Background: Both hyperthermic intraperitoneal chemotherapy (HIPEC) and conventional intraperitoneal chemotherapy (IP) have shown survival benefits in ovarian cancer (OC), but direct comparisons between the two perfusion modalities are lacking. This study aimed to compare effectiveness and safety between HIPEC and conventional IP in OC.
Methods: This retrospective real-world study analyzed 606 patients with stages II-IV OC who received HIPEC or IP following cytoreductive surgery between 2013 and 2024. The primary endpoint was progression-free survival. Overall survival and adverse events were secondary endpoints. The study used inverse probability of treatment propensity-score weighting. We also conducted sensitivity analyses to evaluate result robustness and subgroup analyses to explore potential effect modification.
Results: After a median follow-up of 26 months, disease progression occurred in 40.6% of patients in the HIPEC group and 55.0% in the IP group (hazard ratio [HR] 0.79; P = .103). Mortality rates were 13.2% and 22.5%, respectively (HR 0.83; P = .434), showing no significant differences in progression and survival between the two groups. Exploratory subgroup analyses suggested a trend toward improved progression-free outcomes with HIPEC, particularly among patients with BRCA wild-type or BRCA1-mutated tumors and early postoperative perfusion. Hypoalbuminemia was the most common event in both groups (HIPEC 27.2%; IP 15.6%). HIPEC group had more abdominal distension and wound dehiscence, whereas IP patients experienced nausea and rash more frequently.
Conclusions: HIPEC did not significantly improve survival over conventional IP in the overall population, but showed greater benefit in specific subgroups, underscoring the importance of individualized intraperitoneal chemotherapy strategies in OC.
背景:高温腹腔化疗(HIPEC)和常规腹腔化疗(IP)都显示出卵巢癌(OC)的生存益处,但缺乏两种灌注方式之间的直接比较。本研究旨在比较HIPEC与常规IP治疗OC的有效性和安全性。方法:这项回顾性现实世界研究分析了606例Ⅱ-IV期OC患者,这些患者在2013年至2024年期间接受了HIPEC或IP细胞减少手术。主要终点为无进展生存期。总生存期和不良事件是次要终点。本研究采用治疗倾向-得分加权的逆概率法。我们还进行了敏感性分析来评估结果的稳健性,并进行了亚组分析来探索潜在的效果修正。结果:中位随访26个月后,HIPEC组40.6%的患者出现疾病进展,IP组55.0%(风险比[HR] 0.79; p = 0.103)。死亡率分别为13.2%和22.5% (HR 0.83; p = 0.434),两组间的进展和生存无显著差异。探索性亚组分析表明,HIPEC有改善无进展预后的趋势,特别是在BRCA野生型或brca1突变肿瘤和术后早期灌注的患者中。低白蛋白血症是两组中最常见的事件(HIPEC 27.2%; IP 15.6%)。HIPEC组有更多的腹胀和伤口裂开,而IP组有更多的恶心和皮疹。结论:在总体人群中,HIPEC并没有显著提高常规IP的生存率,但在特定亚组中表现出更大的益处,这强调了个体化腹腔内化疗策略在OC中的重要性。
{"title":"Real-world effectiveness and safety of hyperthermic intraperitoneal chemotherapy and intraperitoneal chemotherapy in ovarian cancer.","authors":"Siyi Zhang, Qian Qie, Yuan Zhang, Ye Liang, Siyu Yang, Yiran Wang, Yuqi Wang, Yuanyuan Zhou, Aparna Singh, Yaling Zhao, Qiling Li","doi":"10.1093/oncolo/oyaf424","DOIUrl":"10.1093/oncolo/oyaf424","url":null,"abstract":"<p><strong>Background: </strong>Both hyperthermic intraperitoneal chemotherapy (HIPEC) and conventional intraperitoneal chemotherapy (IP) have shown survival benefits in ovarian cancer (OC), but direct comparisons between the two perfusion modalities are lacking. This study aimed to compare effectiveness and safety between HIPEC and conventional IP in OC.</p><p><strong>Methods: </strong>This retrospective real-world study analyzed 606 patients with stages II-IV OC who received HIPEC or IP following cytoreductive surgery between 2013 and 2024. The primary endpoint was progression-free survival. Overall survival and adverse events were secondary endpoints. The study used inverse probability of treatment propensity-score weighting. We also conducted sensitivity analyses to evaluate result robustness and subgroup analyses to explore potential effect modification.</p><p><strong>Results: </strong>After a median follow-up of 26 months, disease progression occurred in 40.6% of patients in the HIPEC group and 55.0% in the IP group (hazard ratio [HR] 0.79; P = .103). Mortality rates were 13.2% and 22.5%, respectively (HR 0.83; P = .434), showing no significant differences in progression and survival between the two groups. Exploratory subgroup analyses suggested a trend toward improved progression-free outcomes with HIPEC, particularly among patients with BRCA wild-type or BRCA1-mutated tumors and early postoperative perfusion. Hypoalbuminemia was the most common event in both groups (HIPEC 27.2%; IP 15.6%). HIPEC group had more abdominal distension and wound dehiscence, whereas IP patients experienced nausea and rash more frequently.</p><p><strong>Conclusions: </strong>HIPEC did not significantly improve survival over conventional IP in the overall population, but showed greater benefit in specific subgroups, underscoring the importance of individualized intraperitoneal chemotherapy strategies in OC.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795328","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xubiao Wei, Yabo Jiang, Yong Li, Hong Shi, Weixing Guo, Jie Shi, Zhaochong Zeng, Tianfu Wen, Yang Jiao, Miaomiao Wang, Shuqun Cheng
Background: Radiotherapy (RT) provides meaningful local control for hepatocellular carcinoma (HCC) but is limited by radio-resistance. Preclinical and clinical data suggest that adding programmed death receptor-1 (PD-1) blockade may enhance radiosensitivity in various malignancies. We compared outcomes of stereotactic body radiotherapy (SBRT) plus PD-1 inhibitors versus SBRT alone in unresectable HCC.
Methods: We retrospectively analyzed consecutive patients treated with SBRT (January 2019-December 2024) from three centers. Key exclusions removed confounding from recent systemic/locoregional therapies. Propensity score matching (PSM, 1:1) balanced demographics, liver function, tumor characteristics and prior therapy. Tumor responses were assessed by RECIST 1.1, while survival was estimated by Kaplan-Meier and Cox models.
