Pub Date : 2024-10-21eCollection Date: 2024-01-01DOI: 10.1177/17588359241280689
Sun Min Lim, Stefanie S Schalm, Eun Ji Lee, Sewon Park, Chiara Conti, Yves A Millet, Rich Woessner, Zhuo Zhang, Luz E Tavera-Mendoza, Faith Stevison, Faris Albayya, Thomas A Dineen, John Hsieh, Seung Yeon Oh, Alena Zalutskaya, Julia Rotow, Koichi Goto, Dae-Ho Lee, Mi Ran Yun, Byoung Chul Cho
Introduction: Despite the availability of several epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), most patients with non-small-cell lung cancer (NSCLC) eventually develop resistance to these agents. Notably, EGFR_C797S mutations confer resistance to the third-generation EGFR-TKI osimertinib and no approved post-osimertinib targeted pharmacology options are currently available. BLU-945 is a novel, reversible, and orally available next-generation EGFR-TKI that selectively targets EGFR-activating (EGFRm) and resistance mutations (including EGFR_C797S) with nanomolar potency while sparing wild-type EGFR in vitro.
Methods: In vitro activity of BLU-945 as a single agent and in combination with osimertinib was tested in engineered EGFR-mutant cell lines as well as patient-derived cells and patient-derived organoids. In vivo activity was evaluated in osimertinib-resistant patient-derived xenograft mouse models. Three patient cases from the global, first-in-human, phase I/II SYMPHONY trial (NCT04862780) demonstrating the clinical efficacy of BLU-945 were reported.
Results: In vitro BLU-945 demonstrated inhibited cell viability and growth of EGFR-mutant/osimertinib-resistant cell lines. BLU-945 demonstrated in vivo tumor shrinkage in osimertinib-resistant models of NSCLC (osimertinib second line: EGFR_L858R/C797S and third line: EGFR_ex19del/T790M/C797S and L858R/T790M/C797S) both as monotherapy and in combination with osimertinib. BLU-945 also demonstrated tumor shrinkage in patients from the SYMPHONY trial.
Conclusion: Our findings demonstrate the preclinical and early clinical activity of BLU-945 in EGFRm NSCLC progressing on previous EGFR-TKIs.
{"title":"BLU-945, a potent and selective next-generation EGFR TKI, has antitumor activity in models of osimertinib-resistant non-small-cell lung cancer.","authors":"Sun Min Lim, Stefanie S Schalm, Eun Ji Lee, Sewon Park, Chiara Conti, Yves A Millet, Rich Woessner, Zhuo Zhang, Luz E Tavera-Mendoza, Faith Stevison, Faris Albayya, Thomas A Dineen, John Hsieh, Seung Yeon Oh, Alena Zalutskaya, Julia Rotow, Koichi Goto, Dae-Ho Lee, Mi Ran Yun, Byoung Chul Cho","doi":"10.1177/17588359241280689","DOIUrl":"https://doi.org/10.1177/17588359241280689","url":null,"abstract":"<p><strong>Introduction: </strong>Despite the availability of several epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), most patients with non-small-cell lung cancer (NSCLC) eventually develop resistance to these agents. Notably, <i>EGFR</i>_C797S mutations confer resistance to the third-generation EGFR-TKI osimertinib and no approved post-osimertinib targeted pharmacology options are currently available. BLU-945 is a novel, reversible, and orally available next-generation EGFR-TKI that selectively targets EGFR-activating (<i>EGFR</i>m) and resistance mutations (including EGFR_C797S) with nanomolar potency while sparing wild-type EGFR in vitro.</p><p><strong>Methods: </strong>In vitro activity of BLU-945 as a single agent and in combination with osimertinib was tested in engineered <i>EGFR</i>-mutant cell lines as well as patient-derived cells and patient-derived organoids. In vivo activity was evaluated in osimertinib-resistant patient-derived xenograft mouse models. Three patient cases from the global, first-in-human, phase I/II SYMPHONY trial (NCT04862780) demonstrating the clinical efficacy of BLU-945 were reported.</p><p><strong>Results: </strong>In vitro BLU-945 demonstrated inhibited cell viability and growth of <i>EGFR</i>-mutant/osimertinib-resistant cell lines. BLU-945 demonstrated in vivo tumor shrinkage in osimertinib-resistant models of NSCLC (osimertinib second line: <i>EGFR</i>_L858R/C797S and third line: <i>EGFR</i>_ex19del/T790M/C797S and L858R/T790M/C797S) both as monotherapy and in combination with osimertinib. BLU-945 also demonstrated tumor shrinkage in patients from the SYMPHONY trial.</p><p><strong>Conclusion: </strong>Our findings demonstrate the preclinical and early clinical activity of BLU-945 in <i>EGFR</i>m NSCLC progressing on previous EGFR-TKIs.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241280689"},"PeriodicalIF":4.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497503/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508533","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}
Pub Date : 2024-10-21eCollection Date: 2024-01-01DOI: 10.1177/17588359241290373
[This corrects the article DOI: 10.1177/17588359241267148.].
