Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.501
Filtrer
1.
Nagoya J Med Sci ; 86(2): 304-313, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38962412

RÉSUMÉ

Radiotherapy combined with temozolomide (TMZ+RT) is the primary treatment for high-grade glioma. TMZ is classified as a moderate emetic risk agent and, thus, supportive care for nausea and vomiting is important. In Nagoya University Hospital, all patients are treated with a 5-hydroxy-tryptamine 3 receptor antagonist (5-HT3RA) for the first 3 days. The daily administration of 5-HT3RA is resumed after the 4th day based on the condition of patients during TMZ+RT. Therefore, the present study investigated risk factors for nausea and vomiting in patients requiring the daily administration of 5-HT3RA. Patients with high-grade glioma who received TMZ+RT between January 2014 and December 2019 at our hospital were included. Patients were divided into two groups: a control group (patients who did not resume 5-HT3RA) and resuming 5-HT3RA group (patients who resumed 5-HT3RA after the 4th day), and both groups were compared to identify risk factors for nausea and vomiting during TMZ+RT. There were 78 patients in the control group (68%) and 36 in the resuming 5-HT3RA group (32%). A multivariate analysis of patient backgrounds in the two groups identified age <18 years, PS 2 or more, and occipital lobe tumors as risk factors for nausea and vomiting. Nausea and vomiting were attenuated in 30 patients (83%) in the resuming 5-HT3RA group following the resumption of 5-HT3RA. The results obtained highlight the importance of extracting patients with these risk factors before the initiation of therapy and the early resumption or daily administration of 5-HT3RA according to the condition of each patient.


Sujet(s)
Gliome , Nausée , Antagonistes des récepteurs 5-HT3 de la sérotonine , Témozolomide , Vomissement , Humains , Témozolomide/usage thérapeutique , Témozolomide/administration et posologie , Témozolomide/effets indésirables , Mâle , Antagonistes des récepteurs 5-HT3 de la sérotonine/usage thérapeutique , Antagonistes des récepteurs 5-HT3 de la sérotonine/administration et posologie , Femelle , Vomissement/induit chimiquement , Vomissement/traitement médicamenteux , Adulte d'âge moyen , Gliome/traitement médicamenteux , Gliome/radiothérapie , Facteurs de risque , Sujet âgé , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/radiothérapie , Adulte , Antinéoplasiques alcoylants/usage thérapeutique , Antinéoplasiques alcoylants/effets indésirables , Antinéoplasiques alcoylants/administration et posologie , Chimioradiothérapie/effets indésirables , Chimioradiothérapie/méthodes
2.
Sci Rep ; 14(1): 15646, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977703

RÉSUMÉ

Gamma knife radiosurgery (GKRS) is recommended as the first-line treatment for brain metastases of lung adenocarcinoma (LUAD) in many guidelines, but its specific mechanism is unclear. We aimed to study the changes in the proteome of brain metastases of LUAD in response to the hyperacute phase of GKRS and further explore the mechanism of differentially expressed proteins (DEPs). Cancer tissues were collected from a clinical trial for neoadjuvant stereotactic radiosurgery before surgical resection of large brain metastases (ChiCTR2000038995). Five brain metastasis tissues of LUAD were collected within 24 h after GKRS. Five brain metastasis tissues without radiotherapy were collected as control samples. Proteomics analysis showed that 163 proteins were upregulated and 25 proteins were downregulated. GO and KEGG enrichment analyses showed that the DEPs were closely related to ribosomes. Fifty-three of 70 ribosomal proteins were significantly overexpressed, while none of them were underexpressed. The risk score constructed from 7 upregulated ribosomal proteins (RPL4, RPS19, RPS16, RPLP0, RPS2, RPS26 and RPS25) was an independent risk factor for the survival time of LUAD patients. Overexpression of ribosomal proteins may represent a desperate response to lethal radiotherapy. We propose that targeted inhibition of these ribosomal proteins may enhance the efficacy of GKRS.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du cerveau , Tumeurs du poumon , Protéomique , Radiochirurgie , Protéines ribosomiques , Humains , Protéines ribosomiques/métabolisme , Radiochirurgie/méthodes , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/radiothérapie , Mâle , Femelle , Protéomique/méthodes , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/mortalité , Adénocarcinome pulmonaire/chirurgie , Adénocarcinome pulmonaire/radiothérapie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/radiothérapie , Adulte d'âge moyen , Sujet âgé , Régulation de l'expression des gènes tumoraux , Protéome/métabolisme
3.
Medicine (Baltimore) ; 103(28): e38884, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996135

