首页 > 最新文献

Therapeutic Advances in Medical Oncology最新文献

英文 中文
Identification of a central network hub of key prognostic genes based on correlation between transcriptomics and survival in patients with metastatic solid tumors. 根据转移性实体瘤患者转录组学与生存期之间的相关性,确定关键预后基因的中心网络枢纽。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-17 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241289200
Vladimir Lazar, Eric Raymond, Shai Magidi, Catherine Bresson, Fanny Wunder, Ioana Berindan-Neagoe, Annemilaï Tijeras-Rabaland, Jacques Raynaud, Amir Onn, Michel Ducreux, Gerald Batist, Ulrik Lassen, Fin Cilius Nielsen, Richard L Schilsky, Eitan Rubin, Razelle Kurzrock

Background: Dysregulated pathways in cancer may be hub addicted. Identifying these dysregulated networks for targeting might lead to novel therapeutic options.

Objective: Considering the hypothesis that central hubs are associated with increased lethality, identifying key hub targets within central networks could lead to the development of novel drugs with improved efficacy in advanced metastatic solid tumors.

Design: Exploring transcriptomic data (22,000 gene products) from the WINTHER trial (N = 101 patients with various metastatic cancers), in which both tumor and normal organ-matched tissue were available.

Methods: A retrospective in silico analysis of all genes in the transcriptome was conducted to identify genes different in expression between tumor and normal tissues (paired t-test) and to determine their association with survival outcomes using survival analysis (Cox proportional hazard regression algorithm). Based on the biological relevance of the identified genes, hub targets of interest within central networks were then pinpointed. Patients were grouped based on the expression level of these genes (K-mean clustering), and the association of these groups with survival was examined (Cox proportional hazard regression algorithm, Forest plot, and Kaplan-Meier plot).

Results: We identified four key central hub genes-PLOD3, ARHGAP11A, RNF216, and CDCA8, for which high expression in tumor tissue compared to analogous normal tissue had the most significant correlation with worse outcomes. The correlation was independent of tumor or treatment type. The combination of the four genes showed the highest significance and correlation with the poorer outcome: overall survival (hazard ratio (95% confidence interval (CI)) = 10.5 (3.43-31.9) p = 9.12E-07 log-rank test in a Cox proportional hazard regression model). Findings were validated in independent cohorts.

Conclusion: The expression of PLOD3, ARHGAP11A, RNF216, and CDCA8 constitute, when combined, a prognostic tool, agnostic of tumor type and previous treatments. These genes represent potential targets for intercepting central hub networks in various cancers, offering avenues for novel therapeutic interventions.

背景:癌症中失调的通路可能是上瘾的枢纽。确定这些失调网络的靶点可能会带来新的治疗方案:考虑到中心枢纽与致死率增加有关的假说,确定中心网络中的关键枢纽靶点可能会开发出对晚期转移性实体瘤有更好疗效的新型药物:探索来自 WINTHER 试验(N = 101 名各种转移性癌症患者)的转录组数据(22,000 个基因产物),其中既有肿瘤组织,也有正常器官匹配组织:方法:对转录组中的所有基因进行回顾性硅分析,以确定肿瘤组织和正常组织之间表达不同的基因(配对 t 检验),并通过生存分析(Cox 比例危险回归算法)确定这些基因与生存结果的关系。根据已确定基因的生物学相关性,然后确定中心网络中感兴趣的枢纽靶点。根据这些基因的表达水平对患者进行分组(K-均值聚类),并研究这些分组与生存的关系(Cox比例危险回归算法、Forest图和Kaplan-Meier图):我们发现了四个关键的中心基因--LLOD3、ARHGAP11A、RNF216和CDCA8,与类似的正常组织相比,这些基因在肿瘤组织中的高表达与较差的预后有最显著的相关性。这种相关性与肿瘤或治疗类型无关。这四个基因的组合与较差预后的相关性最高:总生存期(危险比(95% 置信区间 (CI)) = 10.5 (3.43-31.9) p = 9.12E-07 在考克斯比例危险回归模型中的对数rank检验)。研究结果在独立队列中得到了验证:结论:PLOD3、ARHGAP11A、RNF216 和 CDCA8 的表达结合在一起,构成了一种预后工具,与肿瘤类型和既往治疗无关。这些基因是拦截各种癌症中枢网络的潜在靶点,为新型治疗干预提供了途径。
{"title":"Identification of a central network hub of key prognostic genes based on correlation between transcriptomics and survival in patients with metastatic solid tumors.","authors":"Vladimir Lazar, Eric Raymond, Shai Magidi, Catherine Bresson, Fanny Wunder, Ioana Berindan-Neagoe, Annemilaï Tijeras-Rabaland, Jacques Raynaud, Amir Onn, Michel Ducreux, Gerald Batist, Ulrik Lassen, Fin Cilius Nielsen, Richard L Schilsky, Eitan Rubin, Razelle Kurzrock","doi":"10.1177/17588359241289200","DOIUrl":"https://doi.org/10.1177/17588359241289200","url":null,"abstract":"<p><strong>Background: </strong>Dysregulated pathways in cancer may be hub addicted. Identifying these dysregulated networks for targeting might lead to novel therapeutic options.</p><p><strong>Objective: </strong>Considering the hypothesis that central hubs are associated with increased lethality, identifying key hub targets within central networks could lead to the development of novel drugs with improved efficacy in advanced metastatic solid tumors.</p><p><strong>Design: </strong>Exploring transcriptomic data (22,000 gene products) from the WINTHER trial (<i>N</i> = 101 patients with various metastatic cancers), in which both tumor and normal organ-matched tissue were available.</p><p><strong>Methods: </strong>A retrospective in silico analysis of all genes in the transcriptome was conducted to identify genes different in expression between tumor and normal tissues (paired <i>t</i>-test) and to determine their association with survival outcomes using survival analysis (Cox proportional hazard regression algorithm). Based on the biological relevance of the identified genes, hub targets of interest within central networks were then pinpointed. Patients were grouped based on the expression level of these genes (<i>K</i>-mean clustering), and the association of these groups with survival was examined (Cox proportional hazard regression algorithm, Forest plot, and Kaplan-Meier plot).</p><p><strong>Results: </strong>We identified four key central hub genes-<i>PLOD3, ARHGAP11A, RNF216</i>, and <i>CDCA8</i>, for which high expression in tumor tissue compared to analogous normal tissue had the most significant correlation with worse outcomes. The correlation was independent of tumor or treatment type. The combination of the four genes showed the highest significance and correlation with the poorer outcome: overall survival (hazard ratio (95% confidence interval (CI)) = 10.5 (3.43-31.9) <i>p</i> = 9.12E-07 log-rank test in a Cox proportional hazard regression model). Findings were validated in independent cohorts.</p><p><strong>Conclusion: </strong>The expression of <i>PLOD3, ARHGAP11A, RNF216</i>, and <i>CDCA8</i> constitute, when combined, a prognostic tool, agnostic of tumor type and previous treatments. These genes represent potential targets for intercepting central hub networks in various cancers, offering avenues for novel therapeutic interventions.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487509/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475450","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}
引用次数: 0
Age-related differences in progression patterns, follow-up strategies, and postoperative outcomes in locally advanced rectal cancer: insights from a large-scale validated study. 局部晚期直肠癌的进展模式、随访策略和术后结果中与年龄相关的差异:一项大规模验证研究的启示。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-17 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241290129
Yilin Yu, Haixia Wu, Jianjian Qiu, Liang Hong, Shiji Wu, Lingdong Shao, Cheng Lin, Zhiping Wang, Junxin Wu

Background: Locally advanced rectal cancer (LARC) presents significant treatment challenges, particularly as patient age may influence disease progression and treatment response. Understanding the differences in progression patterns and treatment outcomes between older patient (OP) and non-older patient (NOP) is essential for tailoring effective management strategies.

