Craniopharyngioma (CP)

Craniopharyngioma (CP)



A craniopharyngioma (CP) is an embryonic malformation of the sellar region and parasellar region.

Its relation to Rathke’s cleft cyst (RCC) is controversial, and both lesions have been hypothesized to lie on a continuum of ectodermal cystic lesions of the sellar region.


Jakob Erdheim (1874-1937) was a Viennese pathologist who identified and defined a category of pituitary tumors known as craniopharyngiomas. He named these lesions “hypophyseal duct tumors” (Hypophysenganggeschwülste), a term denoting their presumed origin from cell remnants of the hypophyseal duct, the embryological structure through which Rathke’s pouch migrates to form part of the pituitary gland. He described the two histological varieties of these lesions as the adamantinomatous and the squamous-papillary types. He also classified the different topographies of craniopharyngiomas along the hypothalamus-pituitary axis. Finally, he provided the first substantial evidence for the functional role of the hypothalamus in the regulation of metabolism and sexual functions. Erdheim’s monograph on hypophyseal duct tumors elicited interest in the clinical effects and diagnosis of pituitary tumors. It certainly contributed to the development of pituitary surgery and neuroendocrinology. Erdheim’s work was greatly influenced by the philosophy and methods of research introduced to the Medical School of Vienna by the prominent pathologist Carl Rokitansky. Routine practice of autopsies in all patients dying at the Vienna Municipal Hospital (Allgemeines Krankenhaus), as well as the preservation of rare pathological specimens in a huge collection stored at the Pathological-Anatomical Museum, represented decisive policies for Erdheim’s definition of a new category of epithelial hypophyseal growths. Because of the generalized use of the term craniopharyngioma, which replaced Erdheim’s original denomination, his seminal work on hypophyseal duct tumors is only referenced in passing in most articles and monographs on this tumor.

Jakob Erdheim should be recognized as the true father of craniopharyngiomas 1).

Its relation to Rathke’s cleft cyst (RCC) is controversial, and both lesions have been hypothesized to lie on a continuum of cystic ectodermal lesions of the sellar region.

It grows close to the optic nervehypothalamus and pituitary gland.


Craniopharyngiomas frequently grow from remnants of the Rathke pouch, which is located on the cisternal surface of the hypothalamic region. These lesions can also extend elsewhere in the infundibulohypophyseal axis.

These tumors can also grow from the infundibulum or tuber cinereum on the floor of the third ventricle, developing exclusively into the third ventricle.

Genetic and immunological markers show variable expression in different types of CraniopharyngiomaBRAF is implicated in tumorigenesis in papillary Craniopharyngioma (pCP), whereas CTNNB1 and EGFR are often overexpressed in adamantinomatous Craniopharyngioma (aCP) and VEGF is overexpressed in aCP and Craniopharyngioma recurrence. Targeted treatment modalities inhibiting thesepathways can shrink or halt progression of CP. In addition, Epidermal growth factor receptor tyrosine kinase inhibitors may sensitize tumors to radiation therapy. These – drugs show promise in medical management and neoadjuvant therapy for CP. Immunotherapy, including anti-interleukin 6 (IL-6) drugs and interferon treatment, are also effective in managing tumor growth. Ongoing – clinical trials in CP are limited but are testing BRAF/MET inhibitors and IL-6 monoclonal antibodies.

Genetic and immunological markers show variable expression in different subtypes of CP. Several current molecular treatments have shown some success in the management of this disease. Additional clinical trials and targeted therapies will be important to improve CP patient outcomes 2).

Rathke’s cleft cyst.


ependymomapilocytic astrocytomachoroid plexus papilloma (CPP), craniopharyngiomaprimitive neuroectodermal tumor (PNET), choroid plexus carcinoma (CPC), immature teratomaatypical teratoid rhabdoid tumor (AT/RT), anaplastic astrocytoma, and gangliocytoma.


Compared with craniopharyngiomas, sellar gliomas presented with a significantly lower ratio of visual disturbances, growth hormone deficiencies, lesion cystic changes, and calcification. Sellar gliomas had significantly greater effects on the patients’ mentality and anatomical brain stem involvement 3).

Simultaneous sellar-suprasellar craniopharyngioma and intramural clival chordoma, successfully treated by a single staged, extended, fully endoscopic endonasal approach, which required no following adjuvant therapy is reported 4).


1)

Pascual JM, Rosdolsky M, Prieto R, Strauβ S, Winter E, Ulrich W. Jakob Erdheim (1874-1937): father of hypophyseal-duct tumors (craniopharyngiomas). Virchows Arch. 2015 Jun 19. [Epub ahead of print] PubMed PMID: 26089144.
2)

Reyes M, Taghvaei M, Yu S, Sathe A, Collopy S, Prashant GN, Evans JJ, Karsy M. Targeted Therapy in the Management of Modern Craniopharyngiomas. Front Biosci (Landmark Ed). 2022 Apr 20;27(4):136. doi: 10.31083/j.fbl2704136. PMID: 35468695.
3)

Deng S, Li Y, Guan Y, Xu S, Chen J, Zhao G. Gliomas in the Sellar Turcica Region: A Retrospective Study Including Adult Cases and Comparison with Craniopharyngioma. Eur Neurol. 2014 Dec 18;73(3-4):135-143. [Epub ahead of print] PubMed PMID: 25531372.
4)

Iacoangeli M, Rienzo AD, Colasanti R, Scarpelli M, Gladi M, Alvaro L, Nocchi N, Scerrati M. A rare case of chordoma and craniopharyngioma treated by an endoscopic endonasal, transtubercular transclival approach. Turk Neurosurg.2014;24(1):86-9. doi: 10.5137/1019-5149.JTN.7237-12.0. PubMed PMID: 24535799.

Glioblastoma immunotherapy

Glioblastoma immunotherapy



Immunotherapy has shown promising success in a variety of solid tumor types, but efficacy in glioblastoma is yet to be demonstrated. Barriers to the success of immunotherapy in glioblastoma include a heterogeneous tumor cell population, a highly immunosuppressive microenvironment, and the blood-brain barrier, to name a few. Several immunotherapeutic approaches are actively being investigated and developed to overcome these limitations 1)


Immunotherapy approaches include the use of checkpoint inhibitors, chimeric antigen receptor (CAR) T-Cell therapy, vaccine-based approaches, viral vector therapies, and cytokine-based treatment 2)


Future strategies to ameliorate the efficacy of immunotherapy and facilitate clinical translation will be provided to address the unmet medical needs of GBM 3).


With the success of immunotherapy in other aggressive cancers such as advanced melanoma and advanced non-small cell lung cancer, glioblastoma has been brought to the forefront of immunotherapy research 4).


Immunotherapy, has become a promising strategy with the ability to penetrate the blood-brain barrier since the pioneering discovery of lymphatics in the central nervous system.


The anti-tumoral contribution of Gamma delta T cells depends on their activation and differentiation into effectors. This depends on different molecules and membrane receptors, which conditions their physiology. Belghali et al. aimed to determine the phenotypic characteristics of γδT cells in glioblastoma (Glioblastoma) according to five layers of membrane receptors.

Among ten Glioblastoma cases initially enrolled, five of them who had been confirmed by pathological examination and ten healthy controls underwent phenotyping of peripheral γδT cells by flow cytometry, using the following staining: αβTCR, γδTCR, CD3, CD4, CD8, CD16, CD25, CD27, CD28, CD45, CD45RA, CD56, NKG2D, CD272(BTLA) and CD279(PD-1).

Compared to controls, the results showed no significant change in the number of γδT cells. However, they noted a decrease of double-negative (CD4- CD8- ) Tγδ cells and an increase of naive γδT cells, a lack of CD25 expression, a decrease of the expression of CD279, and a remarkable, but not significant increase in the expression of the CD27 and CD28 costimulation markers. Among γδT cell subsets, the number of Vδ2 decreased in Glioblastoma and showed no significant difference in the expression of CD16, CD56, and NKG2D. In contrast, the number of Vδ1 increased in Glioblastoma with overexpression of CD16, CD56, and NKG2D.

The results showed that γδT cells are prone to adopt a pro-inflammatory profile in the Glioblastoma’s context, which suggests that they might be a potential tool to consider in T cell-based glioblastoma immunotherapy. However, this requires additional investigation on a larger sample size 5).


A limited number of phase III trials have been completed for checkpoint inhibitorvaccine, as well as gene therapies, and have been unable to show improvement in survival outcomes. Nevertheless, these trials have also shown these strategies to be safe and promising with further adaptations. Further large-scale studies for chimeric antigen receptors T cell therapies and viral therapies are anticipated. Many current trials are broadening the number of antigens targeted and modulating the microtumor microenvironment to abrogate early mechanisms of resistance. Future Glioblastoma treatment will also likely require synergistic effects by combination regimens 6).


As the pioneer and the main effector cells of immunotherapy, T cells play a key role in tumor immunotherapy.

For glioblastoma, immunotherapy has not been as effective 7) , the T cells in Glioblastoma microenvironment are inhibited by the highly immunosuppressive environment of Glioblastoma, (cold tumor microenvironment) posing huge challenges to T cell-based Glioblastoma immunotherapy 8) 9) 10).

As these tumors do not attract and activate immune cells, approaches based on educating immune cells on killing tumor cells, utilized in “hot” immuno-activating cancers, have not been successful in brain tumor clinical trials. In this context, the use of immune-stimulatory approaches, like therapy with oncolytic viruses (OV), is promising 11)


Xu et al. detailed the management of gliomas and previous studies assessing different immunotherapies in gliomas, despite the fact that the associated clinical trials have not been completed yet. Moreover, several drugs that have undergone clinical trials are listed as novel strategies for future application; however, these clinical trials have indicated limited efficacy in glioma. Therefore, additional studies are warranted to evaluate novel therapeutic approaches in glioma treatment 12).


Earlier forms of immune-based platforms have now given way to more current approaches, including chimeric antigen receptor T-cells, personalized neoantigen vaccines, oncolytic viruses, and checkpoint blockade 13).

Critical to mapping a path forward will be the systematic characterization of the immunobiology of glioblastoma utilizing currently available, state of the art technologies. Therapeutic approaches aimed at driving effector immune cells into the glioblastoma microenvironment as well as overcoming immunosuppressive myeloid cells, physical factors, and cytokines, as well as limiting the potentially detrimental, iatrogenic impact of dexamethasone, will likely be required for the potential of anti-tumor immune responses to be realized for glioblastoma 14).

Patients with glioblastoma (Glioblastoma) exhibit a complex state of immunodeficiency involving multiple mechanisms of local, regional, and systemic immune suppression and tolerance. These pathways are now being identified and their relative contributions explored. Delineating how these pathways are interrelated is paramount to effectively implementing immunotherapy for Glioblastoma 15).


Progress in the development of these therapies for glioblastoma has been slow due to the lack of immunogenic antigen targets that are expressed uniformly and selectively by gliomas.

Trials have revealed promising trends in overall survival and progression free survival for patients with glioblastoma, and have paved the way for ongoing randomized controlled trials 16) 17)


Some clinical trials are reaching phase III. Significant progress has been made in unraveling the molecular and genetic heterogeneity of glioblastoma multiforme and its implications to disease prognosis. There is now consensus related to the critical need to incorporate tumor heterogeneity into the design of therapeutic approaches. Recent data also indicates that an efficacious treatment strategy will need to be combinatorial and personalized to the tumor genetic signature 18).


A recurrent theme of this work is that immunotherapy is not a one-size-fits-all solution. Rather, dynamic, tumor-specific interactions within the tumor microenvironment continually shape the immunologic balance between tumor elimination and escape. High-grade gliomas are a particularly fascinating example. These aggressive, universally fatal tumors are highly resistant to radiation and chemotherapy and inevitably recur after surgical resection. Located in the immune-privileged central nervous system, high-grade gliomas also employ an array of defenses that serve as direct impediments to immune attack. Despite these challenges, vaccines have shown activity against high-grade gliomas and anecdotal, preclinical, and early clinical data bolster the notion that durable remission is possible with immunotherapy. Realizing this potential, however, will require an approach tailored to the unique aspects of glioma biology 19).


Clinical experiences with active specific immunotherapy demonstrate feasibility, safety and most importantly, but incompletely understood, prolonged long-term survival in a fraction of the patients. In relapsed patients, Van Gool et al developed an immunotherapy schedule and categorized patients into clinically defined risk profiles. He learned how to combine immunotherapy with standard multimodal treatment strategies for newly diagnosed glioblastoma multiforme patients. The developmental program allows further improvements related to newest scientific insights. Finally, he developed a mode of care within academic centers to organize cell therapy for experimental clinical trials in a large number of patients 20).


Immunostimulating oligodeoxynucleotides containing unmethylated cytosineguanosine motifs (CpG-ODN) have shown a promising efficacy in several cancer models when injected locally. A previous phase II study of CpG-ODN in patients with Glioblastoma recurrence (Glioblastoma) has suggested some activity and has shown a limited toxicity. This multicentre single-blinded randomised phase II trial was designed to study the efficacy of a local treatment by CpG-ODN in patients with de novo glioblastomas.

Patients with a newly diagnosed glioblastoma underwent large surgical resection and CpG-ODN was randomly administrated locally around the surgical cavity. The patients were then treated according to standard of care (SOC) with radiotherapy and temozolomide. The primary objective was 2-year survival. Secondary outcomes were progression free survival (PFS), and tolerance.

Eighty-one (81) patients were randomly assigned to receive CpG-ODN plus SOC (39 patients) or SOC (42 patients). The 2-year overall survival was 31% (19%; 49%) in the CpG-ODN arm and 26% (16%; 44%) in the SOC arm. The median PFS was 9 months in the CpG-ODN arm and 8.5 months in the SOC arm. The incidence of adverse events was similar in both arms; although fever and post-operative haematoma were more frequent in the CpG-ODN arm.

Local immunotherapy with CpG-ODN injected into the surgical cavity after tumour removal and followed by SOC, although well tolerated, does not improve survival of patients with newly diagnosed Glioblastoma 21).


Epidermal growth factor receptor 3 (EGFRvIII) is present in approximately one-third of glioblastoma (Glioblastoma) patients. It is never found in normal tissues; therefore, it represents a candidate target for glioblastoma immunotherapy. PEPvIII, a peptide sequence from EGFRvIII, was designed to represent a target of glioma and is presented by MHC I/II complexes. Dendritic cells (DCs) have great potential to sensitize CD4+ T and CD8+ T cells to precisely target and eradicate Glioblastoma.

Li et al. show that PEPvIII could be loaded by DCs and presented to T lymphocytes, especially PEPvIII-specific CTLs, to precisely kill U87-EGFRvIII cells. In addition to inhibiting proliferation and inducing the apoptosis of U87-EGFRvIII cells, miR-326 also reduced the expression of TGF-β1 in the tumour environment, resulting in improved efficacy of T cell activation and killing via suppressing the SMO/Gli2 axis, which at least partially reversed the immunosuppressive environment. Furthermore, combining the EGFRvIII-DC vaccine with miR-326 was more effective in killing U87-EGFRvIII cells compared with the administration of either one alone. This finding suggested that a DC-based vaccine combined with miR-326 may induce more powerful anti-tumour immunity against Glioblastoma cells that express a relevant antigen, which provides a promising approach for Glioblastoma immunotherapy 22).

Yuan et al. provided an overview of the basic knowledge underlying immune targeting and promising immunotherapeutic strategies including CAR T cells, oncolytic viruses, cancer vaccines, and checkpoint blockade inhibitors that have been recently investigated in glioblastoma. Current clinical trials and previous clinical trial findings are discussed, shedding light on novel strategies to overcome various limitations and challenges 23).


Rui Y, Green JJ. Overcoming delivery barriers in immunotherapy for glioblastoma. Drug Deliv Transl Res. 2021 May 30. doi: 10.1007/s13346-021-01008-2. Epub ahead of print. PMID: 34053034.


1)

Zaidi SE, Moelker E, Singh K, Mohan A, Salgado MA, Essibayi MA, Hotchkiss K, Shen S, Lee W, Sampson J, Khasraw M. Novel Immunotherapeutic Approaches for the Treatment of Glioblastoma. BioDrugs. 2023 May 31. doi: 10.1007/s40259-023-00598-2. Epub ahead of print. PMID: 37256535.
2)

Sener U, Ruff MW, Campian JL. Immunotherapy in Glioblastoma: Current Approaches and Future Perspectives. Int J Mol Sci. 2022 Jun 24;23(13):7046. doi: 10.3390/ijms23137046. PMID: 35806051; PMCID: PMC9266573.
3)

Bausart M, Préat V, Malfanti A. Immunotherapy for glioblastoma: the promise of combination strategies. J Exp Clin Cancer Res. 2022 Jan 25;41(1):35. doi: 10.1186/s13046-022-02251-2. PMID: 35078492; PMCID: PMC8787896.
4)

Yu MW, Quail DF. Immunotherapy for Glioblastoma: Current Progress and Challenges. Front Immunol. 2021 May 13;12:676301. doi: 10.3389/fimmu.2021.676301. Erratum in: Front Immunol. 2021 Oct 07;12:782687. PMID: 34054867; PMCID: PMC8158294.
5)

Belghali MY, El Moumou L, Hazime R, Brahimi M, El Marrakchi M, Belaid HA, Benali SA, Khouchani M, Ba-M’hamed S, Admou B. Phenotypic characterization of human peripheral γδT-Cell subsets in glioblastoma. Microbiol Immunol. 2022 Jun 19. doi: 10.1111/1348-0421.13016. Epub ahead of print. PMID: 35718749.
6)

Zhang M, Choi J, Lim M. Advances in Immunotherapies for Gliomas. Curr Neurol Neurosci Rep. 2022 Feb 2. doi: 10.1007/s11910-022-01176-9. Epub ahead of print. PMID: 35107784.
7)

Bovenberg MS, Degeling MH, Tannous BA. Cell-based immunotherapy against gliomas: from bench to bedside. Mol Ther. 2013 Jul;21(7):1297-305. doi: 10.1038/mt.2013.80. Epub 2013 May 7. PMID: 23648695; PMCID: PMC3702108.
8)

Wang H, Zhou H, Xu J, Lu Y, Ji X, Yao Y, Chao H, Zhang J, Zhang X, Yao S, Wu Y, Wan J. Different T-cell subsets in glioblastoma multiforme and targeted immunotherapy. Cancer Lett. 2020 Oct 3:S0304-3835(20)30498-5. doi: 10.1016/j.canlet.2020.09.028. Epub ahead of print. PMID: 33022290.
9)

Lim M., Xia Y., Bettegowda C., Weller M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018;15:422–442. doi: 10.1038/s41571-018-0003-5.
10)

Reardon D.A., Wucherpfennig K., Chiocca E.A. Immunotherapy for glioblastoma: On the sidelines or in the game? Discov. Med. 2017;24:201–208.
11)

Iorgulescu JB, Reardon DA, Chiocca EA, Wu CJ. Immunotherapy for glioblastoma: going viral. Nat Med. 2018 Aug;24(8):1094-1096. doi: 10.1038/s41591-018-0142-3. PMID: 30082860; PMCID: PMC6443579.
12)

Xu S, Tang L, Li X, Fan F, Liu Z. Immunotherapy for glioma: current management and future application. Cancer Lett. 2020 Feb 7. pii: S0304-3835(20)30056-2. doi: 10.1016/j.canlet.2020.02.002. [Epub ahead of print] PubMed PMID: 32044356.
13)

Fecci PE, Sampson JH. The current state of immunotherapy for gliomas: an eye toward the future. J Neurosurg. 2019 Sep 1;131(3):657-666. doi: 10.3171/2019.5.JNS181762. Review. PubMed PMID: 31473668.
14)

Reardon DA, Wucherpfennig K, Chiocca EA. Immunotherapy for glioblastoma: on the sidelines or in the game? Discov Med. 2017 Nov;24(133):201-208. PubMed PMID: 29278673.
15)

Jackson CM, Lim M. Immunotherapy for glioblastoma: playing chess, not checkers. Clin Cancer Res. 2018 Apr 24. pii: clincanres.0491.2018. doi: 10.1158/1078-0432.CCR-18-0491. [Epub ahead of print] PubMed PMID: 29691293.
16)

Thomas AA, Fisher JL, Ernstoff MS, Fadul CE. Vaccine-based immunotherapy for glioblastoma. CNS Oncol. 2013 Jul;2(4):331-49. doi: 10.2217/cns.13.29. PubMed PMID: 25054578.
17)

Agrawal NS, Miller R Jr, Lal R, Mahanti H, Dixon-Mah YN, DeCandio ML, Vandergrift WA 3rd, Varma AK, Patel SJ, Banik NL, Lindhorst SM, Giglio P, Das A. Current Studies of Immunotherapy on Glioblastoma. J Neurol Neurosurg. 2014 Apr 5;1(1). pii: 21000104. PubMed PMID: 25346943.
18)

Kamran N, Calinescu A, Candolfi M, Chandran M, Mineharu Y, Assad AS, Koschmann C, Nunez F, Lowenstein P, Castro M. Recent advances and future of immunotherapy for glioblastoma. Expert Opin Biol Ther. 2016 Jul 13. [Epub ahead of print] PubMed PMID: 27411023.
19)

Jackson CM, Lim M, Drake CG. Immunotherapy for Brain Cancer: Recent Progress and Future Promise. Clin Cancer Res. 2014 Apr 25. [Epub ahead of print] PubMed PMID: 24771646.
20)

Van Gool SW. Brain Tumor Immunotherapy: What have We Learned so Far? Front Oncol. 2015 Jun 17;5:98. eCollection 2015. Review. PubMed PMID: 26137448.
21)

Ursu R, Carpentier A, Metellus P, Lubrano V, Laigle-Donadey F, Capelle L, Guyotat J, Langlois O, Bauchet L, Desseaux K, Tibi A, Chinot O, Lambert J, Carpentier AF. Intracerebral injection of CpG oligonucleotide for patients with de novo glioblastoma-A phase II multicentric, randomised study. Eur J Cancer. 2017 Jan 28;73:30-37. doi: 10.1016/j.ejca.2016.12.003. [Epub ahead of print] PubMed PMID: 28142059.
22)

Li J, Wang F, Wang G, Sun Y, Cai J, Liu X, Zhang J, Lu X, Li Y, Chen M, Chen L, Jiang C. Combination epidermal growth factor receptor variant III peptide-pulsed dendritic cell vaccine with miR-326 results in enhanced killing on EGFRvIII-positive cells. Oncotarget. 2017 Feb 17. doi: 10.18632/oncotarget.15445. [Epub ahead of print] PubMed PMID: 28412740.
23)

Yuan B, Wang G, Tang X, Tong A, Zhou L. Immunotherapy of glioblastoma: recent advances and future prospects. Hum Vaccin Immunother. 2022 Mar 28:1-16. doi: 10.1080/21645515.2022.2055417. Epub ahead of print. PMID: 35344682.

Superior sagittal sinus dural arteriovenous fistula

Superior sagittal sinus dural arteriovenous fistula

intracranial dural arteriovenous fistula (dAVF) involving the superior sagittal sinus (SSS) is relatively rare, and its clinical course is usually aggressive. Its concomitance with a tumor has rarely been reported.


Supratentorial dural arteriovenous fistula DVAs mostly drained in the superior sagittal sinus (80%), while all of infratentorial/combined DVAs drained in deep ependymal veins of the 4th ventricle. All the supratentorial dAVFs drained into the superior sagittal sinus, while the infratentorial/combined dAVFs mostly drained in the jugular bulb, Vein of Rosenthal, or transverse-sigmoid sinuses (75%) 1).


Gigliotti et al. reported the first case of a superior sagittal sinus DAVF occurring after surgical resection of a parasagittal meningioma 2).

A case of SSS dAVF due to meningioma invasion, which was treated with sinus reconstruction and endovascular embolization. A 75-year-old man who had undergone tumor resection for parasagittal meningioma 4 years prior presented with intra-ventricular hemorrhage. Computed tomography angiography and magnetic resonance imaging revealed recurrent tumor invasion into the SSS causing occlusion. Cerebral angiography revealed multiple shunts along the occluded segment of the SSS, diffuse deep venous congestion, and cortical reflux. Borden type 3 SSS dAVF was diagnosed. We first performed direct tumor resection, followed by stenting for the occluded SSS and partial embolization of the shunts. After a 6-month interval, transvenous occlusion of the SSS was performed along the stent, resulting in complete obliteration of the dAVF. Sinus reconstruction therapy was effective in the immediate improvement of venous hypertension, obtaining the access route to the fistulas, and eradicating the shunts 3)


A 78-year-old man presented after trauma with basal and cortical subarachnoid hemorrhage (SAH). Computed tomography revealed a parietal bone fracture overlying the superior sagittal sinus (SSS). Catheter angiography performed within 24 hours of the injury demonstrated an SSS dAVF supplied by the middle meningeal artery, adjacent to the fracture.

Lessons: The authors present the case of an acute traumatic dAVF adjacent to a calvarial fracture. In this case, the authors proprose that the underlying pathogenesis is suggestive of direct vessel injury rather than the pathway commonly associated with this pathology 4)


Spontaneous closure of a superior sagittal sinus dural arteriovenous fistula after treatment of subarachnoid hemorrhage and secondary hydrocephalus 5).


A 61-year-old male with a history of meningioma previously managed with subtotal resection and stereotactic radiosurgery presented with progressive right-sided vision loss and bilateral papilledema. Initial imaging suggested possible sinus occlusion. Catheter angiogram revealed a Borden-Shucart grade III DAVF of the superior sagittal sinus and elevated venous pressures and the patient subsequently underwent endovascular transarterial intervention twice. We report on the first case of a superior sagittal sinus DAVF occurring after surgical resection of a parasagittal meningioma 6).


sagittal sinus dural arteriovenous fistula manifesting as dysphonia secondary to vocal cord paresis. The patient presented with a 6-week history of hoarseness. Imaging studies demonstrated findings suggestive of a dural arteriovenous fistula affecting the superior sagittal sinus. Direct laryngoscopy demonstrated paresis of the right vocal fold. We hypothesized that pressure on the vagus nerve from a dilated and arterialized internal jugular vein within the jugular foramen was responsible for the cranial neuropathy. The patient’s dysphonia resolved with embolization of the fistula, and repeat laryngoscopy showed resolution of the vocal fold paresis 7).


Beer-Furlan A, Joshi KC, Dasenbrock HH, Chen M. Endovascular management of complex superior sagittal sinus dural arteriovenous fistula. Neurosurg Focus. 2019 Apr 1;46(Suppl_2):V11. doi: 10.3171/2019.2.FocusVid.18687. PMID: 30939439.


Song W, Sun H, Liu J, Liu L, Liu J. Spontaneous Resolution of Venous Aneurysms After Transarterial Embolization of a Variant Superior Sagittal Sinus Dural Arteriovenous Fistula: Case Report and Literature Review. Neurologist. 2017 Sep;22(5):186-195. doi: 10.1097/NRL.0000000000000137. Review. PubMed PMID: 28859024.


A DAVF of the SSS in a patient who presented uniquely with increasing dizziness and disequilibrium who was treated with a single modality, endovascular embolization with ethyl vinyl alcohol co-polymer (Onyx, EV3, Irvine, CA). The patient underwent staged embolization in 2 sessions with no complications. An angiographic cure was achieved and the patient’s symptoms were ameliorated. Single modality therapy with endovascular embolization of a SSS DAVF can be achieved. Careful attention to technique during embolization with Onyx is required, but complete obliteration is possible without the need for adjunctive surgical resection 8)


A 61-year-old man who had been treated with anticoagulation for a known SSS thrombosis presented with a sudden onset of headache. CT scan revealed an intraventricular hemorrhage and cerebral angiography revealed DAVFs involving the SSS which had severe venous congestion and sinus occlusion. We treated this case with a staged endovascular approach which consisted of stent placement for the occluded sinus and transarterial intravenous embolization resulting in complete eradication of DAVFs. Recanalization of an occluded sinus by stent placement can reduce venous congestion and transarterial intravenous embolization can obliterate dural arteriovenous shunts. This staged strategy is feasible and should be considered a first option of treatment, especially for DAVFs which presented with intracranial hemorrhage and aggressive venous hypertension 9)


A case report and review of the literature of 16 dural arteriovenous fistulas (DAVFs) involving the superior sagittal sinus region are presented. In the case, magnetic resonance angiography detected the DAVF with multiple arterial feeding vessels from both external carotid arteries. The patient was successfully treated endovascularly, with complete occlusion of arterial feeders and a total resolution of symptoms 10).


1)

Agosti E, De Maria L, Panciani PP, Serioli S, Mardighian D, Fontanella MM, Lanzino G. Developmental venous anomaly associated with dural arteriovenous fistula: Etiopathogenesis and hemorrhagic risk. Front Surg. 2023 Mar 21;10:1141857. doi: 10.3389/fsurg.2023.1141857. PMID: 37025268; PMCID: PMC10071040.
2) , 6)

Gigliotti MJ, Patel N, Simon S. Superior sagittal sinus dural arteriovenous fistula caused by treatment of meningioma masquerades as sinus thrombosis. J Cerebrovasc Endovasc Neurosurg. 2021 Sep;23(3):260-265. doi: 10.7461/jcen.2021.E2021.01.002. Epub 2021 Aug 25. PMID: 34428863; PMCID: PMC8497717.
3)

Shima S, Sato S, Kushi K, Okada Y, Niimi Y. Sinus reconstruction therapy for superior sagittal sinus dural arteriovenous fistula caused by parasagittal meningioma invasion: a case report. Neuroradiol J. 2023 May 4:19714009231173103. doi: 10.1177/19714009231173103. Epub ahead of print. PMID: 37142419.
4)

Pryce ML, Chung KHC, Zeineddine HA, Dawes BH. Acute traumatic dural arteriovenous fistula of the superior sagittal sinus: illustrative case. J Neurosurg Case Lessons. 2023 Apr 10;5(15):CASE2392. doi: 10.3171/CASE2392. PMID: 37039291.
5)

Endo H, Ishizuka T, Murahashi T, Oka K, Nakamura H. Spontaneous closure of a superior sagittal sinus dural arteriovenous fistula after treatment of subarachnoid hemorrhage and secondary hydrocephalus. Neurol Sci. 2023 Mar 30. doi: 10.1007/s10072-023-06786-w. Epub ahead of print. PMID: 36995470.
7)

Rinaldo L, Ekbom DC, Lanzino G. Sagittal sinus dural arteriovenous fistula manifesting as unilateral vocal fold paresis. Clin Neurol Neurosurg. 2021 Aug 2;208:106856. doi: 10.1016/j.clineuro.2021.106856. Epub ahead of print. PMID: 34365240.
8)

Chong BW, Demaerschalk BM. Unusual Presentation of a Dural Arteriovenous Fistula of the Superior Sagittal Sinus and Single Modality Therapy with Onyx. Radiol Case Rep. 2015 Nov 6;3(1):158. doi: 10.2484/rcr.v3i1.158. PMID: 27303511; PMCID: PMC4896129.
9)

Ohara N, Toyota S, Kobayashi M, Wakayama A. Superior sagittal sinus dural arteriovenous fistulas treated by stent placement for an occluded sinus and transarterial embolization. A case report. Interv Neuroradiol. 2012 Sep;18(3):333-40. doi: 10.1177/159101991201800314. Epub 2012 Sep 10. PMID: 22958774; PMCID: PMC3442309.
10)

Kurl S, Saari T, Vanninen R, Hernesniemi J. Dural arteriovenous fistulas of superior sagittal sinus: case report and review of literature. Surg Neurol. 1996 Mar;45(3):250-5. doi: 10.1016/0090-3019(95)00361-4. PMID: 8638222.

Glioblastoma

Glioblastoma

J.Sales-Llopis

Neurosurgery Service, Alicante University General Hospital, Spain.

While glioblastoma was historically classified as isocitrate dehydrogenase (IDH)-wildtype and IDH-mutant groups, the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT-NOW) and the World Health Organization Classification of Tumors of the Central Nervous System 2021 clearly updated the nomenclature to reflect glioblastoma to be compatible with wildtype IDH status only. Therefore, glioblastoma is now defined as “a diffuse, astrocytic glioma that is IDH-wildtype and H3-wildtype and has one or more of the following histological or genetic features: microvascular proliferationnecrosisTERT promoter mutationEpidermal growth factor receptor gene amplification, +7/-10 chromosome copy-number changes (CNS WHO grade 4) 1).

see Glioblastoma epidemiology.

Prior malignancies in patients harboring glioblastoma

Patients who develop Glioblastoma following a prior malignancy constitute ~8% of patients with Glioblastoma. Despite significant molecular differences these two cohorts appear to have a similar overall prognosis and clinical course. Thus, whether or not a patient harbors a malignancy prior to diagnosis of Glioblastoma should not exclude him or her from aggressive treatment or for consideration of novel investigational therapies 2).

Genome-wide profiling studies have shown remarkable genomic diversity among glioblastomas.

Molecular studies have helped identify at least 3 different pathways in the development of glioblastomas.

● 1st pathway: dysregulation of growth factor signaling through amplification and mutational activation of receptor tyrosine kinase (RTK) genes. RTKs are transmembrane proteins that act as receptors for an epidermal growth factor (EGF), vascular endothelial growth factor (VEGF) & platelet-derived growth factor (PDGF). They can also act as receptors for cytokines, hormones, and other signaling pathways

● 2nd pathway: activation of the Phosphoinositide 3 kinase (PI3K)/AKT/mTOR, which is an intracellular signaling pathway that is essential in regulating cell survival

● 3rd pathway: inactivation of the p53 and retinoblastoma (Rb) tumor suppressor pathways

Glioblastomas are intrinsic brain tumors thought to originate from a neuroglial stem or progenitor cells. More than 90% of glioblastomas are isocitrate dehydrogenase (IDH)-wildtype tumors. Incidence increases with age, males are more often affected. Beyond rare instances of genetic predisposition and irradiation exposure, there are no known glioblastoma risk factors.

Vessels with different microcirculation patterns are required for glioblastoma (Glioblastoma) growth. However, details of the microcirculation patterns in Glioblastoma remain unclear.

Mei et al. examined the microcirculation patterns of Glioblastoma and analyzed their roles in patient prognosis together with two well-known GMB prognosis factors (O6 -methylguanine DNA methyltransferase [MGMT] promoter methylation status and isocitrate dehydrogenase [IDH] mutations).

Eighty Glioblastoma clinical specimens were collected from patients diagnosed between January 2000 and December 2012. The microcirculation patterns, including endothelium-dependent vessels (EDVs), extracellular matrix-dependent vessels (ECMDVs), Glioblastoma cell-derived vessels (GDVs), and mosaic vessels (MVs), were evaluated by immunohistochemistry (IHC) and immunofluorescence (IF) staining in both Glioblastoma clinical specimens and xenograft tissues. Vascular density assessments and three-dimensional reconstruction were performed. MGMT promoter methylation status was determined by methylation-specific PCR, and IDH1/2 mutations were detected by Sanger sequencing. The relationship between the microcirculation patterns and the patient prognosis was analyzed by the Kaplan-Meier method.

All 4 microcirculation patterns were observed in both Glioblastoma clinical specimens and xenograft tissues. EDVs was detected in all tissue samples, while the other three patterns were observed in a small number of tissue samples (ECMDVs in 27.5%, GDVs in 43.8%, and MVs in 52.5% tissue samples). GDV-positive patients had a median survival of 9.56 months versus 13.60 months for GDV-negative patients (P = 0.015). In MGMT promoter-methylated cohort, GDV-positive patients had a median survival of 6.76 months versus 14.23 months for GDV-negative patients (P = 0.022).

GDVs might be a negative predictor for the survival of Glioblastoma patients, even in those with MGMT promoter methylation 3).

It generally presents with epilepsycognitive declineheadachedysphasia, or progressive hemiparesis4).

Seizures as the presenting symptom of glioblastoma predicted longer survival in adults younger than 60 years. The IDH1 R132H mutation and p53 overexpression (>40%) were associated with seizures at presentation. Seizures showed no relationship with the tumor size or proliferation parameters 5).


1)

Chen J, Han P, Dahiya S. Glioblastoma: Changing concepts in the WHO CNS5 classification. Indian J Pathol Microbiol. 2022 May;65(Supplement):S24-S32. doi: 10.4103/ijpm.ijpm_1109_21. PMID: 35562131.
2)

Zacharia BE, DiStefano N, Mader MM, Chohan MO, Ogilvie S, Brennan C, Gutin P, Tabar V. Prior malignancies in patients harboring glioblastoma: an institutional case-study of 2164 patients. J Neurooncol. 2017 May 27. doi: 10.1007/s11060-017-2512-y. [Epub ahead of print] Review. PubMed PMID: 28551847.
3)

Mei X, Chen YS, Zhang QP, Chen FR, Xi SY, Long YK, Zhang J, Cai HP, Ke C, Wang J, Chen ZP. Association between glioblastoma cell-derived vessels and poor prognosis of the patients. Cancer Commun (Lond). 2020 May 2. doi: 10.1002/cac2.12026. [Epub ahead of print] PubMed PMID: 32359215.
4)

Thomas DGT,Graham DI, McKeran RO,Thomas DGT. The clinical study of gliomas. In: Brain tumours: scientific basis, clinical investigation and current therapy. In: Thomas DGT, Graham DI eds. London: Butterworths, 1980:194–230.
5)

Toledo M, Sarria-Estrada S, Quintana M, Maldonado X, Martinez-Ricarte F, Rodon J, Auger C, Aizpurua M, Salas-Puig J, Santamarina E, Martinez-Saez E. Epileptic features and survival in glioblastomas presenting with seizures. Epilepsy Res. 2016 Dec 26;130:1-6. doi: 10.1016/j.eplepsyres.2016.12.013. [Epub ahead of print] PubMed PMID: 28073027.

Pituitary neuroendocrine tumor

Pituitary neuroendocrine tumor

Pituitary tumors have very few known risk factors, and these are related to genetics. There are no known environmental or lifestyle-related risk factors for pituitary tumors. Though science has suggested that people who are overweight or obese might be at increased risk.


Youn et al. discovered that a 3’untranslated region (3’UTR) variant, rs181031884 of CDKN2B (Asian-specific variant), had significant association with the risk of pituitary neuroendocrine tumor (PA) (Odds ratio = 0.58, P = 0.00003). Also, rs181031884 appeared as an independent causal variant among the significant variants in CDKN2A and CDKN2B, and showed dose-dependent effects on PA.

Although further studies are needed to verify the impact of this variant on pituitary neuroendocrine tumor susceptibility, the results may help to understand CDKN2B polymorphism and the risk of pituitary neuroendocrine tumor 1).

A 45-year-old woman who suffered from limb edema for 2 months. Dong et al. focused on tumor recurrence and other common potential diseases based on the pituitary neuroendocrine tumor history. However, none of the examinations showed any abnormality. Later, her continuous complaints about the family relationship and depressed mood came into sight, and a psychiatry consultation was arranged. Following that, she was diagnosed with major depressive disorder. After several days of Melitracen and tandospirone treatment, the patient’s limb edema dramatically subsided. This is the first case of limb edema associated with depression. This highlights the importance of awareness of mental illness for non-psychiatrists, especially in patients with severe somatic symptoms, but with negative results 2).


A 39-year-old woman reports visual loss (blurred vision) in both eyes for 2 months, the left worse than the right. Refers to headache (twice a week) that subsides with paracetamol In the last 15 days, he has woken up at night several days due to headaches.

Migraines under control by Neurology

Antiphospholipid syndrome antibody/hypercoagulability in follow-up by Hematology

APA is positive at low titer.

Bilateral superior external quadrantanopia

Treatment with Zolmitriptan

Computed tomography

Suprasellar mass is observed that occupies and expands the sella turcica, difficult to define, which produces a break in the continuity of the floor of the sella turcica and a complete occupation of the sphenoid sinus, which also presents an expansion of the same and thinning of its bony walls. It has maximum measurements of 3.5 x 2.8 x 1.2 cm (craniocaudal by transverse by anteroposterior). These findings seem compatible with an aggressive pituitary neuroendocrine tumor

The mass ascends through the sellar diaphragm and compresses the optic chiasm, and in the lateral areas, it completely encompasses both intracavernous carotid arteries and both cavernous sinuses bilaterally.

Brain MRI

A large sellar lesion with a marked isointense suprasellar extension on T1 and heterogeneous on T2 with multiple hyperintense foci on T2, especially the cystic-necrotic suprasellar portion in relation to macroadenoma. The lesion measures approximately 4 cm in diameter craniocaudal, 3.9 anteroposterior, and 4.2 cm transverse. It extends to both cavernous sinuses and surrounds the right internal carotid artery for more than 180° and the left for approximately 180°. It causes a mass effect on the optic chiasm, displacing it superiorly. The pituitary stalk seems to be located anterior to the lesion with a slight right lateralization, although it is difficult to locate it due to the large size of the lesion.

HORMONES

FREE T4 1.7 ng/dL

FSH 5.3 U/L

LH 3.4 U/L

PROLACTIN 8.3ng/mL

ESTRADIOL 32.0 pg/mL

CORTISOL MORNING 10.9 µg/dL

IGF-1 231 ng/mL


Under general anesthesia, orotracheal intubation, and antibiotic prophylaxis with cefazolin 2 gr IV. Supine position with neutral head resting on a donut-type pillow. Preoperative topical intranasal oxymetazoline was applied with lectins.

nasal phase: Right middle turbinate resection. Preparation of a nasoseptal flap with mucosa from the right septum. It is left lodged in the right choana. Posterior septostomy and communication of both nostrils. In the ostium, a tumor is visualized that completely occupies the sphenoid sinus. Wide anterior sphenoidotomy with the help of a laminotome and cutting burr. Profuse bleeding throughout the nasal phase comes from the tumor. Part of the left paramedian septum that was encompassed by the tumor was removed.

Excision phase: Excision of the tumor part contained in the sphenoid sinus until the bony limits of the sella turcica were visualized. With the help of neuronavigation and Doppler, both ICAs were located. Clivus partly eroded. In the most inferior and posterior part, a bone area corresponding to the posterior clinoid is observed, which is moth-eaten and loose, encompassed by a tumor. Intracapsular excision of the tumor is started by way of debulking, and sending tumor samples for AP analysis. The tumor shows a friable consistency and a purplish color compatible with a pituitary neuroendocrine tumor. Central excision until visualizing gradual descent of sellar and arachnoid diagrams in the sellar cavity with contained low-flow fistula. Exeresis in the posterior region until observing the dura mater of the posterior fossa. Excision of the lateral walls and part of the cavernous sinus. Hemostasis with Floseal.

Reconstruction phase: Tachosil is placed covering the arachnoid in the area of ​​the contained fistula. A nasoseptal flap is placed in contact with the bone defect around the sellar opening. The flap is fixed with surgicel and tissucol. Rapid -Rhino binasal tires are left. The free mucosa of the middle turbinate is left covering the part of the septum from which the flap has been removed.


1)

Youn BJ, Cheong HS, Namgoong S, Kim LH, Baek IK, Kim JH, Yoon SJ, Kim EH, Kim SH, Chang JH, Kim SH, Shin HD. Asian-specific 3’UTR variant in CDKN2B associated with risk of pituitary neuroendocrine tumor. Mol Biol Rep. 2022 Sep 12. doi: 10.1007/s11033-022-07796-1. Epub ahead of print. PMID: 36097105.
2)

Dong X, Fang S, Li W, Li X, Zhang S. Deanxit and tandospirone relieved unexplained limb edema in a depressed pituitary neuroendocrine tumor survivor: A case report. Front Psychiatry. 2022 Nov 10;13:965495. doi: 10.3389/fpsyt.2022.965495. PMID: 36440410; PMCID: PMC9685525.

Adamantinomatous craniopharyngioma diagnosis

Adamantinomatous craniopharyngioma diagnosis

Diagnosis of adamantinomatous craniopharyngioma (ACP) is predominantly determined through invasive pathological examination of a neurosurgical biopsy specimen.

▷ ESSENTIAL ◁

Tumor in the sellar region

Squamous non-keratinizing epitheliumbenign

AND

stellate reticulum and/or wet keratin

▷ DESIRABLE ◁

▶ Nuclear immunoreactivity for β-catenin

▶ Mutation in CTNNB1

▶ Absence of BRAF p.V600E mutation


Adamantinomatous craniopharyngiomas typically have a lobulated contour as a result of usually being multiple cystic lesions. Solid components are present, but often form a relatively minor part of the mass and enhance vividly on both CT and MRI. Overall, calcification is very common, but this is only true of the adamantinomatous subtype (~90% are calcified).

These tumors have a predilection to being large, extending superiorly into the third ventricle, encasing vessels, and even adhering to adjacent structures.

Contrast enhancementcyst formation, and calcification are the three characteristic features of craniopharyngiomas on computed tomographic scans. More than 90% of suprasellar craniopharyngiomas exhibit at least two of these three features, thus providing easy radiologic detection. Imaging mnemonic: “90% rule” 90% of adamantinomatous craniopharyngiomas exhibit at least 2 of the following “C” features: cyst formation, prominent calcifications. 1)

T1: iso- to hyperintense to the brain (due to high protein content “motor oil cysts”)

T2: variable but ~80% are mostly or partly T2 hyperintense

T1 C+ (Gd): vivid enhancement

T2: variable or mixed

Difficult to appreciate on conventional imaging

Susceptible sequences may better demonstrate calcification

May show displacement of the A1 segment of the anterior cerebral artery (ACA)

Cyst contents may show a broad lipid spectrum, with an otherwise flat baseline.


Clinical experts can distinguish ACP from Magnetic Resonance Imaging (MRI) with an accuracy of 86%, and 9% of ACP cases are diagnosed this way. Classification using deep learning (DL) provides a solution to support a non-invasive diagnosis of ACP through neuroimaging, but it is still limited in implementation, a major reason being the lack of predictive uncertainty representation. We trained and tested a DL classifier on preoperative MRI from 86 suprasellar tumor patients across multiple institutions. We then applied a Bayesian DL approach to calibrate our previously published ACP classifier, extending beyond point-estimate predictions to predictive distributions. Our original classifier outperforms random forest and XGBoost models in classifying ACP. The calibrated classifier underperformed our previously published results, indicating that the original model was overfitting. The mean values of the predictive distributions were not informative regarding model uncertainty. However, the variance of predictive distributions was indicative of predictive uncertainty. We developed an algorithm to incorporate predicted values and the associated uncertainty to create a classification abstention mechanism. Our model accuracy improved from 80.8% to 95.5%, with a 34.2% abstention rate. We demonstrated that calibration of DL models can be used to estimate predictive uncertainty, which may enable the clinical translation of artificial intelligence to support the non-invasive diagnosis of brain tumors in the future 2).


1)

Johnson LN, Hepler RS, Yee RD, Frazee JG, Simons KB. Magnetic resonance imaging of craniopharyngioma. Am J Ophthalmol. 1986 Aug 15;102(2):242-4. doi: 10.1016/0002-9394(86)90152-2. PMID: 3740186.
2)

Prince EW, Ghosh D, Görg C, Hankinson TC. Uncertainty-Aware Deep Learning Classification of Adamantinomatous Craniopharyngioma from Preoperative MRI. Diagnostics (Basel). 2023 Mar 16;13(6):1132. doi: 10.3390/diagnostics13061132. PMID: 36980440; PMCID: PMC10047069.

Brain invasion in atypical meningioma

Brain invasion in atypical meningioma

J.Sales-Llopis

Neurosurgery Service, Alicante University General Hospital, Alicante, Spain.

The World Health Organization Classification of Tumors of the Central Nervous System 2016 edition added brain invasion (BI) as a criterion for atypical meningioma.

Perry believes that the original definition of brain invasion, which requires a breach of the pial barrier, is still the best method. The two scenarios that are most often mistaken for (or do not show sufficient evidence of) microscopic brain invasion are: 1) an irregular interface with the adjacent brain that nevertheless shows an intervening layer of leptomeninges/collagen and 2) perivascular spread along Virchow-Robin spaces 1).


An accurate assessment of brain invasion is mandatory as brain invasion is a strong predictor of meningioma progression 2). Including brain invasion as a standalone diagnostic criterion for Grade 2, meningiomas had minimal impact on the incidence of specific meningioma-grade tumors. There is strong agreement between the 2007 and 2016 WHO criteria, likely due to the cosegregation of grade elevating features 3)

Go et al. introduced the mechanisms of brain invasion in atypical meningioma and review the genetic factors involved along with epigenetic regulation. First, it is important to understand the three major steps for brain invasion of meningeal cells:

1) degradation of extracellular matrix by proteases, 2) promotion of tumor cell migration to resident cells by adhesion molecules, and 3) neovascularization and supporting cells by growth factors. Second, the genomic landscape of meningiomas should be analyzed by major categories, such as germline mutations in NF2 and somatic mutations in non-NF2 genes (TRAF7KLF4AKT1SMO, and POLR2A). Finally, epigenetic alterations in meningiomas are being studied, with a focus on DNA methylationhistone modification, and RNA interference. Increasing knowledge of the molecular landscape of meningiomas has allowed the identification of prognostic and predictive markers that can guide therapeutic decision-making processes and the timing of follow-up 4).

The volume of peritumoral edema was significantly higher in the brain-invasive meningioma group compared to the non-brain-invasive group. The presence of a complete cleft was a rare finding that was only found in non-brain-invasive meningiomas. The presence of enlarged pial feeding arteries was a rare finding that was only found in brain-invasive meningiomas 5).


For Xiao et al. the combined model (radiomics classifier with BTI4mm ROI + PEV) had greater diagnostic performance than other models 6)

Von Spreckelsen et al. summarize preclinical models studying targeted therapies with potential inhibitory effects 7)

Studies have raised doubts about the prognostic significance of BI in otherwise benign meningiomas.

Invasion of brain tissue by meningiomas has been identified as one key factor for meningioma recurrence. The identification of meningioma tumor tissue surrounded by brain tissue in neurosurgical samples has been touted as a criterion for atypical meningioma (CNS WHO grade 2) but is only rarely seen in the absence of other high-grade features, with brain-invasive otherwise benign (BIOB) meningiomas remaining controversial 8)


Behling et al. were able to show the prognostic impact of CNS invasion in a large comprehensive retrospective meningioma cohort including other established prognostic factors. They discuss the growing experiences that have been gained on this matter, with a focus on the currently nonuniform histopathological assessment, imaging characteristics, and intraoperative sampling as well as the overall outlook on the future role of this potential prognostic factor 9).


Garcia-Segura et al. published that the combination of necrosis and brain invasion is a strong predictor of tumor recurrence and radio-resistance in meningioma, regardless of EOR or adjuvant RT. The findings question the sensibility of brain invasion as an absolute criterion for World health organization grade 2 meningioma status 10).


Nakasu systematically reviewed studies published after 2000 and performed a PRISMA-compliant meta-analysis of the hazard ratios (HRs) for progression-free survival (PFS) between brain-invasive and noninvasive meningiomas. In five studies that included both benign and higher-grade meningiomas, brain invasion was a significant risk factor for recurrence (HR = 2.45, p = 0.0004). However, in 3 studies comparing “brain-invasive meningioma with otherwise benign histology (BIOB)” with grade I meningioma, brain invasion was not a significant predictor of PFS (HR = 1.49, p = 0.23). Among GIIM per the WHO 2000 criteria, brain invasion was a significant predictor of shorter PFS than noninvasive GIIM (HR = 3.40, p = 0.001) but not per the WHO 2016 criteria (HR 1.13, p = 0.54), as the latter includes BIOB. Meta-regression analysis of seven studies of grade II meningioma showed that more frequent BIOB was associated with lower HRs (p < 0.0001). Hence, there is no rationale for brain invasion as a standalone criterion for grade II meningioma, although almost all studies were retrospective and exhibited highly heterogeneous HRs due to differences in brain-tumor interface data availability 11)


Banan et al. investigated the reproducibility of such a prognostic effect.

They identified two cohorts one consisting of 483 patients with meningioma WHO grade I (M°I) or atypical meningioma WHO grade II (M°II) from Hannover Medical School and the other including atypical meningiomas defined according to the classical WHO criteria (M°IIb) from University Hospital Heidelberg. Follow-up data with a median observation time of 38.2 months were available from 308 cases. These included 243 M°I and 65 M°II patients with the latter group consisting of 25 patients with otherwise benign meningiomas with BI (M°IIa) and 40 with M°IIb.

A significant difference in the progression-free interval (PFI) was found between patients with M°I and M°II, M°I and M°IIa, and those with M°I and M°IIb of both cohorts and each separately. However, PFI of M°IIa and M°IIb patients showed no significant difference. In the multivariate regression analysis adjusted for M°I/M°IIa versus M°IIb, sex, age, extent of resection, and tumor location, BI exhibited the strongest risk of relapse (Hazard ratio: 4.95) serving as an independent predictor of PFI (p = 0.002).

The results clearly support the definition of BI as a single criterion of atypia in WHO classification of 2016 12).

Several studies are providing increasing insights into reliable markers to improve the diagnostic and prognostic assessment of meningioma patients. The evidence of brain invasion (BI) signs and its associated variables has been focused on, and currently, scientific research is investing in the study of key aspects, different methods, and approaches to recognize and evaluate BI. This paradigm shift may have significant repercussions for the diagnostic, prognostic, and therapeutic approach to higher-grade meningioma, as long as the evidence of BI may influence patients’ prognosis and inclusion in clinical trials and indirectly impact adjuvant therapy. We intended to review the current knowledge about the impact of BI in meningioma in the most updated literature and explore the most recent implications on both clinical practice and trials and future directions. According to the PRISMA guidelines, systematic research in the most updated platform was performed in order to provide a complete overview of characteristics, preoperative applications, and potential implications of BI in meningiomas. Nineteen articles were included in the present paper and analyzed according to specific research areas. The detection of brain invasion could represent a crucial factor in meningioma patients’ management, and research is flourishing and promising 13)


In 2017 Brokinkel et al. reviewed the current knowledge about brain invasion with emphasis on its implications on current and future clinical practice. We found various definitions of brain invasion and approaches for evaluation in surgically obtained specimens described over the past decades. This heterogeneity is reflected by a weak correlation with prognosis and remains controversial. Similarly, associated clinical factors are largely unknown. Preoperative, imaging-guided detection of brain invasion is unspecific, and intraoperative assessment using standard and new high-magnification microscopic techniques remains imprecise. Despite the increasing knowledge about molecular alterations of the tumor/ brain surface, pharmacotherapeutic options targeting brain invasive meningiomas are lacking. Finally, they summarize the impact of brain invasion on histopathological grading in the WHO classifications of brain tumors since 1979. In conclusion, standardized neurosurgical sampling and neuropathological analyses could improve the diagnostic reliability and reproducibility of future studies. Further research is needed to improve pre-and intraoperative visualization of brain invasion and to develop adjuvant, targeted therapies 14).


1)

Perry, A. (2021). The definition and role of brain invasion in meningioma grading: Still controversial after all these years. Free Neuropathology, 2, 8. https://doi.org/10.17879/freeneuropathology-2021-3276
2)

Picart T, Dumot C, Guyotat J, Pavlov V, Streichenberger N, Vasiljevic A, Fenouil T, Durand A, Jouanneau E, Ducray F, Jacquesson T, Berhouma M, Meyronet D. Clinical and pathological impact of an optimal assessment of brain invasion for grade 2 meningioma diagnosis: lessons from a series of 291 cases. Neurosurg Rev. 2022 Aug;45(4):2797-2809. doi: 10.1007/s10143-022-01792-6. Epub 2022 Apr 29. PMID: 35488071.
3)

Rebchuk AD, Chaharyn BM, Alam A, Hounjet CD, Gooderham PA, Yip S, Makarenko S. The impact of brain invasion criteria on the incidence and distribution of WHO grade 1, 2, and 3 meningiomas. Neuro Oncol. 2022 Sep 1;24(9):1524-1532. doi: 10.1093/neuonc/noac032. PMID: 35139206; PMCID: PMC9435498.
4)

Go KO, Kim YZ. Brain Invasion and Trends in Molecular Research on Meningioma. Brain Tumor Res Treat. 2023 Jan;11(1):47-58. doi: 10.14791/btrt.2022.0044. PMID: 36762808; PMCID: PMC9911709.
5)

Ong T, Bharatha A, Alsufayan R, Das S, Lin AW. MRI predictors for brain invasion in meningiomas. Neuroradiol J. 2020 Sep 14:1971400920953417. doi: 10.1177/1971400920953417. Epub ahead of print. PMID: 32924772.
6)

Xiao D, Zhao Z, Liu J, Wang X, Fu P, Le Grange JM, Wang J, Guo X, Zhao H, Shi J, Yan P, Jiang X. Diagnosis of Invasive Meningioma Based on Brain-Tumor Interface Radiomics Features on Brain MR Images: A Multicenter Study. Front Oncol. 2021 Aug 20;11:708040. doi: 10.3389/fonc.2021.708040. PMID: 34504789; PMCID: PMC8422846.
7) , 8)

von Spreckelsen N, Kesseler C, Brokinkel B, Goldbrunner R, Perry A, Mawrin C. Molecular neuropathology of brain-invasive meningiomas. Brain Pathol. 2022 Mar;32(2):e13048. doi: 10.1111/bpa.13048. PMID: 35213084; PMCID: PMC8877755.
9)

Behling F, Hempel JM, Schittenhelm J. Brain Invasion in Meningioma-A Prognostic Potential Worth Exploring. Cancers (Basel). 2021 Jun 29;13(13):3259. doi: 10.3390/cancers13133259. PMID: 34209798; PMCID: PMC8267840.
10)

Garcia-Segura ME, Erickson AW, Jairath R, Munoz DG, Das S. Necrosis and Brain Invasion Predict Radio-Resistance and Tumor Recurrence in Atypical Meningioma: A Retrospective Cohort Study. Neurosurgery. 2020 Dec 15;88(1):E42-E48. doi: 10.1093/neuros/nyaa348. PMID: 32818240.
11)

Nakasu S, Nakasu Y. Prognostic significance of brain invasion in meningiomas: systematic review and meta-analysis. Brain Tumor Pathol. 2021 Jan 6. doi: 10.1007/s10014-020-00390-y. Epub ahead of print. PMID: 33403457.
12)

Banan R, Abbetmeier-Basse M, Hong B, Dumitru CA, Sahm F, Nakamura M, Krauss JK, Hartmann C. The prognostic significance of clinicopathological features in meningiomas: microscopic brain invasion can predict patient outcome in otherwise benign meningiomas. Neuropathol Appl Neurobiol. 2021 Jan 28. doi: 10.1111/nan.12700. Epub ahead of print. PMID: 33508895.
13)

Brunasso L, Bonosi L, Costanzo R, Buscemi F, Giammalva GR, Ferini G, Valenti V, Viola A, Umana GE, Gerardi RM, Sturiale CL, Albanese A, Iacopino DG, Maugeri R. Updated Systematic Review on the Role of Brain Invasion in Intracranial Meningiomas: What, When, Why? Cancers (Basel). 2022 Aug 27;14(17):4163. doi: 10.3390/cancers14174163. PMID: 36077700; PMCID: PMC9454707.
14)

Brokinkel B, Hess K, Mawrin C. Brain invasion in meningiomas-clinical considerations and impact of neuropathological evaluation: a systematic review. Neuro Oncol. 2017 Oct 1;19(10):1298-1307. doi: 10.1093/neuonc/nox071. PMID: 28419308; PMCID: PMC5596167.

Cingulate gyrus glioma

Cingulate gyrus glioma

J.Sales-LlopisJ.Abarca-OlivasP.González-López

Neurosurgery Department, University General Hospital of Alicante, Spain

A 42-year-old male was admitted for episodes of dizziness and sweating followed by loss of consciousness of at least 15 minutes, Predominantly left retro-ocular headache, intermittent, of months of evolution with worsening in the last 3 days and 2 vomits of food content two days ago. He refers subconjunctival hemorrhage of a long evolution.

The CT showed an extensive hypodense space-occupying lesion in the midline centered in the corpus callosum/cingulate gyrus with a cystic appearance (11 HU of mean attenuation) without clear enhancement or calcifications, measuring approximately 12 cm x 6.8 cm x 9 cm (ap x tra x DC). The lesion is predominantly left and seems to follow the orientation of the cingulate gyrus, positioning itself anterior to the knee of the corpus callosum and superior to the trunk of the corpus callosum, imprinting on it and thinning its posterior third, displacing the splenium of the corpus callosum posterosuperior.


In the MRI the infiltrative neoplastic lesion is centered on the left cingulate gyrus of at least 13 cm x 6 cm x 9.2 cm (ap x tra x cc) that distorts and infiltrates the body and part of the splenium of the corpus callosum, crossing the midline and producing a subfalcine herniation to the right with a deviation of about 9 mm from the midline and compression of the left ventricular atrium.


Left lateral decubitus position, leaving the right cerebral hemisphere in a declining position. Coronal skin incision. Paramedian craniotomy at the left precoronal level. Pedicled dural opening to sagittal sinus. Interhemispheric dissection until reaching the cingulate region where tumor insufflation can be seen. Excision of the anterior half of the cingulate gyrus is performed at the level of the body of the corpus callosum. Resection is limited to that height due to anatomical limitations and the intention to preserve the corpus callosum. Correct vascularization with indocyanine is verified. hemostasis. Hermetic dural closure. Skull replacement with 3 trephine plates. Subcutaneous plane closure with absorbable suture and skin staple.

Low-grade glioma intraoperative PA

Convexity meningioma

Convexity meningioma

J.Sales-Llopis

Neurosurgery Service, Alicante University General Hospital, Spain.

Regorafenib side effects

Regorafenib side effects

 


Some of the most common side effects of regorafenib include:

Fatigue

Diarrhea

Nausea and vomiting

Loss of appetite

Hand-foot syndrome (redness, swelling, and pain on the palms of the hands and soles of the feet)

High blood pressure

Abdominal pain

Headache

Weight loss

Infections


Extensive coagulative necrosis 1).


One patient experienced, after reintervention and during Regorafenib treatment (administered 40 days after surgery), dehiscence of the surgical wound 2)


In patients with progressive WHO grade 3 or 4 gliomas, predominantly with two pretreatment lines or more, regorafenib seems to be effective despite considerable grade 3 or 4 side effects 3).


Treiber et al. described 11 consecutive patients with high-grade glioma recurrence treated with regorafenib at the university medical center in Göttingen. The majority of patients had MGMT promoter methylation (9/11 cases). Regorafenib was given as 2nd line systemic treatment in 6/11 patients and 3rd or higher line treatment in 5/11 patients. The median number of applied cycles was 2 with dosage reductions on 5/11. Response to treatment was observed on 4/11 (PR on 1/11, and SD on 3/11). The Median overall survival for the cohort was 16.1 months, median progression-free survival was 9.0 months, and median time to treatment failure was 3.3 months. Side effects of any CTCAE grade were noted in all patients, hereby 6/11 with CTCAE °III-IV reactions. High-grade side effects were of dermatologic, cardiovascular, and hematologic nature. A mean treatment delay of 57.5 days (range 23-119) was noted between tumor board recommendation and treatment initiation due to the application process for off-label use in this indication. In conclusion, treatment with regorafenib in relapsed high-grade glioma is a feasible treatment option but has to be considered carefully due to the significant side effect profile 4).


Within 12-months of regorafenib treatment, and 16-years since SRS, the patient developed ipsilateral House-Brackmann Grade IV facial weakness. Dramatic VS expansion from 14 to 25 mm in maximum diameter, with new brain stem compression, was seen on MRI. Due to poor prognosis of his gastrointestinal malignancy, he declined surgical resection, and elected for palliative salvage SRS 5).


1) 
Werner JM, Wollring MM, Tscherpel C, Rosen EK, Werr L, Stetter I, Rueß D, Ruge MI, Brunn A, Al Shughri A, Kabbasch C, Fink GR, Langen KJ, Galldiks N. Multimodal imaging findings in patients with glioblastoma with extensive coagulative necrosis related to regorafenib. Neuro Oncol. 2023 Mar 24:noad051. doi: 10.1093/neuonc/noad051. Epub ahead of print. PMID: 36960770.
2) 
Gregucci F, Surgo A, Carbonara R, Laera L, Ciliberti MP, Gentile MA, Caliandro M, Sasso N, Bonaparte I, Fanelli V, Tortora R, Paulicelli E, Surico G, Lombardi G, Signorelli F, Fiorentino A. Radiosurgery and Stereotactic Brain Radiotherapy with Systemic Therapy in Recurrent High-Grade Gliomas: Is It Feasible? Therapeutic Strategies in Recurrent High-Grade Gliomas. J Pers Med. 2022 Aug 20;12(8):1336. doi: 10.3390/jpm12081336. PMID: 36013284; PMCID: PMC9410141.
3) 
Werner JM, Wolf L, Tscherpel C, Bauer EK, Wollring M, Ceccon G, Deckert M, Brunn A, Pappesch R, Goldbrunner R, Fink GR, Galldiks N. Efficacy and tolerability of regorafenib in pretreated patients with progressive CNS grade 3 or 4 gliomas. J Neurooncol. 2022 Jun 18. doi: 10.1007/s11060-022-04066-9. Epub ahead of print. PMID: 35716310.
4) 
Treiber H, von der Brelie C, Malinova V, Mielke D, Rohde V, Chapuy CI. Regorafenib for recurrent high-grade glioma: a unicentric retrospective analysis of feasibility, efficacy, and toxicity. Neurosurg Rev. 2022 Jun 20. doi: 10.1007/s10143-022-01826-z. Epub ahead of print. PMID: 35725846.
5) 
Carlstrom LP, Muñoz-Casabella A, Perry A, Graffeo CS, Link MJ. Dramatic Growth of a Vestibular Schwannoma After 16 Years of Postradiosurgery Stability in Association With Exposure to Tyrosine Kinase Inhibitors. Otol Neurotol. 2021 Dec 1;42(10):e1609-e1613. doi: 10.1097/MAO.0000000000003304. PMID: 34766951; PMCID: PMC8597893.