Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.222
Filter
1.
BMC Cancer ; 24(1): 805, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969990

ABSTRACT

BACKGROUND: Differentiation of glioma and solitary brain metastasis (SBM), which requires biopsy or multi-disciplinary diagnosis, remains sophisticated clinically. Histogram analysis of MR diffusion or molecular imaging hasn't been fully investigated for the differentiation and may have the potential to improve it. METHODS: A total of 65 patients with newly diagnosed glioma or metastases were enrolled. All patients underwent DWI, IVIM, and APTW, as well as the T1W, T2W, T2FLAIR, and contrast-enhanced T1W imaging. The histogram features of apparent diffusion coefficient (ADC) from DWI, slow diffusion coefficient (Dslow), perfusion fraction (frac), fast diffusion coefficient (Dfast) from IVIM, and MTRasym@3.5ppm from APTWI were extracted from the tumor parenchyma and compared between glioma and SBM. Parameters with significant differences were analyzed with the logistics regression and receiver operator curves to explore the optimal model and compare the differentiation performance. RESULTS: Higher ADCkurtosis (P = 0.022), frackurtosis (P<0.001),and fracskewness (P<0.001) were found for glioma, while higher (MTRasym@3.5ppm)10 (P = 0.045), frac10 (P<0.001),frac90 (P = 0.001), fracmean (P<0.001), and fracentropy (P<0.001) were observed for SBM. frackurtosis (OR = 0.431, 95%CI 0.256-0.723, P = 0.002) was independent factor for SBM differentiation. The model combining (MTRasym@3.5ppm)10, frac10, and frackurtosis showed an AUC of 0.857 (sensitivity: 0.857, specificity: 0.750), while the model combined with frac10 and frackurtosis had an AUC of 0.824 (sensitivity: 0.952, specificity: 0.591). There was no statistically significant difference between AUCs from the two models. (Z = -1.14, P = 0.25). CONCLUSIONS: The frac10 and frackurtosis in enhanced tumor region could be used to differentiate glioma and SBM and (MTRasym@3.5ppm)10 helps improving the differentiation specificity.


Subject(s)
Brain Neoplasms , Glioma , Humans , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/secondary , Brain Neoplasms/pathology , Glioma/diagnostic imaging , Glioma/pathology , Female , Male , Middle Aged , Adult , Diagnosis, Differential , Aged , Diffusion Magnetic Resonance Imaging/methods , ROC Curve , Magnetic Resonance Imaging/methods
2.
Breast Cancer Res ; 26(1): 108, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951862

ABSTRACT

BACKGROUND: Metastasis, the spread, and growth of malignant cells at secondary sites within a patient's body, accounts for over 90% of cancer-related mortality. Breast cancer is the most common tumor type diagnosed and the leading cause of cancer lethality in women in the United States. It is estimated that 10-16% breast cancer patients will have brain metastasis. Current therapies to treat patients with breast cancer brain metastasis (BCBM) remain palliative. This is largely due to our limited understanding of the fundamental molecular and cellular mechanisms through which BCBM progresses, which represents a critical barrier for the development of efficient therapies for affected breast cancer patients. METHODS: Previous research in BCBM relied on co-culture assays of tumor cells with rodent neural cells or rodent brain slice ex vivo. Given the need to overcome the obstacle for human-relevant host to study cell-cell communication in BCBM, we generated human embryonic stem cell-derived cerebral organoids to co-culture with human breast cancer cell lines. We used MDA-MB-231 and its brain metastatic derivate MDA-MB-231 Br-EGFP, other cell lines of MCF-7, HCC-1806, and SUM159PT. We leveraged this novel 3D co-culture platform to investigate the crosstalk of human breast cancer cells with neural cells in cerebral organoid. RESULTS: We found that MDA-MB-231 and SUM159PT breast cancer cells formed tumor colonies in human cerebral organoids. Moreover, MDA-MB-231 Br-EGFP cells showed increased capacity to invade and expand in human cerebral organoids. CONCLUSIONS: Our co-culture model has demonstrated a remarkable capacity to discern the brain metastatic ability of human breast cancer cells in cerebral organoids. The generation of BCBM-like structures in organoid will facilitate the study of human tumor microenvironment in culture.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Coculture Techniques , Organoids , Humans , Organoids/pathology , Brain Neoplasms/secondary , Brain Neoplasms/pathology , Female , Breast Neoplasms/pathology , Cell Line, Tumor , Brain/pathology , Cell Communication
3.
Sci Rep ; 14(1): 15646, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977703

ABSTRACT

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.