Results: Of 540 eligible patients, 157 received RT+PD-1 and 383 received RT alone; after PSM, 314 patients remained (157/157). Median follow-up was 29.9 months (IQR 21-36). After matching, RT+PD-1 showed a higher objective response rate (ORR) that approached significance (48.4% vs. 37.6%, p = 0.053). Progression free survival (PFS) was significantly prolonged with RT+PD-1 (median 11.0 vs. 8.5 months; 1-year 69.9% vs. 54.1%; HR 0.75, 95% CI 0.58-0.96, p = 0.024). OS also favored RT+PD-1 (median 33.9 vs. 26.3 months; 3-year 41.9% vs. 23.0%; HR 0.74, 95% CI 0.54-1.02, p = 0.066). On multivariable analysis after PSM, RT+PD-1 independently reduced risks of progression (HR 0.69, p = 0.005) and death (HR 0.70, p = 0.029). Adverse events (AEs) were similar overall, but grade ≥3 AEs were more frequent with RT+PD-1 (15.2% vs. 7.0%, p = 0.020).
Conclusions: In unresectable HCC, adding PD-1 blockade to SBRT enhances anti-tumor activity and improves long-term survival outcomes.
背景:放射治疗(RT)为肝细胞癌(HCC)提供了有意义的局部控制,但受放射耐药的限制。临床前和临床数据表明,添加程序性死亡受体-1 (PD-1)阻断剂可增强各种恶性肿瘤的放射敏感性。我们比较了立体定向放射治疗(SBRT)加PD-1抑制剂与单用SBRT治疗不可切除HCC的结果。方法:我们回顾性分析来自三个中心的连续接受SBRT治疗的患者(2019年1月- 2024年12月)。主要排除了近期系统性/局部治疗的混杂因素。倾向评分匹配(PSM, 1:1)平衡了人口统计学、肝功能、肿瘤特征和既往治疗。采用RECIST 1.1评估肿瘤反应,采用Kaplan-Meier和Cox模型估计生存率。结果:540例符合条件的患者中,157例接受RT+PD-1治疗,383例单独接受RT治疗;PSM后,314例患者存活(157/157)。中位随访时间为29.9个月(IQR 21-36)。匹配后,RT+PD-1的客观有效率(ORR)更高,接近显著性(48.4% vs. 37.6%, p = 0.053)。RT+PD-1组无进展生存期(PFS)显著延长(中位11.0个月vs 8.5个月;1年69.9% vs 54.1%; HR 0.75, 95% CI 0.58-0.96, p = 0.024)。OS也支持RT+PD-1(中位33.9 vs. 26.3个月;3年41.9% vs. 23.0%; HR 0.74, 95% CI 0.54-1.02, p = 0.066)。在PSM后的多变量分析中,RT+PD-1独立降低了进展风险(HR 0.69, p = 0.005)和死亡风险(HR 0.70, p = 0.029)。不良事件(ae)总体上相似,但≥3级ae在RT+PD-1组更常见(15.2%比7.0%,p = 0.020)。结论:在不可切除的HCC中,在SBRT中加入PD-1阻断剂可增强抗肿瘤活性并改善长期生存结果。
{"title":"Enhanced efficacy and long-term survival with SBRT plus PD-1 inhibitors versus SBRT alone in unresectable HCC: a multicenter PSM study.","authors":"Xubiao Wei, Yabo Jiang, Yong Li, Hong Shi, Weixing Guo, Jie Shi, Zhaochong Zeng, Tianfu Wen, Yang Jiao, Miaomiao Wang, Shuqun Cheng","doi":"10.1093/oncolo/oyaf437","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf437","url":null,"abstract":"<p><strong>Background: </strong>Radiotherapy (RT) provides meaningful local control for hepatocellular carcinoma (HCC) but is limited by radio-resistance. Preclinical and clinical data suggest that adding programmed death receptor-1 (PD-1) blockade may enhance radiosensitivity in various malignancies. We compared outcomes of stereotactic body radiotherapy (SBRT) plus PD-1 inhibitors versus SBRT alone in unresectable HCC.</p><p><strong>Methods: </strong>We retrospectively analyzed consecutive patients treated with SBRT (January 2019-December 2024) from three centers. Key exclusions removed confounding from recent systemic/locoregional therapies. Propensity score matching (PSM, 1:1) balanced demographics, liver function, tumor characteristics and prior therapy. Tumor responses were assessed by RECIST 1.1, while survival was estimated by Kaplan-Meier and Cox models.</p><p><strong>Results: </strong>Of 540 eligible patients, 157 received RT+PD-1 and 383 received RT alone; after PSM, 314 patients remained (157/157). Median follow-up was 29.9 months (IQR 21-36). After matching, RT+PD-1 showed a higher objective response rate (ORR) that approached significance (48.4% vs. 37.6%, p = 0.053). Progression free survival (PFS) was significantly prolonged with RT+PD-1 (median 11.0 vs. 8.5 months; 1-year 69.9% vs. 54.1%; HR 0.75, 95% CI 0.58-0.96, p = 0.024). OS also favored RT+PD-1 (median 33.9 vs. 26.3 months; 3-year 41.9% vs. 23.0%; HR 0.74, 95% CI 0.54-1.02, p = 0.066). On multivariable analysis after PSM, RT+PD-1 independently reduced risks of progression (HR 0.69, p = 0.005) and death (HR 0.70, p = 0.029). Adverse events (AEs) were similar overall, but grade ≥3 AEs were more frequent with RT+PD-1 (15.2% vs. 7.0%, p = 0.020).</p><p><strong>Conclusions: </strong>In unresectable HCC, adding PD-1 blockade to SBRT enhances anti-tumor activity and improves long-term survival outcomes.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145991845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Despite recent advances and an improved treatment paradigm, the prognosis in patients with advanced NSCLC remains poor. Novel endpoints to evaluate antitumor activity are needed to expedite safe and effective drug development.
Methods: We performed a retrospective pooled analysis of 10,495 patients with NSCLC using radiological tumor measurements across 22 randomized clinical trials fit to a regression-growth model to derive the g value. We evaluated the g value across both type and line of therapy.
