Pub Date : 2025-12-28DOI: 10.1007/s11523-025-01193-x
Yeokyeong Shin, Jinho Shin, Hyehyun Jeong, Baek-Yeol Ryoo, Kyu-Pyo Kim, Inkeun Park, Dong-Wan Seo, Do Hyun Park, Tae Jun Song, Dongwook Oh, Dae Wook Hwang, Jae Hoon Lee, Ki Byung Song, Song Cheol Kim, Seung-Mo Hong, Changhoon Yoo
Background: Biliary tract cancers (BTCs), including cholangiocarcinoma and gallbladder carcinoma, are aggressive tumors with limited treatment options and poor prognosis. Antibody-drug conjugates (ADCs) targeting membrane proteins, such as claudin-18.2 (CLDN18.2) and trophoblast cell surface antigen 2 (Trop-2), are promising targets for other epithelial cancers. However, their expression patterns and clinical relevance in BTC remain unclear.
Objective: This study aimed to evaluate CLDN18 and Trop-2 expression levels in BTC and their potential as therapeutic targets.
Patients and methods: We retrospectively analyzed 636 patients with BTC who underwent surgical resection at Asan Medical Center between February 1998 and October 2020. Immunohistochemistry was performed using validated antibodies. CLDN18 positivity was defined as membranous staining in ≥ 75% of tumor cells with a moderate-to-strong intensity. Trop-2 expression was quantified by H-score and categorized as low (< 100), medium (100-200), or high (> 200). Associations with clinicopathologic features and survival outcomes were assessed via Kaplan-Meier and Cox regression.
Results: CLDN18 was positive for 5.2% of patients, most frequently in perihilar cholangiocarcinoma (pCCA, 22.2%). Trop-2 high and medium expression were observed in 69.5% and 20.1% of patients, respectively; its expression was low in intrahepatic cholangiocarcinoma (iCCA, 27.3%). CLDN18 expression showed no adverse clinicopathologic features. High expression of Trop-2 was linked to higher T category and more frequent lymphovascular and perineural invasion. Neither biomarker was significantly associated with overall survival (OS) or disease-free survival (DFS).
Conclusions: CLDN18 was highly expressed in specific BTC subtypes, particularly perihilar cholangiocarcinoma (pCCA) and gallbladder cancer, while Trop-2 was broadly expressed. These findings supported the potential of CLDN18.2- and Trop-2-directed therapies in BTC.
{"title":"Claudin-18.2 and Trop-2 as Emerging Biomarkers in Biliary Tract Cancers: Expression Analysis and Therapeutic Potential.","authors":"Yeokyeong Shin, Jinho Shin, Hyehyun Jeong, Baek-Yeol Ryoo, Kyu-Pyo Kim, Inkeun Park, Dong-Wan Seo, Do Hyun Park, Tae Jun Song, Dongwook Oh, Dae Wook Hwang, Jae Hoon Lee, Ki Byung Song, Song Cheol Kim, Seung-Mo Hong, Changhoon Yoo","doi":"10.1007/s11523-025-01193-x","DOIUrl":"https://doi.org/10.1007/s11523-025-01193-x","url":null,"abstract":"<p><strong>Background: </strong>Biliary tract cancers (BTCs), including cholangiocarcinoma and gallbladder carcinoma, are aggressive tumors with limited treatment options and poor prognosis. Antibody-drug conjugates (ADCs) targeting membrane proteins, such as claudin-18.2 (CLDN18.2) and trophoblast cell surface antigen 2 (Trop-2), are promising targets for other epithelial cancers. However, their expression patterns and clinical relevance in BTC remain unclear.</p><p><strong>Objective: </strong>This study aimed to evaluate CLDN18 and Trop-2 expression levels in BTC and their potential as therapeutic targets.</p><p><strong>Patients and methods: </strong>We retrospectively analyzed 636 patients with BTC who underwent surgical resection at Asan Medical Center between February 1998 and October 2020. Immunohistochemistry was performed using validated antibodies. CLDN18 positivity was defined as membranous staining in ≥ 75% of tumor cells with a moderate-to-strong intensity. Trop-2 expression was quantified by H-score and categorized as low (< 100), medium (100-200), or high (> 200). Associations with clinicopathologic features and survival outcomes were assessed via Kaplan-Meier and Cox regression.</p><p><strong>Results: </strong>CLDN18 was positive for 5.2% of patients, most frequently in perihilar cholangiocarcinoma (pCCA, 22.2%). Trop-2 high and medium expression were observed in 69.5% and 20.1% of patients, respectively; its expression was low in intrahepatic cholangiocarcinoma (iCCA, 27.3%). CLDN18 expression showed no adverse clinicopathologic features. High expression of Trop-2 was linked to higher T category and more frequent lymphovascular and perineural invasion. Neither biomarker was significantly associated with overall survival (OS) or disease-free survival (DFS).</p><p><strong>Conclusions: </strong>CLDN18 was highly expressed in specific BTC subtypes, particularly perihilar cholangiocarcinoma (pCCA) and gallbladder cancer, while Trop-2 was broadly expressed. These findings supported the potential of CLDN18.2- and Trop-2-directed therapies in BTC.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145850981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1007/s11523-025-01186-w
Alba Rubio-San-Simón, Lynley V Marshall, Francois Doz, Jaume Mora, Kevin Bielamowicz, Nadege Corradini, Anne-Marie Langevin, Aru Narendran, Amy A Smith, C Michel Zwaan, Deborah Gho, Alison Cardenas, Stephen Fowler, Cecile Guizani, Vanessa Breton, Beate Wulff, Ronald Bernardi, Tanya Trippett, Quentin Campbell-Hewson
Background: MDM2 regulates the P53 pathway and is a promising therapeutic target, particularly in pediatric cancers with wild-type (WT) TP53. Idasanutlin is an investigational MDM2 inhibitor that is highly selective and orally bioavailable. The combination of MDM2 inhibition with cytotoxic chemotherapy or Bcl-2 inhibition has been shown to improve activity in preclinical models.
Objective: iMATRIX idasa was designed to assess the safety, pharmacokinetics, and antitumor activity of idasanutlin in children and young adults with relapsed/refractory solid tumors.
Patients and methods: This multicenter phase I/II study enrolled patients aged < 30 years with extracranial solid tumors. Patients received idasanutlin on days 1-5 of a 28-day cycle as a single agent or in combination with venetoclax or chemotherapy. The single-agent dose escalation utilized a modified continual reassessment method. The primary endpoints included the safety and determination of the maximum tolerated dose, characterization of the pharmacokinetics, and preliminary efficacy.
Results: Of 38 patients (median age 9 years [range: 2-23]), 26 with solid tumors received idasanutlin alone, and 12 with neuroblastoma received it in combination: six with venetoclax and six with chemotherapy (cyclophosphamide/topotecan). In total, 5 of 26 patients (19.2%) treated with single-agent idasanutlin experienced dose-limiting toxicities (including thrombocytopenia, neutropenia, and febrile neutropenia), which established a pediatric recommended phase II dose for idasanutlin of 4.5 mg/kg/day on days 1-5 of each 28-day cycle. The most frequently reported treatment-related adverse events were thrombocytopenia and neutropenia. Tolerable exposures were similar to what has been observed in adults. The objective response rate for patients with WT TP53 neuroblastoma who were treated with idasanutlin in combination with venetoclax or chemotherapy (primary efficacy endpoint) was 11.1% (N = 9; 95% confidence intervals, CI 0.28, 48.25) with one patient having a complete response. No other patients in the overall study population had an objective response.