Subject(s)
Adenocarcinoma of Lung , Brain Neoplasms , Lung Neoplasms , Proteomics , Radiosurgery , Ribosomal Proteins , Humans , Ribosomal Proteins/metabolism , Radiosurgery/methods , Brain Neoplasms/secondary , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Male , Female , Proteomics/methods , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/surgery , Adenocarcinoma of Lung/radiotherapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Middle Aged , Aged , Gene Expression Regulation, Neoplastic , Proteome/metabolism
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1088-1097, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977338

ABSTRACT

OBJECTIVE: To investigate the effect of the aqueous extract of Chuan Xiong Rhizoma (CR) on brain metastasis of melanoma B16F10 cells in mice. METHODS: C57BL/6J mouse models of brain metastasis of melanoma were established by ultrasound-guided intraventricular injection of Luc-labeled B16F10 cells, and brain tumor growth was monitored by in vivo imaging. The mouse models were then randomized for daily gavage of saline or aqueous extract of CR (equivalent crude drug concentration of 1 mg/g). Behavioral tests were used to evaluate the neuroprotective effects of CR in the tumor-bearing mice, and the changes in proteins associated with blood-brain barrier integrity, neuronal cell proliferation and apoptosis, and microglial cell apoptosis and activation were observed using immunofluorescence assay. The efficacy of CR combined with temozolomide (25 mg/kg) against brain metastases of B16F10 cells was observed by in vivo imaging. RESULTS: CR-treated mouse models did not show obvious progression of brain metastases and had a reduced rate of body weight loss and lowered protein expressions of ZO-1, claudin-5, occludin, P-gp, TNF-α, AQP4 and PDGFRß. In the behavioral tests, the CR-treated mice showed prolonged stay on the wooden stick with a shortened time of sticky stick removal. Immunofluorescence assay showed increased proliferation and decreased apoptosis of neuronal cells and microglia in CR-treated mice. CR treatment significantly increased the levels of CD86, CD206, IL-4 and IL-10 and decreased the levels of CD163 and IL-1ß in the microenvironment of brain metastases. The mice receiving combined treatments with CR and temozolomide showed significantly lower intensity of fluorescent signals in the brain than those treated with temozolomide alone. CONCLUSION: CR does not promote brain metastasis of melanoma while inducing opening of the blood-brain barrier, and its combined use with TMZ results in enhanced inhibition against brain metastasis of melanoma B16F10 cells in mice.


Subject(s)
Brain Neoplasms , Drugs, Chinese Herbal , Mice, Inbred C57BL , Temozolomide , Animals , Temozolomide/pharmacology , Mice , Brain Neoplasms/secondary , Brain Neoplasms/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Blood-Brain Barrier/drug effects , Apoptosis/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology
5.
BMJ Open ; 14(7): e078335, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969367

ABSTRACT

BACKGROUND: Patients with advanced non-small-cell lung cancer (NSCLC) with activating mutations in the epidermal growth factor receptor (EGFR) gene are a heterogeneous population who often develop brain metastases (BM). The optimal management of patients with asymptomatic brain metastases is unclear given the activity of newer-generation targeted therapies in the central nervous system. We present a protocol for an individual patient data (IPD) prospective meta-analysis to evaluate whether the addition of stereotactic radiosurgery (SRS) before osimertinib treatment will lead to better control of intracranial metastatic disease. This is a clinically relevant question that will inform practice. METHODS: Randomised controlled trials will be eligible if they include participants with BM arising from EGFR-mutant NSCLC and suitable to receive osimertinib both in the first-line and second-line settings (P); comparisons of SRS followed by osimertinib versus osimertinib alone (I, C) and intracranial disease control included as an endpoint (O). Systematic searches of Medline (Ovid), Embase (Ovid), Cochrane Central Register of Controlled Trials (CENTRAL), CINAHL (EBSCO), PsychInfo, ClinicalTrials.gov and the WHO's International Clinical Trials Registry Platform's Search Portal will be undertaken. An IPD meta-analysis will be performed using methodologies recommended by the Cochrane Collaboration. The primary outcome is intracranial progression-free survival, as determined by response assessment in neuro-oncology-BM criteria. Secondary outcomes include overall survival, time to whole brain radiotherapy, quality of life, and adverse events of special interest. Effect differences will be explored among prespecified subgroups. ETHICS AND DISSEMINATION: Approved by each trial's ethics committee. Results will be relevant to clinicians, researchers, policymakers and patients, and will be disseminated via publications, presentations and media releases. PROSPERO REGISTRATION: CRD42022330532.


