首页 > 最新文献

Cancer research communications最新文献

英文 中文
p53R172H and p53R245W Hotspot Mutations Drive Distinct Transcriptomes in Mouse Mammary Tumors Through a Convergent Transcriptional Mediator. p53R172H 和 p53R245W 热点突变通过一种趋同的转录介质驱动小鼠乳腺肿瘤中不同的转录组。
IF 2 Q3 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2767-9764.CRC-24-0128
Joy M McDaniel, Rhiannon L Morrissey, Denada Dibra, Lalit R Patel, Shunbin Xiong, Yun Zhang, Gilda P Chau, Xiaoping Su, Yuan Qi, Adel K El-Naggar, Guillermina Lozano

Aggressive breast cancers harbor TP53 missense mutations. Tumor cells with TP53 missense mutations exhibit enhanced growth and survival through transcriptional rewiring. To delineate how TP53 mutations in breast cancer contribute to tumorigenesis and progression in vivo, we created a somatic mouse model driven by mammary epithelial cell-specific expression of Trp53 mutations. Mice developed primary mammary tumors reflecting the human molecular subtypes of luminal A, luminal B, HER2-enriched, and triple-negative breast cancer with metastases. Transcriptomic analyses comparing MaPR172H/- or MaPR245W/- mammary tumors to MaP-/- tumors revealed (1) differences in cancer-associated pathways activated in both p53 mutants and (2) Nr5a2 as a novel transcriptional mediator of distinct pathways in p53 mutants. Meta-analyses of human breast tumors corroborated these results. In vitro assays demonstrate mutant p53 upregulates specific target genes that are enriched for Nr5a2 response elements in their promoters. Co-immunoprecipitation studies revealed p53R172H and p53R245W interact with Nr5a2. These findings implicate NR5A2 as a novel mediator of mutant p53 transcriptional activity in breast cancer.

Significance: Our findings implicate NR5A2 as a novel mediator of mutant p53 transcriptional activity in breast cancer. NR5A2 may be an important therapeutic target in hard-to-treat breast cancers such as endocrine-resistant tumors and metastatic triple-negative breast cancers harboring TP53 missense mutations.

侵袭性乳腺癌含有 TP53 错义突变。具有 TP53 错义突变的肿瘤细胞会通过转录重构增强生长和存活能力。为了弄清乳腺癌中的 TP53 基因突变是如何在体内促进肿瘤发生和发展的,我们创建了一种由乳腺上皮细胞特异性表达 Trp53 基因突变驱动的体细胞小鼠模型。小鼠罹患的原发性乳腺肿瘤反映了人类乳腺癌的分子亚型,包括Luminal A、Luminal B、HER2-enriched和三阴性乳腺癌,并伴有转移。将 MaPR172H/- 或 MaPR245W/- 乳腺肿瘤与 MaP-/- 肿瘤进行比较的转录组分析发现:(1)两种 p53 突变体中激活的癌症相关通路存在差异;(2)Nr5a2 是 p53 突变体中不同通路的新型转录介质。人类乳腺肿瘤的元分析证实了这些结果。体外实验表明,突变体 p53 会上调特定的靶基因,这些基因的启动子中富含 Nr5a2 反应元件。共免疫沉淀研究显示 p53R172H 和 p53R245W 与 Nr5a2 相互作用。这些发现表明 NR5A2 是乳腺癌突变 p53 转录活性的新型介导因子。
{"title":"p53R172H and p53R245W Hotspot Mutations Drive Distinct Transcriptomes in Mouse Mammary Tumors Through a Convergent Transcriptional Mediator.","authors":"Joy M McDaniel, Rhiannon L Morrissey, Denada Dibra, Lalit R Patel, Shunbin Xiong, Yun Zhang, Gilda P Chau, Xiaoping Su, Yuan Qi, Adel K El-Naggar, Guillermina Lozano","doi":"10.1158/2767-9764.CRC-24-0128","DOIUrl":"10.1158/2767-9764.CRC-24-0128","url":null,"abstract":"<p><p>Aggressive breast cancers harbor TP53 missense mutations. Tumor cells with TP53 missense mutations exhibit enhanced growth and survival through transcriptional rewiring. To delineate how TP53 mutations in breast cancer contribute to tumorigenesis and progression in vivo, we created a somatic mouse model driven by mammary epithelial cell-specific expression of Trp53 mutations. Mice developed primary mammary tumors reflecting the human molecular subtypes of luminal A, luminal B, HER2-enriched, and triple-negative breast cancer with metastases. Transcriptomic analyses comparing MaPR172H/- or MaPR245W/- mammary tumors to MaP-/- tumors revealed (1) differences in cancer-associated pathways activated in both p53 mutants and (2) Nr5a2 as a novel transcriptional mediator of distinct pathways in p53 mutants. Meta-analyses of human breast tumors corroborated these results. In vitro assays demonstrate mutant p53 upregulates specific target genes that are enriched for Nr5a2 response elements in their promoters. Co-immunoprecipitation studies revealed p53R172H and p53R245W interact with Nr5a2. These findings implicate NR5A2 as a novel mediator of mutant p53 transcriptional activity in breast cancer.</p><p><strong>Significance: </strong>Our findings implicate NR5A2 as a novel mediator of mutant p53 transcriptional activity in breast cancer. NR5A2 may be an important therapeutic target in hard-to-treat breast cancers such as endocrine-resistant tumors and metastatic triple-negative breast cancers harboring TP53 missense mutations.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310746/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141592260","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tumor Vessel Normalization via PFKFB3 Inhibition Alleviates Hypoxia and Increases Tumor Necrosis in Rectal Cancer upon Radiotherapy. 通过抑制 PFKFB3 使肿瘤血管正常化可缓解直肠癌放疗时的缺氧并增加肿瘤坏死。
IF 2 Q3 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2767-9764.CRC-24-0077
Marcus Edelmann, Shuang Fan, Tiago De Oliveira, Tina Goldhardt, Dorothée Sartorius, Teona Midelashvili, Karly Conrads, Niels B Paul, Tim Beißbarth, Johannes R Fleischer, Moritz L Blume, Hanibal Bohnenberger, Natasa Josipovic, Argyris Papantonis, Michael Linnebacher, Leif H Dröge, Michael Ghadimi, Stefan Rieken, Lena-Christin Conradi

Treatment of patients with locally advanced rectal cancer (RC) is based on neoadjuvant chemoradiotherapy followed by surgery. In order to reduce the development of therapy resistance, it is necessary to further improve previous treatment approaches. Recent in vivo experimental studies suggested that the reduction of tumor hypoxia by tumor vessel normalization (TVN), through the inhibition of the glycolytic activator PFKFB3, could significantly improve tumor response to therapy. We have evaluated in vitro and in vivo the effects of the PFKFB3 inhibitor 2E-3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) on cell survival, clonogenicity, migration, invasion, and metabolism using colorectal cancer cells, patient-derived tumor organoid (PDO), and xenograft (PDX). 3PO treatment of colorectal cancer cells increased radiation-induced cell death and reduced cancer cell invasion. Moreover, gene set enrichment analysis shows that 3PO is able to alter the metabolic status of PDOs toward oxidative phosphorylation. Additionally, in vivo neoadjuvant treatment with 3PO induced TVN, alleviated tumor hypoxia, and increased tumor necrosis. Our results support PFKFB3 inhibition as a possible future neoadjuvant addition for patients with RC.

Significance: Novel therapies to better treat colorectal cancer are necessary to improve patient outcomes. Therefore, in this study, we evaluated the combination of a metabolic inhibitor (3PO) and standard radiotherapy in different experimental settings. We have observed that the addition of 3PO increased radiation effects, ultimately improving tumor cell response to therapy.

