532-1. Epub 2018 Nov 1     [PubMed PMID: 30392813]

Level 2 (mid-level) evidence

[40]

Khakban A, Mohammadi T, Lynd LD, Mabbott DJ, Bouffet E, Gastonguay L, Zafari Z, Malkin D, Taylor MD, Marra CA. How do parents and providers trade-off between disability and survival? Preferences in the treatment of pediatric medulloblastoma. Patient preference and adherence. 2018:12():2103-2110. doi: 10.2147/PPA.S168739. Epub 2018 Oct 10     [PubMed PMID: 30349204]