Subject(s)
Acrylamides , Aniline Compounds , Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Radiosurgery , Systematic Reviews as Topic , Humans , Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Brain Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Combined Modality Therapy , ErbB Receptors/genetics , Indoles , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Meta-Analysis as Topic , Mutation , Prospective Studies , Pyrimidines , Radiosurgery/methods , Randomized Controlled Trials as Topic , Research Design
6.
Medicine (Baltimore) ; 103(28): e38884, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996135

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Cranial Irradiation , Hippocampus , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/radiotherapy , Hippocampus/radiation effects , Cranial Irradiation/adverse effects , Cranial Irradiation/methods , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Lung Neoplasms/radiotherapy , Male , Middle Aged , Aged , Female , Magnetic Resonance Imaging
7.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000069

ABSTRACT

Lung cancer is a leading cause of cancer-related morbidity and mortality worldwide. Metastases in the brain are a common hallmark of advanced stages of the disease, contributing to a dismal prognosis. Lung cancer can be broadly classified as either small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC). NSCLC represents the most predominant histology subtype of lung cancer, accounting for the majority of lung cancer cases. Recent advances in molecular genetics, coupled with innovations in small molecule drug discovery strategies, have facilitated both the molecular classification and precision targeting of NSCLC based on oncogenic driver mutations. Furthermore, these precision-based strategies have demonstrable efficacy across the blood-brain barrier, leading to positive outcomes in patients with brain metastases. This review provides an overview of the clinical features of lung cancer brain metastases, as well as the molecular mechanisms that drive NSCLC oncogenesis. We also explore how precision medicine-based strategies can be leveraged to improve NSCLC brain metastases.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Brain Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/drug therapy , Precision Medicine/methods , Mutation , Blood-Brain Barrier/metabolism , Antineoplastic Agents/therapeutic use
8.
BMC Cancer ; 24(1): 866, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026289

ABSTRACT

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).


Subject(s)
Brain Neoplasms , Neoplasm Recurrence, Local , Radiosurgery , Humans , Radiosurgery/methods , Brain Neoplasms/secondary , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Brain Neoplasms/radiotherapy , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/pathology , Prospective Studies , Female , Male , Magnetic Resonance Imaging/methods , Middle Aged , Adult , Aged , Risk Assessment/methods
9.
Cancer Control ; 31: 10732748241266476, 2024.
Article in English | MEDLINE | ID: mdl-39030537

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Humans , Brain Neoplasms/secondary , Brain Neoplasms/radiotherapy , Brain Neoplasms/mortality , Male , Female , Retrospective Studies , Middle Aged , Prognosis , Aged , Lung Neoplasms/radiotherapy , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Adult
12.
Int J Biol Sci ; 20(9): 3285-3301, 2024.
Article in English | MEDLINE | ID: mdl-38993559