RÉSUMÉ

RATIONALE: Brain metastasis is a major concern, and may occur in roughly 50% of patients during the clinical course of small cell lung cancer (SCLC). Because prophylactic cranial irradiation reduces the incidence of brain metastases and improves overall survival, prophylactic cranial irradiation is recommended for SCLC patients without distant metastases or an extensive stage and have responded well to systemic therapy. Hippocampal-avoidance whole-brain radiotherapy (HA-WBRT) is preferred to preserve hippocampal function while minimizing negative cognitive effects. PATIENT CONCERNS: Reducing the dose delivered to the hippocampus below the therapeutic brain dose may increase the risk of hippocampal progression; thus, HA-WBRT may be associated with a risk of perihippocampal recurrence. DIAGNOSIS: Three patients with SCLC received HA-WBRT and developed intracranial failure during clinical follow-up; 3 relapsed with intracranial failure in the perihippocampal region after 12, 13, and 7 months, respectively. INTERVENTION AND OUTCOMES: Compared to the therapeutic brain dose of cases and the underdose region around the HA region, we matched MRI scans of intracranial failure and previous planning scans of simulation and found a deviation of the underdosed region within the perihippocampal failure of approximately 55% to 63%. LESSONS: Perihippocampal failure is a rare clinical outcome in SCLC patients following HA-WBRT. Perihippocampal failure could be caused by an underdose of radiation or by the aggressiveness of the cancer itself. More research into this topic is encouraged.


Sujet(s)
Tumeurs du cerveau , Irradiation crânienne , Hippocampe , Tumeurs du poumon , Carcinome pulmonaire à petites cellules , Humains , Carcinome pulmonaire à petites cellules/radiothérapie , Hippocampe/effets des radiations , Irradiation crânienne/effets indésirables , Irradiation crânienne/méthodes , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/secondaire , Tumeurs du poumon/radiothérapie , Mâle , Adulte d'âge moyen , Sujet âgé , Femelle , Imagerie par résonance magnétique
4.
BMC Cancer ; 24(1): 866, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026289

RÉSUMÉ

BACKGROUND: The identification of viable tumors and radiation necrosis after stereotactic radiosurgery (SRS) is crucial for patient management. Tumor habitat analysis involving the grouping of similar voxels can identify subregions that share common biology and enable the depiction of areas of tumor recurrence and treatment-induced change. This study aims to validate an imaging biomarker for tumor recurrence after SRS for brain metastasis by conducting tumor habitat analysis using multi-parametric MRI. METHODS: In this prospective study (NCT05868928), patients with brain metastases will undergo multi-parametric MRI before SRS, and then follow-up MRIs will be conducted every 3 months until 24 months after SRS. The multi-parametric MRI protocol will include T2-weighted and contrast-enhanced T1-weighted imaging, diffusion-weighted imaging, and dynamic susceptibility contrast imaging. Using k-means voxel-wise clustering, this study will define three structural MRI habitats (enhancing, solid low-enhancing, and nonviable) on T1- and T2-weighted images and three physiologic MRI habitats (hypervascular cellular, hypovascular cellular, and nonviable) on apparent diffusion coefficient maps and cerebral blood volume maps. Using RANO-BM criteria as the reference standard, via Cox proportional hazards analysis, the study will prospectively evaluate associations between parameters of the tumor habitats and the time to recurrence. The DICE similarity coefficients between the recurrence site and tumor habitats will be calculated. DISCUSSION: The tumor habitat analysis will provide an objective and reliable measure for assessing tumor recurrence from brain metastasis following SRS. By identifying subregions for local recurrence, our study could guide the next therapeutic targets for patients after SRS. TRIAL REGISTRATION: This study is registered at ClinicalTrials.gov (NCT05868928).


