Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Bioengineering (Basel) ; 11(5)2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38790322

RESUMEN

Detection and segmentation of brain metastases (BMs) play a pivotal role in diagnosis, treatment planning, and follow-up evaluations for effective BM management. Given the rising prevalence of BM cases and its predominantly multiple onsets, automated segmentation is becoming necessary in stereotactic radiosurgery. It not only alleviates the clinician's manual workload and improves clinical workflow efficiency but also ensures treatment safety, ultimately improving patient care. Recent strides in machine learning, particularly in deep learning (DL), have revolutionized medical image segmentation, achieving state-of-the-art results. This review aims to analyze auto-segmentation strategies, characterize the utilized data, and assess the performance of cutting-edge BM segmentation methodologies. Additionally, we delve into the challenges confronting BM segmentation and share insights gleaned from our algorithmic and clinical implementation experiences.

2.
Cancer ; 130(1): 18-30, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37682730

RESUMEN

BACKGROUND: Immunotherapy (IO) has demonstrated promising results in treating extensive-stage small cell lung cancer (ES-SCLC), and the management of ES-SCLC brain metastases (BMs) is now receiving significant clinical attention. The objective of this study was to evaluate the role of IO in the clinical management of BMs. METHODS: Between January 2020 and December 2021, the study included the records of 250 patients who were diagnosed with ES-SCLC. Overall survival (OS), progression-free survival, intracranial progression-free survival, and the cumulative incidence of BMs were calculated using the Kaplan-Meier method and were compared using the log-rank test. In addition, the Cox regression model was used to analyze prognostic factors. RESULTS: In the entire group, 85 patients had baseline BMs (IO plus chemotherapy [IO + ChT], n = 38; ChT alone, n = 47), and 165 patients (IO + ChT, n = 86; ChT alone, n = 79) did not have BMs at the time of initial diagnosis. The median follow-up was 22.4 months. The OS benefit with first-line antiprogrammed death ligand 1 therapy was maintained regardless of whether patients had BMs (with BMs, 17.97 vs. 13.14 months [p = .03]; without BMs, 18.46 vs. 15.05 months [p = .047]). However, in patients without BMs, IO did not delay the median time to developing brain progression (10.84 vs. 10.74 months; p = .84), and it did not significantly reduce the risk of developing intracranial metastases (the 2-year actuarial risk of developing BMs was 57.0% vs. 50.6%, respectively). CONCLUSIONS: Antiprogrammed death ligand 1 therapy improved OS regardless of the presence of BMs. However, IO did not delay the median time to brain progression or reduce the risk of intracranial metastasis in patients without baseline BMs. The findings of this study have important clinical implications for the future management of BMs from ES-SCLC.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/patología , Estudios Retrospectivos , Ligandos , Neoplasias Encefálicas/secundario
3.
Quant Imaging Med Surg ; 13(12): 8545-8556, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38106281

RESUMEN

Background: Brain metastases (BMs) are common complications in patients with non-small cell lung cancer (NSCLC). The purpose of this study was to investigate whether the metabolic parameters derived from preoperative 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) can predict BM development in patients with surgically resected NSCLC. Methods: We retrospectively reviewed 128 consecutive patients with stage I-IIIA NSCLC who underwent 18F-FDG PET/CT before curative surgery at The First Affiliated Hospital of Jinan University between November 2012 and October 2021. By drawing a volume of interest (VOI), the maximum standardized uptake values (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) of the primary tumor as well as the mean SUV (SUVmean) of the liver and arterial blood were measured. The tumor-to-liver SUV ratio (TLR) and tumor-to-blood SUV ratio (TBR) were also calculated. Receiver operating characteristic curve analysis was used to determine the best cut-off values for positron emission tomography (PET) parameters to predict BM-free survival, and Cox proportional hazards regression analysis was used to assess the predictive value of clinical variables and PET parameters. Results: The median follow-up duration for survival patients was 23.4 months, and 15 patients (11.7%) experienced BM as the initial relapse site. The cumulative rates of BM over the course of 1, 2, and 5 years were 4.5%, 10.5%, and 17.5%, respectively. The optimal cut-off values for the prediction of BM-free survival were 7.7, 4.9, and 4.5 for SUVmax, TLR, and TBR, and 5.5 mL and 16.1 for MTV and TLG, respectively. In the Cox proportional hazards model, the risk of BM was significantly associated with TLR [hazard ratio (HR) =10.712; 95% confidence interval (CI): 2.958-38.801; P<0.001] and MTV (HR =3.150; 95% CI: 0.964-10.293; P=0.020) after adjusting for tumor stage, clinicopathological factors, and other PET parameters. Conclusions: Preoperative TLR and MTV of the primary tumor may be helpful in predicting BM development in patients with surgically resected NSCLC. Tumor metabolic parameters may potentially be used to stratify the risk of BM and determine individualized surveillance strategies.