[This corrects the article DOI: 10.1177/17588359241267148.].
{"title":"Erratum to \"Additional biomarkers for pathological complete response in triple negative breast cancer\".","authors":"","doi":"10.1177/17588359241290373","DOIUrl":"https://doi.org/10.1177/17588359241290373","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1177/17588359241267148.].</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241290373"},"PeriodicalIF":4.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497544/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142508535","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}
Pub Date : 2024-10-18eCollection Date: 2024-01-01DOI: 10.1177/17588359241289202
Wei Xu, Huai Gong, Bolun Li, Xinmin Yin
Aim: To explore clinical features and prognosis of hepatocellular carcinoma (HCC) in hepatitis B virus surface antigen (HBsAg)-serocleared patients and identify risk factors associated with postoperative recurrence after curative hepatectomy.
Methods: Patients who had undergone initial hepatectomy for HCC from January 2010 through December 2022. Clinicopathological data were compared between HBsAg-seropositive and HBsAg-serocleared patients. Furthermore, risk factors associated with early and late postoperative HCC recurrence (early and late recurrences (ER and LR), respectively) were analyzed for HBsAg-serocleared HCC patients treated by curative hepatectomy.
Results: A total of 2184 consecutive patients undergoing initial hepatectomy for HCC were enrolled, including 339 (15.5%) HBsAg-serocleared and 1845 (84.5%) HBsAg-seropositive cases. Tumor characteristics were comparable between the two groups. After curative hepatectomy, the ER rate was lower in the HBsAg-serocleared group than in the HBsAg-seropositive group (16.2% vs 26.3%; p = 0.000). LR rates in the HBsAg-seropositive and HBsAg-serocleared groups were similar (8.3% vs 6.9%, respectively, p = 0.418). Multivariate analysis showed that among HBsAg-serocleared patients, Hong Kong Liver Cancer stage and microvascular invasion were risk factors associated with postoperative ER, while γ-glutamyl transferase level and neutrophil-to-lymphocyte ratio were associated with LR.
Conclusion: HBsAg-serocleared and HBsAg-seropositive HCC patients exhibited similar tumor characteristics. Curative hepatectomy-treated HBsAg-serocleared HCC patients experienced a lower ER rate and better short-term (⩽3 years) overall survival (OS) rates than their HBsAg-seropositive counterparts. LR, very late recurrence, and long-term (4-, and 5-year) OS rates were similar between the two groups.