Results: The g value is inversely associated with survival. Patients receiving targeted therapy had lower g values than patients receiving immune checkpoint inhibitors or chemotherapy. Additionally, patients receiving front-line treatment had a lower g value than patients receiving later line treatment. A lower g value was associated with a higher median OS and a longer median PFS.
Conclusion: The g value, as a volumetric measurement of tumor growth rate, may provide an additional or supplementary approach to assess the potential clinical activity of an agent. Prospective studies are needed to further assess the association of the g value derived from early tumor measurements with long-term outcomes in patients.
Implications for practice: Characterization of the growth rate may provide additional information regarding the potential clinical activity of a drug, which could aid in the selection of therapies for further drug development. While the g value has the potential to provide an early assessment of new therapies in a clinical trial before a time-to-event endpoint is reached, further evaluation through prospective studies and meta-analyses is needed to assess the association of g value with long-term outcomes, such as PFS and OS.
{"title":"An FDA analysis of the association of tumor growth rate, overall survival and progression-free survival in patients with metastatic NSCLC.","authors":"Justin N Malinou, Jiaxin Fan, Joyce Cheng, Yutao Gong, Yuan-Li Shen, Erin Larkins","doi":"10.1093/oncolo/oyag009","DOIUrl":"https://doi.org/10.1093/oncolo/oyag009","url":null,"abstract":"<p><strong>Background: </strong>Despite recent advances and an improved treatment paradigm, the prognosis in patients with advanced NSCLC remains poor. Novel endpoints to evaluate antitumor activity are needed to expedite safe and effective drug development.</p><p><strong>Methods: </strong>We performed a retrospective pooled analysis of 10,495 patients with NSCLC using radiological tumor measurements across 22 randomized clinical trials fit to a regression-growth model to derive the g value. We evaluated the g value across both type and line of therapy.</p><p><strong>Results: </strong>The g value is inversely associated with survival. Patients receiving targeted therapy had lower g values than patients receiving immune checkpoint inhibitors or chemotherapy. Additionally, patients receiving front-line treatment had a lower g value than patients receiving later line treatment. A lower g value was associated with a higher median OS and a longer median PFS.</p><p><strong>Conclusion: </strong>The g value, as a volumetric measurement of tumor growth rate, may provide an additional or supplementary approach to assess the potential clinical activity of an agent. Prospective studies are needed to further assess the association of the g value derived from early tumor measurements with long-term outcomes in patients.</p><p><strong>Implications for practice: </strong>Characterization of the growth rate may provide additional information regarding the potential clinical activity of a drug, which could aid in the selection of therapies for further drug development. While the g value has the potential to provide an early assessment of new therapies in a clinical trial before a time-to-event endpoint is reached, further evaluation through prospective studies and meta-analyses is needed to assess the association of g value with long-term outcomes, such as PFS and OS.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145991888","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Emmanuelle Samalin, Hélène Senellart, Simon Thezenas, Stéphane Jacquot, Stephen Ellis, Faiza Khemissa Akouz, Catherine Fiess, Mohamed Ramdani, Fabienne Portales, Eric Assenat, Marc Ychou, Laurent Mineur, Thibault Mazard
Background: Second-line FOLFOX-bevacizumab treatment is effective in metastatic colorectal cancer (mCRC) treatment after irinotecan-based chemotherapy failure. First- or second-line raltitrexed-oxaliplatin (TOMOX) treatment has an acceptable toxicity profile in mCRC. The aim of study was to evaluated TOMOX- bevacizumab combination as a second-line treatment.
Methods: The BEVATOMOX study was a non-comparative, prospective, randomized, open-label, multicentric phase II trial. Patients with histologically proven unresectable mCRC and progressive metastatic disease after irinotecan-based chemotherapy were randomized (1:2) to receive mFOLFOX6-bevacizumab (control arm, bevacizumab 5 mg/kg, mFOLFOX6 D1 = D15, 12 cycles) or TOMOX-bevacizumab (experimental arm, bevacizumab 7.5 mg/kg, raltitrexed 3 mg/m2 based on creatinine clearance, oxaliplatin 130 mg/m2, D1 = D21, 8 cycles). The primary endpoint was six-month progression-free survival (6PFS). Target enrollment was 30 and 62 patients in the control and experimental arms, respectively.
Results: Eighty-three patients (median age 66 (48-82) years, 63.9% male, 54.2% KRAS mt) were included: 33 in the control and 50 in the experimental arms. The 6PFS rate and median overall survival were 51.5% (95%CI 36-67) and 11.1 months (9.5-16.4) in the control arm vs. 38% (95%CI 26-51) and 9.3 (5.7-11.6) months in the experimental arm. In the experimental arm, left-sided tumors had a longer overall survival vs. right-sided (11.6 vs. 4.6 months, p < 0.001). Grade 3-4 toxicities (mucositis, neutropenia, febrile neutropenia, paresthesia, hand-foot syndrome) were similar between arms.
Conclusion: The TOMOX-Bev combination is a feasible second-line mCRC treatment with an acceptable toxicity profile. Recruitment failure prevents efficacy conclusions, but TOMOX-Bev may be an alternative if fluropyrimidines are contraindicated.
