Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurooncol ; 158(3): 331-339, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35525907

ABSTRACT

PURPOSE: We investigated the prognostic significance of tumor-associated white matter (TA-WM) tracts in glioblastoma (GBM) using magnetic resonance-diffusion tensor imaging (MR-DTI). We hypothesized that (1) TA-WM tracts harbor microscopic disease not targeted through surgery or radiotherapy (RT), and (2) the greater the extent of TA-WM involvement, the worse the survival outcomes. METHODS: We studied a retrospective cohort of 76 GBM patients. TA-WM tracts were identified by MR-DTI fractional anisotropy (FA) maps. For each patient, 22 TA-WM tracts were analyzed and each tract was graded 1-3 based on FA. A TA-WM score (TA-WMS) was computed based on number of involved tracts and corresponding FA grade of involvement. Kaplan-Meier statistics were utilized to determine survival outcomes, log-rank test was used to compare survival between groups, and Cox regression was utilized to determine prognostic variables. RESULTS: For the MGMT-unmethylated cohort, there was a decrease in OS for increasing TA-WMS (median OS 16.5 months for TA-WMS 0-4; 13.6 months for TA-WMS 5-8; 7.3 months for TA-WMS > 9; p = 0.0002). This trend was not observed in the MGMT-methylated cohort. For MGMT-unmethylated patients with TA-WMS > 6 and involvement of tracts passing through brainstem or contralateral hemisphere, median OS was 8.3 months versus median OS 14.1 months with TA-WMS > 6 but not involving aforementioned critical tracts (p = 0.003 log-rank test). For MGMT-unmethylated patients, TA-WMS was predictive of overall survival in multivariate analysis (HR = 1.14, 95% CI 1.03-1.27, p = 0.012) while age, gender, and largest tumor dimension were non-significant. CONCLUSION: Increased TA-WMS and involvement of critical tracts are associated with decreased overall survival in MGMT-unmethylated GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , White Matter , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/therapy , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Diffusion Tensor Imaging , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Prognosis , Promoter Regions, Genetic , Retrospective Studies , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , White Matter/pathology
2.
J Neurooncol ; 155(3): 297-306, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34689306

ABSTRACT

PURPOSE: Survival is dismal for bevacizumab refractory high-grade glioma patients. We prospectively investigated the efficacy of re-irradiation, bevacizumab, and temozolomide in bevacizumab-naïve and bevacizumab-exposed recurrent high-grade glioma, without volume limitations, in a single arm trial. METHODS: Recurrent high-grade glioma patients were stratified based on WHO grade (4 vs. < 4) and prior exposure to bevacizumab (yes vs. no). Eligible patients received radiation using a simultaneous integrated boost technique (55 Gy to enhancing disease, 45 Gy to non-enhancing disease in 25 fractions) with bevacizumab 10 mg/kg every 2 weeks IV and temozolomide 75 mg/m2 daily followed by maintenance bevacizumab 10 mg/kg every 2 weeks and temozolomide 50 mg/m2 daily for 6 weeks then a 2 week holiday until progression. Primary endpoint was overall survival. Quality of life was studied using FACT-Br and FACT-fatigue scales. RESULTS: Fifty-four patients were enrolled. The majority (n = 36, 67%) were bevacizumab pre-exposed GBM. Median OS for all patients was 8.5 months and 7.9 months for the bevacizumab pre-exposed GBM group. Patients ≥ 36 months from initial radiation had a median OS of 13.3 months compared to 7.5 months for those irradiated < 36 months earlier (p < 0.01). FACT-Br and FACT-Fatigue scores initially declined during radiation but returned to pretreatment baseline. Treatment was well tolerated with 5 patients experiencing > grade 3 lymphopenia and 2 with > grade 3 thrombocytopenia. No radiographic or clinical radiation necrosis occurred. CONCLUSIONS: Re-irradiation with bevacizumab and temozolomide is a safe and feasible salvage treatment for patients with large volume bevacizumab-refractory high-grade glioma. Patients further from their initial radiotherapy may derive greater benefit with this regimen.