4.
Quant Imaging Med Surg ; 13(12): 8599-8610, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38106277

RESUMEN

Background: Predicting whether T790M emerges early is crucial to the adjustment of targeted drugs for non-small cell lung cancer (NSCLC) patients. This study aimed to evaluate the risk of T790M resistance in progressive new brain metastases (BMs) based on multisequence magnetic resonance imaging (MRI) radiomics. Methods: This retrospective study included 405 consecutive patients (training cohort: 294 patients; testing cohort: 111 patients) with proven NSCLC with disease progression of new BM. The radiomics features were separately extracted from T2-weighted imaging (T2WI), T2 fluid-attenuated inversion recovery (T2-FLAIR), diffusion-weighted imaging (DWI), and contrast-enhanced T1-weighted imaging (T1-CE) sequence of baseline MRI. Then, we calculated radiomics scores (rad-score) of the 4 sequences respectively and established predictive models (lesion- or patient-level) to evaluate T790M resistance within up to 14 months using random forest classifier. Receiver operating characteristic (ROC) curves and F1 scores were used to validate the performance of two models in both the training and testing cohort. Results: There were significant differences in rad-scores of the four sequences between T790M-positive and negative groups whether in the training or testing cohort (P<0.05). The lesion-level model consisting of rad-scores showed excellent discrimination, with an area under the curve (AUC) and F1-score of 0.879 and 0.798 in the training cohort, and 0.834 and 0.742 in the testing cohort, respectively. The patient-level model also showed a favorable discriminatory ability with an AUC and F1 score of 0.851 and 0.837, which was confirmed with an AUC and F1 score of 0.734 and 0.716 in the testing cohort. Conclusions: The MRI-based radiomics signatures may be new markers to identify patients at high risk of developing resistance in the early period.

7.
Front Immunol ; 13: 955944, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36238280

RESUMEN

Background: Few treatment options are available for brain metastases (BMs) in EGFR-mutant non-small cell lung cancer (NSCLC) that progress with prior EGFR tyrosine kinase inhibitor (EGFR-TKI) therapy. This study aimed to evaluate the efficacy of immune checkpoint inhibitor (ICI) therapy in these patients. Methods: NSCLC patients with confirmed sensitive EGFR mutations and BMs were retrospectively reviewed. All patients experienced failure of EGFR-TKI therapy and were divided into two cohorts based on subsequent treatment. Cohort 1 included patients who received ICI therapy, while cohort 2 included patients treated with chemotherapy. Overall and intracranial objective response rates (ORRs) were used to evaluate the treatment response. Overall and intacranial progression-free survival (PFS) were calculated by Kaplan-Meier analysis and compared with the log-rank test. Univariate and multivariate Cox analyses were used to identify prognostic factors. Results: A total of 53 patients treated with ICI therapy and 40 patients treated with chemotherapy were included in cohorts 1 and 2, respectively. In cohort 1, the overall ORR was 20.8%, with a median overall PFS of 4.2 months. The median intracranial PFS was 5.1 months. Of the 38 patients with measurable intracranial lesions, the intracranial ORR was 21.0%. Patients who received ICI combined with chemotherapy had the highest intracranial ORR of 37.5%. Compared to patients treated with chemotherapy in cohort 2, patients receiving ICI combined with chemotherapy had both longer intracranial PFS (6.4 vs. 5.1 months, p = 0.110) and overall PFS (6.2 vs. 4.6 months, p = 0.054), and these differences approached statistical significance. Univariate and multivariate Cox analyses demonstrated that high disease burden (p = 0.019), prior third-generation EGFR-TKI therapy (p = 0.019), and a poor lung immune prognostic index (LIPI) (p = 0.012) were independent negative predicators of overall PFS and that multiple BMs were negatively correlated with intracranial PFS among patients treated with ICI therapy. Conclusions: Our results suggested that ICI combined with chemotherapy had potent intracranial efficacy and may be a promising treatment candidate in EGFR-mutant NSCLC patients with BMs for whom prior EGFR-TKI therapy failed.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Receptores ErbB/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Estudios Retrospectivos
8.
Front Public Health ; 10: 1000415, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211679