ABSTRACT

Metabolic reprogramming is one of the essential features of tumors that may dramatically contribute to cancer metastasis. Employing liquid chromatography-tandem mass spectrometry-based metabolomics, we analyzed the metabolic profile from 12 pairwise serum samples of NSCLC brain metastasis patients before and after CyberKnife Stereotactic Radiotherapy. We evaluated the histopathological architecture of 144 surgically resected NSCLC brain metastases. Differential metabolites were screened and conducted for functional clustering and annotation. Metabolomic profiling identified a pathway that was enriched in the metabolism of branched-chain amino acids (BCAAs). Pathologically, adenocarcinoma with a solid growth pattern has a higher propensity for brain metastasis. Patients with high BCAT1 protein levels in lung adenocarcinoma tissues were associated with a poor prognosis. We found that brain NSCLC cells had elevated catabolism of BCAAs, which led to a depletion of α-KG. This depletion, in turn, reduced the expression and activity of the m6A demethylase ALKBH5. Thus, ALKBH5 inhibition participated in maintaining the m6A methylation of mesenchymal genes and promoted the occurrence of epithelial-mesenchymal transition (EMT) in NSCLC cells and the proliferation of NSCLC cells in the brain. BCAA catabolism plays an essential role in the metastasis of NSCLC cells.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Epithelial-Mesenchymal Transition , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Epithelial-Mesenchymal Transition/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Male , Female , Amino Acids, Branched-Chain/metabolism , Middle Aged , Cell Line, Tumor , Transaminases
13.
PeerJ ; 12: e17696, 2024.
Article in English | MEDLINE | ID: mdl-39011372

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Cranial Irradiation , Humans , Brain Neoplasms/secondary , Brain Neoplasms/radiotherapy , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Female , Middle Aged , Retrospective Studies , Cranial Irradiation/methods , Adult , Aged , Radiotherapy, Intensity-Modulated/methods , Radiation Dose Hypofractionation , Treatment Outcome
14.
Medicine (Baltimore) ; 103(27): e38794, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968484

ABSTRACT

BACKGROUND: Extracranial metastases occur in <2% of cases of glioblastoma (GBM). When metastases do occur, bone is the most common destination. Herein, we review clinical characteristics of GBM patients with osseous metastases and evaluate both potential risk factors and prognostic significance. METHODS: Using an institutional database, we identified and retrospectively analyzed 6 patients with both GBM and osseous metastases. We collected data on patient demographics, tumor genetics, clinical courses, and outcomes. Given the rarity of metastatic GBM, we conducted historical comparisons using previously published literature. RESULTS: Five patients with osseous metastases (83%) were male, with a median age of 46 years at GBM diagnosis (range: 20-84). All patients had IDH-wildtype, MGMT promoter unmethylated GBM and 5 (83%) had alterations in TP53. All patients underwent surgical resection for GBM followed by radiation with concurrent and adjuvant temozolomide. Four patients (67%) received bevacizumab prior to bone metastasis diagnosis. Bone metastases were discovered at a median of 12.2 months (range: 5.3-35.2) after GBM diagnosis and 4.8 months after starting bevacizumab (range: 3.5-13.2). Three patients (50%) received immunotherapy. After osseous metastasis diagnosis, the median survival was 25 days (range: 13-225). CONCLUSION: In our cohort, most patients were male and young at the time of GBM diagnosis. All patients had IDH-wildtype, MGMT promoter unmethylated GBM, and most had alterations in TP53, which may be important for osseous metastasis. Most patients received bevacizumab, which has been associated with earlier metastasis. Osseous metastases of GBM occur and portend a dismal prognosis in an already aggressive malignancy.


Subject(s)
Bone Neoplasms , Brain Neoplasms , Glioblastoma , Humans , Male , Glioblastoma/genetics , Glioblastoma/secondary , Glioblastoma/pathology , Glioblastoma/therapy , Middle Aged , Female , Adult , Retrospective Studies , Bone Neoplasms/secondary , Bone Neoplasms/genetics , Brain Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Aged , Aged, 80 and over , Young Adult , Prognosis , Bevacizumab/therapeutic use , Tumor Suppressor Protein p53/genetics , DNA Repair Enzymes/genetics , DNA Modification Methylases , Tumor Suppressor Proteins
15.
Methods Mol Biol ; 2811: 113-122, 2024.
Article in English | MEDLINE | ID: mdl-39037653

ABSTRACT

Brain metastasis is a highly complex process, and some cancer cells enter a dormant state after extravasation into the brain. The molecular mechanism of dormancy remains largely unknown and is still under intense investigation. Here, we outline a basic approach to generating and analyzing experimental mouse models to study dormant cancer cells in the brain. Cancer cells stably expressing EGFP and firefly luciferase are injected into the left ventricle of athymic nude mice. After confirmation of brain metastasis by bioluminescence imaging, brain slices are prepared and subjected to Ki67 staining. In addition, a methodology for recovering brain metastatic cancer cells from the mouse brain is described, providing technical tips for unraveling the mysteries of cancer cell dormancy in brain metastasis.


Subject(s)
Brain Neoplasms , Disease Models, Animal , Mice, Nude , Animals , Brain Neoplasms/secondary , Brain Neoplasms/pathology , Mice , Humans , Cell Line, Tumor , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Luminescent Measurements/methods , Brain/pathology , Brain/metabolism
16.
Curr Oncol ; 31(6): 2994-3005, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38920712

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Radiosurgery , Humans , Radiosurgery/methods , Female , Male , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Middle Aged , Aged , Kaplan-Meier Estimate , Aged, 80 and over , Molecular Targeted Therapy/methods
18.
J Assoc Physicians India ; 72(5): 106-108, 2024 May.
Article in English | MEDLINE | ID: mdl-38881122

ABSTRACT

A case describes a 49-year-old male patient who underwent emergency exploratory laparotomy for small intestinal perforation. Peritonitis was present due to perforation of the jejunal tumor. Resection of the jejunal tumor with perforation was performed followed by end-to-end anastomosis of the jejunum. The resected jejunal tumor was identified in the histopathological examination as metastatic from a clear cell variant of squamous cell/large cell carcinoma of the lung. It was associated with metastatic lesions in the brain. Metastasis from the lung carcinoma in the jejunum is a very rare condition predisposing to small intestinal perforation which is also associated with brain metastasis.


Subject(s)
Brain Neoplasms , Intestinal Perforation , Jejunal Neoplasms , Lung Neoplasms , Humans , Male , Middle Aged , Intestinal Perforation/etiology , Intestinal Perforation/surgery , Intestinal Perforation/diagnosis , Jejunal Neoplasms/secondary , Jejunal Neoplasms/surgery , Jejunal Neoplasms/diagnosis , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/diagnosis , Brain Neoplasms/secondary , Carcinoma, Squamous Cell/secondary , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/surgery , Carcinoma, Large Cell/secondary , Carcinoma, Large Cell/surgery , Carcinoma, Large Cell/diagnosis
20.
Medicine (Baltimore) ; 103(25): e38651, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905366

ABSTRACT

This study aimed to evaluate whether palliative surgery for metastatic lesion could provide a survival benefit in metastatic breast cancer (MBC) patients with solitary metastasis. De novo MBC patients with solitary distant lesions were enrolled utilizing the Surveillance, Epidemiology, and End Results (SEER) database. Propensity score matching (PSM) was conducted to form matched pairs of the surgery group and the non-surgery group. The breast cancer-specific survival (BCSS) and overall survival (OS) outcomes between the 2 groups were compared in the following 3 sample models: the entire cohort of MBC (7665 cases); subgroups of patients with different isolated metastatic organs; and subgroups of patients with different molecular subtypes for each isolated metastatic organ. Compared with the Non-surgery group, the surgery group showed better BCSS and OS before PSM (HR = 0.88, 95% CI = 0.79-0.99, P = .04 and HR = 0.85, 95% CI = 0.76-0.95, P = .006, respectively). After PSM, palliative surgery still provided an OS benefit in patients with brain metastasis and lung metastasis (HR = 0.59, 95% CI = 0.37-0.95, P = .01 and HR = 0.64, 95% CI = 0.45-0.90, P = .02, respectively). Likewise, a better BCSS benefit was also found in the subset of patients with brain metastasis (HR = 0.61, 95% CI = 0.38-1.00, P = .01). Further stratification analysis indicated that patients with the luminal A subtype with brain metastasis have a better BCSS (HR = 0.36, 95% CI = 0.16-0.79, P = .04) and OS (HR = 0.37, 95% CI = 0.18-0.75, P = .03) after undergoing palliative surgery than nonsurgical treatment. Our study originality showed that palliative surgery for metastatic lesion could improve survival prognosis in patients with special single-organ metastasis and specific molecular subtypes. More clinical studies are needed to determine whether palliative surgery should be performed in MBC patients.


Subject(s)
Breast Neoplasms , Palliative Care , Propensity Score , SEER Program , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Breast Neoplasms/mortality , Palliative Care/methods , Middle Aged , Aged , Neoplasm Metastasis , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Brain Neoplasms/mortality , Adult , Lung Neoplasms/surgery , Lung Neoplasms/secondary , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...