Subject(s)
Brain Neoplasms , Chemoradiotherapy , Glioma , Re-Irradiation , Bevacizumab/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Fatigue , Glioma/drug therapy , Glioma/radiotherapy , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/radiotherapy , Prospective Studies , Quality of Life , Temozolomide/therapeutic use
3.
J Neurooncol ; 146(1): 121-130, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31741234

ABSTRACT

PURPOSE: Practice patterns vary for adjuvant treatment of 1p/19q-codeleted oligodendroglioma patients. This study evaluates the outcomes of adjuvant (aRT) versus salvage radiation therapy (sRT) in a multi-institutional cohort. METHODS: Oligodendroglioma patients with confirmed 1p/19q codeletion who were treated with RT with or without chemotherapy from 2000 to 2017 at four tertiary centers were retrospectively reviewed. Overall survival (OS), post-RT progression-free survival (PFS), freedom-from-RT (FFRT), and radiation necrosis (RN) rates were determined using Kaplan-Meier analyses. OS1/PFS1 were defined from the initial surgery. OS2/PFS2 were defined from the RT start-date. Multivariable analyses (MVAs) of prognostic factors for OS and PFS were performed with Cox regression. RESULTS: One hundred eighty-six patients were identified: 124(67%) received aRT and 62(33%) received sRT; of sRT patients, 58% were observed after surgery while 42% received chemotherapy without aRT. The median time from initial diagnosis to sRT was 61 months, and 74% had reoperations before sRT. sRT had longer OS1 than aRT (94% vs. 69% at 10 years, p = 0.03) and PFS1 (10-year PFS of 80% vs. 68%, p = 0.03), though sRT was not associated with significantly different OS1/PFS1 on MVAs. Chemotherapy did not delay sRT compared to observation and had worse PFS2 (42% vs. 79% at 5 years, p = 0.08). Higher RT dose was not associated with improved clinical outcomes but was associated with higher symptomatic RN rate (15% vs. 0% at 2 years, p = 0.003). CONCLUSIONS: Delaying RT for selected oligodendroglioma patients appears safe. Adjuvant chemotherapy does not delay sRT longer than observation and may be associated with worse PFS after RT.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , Gene Deletion , Oligodendroglioma/mortality , Practice Patterns, Physicians'/statistics & numerical data , Radiotherapy, Adjuvant/mortality , Salvage Therapy , Adult , Aged , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Oligodendroglioma/radiotherapy , Patient Acceptance of Health Care , Prognosis , Retrospective Studies , Survival Rate , Young Adult
4.
Cancer Immunol Immunother ; 68(6): 999-1009, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31030234

ABSTRACT

AIM: Despite current treatments, high-grade meningiomas continue to have a poor prognosis. Immunotherapy targeting immune checkpoints, such as PD-L1, has demonstrated significant success in controlling numerous malignancies. In this study, we investigate the extent of systemic and local immunosuppression in meningiomas to assess the potential benefit of immune checkpoint inhibitors for the treatment of high-grade meningiomas. METHODS: Peripheral blood was collected from patients undergoing resection of meningiomas (WHO grade I, n = 18; grade II, n = 25; grade III, n = 10). Immunosuppressive myeloid cells (CD45+CD11b+PD-L1+), myeloid-derived suppressor cells (MDSCs) (CD11b+CD33+HLA-DRlow), and regulatory T cells (Tregs) (CD3+CD4+CD25+FoxP3+) were quantified through flow cytometry. Tissue sections from the same patients were assessed for PD-L1 expression and T cell infiltration via immunohistochemistry. RESULTS: Patients with grade III meningiomas demonstrated increased peripheral monocyte PD-L1 compared to patients with grade I/II meningiomas and healthy controls. Peripheral MDSC abundance was increased in grades II and III meningioma patients. PD-L1 staining of meningioma tissue demonstrated increased positivity in grade III meningiomas. Intratumoral PD-L1 was not associated with progression-free survival. High-grade meningiomas had increased T-cell infiltration. However, a significant proportion of these T cells were exhausted PD1+ T cells and immunosuppressive Tregs. CONCLUSIONS: Patients with meningiomas exhibit signs of peripheral immunosuppression, including increased PD-L1 on myeloid cells and elevated MDSC abundance proportional to tumor grade. Additionally, the tumors express substantial PD-L1 proportional to tumor grade. These results suggest a role for immune checkpoint inhibitors targeting the PD-L1/PD-1 pathway in combination with standard therapies for the treatment of high-grade meningiomas.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Immunosuppression Therapy/methods , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/immunology , B7-H1 Antigen/immunology , Female , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Meningeal Neoplasms/immunology , Meningeal Neoplasms/pathology , Meningioma/immunology , Meningioma/pathology , Middle Aged , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Grading , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
5.
J Neurooncol ; 143(1): 157-166, 2019 May.
Article in English | MEDLINE | ID: mdl-30888558