RESUMEN

Background: The extent of the relationship between age and the presence of breast cancer synchronous brain metastases (BCSBMs) and mortality has not yet been well-identified or sufficiently quantified. We aimed to examine the association of age with the presence of BCSBMs and all-cause and cancer-specific mortality outcomes using the SEER database. Methods: Age-associated risk of the presence and survival of BCSBMs were evaluated on a continuous scale (restricted cubic spline, RCS) with logistic or Cox regression models. The main endpoints were the presence of BCSBMs and all-cause mortality or cancer-specific mortality. Cox proportional hazards regression and competing risk models were used in survival analysis. Results: Among 374,132 adult breast cancer patients, 1,441 (0.38%) had BMs. The presence of BCSBMs displayed a U-shaped relationship with age, with the highest point of the curve occurring at the age of 62. In both the younger (age ≤ 61) and older (age ≥ 62) groups, the observed curve showed a nearly linear relationship between age and the presence of BCSBMs. The relationship between age and all-cause mortality (ASM) and cancer-specific mortality (CSM) was linear. Older age at diagnosis was associated with a higher risk of ASM (HR 1.019, 95% CI: 1.013-1.024, p < 0.001) and CSM (HR 1.016, 95% CI: 1.010-1.023, p < 0.001) in multivariable Cox models. Age (sHR 1.007, 95% CI 1-1.013, p = 0.049) was substantially related to a significantly increased risk of CSM in competing risk models. Conclusion: Age had a non-linear U-shaped relationship with the presence of BCSBMs and a linear relationship with BCSBMs mortality.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Adulto , Femenino , Humanos , Modelos de Riesgos Proporcionales , Programa de VERF , Análisis de Supervivencia , Estados Unidos/epidemiología
9.
Heliyon ; 8(9): e10407, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36119883