Sujet(s)
Tumeurs du cerveau , Récidive tumorale locale , Radiochirurgie , Humains , Radiochirurgie/méthodes , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/chirurgie , Tumeurs du cerveau/radiothérapie , Récidive tumorale locale/imagerie diagnostique , Récidive tumorale locale/anatomopathologie , Études prospectives , Femelle , Mâle , Imagerie par résonance magnétique/méthodes , Adulte d'âge moyen , Adulte , Sujet âgé , Appréciation des risques/méthodes
5.
Cancer Control ; 31: 10732748241266476, 2024.
Article de Anglais | MEDLINE | ID: mdl-39030537

RÉSUMÉ

OBJECTIVES: Brain metastases (BMs) are commonly categorized into cystic and solid. However, the difference in the prognosis of patients with either cystic or solid BMs following radiotherapy remains poorly understood. We used a retrospective design to elucidate the disparities in survival between these two patient groups undergoing radiotherapy and to identify factors influencing the overall survival (OS) of patients with BMs. METHODS: This retrospective study encompasses 212 patients diagnosed with BMs. We meticulously analyzed the clinical characteristics, radiation therapy modalities, and risk factors influencing the OS among these patients, categorized by BMs type, post-brain radiation therapy. RESULTS: A statistically significant difference in mOS was observed between the two cohorts (Solid vs Cystic: 23.1 vs 14.6 months). Subgroup analysis unveiled distinctions in mOS, particularly in patients with EGFR-mutant lung adenocarcinoma (Solid vs Cystic: 23.1 vs 6.43 months). The volume of BMs and the biological effective dose (BED) emerged as significantly prognostic factors for patients with cystic BMs. For patients with solid BMs, fraction dose, BED, and the number of BMs were identified as independent prognostic factors for survival. CONCLUSION: Brain radiotherapy shows superior survival benefits for lung cancer patients with solid BMs compared to those with cystic BMs, particularly in EGFR-mutant lung cancer. In particular, patients receiving BED ≥60 Gy have a more favorable prognosis than those receiving BED <60 Gy, regardless of the type of BM (solid or cystic) in lung cancer.


Sujet(s)
Tumeurs du cerveau , Humains , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/mortalité , Mâle , Femelle , Études rétrospectives , Adulte d'âge moyen , Pronostic , Sujet âgé , Tumeurs du poumon/radiothérapie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Adulte
6.
J Med Case Rep ; 18(1): 330, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39030575

RÉSUMÉ

BACKGROUND: Subependymal giant cell astrocytoma is a benign brain tumor that occurs in patients with tuberous sclerosis complex. Surgical removal is the traditional treatment, and expert opinion is strongly against the use of radiotherapy. Recently, success has been reported with the mTor inhibitor everolimus in reducing tumor volume, but regrowth has been observed after dose reduction or cessation. CASE REPORT: We present the case of a 40-year-old Asian female patient treated successfully for growing bilateral subependymal giant cell astrocytoma with fractionated stereotactic radiotherapy before everolimus became available. After a follow-up of 8 years, everolimus was administered for renal angiomyolipoma and the patient was followed up until 13 years after radiotherapy. Successive magnetic resonance imaging demonstrated an 80% volume reduction after radiotherapy that increased to 90% with everolimus. A review of the literature was done leveraging Medline via PubMed, and we assembled a database of 1298 article references and 780 full-text articles in search of evidence for contraindicating radiotherapy in subependymal giant cell astrocytoma. Varying results of single-fraction radiosurgery were described in a total of 13 cases. Only in two published cases was the radiation dose of fractionated radiotherapy mentioned. One single publication mentions an induced secondary brain tumor 8 years after whole-brain radiotherapy. CONCLUSION: There is no evidence of contraindication and exclusion of fractionated radiotherapy in treating subependymal giant cell astrocytoma. Our experience demonstrates that subependymal giant cell astrocytoma, as other benign intracranial tumors, responds slowly but progressively to radiotherapy and suggests that fractionated stereotactic radiotherapy holds promise to consolidate responses obtained with mTor inhibitors avoiding regrowth after cessation.


Sujet(s)
Astrocytome , Tumeurs du cerveau , Évérolimus , Radiochirurgie , Humains , Femelle , Astrocytome/radiothérapie , Astrocytome/chirurgie , Adulte , Tumeurs du cerveau/radiothérapie , Évérolimus/usage thérapeutique , Imagerie par résonance magnétique , Antinéoplasiques/usage thérapeutique , Résultat thérapeutique , Tumeurs du rein/radiothérapie , Tumeurs du rein/anatomopathologie , Complexe de la sclérose tubéreuse/complications
7.
Cancer Imaging ; 24(1): 95, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026377