ABSTRACT

INTRODUCTION: NRG protocols for glioblastoma allow for clinical target volume (CTV) reductions at natural barriers; however, literature examining CTV contouring and the relevant white matter pathways is lacking. This study proposes consensus CTV guidelines, with a focus on areas of controversy while highlighting common errors in glioblastoma target delineation. METHODS: Ten academic radiation oncologists specializing in brain tumor treatment contoured CTVs on four glioblastoma cases. CTV expansions were based on NRG trial guidelines. Contour consensus was assessed and summarized by kappa statistics. A meeting was held to discuss the mathematically averaged contours and form consensus contours and recommendations. RESULTS: Contours of the cavity plus enhancement (mean kappa 0.69) and T2-FLAIR signal (mean kappa 0.74) showed moderate to substantial agreement. Experts were asked to trim off anatomic barriers while respecting pathways of spread to develop their CTVs. Submitted CTV_4600 (mean kappa 0.80) and CTV_6000 (mean kappa 0.81) contours showed substantial to near perfect agreement. Simultaneous truth and performance level estimation (STAPLE) contours were then reviewed and modified by group consensus. Anatomic trimming reduced the amount of total brain tissue planned for radiation targeting by a 13.6% (range 8.7-17.9%) mean proportional reduction. Areas for close scrutiny of target delineation were described, with accompanying recommendations. CONCLUSIONS: Consensus contouring guidelines were established based on expert contours. Careful delineation of anatomic pathways and barriers to spread can spare radiation to uninvolved tissue without compromising target coverage. Further study is necessary to accurately define optimal target volumes beyond isometric expansion techniques for individual patients.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Practice Guidelines as Topic , Radiotherapy Planning, Computer-Assisted/methods , Brain Neoplasms/diagnostic imaging , Clinical Protocols , Glioblastoma/diagnostic imaging , Humans , Magnetic Resonance Imaging
6.
Oncologist ; 23(12): e159-e161, 2018 12.
Article in English | MEDLINE | ID: mdl-30072392

ABSTRACT

Central nervous system metastasis in non-small cell lung cancer remains a therapeutic challenge and confers a poor prognosis. Here we describe a patient with lung adenocarcinoma, parenchymal brain metastases, and leptomeningeal carcinomatosis who demonstrated a sustained response to programmed death 1 inhibition combined with stereotactic radiosurgery.


Subject(s)
Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Meningeal Carcinomatosis/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged , Brain Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Meningeal Carcinomatosis/pathology , Treatment Outcome
7.
J Neurooncol ; 140(2): 395-401, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30084023

