Pub Date : 2024-10-04DOI: 10.1007/s12094-024-03652-9
Lucia Osorio, Tatiana P Grazioso, Guillermo de Velasco, Olatz Etxaniz, Jose Luis Pérez-Gracia, Álvaro Pinto, Ignacio Durán, Enrique Grande, Pablo Borrega Garcia, Martín Lázaro, Laura Rodriguez, Maria Laura Villalobos, Lourdes García, Andrés Cuellar, María Pilar Solís-Hernández, Cristina Pernaut, Juan Francisco Rodríguez-Moreno, Cristina Rodriguez-Antona, Jesús García-Donas
Background and purpose: Despite that incorporating antiangiogenic in combination with immune-checkpoint inhibitors as the standard first-line treatment for advanced clear cell renal cell cancer (ccRCC) yields promising outcomes, these regimens often lead to significant toxicity. However, a subgroup of patients has shown responsiveness to VEGFR tyrosine-kinase inhibitors (TKIs) in monotherapy, leading to the question of whether employing combination therapies can significantly enhance overall survival in all patients over monotherapy. Thus, we aim to identify gene expression signatures that can predict TKI response within subpopulations that might benefit from single-agent therapies, to minimize unnecessary exposure to combination therapies and their associated toxicities, as well as to discover new potential therapeutic targets to improve ccRCC treatment. Based on prior data, the androgen receptor (AR) might meet both conditions.
Patients and methods: We evaluated the association between AR expression, assessed through NanoString® technology-derived mRNA counts, and the clinical outcomes of 98 ccRCC patients treated with first-line antiangiogenics and determined its association with other genes implicated in ccRCC tumorigenesis.
Results: Higher AR-expression correlates significantly with better prognosis and survival based on the MSKCC risk score, and longer PFS. Furthermore, we have identified a gene set signature associated with AR-overexpression and several genes involved in angiogenesis and transcriptional targets of the hypoxia-inducible factor, a cornerstone of ccRCC.
Conclusions: AR-overexpression and its association with other genes could favor a transcriptomic signature set to aid in identifying patients suitable for TKI in monotherapy, rather than aggressive combinations, enhancing thus, precision and personalized therapeutic decisions.
{"title":"Retrospective study assessing the role of the androgen receptor in clear cell renal cell cancer patients treated with VEGFR inhibitors in monotherapy.","authors":"Lucia Osorio, Tatiana P Grazioso, Guillermo de Velasco, Olatz Etxaniz, Jose Luis Pérez-Gracia, Álvaro Pinto, Ignacio Durán, Enrique Grande, Pablo Borrega Garcia, Martín Lázaro, Laura Rodriguez, Maria Laura Villalobos, Lourdes García, Andrés Cuellar, María Pilar Solís-Hernández, Cristina Pernaut, Juan Francisco Rodríguez-Moreno, Cristina Rodriguez-Antona, Jesús García-Donas","doi":"10.1007/s12094-024-03652-9","DOIUrl":"https://doi.org/10.1007/s12094-024-03652-9","url":null,"abstract":"<p><strong>Background and purpose: </strong>Despite that incorporating antiangiogenic in combination with immune-checkpoint inhibitors as the standard first-line treatment for advanced clear cell renal cell cancer (ccRCC) yields promising outcomes, these regimens often lead to significant toxicity. However, a subgroup of patients has shown responsiveness to VEGFR tyrosine-kinase inhibitors (TKIs) in monotherapy, leading to the question of whether employing combination therapies can significantly enhance overall survival in all patients over monotherapy. Thus, we aim to identify gene expression signatures that can predict TKI response within subpopulations that might benefit from single-agent therapies, to minimize unnecessary exposure to combination therapies and their associated toxicities, as well as to discover new potential therapeutic targets to improve ccRCC treatment. Based on prior data, the androgen receptor (AR) might meet both conditions.</p><p><strong>Patients and methods: </strong>We evaluated the association between AR expression, assessed through NanoString<sup>®</sup> technology-derived mRNA counts, and the clinical outcomes of 98 ccRCC patients treated with first-line antiangiogenics and determined its association with other genes implicated in ccRCC tumorigenesis.</p><p><strong>Results: </strong>Higher AR-expression correlates significantly with better prognosis and survival based on the MSKCC risk score, and longer PFS. Furthermore, we have identified a gene set signature associated with AR-overexpression and several genes involved in angiogenesis and transcriptional targets of the hypoxia-inducible factor, a cornerstone of ccRCC.</p><p><strong>Conclusions: </strong>AR-overexpression and its association with other genes could favor a transcriptomic signature set to aid in identifying patients suitable for TKI in monotherapy, rather than aggressive combinations, enhancing thus, precision and personalized therapeutic decisions.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373469","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-04DOI: 10.1007/s12094-024-03734-8
Javier Puente, Alvaro Pinto, Maria José Mendez-Vidal, Xavier García Del Muro, Pablo Maroto, Sergio Vazquez, Raquel Luque-Caro, Urbano Anido, Torsten Strunz-McKendry, Anil Upadhyay, Jose Montes, Aurora Ortiz Nuñez, Judit González Portela, Daniel Castellano
Purpose: Real-world evidence on locally advanced or metastatic urothelial carcinoma (la/mUC) management in Spain is limited. This study describes patient characteristics, treatment patterns, survival, and health care resource utilization (HCRU) in this population.
Methods/patients: This retrospective observational study included all adults with a first diagnosis/record of la/mUC (index date) from January 2015 to June 2020 at nine university hospitals in Spain. Data were collected up to December 31, 2020 (end of study), death, or loss to follow-up. Patient characteristics, treatment patterns, median overall survival (OS) and progression-free survival (PFS) from index date (Kaplan-Meier estimates), and disease-specific HCRU were described.
