Pub Date : 2024-11-28DOI: 10.1136/jitc-2024-009931
Filippo G Dall'Olio, Wael Salem Zrafi, Quentin Blampey, Francois-Xavier Danlos, Matthieu Roulleaux-Dugage, Gabriel Roman, Charles Naltet, Paul-Henry Cournède, Daniel Gautheret, Mihaela Aldea, David Planchard, Fabrice Barlesi, Aurelien Marabelle, Tyler Hulett, Nathalie Chaput-Gras, Benjamin Besse
Introduction: Immune checkpoint blockers (ICBs) revolutionized the treatment of patients with advanced non-small cell lung cancer (NSCLC) but only a fraction of them obtain a response, and clinical benefit from these treatments is often difficult to predict. The aim of our study is to unveil the potential implications of antibody response to previous viral infections in predicting response to ICBs in patients with NSCLC.
Methods: Sera from patients treated with ICBs alone, chemotherapy (CT) or a combination of CT-ICBs were analyzed with VirScan (CDI Labs, USA), a high-throughput method that comprehensively analyzes epitope-level antiviral IgG antibodies via programmable phage display and immunoprecipitation sequencing.Total number of unique positive peptides (tUP) was defined as the total number of non-overlapping positive "is a hit" peptides for each patient.
Results: Overall, 387 patients were included. Of them, 129 were treated with ICBs alone, 66 with CT-ICBs and 195 with CT alone. 90 out of 129 patients treated with ICBs alone received ICBs as a subsequent line of treatment, while CT-ICBs and CT were administered as upfront therapies.A higher tUP was correlated with improved overall survival in patients treated with ICBs, and confirmed in the multivariate model (HR 0.43, 95% CI 0.24, 0.79, p=0.006), while it was not in those treated with CT-ICBs (p=0.8) and CT alone (p=0.1).tUP was not correlated with programmed death-ligand 1 (PD-L1) expression, while at the transcriptome level it was correlated with several immune-related pathways, particularly involving B cells.
Conclusion: A higher number of viral peptides recognized by serum antibodies might reflect increased immune fitness, resulting in improved outcomes in ICBs treated patients with NSCLC.
免疫检查点阻滞剂(ICBs)彻底改变了晚期非小细胞肺癌(NSCLC)患者的治疗,但只有一小部分患者获得了应答,而且这些治疗的临床获益往往难以预测。我们研究的目的是揭示抗体对既往病毒感染的反应在预测非小细胞肺癌患者对ICBs的反应中的潜在意义。方法:使用VirScan(美国CDI实验室)对单独接受ICBs、化疗(CT)或CT-ICBs联合治疗的患者血清进行分析,VirScan是一种高通量方法,通过可编程噬菌体展示和免疫沉淀测序全面分析表位水平抗病毒IgG抗体。唯一阳性肽总数(tUP)定义为每个患者非重叠阳性“命中”肽的总数。结果:共纳入387例患者。其中,单独使用ICBs治疗129例,CT-ICBs治疗66例,单独使用CT治疗195例。在129名单独接受ICBs治疗的患者中,有90名患者接受了ICBs作为后续治疗,而CT-ICBs和CT作为前期治疗。较高的tUP与接受ICBs治疗的患者的总生存率相关,并在多变量模型中得到证实(HR 0.43, 95% CI 0.24, 0.79, p=0.006),而在接受CT-ICBs治疗的患者(p=0.8)和单独接受CT治疗的患者(p=0.1)中则没有。tUP与程序性死亡配体1 (PD-L1)表达无关,而在转录组水平上,它与几种免疫相关途径相关,特别是涉及B细胞。结论:血清抗体识别的病毒肽数量增加可能反映了免疫适应性的增强,从而改善了接受ICBs治疗的非小细胞肺癌患者的预后。
{"title":"Antecedent viral immunization and efficacy of immune checkpoint blockade: an extensive serum antibody profile to predict outcomes in non-small cell lung cancer.","authors":"Filippo G Dall'Olio, Wael Salem Zrafi, Quentin Blampey, Francois-Xavier Danlos, Matthieu Roulleaux-Dugage, Gabriel Roman, Charles Naltet, Paul-Henry Cournède, Daniel Gautheret, Mihaela Aldea, David Planchard, Fabrice Barlesi, Aurelien Marabelle, Tyler Hulett, Nathalie Chaput-Gras, Benjamin Besse","doi":"10.1136/jitc-2024-009931","DOIUrl":"10.1136/jitc-2024-009931","url":null,"abstract":"<p><strong>Introduction: </strong>Immune checkpoint blockers (ICBs) revolutionized the treatment of patients with advanced non-small cell lung cancer (NSCLC) but only a fraction of them obtain a response, and clinical benefit from these treatments is often difficult to predict. The aim of our study is to unveil the potential implications of antibody response to previous viral infections in predicting response to ICBs in patients with NSCLC.</p><p><strong>Methods: </strong>Sera from patients treated with ICBs alone, chemotherapy (CT) or a combination of CT-ICBs were analyzed with VirScan (CDI Labs, USA), a high-throughput method that comprehensively analyzes epitope-level antiviral IgG antibodies via programmable phage display and immunoprecipitation sequencing.Total number of unique positive peptides (tUP) was defined as the total number of non-overlapping positive \"is a hit\" peptides for each patient.</p><p><strong>Results: </strong>Overall, 387 patients were included. Of them, 129 were treated with ICBs alone, 66 with CT-ICBs and 195 with CT alone. 90 out of 129 patients treated with ICBs alone received ICBs as a subsequent line of treatment, while CT-ICBs and CT were administered as upfront therapies.A higher tUP was correlated with improved overall survival in patients treated with ICBs, and confirmed in the multivariate model (HR 0.43, 95% CI 0.24, 0.79, p=0.006), while it was not in those treated with CT-ICBs (p=0.8) and CT alone (p=0.1).tUP was not correlated with programmed death-ligand 1 (PD-L1) expression, while at the transcriptome level it was correlated with several immune-related pathways, particularly involving B cells.</p><p><strong>Conclusion: </strong>A higher number of viral peptides recognized by serum antibodies might reflect increased immune fitness, resulting in improved outcomes in ICBs treated patients with NSCLC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11605809/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142754981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-27DOI: 10.1136/jitc-2024-009861
Qian Zhong, Hongying Hao, Shu Li, Yongling Ning, Hong Li, Xiaoling Hu, Kelly M McMasters, Jun Yan, Chuanlin Ding
Background: The role of B cells in antitumor immunity remains controversial, with studies suggesting the protumor and antitumor activity. This controversy may be due to the heterogeneity in B cell populations, as the balance among the subtypes may impact tumor progression. The immunosuppressive regulatory B cells (Breg) release interleukin 10 (IL-10) but only represent a minor population. Additionally, tumor-specific antibodies (Abs) also exhibit antitumor and protumor functions dependent on the Ab isotype. Transcription factor c-Maf has been suggested to contribute to the regulation of IL-10 in Breg, but the role of B cell c-Maf signaling in antitumor immunity and regulating Ab responses remains unknown.
