May-Lucie Meyer, Fred R Hirsch, Paul A Bunn, Peter Ujhazy, David Fredrickson, Christine D Berg, David P Carbone, Balazs Halmos, Harpreet Singh, Hossein Borghaei, Andrea Ferris, Corey Langer, Sanja Dacic, Tony S Mok, Solange Peters, Bruce E Johnson
Lung cancer, the leading cause of cancer-related deaths globally, remains a pressing health issue despite significant medical advances. The New York Lung Cancer Foundation brought together experts from academia, the pharmaceutical and biotech industries as well as organizational leaders and patient advocates, to thoroughly examine the current state of lung cancer diagnosis, treatment, and research. The goal was to identify areas where our understanding is incomplete and to develop collaborative public health and scientific strategies to generate better patient outcomes, as highlighted in our "Calls to Action." The consortium prioritized 8 different calls to action. These include (1) develop strategies to cure more patients with early-stage lung cancer, (2) investigate carcinogenesis leading to lung cancers in patients without a history of smoking, (3) harness precision medicine for disease interception and prevention, (4) implement solutions to deliver prevention measures and effective therapies to individuals in under-resourced countries, (5) facilitate collaborations with industry to collect and share data and samples, (6) create and maintain open access to big data repositories, (7) develop new immunotherapeutic agents for lung cancer treatment and prevention, and (8) invest in research in both the academic and community settings. These calls to action provide guidance to representatives from academia, the pharmaceutical and biotech industries, organizational and regulatory leaders, and patient advocates to guide ongoing and planned initiatives.
{"title":"Calls to action on lung cancer management and research.","authors":"May-Lucie Meyer, Fred R Hirsch, Paul A Bunn, Peter Ujhazy, David Fredrickson, Christine D Berg, David P Carbone, Balazs Halmos, Harpreet Singh, Hossein Borghaei, Andrea Ferris, Corey Langer, Sanja Dacic, Tony S Mok, Solange Peters, Bruce E Johnson","doi":"10.1093/oncolo/oyae169","DOIUrl":"10.1093/oncolo/oyae169","url":null,"abstract":"<p><p>Lung cancer, the leading cause of cancer-related deaths globally, remains a pressing health issue despite significant medical advances. The New York Lung Cancer Foundation brought together experts from academia, the pharmaceutical and biotech industries as well as organizational leaders and patient advocates, to thoroughly examine the current state of lung cancer diagnosis, treatment, and research. The goal was to identify areas where our understanding is incomplete and to develop collaborative public health and scientific strategies to generate better patient outcomes, as highlighted in our \"Calls to Action.\" The consortium prioritized 8 different calls to action. These include (1) develop strategies to cure more patients with early-stage lung cancer, (2) investigate carcinogenesis leading to lung cancers in patients without a history of smoking, (3) harness precision medicine for disease interception and prevention, (4) implement solutions to deliver prevention measures and effective therapies to individuals in under-resourced countries, (5) facilitate collaborations with industry to collect and share data and samples, (6) create and maintain open access to big data repositories, (7) develop new immunotherapeutic agents for lung cancer treatment and prevention, and (8) invest in research in both the academic and community settings. These calls to action provide guidance to representatives from academia, the pharmaceutical and biotech industries, organizational and regulatory leaders, and patient advocates to guide ongoing and planned initiatives.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"e1634-e1645"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630765/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141604541","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Waleed S Al Amri, Ahmed H Al Amri, Aisha Al Abri, Thomas A Hughes, Fatma Al Lawati
Background: High prevalence of early-onset breast cancer (EOBC) has been reported in Middle Eastern populations. For example, in Oman more than 50% of patients with breast cancer (BC) are under age 45 at diagnosis. Causes for this high incidence are unknown. Germline BRCA gene mutations have been associated with EOBC, however, prevalence of these mutations and how they relate to EOBC in Oman has not been assessed.
Patients and methods: Clinical data were collected for patients with BC treated at Royal Hospital, Oman between 2010 and 2022. Germline BRCA1/2 gene mutations were identified using sequencing and MLPA. Correlation and Kaplan-Meier survival analyses were performed to test relationships among clinico-pathological features, gene mutations, and outcomes.
Results: Total of 1336 Middle Eastern patients with BC were included; 611 were aged <45 at diagnosis (45.7%). No significant correlation was found between BRCA1/2 mutation status and EOBC (P = .229), and the majority of EOBC cases had no family history of BC. EOBC tumors did, however, differ in clinicopathological features; EOBCs were significantly larger (P < .0001), of higher grade (P < .0001), and included more HER2-enriched, and triple negative subtypes (P = .018) compared with later onset cases. Accordingly, survival analyses revealed that EOBC had significantly worse disease-free survival (P = .002). BRCA gene variants showed a distinct range of mutations including, in BRCA2, 3 previously unreported mutations and 4 potential founder recurrent mutations.
Conclusion: Our findings showed that germline BRCA1/2 mutations were not over-represented in EOBC cases in Oman, and therefore are unlikely to be responsible for high EOBC rates.