Results: Among 829 patients, median age at diagnosis was 71 years; 70.2% had ≥ 1 comorbidity, and 52.5% were eligible for cisplatin. Median follow-up was 12.7 months. Most (84.7%) patients received first-line systemic treatment; of these, 46.9% (n = 329) received second-line and 16.6% (n = 116) received third-line therapy. Chemotherapy was the most common treatment in all lines of therapy, followed by programmed cell death protein 1/ligand 1 inhibitors. Median (95% confidence interval) OS and PFS were 18.8 (17.5-21.5) and 9.9 (8.9-10.5) months, respectively. Most patients required ≥ 1 outpatient visit (71.8%), inpatient admission (56.6%), or emergency department visit (56.5%).
Conclusions: Therapeutic patterns were consistent with Spanish guideline recommendations. Chemotherapy had a role in first-line treatment of la/mUC in Spain during the study period. However, the disease burden remains high, and new first-line treatments recommended in the latest European guidelines should be made available to patients in Spain.
{"title":"Real-world treatment patterns, survival outcomes, and health care resource utilization for locally advanced or metastatic urothelial carcinoma in Spain.","authors":"Javier Puente, Alvaro Pinto, Maria José Mendez-Vidal, Xavier García Del Muro, Pablo Maroto, Sergio Vazquez, Raquel Luque-Caro, Urbano Anido, Torsten Strunz-McKendry, Anil Upadhyay, Jose Montes, Aurora Ortiz Nuñez, Judit González Portela, Daniel Castellano","doi":"10.1007/s12094-024-03734-8","DOIUrl":"https://doi.org/10.1007/s12094-024-03734-8","url":null,"abstract":"<p><strong>Purpose: </strong>Real-world evidence on locally advanced or metastatic urothelial carcinoma (la/mUC) management in Spain is limited. This study describes patient characteristics, treatment patterns, survival, and health care resource utilization (HCRU) in this population.</p><p><strong>Methods/patients: </strong>This retrospective observational study included all adults with a first diagnosis/record of la/mUC (index date) from January 2015 to June 2020 at nine university hospitals in Spain. Data were collected up to December 31, 2020 (end of study), death, or loss to follow-up. Patient characteristics, treatment patterns, median overall survival (OS) and progression-free survival (PFS) from index date (Kaplan-Meier estimates), and disease-specific HCRU were described.</p><p><strong>Results: </strong>Among 829 patients, median age at diagnosis was 71 years; 70.2% had ≥ 1 comorbidity, and 52.5% were eligible for cisplatin. Median follow-up was 12.7 months. Most (84.7%) patients received first-line systemic treatment; of these, 46.9% (n = 329) received second-line and 16.6% (n = 116) received third-line therapy. Chemotherapy was the most common treatment in all lines of therapy, followed by programmed cell death protein 1/ligand 1 inhibitors. Median (95% confidence interval) OS and PFS were 18.8 (17.5-21.5) and 9.9 (8.9-10.5) months, respectively. Most patients required ≥ 1 outpatient visit (71.8%), inpatient admission (56.6%), or emergency department visit (56.5%).</p><p><strong>Conclusions: </strong>Therapeutic patterns were consistent with Spanish guideline recommendations. Chemotherapy had a role in first-line treatment of la/mUC in Spain during the study period. However, the disease burden remains high, and new first-line treatments recommended in the latest European guidelines should be made available to patients in Spain.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373468","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-05-14DOI: 10.1007/s12094-024-03508-2
Versha Dahiya, Sanjana Hans, Ruchi Kumari, Gargi Bagchi
Prostate cancer (PCa) is the second most prevalent cancer in men. In 2020, approximately 1,414,259 new cases were reported that accounted for 3,75,324 deaths (Sung et al. in CA 71:209-249, 2021). PCa is often asymptomatic at early stages; hence, routine screening and monitoring based on reliable biomarkers is crucial for early detection and assessment of cancer progression. Early diagnosis of disease is key step in reducing PCa-induced mortality. Biomarkers such as PSA have played vital role in reducing recent PCa deaths. Recent research has identified many other biomarkers and also refined PSA-based tests for non-invasive diagnosis of PCa in patients. Despite progress in screening methods, an important issue that influences treatment is heterogeneity of the cancer in different individuals, necessitating personalized treatment. Currently, focus is to identify biomarkers that can accurately diagnose PCa at early stage, indicate the stage of the disease, metastatic nature and chances of survival based on individual patient profile (Fig. 1). Fig. 1 Graphical abstract.
{"title":"Prostate cancer biomarkers: from early diagnosis to precision treatment.","authors":"Versha Dahiya, Sanjana Hans, Ruchi Kumari, Gargi Bagchi","doi":"10.1007/s12094-024-03508-2","DOIUrl":"10.1007/s12094-024-03508-2","url":null,"abstract":"<p><p>Prostate cancer (PCa) is the second most prevalent cancer in men. In 2020, approximately 1,414,259 new cases were reported that accounted for 3,75,324 deaths (Sung et al. in CA 71:209-249, 2021). PCa is often asymptomatic at early stages; hence, routine screening and monitoring based on reliable biomarkers is crucial for early detection and assessment of cancer progression. Early diagnosis of disease is key step in reducing PCa-induced mortality. Biomarkers such as PSA have played vital role in reducing recent PCa deaths. Recent research has identified many other biomarkers and also refined PSA-based tests for non-invasive diagnosis of PCa in patients. Despite progress in screening methods, an important issue that influences treatment is heterogeneity of the cancer in different individuals, necessitating personalized treatment. Currently, focus is to identify biomarkers that can accurately diagnose PCa at early stage, indicate the stage of the disease, metastatic nature and chances of survival based on individual patient profile (Fig. 1). Fig. 1 Graphical abstract.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140923891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-04-20DOI: 10.1007/s12094-024-03477-6
Zheng Wu, Teng Pan, Wen Li, Yue-Hua Zhang, Sheng-Hu Guo, Ya Liu, Lei Zhang, Zhi-Yu Wang
Background: Transmembrane protein 92 (TMEM92) has been implicated in the facilitation of tumor progression. Nevertheless, comprehensive analyses concerning the prognostic significance of TMEM92, as well as its role in immunological responses across diverse cancer types, remain to be elucidated.