ABSTRACT

PURPOSE: Postoperative stereotactic radiosurgery (SRS) is increasingly utilized following resection of brain metastases (BM); however, there are no volumetric data guiding dose selection. We performed a volumetric analysis to guide cavity SRS dosing for resected BM. METHODS: 83 consecutive patients with gross total resection who underwent postoperative SRS to 90 cavities were identified. The 12 Gy isodose lines (V12total) along with the volume of brain parenchyma receiving 12 Gy excluding cavity fluid, ventricular fluid, and calvarium (V12parenchyma) were contoured. Local recurrence (LR) and radionecrosis (RN) were calculated using cumulative incidence rates. Multivariate analysis (MVA) and cutpoint analysis were conducted. RESULTS: Median follow-up was 12.3 months; median dose was 16 Gy. 1- and 2-year cumulative incidence rates of LR were 7.9% and 11.0%. Radiation dose [hazard ratio (HR) 2.04, p = 0.002] was significantly associated with time to LR on MVA. 1- and 2-year cumulative incidence rates of RN were 2.6% and 5.5% respectively. MVA demonstrated increased risk of RN with a larger V12parenchyma (HR 1.46, p = 0.0496). Cavities ≤ 10 cc showed a low 2-year RN risk (4.3%), but had a modest LR risk (13.9%). A radiation dose ≥ 18 Gy significantly improved LC (HR 4.79, p = 0.01). CONCLUSIONS: V12parenchyma should be examined in postoperative SRS to assess RN risk. Cavities > 10 cc treated with 16 Gy achieved excellent LC and minimal RN at 2 years. Cavities ≤ 10 cc may be better treated with a dose ≥ 18 Gy to significantly improve LC given the low RN rate observed with 16 Gy.


Subject(s)
Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Postoperative Care , Radiosurgery , Adult , Aged , Aged, 80 and over , Brain/diagnostic imaging , Brain/radiation effects , Brain/surgery , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neurosurgical Procedures , Radiotherapy Dosage
8.
J Neurooncol ; 134(1): 197-203, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28547589

ABSTRACT

Patients with head and neck malignancies commonly develop metastatic disease, yet rarely do these carcinomas metastasize to the brain. Stereotactic radiosurgery (SRS) is routinely employed to treat brain metastases (BM). This study was undertaken to examine the efficacy of SRS for BM from primary head and neck carcinomas. From 2000 to 2016, a total of 19 patients with 38 lesions were retrospectively identified. All patients presented with a primary head and neck malignancy and subsequently developed metastatic disease to the brain treated with SRS at our institution. Actuarial rates for overall survival (OS), local control (LC) and distant brain metastases (DBM) were calculated using Kaplan-Meier estimates. Median follow up was 6.8 months and median survival was 15.8 months. Eleven lesions received post-operative SRS to a surgical cavity and 27 lesions received definitive SRS to a metastasis. The median dose prescribed was 18 Gy. One-year actuarial rate for LC was 77.3% (95% confidence interval [CI] 44-92%) while 1 year and 2 year rates of OS were 52.9% (CI 28-73%) and 31.7% (CI 11-55%) respectively. The median time to develop DBM was 8.4 months. Three patients (16%) underwent repeat SRS following development of new BM and three patients (16%) underwent salvage whole brain radiotherapy (WBRT). SRS may be utilized in the treatment of patients with primary head and neck malignancies metastasized to the brain with high efficacy. Patients with well-controlled systemic disease and good performance status may benefit the most from definitive SRS while avoiding WBRT.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/surgery , Carcinoma/pathology , Head and Neck Neoplasms/pathology , Radiosurgery/methods , Brain Neoplasms/diagnostic imaging , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Middle Aged , Retrospective Studies
9.
J Neurooncol ; 133(3): 531-538, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28547593

ABSTRACT

Intracranial metastasis from prostate cancer is rare. As treatment of castration-resistant prostate cancer improves, the incidence of men with intracranial metastasis from prostate cancer is increasing. Radiation therapy for treatment of intracranial metastasis from prostate cancer is systematically reviewed. A comprehensive review examining peer-reviewed, English language articles from 1990 to 2015 was performed on multiple databases, yielding 1274 articles. These articles were reviewed and selected for studies that met the following inclusion criteria: (1) patients with intracranial metastases from prostate cancer; (2) patients underwent radiation therapy as primary or adjuvant therapy; (3) the sample size of patients was larger than 2. All studies that met inclusion criteria utilized whole-brain radiation therapy (WBRT) in at least one patient. Other treatment regimens included stereotactic radiosurgery (SRS), surgical resection followed by WBRT, as well as concurrent cabazitaxel and WBRT. The range of average time from initial diagnosis of prostate cancer to diagnosis of brain metastasis was 29-45 months. The range of reported median survival time after WBRT was 4-9 months, whereas median survivals after SRS ranged from 9 to 13 months. Intracranial metastases from prostate cancer occur late in the disease process, and are increasing as novel therapies for metastatic disease prolong survival. The reviewed literature suggests that outcomes of patients with prostate cancer intracranial metastases appear similar to those of intracranial metastases from other histologies. Prospective examinations of systemic therapies that cross the blood-brain barrier in conjunction with targeted radiotherapy appear warranted for this increasingly common clinical problem.