RÉSUMÉ

BACKGROUND: Radiotherapy is a major therapeutic approach in patients with brain tumors. However, it leads to cognitive impairments. To improve the management of radiation-induced brain sequalae, deformation-based morphometry (DBM) could be relevant. Here, we analyzed the significance of DBM using Jacobian determinants (JD) obtained by non-linear registration of MRI images to detect local vulnerability of healthy cerebral tissue in an animal model of brain irradiation. METHODS: Rats were exposed to fractionated whole-brain irradiation (WBI, 30 Gy). A multiparametric MRI (anatomical, diffusion and vascular) study was conducted longitudinally from 1 month up to 6 months after WBI. From the registration of MRI images, macroscopic changes were analyzed by DBM and microscopic changes at the cellular and vascular levels were evaluated by quantification of cerebral blood volume (CBV) and diffusion metrics including mean diffusivity (MD). Voxel-wise comparisons were performed on the entire brain and in specific brain areas identified by DBM. Immunohistology analyses were undertaken to visualize the vessels and astrocytes. RESULTS: DBM analysis evidenced time-course of local macrostructural changes; some of which were transient and some were long lasting after WBI. DBM revealed two vulnerable brain areas, namely the corpus callosum and the cortex. DBM changes were spatially associated to microstructural alterations as revealed by both diffusion metrics and CBV changes, and confirmed by immunohistology analyses. Finally, matrix correlations demonstrated correlations between JD/MD in the early phase after WBI and JD/CBV in the late phase both in the corpus callosum and the cortex. CONCLUSIONS: Brain irradiation induces local macrostructural changes detected by DBM which could be relevant to identify brain structures prone to radiation-induced tissue changes. The translation of these data in patients could represent an added value in imaging studies on brain radiotoxicity.


Sujet(s)
Lésions encéphaliques , Animaux , Rats , Mâle , Lésions encéphaliques/étiologie , Lésions encéphaliques/imagerie diagnostique , Lésions encéphaliques/anatomopathologie , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/anatomopathologie , Lésions radiques/imagerie diagnostique , Lésions radiques/anatomopathologie , Lésions radiques/étiologie , Encéphale/effets des radiations , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Imagerie par résonance magnétique/méthodes , Lésions radiques expérimentales/imagerie diagnostique , Lésions radiques expérimentales/anatomopathologie , Lésions radiques expérimentales/étiologie , Imagerie par résonance magnétique multiparamétrique/méthodes
10.
BMC Cancer ; 24(1): 837, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003464

RÉSUMÉ

BACKGROUND: This study aimed to compare the survival outcome and side effects in patients with primary high-grade glioma (HGG) who received carbon ion radiotherapy (CIRT) alone or as a boost strategy after photon radiation (photon + CIRTboost). PATIENTS AND METHODS: Thirty-four (34) patients with histologically confirmed HGG and received CIRT alone or Photon + CIRTboost, with concurrent temozolomide between 2020.03-2023.08 in Wuwei Cancer Hospital & Institute, China were retrospectively reviewed. Overall survival (OS), progression-free survival (PFS), and acute and late toxicities were analyzed and compared. RESULTS: Eight WHO grade 3 and 26 grade 4 patients were included in the analysis. The median PFS in the CIRT alone and Photon + CIRTboost groups were 15 and 19 months respectively for all HGG cases, and 15 and 17.5 months respectively for grade 4 cases. The median OS in the CIRT alone and Photon + CIRTboost groups were 28 and 31 months respectively for all HGG cases, and 21 and 19 months respectively for grade 4 cases. No significant difference in these survival outcomes was observed between the CIRT alone and Photon + CIRTboost groups. Only grade 1 acute toxicities were observed in CIRT alone and Photon + CIRTboost groups. CIRT alone group had a significantly lower ratio of acute toxicities compared to Photon + CIRTboost (3/18 vs. 9/16, p = 0.03). No significant difference in late toxicities was observed. CONCLUSION: Both CIRT alone and Photon + CIRTboost with concurrent temozolomide are safe, without significant differences in PFS and OS in HGG patients. It is meaningful to explore whether dose escalation of CIRTboost might improve survival outcomes of HGG patients in future randomized trials.