Methods: In this study, data was sourced from a range of publicly accessible online platforms and databases, including TCGA, GTEx, UCSC Xena, CCLE, cBioPortal, HPA, TIMER2.0, GEPIA, CancerSEA, GDSC, exoRBase, and ImmuCellAI. We systematically analyzed the expression patterns of TMEM92 at both mRNA and protein levels across diverse human organs, tissues, extracellular vesicles (EVs), and cell lines associated with multiple cancer types. Subsequently, analyses were conducted to determine the relationship between TMEM92 and various parameters such as prognosis, DNA methylation, copy number variation (CNV), the tumor microenvironment (TME), immune cell infiltration, genes with immunological relevance, tumor mutational burden (TMB), microsatellite instability (MSI), mismatch repair (MMR), and half-maximal inhibitory concentration (IC50) values.
Results: In the present study, we observed a pronounced overexpression of TMEM92 across a majority of cancer types, which was concomitantly associated with a less favorable prognosis. A notable association emerged between TMEM92 expression and both DNA methylation and CNV. Furthermore, a pronounced relationship was discerned between TMEM92 expression, the TME, and the degree of immune cell infiltration. Intriguingly, while TMEM92 expression displayed a positive correlation with macrophage presence, it inversely correlated with the infiltration level of CD8 + T cells. Concurrently, significant associations were identified between TMEM92 and the major histocompatibility complex, TMB, MSI, and MMR. Results derived from Gene Set Enrichment Analysis and Gene Set Variation Analysis further substantiated the nexus of TMEM92 with both immune and metabolic pathways within the oncogenic context.
Conclusions: These findings expanded the understanding of the roles of TMEM92 in tumorigenesis and progression and suggest that TMEM92 may have an immunoregulatory role in several malignancies.
背景:跨膜蛋白92(TMEM92)与肿瘤进展的促进作用有关。然而,有关 TMEM92 的预后意义及其在不同癌症类型的免疫反应中的作用的全面分析仍有待阐明:本研究的数据来源于一系列可公开访问的在线平台和数据库,包括TCGA、GTEx、UCSC Xena、CCLE、cBioPortal、HPA、TIMER2.0、GEPIA、CancerSEA、GDSC、exoRBase和ImmuCellAI。我们系统分析了 TMEM92 在与多种癌症类型相关的不同人体器官、组织、细胞外囊泡 (EV) 和细胞系中的 mRNA 和蛋白质水平的表达模式。随后,我们分析了TMEM92与预后、DNA甲基化、拷贝数变异(CNV)、肿瘤微环境(TME)、免疫细胞浸润、免疫相关基因、肿瘤突变负荷(TMB)、微卫星不稳定性(MSI)、错配修复(MMR)和半数最大抑制浓度(IC50)值等各种参数之间的关系:在本研究中,我们观察到 TMEM92 在大多数癌症类型中明显过表达,这与预后较差有关。TMEM92 的表达与 DNA 甲基化和 CNV 之间存在明显的关联。此外,TMEM92 的表达、TME 和免疫细胞浸润程度之间也有明显的关系。耐人寻味的是,虽然 TMEM92 的表达与巨噬细胞的存在呈正相关,但却与 CD8 + T 细胞的浸润程度成反比。同时,还发现 TMEM92 与主要组织相容性复合体、TMB、MSI 和 MMR 之间存在重要关联。基因组富集分析(Gene Set Enrichment Analysis)和基因组变异分析(Gene Set Variation Analysis)得出的结果进一步证实了TMEM92与致癌背景下的免疫和代谢途径之间的联系:这些发现拓展了人们对 TMEM92 在肿瘤发生和发展过程中的作用的认识,并表明 TMEM92 在多种恶性肿瘤中可能具有免疫调节作用。
{"title":"Comprehensive pan-cancer analysis reveals prognostic implications of TMEM92 in the tumor immune microenvironment.","authors":"Zheng Wu, Teng Pan, Wen Li, Yue-Hua Zhang, Sheng-Hu Guo, Ya Liu, Lei Zhang, Zhi-Yu Wang","doi":"10.1007/s12094-024-03477-6","DOIUrl":"10.1007/s12094-024-03477-6","url":null,"abstract":"<p><strong>Background: </strong>Transmembrane protein 92 (TMEM92) has been implicated in the facilitation of tumor progression. Nevertheless, comprehensive analyses concerning the prognostic significance of TMEM92, as well as its role in immunological responses across diverse cancer types, remain to be elucidated.</p><p><strong>Methods: </strong>In this study, data was sourced from a range of publicly accessible online platforms and databases, including TCGA, GTEx, UCSC Xena, CCLE, cBioPortal, HPA, TIMER2.0, GEPIA, CancerSEA, GDSC, exoRBase, and ImmuCellAI. We systematically analyzed the expression patterns of TMEM92 at both mRNA and protein levels across diverse human organs, tissues, extracellular vesicles (EVs), and cell lines associated with multiple cancer types. Subsequently, analyses were conducted to determine the relationship between TMEM92 and various parameters such as prognosis, DNA methylation, copy number variation (CNV), the tumor microenvironment (TME), immune cell infiltration, genes with immunological relevance, tumor mutational burden (TMB), microsatellite instability (MSI), mismatch repair (MMR), and half-maximal inhibitory concentration (IC50) values.</p><p><strong>Results: </strong>In the present study, we observed a pronounced overexpression of TMEM92 across a majority of cancer types, which was concomitantly associated with a less favorable prognosis. A notable association emerged between TMEM92 expression and both DNA methylation and CNV. Furthermore, a pronounced relationship was discerned between TMEM92 expression, the TME, and the degree of immune cell infiltration. Intriguingly, while TMEM92 expression displayed a positive correlation with macrophage presence, it inversely correlated with the infiltration level of CD8 + T cells. Concurrently, significant associations were identified between TMEM92 and the major histocompatibility complex, TMB, MSI, and MMR. Results derived from Gene Set Enrichment Analysis and Gene Set Variation Analysis further substantiated the nexus of TMEM92 with both immune and metabolic pathways within the oncogenic context.</p><p><strong>Conclusions: </strong>These findings expanded the understanding of the roles of TMEM92 in tumorigenesis and progression and suggest that TMEM92 may have an immunoregulatory role in several malignancies.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140874946","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-05-16DOI: 10.1007/s12094-024-03501-9
Maria Gonzalez-Cao, Teresa Puertolas, Jose Luis Manzano, Cayetana Maldonado, Oriol Yelamos, Miguel Ángel Berciano-Guerrero, Pablo Cerezuela, Juan Martin-Liberal, Eva Muñoz-Couselo, Enrique Espinosa, Ana Drozdowskyj, Alfonso Berrocal, Ainara Soria, Ivan Marquez-Rodas, Salvador Martin-Algarra, Maria Quindos, Susana Puig
Background: The development of highly active drugs has improved the survival of melanoma patients, but elevated drug prices place a significant burden on health care systems. In Spain, the public health care system is transferred to the 17 autonomous communities (AACC). The objective of this study is to describe the situation of drug access for melanoma patients in Spain and how this decentralized system is affecting equity.
Methods: From July to September 2023, a cross-sectional survey was sent to members of the Spanish Multidisciplinary Melanoma Group (GEM Group). The questionnaire consulted about the real access to new drugs in each hospital. The responses were collected anonymously and analyzed according to several variables, including the AACC.
Results: The survey was answered by 50 physicians in 15 AACC. No major differences on access between AACC were observed for indications that are reimbursed by the Spanish Health Care System (adjuvant immunotherapy for stage IIIC-IIID and resected stage IV melanoma). Important differences in drug access were observed among AACC and among centers within the same AACC, for most of the EMA indications that are not reimbursed (adjuvant immunotherapy for stages IIB-IIC-IIIA-IIIB) or that are not fully reimbursed (ipilimumab plus nivolumab in advanced stage). Homogeneously, access to adjuvant targeted drugs, TIL therapy and T-VEC, is extremely low or non-existing in all AACC.
Conclusions: For most indications that reimbursement is restricted out of the EMA indication, a great diversity on access was found throughout the different hospitals in Spain, including heterogeneity intra-AACC.
{"title":"Access to melanoma drugs in Spain: a cross-sectional survey.","authors":"Maria Gonzalez-Cao, Teresa Puertolas, Jose Luis Manzano, Cayetana Maldonado, Oriol Yelamos, Miguel Ángel Berciano-Guerrero, Pablo Cerezuela, Juan Martin-Liberal, Eva Muñoz-Couselo, Enrique Espinosa, Ana Drozdowskyj, Alfonso Berrocal, Ainara Soria, Ivan Marquez-Rodas, Salvador Martin-Algarra, Maria Quindos, Susana Puig","doi":"10.1007/s12094-024-03501-9","DOIUrl":"10.1007/s12094-024-03501-9","url":null,"abstract":"<p><strong>Background: </strong>The development of highly active drugs has improved the survival of melanoma patients, but elevated drug prices place a significant burden on health care systems. In Spain, the public health care system is transferred to the 17 autonomous communities (AACC). The objective of this study is to describe the situation of drug access for melanoma patients in Spain and how this decentralized system is affecting equity.</p><p><strong>Methods: </strong>From July to September 2023, a cross-sectional survey was sent to members of the Spanish Multidisciplinary Melanoma Group (GEM Group). The questionnaire consulted about the real access to new drugs in each hospital. The responses were collected anonymously and analyzed according to several variables, including the AACC.</p><p><strong>Results: </strong>The survey was answered by 50 physicians in 15 AACC. No major differences on access between AACC were observed for indications that are reimbursed by the Spanish Health Care System (adjuvant immunotherapy for stage IIIC-IIID and resected stage IV melanoma). Important differences in drug access were observed among AACC and among centers within the same AACC, for most of the EMA indications that are not reimbursed (adjuvant immunotherapy for stages IIB-IIC-IIIA-IIIB) or that are not fully reimbursed (ipilimumab plus nivolumab in advanced stage). Homogeneously, access to adjuvant targeted drugs, TIL therapy and T-VEC, is extremely low or non-existing in all AACC.</p><p><strong>Conclusions: </strong>For most indications that reimbursement is restricted out of the EMA indication, a great diversity on access was found throughout the different hospitals in Spain, including heterogeneity intra-AACC.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140946353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-05-17DOI: 10.1007/s12094-024-03516-2
Dong-Feng Ge, Yang-Kun Wang, Ying-Ying Li, Xing-Hai Liao, Chao-Ya Zhu, Bo Jiang, Su-Nan Wang
Objective: The objective of this study is to assess the clinical pathological attributes of Hepatoid Adenocarcinoma of the Stomach (HAS) and to delineate the differential diagnostic considerations about it.
Method: The investigation involved analyzing 31 HAS cases using histomorphological assessment, immunohistochemical profiling, and relevant gene detection methodologies.
Results: Among the 31 HAS cases, 9 (29.0%) were of trabecular hepatoid adenocarcinoma of the stomach, 7 (22.6%) were of glandular hepatoid adenocarcinoma of the stomach, 4 (12.9%) were of nesting hepatoid adenocarcinoma of the stomach, 3 (9.7%) were of clear cell hepatoid adenocarcinoma of the stomach, and 8 (25.8%) were of diverse hepatoid adenocarcinoma of the stomach. Of these 31 cases, 24 were male, accounting for 77.4% of the cases. Serum alpha-fetoprotein (AFP) levels were notably elevated, with radioimmunoassay results reaching 1240 ng/ml; 28 out of 31 cases had AFP levels below 25 µg/l, accounting for 90.3%. Related genes: HER2 protein indicated positive expression on the cell membrane in 35.5% (11/31) of the cases; HER2 gene amplification detected by the FISH technique was 12.9% (4/31). Tumoral stromal lymphocytes exhibited a PD-1 positive expression rate of 58.1% (18/31). In gastric cancer tissues, the PD-L1 positive rate was 45.1% (14/31).
Conclusion: HAS represents a distinctive subtype of gastric cancer with a propensity for mimicking other forms of tumors, underscoring the significance of discerning its unique histopathological attributes for accurate differential diagnosis and tailored therapeutic interventions.
{"title":"Assessing clinical pathological characteristics and gene expression patterns associated with hepatoid adenocarcinoma of the stomach.","authors":"Dong-Feng Ge, Yang-Kun Wang, Ying-Ying Li, Xing-Hai Liao, Chao-Ya Zhu, Bo Jiang, Su-Nan Wang","doi":"10.1007/s12094-024-03516-2","DOIUrl":"10.1007/s12094-024-03516-2","url":null,"abstract":"<p><strong>Objective: </strong>The objective of this study is to assess the clinical pathological attributes of Hepatoid Adenocarcinoma of the Stomach (HAS) and to delineate the differential diagnostic considerations about it.</p><p><strong>Method: </strong>The investigation involved analyzing 31 HAS cases using histomorphological assessment, immunohistochemical profiling, and relevant gene detection methodologies.</p><p><strong>Results: </strong>Among the 31 HAS cases, 9 (29.0%) were of trabecular hepatoid adenocarcinoma of the stomach, 7 (22.6%) were of glandular hepatoid adenocarcinoma of the stomach, 4 (12.9%) were of nesting hepatoid adenocarcinoma of the stomach, 3 (9.7%) were of clear cell hepatoid adenocarcinoma of the stomach, and 8 (25.8%) were of diverse hepatoid adenocarcinoma of the stomach. Of these 31 cases, 24 were male, accounting for 77.4% of the cases. Serum alpha-fetoprotein (AFP) levels were notably elevated, with radioimmunoassay results reaching 1240 ng/ml; 28 out of 31 cases had AFP levels below 25 µg/l, accounting for 90.3%. Related genes: HER2 protein indicated positive expression on the cell membrane in 35.5% (11/31) of the cases; HER2 gene amplification detected by the FISH technique was 12.9% (4/31). Tumoral stromal lymphocytes exhibited a PD-1 positive expression rate of 58.1% (18/31). In gastric cancer tissues, the PD-L1 positive rate was 45.1% (14/31).</p><p><strong>Conclusion: </strong>HAS represents a distinctive subtype of gastric cancer with a propensity for mimicking other forms of tumors, underscoring the significance of discerning its unique histopathological attributes for accurate differential diagnosis and tailored therapeutic interventions.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140959495","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-04-20DOI: 10.1007/s12094-024-03487-4
Manuel Valladares-Ayerbes, Maria José Safont, Encarnación González Flores, Pilar García-Alfonso, Enrique Aranda, Ana-Maria López Muñoz, Esther Falcó Ferrer, Luís Cirera Nogueras, Nuria Rodríguez-Salas, Jorge Aparicio, Marta Llanos Muñoz, Paola Patricia Pimentel Cáceres, Oscar Alfredo Castillo Trujillo, Rosario Vidal Tocino, Mercedes Salgado Fernández, Antonieta Salud-Salvia, Bartomeu Massuti Sureda, Rocio Garcia-Carbonero, Maria Ángeles Vicente Conesa, Ariadna Lloansí Vila
Purpose: RAS (KRAS/NRAS) mutational status on a tumor biopsy is mandatory to guide the best treatment in metastatic colorectal cancer (mCRC). Determining the RAS mutational status by tumor-tissue biopsy is essential in guiding the optimal treatment decision for mCRC. RAS mutations are negative predictive factors for the use of EGFR monoclonal antibodies. Cell-free DNA (cfDNA) analysis enables minimally invasive monitoring of tumor evolution.
Methods/patients: PERSEIDA was an observational, prospective study assessing cfDNA RAS, BRAF and EGFR mutations (using Idylla™) in first-line mCRC, RAS wild-type (baseline tumor-tissue biopsy) patients (cohort 2). Plasma samples were collected before first-line treatment, after 20 ± 2 weeks, and at disease progression.
Results: 117 patients were included (103 received panitumumab + chemotherapy as first-line treatment). At baseline, 7 (6.8%) patients had RAS mutations, 4 (3.9%) BRAF mutations and no EGFR mutations were detected (cfDNA, panitumumab + chemotherapy subpopulation [panitumumab + Ch]). The baseline RAS mutational status concordance between tissue and liquid biopsies was 94.0% (93.2%, panitumumab + Ch). At 20 weeks, only one patient in the study (included in the panitumumab + Ch) had an emerging cfDNA RAS mutation. No emerging BRAF or EGFR mutations were reported. At disease progression, 6 patients had emergent mutations not present at baseline (RAS conversion rate: 13.3% [6/45]; 15.0% [6/40], panitumumab + Ch).
Conclusions: The concordance rate between liquid and solid biopsies at baseline was very high, as previously reported, while our results suggest a considerable emergence of RAS mutations during disease progression. Thus, the dynamics of the genomic landscape in ctDNA may provide relevant information for the management of mCRC patients.
{"title":"Sequential RAS mutations evaluation in cell-free DNA of patients with tissue RAS wild-type metastatic colorectal cancer: the PERSEIDA (Cohort 2) study.","authors":"Manuel Valladares-Ayerbes, Maria José Safont, Encarnación González Flores, Pilar García-Alfonso, Enrique Aranda, Ana-Maria López Muñoz, Esther Falcó Ferrer, Luís Cirera Nogueras, Nuria Rodríguez-Salas, Jorge Aparicio, Marta Llanos Muñoz, Paola Patricia Pimentel Cáceres, Oscar Alfredo Castillo Trujillo, Rosario Vidal Tocino, Mercedes Salgado Fernández, Antonieta Salud-Salvia, Bartomeu Massuti Sureda, Rocio Garcia-Carbonero, Maria Ángeles Vicente Conesa, Ariadna Lloansí Vila","doi":"10.1007/s12094-024-03487-4","DOIUrl":"10.1007/s12094-024-03487-4","url":null,"abstract":"<p><strong>Purpose: </strong>RAS (KRAS/NRAS) mutational status on a tumor biopsy is mandatory to guide the best treatment in metastatic colorectal cancer (mCRC). Determining the RAS mutational status by tumor-tissue biopsy is essential in guiding the optimal treatment decision for mCRC. RAS mutations are negative predictive factors for the use of EGFR monoclonal antibodies. Cell-free DNA (cfDNA) analysis enables minimally invasive monitoring of tumor evolution.</p><p><strong>Methods/patients: </strong>PERSEIDA was an observational, prospective study assessing cfDNA RAS, BRAF and EGFR mutations (using Idylla™) in first-line mCRC, RAS wild-type (baseline tumor-tissue biopsy) patients (cohort 2). Plasma samples were collected before first-line treatment, after 20 ± 2 weeks, and at disease progression.</p><p><strong>Results: </strong>117 patients were included (103 received panitumumab + chemotherapy as first-line treatment). At baseline, 7 (6.8%) patients had RAS mutations, 4 (3.9%) BRAF mutations and no EGFR mutations were detected (cfDNA, panitumumab + chemotherapy subpopulation [panitumumab + Ch]). The baseline RAS mutational status concordance between tissue and liquid biopsies was 94.0% (93.2%, panitumumab + Ch). At 20 weeks, only one patient in the study (included in the panitumumab + Ch) had an emerging cfDNA RAS mutation. No emerging BRAF or EGFR mutations were reported. At disease progression, 6 patients had emergent mutations not present at baseline (RAS conversion rate: 13.3% [6/45]; 15.0% [6/40], panitumumab + Ch).</p><p><strong>Conclusions: </strong>The concordance rate between liquid and solid biopsies at baseline was very high, as previously reported, while our results suggest a considerable emergence of RAS mutations during disease progression. Thus, the dynamics of the genomic landscape in ctDNA may provide relevant information for the management of mCRC patients.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11410833/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140861268","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-04-25DOI: 10.1007/s12094-024-03436-1
Marilina Santero, Carolina Requeijo, Maria Jesus Quintana, Dulce Rodríguez, David Bottaro, Ismael Macias, Carles Pericay, Natalie Farina, Jesus Manuel Blanco, Iratxe Urreta-Barallobre, Laura Punti, Maria Angeles Nava, Xavier Bonfill Cosp
Aim: To assess the appropriateness of systemic oncological treatments (SOT) provided to patients diagnosed with advanced esophageal cancer (EC) across a group of participating hospitals.
Methods: Multicenter, retrospective cohort study in five Spanish hospitals including newly confirmed advanced EC cases between July 1, 2014, and June 30, 2016, with a 5-year follow-up.
Results: We identified 157 patients fulfilling the inclusion criteria (median age: 65 years, 85.9% males). Most patients, 125 (79.6%) were treated at least with one active treatment, and 33% received two or more lines of SOT. The 1-, 2- and 5-year overall survival rates were 30.3% [95%CI: 23.8, 38.7], 14.0% [95%CI: 9.3, 21.0], and 7.1% [95% CI: 3.8, 13.1] respectively, and the median survival time 8 months (95% CI: 6, 19) for stages IIIb IIIc and 7 months (95% CI: 5, 9) for stage IV. Clinical stage, receiving more than one line of SOT, and treatment with radiotherapy accelerated the time to death (0.4, 0.9-, and 0.8-times shorter survival respectively, p < 0.05). Better performance status (ECOG < 2) extended survival time by 2.2 times (p = 0.04). Age < 65 years (OR 9.4, 95% CI 3.2, 31.4, p < 0.001), and being treated in one particular hospital (OR 0.2, 95% CI 0.0, 0.8, p < 0.01) were associated with the administration of two or more lines of SOT. Altogether, 18.9% and 9.0% of patients received chemotherapy in the last four and two weeks of life, respectively. Moreover, 2.5% of patients were prescribed a new line of chemotherapy during the last month of life. The proportion of all patients who did not have access to palliative care reached 29.3%, and among those who had access to it, 34.2% initiated it in the last month of life.
Conclusion: A high proportion of advanced EC patients receive many treatments not based on sound evidence and they do not benefit enough from palliative care services. The most accepted appropriateness indicators point out that some of the analyzed patients could have been overtreated. This study provides important insights into the quality of care provided to advanced EC, and furthermore, for giving valuable insight and opportunities for improvement.
目的:评估参与研究的医院为确诊为晚期食管癌(EC)患者提供全身肿瘤治疗(SOT)的适当性:在西班牙五家医院开展多中心回顾性队列研究,包括2014年7月1日至2016年6月30日期间新确诊的晚期食管癌病例,并进行为期5年的随访:我们发现157名患者符合纳入标准(中位年龄:65岁,85.9%为男性)。大多数患者(125人,79.6%)至少接受了一种积极治疗,33%的患者接受了两种或两种以上的SOT治疗。1年、2年和5年总生存率分别为30.3%[95%CI:23.8,38.7]、14.0%[95%CI:9.3,21.0]和7.1%[95%CI:3.8,13.1],中位生存时间IIIb IIIc期为8个月(95%CI:6,19),IV期为7个月(95%CI:5,9)。临床分期、接受一种以上的 SOT 治疗和放疗会加快患者的死亡时间(生存期分别缩短 0.4、0.9 和 0.8 倍,P 结论:很高比例的晚期肺癌患者接受了许多并非基于可靠证据的治疗,他们从姑息治疗服务中获益不足。最被认可的适当性指标指出,一些被分析的患者可能接受了过度治疗。这项研究为了解晚期心血管疾病患者的护理质量提供了重要依据,同时也提供了宝贵的改进意见和机会。
{"title":"How appropriate is treating patients diagnosed with advanced esophageal cancer with anticancer drugs? A multicenter retrospective cohort Spanish study.","authors":"Marilina Santero, Carolina Requeijo, Maria Jesus Quintana, Dulce Rodríguez, David Bottaro, Ismael Macias, Carles Pericay, Natalie Farina, Jesus Manuel Blanco, Iratxe Urreta-Barallobre, Laura Punti, Maria Angeles Nava, Xavier Bonfill Cosp","doi":"10.1007/s12094-024-03436-1","DOIUrl":"10.1007/s12094-024-03436-1","url":null,"abstract":"<p><strong>Aim: </strong>To assess the appropriateness of systemic oncological treatments (SOT) provided to patients diagnosed with advanced esophageal cancer (EC) across a group of participating hospitals.</p><p><strong>Methods: </strong>Multicenter, retrospective cohort study in five Spanish hospitals including newly confirmed advanced EC cases between July 1, 2014, and June 30, 2016, with a 5-year follow-up.</p><p><strong>Results: </strong>We identified 157 patients fulfilling the inclusion criteria (median age: 65 years, 85.9% males). Most patients, 125 (79.6%) were treated at least with one active treatment, and 33% received two or more lines of SOT. The 1-, 2- and 5-year overall survival rates were 30.3% [95%CI: 23.8, 38.7], 14.0% [95%CI: 9.3, 21.0], and 7.1% [95% CI: 3.8, 13.1] respectively, and the median survival time 8 months (95% CI: 6, 19) for stages IIIb IIIc and 7 months (95% CI: 5, 9) for stage IV. Clinical stage, receiving more than one line of SOT, and treatment with radiotherapy accelerated the time to death (0.4, 0.9-, and 0.8-times shorter survival respectively, p < 0.05). Better performance status (ECOG < 2) extended survival time by 2.2 times (p = 0.04). Age < 65 years (OR 9.4, 95% CI 3.2, 31.4, p < 0.001), and being treated in one particular hospital (OR 0.2, 95% CI 0.0, 0.8, p < 0.01) were associated with the administration of two or more lines of SOT. Altogether, 18.9% and 9.0% of patients received chemotherapy in the last four and two weeks of life, respectively. Moreover, 2.5% of patients were prescribed a new line of chemotherapy during the last month of life. The proportion of all patients who did not have access to palliative care reached 29.3%, and among those who had access to it, 34.2% initiated it in the last month of life.</p><p><strong>Conclusion: </strong>A high proportion of advanced EC patients receive many treatments not based on sound evidence and they do not benefit enough from palliative care services. The most accepted appropriateness indicators point out that some of the analyzed patients could have been overtreated. This study provides important insights into the quality of care provided to advanced EC, and furthermore, for giving valuable insight and opportunities for improvement.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11410856/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140873562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Despite the generally favourable prognoses observed in patients with ALK-positive non-small cell lung cancer (NSCLC), there remains significant variability in clinical outcomes. The objective of this study is to enhance patient stratification by examining both the specific sites of gene fusion and the presence of co-occurring mutations.
Methods: We collected retrospective clinical and pathological data on ALK-positive patients with locally advanced or metastatic disease. ALK fusion variants and concomitant mutations were identified through next-generation sequencing technology. We then assessed treatment efficacy via tumor response and survival metrics.
Results: This study included a total of 59 patients, with 49 harboring echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusions and 10 presenting with rare fusions. The median follow-up period was 33 months. Clinical outcomes between non-EML4-ALK and EML4-ALK patients were comparable. Among the EML4-ALK cohort, patients with longer variants (v1, v2, v8) demonstrated superior progression-free survival (PFS) (median PFS: 34 months vs. 11 months; hazard ratio [HR]: 2.28; P = 0.05) compared to those with shorter variants (v3, v5). Furthermore, patients treated with second-generation ALK inhibitors (ALKi) displayed a progression-free survival advantage (median PFS: not reached [NR] vs. 9 months; HR: 5.37; P = 0.013). Baseline TP53 co-mutation were linked with a substantially shorter OS (median OS,37 months vs. NR; HR 2.74; P = 0.047).
Conclusions: In ALK+ NSCLC, longer EML4-ALK variants correlate with improved prognosis and enhanced response to second-generation ALKi, while TP53 co-mutations indicate a negative prognosis.
{"title":"Clinical difference on the variants and co-mutation in a Chinese cohort with ALK-positive advanced non-small cell lung cancer.","authors":"Ying Fu, Qing Liu, Xiaohan Wang, Liangchao Sun, Xiao Han, Xue Meng","doi":"10.1007/s12094-024-03481-w","DOIUrl":"10.1007/s12094-024-03481-w","url":null,"abstract":"<p><strong>Purpose: </strong>Despite the generally favourable prognoses observed in patients with ALK-positive non-small cell lung cancer (NSCLC), there remains significant variability in clinical outcomes. The objective of this study is to enhance patient stratification by examining both the specific sites of gene fusion and the presence of co-occurring mutations.</p><p><strong>Methods: </strong>We collected retrospective clinical and pathological data on ALK-positive patients with locally advanced or metastatic disease. ALK fusion variants and concomitant mutations were identified through next-generation sequencing technology. We then assessed treatment efficacy via tumor response and survival metrics.</p><p><strong>Results: </strong>This study included a total of 59 patients, with 49 harboring echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusions and 10 presenting with rare fusions. The median follow-up period was 33 months. Clinical outcomes between non-EML4-ALK and EML4-ALK patients were comparable. Among the EML4-ALK cohort, patients with longer variants (v1, v2, v8) demonstrated superior progression-free survival (PFS) (median PFS: 34 months vs. 11 months; hazard ratio [HR]: 2.28; P = 0.05) compared to those with shorter variants (v3, v5). Furthermore, patients treated with second-generation ALK inhibitors (ALKi) displayed a progression-free survival advantage (median PFS: not reached [NR] vs. 9 months; HR: 5.37; P = 0.013). Baseline TP53 co-mutation were linked with a substantially shorter OS (median OS,37 months vs. NR; HR 2.74; P = 0.047).</p><p><strong>Conclusions: </strong>In ALK+ NSCLC, longer EML4-ALK variants correlate with improved prognosis and enhanced response to second-generation ALKi, while TP53 co-mutations indicate a negative prognosis.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140867468","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-01Epub Date: 2024-04-28DOI: 10.1007/s12094-024-03459-8
Enzhao Zhu, Linmei Zhang, Yixian Liu, Tianyu Ji, Jianmeng Dai, Ruichen Tang, Jiayi Wang, Chunyu Hu, Kai Chen, Qianyi Yu, Qiuyi Lu, Zisheng Ai
Background: The survival advantage of neoadjuvant systemic therapy (NST) for breast cancer patients remains controversial, especially when considering the heterogeneous characteristics of individual patients.
Objective: To discern the variability in responses to breast cancer treatment at the individual level and propose personalized treatment recommendations utilizing deep learning (DL).
Methods: Six models were developed to offer individualized treatment suggestions. Outcomes for patients whose actual treatments aligned with model recommendations were compared to those whose did not. The influence of certain baseline features of patients on NST selection was visualized and quantified by multivariate logistic regression and Poisson regression analyses.
Results: Our study included 94,487 female breast cancer patients. The Balanced Individual Treatment Effect for Survival data (BITES) model outperformed other models in performance, showing a statistically significant protective effect with inverse probability treatment weighting (IPTW)-adjusted baseline features [IPTW-adjusted hazard ratio: 0.51, 95% confidence interval (CI), 0.41-0.64; IPTW-adjusted risk difference: 21.46, 95% CI 18.90-24.01; IPTW-adjusted difference in restricted mean survival time: 21.51, 95% CI 19.37-23.80]. Adherence to BITES recommendations is associated with reduced breast cancer mortality and fewer adverse effects. BITES suggests that patients with TNM stage IIB, IIIB, triple-negative subtype, a higher number of positive axillary lymph nodes, and larger tumors are most likely to benefit from NST.
Conclusions: Our results demonstrated the potential of BITES to aid in clinical treatment decisions and offer quantitative treatment insights. In our further research, these models should be validated in clinical settings and additional patient features as well as outcome measures should be studied in depth.
背景:乳腺癌患者接受新辅助全身治疗(NST)的生存优势仍存在争议:乳腺癌患者新辅助系统疗法(NST)的生存优势仍存在争议,尤其是考虑到个体患者的异质性特征时:利用深度学习(DL)辨别个体对乳腺癌治疗反应的差异性,并提出个性化治疗建议:方法:开发了六个模型来提供个性化治疗建议。将实际治疗与模型建议一致的患者的治疗结果与不一致的患者的治疗结果进行比较。通过多变量逻辑回归和泊松回归分析,对患者的某些基线特征对 NST 选择的影响进行了可视化和量化:我们的研究纳入了 94,487 名女性乳腺癌患者。生存数据的平衡个体治疗效果(BITES)模型在性能上优于其他模型,在反概率治疗加权(IPTW)调整基线特征后,显示出具有统计学意义的保护效果[IPTW调整后的危险比:0.51,95%置信区间(CI),0.41-0.64;IPTW调整后的风险差异:21.46,95% CI 18.90-24.01;IPTW调整后的受限平均生存时间差异:21.51,95% CI 19.37-23.80]。遵守 BITES 建议与降低乳腺癌死亡率和减少不良反应有关。BITES建议TNM分期为IIB、IIIB、三阴亚型、腋窝淋巴结阳性数目较多、肿瘤较大的患者最有可能从NST中获益:我们的研究结果表明,BITES 具备辅助临床治疗决策和提供定量治疗见解的潜力。在我们的进一步研究中,这些模型应在临床环境中得到验证,并应深入研究更多的患者特征和疗效指标。
{"title":"Determining individual suitability for neoadjuvant systemic therapy in breast cancer patients through deep learning.","authors":"Enzhao Zhu, Linmei Zhang, Yixian Liu, Tianyu Ji, Jianmeng Dai, Ruichen Tang, Jiayi Wang, Chunyu Hu, Kai Chen, Qianyi Yu, Qiuyi Lu, Zisheng Ai","doi":"10.1007/s12094-024-03459-8","DOIUrl":"10.1007/s12094-024-03459-8","url":null,"abstract":"<p><strong>Background: </strong>The survival advantage of neoadjuvant systemic therapy (NST) for breast cancer patients remains controversial, especially when considering the heterogeneous characteristics of individual patients.</p><p><strong>Objective: </strong>To discern the variability in responses to breast cancer treatment at the individual level and propose personalized treatment recommendations utilizing deep learning (DL).</p><p><strong>Methods: </strong>Six models were developed to offer individualized treatment suggestions. Outcomes for patients whose actual treatments aligned with model recommendations were compared to those whose did not. The influence of certain baseline features of patients on NST selection was visualized and quantified by multivariate logistic regression and Poisson regression analyses.</p><p><strong>Results: </strong>Our study included 94,487 female breast cancer patients. The Balanced Individual Treatment Effect for Survival data (BITES) model outperformed other models in performance, showing a statistically significant protective effect with inverse probability treatment weighting (IPTW)-adjusted baseline features [IPTW-adjusted hazard ratio: 0.51, 95% confidence interval (CI), 0.41-0.64; IPTW-adjusted risk difference: 21.46, 95% CI 18.90-24.01; IPTW-adjusted difference in restricted mean survival time: 21.51, 95% CI 19.37-23.80]. Adherence to BITES recommendations is associated with reduced breast cancer mortality and fewer adverse effects. BITES suggests that patients with TNM stage IIB, IIIB, triple-negative subtype, a higher number of positive axillary lymph nodes, and larger tumors are most likely to benefit from NST.</p><p><strong>Conclusions: </strong>Our results demonstrated the potential of BITES to aid in clinical treatment decisions and offer quantitative treatment insights. In our further research, these models should be validated in clinical settings and additional patient features as well as outcome measures should be studied in depth.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140868112","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}