局部晚期直肠癌(RC)患者的治疗以新辅助化放疗为基础,然后进行手术。为了减少耐药性的产生,有必要进一步改进以往的治疗方法。最近的体内实验研究表明,通过抑制糖酵解活化剂 PFKFB3,使肿瘤血管正常化(TVN),从而减少肿瘤缺氧,可显著改善肿瘤对治疗的反应。我们利用结直肠癌细胞、患者衍生肿瘤器官组织(PDO)和异种移植(PDX),在体外和体内评估了 PFKFB3 抑制剂 2E-3-(3-吡啶基)-1-(4-吡啶基)-2-丙烯-1-酮(3PO)对细胞存活、克隆性、迁移、侵袭和代谢的影响。3PO 处理结直肠癌细胞可增加辐射诱导的细胞死亡,并减少癌细胞的侵袭。此外,基因组富集分析(Gene Set Enrichment Analysis)显示,3PO能够改变PDO的代谢状态,使其趋向氧化磷酸化。此外,在体内使用 3PO 进行新辅助治疗可诱导 TVN、缓解肿瘤缺氧并增加肿瘤坏死。我们的研究结果支持将 PFKFB3 抑制作为直肠癌患者未来可能的新辅助治疗药物。
{"title":"Tumor Vessel Normalization via PFKFB3 Inhibition Alleviates Hypoxia and Increases Tumor Necrosis in Rectal Cancer upon Radiotherapy.","authors":"Marcus Edelmann, Shuang Fan, Tiago De Oliveira, Tina Goldhardt, Dorothée Sartorius, Teona Midelashvili, Karly Conrads, Niels B Paul, Tim Beißbarth, Johannes R Fleischer, Moritz L Blume, Hanibal Bohnenberger, Natasa Josipovic, Argyris Papantonis, Michael Linnebacher, Leif H Dröge, Michael Ghadimi, Stefan Rieken, Lena-Christin Conradi","doi":"10.1158/2767-9764.CRC-24-0077","DOIUrl":"10.1158/2767-9764.CRC-24-0077","url":null,"abstract":"<p><p>Treatment of patients with locally advanced rectal cancer (RC) is based on neoadjuvant chemoradiotherapy followed by surgery. In order to reduce the development of therapy resistance, it is necessary to further improve previous treatment approaches. Recent in vivo experimental studies suggested that the reduction of tumor hypoxia by tumor vessel normalization (TVN), through the inhibition of the glycolytic activator PFKFB3, could significantly improve tumor response to therapy. We have evaluated in vitro and in vivo the effects of the PFKFB3 inhibitor 2E-3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) on cell survival, clonogenicity, migration, invasion, and metabolism using colorectal cancer cells, patient-derived tumor organoid (PDO), and xenograft (PDX). 3PO treatment of colorectal cancer cells increased radiation-induced cell death and reduced cancer cell invasion. Moreover, gene set enrichment analysis shows that 3PO is able to alter the metabolic status of PDOs toward oxidative phosphorylation. Additionally, in vivo neoadjuvant treatment with 3PO induced TVN, alleviated tumor hypoxia, and increased tumor necrosis. Our results support PFKFB3 inhibition as a possible future neoadjuvant addition for patients with RC.</p><p><strong>Significance: </strong>Novel therapies to better treat colorectal cancer are necessary to improve patient outcomes. Therefore, in this study, we evaluated the combination of a metabolic inhibitor (3PO) and standard radiotherapy in different experimental settings. We have observed that the addition of 3PO increased radiation effects, ultimately improving tumor cell response to therapy.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310748/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141617712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Tumor Microbiome as a Predictor of Outcomes in Patients with Metastatic Melanoma Treated with Immune Checkpoint Inhibitors. 肿瘤微生物组是预测接受免疫检查点抑制剂治疗的转移性黑色素瘤患者预后的指标。
IF 2 Q3 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2767-9764.CRC-23-0170
Caroline E Dravillas, Samuel S Coleman, Rebecca Hoyd, Griffin Caryotakis, Louis Denko, Carlos H F Chan, Michelle L Churchman, Nicholas Denko, Rebecca D Dodd, Islam Eljilany, Sheetal Hardikar, Marium Husain, Alexandra P Ikeguchi, Ning Jin, Qin Ma, Martin D McCarter, Afaf E G Osman, Lary A Robinson, Eric A Singer, Gabriel Tinoco, Cornelia M Ulrich, Yousef Zakharia, Daniel Spakowicz, Ahmad A Tarhini, Aik Choon Tan

Emerging evidence supports the important role of the tumor microbiome in oncogenesis, cancer immune phenotype, cancer progression, and treatment outcomes in many malignancies. In this study, we investigated the metastatic melanoma tumor microbiome and its potential roles in association with clinical outcomes, such as survival, in patients with metastatic disease treated with immune checkpoint inhibitors (ICI). Baseline tumor samples were collected from 71 patients with metastatic melanoma before treatment with ICIs. Bulk RNA sequencing (RNA-seq) was conducted on the formalin-fixed, paraffin-embedded and fresh frozen tumor samples. Durable clinical benefit (primary clinical endpoint) following ICIs was defined as overall survival >24 months and no change to the primary drug regimen (responders). We processed RNA-seq reads to carefully identify exogenous sequences using the {exotic} tool. The age of the 71 patients with metastatic melanoma ranged from 24 to 83 years, 59% were male, and 55% survived >24 months following the initiation of ICI treatment. Exogenous taxa were identified in the tumor RNA-seq, including bacteria, fungi, and viruses. We found differences in gene expression and microbe abundances in immunotherapy-responsive versus nonresponsive tumors. Responders showed significant enrichment of bacteriophages in the phylum Uroviricota, and nonresponders showed enrichment of several bacteria, including Campylobacter jejuni. These microbes correlated with immune-related gene expression signatures. Finally, we found that models for predicting prolonged survival with immunotherapy using both microbe abundances and gene expression outperformed models using either dataset alone. Our findings warrant further investigation and potentially support therapeutic strategies to modify the tumor microbiome in order to improve treatment outcomes with ICIs.

Significance: We analyzed the tumor microbiome and interactions with genes and pathways in metastatic melanoma treated with immunotherapy and identified several microbes associated with immunotherapy response and immune-related gene expression signatures. Machine learning models that combined microbe abundances and gene expression outperformed models using either dataset alone in predicting immunotherapy responses.