Methods: Conditional B cell c-Maf knockout (KO) and control mice were used to establish a KPC pancreatic cancer model and B16.F10 melanoma model. Tumor progression was evaluated. B cell and T cell phenotypes were determined by flow cytometry, mass cytometry, and cytokine/chemokine profiling. Differentially expressed genes in B cells were examined by using RNA sequencing (RNA-seq). Peripheral blood samples were collected from healthy donors and patients with melanoma for B cell phenotyping.
Results: Compared with B cells from the spleen and lymph nodes (LN), B cells in the pancreas exhibited significantly less follicular phenotype and higher IL-10 production in naïve mice. c-Maf deficiency resulted in a significant reduction of CD9+ IL-10-producing Breg in the pancreas. Pancreatic ductal adenocarcinoma (PDAC) progression resulted in the accumulation of circulating B cells with the follicular phenotype and less IL-10 production in the pancreas. Notably, B cell c-Maf deficiency delayed PDAC tumor progression and resulted in proinflammatory B cells. Further, tumor volume reduction and increased effective T cells in the tumor-draining LN were observed in B cell c-Maf KO mice in the B16.F10 melanoma model. RNA-seq analysis of isolated B cells revealed that B cell c-Maf signaling modulates immunoglobulin-associated genes and tumor-specific Ab production. We furthermore demonstrated c-Maf-positive B cell subsets and an increase of IL-10-producing B cells after incubation with IL-4 and CD40L in the peripheral blood of patients with melanoma.
Conclusion: Our study highlights that B cell c-Maf signaling drives tumor progression through the modulation of Breg, inflammatory responses, and tumor-specific Ab responses.
背景:B细胞在抗肿瘤免疫中的作用仍然存在争议,有研究表明B细胞具有抗肿瘤活性和抗肿瘤活性。这种争议可能是由于B细胞群的异质性,因为亚型之间的平衡可能影响肿瘤的进展。免疫抑制调节性B细胞(Breg)释放白细胞介素10 (IL-10),但仅代表少数群体。此外,肿瘤特异性抗体(Abs)也表现出依赖于Ab同型的抗肿瘤和抗肿瘤功能。转录因子c-Maf已被认为有助于Breg中IL-10的调节,但B细胞c-Maf信号在抗肿瘤免疫和调节Ab反应中的作用尚不清楚。方法:采用条件B细胞c-Maf敲除(KO)小鼠和对照小鼠建立KPC胰腺癌模型和B16。F10黑色素瘤模型。评估肿瘤进展。B细胞和T细胞表型通过流式细胞术、质量细胞术和细胞因子/趋化因子谱测定。采用RNA测序(RNA-seq)技术检测B细胞差异表达基因。采集健康供体和黑色素瘤患者外周血样本进行B细胞表型分析。结果:与脾脏和淋巴结(LN)的B细胞相比,胰腺B细胞在naïve小鼠中表现出更少的滤泡表型和更高的IL-10产量。c-Maf缺乏导致胰腺中产生CD9+ il -10的Breg显著减少。胰腺导管腺癌(PDAC)的进展导致胰腺中滤泡型循环B细胞的积累和IL-10的产生减少。值得注意的是,B细胞c-Maf缺乏延迟PDAC肿瘤进展并导致促炎B细胞。此外,在B16的B细胞c-Maf KO小鼠中观察到肿瘤体积缩小和肿瘤引流LN中有效T细胞增加。F10黑色素瘤模型。分离B细胞的RNA-seq分析显示,B细胞c-Maf信号调节免疫球蛋白相关基因和肿瘤特异性抗体的产生。我们进一步证实了黑色素瘤患者外周血中c- maf阳性B细胞亚群和IL-4和CD40L孵育后产生il -10的B细胞增加。结论:我们的研究强调了B细胞c-Maf信号通过调节Breg、炎症反应和肿瘤特异性Ab反应来驱动肿瘤进展。
{"title":"B cell c-Maf signaling promotes tumor progression in animal models of pancreatic cancer and melanoma.","authors":"Qian Zhong, Hongying Hao, Shu Li, Yongling Ning, Hong Li, Xiaoling Hu, Kelly M McMasters, Jun Yan, Chuanlin Ding","doi":"10.1136/jitc-2024-009861","DOIUrl":"10.1136/jitc-2024-009861","url":null,"abstract":"<p><strong>Background: </strong>The role of B cells in antitumor immunity remains controversial, with studies suggesting the protumor and antitumor activity. This controversy may be due to the heterogeneity in B cell populations, as the balance among the subtypes may impact tumor progression. The immunosuppressive regulatory B cells (Breg) release interleukin 10 (IL-10) but only represent a minor population. Additionally, tumor-specific antibodies (Abs) also exhibit antitumor and protumor functions dependent on the Ab isotype. Transcription factor c-Maf has been suggested to contribute to the regulation of IL-10 in Breg, but the role of B cell c-Maf signaling in antitumor immunity and regulating Ab responses remains unknown.</p><p><strong>Methods: </strong>Conditional B cell c-Maf knockout (KO) and control mice were used to establish a KPC pancreatic cancer model and B16.F10 melanoma model. Tumor progression was evaluated. B cell and T cell phenotypes were determined by flow cytometry, mass cytometry, and cytokine/chemokine profiling. Differentially expressed genes in B cells were examined by using RNA sequencing (RNA-seq). Peripheral blood samples were collected from healthy donors and patients with melanoma for B cell phenotyping.</p><p><strong>Results: </strong>Compared with B cells from the spleen and lymph nodes (LN), B cells in the pancreas exhibited significantly less follicular phenotype and higher IL-10 production in naïve mice. c-Maf deficiency resulted in a significant reduction of CD9<sup>+</sup> IL-10-producing Breg in the pancreas. Pancreatic ductal adenocarcinoma (PDAC) progression resulted in the accumulation of circulating B cells with the follicular phenotype and less IL-10 production in the pancreas. Notably, B cell c-Maf deficiency delayed PDAC tumor progression and resulted in proinflammatory B cells. Further, tumor volume reduction and increased effective T cells in the tumor-draining LN were observed in B cell c-Maf KO mice in the B16.F10 melanoma model. RNA-seq analysis of isolated B cells revealed that B cell c-Maf signaling modulates immunoglobulin-associated genes and tumor-specific Ab production. We furthermore demonstrated c-Maf-positive B cell subsets and an increase of IL-10-producing B cells after incubation with IL-4 and CD40L in the peripheral blood of patients with melanoma.</p><p><strong>Conclusion: </strong>Our study highlights that B cell c-Maf signaling drives tumor progression through the modulation of Breg, inflammatory responses, and tumor-specific Ab responses.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603694/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750915","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: CameL phase 3 study demonstrated the superiority of camrelizumab plus chemotherapy over chemotherapy alone for progression-free survival in patients with previously untreated advanced non-squamous non-small-cell lung cancer (NSCLC) without EGFR/ALK alterations. Here, we present the 5-year outcomes.
Methods: Patients were randomized (1:1) and received 4-6 cycles of camrelizumab plus carboplatin and pemetrexed (n=205) or carboplatin and pemetrexed (n=207) every 3 weeks, followed by maintenance camrelizumab plus pemetrexed or pemetrexed only. Crossover from chemotherapy group to camrelizumab monotherapy was permitted after disease progression.
Results: Median time from randomization to data cut-off was 65.2 months (range, 59.7-72.2). HR for overall survival (OS) was 0.74 (95% CI 0.58 to 0.93; one-sided p=0.0043), and was 0.62 (95% CI 0.49 to 0.79; one-sided p<0.0001) after adjustment for crossover. Five-year OS rates were 31.2% (95% CI 24.7% to 37.9%) with camrelizumab plus chemotherapy versus 19.3% (95% CI 13.9% to 25.3%) with chemotherapy alone. Among the 33 patients who completed 2 years of camrelizumab, 5-year OS rate was 84.3% (95% CI 66.4% to 93.2%), and 5-year duration of response rate was 46.5% (95% CI 24.9% to 65.6%) in the 32 responders. No new safety signals were noted.
Conclusions: Camrelizumab plus carboplatin and pemetrexed as first-line therapy continued to demonstrate long-term OS benefit over carboplatin and pemetrexed, with manageable toxicity. Patients who completed 2 years of camrelizumab had enduring response and impressive OS. Current 5-year updated analysis further supports camrelizumab plus carboplatin and pemetrexed as a standard-of-care for previously untreated advanced non-squamous NSCLC without EGFR/ALK alterations.
Trial registration number: NCT03134872.
背景:CameL iii期研究表明,camrelizumab联合化疗比单独化疗更能提高未接受EGFR/ALK改变的晚期非鳞状非小细胞肺癌(NSCLC)患者的无进展生存期。在这里,我们展示了5年的结果。方法:患者以1:1的比例随机分组,每3周接受4-6个周期的camrelizumab +卡铂+培美曲塞(n=205)或卡铂+培美曲塞(n=207),随后进行camrelizumab +培美曲塞或仅培美曲塞的维持治疗。在疾病进展后,允许从化疗组切换到camrelizumab单药治疗。结果:从随机分组到数据截止的中位时间为65.2个月(范围59.7-72.2)。总生存期(OS)的HR为0.74 (95% CI 0.58 ~ 0.93;单侧p=0.0043),为0.62 (95% CI 0.49 ~ 0.79;单侧结论:Camrelizumab联合卡铂和培美曲塞作为一线治疗继续显示出长期的OS优于卡铂和培美曲塞,毒性可控。完成2年camrelizumab治疗的患者有持久的反应和令人印象深刻的OS。目前的5年更新分析进一步支持camrelizumab联合卡铂和培美曲塞作为先前未治疗的晚期非鳞状NSCLC的标准治疗,无EGFR/ALK改变。试验注册号:NCT03134872。
{"title":"Camrelizumab plus carboplatin and pemetrexed as first-line therapy for advanced non-squamous non-small-cell lung cancer: 5-year outcomes of the CameL randomized phase 3 study.","authors":"Caicun Zhou, Gongyan Chen, Yunchao Huang, Jianying Zhou, LiZhu Lin, Jifeng Feng, Zhehai Wang, Yongqian Shu, Jianhua Shi, Yi Hu, QiMing Wang, Ying Cheng, Fengying Wu, Jianhua Chen, Xiaoyan Lin, Yongsheng Wang, Jianan Huang, Jiuwei Cui, Lejie Cao, Yunpeng Liu, Yiping Zhang, Yueyin Pan, Jun Zhao, LiPing Wang, Jianhua Chang, Qun Chen, Xiubao Ren, Wei Zhang, Yun Fan, Zhiyong He, Jian Fang, Kangsheng Gu, XiaoRong Dong, Faguang Jin, Hongjun Gao, Guangyu An, Cuimin Ding, Xiaodong Jiang, Jianping Xiong, Xiangdong Zhou, Sheng Hu, Ping Lu, Anwen Liu, Shuliang Guo, Jianjin Huang, Chengchu Zhu, Jian Zhao, Beili Gao, Yinglan Chen, Chengping Hu, Jian Zhang, Hongmei Zhang, Hui Zhao, Zhigao Wang, Xinjing Ma, Wei Shi","doi":"10.1136/jitc-2024-009240","DOIUrl":"10.1136/jitc-2024-009240","url":null,"abstract":"<p><strong>Background: </strong>CameL phase 3 study demonstrated the superiority of camrelizumab plus chemotherapy over chemotherapy alone for progression-free survival in patients with previously untreated advanced non-squamous non-small-cell lung cancer (NSCLC) without <i>EGFR</i>/<i>ALK</i> alterations. Here, we present the 5-year outcomes.</p><p><strong>Methods: </strong>Patients were randomized (1:1) and received 4-6 cycles of camrelizumab plus carboplatin and pemetrexed (n=205) or carboplatin and pemetrexed (n=207) every 3 weeks, followed by maintenance camrelizumab plus pemetrexed or pemetrexed only. Crossover from chemotherapy group to camrelizumab monotherapy was permitted after disease progression.</p><p><strong>Results: </strong>Median time from randomization to data cut-off was 65.2 months (range, 59.7-72.2). HR for overall survival (OS) was 0.74 (95% CI 0.58 to 0.93; one-sided p=0.0043), and was 0.62 (95% CI 0.49 to 0.79; one-sided p<0.0001) after adjustment for crossover. Five-year OS rates were 31.2% (95% CI 24.7% to 37.9%) with camrelizumab plus chemotherapy versus 19.3% (95% CI 13.9% to 25.3%) with chemotherapy alone. Among the 33 patients who completed 2 years of camrelizumab, 5-year OS rate was 84.3% (95% CI 66.4% to 93.2%), and 5-year duration of response rate was 46.5% (95% CI 24.9% to 65.6%) in the 32 responders. No new safety signals were noted.</p><p><strong>Conclusions: </strong>Camrelizumab plus carboplatin and pemetrexed as first-line therapy continued to demonstrate long-term OS benefit over carboplatin and pemetrexed, with manageable toxicity. Patients who completed 2 years of camrelizumab had enduring response and impressive OS. Current 5-year updated analysis further supports camrelizumab plus carboplatin and pemetrexed as a standard-of-care for previously untreated advanced non-squamous NSCLC without <i>EGFR</i>/<i>ALK</i> alterations.</p><p><strong>Trial registration number: </strong>NCT03134872.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603811/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Recurrent or metastatic cervical cancer (r/m CC) presents limited treatment options for patients failed or progressed quickly following first-line therapy. This study investigated the potential of sintilimab with a prophylactic human papillomavirus (HPV) quadrivalent vaccine as a second-line treatment for r/m CC.
Methods: In this phase 2 clinical trial, patients with r/m CC previously unresponsive or intolerant to standard treatments for metastatic or recurrent lesions were enrolled. Participants received sintilimab (3 mg/kg for body weight <60 kg; 200 mg for ≥60 kg) every 3 weeks until 24 months or 35 cycles and 3 doses of the HPV quadrivalent vaccine (initial dose prior to sintilimab initiation, with subsequent doses at 2 and 6 months). The primary endpoint was the objective response rate (ORR). A Simon two-stage optimal design was used.
Results: From October 2019 to October 2022, 13 patients with r/m CC were enrolled. ORR achieved 53.8% (95% CI 25.1% to 80.8%), and the disease control rate was 76.9% (95% CI 46.2% to 95.0%). Median follow-up duration was 16.07 months (range: 3.64-48.2 months), and median progressive free survival was 7.16 months (95% CI 1.91 -not applicable (NA)). The median overall survival (OS) was not reached (95% CI 9.89 -NA). Hypothyroidism (15.6%) was the most common treatment-related adverse event (AE). No grade 3 or above AEs were observed.
Conclusions: This study suggests the combination of sintilimab plus prophylactic HPV vaccine offers a potentially promising therapeutic strategy for patients with r/m CC unresponsive or intolerant to standard therapies.Trial registration numberNCT04096911.
目的:复发性或转移性宫颈癌(r/m CC)对一线治疗失败或进展迅速的患者提供有限的治疗选择。本研究探讨了辛替单抗与预防性人乳头瘤病毒(HPV)四价疫苗作为r/m CC二线治疗的潜力。方法:在这项2期临床试验中,纳入了先前对转移性或复发性病变的标准治疗无反应或不耐受的r/m CC患者。结果:从2019年10月到2022年10月,纳入了13例r/m CC患者。ORR为53.8% (95% CI 25.1% ~ 80.8%),疾病控制率为76.9% (95% CI 46.2% ~ 95.0%)。中位随访时间为16.07个月(3.64-48.2个月),中位进展自由生存期为7.16个月(95% CI 1.91 -不适用(NA))。中位总生存期(OS)未达到(95% CI 9.89 -NA)。甲状腺功能减退(15.6%)是最常见的治疗相关不良事件(AE)。未见3级及以上ae。结论:这项研究表明,辛替单抗联合预防性HPV疫苗为对标准治疗无反应或不耐受的r/m CC患者提供了一种潜在的治疗策略。试验注册号:bernct04096911。
{"title":"Sintilimab plus HPV vaccine for recurrent or metastatic cervical cancer.","authors":"Buhai Wang, Yichen Liang, Yuechao Wu, Qiuxian Li, Yichun Zeng, Liqin Liu, Wenmiao Cao, Xiaoru Geng, Yuxiang Huang, Yinxia Wu, Jiulin Pan, Xian Zhang, J Juan Gu","doi":"10.1136/jitc-2024-009898","DOIUrl":"10.1136/jitc-2024-009898","url":null,"abstract":"<p><strong>Purpose: </strong>Recurrent or metastatic cervical cancer (r/m CC) presents limited treatment options for patients failed or progressed quickly following first-line therapy. This study investigated the potential of sintilimab with a prophylactic human papillomavirus (HPV) quadrivalent vaccine as a second-line treatment for r/m CC.</p><p><strong>Methods: </strong>In this phase 2 clinical trial, patients with r/m CC previously unresponsive or intolerant to standard treatments for metastatic or recurrent lesions were enrolled. Participants received sintilimab (3 mg/kg for body weight <60 kg; 200 mg for ≥60 kg) every 3 weeks until 24 months or 35 cycles and 3 doses of the HPV quadrivalent vaccine (initial dose prior to sintilimab initiation, with subsequent doses at 2 and 6 months). The primary endpoint was the objective response rate (ORR). A Simon two-stage optimal design was used.</p><p><strong>Results: </strong>From October 2019 to October 2022, 13 patients with r/m CC were enrolled. ORR achieved 53.8% (95% CI 25.1% to 80.8%), and the disease control rate was 76.9% (95% CI 46.2% to 95.0%). Median follow-up duration was 16.07 months (range: 3.64-48.2 months), and median progressive free survival was 7.16 months (95% CI 1.91 -not applicable (NA)). The median overall survival (OS) was not reached (95% CI 9.89 -NA). Hypothyroidism (15.6%) was the most common treatment-related adverse event (AE). No grade 3 or above AEs were observed.</p><p><strong>Conclusions: </strong>This study suggests the combination of sintilimab plus prophylactic HPV vaccine offers a potentially promising therapeutic strategy for patients with r/m CC unresponsive or intolerant to standard therapies.Trial registration numberNCT04096911.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603683/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-27DOI: 10.1136/jitc-2024-010157
Jun Cao, Bingbing Su, Chi Zhang, Rui Peng, Daoyuan Tu, Qiangwei Deng, Guoqing Jiang, Shengjie Jin, Qian Wang, Dou-Sheng Bai
Background: Resistance to immune checkpoint inhibitors (ICIs) significantly limits the efficacy of immunotherapy in patients with hepatocellular carcinoma (HCC). However, the mechanisms underlying immunotherapy resistance remain poorly understood. Our aim was to clarify the role of membrane-associated ring-CH-type finger 3 (MARCHF3) in HCC within the framework of anti-programmed cell death protein-1 (PD-1) therapy.
Methods: MARCHF3 was identified in the transcriptomic profiles of HCC tumors exhibiting different responses to ICIs. In humans, the correlation between MARCHF3 expression and the tumor microenvironment (TME) was assessed via multiplex immunohistochemistry. In addition, MARCHF3 expression in tumor cells and immune cell infiltration were assessed by flow cytometry.
Results: MARCHF3 was significantly upregulated in tumors from patients who responded to ICIs. Increased MARCHF3 expression in HCC cells promoted dendritic cell (DC) maturation and stimulated CD8+ T-cell activation, thereby augmenting tumor control. Mechanistically, we identified MARCHF3 as a pivotal regulator of the DNA damage response. It directly interacted with Poly(ADP-Ribose) Polymerase 1 (PARP1) via K48-linked ubiquitination, leading to PARP1 degradation. This process promoted the release of double-strand DNA and activated cCAS-STING in DCs, thereby initiating DC-mediated antigen cross-presentation and CD8+ T-cell activation. Additionally, ATF4 transcriptionally regulated MARCHF3 expression. Notably, the PARP1 inhibitor olaparib augmented the efficacy of anti-PD-1 immunotherapy in both subcutaneous and orthotopic HCC mouse models.
Conclusions: MARCHF3 has emerged as a pivotal regulator of the immune landscape in the HCC TME and is a potent predictive biomarker for HCC. Combining interventions targeting the DNA damage response with ICIs is a promising treatment strategy for HCC.
{"title":"Degradation of PARP1 by MARCHF3 in tumor cells triggers cCAS-STING activation in dendritic cells to regulate antitumor immunity in hepatocellular carcinoma.","authors":"Jun Cao, Bingbing Su, Chi Zhang, Rui Peng, Daoyuan Tu, Qiangwei Deng, Guoqing Jiang, Shengjie Jin, Qian Wang, Dou-Sheng Bai","doi":"10.1136/jitc-2024-010157","DOIUrl":"10.1136/jitc-2024-010157","url":null,"abstract":"<p><strong>Background: </strong>Resistance to immune checkpoint inhibitors (ICIs) significantly limits the efficacy of immunotherapy in patients with hepatocellular carcinoma (HCC). However, the mechanisms underlying immunotherapy resistance remain poorly understood. Our aim was to clarify the role of membrane-associated ring-CH-type finger 3 (MARCHF3) in HCC within the framework of anti-programmed cell death protein-1 (PD-1) therapy.</p><p><strong>Methods: </strong>MARCHF3 was identified in the transcriptomic profiles of HCC tumors exhibiting different responses to ICIs. In humans, the correlation between MARCHF3 expression and the tumor microenvironment (TME) was assessed via multiplex immunohistochemistry. In addition, MARCHF3 expression in tumor cells and immune cell infiltration were assessed by flow cytometry.</p><p><strong>Results: </strong>MARCHF3 was significantly upregulated in tumors from patients who responded to ICIs. Increased MARCHF3 expression in HCC cells promoted dendritic cell (DC) maturation and stimulated CD8<sup>+</sup> T-cell activation, thereby augmenting tumor control. Mechanistically, we identified MARCHF3 as a pivotal regulator of the DNA damage response. It directly interacted with Poly(ADP-Ribose) Polymerase 1 (PARP1) via K48-linked ubiquitination, leading to PARP1 degradation. This process promoted the release of double-strand DNA and activated cCAS-STING in DCs, thereby initiating DC-mediated antigen cross-presentation and CD8<sup>+</sup> T-cell activation. Additionally, ATF4 transcriptionally regulated MARCHF3 expression. Notably, the PARP1 inhibitor olaparib augmented the efficacy of anti-PD-1 immunotherapy in both subcutaneous and orthotopic HCC mouse models.</p><p><strong>Conclusions: </strong>MARCHF3 has emerged as a pivotal regulator of the immune landscape in the HCC TME and is a potent predictive biomarker for HCC. Combining interventions targeting the DNA damage response with ICIs is a promising treatment strategy for HCC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11605840/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Ovarian clear cell carcinoma (OCCC) is a rare and chemo-resistant subtype of ovarian cancer. While immunotherapy has demonstrated effectiveness in some OCCC cases, the mechanisms for heterogeneous immunoreactivity and potential combinatory strategies remain unclear.
Methods: Tumor samples from 13 patients with OCCC underwent single-cell mRNA-seq and TCR-seq to generate 1 40 683 cells transcriptome, while additionally 31 formalin-fixed paraffin-embedded samples were used for immunohistochemistry. Spatial transcriptomics of two OCCC samples and bulk RNA-seq of 58 patients were incorporated for spatial and interpatient level explorations. Serum tumor markers and radiologic images of three patients with OCCC who received combinatory VEGF and PD-1 inhibition were retrospectively analyzed.
Results: OCCC exhibited a dynamic immune architecture shaped by genetic and therapeutic pressure. ARID1A mutation linked to baseline immune activation, correlated with an enrichment of neoantigen-reactive CXCL13+ CTLA4+ CD8+ T cells (p<0.001) and enhanced FASLG-FAS interactions. Recurrent OCCC was fibrotic, angiogenic, and immunosuppressive, exhibiting metabolic reprogramming towards activated activity in fatty acid metabolism. High CD36 (log-rank p=0.012, HR: 4.515) and CD47 expression (log-rank p=0.037, HR: 3.246) indicated worse progression-free survival. Treatment with bevacizumab increased intratumoral T cell infiltration and activated T cell interferon-γ signaling. Retrospective analysis of clinical cases revealed that combination therapy with anti-VEGF (vascular endothelial growth factor) and anti-PD-1 agents exerted clinical benefits in patients with OCCC with persistent, recurrent, and metastatic disease.
Conclusions: ARID1A mutation correlated with OCCC baseline immune activation. Stromal reconstruction and tumor metabolic reprogramming functioned as key processes of OCCC dynamic progression. VEGF inhibition remodeled OCCC stroma, restored T cell function and potentiated immunotherapy. CD36 and CD47 might be potential therapeutic targets for recurrent OCCC.
{"title":"Genetic and therapeutic heterogeneity shape the baseline and longitudinal immune ecosystem of ovarian clear cell carcinoma.","authors":"Siyu Xia, Lihua Chen, Min Yu, Jiana Li, Jiaxin Chen, Fei Xu, Mengdong Ni, Chaohua Liu, Xiaohua Wu, Xiaojun Chen, Jiajia Li","doi":"10.1136/jitc-2024-010069","DOIUrl":"10.1136/jitc-2024-010069","url":null,"abstract":"<p><strong>Background: </strong>Ovarian clear cell carcinoma (OCCC) is a rare and chemo-resistant subtype of ovarian cancer. While immunotherapy has demonstrated effectiveness in some OCCC cases, the mechanisms for heterogeneous immunoreactivity and potential combinatory strategies remain unclear.</p><p><strong>Methods: </strong>Tumor samples from 13 patients with OCCC underwent single-cell mRNA-seq and TCR-seq to generate 1 40 683 cells transcriptome, while additionally 31 formalin-fixed paraffin-embedded samples were used for immunohistochemistry. Spatial transcriptomics of two OCCC samples and bulk RNA-seq of 58 patients were incorporated for spatial and interpatient level explorations. Serum tumor markers and radiologic images of three patients with OCCC who received combinatory VEGF and PD-1 inhibition were retrospectively analyzed.</p><p><strong>Results: </strong>OCCC exhibited a dynamic immune architecture shaped by genetic and therapeutic pressure. <i>ARID1A</i> mutation linked to baseline immune activation, correlated with an enrichment of neoantigen-reactive CXCL13<sup>+</sup> CTLA4<sup>+</sup> CD8<sup>+</sup> T cells (p<0.001) and enhanced FASLG-FAS interactions. Recurrent OCCC was fibrotic, angiogenic, and immunosuppressive, exhibiting metabolic reprogramming towards activated activity in fatty acid metabolism. High CD36 (log-rank p=0.012, HR: 4.515) and CD47 expression (log-rank p=0.037, HR: 3.246) indicated worse progression-free survival. Treatment with bevacizumab increased intratumoral T cell infiltration and activated T cell interferon-γ signaling. Retrospective analysis of clinical cases revealed that combination therapy with anti-VEGF (vascular endothelial growth factor) and anti-PD-1 agents exerted clinical benefits in patients with OCCC with persistent, recurrent, and metastatic disease.</p><p><strong>Conclusions: </strong><i>ARID1A</i> mutation correlated with OCCC baseline immune activation. Stromal reconstruction and tumor metabolic reprogramming functioned as key processes of OCCC dynamic progression. VEGF inhibition remodeled OCCC stroma, restored T cell function and potentiated immunotherapy. CD36 and CD47 might be potential therapeutic targets for recurrent OCCC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603735/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-27DOI: 10.1136/jitc-2024-009602corr1
{"title":"Correction: Metabolic reprogramming of hypoxic tumor-associated macrophages through CSF-1R targeting favors treatment efficiency in colorectal cancers.","authors":"","doi":"10.1136/jitc-2024-009602corr1","DOIUrl":"10.1136/jitc-2024-009602corr1","url":null,"abstract":"","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603823/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-27DOI: 10.1136/jitc-2024-010080
Wenqiang Yan, Yu Xiong, Rui Lv, Chenxing Du, Tengteng Yu, Shuaishuai Zhang, Weiwei Sui, Shuhui Deng, Jigang Xiao, Yan Xu, Dehui Zou, Lugui Qiu, Gang An
B-cell maturation antigen(BCMA)-directed chimeric antigen receptor (CAR)-T-cell therapy has significantly improved the treatment of relapsed or refractory multiple myeloma (MM). Nevertheless, the uncommon phenomenon of biphasic CAR-T cell expansion in vivo and its related severe toxicities have not been methodically described and studied. Herein, we report a case of patients with MM who experienced two CAR-T cell expansion peaks and subsequently developed multiple severe toxicities following BCMA CAR-T cell infusion. The first expansion peak occurred on Day 7, accompanied by grade 3 cytokine release syndrome. The second peak occurred on Day 28, associated with severe immune effector cell-associated hematotoxicity (ICAHT), immune effector-cell associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), and polymicrobial infections. Both ICAHT and IEC-HS were refractory to our standard treatments; however, human umbilical cord mesenchymal stem cell infusion exhibited some efficacy in improving cytopenia. Despite the administration of a broad-spectrum anti-infective regimen, cytomegalovirus viremia remained uncontrollable, resulting in the development of central nervous system infection, neurological symptoms, and ultimately death. Additionally, we also employed high-dimensional 33-color spectral flow cytometry to describe the dynamic changes in immune cell components and functions between the two expansion peaks. Collectively, this case provides novel insights into the biphasic CAR-T expansion and related immune effector cell-associated toxicities.
{"title":"Uncommon biphasic CAR-T expansion induces hemophagocytic lymphohistiocytosis-like syndrome and fatal multiple infections following BCMA CAR-T cell therapy: a case report.","authors":"Wenqiang Yan, Yu Xiong, Rui Lv, Chenxing Du, Tengteng Yu, Shuaishuai Zhang, Weiwei Sui, Shuhui Deng, Jigang Xiao, Yan Xu, Dehui Zou, Lugui Qiu, Gang An","doi":"10.1136/jitc-2024-010080","DOIUrl":"10.1136/jitc-2024-010080","url":null,"abstract":"<p><p>B-cell maturation antigen(BCMA)-directed chimeric antigen receptor (CAR)-T-cell therapy has significantly improved the treatment of relapsed or refractory multiple myeloma (MM). Nevertheless, the uncommon phenomenon of biphasic CAR-T cell expansion in vivo and its related severe toxicities have not been methodically described and studied. Herein, we report a case of patients with MM who experienced two CAR-T cell expansion peaks and subsequently developed multiple severe toxicities following BCMA CAR-T cell infusion. The first expansion peak occurred on Day 7, accompanied by grade 3 cytokine release syndrome. The second peak occurred on Day 28, associated with severe immune effector cell-associated hematotoxicity (ICAHT), immune effector-cell associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), and polymicrobial infections. Both ICAHT and IEC-HS were refractory to our standard treatments; however, human umbilical cord mesenchymal stem cell infusion exhibited some efficacy in improving cytopenia. Despite the administration of a broad-spectrum anti-infective regimen, cytomegalovirus viremia remained uncontrollable, resulting in the development of central nervous system infection, neurological symptoms, and ultimately death. Additionally, we also employed high-dimensional 33-color spectral flow cytometry to describe the dynamic changes in immune cell components and functions between the two expansion peaks. Collectively, this case provides novel insights into the biphasic CAR-T expansion and related immune effector cell-associated toxicities.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11603806/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750921","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Despite the success of immune checkpoint inhibitor (ICI)-based combination therapies in hepatocellular carcinoma (HCC), its effectiveness remains confined to a subset of patients. The development of reliable, predictive markers is important for accurate patient stratification and further mechanistic understanding of therapy response.
Methods: We comprehensively analyzed paired single-cell RNA transcriptome and T-cell repertoire profiles from 14 HCC ascites samples, collected from 7 patients before and after treatment with the combination of sintilimab (anti-PD-1) and bevacizumab (anti-VEGF).
Results: We identify a widespread convergence on interferon (IFN) signaling across various immune cell lineages in treatment-responsive patients with HCC, indicating a common transcriptional state transition in the immune microenvironment linked to immunotherapy response in HCC. Strong IFN signaling marks CD8+ T cells with larger clonal expansion and enhanced cytotoxicity, macrophages toward M1-like polarization and strong T-cell recruitment ability, dendritic cells with increased antigen presentation capacity, as well as highly cytotoxic natural killer cells and activated B cells. By translating our finding to cohorts of patients with HCC, we demonstrate the specificity of IFN-signaling in the prognosis of patients with HCC and its ability to predict immunotherapy response.
Conclusions: This study provides a unique single-cell resource with clonal and longitudinal resolution during ICI therapy and reveals IFN signaling as a biomarker of immunotherapy response in HCC, suggesting a beneficial effect by combining IFN inducers with ICIs for patients with HCC.
{"title":"Integrated single-cell transcriptome and TCR profiles of hepatocellular carcinoma highlight the convergence on interferon signaling during immunotherapy.","authors":"Tianhao Li, Shengnan Guo, Chang Xu, Mingjie Zhang, Cheng Lyu, Huanhuan Xu, Zepeng Hou, Mingshuo Zhang, Xiaobo Li, Jing Ren, Changqing Liu, Dan Kong, Dapeng Hao, Guangyu Wang","doi":"10.1136/jitc-2024-010534","DOIUrl":"10.1136/jitc-2024-010534","url":null,"abstract":"<p><strong>Background: </strong>Despite the success of immune checkpoint inhibitor (ICI)-based combination therapies in hepatocellular carcinoma (HCC), its effectiveness remains confined to a subset of patients. The development of reliable, predictive markers is important for accurate patient stratification and further mechanistic understanding of therapy response.</p><p><strong>Methods: </strong>We comprehensively analyzed paired single-cell RNA transcriptome and T-cell repertoire profiles from 14 HCC ascites samples, collected from 7 patients before and after treatment with the combination of sintilimab (anti-PD-1) and bevacizumab (anti-VEGF).</p><p><strong>Results: </strong>We identify a widespread convergence on interferon (IFN) signaling across various immune cell lineages in treatment-responsive patients with HCC, indicating a common transcriptional state transition in the immune microenvironment linked to immunotherapy response in HCC. Strong IFN signaling marks CD8<sup>+</sup> T cells with larger clonal expansion and enhanced cytotoxicity, macrophages toward M1-like polarization and strong T-cell recruitment ability, dendritic cells with increased antigen presentation capacity, as well as highly cytotoxic natural killer cells and activated B cells. By translating our finding to cohorts of patients with HCC, we demonstrate the specificity of IFN-signaling in the prognosis of patients with HCC and its ability to predict immunotherapy response.</p><p><strong>Conclusions: </strong>This study provides a unique single-cell resource with clonal and longitudinal resolution during ICI therapy and reveals IFN signaling as a biomarker of immunotherapy response in HCC, suggesting a beneficial effect by combining IFN inducers with ICIs for patients with HCC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11590841/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142710274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-24DOI: 10.1136/jitc-2024-010074
Qiyan Cai, Yi Que, Xing Zhang
Soft tissue sarcoma is characterized by its rarity and complexity, making it more difficult to conduct large clinical trials compared with other solid tumors. Also known as 'cold tumors,' sarcomas, especially advanced sarcomas, have poor responses to immunotherapy. Based on that, the results of two groundbreaking phase 2 clinical trials about neoadjuvant immunotherapy in patients with liposarcoma or undifferentiated pleomorphic sarcoma are encouraging. In this paper, we discuss the results of these clinical trials and the challenges we are facing to conduct neoadjuvant immunotherapy in sarcomas and call for further research to promote the development of it.
{"title":"Neoadjuvant immunotherapy in the evolving landscape of sarcoma treatment.","authors":"Qiyan Cai, Yi Que, Xing Zhang","doi":"10.1136/jitc-2024-010074","DOIUrl":"10.1136/jitc-2024-010074","url":null,"abstract":"<p><p>Soft tissue sarcoma is characterized by its rarity and complexity, making it more difficult to conduct large clinical trials compared with other solid tumors. Also known as 'cold tumors,' sarcomas, especially advanced sarcomas, have poor responses to immunotherapy. Based on that, the results of two groundbreaking phase 2 clinical trials about neoadjuvant immunotherapy in patients with liposarcoma or undifferentiated pleomorphic sarcoma are encouraging. In this paper, we discuss the results of these clinical trials and the challenges we are facing to conduct neoadjuvant immunotherapy in sarcomas and call for further research to promote the development of it.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11590830/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142710282","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}