Sujet(s)
Gliome , Radiothérapie par ions lourds , Photons , Humains , Adulte d'âge moyen , Études rétrospectives , Radiothérapie par ions lourds/effets indésirables , Radiothérapie par ions lourds/méthodes , Femelle , Mâle , Gliome/radiothérapie , Gliome/mortalité , Gliome/anatomopathologie , Photons/usage thérapeutique , Photons/effets indésirables , Adulte , Sujet âgé , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/mortalité , Témozolomide/usage thérapeutique , Grading des tumeurs , Jeune adulte , Survie sans progression , Résultat thérapeutique
11.
Cells ; 13(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38994929

RÉSUMÉ

Standard-of-care treatment for Glioblastoma Multiforme (GBM) is comprised of surgery and adjuvant chemoradiation. Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated disease-modifying activity in GBM and holds great promise. Radiation, a standard-of-care treatment for GBM, has well-known immunomodulatory properties and may overcome the immunosuppressive tumor microenvironment (TME); however, radiation dose optimization and integration with CAR T cell therapy is not well defined. Murine immunocompetent models of GBM were treated with titrated doses of stereotactic radiosurgery (SRS) of 5, 10, and 20 Gray (Gy), and the TME was analyzed using Nanostring. A conditioning dose of 10 Gy was determined based on tumor growth kinetics and gene expression changes in the TME. We demonstrate that a conditioning dose of 10 Gy activates innate and adaptive immune cells in the TME. Mice treated with 10 Gy in combination with mCAR T cells demonstrated enhanced antitumor activity and superior memory responses to rechallenge with IL13Rα2-positive tumors. Furthermore, 10 Gy plus mCAR T cells also protected against IL13Rα2-negative tumors through a mechanism that was, in part, c-GAS-STING pathway-dependent. Together, these findings support combination conditioning with low-dose 10 Gy radiation in combination with mCAR T cells as a therapeutic strategy for GBM.


Sujet(s)
Glioblastome , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Glioblastome/thérapie , Glioblastome/immunologie , Glioblastome/radiothérapie , Glioblastome/anatomopathologie , Animaux , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Souris , Microenvironnement tumoral/immunologie , Humains , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/radiothérapie , Lymphocytes T/immunologie , Souris de lignée C57BL , Immunomodulation , Femelle
12.
PeerJ ; 12: e17696, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011372

RÉSUMÉ

Background: The effect of whole-brain radiation therapy (WBRT) plus simultaneous integrated boost (SIB) in brain metastasis from breast cancers has not been demonstrated. Method: In this single-center retrospective study, we reviewed consecutive breast cancer patients who developed brain metastasis and were treated with hypofractionated radiation therapy plus WBRT using intensity-modulated radiation therapy (IMRT)-SIB approaches. We analyzed clinical outcomes, prognostic factors and patterns of treatment failure. Result: A total of 27 patients were eligible for analysis. Four (14.8%) patients achieved clinical complete response and 14 (51.9%) had partial response of brain lesions. The other nine patients were not evaluated for brain tumor response. The median brain progression-free survival was 8.60 (95% CI [6.43-13.33]) months and the median overall survival was 16.8 (95% CI [13.3-27.7]) months. Three patients had in-field failure, five had out-field failure and two had in-field and out-field failure. Conclusion: WBRT plus SIB led to improved tumor control and clinical outcome in breast cancer patients with brain metastasis.


Sujet(s)
Tumeurs du cerveau , Tumeurs du sein , Irradiation crânienne , Humains , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/radiothérapie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/radiothérapie , Femelle , Adulte d'âge moyen , Études rétrospectives , Irradiation crânienne/méthodes , Adulte , Sujet âgé , Radiothérapie conformationnelle avec modulation d'intensité/méthodes , Hypofractionnement de dose , Résultat thérapeutique
13.
Curr Oncol ; 31(6): 2994-3005, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38920712

RÉSUMÉ

Randomised control trial data support the use of stereotactic radiosurgery (SRS) in up to 4 brain metastases (BMs), with non-randomised prospective data complementing this for up to 10 BMs. There is debate in the neuro-oncology community as to the appropriateness of SRS in patients with >10 BMs. We present data from a large single-centre cohort, reporting survival in those with >10 BMs and in a >20 BMs subgroup. A total of 1181 patients receiving SRS for BMs were included. Data were collected prospectively from the time of SRS referral. Kaplan-Meier graphs and logrank tests were used to compare survival between groups. Multivariate analysis was performed using the Cox proportional hazards model to account for differences in group characteristics. Median survival with 1 BM (n = 379), 2-4 BMs (n = 438), 5-10 BMs (n = 236), and >10 BMs (n = 128) was 12.49, 10.22, 10.68, and 10.09 months, respectively. Using 2-4 BMs as the reference group, survival was not significantly different in those with >10 BMs in either our univariable (p = 0.6882) or multivariable analysis (p = 0.0564). In our subgroup analyses, median survival for those with >20 BMs was comparable to those with 2-4 BMs (10.09 vs. 10.22 months, p = 0.3558). This study contributes a large dataset to the existing literature on SRS for those with multi-metastases and supports growing evidence that those with >10 BMs should be considered for SRS.