{"title":"The BEVATOMOX phase II trial: raltitrexed/oxaliplatin/bevacizumab vs mFOLFOX6/bevacizumab in 2nd-line metastatic colorectal cancer.","authors":"Emmanuelle Samalin, Hélène Senellart, Simon Thezenas, Stéphane Jacquot, Stephen Ellis, Faiza Khemissa Akouz, Catherine Fiess, Mohamed Ramdani, Fabienne Portales, Eric Assenat, Marc Ychou, Laurent Mineur, Thibault Mazard","doi":"10.1093/oncolo/oyag006","DOIUrl":"https://doi.org/10.1093/oncolo/oyag006","url":null,"abstract":"<p><strong>Background: </strong>Second-line FOLFOX-bevacizumab treatment is effective in metastatic colorectal cancer (mCRC) treatment after irinotecan-based chemotherapy failure. First- or second-line raltitrexed-oxaliplatin (TOMOX) treatment has an acceptable toxicity profile in mCRC. The aim of study was to evaluated TOMOX- bevacizumab combination as a second-line treatment.</p><p><strong>Methods: </strong>The BEVATOMOX study was a non-comparative, prospective, randomized, open-label, multicentric phase II trial. Patients with histologically proven unresectable mCRC and progressive metastatic disease after irinotecan-based chemotherapy were randomized (1:2) to receive mFOLFOX6-bevacizumab (control arm, bevacizumab 5 mg/kg, mFOLFOX6 D1 = D15, 12 cycles) or TOMOX-bevacizumab (experimental arm, bevacizumab 7.5 mg/kg, raltitrexed 3 mg/m2 based on creatinine clearance, oxaliplatin 130 mg/m2, D1 = D21, 8 cycles). The primary endpoint was six-month progression-free survival (6PFS). Target enrollment was 30 and 62 patients in the control and experimental arms, respectively.</p><p><strong>Results: </strong>Eighty-three patients (median age 66 (48-82) years, 63.9% male, 54.2% KRAS mt) were included: 33 in the control and 50 in the experimental arms. The 6PFS rate and median overall survival were 51.5% (95%CI 36-67) and 11.1 months (9.5-16.4) in the control arm vs. 38% (95%CI 26-51) and 9.3 (5.7-11.6) months in the experimental arm. In the experimental arm, left-sided tumors had a longer overall survival vs. right-sided (11.6 vs. 4.6 months, p < 0.001). Grade 3-4 toxicities (mucositis, neutropenia, febrile neutropenia, paresthesia, hand-foot syndrome) were similar between arms.</p><p><strong>Conclusion: </strong>The TOMOX-Bev combination is a feasible second-line mCRC treatment with an acceptable toxicity profile. Recruitment failure prevents efficacy conclusions, but TOMOX-Bev may be an alternative if fluropyrimidines are contraindicated.</p><p><strong>Trial registration number: </strong>ClinicalTrials.gov, NCT01532804.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971599","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Paola Di Nardo, Marco de Scordilli, Fabiola Giudici, Debora Basile, Brenno Pastò, Simone Rota, Sara Torresan, Martina Bortolot, Luisa Foltran, Michela Guardascione, Arianna Fumagalli, Claudia Noto, Elena Ongaro, Angela Buonadonna, Fabio Puglisi
Background: The optimal first-line treatment for RASwild-type metastatic colorectal cancer remains undetermined. Several studies have compared the efficacy of different first-line regimens, including doublet- or triplet-chemotherapy(CT) alone or in combination with targeted therapies (anti-EGFR/anti-VEGF), without conclusive results.
Methods: We conducted a systematic review and meta-analysis of phase II/III randomized clinical trials(RCT) comparing triplet-CT+anti-EGFRs with alternative first-line regimens in RASwild-type patients. Pairwise- and network-meta-analyses were performed to assess ORR. Furthermore, we evaluated PFS and OS with pairwise-metanalyses.
Results: A total of 1,283 patients across seven RCT were included. Four treatment arms were analyzed: Arm A (triplet-CT+anti-EGFR), Arm B (doublet-CT+anti-EGFR), Arm C (triplet alone), and Arm D (triplet+anti-VEGF). Arms A, B e D demonstrated higher ORR compared to Arm C, while no significant differences were found among Arms A, B, and D (OR 1.05, 95% CI 0.73-1.49; p = 0.804, for Arm B in comparison to Arm A; OR 0.80, 95% CI 0.52-1.25; p = 0.328, for Arm D in comparison to Arm A). Pairwise-meta-analysis revealed significantly lower ORR for Arm C compared to Arm A (OR 4.23,95% CI 2.06-8.68, p = 0.002). P-scores ranked Arm B highest for effectiveness (0.808), followed by Arm A (0.746), then Arm D (0.444) and lastly Arm C (0.002) The pooled HR for OS demonstrated a superiority for arm A (0.82, 95% CI 0.70-0.97, p = 0.022).
Conclusions: Triplet-CT+anti-EGFR demonstrated no clear ORR advantage over other targeted regimens but was superior to triplet-CT alone. Preliminary data indicate a potential OS benefit. Due to increased toxicity, routine use of triplet-CT+anti-EGFR should be carefully evaluated.
背景:ras野生型转移性结直肠癌的最佳一线治疗方案仍未确定。一些研究比较了不同一线方案的疗效,包括单用双重或三重化疗(CT)或联合靶向治疗(抗egfr /抗vegf),但没有结论性结果。方法:我们对II/III期随机临床试验(RCT)进行了系统回顾和荟萃分析,比较了raswild型患者的三重ct +抗egfr与其他一线方案。进行两两和网络meta分析来评估ORR。此外,我们用双元分析评估PFS和OS。结果:7项随机对照试验共纳入1283例患者。分析了四个治疗组:A组(三重联体- ct +抗egfr), B组(双重联体- ct +抗egfr), C组(单独三重联体)和D组(三重联体+抗vegf)。与C组相比,A组、B组和D组的ORR更高,而A组、B组和D组之间无显著差异(OR为1.05,95% CI 0.73-1.49; p = 0.804, B组与A组相比;OR为0.80,95% CI 0.52-1.25; p = 0.328, D组与A组相比)。双元分析显示,与A组相比,C组的ORR显著降低(OR 4.23,95% CI 2.06-8.68, p = 0.002)。p -评分显示,B组疗效最高(0.808),其次是A组(0.746),然后是D组(0.444),最后是C组(0.002)。A组OS的综合HR显示出优势(0.82,95% CI 0.70-0.97, p = 0.022)。结论:与其他靶向治疗方案相比,Triplet-CT+抗egfr没有明显的ORR优势,但优于单独使用Triplet-CT。初步数据表明潜在的操作系统优势。由于毒性增加,应仔细评估常规使用三重ct +抗egfr。
{"title":"Triplet CT combined with anti-EGFR treatment in RAS wild-type colorectal cancer; a network metanalysis.","authors":"Paola Di Nardo, Marco de Scordilli, Fabiola Giudici, Debora Basile, Brenno Pastò, Simone Rota, Sara Torresan, Martina Bortolot, Luisa Foltran, Michela Guardascione, Arianna Fumagalli, Claudia Noto, Elena Ongaro, Angela Buonadonna, Fabio Puglisi","doi":"10.1093/oncolo/oyag011","DOIUrl":"https://doi.org/10.1093/oncolo/oyag011","url":null,"abstract":"<p><strong>Background: </strong>The optimal first-line treatment for RASwild-type metastatic colorectal cancer remains undetermined. Several studies have compared the efficacy of different first-line regimens, including doublet- or triplet-chemotherapy(CT) alone or in combination with targeted therapies (anti-EGFR/anti-VEGF), without conclusive results.</p><p><strong>Methods: </strong>We conducted a systematic review and meta-analysis of phase II/III randomized clinical trials(RCT) comparing triplet-CT+anti-EGFRs with alternative first-line regimens in RASwild-type patients. Pairwise- and network-meta-analyses were performed to assess ORR. Furthermore, we evaluated PFS and OS with pairwise-metanalyses.