越来越多的证据支持肿瘤微生物组在许多恶性肿瘤的肿瘤发生、癌症免疫表型、癌症进展和治疗结果中的重要作用。在这项研究中,我们调查了接受免疫检查点抑制剂(ICIs)治疗的转移性黑色素瘤患者的转移性黑色素瘤肿瘤微生物组及其与生存期等临床结果相关的潜在作用。71名转移性黑色素瘤患者在接受ICIs治疗前采集了基线肿瘤样本。对福尔马林固定石蜡包埋(FFPE)和新鲜冷冻(FF)肿瘤样本进行了大量RNA-seq分析。使用 ICIs 后的持久临床获益(主要临床终点)被定义为总生存期大于 24 个月且不改变主要用药方案(应答者)。我们使用{exotic}工具处理RNA-seq读数,仔细识别外源序列。71名转移性黑色素瘤患者的年龄从24岁到83岁不等,59%为男性,55%的患者在接受ICI治疗后存活时间超过24个月。肿瘤 RNA 序列中发现了外源分类群,包括细菌、真菌和病毒。我们发现免疫疗法应答肿瘤与非应答肿瘤的基因表达和微生物丰度存在差异。有反应者明显富集了噬菌体门中的噬菌体,而无反应者则富集了包括空肠弯曲杆菌在内的多种细菌。这些微生物与免疫相关基因表达特征相关。最后,我们发现利用微生物丰度和基因表达预测免疫疗法延长生存期的模型优于单独使用其中一种数据集的模型。我们的发现值得进一步研究,并有可能支持改变肿瘤微生物组以改善 ICIs 治疗效果的治疗策略。
{"title":"The Tumor Microbiome as a Predictor of Outcomes in Patients with Metastatic Melanoma Treated with Immune Checkpoint Inhibitors.","authors":"Caroline E Dravillas, Samuel S Coleman, Rebecca Hoyd, Griffin Caryotakis, Louis Denko, Carlos H F Chan, Michelle L Churchman, Nicholas Denko, Rebecca D Dodd, Islam Eljilany, Sheetal Hardikar, Marium Husain, Alexandra P Ikeguchi, Ning Jin, Qin Ma, Martin D McCarter, Afaf E G Osman, Lary A Robinson, Eric A Singer, Gabriel Tinoco, Cornelia M Ulrich, Yousef Zakharia, Daniel Spakowicz, Ahmad A Tarhini, Aik Choon Tan","doi":"10.1158/2767-9764.CRC-23-0170","DOIUrl":"10.1158/2767-9764.CRC-23-0170","url":null,"abstract":"<p><p>Emerging evidence supports the important role of the tumor microbiome in oncogenesis, cancer immune phenotype, cancer progression, and treatment outcomes in many malignancies. In this study, we investigated the metastatic melanoma tumor microbiome and its potential roles in association with clinical outcomes, such as survival, in patients with metastatic disease treated with immune checkpoint inhibitors (ICI). Baseline tumor samples were collected from 71 patients with metastatic melanoma before treatment with ICIs. Bulk RNA sequencing (RNA-seq) was conducted on the formalin-fixed, paraffin-embedded and fresh frozen tumor samples. Durable clinical benefit (primary clinical endpoint) following ICIs was defined as overall survival >24 months and no change to the primary drug regimen (responders). We processed RNA-seq reads to carefully identify exogenous sequences using the {exotic} tool. The age of the 71 patients with metastatic melanoma ranged from 24 to 83 years, 59% were male, and 55% survived >24 months following the initiation of ICI treatment. Exogenous taxa were identified in the tumor RNA-seq, including bacteria, fungi, and viruses. We found differences in gene expression and microbe abundances in immunotherapy-responsive versus nonresponsive tumors. Responders showed significant enrichment of bacteriophages in the phylum Uroviricota, and nonresponders showed enrichment of several bacteria, including Campylobacter jejuni. These microbes correlated with immune-related gene expression signatures. Finally, we found that models for predicting prolonged survival with immunotherapy using both microbe abundances and gene expression outperformed models using either dataset alone. Our findings warrant further investigation and potentially support therapeutic strategies to modify the tumor microbiome in order to improve treatment outcomes with ICIs.</p><p><strong>Significance: </strong>We analyzed the tumor microbiome and interactions with genes and pathways in metastatic melanoma treated with immunotherapy and identified several microbes associated with immunotherapy response and immune-related gene expression signatures. Machine learning models that combined microbe abundances and gene expression outperformed models using either dataset alone in predicting immunotherapy responses.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11307144/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141629419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Estrogen Receptor Blockade Potentiates Immunotherapy for Liver Metastases by Altering the Liver Immunosuppressive Microenvironment. 雌激素受体阻断剂通过改变肝脏免疫抑制微环境,增强肝转移瘤的免疫疗法。
IF 2 Q3 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2767-9764.CRC-24-0196
Yasmine Benslimane, Kevin Amalfi, Sara Lapin, Stephanie Perrino, Pnina Brodt

Liver metastases (LM) remain a major cause of cancer-related death and are a major clinical challenge. LM and the female sex are predictors of a poorer response to immunotherapy but the underlying mechanisms remain unclear. We previously reported on a sexual dimorphism in the control of the tumor microenvironment (TME) of colorectal carcinoma liver metastases (CRCLM) and identified estrogen as a regulator of an immunosuppressive TME in the liver. Here we aimed to assess the effect of estrogen deprivation on the cytokine/chemokine profile associated with CRCLM, using a multiplex cytokine array and the RNAscope technology, and its effects on the innate and adaptive immune responses in the liver. We also evaluated the benefit of combining the selective estrogen-receptor degrader Fulvestrant with immune checkpoint blockade for the treatment of CRCLM. We show that estrogen depletion altered the cytokine/chemokine repertoire of the liver, decreased macrophage polarization, as reflected in reduced accumulation of tumor infiltrating M2 macrophages and increased the accumulation of CCL5+/CCR5+ CD8+ T and NKT cells in the liver TME. Similar results were obtained in a murine pancreatic ductal adenocarcinoma model. Importantly, treatment with Fulvestrant also increased the accumulation of CD8+CCL5+, CD8+CCR5+ T and NK cells in the liver TME and enhanced the therapeutic benefit of anti-PD1 immunotherapy, resulting in a significant reduction in the outgrowth of LM. Taken together, our results show that estrogen regulates immune cell recruitment to the liver and suggest that inhibition of estrogen action could potentiate the tumor-inhibitory effect of immunotherapy in hormone-independent and immunotherapy-resistant metastatic cancer.

Significance: The immune microenvironment of the liver plays a major role in controlling the expansion of hepatic metastases and is regulated by estrogen. We show that treatment of tumor-bearing mice with an estrogen receptor degrader potentiated an anti-metastatic effect of immunotherapy. Our results provide mechanistic insight into clinical findings and a rationale for evaluating the efficacy of combination anti-estrogen and immunotherapy for prevention and/or treatment of hepatic metastases in female patients.

肝转移(LM)仍然是癌症相关死亡的主要原因之一,也是一项重大的临床挑战。肝转移和女性性别是免疫疗法反应较差的预测因素,但其潜在机制仍不清楚。我们以前曾报道过 CRC 肝转移瘤(CRCLM)肿瘤微环境(TME)控制中的性别二态性,并确定雌激素是肝脏中免疫抑制性 TME 的调节剂。在这里,我们旨在利用多重细胞因子阵列和 RNAscope 技术评估雌激素剥夺对与 CRCLM 相关的细胞因子/趋化因子谱的影响,及其对肝脏中先天性和适应性免疫反应的影响。我们还评估了选择性雌激素受体降解剂氟维司群与免疫检查点阻断相结合治疗 CRCLM 的益处。我们发现,雌激素降解改变了肝脏的细胞因子/趋化因子谱系,降低了巨噬细胞的极化,这反映在肿瘤浸润的 M2 巨噬细胞的聚集减少,并增加了肝脏 TME 中 CCL5+/CCR5+ CD8+ T 细胞和 NKT 细胞的聚集。在小鼠胰腺导管腺癌模型中也获得了类似的结果。重要的是,氟维司群还能增加肝脏TME中CD8+CCL5+、CD8+CCR5+ T细胞和NK细胞的积累,增强抗PD1免疫疗法的治疗效果,从而显著减少LM的生长。综上所述,我们的研究结果表明,雌激素可调控免疫细胞向肝脏的招募,并提示抑制雌激素的作用可增强免疫疗法对激素依赖性和IT耐药转移性癌症的抑瘤效果。
{"title":"Estrogen Receptor Blockade Potentiates Immunotherapy for Liver Metastases by Altering the Liver Immunosuppressive Microenvironment.","authors":"Yasmine Benslimane, Kevin Amalfi, Sara Lapin, Stephanie Perrino, Pnina Brodt","doi":"10.1158/2767-9764.CRC-24-0196","DOIUrl":"10.1158/2767-9764.CRC-24-0196","url":null,"abstract":"<p><p>Liver metastases (LM) remain a major cause of cancer-related death and are a major clinical challenge. LM and the female sex are predictors of a poorer response to immunotherapy but the underlying mechanisms remain unclear. We previously reported on a sexual dimorphism in the control of the tumor microenvironment (TME) of colorectal carcinoma liver metastases (CRCLM) and identified estrogen as a regulator of an immunosuppressive TME in the liver. Here we aimed to assess the effect of estrogen deprivation on the cytokine/chemokine profile associated with CRCLM, using a multiplex cytokine array and the RNAscope technology, and its effects on the innate and adaptive immune responses in the liver. We also evaluated the benefit of combining the selective estrogen-receptor degrader Fulvestrant with immune checkpoint blockade for the treatment of CRCLM. We show that estrogen depletion altered the cytokine/chemokine repertoire of the liver, decreased macrophage polarization, as reflected in reduced accumulation of tumor infiltrating M2 macrophages and increased the accumulation of CCL5+/CCR5+ CD8+ T and NKT cells in the liver TME. Similar results were obtained in a murine pancreatic ductal adenocarcinoma model. Importantly, treatment with Fulvestrant also increased the accumulation of CD8+CCL5+, CD8+CCR5+ T and NK cells in the liver TME and enhanced the therapeutic benefit of anti-PD1 immunotherapy, resulting in a significant reduction in the outgrowth of LM. Taken together, our results show that estrogen regulates immune cell recruitment to the liver and suggest that inhibition of estrogen action could potentiate the tumor-inhibitory effect of immunotherapy in hormone-independent and immunotherapy-resistant metastatic cancer.</p><p><strong>Significance: </strong>The immune microenvironment of the liver plays a major role in controlling the expansion of hepatic metastases and is regulated by estrogen. We show that treatment of tumor-bearing mice with an estrogen receptor degrader potentiated an anti-metastatic effect of immunotherapy. Our results provide mechanistic insight into clinical findings and a rationale for evaluating the efficacy of combination anti-estrogen and immunotherapy for prevention and/or treatment of hepatic metastases in female patients.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11306998/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141617710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mathematical Modeling of Tumor Growth in Preclinical Mouse Models with Applications in Biomarker Discovery and Drug Mechanism Studies. 临床前小鼠模型中肿瘤生长的数学建模及其在生物标记物发现和药物机制研究中的应用。
IF 2 Q3 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2767-9764.CRC-24-0059
Huajun Zhou, Binchen Mao, Sheng Guo