{"title":"BRCA1/2 mutations and outcomes among Middle Eastern patients with early-onset breast cancer in Oman.","authors":"Waleed S Al Amri, Ahmed H Al Amri, Aisha Al Abri, Thomas A Hughes, Fatma Al Lawati","doi":"10.1093/oncolo/oyae214","DOIUrl":"10.1093/oncolo/oyae214","url":null,"abstract":"<p><strong>Background: </strong>High prevalence of early-onset breast cancer (EOBC) has been reported in Middle Eastern populations. For example, in Oman more than 50% of patients with breast cancer (BC) are under age 45 at diagnosis. Causes for this high incidence are unknown. Germline BRCA gene mutations have been associated with EOBC, however, prevalence of these mutations and how they relate to EOBC in Oman has not been assessed.</p><p><strong>Patients and methods: </strong>Clinical data were collected for patients with BC treated at Royal Hospital, Oman between 2010 and 2022. Germline BRCA1/2 gene mutations were identified using sequencing and MLPA. Correlation and Kaplan-Meier survival analyses were performed to test relationships among clinico-pathological features, gene mutations, and outcomes.</p><p><strong>Results: </strong>Total of 1336 Middle Eastern patients with BC were included; 611 were aged <45 at diagnosis (45.7%). No significant correlation was found between BRCA1/2 mutation status and EOBC (P = .229), and the majority of EOBC cases had no family history of BC. EOBC tumors did, however, differ in clinicopathological features; EOBCs were significantly larger (P < .0001), of higher grade (P < .0001), and included more HER2-enriched, and triple negative subtypes (P = .018) compared with later onset cases. Accordingly, survival analyses revealed that EOBC had significantly worse disease-free survival (P = .002). BRCA gene variants showed a distinct range of mutations including, in BRCA2, 3 previously unreported mutations and 4 potential founder recurrent mutations.</p><p><strong>Conclusion: </strong>Our findings showed that germline BRCA1/2 mutations were not over-represented in EOBC cases in Oman, and therefore are unlikely to be responsible for high EOBC rates.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"e1714-e1722"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630774/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142074598","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marwan Fakih, Jaideep Sandhu, Xiaochen Li, Chongkai Wang
Background: There have been conflicting reports on the predictive impact of metastatic disease sites on the response to checkpoint inhibitors (CPI) in microsatellite instability (MSI) metastatic colorectal cancers (mCRC). Recent studies have highlighted peritoneal metastases, ascites, and liver metastases as possible indicators of resistance to CPI.
Methods: We performed a detailed analysis of high microsatellite instability (MSI-H) mCRC treated with programmed cell death (PD-1) or PD-1/cytotoxic T-lymphocyte-associated protein 4 CPI in a single center. Overall response rate (ORR), progression-free survival (PFS), overall survival (OS), and stable disease but with complete pathological response upon resection (SDcPR) were analyzed by the presence of liver metastases, peritoneal metastases, or absence of either. The impact of number and size of liver metastases on clinical outcomes were also interrogated.
Results: Thirty-five patients with MSI mCRC were included in the analysis. Patients with peritoneal metastatic disease had lower ORR and shorter PFS compared to patients without liver and peritoneal metastases. Contrary to recent reports, ORR and ORR + SDcPR rates were high in patients with liver metastases, at 58% and 66%, respectively. In the liver metastases category, a better response rate was noted for patients with<5 lesions compared to patients with more than 5 lesions. Patients who responded had a higher median tumor mutation burden than patients with progressive disease.
Conclusions: In MSI mCRC, no single clinical characteristic was sufficient to preclude CPI response. Peritoneal metastatic disease was associated with numerically lower ORR and shorter PFS. In contrast, liver metastases do not predict poor outcome.
{"title":"Liver metastases and peritoneal metastases and response to checkpoint inhibitors in metastatic colorectal cancer with microsatellite instability.","authors":"Marwan Fakih, Jaideep Sandhu, Xiaochen Li, Chongkai Wang","doi":"10.1093/oncolo/oyae249","DOIUrl":"10.1093/oncolo/oyae249","url":null,"abstract":"<p><strong>Background: </strong>There have been conflicting reports on the predictive impact of metastatic disease sites on the response to checkpoint inhibitors (CPI) in microsatellite instability (MSI) metastatic colorectal cancers (mCRC). Recent studies have highlighted peritoneal metastases, ascites, and liver metastases as possible indicators of resistance to CPI.</p><p><strong>Methods: </strong>We performed a detailed analysis of high microsatellite instability (MSI-H) mCRC treated with programmed cell death (PD-1) or PD-1/cytotoxic T-lymphocyte-associated protein 4 CPI in a single center. Overall response rate (ORR), progression-free survival (PFS), overall survival (OS), and stable disease but with complete pathological response upon resection (SDcPR) were analyzed by the presence of liver metastases, peritoneal metastases, or absence of either. The impact of number and size of liver metastases on clinical outcomes were also interrogated.</p><p><strong>Results: </strong>Thirty-five patients with MSI mCRC were included in the analysis. Patients with peritoneal metastatic disease had lower ORR and shorter PFS compared to patients without liver and peritoneal metastases. Contrary to recent reports, ORR and ORR + SDcPR rates were high in patients with liver metastases, at 58% and 66%, respectively. In the liver metastases category, a better response rate was noted for patients with<5 lesions compared to patients with more than 5 lesions. Patients who responded had a higher median tumor mutation burden than patients with progressive disease.</p><p><strong>Conclusions: </strong>In MSI mCRC, no single clinical characteristic was sufficient to preclude CPI response. Peritoneal metastatic disease was associated with numerically lower ORR and shorter PFS. In contrast, liver metastases do not predict poor outcome.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"1052-1058"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630756/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142332559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Adam Brufsky, Xianchen Liu, Benjamin Li, Lynn McRoy, Connie Chen, Doris Makari, Rachel M Layman, Hope S Rugo
Background: Patients with cardiovascular disease (CVD) comorbidities are often excluded from participating in breast cancer clinical trials. Consequently, data to inform treatment decisions for patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer (mBC) and CVD are limited.