</p><p><strong>Results: </strong>A total of 1,283 patients across seven RCT were included. Four treatment arms were analyzed: Arm A (triplet-CT+anti-EGFR), Arm B (doublet-CT+anti-EGFR), Arm C (triplet alone), and Arm D (triplet+anti-VEGF). Arms A, B e D demonstrated higher ORR compared to Arm C, while no significant differences were found among Arms A, B, and D (OR 1.05, 95% CI 0.73-1.49; p = 0.804, for Arm B in comparison to Arm A; OR 0.80, 95% CI 0.52-1.25; p = 0.328, for Arm D in comparison to Arm A). Pairwise-meta-analysis revealed significantly lower ORR for Arm C compared to Arm A (OR 4.23,95% CI 2.06-8.68, p = 0.002). P-scores ranked Arm B highest for effectiveness (0.808), followed by Arm A (0.746), then Arm D (0.444) and lastly Arm C (0.002) The pooled HR for OS demonstrated a superiority for arm A (0.82, 95% CI 0.70-0.97, p = 0.022).</p><p><strong>Conclusions: </strong>Triplet-CT+anti-EGFR demonstrated no clear ORR advantage over other targeted regimens but was superior to triplet-CT alone. Preliminary data indicate a potential OS benefit. Due to increased toxicity, routine use of triplet-CT+anti-EGFR should be carefully evaluated.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145971051","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The age-specific distribution of frailty and its domain-level characteristics remain poorly understood across the adult population of patients with cancer. We aimed to quantify the frailty prevalence and geriatric assessment (GA) impairment patterns in adults and to examine their prognostic relevance in newly diagnosed patients with cancer.
Material and methods: This multicenter, cross-sectional cohort enrolled 2,501 adults (≥20 years) before therapy in 2021-2023. GA covered eight domains (function, comorbidity, cognition, mood, nutrition, polypharmacy, falls, and social support). Patients were grouped into six age bands (20-39 to ≥ 80) and labeled fit (0 deficits), prefrail (1), or frail (≥2). We analyzed age-specific geriatric impairment patterns and overall survival (OS).
Results: The mean number of GA deficits increased significantly across the six ordered age bands. Frailty was common and increased with age: 40.0% (20-39), 42.3% (40-49), 56.2% (70-79), and 74.4% (≥80). Malnutrition was the most frequent deficit (59.1% overall), affecting 51% of patients aged 20-39 years, peaking at 63.8% in the 70-79-year cohort. Older age groups showed steeper increases in comorbidities, cognitive impairment, and functional decline. Polypharmacy and depressed mood were frequent, but varied less with age; inadequate social support was uniformly low. In multivariable models, prefrailty and frailty predicted worse OS. Age-stratified analyses of 40-49, 50-59, and 70-79-year cohorts showed similar associations.
Conclusion: Frailty is prevalent across adults of all ages with distinct, age-associated GA profiles. Nutritional deficits were the most prevalent impairment even among younger adults, whereas functional, comorbidity, and cognitive burdens escalate in older patients with cancer. Routine pretreatment GA for all adults can identify vulnerabilities and enable age-tailored supportive interventions.
{"title":"High Burden of Geriatric Assessment Impairments Across the Adult Age Spectrum in Patients with Cancer.","authors":"Chin-Tung Nien, Chieh-Ying Chang, Chang-Hsien Lu, Kun-Yun Yeh, Yu-Shin Hung, Wen-Chi Chou","doi":"10.1093/oncolo/oyag010","DOIUrl":"https://doi.org/10.1093/oncolo/oyag010","url":null,"abstract":"<p><strong>Background: </strong>The age-specific distribution of frailty and its domain-level characteristics remain poorly understood across the adult population of patients with cancer. We aimed to quantify the frailty prevalence and geriatric assessment (GA) impairment patterns in adults and to examine their prognostic relevance in newly diagnosed patients with cancer.</p><p><strong>Material and methods: </strong>This multicenter, cross-sectional cohort enrolled 2,501 adults (≥20 years) before therapy in 2021-2023. GA covered eight domains (function, comorbidity, cognition, mood, nutrition, polypharmacy, falls, and social support). Patients were grouped into six age bands (20-39 to ≥ 80) and labeled fit (0 deficits), prefrail (1), or frail (≥2). We analyzed age-specific geriatric impairment patterns and overall survival (OS).</p><p><strong>Results: </strong>The mean number of GA deficits increased significantly across the six ordered age bands. Frailty was common and increased with age: 40.0% (20-39), 42.3% (40-49), 56.2% (70-79), and 74.4% (≥80). Malnutrition was the most frequent deficit (59.1% overall), affecting 51% of patients aged 20-39 years, peaking at 63.8% in the 70-79-year cohort. Older age groups showed steeper increases in comorbidities, cognitive impairment, and functional decline. Polypharmacy and depressed mood were frequent, but varied less with age; inadequate social support was uniformly low. In multivariable models, prefrailty and frailty predicted worse OS. Age-stratified analyses of 40-49, 50-59, and 70-79-year cohorts showed similar associations.</p><p><strong>Conclusion: </strong>Frailty is prevalent across adults of all ages with distinct, age-associated GA profiles. Nutritional deficits were the most prevalent impairment even among younger adults, whereas functional, comorbidity, and cognitive burdens escalate in older patients with cancer. Routine pretreatment GA for all adults can identify vulnerabilities and enable age-tailored supportive interventions.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145960580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dara Bracken-Clarke, Danielle M Pastor, Jennifer L Marté, Renee N Donahue, James W Hodge, Lisa Cordes, Charalampos S Floudas, Nicholas P Tschernia, Fatima Karzai, Isaac Brownell, Evrim B Turkbey, Patrick Soon-Shiong, Jeffrey Schlom, James L Gulley, Hoyoung M Maeng, Julius Strauss, Jason M Redman
Background: Despite successes in other malignancies, immune checkpoint blockade (ICB) has not demonstrated reproducible efficacy in mismatch repair-proficient (pMMR) colorectal cancer (CRC). Preclinical studies indicate that multitargeted combination immunotherapy may sensitize resistant tumors to immune-mediated killing. This trial was designed to test this hypothesis by combining agents to induce and augment an immune response in pMMR CRC.