Oncology drug efficacy is evaluated in mouse models by continuously monitoring tumor volumes, which can be mathematically described by growth kinetic models. Although past studies have investigated various growth models, their reliance on small datasets raises concerns about whether their findings are truly representative of tumor growth in diverse mouse models under different vehicle or drug treatments. In this study, we systematically evaluated six parametric models (exponential, exponential quadratic, monomolecular, logistic, Gompertz, and von Bertalanffy) and the semiparametric generalized additive model (GAM) on fitting tumor volume data from more than 30,000 mice in 930 experiments conducted in patient-derived xenografts, cell line-derived xenografts, and syngeneic models. We found that the exponential quadratic model is the best parametric model and can adequately model 87% studies, higher than other models including von Bertalanffy (82%) and Gompertz (80%) models; the latter is often considered the standard growth model. At the mouse group level, 7.5% of growth data could not be fit by any parametric model and were fitted by GAM. We show that endpoint gain integrated in time, a GAM-derived efficacy metric, is equivalent to exponential growth rate, a metric we previously proposed and conveniently calculated by simple algebra. Using five studies on paclitaxel, anti-PD1 antibody, cetuximab, irinotecan, and sorafenib, we showed that exponential and exponential quadratic models achieve similar performance in uncovering drug mechanism and biomarkers. We also compared exponential growth rate-based association analysis and exponential modeling approach in biomarker discovery and found that they complement each other. Modeling methods herein are implemented in an open-source R package freely available at https://github.com/hjzhou988/TuGroMix.

Significance: We present a general strategy for mathematically modeling tumor growth in mouse models using data from 30,000 mice and show that modeling and nonmodeling approaches are complementary in biomarker discovery and drug mechanism studies.

肿瘤药物疗效是通过连续监测肿瘤体积在小鼠模型中进行评估的,肿瘤体积可以用生长动力学模型进行数学描述。过去的研究研究了各种生长模型,但它们对小数据集的依赖性使人担心它们的研究结果是否真正代表了不同载体或药物治疗下不同小鼠模型的肿瘤生长情况。在此,我们系统地评估了六个参数模型(指数模型、指数二次模型、单分子模型、逻辑模型、贡珀茨模型和冯-贝塔朗菲模型)和半参数广义相加模型(GAM),这些模型拟合了在患者衍生异种移植、细胞系衍生异种移植和共生模型中进行的 930 次实验中 30,000 多只小鼠的肿瘤体积数据。我们发现,指数二次模型是最佳参数模型,能充分模拟 87% 的研究,高于其他模型,包括 von Bertalanffy 模型(82%)和 Gompertz 模型(80%),后者通常被认为是标准生长模型。在小鼠群体水平上,7.5%的生长数据无法用任何参数模型拟合,只能用 GAM 拟合。我们的研究表明,由 GAM 得出的疗效指标 eGaIT 等同于我们之前提出的指标 eGR,后者可以通过简单的代数方便地计算出来。通过对紫杉醇、抗 PD-1 抗体、西妥昔单抗、伊立替康和索拉非尼的五项研究,我们发现指数模型和指数二次模型在揭示药物机制和生物标记物方面具有相似的性能。我们还比较了生物标记物发现中基于 eGR 的关联分析和指数建模方法,发现它们可以互补。本文中的建模方法是在 https://github.com/hjzhou988/TuGroMix 免费提供的开源 R 软件包中实现的。
{"title":"Mathematical Modeling of Tumor Growth in Preclinical Mouse Models with Applications in Biomarker Discovery and Drug Mechanism Studies.","authors":"Huajun Zhou, Binchen Mao, Sheng Guo","doi":"10.1158/2767-9764.CRC-24-0059","DOIUrl":"10.1158/2767-9764.CRC-24-0059","url":null,"abstract":"<p><p>Oncology drug efficacy is evaluated in mouse models by continuously monitoring tumor volumes, which can be mathematically described by growth kinetic models. Although past studies have investigated various growth models, their reliance on small datasets raises concerns about whether their findings are truly representative of tumor growth in diverse mouse models under different vehicle or drug treatments. In this study, we systematically evaluated six parametric models (exponential, exponential quadratic, monomolecular, logistic, Gompertz, and von Bertalanffy) and the semiparametric generalized additive model (GAM) on fitting tumor volume data from more than 30,000 mice in 930 experiments conducted in patient-derived xenografts, cell line-derived xenografts, and syngeneic models. We found that the exponential quadratic model is the best parametric model and can adequately model 87% studies, higher than other models including von Bertalanffy (82%) and Gompertz (80%) models; the latter is often considered the standard growth model. At the mouse group level, 7.5% of growth data could not be fit by any parametric model and were fitted by GAM. We show that endpoint gain integrated in time, a GAM-derived efficacy metric, is equivalent to exponential growth rate, a metric we previously proposed and conveniently calculated by simple algebra. Using five studies on paclitaxel, anti-PD1 antibody, cetuximab, irinotecan, and sorafenib, we showed that exponential and exponential quadratic models achieve similar performance in uncovering drug mechanism and biomarkers. We also compared exponential growth rate-based association analysis and exponential modeling approach in biomarker discovery and found that they complement each other. Modeling methods herein are implemented in an open-source R package freely available at https://github.com/hjzhou988/TuGroMix.</p><p><strong>Significance: </strong>We present a general strategy for mathematically modeling tumor growth in mouse models using data from 30,000 mice and show that modeling and nonmodeling approaches are complementary in biomarker discovery and drug mechanism studies.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11360417/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141891150","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Tumor Microbiome Reacts to Hypoxia and Can Influence Response to Radiation Treatment in Colorectal Cancer. 肿瘤微生物群对缺氧有反应,并能影响结直肠癌患者对放射治疗的反应。
IF 2 Q3 ONCOLOGY Pub Date : 2024-07-01 DOI: 10.1158/2767-9764.CRC-23-0367
Martin Benej, Rebecca Hoyd, McKenzie Kreamer, Caroline E Wheeler, Dennis J Grencewicz, Fouad Choueiry, Carlos H F Chan, Yousef Zakharia, Qin Ma, Rebecca D Dodd, Cornelia M Ulrich, Sheetal Hardikar, Michelle L Churchman, Ahmad A Tarhini, Lary A Robinson, Eric A Singer, Alexandra P Ikeguchi, Martin D McCarter, Gabriel Tinoco, Marium Husain, Ning Jin, Aik C Tan, Afaf E G Osman, Islam Eljilany, Gregory Riedlinger, Bryan P Schneider, Katarina Benejova, Martin Kery, Ioanna Papandreou, Jiangjiang Zhu, Nicholas Denko, Daniel Spakowicz

Tumor hypoxia has been shown to predict poor patient outcomes in several cancer types, partially because it reduces radiation's ability to kill cells. We hypothesized that some of the clinical effects of hypoxia could also be due to its impact on the tumor microbiome. Therefore, we examined the RNA sequencing data from the Oncology Research Information Exchange Network database of patients with colorectal cancer treated with radiotherapy. We identified microbial RNAs for each tumor and related them to the hypoxic gene expression scores calculated from host mRNA. Our analysis showed that the hypoxia expression score predicted poor patient outcomes and identified tumors enriched with certain microbes such as Fusobacterium nucleatum. The presence of other microbes, such as Fusobacterium canifelinum, predicted poor patient outcomes, suggesting a potential interaction between hypoxia, the microbiome, and radiation response. To experimentally investigate this concept, we implanted CT26 colorectal cancer cells into immune-competent BALB/c and immune-deficient athymic nude mice. After growth, in which tumors passively acquired microbes from the gastrointestinal tract, we harvested tumors, extracted nucleic acids, and sequenced host and microbial RNAs. We stratified tumors based on their hypoxia score and performed a metatranscriptomic analysis of microbial gene expression. In addition to hypoxia-tropic and -phobic microbial populations, analysis of microbial gene expression at the strain level showed expression differences based on the hypoxia score. Thus, hypoxia gene expression scores seem to associate with different microbial populations and elicit an adaptive transcriptional response in intratumoral microbes, potentially influencing clinical outcomes.

Significance: Tumor hypoxia reduces radiotherapy efficacy. In this study, we explored whether some of the clinical effects of hypoxia could be due to interaction with the tumor microbiome. Hypoxic gene expression scores associated with certain microbes and elicited an adaptive transcriptional response in others that could contribute to poor clinical outcomes.

在几种癌症类型中,肿瘤缺氧已被证明可预测患者的不良预后,部分原因是缺氧会降低辐射杀死细胞的能力。我们假设,缺氧的一些临床影响也可能是由于它对肿瘤微生物组的影响。因此,我们研究了肿瘤研究信息交换网络(ORIEN)数据库中接受放疗的结直肠癌(CRC)患者的 RNA-seq 数据。我们确定了每个肿瘤的微生物 RNA,并将它们与根据宿主 mRNA 计算出的缺氧基因表达评分联系起来。我们的分析表明,缺氧基因表达得分预示着患者的不良预后,并确定了富含某些微生物(如核酸镰刀菌)的肿瘤。其他微生物(如镰刀菌)的存在也预示着患者的预后较差,这表明缺氧、微生物组和辐射反应之间可能存在相互作用。为了对这一概念进行实验研究,我们将 CT26 CRC 细胞植入免疫功能正常的 BALB/c 和免疫功能缺陷的无胸腺裸鼠体内。在肿瘤生长过程中,肿瘤被动地从胃肠道中获取微生物,之后我们收获肿瘤,提取核酸,并对宿主和微生物的 RNA 进行测序。我们根据肿瘤的缺氧评分对其进行分层,并对微生物基因表达进行了元转录组学分析。除了耐缺氧和怕缺氧的微生物种群外,菌株水平的微生物基因表达分析也显示了基于缺氧评分的表达差异。因此,缺氧似乎与不同的微生物群相关联,并引起瘤内微生物的适应性转录反应,从而可能影响临床结果。
{"title":"The Tumor Microbiome Reacts to Hypoxia and Can Influence Response to Radiation Treatment in Colorectal Cancer.","authors":"Martin Benej, Rebecca Hoyd, McKenzie Kreamer, Caroline E Wheeler, Dennis J Grencewicz, Fouad Choueiry, Carlos H F Chan, Yousef Zakharia, Qin Ma, Rebecca D Dodd, Cornelia M Ulrich, Sheetal Hardikar, Michelle L Churchman, Ahmad A Tarhini, Lary A Robinson, Eric A Singer, Alexandra P Ikeguchi, Martin D McCarter, Gabriel Tinoco, Marium Husain, Ning Jin, Aik C Tan, Afaf E G Osman, Islam Eljilany, Gregory Riedlinger, Bryan P Schneider, Katarina Benejova, Martin Kery, Ioanna Papandreou, Jiangjiang Zhu, Nicholas Denko, Daniel Spakowicz","doi":"10.1158/2767-9764.CRC-23-0367","DOIUrl":"10.1158/2767-9764.CRC-23-0367","url":null,"abstract":"<p><p>Tumor hypoxia has been shown to predict poor patient outcomes in several cancer types, partially because it reduces radiation's ability to kill cells. We hypothesized that some of the clinical effects of hypoxia could also be due to its impact on the tumor microbiome. Therefore, we examined the RNA sequencing data from the Oncology Research Information Exchange Network database of patients with colorectal cancer treated with radiotherapy. We identified microbial RNAs for each tumor and related them to the hypoxic gene expression scores calculated from host mRNA. Our analysis showed that the hypoxia expression score predicted poor patient outcomes and identified tumors enriched with certain microbes such as Fusobacterium nucleatum. The presence of other microbes, such as Fusobacterium canifelinum, predicted poor patient outcomes, suggesting a potential interaction between hypoxia, the microbiome, and radiation response. To experimentally investigate this concept, we implanted CT26 colorectal cancer cells into immune-competent BALB/c and immune-deficient athymic nude mice. After growth, in which tumors passively acquired microbes from the gastrointestinal tract, we harvested tumors, extracted nucleic acids, and sequenced host and microbial RNAs. We stratified tumors based on their hypoxia score and performed a metatranscriptomic analysis of microbial gene expression. In addition to hypoxia-tropic and -phobic microbial populations, analysis of microbial gene expression at the strain level showed expression differences based on the hypoxia score. Thus, hypoxia gene expression scores seem to associate with different microbial populations and elicit an adaptive transcriptional response in intratumoral microbes, potentially influencing clinical outcomes.</p><p><strong>Significance: </strong>Tumor hypoxia reduces radiotherapy efficacy. In this study, we explored whether some of the clinical effects of hypoxia could be due to interaction with the tumor microbiome. Hypoxic gene expression scores associated with certain microbes and elicited an adaptive transcriptional response in others that could contribute to poor clinical outcomes.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11234499/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141433488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Phase II Trial of the WEE1 Inhibitor Adavosertib in SETD2-Altered Advanced Solid Tumor Malignancies (NCI 10170). WEE1抑制剂Adavosertib治疗SETD2改变的晚期实体瘤恶性肿瘤的II期试验(NCI 10170)。
IF 2 Q3 ONCOLOGY Pub Date : 2024-07-01 DOI: 10.1158/2767-9764.CRC-24-0213
Edward Maldonado, W Kimryn Rathmell, Geoffrey I Shapiro, Naoko Takebe, Jordi Rodon, Devalingam Mahalingam, Nikolaos A Trikalinos, Arash R Kalebasty, Mamta Parikh, Scott A Boerner, Celene Balido, Gregor Krings, Timothy F Burns, Emily K Bergsland, Pamela N Munster, Alan Ashworth, Patricia LoRusso, Rahul R Aggarwal

We sought to evaluate the efficacy of WEE1 inhibitor adavosertib in patients with solid tumor malignancies (cohort A) and clear cell renal cell carcinoma (ccRCC; cohort B). NCT03284385 was a parallel cohort, Simon two-stage, phase II study of adavosertib (300 mg QDAY by mouth on days 1-5 and 8-12 of each 21-day cycle) in patients with solid tumor malignancies harboring a pathogenic SETD2 mutation. The primary endpoint was the objective response rate. Correlative assays evaluated the loss of H3K36me3 by IHC, a downstream consequence of SETD2 loss, in archival tumor tissue. Eighteen patients were enrolled (9/cohort). The median age was 60 years (range 45-74). The median duration of treatment was 1.28 months (range 0-24+). No objective responses were observed in either cohort; accrual was halted following stage 1. Minor tumor regressions were observed in 4/18 (22%) evaluable patients. Stable disease (SD) was the best overall response in 10/18 (56%) patients, including three patients with SD > 4 months. One patient with ccRCC remains on treatment for >24 months. The most common adverse events of any grade were nausea (59%), anemia (41%), diarrhea (41%), and neutropenia (41%). Nine patients (50%) experienced a Grade ≥3 adverse event. Of eight evaluable archival tissue samples, six (75%) had a loss of H3K36me3 by IHC. Adavosertib failed to exhibit objective responses in SETD2-altered ccRCC and other solid tumor malignancies although prolonged SD was observed in a subset of patients. Combination approaches may yield greater depth of tumor response.

Significance: WEE1 inhibition with adavosertib monotherapy demonstrated limited clinical activity in patients with SETD2-altered solid tumors despite compelling preclinical data indicating a synthetic lethal effect, which did not translate into robust tumor regression. Loss of the H3K36me3 trimethylation mark caused by SETD2-deficiency was confirmed in the majority of evaluable tumors. A subset of patients derived clinical benefit as manifested by minor tumor regressions and prolonged SD.

目的:评估WEE1抑制剂adavosertib在实体瘤恶性肿瘤(A组)和透明细胞肾细胞癌(ccRCC;B组)患者(pts)中的疗效:NCT03284385是一项平行队列、西蒙两阶段II期研究,针对携带致病性SETD2突变的实体瘤恶性肿瘤患者进行阿达韦塞替布治疗(每21天周期的第1-5天和第8-12天口服300毫克QDAY)。主要终点是客观反应率(ORR)。相关检测通过免疫组化(IHC)评估了档案肿瘤组织中H3K36me3的缺失情况,这是SETD2缺失的下游结果:18名患者入组(9人/组)。中位年龄为 60 岁(45 - 74 岁不等)。中位治疗时间为 1.28 个月(0 - 24 个月)。两组患者均未观察到客观反应;第一阶段结束后,招募工作停止。在 4/18(22%)名可评估患者中观察到轻微的肿瘤消退。10/18例(56%)患者的最佳总体反应是病情稳定(SD),其中3例患者病情稳定时间超过4个月。一名ccRCC患者的治疗时间超过24个月。最常见的任何级别的不良事件(AE)是恶心(59%)、贫血(41%)、腹泻(41%)和中性粒细胞减少(41%)。9名患者(50%)发生了≥3级的不良反应。在8份可评估的存档组织样本中,6份(75%)经IHC检测出现H3K36me3缺失:阿达伐他替尼未能对SETD2改变的ccRCC和其他实体瘤恶性肿瘤产生客观反应,但在一部分患者中观察到了长期稳定的疾病。联合疗法可能会产生更深层次的肿瘤反应。
{"title":"A Phase II Trial of the WEE1 Inhibitor Adavosertib in SETD2-Altered Advanced Solid Tumor Malignancies (NCI 10170).","authors":"Edward Maldonado, W Kimryn Rathmell, Geoffrey I Shapiro, Naoko Takebe, Jordi Rodon, Devalingam Mahalingam, Nikolaos A Trikalinos, Arash R Kalebasty, Mamta Parikh, Scott A Boerner, Celene Balido, Gregor Krings, Timothy F Burns, Emily K Bergsland, Pamela N Munster, Alan Ashworth, Patricia LoRusso, Rahul R Aggarwal","doi":"10.1158/2767-9764.CRC-24-0213","DOIUrl":"10.1158/2767-9764.CRC-24-0213","url":null,"abstract":"<p><p>We sought to evaluate the efficacy of WEE1 inhibitor adavosertib in patients with solid tumor malignancies (cohort A) and clear cell renal cell carcinoma (ccRCC; cohort B). NCT03284385 was a parallel cohort, Simon two-stage, phase II study of adavosertib (300 mg QDAY by mouth on days 1-5 and 8-12 of each 21-day cycle) in patients with solid tumor malignancies harboring a pathogenic SETD2 mutation. The primary endpoint was the objective response rate. Correlative assays evaluated the loss of H3K36me3 by IHC, a downstream consequence of SETD2 loss, in archival tumor tissue. Eighteen patients were enrolled (9/cohort). The median age was 60 years (range 45-74). The median duration of treatment was 1.28 months (range 0-24+). No objective responses were observed in either cohort; accrual was halted following stage 1. Minor tumor regressions were observed in 4/18 (22%) evaluable patients. Stable disease (SD) was the best overall response in 10/18 (56%) patients, including three patients with SD > 4 months. One patient with ccRCC remains on treatment for >24 months. The most common adverse events of any grade were nausea (59%), anemia (41%), diarrhea (41%), and neutropenia (41%). Nine patients (50%) experienced a Grade ≥3 adverse event. Of eight evaluable archival tissue samples, six (75%) had a loss of H3K36me3 by IHC. Adavosertib failed to exhibit objective responses in SETD2-altered ccRCC and other solid tumor malignancies although prolonged SD was observed in a subset of patients. Combination approaches may yield greater depth of tumor response.</p><p><strong>Significance: </strong>WEE1 inhibition with adavosertib monotherapy demonstrated limited clinical activity in patients with SETD2-altered solid tumors despite compelling preclinical data indicating a synthetic lethal effect, which did not translate into robust tumor regression. Loss of the H3K36me3 trimethylation mark caused by SETD2-deficiency was confirmed in the majority of evaluable tumors. A subset of patients derived clinical benefit as manifested by minor tumor regressions and prolonged SD.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141452259","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stratification to Neoadjuvant Radiotherapy in Rectal Cancer by Regimen and Transcriptional Signatures. 根据治疗方案和转录信号对直肠癌新辅助放疗进行分层
IF 2 Q3 ONCOLOGY Pub Date : 2024-07-01 DOI: 10.1158/2767-9764.CRC-23-0502
Umair Mahmood, Andrew Blake, Sanjay Rathee, Leslie Samuel, Graeme Murray, David Sebag-Montefiore, Simon Gollins, Nicholas P West, Rubina Begum, Simon P Bach, Susan D Richman, Phil Quirke, Keara L Redmond, Manuel Salto-Tellez, Viktor H Koelzer, Simon J Leedham, Ian Tomlinson, Philip D Dunne, Francesca M Buffa, Tim S Maughan, Enric Domingo

Response to neoadjuvant radiotherapy (RT) in rectal cancer has been associated with immune and stromal features that are captured by transcriptional signatures. However, how such associations perform across different chemoradiotherapy regimens and within individual consensus molecular subtypes (CMS) and how they affect survival remain unclear. In this study, gene expression and clinical data of pretreatment biopsies from nine cohorts of primary rectal tumors were combined (N = 826). Exploratory analyses were done with transcriptomic signatures for the endpoint of pathologic complete response (pCR), considering treatment regimen or CMS subtype. Relevant findings were tested for overall survival and recurrence-free survival. Immune and stromal signatures were strongly associated with pCR and lack of pCR, respectively, in RT and capecitabine (Cap)/5-fluorouracil (5FU)-treated patients (N = 387), in which the radiosensitivity signature (RSS) showed the strongest association. Upon addition of oxaliplatin (Ox; N = 123), stromal signatures switched direction and showed higher chances to achieve pCR than without Ox (p for interaction 0.02). Among Cap/5FU patients, most signatures performed similarly across CMS subtypes, except cytotoxic lymphocytes that were associated with pCR in CMS1 and CMS4 cases compared with other CMS subtypes (p for interaction 0.04). The only variables associated with survival were pCR and RSS. Although the frequency of pCR across different chemoradiation regimens is relatively similar, our data suggest that response rates may differ depending on the biological landscape of rectal cancer. Response to neoadjuvant RT in stroma-rich tumors may potentially be improved by the addition of Ox. RSS in preoperative biopsies provides predictive information for response specifically to neoadjuvant RT with 5FU.

Significance: Rectal cancers with stromal features may respond better to RT and 5FU/Cap with the addition of Ox. Within patients not treated with Ox, high levels of cytotoxic lymphocytes associate with response only in immune and stromal tumors. Our analyses provide biological insights about the outcome by different radiotherapy regimens in rectal cancer.

直肠癌对新辅助放疗(RT)的反应与转录特征捕捉到的免疫和基质特征有关。然而,这种关联在不同的化放疗方案和个体共识分子亚型(CMS)中的表现如何,以及它们如何影响生存率仍不清楚。在这项研究中,合并了来自九个原发性直肠肿瘤队列的预处理活检的基因表达和临床数据(N = 826)。在考虑治疗方案或CMS亚型的情况下,对病理完全反应(pCR)终点的转录组特征进行了探索性分析。对总生存期和无复发生存期的相关结果进行了检验。在 RT 和卡培他滨(Cap)/5-氟尿嘧啶(5FU)治疗的患者(N = 387)中,免疫和基质特征分别与病理完全应答和无病理完全应答密切相关,其中放射敏感性特征(RSS)显示出最强的关联性。添加奥沙利铂(Ox;N = 123)后,基质特征改变了方向,与未添加奥沙利铂时相比,获得 pCR 的几率更高(交互作用 p 为 0.02)。在Cap/5FU患者中,除了细胞毒性淋巴细胞与其他CMS亚型相比与CMS1和CMS4病例的pCR相关(交互作用的p为0.04)外,大多数CMS亚型的特征表现相似。唯一与生存率相关的变量是 pCR 和 RSS。虽然不同化疗方案的 pCR 频率相对相似,但我们的数据表明,反应率可能因直肠癌的生物学特征而异。对富含基质的肿瘤进行新辅助 RT 有助于改善对 Ox 的反应。术前活检中的RSS提供了对5FU新辅助RT反应的预测信息:具有基质特征的直肠癌在加用 Ox 后可能会对 RT 和 5FU/Cap 产生更好的反应。在未接受 Ox 治疗的患者中,高水平的细胞毒性淋巴细胞仅与免疫性肿瘤和间质肿瘤的反应有关。我们的分析为直肠癌不同放疗方案的疗效提供了生物学启示。
{"title":"Stratification to Neoadjuvant Radiotherapy in Rectal Cancer by Regimen and Transcriptional Signatures.","authors":"Umair Mahmood, Andrew Blake, Sanjay Rathee, Leslie Samuel, Graeme Murray, David Sebag-Montefiore, Simon Gollins, Nicholas P West, Rubina Begum, Simon P Bach, Susan D Richman, Phil Quirke, Keara L Redmond, Manuel Salto-Tellez, Viktor H Koelzer, Simon J Leedham, Ian Tomlinson, Philip D Dunne, Francesca M Buffa, Tim S Maughan, Enric Domingo","doi":"10.1158/2767-9764.CRC-23-0502","DOIUrl":"10.1158/2767-9764.CRC-23-0502","url":null,"abstract":"<p><p>Response to neoadjuvant radiotherapy (RT) in rectal cancer has been associated with immune and stromal features that are captured by transcriptional signatures. However, how such associations perform across different chemoradiotherapy regimens and within individual consensus molecular subtypes (CMS) and how they affect survival remain unclear. In this study, gene expression and clinical data of pretreatment biopsies from nine cohorts of primary rectal tumors were combined (N = 826). Exploratory analyses were done with transcriptomic signatures for the endpoint of pathologic complete response (pCR), considering treatment regimen or CMS subtype. Relevant findings were tested for overall survival and recurrence-free survival. Immune and stromal signatures were strongly associated with pCR and lack of pCR, respectively, in RT and capecitabine (Cap)/5-fluorouracil (5FU)-treated patients (N = 387), in which the radiosensitivity signature (RSS) showed the strongest association. Upon addition of oxaliplatin (Ox; N = 123), stromal signatures switched direction and showed higher chances to achieve pCR than without Ox (p for interaction 0.02). Among Cap/5FU patients, most signatures performed similarly across CMS subtypes, except cytotoxic lymphocytes that were associated with pCR in CMS1 and CMS4 cases compared with other CMS subtypes (p for interaction 0.04). The only variables associated with survival were pCR and RSS. Although the frequency of pCR across different chemoradiation regimens is relatively similar, our data suggest that response rates may differ depending on the biological landscape of rectal cancer. Response to neoadjuvant RT in stroma-rich tumors may potentially be improved by the addition of Ox. RSS in preoperative biopsies provides predictive information for response specifically to neoadjuvant RT with 5FU.</p><p><strong>Significance: </strong>Rectal cancers with stromal features may respond better to RT and 5FU/Cap with the addition of Ox. Within patients not treated with Ox, high levels of cytotoxic lymphocytes associate with response only in immune and stromal tumors. Our analyses provide biological insights about the outcome by different radiotherapy regimens in rectal cancer.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11257085/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141635975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic Induction of Cancer-Testis Antigens and Endogenous Retroviruses at Single-Cell Level Enhances Immune Recognition and Response in Glioma. 在单细胞水平对癌症睾丸抗原和内源性逆转录病毒进行表观遗传诱导,可增强胶质瘤的免疫识别和反应。
IF 2 Q3 ONCOLOGY Pub Date : 2024-07-01 DOI: 10.1158/2767-9764.CRC-23-0566
Thomas J Lai, Lu Sun, Kevin Li, Terry J Prins, Janet Treger, Tie Li, Matthew Z Sun, David A Nathanson, Linda M Liau, Albert Lai, Robert M Prins, Richard G Everson

Glioblastoma (GBM) is the most common malignant primary brain tumor and remains incurable. Previous work has shown that systemic administration of Decitabine (DAC) induces sufficient expression of cancer-testis antigens (CTA) in GBM for targeting by adoptive T-cell therapy in vivo. However, the mechanisms by which DAC enhances immunogenicity in GBM remain to be elucidated. Using New York esophageal squamous cell carcinoma 1 (NY-ESO-1) as a representative inducible CTA, we demonstrate in patient tissue, immortalized glioma cells, and primary patient-derived gliomaspheres that basal CTA expression is restricted by promoter hypermethylation in gliomas. DAC treatment of glioma cells specifically inhibits DNA methylation silencing to render NY-ESO-1 and other CTA into inducible tumor antigens at single-cell resolution. Functionally, NY-ESO-1 T-cell receptor-engineered effector cell targeting of DAC-induced antigen in primary glioma cells promotes specific and polyfunctional T-cell cytokine profiles. In addition to induction of CTA, DAC concomitantly reactivates tumor-intrinsic human endogenous retroviruses, interferon response signatures, and MHC-I. Overall, we demonstrate that DAC induces targetable tumor antigen and enhances T-cell functionality against GBM, ultimately contributing to the improvement of targeted immune therapies in glioma.

Significance: This study dissects the tumor-intrinsic epigenetic and transcriptional mechanisms underlying enhanced T-cell functionality targeting decitabine-induced cancer-testis antigens in glioma. Our findings demonstrate concomitant induction of tumor antigens, reactivation of human endogenous retroviruses, and stimulation of interferon signaling as a mechanistic rationale to epigenetically prime human gliomas to immunotherapeutic targeting.

胶质母细胞瘤(GBM)是最常见的恶性原发性脑肿瘤,至今仍无法治愈。先前的研究表明,全身给药地西他滨(DAC)可诱导 GBM 中的癌-睾丸抗原(CTA)充分表达,从而使体内的采用性 T 细胞疗法具有靶向性。然而,DAC增强GBM免疫原性的机制仍有待阐明。我们使用 NY-ESO-1 作为诱导型 CTA 的代表,在患者组织、永生化胶质瘤细胞和原发性患者来源胶质瘤球中证明,胶质瘤中 CTA 的基础表达受到启动子超甲基化的限制。对胶质瘤细胞进行 DAC 处理可特异性抑制 DNA 甲基化沉默,从而使 NY-ESO-1 和其他 CTA 在单细胞分辨率下成为可诱导的肿瘤抗原。在功能上,NY-ESO-1 TCR 工程效应细胞靶向原发性胶质瘤细胞中 DAC 诱导的抗原,可促进特异性和多功能 T 细胞细胞因子谱。除了诱导 CTA 外,DAC 还能同时重新激活肿瘤内在人类内源性逆转录病毒、干扰素反应特征和 MHC-I。总之,我们证明了 DAC 能诱导可靶向的肿瘤抗原并增强 T 细胞对抗 GBM 的功能,最终有助于改善胶质瘤的靶向免疫疗法。
{"title":"Epigenetic Induction of Cancer-Testis Antigens and Endogenous Retroviruses at Single-Cell Level Enhances Immune Recognition and Response in Glioma.","authors":"Thomas J Lai, Lu Sun, Kevin Li, Terry J Prins, Janet Treger, Tie Li, Matthew Z Sun, David A Nathanson, Linda M Liau, Albert Lai, Robert M Prins, Richard G Everson","doi":"10.1158/2767-9764.CRC-23-0566","DOIUrl":"10.1158/2767-9764.CRC-23-0566","url":null,"abstract":"<p><p>Glioblastoma (GBM) is the most common malignant primary brain tumor and remains incurable. Previous work has shown that systemic administration of Decitabine (DAC) induces sufficient expression of cancer-testis antigens (CTA) in GBM for targeting by adoptive T-cell therapy in vivo. However, the mechanisms by which DAC enhances immunogenicity in GBM remain to be elucidated. Using New York esophageal squamous cell carcinoma 1 (NY-ESO-1) as a representative inducible CTA, we demonstrate in patient tissue, immortalized glioma cells, and primary patient-derived gliomaspheres that basal CTA expression is restricted by promoter hypermethylation in gliomas. DAC treatment of glioma cells specifically inhibits DNA methylation silencing to render NY-ESO-1 and other CTA into inducible tumor antigens at single-cell resolution. Functionally, NY-ESO-1 T-cell receptor-engineered effector cell targeting of DAC-induced antigen in primary glioma cells promotes specific and polyfunctional T-cell cytokine profiles. In addition to induction of CTA, DAC concomitantly reactivates tumor-intrinsic human endogenous retroviruses, interferon response signatures, and MHC-I. Overall, we demonstrate that DAC induces targetable tumor antigen and enhances T-cell functionality against GBM, ultimately contributing to the improvement of targeted immune therapies in glioma.</p><p><strong>Significance: </strong>This study dissects the tumor-intrinsic epigenetic and transcriptional mechanisms underlying enhanced T-cell functionality targeting decitabine-induced cancer-testis antigens in glioma. Our findings demonstrate concomitant induction of tumor antigens, reactivation of human endogenous retroviruses, and stimulation of interferon signaling as a mechanistic rationale to epigenetically prime human gliomas to immunotherapeutic targeting.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11275559/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141297488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
E2F1-Associated Purine Synthesis Pathway Is a Major Component of the MET-DNA Damage Response Network. E2F1相关的嘌呤合成途径是MET-DNA损伤反应网络的主要组成部分。
IF 2 Q3 ONCOLOGY Pub Date : 2024-07-01 DOI: 10.1158/2767-9764.CRC-23-0370
Michaela Poliaková Turan, Rahel Riedo, Matúš Medo, Chiara Pozzato, Manja Friese-Hamim, Jonas P Koch, Si'Ana A Coggins, Qun Li, Baek Kim, Joachim Albers, Daniel M Aebersold, Nicola Zamboni, Yitzhak Zimmer, Michaela Medová

Various lines of investigation support a signaling interphase shared by receptor tyrosine kinases and the DNA damage response. However, the underlying network nodes and their contribution to the maintenance of DNA integrity remain unknown. We explored MET-related metabolic pathways in which interruption compromises proper resolution of DNA damage. Discovery metabolomics combined with transcriptomics identified changes in pathways relevant to DNA repair following MET inhibition (METi). METi by tepotinib was associated with the formation of γH2AX foci and with significant alterations in major metabolic circuits such as glycolysis, gluconeogenesis, and purine, pyrimidine, amino acid, and lipid metabolism. 5'-Phosphoribosyl-N-formylglycinamide, a de novo purine synthesis pathway metabolite, was consistently decreased in in vitro and in vivo MET-dependent models, and METi-related depletion of dNTPs was observed. METi instigated the downregulation of critical purine synthesis enzymes including phosphoribosylglycinamide formyltransferase, which catalyzes 5'-phosphoribosyl-N-formylglycinamide synthesis. Genes encoding these enzymes are regulated through E2F1, whose levels decrease upon METi in MET-driven cells and xenografts. Transient E2F1 overexpression prevented dNTP depletion and the concomitant METi-associated DNA damage in MET-driven cells. We conclude that DNA damage following METi results from dNTP reduction via downregulation of E2F1 and a consequent decline of de novo purine synthesis.

Significance: Maintenance of genome stability prevents disease and affiliates with growth factor receptor tyrosine kinases. We identified de novo purine synthesis as a pathway in which key enzymatic players are regulated through MET receptor and whose depletion via MET targeting explains MET inhibition-associated formation of DNA double-strand breaks. The mechanistic importance of MET inhibition-dependent E2F1 downregulation for interference with DNA integrity has translational implications for MET-targeting-based treatment of malignancies.

各种研究都支持受体酪氨酸激酶和 DNA 损伤反应共享信号间期。然而,潜在的网络节点及其对维护 DNA 完整性的贡献仍然未知。我们探索了与 MET 相关的代谢通路,这些通路的中断会影响 DNA 损伤的适当解决。发现代谢组学与转录组学相结合,确定了MET抑制(METi)后DNA修复相关通路的变化。特博替尼的 METi 与 γH2AX 病灶的形成以及糖酵解、糖代谢、嘌呤、嘧啶、氨基酸和脂质代谢等主要代谢回路的显著改变有关。5'-磷酸核糖基-N-甲酰基甘氨酰胺(FGAR)是一种嘌呤合成途径的新代谢物,在体外和体内 MET 依赖性模型中持续减少,并观察到与 METi 相关的 dNTPs 消耗。METi 导致关键的嘌呤合成酶下调,包括催化 FGAR 合成的磷酸核糖基甘氨酰胺甲酰转移酶(GART)。编码这些酶的基因通过 E2F1 调节,而在 METi 驱动的细胞和异种移植中,E2F1 的水平会随着 METi 而降低。在MET驱动的细胞中,瞬时E2F1过表达可防止dNTPs耗竭和随之而来的METi相关DNA损伤。我们的结论是,METi 导致的 DNA 损伤是通过下调 E2F1 导致的 dNTPs 减少以及随之而来的新嘌呤合成下降造成的。
{"title":"E2F1-Associated Purine Synthesis Pathway Is a Major Component of the MET-DNA Damage Response Network.","authors":"Michaela Poliaková Turan, Rahel Riedo, Matúš Medo, Chiara Pozzato, Manja Friese-Hamim, Jonas P Koch, Si'Ana A Coggins, Qun Li, Baek Kim, Joachim Albers, Daniel M Aebersold, Nicola Zamboni, Yitzhak Zimmer, Michaela Medová","doi":"10.1158/2767-9764.CRC-23-0370","DOIUrl":"10.1158/2767-9764.CRC-23-0370","url":null,"abstract":"<p><p>Various lines of investigation support a signaling interphase shared by receptor tyrosine kinases and the DNA damage response. However, the underlying network nodes and their contribution to the maintenance of DNA integrity remain unknown. We explored MET-related metabolic pathways in which interruption compromises proper resolution of DNA damage. Discovery metabolomics combined with transcriptomics identified changes in pathways relevant to DNA repair following MET inhibition (METi). METi by tepotinib was associated with the formation of γH2AX foci and with significant alterations in major metabolic circuits such as glycolysis, gluconeogenesis, and purine, pyrimidine, amino acid, and lipid metabolism. 5'-Phosphoribosyl-N-formylglycinamide, a de novo purine synthesis pathway metabolite, was consistently decreased in in vitro and in vivo MET-dependent models, and METi-related depletion of dNTPs was observed. METi instigated the downregulation of critical purine synthesis enzymes including phosphoribosylglycinamide formyltransferase, which catalyzes 5'-phosphoribosyl-N-formylglycinamide synthesis. Genes encoding these enzymes are regulated through E2F1, whose levels decrease upon METi in MET-driven cells and xenografts. Transient E2F1 overexpression prevented dNTP depletion and the concomitant METi-associated DNA damage in MET-driven cells. We conclude that DNA damage following METi results from dNTP reduction via downregulation of E2F1 and a consequent decline of de novo purine synthesis.</p><p><strong>Significance: </strong>Maintenance of genome stability prevents disease and affiliates with growth factor receptor tyrosine kinases. We identified de novo purine synthesis as a pathway in which key enzymatic players are regulated through MET receptor and whose depletion via MET targeting explains MET inhibition-associated formation of DNA double-strand breaks. The mechanistic importance of MET inhibition-dependent E2F1 downregulation for interference with DNA integrity has translational implications for MET-targeting-based treatment of malignancies.</p>","PeriodicalId":72516,"journal":{"name":"Cancer research communications","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11288008/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141494445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer research communications
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1