Objective: We compared the effectiveness of first-line palbociclib plus an aromatase inhibitor (AI) vs an AI alone and evaluated palbociclib treatment patterns in patients with HR+/HER2- mBC and CVD in routine clinical practice.
Methods: Data from the Flatiron Health Analytic Database were captured for patients with HR+/HER2- mBC and CVD who initiated first-line treatment with palbociclib plus an AI or an AI alone between February 2015 and March 2020 (data cutoff: September 30, 2020). Overall survival (OS), real-world progression-free survival (PFS), and treatment patterns were evaluated.
Results: Of the 469 patients with identifiable CVD, 160 received palbociclib plus an AI, and 309 received an AI alone. After stabilized inverse probability treatment weighting, both median OS (40.7 vs 26.5 months; hazard ratio [HR], 0.732 [95% CI, 0.537-0.997]; P = .048) and median real-world PFS (20.0 vs 12.5 months; HR, 0.679 [95% CI, 0.512-0.900]; P = .007) were significantly prolonged in patients treated with palbociclib plus an AI vs an AI alone. Among patients with a documented palbociclib starting dose, 78.5% started palbociclib at 125 mg/day, and 38.6% experienced dose adjustment.
Conclusions: In this real-world analysis, first-line palbociclib plus an AI was associated with improved effectiveness compared with an AI alone in patients with HR+/HER2- mBC and CVD.
背景:合并心血管疾病(CVD)的患者通常被排除在乳腺癌临床试验之外。因此,为激素受体阳性/人表皮生长因子受体 2 阴性(HR+/HER2-)转移性乳腺癌(mBC)和心血管疾病患者的治疗决策提供参考的数据非常有限:我们比较了一线帕博西尼(palbociclib)联合芳香化酶抑制剂(AI)与单独使用AI的疗效,并评估了常规临床实践中HR+/HER2- mBC和心血管疾病患者的帕博西尼治疗模式:从Flatiron健康分析数据库中获取了HR+/HER2- mBC和CVD患者的数据,这些患者在2015年2月至2020年3月期间(数据截止日期:2020年9月30日)开始接受palbociclib联合AI或单独AI的一线治疗。对总生存期(OS)、实际无进展生存期(PFS)和治疗模式进行了评估:结果:在469例可确定患有心血管疾病的患者中,160例接受了palbociclib加AI治疗,309例单独接受了AI治疗。经过稳定的逆概率治疗加权后,palbociclib联合AI与单用AI相比,中位OS(40.7个月 vs 26.5个月;危险比[HR],0.732 [95% CI,0.537-0.997];P = .048)和中位实际PFS(20.0个月 vs 12.5个月;HR,0.679 [95% CI,0.512-0.900];P = .007)均显著延长。在有palbociclib起始剂量记录的患者中,78.5%的患者从125 mg/天开始服用palbociclib,38.6%的患者经历了剂量调整:在这项真实世界分析中,对于HR+/HER2- mBC和心血管疾病患者,一线palbociclib加一种AI比单独使用AI更有效:NCT05361655(ClinicalTrials.gov)。
{"title":"Palbociclib plus aromatase inhibitors in patients with metastatic breast cancer and cardiovascular diseases: real-world effectiveness.","authors":"Adam Brufsky, Xianchen Liu, Benjamin Li, Lynn McRoy, Connie Chen, Doris Makari, Rachel M Layman, Hope S Rugo","doi":"10.1093/oncolo/oyae273","DOIUrl":"10.1093/oncolo/oyae273","url":null,"abstract":"<p><strong>Background: </strong>Patients with cardiovascular disease (CVD) comorbidities are often excluded from participating in breast cancer clinical trials. Consequently, data to inform treatment decisions for patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer (mBC) and CVD are limited.</p><p><strong>Objective: </strong>We compared the effectiveness of first-line palbociclib plus an aromatase inhibitor (AI) vs an AI alone and evaluated palbociclib treatment patterns in patients with HR+/HER2- mBC and CVD in routine clinical practice.</p><p><strong>Methods: </strong>Data from the Flatiron Health Analytic Database were captured for patients with HR+/HER2- mBC and CVD who initiated first-line treatment with palbociclib plus an AI or an AI alone between February 2015 and March 2020 (data cutoff: September 30, 2020). Overall survival (OS), real-world progression-free survival (PFS), and treatment patterns were evaluated.</p><p><strong>Results: </strong>Of the 469 patients with identifiable CVD, 160 received palbociclib plus an AI, and 309 received an AI alone. After stabilized inverse probability treatment weighting, both median OS (40.7 vs 26.5 months; hazard ratio [HR], 0.732 [95% CI, 0.537-0.997]; P = .048) and median real-world PFS (20.0 vs 12.5 months; HR, 0.679 [95% CI, 0.512-0.900]; P = .007) were significantly prolonged in patients treated with palbociclib plus an AI vs an AI alone. Among patients with a documented palbociclib starting dose, 78.5% started palbociclib at 125 mg/day, and 38.6% experienced dose adjustment.</p><p><strong>Conclusions: </strong>In this real-world analysis, first-line palbociclib plus an AI was associated with improved effectiveness compared with an AI alone in patients with HR+/HER2- mBC and CVD.</p><p><strong>Trial registration: </strong>NCT05361655 (ClinicalTrials.gov).</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"1032-1043"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630750/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142481144","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yilin Wu, Yanling Li, Bin Chen, Ying Zhang, Wanying Xing, Baoliang Guo, Wan Wang
Background: Neoadjuvant treatment has been developed as a systematic approach for patients with early breast cancer and has resulted in improved breast-conserving rate and survival. However, identifying treatment-sensitive patients at the early phase of therapy remains a problem, hampering disease management and raising the possibility of disease progression during treatment.