Methods: In this single-center, phase 1/2 trial, eligible patients had pMMR, RECIST v1.1-measurable metastatic CRC, ≥2 prior lines of therapy and no prior immunotherapy. The primary endpoint was objective response rate. Patients received triplet therapy: CV-301 (CEA/MUC-1 vaccine), N-803 (IL-15 receptor superagonist), bintrafusp alfa (bifunctional anti-PD-L1/TGFβ "trap") or quadruplet therapy (adding PDS01ADC [tumor-targeted IL-12 immunocytokine]).
Results: Between September 2020 and September 2022, 32 patients enrolled to triplet (n = 12) and quadruplet (n = 20) therapy. The combinations had toxicity profiles consistent with each individual agent included. One complete response durable for 29+ months occurred in a patient with RAS wild-type CRC metastatic to lymph nodes who received quadruplet therapy.
Conclusion: Quadruplet therapy produced one objective response in unselected mCRC patients (n = 20). There were no responses with triplet therapy (n = 12). The role of ICB-based combination immunotherapy in pMMR CRC remains unclear. NCT04491955.
{"title":"The Quadruple Immunotherapy for Colorectal Cancer (QuICC) Trial for Mismatch Repair-Proficient Metastatic Colorectal Cancer.","authors":"Dara Bracken-Clarke, Danielle M Pastor, Jennifer L Marté, Renee N Donahue, James W Hodge, Lisa Cordes, Charalampos S Floudas, Nicholas P Tschernia, Fatima Karzai, Isaac Brownell, Evrim B Turkbey, Patrick Soon-Shiong, Jeffrey Schlom, James L Gulley, Hoyoung M Maeng, Julius Strauss, Jason M Redman","doi":"10.1093/oncolo/oyag008","DOIUrl":"https://doi.org/10.1093/oncolo/oyag008","url":null,"abstract":"<p><strong>Background: </strong>Despite successes in other malignancies, immune checkpoint blockade (ICB) has not demonstrated reproducible efficacy in mismatch repair-proficient (pMMR) colorectal cancer (CRC). Preclinical studies indicate that multitargeted combination immunotherapy may sensitize resistant tumors to immune-mediated killing. This trial was designed to test this hypothesis by combining agents to induce and augment an immune response in pMMR CRC.</p><p><strong>Methods: </strong>In this single-center, phase 1/2 trial, eligible patients had pMMR, RECIST v1.1-measurable metastatic CRC, ≥2 prior lines of therapy and no prior immunotherapy. The primary endpoint was objective response rate. Patients received triplet therapy: CV-301 (CEA/MUC-1 vaccine), N-803 (IL-15 receptor superagonist), bintrafusp alfa (bifunctional anti-PD-L1/TGFβ \"trap\") or quadruplet therapy (adding PDS01ADC [tumor-targeted IL-12 immunocytokine]).</p><p><strong>Results: </strong>Between September 2020 and September 2022, 32 patients enrolled to triplet (n = 12) and quadruplet (n = 20) therapy. The combinations had toxicity profiles consistent with each individual agent included. One complete response durable for 29+ months occurred in a patient with RAS wild-type CRC metastatic to lymph nodes who received quadruplet therapy.</p><p><strong>Conclusion: </strong>Quadruplet therapy produced one objective response in unselected mCRC patients (n = 20). There were no responses with triplet therapy (n = 12). The role of ICB-based combination immunotherapy in pMMR CRC remains unclear. NCT04491955.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rosalba Torrisi, Federica Giugliano, Laura Giordano, Giuseppe Saltalamacchia, Flavia Jacobs, Ambra Carnevale Schianca, Monica Milano, Nadia Bianco, Claudia Anna Sangalli, Rita De Sanctis, Giovanna Masci, Giuseppe Curigliano, Armando Santoro, Elisabetta Munzone
Patients and methods: we retrospectively collected data of patients with HR+/HER2- advanced breast cancer (ABC) treated with endocrine therapy (ET) and a CDK4/6 inhibitor (CDK4/6i) aiming to describe the patterns of post-progression outcomes.
Results: Among 452 evaluable patients 325 were treated in the first-line setting. Median progression free-survival (mPFS) was 22.8 months overall and 29.7 months in patients treated in first-line setting. Factors associated with outcomes in multivariate analysis were the line of CDK4/6i therapy, de novo vs recurrent disease, visceral vs bone-only metastases, and primary endocrine resistance.A total of 300 patients progressed and 250 overall and 156 in the first-line cohort received a subsequent treatment. Visceral progression and CDK4/6i duration <12 months were associated with a higher likelihood of receiving anthracycline or taxanes (AT) as compared to ET ±everolimus (EET). Post-progression PFS (PPFS) and post-progression OS (PPOS) were statistically significantly better with EET and capecitabine (C) arms over AT overall and in patients with visceral progression. Multivariate analysis confirmed a significant advantage for EET and C, while visceral progression retained a significant impact only on PPOS. After progression to the 1st post-CDK4/6i treatment C obtained a significant better PPOS as compared to other treatments.
Conclusion: we showed in a large real-world series that most patients with HR+/HER2- ABC failing CDK4/6i and ET unselected for the occurrence of molecular mutations retain endocrine sensitivity and may benefit of a subsequent ET ± a targeted therapy delaying the need for chemotherapy regardless of site of progression and prior CDK4/6i therapy duration.
