Annals of OncologyPub Date : 2024-12-17DOI: 10.1016/j.annonc.2024.12.004
D Daoudlarian, A Segot, S Latifyan, R Bartolini, V Joo, N Mederos, H Bouchaab, R Demicheli, K Abdelhamid, N Ferahta, J Doms, G Stalder, A Noto, L Mencarelli, V Mosimann, D Berthold, A Stravodimou, C Sartori, K Shabafrouz, J A Thompson, Y Wang, S Peters, G Pantaleo, M Obeid
{"title":"Tocilizumab and immune signatures for targeted management of cytokine release syndrome in immune checkpoint therapy.","authors":"D Daoudlarian, A Segot, S Latifyan, R Bartolini, V Joo, N Mederos, H Bouchaab, R Demicheli, K Abdelhamid, N Ferahta, J Doms, G Stalder, A Noto, L Mencarelli, V Mosimann, D Berthold, A Stravodimou, C Sartori, K Shabafrouz, J A Thompson, Y Wang, S Peters, G Pantaleo, M Obeid","doi":"10.1016/j.annonc.2024.12.004","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.004","url":null,"abstract":"<p><strong>Background: </strong>This study aimed to identify specific biomarkers in oncology patients experiencing immune-related cytokine release syndrome (irCRS)-like symptoms during immune checkpoint inhibitor (ICI) therapy, including severe cases like hemophagocytic lymphohistiocytosis (irHLH), and to distinguish these from sepsis. A secondary objective was to retrospectively analyze the efficacy of tocilizumab (TCZ) in treating corticosteroid (CS)-refractory high-grade irCRS.</p><p><strong>Patients and methods: </strong>A cohort of 35 patients presenting with irCRS-like symptoms was studied, including 9 with irHLH-like manifestations and 8 with sepsis. Immune profiling was performed using 48 mass cytometry markers, along with an analysis of 45 serum biomarkers, including 27 cytokines and 18 additional markers from the HScore. Twelve patients with high-grade irCRS refractory to CS were treated with TCZ.</p><p><strong>Results: </strong>24 biomarkers significantly distinguished between irHLH and Grade 3 irCRS (P=0.0027-0.0455). Hepatocyte growth factor (HGF) and ferritin had superior predictive values compared to the traditional HScore, both with a positive predictive value (PPV) and negative predictive value (NPV) of 100%. CXCL9 differentiated irHLH from Grade 3 irCRS and predicted the need for TCZ treatment intensification (PPV=90%, NPV=100%). Additional biomarkers, including leukocyte count, neutrophils, ferritin, IL-6, IL-7, EGF, fibrinogen, and GM-CSF, discriminated sepsis from high-grade irCRS (PPV=75-80%, NPV=100%). Elevated frequencies of CXCR5+ or CCR4+ CD8 memory cells, CD38+ intermediate monocytes, and CD62L+ neutrophils were observed in high-grade irCRS compared to sepsis. All 12 patients with high-grade irCRS refractory to CS treated with TCZ experienced complete resolution.</p><p><strong>Conclusions: </strong>This study highlights the importance of specific immunologic biomarkers in determining irCRS severity, predicting outcomes, and distinguishing between irHLH, irCRS, and sepsis. It also demonstrates the efficacy of TCZ in managing high-grade irCRS, underscoring the need for personalized therapeutic strategies based on these biomarkers.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142863165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-14DOI: 10.1016/j.annonc.2024.12.007
G Liu, S V Bratman, D D De Carvalho, A-R Hartman
{"title":"Response Letter to the Editor.","authors":"G Liu, S V Bratman, D D De Carvalho, A-R Hartman","doi":"10.1016/j.annonc.2024.12.007","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.007","url":null,"abstract":"","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142833765","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-13DOI: 10.1016/j.annonc.2024.12.006
B Martínez-Castedo, D G Camblor, J Martín-Arana, J A Carbonell-Asins, B García-Micó, V Gambardella, M Huerta, S Roselló, D Roda, F Gimeno-Valiente, A Cervantes, N Tarazona
{"title":"Minimal residual disease in colorectal cancer. Tumor-informed versus tumor-agnostic approaches: unraveling the optimal strategy.","authors":"B Martínez-Castedo, D G Camblor, J Martín-Arana, J A Carbonell-Asins, B García-Micó, V Gambardella, M Huerta, S Roselló, D Roda, F Gimeno-Valiente, A Cervantes, N Tarazona","doi":"10.1016/j.annonc.2024.12.006","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.006","url":null,"abstract":"<p><strong>Background: </strong>Circulating tumor DNA (ctDNA) analysis has emerged as a minimally invasive tool for detecting minimal residual disease (MRD) in colorectal cancer (CRC) patients. This enables dynamic risk stratification, earlier recurrence detection, and optimized post-surgical treatment. Two primary methodologies have been developed for ctDNA-based MRD detection: tumor-informed strategies, which identify tumor-specific mutations through initial tissue sequencing to guide ctDNA monitoring, and tumor-agnostic approaches, which utilize predefined panels to detect common cancer-associated genomic or epigenomic alterations directly from plasma without prior tissue analysis. The debate over which is superior in terms of sensitivity, specificity, cost-effectiveness, and clinical feasibility remains unresolved.</p><p><strong>Design: </strong>This review summarizes studies published up to November 2024, exploring the utility and performance of tumor-informed and tumor-agnostic approaches for ctDNA analysis in CRC. We evaluate the strengths and limitations of each methodology, focusing on sensitivity, specificity, and clinical outcomes.</p><p><strong>Results: </strong>Both strategies demonstrate clinical utility in postoperative risk stratification and guiding adjuvant chemotherapy decisions in CRC patients. Tumor-informed approaches generally exhibit superior sensitivity and specificity for recurrence prediction, attributed to their personalized tumor profile designs. However, these methods are limited by the need for prior tissue sequencing and higher associated costs. In contrast, tumor-agnostic approaches offer broader applicability due to their reliance on plasma-only analysis, although with relatively lower sensitivity. Technological advancements, including fragmentomics and multi-omic integrations, are expanding the capabilities of ctDNA-based MRD detection, enhancing the performance of both approaches.</p><p><strong>Conclusions: </strong>While tumor-informed strategies currently offer higher precision in MRD detection, tumor-agnostic approaches are gaining traction due to their convenience and improving performance metrics. The integration of novel technologies in ongoing clinical trials may redefine the optimal approach for MRD detection in CRC, paving the way for more personalized and adaptive patient management strategies.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142827223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-11DOI: 10.1016/j.annonc.2024.11.017
L Goyal, D DiToro, A Hollebecque, J A Bridgewater, M Shimura, A Kano, S Okamura, J L Silhavy, V Wacheck, A Halim, F Meric-Bernstam
{"title":"Genomic correlates of response and resistance to the irreversible FGFR1-4 inhibitor futibatinib based on biopsy and circulating tumor DNA profiling.","authors":"L Goyal, D DiToro, A Hollebecque, J A Bridgewater, M Shimura, A Kano, S Okamura, J L Silhavy, V Wacheck, A Halim, F Meric-Bernstam","doi":"10.1016/j.annonc.2024.11.017","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.11.017","url":null,"abstract":"<p><strong>Background: </strong>Futibatinib is the only covalent inhibitor of FGFR1-4 to gain regulatory approval in oncology. Here, we present genomic analyses of tissue biopsies and circulating tumor DNA (ctDNA) from patients with one of nearly 20 tumor types treated with futibatinib in the phase I/II FOENIX study.</p><p><strong>Patients and methods: </strong>Eligible patients included those with ctDNA samples collected per protocol at baseline and/or progression on futibatinib in the phase Ib portion of the study for FGF/FGFR-altered advanced solid tumors or the phase II portion of the study for FGFR2 fusion/rearrangement-positive cholangiocarcinoma. Assessments included analytical concordance between tumor and ctDNA analyses for detection of FGFR alterations, association of ctDNA-detected co-occurring genomic alterations with response to futibatinib, and determination of patterns of acquired resistance following progression on futibatinib.</p><p><strong>Results: </strong>Among 300 patients treated with futibatinib, 226 were eligible for this analysis, including 139 (62%) with cholangiocarcinoma. Among patients with known FGFR2 fusions/rearrangements, FGFR1 fusions, FGFR3 fusions, or FGFR2 amplifications per tissue analysis, detection rates in ctDNA for these aberrations were 84%, 0%, 11%, and 59%, respectively. Objective response rates on futibatinib were not significantly different between patients with TP53 altered versus unaltered solid tumors; progression-free survival was reduced in patients with CDKN2B altered versus unaltered cholangiocarcinoma (median, 4.8 versus 11.0 months; P=0.03). Acquired resistance to futibatinib was frequently polyclonal and driven by an array of mutations within the relevant FGFR kinase domain, predominantly V565L, V565F, and N550K variants.</p><p><strong>Conclusions: </strong>In this largest and most systematic analysis of acquired resistance to an FGFR inhibitor from prospective clinical trials, emergence of secondary FGFR2 kinase domain mutations was observed in most patients receiving clinical benefit to futibatinib. ctDNA analysis shows clinically relevant potential as a noninvasive method for assessing genomic profiles, identifying patients who may benefit from FGFR inhibitor treatment, and exploring acquired resistance mechanisms.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821787","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-11DOI: 10.1016/j.annonc.2024.12.003
R J Motzer, C Porta, M Eto, T E Hutson, S Y Rha, J R Merchan, E Winquist, H Gurney, V Grünwald, S George, J Markensohn, J E Burgents, R Cristescu, P Sachdev, Y Narita, J Huang, Z Zhao, C E Okpara, Y Minoshima, T K Choueiri
{"title":"Biomarker analyses from the phase 3 randomized CLEAR trial: Lenvatinib plus pembrolizumab versus sunitinib in advanced renal cell carcinoma.","authors":"R J Motzer, C Porta, M Eto, T E Hutson, S Y Rha, J R Merchan, E Winquist, H Gurney, V Grünwald, S George, J Markensohn, J E Burgents, R Cristescu, P Sachdev, Y Narita, J Huang, Z Zhao, C E Okpara, Y Minoshima, T K Choueiri","doi":"10.1016/j.annonc.2024.12.003","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.003","url":null,"abstract":"<p><strong>Background: </strong>In CLEAR, lenvatinib + pembrolizumab (L+P) significantly improved efficacy versus sunitinib in first-line treatment of patients with advanced renal cell carcinoma (RCC). We report results from CLEAR biomarker analyses.</p><p><strong>Methods: </strong>PD-L1 immunohistochemistry (IHC) and next-generation sequencing assays (whole exome sequencing/RNA-sequencing) were performed on archival tumor specimens. For IHC-derived/RNA-sequencing analyses, a continuous analysis was performed adjusting by Karnofsky performance status (KPS) score for: PD-L1 CPS versus best overall response (BOR)/PFS; and each gene-signature score (T-cell inflamed gene-expression profile [Tcell<sub>inf</sub>GEP]/non-Tcell<sub>inf</sub>GEP signatures including proliferation and angiogenesis) versus BOR/PFS. Association between mutation status of RCC driver genes and PFS were analyzed for genes for which ≥20 patients per arm had oncogenic alterations. Association of molecular subtypes with outcome was evaluated with baseline KPS adjustments. The set of biomarkers evaluated and statistical significance criteria for PD-L1 CPS, gene signature scores, and molecular subtypes were pre-specified.</p><p><strong>Results: </strong>Within-arm analyses using continuous values showed no association between PD-L1 levels and BOR/PFS for either treatment. PFS hazard ratios between arms were similar regardless of mutant or wildtype subgroups of RCC driver genes (VHL, PBRM1, SETD2, BAP1, KDM5C). No associations between PFS and gene-signature scores were observed for L+P. With sunitinib, high proliferation and MYC signature scores showed shorter PFS; high angiogenesis and microvessel density signature scores showed longer PFS. Six new molecular subtypes were defined. Tumors of patients with favorable/intermediate risk were enriched in angiogenesis and angiogenesis/stromal clusters; those with poor risk were enriched in proliferative and unclassified (low-Tcell<sub>inf</sub>GEP/low-angiogenesis/low-proliferation) clusters. No association between molecular subtypes and PFS for L+P/sunitinib was observed (after adjustment for KPS and gene signatures that were individually associated with PFS).</p><p><strong>Conclusions: </strong>Improvements in ORR and PFS for L+P versus sunitinib in aRCC were observed consistently across a range of biomarker subgroups defined using RCC driver mutations, PD-L1, gene expression signatures, and molecular subtypes.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-09DOI: 10.1016/j.annonc.2024.12.002
L Zhou, K W Yang, S Zhang, X Q Yan, S M Li, H Y Xu, J Li, Y Q Liu, B X Tang, Z H Chi, L Si, C L Cui, H Q Guo, Z S He, J Guo, X N Sheng
{"title":"Disitamab vedotin plus toripalimab in patients with locally advanced or metastatic urothelial carcinoma (RC48-C014): a phase 1b/2 dose-escalation and dose-expansion study.","authors":"L Zhou, K W Yang, S Zhang, X Q Yan, S M Li, H Y Xu, J Li, Y Q Liu, B X Tang, Z H Chi, L Si, C L Cui, H Q Guo, Z S He, J Guo, X N Sheng","doi":"10.1016/j.annonc.2024.12.002","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.002","url":null,"abstract":"<p><strong>Background: </strong>HER2-targeted antibody-drug conjugates (ADCs) such as disitamab vedotin (DV) and trastuzumab deruxtecan (T-Dxd) have emerged as effective treatment options and received regulatory approvals for HER2 expressing locally advanced or metastatic urothelial carcinoma (la/mUC). In addition, ADCs in combination with immunotherapy have demonstrated anti-tumor activity. The current study aimed to evaluate the combination of DV and toripalimab in patients with la/mUC.</p><p><strong>Patients and methods: </strong>This open-label phase 1b/2 study enrolled patients with untreated or chemo-refractory la/mUC. During the dose escalation phase, DV was administered at escalating doses of 1.5 and 2.0 mg/kg in combination with toripalimab 3.0 mg/kg once every two weeks. Primary endpoints were safety and the recommended phase 2 dose (RP2D). Secondary endpoints included objective response rate (ORR), progression-free survival (PFS), and overall survival (OS).</p><p><strong>Results: </strong>From August 2020 to December 2021, a total of forty-one patients were enrolled, including six in the dose-escalation phase, and thirty-five in the dose-expansion phase. Sixty-one percent of patients were treatment naïve. No dose-limiting toxicity was observed. The RP2D was determined as DV (2.0 mg/kg) plus toripalimab (3.0 mg/kg). By the data cutoff date of March 1, 2024, the confirmed ORR was 73.2%. The median PFS was 9.3 months, and the median OS was 33.1 months. The most common treatment-related adverse events (TRAEs) were aspartate aminotransferase increased (65.9%), alanine aminotransferase increased (63.4%), and peripheral sensory neuropathy (63.4%). Grade 3 or higher TRAEs occurred in 51.2% of patients, with the most common being gamma-glutamyltransferase increased (12.2%), asthenia (9.8%), and alanine aminotransferase increased (7.3%). One treatment-related death (due to pneumonitis) was reported.</p><p><strong>Conclusions: </strong>The combination of DV and toripalimab demonstrated promising response rate and overall survival results with a manageable safety profile in HER2 unselected la/mUC patients. This combination represents a promising first-line option for la/mUC. Randomized phase III study is currently ongoing.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142811734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-09DOI: 10.1016/j.annonc.2024.10.826
A Bercz, J J Smith, P B Romesser
{"title":"Reply to the Letter to the Editor 'Circulating tumor DNA after definitive therapy for locally advanced rectal cancer' by Drs Sorscher and Rocha Lima.","authors":"A Bercz, J J Smith, P B Romesser","doi":"10.1016/j.annonc.2024.10.826","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.10.826","url":null,"abstract":"","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142806073","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-06DOI: 10.1016/j.annonc.2024.12.001
A González-Martín, I A Malinowska, B J Monk
{"title":"Reply to Letter to the Editor \"Long-term outcomes in the PRIMA trial: a closer look at PFS and OS\" by Wu et al.","authors":"A González-Martín, I A Malinowska, B J Monk","doi":"10.1016/j.annonc.2024.12.001","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.001","url":null,"abstract":"","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142794327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-03DOI: 10.1016/j.annonc.2024.11.016
A Raimondi, S Lonardi, S Murgioni, G G Cardellino, S Tamberi, A Strippoli, F Palermo, G De Manzoni, M Bencivenga, A Bittoni, C Chiodoni, D Lorenzini, K Todoerti, P Manca, S Sangaletti, M Prisciandaro, G Randon, F Nichetti, F Bergamo, S Brich, A Belfiore, A Bertolotti, D Stetco, A Guidi, T Torelli, A Vingiani, R P Joshi, M Khoshdeli, N Beaubier, M C Stumpe, F Nappo, A G Leone, C C Pircher, G Leoncini, G Sabella, L Airo' Farulla, A Alessi, F Morano, A Martinetti, M Niger, M Fassan, M Di Maio, K Kaneva, M Milione, H Nimeiri, C Sposito, L Agnelli, V Mazzaferro, M Di Bartolomeo, F Pietrantonio
{"title":"Tremelimumab and durvalumab as neoadjuvant or non-operative management strategy of patients with microsatellite instability-high resectable gastric or gastroesophageal junction adenocarcinoma: the INFINITY study by GONO.","authors":"A Raimondi, S Lonardi, S Murgioni, G G Cardellino, S Tamberi, A Strippoli, F Palermo, G De Manzoni, M Bencivenga, A Bittoni, C Chiodoni, D Lorenzini, K Todoerti, P Manca, S Sangaletti, M Prisciandaro, G Randon, F Nichetti, F Bergamo, S Brich, A Belfiore, A Bertolotti, D Stetco, A Guidi, T Torelli, A Vingiani, R P Joshi, M Khoshdeli, N Beaubier, M C Stumpe, F Nappo, A G Leone, C C Pircher, G Leoncini, G Sabella, L Airo' Farulla, A Alessi, F Morano, A Martinetti, M Niger, M Fassan, M Di Maio, K Kaneva, M Milione, H Nimeiri, C Sposito, L Agnelli, V Mazzaferro, M Di Bartolomeo, F Pietrantonio","doi":"10.1016/j.annonc.2024.11.016","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.11.016","url":null,"abstract":"<p><strong>Background: </strong>In resectable gastric/gastroesophageal junction adenocarcinoma (GAC/GEJAC), microsatellite instability (MSI-H) confers improved survival, but limited benefit from chemotherapy. Immunotherapy may eliminate the need of chemotherapy or surgery.</p><p><strong>Patients and methods: </strong>INFINITY is a multicentre, multicohort phase II trial (NCT04817826) investigating in Cohort 1 the activity and safety of tremelimumab+durvalumab (T300/D) as neoadjuvant treatment for dMMR/MSI-H, resectable GAC/GEJAC. Primary endpoint was pathologic complete response (pCR) rate; Secondary endpoints: progression-free survival (PFS), overall survival (OS), quality of life (QoL), and translational analyses. In Cohort 2, the activity and safety of T300/D was explored as definitive treatment in patients achieving clinical complete response (cCR). Primary endpoint was 2-year cCR rate, Secondary endpoints: PFS, OS, QoL, gastrectomy-free survival (GFS) and translational analyses.</p><p><strong>Results: </strong>In Cohort 1, 18 patients were recruited and 15 evaluable. pCR and major-complete pathological response were 60% and 80%. Since pCR rate in T4 tumors was 17%, this subgroup of patients was excluded from enrollment in Cohort 2. At 28.1 months median follow-up, 24-month GC-specific PFS and OS rates were 85% and 92%. In Cohort 2, 18 patients were enrolled and 17 evaluable, 13 had cCR and started non-operative management (NOM). At 11.5 months median follow up, 1 patient had local regrowth and underwent salvage surgery, 12-month GFS was 64.2%.</p><p><strong>Conclusions: </strong>The INFINITY study provided promising activity results of a chemo-free T300/D combination regimen as pre-operative treatment in dMMR/MSI GAC/GEJAC and the first available feasibility results of a NOM strategy in this disease setting, worth of further validation in larger cohorts.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142783900","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Annals of OncologyPub Date : 2024-12-03DOI: 10.1016/j.annonc.2024.11.014
A Di Federico, L Hong, A Elkrief, R Thummalapalli, A J Cooper, B Ricciuti, S Digumarthy, J V Alessi, P Gogia, F Pecci, M Makarem, M M Gandhi, E Garbo, A Saini, A De Giglio, V Favorito, S Scalera, L Cipriani, D Marinelli, D Haradon, T Nguyen, J Haradon, E Voligny, V Vaz, F Gelsomino, F Sperandi, B Melotti, M Ladanyi, J Zhang, D L Gibbons, J V Heymach, M Nishino, J Lindsay, S J Rodig, K Pfaff, L M Sholl, X Wang, B E Johnson, P A Jänne, N Rekhtman, M Maugeri-Saccà, R S Heist, A Ardizzoni, M M Awad, K C Arbour, A J Schoenfeld, N I Vokes, J Luo
{"title":"Lung adenocarcinomas with mucinous histology: clinical, genomic, and immune microenvironment characterization and outcomes to immunotherapy-based treatments and KRAS<sup>G12C</sup> inhibitors.","authors":"A Di Federico, L Hong, A Elkrief, R Thummalapalli, A J Cooper, B Ricciuti, S Digumarthy, J V Alessi, P Gogia, F Pecci, M Makarem, M M Gandhi, E Garbo, A Saini, A De Giglio, V Favorito, S Scalera, L Cipriani, D Marinelli, D Haradon, T Nguyen, J Haradon, E Voligny, V Vaz, F Gelsomino, F Sperandi, B Melotti, M Ladanyi, J Zhang, D L Gibbons, J V Heymach, M Nishino, J Lindsay, S J Rodig, K Pfaff, L M Sholl, X Wang, B E Johnson, P A Jänne, N Rekhtman, M Maugeri-Saccà, R S Heist, A Ardizzoni, M M Awad, K C Arbour, A J Schoenfeld, N I Vokes, J Luo","doi":"10.1016/j.annonc.2024.11.014","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.11.014","url":null,"abstract":"<p><strong>Background: </strong>Approximately 10% of lung adenocarcinomas (LUAD) have mucinous histology (LUADMuc), which is associated to a light/absent smoking history and a high prevalence of KRAS mutations. We sought to characterize LUADMuc by comparing it to LUAD without mucinous histology (LUADnon-muc) and determine the relative benefit of current treatments.</p><p><strong>Patients and methods: </strong>Patients with LUAD from five institutions and TCGA PanCancer Atlas classified as LUADMuc or LUADnon-muc were included. Clinicopathologic, genomic, immunophenotypic, transcriptional features, and treatment outcomes were compared between LUADMuc and LUADnon-muc.</p><p><strong>Results: </strong>Of 4,082 patients with LUAD, 9.9% had LUAD<sup>Muc</sup>. Compared to LUAD<sup>non-muc</sup>, patients with LUAD<sup>Muc</sup> had a lighter smoking history (median: 15 vs 20 pack-years, P=0.008), lower PD-L1 TPS (median: 0% vs 5%, P<0.0001), and lower tumor mutation burden (median: 6.8 vs 8.5 mutations/megabase, P<0.0001). Mutations in KRAS, NKX2-1 (TTF-1), STK11, SMARCA4, GNAS, and ALK rearrangements were enriched in LUAD<sup>Muc</sup>, while TP53, EGFR, BRAF, and MET mutations were enriched in LUAD<sup>non-muc</sup>. At stage IV diagnosis, LUAD<sup>Muc</sup> was more likely to have contralateral lung metastasis (55.2% vs 36.9%, P<0.0001) and less likely to have brain metastases (23.3% vs 41.9%, P<0.0001). Compared to LUAD<sup>non-muc</sup>, LUAD<sup>Muc</sup> cases showed lower intratumor CD8<sup>+</sup>, PD-1<sup>+</sup>, CD8<sup>+</sup>PD-1<sup>+</sup>, and FOXP3<sup>+</sup> cells. Among metastatic cases receiving ICIs, compared to LUAD<sup>non-muc</sup> (N=1,511), LUAD<sup>Muc</sup> (N=112) had lower objective response rate (ORR, 8.4% vs 25.9%, P<0.0001), and shorter median progression-free survival (mPFS, 2.6 vs 3.9 months, P<0.0001) and overall survival (mOS, 9.9 vs 17.2 months, P<0.0001). Similarly, patients with LUAD<sup>Muc</sup> had worse outcomes to chemo-immunotherapy. LUAD<sup>Muc</sup> (N=18) and LUAD<sup>non-muc</sup> (N=150) had similar ORR (16.7% vs 34.9%, P=0.12) and mPFS (4.6 vs 5.6 months, P=0.17) to treatment with KRAS<sup>G12C</sup> inhibitors, but LUAD<sup>Muc</sup> had shorter mOS (6.8 vs 10.8 months, P=0.018).</p><p><strong>Conclusion: </strong>LUAD<sup>Muc</sup> represents a distinct LUAD subpopulation with unique clinicopathologic, genomic, immunophenotypic, and transcriptional features, achieving worse outcomes to standard immunotherapy-based treatments.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142783899","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}