{"title":"Hepatocellular carcinoma in HBsAg seroclearance: clinical features, recurrence, and prognosis following curative hepatectomy.","authors":"Wei Xu, Huai Gong, Bolun Li, Xinmin Yin","doi":"10.1177/17588359241289202","DOIUrl":"10.1177/17588359241289202","url":null,"abstract":"<p><strong>Aim: </strong>To explore clinical features and prognosis of hepatocellular carcinoma (HCC) in hepatitis B virus surface antigen (HBsAg)-serocleared patients and identify risk factors associated with postoperative recurrence after curative hepatectomy.</p><p><strong>Methods: </strong>Patients who had undergone initial hepatectomy for HCC from January 2010 through December 2022. Clinicopathological data were compared between HBsAg-seropositive and HBsAg-serocleared patients. Furthermore, risk factors associated with early and late postoperative HCC recurrence (early and late recurrences (ER and LR), respectively) were analyzed for HBsAg-serocleared HCC patients treated by curative hepatectomy.</p><p><strong>Results: </strong>A total of 2184 consecutive patients undergoing initial hepatectomy for HCC were enrolled, including 339 (15.5%) HBsAg-serocleared and 1845 (84.5%) HBsAg-seropositive cases. Tumor characteristics were comparable between the two groups. After curative hepatectomy, the ER rate was lower in the HBsAg-serocleared group than in the HBsAg-seropositive group (16.2% vs 26.3%; <i>p</i> = 0.000). LR rates in the HBsAg-seropositive and HBsAg-serocleared groups were similar (8.3% vs 6.9%, respectively, <i>p</i> = 0.418). Multivariate analysis showed that among HBsAg-serocleared patients, Hong Kong Liver Cancer stage and microvascular invasion were risk factors associated with postoperative ER, while γ-glutamyl transferase level and neutrophil-to-lymphocyte ratio were associated with LR.</p><p><strong>Conclusion: </strong>HBsAg-serocleared and HBsAg-seropositive HCC patients exhibited similar tumor characteristics. Curative hepatectomy-treated HBsAg-serocleared HCC patients experienced a lower ER rate and better short-term (⩽3 years) overall survival (OS) rates than their HBsAg-seropositive counterparts. LR, very late recurrence, and long-term (4-, and 5-year) OS rates were similar between the two groups.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241289202"},"PeriodicalIF":4.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526261/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558856","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}
Pub Date : 2024-10-18eCollection Date: 2024-01-01DOI: 10.1177/17588359241287658
Loredana G Marcu, David C Marcu
Medical oncology, through conventional chemotherapy as well as targeted drugs, remains an important component of cancer patient management, particularly for systemic disease. Despite advances in all areas of medical oncology, certain challenges persist in the form of drug resistance and severe normal tissue toxicity. These unwanted effects can be counteracted through a patient-tailored treatment approach, which in chemotherapy is translated as pharmacogenomics. This research field investigates the way genetic makeup influences a patient's response to various drugs with the aim to minimize trial-and-error associated with drug administration. The paper introduces the role, advances and challenges of pharmacogenomics, highlighting the importance of Big Data mining to reveal the mechanisms behind drug-gene pair interaction for better patient outcomes. International consortiums have prioritized their focus on the clinical implementation of pharmacogenomics while tackling the challenges ahead: data standardization, ethical aspects and the education of physicians and patients alike to comprehend the power of pharmacogenomics to transform medical oncology.
{"title":"Pharmacogenomics and Big Data in medical oncology: developments and challenges.","authors":"Loredana G Marcu, David C Marcu","doi":"10.1177/17588359241287658","DOIUrl":"10.1177/17588359241287658","url":null,"abstract":"<p><p>Medical oncology, through conventional chemotherapy as well as targeted drugs, remains an important component of cancer patient management, particularly for systemic disease. Despite advances in all areas of medical oncology, certain challenges persist in the form of drug resistance and severe normal tissue toxicity. These unwanted effects can be counteracted through a patient-tailored treatment approach, which in chemotherapy is translated as pharmacogenomics. This research field investigates the way genetic makeup influences a patient's response to various drugs with the aim to minimize trial-and-error associated with drug administration. The paper introduces the role, advances and challenges of pharmacogenomics, highlighting the importance of Big Data mining to reveal the mechanisms behind drug-gene pair interaction for better patient outcomes. International consortiums have prioritized their focus on the clinical implementation of pharmacogenomics while tackling the challenges ahead: data standardization, ethical aspects and the education of physicians and patients alike to comprehend the power of pharmacogenomics to transform medical oncology.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241287658"},"PeriodicalIF":4.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526290/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558858","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}
Pub Date : 2024-10-17eCollection Date: 2024-01-01DOI: 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.
{"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":"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":"16 ","pages":"17588359241289200"},"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}
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.
{"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":"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":"16 ","pages":"17588359241290129"},"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}
Background: Bevacizumab is extensively used in the treatment of advanced non-small-cell lung cancer (NSCLC). Numerous clinical trials have proven the clinical efficacies of bevacizumab biosimilars (BB).
Objective: Our study aimed to compare the clinical outcomes between bevacizumab reference product (RP) and BB among advanced NSCLC patients in a real-world setting.
Design: We retrospectively analyzed stage IV metastatic NSCLC patients who were treated with bevacizumab as part of a combination therapy. Patients were categorized into chemotherapy (CT) and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) groups. We compared the patients' characteristics, treatment efficacy, and adverse events between RP and BB in the two treatment groups.
Methods: From January 2020 to July 2022, a total of 171 patients who underwent combination therapy with bevacizumab were screened. Seventy-nine of these patients met the study's inclusion criteria and were enrolled in the final analysis. We utilized the Kaplan-Meier method to estimate progression-free survival (PFS) and the log-rank test to compare PFS between groups. The Cox proportional hazards model was used to identify predictors of PFS.
Results: Within the CT cohort, 34 patients were treated with RP in combination with platinum and pemetrexed, and 25 patients received a combination regimen with BB. The median PFS was 6.9 months in the RP group and 8.9 months in the BB group (p = 0.255). Within the EGFR-TKI cohort, 20 patients with EGFR-mutant NSCLC received first-line treatment with EGFR-TKI plus bevacizumab. Of these patients, 9 were treated with a combination regimen that included RP, and 11 patients received EGFR-TKI in combination with BB. The median PFS was 18.4 months for the RP group and 13.6 months for the BB group (p = 0.363).
Conclusion: In our advanced NSCLC patients, we found no difference in clinical outcomes when receiving treatment with RP or BB. Given a combination regimen, BB was as effective as RP together with either CT or EGFR-TKIs.
{"title":"Real-world clinical efficacy of bevacizumab biosimilar in patients with advanced non-small-cell lung cancer.","authors":"Wei-Fan Ou, Kuo-Hsuan Hsu, Jeng-Sen Tseng, Po-Hsin Lee, Kun-Chieh Chen, Yen-Hsiang Huang, Gee-Chen Chang, Tsung-Ying Yang","doi":"10.1177/17588359241290718","DOIUrl":"10.1177/17588359241290718","url":null,"abstract":"<p><strong>Background: </strong>Bevacizumab is extensively used in the treatment of advanced non-small-cell lung cancer (NSCLC). Numerous clinical trials have proven the clinical efficacies of bevacizumab biosimilars (BB).</p><p><strong>Objective: </strong>Our study aimed to compare the clinical outcomes between bevacizumab reference product (RP) and BB among advanced NSCLC patients in a real-world setting.</p><p><strong>Design: </strong>We retrospectively analyzed stage IV metastatic NSCLC patients who were treated with bevacizumab as part of a combination therapy. Patients were categorized into chemotherapy (CT) and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) groups. We compared the patients' characteristics, treatment efficacy, and adverse events between RP and BB in the two treatment groups.</p><p><strong>Methods: </strong>From January 2020 to July 2022, a total of 171 patients who underwent combination therapy with bevacizumab were screened. Seventy-nine of these patients met the study's inclusion criteria and were enrolled in the final analysis. We utilized the Kaplan-Meier method to estimate progression-free survival (PFS) and the log-rank test to compare PFS between groups. The Cox proportional hazards model was used to identify predictors of PFS.</p><p><strong>Results: </strong>Within the CT cohort, 34 patients were treated with RP in combination with platinum and pemetrexed, and 25 patients received a combination regimen with BB. The median PFS was 6.9 months in the RP group and 8.9 months in the BB group (<i>p</i> = 0.255). Within the EGFR-TKI cohort, 20 patients with <i>EGFR</i>-mutant NSCLC received first-line treatment with EGFR-TKI plus bevacizumab. Of these patients, 9 were treated with a combination regimen that included RP, and 11 patients received EGFR-TKI in combination with BB. The median PFS was 18.4 months for the RP group and 13.6 months for the BB group (<i>p</i> = 0.363).</p><p><strong>Conclusion: </strong>In our advanced NSCLC patients, we found no difference in clinical outcomes when receiving treatment with RP or BB. Given a combination regimen, BB was as effective as RP together with either CT or EGFR-TKIs.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241290718"},"PeriodicalIF":4.3,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526195/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558859","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}
Pub Date : 2024-10-17eCollection Date: 2024-01-01DOI: 10.1177/17588359241290733
Lisha Ye, Wenjing Wang, Huihui Li, Yongling Ji, Xiuning Le, Xiaoling Xu
Non-small cell lung cancer (NSCLC) represents the most prevalent histological subtype of lung cancer. Within this disease, the MET gene emerges as a critical therapeutic target, exhibiting various forms of dysregulation. Although MET tyrosine kinase inhibitors, HGF/c-MET targeting antibodies, and antibody-drug conjugates constitute the primary treatment modalities for patients with MET-altered NSCLC, numerous questions remain regarding their optimal application. The advent of immunotherapy holds promise for enhancing therapeutic outcomes in patients with MET-altered NSCLC. MET mutations can reshape the tumor immune microenvironment of NSCLC by reducing tumor immunogenicity, inducing exhaustion in immune-activated cells, and promoting immune evasion, which are crucial for modulating treatment responses. Furthermore, we emphasize the promising synergy of immunotherapy with emerging treatments and the challenges and opportunities in refining these approaches to improve patient outcomes.
{"title":"Targeting the MET gene: unveiling therapeutic opportunities in immunotherapy within the tumor immune microenvironment of non-small cell lung cancer.","authors":"Lisha Ye, Wenjing Wang, Huihui Li, Yongling Ji, Xiuning Le, Xiaoling Xu","doi":"10.1177/17588359241290733","DOIUrl":"10.1177/17588359241290733","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) represents the most prevalent histological subtype of lung cancer. Within this disease, the MET gene emerges as a critical therapeutic target, exhibiting various forms of dysregulation. Although MET tyrosine kinase inhibitors, HGF/c-MET targeting antibodies, and antibody-drug conjugates constitute the primary treatment modalities for patients with MET-altered NSCLC, numerous questions remain regarding their optimal application. The advent of immunotherapy holds promise for enhancing therapeutic outcomes in patients with MET-altered NSCLC. MET mutations can reshape the tumor immune microenvironment of NSCLC by reducing tumor immunogenicity, inducing exhaustion in immune-activated cells, and promoting immune evasion, which are crucial for modulating treatment responses. Furthermore, we emphasize the promising synergy of immunotherapy with emerging treatments and the challenges and opportunities in refining these approaches to improve patient outcomes.</p>","PeriodicalId":23053,"journal":{"name":"Therapeutic Advances in Medical Oncology","volume":"16 ","pages":"17588359241290733"},"PeriodicalIF":4.3,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526239/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558860","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}
Pub Date : 2024-10-17eCollection Date: 2024-01-01DOI: 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.
{"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":"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":"16 ","pages":"17588359241289648"},"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}
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":"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":"16 ","pages":"17588359241289671"},"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}