Objectives: We aimed to explore the differences of progression pattern, postoperative treatment, and survival outcome between OP and NOP groups in LARC.

Design/methods: The random survival forest model was used to determine the probability of time-to-event occurrence every 3 months. Patients in the NOP and OP group were both categorized into three risk groups based on progression-free survival nomogram scores. We employed inverse probability of treatment weighting (IPTW) analysis and the Surveillance, Epidemiology, and End Results (SEER) database to verify our findings.

Results: Our results revealed that Groups 1, 2, and 3 experienced peaks in progression within the first 24 months in NOP group. As for OP group, Group 4 reached a progression peak at the 18th month, Group 5 at the 12th month, and Group 6 at the 9th month. In NOP group, high-risk patients who underwent postoperative chemotherapy had significantly improved overall survival compared to those who did not. Additionally, postoperative chemotherapy did not significantly improve prognosis for patients in low-, moderate-, or high-risk groups of OP group. Finally, the validation results of IPTW analysis and SEER database showed compliance with our findings.

Conclusion: For NOP group, we recommended close follow-up during the first 2 years. As for OP group, it was suggested to conduct close follow-up at the 18th, 12th, and 9th month for low-, moderate-, and high-risk groups, respectively. Furthermore, postoperative chemotherapy can provide survival benefits for patients in high-risk group of NOP group. However, OP group patients should be informed that the potential benefits of postoperative chemotherapy may be minimal.

背景:局部晚期直肠癌(LARC)给治疗带来了巨大挑战,尤其是患者年龄可能会影响疾病进展和治疗反应。了解老年患者(OP)和非老年患者(NOP)在疾病进展模式和治疗效果方面的差异,对于制定有效的治疗策略至关重要:目的:我们旨在探讨 LARC 患者中 OP 组和 NOP 组在进展模式、术后治疗和生存结果方面的差异:设计/方法:采用随机生存森林模型确定每 3 个月事件发生的时间概率。NOP组和OP组患者均根据无进展生存期提名图评分分为三个风险组。我们采用了反向治疗概率加权(IPTW)分析和监测、流行病学和最终结果(SEER)数据库来验证我们的研究结果:结果显示,NOP 组的第 1、2 和 3 组在最初 24 个月内出现了进展高峰。至于 OP 组,第 4 组在第 18 个月达到进展高峰,第 5 组在第 12 个月,第 6 组在第 9 个月。在 NOP 组中,与未接受术后化疗的患者相比,接受术后化疗的高危患者的总生存率明显提高。此外,术后化疗并不能明显改善 OP 组低、中、高危患者的预后。最后,IPTW分析和SEER数据库的验证结果显示与我们的研究结果一致:结论:对于 NOP 组,我们建议在头两年进行密切随访。结论:对于 NOP 组,我们建议在最初 2 年进行密切随访;对于 OP 组,建议分别在第 18 个月、第 12 个月和第 9 个月对低危、中危和高危组进行密切随访。此外,术后化疗可为 NOP 组的高危患者带来生存益处。但应告知 OP 组患者,术后化疗的潜在益处可能微乎其微。
{"title":"Age-related differences in progression patterns, follow-up strategies, and postoperative outcomes in locally advanced rectal cancer: insights from a large-scale validated study.","authors":"Yilin Yu, Haixia Wu, Jianjian Qiu, Liang Hong, Shiji Wu, Lingdong Shao, Cheng Lin, Zhiping Wang, Junxin Wu","doi":"10.1177/17588359241290129","DOIUrl":"https://doi.org/10.1177/17588359241290129","url":null,"abstract":"<p><strong>Background: </strong>Locally advanced rectal cancer (LARC) presents significant treatment challenges, particularly as patient age may influence disease progression and treatment response. Understanding the differences in progression patterns and treatment outcomes between older patient (OP) and non-older patient (NOP) is essential for tailoring effective management strategies.</p><p><strong>Objectives: </strong>We aimed to explore the differences of progression pattern, postoperative treatment, and survival outcome between OP and NOP groups in LARC.</p><p><strong>Design/methods: </strong>The random survival forest model was used to determine the probability of time-to-event occurrence every 3 months. Patients in the NOP and OP group were both categorized into three risk groups based on progression-free survival nomogram scores. We employed inverse probability of treatment weighting (IPTW) analysis and the Surveillance, Epidemiology, and End Results (SEER) database to verify our findings.</p><p><strong>Results: </strong>Our results revealed that Groups 1, 2, and 3 experienced peaks in progression within the first 24 months in NOP group. As for OP group, Group 4 reached a progression peak at the 18th month, Group 5 at the 12th month, and Group 6 at the 9th month. In NOP group, high-risk patients who underwent postoperative chemotherapy had significantly improved overall survival compared to those who did not. Additionally, postoperative chemotherapy did not significantly improve prognosis for patients in low-, moderate-, or high-risk groups of OP group. Finally, the validation results of IPTW analysis and SEER database showed compliance with our findings.</p><p><strong>Conclusion: </strong>For NOP group, we recommended close follow-up during the first 2 years. As for OP group, it was suggested to conduct close follow-up at the 18th, 12th, and 9th month for low-, moderate-, and high-risk groups, respectively. Furthermore, postoperative chemotherapy can provide survival benefits for patients in high-risk group of NOP group. However, OP group patients should be informed that the potential benefits of postoperative chemotherapy may be minimal.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487512/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475436","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}
引用次数: 0
Expert consensus on the use of third-generation EGFR-TKIs in EGFR-mutated advanced non-small cell lung cancer with various T790M mutations post-resistance to first-/second-generation EGFR-TKIs. 关于对第一代/第二代表皮生长因子受体-TKIs耐药后出现各种T790M突变的表皮生长因子受体突变晚期非小细胞肺癌使用第三代表皮生长因子受体-TKIs的专家共识。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-17 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241289648
Jietao Ma, Letian Huang, Chengbo Han

Third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have emerged as the mainstay of treatment for advanced EGFR-mutant advanced non-small cell lung cancer (NSCLC), effectively overcoming the problems of acquired threonine-to-methionine (T790M) mutations associated with the first- or second-generation TKIs. Evidence from several studies suggests that these agents, including osimertinib and aumolertinib, also show potential benefits in T790M-negative or unknown populations, particularly those with brain metastases, where the high permeability of the blood-brain barrier allows effective control of intracranial lesions. Despite the encouraging results, further high-quality research, including prospective trials, is warranted to fully elucidate the efficacy profiles of these third-generation TKIs in T790M-negative or unknown NSCLC patients after first- or second-line TKI failure. The present expert consensus highlights the evolving role of third-generation EGFR-TKIs in overcoming therapeutic resistance and optimizing patient outcomes.

第三代表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKIs)已成为治疗晚期表皮生长因子受体突变晚期非小细胞肺癌(NSCLC)的主要药物,有效克服了第一代或第二代 TKIs 所带来的获得性苏氨酸-蛋氨酸(T790M)突变问题。几项研究的证据表明,包括奥西替尼(osimertinib)和奥莫替尼(aumolertinib)在内的这些药物在T790M阴性或未知人群中也显示出潜在的优势,尤其是那些脑转移患者,因为血脑屏障的高通透性可以有效控制颅内病变。尽管结果令人鼓舞,但仍需进一步开展高质量的研究,包括前瞻性试验,以全面阐明这些第三代 TKIs 在一线或二线 TKI 治疗失败后的 T790M 阴性或未知 NSCLC 患者中的疗效。本专家共识强调了第三代表皮生长因子受体抑制剂(EGFR-TKIs)在克服耐药性和优化患者预后方面不断发展的作用。
{"title":"Expert consensus on the use of third-generation EGFR-TKIs in EGFR-mutated advanced non-small cell lung cancer with various T790M mutations post-resistance to first-/second-generation EGFR-TKIs.","authors":"Jietao Ma, Letian Huang, Chengbo Han","doi":"10.1177/17588359241289648","DOIUrl":"https://doi.org/10.1177/17588359241289648","url":null,"abstract":"<p><p>Third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have emerged as the mainstay of treatment for advanced EGFR-mutant advanced non-small cell lung cancer (NSCLC), effectively overcoming the problems of acquired threonine-to-methionine (T790M) mutations associated with the first- or second-generation TKIs. Evidence from several studies suggests that these agents, including osimertinib and aumolertinib, also show potential benefits in T790M-negative or unknown populations, particularly those with brain metastases, where the high permeability of the blood-brain barrier allows effective control of intracranial lesions. Despite the encouraging results, further high-quality research, including prospective trials, is warranted to fully elucidate the efficacy profiles of these third-generation TKIs in T790M-negative or unknown NSCLC patients after first- or second-line TKI failure. The present expert consensus highlights the evolving role of third-generation EGFR-TKIs in overcoming therapeutic resistance and optimizing patient outcomes.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492187/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475449","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}
引用次数: 0
CD34 as a potential prognostic indicator for camrelizumab response in advanced non-small-cell lung cancer: insights from digital spatial profiling. CD34 作为晚期非小细胞肺癌康瑞珠单抗反应的潜在预后指标:数字空间图谱分析的启示。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-17 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241289671
Xinyi Huang, Baoqing Tian, Ziyuan Ren, Jingxin Zhang, Weiwei Yan, You Mo, Jupeng Yuan, Yujiao Ma, Ruiyang Wang, Rufei Liu, Minxin Chen, Jinming Yu, Dawei Chen

Background: Given that only a small subset of patients with advanced non-small-cell lung cancer (aNSCLC) benefit from immune checkpoint inhibitors (ICIs), the effectiveness of ICIs is often compromised by the complex interplay within the tumor microenvironment (TME).

Objectives: To identify predictive biomarkers associated with ICI resistance at a multi-omics spatial level.

Design: A total of eight aNSCLC patients who received first-line anti-programmed cell death protein-1 (PD-1) monoclonal antibody camrelizumab at Shandong Cancer Hospital and Institute between 2021 and 2022 were included in the discovery cohort. An additional validation cohort of 45 samples from camrelizumab-treated aNSCLC patients was also enrolled.

Methods: NanoString GeoMx® digital spatial profiling was conducted at the transcriptomic and proteomic level within pan-cytokeratin (panCK+), CD45+, and CD68+ compartments. For validation, multiplex immunofluorescence (mIF) staining was performed.

Results: Distinct spatial expression patterns and levels of immune infiltration were observed between tumor and leukocyte compartments. Higher CD34 expression in the macrophage compartment correlated with poorer prognosis and response to camrelizumab (p < 0.05). mIF validation confirmed the association of elevated CD34 expression level with reduced progression-free survival (PFS; hazard ratio (HR) = 5.011, 95% confidence interval: 1.057-23.752, p = 0.042), outperforming traditional tumor markers in predictive accuracy.

Conclusion: Our findings identify CD34 as a novel spatial biomarker for anti-PD-1 therapy efficacy, potentially guiding the selection of aNSCLC patients who are more likely to benefit from ICI treatment.

Trial registration: ChiCTR2000040416.

背景:鉴于只有一小部分晚期非小细胞肺癌(aNSCLC)患者从免疫检查点抑制剂(ICIs)中获益,ICIs的有效性往往会受到肿瘤微环境(TME)内复杂相互作用的影响:在多组学空间水平上确定与 ICI 耐药性相关的预测性生物标志物:设计:发现队列中纳入了2021年至2022年期间在山东省肿瘤医院和研究所接受一线抗程序性细胞死亡蛋白-1(PD-1)单克隆抗体康瑞珠单抗治疗的8例ANSCLC患者。另外还加入了一个验证队列,该队列中有45个样本来自接受过康瑞珠单抗治疗的非小细胞肺癌患者:方法:在泛细胞角蛋白(panCK+)、CD45+和CD68+区段内进行了转录组和蛋白质组水平的NanoString GeoMx®数字空间谱分析。为了进行验证,还进行了多重免疫荧光(mIF)染色:结果:在肿瘤和白细胞间观察到了不同的空间表达模式和免疫浸润水平。巨噬细胞分区中 CD34 的高表达与较差的预后和对坎瑞珠单抗的反应相关(p p = 0.042),其预测准确性优于传统的肿瘤标志物:我们的研究结果表明,CD34是抗PD-1疗法疗效的新型空间生物标志物,可能指导选择更有可能从ICI治疗中获益的非小细胞肺癌患者:ChiCTR2000040416。
{"title":"CD34 as a potential prognostic indicator for camrelizumab response in advanced non-small-cell lung cancer: insights from digital spatial profiling.","authors":"Xinyi Huang, Baoqing Tian, Ziyuan Ren, Jingxin Zhang, Weiwei Yan, You Mo, Jupeng Yuan, Yujiao Ma, Ruiyang Wang, Rufei Liu, Minxin Chen, Jinming Yu, Dawei Chen","doi":"10.1177/17588359241289671","DOIUrl":"https://doi.org/10.1177/17588359241289671","url":null,"abstract":"<p><strong>Background: </strong>Given that only a small subset of patients with advanced non-small-cell lung cancer (aNSCLC) benefit from immune checkpoint inhibitors (ICIs), the effectiveness of ICIs is often compromised by the complex interplay within the tumor microenvironment (TME).</p><p><strong>Objectives: </strong>To identify predictive biomarkers associated with ICI resistance at a multi-omics spatial level.</p><p><strong>Design: </strong>A total of eight aNSCLC patients who received first-line anti-programmed cell death protein-1 (PD-1) monoclonal antibody camrelizumab at Shandong Cancer Hospital and Institute between 2021 and 2022 were included in the discovery cohort. An additional validation cohort of 45 samples from camrelizumab-treated aNSCLC patients was also enrolled.</p><p><strong>Methods: </strong>NanoString GeoMx<sup>®</sup> digital spatial profiling was conducted at the transcriptomic and proteomic level within pan-cytokeratin (panCK<sup>+</sup>), CD45<sup>+</sup>, and CD68<sup>+</sup> compartments. For validation, multiplex immunofluorescence (mIF) staining was performed.</p><p><strong>Results: </strong>Distinct spatial expression patterns and levels of immune infiltration were observed between tumor and leukocyte compartments. Higher CD34 expression in the macrophage compartment correlated with poorer prognosis and response to camrelizumab (<i>p</i> < 0.05). mIF validation confirmed the association of elevated CD34 expression level with reduced progression-free survival (PFS; hazard ratio (HR) = 5.011, 95% confidence interval: 1.057-23.752, <i>p</i> = 0.042), outperforming traditional tumor markers in predictive accuracy.</p><p><strong>Conclusion: </strong>Our findings identify CD34 as a novel spatial biomarker for anti-PD-1 therapy efficacy, potentially guiding the selection of aNSCLC patients who are more likely to benefit from ICI treatment.</p><p><strong>Trial registration: </strong>ChiCTR2000040416.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11489950/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475437","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}
引用次数: 0
Should we be afraid of radiotherapy for hemorrhagic brain metastases? A narrative review. 我们应该害怕出血性脑转移瘤的放射治疗吗?叙述性综述。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-15 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241289203
Aleksandra Łupicka, Weronika Kowalczyk, Bartosz Cyman, Mateusz Spałek

Brain metastases (BM) are the most common intracranial malignancies. They are responsible for death as well as impairment of quality of life and cognitive function. In some cases, BMs can cause intracranial hemorrhage, which is not only responsible for the acute onset of either a new focal neurological deficit or worsening of a preexisting focal deficit but also poses a new challenge in treatment planning and clinical management. The aim of this study was to evaluate the available treatment modalities and their efficacy in hemorrhagic brain metastases (HBMs) with special attention to radiotherapy. In this review, we searched PubMed, BMJ, NCBI, Springer, BMC Cancer, Cochrane, and Google Scholar for articles containing data on the diagnosis and treatment of patients with HBMs, excluding the pediatric population. Treatment strategies consist of neurosurgery, whole brain radiotherapy, and stereotactic techniques (fractionated stereotactic radiosurgery (fSRS)/stereotactic radiosurgery (SRS)). Although the optimal treatment strategy for HBMs has not been established, we found no convincing evidence that radiotherapy, especially fSRS/SRS, is contraindicated in HBMs. We concluded that fSRS/SRS is a promising option for patients with HBM, particularly when surgical intervention poses risks.

{"title":"Should we be afraid of radiotherapy for hemorrhagic brain metastases? A narrative review.","authors":"Aleksandra Łupicka, Weronika Kowalczyk, Bartosz Cyman, Mateusz Spałek","doi":"10.1177/17588359241289203","DOIUrl":"https://doi.org/10.1177/17588359241289203","url":null,"abstract":"<p><p>Brain metastases (BM) are the most common intracranial malignancies. They are responsible for death as well as impairment of quality of life and cognitive function. In some cases, BMs can cause intracranial hemorrhage, which is not only responsible for the acute onset of either a new focal neurological deficit or worsening of a preexisting focal deficit but also poses a new challenge in treatment planning and clinical management. The aim of this study was to evaluate the available treatment modalities and their efficacy in hemorrhagic brain metastases (HBMs) with special attention to radiotherapy. In this review, we searched PubMed, BMJ, NCBI, Springer, BMC Cancer, Cochrane, and Google Scholar for articles containing data on the diagnosis and treatment of patients with HBMs, excluding the pediatric population. Treatment strategies consist of neurosurgery, whole brain radiotherapy, and stereotactic techniques (fractionated stereotactic radiosurgery (fSRS)/stereotactic radiosurgery (SRS)). Although the optimal treatment strategy for HBMs has not been established, we found no convincing evidence that radiotherapy, especially fSRS/SRS, is contraindicated in HBMs. We concluded that fSRS/SRS is a promising option for patients with HBM, particularly when surgical intervention poses risks.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11481081/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475455","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}
引用次数: 0
Real-world efficacy of PD-1/PD-L1 inhibitors in patients with advanced pulmonary neuroendocrine carcinoma: a single-center analysis. PD-1/PD-L1抑制剂对晚期肺神经内分泌癌患者的实际疗效:单中心分析。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-14 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241288130
Wanchen Zhai, Ying Yu, Haicheng Wu, Qian Zhang, Yunfei Chen, Yehao Yang, Yun Fan

Background: Immunotherapy blocking programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) has revolutionized the treatment of extensive-stage small-cell lung cancer (SCLC), but only with limited real-world efficacy data; evidence from immunotherapy for other pulmonary neuroendocrine carcinoma (PNEC) is scarce.

Objective: The purpose of this study is to evaluate the efficacy of receiving PD-1/PD-L1 inhibitors in patients with advanced PNEC and explore factors related to survival prognosis, providing clues for treatment for patients with advanced PNEC.

Methods: In all, 203 patients with advanced PNEC who received PD-1/PD-L1 inhibitors between January 2019 and December 2021 were retrospectively analyzed. Kaplan-Meier curves were constructed for progression-free survival (PFS) and overall survival (OS).

Results: For the 203 patients, the objective response rate (ORR) was 48.3%, the disease control rate (DCR) was 83.3%, the median PFS (mPFS) was 6.0 months, and the median OS (mOS) was 13.1 months. Among them, the histology was 166 SCLC, 13 large-cell neuroendocrine carcinoma, and 24 other unspecified PNEC. Histologically, no significant difference was observed in PFS (p = 0.240) or OS (p = 0.845). In first-line (1L) treatment (N = 125), patients received chemoimmunotherapy and had an ORR of 64.8%, DCR of 92.0%, mPFS of 6.6 months, and mOS of 14.9 months. In second-line (2L) or later-line setting, the ORR, DCR, mPFS, and mOS were 21.8%, 69.2%, 4.4, and 9.4 months; immunotherapy plus small-molecule antiangiogenic agents showed significantly greater PFS than immunotherapy monotherapy or chemoimmunotherapy (6.4 vs 1.4 vs 3.7 months, p = 0.041). Patients without liver metastasis had superior PFS (7.0 vs 5.1 months, p < 0.001) and OS (19.2 vs 9.6 months, p < 0.001) than those with liver metastasis.

Conclusion: In clinical practice, PD-1/PD-L1 inhibitors are effective in patients with advanced PNEC, regardless of the pathological histology. The efficacy of 1L immunochemotherapy is worthy of recognition, and the addition of small-molecule antiangiogenic agents to immunotherapy in 2L or later-line treatment provides a better survival trend.

Design: Retrospective study.

背景:阻断程序性死亡-1(PD-1)/程序性死亡配体-1(PD-L1)的免疫疗法彻底改变了广泛期小细胞肺癌(SCLC)的治疗,但实际疗效数据有限;其他肺神经内分泌癌(PNEC)的免疫疗法证据也很少:本研究旨在评估晚期PNEC患者接受PD-1/PD-L1抑制剂的疗效,并探讨与生存预后相关的因素,为晚期PNEC患者的治疗提供线索:回顾性分析了2019年1月至2021年12月期间接受PD-1/PD-L1抑制剂治疗的203例晚期PNEC患者。构建了无进展生存期(PFS)和总生存期(OS)的卡普兰-梅耶曲线:203例患者的客观反应率(ORR)为48.3%,疾病控制率(DCR)为83.3%,中位PFS(mPFS)为6.0个月,中位OS(mOS)为13.1个月。其中,组织学类型为SCLC的有166例,大细胞神经内分泌癌13例,其他未指定类型的PNEC 24例。从组织学角度看,PFS(P = 0.240)和OS(P = 0.845)无明显差异。在一线(1L)治疗中(N = 125),患者接受化疗免疫疗法,ORR 为 64.8%,DCR 为 92.0%,mPFS 为 6.6 个月,mOS 为 14.9 个月。在二线(2L)或后线治疗中,ORR、DCR、mPFS和mOS分别为21.8%、69.2%、4.4和9.4个月;免疫治疗联合小分子抗血管生成药物的PFS显著高于免疫治疗单药或化学免疫治疗(6.4 vs 1.4 vs 3.7个月,P = 0.041)。没有肝转移的患者的 PFS 更优(7.0 个月 vs 5.1 个月,p p 结论:在临床实践中,无论病理组织学如何,PD-1/PD-L1抑制剂对晚期PNEC患者均有效。1L免疫化疗的疗效值得肯定,在2L或更晚一线治疗中,在免疫治疗的基础上加用小分子抗血管生成药物,可获得更好的生存趋势:设计:回顾性研究。
{"title":"Real-world efficacy of PD-1/PD-L1 inhibitors in patients with advanced pulmonary neuroendocrine carcinoma: a single-center analysis.","authors":"Wanchen Zhai, Ying Yu, Haicheng Wu, Qian Zhang, Yunfei Chen, Yehao Yang, Yun Fan","doi":"10.1177/17588359241288130","DOIUrl":"https://doi.org/10.1177/17588359241288130","url":null,"abstract":"<p><strong>Background: </strong>Immunotherapy blocking programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) has revolutionized the treatment of extensive-stage small-cell lung cancer (SCLC), but only with limited real-world efficacy data; evidence from immunotherapy for other pulmonary neuroendocrine carcinoma (PNEC) is scarce.</p><p><strong>Objective: </strong>The purpose of this study is to evaluate the efficacy of receiving PD-1/PD-L1 inhibitors in patients with advanced PNEC and explore factors related to survival prognosis, providing clues for treatment for patients with advanced PNEC.</p><p><strong>Methods: </strong>In all, 203 patients with advanced PNEC who received PD-1/PD-L1 inhibitors between January 2019 and December 2021 were retrospectively analyzed. Kaplan-Meier curves were constructed for progression-free survival (PFS) and overall survival (OS).</p><p><strong>Results: </strong>For the 203 patients, the objective response rate (ORR) was 48.3%, the disease control rate (DCR) was 83.3%, the median PFS (mPFS) was 6.0 months, and the median OS (mOS) was 13.1 months. Among them, the histology was 166 SCLC, 13 large-cell neuroendocrine carcinoma, and 24 other unspecified PNEC. Histologically, no significant difference was observed in PFS (<i>p</i> = 0.240) or OS (<i>p</i> = 0.845). In first-line (1L) treatment (<i>N</i> = 125), patients received chemoimmunotherapy and had an ORR of 64.8%, DCR of 92.0%, mPFS of 6.6 months, and mOS of 14.9 months. In second-line (2L) or later-line setting, the ORR, DCR, mPFS, and mOS were 21.8%, 69.2%, 4.4, and 9.4 months; immunotherapy plus small-molecule antiangiogenic agents showed significantly greater PFS than immunotherapy monotherapy or chemoimmunotherapy (6.4 vs 1.4 vs 3.7 months, <i>p</i> = 0.041). Patients without liver metastasis had superior PFS (7.0 vs 5.1 months, <i>p</i> < 0.001) and OS (19.2 vs 9.6 months, <i>p</i> < 0.001) than those with liver metastasis.</p><p><strong>Conclusion: </strong>In clinical practice, PD-1/PD-L1 inhibitors are effective in patients with advanced PNEC, regardless of the pathological histology. The efficacy of 1L immunochemotherapy is worthy of recognition, and the addition of small-molecule antiangiogenic agents to immunotherapy in 2L or later-line treatment provides a better survival trend.</p><p><strong>Design: </strong>Retrospective study.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11475206/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475454","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}
引用次数: 0
Prognostic value of immune-inflammatory and nutrition indicators in non-metastatic nasopharyngeal carcinoma with negative plasma Epstein-Barr virus DNA. 血浆 Epstein-Barr 病毒 DNA 阴性的非转移性鼻咽癌的免疫炎症和营养指标的预后价值。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-14 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241286489
Youliang Weng, Lishui Wu, Ying Li, Jing Wang, Zijie Wu, Xinyi Hong, Xiaoyong Liu, Jinghua Lai, Jun Lu, Sufang Qiu

Background: Plasma Epstein-Barr virus (EBV) DNA has been identified as a significant prognostic marker for nasopharyngeal carcinoma (NPC), yet there is limited research on the prognosis of NPC patients with negative EBV DNA.

Objectives: We explore the prognostic value of comprehensive immune-inflammatory and nutritional indicators to offer personalized treatment recommendations and prognosis predictions for non-metastatic NPC patients with negative EBV DNA.

Design: This was a retrospective study.

Methods: This study retrospectively analyzed 257 non-metastatic NPC patients with negative EBV DNA between January 2015 and December 2019. The Kaplan-Meier survival curves evaluated survival endpoints, and group discrepancies were assessed with log-rank tests. Principal component analysis (PCA) reduced data dimensionality. Univariate and multivariate Cox regression analyses identified significant prognostic variables. Risk stratification was performed based on recursive partitioning analysis (RPA). A robust prognostic model was constructed by nomogram and evaluated by calibration curves, decision curves, and the time-dependent area under the curve analysis.

Results: PCA was employed to compute the immune-inflammation index (III) and nutrition index (NI). Multivariate Cox regression analysis revealed lactate dehydrogenase, III, and NI as significant prognostic variables for overall survival (OS). Utilizing RPA, we stratified the risk into three categories: low-risk group (low III + high NI), middle-risk group (low III + low NI), and high-risk group (high III). Both the middle- (p = 0.025) and high-risk groups (p < 0.001) exhibited poorer OS compared with the low-risk group. The nomogram model exhibited superior predictive accuracy compared to tumor lymph node metastasis stage alone (C-index: 0.774 vs 0.679).

Conclusion: Our study validated the prognostic significance of III and NI in non-metastatic NPC patients with negative EBV DNA. Additionally, a clinical risk stratification was constructed to offer valuable insights into the individualized treatment of these patients.

背景:血浆爱泼斯坦-巴尔病毒(EBV)DNA已被确定为鼻咽癌(NPC)的一个重要预后标志物,但对EBV DNA阴性的鼻咽癌患者的预后研究却很有限:我们探讨了免疫炎症和营养综合指标的预后价值,以便为EBV DNA阴性的非转移性鼻咽癌患者提供个性化治疗建议和预后预测:这是一项回顾性研究:本研究回顾性分析了2015年1月至2019年12月期间257例EBV DNA阴性的非转移性鼻咽癌患者。Kaplan-Meier 生存曲线评估了生存终点,并用对数秩检验评估了组间差异。主成分分析(PCA)降低了数据维度。单变量和多变量考克斯回归分析确定了重要的预后变量。根据递归分区分析(RPA)进行了风险分层。通过提名图构建了一个稳健的预后模型,并通过校准曲线、决策曲线和随时间变化的曲线下面积分析进行了评估:结果:采用 PCA 计算了免疫炎症指数(III)和营养指数(NI)。多变量考克斯回归分析显示,乳酸脱氢酶、III和NI是影响总生存期(OS)的重要预后变量。利用 RPA,我们将风险分为三类:低风险组(低 III + 高 NI)、中风险组(低 III + 低 NI)和高风险组(高 III)。中危组(P = 0.025)和高危组(P = 0.025 结论:低危组和高危组的预后均好于中危组和高危组:我们的研究验证了 III 和 NI 在 EBV DNA 阴性的非转移性鼻咽癌患者中的预后意义。此外,我们还构建了临床风险分层,为这些患者的个体化治疗提供了有价值的见解。
{"title":"Prognostic value of immune-inflammatory and nutrition indicators in non-metastatic nasopharyngeal carcinoma with negative plasma Epstein-Barr virus DNA.","authors":"Youliang Weng, Lishui Wu, Ying Li, Jing Wang, Zijie Wu, Xinyi Hong, Xiaoyong Liu, Jinghua Lai, Jun Lu, Sufang Qiu","doi":"10.1177/17588359241286489","DOIUrl":"https://doi.org/10.1177/17588359241286489","url":null,"abstract":"<p><strong>Background: </strong>Plasma Epstein-Barr virus (EBV) DNA has been identified as a significant prognostic marker for nasopharyngeal carcinoma (NPC), yet there is limited research on the prognosis of NPC patients with negative EBV DNA.</p><p><strong>Objectives: </strong>We explore the prognostic value of comprehensive immune-inflammatory and nutritional indicators to offer personalized treatment recommendations and prognosis predictions for non-metastatic NPC patients with negative EBV DNA.</p><p><strong>Design: </strong>This was a retrospective study.</p><p><strong>Methods: </strong>This study retrospectively analyzed 257 non-metastatic NPC patients with negative EBV DNA between January 2015 and December 2019. The Kaplan-Meier survival curves evaluated survival endpoints, and group discrepancies were assessed with log-rank tests. Principal component analysis (PCA) reduced data dimensionality. Univariate and multivariate Cox regression analyses identified significant prognostic variables. Risk stratification was performed based on recursive partitioning analysis (RPA). A robust prognostic model was constructed by nomogram and evaluated by calibration curves, decision curves, and the time-dependent area under the curve analysis.</p><p><strong>Results: </strong>PCA was employed to compute the immune-inflammation index (III) and nutrition index (NI). Multivariate Cox regression analysis revealed lactate dehydrogenase, III, and NI as significant prognostic variables for overall survival (OS). Utilizing RPA, we stratified the risk into three categories: low-risk group (low III + high NI), middle-risk group (low III + low NI), and high-risk group (high III). Both the middle- (<i>p</i> = 0.025) and high-risk groups (<i>p</i> < 0.001) exhibited poorer OS compared with the low-risk group. The nomogram model exhibited superior predictive accuracy compared to tumor lymph node metastasis stage alone (C-index: 0.774 vs 0.679).</p><p><strong>Conclusion: </strong>Our study validated the prognostic significance of III and NI in non-metastatic NPC patients with negative EBV DNA. Additionally, a clinical risk stratification was constructed to offer valuable insights into the individualized treatment of these patients.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11472370/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475453","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}
引用次数: 0
Immune signatures of patients with advanced non-small-cell lung cancer for efficacy prediction after immunotherapy. 用于预测免疫疗法疗效的晚期非小细胞肺癌患者免疫特征。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-09 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241284946
Yung-Hung Luo, Chia-I Shen, Chi-Lu Chiang, Yuh-Min Chen

Background: Programmed cell death protein 1 ligand 1 (PD-L1) expression alone may not be the optimal predictor of immunotherapy (IO) efficacy in advanced non-small cell lung cancer (NSCLC). Evaluation of circulating immune signatures using mass cytometry is a promising technique for predicting IO response and prognosis. The utility of circulating immune signatures for efficacy prediction after IO in advanced NSCLC remains to be elucidated.

Objectives: To assess the feasibility of circulating immune cells and cytokines in predicting tumor response to IO in advanced NSCLC.

Design: A prospective observational study.

Methods: To investigate dynamic changes in immune signatures, blood specimens were prospectively collected from patients with NSCLC at baseline and following chemotherapy (C/T) and/or IO. Mass cytometry and enzyme-linked immunosorbent assay were used to characterize immune signatures and cytokine patterns to identify correlations between immune profiles and treatment efficacy.

Results: The study enrolled 45 patients. The proportion of circulating natural killer (NK) cells and CD8+ T cells significantly increased after IO alone treatment. Cell levels of PD-1+CD8+ T cells, PD-1+CD4+ T cells, TIM-3+CD8+ T cells, LAG-3+ NK cells, and LAG-3+CD8+ T cells significantly decreased in patients with treatment response to IO alone. Tumor necrosis factor-alpha (TNF-α) levels significantly increased after IO alone treatment. Patients with high PD-1+CD8+ T cells before IO alone treatment had lower overall survival (OS) compared to those with low levels. Patients with high LAG-3+CD8+ T cells before chemotherapy plus immunotherapy treatment had lower OS compared to those with low levels.

Conclusion: Responses to IO in NSCLC were correlated with declines in specific exhausted T cells, suggesting that IO may exert therapeutical efficacy by decreasing circulating exhausted T cells, which were associated with poorer survival, while also increasing TNF-α. These results highlight the prognostic value of monitoring changes in circulating exhausted T cells to predict IO response and survival outcomes in advanced lung cancer.

背景:仅凭程序性细胞死亡蛋白 1 配体 1(PD-L1)的表达可能无法预测晚期非小细胞肺癌(NSCLC)的免疫疗法(IO)疗效。使用质控细胞仪评估循环免疫特征是预测 IO 反应和预后的一项很有前景的技术。循环免疫特征在晚期非小细胞肺癌IO后疗效预测中的作用仍有待阐明:评估循环免疫细胞和细胞因子预测晚期NSCLC患者IO后肿瘤反应的可行性:设计:前瞻性观察研究:为了研究免疫特征的动态变化,前瞻性地收集了NSCLC患者在基线和化疗(C/T)和/或IO后的血液标本。采用质控细胞仪和酶联免疫吸附测定法描述免疫特征和细胞因子模式,以确定免疫特征与治疗效果之间的相关性:研究共招募了 45 名患者。单用 IO 治疗后,循环中自然杀伤(NK)细胞和 CD8+ T 细胞的比例显著增加。对单用IO治疗有反应的患者中,PD-1+CD8+ T细胞、PD-1+CD4+ T细胞、TIM-3+CD8+ T细胞、LAG-3+ NK细胞和LAG-3+CD8+ T细胞的细胞水平明显下降。单用 IO 治疗后,肿瘤坏死因子-α(TNF-α)水平明显升高。与PD-1+CD8+ T细胞水平低的患者相比,单用IO治疗前PD-1+CD8+ T细胞水平高的患者总生存期(OS)较低。化疗加免疫疗法治疗前LAG-3+CD8+ T细胞水平高的患者的总生存期低于LAG-3+CD8+ T细胞水平低的患者:结论:NSCLC患者对IO的反应与特异性衰竭T细胞的减少有关,这表明IO可通过减少循环中与生存率降低有关的衰竭T细胞来发挥疗效,同时还可增加TNF-α。这些结果凸显了监测循环衰竭T细胞的变化以预测晚期肺癌的IO反应和生存结果的预后价值。
{"title":"Immune signatures of patients with advanced non-small-cell lung cancer for efficacy prediction after immunotherapy.","authors":"Yung-Hung Luo, Chia-I Shen, Chi-Lu Chiang, Yuh-Min Chen","doi":"10.1177/17588359241284946","DOIUrl":"10.1177/17588359241284946","url":null,"abstract":"<p><strong>Background: </strong>Programmed cell death protein 1 ligand 1 (PD-L1) expression alone may not be the optimal predictor of immunotherapy (IO) efficacy in advanced non-small cell lung cancer (NSCLC). Evaluation of circulating immune signatures using mass cytometry is a promising technique for predicting IO response and prognosis. The utility of circulating immune signatures for efficacy prediction after IO in advanced NSCLC remains to be elucidated.</p><p><strong>Objectives: </strong>To assess the feasibility of circulating immune cells and cytokines in predicting tumor response to IO in advanced NSCLC.</p><p><strong>Design: </strong>A prospective observational study.</p><p><strong>Methods: </strong>To investigate dynamic changes in immune signatures, blood specimens were prospectively collected from patients with NSCLC at baseline and following chemotherapy (C/T) and/or IO. Mass cytometry and enzyme-linked immunosorbent assay were used to characterize immune signatures and cytokine patterns to identify correlations between immune profiles and treatment efficacy.</p><p><strong>Results: </strong>The study enrolled 45 patients. The proportion of circulating natural killer (NK) cells and CD8<sup>+</sup> T cells significantly increased after IO alone treatment. Cell levels of PD-1<sup>+</sup>CD8<sup>+</sup> T cells, PD-1<sup>+</sup>CD4<sup>+</sup> T cells, TIM-3<sup>+</sup>CD8<sup>+</sup> T cells, LAG-3<sup>+</sup> NK cells, and LAG-3<sup>+</sup>CD8<sup>+</sup> T cells significantly decreased in patients with treatment response to IO alone. Tumor necrosis factor-alpha (TNF-α) levels significantly increased after IO alone treatment. Patients with high PD-1<sup>+</sup>CD8<sup>+</sup> T cells before IO alone treatment had lower overall survival (OS) compared to those with low levels. Patients with high LAG-3<sup>+</sup>CD8<sup>+</sup> T cells before chemotherapy plus immunotherapy treatment had lower OS compared to those with low levels.</p><p><strong>Conclusion: </strong>Responses to IO in NSCLC were correlated with declines in specific exhausted T cells, suggesting that IO may exert therapeutical efficacy by decreasing circulating exhausted T cells, which were associated with poorer survival, while also increasing TNF-α. These results highlight the prognostic value of monitoring changes in circulating exhausted T cells to predict IO response and survival outcomes in advanced lung cancer.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11465298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142401366","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}
引用次数: 0
Circulating tumor cells in pancreatic cancer: more than liquid biopsy. 胰腺癌中的循环肿瘤细胞:不仅仅是液体活检。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-09 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241284935
Zeru Li, Cheng Qin, Bangbo Zhao, Tianyu Li, Yutong Zhao, Xiangyu Zhang, Weibin Wang

Circulating tumor cells (CTCs) are tumor cells that slough off the primary lesions and extravasate into the bloodstream. By forming CTC clusters and interacting with other circulating cells (platelets, NK cells, macrophage, etc.), CTCs are able to survive in the circulatory system of tumor patients and colonize to metastatic organs. In recent years, the potential of CTCs in diagnosis, prognostic assessment, and individualized therapy of various types of tumors has been gradually explored, while advances in biotechnology have made it possible to extract CTCs from patient blood samples. These biological features of CTCs provide us with new insights into cancer vulnerabilities. With the advent of new immunotherapies and personalized medicines, disrupting the heterotypical interaction between CTCs and circulatory cells as well as direct CTCs targeting hold great promise. Pancreatic cancer (PC) is one of the most malignant cancers, in part because of early metastasis, difficult diagnosis, and limited treatment options. Although there is significant potential for CTCs as a biomarker to impact PC from diagnosis to therapy, there still remain a number of challenges to the routine implementation of CTCs in the clinical management of PC. In this review, we summed up the progress made in understanding biological characteristics and exceptional technological advances of CTCs and provided insight into exploiting these developments to design future clinical tools for improving the diagnosis and treatment of PC.

循环肿瘤细胞(CTC)是指从原发病灶脱落并外渗到血液中的肿瘤细胞。通过形成 CTC 簇并与其他循环细胞(血小板、NK 细胞、巨噬细胞等)相互作用,CTCs 能够在肿瘤患者的循环系统中存活并定植到转移器官。近年来,CTCs 在各类肿瘤的诊断、预后评估和个体化治疗方面的潜力逐渐被发掘出来,而生物技术的进步也使得从患者血液样本中提取 CTCs 成为可能。CTCs 的这些生物学特征为我们了解癌症的弱点提供了新的视角。随着新型免疫疗法和个性化药物的出现,破坏 CTC 与循环细胞之间的异型相互作用以及直接靶向 CTC 都大有可为。胰腺癌(PC)是恶性程度最高的癌症之一,部分原因是转移早、诊断难、治疗方案有限。虽然 CTCs 作为一种生物标记物对 PC 从诊断到治疗的影响潜力巨大,但在 PC 的临床管理中常规应用 CTCs 仍面临诸多挑战。在这篇综述中,我们总结了在了解 CTCs 的生物学特性和特殊技术进步方面取得的进展,并深入探讨了如何利用这些进展设计未来的临床工具,以改善 PC 的诊断和治疗。
{"title":"Circulating tumor cells in pancreatic cancer: more than liquid biopsy.","authors":"Zeru Li, Cheng Qin, Bangbo Zhao, Tianyu Li, Yutong Zhao, Xiangyu Zhang, Weibin Wang","doi":"10.1177/17588359241284935","DOIUrl":"https://doi.org/10.1177/17588359241284935","url":null,"abstract":"<p><p>Circulating tumor cells (CTCs) are tumor cells that slough off the primary lesions and extravasate into the bloodstream. By forming CTC clusters and interacting with other circulating cells (platelets, NK cells, macrophage, etc.), CTCs are able to survive in the circulatory system of tumor patients and colonize to metastatic organs. In recent years, the potential of CTCs in diagnosis, prognostic assessment, and individualized therapy of various types of tumors has been gradually explored, while advances in biotechnology have made it possible to extract CTCs from patient blood samples. These biological features of CTCs provide us with new insights into cancer vulnerabilities. With the advent of new immunotherapies and personalized medicines, disrupting the heterotypical interaction between CTCs and circulatory cells as well as direct CTCs targeting hold great promise. Pancreatic cancer (PC) is one of the most malignant cancers, in part because of early metastasis, difficult diagnosis, and limited treatment options. Although there is significant potential for CTCs as a biomarker to impact PC from diagnosis to therapy, there still remain a number of challenges to the routine implementation of CTCs in the clinical management of PC. In this review, we summed up the progress made in understanding biological characteristics and exceptional technological advances of CTCs and provided insight into exploiting these developments to design future clinical tools for improving the diagnosis and treatment of PC.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11483845/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475448","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}
引用次数: 0
Tyrosine kinase inhibitors in patients with neuroendocrine neoplasms: a systematic literature review. 神经内分泌肿瘤患者使用酪氨酸激酶抑制剂:系统性文献综述。
IF 4.3 2区 医学 Q2 ONCOLOGY Pub Date : 2024-10-09 eCollection Date: 2024-01-01 DOI: 10.1177/17588359241286751
Rodrigo G Taboada, Felicia P Cavalher, Juliana F Rego, Rachel P Riechelmann

Background: Several tyrosine kinase receptors inhibitors (TKIs) have demonstrated antiproliferative effects in well-differentiated neuroendocrine tumors (NETs). We aimed to summarize and appraise the current evidence of the efficacy of TKIs in patients with different types of NETs.

Methods: We performed a systematic review of clinical trials of TKIs in patients with advanced gastroenteropancreatic or lung NETs (PROSPERO registration number: CRD42024507379). Population characteristics, efficacy, and safety results were summarized by type of NET.

Results: Twenty-eight studies were eligible, totaling 2284 patients. While sunitinib remains the only Food and Drug Administration-approved TKI in patients with NETs (for patients with pancreatic well-differentiated NETs), recent placebo-controlled randomized trials have demonstrated improved response rates and progression-free survival for patients with progressive and pre-treated well-differentiated pancreatic (cabozantinib or surufatinib) or gastrointestinal (GI) NETs (pazopanib, cabozantinib, or surufatinib). There is limited evidence to support the use of a TKI in patients with lung or grade 3 NETs. The toxicity associated with TKIs follows a class effect, with a significant proportion of patients experiencing fatigue, hypertension, and hand-foot skin reactions.

Conclusion: TKIs are effective therapies in patients with pancreatic or GI well-differentiated NETs and should be part of the therapeutical sequencing of these patients.

背景:几种酪氨酸激酶受体抑制剂(TKIs)对分化良好的神经内分泌肿瘤(NETs)具有抗增殖作用。我们旨在总结和评估目前TKIs在不同类型NET患者中的疗效证据:我们对TKIs治疗晚期胃肠胰或肺NET患者的临床试验进行了系统性回顾(PROSPERO注册号:CRD42024507379)。按NET类型总结了人群特征、疗效和安全性结果:共有 28 项研究符合条件,共计 2284 名患者。尽管舒尼替尼仍是唯一获得美国食品药品管理局批准的治疗NET患者的TKI(用于胰腺分化良好的NET患者),但最近的安慰剂对照随机试验显示,进展期和预处理良好的胰腺(卡博赞替尼或素鲁法替尼)或胃肠道(GI)NET(帕唑帕尼、卡博赞替尼或素鲁法替尼)患者的应答率和无进展生存期均有所改善。支持肺癌或 3 级 NET 患者使用 TKI 的证据有限。与TKIs相关的毒性具有同类效应,相当一部分患者会出现疲劳、高血压和手足皮肤反应:TKIs对胰腺或消化道分化良好的NET患者是有效的疗法,应成为这些患者治疗顺序的一部分。
{"title":"Tyrosine kinase inhibitors in patients with neuroendocrine neoplasms: a systematic literature review.","authors":"Rodrigo G Taboada, Felicia P Cavalher, Juliana F Rego, Rachel P Riechelmann","doi":"10.1177/17588359241286751","DOIUrl":"https://doi.org/10.1177/17588359241286751","url":null,"abstract":"<p><strong>Background: </strong>Several tyrosine kinase receptors inhibitors (TKIs) have demonstrated antiproliferative effects in well-differentiated neuroendocrine tumors (NETs). We aimed to summarize and appraise the current evidence of the efficacy of TKIs in patients with different types of NETs.</p><p><strong>Methods: </strong>We performed a systematic review of clinical trials of TKIs in patients with advanced gastroenteropancreatic or lung NETs (PROSPERO registration number: CRD42024507379). Population characteristics, efficacy, and safety results were summarized by type of NET.</p><p><strong>Results: </strong>Twenty-eight studies were eligible, totaling 2284 patients. While sunitinib remains the only Food and Drug Administration-approved TKI in patients with NETs (for patients with pancreatic well-differentiated NETs), recent placebo-controlled randomized trials have demonstrated improved response rates and progression-free survival for patients with progressive and pre-treated well-differentiated pancreatic (cabozantinib or surufatinib) or gastrointestinal (GI) NETs (pazopanib, cabozantinib, or surufatinib). There is limited evidence to support the use of a TKI in patients with lung or grade 3 NETs. The toxicity associated with TKIs follows a class effect, with a significant proportion of patients experiencing fatigue, hypertension, and hand-foot skin reactions.</p><p><strong>Conclusion: </strong>TKIs are effective therapies in patients with pancreatic or GI well-differentiated NETs and should be part of the therapeutical sequencing of these patients.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":null,"pages":null},"PeriodicalIF":4.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11483706/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142485923","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}
引用次数: 0
期刊
Therapeutic Advances in Medical Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1