Methods: In this retrospective analysis, we collected 2-deoxy-2-[F-18] fluoro-d-glucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) images of primary tumor sites and axillary areas and reciprocal clinical pathological data from 121 patients who underwent neoadjuvant treatment and surgery in our center. The univariate and multivariate logistic regression analyses were performed to investigate features associated with pathological complete response (pCR). An 18F-FDG PET/CT-based prediction model was trained, and the performance was evaluated by receiver operating characteristic curves (ROC).
Results: The maximum standard uptake values (SUVmax) of 18F-FDG PET/CT were a powerful indicator of tumor status. The SUVmax values of axillary areas were closely related to metastatic lymph node counts (R = 0.62). Moreover, the early SUVmax reduction rates (between baseline and second cycle of neoadjuvant treatment) were statistically different between pCR and non-pCR patients. The early SUVmax reduction rates-based model showed great ability to predict pCR (AUC = 0.89), with all molecular subtypes (HR+HER2-, HR+HER2+, HR-HER2+, and HR-HER2-) considered.
Conclusion: Our research proved that the SUVmax reduction rate of 18F-FDG PET/CT contributed to the early prediction of pCR, providing rationales for utilizing PET/CT in NAT in the future.
{"title":"18F-FDG PET/CT for early prediction of pathological complete response in breast cancer neoadjuvant therapy: a retrospective analysis.","authors":"Yilin Wu, Yanling Li, Bin Chen, Ying Zhang, Wanying Xing, Baoliang Guo, Wan Wang","doi":"10.1093/oncolo/oyae185","DOIUrl":"10.1093/oncolo/oyae185","url":null,"abstract":"<p><strong>Background: </strong>Neoadjuvant treatment has been developed as a systematic approach for patients with early breast cancer and has resulted in improved breast-conserving rate and survival. However, identifying treatment-sensitive patients at the early phase of therapy remains a problem, hampering disease management and raising the possibility of disease progression during treatment.</p><p><strong>Methods: </strong>In this retrospective analysis, we collected 2-deoxy-2-[F-18] fluoro-d-glucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) images of primary tumor sites and axillary areas and reciprocal clinical pathological data from 121 patients who underwent neoadjuvant treatment and surgery in our center. The univariate and multivariate logistic regression analyses were performed to investigate features associated with pathological complete response (pCR). An 18F-FDG PET/CT-based prediction model was trained, and the performance was evaluated by receiver operating characteristic curves (ROC).</p><p><strong>Results: </strong>The maximum standard uptake values (SUVmax) of 18F-FDG PET/CT were a powerful indicator of tumor status. The SUVmax values of axillary areas were closely related to metastatic lymph node counts (R = 0.62). Moreover, the early SUVmax reduction rates (between baseline and second cycle of neoadjuvant treatment) were statistically different between pCR and non-pCR patients. The early SUVmax reduction rates-based model showed great ability to predict pCR (AUC = 0.89), with all molecular subtypes (HR+HER2-, HR+HER2+, HR-HER2+, and HR-HER2-) considered.</p><p><strong>Conclusion: </strong>Our research proved that the SUVmax reduction rate of 18F-FDG PET/CT contributed to the early prediction of pCR, providing rationales for utilizing PET/CT in NAT in the future.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"e1646-e1655"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630790/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141753350","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Surgery and radiotherapy are primary nonconservative treatments for prostate cancer (PCa). However, personalizing treatment options between these treatment modalities is challenging due to unclear criteria. We developed an artificial intelligence (AI)-based model that can identify patients with localized PCa who would benefit more from either radiotherapy or surgery, thereby providing personalized clinical decision-making.
Material and methods: Data from consecutive patients with localized PCa who received radiotherapy or surgery with complete records of clinicopathological variables and follow-up results in 12 registries of the Surveillance, Epidemiology, and End Results database were analyzed. Patients from 7 registries were randomly assigned to training (TD) and internal validation datasets (IVD) at a 9:1 ratio. The remaining 5 registries constituted the external validation dataset (EVD). TD was divided into training-radiotherapy (TRD) and training-surgery (TSD) datasets, and IVD was divided into internal-radiotherapy (IRD) and internal-surgery (ISD) datasets. Six models for radiotherapy and surgery were trained using TRD and TSD to predict radiotherapy survival probability (RSP) and surgery survival probability (SSP), respectively. The models with the highest concordance index (C-index) on IRD and ISD were chosen to form the final treatment recommendation model (FTR). FTR recommendations were based on the higher value between RSP and SSP. Kaplan-Meier curves were generated for patients receiving recommended (consistent group) and nonrecommended treatments (inconsistent group), which were compared using the log-rank test.
Results: The study included 118 236 patients, categorized into TD (TRD: 44 621; TSD: 41 500), IVD (IRD: 4949; ISD: 4621), and EVD (22 545). Both radiotherapy and surgery models accurately predicted RSP and SSP (C-index: 0.735-0.787 and 0.769-0.797, respectively). The consistent group exhibited higher survival rates than the inconsistent group, particularly among patients not suitable for active surveillance (P < .001).
Conclusion: FTR accurately identifies patients with localized PCa who would benefit more from either radiotherapy or surgery, offering clinicians an effective AI tool to make informed choices between these 2 treatments.
{"title":"Artificial intelligence-based personalized clinical decision-making for patients with localized prostate cancer: surgery versus radiotherapy.","authors":"Yuwei Liu, Litao Zhao, Jiangang Liu, Liang Wang","doi":"10.1093/oncolo/oyae184","DOIUrl":"10.1093/oncolo/oyae184","url":null,"abstract":"<p><strong>Background: </strong>Surgery and radiotherapy are primary nonconservative treatments for prostate cancer (PCa). However, personalizing treatment options between these treatment modalities is challenging due to unclear criteria. We developed an artificial intelligence (AI)-based model that can identify patients with localized PCa who would benefit more from either radiotherapy or surgery, thereby providing personalized clinical decision-making.</p><p><strong>Material and methods: </strong>Data from consecutive patients with localized PCa who received radiotherapy or surgery with complete records of clinicopathological variables and follow-up results in 12 registries of the Surveillance, Epidemiology, and End Results database were analyzed. Patients from 7 registries were randomly assigned to training (TD) and internal validation datasets (IVD) at a 9:1 ratio. The remaining 5 registries constituted the external validation dataset (EVD). TD was divided into training-radiotherapy (TRD) and training-surgery (TSD) datasets, and IVD was divided into internal-radiotherapy (IRD) and internal-surgery (ISD) datasets. Six models for radiotherapy and surgery were trained using TRD and TSD to predict radiotherapy survival probability (RSP) and surgery survival probability (SSP), respectively. The models with the highest concordance index (C-index) on IRD and ISD were chosen to form the final treatment recommendation model (FTR). FTR recommendations were based on the higher value between RSP and SSP. Kaplan-Meier curves were generated for patients receiving recommended (consistent group) and nonrecommended treatments (inconsistent group), which were compared using the log-rank test.</p><p><strong>Results: </strong>The study included 118 236 patients, categorized into TD (TRD: 44 621; TSD: 41 500), IVD (IRD: 4949; ISD: 4621), and EVD (22 545). Both radiotherapy and surgery models accurately predicted RSP and SSP (C-index: 0.735-0.787 and 0.769-0.797, respectively). The consistent group exhibited higher survival rates than the inconsistent group, particularly among patients not suitable for active surveillance (P < .001).</p><p><strong>Conclusion: </strong>FTR accurately identifies patients with localized PCa who would benefit more from either radiotherapy or surgery, offering clinicians an effective AI tool to make informed choices between these 2 treatments.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"e1692-e1700"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630763/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141857217","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Laura H Rosenberger, Richard F Riedel, Emilia J Diego, Amanda L Nash, Juneko E Grilley-Olson, Natalie A Danziger, Ethan S Sokol, Jeffrey S Ross, Sarah L Sammons
Background: Malignant phyllodes tumors (MPT) are rare fibroepithelial breast cancers with no known effective systemic therapy; metastatic progression portends a dismal prognosis. We sought to describe the genomic landscape of MPTs through genomic profiling and immunotherapeutic biomarker analysis.
Materials and methods: Cases of sequenced MPT were identified from a Clinical Laboratory Improvement Amendments-certified, College of American Pathologists-accredited laboratory (Foundation Medicine). All cases underwent genomic profiling using adaptor ligation-based, next-generation sequencing assay of 324 genes. Tumor agnostic immunotherapy biomarkers, microsatellite instability, tumor mutational burden (TMB), and programmed death-ligand 1 (PD-L1) expression were evaluated. Fisher's Exact Tests and analysis of variance were used to test for differences between groups and for continuous variables as appropriate.
Results: Of 135 MPT cases identified; 94 (69.6%) were localized/locally recurrent and 41 (30.4%) were metastatic. Median age was 54 years (range 14-86). The median TMB was 2.5 mut/Mb and 3 were TMB-high (≥10 mut/Mb). 21.4% were PD-L1+ via Dako 22C3 assay (CPS ≥1). Most commonly altered genes included TERT-promoter (69.7%), CDKN2A (45.9%), TP53 (37.8%), NF1 (35.6%), CDKN2B (33.3%), MED12 (28.9%), MTAP (27.7%), KMT2D (22.2%), PIK3CA (20.0%), PTEN (18.5%), and RB1 (18.5%). Several tumors harboring genomic alterations with US Food and Drug Administration-approved indications in other tumor types were found including NF1, PIK3CA, EGFR Exon 19/20 insertions, and BRAF V600E mutations.
Conclusions: In the largest genomic evaluation of MPT to date, multiple clinically actionable mutations were found. Routine sequencing of metastatic MPT may provide additional information to guide treatment decisions and clinical trial enrollment.
{"title":"Genomic landscape of malignant phyllodes tumors reveals multiple targetable opportunities.","authors":"Laura H Rosenberger, Richard F Riedel, Emilia J Diego, Amanda L Nash, Juneko E Grilley-Olson, Natalie A Danziger, Ethan S Sokol, Jeffrey S Ross, Sarah L Sammons","doi":"10.1093/oncolo/oyae218","DOIUrl":"10.1093/oncolo/oyae218","url":null,"abstract":"<p><strong>Background: </strong>Malignant phyllodes tumors (MPT) are rare fibroepithelial breast cancers with no known effective systemic therapy; metastatic progression portends a dismal prognosis. We sought to describe the genomic landscape of MPTs through genomic profiling and immunotherapeutic biomarker analysis.</p><p><strong>Materials and methods: </strong>Cases of sequenced MPT were identified from a Clinical Laboratory Improvement Amendments-certified, College of American Pathologists-accredited laboratory (Foundation Medicine). All cases underwent genomic profiling using adaptor ligation-based, next-generation sequencing assay of 324 genes. Tumor agnostic immunotherapy biomarkers, microsatellite instability, tumor mutational burden (TMB), and programmed death-ligand 1 (PD-L1) expression were evaluated. Fisher's Exact Tests and analysis of variance were used to test for differences between groups and for continuous variables as appropriate.</p><p><strong>Results: </strong>Of 135 MPT cases identified; 94 (69.6%) were localized/locally recurrent and 41 (30.4%) were metastatic. Median age was 54 years (range 14-86). The median TMB was 2.5 mut/Mb and 3 were TMB-high (≥10 mut/Mb). 21.4% were PD-L1+ via Dako 22C3 assay (CPS ≥1). Most commonly altered genes included TERT-promoter (69.7%), CDKN2A (45.9%), TP53 (37.8%), NF1 (35.6%), CDKN2B (33.3%), MED12 (28.9%), MTAP (27.7%), KMT2D (22.2%), PIK3CA (20.0%), PTEN (18.5%), and RB1 (18.5%). Several tumors harboring genomic alterations with US Food and Drug Administration-approved indications in other tumor types were found including NF1, PIK3CA, EGFR Exon 19/20 insertions, and BRAF V600E mutations.</p><p><strong>Conclusions: </strong>In the largest genomic evaluation of MPT to date, multiple clinically actionable mutations were found. Routine sequencing of metastatic MPT may provide additional information to guide treatment decisions and clinical trial enrollment.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"1024-1031"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630793/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142082623","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The impact of sidedness on survival of later-line treatment in patients with metastatic colorectal cancer (mCRC) is undetermined. This study aimed to investigate the association between sidedness and survival among chemotherapy refractory patients with mCRC treated with trifluridine/tipiracil (TAS-102) or regorafenib or both.
Patients and methods: Patients with mCRC treated with TAS-102 or regorafenib between 2015 and 2020 was retrospectively collected. Patients were stratified into TAS-102 first and regorafenib first, then subdivided into TAS-102 followed by regorafenib (T-R) and regorafenib followed by TAS-102 (R-T) groups. The oncologic outcomes were presented with time-to-treatment failure (TTF) and overall survival (OS).
Results: After matching, 376 TAS-102 patients and 376 regorafenib patients were included for outcomes comparison. TTF had insignificant differences while OS was significantly different between TAS-102 and regorafenib groups. Median TTF and OS were 1.9 months versus 2.0 months (P = .701) and 9.1 months versus 7.0 months (P = .008) in TAS-102 and regorafenib, respectively. The OS benefits were consistent regardless primary tumor location. Subgroup analysis with 174 T-R patients and 174 R-T patients was investigated for treatment sequences. TTF and OS had significant differences in both groups. Median TTF and OS were 8.5 months versus 6.3 months (P = .001) and 14.4 months versus 12.6 months (P = .035) in T-R and R-T groups, respectively. The TTF and OS benefits were persisted regardless primary tumor location.
Conclusion: TAS-102 first provided a better survival benefit in chemotherapy refractory patients with mCRC across all sidedness. Further prospective studies are warranted to validate our conclusions.
{"title":"Association between sidedness and survival among chemotherapy refractory metastatic colorectal cancer patients treated with trifluridine/tipiracil or regorafenib.","authors":"Kai-Yuan Hsiao, Hsin-Pao Chen, Kun-Ming Rau, Kuang-Wen Liu, Ben-Chang Shia, Wei-Shan Chang, Hao-Yun Liang, Meng-Che Hsieh","doi":"10.1093/oncolo/oyae235","DOIUrl":"10.1093/oncolo/oyae235","url":null,"abstract":"<p><strong>Background: </strong>The impact of sidedness on survival of later-line treatment in patients with metastatic colorectal cancer (mCRC) is undetermined. This study aimed to investigate the association between sidedness and survival among chemotherapy refractory patients with mCRC treated with trifluridine/tipiracil (TAS-102) or regorafenib or both.</p><p><strong>Patients and methods: </strong>Patients with mCRC treated with TAS-102 or regorafenib between 2015 and 2020 was retrospectively collected. Patients were stratified into TAS-102 first and regorafenib first, then subdivided into TAS-102 followed by regorafenib (T-R) and regorafenib followed by TAS-102 (R-T) groups. The oncologic outcomes were presented with time-to-treatment failure (TTF) and overall survival (OS).</p><p><strong>Results: </strong>After matching, 376 TAS-102 patients and 376 regorafenib patients were included for outcomes comparison. TTF had insignificant differences while OS was significantly different between TAS-102 and regorafenib groups. Median TTF and OS were 1.9 months versus 2.0 months (P = .701) and 9.1 months versus 7.0 months (P = .008) in TAS-102 and regorafenib, respectively. The OS benefits were consistent regardless primary tumor location. Subgroup analysis with 174 T-R patients and 174 R-T patients was investigated for treatment sequences. TTF and OS had significant differences in both groups. Median TTF and OS were 8.5 months versus 6.3 months (P = .001) and 14.4 months versus 12.6 months (P = .035) in T-R and R-T groups, respectively. The TTF and OS benefits were persisted regardless primary tumor location.</p><p><strong>Conclusion: </strong>TAS-102 first provided a better survival benefit in chemotherapy refractory patients with mCRC across all sidedness. Further prospective studies are warranted to validate our conclusions.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"e1669-e1679"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630785/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142156700","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jaime Shaw, Xerxes Pundole, Akhila Balasubramanian, Erik S Anderson, Malaika Pastel, D Gwyn Bebb, Tony Jiang, Pablo Martinez, Suresh S Ramalingam, Hossein Borghaei
Background: The landscape of small cell lung cancer (SCLC) has changed since the 2019 and 2020 approvals of anti-PD-L1 atezolizumab and durvalumab for first-line (1L) treatment in combination with chemotherapy. We studied treatment patterns and real-world overall survival (rwOS) following 1L-3L therapy.
Patients and methods: A nationwide electronic health record (EHR)-derived de-identified database was used to describe treatment patterns, characteristics, and survival of patients with extensive-stage (ES)-SCLC by 1L anti-PD-L1 treatment. Patients with ES-SCLC who initiated ≥1 line of systemic therapy from 2013 to 2021, with potential follow-up through 2022, were included.
Results: Among 9952 patients with SCLC, there were 4308 patients with ES-SCLC treated during the study period who met eligibility criteria. Etoposide + platinum (EP) chemotherapy was most common in the 1L, with addition of anti-PD-L1 therapy to most regimens by 2019. Second-line regimens varied by platinum sensitivity status and shifted from topotecan to lurbinectedin over time. Median rwOS following 1L therapy was 8.3 months (95% CI, 7.9-8.8) in those treated with 1L anti-PD-L1 and 8.0 months (95% CI, 7.8-8.2) in those who were not. Following 2L and 3L, median rwOS was 5.6 (95% CI, 4.9-6.3) and 4.9 months (95% CI, 3.4-6.0), respectively, among 1L anti-PD-L1-treated, and 4.5 (95% CI, 4.2-4.9) and 4.0 months (95% CI, 3.7-4.5), respectively, among those who were not.
Conclusion: Despite the introduction of frontline anti-PD-L1 therapy, survival remains dismal among patients with ES-SCLC treated in the real-world setting.
{"title":"Recent treatment patterns and real-world survival following first-line anti-PD-L1 treatment for extensive-stage small cell lung cancer.","authors":"Jaime Shaw, Xerxes Pundole, Akhila Balasubramanian, Erik S Anderson, Malaika Pastel, D Gwyn Bebb, Tony Jiang, Pablo Martinez, Suresh S Ramalingam, Hossein Borghaei","doi":"10.1093/oncolo/oyae234","DOIUrl":"10.1093/oncolo/oyae234","url":null,"abstract":"<p><strong>Background: </strong>The landscape of small cell lung cancer (SCLC) has changed since the 2019 and 2020 approvals of anti-PD-L1 atezolizumab and durvalumab for first-line (1L) treatment in combination with chemotherapy. We studied treatment patterns and real-world overall survival (rwOS) following 1L-3L therapy.</p><p><strong>Patients and methods: </strong>A nationwide electronic health record (EHR)-derived de-identified database was used to describe treatment patterns, characteristics, and survival of patients with extensive-stage (ES)-SCLC by 1L anti-PD-L1 treatment. Patients with ES-SCLC who initiated ≥1 line of systemic therapy from 2013 to 2021, with potential follow-up through 2022, were included.</p><p><strong>Results: </strong>Among 9952 patients with SCLC, there were 4308 patients with ES-SCLC treated during the study period who met eligibility criteria. Etoposide + platinum (EP) chemotherapy was most common in the 1L, with addition of anti-PD-L1 therapy to most regimens by 2019. Second-line regimens varied by platinum sensitivity status and shifted from topotecan to lurbinectedin over time. Median rwOS following 1L therapy was 8.3 months (95% CI, 7.9-8.8) in those treated with 1L anti-PD-L1 and 8.0 months (95% CI, 7.8-8.2) in those who were not. Following 2L and 3L, median rwOS was 5.6 (95% CI, 4.9-6.3) and 4.9 months (95% CI, 3.4-6.0), respectively, among 1L anti-PD-L1-treated, and 4.5 (95% CI, 4.2-4.9) and 4.0 months (95% CI, 3.7-4.5), respectively, among those who were not.</p><p><strong>Conclusion: </strong>Despite the introduction of frontline anti-PD-L1 therapy, survival remains dismal among patients with ES-SCLC treated in the real-world setting.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"1079-1089"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630770/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142332561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Andreas Reichinger, Leo Essl, Paul Kerschner, Jonathan Burghofer, Gerald Webersinke, Holger Rumpold, Bernhard Doleschal
Background: Biliary tract cancers (BTCs) are a diverse group of malignancies with varied genetic backgrounds. The prevalence of intrahepatic cholangiocarcinoma (iCC) is increasing, particularly in Western countries. Despite advancements in treatments, the prognosis for BTC remains poor. Recent molecular profiling has revealed that up to 40% of iCC cases have targetable genetic alterations. MET amplification, although rare, presents a significant target for therapy.
Case presentation: A 25-year-old female with a history of ulcerative colitis presented with shoulder pain and a positron emission tomography-computed tomography (PET-CT) scan revealed an enlarged liver and multiple metastases. Histopathological analysis diagnosed poorly differentiated adenocarcinoma. First-line therapy with Cisplatin, Gemcitabine, and Durvalumab resulted in disease progression. Molecular profiling identified a TP53 mutation and MET amplification. Based on these findings, Tepotinib was initiated. Tepotinib treatment led to a significant reduction in tumor size and normalization of CA 19-9 levels within 2 months, achieving a complete metabolic remission lasting up to 17 months. The treatment was well tolerated with minimal side effects.
Discussion: MET-amplified BTCs are exceedingly rare, and evidence for targeted treatment is limited. This case demonstrates the efficacy of Tepotinib in a young patient with MET-amplified iCC, showing a long-term response and suggesting a potential new standard treatment option for this molecularly defined entity. This case also highlights the aggressive nature of MET-amplified tumors and the need for targeted second-line therapies.
Conclusion: Tepotinib showed remarkable efficacy in treating MET-amplified intrahepatic cholangiocarcinoma, underscoring the importance of molecular profiling in BTCs and suggesting a potential new therapeutic approach for this rare cancer subtype.
背景:胆道癌(BTC)是一组遗传背景各不相同的恶性肿瘤。肝内胆管癌(iCC)的发病率正在上升,尤其是在西方国家。尽管治疗手段不断进步,但肝内胆管癌的预后仍然很差。最近的分子图谱分析表明,多达 40% 的 iCC 病例存在可靶向的基因改变。MET扩增虽然罕见,但却是治疗的一个重要靶点:正电子发射计算机断层扫描(PET-CT)显示肝脏肿大并有多处转移。组织病理分析诊断为分化不良腺癌。使用顺铂、吉西他滨和Durvalumab的一线治疗导致疾病进展。分子分析发现了 TP53 突变和 MET 扩增。基于这些发现,患者开始接受特泊替尼治疗。特泊替尼治疗使肿瘤体积显著缩小,CA 19-9水平在2个月内恢复正常,实现了长达17个月的完全代谢缓解。患者对治疗的耐受性良好,副作用极小:讨论:MET扩增的BTC极为罕见,靶向治疗的证据有限。本病例展示了特泊替尼对一名年轻的MET扩增iCC患者的疗效,显示了长期的应答,为这种分子定义的实体提供了一种潜在的新标准治疗方案。该病例还强调了MET扩增肿瘤的侵袭性以及二线靶向治疗的必要性:结论:特泊替尼治疗MET扩增的肝内胆管癌疗效显著,突显了BTCs分子图谱分析的重要性,并为这种罕见的癌症亚型提供了一种潜在的新治疗方法。
{"title":"Exceptional sustained long-term complete response to Tepotinib in a MET-amplified advanced intrahepatic biliary tract cancer failing Durvalumab plus Cisplatin and Gemcitabine.","authors":"Andreas Reichinger, Leo Essl, Paul Kerschner, Jonathan Burghofer, Gerald Webersinke, Holger Rumpold, Bernhard Doleschal","doi":"10.1093/oncolo/oyae265","DOIUrl":"10.1093/oncolo/oyae265","url":null,"abstract":"<p><strong>Background: </strong>Biliary tract cancers (BTCs) are a diverse group of malignancies with varied genetic backgrounds. The prevalence of intrahepatic cholangiocarcinoma (iCC) is increasing, particularly in Western countries. Despite advancements in treatments, the prognosis for BTC remains poor. Recent molecular profiling has revealed that up to 40% of iCC cases have targetable genetic alterations. MET amplification, although rare, presents a significant target for therapy.</p><p><strong>Case presentation: </strong>A 25-year-old female with a history of ulcerative colitis presented with shoulder pain and a positron emission tomography-computed tomography (PET-CT) scan revealed an enlarged liver and multiple metastases. Histopathological analysis diagnosed poorly differentiated adenocarcinoma. First-line therapy with Cisplatin, Gemcitabine, and Durvalumab resulted in disease progression. Molecular profiling identified a TP53 mutation and MET amplification. Based on these findings, Tepotinib was initiated. Tepotinib treatment led to a significant reduction in tumor size and normalization of CA 19-9 levels within 2 months, achieving a complete metabolic remission lasting up to 17 months. The treatment was well tolerated with minimal side effects.</p><p><strong>Discussion: </strong>MET-amplified BTCs are exceedingly rare, and evidence for targeted treatment is limited. This case demonstrates the efficacy of Tepotinib in a young patient with MET-amplified iCC, showing a long-term response and suggesting a potential new standard treatment option for this molecularly defined entity. This case also highlights the aggressive nature of MET-amplified tumors and the need for targeted second-line therapies.</p><p><strong>Conclusion: </strong>Tepotinib showed remarkable efficacy in treating MET-amplified intrahepatic cholangiocarcinoma, underscoring the importance of molecular profiling in BTCs and suggesting a potential new therapeutic approach for this rare cancer subtype.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":"1090-1094"},"PeriodicalIF":4.8,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142362466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}