RESUMEN

The third-generation epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) established a new standard for EGFR mutation positive non-small cell lung cancer (NSCLC) treatment. Brain metastases (BMS) are common in NSCLCs with poor prognosis, and patients with BMS who carry uncommon mutations is lack of treatment options. Aumolertinib is the first third-generation EGFR TKI in China and the second in the global context. There are few reports of the efficacy of aumolertinib in treating NSCLC patients with BMS who harboring uncommon EGFR mutations, which is needs to be investigated. Here we reported two cases of aumolertinib in treating NSCLC patients with BMS harboring EGFR G719X/L861Q mutations. The first one was diagnosed with poorly differentiated stage IVB adenocarcinoma with brain metastases. Genetic tests showed mutations in exons of EGFR 18 (G719D) and 21 (L861Q) initially. The patient was administered with pemetrexed 0.8 g and lobaplatin 30 mg, and aumolertinib (110 mg QD) combined with one-month of WBRT(Whole Brain Radiation Therapy) (42 Gy in 7 fractions). In order to avoid the side effects of radiotherapy on brain, radiotherapy was discontinued on February 5, 2020. The regime was continued with pemetrexed 0.75 g, lobaplatin 30 mg, bevacizumab 400 mg, and aumolertinib 110 mg QD. The four-drug combination regimen lasted for 6 months until serum tumor markers were elevated slightly. Then lobaplatin was replaced with nedaplatin, and the new four-drug combination regimen (pemetrexed 0.75 g, nedaplatin 90mg, bevacizumab 400 mg, and aumolertinib 110mg QD) was used with a one-month cycle for 3 cycles. Aumolertinib was administered daily throughout the four-drug combination, with the rest administered on day 1 of this treatment cycle. Chest CT scan were performed each month, which showed no progressed of lung disease. The patient had a progression-free survival of 13 months and is still being treated with aumolertinib. The second patient was diagnosed as right lung adenocarcinoma (T3N2M1) IVB secondary bone and brain malignancy carrying EGFR 18 (G719A) and 21 (L861Q) exon mutation. Aumolertinib combined with local radiotherapy was used after failure of afatinib combined with radiotherapy. Follow-up chest CT and brain MRI revealed that the lung lesions were partially relieved. Furthermore, the multiple nodules in the brain were relieved and cerebral edema was reduced. The aumolertinib monotherapy was continued, and follow-up imaging showed no disease progression. PFS was 13 months during the treatment with aumolertinib. The two cases showed good efficacy of aumolertinib in treating patients with brain metastases (BMS) that harbored EGFR 18 (G719X) and 21 (L861Q) mutations.

10.
Front Oncol ; 12: 920393, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35912214

RESUMEN

Introduction: There is a cumulative risk of 20-40% of developing brain metastases (BM) in solid cancers. Stereotactic radiotherapy (SRT) enables the application of high focal doses of radiation to a volume and is often used for BM treatment. However, SRT can cause adverse radiation effects (ARE), such as radiation necrosis, which sometimes cause irreversible damage to the brain. It is therefore of clinical interest to identify patients at a high risk of developing ARE. We hypothesized that models trained with radiomics features, deep learning (DL) features, and patient characteristics or their combination can predict ARE risk in patients with BM before SRT. Methods: Gadolinium-enhanced T1-weighted MRIs and characteristics from patients treated with SRT for BM were collected for a training and testing cohort (N = 1,404) and a validation cohort (N = 237) from a separate institute. From each lesion in the training set, radiomics features were extracted and used to train an extreme gradient boosting (XGBoost) model. A DL model was trained on the same cohort to make a separate prediction and to extract the last layer of features. Different models using XGBoost were built using only radiomics features, DL features, and patient characteristics or a combination of them. Evaluation was performed using the area under the curve (AUC) of the receiver operating characteristic curve on the external dataset. Predictions for individual lesions and per patient developing ARE were investigated. Results: The best-performing XGBoost model on a lesion level was trained on a combination of radiomics features and DL features (AUC of 0.71 and recall of 0.80). On a patient level, a combination of radiomics features, DL features, and patient characteristics obtained the best performance (AUC of 0.72 and recall of 0.84). The DL model achieved an AUC of 0.64 and recall of 0.85 per lesion and an AUC of 0.70 and recall of 0.60 per patient. Conclusion: Machine learning models built on radiomics features and DL features extracted from BM combined with patient characteristics show potential to predict ARE at the patient and lesion levels. These models could be used in clinical decision making, informing patients on their risk of ARE and allowing physicians to opt for different therapies.