Subject(s)
Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Prostatic Neoplasms/pathology , Humans , Male , Prostatic Neoplasms/radiotherapy
10.
J Neurooncol ; 134(2): 331-338, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28643151

ABSTRACT

A recent nomogram for glioblastoma (GBM) was designed to incorporate methylguanine-DNA methyltransferase (MGMT) methylation status in trial patients receiving temozolomide. Since clinical trial patients are strictly selected, compared to the general population, we performed a multi-institutional, external, independent assessment of the nomogram. Consecutive adult patients with supratentorial GBM diagnosed between June 2007 and December 2014 who initiated TMZ-based concurrent chemoradiotherapy (CRT) and were not enrolled on RTOG 0525 or 0825 were eligible. We collected age, gender, MGMT status, performance status, resection extent, race, and tumor site and Cox regression analysis of overall survival (OS) was conducted with the 1-year nomogram-predicted survival (NPS). The predictive accuracy was quantified by the concordance index (c-index) as well as by separating patients into quintile-groups of the population distribution of NPS and comparing mean NPS and observed OS. Of 514 patients with GBM, 309 had all nomogram factors. Median OS was 18.7 months. NPS and observed OS demonstrated a c-index of 0.695. On univariate analysis, the NPS and all included factors except gender were significant. On multivariable analysis (MVA) the only significant factor for worse survival was lower NPS. When separated into quintile-groups of NPS, the observed survival was slightly better than the predicted survival for all but the worst prognostic group. Our multi-institutional cohort provides independent external validation of a novel GBM nomogram incorporating MGMT methylation status. No individual factor included in the nomogram retained significance on MVA after adjusting for NPS.


Subject(s)
Brain Neoplasms/diagnosis , DNA Methylation , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioblastoma/diagnosis , Nomograms , Tumor Suppressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Chemoradiotherapy , Clinical Trials as Topic , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neurosurgical Procedures , Proportional Hazards Models , Retrospective Studies , Tumor Suppressor Proteins/genetics , Young Adult
11.
J Neurooncol ; 121(1): 151-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25270349

ABSTRACT

Choroid plexus tumors (CPTs) are rare neoplasms of the central nervous system whose optimal management is not well defined. The Surveillance Epidemiology and End Results (SEER) Database from 1978 to 2009 was queried to define population-based outcomes for all patients with CPTs. Patient demographic data, histological classification (choroid plexus papilloma [CPP], atypical CPP [aCPP], and choroid plexus carcinoma [CPC]), extent of surgery, and use of radiation therapy (RT) as part of an initial course of therapy were analyzed for impact on overall survival (OS) and cause-specific survival (CSS). Chemotherapy data were not available within the SEER database. A total of 349 patients with CPTs were identified (120 CPCs, 26 aCPPs, and 203 CPPs). Patients with CPC presented at a younger age (median 3, mean 14.8 years) relative to CPP (median 25, mean 28.4 years; p < 0.0001). Histology was a significant predictor of OS, with 5-year OS rates of 90, 77, and 58 % for CPP, aCPP, and CPC, respectively. Older age and male sex were prognostic for worse OS and CSS for CPP. Only extent of surgery had a significant impact on survival for CPC. The use of adjuvant RT in patients with CPC undergoing surgery was not associated with a significantly improved OS (p = 0.17). For patients undergoing GTR without RT as part of an initial course of therapy, estimated 5- and 10-year OS were 70 % (±7 %) and 67 % (±8 %), respectively. Prospective data are required to define the optimal combination of surgery with adjuvant therapies, including chemotherapy.