Sujet(s)
Tumeurs du cerveau , Radiochirurgie , Humains , Radiochirurgie/méthodes , Femelle , Mâle , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/chirurgie , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/radiothérapie , Adulte d'âge moyen , Sujet âgé , Estimation de Kaplan-Meier , Sujet âgé de 80 ans ou plus , Thérapie moléculaire ciblée/méthodes
15.
Sci Rep ; 14(1): 13950, 2024 06 17.
Article de Anglais | MEDLINE | ID: mdl-38886395

RÉSUMÉ

Tumor-to-normal ratio (T/N) measurement of 18F-FBPA is crucial for patient eligibility to receive boron neutron capture therapy. This study aims to compare the difference in standard uptake value ratios on brain tumors and normal brains using PET/MR ZTE and atlas-based attenuation correction with the current standard PET/CT attenuation correction. Regarding the normal brain uptake, the difference was not significant between PET/CT and PET/MR attenuation correction methods. The T/N ratio of PET/CT-AC, PET/MR ZTE-AC and PET/MR AB-AC were 2.34 ± 0.95, 2.29 ± 0.88, and 2.19 ± 0.80, respectively. The T/N ratio comparison showed no significance using PET/CT-AC and PET/MR ZTE-AC. As for the PET/MRI AB-AC, significantly lower T/N ratio was observed (- 5.18 ± 9.52%; p < 0.05). The T/N difference between ZTE-AC and AB-AC was also significant (4.71 ± 5.80%; p < 0.01). Our findings suggested PET/MRI imaging using ZTE-AC provided superior quantification on 18F-FBPA-PET compared to atlas-based AC. Using ZTE-AC on 18F-FBPA-PET /MRI might be crucial for BNCT pre-treatment planning.


Sujet(s)
Thérapie par capture de neutrons par le bore , Tumeurs du cerveau , Imagerie par résonance magnétique , Tomographie par émission de positons couplée à la tomodensitométrie , Humains , Thérapie par capture de neutrons par le bore/méthodes , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/imagerie diagnostique , Femelle , Mâle , Imagerie par résonance magnétique/méthodes , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Adulte d'âge moyen , Tomographie par émission de positons/méthodes , Adulte , Sujet âgé , Encéphale/imagerie diagnostique , Radio-isotopes du fluor , Composés du bore , Phénylalanine/analogues et dérivés
16.
Semin Radiat Oncol ; 34(3): 337-343, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38880542

RÉSUMÉ

Radiation treatment has been the cornerstone in cancer management. However, long term treatment-related morbidity always accompanies tumor control which has significant impact on quality of life of the patient who has survived the cancer. Spatially fractionated radiation has the potential to achieve both cure and to avoid dreaded long term sequelae. The first ever randomized study of mini-beam radiation treatment (MBRT) of canine brain tumor has clearly shown the ability to achieve this goal. Dogs have gyrencephalic brains functionally akin to human brain. We here report long term follow-up and final outcome of the dogs, revealing both tumor control and side effects on normal brain. The results augur potential for conducting human studies with MBRT.