11.
Diagnostics (Basel) ; 12(7)2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35885491

RESUMEN

Background: Brain metastases (BMs), particularly synchronous brain metastases, in colorectal cancer (CRC) patients are uncommon. The survival benefit of primary tumor resection (PTR) in patients with metastatic colorectal cancer is controversial. Whether PTR can bring survival benefits to patients with BMs of CRC has not been reported. Methods: From 2010 to 2016, 581 CRC patients with BMs from the Surveillance, Epidemiology, and End Results (SEER) database were divided into PTR and non-PTR groups. The log-rank test was used to compare the survival distributions. The Kaplan-Meier method was used to estimate survival. By controlling additional prognostic factors, a Cox proportional multivariate regression analysis was used to estimate the survival benefit of PTR. Results: The median overall survival for CRC patients with synchronous BMs was 3 months, with a 1-year survival rate of 27.2% and a 2-year survival rate of 12.8%. The PTR group contained 171 patients (29.4%), whereas the non-PTR group had 410 patients (70.6%). Patients who underwent PTR had a 1-year survival rate of 40.2% compared to 21.7% in those who did not (p < 0.0001). Cox proportional analysis showed that patients ≥60 years (hazard ratio [HR] 1.718, 95% confidence interval [CI] 1.423−2.075, p < 0.0001) had a shorter OS than patients < 60 years of age. OS was better in CEA-negative than in CEA-positive patients (HR 0.652, 95% CI 0.472−0.899, p = 0.009). Patients in whom the primary tumor was removed had considerably improved prognoses (HR 0.654, 95% CI 0.531−0.805, p < 0.0001). Subgroup analysis revealed that the PTR group achieved a survival advantage except for patients with CEA negative. Conclusions: Patients with synchronous BMs from CRC may benefit from primary tumor resection (PTR). Age, CEA level, and PTR were independent prognostic risk factors for CRC patients with synchronous BMs.

12.
Front Oncol ; 12: 859071, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35493999

RESUMEN

In this review, we provide the state of the art about brain metastases (BMs) from gestational trophoblastic neoplasia (GTN), a rare condition. Data concerning the epidemiology, clinical presentation, innovations in therapeutic modalities, and outcomes of GTN BMs are comprehensively presented with particular attention to the role of radiotherapy, neurosurgery, and the most recent chemotherapy regimens. Good response rates have been achieved thanks to multi-agent chemotherapy, but brain involvement by GTNs entails significant risks for patients' health since sudden and extensive intracranial hemorrhages are possible. Moreover, despite the evolution of treatment protocols, a small proportion of these patients ultimately develops a resistant disease. To tackle this unmet clinical need, immunotherapy has been recently proposed. The role of this novel option for this subset of patients as well as the achieved results so far are also discussed.

13.
Front Immunol ; 13: 852811, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35422812

RESUMEN

Brain metastases (BMs) in non-small-cell lung cancer (NSCLC) patients are associated with significant morbidity and poor prognosis. Immune checkpoint inhibitors (ICIs) have resulted in a paradigm shift in the management of advanced NSCLC. However, the value of ICIs in NSCLC patients with BMs remains unclear because patients with BMs are routinely excluded in numerous prospective trials on ICIs. Here, starting from the mechanisms of ICIs for BMs, we will reveal the value of ICIs by reviewing the efficacy and adverse effects of ICIs monotherapy as well as promising combination strategies, such as combinations with chemotherapy, radiotherapy, and anti-angiogenic drugs, etc. In addition, the methods of patient selection and response assessment will be summarized to assist clinical practice and further studies.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Estudios Prospectivos
14.
Front Neurol ; 13: 823731, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35370912