{"title":"Real-world patterns of post-progression treatment and outcomes in patients with HR+/HER2- advanced breast cancer treated with CDK4/6 inhibitors.","authors":"Rosalba Torrisi, Federica Giugliano, Laura Giordano, Giuseppe Saltalamacchia, Flavia Jacobs, Ambra Carnevale Schianca, Monica Milano, Nadia Bianco, Claudia Anna Sangalli, Rita De Sanctis, Giovanna Masci, Giuseppe Curigliano, Armando Santoro, Elisabetta Munzone","doi":"10.1093/oncolo/oyag003","DOIUrl":"https://doi.org/10.1093/oncolo/oyag003","url":null,"abstract":"<p><strong>Patients and methods: </strong>we retrospectively collected data of patients with HR+/HER2- advanced breast cancer (ABC) treated with endocrine therapy (ET) and a CDK4/6 inhibitor (CDK4/6i) aiming to describe the patterns of post-progression outcomes.</p><p><strong>Results: </strong>Among 452 evaluable patients 325 were treated in the first-line setting. Median progression free-survival (mPFS) was 22.8 months overall and 29.7 months in patients treated in first-line setting. Factors associated with outcomes in multivariate analysis were the line of CDK4/6i therapy, de novo vs recurrent disease, visceral vs bone-only metastases, and primary endocrine resistance.A total of 300 patients progressed and 250 overall and 156 in the first-line cohort received a subsequent treatment. Visceral progression and CDK4/6i duration <12 months were associated with a higher likelihood of receiving anthracycline or taxanes (AT) as compared to ET ±everolimus (EET). Post-progression PFS (PPFS) and post-progression OS (PPOS) were statistically significantly better with EET and capecitabine (C) arms over AT overall and in patients with visceral progression. Multivariate analysis confirmed a significant advantage for EET and C, while visceral progression retained a significant impact only on PPOS. After progression to the 1st post-CDK4/6i treatment C obtained a significant better PPOS as compared to other treatments.</p><p><strong>Conclusion: </strong>we showed in a large real-world series that most patients with HR+/HER2- ABC failing CDK4/6i and ET unselected for the occurrence of molecular mutations retain endocrine sensitivity and may benefit of a subsequent ET ± a targeted therapy delaying the need for chemotherapy regardless of site of progression and prior CDK4/6i therapy duration.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949085","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Biliary tract cancers (BTCs), including gallbladder cancer (GBC) and cholangiocarcinoma (CCA), are rare but aggressive malignancies with distinct molecular landscapes and poor prognosis. Genomic profiling has revealed significant molecular alterations, but the genomic landscape of BTC in the Indian population remains underexplored. This study aims to comprehensively characterize the mutation landscape of BTC in the Indian population.
Methods: A total of 154 BTC cases, including 69 CCA and 85 GBC, were retrospectively analyzed using data collected from various targeted sequencing panels. Somatic mutations, copy number variations (CNVs), and gene fusions in key oncogenic and tumor suppressor genes were identified from these panel reports. Downstream analyses were performed to derive key biological insights, including pathway enrichment and mutual exclusivity and co-occurrence analyses of genomic alterations.
Results: TP53 was the most frequently mutated gene (53%), followed by KRAS (18%), ARID1A (9%), IDH1 (7%), and PIK3CA (7%). Recurrent amplifications were observed in MYC (12%) and ERBB2 (9%). Pathway enrichment analysis revealed significant dysregulation in the PI3K-AKT-mTOR, Notch, and Wnt/β-catenin signaling pathways. Notably, IDH1 mutations were primarily observed in CCA, while STK11 mutations were exclusive to GBC, highlighting distinct molecular characteristics between the two subtypes. PD-L1-negative tumors exhibited distinct genomic alterations, notably SMAD4 mutations, which were associated with reduced PD-L1 expression. This loss of SMAD4, involved in TGF-β signaling, could impair immune response regulation and facilitate immune evasion.
Conclusions: This study provides a comprehensive molecular profiling of BTCs in the Indian population, revealing key genomic alterations, subtype-specific differences, and associations with immune features. The findings underscore the importance of molecular profiling in guiding personalized treatment strategies.
{"title":"Actionable Genomic Landscape of Biliary Tract Cancer in the Indian Population.","authors":"Sewanti Limaye, Aditya Shreenivas, Darshana Patil, Soumil Vyas, Irene A George, Janani Sambath, Shambhavi Singh, Chetan Madre, Anjali Parab, Pritam Kataria, Darshit Shah, Niyati Shah, Shaheenah Dawood, Nitesh Rohatgi, Ruturaj Deshpande, Aakriti Datta, Humaid Al Shamsi, Andrew Gaya, Ashok Kumar Vaid, Shriniwas Kulkarni, Senthil Rajappa, Damian Rieke, Prashant Kumar, Rajan Datar, Milind Javle","doi":"10.1093/oncolo/oyaf430","DOIUrl":"https://doi.org/10.1093/oncolo/oyaf430","url":null,"abstract":"<p><strong>Background: </strong>Biliary tract cancers (BTCs), including gallbladder cancer (GBC) and cholangiocarcinoma (CCA), are rare but aggressive malignancies with distinct molecular landscapes and poor prognosis. Genomic profiling has revealed significant molecular alterations, but the genomic landscape of BTC in the Indian population remains underexplored. This study aims to comprehensively characterize the mutation landscape of BTC in the Indian population.</p><p><strong>Methods: </strong>A total of 154 BTC cases, including 69 CCA and 85 GBC, were retrospectively analyzed using data collected from various targeted sequencing panels. Somatic mutations, copy number variations (CNVs), and gene fusions in key oncogenic and tumor suppressor genes were identified from these panel reports. Downstream analyses were performed to derive key biological insights, including pathway enrichment and mutual exclusivity and co-occurrence analyses of genomic alterations.</p><p><strong>Results: </strong>TP53 was the most frequently mutated gene (53%), followed by KRAS (18%), ARID1A (9%), IDH1 (7%), and PIK3CA (7%). Recurrent amplifications were observed in MYC (12%) and ERBB2 (9%). Pathway enrichment analysis revealed significant dysregulation in the PI3K-AKT-mTOR, Notch, and Wnt/β-catenin signaling pathways. Notably, IDH1 mutations were primarily observed in CCA, while STK11 mutations were exclusive to GBC, highlighting distinct molecular characteristics between the two subtypes. PD-L1-negative tumors exhibited distinct genomic alterations, notably SMAD4 mutations, which were associated with reduced PD-L1 expression. This loss of SMAD4, involved in TGF-β signaling, could impair immune response regulation and facilitate immune evasion.</p><p><strong>Conclusions: </strong>This study provides a comprehensive molecular profiling of BTCs in the Indian population, revealing key genomic alterations, subtype-specific differences, and associations with immune features. The findings underscore the importance of molecular profiling in guiding personalized treatment strategies.</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949101","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiaojun Liu, Yanchun Meng, Yiqun Du, Yuxin Mu, Gang Li, Hengyu Li, Xiaoxiang Guan, Jian Zhang
Background: Chemotherapy-induced nausea and vomiting (CINV) significantly impact the patients' quality of life. Whether the granisetron transdermal delivery system (GTDS) offers better protection than palonosetron against long-delayed (120-168 h) CINV after highly or moderately emetogenic chemotherapy (HEC/MEC) had not been prospectively tested.