Subject(s)
Choroid Plexus Neoplasms/epidemiology , Choroid Plexus Neoplasms/therapy , Adolescent , Adult , Aged , Child , Child, Preschool , Combined Modality Therapy , Databases, Factual , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Survival Analysis , Treatment Outcome , Young Adult
14.
Neuro Oncol ; 25(7): 1323-1330, 2023 07 06.
Article in English | MEDLINE | ID: mdl-36734195

ABSTRACT

BACKGROUND: Brain metastases are the most common intracranial tumors in adults and are associated with significant morbidity and mortality. Whole-brain radiotherapy (WBRT) is used frequently in patients for palliation, but can result in neurocognitive deficits. While dose-dependent injury to individual areas such as the hippocampus has been demonstrated, global structural shape changes after WBRT remain to be studied. METHODS: We studied healthy controls and patients with brain metastases and examined MRI brain anatomic surface data before and after WBRT. We implemented a validated graph convolutional neural network model to estimate patient's "brain age". We further developed a mixed-effects linear model to compare the estimated age of the whole brain and substructures before and after WBRT. RESULTS: 4220 subjects were analyzed (4148 healthy controls and 72 patients). The median radiation dose was 30 Gy (range 25-37.5 Gy). The whole brain and substructures underwent structural change resembling rapid aging in radiated patients compared to healthy controls; the whole brain "aged" 9.32 times faster, the cortex 8.05 times faster, the subcortical structures 12.57 times faster, and the hippocampus 10.14 times faster. In a subset analysis, the hippocampus "aged" 8.88 times faster in patients after conventional WBRT versus after hippocampal avoidance (HA)-WBRT. CONCLUSIONS: Our findings suggest that WBRT causes the brain and its substructures to undergo structural changes at a pace up to 13x of the normal aging pace, where hippocampal avoidance offers focal structural protection. Correlating these structural imaging changes with neurocognitive outcomes following WBRT or HA-WBRT would benefit from future analysis.


Subject(s)
Brain Neoplasms , Deep Learning , Radiotherapy, Intensity-Modulated , Adult , Humans , Radiotherapy, Intensity-Modulated/adverse effects , Cranial Irradiation/adverse effects , Cranial Irradiation/methods , Brain , Brain Neoplasms/pathology , Hippocampus/pathology , Aging , Radiotherapy Dosage
15.
J Arthroplasty ; 27(7): 1349-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22245125

ABSTRACT

Recent reports have noted higher rates of heterotopic ossification (HO) with surface replacement arthroplasty (SRA) than with traditional total hip arthroplasty in the absence of postoperative HO prophylaxis. This study reports rates and grades of HO in 44 SRA patients with at least 1 year of follow-up. Heterotopic ossification prophylaxis was used in 32 (73%) of 44 cases. Heterotopic ossification prophylaxis consisted of radiotherapy (22/32), nonsteroidal anti-inflammatory drugs (8/32), or both (2/32). One case of clinically significant HO was documented in the no-prophylaxis group. This strategy of selective HO prophylaxis in patients felt by orthopedic surgeons to be at high risk of HO resulted in low rates of clinically relevant HO after SRA (1/44, 2.3%). Further study is needed to establish optimal selection criteria for HO prophylaxis after SRA.


Subject(s)
Arthroplasty, Replacement, Hip/adverse effects , Ossification, Heterotopic/epidemiology , Ossification, Heterotopic/prevention & control , Osteoarthritis, Hip/surgery , Radiotherapy/methods , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibiotic Prophylaxis , Celecoxib , Female , Follow-Up Studies , Humans , Incidence , Indomethacin/therapeutic use , Male , Middle Aged , Pyrazoles/therapeutic use , Risk Factors , Sulfonamides/therapeutic use , Treatment Outcome
16.
World Neurosurg ; 165: e380-e385, 2022 09.
Article in English | MEDLINE | ID: mdl-35724885

ABSTRACT

OBJECTIVE: Gamma Knife (GK) stereotactic radiosurgery (SRS) is increasingly used as an initial treatment for patients with 10 or more brain metastases. However, the clinical and dosimetric consequences of this practice are not well established. METHODS: We performed a single-institution, retrospective analysis of 30 patients who received Gamma Knife SRS for 10 or more brain metastases in 1 session. We utilized MIM Software to contour the whole brain and accumulated the doses from all treated lesions to determine the mean dose delivered to the whole brain. Patient outcomes were determined from chart review. RESULTS: Our cohort had a median number of 13 treated lesions (range 10-26 lesions) for a total of 427 treated lesions. The mean dose to the whole brain was determined to be 1.8 ± 0.91 Gy (range 0.70-3.8 Gy). The mean dose to the whole brain did not correlate with the number of treated lesions (Pearson r = 0.23, P = 0.21), but was closely associated with tumor volume (Pearson r = 0.95, P < 0.0001). There were no significant correlations between overall survival and number of lesions or aggregate tumor volume. Fourteen patients (47%) underwent additional SRS sessions and 6 patients (20%) underwent whole-brain radiotherapy with a median of 6.6 months (range 3.0-50 months) after SRS. Two patients (6.6%) developed grade 2 radionecrosis following SRS beyond earlier whole-brain radiotherapy. CONCLUSION: The mean dose to the whole brain in patients treated with Gamma Knife SRS for 10 or more brain metastases remained low with an acceptable rate of radionecrosis. This strategy allowed the majority of patients to avoid subsequent whole-brain radiotherapy.


Subject(s)
Brain Neoplasms , Radiation Injuries , Radiosurgery , Brain , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Humans , Radiation Injuries/etiology , Radiosurgery/adverse effects , Retrospective Studies , Treatment Outcome
17.
Phys Imaging Radiat Oncol ; 24: 71-75, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36217428

ABSTRACT

This study aimed to assess the incidental radiation exposure of the hippocampus (HC) in locoregionally-advanced oropharyngeal cancer patients undergoing volumetric modulated arc therapy and the feasibility of HC-sparing plan optimization. The initial plans were generated without dose-volume constraints to the HC and were compared with the HC-sparing plans. The incidental Dmean_median doses to the bilateral, ipsilateral and contralateral HC were 2.9, 3.1, and 2.5 Gy in the initial plans and 1.4, 1.6, and 1.3 Gy with HC-sparing. It was feasible to reduce the HC dose with HC-sparing plan optimization without compromising target coverage and/or dose constraints to other OARs.

18.
Cancer ; 117(12): 2629-36, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21656740

ABSTRACT

BACKGROUND: Postprostatectomy adjuvant or salvage radiotherapy, when using standard fractionation, requires 6.5 to 8 weeks of treatment. The authors report on the safety and efficacy of an expedited radiotherapy course for salvage prostate radiotherapy. METHODS: A total of 108 consecutive patients were treated with salvage radiation therapy to 65 grays (Gy) in 26 fractions of 2.5 Gy. Median follow-up was 32.4 months. Median presalvage prostate-specific antigen (PSA) was 0.44 (range, 0.05-9.50). Eighteen (17%) patients received androgen deprivation after surgery or concurrently with radiation. RESULTS: The actuarial freedom from biochemical failure for the entire group at 4 years was 67% ± 5.3%. An identical 67% control rate was seen at 5 years for the first 50 enrolled patients, whose median follow-up was longer at 43 months. One acute grade 3 genitourinary toxicity occurred, with no acute grade 3 gastrointestinal and no late grade 3 toxicities observed. On univariate analysis, higher Gleason score (P = .006), PSA doubling time ≤12 months (P = .03), perineural invasion (P = .06), and negative margins (P = .06) showed association with unsuccessful salvage. On multivariate analysis, higher Gleason score (P = .057) and negative margins (P = .088) retained an association with biochemical failure. CONCLUSIONS: Hypofractionated radiotherapy (65 Gy in 2.5 Gy fractions in about 5 weeks) reduces the length of treatment by from 1-½ to 3 weeks relative to other treatment schedules commonly used, produces low rates of toxicity, and demonstrates encouraging efficacy at 4 to 5 years. Hypofractionation may provide a convenient, resource-efficient, and well-tolerated salvage approach for the estimated 20,000 to 35,000 US men per year experiencing biochemical recurrence after prostatectomy.


Subject(s)
Dose Fractionation, Radiation , Neoplasm Recurrence, Local/radiotherapy , Prostatectomy , Prostatic Neoplasms/radiotherapy , Salvage Therapy/methods , Aged , Combined Modality Therapy , Humans , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Radiotherapy Planning, Computer-Assisted , Urogenital System/radiation effects
19.
Exp Cell Res ; 316(7): 1083-100, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20064507

ABSTRACT

The epidermal growth factor (EGF) family of receptor tyrosine kinases consists of four members: EGFR (HER1/ErbB1), HER2/neu (ErbB2), HER3 (ErbB3) and HER4 (ErbB4). Receptor activation via ligand binding leads to downstream signaling that influence cell proliferation, angiogenesis, invasion and metastasis. Aberrant expression or activity of EGFR and HER2 have been strongly linked to the etiology of several human epithelial cancers including but not limited to head and neck squamous cell carcinoma (HNSCC), non-small cell lung cancer (NSCLC), colorectal cancer (CRC), and breast cancer. With this, intense efforts have been made to inhibit the activity of the EGFR and HER2 by designing antibodies against the ligand binding domains (cetuximab, panitumumab and trastuzumab) or small molecules against the tyrosine kinase domains (erlotinib, gefitinib, and lapatinib). Both approaches have shown considerable clinical promise. However, increasing evidence suggests that the majority of patients do not respond to these therapies, and those who show initial response ultimately become refractory to treatment. While mechanisms of resistance to tyrosine kinase inhibitors have been extensively studied, resistance to monoclonal antibodies is less well understood, both in the laboratory and in the clinical setting. In this review, we discuss resistance to antibody-based therapies against the EGFR and HER2, similarities between these resistance profiles, and strategies to overcome resistance to HER family targeting monoclonal antibody therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Drug Resistance, Neoplasm/physiology , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/immunology , ErbB Receptors/immunology , ErbB Receptors/metabolism , ErbB Receptors/physiology , Humans , Models, Biological , Multigene Family/immunology , Multigene Family/physiology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Protein Multimerization/immunology , Protein Multimerization/physiology , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/physiology
20.
Ann Palliat Med ; 9(5): 2800-2808, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32787372

ABSTRACT

BACKGROUND: Early palliative care referral for patients with advanced cancer has demonstrable benefits but is underutilized. We sought to characterize medical oncologists' perceptions about palliative care referral in their clinical practices. METHODS: We conducted 4 focus groups with a national sample of medical oncologists to elicit perspectives about the optimal timing of and barriers to palliative care referral for patients with cancer. We used qualitative content analysis to uncover themes related to early integration of palliative care into standard oncologic practice. RESULTS: Study participants readily acknowledged the evidence supporting early palliative care referral. However, medical oncologists identified patient-centered and physician-centered barriers to widespread adoption of early palliative care. Patient-centered barriers included patients' and families' perceptions or misperceptions of the role of palliative care. Additionally, physicians themselves described acting as a barrier to palliative care referral because they were concerned that palliative care physicians may interfere with the plan of care, or offer options that were not endorsed by the medical oncologist. Medical oncologists depicted themselves having authority over the timing of palliative care referral, and as granting limited autonomy to other clinical team members in counseling patients about advanced care planning. CONCLUSIONS: Medical oncologists are hesitant to adopt the practice of early palliative care referral because they are concerned that other physicians may disrupt a patient's treatment plan. Physician-centered barriers may delay integration of palliative care, and future efforts to promote a collaborative approach to advanced care planning may improve patient-centered outcomes through access to early palliative care.


Subject(s)
Hospice and Palliative Care Nursing , Neoplasms , Oncologists , Physicians , Attitude of Health Personnel , Humans , Palliative Care
SELECTION OF CITATIONS
SEARCH DETAIL