RESUMEN

Objectives: Dynamic susceptibility contrast perfusion weighted imaging (DSC-PWI) plays an important role in the differential diagnosis between radionecrosis and recurrence of brain metastases (BMs) after gamma knife radiosurgery (GKRS). While the perfusion condition of preliminary hyperperfusion and hypoperfusion BMs when recur has not been studied, as well the separating performance of quantitative DSC-PWI in both kinds of BMs. Methods: From February 2017 to October 2019, quantitative DSC-PWI was performed in patients with untreated BMs in this observational study. Patients were assigned to hyperperfusion and hypoperfusion group according the quantitative cerebral blood volume (qCBV). During follow-up after GKRS, patients with a diagnostic pitfall of radionecrosis and recurrence accepted second quantitative DSC-PWI. Final diagnosis was based on the histological results or follow-up results. Receiver operating curve analysis was used to explore the performance of qCBV. Results: Twenty-nine patients (mean age: 61.3 ± 9.4 years old; male/female: 13/16) were assigned to the group of hypoperfusion group, and 26 patients (mean age: 58 ± 10.4 years old; male/female: 14/12) to hyperperfusion group. The mean qCBV values between hypoperfusion and hyperperfusion groups when recurred were not significantly different (3.17 ± 0.53 ml/100 g vs. 3.27 ± 0.47 ml/100 g, p = 0.63). qCBV was feasible to separate radionecrosis and recurrence in both groups (AUC=0.94 and AUC=0.93, separately). Conclusion: Both premilitary hyperperfusion and hypoperfusion BMs would transform to a high microvascular density when recurs. qCBV is feasible to distinguish radionecrosis and recurrence among both kinds of BMs after GKRS.

15.
Transl Lung Cancer Res ; 11(2): 173-187, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35280308

RESUMEN

Background: Despite the emergence of programmed death 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors in the treatment of non-small cell lung cancer (NSCLC) patients with brain metastases (BMs), knowledge gaps remain regarding the impact and timing of cranial radiotherapy for patients receiving anti-PD-1/PD-L1 therapy. Methods: Data were collected from 461 consecutive patients who received anti-PD-1/PD-L1 therapy for metastatic NSCLC at three institutions between June 2017 and September 2020. Intracranial progressive disease (PD) at the original disease sites, new sites, or both sites were classified as original-site PD (OPD), new-site PD (NPD), and original-and-new-site PD (ONPD), respectively. Patients with baseline BMs were categorized based on whether they received upfront cranial radiotherapy (uCRT) at any time point between the introduction of anti-PD-1/PD-L1 therapy and the first subsequent progression. Results: Of the 461 patients enrolled, 110 (23.9%) had BMs at baseline. The presence of BMs did not show independent prognostic value for progression-free survival (PFS) or overall survival (OS). During a median follow-up of 13.2 months, 96 patients with BMs developed PD, of whom 53 (55.2%) experienced intracranial PD. OPD, NPD, and ONPD were observed in 50.9%, 18.9%, and 30.2% of patients, respectively. Patients who received uCRT exhibited a longer median OS than those with BMs who did not receive uCRT (25.4 vs. 14.6 months, HR: 0.52, 95% CI: 0.29-0.91, P=0.041); this survival advantage was more prominent in patients with 1-4 BMs (median OS, 25.4 vs. 17.0 months, HR: 0.42, 95% CI: 0.22-0.81, P=0.024), and uCRT was independently associated with OS among these patients. Conclusions: The presence of BMs at baseline was not associated with poorer OS in patients with metastatic NSCLC treated with anti-PD-1/PD-L1 therapy. Intracranial progression on PD-l/PD-L1 inhibitors predominately occurred at the original BM sites. The use of uCRT may improve OS, especially in NSCLC patients with 1-4 BMs.