Methods: A multicenter, randomized clinical study was conducted in China. Patients scheduled to receive either HEC or MEC were randomly assigned (1:1) to GTDS or palonosetron, each in combination with a neurokinin-1 receptor antagonist (NK1-RA) and dexamethasone. The primary endpoint was the complete response (CR; no vomiting and no rescue medication) rate during the long-delayed phase (120-168 hours), stratified by HEC and MEC categories, to demonstrate the superiority of GTDS over palonosetron.
Results: Overall, 150 patients received either GTDS or palonosetron respectively. We found that the GTDS group demonstrated a significantly higher long-delayed CR rate (97.3%) than the palonosetron group (92%) (p = 0.04). This advantage was driven predominantly by the HEC subgroup (GTDS 97.5% vs palonosetron 90.8%, p = 0.028). No significant differences were observed between the groups for the acute (0-24 h, 92.7% vs 90.0%; p = 0.412), delayed (24-120 h, 80.0% vs 76.0%; p = 0.403), extended-delayed (24-168 h, 80.7% vs 75.3%; p = 0.265), or overall (0-168 h, 78.0% vs 74.0%; p = 0.417) phases.
Conclusion: A GTDS-based triple antiemetic regimen can effectively control CINV associated with HEC or MEC. It provides a convenient alternative route for delivering granisetron for up to 7 days, with superior efficacy in controlling long-delayed CINV.
背景:化疗引起的恶心和呕吐(CINV)严重影响患者的生活质量。格拉司琼透皮给药系统(GTDS)是否比帕洛诺司琼更好地保护高或中度致吐性化疗(HEC/MEC)后的长延迟(120-168 h) CINV,目前尚未进行前瞻性试验。方法:在中国进行一项多中心随机临床研究。计划接受HEC或MEC的患者被随机(1:1)分配到GTDS或帕洛诺司琼组,每组均联合神经球蛋白-1受体拮抗剂(NK1-RA)和地塞米松。主要终点是长延迟期(120-168小时)的完全缓解(CR,无呕吐和无抢救用药)率,按HEC和MEC分类分层,以证明GTDS优于帕洛诺司琼。结果:总体而言,150例患者分别接受了GTDS或帕洛诺司琼治疗。我们发现GTDS组的长延迟CR率(97.3%)明显高于帕洛诺司琼组(92%)(p = 0.04)。这一优势主要由HEC亚组驱动(GTDS 97.5% vs帕洛诺司琼90.8%,p = 0.028)。急性期(0-24 h, 92.7% vs 90.0%, p = 0.412)、延迟期(24-120 h, 80.0% vs 76.0%, p = 0.403)、延长-延迟期(24-168 h, 80.7% vs 75.3%, p = 0.265)或总期(0-168 h, 78.0% vs 74.0%, p = 0.417)组间无显著差异。结论:以gtds为基础的三联止吐方案可有效控制伴有HEC或MEC的CINV。它提供了一种方便的替代途径给药格拉司琼长达7天,在控制长延迟CINV方面具有优越的疗效。试验注册:clinicaltrials.gov标识符:NCT04912271(内部伦理号:YBCSG-21-04)。
{"title":"Granisetron Transdermal Delivery System Versus Palonosetron in the Prevention of Long-delayed Nausea and Vomiting: A Phase III Randomized Trial.","authors":"Xiaojun Liu, Yanchun Meng, Yiqun Du, Yuxin Mu, Gang Li, Hengyu Li, Xiaoxiang Guan, Jian Zhang","doi":"10.1093/oncolo/oyag007","DOIUrl":"https://doi.org/10.1093/oncolo/oyag007","url":null,"abstract":"<p><strong>Background: </strong>Chemotherapy-induced nausea and vomiting (CINV) significantly impact the patients' quality of life. Whether the granisetron transdermal delivery system (GTDS) offers better protection than palonosetron against long-delayed (120-168 h) CINV after highly or moderately emetogenic chemotherapy (HEC/MEC) had not been prospectively tested.</p><p><strong>Methods: </strong>A multicenter, randomized clinical study was conducted in China. Patients scheduled to receive either HEC or MEC were randomly assigned (1:1) to GTDS or palonosetron, each in combination with a neurokinin-1 receptor antagonist (NK1-RA) and dexamethasone. The primary endpoint was the complete response (CR; no vomiting and no rescue medication) rate during the long-delayed phase (120-168 hours), stratified by HEC and MEC categories, to demonstrate the superiority of GTDS over palonosetron.</p><p><strong>Results: </strong>Overall, 150 patients received either GTDS or palonosetron respectively. We found that the GTDS group demonstrated a significantly higher long-delayed CR rate (97.3%) than the palonosetron group (92%) (p = 0.04). This advantage was driven predominantly by the HEC subgroup (GTDS 97.5% vs palonosetron 90.8%, p = 0.028). No significant differences were observed between the groups for the acute (0-24 h, 92.7% vs 90.0%; p = 0.412), delayed (24-120 h, 80.0% vs 76.0%; p = 0.403), extended-delayed (24-168 h, 80.7% vs 75.3%; p = 0.265), or overall (0-168 h, 78.0% vs 74.0%; p = 0.417) phases.</p><p><strong>Conclusion: </strong>A GTDS-based triple antiemetic regimen can effectively control CINV associated with HEC or MEC. It provides a convenient alternative route for delivering granisetron for up to 7 days, with superior efficacy in controlling long-delayed CINV.</p><p><strong>Trial registration: clinicaltrials.gov identifier: </strong>NCT04912271 (in-house ethic number: YBCSG-21-04).</p>","PeriodicalId":54686,"journal":{"name":"Oncologist","volume":" ","pages":""},"PeriodicalIF":4.2,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949142","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}