Sujet(s)
Tumeurs du cerveau , Photons , Chiens , Tumeurs du cerveau/radiothérapie , Photons/usage thérapeutique , Animaux , Humains , Fractionnement de la dose d'irradiation
17.
Comput Biol Med ; 177: 108637, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38824789

RÉSUMÉ

Radiotherapy is a preferred treatment for brain metastases, which kills cancer cells via high doses of radiation meanwhile hardly avoiding damage to surrounding healthy cells. Therefore, the delineation of organs-at-risk (OARs) is vital in treatment planning to minimize radiation-induced toxicity. However, the following aspects make OAR delineation a challenging task: extremely imbalanced organ sizes, ambiguous boundaries, and complex anatomical structures. To alleviate these challenges, we imitate how specialized clinicians delineate OARs and present a novel cascaded multi-OAR segmentation framework, called OAR-SegNet. OAR-SegNet comprises two distinct levels of segmentation networks: an Anatomical-Prior-Guided network (APG-Net) and a Point-Cloud-Guided network (PCG-Net). Specifically, APG-Net handles segmentation for all organs, where multi-view segmentation modules and a deep prior loss are designed under the guidance of prior knowledge. After APG-Net, PCG-Net refines small organs through the mini-segmentation and the point-cloud alignment heads. The mini-segmentation head is further equipped with the deep prior feature. Extensive experiments were conducted to demonstrate the superior performance of the proposed method compared to other state-of-the-art medical segmentation methods.


Sujet(s)
Tumeurs du cerveau , Planification de radiothérapie assistée par ordinateur , Humains , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/imagerie diagnostique , Planification de radiothérapie assistée par ordinateur/méthodes , Organes à risque , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Traitement d'image par ordinateur/méthodes
18.
Medicine (Baltimore) ; 103(23): e38464, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38847695

RÉSUMÉ

RATIONALE: Gamma knife stereotactic radiosurgery (GKRS) is a recognized safe and effective treatment for brain metastasis; however, some complications can present significant clinical challenges. This case report highlights a rare occurrence of cerebrospinal fluid (CSF) leakage and pneumocranium following GKRS, emphasizing the need for awareness and prompt management of these complications. PATIENT CONCERNS: A 35-year-old male with a history of malignant neoplasm of the lip in 2015 and perineural spread of malignancy into the left cavernous sinus was treated with GKRS in 2017. The patient was admitted emergently 39 days after discharge due to persistent headache and dizziness. DIAGNOSES: Brain computed tomography (CT) revealed diffuse bilateral pneumocranium alongside an observation of CSF leakage. INTERVENTIONS: A surgical procedure involving a left frontal-temporal craniotomy was performed to excise a residual skull base tumor and repair the dura, guided by a navigator system. The conclusive pathological assessment revealed the presence of squamous cell carcinoma markers. OUTCOMES: The patient exhibited excellent tolerance to the entire procedure and experienced a prompt and uneventful recovery process. After surgery, the symptoms alleviated and CSF leak stopped. The follow-up image showed the pneumocranium resolved. LESSONS: Pneumocranium due to early-stage post-GKRS is uncommon. The rapid tumor shrinkage and timing of brain metastasis spreading through the dura can lead to CSF leak and pneumocranium. We reviewed current treatment options and presented a successful craniotomy-based dura repair case.


Sujet(s)
Tumeurs du cerveau , Radiochirurgie , Adulte , Humains , Mâle , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/chirurgie , Fuite de liquide cérébrospinal/étiologie , Pneumocéphale/étiologie , Complications postopératoires/étiologie , Radiochirurgie/effets indésirables , Radiochirurgie/méthodes , Tomodensitométrie
19.
Cell Mol Life Sci ; 81(1): 247, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38829550

RÉSUMÉ

BACKGROUND: The high degree of intratumoral genomic heterogeneity is a major obstacle for glioblastoma (GBM) tumors, one of the most lethal human malignancies, and is thought to influence conventional therapeutic outcomes negatively. The proneural-to-mesenchymal transition (PMT) of glioma stem cells (GSCs) confers resistance to radiation therapy in glioblastoma patients. POLD4 is associated with cancer progression, while the mechanisms underlying PMT and tumor radiation resistance have remained elusive. METHOD: Expression and prognosis of the POLD family were analyzed in TCGA, the Chinese Glioma Genome Atlas (CGGA) and GEO datasets. Tumorsphere formation and in vitro limiting dilution assay were performed to investigate the effect of UCHL3-POLD4 on GSC self-renewal. Apoptosis, TUNEL, cell cycle phase distribution, modification of the Single Cell Gel Electrophoresis (Comet), γ-H2AX immunofluorescence, and colony formation assays were conducted to evaluate the influence of UCHL3-POLD4 on GSC in ionizing radiation. Coimmunoprecipitation and GST pull-down assays were performed to identify POLD4 protein interactors. In vivo, intracranial xenograft mouse models were used to investigate the molecular effect of UCHL3, POLD4 or TCID on GCS. RESULT: We determined that POLD4 was considerably upregulated in MES-GSCs and was associated with a meagre prognosis. Ubiquitin carboxyl terminal hydrolase L3 (UCHL3), a DUB enzyme in the UCH protease family, is a bona fide deubiquitinase of POLD4 in GSCs. UCHL3 interacted with, depolyubiquitinated, and stabilized POLD4. Both in vitro and in vivo assays indicated that targeted depletion of the UCHL3-POLD4 axis reduced GSC self-renewal and tumorigenic capacity and resistance to IR treatment by impairing homologous recombination (HR) and nonhomologous end joining (NHEJ). Additionally, we proved that the UCHL3 inhibitor TCID induced POLD4 degradation and can significantly enhance the therapeutic effect of IR in a gsc-derived in situ xenograft model. CONCLUSION: These findings reveal a new signaling axis for GSC PMT regulation and highlight UCHL3-POLD4 as a potential therapeutic target in GBM. TCID, targeted for reducing the deubiquitinase activity of UCHL3, exhibited significant synergy against MES GSCs in combination with radiation.