16.
J Thorac Dis ; 14(2): 455-463, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35280489

RESUMEN

Background: Brain metastases (BMs) develop in 20-65% of non-small cell lung cancer (NSCLC) patients and are associated with a poor prognosis. Apatinib, a tyrosine kinase inhibitor (TKI) that selectively inhibits the vascular endothelial growth factor receptor 2, is safe and significantly prolongs the survival of chemotherapy-refractory gastric cancer patients. This retrospective study evaluated the safety and efficacy of apatinib combined with concurrent brain radiotherapy in NSCLC patients with BMs. Methods: This trial enrolled patients with non-recurrent BM from histologically-confirmed NSCLC without any limits regarding the BM size/quantity. Eligibility criteria were patients 18-75 years old with measurable BM from histologically-confirmed NSCLC (including both newly-diagnosed and previously treated NSCLC) and expected survival time greater than 3 months. Oral apatinib (500 or 250 mg/day) was started within 1 week prior to commencing whole brain radiotherapy with simultaneous integrated boost (WBRT-SIB) and continued until one week after radiotherapy completion. In addition to toxicities, analyzed outcomes included intracranial overall response rate (iORR), intracranial disease control rate (iDCR), intracranial progression free survival (iPFS), and overall survival (OS). Results: From July 2016 to January 2020, 16 patients were enrolled in this retrospective study. After 3 months of brain radiotherapy, the iORR was 75%, the iDCR was 100%, and the brain edema index (EI) was significantly reduced compared to that before brain radiation therapy (4.2 vs. 1.9; P=0.02). The median iPFS was 16.5 months [95% confidence interval (CI): 15.1-37.4 months]. The median OS was 26 months (95% CI: 17.0-54.0 months). Most of the patients tolerated apatinib well, but 7 patients had side effects, most commonly grade 1 or 2. Only 2 patients experienced grade 3 adverse events (hypertension and oral mucositis), and no grade 4 or 5 toxicities were observed. Conclusions: Apatinib combined with WBRT-SIB appears to be safe and effective in treating BMs in NSCLC patients.

17.
Chin Clin Oncol ; 11(2): 12, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34775779

RESUMEN

OBJECTIVE: The purpose of this narrative review is to survey the current literature and provide treatment recommendations for the management of patients with brain metastases (BMs) from small cell lung cancer (SCLC). BACKGROUND: BMs are very common in patients with SCLC and management has changed significantly in recent years. Radiation techniques and systemic therapies have evolved and this review will highlight these recent studies and implications on clinical practice. METHODS: This narrative review covers prophylaxis and treatment of established BMs in patients with SCLC. Studies included are based on Medline, PubMed, Google scholar searches, abstracts from conference proceedings and references from published papers. CONCLUSIONS: Radiation therapy continue to have an important role in the prophylaxis and treatment of patients with BMs. Radiation technologies have resulted in significant reduction of toxicities and improved quality of life (QoL). There continues to be many unanswered questions and results of currently accruing trials will hopefully fill some of these gaps and refine the role of radiation therapy in the management of this condition.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Pulmonares , Radiocirugia , Carcinoma Pulmonar de Células Pequeñas , Neoplasias Encefálicas/secundario , Irradiación Craneana , Humanos , Neoplasias Pulmonares/patología , Calidad de Vida , Radiocirugia/métodos , Carcinoma Pulmonar de Células Pequeñas/radioterapia
18.
Artículo en Inglés | MEDLINE | ID: mdl-38751521

RESUMEN

Breast cancer is one of the most common malignancies among women worldwide. According to the International Agency for Research on Cancer, breast cancer affected more Chinese women than any other cancer in 2020. The brain is an increasingly common metastatic sites of breast cancer. Although the risk of developing brain metastases (BMs) is lower in breast cancer than in lung cancer and melanoma, due to its high prevalence, it is the second most common cause of BM among solid tumors, being second only to lung cancer. The incidence of breast cancer brain metastasis (BCBM) differs by molecular subtype. Half of patients with advanced human epidermal growth factor receptor-2 (HER2)-positive and one-third of patients with triple-negative breast cancer (TNBC) develop BM. The clinical manifestations of leptomeningeal metastasis (LM) are often non-specific and may manifest as a variety of signs and symptoms, mainly including brain parenchyma involvement and meningeal irritation syndromes cranial nerve involvement, increased intracranial pressure, and progressive brain dysfunction. Therefore, the Chinese Society of Clinical Oncology (CSCO) Breast Cancer Committee has developed this expert consensus on BM, in an effort to improve the overall prognosis of BCBM and promote the standardized diagnosis and treatment of this disease. During the development of this expert consensus, we carried out a comprehensive literature review and referred to some of the most authoritative guidelines in China and abroad. In this consensus, we will discuss clinical manifestations, imaging examinations, pathological diagnosis, treatments, prognosis, follow-up and monitoring. We hope this consensus will be of help to all the clinicians majored in breast cancer and other similar professions.

19.
Ann Palliat Med ; 10(9): 9974-9983, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34628921

RESUMEN

BACKGROUND: Data focusing on the synergistic effect of programmed cell death 1 (PD-1) inhibitors and brain radiotherapy for brain metastases (BMs) in lung cancer is scarce. METHODS: A total of 60 lung cancer patients receiving PD-1 inhibitors with or without brain radiotherapy were identified in this retrospective study. The primary endpoints were intracranial progression-free survival (iPFS), extracranial progression-free survival (PFS), and overall survival (OS) among three groups. RESULTS: Twenty-one patients received PD-1 inhibitors and concurrent brain radiotherapy, 20 patients were treated with PD-1 inhibitors and non-concurrent brain radiotherapy, and the other 19 patients were treated with PD-1 inhibitors alone. Patients in the concurrent group achieved a higher intracranial objective response rate (iORR, 61.1% vs. 29.4% vs. 25.0%) and a higher intracranial disease control rate (iDCR, 83.3% vs. 58.8% vs. 56.3%) compared with those in the non-concurrent group and PD-1 inhibitors alone group. The median iPFS was significantly longer in the concurrent group than the non-concurrent group and the PD-1 inhibitors alone group (9.8, 5.7, and 4.8 months, P=0.039, respectively). The median PFS were 9.2, 5.7 and 4.6 months (P=0.347) in the concurrent group, non-concurrent group and PD-1 inhibitors alone group. And the median OS were not reached, 12.1 and 6.9 months (P=0.206), respectively. Multivariate analysis revealed that the lack of concurrent brain radiotherapy was independently associated with a shorter iPFS. CONCLUSIONS: PD-1 inhibitors with concurrent brain radiotherapy achieved a higher iORR, iDCR, and iPFS in lung cancer patients with treated or newly diagnosed BMs.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Pulmonares , Apoptosis , Encéfalo , Neoplasias Encefálicas/radioterapia , Humanos , Neoplasias Pulmonares/radioterapia , Estudios Retrospectivos
20.
Ann Palliat Med ; 10(2): 2379-2386, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33725780

RESUMEN

The brain is a common metastatic site of small cell lung cancer (SCLC), but systematic treatment options are limited by the blood-brain barrier. Currently, the optimal treatment regimen remains controversial, especially for patients already treated by brain radiotherapy. Anlotinib is a novel oral multitarget tyrosine kinase inhibitor which has shown significant improvement in progression-free survival and overall survival in third-line or beyond therapy of advanced SCLC in a randomized, double-blind phase II study (ALTER1202 trial) based on a Chinese population sample. Emerging data has also suggested that immunotherapy, such as the programmed death ligand 1 (PD-L1) inhibitor, has a relatively high response rate in brain metastatic SCLC, although there is a lack of large sample-size studies. Integrating anlotinib and immunotherapy for recurrent or relapsing brain metastases (BMs) of SCLC has not been previously reported, but it is possible that these two treatments may have synergistic effects and provide even better outcomes. Here, we present a case of stage III SCLC who developed lung and BMs after concurrent chemoradiotherapy (cCRT) and achieved radiographic locally complete regression following whole brain irradiation (WBI) with a simultaneous integrated boost (SIB) technique. Durvalumab was delivered as maintenance therapy. Asymptomatic multifocal recurrence of BMs occurred after the administration of the second dose of durvalumab. After administration of combined durvalumab and anlotinib, the BMs achieved near-complete regression and no severe toxicity was reported. This suggests a potential synergistic effect of combined durvalumab and anlotinib in previously treated BMs in a patient with SCLC and may provide a direction for future clinical decisions.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Anticuerpos Monoclonales , Encéfalo , Quimioradioterapia , Humanos , Indoles , Pulmón , Neoplasias Pulmonares/tratamiento farmacológico , Quinolinas , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA