Pub Date : 2025-01-01Epub Date: 2024-11-26DOI: 10.1016/j.annonc.2024.09.007
M Roulleaux Dugage, O Lambotte, X Mariette, A Marabelle
{"title":"Breaking the link: IL-6 targeting to uncouple toxicity and efficacy in patients treated with immune checkpoint blockade.","authors":"M Roulleaux Dugage, O Lambotte, X Mariette, A Marabelle","doi":"10.1016/j.annonc.2024.09.007","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.09.007","url":null,"abstract":"","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":"36 1","pages":"6-9"},"PeriodicalIF":56.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142926308","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}
Pub Date : 2025-01-01Epub Date: 2024-10-10DOI: 10.1016/j.annonc.2024.10.002
D Lorusso, N Colombo, C Dubot, M V Cáceres, K Hasegawa, R Shapira-Frommer, P Salman, E Yañez, M Gümüş, M Olivera, V Samouëlian, V Castonguay, A Arkhipov, K Li, S Toker, C Tekin, K S Tewari, B J Monk
Background: In KEYNOTE-826 (NCT03635567), pembrolizumab plus chemotherapy (±bevacizumab) significantly improved overall survival (OS) and progression-free survival (PFS) in patients with persistent, recurrent, or metastatic cervical cancer. This exploratory analysis examined outcomes in patient subgroups defined by bevacizumab use.
Patients and methods: Eligible adult patients had persistent, recurrent, or metastatic squamous cell carcinoma, adenosquamous carcinoma, or adenocarcinoma of the cervix not previously treated with chemotherapy and not amenable to curative treatment; measurable disease per RECIST v1.1; and an Eastern Cooperative Oncology Group performance status ≤1. Patients were randomly allocated 1 : 1 to pembrolizumab 200 mg every 3 weeks or placebo for up to 35 cycles plus chemotherapy (±bevacizumab 15 mg/kg). Dual primary endpoints were OS and PFS per RECIST v1.1 by investigator assessment. Outcomes were assessed in subgroups defined by bevacizumab use. Hazard ratios (HRs) and 95% confidence intervals (CIs) were based on a stratified Cox regression model.
Results: A total of 617 patients were randomly assigned [pembrolizumab arm, n = 308 (63.6% with bevacizumab); placebo arm, n = 309 (62.5% with bevacizumab)]. The most common reason for bevacizumab exclusion was medical contraindication (75.9%). Among patients who received bevacizumab, HRs (95% CIs) for PFS favored the pembrolizumab arm in the programmed cell death-ligand 1 combined positive score ≥1 [0.56 (0.43-0.73)] and all-comer [0.57 (0.45-0.73)] populations; OS results were 0.60 (0.45-0.79) and 0.61 (0.47-0.80), respectively. Among patients who did not receive bevacizumab, HRs (95% CIs) for PFS also favored the pembrolizumab arm in the programmed cell death-ligand 1 combined positive score ≥1 [0.61 (0.44-0.85)] and all-comer [0.69 (0.50-0.94)] populations; OS results were 0.61 (0.44-0.85) and 0.67 (0.49-0.91), respectively. Among patients who received bevacizumab, grade ≥3 treatment-related adverse events occurred in 74.0% of patients in the pembrolizumab arm and 66.8% in the placebo arm.
Conclusion: Pembrolizumab plus chemotherapy prolonged PFS and OS and had manageable safety compared with placebo plus chemotherapy in patient subgroups defined by bevacizumab use.
{"title":"Pembrolizumab plus chemotherapy for advanced and recurrent cervical cancer: final analysis according to bevacizumab use in the randomized KEYNOTE-826 study.","authors":"D Lorusso, N Colombo, C Dubot, M V Cáceres, K Hasegawa, R Shapira-Frommer, P Salman, E Yañez, M Gümüş, M Olivera, V Samouëlian, V Castonguay, A Arkhipov, K Li, S Toker, C Tekin, K S Tewari, B J Monk","doi":"10.1016/j.annonc.2024.10.002","DOIUrl":"10.1016/j.annonc.2024.10.002","url":null,"abstract":"<p><strong>Background: </strong>In KEYNOTE-826 (NCT03635567), pembrolizumab plus chemotherapy (±bevacizumab) significantly improved overall survival (OS) and progression-free survival (PFS) in patients with persistent, recurrent, or metastatic cervical cancer. This exploratory analysis examined outcomes in patient subgroups defined by bevacizumab use.</p><p><strong>Patients and methods: </strong>Eligible adult patients had persistent, recurrent, or metastatic squamous cell carcinoma, adenosquamous carcinoma, or adenocarcinoma of the cervix not previously treated with chemotherapy and not amenable to curative treatment; measurable disease per RECIST v1.1; and an Eastern Cooperative Oncology Group performance status ≤1. Patients were randomly allocated 1 : 1 to pembrolizumab 200 mg every 3 weeks or placebo for up to 35 cycles plus chemotherapy (±bevacizumab 15 mg/kg). Dual primary endpoints were OS and PFS per RECIST v1.1 by investigator assessment. Outcomes were assessed in subgroups defined by bevacizumab use. Hazard ratios (HRs) and 95% confidence intervals (CIs) were based on a stratified Cox regression model.</p><p><strong>Results: </strong>A total of 617 patients were randomly assigned [pembrolizumab arm, n = 308 (63.6% with bevacizumab); placebo arm, n = 309 (62.5% with bevacizumab)]. The most common reason for bevacizumab exclusion was medical contraindication (75.9%). Among patients who received bevacizumab, HRs (95% CIs) for PFS favored the pembrolizumab arm in the programmed cell death-ligand 1 combined positive score ≥1 [0.56 (0.43-0.73)] and all-comer [0.57 (0.45-0.73)] populations; OS results were 0.60 (0.45-0.79) and 0.61 (0.47-0.80), respectively. Among patients who did not receive bevacizumab, HRs (95% CIs) for PFS also favored the pembrolizumab arm in the programmed cell death-ligand 1 combined positive score ≥1 [0.61 (0.44-0.85)] and all-comer [0.69 (0.50-0.94)] populations; OS results were 0.61 (0.44-0.85) and 0.67 (0.49-0.91), respectively. Among patients who received bevacizumab, grade ≥3 treatment-related adverse events occurred in 74.0% of patients in the pembrolizumab arm and 66.8% in the placebo arm.</p><p><strong>Conclusion: </strong>Pembrolizumab plus chemotherapy prolonged PFS and OS and had manageable safety compared with placebo plus chemotherapy in patient subgroups defined by bevacizumab use.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":"65-75"},"PeriodicalIF":56.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142405946","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}
Pub Date : 2025-01-01Epub Date: 2024-09-17DOI: 10.1016/j.annonc.2024.09.008
C S Groeneveld, C Pfister, S Culine, V Harter, C Krucker, J Fontugne, V Dixon, N Sirab, I Bernard-Pierrot, A de Reyniès, F Radvanyi, Y Allory
Background: Neoadjuvant chemotherapy (NAC) is the standard treatment for muscle-invasive bladder cancer (MIBC), yet 40% of patients progress, emphasizing the need for biomarkers predictive for response or chemoresistance. Gene expression-based subtypes may serve as biomarkers, though which subtypes will respond, notably when it comes to the basal subtype, remains contentious.
Patients and methods: This post hoc study analyzed 300 NAC-treated patients enrolled in the GETUG/AFU VESPER trial, with transurethral diagnostic formalin-fixed paraffin-embedded tissue which underwent pathological review before being sequenced. 'Mixed' subtype was defined for tumors displaying at least two different Consensus molecular subtypes in separate regions. We evaluated the association between molecular subtypes and outcome after NAC. Tumors with remaining tissue at cystectomy (n = 83) were compared with pre-treatment tumors.
Results: Cases were classified basal/squamous (Ba/Sq) (n = 84), luminal unstable (n = 57), stroma-rich (n = 53), mixed (n = 48), luminal papillary (n = 39), luminal non-specific (n = 18), and neuroendocrine-like (n = 1), with 30/48 mixed cases including a Ba/Sq component. Compared with other molecular subtypes in a multivariate Cox model, Ba/Sq (pure or mixed) patients had an increased hazard ratio (HR) of progression-free survival [HR 2.0, 95% confidence interval (CI) 1.36-3.0]. Mixed tumors were associated with decreased metabolic activity that could account for chemoresistance. Ba/Sq and mixed non-responders mostly maintained their subtype at cystectomy and have fewer myeloid dendritic cells after NAC. Tumors classified luminal papillary at transurethral resection of the urinary bladder tumor exhibited an increase in T CD4+ and macrophage signatures after NAC. Other subtypes did not show significant immune changes after NAC. Our study design relied on detailed pathological review, which precluded evaluating the mixed subtype in published datasets. Furthermore, the sample size for post-NAC analyses constrained the statistical power of these findings.
Conclusions: Our findings underscore the importance of recognizing intra-tumor heterogeneity in MIBC and its role in chemoresistance associated with Ba/Sq subtype, and provide valuable insights that could help future treatment development and improve patient outcomes.
{"title":"Basal/squamous and mixed subtype bladder cancers present poor outcomes after neoadjuvant chemotherapy in the VESPER trial.","authors":"C S Groeneveld, C Pfister, S Culine, V Harter, C Krucker, J Fontugne, V Dixon, N Sirab, I Bernard-Pierrot, A de Reyniès, F Radvanyi, Y Allory","doi":"10.1016/j.annonc.2024.09.008","DOIUrl":"10.1016/j.annonc.2024.09.008","url":null,"abstract":"<p><strong>Background: </strong>Neoadjuvant chemotherapy (NAC) is the standard treatment for muscle-invasive bladder cancer (MIBC), yet 40% of patients progress, emphasizing the need for biomarkers predictive for response or chemoresistance. Gene expression-based subtypes may serve as biomarkers, though which subtypes will respond, notably when it comes to the basal subtype, remains contentious.</p><p><strong>Patients and methods: </strong>This post hoc study analyzed 300 NAC-treated patients enrolled in the GETUG/AFU VESPER trial, with transurethral diagnostic formalin-fixed paraffin-embedded tissue which underwent pathological review before being sequenced. 'Mixed' subtype was defined for tumors displaying at least two different Consensus molecular subtypes in separate regions. We evaluated the association between molecular subtypes and outcome after NAC. Tumors with remaining tissue at cystectomy (n = 83) were compared with pre-treatment tumors.</p><p><strong>Results: </strong>Cases were classified basal/squamous (Ba/Sq) (n = 84), luminal unstable (n = 57), stroma-rich (n = 53), mixed (n = 48), luminal papillary (n = 39), luminal non-specific (n = 18), and neuroendocrine-like (n = 1), with 30/48 mixed cases including a Ba/Sq component. Compared with other molecular subtypes in a multivariate Cox model, Ba/Sq (pure or mixed) patients had an increased hazard ratio (HR) of progression-free survival [HR 2.0, 95% confidence interval (CI) 1.36-3.0]. Mixed tumors were associated with decreased metabolic activity that could account for chemoresistance. Ba/Sq and mixed non-responders mostly maintained their subtype at cystectomy and have fewer myeloid dendritic cells after NAC. Tumors classified luminal papillary at transurethral resection of the urinary bladder tumor exhibited an increase in T CD4+ and macrophage signatures after NAC. Other subtypes did not show significant immune changes after NAC. Our study design relied on detailed pathological review, which precluded evaluating the mixed subtype in published datasets. Furthermore, the sample size for post-NAC analyses constrained the statistical power of these findings.</p><p><strong>Conclusions: </strong>Our findings underscore the importance of recognizing intra-tumor heterogeneity in MIBC and its role in chemoresistance associated with Ba/Sq subtype, and provide valuable insights that could help future treatment development and improve patient outcomes.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":"89-98"},"PeriodicalIF":56.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142279653","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}
Pub Date : 2024-12-18DOI: 10.1016/j.annonc.2024.12.008
T K Choueiri, K Penkov, H Uemura, M T Campbell, S Pal, C Kollmannsberger, J L Lee, B Venugopal, A J M van den Eertwegh, S Negrier, H Gurney, L Albiges, R Berger, J B A G Haanen, V Oyervides Juárez, B I Rini, J Larkin, F Nolè, M Schmidinger, M B Atkins, Y Tomita, B Ellers-Lenz, J Hoffman, R Sandner, J Wang, A di Pietro, R J Motzer
Background: In the phase 3 JAVELIN Renal 101 trial (NCT02684006), first-line treatment with avelumab + axitinib resulted in significantly longer progression-free survival (PFS) and a higher objective response rate (ORR) vs sunitinib in patients with advanced renal cell carcinoma (aRCC). We report the final analysis, including the primary analysis of overall survival (OS).
Patients and methods: Patients with untreated aRCC (any prognostic risk score) were enrolled. Primary endpoints were OS and PFS in the programmed death ligand 1-positive (PD-L1+) population. ORR, duration of response (DOR), safety, and patient-reported outcomes (PROs) were also assessed.
Results: Minimum follow-up was 68 months in all patients. Median OS (95% CI) with avelumab + axitinib vs sunitinib, respectively, was 43.2 (36.5-51.7) vs 36.2 (29.8-44.2) months in the PD-L1+ population (hazard ratio [HR], 0.86 [95% CI, 0.701-1.057]; P=0.0755) and 44.8 (39.7-51.1) vs 38.9 (31.4-45.2) months in the overall population (HR, 0.88 [95% CI, 0.749-1.039]; P=0.0669). Investigator-assessed PFS remained prolonged with avelumab + axitinib vs sunitinib (5-year event-free rate [95% CI] in the overall population, 12.0% [8.9%-15.6%] vs 4.4% [2.5%-7.3%]). ORR (95% CI) in the overall population was 59.7% (55.0%-64.3%) with avelumab + axitinib vs 32.0% (27.7%-36.5%) with sunitinib; DOR (95% CI) was ≥5 years in 16.4% (12.0%-21.4%) vs 9.2% (4.6%-15.7%), respectively. Rates of grade ≥3 treatment-related adverse events were 66.8% vs 61.5%, respectively. PROs were similar between arms.
Conclusions: JAVELIN Renal 101 provides the longest follow-up to date for immune checkpoint inhibitor + tyrosine kinase inhibitor combination treatment from a phase 3 trial in aRCC. OS analyses favored avelumab + axitinib vs sunitinib but did not reach statistical significance; subsequent treatment may have impacted results. Avelumab + axitinib provided long-term efficacy benefits vs sunitinib, including prolonged PFS, a nearly doubled ORR, and more durable responses, with a manageable long-term safety profile.
{"title":"Avelumab + axitinib vs sunitinib as first-line treatment for patients with advanced renal cell carcinoma: final analysis of the phase 3 JAVELIN Renal 101 trial.","authors":"T K Choueiri, K Penkov, H Uemura, M T Campbell, S Pal, C Kollmannsberger, J L Lee, B Venugopal, A J M van den Eertwegh, S Negrier, H Gurney, L Albiges, R Berger, J B A G Haanen, V Oyervides Juárez, B I Rini, J Larkin, F Nolè, M Schmidinger, M B Atkins, Y Tomita, B Ellers-Lenz, J Hoffman, R Sandner, J Wang, A di Pietro, R J Motzer","doi":"10.1016/j.annonc.2024.12.008","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.008","url":null,"abstract":"<p><strong>Background: </strong>In the phase 3 JAVELIN Renal 101 trial (NCT02684006), first-line treatment with avelumab + axitinib resulted in significantly longer progression-free survival (PFS) and a higher objective response rate (ORR) vs sunitinib in patients with advanced renal cell carcinoma (aRCC). We report the final analysis, including the primary analysis of overall survival (OS).</p><p><strong>Patients and methods: </strong>Patients with untreated aRCC (any prognostic risk score) were enrolled. Primary endpoints were OS and PFS in the programmed death ligand 1-positive (PD-L1+) population. ORR, duration of response (DOR), safety, and patient-reported outcomes (PROs) were also assessed.</p><p><strong>Results: </strong>Minimum follow-up was 68 months in all patients. Median OS (95% CI) with avelumab + axitinib vs sunitinib, respectively, was 43.2 (36.5-51.7) vs 36.2 (29.8-44.2) months in the PD-L1+ population (hazard ratio [HR], 0.86 [95% CI, 0.701-1.057]; P=0.0755) and 44.8 (39.7-51.1) vs 38.9 (31.4-45.2) months in the overall population (HR, 0.88 [95% CI, 0.749-1.039]; P=0.0669). Investigator-assessed PFS remained prolonged with avelumab + axitinib vs sunitinib (5-year event-free rate [95% CI] in the overall population, 12.0% [8.9%-15.6%] vs 4.4% [2.5%-7.3%]). ORR (95% CI) in the overall population was 59.7% (55.0%-64.3%) with avelumab + axitinib vs 32.0% (27.7%-36.5%) with sunitinib; DOR (95% CI) was ≥5 years in 16.4% (12.0%-21.4%) vs 9.2% (4.6%-15.7%), respectively. Rates of grade ≥3 treatment-related adverse events were 66.8% vs 61.5%, respectively. PROs were similar between arms.</p><p><strong>Conclusions: </strong>JAVELIN Renal 101 provides the longest follow-up to date for immune checkpoint inhibitor + tyrosine kinase inhibitor combination treatment from a phase 3 trial in aRCC. OS analyses favored avelumab + axitinib vs sunitinib but did not reach statistical significance; subsequent treatment may have impacted results. Avelumab + axitinib provided long-term efficacy benefits vs sunitinib, including prolonged PFS, a nearly doubled ORR, and more durable responses, with a manageable long-term safety profile.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870677","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}
Pub Date : 2024-12-18DOI: 10.1016/j.annonc.2024.12.011
L Goyal, D DiToro, F Facchinetti, E E Martin, P Peng, I Baiev, R Iyer, J Maurer, S Reyes, K Zhang, U Majeed, J E Berchuck, C T Chen, C Walmsley, C Pinto, D Vasseur, J D Gordan, K Mody, M Borad, T Karasic, N Damjanov, B P Danysh, E Wehrenberg-Klee, A R Kambadakone, S K Saha, I D Hoffman, K J Nelson, S Iyer, X Qiang, C Sun, H Wang, L Li, M Javle, B Lin, W Harris, A X Zhu, J M Cleary, K T Flaherty, T Harris, R T Shroff, I Leshchiner, L Parida, R K Kelley, J Fan, J R Stone, N V Uboha, H Hirai, H Sootome, F Wu, D C Bensen, A Hollebecque, L Friboulet, J K Lennerz, G Getz, D Juric
Background: Fibroblast growth factor receptor (FGFR) inhibitors have significantly improved outcomes for patients with FGFR-altered cholangiocarcinoma, leading to their regulatory approval in multiple countries. However, as with many targeted therapies, acquired resistance limits their efficacy. A comprehensive, multimodal approach is crucial to characterizing resistance patterns to FGFR inhibitors.
Patients and methods: This study integrated data from six investigative strategies: cell-free DNA, tissue biopsy, rapid autopsy, statistical genomics, in vitro and in vivo studies, and pharmacology. We characterized the diversity, clonality, frequency, and mechanisms of acquired resistance to FGFR inhibitors in patients with FGFR-altered cholangiocarcinoma. Clinical samples were analyzed longitudinally as part of routine care across 10 institutions.
Results: Among 138 patients evaluated, 77 met eligibility, yielding a total of 486 clinical samples. Patients with clinical benefit exhibited a significantly higher rate of FGFR2 kinase domain mutations compared to those without clinical benefit (65% vs 10%, p<0.0001). We identified 26 distinct FGFR2 kinase domain mutations, with 63% of patients harboring multiple. While IC50 assessments indicated strong potency of pan-FGFR inhibitors against common resistance mutations, pharmacokinetic studies revealed that low clinically achievable drug concentrations may underly polyclonal resistance. Molecular brake and gatekeeper mutations predominated, with 94% of patients with FGFR2 mutations exhibiting one or both, whereas mutations at the cysteine residue targeted by covalent inhibitors were rare. Statistical genomics and functional studies demonstrated that mutation frequencies were driven by their combined effects on drug binding and kinase activity rather than intrinsic mutational processes.
Conclusion: Our multimodal analysis led to a model characterizing the biology of acquired resistance, informing the rational design of next-generation FGFR inhibitors. FGFR inhibitors should be small, high-affinity, and selective for specific FGFR family members. Tinengotinib, a novel small molecule inhibitor with these characteristics, exhibited preclinical and clinical activity against key resistance mutations. This integrated approach offers a blueprint for advancing drug resistance research across cancer types.
背景:成纤维细胞生长因子受体(FGFR)抑制剂显著改善了FGFR改变的胆管癌患者的预后,导致其在多个国家获得监管部门的批准。然而,与许多靶向治疗一样,获得性耐药性限制了它们的疗效。一个全面的、多模式的方法对于表征对FGFR抑制剂的耐药模式至关重要。患者和方法:本研究整合了六种调查策略的数据:无细胞DNA、组织活检、快速尸检、统计基因组学、体外和体内研究以及药理学。我们描述了FGFR改变的胆管癌患者对FGFR抑制剂获得性耐药的多样性、克隆性、频率和机制。临床样本作为10家机构常规护理的一部分进行纵向分析。结果:138例患者中,77例符合入选条件,共获得486份临床样本。与没有临床获益的患者相比,有临床获益的患者表现出显著更高的FGFR2激酶结构域突变率(65% vs 10%)。结论:我们的多模态分析导致了一个表征获得性耐药生物学的模型,为下一代FGFR抑制剂的合理设计提供了信息。FGFR抑制剂应该是小的、高亲和力的、对特定FGFR家族成员有选择性的。Tinengotinib是一种具有这些特征的新型小分子抑制剂,对关键耐药突变具有临床前和临床活性。这种综合方法为推进跨癌症类型的耐药性研究提供了蓝图。
{"title":"A Model for Decoding Resistance in Precision Oncology: Acquired Resistance to FGFR inhibitors in Cholangiocarcinoma.","authors":"L Goyal, D DiToro, F Facchinetti, E E Martin, P Peng, I Baiev, R Iyer, J Maurer, S Reyes, K Zhang, U Majeed, J E Berchuck, C T Chen, C Walmsley, C Pinto, D Vasseur, J D Gordan, K Mody, M Borad, T Karasic, N Damjanov, B P Danysh, E Wehrenberg-Klee, A R Kambadakone, S K Saha, I D Hoffman, K J Nelson, S Iyer, X Qiang, C Sun, H Wang, L Li, M Javle, B Lin, W Harris, A X Zhu, J M Cleary, K T Flaherty, T Harris, R T Shroff, I Leshchiner, L Parida, R K Kelley, J Fan, J R Stone, N V Uboha, H Hirai, H Sootome, F Wu, D C Bensen, A Hollebecque, L Friboulet, J K Lennerz, G Getz, D Juric","doi":"10.1016/j.annonc.2024.12.011","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.011","url":null,"abstract":"<p><strong>Background: </strong>Fibroblast growth factor receptor (FGFR) inhibitors have significantly improved outcomes for patients with FGFR-altered cholangiocarcinoma, leading to their regulatory approval in multiple countries. However, as with many targeted therapies, acquired resistance limits their efficacy. A comprehensive, multimodal approach is crucial to characterizing resistance patterns to FGFR inhibitors.</p><p><strong>Patients and methods: </strong>This study integrated data from six investigative strategies: cell-free DNA, tissue biopsy, rapid autopsy, statistical genomics, in vitro and in vivo studies, and pharmacology. We characterized the diversity, clonality, frequency, and mechanisms of acquired resistance to FGFR inhibitors in patients with FGFR-altered cholangiocarcinoma. Clinical samples were analyzed longitudinally as part of routine care across 10 institutions.</p><p><strong>Results: </strong>Among 138 patients evaluated, 77 met eligibility, yielding a total of 486 clinical samples. Patients with clinical benefit exhibited a significantly higher rate of FGFR2 kinase domain mutations compared to those without clinical benefit (65% vs 10%, p<0.0001). We identified 26 distinct FGFR2 kinase domain mutations, with 63% of patients harboring multiple. While IC50 assessments indicated strong potency of pan-FGFR inhibitors against common resistance mutations, pharmacokinetic studies revealed that low clinically achievable drug concentrations may underly polyclonal resistance. Molecular brake and gatekeeper mutations predominated, with 94% of patients with FGFR2 mutations exhibiting one or both, whereas mutations at the cysteine residue targeted by covalent inhibitors were rare. Statistical genomics and functional studies demonstrated that mutation frequencies were driven by their combined effects on drug binding and kinase activity rather than intrinsic mutational processes.</p><p><strong>Conclusion: </strong>Our multimodal analysis led to a model characterizing the biology of acquired resistance, informing the rational design of next-generation FGFR inhibitors. FGFR inhibitors should be small, high-affinity, and selective for specific FGFR family members. Tinengotinib, a novel small molecule inhibitor with these characteristics, exhibited preclinical and clinical activity against key resistance mutations. This integrated approach offers a blueprint for advancing drug resistance research across cancer types.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870671","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}
Pub Date : 2024-12-18DOI: 10.1016/j.annonc.2024.12.010
Aranzazu Fernandez-Martinez, Maki Tanioka, Sung Gwe Ahn, Paola Zagami, Tomás Pascual, Mattia Rediti, Gong Tang, Katherine A Hoadley, David Venet, Naim U Rashid, Patricia A Spears, Serena Di Cosimo, Evandro de Azambuja, Anup Choudhury, Priya Rastogi, Md N Islam, Javier Cortes, Antonio Llombart-Cussac, Sandra M Swain, Christos Sotiriou, Aleix Prat, Charles M Perou, Lisa A Carey
Background: In HER2+ early breast cancer (EBC), we investigated tumor and immune changes during neoadjuvant treatment and their impact on residual disease (RD) biology and prognostic implications across 4 neoadjuvant studies of trastuzumab with or without lapatinib, and with or without chemotherapy: CALGB 40601, PAMELA, NeoALTTO and NSABP B-41.
Patients and methods: We compared tumor and immune gene expression changes during neoadjuvant treatment and their association with with event-free survival (EFS) by uni- and multivariable Cox regression models in different cohorts and timepoints: 452 RD samples at baseline including 169 with a paired RD, and biomarker changes during neoadjuvant therapy, evaluating model performance via the c-index.
Results: Analysis of 169 paired tumor samples revealed a shift in intrinsic subtype proportions from HER2-Enriched at baseline (50.3%) to Normal-like (49.1%) followed by Luminal A (18.9%) in RD. This luminal phenotypic change was supported by decreased correlation to the HER2-Enriched centroid, ERBB2, and HER2 amplicon genes and increased correlation to the Luminal A centroid (Wilcoxon-test p-value < 0.001). Additionally, RD showed relative immune activation marked by significant increases in B cell, CD8 T cell, and NK cell signatures (Wilcoxon-test p-value < 0.05). In multivariable Cox models, intrinsic subtypes at baseline provided more prognostic information, while immune gene expression signatures provided more prognostic information in RD. Notably, the best multivariable EFS model (c-index = 0.77) integrated the IgG signature from RD samples (adjusted hazard ratio 0.45, 95% CI 0.30-0.67, adjusted p-value 0.002).
Conclusions: In patients with HER2+ EBC and RD, tumor biomarkers provide more prognostic information at baseline. In contrast, immune biomarkers perform better for EFS prognosis in RD.
{"title":"Prognostic value of residual disease (RD) biology and gene expression changes during the neoadjuvant treatment in patients with HER2+ early breast cancer (EBC).","authors":"Aranzazu Fernandez-Martinez, Maki Tanioka, Sung Gwe Ahn, Paola Zagami, Tomás Pascual, Mattia Rediti, Gong Tang, Katherine A Hoadley, David Venet, Naim U Rashid, Patricia A Spears, Serena Di Cosimo, Evandro de Azambuja, Anup Choudhury, Priya Rastogi, Md N Islam, Javier Cortes, Antonio Llombart-Cussac, Sandra M Swain, Christos Sotiriou, Aleix Prat, Charles M Perou, Lisa A Carey","doi":"10.1016/j.annonc.2024.12.010","DOIUrl":"10.1016/j.annonc.2024.12.010","url":null,"abstract":"<p><strong>Background: </strong>In HER2+ early breast cancer (EBC), we investigated tumor and immune changes during neoadjuvant treatment and their impact on residual disease (RD) biology and prognostic implications across 4 neoadjuvant studies of trastuzumab with or without lapatinib, and with or without chemotherapy: CALGB 40601, PAMELA, NeoALTTO and NSABP B-41.</p><p><strong>Patients and methods: </strong>We compared tumor and immune gene expression changes during neoadjuvant treatment and their association with with event-free survival (EFS) by uni- and multivariable Cox regression models in different cohorts and timepoints: 452 RD samples at baseline including 169 with a paired RD, and biomarker changes during neoadjuvant therapy, evaluating model performance via the c-index.</p><p><strong>Results: </strong>Analysis of 169 paired tumor samples revealed a shift in intrinsic subtype proportions from HER2-Enriched at baseline (50.3%) to Normal-like (49.1%) followed by Luminal A (18.9%) in RD. This luminal phenotypic change was supported by decreased correlation to the HER2-Enriched centroid, ERBB2, and HER2 amplicon genes and increased correlation to the Luminal A centroid (Wilcoxon-test p-value < 0.001). Additionally, RD showed relative immune activation marked by significant increases in B cell, CD8 T cell, and NK cell signatures (Wilcoxon-test p-value < 0.05). In multivariable Cox models, intrinsic subtypes at baseline provided more prognostic information, while immune gene expression signatures provided more prognostic information in RD. Notably, the best multivariable EFS model (c-index = 0.77) integrated the IgG signature from RD samples (adjusted hazard ratio 0.45, 95% CI 0.30-0.67, adjusted p-value 0.002).</p><p><strong>Conclusions: </strong>In patients with HER2+ EBC and RD, tumor biomarkers provide more prognostic information at baseline. In contrast, immune biomarkers perform better for EFS prognosis in RD.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870818","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}
Pub Date : 2024-12-18DOI: 10.1016/j.annonc.2024.12.009
P J Bröckelmann, H Müller, M Fuchs, S Gillessen, D A Eichenauer, S Borchmann, A S Jacob, K Behringer, J Momotow, J Ferdinandus, B Böll, X Yang, C Kobe, H-T Eich, C Baues, W Klapper, A Engert, P Borchmann, B von Tresckow
Background: We aimed to evaluate the correlation of progression-free (PFS) and overall survival (OS) after first-line treatment of classical Hodgkin lymphoma (HL) and the potential of PFS to serve as a surrogate parameter for OS.
Patients and methods: We analyzed individual patient data obtained during and after treatment with polychemotherapy within nine randomized phase III trials (GHSG HD7-HD15) between 01/93 - 08/18. Effects of 16 experimental treatments on PFS and OS on trial level were evaluated by estimation of the treatment effects with Cox proportional hazards (PH) regression and a linear weighted least squares regression. On the patient level, marginal Cox PH models for multiple endpoints were applied according to the Wei-Lin-Weissfeld method.
Results: At least one PFS and OS event was recorded in 1,682 and 1,064 of 10,605 patients, respectively. At trial level there was a strong correlation of treatment effects on PFS and OS (weighted Pearson r= 0.72, R2= 0.54, P< 0.001). At patient level, moderate to strong correlation of treatment effects on PFS and OS was confirmed with Pearson r ranging between 0.61 - 0.85 (each P< 0.001) and overall r= 0.74. A regression model accounting for different types of experimental treatments and historical progress over trial generations reached very strong correlation (R2= 0.93). Applying this model to data from the contemporary first-line ECHELON-1 trial allowed for prediction of OS from PFS (prognosticated ln[HR(OS)]= -0.68 as compared to observed ln[HR(0.59)]= -0.53).
Conclusion: In first-line trials of HL, PFS and OS as well as treatment effects and prognostic effects on PFS and OS are strongly correlated. PFS thereby predicts treatment effects on OS to a high degree and many years before OS can be reliably evaluated.
{"title":"Correlation Between Progression-Free and Overall Survival in Patients with Hodgkin Lymphoma: A Comprehensive Analysis of Individual Patient Data from Randomized GHSG Trials.","authors":"P J Bröckelmann, H Müller, M Fuchs, S Gillessen, D A Eichenauer, S Borchmann, A S Jacob, K Behringer, J Momotow, J Ferdinandus, B Böll, X Yang, C Kobe, H-T Eich, C Baues, W Klapper, A Engert, P Borchmann, B von Tresckow","doi":"10.1016/j.annonc.2024.12.009","DOIUrl":"https://doi.org/10.1016/j.annonc.2024.12.009","url":null,"abstract":"<p><strong>Background: </strong>We aimed to evaluate the correlation of progression-free (PFS) and overall survival (OS) after first-line treatment of classical Hodgkin lymphoma (HL) and the potential of PFS to serve as a surrogate parameter for OS.</p><p><strong>Patients and methods: </strong>We analyzed individual patient data obtained during and after treatment with polychemotherapy within nine randomized phase III trials (GHSG HD7-HD15) between 01/93 - 08/18. Effects of 16 experimental treatments on PFS and OS on trial level were evaluated by estimation of the treatment effects with Cox proportional hazards (PH) regression and a linear weighted least squares regression. On the patient level, marginal Cox PH models for multiple endpoints were applied according to the Wei-Lin-Weissfeld method.</p><p><strong>Results: </strong>At least one PFS and OS event was recorded in 1,682 and 1,064 of 10,605 patients, respectively. At trial level there was a strong correlation of treatment effects on PFS and OS (weighted Pearson r= 0.72, R<sup>2</sup>= 0.54, P< 0.001). At patient level, moderate to strong correlation of treatment effects on PFS and OS was confirmed with Pearson r ranging between 0.61 - 0.85 (each P< 0.001) and overall r= 0.74. A regression model accounting for different types of experimental treatments and historical progress over trial generations reached very strong correlation (R<sup>2</sup>= 0.93). Applying this model to data from the contemporary first-line ECHELON-1 trial allowed for prediction of OS from PFS (prognosticated ln[HR(OS)]= -0.68 as compared to observed ln[HR(0.59)]= -0.53).</p><p><strong>Conclusion: </strong>In first-line trials of HL, PFS and OS as well as treatment effects and prognostic effects on PFS and OS are strongly correlated. PFS thereby predicts treatment effects on OS to a high degree and many years before OS can be reliably evaluated.</p>","PeriodicalId":8000,"journal":{"name":"Annals of Oncology","volume":" ","pages":""},"PeriodicalIF":56.7,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870734","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}
Pub 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
Background: 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.
Patients and methods: 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.
Results: 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.
Conclusions: 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.
{"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}
Pub 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}