Sujet(s)
Cellules souches tumorales , Radiotolérance , Ubiquitin thiolesterase , Humains , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Radiotolérance/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/effets des radiations , Animaux , Souris , Lignée cellulaire tumorale , Gliome/anatomopathologie , Gliome/génétique , Gliome/radiothérapie , Gliome/métabolisme , Apoptose/génétique , Apoptose/effets des radiations , Ubiquitination , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/radiothérapie , Souris nude , Phénotype , Régulation de l'expression des gènes tumoraux , Pronostic
20.
BMC Cancer ; 24(1): 736, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879476

RÉSUMÉ

BACKGROUND: Glioblastoma (GBM) is the most common and aggressive primary brain cancer. The treatment of GBM consists of a combination of surgery and subsequent oncological therapy, i.e., radiotherapy, chemotherapy, or their combination. If postoperative oncological therapy involves irradiation, magnetic resonance imaging (MRI) is used for radiotherapy treatment planning. Unfortunately, in some cases, a very early worsening (progression) or return (recurrence) of the disease is observed several weeks after the surgery and is called rapid early progression (REP). Radiotherapy planning is currently based on MRI for target volumes definitions in many radiotherapy facilities. However, patients with REP may benefit from targeting radiotherapy with other imaging modalities. The purpose of the presented clinical trial is to evaluate the utility of 11C-methionine in optimizing radiotherapy for glioblastoma patients with REP. METHODS: This study is a nonrandomized, open-label, parallel-setting, prospective, monocentric clinical trial. The main aim of this study was to refine the diagnosis in patients with GBM with REP and to optimize subsequent radiotherapy planning. Glioblastoma patients who develop REP within approximately 6 weeks after surgery will undergo 11C-methionine positron emission tomography (PET/CT) examinations. Target volumes for radiotherapy are defined using both standard planning T1-weighted contrast-enhanced MRI and PET/CT. The primary outcome is progression-free survival defined using RANO criteria and compared to a historical cohort with REP treated without PET/CT optimization of radiotherapy. DISCUSSION: PET is one of the most modern methods of molecular imaging. 11C-Methionine is an example of a radiolabelled (carbon 11) amino acid commonly used in the diagnosis of brain tumors and in the evaluation of response to treatment. Optimized radiotherapy may also have the potential to cover those regions with a high risk of subsequent progression, which would not be identified using standard-of-care MRI for radiotherapy planning. This is one of the first study focused on radiotherapy optimization for subgroup of patinets with REP. TRIAL REGISTRATION: NCT05608395, registered on 8.11.2022 in clinicaltrials.gov; EudraCT Number: 2020-000640-64, registered on 26.5.2020 in clinicaltrialsregister.eu. Protocol ID: MOU-2020-01, version 3.2, date 18.09.2020.


Sujet(s)
Tumeurs du cerveau , Évolution de la maladie , Glioblastome , Méthionine , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/diagnostic , Radio-isotopes du carbone , Glioblastome/imagerie diagnostique , Glioblastome/thérapie , Glioblastome/diagnostic , Glioblastome/radiothérapie , Imagerie par résonance magnétique/méthodes , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Études prospectives , Radiopharmaceutiques/usage thérapeutique , Planification de radiothérapie assistée par ordinateur/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE