[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (6)

Search Parameters:
Keywords = neurocytoma

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 885 KiB  
Article
Surgical Treatment of Pediatric Incidentally Found Brain Tumors: A Single-Center Experience
by Lukasz Antkowiak, Mikolaj Zimny, Krzysztof Starszak, Ryszard Sordyl and Marek Mandera
Brain Sci. 2023, 13(5), 746; https://doi.org/10.3390/brainsci13050746 - 29 Apr 2023
Cited by 1 | Viewed by 1815
Abstract
There remains much debate about the correct management of incidentally found brain tumors in the pediatric population. This study aimed to evaluate the efficacy and safety of surgical treatment of incidentally found pediatric brain tumors. A retrospective analysis of pediatric patients who underwent [...] Read more.
There remains much debate about the correct management of incidentally found brain tumors in the pediatric population. This study aimed to evaluate the efficacy and safety of surgical treatment of incidentally found pediatric brain tumors. A retrospective analysis of pediatric patients who underwent surgical resection of incidentally found brain tumors between January 2010 and April 2016 was performed. A total of seven patients were included. The median age at the time of diagnosis was 9.7 years. The reasons for performing neuroimaging were as follows: impeded speech development (n = 2), shunt control (n = 1), paranasal sinuses control (n = 1), behavior changes (n = 1), head trauma (n = 1), and preterm birth (n = 1). Five patients underwent gross total tumor resection (71.4%), while subtotal resection was performed in two patients (28.6%). There was no surgery-related morbidity. Patients were followed up for a mean of 79 months. One patient with atypical neurocytoma experienced tumor recurrence 45 months following primary resection. All patients remained neurologically intact. The majority of pediatric incidentally found brain tumors were histologically benign. Surgery remains a safe therapeutic approach associated with favorable long-term outcomes. Considering the expected long lifetime of pediatric patients, as well as the psychological burden associated with having a brain tumor as a child, surgical resection can be considered an initial approach. Full article
(This article belongs to the Section Neurosurgery and Neuroanatomy)
Show Figures

Figure 1

Figure 1
<p>A 14-year-old girl with right-sided thalamic ganglioglioma. (<b>A</b>) A preoperative axial non-contrast T1 MRI scan showing an isodose solid mass located within the right thalamus with an adherent cystic component, causing a mass effect and slight midline shift. (<b>B</b>) The preoperative axial postcontrast T1 MRI scan revealed a heterogeneous solid mass enhancement. (<b>C</b>) A postoperative axial postcontrast T1 MRI scan reflecting subtotal excision of the tumor, with reduced size of the cystic component, relieved mass effect, and no evident midline shift.</p>
Full article ">Figure 2
<p>A 21-month-old boy with choroid plexus papilloma of the left lateral ventricle. (<b>A</b>) A preoperative axial non-contrast T1 MRI scan with a hypodense mass located within the left occipital horn of the lateral ventricle with cystic components. (<b>B</b>) The preoperative axial postcontrast T1 MRI scan showed homogeneous intraventricular mass enhancement, reflective of choroid plexus papilloma. (<b>C</b>) A postoperative axial postcontrast T1 MRI scan reflecting complete excision of the intraventricular mass, with the remaining cystic component.</p>
Full article ">
15 pages, 2418 KiB  
Article
Whole Exome Sequencing Identifies PHF14 Mutations in Neurocytoma and Predicts Responsivity to the PDGFR Inhibitor Sunitinib
by Dongyun Zhang, William H. Yong, Masoud Movassaghi, Fausto J. Rodriguez, Issac Yang, Paul McKeever, Jiang Qian, Jian Yi Li, Qinwen Mao, Kathy L. Newell, Richard M. Green, Cynthia T. Welsh and Anthony P. Heaney
Biomedicines 2022, 10(11), 2842; https://doi.org/10.3390/biomedicines10112842 - 8 Nov 2022
Cited by 2 | Viewed by 2740
Abstract
Neurocytomas are rare low-grade brain tumors predominantly affecting young adults, but their cellular origin and molecular pathogenesis is largely unknown. We previously reported a sellar neurocytoma that secreted excess arginine vasopressin causing syndrome of inappropriate anti-diuretic hormone (SIADH). Whole exome sequencing in 21 [...] Read more.
Neurocytomas are rare low-grade brain tumors predominantly affecting young adults, but their cellular origin and molecular pathogenesis is largely unknown. We previously reported a sellar neurocytoma that secreted excess arginine vasopressin causing syndrome of inappropriate anti-diuretic hormone (SIADH). Whole exome sequencing in 21 neurocytoma tumor tissues identified somatic mutations in the plant homeodomain finger protein 14 (PHF14) in 3/21 (14%) tumors. Of these mutations, two were missense mutations and 4 caused splicing site losses, resulting in PHF14 dysfunction. Employing shRNA-mediated knockdown and CRISPR/Cas9-based knockout approaches, we demonstrated that loss of PHF14 increased proliferation and colony formation in five different human, mouse and rat mesenchymal and differentiated cell lines. Additionally, we demonstrated that PHF14 depletion resulted in upregulation of platelet derived growth factor receptor-alpha (PDGFRα) mRNA and protein in neuroblastoma SHSY-5Y cells and led to increased sensitivity to treatment with the PDGFR inhibitor Sunitinib. Furthermore, in a neurocytoma primary culture harboring splicing loss PHF14 mutations, overexpression of wild-type PHF14 and sunitinib treatment inhibited cell proliferation. Nude mice, inoculated with PHF14 knockout SHSY-5Y cells developed earlier and larger tumors than control cell-inoculated mice and Sunitinib administration caused greater tumor suppression in mice harboring PHF-14 knockout than control SHSY-5Y cells. Altogether our studies identified mutations of PHF14 in 14% of neurocytomas, demonstrate it can serve as an alternative pathway for certain cancerous behavior, and suggest a potential role for Sunitinib treatment in some patients with residual/recurrent neurocytoma. Full article
(This article belongs to the Special Issue 10th Anniversary of Biomedicines—Novel Targets for Cranial Tumors)
Show Figures

Figure 1

Figure 1
<p>Identification of homeodomain finger protein 14 (PHF14) mutations in neurocytomas by whole exome sequencing. (<b>A</b>) Neurocytoma formalin-fixed, paraffin-embedded (FFPE) and frozen tissues (<span class="html-italic">n</span> = 54 total) were retrospectively collected from 11 centers across United States and Canada. Genomic DNA was extracted from 38 neurocytoma tissues with tumor area &gt;95% and diameter &gt;5 mm and was of suitable quality for whole exome sequencing in 21 neurocytoma tumors (FFPE samples <span class="html-italic">n</span> = 17, frozen samples <span class="html-italic">n</span> = 4). (<b>B</b>) Demographics and subtypes of 21 sequenced neurocytoma samples and 5 normal cerebellar controls. (<b>C</b>) Summary of the six PHF14 mutations detected by WES in three neurocytoma samples compared.</p>
Full article ">Figure 2
<p>PHF14 Depletion Enhances Cell Proliferation and Anchorage Independent Cell Growth. (<b>A</b>) A conserved 19-bp region in the human, mouse and rat PHF14 gene in Exon 10 (CGC ATG ATT CAA ATT CAG GAA) was selected as shRNA target to knockdown PHF14 expression. Five cell lines originating from human, mouse and rat were used to evaluate the biological effect of PHF14 depletion. PHF14 knockdown stable transfectants were established by puromycin selection, and the knockdown efficiency was determined by Western Blot using anti-PHF14 antibody. The densitometric analyses of the protein bands vs. the individual loading controls were shown under individual blot using the ImageQuant 5.2 software (GE Healthcare, Pittsburgh, PA). (<b>B</b>) Cell proliferation rate was enhanced in shRNA PHF4 knockdown transfectants compared to Nonsense controls as measured by CellTiter-Glo<sup>®</sup> luminescent cell viability assay in a variety of cell lines. (<b>C</b>) Soft agar assay demonstrated that PHF14 knockdown induced an increase in colony size in human neuroblastoma SHSY-5Y cells. (<b>D</b>) A single guiding RNA (sgRNA) targeting a 20-bp region (TGG ATC GCA GCT CCA AGA GG) in PHF14 Exon 1 was designed for CRISPR-Cas9 mediated genetic editing. The knockout efficiency was confirmed by Western Blot. PHF14 knockout in human neuroblastoma SHSY-5Y cells promoted cell proliferation as measured by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. PHF14 knockout increased colony formation in soft agar assay. The results shown were representative of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2 Cont.
<p>PHF14 Depletion Enhances Cell Proliferation and Anchorage Independent Cell Growth. (<b>A</b>) A conserved 19-bp region in the human, mouse and rat PHF14 gene in Exon 10 (CGC ATG ATT CAA ATT CAG GAA) was selected as shRNA target to knockdown PHF14 expression. Five cell lines originating from human, mouse and rat were used to evaluate the biological effect of PHF14 depletion. PHF14 knockdown stable transfectants were established by puromycin selection, and the knockdown efficiency was determined by Western Blot using anti-PHF14 antibody. The densitometric analyses of the protein bands vs. the individual loading controls were shown under individual blot using the ImageQuant 5.2 software (GE Healthcare, Pittsburgh, PA). (<b>B</b>) Cell proliferation rate was enhanced in shRNA PHF4 knockdown transfectants compared to Nonsense controls as measured by CellTiter-Glo<sup>®</sup> luminescent cell viability assay in a variety of cell lines. (<b>C</b>) Soft agar assay demonstrated that PHF14 knockdown induced an increase in colony size in human neuroblastoma SHSY-5Y cells. (<b>D</b>) A single guiding RNA (sgRNA) targeting a 20-bp region (TGG ATC GCA GCT CCA AGA GG) in PHF14 Exon 1 was designed for CRISPR-Cas9 mediated genetic editing. The knockout efficiency was confirmed by Western Blot. PHF14 knockout in human neuroblastoma SHSY-5Y cells promoted cell proliferation as measured by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. PHF14 knockout increased colony formation in soft agar assay. The results shown were representative of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>PHF14 Depletion Elevates platelet derived growth factor receptor-alpha (PDGFRα) Expression and Increases Responsivity to the PDGFR Inhibitor Sunitinib. (<b>A</b>–<b>B</b>) ShRNA-directed PHF14 knockdown increased PDGFRα expression at both mRNA (<b>A</b>) and protein (<b>B</b>) levels by Real Time PCR (<b>A</b>) and Western Blot (<b>B</b>) in human neuroblastoma SHSY-5Y cells and mouse embryonic fibroblasts NIH3T3 cells. (<b>C</b>) Increased PDGFRα expression at both protein and mRNA levels was confirmed in PHF14 knockout human neuroblastoma SHSY-5Y cells. (<b>D</b>) The anti-proliferation effects of PDGFR inhibitors, Imatinib and Sunitinib, were evaluated in SHSY-5Y cells by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. (<b>E</b> and <b>F</b>) PHF4 knockdown sensitized the anti-proliferation effect of PDGFR inhibitor Sunitinib (<b>F</b>), but not Imatinib (<b>E</b>) in human neuroblastoma SHSY-5Y cells as analyzed by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. (<b>G</b>) The sensitization to Sunitinib treatment was reproduced in PHF14 knockout SHSY-5Y cells. The results shown were representative of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.005, compared with media control. # <span class="html-italic">p</span> &lt; 0.05; ## <span class="html-italic">p</span> &lt; 0.01 compared with Nonsense control.</p>
Full article ">Figure 4
<p>PHF14 Inhibits Cell Growth in Neurocytoma Primary Culture. (<b>A</b>) Typical morphologic appearance of neurocytoma primary cultures in serum free, insulin, transferrin, selenium and fibronectin (ITSFn) media (Top Panel) and serum supplemented ITFSn media (Bottom Panel) for 3 weeks to compare cell morphology changes. (<b>B</b>) Detection of four splicing loss mutations of PHF14 in the neurocytoma primary cultures by whole exome sequencing (WES). (<b>C</b>) Comparison of the transfection efficiency of Lipofectamine 2000 and transduction efficiency of lentivirus by introducing GFP-expressing vector. Wild type PHF14 was introduced into neurocytoma primary culture using lentivirus. The transduction efficiency was evaluated by Real Time PCR to detect PHF14 overexpression. Cell viability was detected by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. (<b>D</b>) Neurocytoma primary culture was treated with Sunitinib, MEK-162 and Metformin. Cell viability was detected by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. Data shown are representative of at least three independently conducted experiments. Bars indicate the mean ± standard error of the mean of triplicate tests. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 4 Cont.
<p>PHF14 Inhibits Cell Growth in Neurocytoma Primary Culture. (<b>A</b>) Typical morphologic appearance of neurocytoma primary cultures in serum free, insulin, transferrin, selenium and fibronectin (ITSFn) media (Top Panel) and serum supplemented ITFSn media (Bottom Panel) for 3 weeks to compare cell morphology changes. (<b>B</b>) Detection of four splicing loss mutations of PHF14 in the neurocytoma primary cultures by whole exome sequencing (WES). (<b>C</b>) Comparison of the transfection efficiency of Lipofectamine 2000 and transduction efficiency of lentivirus by introducing GFP-expressing vector. Wild type PHF14 was introduced into neurocytoma primary culture using lentivirus. The transduction efficiency was evaluated by Real Time PCR to detect PHF14 overexpression. Cell viability was detected by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. (<b>D</b>) Neurocytoma primary culture was treated with Sunitinib, MEK-162 and Metformin. Cell viability was detected by CellTiter-Glo<sup>®</sup> luminescent cell viability assay. Data shown are representative of at least three independently conducted experiments. Bars indicate the mean ± standard error of the mean of triplicate tests. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 5
<p>Evaluation of the Tumor Inhibitory Effect of PHF14 and Sunitinib Using in vivo Xenograft Animal Model. (<b>A</b>,<b>B</b>) Comparison of tumor development in PHF14 knockout cells (<span class="html-italic">n</span> = 10) and control neuroblastoma cells (<span class="html-italic">n</span> = 10) in an in vivo xenograft model of neuroblastoma. (<b>C</b>,<b>D</b>) To determine the effects of Sunitinib treatment on tumor growth in vivo, we administrated Sunitinib (dissolved in 0.5% CMC) by oral gavage following SHSY-5Y cells inoculation. The tumor growth was monitored and measured daily, and the animals were euthanized on day 21 after drug treatment due to deteriorating animal health condition. Tumors were excised, and weighed. Data were analyzed by two-tailed unpaired <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.005; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5 Cont.
<p>Evaluation of the Tumor Inhibitory Effect of PHF14 and Sunitinib Using in vivo Xenograft Animal Model. (<b>A</b>,<b>B</b>) Comparison of tumor development in PHF14 knockout cells (<span class="html-italic">n</span> = 10) and control neuroblastoma cells (<span class="html-italic">n</span> = 10) in an in vivo xenograft model of neuroblastoma. (<b>C</b>,<b>D</b>) To determine the effects of Sunitinib treatment on tumor growth in vivo, we administrated Sunitinib (dissolved in 0.5% CMC) by oral gavage following SHSY-5Y cells inoculation. The tumor growth was monitored and measured daily, and the animals were euthanized on day 21 after drug treatment due to deteriorating animal health condition. Tumors were excised, and weighed. Data were analyzed by two-tailed unpaired <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.005; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
10 pages, 3832 KiB  
Article
Diffuse GFAP Immunopositivity in the Oligodendrocyte-like Component of Pilocytic Astrocytoma Distinguishes It from Mimickers
by Murad Alturkustani
Diagnostics 2022, 12(7), 1632; https://doi.org/10.3390/diagnostics12071632 - 5 Jul 2022
Cited by 3 | Viewed by 2553
Abstract
Pilocytic astrocytoma with a predominant oligodendrocyte-like component can be difficult to distinguish from oligodendroglioma, dysembryoplastic neuroepithelial tumors (DNTs), central neurocytoma, and ependymoma (clear cell phenotype). The utility of GFAP immunostaining in this context is not well discussed. All cases with a diagnosis of [...] Read more.
Pilocytic astrocytoma with a predominant oligodendrocyte-like component can be difficult to distinguish from oligodendroglioma, dysembryoplastic neuroepithelial tumors (DNTs), central neurocytoma, and ependymoma (clear cell phenotype). The utility of GFAP immunostaining in this context is not well discussed. All cases with a diagnosis of pilocytic astrocytoma were retrieved from the pathological archives along with the following information: age, sex, and pathological description. The GFAP immunostaining was scored as score 1 (<25%), score 2 (25–50%), score 3 (50–75%), and score 4 (>75%). The comparison group included oligodendrogliomas, DNTs, ependymomas, and central neurocytomas. All 26 cases (16 males and 10 females) of pilocytic astrocytoma showed strong and diffuse (score 4) GFAP immunostaining in the neoplastic cells of both the solid fibrillary and oligodendrocyte-like components. The staining pattern in the neoplastic round cells in the oligodendrocyte-like areas was perinuclear cytoplasmic with no processes. In the comparison group, GFAP immunostaining was mostly restricted to the reactive astrocytes in the background. Focal areas of the neoplastic cells showed scores of 1–3 in the neoplastic cells, but the staining pattern was different from those in pilocytic astrocytoma. In the setting of tumors with predominant oligodendrocyte-like areas, the GFAP immunostaining score and pattern help distinguish pilocytic astrocytoma from its mimickers. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Definition of the oligodendrocyte-like areas and GFAP scores. (<b>A</b>) Oligodendroglioma with round neoplastic cells. (<b>B</b>) Oligodendrocyte-like area in pilocytic astrocytoma. GFAP scoring system (<b>C</b>–<b>F</b>). (<b>C</b>) Score 1: GFAP-staining cells in less than 25% of neoplastic cells. Most stained cells here are reactive astrocytes (arrows), whereas most neoplastic cells are negative (arrowheads). (<b>D</b>) Score 2: GFAP-staining cells in 25–50% of the neoplastic cells. (<b>E</b>) Score 3: GFAP staining in 50–75% of the neoplastic cells (arrowheads). (<b>F</b>) Score 4: GFAP staining in more than 75% of the neoplastic cells. Scale bars: 50 µm (<b>A</b>–<b>F</b>).</p>
Full article ">Figure 2
<p>GFAP staining in pilocytic astrocytoma. (<b>A</b>) Solid fibrillary area in pilocytic astrocytoma with Rosenthal fibers (arrowheads). (<b>B</b>) Diffuse and strong GFAP immunostaining in the elongated neoplastic cells. The Rosenthal fibers are within the astrocytic processes (arrowheads). (<b>C</b>) Microcystic area with oligodendrocyte-like neoplastic cells. (<b>D</b>) Diffuse and strong GFAP immunostaining (score 4) in the round cells. The immunostaining is mainly a cytoplasmic rim around the nucleus with no processes. (<b>E</b>) The granular cell layer of the cerebellum appears as round blue cells with perinuclear clearing (arrowheads). (<b>F</b>) GFAP staining in the granular cell layer highlights background staining, whereas the round cells are immunonegative for the GFAP immunostain (arrowheads). Scale bars: 50 µm (<b>A</b>–<b>F</b>).</p>
Full article ">Figure 3
<p>GFAP staining in oligodendrocyte-like areas in other tumors. (<b>A</b>) Oligodendroglioma with an area rich in neoplastic gliofibrillary oligodendrocytes and minigemistocytes. (<b>B</b>) GFAP highlights the gliofibrillary oligodendrocytes (arrowheads) and minigemistocytes (arrows). (<b>C</b>) Round neoplastic cells with perinuclear clearing in DNT. (<b>D</b>) GFAP is mainly in the background’s reactive astrocytes (arrows), whereas the round neoplastic cells are immunonegative (arrowheads). (<b>E</b>) Central neurocytoma with round neoplastic cells with neuropil in the background (arrowhead). (<b>F</b>) GFAP immunostain is immunopositive in scattered neoplastic cells (arrowheads) and the reactive astrocytes in the background (arrows). (<b>G</b>) Round neoplastic cells in ependymoma (clear cell phenotype). (<b>H</b>) GFAP highlights neoplastic cells with perivascular accentuation for GFAP (arrowheads). Scale bars: 50 µm (<b>A</b>–<b>H</b>).</p>
Full article ">
19 pages, 2672 KiB  
Review
Cyclic Metronomic Chemotherapy for Pediatric Tumors: Six Case Reports and a Review of the Literature
by Benjamin Carcamo and Giulio Francia
J. Clin. Med. 2022, 11(10), 2849; https://doi.org/10.3390/jcm11102849 - 18 May 2022
Cited by 5 | Viewed by 2792
Abstract
We report a retrospective case series of six Hispanic children with tumors treated with metronomic chemotherapy. The six cases comprised one rhabdoid tumor of the kidney, one ependymoma, two medulloblastomas, one neuroblastoma, and a type II neurocytoma of the spine. Treatment included oral [...] Read more.
We report a retrospective case series of six Hispanic children with tumors treated with metronomic chemotherapy. The six cases comprised one rhabdoid tumor of the kidney, one ependymoma, two medulloblastomas, one neuroblastoma, and a type II neurocytoma of the spine. Treatment included oral cyclophosphamide daily for 21 days alternating with oral etoposide daily for 21 days in a backbone of daily valproic acid and celecoxib. In one case, celecoxib was substituted with sulindac. Of the six patients, three showed complete responses, and all patients showed some response to metronomic therapy with only minor hematologic toxicity. One patient had hemorrhagic gastritis likely associated with NSAIDs while off prophylactic antacids. These data add to a growing body of evidence suggesting that continuous doses of valproic acid and celecoxib coupled with alternating metronomic chemotherapy of agents such as etoposide and cyclophosphamide can produce responses in pediatric tumors relapsing to conventional dose chemotherapy. Full article
(This article belongs to the Special Issue Clinical Applications of Metronomic Chemotherapy)
Show Figures

Figure 1

Figure 1
<p>Patient 1: CT with contrast shows a large 11 × 7 cm left renal tumor with retroperitoneal infiltration, regional metastatic retroperitoneal adenopathy, and extension to renal vein and inferior vena cava. The patient also had innumerable solid circumscribed masses throughout the lung parenchyma bilaterally (not shown).</p>
Full article ">Figure 2
<p>Patient 2: MRI shows confluent encephalomalacia gliosis in the left temporal and occipital lobes. There was no evidence of mass or pathologic enhancement 8 years from diagnosis and 4 years off therapy.</p>
Full article ">Figure 3
<p>Patient 4: (<b>A</b>) image shows and enhancing tumor in the anterior horn of the left ventricle when she presented with first relapse of medulloblastoma. She achieved complete remission with salvage therapy; (<b>B</b>) image shows an enhancing tumor involving the anterior aspect and floor of the fourth ventricle (arrows) when she presented with second relapse; (<b>C</b>) image shows response to 3 months of metronomic therapy with decreased size and intensity of enhancing lesions; (<b>D</b>) image shows interval progression of the tumor seen in T2 FLAIR resulting in a change from temozolomide to etoposide at 4 months of treatment; (<b>E</b>) image shows resolution of the tumor mass and T2 FLAIR changes at 8 months of metronomic therapy; (<b>F</b>) image shows progressive disease at 10 months of treatment.</p>
Full article ">Figure 3 Cont.
<p>Patient 4: (<b>A</b>) image shows and enhancing tumor in the anterior horn of the left ventricle when she presented with first relapse of medulloblastoma. She achieved complete remission with salvage therapy; (<b>B</b>) image shows an enhancing tumor involving the anterior aspect and floor of the fourth ventricle (arrows) when she presented with second relapse; (<b>C</b>) image shows response to 3 months of metronomic therapy with decreased size and intensity of enhancing lesions; (<b>D</b>) image shows interval progression of the tumor seen in T2 FLAIR resulting in a change from temozolomide to etoposide at 4 months of treatment; (<b>E</b>) image shows resolution of the tumor mass and T2 FLAIR changes at 8 months of metronomic therapy; (<b>F</b>) image shows progressive disease at 10 months of treatment.</p>
Full article ">Figure 3 Cont.
<p>Patient 4: (<b>A</b>) image shows and enhancing tumor in the anterior horn of the left ventricle when she presented with first relapse of medulloblastoma. She achieved complete remission with salvage therapy; (<b>B</b>) image shows an enhancing tumor involving the anterior aspect and floor of the fourth ventricle (arrows) when she presented with second relapse; (<b>C</b>) image shows response to 3 months of metronomic therapy with decreased size and intensity of enhancing lesions; (<b>D</b>) image shows interval progression of the tumor seen in T2 FLAIR resulting in a change from temozolomide to etoposide at 4 months of treatment; (<b>E</b>) image shows resolution of the tumor mass and T2 FLAIR changes at 8 months of metronomic therapy; (<b>F</b>) image shows progressive disease at 10 months of treatment.</p>
Full article ">Figure 4
<p>Patient 6: (<b>A</b>) MRI shows a 4.5 cm homogeneously enhancing expansile intramedullary tumor involving the medulla and upper cervical cord down to the level of C3–C4 (arrow) with an elongated syrinx extending inferiorly to the T3–T4 level; (<b>B</b>) the tumor was removed and treated with radiation but came back, for which it was treated with nine cycles of topotecan–ifosfamide–carboplatin with no significant change in tumor size (arrow) but significant toxicity, for which treatment was changed to metronomic chemotherapy; (<b>C</b>) after 3 months on metronomic chemotherapy, the patient recovered from toxicity and the tumor was slightly decreased; (<b>D</b>) the tumor was stable at the end of 4 years of metronomic chemotherapy.</p>
Full article ">Figure 4 Cont.
<p>Patient 6: (<b>A</b>) MRI shows a 4.5 cm homogeneously enhancing expansile intramedullary tumor involving the medulla and upper cervical cord down to the level of C3–C4 (arrow) with an elongated syrinx extending inferiorly to the T3–T4 level; (<b>B</b>) the tumor was removed and treated with radiation but came back, for which it was treated with nine cycles of topotecan–ifosfamide–carboplatin with no significant change in tumor size (arrow) but significant toxicity, for which treatment was changed to metronomic chemotherapy; (<b>C</b>) after 3 months on metronomic chemotherapy, the patient recovered from toxicity and the tumor was slightly decreased; (<b>D</b>) the tumor was stable at the end of 4 years of metronomic chemotherapy.</p>
Full article ">Figure 5
<p>Schematic of future planned studies for pediatric cases eligible for metronomic chemotherapy. Proteomic and metabolic pathway analysis, where feasible, will be used to determine eligibility of patients to receive specific tyrosine kinase inhibitor (TKI) therapy coupled with metronomic chemotherapy, which involves alternating cycles of metronomic etoposide with metronomic cyclophosphamide. Patients not selected or not eligible for TKI-based therapies will receive valproic acid (VA) and celecoxib with metronomic chemotherapy as outlined in this manuscript.</p>
Full article ">
7 pages, 527 KiB  
Article
Impact of Adjuvant Radiotherapy in Patients with Central Neurocytoma: A Multicentric International Analysis
by Laith Samhouri, Mohamed A. M. Meheissen, Ahmad K. H. Ibrahimi, Abdelatif Al-Mousa, Momen Zeineddin, Yasser Elkerm, Zeyad M. A. Hassanein, Abdelsalam Attia Ismail, Hazem Elmansy, Motasem M. Al-Hanaqta, Omar A. AL-Azzam, Amr Abdelaziz Elsaid, Christopher Kittel, Oliver Micke, Walter Stummer, Khaled Elsayad and Hans Theodor Eich
Cancers 2021, 13(17), 4308; https://doi.org/10.3390/cancers13174308 - 26 Aug 2021
Cited by 10 | Viewed by 2757
Abstract
Background: Central neurocytoma (CN) is a rare tumor accounting for <0.5% of all intracranial tumors. Surgery ± radiotherapy is the mainstay treatment. This international multicentric study aims to evaluate the outcomes of CNs patients after multimodal therapies and identify predictive factors. Patients and [...] Read more.
Background: Central neurocytoma (CN) is a rare tumor accounting for <0.5% of all intracranial tumors. Surgery ± radiotherapy is the mainstay treatment. This international multicentric study aims to evaluate the outcomes of CNs patients after multimodal therapies and identify predictive factors. Patients and methods: We retrospectively identified 33 patients with CN treated between 2005 and 2019. Treatment characteristics and outcomes were assessed. Results: All patients with CN underwent surgical resection. Radiotherapy was delivered in 19 patients. The median radiation dose was 54 Gy (range, 50–60 Gy). The median follow-up time was 56 months. The 5-year OS and 5-year PFS were 90% and 76%, respectively. Patients who received radiotherapy had a significantly longer PFS than patients without RT (p = 0.004) and a trend towards longer OS. In addition, complete response after treatments was associated with longer PFS (p = 0.07). Conclusions: Using RT seems to be associated with longer survival rates with an acceptable toxicity profile. Full article
(This article belongs to the Special Issue Cancer Radiotherapy)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier estimate of progression-free survival (<b>A</b>) and overall survival (<b>B</b>) in central neurocytoma patients according to radiotherapy administration (N = 33).</p>
Full article ">
11 pages, 3906 KiB  
Review
Hypothalamic Endocrine Tumors: An Update
by Sylvia L. Asa and Ozgur Mete
J. Clin. Med. 2019, 8(10), 1741; https://doi.org/10.3390/jcm8101741 - 20 Oct 2019
Cited by 15 | Viewed by 5430
Abstract
The hypothalamus is the site of synthesis and secretion of a number of endocrine peptides that are involved in the regulation of hormonal activity of the pituitary and other endocrine targets. Tumors of the hypothalamus have been recognized to have both structural and [...] Read more.
The hypothalamus is the site of synthesis and secretion of a number of endocrine peptides that are involved in the regulation of hormonal activity of the pituitary and other endocrine targets. Tumors of the hypothalamus have been recognized to have both structural and functional effects including hormone hypersecretion. The classification of these tumors has advanced over the last few years, and biomarkers are now available to classify these tumors and provide accurate structure–function correlations. This review provides an overview of tumors in this region that is critical to metabolic homeostasis with a focus on advances in the diagnosis of gangliocytomas, neurocytomas, and pituicytomas that are unique to this region. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Therapy of Pituitary Disorders)
Show Figures

Figure 1

Figure 1
<p>Hypothalamic gangliocytoma associated with a somatotroph tumor. (<b>a</b>)Hematoxylin and eosin (H&amp;E) stain identifies two cell populations; (<b>b</b>) Neurofilament highlights the neurons; (<b>c</b>) Growth hormone-releasing hormone (GHRH) is localized to the neurons; (<b>d</b>) PIT1 stains the nuclei of somatotroph, but not neurons (arrows).</p>
Full article ">Figure 2
<p>Hypothalamic neurocytoma. (<b>a</b>) H&amp;E identifies small round cells in abundant neuropil with a dilated neuronal axon resembling a Herring body; (<b>b</b>) Neurofilament highlights the neurons and neuropil; (<b>c</b>) TTF1 decorates some of the neurons; (<b>d</b>) Vasopressin is expressed by the tumor cells and highlights an axonal terminal known as a Herring body.</p>
Full article ">Figure 3
<p>Pituicytomas. (<b>a</b>) H&amp;E of a classical pituicytoma shows spindle-shaped cells forming fascicles; (<b>b</b>) An oncocytic variant that is composed of round cells with granular eosinophilic cytoplasm; (<b>c</b>) TTF1 decorates the nuclei of the tumor cells; (<b>d</b>) S100 is expressed by the tumor cells.</p>
Full article ">
Back to TopTop