Conclusions: The safety profile and tolerable exposure of idasanutlin in pediatrics was similar to that reported in adults. Limited clinical activity was observed in patients with solid tumors. On the basis of the negative benefit-risk assessment, the study was terminated, and the overall pediatric development program for idasanutlin was discontinued.
Clinical trial registration: ClinicalTrials.gov NCT04029688, registered 19 July 2019.
{"title":"Idasanutlin in Combination with Chemotherapy or Venetoclax in Pediatric and Young Adult Patients with Relapsed/Refractory Solid Tumors (iMATRIX Idasa): Results of a Phase I/II, Multicenter, Multi-arm Study.","authors":"Alba Rubio-San-Simón, Lynley V Marshall, Francois Doz, Jaume Mora, Kevin Bielamowicz, Nadege Corradini, Anne-Marie Langevin, Aru Narendran, Amy A Smith, C Michel Zwaan, Deborah Gho, Alison Cardenas, Stephen Fowler, Cecile Guizani, Vanessa Breton, Beate Wulff, Ronald Bernardi, Tanya Trippett, Quentin Campbell-Hewson","doi":"10.1007/s11523-025-01186-w","DOIUrl":"https://doi.org/10.1007/s11523-025-01186-w","url":null,"abstract":"<p><strong>Background: </strong>MDM2 regulates the P53 pathway and is a promising therapeutic target, particularly in pediatric cancers with wild-type (WT) TP53. Idasanutlin is an investigational MDM2 inhibitor that is highly selective and orally bioavailable. The combination of MDM2 inhibition with cytotoxic chemotherapy or Bcl-2 inhibition has been shown to improve activity in preclinical models.</p><p><strong>Objective: </strong>iMATRIX idasa was designed to assess the safety, pharmacokinetics, and antitumor activity of idasanutlin in children and young adults with relapsed/refractory solid tumors.</p><p><strong>Patients and methods: </strong>This multicenter phase I/II study enrolled patients aged < 30 years with extracranial solid tumors. Patients received idasanutlin on days 1-5 of a 28-day cycle as a single agent or in combination with venetoclax or chemotherapy. The single-agent dose escalation utilized a modified continual reassessment method. The primary endpoints included the safety and determination of the maximum tolerated dose, characterization of the pharmacokinetics, and preliminary efficacy.</p><p><strong>Results: </strong>Of 38 patients (median age 9 years [range: 2-23]), 26 with solid tumors received idasanutlin alone, and 12 with neuroblastoma received it in combination: six with venetoclax and six with chemotherapy (cyclophosphamide/topotecan). In total, 5 of 26 patients (19.2%) treated with single-agent idasanutlin experienced dose-limiting toxicities (including thrombocytopenia, neutropenia, and febrile neutropenia), which established a pediatric recommended phase II dose for idasanutlin of 4.5 mg/kg/day on days 1-5 of each 28-day cycle. The most frequently reported treatment-related adverse events were thrombocytopenia and neutropenia. Tolerable exposures were similar to what has been observed in adults. The objective response rate for patients with WT TP53 neuroblastoma who were treated with idasanutlin in combination with venetoclax or chemotherapy (primary efficacy endpoint) was 11.1% (N = 9; 95% confidence intervals, CI 0.28, 48.25) with one patient having a complete response. No other patients in the overall study population had an objective response.</p><p><strong>Conclusions: </strong>The safety profile and tolerable exposure of idasanutlin in pediatrics was similar to that reported in adults. Limited clinical activity was observed in patients with solid tumors. On the basis of the negative benefit-risk assessment, the study was terminated, and the overall pediatric development program for idasanutlin was discontinued.</p><p><strong>Clinical trial registration: </strong>ClinicalTrials.gov NCT04029688, registered 19 July 2019.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145725536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1007/s11523-025-01191-z
Khalid Jazieh, Jill Tsai, Sheila Solomon, Mojun Zhu, Katrina S Pedersen, Martin E Fernandez-Zapico, Hao Xie
Background: KRASG12D is one of the most prevalent driver mutations in patients with pancreatic ductal adenocarcinoma (PDAC) and colorectal adenocarcinoma (CRC). Although this genetic alteration is associated with poor prognosis and resistance to chemotherapy, additional genomic features may contribute to the behavior of KRAS-mutant PDACs and CRCs.
Objective: Here, we aimed at defining the landscape of these additional genomic features of KRASG12D-mutant PDAC and CRC, and their impact on clinical outcomes.
Patients and methods: This retrospective analysis utilized circulating tumor DNA data from two cohorts with advanced CRC and PDAC: a national cohort from Guardant (n = 27,497) and a Mayo Clinic cohort (n = 1434). Patients were categorized into three groups: KRASG12D alone, KRASG12D with putative resistance alterations, and KRAS not detected (ND). Genomic co-occurrences were summarized. Overall survival (OS) was compared among groups using Kaplan-Meier and multivariable survival analysis.
Results: Among patients with KRASG12D mutation, additional oncogenic alterations were detected in 34.5% of CRC and 11.5% of PDAC in the national cohort; 38.9% of CRC, and 17.4% of PDAC in the Mayo cohort. Common additional oncogenic alterations included EGFR amplifications, additional KRAS point mutations, and alterations in NRAS, BRAF, and PIK3CA. Patients with KRASG12D and these alterations had significantly shorter median OS compared with those with KRASG12D alone and KRAS ND for CRC (p < 0.0001) and PDAC (p < 0.0001). Presence of KRASG12D and additional oncogenic alterations was the only variable significantly associated with OS outcomes in both CRC and PDAC.
Conclusions: We described the genomic landscape of KRASG12D-mutant CRC and PDAC, demonstrating that cases often have additional oncogenic alterations linked to resistance to KRAS inhibition. These alterations are also associated with a worse prognosis. Recognizing these alterations may inform new therapeutic strategies. Further studies are warranted to validate these findings in ongoing clinical trials.
{"title":"Translational Relevance of the Genomic Landscape of KRAS<sup>G12D</sup>-Mutant Colorectal and Pancreatic Cancers.","authors":"Khalid Jazieh, Jill Tsai, Sheila Solomon, Mojun Zhu, Katrina S Pedersen, Martin E Fernandez-Zapico, Hao Xie","doi":"10.1007/s11523-025-01191-z","DOIUrl":"https://doi.org/10.1007/s11523-025-01191-z","url":null,"abstract":"<p><strong>Background: </strong>KRAS<sup>G12D</sup> is one of the most prevalent driver mutations in patients with pancreatic ductal adenocarcinoma (PDAC) and colorectal adenocarcinoma (CRC). Although this genetic alteration is associated with poor prognosis and resistance to chemotherapy, additional genomic features may contribute to the behavior of KRAS-mutant PDACs and CRCs.</p><p><strong>Objective: </strong>Here, we aimed at defining the landscape of these additional genomic features of KRAS<sup>G12D</sup>-mutant PDAC and CRC, and their impact on clinical outcomes.</p><p><strong>Patients and methods: </strong>This retrospective analysis utilized circulating tumor DNA data from two cohorts with advanced CRC and PDAC: a national cohort from Guardant (n = 27,497) and a Mayo Clinic cohort (n = 1434). Patients were categorized into three groups: KRAS<sup>G12D</sup> alone, KRAS<sup>G12D</sup> with putative resistance alterations, and KRAS not detected (ND). Genomic co-occurrences were summarized. Overall survival (OS) was compared among groups using Kaplan-Meier and multivariable survival analysis.</p><p><strong>Results: </strong>Among patients with KRAS<sup>G12D</sup> mutation, additional oncogenic alterations were detected in 34.5% of CRC and 11.5% of PDAC in the national cohort; 38.9% of CRC, and 17.4% of PDAC in the Mayo cohort. Common additional oncogenic alterations included EGFR amplifications, additional KRAS point mutations, and alterations in NRAS, BRAF, and PIK3CA. Patients with KRAS<sup>G12D</sup> and these alterations had significantly shorter median OS compared with those with KRAS<sup>G12D</sup> alone and KRAS ND for CRC (p < 0.0001) and PDAC (p < 0.0001). Presence of KRAS<sup>G12D</sup> and additional oncogenic alterations was the only variable significantly associated with OS outcomes in both CRC and PDAC.</p><p><strong>Conclusions: </strong>We described the genomic landscape of KRAS<sup>G12D</sup>-mutant CRC and PDAC, demonstrating that cases often have additional oncogenic alterations linked to resistance to KRAS inhibition. These alterations are also associated with a worse prognosis. Recognizing these alterations may inform new therapeutic strategies. Further studies are warranted to validate these findings in ongoing clinical trials.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145725656","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1007/s11523-025-01190-0
Carolina Colli Cruz, Maria Julia Moura Nascimento Santos, Sharada Wali, Cristina Natha, Rachel Mortan, Rohan Ahuja, Jarrett Rong, Tanvi Gupta, Irene Jeong-Ah Lee, Varun Vemulapalli, Sean Ngo, Kei Takigawa, Krishnavathana Varatharajalu, Karen C Kim, Kathryn Bollin, Stephane Champiat, Mehnaz A Shafi, Anusha S Thomas, Yinghong Wang
Background: Immune-mediated gastroenteritis (IMG) has been commonly managed with proton pump inhibitors (PPIs), though their effectiveness is unclear. Histological analysis has shown depletion of parietal cells in the gastric mucosa during IMG, which inhibits PPI action.
Objective: Extending on a small cohort study, we assessed the role of PPIs, and alternatives such as corticosteroids, for managing IMG.
Patients and methods: This was a retrospective study at a tertiary care cancer center, including patients with malignancy who received an immune-checkpoint inhibitor (ICI) between 2010 and 2024 and developed IMG.
Results: A total of 399 patients were included, of whom 281 (70.4%) received PD-1/PD-L1 inhibitors. Of these, 190 (47.6%) had exclusive IMG, and 69 (36.3%) were treated with PPIs. PPI use did not significantly impact clinical outcomes. In contrast, 156 (39.1%) received corticosteroids, showing improved clinical outcomes (75.8% vs 65%; p = 0.027) and a trend towards faster symptom resolution (41.5 vs 53 days; p = 0.064). Endoscopic remission was achieved in 71.1% of the steroid group and 36.7% of the non-steroid group. ICI discontinuation was more frequent with steroids (73.5% vs 50.7%; p < 0.0001), as was symptom recurrence within 6 months (16.7% vs 3.6%; p < 0.0001). All-cause mortality was higher in the non-steroid group (51.5% vs 41%; p = 0.042), which had a shorter follow-up period (0.7 vs 1.1 years; p = 0.004). Binary logistic regression showed that steroid use (OR 1.7, 95% CI 1.06-2.7; p = 0.027) and ICI discontinuation (OR 1.9, 95% CI 1.1-3.0; p = 0.007) were associated with clinical improvement.
Conclusion: Our findings show faster clinical improvement and higher endoscopic remission rates with steroids, while PPIs demonstrated no significant effectiveness.
背景:免疫介导性胃肠炎(IMG)通常使用质子泵抑制剂(PPIs)治疗,尽管其有效性尚不清楚。组织学分析显示胃粘膜壁细胞在IMG过程中耗竭,这抑制了PPI的作用。目的:在一项小型队列研究的基础上,我们评估了PPIs和皮质类固醇等替代药物在管理IMG方面的作用。患者和方法:这是一项在三级保健癌症中心进行的回顾性研究,包括2010年至2024年间接受免疫检查点抑制剂(ICI)治疗并发展为IMG的恶性肿瘤患者。结果:共纳入399例患者,其中281例(70.4%)接受了PD-1/PD-L1抑制剂治疗。其中,190例(47.6%)患有排他性IMG, 69例(36.3%)接受PPIs治疗。使用PPI对临床结果没有显著影响。相比之下,156例(39.1%)接受皮质类固醇治疗,临床结果得到改善(75.8% vs 65%, p = 0.027),症状缓解趋势更快(41.5 vs 53天,p = 0.064)。内镜下缓解在71.1%的类固醇组和36.7%的非类固醇组。结论:我们的研究结果显示类固醇治疗的临床改善更快,内镜下缓解率更高,而PPIs没有明显的疗效。
{"title":"Beyond Proton Pump Inhibitors: Evaluating Treatment Strategies for Immune-Mediated Gastroenteritis from Cancer Immunotherapy.","authors":"Carolina Colli Cruz, Maria Julia Moura Nascimento Santos, Sharada Wali, Cristina Natha, Rachel Mortan, Rohan Ahuja, Jarrett Rong, Tanvi Gupta, Irene Jeong-Ah Lee, Varun Vemulapalli, Sean Ngo, Kei Takigawa, Krishnavathana Varatharajalu, Karen C Kim, Kathryn Bollin, Stephane Champiat, Mehnaz A Shafi, Anusha S Thomas, Yinghong Wang","doi":"10.1007/s11523-025-01190-0","DOIUrl":"https://doi.org/10.1007/s11523-025-01190-0","url":null,"abstract":"<p><strong>Background: </strong>Immune-mediated gastroenteritis (IMG) has been commonly managed with proton pump inhibitors (PPIs), though their effectiveness is unclear. Histological analysis has shown depletion of parietal cells in the gastric mucosa during IMG, which inhibits PPI action.</p><p><strong>Objective: </strong>Extending on a small cohort study, we assessed the role of PPIs, and alternatives such as corticosteroids, for managing IMG.</p><p><strong>Patients and methods: </strong>This was a retrospective study at a tertiary care cancer center, including patients with malignancy who received an immune-checkpoint inhibitor (ICI) between 2010 and 2024 and developed IMG.</p><p><strong>Results: </strong>A total of 399 patients were included, of whom 281 (70.4%) received PD-1/PD-L1 inhibitors. Of these, 190 (47.6%) had exclusive IMG, and 69 (36.3%) were treated with PPIs. PPI use did not significantly impact clinical outcomes. In contrast, 156 (39.1%) received corticosteroids, showing improved clinical outcomes (75.8% vs 65%; p = 0.027) and a trend towards faster symptom resolution (41.5 vs 53 days; p = 0.064). Endoscopic remission was achieved in 71.1% of the steroid group and 36.7% of the non-steroid group. ICI discontinuation was more frequent with steroids (73.5% vs 50.7%; p < 0.0001), as was symptom recurrence within 6 months (16.7% vs 3.6%; p < 0.0001). All-cause mortality was higher in the non-steroid group (51.5% vs 41%; p = 0.042), which had a shorter follow-up period (0.7 vs 1.1 years; p = 0.004). Binary logistic regression showed that steroid use (OR 1.7, 95% CI 1.06-2.7; p = 0.027) and ICI discontinuation (OR 1.9, 95% CI 1.1-3.0; p = 0.007) were associated with clinical improvement.</p><p><strong>Conclusion: </strong>Our findings show faster clinical improvement and higher endoscopic remission rates with steroids, while PPIs demonstrated no significant effectiveness.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145744555","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1007/s11523-025-01188-8
Susu Zhou, Devashish Desai, Noriko Kishi, Sam Benjamin, Che-Kai Tsao
<p><strong>Background: </strong>PARP inhibitor (PARPi)-based therapy is a well-established treatment modality for metastatic castration-resistant prostate cancer (mCRPC) harboring homologous recombination repair (HRR) deficiencies. However, its clinical efficacy in mCRPC without HRR alterations remains undefined.</p><p><strong>Objective: </strong>This study aimed to evaluate the efficacy of PARPis in HRR-proficient mCRPC.</p><p><strong>Methods: </strong>A systematic database search was conducted to identify clinical trials evaluating the efficacy of PARPi-based therapies in patients with HRR-proficient mCRPC. Searches were performed using PubMed, Embase Cochrane Library, Web of Science, and relevant international conference proceedings. Both single-arm and pairwise meta-analytical approaches were employed to assess the efficacy of PARPis in HRR-proficient mCRPC, while concurrently comparing the estimates with those from HRR-deficient populations within the same trials.</p><p><strong>Results: </strong>A total of eight single-arm trials and eight randomized controlled trials were included, comprising 3314 patients, of whom 1727 were HRR-proficient. In the single-arm meta-analysis, the pooled prostate-specific antigen (PSA) response (≥ 50% decrease from baseline) rate was 18% (95% CI 9-29, I<sup>2</sup> = 82%) in the HRR-proficient subpopulation, and 46% (95% CI 35-59, I<sup>2</sup> = 63%) in the HRR-deficient subpopulation, with a significant difference between subpopulations (p = 0.003). The pooled 12-month progression-free survival (PFS) rate was 42% (95% CI 29-57, I<sup>2</sup> = 93%) in the HRR-proficient subpopulation and 57% (95% CI 48-67, I<sup>2</sup> = 72%) in the HRR-deficient subpopulation. No significant difference was observed between the two subpopulations (p = 0.177). Subgroup analyses based on treatment regimens (PARPi monotherapy, PARPi plus androgen receptor axis-targeted (ARAT) agents, PARPi plus immune checkpoint inhibitors, and PARPi plus others) also revealed no significant difference in efficacy between HRR-proficient and HRR-deficient subpopulations. In the pairwise meta-analysis, the addition of PARPi was associated with a significantly longer PFS compared with treatment without PARPi in both HRR-proficient and HRR-deficient subpopulations, with hazard ratios of 0.69 (95% CI 0.60-0.80, I<sup>2</sup> = 32%) and 0.60 (95% CI 0.50-0.72, I<sup>2</sup> = 16%), respectively. Also, there was no statistically significant difference (p = 0.22) between the two subpopulations. PARPi plus ARAT showed a trend toward improved overall survival in HRR-proficient patients, with a more pronounced benefit observed in HRR-deficient patients.</p><p><strong>Conclusions: </strong>The use of PARPis in the treatment of CRPC demonstrated a significant improvement in PFS irrespective of HRR status. Even in the HRR-proficient subpopulation, PARPi-based therapy conferred encouraging PFS benefits, underscoring its potential clinical relevance beyond HRR
背景:PARP抑制剂(PARPi)为基础的治疗是转移性去势抵抗性前列腺癌(mCRPC)同源重组修复(HRR)缺陷的一种成熟的治疗方式。然而,其在无HRR改变的mCRPC中的临床疗效尚不明确。目的:本研究旨在评价PARPis在hrr熟练的mCRPC中的疗效。方法:进行系统的数据库检索,以确定评估基于parpi的治疗对hrr精通的mCRPC患者疗效的临床试验。检索使用PubMed, Embase Cochrane Library, Web of Science和相关的国际会议记录。采用单臂和两两荟萃分析方法来评估PARPis在hrr熟练的mCRPC中的疗效,同时将估计结果与同一试验中hrr缺乏人群的估计结果进行比较。结果:共纳入8项单臂试验和8项随机对照试验,共3314例患者,其中1727例hrr精通。在单臂荟萃分析中,hrr精通亚群的总前列腺特异性抗原(PSA)应答率(比基线降低≥50%)为18% (95% CI 9-29, I2 = 82%), hrr缺乏亚群的总PSA应答率为46% (95% CI 35-59, I2 = 63%),亚群间差异显著(p = 0.003)。总的12个月无进展生存率(PFS)在hrr精通亚群中为42% (95% CI 29-57, I2 = 93%),在hrr缺乏亚群中为57% (95% CI 48-67, I2 = 72%)。两个亚群间差异无统计学意义(p = 0.177)。基于治疗方案的亚组分析(PARPi单药、PARPi加雄激素受体轴靶向(ARAT)药物、PARPi加免疫检查点抑制剂和PARPi加其他药物)也显示,hrr精通和hrr缺乏亚群之间的疗效无显著差异。在两两荟萃分析中,在hrr精通和hrr缺乏亚群中,与不使用PARPi治疗相比,添加PARPi与PFS显著延长相关,风险比分别为0.69 (95% CI 0.60-0.80, I2 = 32%)和0.60 (95% CI 0.50-0.72, I2 = 16%)。两个亚群之间也无统计学差异(p = 0.22)。PARPi + ARAT在hrr熟练的患者中显示出改善总生存的趋势,在hrr缺乏的患者中观察到更明显的益处。结论:无论HRR状况如何,使用PARPis治疗CRPC均可显著改善PFS。即使在hrr精通的亚群中,基于parpi的治疗也带来了令人鼓舞的PFS益处,强调了其在hrr缺乏环境之外的潜在临床相关性。进一步的生物标志物分析是必要的,以完善患者选择和优化治疗策略。
{"title":"Can PARP Inhibitors Benefit Patients with Homologous Recombination Repair-Proficient Castration-Resistant Prostate Cancer? A Meta-analysis.","authors":"Susu Zhou, Devashish Desai, Noriko Kishi, Sam Benjamin, Che-Kai Tsao","doi":"10.1007/s11523-025-01188-8","DOIUrl":"https://doi.org/10.1007/s11523-025-01188-8","url":null,"abstract":"<p><strong>Background: </strong>PARP inhibitor (PARPi)-based therapy is a well-established treatment modality for metastatic castration-resistant prostate cancer (mCRPC) harboring homologous recombination repair (HRR) deficiencies. However, its clinical efficacy in mCRPC without HRR alterations remains undefined.</p><p><strong>Objective: </strong>This study aimed to evaluate the efficacy of PARPis in HRR-proficient mCRPC.</p><p><strong>Methods: </strong>A systematic database search was conducted to identify clinical trials evaluating the efficacy of PARPi-based therapies in patients with HRR-proficient mCRPC. Searches were performed using PubMed, Embase Cochrane Library, Web of Science, and relevant international conference proceedings. Both single-arm and pairwise meta-analytical approaches were employed to assess the efficacy of PARPis in HRR-proficient mCRPC, while concurrently comparing the estimates with those from HRR-deficient populations within the same trials.</p><p><strong>Results: </strong>A total of eight single-arm trials and eight randomized controlled trials were included, comprising 3314 patients, of whom 1727 were HRR-proficient. In the single-arm meta-analysis, the pooled prostate-specific antigen (PSA) response (≥ 50% decrease from baseline) rate was 18% (95% CI 9-29, I<sup>2</sup> = 82%) in the HRR-proficient subpopulation, and 46% (95% CI 35-59, I<sup>2</sup> = 63%) in the HRR-deficient subpopulation, with a significant difference between subpopulations (p = 0.003). The pooled 12-month progression-free survival (PFS) rate was 42% (95% CI 29-57, I<sup>2</sup> = 93%) in the HRR-proficient subpopulation and 57% (95% CI 48-67, I<sup>2</sup> = 72%) in the HRR-deficient subpopulation. No significant difference was observed between the two subpopulations (p = 0.177). Subgroup analyses based on treatment regimens (PARPi monotherapy, PARPi plus androgen receptor axis-targeted (ARAT) agents, PARPi plus immune checkpoint inhibitors, and PARPi plus others) also revealed no significant difference in efficacy between HRR-proficient and HRR-deficient subpopulations. In the pairwise meta-analysis, the addition of PARPi was associated with a significantly longer PFS compared with treatment without PARPi in both HRR-proficient and HRR-deficient subpopulations, with hazard ratios of 0.69 (95% CI 0.60-0.80, I<sup>2</sup> = 32%) and 0.60 (95% CI 0.50-0.72, I<sup>2</sup> = 16%), respectively. Also, there was no statistically significant difference (p = 0.22) between the two subpopulations. PARPi plus ARAT showed a trend toward improved overall survival in HRR-proficient patients, with a more pronounced benefit observed in HRR-deficient patients.</p><p><strong>Conclusions: </strong>The use of PARPis in the treatment of CRPC demonstrated a significant improvement in PFS irrespective of HRR status. Even in the HRR-proficient subpopulation, PARPi-based therapy conferred encouraging PFS benefits, underscoring its potential clinical relevance beyond HRR","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145669892","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1007/s11523-025-01187-9
Erick F Saldanha, Mariana M Noronha, Pedro C A Reis, Pedro Robson Costa Passos, Valbert O C Filho, Anelise P Cappellaro, Luiz Felipe Costa Almeida, Jean Henri Maselli-Shoueri, Carlos Diego Holanda Lopes, Luis Felipe Leite, Mauricio F Ribeiro, Daniel V Araujo
Background: Tebentafusp is the first systemic therapy to improve survival outcomes for patients with HLA-A*02:01-positive metastatic uveal melanoma (mUM). However, outside the pivotal study, limited data for tebentafusp are reported in literature.
Objective: To evaluate the efficacy and safety of tebentafusp in patients with human leukocyte antigen (HLA)-A*02:01-positive mUM across cohort studies and clinical trials.
Patients and methods: PubMed, EMBASE, Cochrane, and Web of Science (up to July 2025) were surveyed for studies evaluating tebentafusp in patients with HLA-A*02:01-positive mUM. A meta-analysis using a random-effects model and the inverse variance method was conducted; Kaplan-Meier curves, where available, were used to recreate the time-to-event data. The primary objective was efficacy, including objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). The secondary objective was to evaluate all-grade and severe (grade ≥ 3 or higher) adverse events (AEs).
Results: A total of 997 patients from 12 cohorts were included. Using reconstructed time-to-event outcomes from published Kaplan-Meier curves, the estimated median PFS was 3.9 months (95% confidence interval [CI] 3.77-4.92). The median OS was 21.2 months (95% CI 19.2-23.1). When stratified by study design, the median OS was similar between prospective and retrospective studies: 21.2 months (95% CI 19.2-23.8) and 21.1 months (95% CI 18.1-33.6), respectively. Likewise, median progression-free survival (PFS) was comparable between prospective and retrospective studies: 3.8 months (95% CI 3.25-5.5) and 3.9 months (95% CI 3.8-4.9), respectively. The rate of cytokine- and cutaneous-mediated events was 67% (95% CI 45-84; I2 = 94.4%) and 64% (95% CI 28-89; I2 = 97.3%), respectively. Severe AEs for cytokine-mediated events were 3% (95% CI 1-13; I2 = 87.6%), and cutaneous-mediated events were 11% (95% CI 8-14; I2 = 0%). The discontinuation rate was 2% (95% CI 1-4; I2 = 0%).
Conclusions: Tebentafusp for patients with HLA-A*02:01-positive mUM is associated with improved survival outcomes and manageable toxicity. These findings support tebentafusp as the standard of care for this patient population.
背景:Tebentafusp是首个改善HLA-A*02:01阳性转移性葡萄膜黑色素瘤(mUM)患者生存结局的全身疗法。然而,在关键研究之外,文献报道了有限的tebentafusp数据。目的:通过队列研究和临床试验,评价替本他福普治疗人白细胞抗原(HLA)-A*02:01阳性mUM患者的疗效和安全性。患者和方法:PubMed, EMBASE, Cochrane和Web of Science(截至2025年7月)进行了调查,以评估tebentafusp对HLA-A*02:01阳性mUM患者的影响。采用随机效应模型和反方差法进行meta分析;Kaplan-Meier曲线(如果有的话)被用来重建事件发生的时间数据。主要目标是疗效,包括客观缓解率(ORR)、无进展生存期(PFS)和总生存期(OS)。次要目的是评估所有级别和严重(≥3级或更高)不良事件(ae)。结果:12个队列共纳入997例患者。根据已发表的Kaplan-Meier曲线重建的事件发生时间结果,估计中位PFS为3.9个月(95%可信区间[CI] 3.77-4.92)。中位OS为21.2个月(95% CI 19.2-23.1)。当按研究设计分层时,前瞻性和回顾性研究的中位总生存期相似:分别为21.2个月(95% CI 19.2-23.8)和21.1个月(95% CI 18.1-33.6)。同样,中位无进展生存期(PFS)在前瞻性和回顾性研究之间具有可比性:分别为3.8个月(95% CI 3.25-5.5)和3.9个月(95% CI 3.8-4.9)。细胞因子和皮肤介导的事件发生率分别为67% (95% CI 45-84; I2 = 94.4%)和64% (95% CI 28-89; I2 = 97.3%)。细胞因子介导事件的严重ae为3% (95% CI 1-13; I2 = 87.6%),皮肤介导事件的严重ae为11% (95% CI 8-14; I2 = 0%)。停药率为2% (95% CI 1-4; I2 = 0%)。结论:Tebentafusp用于HLA-A*02:01阳性mUM患者可改善生存结果和控制毒性。这些发现支持tebentafusp作为该患者群体的标准护理。
{"title":"Tebentafusp in Metastatic Uveal Melanoma: A Meta-analysis.","authors":"Erick F Saldanha, Mariana M Noronha, Pedro C A Reis, Pedro Robson Costa Passos, Valbert O C Filho, Anelise P Cappellaro, Luiz Felipe Costa Almeida, Jean Henri Maselli-Shoueri, Carlos Diego Holanda Lopes, Luis Felipe Leite, Mauricio F Ribeiro, Daniel V Araujo","doi":"10.1007/s11523-025-01187-9","DOIUrl":"https://doi.org/10.1007/s11523-025-01187-9","url":null,"abstract":"<p><strong>Background: </strong>Tebentafusp is the first systemic therapy to improve survival outcomes for patients with HLA-A*02:01-positive metastatic uveal melanoma (mUM). However, outside the pivotal study, limited data for tebentafusp are reported in literature.</p><p><strong>Objective: </strong>To evaluate the efficacy and safety of tebentafusp in patients with human leukocyte antigen (HLA)-A*02:01-positive mUM across cohort studies and clinical trials.</p><p><strong>Patients and methods: </strong>PubMed, EMBASE, Cochrane, and Web of Science (up to July 2025) were surveyed for studies evaluating tebentafusp in patients with HLA-A*02:01-positive mUM. A meta-analysis using a random-effects model and the inverse variance method was conducted; Kaplan-Meier curves, where available, were used to recreate the time-to-event data. The primary objective was efficacy, including objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). The secondary objective was to evaluate all-grade and severe (grade ≥ 3 or higher) adverse events (AEs).</p><p><strong>Results: </strong>A total of 997 patients from 12 cohorts were included. Using reconstructed time-to-event outcomes from published Kaplan-Meier curves, the estimated median PFS was 3.9 months (95% confidence interval [CI] 3.77-4.92). The median OS was 21.2 months (95% CI 19.2-23.1). When stratified by study design, the median OS was similar between prospective and retrospective studies: 21.2 months (95% CI 19.2-23.8) and 21.1 months (95% CI 18.1-33.6), respectively. Likewise, median progression-free survival (PFS) was comparable between prospective and retrospective studies: 3.8 months (95% CI 3.25-5.5) and 3.9 months (95% CI 3.8-4.9), respectively. The rate of cytokine- and cutaneous-mediated events was 67% (95% CI 45-84; I<sup>2</sup> = 94.4%) and 64% (95% CI 28-89; I<sup>2</sup> = 97.3%), respectively. Severe AEs for cytokine-mediated events were 3% (95% CI 1-13; I<sup>2</sup> = 87.6%), and cutaneous-mediated events were 11% (95% CI 8-14; I<sup>2</sup> = 0%). The discontinuation rate was 2% (95% CI 1-4; I<sup>2</sup> = 0%).</p><p><strong>Conclusions: </strong>Tebentafusp for patients with HLA-A*02:01-positive mUM is associated with improved survival outcomes and manageable toxicity. These findings support tebentafusp as the standard of care for this patient population.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145669850","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-11-15DOI: 10.1007/s11523-025-01183-z
Eve Merry, Charlie Gourley
Exploiting DNA damage repair (DDR) vulnerabilities has become a major focus in cancer drug development following the clinical success of poly (ADP-ribose) polymerase (PARP) inhibitors. Beyond PARP, inhibitors of multiple other DDR proteins have progressed from preclinical development to early-phase clinical trials. DNA damage repair inhibitors have shown promise both as a selective therapeutic strategy in genomically selected cancers harbouring specific genetic vulnerabilities, and as a potential treatment approach to overcome innate and acquired PARP inhibitor resistance. This review summarises the most recent DDR inhibitor clinical evidence, focusing on DDR signalling targets; ATR (ataxia telangiectasia and Rad3-related protein serine/threonine kinase), ATM (ataxia telangiectasia mutated kinase), DNA-PK (DNA-dependent protein kinase), CHK1 (checkpoint kinase 1), WEE1 and recently emerging DNA repair targets; RAD51, PolƟ (DNA polymerase theta), WRN (Werner syndrome helicase), USP1 (ubiquitin specific peptidase 1) and PARG (poly(ADP-ribose) glycohydrolase). We highlight both clinical successes and failures of DDR inhibitors as monotherapy or in combination with chemotherapy, PARP and other DDR inhibitors. The challenges that must be addressed to see the true potential of these agents are discussed. Finally, we consider future directions and the necessity for integration of biomarkers and genomic profiling in DDR inhibitor clinical trials to optimally identify patients with tumours genetically vulnerable, and so crucially, take advantage of the selective therapeutic opportunity provided by DDR inhibition.
{"title":"Targeting DNA Damage Repair Pathways Beyond PARP Inhibition.","authors":"Eve Merry, Charlie Gourley","doi":"10.1007/s11523-025-01183-z","DOIUrl":"10.1007/s11523-025-01183-z","url":null,"abstract":"<p><p>Exploiting DNA damage repair (DDR) vulnerabilities has become a major focus in cancer drug development following the clinical success of poly (ADP-ribose) polymerase (PARP) inhibitors. Beyond PARP, inhibitors of multiple other DDR proteins have progressed from preclinical development to early-phase clinical trials. DNA damage repair inhibitors have shown promise both as a selective therapeutic strategy in genomically selected cancers harbouring specific genetic vulnerabilities, and as a potential treatment approach to overcome innate and acquired PARP inhibitor resistance. This review summarises the most recent DDR inhibitor clinical evidence, focusing on DDR signalling targets; ATR (ataxia telangiectasia and Rad3-related protein serine/threonine kinase), ATM (ataxia telangiectasia mutated kinase), DNA-PK (DNA-dependent protein kinase), CHK1 (checkpoint kinase 1), WEE1 and recently emerging DNA repair targets; RAD51, PolƟ (DNA polymerase theta), WRN (Werner syndrome helicase), USP1 (ubiquitin specific peptidase 1) and PARG (poly(ADP-ribose) glycohydrolase). We highlight both clinical successes and failures of DDR inhibitors as monotherapy or in combination with chemotherapy, PARP and other DDR inhibitors. The challenges that must be addressed to see the true potential of these agents are discussed. Finally, we consider future directions and the necessity for integration of biomarkers and genomic profiling in DDR inhibitor clinical trials to optimally identify patients with tumours genetically vulnerable, and so crucially, take advantage of the selective therapeutic opportunity provided by DDR inhibition.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":"937-953"},"PeriodicalIF":4.0,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145522869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-10-28DOI: 10.1007/s11523-025-01181-1
Chin Hang Yiu, Adrian Lee, Christine Y Lu
Introduction: Immune checkpoint inhibitors are a standard first-line treatment for metastatic renal cell carcinoma (RCC), with combination ipilimumab and nivolumab (ipi-nivo) widely recommended. However, real-world data on its use in Australia remain limited.
Objective: To evaluate real-world outcomes of ipi-nivo for metastatic RCC in the Australian healthcare setting.
Methods: We conducted a retrospective cohort study using nationally linked Pharmaceutical Benefits Scheme data and National Death Index records accessed via the Australian Bureau of Statistics DataLab. Patients initiating ipi-nivo for stage IV clear cell RCC between 1 July 2019 and 30 June 2023 were included. Kaplan-Meier analyses and multivariate Cox regression were used to estimate overall survival (OS) and time to treatment discontinuation (TTD). Immune-related adverse events (irAEs) were inferred from incident dispensing of corticosteroids and levothyroxine. Subgroup analyses were performed by age (18-64 versus ≥ 65 years) and sex.
Results: Among 1334 patients, median OS was 30.0 months with 12- and 24-month survival rates of 72.9% (95% confidence interval [CI] 70.5-75.3) and 56.4% (95% CI 53.7-59.2), respectively. Median TTD was 4.9 months, and 38.5% of patients discontinued treatment during the induction phase. Incident corticosteroid and levothyroxine use occurred in 24.2% and 11.2% of patients, respectively. No significant differences in OS, TTD, or irAE proxies were observed by age or sex.
Conclusions: In this national, population-based study, real-world survival outcomes with ipi-nivo for metastatic RCC were shorter than those reported in clinical trials. These findings provide population-level benchmarks from a universal healthcare system and highlight the limitations of applying trial outcomes to unselected real-world populations. By contributing robust, large-scale data from routine practice, this study adds to the global evidence base and supports the integration of real-world data into clinical and policy decisions around immunotherapy.
免疫检查点抑制剂是转移性肾细胞癌(RCC)的标准一线治疗方法,ipilimumab和nivolumab (ipi-nivo)联合被广泛推荐。然而,关于它在澳大利亚使用的真实数据仍然有限。目的:评估ipi-nivo在澳大利亚医疗机构治疗转移性肾细胞癌的实际结果。方法:我们进行了一项回顾性队列研究,使用了通过澳大利亚统计局数据实验室获得的全国药品福利计划数据和全国死亡指数记录。纳入了2019年7月1日至2023年6月30日期间接受ipi-nivo治疗IV期透明细胞RCC的患者。Kaplan-Meier分析和多变量Cox回归用于估计总生存期(OS)和治疗停止时间(TTD)。免疫相关不良事件(irAEs)是从皮质类固醇和左甲状腺素的事件分配推断出来的。按年龄(18-64岁对≥65岁)和性别进行亚组分析。结果:1334例患者中位OS为30.0个月,12月和24月生存率分别为72.9%(95%可信区间[CI] 70.5-75.3)和56.4% (95% CI 53.7-59.2)。中位TTD为4.9个月,38.5%的患者在诱导期停止治疗。使用皮质类固醇和左旋甲状腺素的发生率分别为24.2%和11.2%。OS、TTD或irAE指标在年龄或性别方面无显著差异。结论:在这项全国性的、基于人群的研究中,ipi-nivo治疗转移性RCC的真实生存结果比临床试验中报道的要短。这些发现提供了全民医疗保健系统的人口水平基准,并强调了将试验结果应用于未选择的现实世界人群的局限性。通过提供来自常规实践的可靠的大规模数据,本研究增加了全球证据基础,并支持将真实世界的数据整合到免疫治疗的临床和政策决策中。
{"title":"Real-World Outcomes of Ipilimumab Plus Nivolumab for Metastatic Renal Cell Carcinoma: A National Population-Based Cohort Study.","authors":"Chin Hang Yiu, Adrian Lee, Christine Y Lu","doi":"10.1007/s11523-025-01181-1","DOIUrl":"10.1007/s11523-025-01181-1","url":null,"abstract":"<p><strong>Introduction: </strong>Immune checkpoint inhibitors are a standard first-line treatment for metastatic renal cell carcinoma (RCC), with combination ipilimumab and nivolumab (ipi-nivo) widely recommended. However, real-world data on its use in Australia remain limited.</p><p><strong>Objective: </strong>To evaluate real-world outcomes of ipi-nivo for metastatic RCC in the Australian healthcare setting.</p><p><strong>Methods: </strong>We conducted a retrospective cohort study using nationally linked Pharmaceutical Benefits Scheme data and National Death Index records accessed via the Australian Bureau of Statistics DataLab. Patients initiating ipi-nivo for stage IV clear cell RCC between 1 July 2019 and 30 June 2023 were included. Kaplan-Meier analyses and multivariate Cox regression were used to estimate overall survival (OS) and time to treatment discontinuation (TTD). Immune-related adverse events (irAEs) were inferred from incident dispensing of corticosteroids and levothyroxine. Subgroup analyses were performed by age (18-64 versus ≥ 65 years) and sex.</p><p><strong>Results: </strong>Among 1334 patients, median OS was 30.0 months with 12- and 24-month survival rates of 72.9% (95% confidence interval [CI] 70.5-75.3) and 56.4% (95% CI 53.7-59.2), respectively. Median TTD was 4.9 months, and 38.5% of patients discontinued treatment during the induction phase. Incident corticosteroid and levothyroxine use occurred in 24.2% and 11.2% of patients, respectively. No significant differences in OS, TTD, or irAE proxies were observed by age or sex.</p><p><strong>Conclusions: </strong>In this national, population-based study, real-world survival outcomes with ipi-nivo for metastatic RCC were shorter than those reported in clinical trials. These findings provide population-level benchmarks from a universal healthcare system and highlight the limitations of applying trial outcomes to unselected real-world populations. By contributing robust, large-scale data from routine practice, this study adds to the global evidence base and supports the integration of real-world data into clinical and policy decisions around immunotherapy.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":"1003-1014"},"PeriodicalIF":4.0,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145393115","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-10-27DOI: 10.1007/s11523-025-01180-2
Zaid Muhanna, Muntaser Al Zyoud, Ahmad Issa, Muhammad Awidi
Background: Osimertinib, a third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), is the standard first-line therapy for EGFR-mutant non-small cell lung cancer (NSCLC). Emerging evidence suggests that it may be associated with increased cardiotoxicity; however, current evidence is conflicting, and a small sample size and a lack of direct comparisons with other EGFR-TKIs limit existing studies.
Objective: We aim to examine the incidence of cardiovascular toxicity with osimertinib compared with other EGFR-TKIs in a real-world setting.
Patients and methods: A retrospective cohort study was conducted via the Trinetx global federated health research platform to compare the incidence of cardiotoxicity between osimertinib and other EGFR-TKIs over a 5-year follow-up. Patients were matched using 1:1 propensity score matching (PSM). Outcomes included cardiomyopathies, arrhythmias, ischemic heart disease (IHD), and heart failure (HF), assessed using ICD-10 codes.
Results: Following PSM, each arm consisted of 7331 patients; the arms were well balanced. Osimertinib was associated with increased risk of cardiomyopathy (2.8% vs 0.8%; HR: 3.143 [95% CI 2.342-4.218]), IHD (8.7% vs 5.5%; HR: 1.432 [95% CI 1.248-1.643]), and HF (6.2% vs 4%; HR: 1.41 [95% CI 1.210-1.644]). A similar incidence of arrhythmia was observed (9% vs 8.5%; HR: 0.938 [95% CI 0.831-1.059]); however, it increased after 3 years. Comparisons with individual EGFR-TKIs showed varying results.
Conclusion: In this large, real-world study, osimertinib was associated with elevated cardiotoxicity risk. These findings underscore the need for serial monitoring of patients receiving osimertinib and for future studies comparing cardioprotective drugs.
背景:奥西替尼是第三代表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI),是egfr突变型非小细胞肺癌(NSCLC)的标准一线治疗药物。新出现的证据表明,它可能与心脏毒性增加有关;然而,目前的证据是相互矛盾的,小样本量和缺乏与其他egfr - tki的直接比较限制了现有的研究。目的:我们的目的是在现实环境中比较奥西替尼与其他EGFR-TKIs的心血管毒性发生率。患者和方法:通过Trinetx全球联合健康研究平台进行了一项回顾性队列研究,比较了奥西替尼和其他EGFR-TKIs在5年随访期间的心脏毒性发生率。采用1:1倾向评分匹配(PSM)对患者进行匹配。结果包括心肌病、心律失常、缺血性心脏病(IHD)和心力衰竭(HF),使用ICD-10代码进行评估。结果:PSM后,每组7331例患者;手臂很平衡。奥西替尼与心肌病(2.8% vs 0.8%;风险比:3.143 [95% CI 2.342-4.218])、IHD (8.7% vs 5.5%;风险比:1.432 [95% CI 1.248-1.643])和HF (6.2% vs 4%;风险比:1.41 [95% CI 1.210-1.644])相关。心律失常发生率相似(9% vs 8.5%; HR: 0.938 [95% CI 0.831-1.059]);然而,3年后增加了。与个体egfr - tki的比较显示出不同的结果。结论:在这项大型现实世界研究中,奥西替尼与心脏毒性风险升高相关。这些发现强调需要对接受奥西替尼的患者进行连续监测,并对未来的心脏保护药物进行比较研究。
{"title":"Cardiotoxicity Profiles of Osimertinib Compared with Other EGFR Tyrosine Kinase Inhibitors: A Real-World Comparative Incidence Analysis.","authors":"Zaid Muhanna, Muntaser Al Zyoud, Ahmad Issa, Muhammad Awidi","doi":"10.1007/s11523-025-01180-2","DOIUrl":"10.1007/s11523-025-01180-2","url":null,"abstract":"<p><strong>Background: </strong>Osimertinib, a third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), is the standard first-line therapy for EGFR-mutant non-small cell lung cancer (NSCLC). Emerging evidence suggests that it may be associated with increased cardiotoxicity; however, current evidence is conflicting, and a small sample size and a lack of direct comparisons with other EGFR-TKIs limit existing studies.</p><p><strong>Objective: </strong>We aim to examine the incidence of cardiovascular toxicity with osimertinib compared with other EGFR-TKIs in a real-world setting.</p><p><strong>Patients and methods: </strong>A retrospective cohort study was conducted via the Trinetx global federated health research platform to compare the incidence of cardiotoxicity between osimertinib and other EGFR-TKIs over a 5-year follow-up. Patients were matched using 1:1 propensity score matching (PSM). Outcomes included cardiomyopathies, arrhythmias, ischemic heart disease (IHD), and heart failure (HF), assessed using ICD-10 codes.</p><p><strong>Results: </strong>Following PSM, each arm consisted of 7331 patients; the arms were well balanced. Osimertinib was associated with increased risk of cardiomyopathy (2.8% vs 0.8%; HR: 3.143 [95% CI 2.342-4.218]), IHD (8.7% vs 5.5%; HR: 1.432 [95% CI 1.248-1.643]), and HF (6.2% vs 4%; HR: 1.41 [95% CI 1.210-1.644]). A similar incidence of arrhythmia was observed (9% vs 8.5%; HR: 0.938 [95% CI 0.831-1.059]); however, it increased after 3 years. Comparisons with individual EGFR-TKIs showed varying results.</p><p><strong>Conclusion: </strong>In this large, real-world study, osimertinib was associated with elevated cardiotoxicity risk. These findings underscore the need for serial monitoring of patients receiving osimertinib and for future studies comparing cardioprotective drugs.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":"1015-1022"},"PeriodicalIF":4.0,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145378752","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-10-14DOI: 10.1007/s11523-025-01175-z
Richard S Finn, Hope S Rugo, Javier Cortes, Sibylle Loibl, Grace Foley, Eric Gauthier, Yao Wang, Sindy Kim, Lars Anders, Dennis J Slamon
The initial approval of palbociclib in 2015 marked a major advancement in the treatment of hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) advanced breast cancer (ABC). As the first-in-class cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, palbociclib introduced a new therapeutic strategy for this disease subtype by targeting the CDK4/6:cyclin-D:Rb pathway to halt cell cycle progression from the G1 to the S phase. The PALOMA clinical trials demonstrated a significant improvement in progression-free survival for palbociclib in combination with endocrine therapy (ET), representing clinically meaningful and consistent progression-free survival benefits across all studies in patients with HR+/HER2- ABC. Although the PALOMA clinical trials did not demonstrate a statistically significant overall survival (OS; secondary endpoint) benefit for palbociclib combined with ET in HR+/HER2- ABC over ET monotherapy, several real-world studies have reported substantial OS improvements in diverse patient populations. Despite the significant improvement in clinical outcomes, there remain challenges, particularly regarding proposed resistance mechanisms such as RB1 loss and CDK2 activation, which may diminish the long-term effectiveness of CDK4/6 inhibitors. Moreover, although the toxicity profiles of CDK4/6 inhibitors, such as the risks of myelosuppression and gastrointestinal side effects, necessitate careful patient monitoring, there is an opportunity to refine their use and explore strategies for broader applicability. For these reasons, further research into next-generation CDK inhibitors and combination therapies are critical and ongoing. This review explores the discovery of CDK inhibitors, the development of palbociclib from preclinical studies to its global approval, and future drug development strategies to overcome resistance and enhance patient outcomes.
{"title":"A Decade After Approval of the First CDK4/6 Inhibitor: A Look Back at Palbociclib's Journey from Discovery to Approval and What's Next in CDK Inhibition in Breast Cancer.","authors":"Richard S Finn, Hope S Rugo, Javier Cortes, Sibylle Loibl, Grace Foley, Eric Gauthier, Yao Wang, Sindy Kim, Lars Anders, Dennis J Slamon","doi":"10.1007/s11523-025-01175-z","DOIUrl":"10.1007/s11523-025-01175-z","url":null,"abstract":"<p><p>The initial approval of palbociclib in 2015 marked a major advancement in the treatment of hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) advanced breast cancer (ABC). As the first-in-class cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, palbociclib introduced a new therapeutic strategy for this disease subtype by targeting the CDK4/6:cyclin-D:Rb pathway to halt cell cycle progression from the G1 to the S phase. The PALOMA clinical trials demonstrated a significant improvement in progression-free survival for palbociclib in combination with endocrine therapy (ET), representing clinically meaningful and consistent progression-free survival benefits across all studies in patients with HR+/HER2- ABC. Although the PALOMA clinical trials did not demonstrate a statistically significant overall survival (OS; secondary endpoint) benefit for palbociclib combined with ET in HR+/HER2- ABC over ET monotherapy, several real-world studies have reported substantial OS improvements in diverse patient populations. Despite the significant improvement in clinical outcomes, there remain challenges, particularly regarding proposed resistance mechanisms such as RB1 loss and CDK2 activation, which may diminish the long-term effectiveness of CDK4/6 inhibitors. Moreover, although the toxicity profiles of CDK4/6 inhibitors, such as the risks of myelosuppression and gastrointestinal side effects, necessitate careful patient monitoring, there is an opportunity to refine their use and explore strategies for broader applicability. For these reasons, further research into next-generation CDK inhibitors and combination therapies are critical and ongoing. This review explores the discovery of CDK inhibitors, the development of palbociclib from preclinical studies to its global approval, and future drug development strategies to overcome resistance and enhance patient outcomes.</p>","PeriodicalId":22195,"journal":{"name":"Targeted Oncology","volume":" ","pages":"917-936"},"PeriodicalIF":4.0,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12669314/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145293834","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}